WO2003082893A2 - Theraputic methods and uses of sapogenins and their derivatives - Google Patents

Theraputic methods and uses of sapogenins and their derivatives Download PDF

Info

Publication number
WO2003082893A2
WO2003082893A2 PCT/GB2003/001380 GB0301380W WO03082893A2 WO 2003082893 A2 WO2003082893 A2 WO 2003082893A2 GB 0301380 W GB0301380 W GB 0301380W WO 03082893 A2 WO03082893 A2 WO 03082893A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
pharmaceutically acceptable
group
absent
acceptable salts
Prior art date
Application number
PCT/GB2003/001380
Other languages
French (fr)
Other versions
WO2003082893A3 (en
WO2003082893A8 (en
Inventor
Daryl Rees
Phil Gunning
Antonia Orsi
Zongqin Xia
Yaer Hu
Original Assignee
Phytopharm Plc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from ARP020101170A external-priority patent/AR033079A1/en
Priority to CN038071886A priority Critical patent/CN1642558B/en
Priority to EP03722713A priority patent/EP1490071B1/en
Priority to DE60326437T priority patent/DE60326437D1/en
Priority to IL16416103A priority patent/IL164161A0/en
Priority to JP2003580357A priority patent/JP4573157B2/en
Priority to KR1020047014567A priority patent/KR101130212B1/en
Priority to CA2477787A priority patent/CA2477787C/en
Priority to NZ535093A priority patent/NZ535093A/en
Priority to KR1020127008441A priority patent/KR20120128596A/en
Application filed by Phytopharm Plc filed Critical Phytopharm Plc
Priority to BR0308788-3A priority patent/BR0308788A/en
Priority to MXPA04009321A priority patent/MXPA04009321A/en
Priority to AU2003229877A priority patent/AU2003229877B8/en
Priority to US10/507,000 priority patent/US20050130948A1/en
Publication of WO2003082893A2 publication Critical patent/WO2003082893A2/en
Publication of WO2003082893A8 publication Critical patent/WO2003082893A8/en
Publication of WO2003082893A3 publication Critical patent/WO2003082893A3/en
Priority to NO20044468A priority patent/NO20044468L/en
Priority to HR20041009A priority patent/HRP20041009A2/en
Priority to HK04110420.5A priority patent/HK1067546A1/en
Priority to AU2008207565A priority patent/AU2008207565A1/en
Priority to US13/085,225 priority patent/US20110190249A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/655Azo (—N=N—), diazo (=N2), azoxy (>N—O—N< or N(=O)—N<), azido (—N3) or diazoamino (—N=N—N<) compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • 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
    • A61P25/08Antiepileptics; Anticonvulsants
    • 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
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J71/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton is condensed with a heterocyclic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J71/00Steroids in which the cyclopenta(a)hydrophenanthrene skeleton is condensed with a heterocyclic ring
    • C07J71/0005Oxygen-containing hetero ring
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to therapeutic methods and uses of sapogenins, related compounds and their derivatives.
  • the uses of the sapogenins, related compounds and their derivatives are in the treatment of non-cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor- sensory neurodegeneration, or receptor dysfunction or loss.
  • the invention relates to compositions for use in such treatments.
  • Cognitive dysfunction is a characteristic of dementia conditions and syndromes, such as Alzheimer's disease (AD), senile dementia of the Alzheimer's type (SDAT), Lewy body dementia and vascular dementia.
  • a lesser degree of cognitive dysfunction is also a characteristic of certain non-dementia conditions and syndromes, such as mild cognitive impairment (MCI), age-associated memory impairment (AAMI), autism and neuroimpairment.
  • MCI mild cognitive impairment
  • AAMI age-associated memory impairment
  • autism neuroimpairment.
