EP1027335A1 - Derives d'ortho-hydroxypyridinone utilises comme agents chelateurs du fer et comme agents antioxydants - Google Patents

Derives d'ortho-hydroxypyridinone utilises comme agents chelateurs du fer et comme agents antioxydants

Info

Publication number
EP1027335A1
EP1027335A1 EP98950227A EP98950227A EP1027335A1 EP 1027335 A1 EP1027335 A1 EP 1027335A1 EP 98950227 A EP98950227 A EP 98950227A EP 98950227 A EP98950227 A EP 98950227A EP 1027335 A1 EP1027335 A1 EP 1027335A1
Authority
EP
European Patent Office
Prior art keywords
compound according
alkyl
methyl
hydroxy
butyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP98950227A
Other languages
German (de)
English (en)
Inventor
David Cerebrus Limited BEBBINGTON
Nat Cerebrus Limited MONCK
Suneel Cerebrus Limited GAUR
Alan Cerebrus Limited PALMER
Richard Cerebrus Limited PORTER
Craig Cerebrus Limited MALCOLM
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ligand UK Research Ltd
Original Assignee
Cerebrus Pharmaceuticals Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cerebrus Pharmaceuticals Ltd filed Critical Cerebrus Pharmaceuticals Ltd
Publication of EP1027335A1 publication Critical patent/EP1027335A1/fr
Withdrawn legal-status Critical Current

