WO2016079502A1 - Metal chelating compounds for use in treating diseases - Google Patents

Metal chelating compounds for use in treating diseases Download PDF

Info

Publication number
WO2016079502A1
WO2016079502A1 PCT/GB2015/053492 GB2015053492W WO2016079502A1 WO 2016079502 A1 WO2016079502 A1 WO 2016079502A1 GB 2015053492 W GB2015053492 W GB 2015053492W WO 2016079502 A1 WO2016079502 A1 WO 2016079502A1
Authority
WO
WIPO (PCT)
Prior art keywords
heterocycle
group
compound
hydroxypyridin
aryl
Prior art date
Application number
PCT/GB2015/053492
Other languages
French (fr)
Inventor
David Tetard
Francis William LEWIS
Original Assignee
University Of Northumbria
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 Of Northumbria filed Critical University Of Northumbria
Publication of WO2016079502A1 publication Critical patent/WO2016079502A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/89Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members with hetero atoms directly attached to the ring nitrogen atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates to heterocyclic compounds for use in the treatment of diseases.
  • the invention relates to heterocyclic compounds for use in the treatment of neurodegenerative diseases, such as Parkinson's or Alzheimer's disease.
  • Neurodegeneration is the progressive loss of structure or function, and in some cases death, of neurons responsible for processing and transmitting information in the human body through chemical or electrical signals. As a consequence of such progressive loss and death of neurons, diseases such as Parkinson's and Alzheimer's may result.
  • a common cause of neurodegenerative diseases is disregulation and subsequent accumulation of metals in certain regions of the brain, in particular iron(III) but also copper and zinc, which in excess cause oxidative damage to neurons. While the origins are not fully understood, metal accumulation is due to metal dyshomeostasis, i.e. a breakdown in the mechanism through which the body regulates metal ion concentrations. As a downstream consequence of metal disregulation, displaced metals detrimentally interact with proteins present in the brain, for example, beta amyloid in the case of Alzheimer's disease and tau protein and alpha-synuclein in the case of Parkinson's disease. Accordingly, excessive metal accumulation causes increased interactions with these proteins, leading to accumulation of protein-metal aggregates which are one of the causes of nerve cell death and, therefore, neurodegeneration.
  • Parkinson patients Following the onset of neurodegenerative diseases such as Parkinson's or Alzheimer's, steps are taken to manage the disease, thereby providing symptomatic relief through the use of pharmaceutical drugs.
  • surgical procedures may be offered to Parkinson patients including deep brain stimulation, in which a brain pacemaker is implanted to send electrical impulses to specific parts of the brain to control symptoms, or lesional surgery, which involves the intentional creation of lesions to suppress over activity of specific subcortical areas.
  • symptomatic treatment is largely ineffective for Alzheimer's patients and so in most situations palliative care is provided.
  • metal chelation may be used to reduce excessive metal accumulation in regions of the brain, thereby reducing neurodegeneration and so delaying or halting the progression of neurodegenerative diseases.
  • 3-hydroxypyridin-4-one based compounds has been proposed for use as iron, copper and/or zinc chelators.
  • 5-chloro- 7-iodo-quinolin-8-ol, the antifungal and antiprotozoal drug clioquinol has been proposed for use in treating neurodegenerative diseases.
  • the majority of the metal chelators proposed to date suffer serious drawbacks, such as poor target specificity, poor penetration of the blood-brain barrier and clinical safety, limiting their widespread use in the treatment and prevention of such neurodegenerative diseases.
  • clioquinol is controversial since 10,000 people in Japan developed subacute myelo-optico neuropathy when using it.
  • known metal chelators such as clioquinol and related 8-hydroxyquinolines may not fully stabilise iron in the 3 + oxidation state, thus providing a risk of Fenton chemistry and associated oxidative stress.
  • Deferiprone a drug used to treat thalassaemia major, a blood disorder caused by the weakening and destruction of red blood cells.
  • Deferiprone chelates iron, reducing iron overload, a complication caused by thalassaemia major, thereby preventing the patient from accumulating potentially fatal iron levels.
  • Deferiprone is the subject of ongoing clinical trials for the treatment of neurodegenerative diseases; it has also been commercially available for use in treatment of iron overload for some time now, buy typically has a 85% efficacy at treating iron overload.
  • Other drugs used in medical applications as chelating agents to remove excess iron from the body include desferrioxamine, a hexadentate ligand, which has the drawback of being orally inactive as it is too hydrophilic and so must be injected to cross the blood- brain barrier, thus leading to poor patient compliance.
  • a heterocycle containing at least one electronegative atom and comprising a hydroxyl group or substituted hydroxyl group at the 1-position and the carbon at the 2- position being a carbonyl group for use as a medicament.
  • heterocycles of the present invention strongly chelate to metals present in the brain, including metals in the 3 + oxidation state, thus rendering the metals redox inactive, preventing oxidative damage to neurons through the production of free radicals via the Fenton reaction and/or Haber-Weiss reaction and, therefore preventing neuro degeneration. Additionally, chelation with the heterocycles of the present invention increases the solubility of the metal, thus increasing distribution of the metal away from the brain.
  • heterocycles of the present invention suitable for use in a wide range of medical applications, for example, as antimicrobial agents, antifungal agents, biostatic agents, drugs against iron overload, cancer and malaria.
  • 3-hydroxypyridin-4-one hereafter called "3,4-substituted heterocycles”
  • 1,2- substituted heterocycles have different properties, for example pKa, to 3,4- substituted heterocycles and so it is impossible to predict the effect of moving groups within a heterocycle.
  • the 1,2 -substituted heterocycles of the present invention are non-toxic to nerve cells, thus suggesting that they may be clinically safe, and provide effective chelation to metals present in the brain, thus delaying, halting and, in some cases, potentially reversing the damage cause by excess accumulation of the metals. Accordingly, the heterocycles of the present invention offer an effective treatment for neurodegenerative diseases such as Parkinson's and Alzheimer's.
  • the presence of a hydroxyl group or substituted hydroxyl group and the carbonyl group provide a bidentate ligand for chelation to metal ions such as iron, copper and zinc present in the brain.
  • the 1,2 -substituted heterocycles of the present invention have lower pKa's than other derivatives of the substituted heterocycles, such as 3,4-substituted heterocycles, due to the alpha effect of the hydroxyl group or substituted hydroxyl group being attached directly to the electronegative atom of the heterocyclic ring. Therefore, at physiological pH the hydrogen or cation of the hydroxyl group or substituted hydroxyl group is more easily lost for efficient chelation.
  • the metal complexes once formed between the 1,2 -substituted heterocycles of the present invention and the metal ions present in the brain are thermodynamically stronger complexes and so are less likely to dissociate.
  • the heterocycle of the present invention provides a bidentate ligand for chelation to Fe(III), thereby providing an effective treatment for diseases caused by excess Fe(III) accumulation in the human and animal body.
  • the substituted form of the hydroxyl group may either be an ester or comprise an alkali metal such as sodium, potassium or lithium, an alkaline earth metal such as magnesium or calcium, an ammonium salt, a phosphonium salt or a sulfonium salt.
  • the substituted form dissociates in situ to enable coordinate bonds to form between the heterocycle ligand and the metal ion.
  • the substituted form of the hydroxyl group may be a protecting group which can be easily cleaved in the body by enzymatic action or other means to reveal the oxygen of the hydroxyl group.
  • protecting groups include an acetate, ether, silyl ether, carbonate ester, carbamate ester, phosphate ester or sulfonate ester.
  • the protecting group is a carbamate ester, this may provide the additional benefit in use of inhibiting the acetylcholinesterase (AChE) enzyme which converts the neurotransmitter acetylcholine to choline and which is overexpressed in Alzheimer's disease.
  • the ester or protecting group form of the heterocycle may be favoured to assist the heterocycle of the present invention to cross the barrier between the blood and the brain following administration, where the heterocycle is required as a treatment for neurodegenerative diseases.
  • the heterocycle may further comprise at least one additional substituent selected to provide this functionality.
  • the heterocycle contains at least one oxygen atom, at least one nitrogen atom or a combination thereof. More preferably, the heterocycle contains at least one nitrogen atom.
  • the heterocycle is a 4-, 5-, 6-, or 7-membered ring.
  • the heterocycle is a 5- or 6-membered ring. More preferably the heterocycle is a 6-membered ring.
  • the heterocycle may be saturated or unsaturated.
  • the heterocycle may be substituted with or fused to one or more cyclic compounds or two or more heterocycles of the present invention may be linked together to form a bis, tris, tetrakis, etc. heterocycle linked by any suitable linking group, as will be appreciated in the art, for example " OCH2CH2O, thus forming tetradentate, hexadentate, etc chelators.
  • the heterocycle is unsaturated.
  • the 1,2 -substituted heterocycles in accordance with the preferred embodiments of the present invention chelate strongly to iron(III) and selectively in the presence of other metals, such as copper(II) and zinc(II). This is particularly beneficial where the heterocycles are intended for use in treating neurodegenerative diseases as iron(III) is redox active and is believed to play a significant role in neuro degeneration. Accordingly, these heterocycles may further overcome the problem of poor target specificity.
  • heterocycles in accordance with the first aspect of the present invention include:
  • each of the structures exemplified above provide a hydroxyl group, which may be provided in a substituted form, directly attached to the electronegative atom of the heterocyclic ring and located adjacent to a carbonyl group, thus providing effective bidentate chelation to metal ions in the brain.
  • the heterocycle of the present invention may be a hydroxypyridinone, hydroxypyranone, hydroxypyrazinone, hydroxypyridazinone or hydroxypyrimidinone.