  • Non-cognitive neurodegeneration i.e. neurodegeneration in the absence of cognitive dysfunction
  • non-cognitive neuromuscular degeneration i.e. neuromuscular degeneration in the absence of cognitive dysfunction
  • motor-sensory neurodegeneration are characteristic of conditions and syndromes such as Parkinson's disease, muscular dystrophy including facioscapulohumeral muscular dystrophy (FSH), Duchenne muscular dystrophy, Becker muscular dystrophy and Bruce's muscular dystrophy, Fuchs' dystrophy, myotonic dystrophy, corneal dystrophy, reflex sympathetic dystrophy syndrome (RSDSA), neurovascular dystrophy, myasthenia gravis, Lambert Eaton disease, Huntington's disease, amyotrophic lateral sclerosis (ALS) and multiple sclerosis.
  • FSH facioscapulohumeral muscular dystrophy
  • Duchenne muscular dystrophy Becker muscular dystrophy and Bruce's muscular dystrophy
  • Fuchs' dystrophy myotonic dystrophy
  • Receptor dysfunction or loss - particularly dysfunction or loss of nicotinic and/or muscarinic acetylcholine receptors and/or dopamine receptors and/or adrenoceptors - is a characteristic of some or all of the above conditions and syndromes.
  • Receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment is also a characteristic of conditions and syndromes such as postural hypotension, chronic fatigue syndrome, asthma, susceptibility to heart failure and macular degeneration.
  • WO-A-99/16786 the disclosure of which is incorporated herein by reference, suggests the use of certain saponins and sapogenins in the treatment of dementia.
  • WO-A-99/48482, WO-A-99/48507, WO-A-01/23406, WO-A-01/23407, WO-A-01/23408, the disclosures of which are incorporated herein by reference, relate to the use of certain saponins, sapogenins and derivatives thereof in the treatment of cognitive dysfunction and allied conditions.
  • cognitive dysfunction is not a primary symptom, although it may be present as one of a number of possible secondary symptoms.
  • these conditions are not viral diseases or dementias.
  • a treatment for cognitive dysfunction e.g. dementia
  • the present invention is based upon our finding that certain sapogenins and their derivatives, including saponins, have a surprising disease-modifying activity against non- cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration as well as against receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment, and thus actively prevent and reverse the conditions.
  • This finding enables improved treatment of certain non-viral, spectrum and non-spectrum, disorders in which cognitive dysfunction is not a primary symptom.
  • the active agents in the treatment or prevention of, or in the preparation of compositions (e.g. pharmaceutical compositions, foodstuffs, food supplements and beverages) for the treatment or prevention of, (i) non-cognitive neurodegeneration, (ii) non- cognitive neuromuscular degeneration, (iii) motor-sensory neurodegeneration (iv) receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment, in human and non-human animals suffering therefrom or susceptible thereto.
  • compositions e.g. pharmaceutical compositions, foodstuffs, food supplements and beverages
  • active agents refers to compounds of the following general formulae I, II and HI, sapogenin derivatives as defined below by reference to derivatives bearing at least one defined X radical substituent, sugar-substituted derivatives of such compounds as defined below, all their stereoisomers and racemic mixtures, all their pharmaceutically acceptable pro-drugs and salts, and all mixtures and combinations thereof:
  • R 33 or R 14 alkyl group, and the stereochemistry of R 25 is in the ⁇ orientation;
  • sapogenin derivatives particularly but not exclusively steroidal spirostane sapogenin derivatives, bearing at least one X radical substituent, wherein X is chosen from the group consisting of :
  • the carbon atom at the 3-position i.e. the carbon to which R 3 is attached, or the corresponding position in the sapogenin derivatives defined above without a formula
  • the 3- position carbon atom, the 26-position i.e. the carbon to which R 34 is attached
  • each of the carbon atoms at the 3- and 26-positions carries an O-sugar moiety wherein the sugar group is a mono-, di- or tri-saccharide.
  • sugar-carrying derivative forms of sapogenin active agents are generally referred to in the art as saponins.
  • the expression "carbohydrate” used herein includes particularly such sugar groups.