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
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/69Two or more oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • Stroke is the third leading cause of death in major industrialised countries and the commonest cause of permanent disability (Hunter et al, Trends in Pharmacological Sciences, 1995, 16, 123-128). Each year, in the US and Europe, approximately 1 million people suffer acute stroke (Dorman et al, CNS Drugs, 1996, 5, 457-474). Between 25% and 35% of these patients die within the first three weeks, and of the survivors 25% to 0 50%) will be totally dependant on family or institutional care for the rest of their lives.
  • Stroke is defined as an interruption of the blood flow to the brain or leakage of blood out of the brain, resulting in oxygen deprivation (ischaemia) and subsequent neuronal cell 0 death. Strokes can be divided into two classes, ischaemic and haemorragic. The former accounts for approximately 83% of all strokes and is caused by thrombosis (65%o) and/or detachment of a previously formed clot (embolus, 18%>). Haemorrhagic strokes, which account for the remaining 17%> of all strokes, can be subdivided into subarachnoid haemorrhage (7%) and cerebral haemorrhage (10%).
  • Altepase ® tissue plasminogen activator, rTPA
  • rTPA tissue plasminogen activator
  • Therapeutic thrombolysis can, however, be complicated by a) systemic haemorrhage, b) intracerebral haemorrhage, c) distal embolism of a partially digested clot leading to secondary infarction and d) cerebral oedema secondary to reperfusion injury. It is, therefore, necessary to exclude the possibility of haemorrhagic stroke by computerised tomographic (CT) scanning of patients before administering Alteplase.
  • CT computerised tomographic
  • R 1 , R 2 and R 3 are independently selected from H and alkyl; wherein X is O, S, NR 4 or a direct bond, wherein R 4 is H or alkyl; wherein Z is a saturated hydrocarbyl chain comprising from 1 to 10 carbon atoms; wherein q is 1, 2 or 3, wherein if q is 2 or 3, then each A can be the same or different; wherein the or each R 5 is independently selected from H or alkyl; wherein the or each R 6 is independently selected from alkyl; wherein n is 1 to 5; wherein p is 0 to 4; and wherein the sum of n and p is less than 6, or a pharmaceutically acceptable salt thereof.
  • the compounds of the present invention which have a combined antioxidant and iron-chelating activity can be used for treating oxidative stress, particularly oxidative damage to the central nervous system.
  • the compounds of the present invention are surprisingly more effective in vitro, especially at low concentrations, than the simultaneous use of the separate hydroxypyridone iron-chelating compound and the phenolic antioxidant compound.
  • alkyl means a branched or unbranched, cyclic or acyclic, saturated or unsaturated (e.g. alkenyl or alkynyl) hydrocarbyl radical.
  • the alkyl group is preferably C 3 to C, 2 , more preferably C 5 to C, resume, more preferably C 5 to C 7 .
  • the alkyl group is preferably C, to C 10 , more preferably C, to C 6 , more preferably methyl, ethyl, propyl, isopropyl, butyl, t-butyl, pentyl and iso-pentyl.
  • aminocarbonyl e.g. aminocarbonyl, mono- or dialkylaminocarbonyl, aminocarbonylalkyl, mono- or dialkylaminocarbonylalkyl, arylaminocarbonyl
  • nitrogen containing groups such as amines (e.g. amino, mono- or dialkylamino, aminoalkyl, mono- or dialkylaminoalkyl).
  • halogen means a fluorine, chlorine, bromine or iodine radical, preferably a fluorine or chlorine radical.
  • salts may be prepared from pharmaceutically acceptable non-toxic acids and bases including inorganic and organic acids and bases.
  • acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, dichloroacetic, furnaric, gluconic, glutamic, hippuric, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, oxalic, p-toluenesulfonic and the like.
  • hydrochloric, hydrobromic, phosphoric, sulfuric and methanesulfonic acids particularly preferred are hydrochloric, hydrobromic, phosphoric, sulfuric and methanesulfonic acids, and most particularly preferred is the methanesulfonate salt.
  • Acceptable base salts include alkali metal (e.g. sodium, potassium), alkaline earth metal (e.g. calcium, magnesium) and aluminium salts.
  • R 2 and R 3 are independently selected from H, unsubstituted alkyl, CH 2 OR 7 , CH 2 OCOR 7 , COOR 7 , CH 2 NHR 7 , CH 2 NHCOR 7 and CONHR 7 wherein R 7 is H or alkyl.
  • R 1 is selected from H and unsubstituted alkyl. More preferably, R 1 , R 2 and R 3 are independently selected from H and unsubstituted alkyl. More preferably, R 1 and R 2 are H and R 3 is unsubstituted alkyl. It is further preferred that R 3 is methyl.
  • R 1 , R 2 and R 3 are independently selected from H, unsubstituted alkyl, CH 2 OR 7 , CH 2 OCOR 7 , COOR 7 , CH 2 NHR 7 , CH 2 NHCOR 7 and CONHR 7 wherein R 7 is H or alkyl.
  • R 1 , R 2 and R 3 are independently selected from H and unsubstituted alkyl. More preferably, R ! , R 2 and R 3 are H.
  • the present invention provides compounds wherein A is Al.
  • the present invention provides compounds wherein Z is (CH 2 ) m wherein m is 1 to 10.
  • the present invention provides compounds wherein Z is a hydrocarbyl chain having from 2 to 10 carbon atoms, preferably from 2 to 6 carbon atoms, more preferably 2, 3 or 4 carbon atoms.
  • the present invention provides compounds wherein Z is a hydrocarbyl chain having from 1 to 6 carbon atoms, preferably from 2 to 6 carbon atoms, more preferably 2, 3 or 4 carbon atoms.
  • the saturated hydrocarbyl chain Z may be branched or unbranched, optionally substituted by one or more alkyl groups, and may be cyclic.
  • cyclic means either that Z may comprise a cyclic hydrocarbyl group of from 3 to 10 carbon atoms, preferably 5, 6 or 7 carbon atoms; or that a cyclic group is present as a result of cyclisation of R 5 or R 6 onto Z; or that, where X is NR 4 and R 4 is alkyl, a cyclic group is present as a result of cyclisation of R 4 onto Z. It is preferred that a cyclic group formed as a result of cyclisation of R 4 , R 5 or R 6 onto Z is a 5, 6 or 7-membered ring.
  • Z is an unbranched hydrocarbyl chain.
  • the present invention provides compounds wherein X is O, S or a direct bond, more preferably X is O or a direct bond.
  • R 4 is preferably alkyl
  • R 4 when X is NR 4 and R 4 is alkyl, R 4 may be cyclized onto the chain defined as Z.
  • R 4 may be cyclized onto the chain defined as Z.
  • An example of a compound of formula (1) where R 4 is cyclized onto the chain defined as Z is:
  • R 5 is preferably selected from H and C,. 10 alkyl. Where R s is C,. 10 alkyl, R 5 is preferably methyl. Most preferably, R 5 is H. It is preferred that at least one R 5 is H.
  • the present invention provides compounds wherein n is 1 to 3, more preferably n is 1 or 2.
  • R 5 is H
  • OR 5 is positioned in the ortho or para position in the ring with respect to X. More preferably OR 5 is positioned in the para position with respect to X.