  • the heterocycle is a nitrogen containing six membered heterocycle having the general formula:
  • R 1 is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group or a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation; and
  • R 2 and/or R 3 are absent or comprises at least one additional substituent selected to provide additional functionality to the heterocycle or a pharmaceutically acceptable salt and/or an isomer thereof.
  • the protecting group may be any of the types described above.
  • the group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation may be selected from carbonates, carbamates, glycosides, benzyl ethers or one or more protecting group as defined above.
  • R 1 may also be substituent selected to provide additional functionality to the heterocycle, as discussed in further detail below.
  • at least one of X, Y, Z and X' are carbon atoms, more preferably at least Z and X' is a carbon atom.
  • X and/or Y is a nitrogen atom.
  • all of X, Y, Z and X' are carbon atoms.
  • At least one additional substituent may be present or absent depending upon whether the compound requires modifications to be suitable for use in the human body or further reduce toxicity, if necessary.
  • the selection of a suitable substituent requires a simple design modification rather than any inventive skill, depending on the intended use of the compound.
  • the nitrogen containing six membered heterocycle may comprise from 1 to 4 additional substituents at the 3-, 4-, 5- and/or 6- positions.
  • R 2 or R 3 i.e. the from 1 to 4 additional substituents, may be independently selected from the groups comprising:
  • substituted or unsubstituted alkyl, heteroalkyl, alkenyl, heteroalkenyl or alkynyl which may be linear, branched or cyclic;
  • the additional substituents may be linked to form a cyclic structure.
  • the additional substituents may be linked together via a linking group, such as " OCH2CH2O, to form a bis-, tris- or tetrakis structure.
  • the heterocycle of the present invention may be a pharmaceutically acceptable salt.
  • Pharmaceutically or physiologically acceptable salts are particularly suitable for medical applications because of their greater aqueous solubility relative to the parent compounds. Such salts must clearly have a physiologically acceptable anion or cation.
  • physiologically acceptable salts of the compounds of the present invention include acid addition salts formed with inorganic acids such as hydrochloric, hydrobromic, hydroiodic, phosphoric, metaphosphoric, nitric and sulfuric acids, and with organic acids, such as tartaric, acetic, trifluoroacetic, citric, malic, lactic, fumaric, benzoic, formic, propionic, glycolic, gluconic, maleic, succinic, camphorsulfonic, isothionic, mucic, gentisic, isonicotinic, saccharic, glucuronic, furoic, glutamic, ascorbic, anthranilic, salicylic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, pantothenic, stearic, sulfinilic, alginic, galacturonic and arylsulfonic, for example benzenesul, in
  • Salts having a non-physiologically acceptable anion or cation are within the scope of the invention as useful intermediates for the preparation of physiologically acceptable salts and/or for use in non-therapeutic, for example, in vitro, situations.
  • the heterocycle may exist as an isomer of formula I, preferably a stereoisomer thereof including but not limited to diastereomeric, geometric and optical isomers, such as cis- and trans- forms, £-and Z- forms, D- and L- forms, (+) and (-) forms, and combinations thereof.
  • the nitrogen containing six membered heterocycle has the formula:
  • X and Y are independently selected from C or N;
  • R 1 is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle, as defined above;
  • R 3 and/or R 4 are each independently absent or present on the ring and comprise at least one additional substituent selected to provide additional functionality to the heterocycle, as defined above; or a pharmaceutically acceptable salt and/or an isomer thereof.
  • the additional substituent may be any of the groups described above.
  • the nitrogen containing six membered heterocycle may comprise from 1 to 3 additional substituents at the 3-, 4-, and/or 5- position, independently selected from the aforementioned groups.
  • At least X may be N.
  • R 2 and/or R 3 are at the 3-, 5- or 6- position in the nitrogen containing six membered ring.
  • the heterocycle of the present invention is intended for use in the treatment of neurodegenerative diseases, preferably Parkinson's disease or Alzheimer's disease.
  • a neurodegenerative disease comprising the steps of:
  • composition comprising a heterocycle containing at least one electronegative atom and comprising a hydroxyl group or salt derived therefrom at the 1-position and a carbonyl group at the 2-position; and administering an effective dose of the composition to a patient.
  • the "effective dose” of the composition is an amount sufficient to provide therapeutic relief or efficacy without any toxic side-effects. This is dependent upon the 1,2 -substituted heterocycle provided and may be readily calculated.
  • the composition may further comprise a pharmaceutically acceptable excipient.
  • the composition may comprise one or more pharmaceutically acceptable excipients well known in the art.
  • the composition may comprise one or more known active pharmaceutical ingredient used for the treatment of symptoms of neurodegenerative disease, such as Alzheimer's or Parkinson's, which complement the activity of the 1,2 -substituted heterocycles of the present invention. Examples of such known active pharmaceutical ingredients include additional chelating compounds or antioxidants.
  • R is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle, as defined above in relation to the first aspect of the present invention;
  • R 2 is H, an alkyl, an acyl, an aryl, a sulfanyl, an antioxidant moiety or any other functional groups, such as groups which help deliver the nitrogen containing six membered heterocycle to a certain region of the nerve cell or assist in crossing the blood-brain barrier; and
  • R 3 is absent or present at at least one of the 3-, 4- or 5 -positions on the ring and comprises at least one additional substituent selected to provide additional functionality to the heterocycle or a pharmaceutically acceptable salt and/or an isomer thereof.
  • the additional substituent may be any of the groups described above in relation to the preferred embodiment of the first aspect of the present invention.
  • the nitrogen containing six membered heterocycle may comprise from 1 to 3 additional substituents at the 3-, 4-, and/or 5- position, independently selected from the aforementioned groups.
  • the heterocycle in accordance with the third aspect of the present invention which is a previously unknown heterocycle, is particularly effective at chelating to excess metals in the brain and so reducing oxidative damage to neurons.
  • this heterocycle provides a 5% cell viability compared to a control at concentrations of ⁇ , in the presence of 6- hydroxydopamine (6-OHDA), a synthetic compound used to selectively destroy neurons and induce Parkinsonism. This is an improvement of over 111% when compared to the results achieved with commercially available drugs (such as deferiprone) at the same concentration.
  • 6-OHDA 6- hydroxydopamine
  • the heterocycle of the third aspect of the present invention provides a more effective treatment for neurodegenerative diseases.
  • R is H and so the nitrogen containing six membered heterocycle is l-hydroxy-6-(hydroxymethyl)pyridin-2- one.
  • the nitrogen containing six membered heterocycle has the formula:
  • R is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle, as defined above in relation to the first aspect of the present invention;
  • the nitrogen containing six membered heterocycle of formula (IV) may further comprise one or more additional substituents at the 3-, 4- or 5- position independently selected from the groups described in relation to the first aspect of the present invention to provide functionality to the heterocycle.
  • a fourth aspect of the present invention there is provide a method of forming the nitrogen containing six membered heterocycle of the third aspect comprising the steps of:
  • the l-hydroxypyridin-2-one-6-carboxylic acid methyl ester is provided by reaction of a l-hydroxypyridin-2-one-6-carboxylic acid with thionyl chloride in methanol. Further, the l-hydroxypyridin-2-one-6-carboxylic acid may be synthesised by treatment of 6-hydroxypicolinic acid with peracetic acid in acetic acid.
  • the protecting group may be an allyl, benzyl, silyl or sulfonate ester group, or any other protecting group known in the art.
  • the protecting group is an allyl group for ease of subsequent removal and also to avoid the risk of cleavage of the benzylic C-0 bond (i.e., the C-0 bond of the 6-hydroxymethyl group) under standard hydrogenation conditions.
  • Reduction of the ester group of the l-hydroxypyridin-2-one-6-carboxylic acid methyl ester may be accomplished by reaction with a reducing agent, for example, a reducing agent comprising potassium, calcium, barium, sodium or magnesium and/or borohydride.
  • a reducing agent comprising potassium, calcium, barium, sodium or magnesium and/or borohydride.
  • the reducing agent is sodium borohydride.
  • Deprotection of the N-hydroxyl group of the protected l-hydroxy-6-hydroxymethyl- pyridin-2-one by removal of the protecting group may be accomplished in the presence of palladium on activated carbon and trifluoroacetic acid.
  • the nitrogen containing six membered heterocycle of the third aspect for use as a medicament.
  • the nitrogen containing six membered heterocycle is intended for use in the treatment of neurodegenerative diseases, more preferably Parkinson's disease or Alzheimer's disease.
  • a neurodegenerative disease comprising the steps of:
  • composition comprising the nitrogen containing six membered heterocycle of the third aspect of the present invention; and administering an effective dose of the composition to a patient.
  • the preferred features and embodiments of the various aspects of the present invention apply mutatis mutandis to the other aspects of the present invention.
  • Figures 1 to 12 graphically show the results of the analysis of the neuroprotection of the tested compounds, as detailed below, to 6-OHDA toxicity;
  • Figures 13 to 16 graphically show data obtained via analysis of the neuroprotection of the l-hydroxypyridin-2-one compound in accordance with the fifth embodiment of the present invention and its analogues;
  • Figure 17 shows a comparison of the metal chelation ability of the compound of the fifth embodiment of the present invention, its analogues and the known 3- hydroxypyridin-4-one compound, Deferiprone (DFP); and
  • Figure 18 illustrates the changes to known markers of Parkinson's disease toxicity with deferiprone (DFP), the compound of the fifth embodiment of the present invention and its analogues.