  • the active agents used in the present invention are preferably the non-estrogenic steroidal sapogenins, saponins, and derivatives thereof within the terms of the above definition, including all physiologically acceptable pro-drugs and salts thereof.
  • the active agents may be naturally occurring or non-naturally occurring.
  • Non-naturally occurring active agents may suitably be prepared by modification of side groups and/or side atoms of naturally occurring compounds, as described below and as known in the art.
  • the invention also provides corresponding methods for the treatment of human and non- human animals, and compositions containing the active agents for use in the said treatment methods.
  • the active agents of the invention may, if desired, be co-administered with one or more additional active agent, for example one or more agent selected from, but not limited to, cholinesterase inhibitors, dopamine agonists (e.g. L-dopa), COMT inhibitors, MAO-B inhibitors, anti-cholinergics, acetylcholine agonists, serotonin agonists, AMPA receptor agonists, GABA receptor agonists, NMDA receptor agonists, ⁇ -adrenoceptor agonists, digoxin, dobutamine, anti-inflammatories, neurotrophic factors, statins, adenosine A2a receptor antagonists, aldose reductase inhibitors, immunomodulators, cannabinoid agonists, interferon ⁇ or tricyclic anti-depressants.
  • cholinesterase inhibitors e.g. L-dopa
  • COMT inhibitors e.g. L-dopa
  • the active agents may be applied therapeutically or prophylactically to human and non-human animals suffering from, or susceptible to, conditions and diseases that are characterised by non-cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration, or receptor dysfunction or loss. Examples of such conditions and diseases are provided below.
  • the present invention therefore also provides the use of the active agents (as herein defined) in the treatment or prevention of, or in the preparation of compositions (e.g. pharmaceutical compositions, foodstuffs, food supplements and beverages) for the treatment or prevention of, one or more of said conditions and diseases in human and non- human animals suffering therefrom or susceptible thereto.
  • active agents as herein defined
  • compositions e.g. pharmaceutical compositions, foodstuffs, food supplements and beverages
  • R alkyl or acyl group
  • R 33 or R 14 alkyl group, and the stereochemistry of R 25 is in the ⁇ orientation;
  • R 33 or R i4 alkyl group, and the stereochemistry of R 25 is in the ⁇ orientation;
  • R 33 or R 14 alkyl group, and the stereochemistry of R 25 is in the ⁇ orientation;
  • R 33 or R 14 alkyl group, and the stereochemistry of R 25 is in the ⁇ orientation;
  • R 36 H or -OH represents a single bond
  • R 36 H or OH represents a single bond
  • Substituted sapogenins particularly but not exclusively steroidal spirostane sapogenins, wherein at least one OH-group of the sapogenin is substituted with X, chosen from the group consisting of :
  • Sapogenins defined above particularly but not exclusively steroidal spirostane sapogenins, wherein in the definition of X the halo atom is a fluoro atom;
  • Substituted sapogenins selected from :
  • Substituted sapogenins wherein the parent sapogenin which is then substituted with at least one X radical as defined above is selected from sarsasapogenin, episarsasapogenin, smilagenin, epismilagenin, andmilarogenin-D;
  • group R is selected from hydrogen; alkylcarbonyl; alkoxycarbonyl; alkyl- carbamoyl; or arylcarbonyl; or sulpho (H0 3 S); phosphono ((HO) 2 P(0)-); or a mono-, di- or tri-saccharide; wherein any alkyl group is optionally substituted with aryl, amino, mono- or di-alkyl-amino, a carboxylic acid residue (-COOH), or any combination thereof;
  • Optional amino, mono-alkyl-amino and di-alkyl-amino substituents of alkyl groups, where present, are preferably a mono-substituent at the ⁇ position of the alkyl group.
  • Optional -COOH substituents of alkyl groups, where present, may be at the terminal or any other position of the alkyl group.