  • n is greater than 1, it is preferred that the OR 3 groups are positioned ortho to each other to give, for example, a compound of formula:
  • the or each R 6 is preferably independently selected from C,. 10 alkyl, preferably C M alkyl, most preferably methyl, isopropyl or t-butyl.
  • the present invention provides compounds wherein p is 2, 3 or 4.
  • alkyl groups represented by R 6 is/are preferably positioned ortho to OR 5 , preferably in the meta-position of the ring with respect to X to give, for example, a compound of formula:-
  • alkyl groups represented by R 6 are preferably .positioned to give, for example where three R 6 are methyl, a compound of formula: -
  • R 5 or R 6 can be cyclized on to the chain defined as Z to form a ring.
  • An example of a compound of formula (1) where R 5 is cyclized on to the chain defined as Z and where X is a direct bond is:
  • A is Al, R 1 and R 2 are H, R 3 is methyl, m is 2 or 3, X is a direct bond, R 5 is H, R 6 is t-butyl, n is 1 and p is 2.
  • the compound of formula (1) is l-(3-(3,5-di-tert-butyl-4- hydroxyphenyl)propyl)-3-hydroxy-2-methyl-4(lH)-pyridinone (2a):
  • A is Al, R 1 and R 2 are ⁇ , R 3 is methyl, m is 3, X is O, R 5 is ⁇ , R 6 is methyl, n is 2 and p is 4.
  • the compound of formula (1) is l-(2-(2,3-dihydro-5-hydroxy-4,6,7- trimethylbenzofuran-2-yl)ethyl)-3-hydroxy-2-methyl-4(lH)-pyridinone (2b):
  • the compounds of the present invention may be prepared using standard synthetic chemistry.
  • a general method for the synthesis of compounds where q- ⁇ and A is a 3-hydroxy-4(lH)- pyridinone moiety comprises condensation of the primary amine of the respective antioxidant unit (3) with 3-benzyloxy-2-methyl-4-pyrone (4), followed by removal of the benzyl protecting group, as illustrated in Reaction Scheme 1.
  • condensation of (4) with simple primary amines see Dobbin et al, J. Med. Chem., 1993, 36, 2448-2458).
  • the primary amines, of the respective antioxidants, (3) can be prepared using standard synthetic chemistry.
  • the -OH of the antioxidant unit can be optionally protected during synthetic manipulation (for example, as a benzyl ether). Deprotection to reveal the -OH of the antioxidant unit can be carried out simultaneously with removal of the benzyl protecting group of the hydroxy pyridone unit, in the last step of the sequence.
  • an alternative electrophilic alkylating derivative of the antioxidant unit for example, mesylate or tosylate
  • the primary halides, (mesylates and tosylates) of the antioxidants, (6) can be prepared using standard synthetic chemistry.
  • the -OH of the antioxidant unit can be optionally protected during synthetic manipulation (for example, as a benzyl ether). Deprotection to reveal the -OH of the antioxidant unit can be carried out simultaneously with removal of the methyl protecting group of the hydroxy pyridone unit, in the last step of the sequence.
  • the bis-primary amine (7) may be prepared using standard synthetic chemistry.
  • Reaction Scheme 4 illustrates a method of preparation of bis-primary amine (8).
  • a compound is an intermediate in the synthesis of a compound of the type exemplified as Example 11 herein.
  • the bis-primary amine (8) may be prepared from bromide (9) via reaction with dimethylmalonate (in the presence of a suitable base, for example NaH) to produce the dimethylester, followed by reduction (using for example BH 3 .Me 2 S) to produce the diol (10).
  • Diol (10) may then be converted to the dimesylate (using methanesulphonyl chloride and a suitable base, for example triethylamine), reacted with sodium azide, and reduced (using for example H 2 and Pd/C) to produce the bis-primary amine (8) (Palmer et al J. Med Chem., 1990, 33, 3008-3014).
  • a suitable base for example triethylamine
  • the bis-primary amine (8) may be prepared from the bis-diamide (11) via reduction (with for example lithium aluminium hydride, or borane), as illustrated in Reaction Scheme 5.
  • the bis-amide (11) may be prepared using standard synthetic chemistry procedures.
  • the bis-diamide (11) may be prepared from the benzyl bromide (9); via reaction with malonamide in the presence of a suitable base (for example NaOH in liquid ammonia) (Asami et al Sci. Rep. Res. Inst., 1957, 335-337); or alternatively via reaction with diethyl malonate in the presence of a suitable base (for example NaH in DMF) to produce the diester, followed by di-amidation (using for example ammonia).
  • a suitable base for example NaOH in liquid ammonia
  • a suitable base for example NaH in DMF
  • bis-diamide (11) may be prepared from benzaldehyde (12), via reaction with diethylmalonate in the presence of a suitable base (for example piperidine in EtOH) to produce the diester olefin, followed by reduction of the olefin (using for example H 2 and Pd/C), and finally di-amidation (using for example ammonia in EtOH) (Sekiya et al. Chem. Pharm. Bull., 1964, 12, 674-677); or via reaction with malononitrile in the presence of a suitable base (for example piperidine in EtOH) to produce the dinitrile olefin (Gazit et al, J. Med.
  • a suitable base for example piperidine in EtOH
  • Reaction Scheme 7 illustrates a method of preparation of a bis-primary amine (13).
  • a bis-primary amine 13
  • Such a compound is an intermediate in the synthesis of a compound of the type exemplified as Example 12 herein.
  • the bis-primary amine (13) may be prepared from aldehyde (12) via reaction with nitromethane in the presence of a suitable base (using for example catalytic butyl amine) to produce the dinitro compound (14) (Cassels et al. Rev. Latinoam. Quim. 1988, 1_9, 25-8), followed by reduction (with for example with lithium aluminium hydride, or H 2 and Pd/C, or H 2 and Raney-nickel).
  • the bis-primary amine (13) may be prepared from aldehyde (12) via reaction with cyanoacetic acid in the presence of a suitable base (using for example pyridine and sodium acetate in toluene) to produce the dinitrile (Erion et al J. Med. Chem., 1993, 36, 3771-3783), followed by hydrolysis (using for example aqueous sulphuric acid) to produce the bis-amide (15), and finally Hofmann rearrangement (using for example NaOH and bromine) to produce the bis- primary amine (13) (Weinhardt et al. J. Med Chem., 1985, 28, 694-698). Reaction Scheme 7
  • bis-primary amine (13) may be prepared from chloroamide (16) via reaction with sodium cyanide (in a suitable solvent such as DMF), followed by reduction (using for example lithium aluminium hydride) (Jahn et al, Can. J. Chem., 1988, 66, 123-131), as illustrated in Reaction Scheme 8.
  • the compounds of the present invention may contain one or more asymmetric carbon atoms, so that the compounds exist in different stereoisomeric forms.
  • the compounds can be, for example, racemates or optically active forms.
  • the optically active forms can be obtained by resolution of the racemates or by asymmetric syntheses.