  • l-hydroxy-2(i/f)-pyrazinone compound in accordance with a first embodiment synthesised by the reaction of glycine hydroxyamic acid and pyruvaldehyde in water in at an alkaline pH.
  • N-oxide [11] and pyridone [12] were synthesised, as described in detail below, having the following formulae:
  • Aqueous hydrochloric acid (37 %, 10 M) was then added dropwise to bring the solution to pH 3 and the solution was evaporated to a small volume.
  • the solution was diluted with water (40 mL) and extracted with chloroform (3 x 100 mL). The combined organic extracts were dried and evaporated to afford a light brown solid.
  • the solid was triturated with ether (50 mL) and filtered and washed with ether (50 mL) to afford 6-methyl-l-hydroxy-2(i//)- pyrazinone [15] as a light brow g, 11 %), having the formula:
  • SH-SY-5Y neuroblastoma cells were plated at 20,000 cells/well in a 96 well plate and left to adhere to the well surface and culture overnight in 50% advanced minimum essential medium (MEM), 50% Ham's F12 medium, 1% L-Glutamine and 2% fetal bovine serum (FBS). The media was then replaced with 100 ⁇ of FBS free MEM/Ham's F12 containing varying concentrations of the tested compound ranging from 0 to 300 ⁇ . After a further 24 hour incubation at 37 °C in a 5% C0 2 environment cell viability was measured using the tetrazolium dye 3-(4,5- dimethylthiazol-2-yl)-2.5-diphenyltetrazolium bromide (MTT).
  • MEM advanced minimum essential medium
  • FBS fetal bovine serum
  • the cell toxicity of the tested compound on normal healthy neurons was calculated to obtain a non-toxic concentration. This was done by adding increasing concentrations of the tested compound to the cells to calculate the concentration which may be applied before the cells die due to toxicity, if any, of the tested compounds.
  • Cell viability was calculated as a % compared to the untreated control, i.e. in the absence of the tested compound.
  • the cell viability was measured in the presence of 6-hydroxydopamine hydrobromide (6-OHDA), which synthetically induces Parkinson's disease in nerve cells, and the tested compound at the non-toxic concentration found in order to assess the efficacy in preventing neuro degeneration.
  • 6-OHDA 6-hydroxydopamine hydrobromide
  • SH-SY-5Y neuroblastoma cells were plated at 20,000 cells/well in a 96 well plate and left to adhere to the well surface and culture overnight in 50% advanced minimum essential medium (MEM), 50% Ham's F12 medium, 1% L-Glutamine and 2% fetal bovine serum (FBS).
  • MEM advanced minimum essential medium
  • FBS fetal bovine serum
  • 6-OHDA 6-hydroxydopamine hydrobromide
  • cell viability was measured using the tetrazolium dye 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT).
  • MTT 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
  • 11 of MTT 5 mg/mL was added to the 100 media in the well and incubated for 3 hours at 37 °C.
  • an equal volume of solubilizing solution 24 mL isopropyl alcohol, 1 mL HC1
  • Absorption was measured at 570 nm using a spectrophotometer.
  • Cell viability was calculated as a % compared to the untreated control, i.e. in the absence of 6-OHDA or the tested compound.
  • Calcein-AM to measure neuronal cytoplasmic labile iron pool was adapted from a previously reported procedure disclosed in Free Radic. Biol. Med. 2002, 33, 1037-1046.
  • SH-SY5Y cells plated at 20,000 cells/well in a black 96-well microplate were treated with ferric ammonium citrate (FAC; 50 ⁇ ) for 6 hrs in serum-free media.
  • FAC ferric ammonium citrate
  • Cells were washed twice with phosphate buffered saline (PBS) after which Calcein-AM (60 nM) was added.
  • PBS phosphate buffered saline
  • SH-SY5Y's prepared 6-well plates were incubated in serum-free media with FAC (50 ⁇ ) for 24 hrs. After iron loading, iron containing media was replaced with fresh serum-free media alone or in the presence of the compound of interest (100 ⁇ ) and incubated for a further 12 hrs.
  • FAC 50 ⁇
  • SH-SY5Y's prepared in 6-well plates were incubated in serum- free media with 6-OHDA (50 ⁇ ) with or without the compound (100 ⁇ ) for 24 hrs.
  • the data tables below show the results of the analysis of the neuroprotection of the tested compounds to 6-OHDA toxicity.
  • the compounds of the present invention provide comparable cell survival in the presence of 6-OHDA compared to the known compounds and higher non-toxic concentrations in the control. These results show that the treatment of healthy neuroblastoma cells (i.e. the control) with 50 ⁇ 6-OHDA typically results in the death of approximately 50% of the cells. This is due to the oxidative stress and metal dysregulation, etc. initiated by 6-OHDA. Further, at relatively low concentrations, i.e.
  • the known 3- hydroxypyridin-4-one compound (Deferiprone) and the compound of the present invention exhibit similar % cell viability in the 6-OHDA model of Parkinson's disease when compared to the control.
  • Figures 1 to 12 graphically show the data in relation to the tables given below, and the compound identification numbers relate to the identifying numbers described below in relation to the data tables.
  • the y-axis indicates percentage cell viability as compared to the control.
  • Figures 13 to 16 show pooled data obtained via analysis of the neuroprotection of the l-hydroxypyridin-2-one compound in accordance with the fifth embodiment of the present invention, and its analogues which lack the cyclic hydroxamic acid moiety and thus do not have the ability to chelate metals.
  • FIG 17 shows that l-hydroxypyridin-2-one compound in accordance with the fifth embodiment of the present invention (Compound 17; Compound [9]) has comparable ability to bind the labile pool of iron (LIP) in the cell as the known 3- hydroxypyridin-4-one compound, Deferiprone (DFP).
  • LIP labile pool of iron
  • DFP Deferiprone
  • Calcein-AM is a fluorescent dye whose fluorescence is quenched by excess labile iron in the cell.
  • the Calcein-AM assay indicates that the compound of the present invention is as efficient as Deferiprone (DFP) at causing Calcein fluorescence (which was originally quenched by the LIP upon incubation of the cells with 6-OHDA or iron), whereas the non-chelating compounds [8] (Compound 17A), [11] (Compound FL8) and [12] (Compound FL9) are unable to reduce LIP quenched calcein fluorescence.
  • DFP Deferiprone
  • ferritin and transferrin receptor expression is restored to levels seen in a healthy neuron following incubation with iron in the presence of DFP and compound [9], while compound [12] (Compound FL9) has no effect.
  • ferritin and transferrin receptor expression are altered by 6-OHDA in a similar way as iron incubation and accordingly, both DFP and compound [9] reduce this response back to control levels whereas compound [12] (Compound FL9) is unable to do so.
  • Figure 18 illustrates the changes to known markers of Parkinson's disease toxicity with deferiprone (DFP) and compound [9] (Compound 17).
  • DFP deferiprone
  • Compound 17 Tyrosine hydroxylase and Synaptophysin levels are both reduced in Parkinson's disease and are similarly reduced in neurons after incubation with Iron (Figure 18A) and 6-OHDA ( Figure 18B). While DFP and compound [9] are able to negate the reduced expression of both proteins to levels seen in a healthy neuron after incubation with iron ( Figure 18A) or 6-OHDA ( Figure 18B), the non-chelating compound [12] (Compound FL9) is unable to do so.
  • the compounds of the present invention offer an effective treatment for neurodegenerative disease based on strong metal chelation.
  • the compounds have been shown to reverse the damage caused by excess accumulation of metals. It follows that the compounds of the present invention will also be an effective treatment for other neurodegenerative diseases caused by disregulation/accumulation of metals in certain regions of the brain and central nervous system, such as Alzheimer's disease, Huntington's disease, macular degeneration, Friedreich's ataxia, amyotrophic lateral sclerosis and neurodegeneration with brain iron accumulation.
  • neurodegenerative diseases caused by disregulation/accumulation of metals in certain regions of the brain and central nervous system, such as Alzheimer's disease, Huntington's disease, macular degeneration, Friedreich's ataxia, amyotrophic lateral sclerosis and neurodegeneration with brain iron accumulation.
  • Table 1 shows compound toxicity in absence of 6-OHDA for compound [9],
  • Table 2 shows cell rescue effect in presence of 6-OHDA for compound [9].
  • Compound [9] is also identified as Compound 17 in the Tables and Figures.
  • Table 3 shows compound toxicity in absence of 6-OHDA for compound [16]
  • Table 4 shows cell rescue effect in presence of 6-OHDA for compound [16].
  • Compound [16] is also identified as Compound FLl in the Tables and Figures.
  • Table 5 shows compound toxicity in absence of 6-OHDA for compound [15]
  • Table 6 shows cell rescue effect in presence of 6-OHDA for compound [15].
  • Compound [15] is identified as Compound FL2 in the Tables and Figures.
  • Table 7 shows compound toxicity in absence of 6-OHDA for compound [5]
  • Table 8 shows cell rescue effect in presence of 6-OHDA for compound [5]
  • Compound [5] is also identified as Compound 7 in the Tables and Figures.
  • Table 9 shows compound toxicity in absence of 6-OHDA for compound [6]
  • Table 10 shows cell rescue effect in presence of 6-OHDA for compound [6].
  • Compound [6] is also identified as Compound 8 in the Tables and Figures.
  • Table 11 shows compound toxicity in absence of 6-OHDA for compound [3]
  • Table 12 shows cell rescue effect in presence of 6-OHDA for compound [3].
  • Compound [3] is also identified as Compound 14 in the Tables and Figures.
  • Table 13 shows compound toxicity in absence of 6-OHDA for compound [8]
  • Table 14 shows cell rescue effect in presence of 6-OHDA for compound [8].
  • Compound [8] is also identified as Compound 17A in the Tables and Figures.
  • Table 15 shows compound toxicity in absence of 6-OHDA for compound [11]
  • Table 16 shows cell rescue effect in presence of 6-OHDA for compound [11].
  • Compound [11] is also identified as Compound FL8 in the Tables and Figures.
  • Table 17 shows compound toxicity in absence of 6-OHDA for compound [12]
  • Table 18 shows cell rescue effect in presence of 6-OHDA for compound [12].
  • Compound [12] is also identified as Compound FL9 in the Tables and Figures.
  • Table 19 shows compound toxicity in absence of 6-OHDA for 3-hydroxypyridin-4- one (Deferiprone), Table 20 shows cell rescue effect in presence of 6-OHDA for Deferiprone. This compound is also identified as Compound 11 in the Tables and Figures, and is shown below.
  • Table 21 shows compound toxicity in absence of 6-OHDA for compound [17]
  • Table 22 shows cell rescue effect in presence of 6-OHDA for compound [17]. This compound is also identified as Compound 2 in the Tables and Figures, and is shown below.
  • Table 23 shows compound toxicity in absence of 6-OHDA for 3-hydroxypyran-4-one (Maltol), Table 24 shows cell rescue effect in presence of 6-OHDA for Maltol. This compound is also identified as Compound 10 in the Tables and Figures, and is shown below.