  • Alkyl means an aliphatic hydrocarbon group which may be straight or branched having about 1 to about 20 carbon atoms in the chain. Preferred alkyl groups have 1 to about 12 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl are attached to a linear alkyl chain. "Lower alkyl” means about 1 to about 4 carbon atoms in the chain which may be straight or branched. Exemplary alkyl groups include methyl, ethyl, n-propyl, t-propyl, M-butyl, t-butyl, _?-butyl, n-pentyl, 3-pentyl.
  • Aryl means any group comprising an aromatic ring or system of fused rings, and preferably contains up to 12 carbon atoms.
  • An exemplary aryl group is the phenyl group.
  • An aryl group may optionally be mono- or poly-substituted, for example by substituents independently selected from halo (e.g. chloro or bromo), alkyl, cycloalkyl, hydroxy, alkoxy, amino, nitro, acylamino, carboxy and alkoxycarbonyl.
  • Carboxylic acid residue means the group -COOH.
  • acyl means an H-CO- or Alkyl-CO- group wherein the alkyl group is as defined below.
  • Preferred acyls contain a lower alkyl.
  • Exemplary acyl groups include formyl. acetyl, propanoyl, 2-methylpropanoyl, butanoyl and palmitoyl;
  • Optionally substituted means that the said group may be substituted with one or more substituents, which may be the same or different, preferably one or more substituents which individually have a size which is small (e.g. less than about 20% of the largest molecular dimension) in relation to the parent group being substituted; suitable substituents include halo (e.g. chloro or bromo), alkyl, cycloalkyl, hydroxy, alkoxy, amino, acylamino, aryl, aroylamino, carboxy, alkoxycarbonyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, and optionally substituted carbamoyl, preferably subject to the size limitation set out above ;
  • substituents include halo (e.g. chloro or bromo), alkyl, cycloalkyl, hydroxy, alkoxy, amino, acylamino, aryl, aroylamino, carboxy, alkoxycarbonyl, aral
  • “Pharmaceutically acceptable” means it is, within the scope of sound medical and veterinary judgement, suitable for use in contact with the cells of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable prodrugs” means those prodrugs of the compounds which are, within the scope of sound medical and veterinary judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds.
  • prodrug means compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood.
  • Functional groups which may be rapidly transformed, by metabolic cleavage, in vivo form a class of groups reactive with the carboxyl group. Because of the ease with which the metabolically cleavable groups of the compounds are cleaved in vivo, the compounds bearing such groups act as pro-drugs.
  • a thorough discussion of prodrugs is provided in the following: Design of Prodrugs, H. Bundgaard, ed., Elsevier, 1985; Methods in Enzymology, K.
  • “Pharmaceutically acceptable salts” means the relatively non-toxic, inorganic and organic acid addition salts, and base addition salts, of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds.
  • acid addition salts can be prepared by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. See, for example S. M. Berge, et al., Pharmaceutical Salts, J. Pharm. Sci., 66: p.l- 19 (1977) which is incorporated herein by reference.
  • Base addition salts can also be prepared by separately reacting the purified compound in its acid form with a suitable organic or inorganic base and isolating the salt thus formed.
  • Base addition salts include pharmaceutically acceptable metal and amine salts.
  • suitable acid addition salts are those formed with acids selected from hydrochloric, sulphuric, phosphoric and nitric acids.
  • suitable base addition salts are those formed with bases selected from sodium hydroxide, potassium hydroxide and ammonium hydroxide.
  • a particularly preferred class of active agents are the compounds of the general formula la.
  • the C 25 methyl group is in the S configuration; these compounds of the invention are sarsasapogenin and episarsasapogenin or derivatives thereof.
  • the C 25 methyl group is in the R configuration; these compounds of the invention are smilagenin and epismilagenin or derivatives thereof.
  • -OR may, for example, be selected from the following (unless excluded by proviso):, hydroxy, cathylatc (ethoxycarbonyloxy), acetate, succinate, cinnamate, ferulate, propionate, butyrate, valerate, isovalerate, caproate, isocaproate, diethylacetate, octanoate, decanoate, laurate, myristate, palmitate, stearate, benzoate, phenylacetate, phenylpropionate, cinnamate, p-nitrobenzoyloxy, 3,5-dinitrobenzoyloxy, p- chlorobenzoyloxy, 2,4-dichlorobenzoyloxy, p-bromobenzoyloxy, m-bromobenzoyloxy.