  • the compounds of the present invention may also be prepared in a prodrug form wherein some or all the free -OH groups of the preferred compounds are derivatised (for example, via an ester, amide or phosphate bond) with a suitable group (the group may contain, for example, an alkyl, aryl, phosphate, sugar, amine, glycol, sulfonate or acid function) which is suitably labile so as it will be removed / cleaved (eg. by hydrolysis) to reveal the preferred compound sometime after administration or when exposed to the desired biological environment.
  • a suitable group the group may contain, for example, an alkyl, aryl, phosphate, sugar, amine, glycol, sulfonate or acid function
  • labile eg. by hydrolysis
  • compounds of the present invention may also contain additional non-covalently linked components such as dextrans or cyclodextrins, which aid stability and dispersion, and decrease metabolism of the active ingredient.
  • additional non-covalently linked components such as dextrans or cyclodextrins, which aid stability and dispersion, and decrease metabolism of the active ingredient.
  • a compound of the present invention in the manufacture of a medicament for the treatment of a condition resulting in oxidative stress, particularly oxidative damage of the central nervous system.
  • treatment includes prophylaxis.
  • Diseases, disorders and medical treatments/procedures resulting in oxidative stress include: aging; acute intermittent porphyria; adriamycin-induced cardiomyopathy; AIDS dementia and HIV-l induced neurotoxicity; Alzheimer's disease; atherosclerosis; cateract; cerebral ischaemia; cerebral palsy; cerebral tumour; chemotherapy-induced organ damage; cisplatin-induced nephrotoxicity; coronary artery bypass surgery; diabetic neuropathy; Down's syndrome; drowning; epilepsy and post-traumatic epilepsy; Friedrich's ataxia; frontotemporal dementia; glaucoma; glomerulopathy; haemochromatosis; haemodialysis; haemolysis; haemolytic uraemic syndrome (Weil's disease); haemorrhagic stroke; heart attack and reperfusion injury; Huntington's disease; Lewy body disease; intermittent claudication; ischaemic stroke; inflammatory bowel disease; macular
  • compounds of the present invention may also be used to potentiate the effects of other treatments, for example to potentiate the neuroprotective effects of brain derived nerve growth factor.
  • the invention is particularly directed to conditions which induce oxidative damage of the central nervous system, including acute and chronic neurological disorders such as traumatic brain injury, spinal cord injury, cerebral ischaemia, stroke (ischaemic and haemorragic), subharrachnoid haemorrage/cerebral vasospasm, cerebral tumour, Alzheimer's disease, Huntington's disease, Parkinson's disease, Friedrich's ataxia, motor
  • the invention further provides a method of treating a condition resulting in oxidative stress, particularly oxidative damage of the central nervous system, comprising administering to a patient in need of such treatment an effective dose of a compound of the 20 present invention.
  • the invention further provides a pharmaceutical composition comprising a compound of the present invention in combination with a pharmaceutically acceptable carrier or excipient and a method of making such a composition comprising combining a compound 25 of the present invention with a pharmaceutically acceptable carrier or excipient.
  • Compounds of the present invention may be administered in a form suitable for oral use, for example a tablet, capsule, granule powder, aqueous or oily solution, suspension or emulsion; for topical use including transmucosal and transdermal use, for example a cream, 30 ointment, gel, aqueous or oil solution or suspension, salve, patch or plaster; for nasal use, for a example a snuff, nasal spray or nasal drops; for vaginal or rectal use, for example a suppository; for administration by inhalation, for example a finely divided powder or a liquid aerosol; for sub-lingual or buccal use, for example a tablet or capsule; or for parenteral use (including intravenous, subcutaneous, intramuscular, intravascular, intrathecal or infusion), for example a sterile aqueous or oil solution or suspension.
  • parenteral use including intravenous, subcutaneous, intramuscular, intravascular, intrathecal or infusion
  • compositions may be prepared in a conventional manner using conventional excipients, using standard techniques well known to those skilled in the art of pharmacy.
  • the compound is administered orally for chronic disorders such as Alzheimer's and Parkinson's disease, and intravenously for acute disorders such as stroke and TBI.
  • the compounds of the invention will generally be provided in the form of tablets or capsules or as an aqueous solution or suspension.
  • Tablets for oral use may include the active ingredient mixed with pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavouring agents, colouring agents and preservatives.
  • suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, cyclodextrin, and lactose, while corn starch and alginic acid are suitable disintegrating agents.
  • Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • Capsules for oral use include hard gelatin capsules in which the active ingredient is mixed with a solid diluent, and soft gelatin capsules wherein the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.
  • the compounds of the invention will generally be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity.
  • Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride.
  • Aqueous suspensions according to the invention may include suspending agents such as cellulose derivatives, lipids, sodium alginate, polyvinyl- pyrrolidone, cyclodextrins, gum tragacanth, polyethylene glycol, propylene glycol, N,N- dimethylacetamide, cremophors, polysorbates, liposomes and wetting agents such as lecithin.
  • l-(3-(3,5-di-tert-butyl-4-hydroxyphenyl)propyl)-3-hydroxy- 2-methyl-4(lH)-pyridinone (2a) is particularly soluble in hydroxypropyl- ⁇ -cyclodextrin.
  • Suitable preservatives for aqueous suspensions include ethyl and n-propyl p- hydroxybenzoate.
  • dosage levels used may vary over quite a wide range depending upon the compound used, the severity of the symptoms exhibited by the patient and the patient's body weight.
  • Figure 1 shows the protective effect of a compound of the present invention on cerebellar granular cells exposed to IAA-induced oxidative damage.
  • Figure 2 shows the effect of compounds of the present invention on intracellular oxidation of dichlorodihydrofluorescin (DCF ⁇ ) to dichlorofluorecin (DCF). IAA-stimulated fluorescence values are given as a function of concentration of the test compound.
  • Figure 3 and Figure 4 show the in vivo activity of compounds of the present invention in the malonic acid lesion model of oxidative stress.
  • Methanesulphonic acid (175 ⁇ L, 2.7 mmol) was added dropwise to l-(3-(3,5-di-tert-butyl- 4-hydroxyphenyl)propyl)-3-hydroxy-2-methyl-4(lH)-pyridinone (1.0 g, 2.7 mmol) in Et j O (50 mL) and C ⁇ 2 C1 2 (50 mL). The mixture was stirred for 1.5 h, concentrated in vacuo, suspended in CHC1 3 and the solid collected by filtration to give the title compound (1.1 g,