Landscapes

  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Neurosurgery (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to heterocyclic compounds for use in the treatment of diseases. In particular, the invention relates to heterocyclic compounds for use in the treatment of neurodegenerative diseases, such as Parkinson's or Alzheimer's disease, and methods of making and using the same. The heterocycle compounds contain at least one electronegative atom and comprise a hydroxyl group or substituted hydroxyl group at the 1-position and the carbon at the 2-position being a carbonyl group.

Description

METAL CHELATING COMPOUNDS FOR USE IN TREATING DISEASES
The present invention relates to heterocyclic compounds for use in the treatment of diseases. In particular, the invention relates to heterocyclic compounds for use in the treatment of neurodegenerative diseases, such as Parkinson's or Alzheimer's disease.
Neurodegeneration is the progressive loss of structure or function, and in some cases death, of neurons responsible for processing and transmitting information in the human body through chemical or electrical signals. As a consequence of such progressive loss and death of neurons, diseases such as Parkinson's and Alzheimer's may result.
A common cause of neurodegenerative diseases is disregulation and subsequent accumulation of metals in certain regions of the brain, in particular iron(III) but also copper and zinc, which in excess cause oxidative damage to neurons. While the origins are not fully understood, metal accumulation is due to metal dyshomeostasis, i.e. a breakdown in the mechanism through which the body regulates metal ion concentrations. As a downstream consequence of metal disregulation, displaced metals detrimentally interact with proteins present in the brain, for example, beta amyloid in the case of Alzheimer's disease and tau protein and alpha-synuclein in the case of Parkinson's disease. Accordingly, excessive metal accumulation causes increased interactions with these proteins, leading to accumulation of protein-metal aggregates which are one of the causes of nerve cell death and, therefore, neurodegeneration.
Following the onset of neurodegenerative diseases such as Parkinson's or Alzheimer's, steps are taken to manage the disease, thereby providing symptomatic relief through the use of pharmaceutical drugs. Alternatively but less commonly, surgical procedures may be offered to Parkinson patients including deep brain stimulation, in which a brain pacemaker is implanted to send electrical impulses to specific parts of the brain to control symptoms, or lesional surgery, which involves the intentional creation of lesions to suppress over activity of specific subcortical areas. However, symptomatic treatment is largely ineffective for Alzheimer's patients and so in most situations palliative care is provided.
At present, there are no widely used measures effective in delaying or preventing the progression of neurodegenerative diseases such as Parkinson's or Alzheimer's. Further, no measure has been proposed which has the potential to reverse neuro degeneration, thus providing the possibility of an effective cure for such diseases.
It has previously been proposed that metal chelation may be used to reduce excessive metal accumulation in regions of the brain, thereby reducing neurodegeneration and so delaying or halting the progression of neurodegenerative diseases. For example, the use of 3-hydroxypyridin-4-one based compounds has been proposed for use as iron, copper and/or zinc chelators. Additionally, 5-chloro- 7-iodo-quinolin-8-ol, the antifungal and antiprotozoal drug clioquinol, has been proposed for use in treating neurodegenerative diseases. However, the majority of the metal chelators proposed to date suffer serious drawbacks, such as poor target specificity, poor penetration of the blood-brain barrier and clinical safety, limiting their widespread use in the treatment and prevention of such neurodegenerative diseases. Further, the use of clioquinol is controversial since 10,000 people in Japan developed subacute myelo-optico neuropathy when using it. In addition, known metal chelators such as clioquinol and related 8-hydroxyquinolines may not fully stabilise iron in the 3+ oxidation state, thus providing a risk of Fenton chemistry and associated oxidative stress. As such, there is no commercially available metal chelator drug used to treat neurodegenerative diseases.
To date, the only commercially available drug based on 3-hydroxypyridin-4-one is deferiprone, a drug used to treat thalassaemia major, a blood disorder caused by the weakening and destruction of red blood cells. Deferiprone chelates iron, reducing iron overload, a complication caused by thalassaemia major, thereby preventing the patient from accumulating potentially fatal iron levels. Deferiprone is the subject of ongoing clinical trials for the treatment of neurodegenerative diseases; it has also been commercially available for use in treatment of iron overload for some time now, buy typically has a 85% efficacy at treating iron overload. Other drugs used in medical applications as chelating agents to remove excess iron from the body include desferrioxamine, a hexadentate ligand, which has the drawback of being orally inactive as it is too hydrophilic and so must be injected to cross the blood- brain barrier, thus leading to poor patient compliance.
There exists a need to provide an effective treatment for neurodegenerative diseases based on metal chelation which effectively reduces metal accumulation, in particular iron accumulation, and restores metal ion homeostasis in the brain to delay or halt the progression of neurodegenerative diseases, which does not suffer the drawbacks of previously proposed metal chelators.
Accordingly, in a first aspect of the present invention, there is provided a heterocycle containing at least one electronegative atom and comprising a hydroxyl group or substituted hydroxyl group at the 1-position and the carbon at the 2- position being a carbonyl group for use as a medicament.
Without being bound to a specific mechanism of action, it is believed that the heterocycles of the present invention (hereafter called "1,2 -substituted heterocycles") strongly chelate to metals present in the brain, including metals in the 3+ oxidation state, thus rendering the metals redox inactive, preventing oxidative damage to neurons through the production of free radicals via the Fenton reaction and/or Haber-Weiss reaction and, therefore preventing neuro degeneration. Additionally, chelation with the heterocycles of the present invention increases the solubility of the metal, thus increasing distribution of the metal away from the brain. These surprising effects also render the heterocycles of the present invention suitable for use in a wide range of medical applications, for example, as antimicrobial agents, antifungal agents, biostatic agents, drugs against iron overload, cancer and malaria. This is surprising despite previous proposals to use 3-hydroxypyridin-4-one (hereafter called "3,4-substituted heterocycles") as it is appreciated that 1,2- substituted heterocycles have different properties, for example pKa, to 3,4- substituted heterocycles and so it is impossible to predict the effect of moving groups within a heterocycle. In addition, it has been found that the 1,2 -substituted heterocycles of the present invention are non-toxic to nerve cells, thus suggesting that they may be clinically safe, and provide effective chelation to metals present in the brain, thus delaying, halting and, in some cases, potentially reversing the damage cause by excess accumulation of the metals. Accordingly, the heterocycles of the present invention offer an effective treatment for neurodegenerative diseases such as Parkinson's and Alzheimer's.
The presence of a hydroxyl group or substituted hydroxyl group and the carbonyl group provide a bidentate ligand for chelation to metal ions such as iron, copper and zinc present in the brain. In particular, the 1,2 -substituted heterocycles of the present invention have lower pKa's than other derivatives of the substituted heterocycles, such as 3,4-substituted heterocycles, due to the alpha effect of the hydroxyl group or substituted hydroxyl group being attached directly to the electronegative atom of the heterocyclic ring. Therefore, at physiological pH the hydrogen or cation of the hydroxyl group or substituted hydroxyl group is more easily lost for efficient chelation. Further, as a consequence of the lower pKa, the metal complexes once formed between the 1,2 -substituted heterocycles of the present invention and the metal ions present in the brain are thermodynamically stronger complexes and so are less likely to dissociate.
Additionally, the presence of a hydroxyl or substituted hydroxyl group and a carbonyl group at adjacent positions on the heterocyclic ring provide effective chelation of metal ions present in the brain as they provide two hard Lewis bases for chelation to a hard Lewis acid (i.e. metal ions such as Fe(III)) and due to the enhanced entropy of binding. In one or more embodiments of the present invention, the heterocycle of the present invention provides a bidentate ligand for chelation to Fe(III), thereby providing an effective treatment for diseases caused by excess Fe(III) accumulation in the human and animal body.
The substituted form of the hydroxyl group may either be an ester or comprise an alkali metal such as sodium, potassium or lithium, an alkaline earth metal such as magnesium or calcium, an ammonium salt, a phosphonium salt or a sulfonium salt. The substituted form dissociates in situ to enable coordinate bonds to form between the heterocycle ligand and the metal ion. Alternatively or additionally, the substituted form of the hydroxyl group may be a protecting group which can be easily cleaved in the body by enzymatic action or other means to reveal the oxygen of the hydroxyl group. Examples of such protecting groups include an acetate, ether, silyl ether, carbonate ester, carbamate ester, phosphate ester or sulfonate ester. Where the protecting group is a carbamate ester, this may provide the additional benefit in use of inhibiting the acetylcholinesterase (AChE) enzyme which converts the neurotransmitter acetylcholine to choline and which is overexpressed in Alzheimer's disease. In particular, the ester or protecting group form of the heterocycle may be favoured to assist the heterocycle of the present invention to cross the barrier between the blood and the brain following administration, where the heterocycle is required as a treatment for neurodegenerative diseases. However, the heterocycle may further comprise at least one additional substituent selected to provide this functionality.
Preferably, the heterocycle contains at least one oxygen atom, at least one nitrogen atom or a combination thereof. More preferably, the heterocycle contains at least one nitrogen atom.
Preferably the heterocycle is a 4-, 5-, 6-, or 7-membered ring. In one preferred embodiment of the present invention, the heterocycle is a 5- or 6-membered ring. More preferably the heterocycle is a 6-membered ring. The heterocycle may be saturated or unsaturated. Alternatively and/or additionally, the heterocycle may be substituted with or fused to one or more cyclic compounds or two or more heterocycles of the present invention may be linked together to form a bis, tris, tetrakis, etc. heterocycle linked by any suitable linking group, as will be appreciated in the art, for example "OCH2CH2O, thus forming tetradentate, hexadentate, etc chelators. In a preferred embodiment, the heterocycle is unsaturated.
The 1,2 -substituted heterocycles in accordance with the preferred embodiments of the present invention chelate strongly to iron(III) and selectively in the presence of other metals, such as copper(II) and zinc(II). This is particularly beneficial where the heterocycles are intended for use in treating neurodegenerative diseases as iron(III) is redox active and is believed to play a significant role in neuro degeneration. Accordingly, these heterocycles may further overcome the problem of poor target specificity.
Examples of the heterocycles in accordance with the first aspect of the present invention include:
Figure imgf000007_0001
Figure imgf000008_0001
Figure imgf000008_0002
As will be appreciated, each of the structures exemplified above provide a hydroxyl group, which may be provided in a substituted form, directly attached to the electronegative atom of the heterocyclic ring and located adjacent to a carbonyl group, thus providing effective bidentate chelation to metal ions in the brain. Alternatively, and/or additionally, the heterocycle of the present invention may be a hydroxypyridinone, hydroxypyranone, hydroxypyrazinone, hydroxypyridazinone or hydroxypyrimidinone.
In a preferred embodiment of the present invention, the heterocycle is a nitrogen containing six membered heterocycle having the general formula:
Figure imgf000009_0001
(I) wherein X, Y, Z and X' are independently selected from the group consisting of C, N, 0 or S;
R1 is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group or a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation; and
R2 and/or R3 are absent or comprises at least one additional substituent selected to provide additional functionality to the heterocycle or a pharmaceutically acceptable salt and/or an isomer thereof.
The protecting group may be any of the types described above. The group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation may be selected from carbonates, carbamates, glycosides, benzyl ethers or one or more protecting group as defined above. Alternatively and/or additionally, R1 may also be substituent selected to provide additional functionality to the heterocycle, as discussed in further detail below. Preferably, at least one of X, Y, Z and X' are carbon atoms, more preferably at least Z and X' is a carbon atom. In one embodiment, X and/or Y is a nitrogen atom. In an alternative embodiment, all of X, Y, Z and X' are carbon atoms.