  • p- methoxybenzoyloxy phthalyl, glycinate, alaninate, valinate, phenylalaninate, isoleucinate, methioninate, argininate, asparaginate, aspartate, cysteinate, glutamate, histidinate, lysinate, prolinate, serinate, threoninate, tryptophanate, tyrosinate, fumarate or maleate.
  • suitable active agents include 16,22-epoxycoprostan-3 ⁇ -ol, smilagenone, coprosterol and pharmaceutically acceptable pro-drugs and salts thereof.
  • the present invention thus enables and provides a method for treating or preventing non- cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration or receptor dysfunction or loss in the absence of cognitive, neural or neuromuscular impairment (particularly but not exclusively in relation to the specific disease states mentioned above) in a human or non-human animal in need thereof, which comprises administering to the said human or non-human animal an effective dosage of an active agent (as defined herein) or a pharmaceutically acceptable salt thereof.
  • an active agent as defined herein
  • the active agent may be administered in the form of a composition comprising the active agent and any suitable additional component.
  • the composition may, for example, be a pharmaceutical composition (medicament), a foodstuff, food supplement or beverage.
  • Such a composition may contain a mixture of the specified compounds, and/or of their pharmaceutically acceptable salts.
  • composition in the context of this invention means a composition comprising an active agent and comprising additionally pharmaceutically acceptable carriers, diluents, adjuvants, excipients, or vehicles, such as preserving agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • pharmaceutically acceptable carriers such as preserving agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
  • the dosage of the active agent will vary widely, depending on the severity of the symptoms to be treated or prevented. The selection of appropriate dosages is within the ability of one of ordinary skill in this art, without undue burden.
  • the dosage of the active agent may, for example, be greater than about 0.1 mg/kg body weight, for example greater than about 0.3 mg/kg body weight, preferably administered once per day. More typically, the dosage will be between about 1 and about 25 mg/kg, e.g. between about 1 and about 10 mg/kg, preferably administered once per day. For human use, the dosage may conveniently be between about 70 and about 700 mg per day.
  • “Pharmaceutically acceptable dosage forms” means dosage forms of the compounds or compositions of the invention, and includes, for example, tablets, dragees, powders, elixirs, syrups, liquid preparations, including suspensions, sprays, inhalants, tablets, lozenges, emulsions, solutions, granules, capsules and suppositories, as well as liquid preparations for injections, including liposome preparations.
  • Techniques and formulations generally may be found in Remington, Pharmaceutical Sciences, Mack Publishing Co., Easton, PA, latest edition. In general, reference herein to the presence of one of a specified group of compounds includes within its scope the presence of a mixture of two or more of such compounds.
  • This invention provides for the treatment of (i) non-cognitive neurodegeneration, (ii) non- cognitive neuromuscular degeneration, (iii) motor-sensory neurodegeneration, or (iv) receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment, in a human or non-human animal subject suffering from, or susceptible to, any of: Parkinson's disease, postencephalitic Parkinsonism, depression, schizophrenia, muscular dystrophy including facioscapulohumeral muscular dystrophy (FSH), Duchenne muscular dystrophy, Becker muscular dystrophy and Brace's muscular dystrophy, Fuchs' dystrophy, myotonic dystrophy, corneal dystrophy, reflex sympathetic dystrophy syndrome (RSDSA), neurovascular dystrophy, myasthenia gravis, Lambert Eaton disease, Huntington's disease, motor neurone diseases including amyotrophic lateral sclerosis (ALS), multiple sclerosis, postural hypotension, traumatic neurodegeneration e.g.