  • BH 3 -Me 2 S (130 mmol) is added to a mixture of dimethyl 2-(3,5-di-tert-butyl-4- hydroxybenzyl)malonate (40 mmol) and THF (100 mL) under Ar. The resulting solution is heated under reflux for 40 h. MeOH is added slowly to the cooled solution to destroy excess reagent. The mixture is diluted with brine, extracted with EtOAc and concentrated in vacuo. The residue is purified by chromatography [SiO 2 ; EtOAc-Hexane] to give the product.
  • reaction is cooled, adjusted to pH 3-4 with 1.0-M HCl, concentrated in vacuo, extracted with CHC1 3 , dried (MgSO 4 ), concentrated in vacuo and purified by chromatography [SiO,; CH 2 Cl 2 -MeOH (95:5)] to give the product in low yield.
  • Methanesulphonic acid (175 ⁇ L, 2.7 mmol) is added dropwise to l-(3-(3,5-di-tert-butyl-4- hydroxyphenyl)-2-(l,4-dihydro-3-hydroxy-2-methyl-4-oxo-l-pyridinylmethyl)propyl)-3- hydroxy-2-methyl-4(lH)-pyridinone (1.3 mmol) in Et,O (50 mL) and C ⁇ 2 C1 2 (50 mL). The mixture is stirred for 1.5 h, concentrated in vacuo, suspended in CHC1 3 and the solid collected by filtration to give the title compound.
  • Methanesulphonic acid (2 eq) is added dropwise to l-(2-(3,5-di-tert-butyl-4- hydroxyphenyl)-2-( 1 ,4-dihydro-3 -hydroxy-2-methyl-4-oxo- 1 -pyridinylmethyl)ethyl)-3- hydroxy-2-methyl-4(lH)-pyridinone in Et 2 O and C ⁇ 2 C1 2 .
  • the mixture is stirred for 1.5 h, concentrated in vacuo, suspended in CHC1 3 and the solid collected by filtration to give the title compound.
  • Lipid peroxidation in rat brain homogenates is a general procedure used to measure the antioxidant capacity of molecules in a biological environment (Das N.P. and Ratty A.K., Biochem. Med. Metab. Biol. 1987, 37, 256-264). Compounds of the present invention have been shown to be potent inhibitors of lipid peroxidation.
  • Iodoacetate via its alkylation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), is a potent inhibitor of glycolysis and hence energy production in cells.
  • GPDH glyceraldehyde-3-phosphate dehydrogenase
  • the compounds of the present invention have been shown to protect cerebellar granule cells from this chemical induced oxidative stress. Furthermore, synergistic behaviour has been demonstrated for the compound of Example 1 over the combination of a single action Fe- chelator and single action antioxidant.
  • Malonic acid is an inhibitor of succinate dehydrogenase. It depletes intracellular ATP production, and intrastriatal injection has previously been demonstrated to cause a NMDA receptor mediated lesion (Greene et al, J. Neurochem. 1993, 61, 1151-5). Compounds of the present invention have shown neuroprotective ability in this animal model of oxidative stress.
  • Rat cortex was homogenised in 20 volumes of ice cold 0.32-M sucrose and centrifuged at 1,000 g for 10 min. The pellet was discarded whilst the resulting supernatant was centrifuged at 15,000 g for 20 min at 4°C to yield p2 pellets. The pellet was resuspended in ice-cold 50.0-mM phosphate buffer and centrifuged at 30,000 g for 20 min at 4°C. The pellet was resuspended in 30 vols of 50.0-mM phosphate buffer and used immediately in the assay.
  • Assays contained 500- ⁇ M L-ascorbic acid to induce lipid peroxidation, plus various concentrations of test compound, with the tissue preparation in a total volume of 500 ⁇ L. This was incubated at 37°C for 30 min. Lipid peroxidation was measured by adding to the reaction mixture, 300 ⁇ L 40% (w/v) trichloroacetic acid, 150 ⁇ L 5.0-M HCl and 300 ⁇ L 2%> (w/v) 2-thiobarbituric acid (TBA). Samples were then heated at 90°C in a water bath for 15 min, and centrifuged at 15,000 g for 10 min. The pink colour of the supernatant was assessed spectrophotometrically at a wavelength of 532 nm.
  • MDA malondialdehyde
  • IC 50 values ( ⁇ M) and presented in Table 1 below.
  • the IC 50 values show the concentration of test compound required to inhibit the lipid peroxidation by 50%>. Table 1.
  • CGC cerebellar granule cell
  • IAA Iodoacetate
  • BSS balanced salt solution
  • HEPES N-[2-hydroxyethyl]piperazine-N ' -[2-ethanesulfonic acid]
  • HEPES N-[2-hydroxyethyl]piperazine-N ' -[2-ethanesulfonic acid]
  • any neuroprotective agents were made up in pre-warmed tissue culture media and allowed to equilibrate in a controlled environment (5%o CO 2 , 95%> air).
  • the assay was initiated by aspiration of the maintenance media, which was replaced by either BSS (control) or 30- ⁇ M IAA in BSS, both solutions containing 10- ⁇ M of the NMDA receptor antagonist MK-801.
  • the exposure to IAA was for 30 min only at 37 °C, after which time the BSS was aspirated and replaced with fresh, pre-equilibriated maintenance media containing various concentrations of test compound. All conditions were performed at least in duplicate in each 96 well plate. - The final volume for each well was always 200- ⁇ L.
  • Results are expressed as EC 50 values ( ⁇ M) and presented in Table 2 below.
  • Figure 1 shows the protective effect from 30- ⁇ M IAA toxicity by 1- ⁇ M concentrations of test compound.
  • CGC are exposed to 30- ⁇ M IAA for 30 mins in a physiological salt solution. This is replaced with maintenance media containing 1- ⁇ M test compound and the cells are then tested for viability 24 hrs later.
  • the compounds tested were Example 1, compound I (below), compound ⁇ (below) and a mixture of compounds I and II
  • DCFH-DA oxidant-sensitive fluorescent dye 2',7'-dichlorodihydrofluorescein diacetate
  • DCFH-DA oxidant-sensitive fluorescent dye 2',7'-dichlorodihydrofluorescein diacetate
  • Results are expressed as EC 50 values ( ⁇ M) and presented in Table 3 below.
  • the EC 50 value gives the effective concentration of test compound required to block the oxidation of DCFH to DCF by 50%.
  • the EC 50 value is derived from Figure 2 by extrapolating from the point at which the fluoresence value is reduced to 50% of its original maximum value.
  • Malonic acid is a competitive inhibitor of succinate dehydrogenase, a key enzyme in both the tricarboxyhc acid cycle and oxidative phosphorylation.
  • Injection of malonic acid into the striatum causes ATP depletion, resulting in an excitotoxic lesion (Greene et al, J. Neurochem., 1993, 61, 1151-1154).
  • 2 ⁇ L of a 0.5-M malonic acid solution is injected into the right striatum of rats, with or without test compounds. 24 hours after surgery the animals are sacrificed and the size of the lesion measured using TTC histochemistry.
  • Example 1 The observed activity of the compounds of Example 1 and Example 7 are displayed in Figures 3 and 4, respectively.
  • 2.2 ng of the compound of Example 1 is the equivalent of 2 ⁇ L of a 3.0- ⁇ M solution.
  • 7.4 ng of the compound of either Example 1 or Example 7 is the equivalent of 2 ⁇ L of a 10.0- ⁇ M solution.
  • 74 ng of the compound of Example 7 is the equivalent of 2 ⁇ L of a 100.0- ⁇ M solution.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (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)
  • Psychology (AREA)
  • Vascular Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pyridine Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