At least one additional substituent may be present or absent depending upon whether the compound requires modifications to be suitable for use in the human body or further reduce toxicity, if necessary. As will be appreciated, the selection of a suitable substituent requires a simple design modification rather than any inventive skill, depending on the intended use of the compound.
The nitrogen containing six membered heterocycle may comprise from 1 to 4 additional substituents at the 3-, 4-, 5- and/or 6- positions.
For example, R2 or R3, i.e. the from 1 to 4 additional substituents, may be independently selected from the groups comprising:
F, CI, Br or I;
OH, OR, OCOR, NH2, NHR4, NR4R5, +NR4R5R6, C02R4, C0NHR4, CONR4R5 or
C=0R4;
R4OR5;
a substituted or unsubstituted alkyl, heteroalkyl, alkenyl, heteroalkenyl or alkynyl, which may be linear, branched or cyclic;
a substituted or unsubstituted aryl or heteroaryl;
S03-, X+, SO3H, SH or SR4;
P=0(OH)2 or salts P=0(OR4)2 or P=0(OR4)(OR5);
S=0R4 or 0=S=0R4;
B(OH)2 or B(OR4)(OR5);
CN;
N02; or
C(=NH)(NH2), C=SR4;
wherein R4, R5 and R6 may be independently selected from the group consisting of: H; OH; 0(CH2)XCH3; (CH2)XCH3; a alkyl, heteroalkyl, alkenyl, heteroalkenyl or alkynyl, which may be linear, branched or cyclic; a methylene moiety; an acyl; a sulfanyl; an aryl; a heteroaryl; or an antioxidant moiety; where x = 0 to 12, and X+ may be independently selected from the group consisting of Na+, K+, Li+, Mg2+, Ca2+ or an organic cation.
In one or more embodiments, where at least two additional substituents are present, the additional substituents may be linked to form a cyclic structure. Alternatively and/or additionally, the additional substituents may be linked together via a linking group, such as "OCH2CH2O, to form a bis-, tris- or tetrakis structure.
The heterocycle of the present invention may be a pharmaceutically acceptable salt. Pharmaceutically or physiologically acceptable salts are particularly suitable for medical applications because of their greater aqueous solubility relative to the parent compounds. Such salts must clearly have a physiologically acceptable anion or cation. Suitably physiologically acceptable salts of the compounds of the present invention include acid addition salts formed with inorganic acids such as hydrochloric, hydrobromic, hydroiodic, phosphoric, metaphosphoric, nitric and sulfuric acids, and with organic acids, such as tartaric, acetic, trifluoroacetic, citric, malic, lactic, fumaric, benzoic, formic, propionic, glycolic, gluconic, maleic, succinic, camphorsulfonic, isothionic, mucic, gentisic, isonicotinic, saccharic, glucuronic, furoic, glutamic, ascorbic, anthranilic, salicylic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, pantothenic, stearic, sulfinilic, alginic, galacturonic and arylsulfonic, for example benzenesulfonic and p-toluenesulfonic, acids; base addition salts formed with alkali metals and alkaline earth metals and organic bases such as Ν,Ν-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine), lysine and procaine; and internally formed salts. Salts having a non-physiologically acceptable anion or cation are within the scope of the invention as useful intermediates for the preparation of physiologically acceptable salts and/or for use in non-therapeutic, for example, in vitro, situations. As will be appreciated, the heterocycle may exist as an isomer of formula I, preferably a stereoisomer thereof including but not limited to diastereomeric, geometric and optical isomers, such as cis- and trans- forms, £-and Z- forms, D- and L- forms, (+) and (-) forms, and combinations thereof.
In one or more preferred embodiments, the nitrogen containing six membered heterocycle has the formula:
Figure imgf000012_0001
wherein X and Y are independently selected from C or N;
R1 is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle, as defined above;
R2 is an alkyl, an aryl, an heteroaryl, OH, OR', SH, SR', S(0)R', S02R', NH, NR', sulfinyl, sulfonyl, F, CI, Br, I, C=0, COOR', C=NR' or R5OR6, wherein R' is H, alkyl, cycloalkyl, heterocycloalkyl, acyl, aryl or heteroaryl, R5 is (CH2)X wherein x = 0 to 100 and R6 is H, an alkyl, an acyl, an aryl, a sulfinyl, a sulfonyl, an antioxidant moiety or any other functional groups, such as groups which help deliver the nitrogen containing six membered heterocycle to a certain region of the nerve cell or assist in crossing the blood-brain barrier; and
R3 and/or R4 are each independently absent or present on the ring and comprise at least one additional substituent selected to provide additional functionality to the heterocycle, as defined above; or a pharmaceutically acceptable salt and/or an isomer thereof.
The additional substituent may be any of the groups described above. Further, the nitrogen containing six membered heterocycle may comprise from 1 to 3 additional substituents at the 3-, 4-, and/or 5- position, independently selected from the aforementioned groups.
At least X may be N. Preferably R2 and/or R3 are at the 3-, 5- or 6- position in the nitrogen containing six membered ring. In a preferred embodiment, R2 and/or R3 are selected from the group consisting of: H, an alkyl, such as CH3,; cycloalkyl, such as R2-(CH2)x-R3 where x = 0 to 6, COOR', such as COOH; and R6OR7, such as CH2OH.
In a preferred embodiment of the present invention, the heterocycle of the present invention is intended for use in the treatment of neurodegenerative diseases, preferably Parkinson's disease or Alzheimer's disease.
In a second aspect of the present invention, there is provided a method for treating a neurodegenerative disease comprising the steps of:
providing a composition comprising a heterocycle containing at least one electronegative atom and comprising a hydroxyl group or salt derived therefrom at the 1-position and a carbonyl group at the 2-position; and administering an effective dose of the composition to a patient.
The "effective dose" of the composition is an amount sufficient to provide therapeutic relief or efficacy without any toxic side-effects. This is dependent upon the 1,2 -substituted heterocycle provided and may be readily calculated.
As will be appreciated, the preferred features and embodiments of the first aspect of the present invention apply mutatis mutandis to the second aspect of the present invention. The composition may further comprise a pharmaceutically acceptable excipient. For example, the composition may comprise one or more pharmaceutically acceptable excipients well known in the art. Additionally or alternatively, the composition may comprise one or more known active pharmaceutical ingredient used for the treatment of symptoms of neurodegenerative disease, such as Alzheimer's or Parkinson's, which complement the activity of the 1,2 -substituted heterocycles of the present invention. Examples of such known active pharmaceutical ingredients include additional chelating compounds or antioxidants.
In a third aspect of the present invention, there is provided a nitrogen containing six membered heterocycle having the formula:
Figure imgf000014_0001
(III) wherein R is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle, as defined above in relation to the first aspect of the present invention;
R1 is (CH2)x, wherein x = 0 to 100;
R2 is H, an alkyl, an acyl, an aryl, a sulfanyl, an antioxidant moiety or any other functional groups, such as groups which help deliver the nitrogen containing six membered heterocycle to a certain region of the nerve cell or assist in crossing the blood-brain barrier; and
R3 is absent or present at at least one of the 3-, 4- or 5 -positions on the ring and comprises at least one additional substituent selected to provide additional functionality to the heterocycle or a pharmaceutically acceptable salt and/or an isomer thereof.
The additional substituent may be any of the groups described above in relation to the preferred embodiment of the first aspect of the present invention. Further, the nitrogen containing six membered heterocycle may comprise from 1 to 3 additional substituents at the 3-, 4-, and/or 5- position, independently selected from the aforementioned groups.
It has been found that the heterocycle in accordance with the third aspect of the present invention, which is a previously unknown heterocycle, is particularly effective at chelating to excess metals in the brain and so reducing oxidative damage to neurons. In particular, testing has shown that this heterocycle provides a 5% cell viability compared to a control at concentrations of ΙΟΟμΜ, in the presence of 6- hydroxydopamine (6-OHDA), a synthetic compound used to selectively destroy neurons and induce Parkinsonism. This is an improvement of over 111% when compared to the results achieved with commercially available drugs (such as deferiprone) at the same concentration. Accordingly, the heterocycle of the third aspect of the present invention provides a more effective treatment for neurodegenerative diseases.
In a preferred embodiment of the present invention, R is H and so the nitrogen containing six membered heterocycle is l-hydroxy-6-(hydroxymethyl)pyridin-2- one.
In one or more embodiments of the present invention, the nitrogen containing six membered heterocycle has the formula:
Figure imgf000015_0001
(IV) wherein R is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle, as defined above in relation to the first aspect of the present invention;
or a pharmaceutically acceptable salt and/or an isomer thereof.
The nitrogen containing six membered heterocycle of formula (IV) may further comprise one or more additional substituents at the 3-, 4- or 5- position independently selected from the groups described in relation to the first aspect of the present invention to provide functionality to the heterocycle.
In a fourth aspect of the present invention, there is provide a method of forming the nitrogen containing six membered heterocycle of the third aspect comprising the steps of:
providing a l-hydroxypyridin-2-one-6-carboxylic acid methyl ester; reacting the l-hydroxypyridin-2-one-6-carboxylic acid methyl ester with a protecting group to protect the N-hydroxyl group, thereby forming a protected l-hydroxypyridin-2-one-6-carboxylic acid methyl ester;
reduction of the methyl ester group of the protected 1-hydroxypyridin- 2-one-6-carboxylic acid methyl ester to form protected l-hydroxy-6- hydroxymethyl-pyridin-2-one; and
deprotecting of the N-hydroxyl group of the protected l-hydroxy-6- hydroxymethyl-pyridin-2-one by removal of the protecting group to form the nitrogen containing heterocycle of the third aspect of the present invention.
In one embodiment, the l-hydroxypyridin-2-one-6-carboxylic acid methyl ester is provided by reaction of a l-hydroxypyridin-2-one-6-carboxylic acid with thionyl chloride in methanol. Further, the l-hydroxypyridin-2-one-6-carboxylic acid may be synthesised by treatment of 6-hydroxypicolinic acid with peracetic acid in acetic acid.
The protecting group may be an allyl, benzyl, silyl or sulfonate ester group, or any other protecting group known in the art. In a preferred embodiment, the protecting group is an allyl group for ease of subsequent removal and also to avoid the risk of cleavage of the benzylic C-0 bond (i.e., the C-0 bond of the 6-hydroxymethyl group) under standard hydrogenation conditions.
Reduction of the ester group of the l-hydroxypyridin-2-one-6-carboxylic acid methyl ester may be accomplished by reaction with a reducing agent, for example, a reducing agent comprising potassium, calcium, barium, sodium or magnesium and/or borohydride. Preferably, the reducing agent is sodium borohydride.
Deprotection of the N-hydroxyl group of the protected l-hydroxy-6-hydroxymethyl- pyridin-2-one by removal of the protecting group may be accomplished in the presence of palladium on activated carbon and trifluoroacetic acid.
In a fifth aspect of the present invention, there is provided the nitrogen containing six membered heterocycle of the third aspect for use as a medicament.
Preferably, the nitrogen containing six membered heterocycle is intended for use in the treatment of neurodegenerative diseases, more preferably Parkinson's disease or Alzheimer's disease.
In a sixth aspect of the present invention, there is provided a method for treating a neurodegenerative disease comprising the steps of:
providing a composition comprising the nitrogen containing six membered heterocycle of the third aspect of the present invention; and administering an effective dose of the composition to a patient. As will be appreciated, the preferred features and embodiments of the various aspects of the present invention apply mutatis mutandis to the other aspects of the present invention.
Various embodiments of the present invention will now be described more particularly by way of examples, with reference to the accompanying figures in which:
Figures 1 to 12 graphically show the results of the analysis of the neuroprotection of the tested compounds, as detailed below, to 6-OHDA toxicity;
Figures 13 to 16 graphically show data obtained via analysis of the neuroprotection of the l-hydroxypyridin-2-one compound in accordance with the fifth embodiment of the present invention and its analogues;
Figure 17 shows a comparison of the metal chelation ability of the compound of the fifth embodiment of the present invention, its analogues and the known 3- hydroxypyridin-4-one compound, Deferiprone (DFP); and
Figure 18 illustrates the changes to known markers of Parkinson's disease toxicity with deferiprone (DFP), the compound of the fifth embodiment of the present invention and its analogues.
Compounds of the Present Invention
l-hydroxy-2(i/f)-pyrazinone compound in accordance with a first embodiment synthesised by the reaction of glycine hydroxyamic acid and pyruvaldehyde in water in at an alkaline pH.
Figure imgf000018_0001