La présente invention concerne un composé de la formule (1): dans laquelle A est: ou: où R?1, R2 et R3¿ sont choisis indépendamment entre H et alkyle; X est O, S, NR4 ou une liaison directe, R4 étant H ou alkyle; Z est une chaîne hydrocarbyle saturée renfermant entre 1 et 10 atomes de carbone; q est égal à 1, 2 ou 3, chaque A pouvant être le même ou différent si q est égal à 2 ou 3; R5 ou chaque R5 est choisi indépendamment entre H ou alkyle; R6 ou chaque R6 est choisi indépendamment parmi les alkyles; n est compris entre 1 et 5; p est compris entre 0 et 4; la somme de n et p est inférieure à 6; ou un sel pharmaceutiquement acceptable de ce composé, et l'invention concerne également l'utilisation thérapeutique de ces derniers, en particulier dans le traitement d'un état aboutissant à un stress oxydatif.
EP98950227A 1997-10-31 1998-10-30 Derives d'ortho-hydroxypyridinone utilises comme agents chelateurs du fer et comme agents antioxydants Withdrawn EP1027335A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
GBGB9723078.3A GB9723078D0 (en) 1997-10-31 1997-10-31 Chemical compounds
GB9723078 1997-10-31
PCT/GB1998/003244 WO1999023075A1 (fr) 1997-10-31 1998-10-30 Derives d'ortho-hydroxypyridinone utilises comme agents chelateurs du fer et comme agents antioxydants