[15] l-hydroxypyridin-2-one compound in accordance with a second embodiment, synthesised in accordance with the method disclosed in /. Med. Chem. 2002, 45, 3963-3971, in which acetic acid was employed as the solvent instead of trifluoroacetic acid/trifluoroacetic anh dride for safety reasons.
Figure imgf000019_0001
l-hydroxypyridin-2-one compound in accordance with a third embodiment, synthesised in accordance with the method disclosed in /. Labelled Cpd. Radiopharm. 2001, 44, 13-19.
Figure imgf000019_0002
l-hydroxypyridin-2-one compound in accordance with a fourth embodiment.
Figure imgf000019_0003
[3]
This compound was synthesised via the procedure described in detail below. l-hydroxypyridin-2-one compound in accordance with a fifth embodiment
Figure imgf000019_0004
[9] This compound, which is in accordance with a third aspect of the present invention, was synthesised via the procedure described in detail below.
Prior art compounds
Compounds known in the art were provided for comparison to the compounds in accordance with the present invention. The prior art compounds are commercially available or were synthesised in accordance with techniques described in the art, having the following formulae: l-hydroxy-2(iH)-pyrazinone compound, synthesised in accordance with the method disclosed in Chem. Phar 36, 2323-2330.
Figure imgf000020_0001
[16]
3-hydroxypyridin-2-one compound, synthesised in accordance with the method disclosed in Tetrahedron, 1995, 55, 1129-1142.
Figure imgf000020_0002
3-hydroxypyran-4-one compound (maltol, commercially available from Sigma- Aldrich).
Figure imgf000021_0001
3 -hydroxypyridin-4-one compound (deferiprone, commercially available from Sigma-Aldrich).
Figure imgf000021_0002
In addition, an N-oxide [11] and pyridone [12] were synthesised, as described in detail below, having the following formulae:
Figure imgf000021_0003
Figure imgf000021_0004
These compounds are considered structural analogues of the l-hydroxypyridin-2 - one compound in accordance with the fifth embodiment of the present invention but lack the cyclic hydroxamic acid metal chelating functionality. Accordingly, the screening results for these compounds provide information on the extent to which drug efficacy in l-hydroxypyridin-2 -one compounds is due to their metal chelating ability. Experimental
General Procedures. All solvents and reagents were purchased from Sigma-Aldrich, Acros Organics or Alfa-Aesar and used without further purification unless otherwise specified. Reactions were monitored by TLC using silica gel with UV254 fluorescent indicator. Organic extracts of reaction products were dried over anhydrous magnesium sulfate.
Synthesis of l-hydroxypyridin-2-one in accordance with the fourth embodiment
Formation of 2-chloropyridine-3-carboxylic acid N-oxide:
To a solution of 2-chloronicotinic acid [1] (12.00 g, 76 mmol), having the formula:
Figure imgf000022_0001
in glacial acetic acid (90 mL) was added peroxyacetic acid (36-40 %, 35 mL). The solution was carefully raised to 80 °C and stirring was continued for 10 hours. The solution was cooled to room temperature and the resulting precipitate was filtered and washed with diethyl ether. The solid was allowed to dry in air to afford 2-chloropyridine-3-carboxylic acid N-oxide [2] as a white powder (5.56 g, 42 %), having the formula:
Figure imgf000023_0001
[2]
Formation of l-hydroxy-2-oxo-l,2-dihydropyridine-3-carboxylic acid:
A solution of N-oxide [2] (7.85 g, 45 mmol) in aqueous potassium hydroxide (10 %, 125 mL) was stirred at 70 °C for 3 days. The solution was then cooled to 0 °C and concentrated hydrochloric acid was added to pH 1. The precipitated solid was collected by filtration and washed with water. The crude solid was then recrystallised from methanol to afford l-hydroxy-2-oxo- l,2-dihydropyridine-3-carboxylic acid [3] as white needles (6.51 g, 92 %), having the formula:
Figure imgf000023_0002
[3]
Synthesis of l-hydroxypyridin-2-one in accordance with the fifth embodiment
Formation of l-hydroxy-6-oxo-l,6-dihydropyridine-2-carboxylic acid:
To a suspension of 6-hydroxypicolinic acid [4] (26.20 g, 188 mmol) having the formula:
Figure imgf000023_0003
in glacial acetic acid (160 mL) was carefully added peroxyacetic acid (36-40 %, 80 mL). The temperature was carefully raised to 80 °C and stirring was continued for 12 hours. The flask was allowed to cool to room temperature and the resulting solid precipitate was collected by filtration and washed with diethyl ether, affording l-Hydroxy-6-oxo-l,6-dihydropyridine-2-carboxylic acid [5] as a cream solid (18.17 g, 77 %), having the formula:
Figure imgf000024_0001
Formation of methyl l-hydroxy-6-oxo-l,6-dihydropyridine-2-carboxylate: To a suspension of acid [5] (15.73 g, 101 mmol) in methanol (200 mL) at 0 °C was added thionyl chloride (31.00 g, 426 mmol) dropwise. The mixture was heated under reflux for 4 hours. The solution was then allowed to cool to room temperature and the solvent was removed in vacuo to afford methyl 1- hydroxy-6-oxo-l,6-dihydropyridine-2-carboxylate [6] as a cream solid (16.47 g, 96 %), having the formula:
Figure imgf000024_0002
Formation of methyl 6-oxo-l-(allyloxy)-l,6-dihydropyridine-2-carboxylate: To a solution of [6] (16.47 g, 97 mmol) in acetonitrile (200 mL) was added potassium carbonate (32.11 g, 232 mmol), followed by allyl bromide (28.10 g, 232 mmol). The flask was heated under reflux for 4 hours before the reaction mixture was filtered and the solvent removed under high vacuum. The residue was dissolved in toluene (100 mL) and the solvent was evaporated in vacuo to afford methyl 6-oxo-l-(allyloxy)-l,6-dihydropyridine-2-carboxylate
[7] as a white crystalline solid (19.12 g, 94 %), having the formula:
Figure imgf000025_0001
Formation of 6-(hydroxymethyl)-l-(allyloxy)pyridin-2 (lH)-one:
To a suspension of [7] (19.12 g, 92 mmol) in tetrahydrofuran (200 mL) was added solid sodium borohydride (25.07 g, 663 mmol) in small portions. The solution was heated under reflux for 15 minutes. Methanol (14 mL) was then added dropwise at reflux over 2 hours. The solution was then cooled to 0 °C, quenched by careful addition of saturated aqueous ammonium chloride (25 mL) and stirring was continued for 15 minutes. The solvents were removed in vacuo and the residue was extracted with dichloromethane (3 25 mL). The combined organic extracts were dried and evaporated to afford 6- (hydroxymethyl)-l-(allyloxy)pyridin-2(lH)-one [8] an off-white solid (10.83 g, 65 %), having the formula:
Figure imgf000025_0002
[8] Formation of l-hydroxy-6-(hydroxymethyl)pyridin-2 (lH)-one:
To a solution of compound [8] (0.21 g, 1.06 mmol) in 20 % water in dioxane (10 mL) was added trifluoroacetic acid (0.11 mL). The solution was heated under reflux for 1 hour. Palladium on activated carbon (10% Pd/C, 0.02 g) was then added and the mixture was heated under reflux for a further 20 hours. The mixture was filtered and the filtrate was evaporated in vacuo to afford l-hydroxy-6-(hydroxymethyl)pyridin-2 (lH)-one [9] as a brown solid (0.08 g, 60 %), having the formula:
Figure imgf000026_0001
Synthesis of 2- (hydroxymethyl) pyridine TV-oxide
To a solution of m-CPBA (14.67 g, 50 wt % in Η2Ο, 42.5 mmol) in chloroform (30 mL) was slowly added a solution of 2-(hydroxymethyl)pyridine [10] (3.85 g, 35.25 mmol) in chloroform (15 mL), having the formula:
Figure imgf000026_0002
The solution was heated at 65 °C for 20 hours. The solution was then allowed to cool to room temperature and was dried and evaporated. The resulting residue was triturated with diethyl ether (150 mL) and the suspension was heated under reflux for 30 minutes. The insoluble solid was filtered and washed with diethyl ether (20 mL) and allowed to dry in air to afford 2- (hydroxymethyl)pyridine N-oxide [11] as a white solid (2.77 g, 63 %), having the formula:
Figure imgf000027_0001
[11]
Synthesis of 6-(hydroxymethyl)pyridin-2(lH)-one
To a suspension of acid [4] (1.00 g, 7.19 mmol) in dry tetrahydrofuran (35 mL) was added a solution of borane in tetrahydrofuran (35.9 mL, 1 M, 5 eq) dropwise. After the addition was complete, the flask was heated to 65 °C for 24 hours. The solution was then cooled to 0 °C and methanol (30 mL) was added. The flask was heated at 65 ° overnight and the solvent was evaporated. Methanol (50 mL) was added and the solvent was again evaporated. The resulting solid was triturated with acetone (30 mL), and then filtered and washed with methanol (5 mL) and acetone (40 mL) to afford 6- (hydroxymethyl)pyridin-2 (lH)-one [12] as a white solid (0.47 g, 52 %), having the formula:
Figure imgf000027_0002
Η [12]
Synthesis of 6-methyl-l-hydroxy-2(lH)-pyrazinone
Formation of glycine hydroxamic acid:
To a solution of glycine ethyl ester hydrochloride [13] (6.02 g, 43.129 mmol), having the formula:
Figure imgf000028_0001
in water (4 mL) was added hydroxylamine hydrochloride (2.997 g, 43.129 mmol). The solution was cooled to 0 °C and aqueous sodium hydroxide (11.8 mL, 12 M, 3.3 equiv) was added dropwise over 10 minutes. The solution was stirred at 0 °C for 30 minutes and was then allowed to warm to room temperature and stirring was continued for 24 hours. Aqueous hydrochloric acid (37 %, 4.3 mL, 10 M, 1 equiv) was then added to bring the solution to pH 2 and the solution was cooled to 0 °C. The precipitated solid was filtered and washed with cold water (10 mL) and was allowed to dry in air to afford glycine hydroxamic acid [14] as a white solid (2.49 g, 64 %), having the formula:
Figure imgf000028_0002
[14]
Formation of 6-methyl-l-hydroxy-2(i//)-pyrazinone:
To a solution of glycine hydroxamic acid [14] (0.50 g, 5.555 mmol) in methanol (40 mL) and water (20 mL) at 0 °C was added a solution of pyruvaldehyde (1.15 g, 40 wt. % in water, 1.15 equiv) in methanol (15 mL). Aqueous sodium hydroxide (2.0 mL, 2 M) was added dropwise over 15 minutes and the solution was stirred at 0 °C for 1 hour. The solution was then allowed to warm to room temperature and stirring was continued for 24 hours. Aqueous hydrochloric acid (37 %, 10 M) was then added dropwise to bring the solution to pH 3 and the solution was evaporated to a small volume. The solution was diluted with water (40 mL) and extracted with chloroform (3 x 100 mL). The combined organic extracts were dried and evaporated to afford a light brown solid. The solid was triturated with ether (50 mL) and filtered and washed with ether (50 mL) to afford 6-methyl-l-hydroxy-2(i//)- pyrazinone [15] as a light brow g, 11 %), having the formula:
Figure imgf000028_0003
[15] Synthesis of 5,6-dimethyl-l-hydroxy-2(lH)-pyrazinone
To a solution of glycine hydroxamic acid [14] (0.70 g, 7.777 mmol) in methanol (50 mL) and water (30 mL) at 0 °C was added a solution of butane-2,3-dione (o.78 mL, 1.15 equiv) in methanol (10 mL). Aqueous sodium hydroxide (1.0 mL, 2 M) was added dropwise over 5 minutes and the solution was stirred at 0 °C for 1 hour. The solution was then allowed to warm to room temperature and stirring was continued for 24 hours. Aqueous hydrochloric acid (37 %, 10 M) was then added dropwise to bring the solution to pH 3 and the solution was evaporated to a small volume. The solution was diluted with water (20 mL) and extracted with chloroform (5 x 50 mL). The combined organic extracts were dried and evaporated to afford a light brown solid. The solid was triturated with acetone (10 mL) and filtered and washed with acetone to afford 5,6-dimethyl-l-hydroxy-2(i//)-pyrazinone [16] as a light brown solid (0.26 g, 24 %), having the formula:
Figure imgf000029_0001
[16]
Testing Methodology
Analysis of tested compound toxicity in SH-SY-5Y neuroblastoma cells
SH-SY-5Y neuroblastoma cells were plated at 20,000 cells/well in a 96 well plate and left to adhere to the well surface and culture overnight in 50% advanced minimum essential medium (MEM), 50% Ham's F12 medium, 1% L-Glutamine and 2% fetal bovine serum (FBS). The media was then replaced with 100 μί^εΐΐ of FBS free MEM/Ham's F12 containing varying concentrations of the tested compound ranging from 0 to 300 μΜ. After a further 24 hour incubation at 37 °C in a 5% C02 environment cell viability was measured using the tetrazolium dye 3-(4,5- dimethylthiazol-2-yl)-2.5-diphenyltetrazolium bromide (MTT). For this colorimetric assay, 11 μΐ. of MTT (5 mg/mL) was added to the 100 μΐ. media in the well and incubated for 3 hours at 37 °C. After this time an equal volume of solubilizing solution (24 mL isopropyl alcohol, 1 mL HC1) was added to each well and thoroughly mixed to lyse the cells. Absorption was measured at 570 nm using a Tecan Sunrise spectrophotometer; model Sunrise-basic Tecan.
The cell toxicity of the tested compound on normal healthy neurons was calculated to obtain a non-toxic concentration. This was done by adding increasing concentrations of the tested compound to the cells to calculate the concentration which may be applied before the cells die due to toxicity, if any, of the tested compounds.
Cell viability was calculated as a % compared to the untreated control, i.e. in the absence of the tested compound. In particular, the cell viability was measured in the presence of 6-hydroxydopamine hydrobromide (6-OHDA), which synthetically induces Parkinson's disease in nerve cells, and the tested compound at the non-toxic concentration found in order to assess the efficacy in preventing neuro degeneration. The results for the cell viability of the tested compounds are as follows:
Analysis of tested compound neuroprotection to 6-OHDA toxicity in SH-SY-5Y neuroblastoma cells.
SH-SY-5Y neuroblastoma cells were plated at 20,000 cells/well in a 96 well plate and left to adhere to the well surface and culture overnight in 50% advanced minimum essential medium (MEM), 50% Ham's F12 medium, 1% L-Glutamine and 2% fetal bovine serum (FBS). Just before addition to cells, 6-hydroxydopamine hydrobromide (6-OHDA) was made to a stock concentration of 10 mM and stored in the dark at 4 °C. Media was then replaced with 100 μΙ/weW FBS free MEM/Ham's F12 containing 50 μΜ 6-OHDA and varying concentration of tested compound ranging from 0 to 300 μΜ. After a further 24 hour incubation at 37 °C in a 5% C02 environment cell viability was measured using the tetrazolium dye 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT). For this colorimetric assay, 11 of MTT (5 mg/mL) was added to the 100 media in the well and incubated for 3 hours at 37 °C. After this time an equal volume of solubilizing solution (24 mL isopropyl alcohol, 1 mL HC1) was added to each well and thoroughly mixed to lyse the cells. Absorption was measured at 570 nm using a spectrophotometer. Cell viability was calculated as a % compared to the untreated control, i.e. in the absence of 6-OHDA or the tested compound.
Measurement of the neuronal cytoplasmic labile iron pool using Calcein-AM assay.
The use of Calcein-AM to measure neuronal cytoplasmic labile iron pool was adapted from a previously reported procedure disclosed in Free Radic. Biol. Med. 2002, 33, 1037-1046. In brief, SH-SY5Y cells plated at 20,000 cells/well in a black 96-well microplate were treated with ferric ammonium citrate (FAC; 50 μΜ) for 6 hrs in serum-free media. Cells were washed twice with phosphate buffered saline (PBS) after which Calcein-AM (60 nM) was added. Fluorescence at an excitation of 485 nm and emission of 535 nm measurements were started immediately and taken every minute using a Biotek Fluorescence microplate reader for 10 min or until a consistent minimum reading had been reached. In triplicate, the compounds of interest (100 μΜ) were rapidly added and fluorescence measurements started again immediately. Readings were continued for a further 5 min or until a plateau had been reached. The percentage labile iron pool chelated was calculated as the AF (the difference in Fluorescence from before and after compound addition) compared to deferiprone (DFP) as a known lipid permeable chelator.
Analysis of iron and 6-OHDA changes to iron response proteins.
For investigating compound effect on iron response protein expression after iron loading, SH-SY5Y's prepared 6-well plates, were incubated in serum-free media with FAC (50 μΜ) for 24 hrs. After iron loading, iron containing media was replaced with fresh serum-free media alone or in the presence of the compound of interest (100 μΜ) and incubated for a further 12 hrs. To evaluate iron response protein expression to each compound on 6-OHDA, a similar procedure to the MTT assay was followed. Therefore, SH-SY5Y's prepared in 6-well plates, were incubated in serum- free media with 6-OHDA (50 μΜ) with or without the compound (100 μΜ) for 24 hrs. After each experimental condition, cells were washed twice with cold PBS, before collection and homogenizing in RIPA buffer (150 mM NaCl, 1% (v/v) Nonidet P-40, 1% (w/v) sodium deoxycholate, 0.1% (v/v) SDS, 25 mM Tris-HCl, pH 7.6) with EDTA-free protease inhibitor cocktail (Complete; Roche). Lysates were clarified by centrifugation at 14,000 x g for 15 min.
Western blot analysis.
As determined by BCA assay, 10 μg of the protein from each experimental condition was separated either on 10% PAGE (Tris-Glycine, BioRad) for 22C11 and Transferrin (TfR) or 4-20% PAGE (Tris-Glycine, BioRad) for Ferritin (FT), Tyrosine Hydroxylase (TH) and a-Synuclein (Syn). Resolved proteins transferred to polyvinylidene difluoride membranes (Hybond-P, Pierce) were probed with mouse anti-APP (1:1000, 22C11, in house), mouse anti-TfR (1:2000, H68.4, Invitrogen), rabbit anti-Ferritin (1:1000, Cell Signaling Technology), mouse anti-TH (1:1000, AB152, Millipore) or mouse anti-Syn (1:1000, D35E4, Cell Signaling Technology) and the appropriate secondary antibodies. The load control was mouse anti- -actin (1:5,000, AC15, Sigma). Proteins of interest were visualized with ECL (Pierce) and a LAS-3000 Imaging suite, and analyzed using Multi Gauge (Fuji). Densitometry using Image J (NIH) was performed in triplicate on 3 separate experiments. All quantitation was standardized against β-actin levels.
Results
The data tables below show the results of the analysis of the neuroprotection of the tested compounds to 6-OHDA toxicity. The compounds of the present invention provide comparable cell survival in the presence of 6-OHDA compared to the known compounds and higher non-toxic concentrations in the control. These results show that the treatment of healthy neuroblastoma cells (i.e. the control) with 50 μΜ 6-OHDA typically results in the death of approximately 50% of the cells. This is due to the oxidative stress and metal dysregulation, etc. initiated by 6-OHDA. Further, at relatively low concentrations, i.e. 3 μΜ to 30 μΜ, the known 3- hydroxypyridin-4-one compound (Deferiprone) and the compound of the present invention exhibit similar % cell viability in the 6-OHDA model of Parkinson's disease when compared to the control. At a concentration of 100 μΜ, the compound of the present invention exhibits cell viability comparable to the known 3-hydroxypyridin- 4-one. This reduction in cell death for the present invention is believed to occur as a result of the compound of the present invention binding to metal ions present, particularly iron(III), which prevents Fenton chemistry, stops the onset of oxidative stress and prevents metal-protein interactions.
Figures 1 to 12 graphically show the data in relation to the tables given below, and the compound identification numbers relate to the identifying numbers described below in relation to the data tables. The y-axis indicates percentage cell viability as compared to the control.
Figures 13 to 16 show pooled data obtained via analysis of the neuroprotection of the l-hydroxypyridin-2-one compound in accordance with the fifth embodiment of the present invention, and its analogues which lack the cyclic hydroxamic acid moiety and thus do not have the ability to chelate metals.
The results shown in Figures 13 to 16 show that, while the structural analogues may show a slight initial improvement on cell viability compared to the cells in the presence of 50 μΜ 6-OHDA, this improvement does not increase with concentration of the tested compound. Therefore, these compounds do not demonstrate a cell rescue effect, presumably as these compounds are unable to effectively chelate the free metal ions present, such as iron. In contrast, the results shown in Figure 13 show that the compound in accordance with the fifth embodiment of the present invention provides a greater improvement on cell viability, which increases as the concentration of the compound is increased. Therefore, these results demonstrate that the compounds of the present invention provide a cell rescue effect as a result of the strong chelation between the compound and the free metal ions.
Figure 17 shows that l-hydroxypyridin-2-one compound in accordance with the fifth embodiment of the present invention (Compound 17; Compound [9]) has comparable ability to bind the labile pool of iron (LIP) in the cell as the known 3- hydroxypyridin-4-one compound, Deferiprone (DFP). With reference to Figure 17A Calcein-AM is a fluorescent dye whose fluorescence is quenched by excess labile iron in the cell. The Calcein-AM assay indicates that the compound of the present invention is as efficient as Deferiprone (DFP) at causing Calcein fluorescence (which was originally quenched by the LIP upon incubation of the cells with 6-OHDA or iron), whereas the non-chelating compounds [8] (Compound 17A), [11] (Compound FL8) and [12] (Compound FL9) are unable to reduce LIP quenched calcein fluorescence. With reference to Figure 17B, the expression of the iron responsive proteins ferritin and transferrin receptor changes upon incubation of the cells with iron. Corresponding to Figure 17A, ferritin and transferrin receptor expression is restored to levels seen in a healthy neuron following incubation with iron in the presence of DFP and compound [9], while compound [12] (Compound FL9) has no effect. With reference to Figure 17C, ferritin and transferrin receptor expression are altered by 6-OHDA in a similar way as iron incubation and accordingly, both DFP and compound [9] reduce this response back to control levels whereas compound [12] (Compound FL9) is unable to do so.
These results show that the neuroprotective mode of action of the compounds of the present invention is chelation of the labile iron pool inside the cell.
Figure 18 illustrates the changes to known markers of Parkinson's disease toxicity with deferiprone (DFP) and compound [9] (Compound 17). Tyrosine hydroxylase and Synaptophysin levels are both reduced in Parkinson's disease and are similarly reduced in neurons after incubation with Iron (Figure 18A) and 6-OHDA (Figure 18B). While DFP and compound [9] are able to negate the reduced expression of both proteins to levels seen in a healthy neuron after incubation with iron (Figure 18A) or 6-OHDA (Figure 18B), the non-chelating compound [12] (Compound FL9) is unable to do so.
These results further show that the neuroprotective mode of action of the compounds of the present invention is chelation of the labile iron pool inside the cell.
Accordingly, in the 6-hydroxydopamine hydrobromide (6-OHDA) cellular model of Parkinson's disease, it has been shown that the compounds of the present invention offer an effective treatment for neurodegenerative disease based on strong metal chelation. This effectively stops the Fenton chemistry that leads to the formation of free-radical species and reactive oxygen species which lead to oxidative stress, reduces metal accumulation in the neuroblastoma cells and reduces metal interactions with alpha-synuclein and/or tau protein, which in turn reduces the accumulation of protein-metal aggregates capable of causing cell death. Therefore, the compounds of the present invention have been shown to be effective in delaying or halting the progression of Parkinson's disease to a similar extent to previously proposed metal chelators, thus providing an alternative. Further, in some situations, the compounds have been shown to reverse the damage caused by excess accumulation of metals. It follows that the compounds of the present invention will also be an effective treatment for other neurodegenerative diseases caused by disregulation/accumulation of metals in certain regions of the brain and central nervous system, such as Alzheimer's disease, Huntington's disease, macular degeneration, Friedreich's ataxia, amyotrophic lateral sclerosis and neurodegeneration with brain iron accumulation. Data Tables
(Key: Abs. = absorbance, AVG = average, StDev = standard deviation, 6-OHDA = 6- hydroxydopamine, n/a = not measured)
Table 1 shows compound toxicity in absence of 6-OHDA for compound [9],
Table 2 shows cell rescue effect in presence of 6-OHDA for compound [9].
Compound [9] is also identified as Compound 17 in the Tables and Figures.
Figure imgf000036_0001
[9]
Table 3 shows compound toxicity in absence of 6-OHDA for compound [16], Table 4 shows cell rescue effect in presence of 6-OHDA for compound [16].
Compound [16] is also identified as Compound FLl in the Tables and Figures.
Figure imgf000036_0002
[16]
Table 5 shows compound toxicity in absence of 6-OHDA for compound [15], Table 6 shows cell rescue effect in presence of 6-OHDA for compound [15].
Compound [15] is identified as Compound FL2 in the Tables and Figures.
Figure imgf000036_0003
[15]
Table 7 shows compound toxicity in absence of 6-OHDA for compound [5], Table 8 shows cell rescue effect in presence of 6-OHDA for compound [5] . Compound [5] is also identified as Compound 7 in the Tables and Figures.
Figure imgf000037_0001
Table 9 shows compound toxicity in absence of 6-OHDA for compound [6], Table 10 shows cell rescue effect in presence of 6-OHDA for compound [6]. Compound [6] is also identified as Compound 8 in the Tables and Figures.
Figure imgf000037_0002
Table 11 shows compound toxicity in absence of 6-OHDA for compound [3], Table 12 shows cell rescue effect in presence of 6-OHDA for compound [3]. Compound [3] is also identified as Compound 14 in the Tables and Figures.
Figure imgf000037_0003
Table 13 shows compound toxicity in absence of 6-OHDA for compound [8], Table 14 shows cell rescue effect in presence of 6-OHDA for compound [8]. Compound [8] is also identified as Compound 17A in the Tables and Figures.
Figure imgf000038_0001
Table 15 shows compound toxicity in absence of 6-OHDA for compound [11], Table 16 shows cell rescue effect in presence of 6-OHDA for compound [11]. Compound [11] is also identified as Compound FL8 in the Tables and Figures.
Figure imgf000038_0002
[11]
Table 17 shows compound toxicity in absence of 6-OHDA for compound [12], Table 18 shows cell rescue effect in presence of 6-OHDA for compound [12]. Compound [12] is also identified as Compound FL9 in the Tables and Figures.
Figure imgf000038_0003
Table 19 shows compound toxicity in absence of 6-OHDA for 3-hydroxypyridin-4- one (Deferiprone), Table 20 shows cell rescue effect in presence of 6-OHDA for Deferiprone. This compound is also identified as Compound 11 in the Tables and Figures, and is shown below.
Figure imgf000039_0001
Table 21 shows compound toxicity in absence of 6-OHDA for compound [17], Table 22 shows cell rescue effect in presence of 6-OHDA for compound [17]. This compound is also identified as Compound 2 in the Tables and Figures, and is shown below.
Figure imgf000039_0002
[17]
Table 23 shows compound toxicity in absence of 6-OHDA for 3-hydroxypyran-4-one (Maltol), Table 24 shows cell rescue effect in presence of 6-OHDA for Maltol. This compound is also identified as Compound 10 in the Tables and Figures, and is shown below.
Figure imgf000039_0003
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Table 4.
Figure imgf000043_0001
Figure imgf000044_0001
Table 6.
Figure imgf000045_0001
Figure imgf000046_0001
Table 8.
Figure imgf000047_0001
Figure imgf000048_0001
Table 10.
Figure imgf000049_0001
Figure imgf000050_0001
Table 12.
Figure imgf000051_0001
Figure imgf000052_0001
Table 14.
Figure imgf000053_0001
Figure imgf000054_0001
Table 16.
Figure imgf000055_0001
Figure imgf000056_0001
Table 18.
Figure imgf000057_0001
Figure imgf000058_0001
Table 20.
Figure imgf000059_0001
Figure imgf000060_0001
Table 22.
Figure imgf000061_0001
Figure imgf000062_0001
Table 24.
Figure imgf000063_0001