Publications (1)

Publication Number Publication Date
EP1027335A1 true EP1027335A1 (fr) 2000-08-16

Family

ID=10821419

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98950227A Withdrawn EP1027335A1 (fr) 1997-10-31 1998-10-30 Derives d'ortho-hydroxypyridinone utilises comme agents chelateurs du fer et comme agents antioxydants

Country Status (5)

Country Link
EP (1) EP1027335A1 (fr)
JP (1) JP2001521924A (fr)
AU (1) AU9638098A (fr)
GB (1) GB9723078D0 (fr)
WO (1) WO1999023075A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111170936A (zh) * 2020-01-19 2020-05-19 杭州泽德医药科技有限公司 3,4-二羟基-n-(1’-苄基-2’-羟乙基)-2-甲基吡啶氯化物及制备和应用

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1006108A1 (fr) * 1998-12-01 2000-06-07 Cerebrus Pharmaceuticals Limited Dérivés de 3-hydroxy-2(1H)-pyridinone ou 3-hydroxy-4(1H)-pyridinone et utilisation comme des capteurs des espèces d'oxygène réactifs (ROS)
EP1006112A1 (fr) * 1998-12-01 2000-06-07 Cerebrus Pharmaceuticals Limited Dérivés de 3-Hydroxy-2(1H)-pyridinone ou 3-hydroxy-4(1H)-pyridinone et utilisation en tant que capteurs d'espèces d'oxygène réaktifs (ROS)
US7482366B2 (en) 2001-12-21 2009-01-27 X-Ceptor Therapeutics, Inc. Modulators of LXR
ES2421511T3 (es) 2001-12-21 2013-09-03 X Ceptor Therapeutics Inc Moduladores de LXR
AU2003210850A1 (en) 2002-02-05 2003-09-02 Bristol-Myers Squibb Company N-substituted3-hydroxy-4-pyridinones and pharmaceuticals containing thereof
US8058442B2 (en) 2002-11-07 2011-11-15 Technion Research And Development Foundation Ltd. Neuroprotective iron chelators and pharmaceutical compositions comprising them
KR20130044381A (ko) * 2010-03-04 2013-05-02 머크 샤프 앤드 돔 코포레이션 카테콜 o-메틸 트랜스퍼라제의 억제제 및 정신병적 장애의 치료에서의 그의 용도
CN110407741B (zh) * 2018-04-26 2023-03-21 启元生物(杭州)有限公司 一种抗炎化合物及其制备和应用

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308055D0 (en) * 1983-03-24 1983-05-05 Hider R C Pharmaceutical compositions
US5336482A (en) * 1991-12-05 1994-08-09 The Du Pont Merck Pharmaceutical Company Technetium-99m complexes with N-substituted 3-hydroxy-4-pyridinones
US5624901A (en) * 1994-04-15 1997-04-29 The Regents Of The University Of California 3-hydroxy-2(1H)-pyridinone chelating agents
ES2244967T3 (es) * 1995-09-29 2005-12-16 Novartis Ag Nuevas hidroxipiridinonas.
GB9625638D0 (en) * 1996-12-10 1997-01-29 Cenes Ltd Therapeutic antioxidants for alzheimer's diease

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9923075A1 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111170936A (zh) * 2020-01-19 2020-05-19 杭州泽德医药科技有限公司 3,4-二羟基-n-(1’-苄基-2’-羟乙基)-2-甲基吡啶氯化物及制备和应用

Also Published As

Publication number Publication date
WO1999023075A1 (fr) 1999-05-14
JP2001521924A (ja) 2001-11-13
AU9638098A (en) 1999-05-24
GB9723078D0 (en) 1998-01-07

Similar Documents

Publication Publication Date Title
EP1006112A1 (fr) Dérivés de 3-Hydroxy-2(1H)-pyridinone ou 3-hydroxy-4(1H)-pyridinone et utilisation en tant que capteurs d'espèces d'oxygène réaktifs (ROS)
CA2417475C (fr) Azabicyclic compounds, preparation thereof and use as medicines, in particular as antibacterial agents
JP5838157B2 (ja) Hdac阻害剤およびそれを用いる治療法
US7410956B2 (en) Caspase inhibitor prodrugs
KR20170008320A (ko) 니코틴아미드 리보시드 유사체 및 그의 제약 조성물 및 용도
GB2345058A (en) Hydroxypyridone compounds useful in the treatment of oxidative damage to the central nervous system
PL154186B1 (en) Method for manufacturing arylic derivatives of the hydroxamic acid
WO2014093231A2 (fr) Composés hybrides et leurs procédés de fabrication et d'utilisation
KR101539588B1 (ko) (6,7-디히드로-2-니트로-5h-이미다졸[2,1-b][1,3]옥사진-6-일)아미드 화합물, 그의 제조방법 및 용도
JP2004518628A (ja) 窒素含有化合物およびそれらのグリシン輸送阻害薬としての使用法
EP2242492B1 (fr) Agents antitumoraux avec une structure de benzophénanthridine et formulations les contenant
WO1999023075A1 (fr) Derives d'ortho-hydroxypyridinone utilises comme agents chelateurs du fer et comme agents antioxydants
EP1006108A1 (fr) Dérivés de 3-hydroxy-2(1H)-pyridinone ou 3-hydroxy-4(1H)-pyridinone et utilisation comme des capteurs des espèces d'oxygène réactifs (ROS)
US20050176763A1 (en) 3-Thia-4-arylquinolin-2-one potassium channel modulators
US20210206713A1 (en) Compounds and methods for the treatment of neurodegenerative diseases
CN116724023A (zh) 作为药物活性化合物的大麻素衍生物及其制备方法
JPH09507846A (ja) フェニルメチルヘキサンアミドおよびその使用
US20140148432A1 (en) Compounds for the Treatment of Neurological Disorders
EP0499882A1 (fr) Compositions pharmaceutiques contenant des dérivés de la N-acétyl-cysteine pour le traitement de la cataracte
JP2007508301A5 (fr)
RU2685502C2 (ru) Терапевтические агенты для применения в профилактике и/или лечении гиперкинетических двигательных расстройств
KR101115196B1 (ko) 2-아미노벤조일 유도체
PL208284B1 (pl) Pochodne benzo[g]chinoliny, kompozycje je zawierające oraz zastosowanie takich pochodnych
CA3176909A1 (fr) Compositions et methodes pour la prevention et/ou le traitement d'une maladie mitochondriale, notamment l'ataxie de friedreich
WO2012001438A1 (fr) Utilisation de dérivés d'amide d'acide kynurénique pour le traitement de la maladie de huntington

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20000428

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: VERNALIS RESEARCH LIMITED

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20020501