Claims

Claims
1. A heterocycle compound containing at least one electronegative atom and comprising a hydroxyl group or substituted hydroxyl group at the 1 -position and the carbon at the 2 -position being a carbonyl group for use as a medicament.
2. The heterocycle of claim 1 wherein the compound contains at least one oxygen atom, at least one nitrogen atom or a combination thereof.
3. The heterocycle of claim 1 or 2 wherein the compound contains at least one nitrogen atom.
4. The heterocycle of any preceding claim, wherein the compound is a nitrogen containing six membered heterocycle having the general formula:
Figure imgf000064_0001
(I) wherein X, Y, Z and X' are independently selected from the group consisting of C, N, 0 or S;
R1 is selected from the group comprising H, an alkyl, an aryl, a benzyl, an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle; and
R2 and/or R3 are absent or comprises at least one additional substituent selected to provide additional functionality to the heterocycle or a pharmaceutically acceptable salt and/or an isomer thereof.
5. The heterocycle of claim 4, wherein at least one of X, Y, Z and X' are carbon atoms.
6. The heterocycle of claim 4 or 5, which further comprises from 1 to 4 additional substituents, which are independently selected from the groups comprising:
F, CI, Br or I;
OH, OR, OCOR, NH2, NHR4, NR4R5, +NR4R5R6, C02R4, C0NHR4, CONR4R5 or C=0R4;
R4OR5;
a substituted or unsubstituted alkyl, heteroalkyl, alkenyl, heteroalkenyl or alkynyl, which may be linear, branched or cyclic;
a substituted or unsubstituted aryl or heteroaryl;
S03-, X+, SO3H, SH or SR4;
P=0(OH)2 or salts P=0(OR4)2 or P=0(OR4)(OR5);
S=0R4 or 0=S=0R4;
B(OH)2 or B(OR4)(OR5);
CN;
N02; or
C(=NH)(NH2), C=SR4;
wherein R4, R5 and R6 may be independently selected from the group consisting of: H; OH; 0(CH2)XCH3; (CH2)XCH3; a alkyl, heteroalkyl, alkenyl, heteroalkenyl or alkynyl, which may be linear, branched or cyclic; a methylene moiety; an acyl; a sulfanyl; an aryl; a heteroaryl; or an antioxidant moiety; where x = 0 to 12, and X+ may be independently selected from the group consisting of Na+, K+, Li+, Mg2+, Ca2+ or an organic cation.
7. The heterocycle of any one of claims 4 to 6, wherein at least two additional substituents are present, and the additional substituents may be linked to form a cyclic structure. The heterocycle of claim any of claim 4 to 7, wherein the additional substituents may be linked together via a linking group, such as "OCH2CH2O, to form a bis-, tris- or tetrakis structure.
The heterocycle of any one of claims 1 to 8, wherein the heterocycle is a nitrogen containing six membered heterocycle having the formula:
Figure imgf000066_0001
wherein X and Y are independently selected from C or N;
R1 is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle;
R2 is an alkyl, an aryl, an heteroaryl, OH, OR', SH, SR', S(0)R', S02R', NH, NR', sulfinyl, sulfonyl, F, CI, Br, I, C=0, COOR', C=NR' or R5OR6, wherein R' is H, alkyl, cycloalkyl, heterocycloalkyl, acyl, aryl or heteroaryl, R5 is (CH2)X wherein x = 0 to 100 and R6 is H, an alkyl, an acyl, an aryl, a sulfinyl, a sulfonyl, an antioxidant moiety or any other functional groups, such as groups which help deliver the nitrogen containing six membered heterocycle to a certain region of the nerve cell or assist in crossing the blood-brain barrier; and
R3 and/or R4 are each independently absent or present on the ring and comprise at least one additional substituent selected to provide additional functionality to the heterocycle; or a pharmaceutically acceptable salt and/or an isomer thereof.
10. The heterocycle of any one of claims 1 to 9, wherein the heterocycle a nitrogen containing six membered heterocycle having the formula:
Figure imgf000067_0001
(III) wherein R is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle;
R1 is (CH2)x, wherein x = 0 to 100;
R2 is H, an alkyl, an acyl, an aryl, a sulfanyl, an antioxidant moiety or any other functional groups, such as groups which help deliver the nitrogen containing six membered heterocycle to a certain region of the nerve cell or assist in crossing the blood-brain barrier; and
R3 is absent or present at at least one of the 3-, 4- or 5 -positions on the ring and comprises at least one additional substituent selected to provide additional functionality to the heterocycle
or a pharmaceutically acceptable salt and/or an isomer thereof.
11. The heterocycle according to claim 10, wherein R is H and so the nitrogen containing six membered heterocycle is l-hydroxy-6- (hydroxymethyl)pyridin-2-one or a pharmaceutically acceptable salt and/or an isomer thereof.
12. The heterocycle according to any one of claims 1 to 10, wherein the heterocycle is a nitrogen containing six membered heterocycle having the formula:
Figure imgf000068_0001
(IV) wherein R is selected from the group comprising H, an alkyl, an aryl, a benzyl an ester, an amide, a carbamate, a carbonate, a sulfinate, a sulfonate, a phosphonate, an alkali metal, an alkaline earth metal, an ammonium salt, a phosphonium salt, a sulfonium salt, an O-glycoside, an antioxidant, a protecting group, a group capable of being cleaved enzymatically in the cell to reveal the oxygen for chelation or a substituent selected to provide additional functionality to the heterocycle;
or a pharmaceutically acceptable salt and/or an isomer thereof.
13. A method of forming the nitrogen containing six membered heterocycle according to claim 10 to 12 comprising the steps of:
providing a l-hydroxypyridin-2-one-6-carboxylic acid methyl ester; reacting the l-hydroxypyridin-2-one-6-carboxylic acid methyl ester with a protecting group to protect the N-hydroxyl group, thereby forming a protected l-hydroxypyridin-2-one-6-carboxylic acid methyl ester;
reduction of the methyl ester group of the protected 1-hydroxypyridin- 2-one-6-carboxylic acid methyl ester to form protected l-hydroxy-6- hydroxymethyl-pyridin-2-one; and
deprotecting of the N-hydroxyl group of the protected l-hydroxy-6- hydroxymethyl-pyridin-2-one by removal of the protecting group to form the nitrogen containing heterocycle of any of claims 10 to 12.
14. The method of claim 13 wherein the l-hydroxypyridin-2-one-6-carboxylic acid methyl ester is provided by reaction of a l-hydroxypyridin-2-one-6- carboxylic acid with thionyl chloride in methanol, or the l-hydroxypyridin-2- one-6-carboxylic acid is synthesised by treatment of 6-hydroxypicolinic acid with peracetic acid in acetic acid.
15. The method of claim 13 or 14 wherein, the protecting group is an allyl, benzyl, silyl or sulfonate ester group, or any other protecting group known in the art.
16. The method of claim 15, wherein the protecting group is an allyl.
17. The method of any one of claims 13 to 16, wherein reduction of the ester group of the l-hydroxypyridin-2-one-6-carboxylic acid methyl ester is accomplished by reaction with a reducing agent, such as a reducing agent comprising potassium, calcium, barium, sodium or magnesium and/or borohydride.
18. The method of any one of claims 13 to 17 wherein the deprotection of the N- hydroxyl group of the protected l-hydroxy-6-hydroxymethyl-pyridin-2-one by removal of the protecting group is accomplished in the presence of palladium on activated carbon and trifluoroacetic acid.
19. The heterocycle of any preceding claim for use in the treatment of a neurodegenerative disease.
20. The heterocycle of any preceding claim for use in treatment of Parkinson's disease or Alzheimer's disease.
21. A method for treating a neurodegenerative disease comprising the steps of: providing a composition comprising a heterocycle compound according to any one of claim 1 to 12; and
administering an effective dose of the composition to a patient.
22. The method of claim 21, wherein the composition further comprises a pharmaceutically acceptable excipient.
23. The method of claim 22 or 23, wherein the composition comprises one or more known active pharmaceutical ingredient used in the treatment of symptoms of neurodegenerative disease, such as known active pharmaceutical ingredients including additional chelating compounds or antioxidants.
24. A composition for the treatment of a neurodegenerative disease comprising a heterocycle in accordance with any one of claims 1 to 12, optionally a pharmaceutically acceptable excipient and optionally one or more known pharmaceutical ingredients.
PCT/GB2015/053492 2014-11-17 2015-11-17 Metal chelating compounds for use in treating diseases WO2016079502A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB1420348.3 2014-11-17
GBGB1420348.3A GB201420348D0 (en) 2014-11-17 2014-11-17 Compounds for treating neurodegenerative diseases

Publications (1)

Publication Number Publication Date
WO2016079502A1 true WO2016079502A1 (en) 2016-05-26

Family

ID=52248427

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2015/053492 WO2016079502A1 (en) 2014-11-17 2015-11-17 Metal chelating compounds for use in treating diseases

Country Status (2)

Country Link
GB (1) GB201420348D0 (en)
WO (1) WO2016079502A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111945181A (en) * 2020-08-17 2020-11-17 湖南科技学院 Electrochemical synthesis method of 3-alkylselenoquinolinone compound

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0241918A2 (en) * 1986-04-18 1987-10-21 Hoechst Aktiengesellschaft 1-Hydroxy-2-pyridinones, process for their preparation and medicines containing them, as well as intermediates formed by the preparation of 1-hydroxy-2-pyridinones
GB2345058A (en) * 1998-12-01 2000-06-28 Cerebrus Pharm Ltd Hydroxypyridone compounds useful in the treatment of oxidative damage to the central nervous system
WO2004041151A2 (en) * 2002-11-07 2004-05-21 Technion Research And Development Foundation Ltd. Neuroprotective iron chelators and pharmaceutical compositions comprising them
WO2009103950A1 (en) * 2008-02-19 2009-08-27 Btg International Limited Fluorinated pyridin-4-0nes
WO2011153197A1 (en) * 2010-06-01 2011-12-08 Biotheryx, Inc. Hydroxypyridone derivatives, pharmaceutical compositions thereof, and their therapeutic use for treating proliferative diseases
WO2013143477A1 (en) * 2012-03-28 2013-10-03 Sun Weixin 1,4-dihydroxy-6-methyl-2-pyridone compound, and preparation method and application thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0241918A2 (en) * 1986-04-18 1987-10-21 Hoechst Aktiengesellschaft 1-Hydroxy-2-pyridinones, process for their preparation and medicines containing them, as well as intermediates formed by the preparation of 1-hydroxy-2-pyridinones
GB2345058A (en) * 1998-12-01 2000-06-28 Cerebrus Pharm Ltd Hydroxypyridone compounds useful in the treatment of oxidative damage to the central nervous system
WO2004041151A2 (en) * 2002-11-07 2004-05-21 Technion Research And Development Foundation Ltd. Neuroprotective iron chelators and pharmaceutical compositions comprising them
WO2009103950A1 (en) * 2008-02-19 2009-08-27 Btg International Limited Fluorinated pyridin-4-0nes
WO2011153197A1 (en) * 2010-06-01 2011-12-08 Biotheryx, Inc. Hydroxypyridone derivatives, pharmaceutical compositions thereof, and their therapeutic use for treating proliferative diseases
WO2013143477A1 (en) * 2012-03-28 2013-10-03 Sun Weixin 1,4-dihydroxy-6-methyl-2-pyridone compound, and preparation method and application thereof

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
HOSSEIN HELI ET AL: "Advances in iron chelation: an update", EXPERT OPINION ON THERAPEUTIC PATENTS., vol. 21, no. 6, 1 June 2011 (2011-06-01), GB, pages 819 - 856, XP055242680, ISSN: 1354-3776, DOI: 10.1517/13543776.2011.569493 *
JIDE XU ET AL: "Synthesis and Initial Evaluation for In Vivo Chelation of Pu(IV) of a Mixed Octadentate Spermine-Based Ligand Containing 4-Carbamoyl-3-hydroxy-1-methyl-2(1 H )-pyridinone and 6-Carbamoyl-1-hydroxy-2(1 H )-pyridinone", JOURNAL OF MEDICINAL CHEMISTRY, vol. 45, no. 18, 1 August 2002 (2002-08-01), US, pages 3963 - 3971, XP055242703, ISSN: 0022-2623, DOI: 10.1021/jm010564t *
NOAH BIRCH ET AL: "Iron chelators as therapeutic iron depletion agents", EXPERT OPINION ON THERAPEUTIC PATENTS., vol. 16, no. 11, 1 November 2006 (2006-11-01), GB, pages 1533 - 1556, XP055242688, ISSN: 1354-3776, DOI: 10.1517/13543776.16.11.1533 *
THORSON MEGAN K ET AL: "Inhibition of the lymphoid tyrosine phosphatase: The effect of zinc(II) ions and chelating ligand fragments on enzymatic activity", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 24, no. 16, 16 June 2014 (2014-06-16), pages 4019 - 4022, XP029041692, ISSN: 0960-894X, DOI: 10.1016/J.BMCL.2014.06.016 *
ZHEN LIU ET AL: "Inhibition of Cancer-Associated Mutant Isocitrate Dehydrogenases: Synthesis, Structure-Activity Relationship, and Selective Antitumor Activity", JOURNAL OF MEDICINAL CHEMISTRY, vol. 57, no. 20, 23 October 2014 (2014-10-23), US, pages 8307 - 8318, XP055242812, ISSN: 0022-2623, DOI: 10.1021/jm500660f *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111945181A (en) * 2020-08-17 2020-11-17 湖南科技学院 Electrochemical synthesis method of 3-alkylselenoquinolinone compound

Also Published As

Publication number Publication date
GB201420348D0 (en) 2014-12-31

Similar Documents

Publication Publication Date Title
TWI827646B (en) Ptpn11 inhibitors
JP6824876B2 (en) Amidine derivative compositions and their use
KR101122782B1 (en) Neurologically-active compounds
WO2008074068A1 (en) Substituted quinoline derivatives as antiamyloidogeneic agents
JP2014012706A (en) Compound having anticancer activity
US20180111904A1 (en) Dimeric quinacrine derivatives as autophagy inhibitors for cancer therapy
KR20220029681A (en) Methods of Treatment of Idiopathic Pulmonary Fibrosis
CA3009850A1 (en) Deuterated compounds for treating cancer and related diseases and conditions, and compositions and methods thereof
US20160008336A1 (en) Hdac inhibitors for the treatment of traumatic brain injury
US8633174B2 (en) Compounds for use in the treatment of cancer
JP2021512075A (en) Inhibition of transient receptor potential A1 ion channels
CN103391932A (en) Heterocyclic compounds for the inhibition of pask
WO2015031109A1 (en) Inhibition of wnt, tgf beta and hippo signaling pathways to treat cancer, organ fibrosis and metabolic disorders
CN112261941A (en) Aminopyrazines and related compounds useful as mitochondrial uncouplers
KR20030063360A (en) Modified prodrug forms of AP/AMP
WO2016079502A1 (en) Metal chelating compounds for use in treating diseases
CA2883443A1 (en) Compositions and methods for drug-sensitization or inhibition of a cancer cell
EP4154890A1 (en) Pharmaceutical composition for preventing or treating pancreatic cancer associated with ron mutation and method using same
CN112654626A (en) Compound and use thereof
JP2021503442A (en) Kinase inhibitors for the treatment of central and peripheral nervous system disorders
EP3012248A1 (en) Substance having tyrosine kinase inhibitory activity and preparation method and use thereof
WO2016160706A1 (en) Inhibitors of collagen prolyl 4-hydroxylase
US10689394B2 (en) Thieno[2,3-b]pyridine derivative, quinoline derivative, and use thereof
CN111247148B (en) Wnt pathway modulators
WO2024112397A1 (en) Combination therapies comprising kras inhibitors and sph2 inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15804183

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15804183

Country of ref document: EP

Kind code of ref document: A1