  • ALS amy
  • traumatic head injury or spinal cord injury Batten's disease, Cockayne syndrome, Down syndrome, corticobasal ganglionic degeneration, multiple system atrophy, cerebral atrophy, olivopontocerebellar atrophy, dentatorubral atrophy, pallidoluysian atrophy, spinobulbar atrophy, optic neuritis, sclerosing pan-encephalitis (SSPE), attention deficit disorder, post-viral encephalitis, post-poliomyelitis syndrome, Fahr's syndrome, Joubert syndrome, Guillain-Barre syndrome, lissencephaly, Moyamoya disease, neuronal migration disorders, autistic syndrome, polyglutamine disease, Niemann- Pick disease, progressive multifocal leukoencephalopathy, pseudotumor cerebri, Refsum disease, Zellweger syndrome, supranuclear palsy, Friedreich's ataxia, spinocerebellar ataxia type 2, Rhett syndrome, Shy
  • the invention therefore includes methods of treating or preventing the above diseases and conditions in a human or non-human animal suffering therefrom or susceptible thereto, which comprises administering to the said human or non-human animal an effective amount of an active agent as defined herein, as well as uses of the active agents in the preparation of compositions for said treatment or prevention.
  • the present invention may be subject to the proviso that either symptoms of cognitive dysfunction are absent, or any symptoms of cognitive dysfunction presented by a subject to be treated are secondary or ancillary to the symptoms of non-cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration or receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment.
  • Smilagenin, epismilagenin and sarsasapogenin are commercially available materials. Suppliers include, for example, Sigma Aldrich, Research Plus Inc. and Steraloids Inc. Preparative methods for these materials are also to be found in the literature (e.g. a preparation of episarsasapogenin is given in JACS p.5225 (1959)).
  • Sarsasapogenone can be prepared using the method of Lajis et al, Steroids, 1993, 58, 387-- 389.
  • unsubstituted saponins and sapogenins may occur naturally in a range of plant species, notably plants of the genus Smilax, Asparagus, Anemarrhena, Yucca or Agave.
  • smilagenin or sarsasapogenin is used in accordance with this invention, it may be in the form of a plant extract, or dry powdered plant material, derived from a plant . of the genus Smilax, Asparagus, Anemarrhena, Yucca or Agave.
  • the preferred reaction is a nucleophilic substitution reaction, in which a compound having OH at the 3-position is reacted with a compound of formula
  • R is selected from alkylcarbonyl; alkoxycarbonyl; alkylcarbamoyl; or arylcarbonyl; or wherein any alkyl group is optionally substituted with aryl, amino, mono- alkyl-amino, di-alkyl-amino, a carboxylic acid residue (-COOH), or any combination thereof; and L is a leaving group, under conditions suitable for nucleophilic substitution.
  • the compound L-R may, for example, be a carboxylic acid or, if appropriate, an anhydride, or an acyl halide (e.g. an acyl chloride).
  • R is a cathylate (ethoxycarbonyl) moiety
  • the compound L-R may suitably be ethyl chloroformate.
  • the reaction is suitably performed in a base such as pyridine, optionally in the presence of an acid such as hydrochloric acid.
  • Dihydrosarsasapogenin may be made using the method described in Marker and Rohrmann (1939), Sterols LIJI; The structure of the side chain of sarsasapogenin, J. Am. Chem. Soc. 61, pp 846-851. 16,22-Epoxycoprostan-3 ⁇ -ol may be made using the method described in Scheer et al, (1955), The C-25 isomerism of smilagenin and sarsasapogenin: J. Am. Chem. Soc. 77, pp 641-646.
  • an alkoxycarbonyl protecting group for protecting amino substituents in compounds of formula L-R wherein R is amino-substituted, it is preferred to use an alkoxycarbonyl protecting group, whereby the amino function is present as an alkoxycarbonylamino group (preferably t.-butoxycarbonylamino) during the synthetic steps, until deprotection in acid conditions in a dry solvent.
  • the compound thus prepared may be recovered from the reaction mixture by conventional means.
  • the compound may be recovered by distilling off the solvent from the reaction mixture or, if necessary after distilling off the solvent from the reaction mixture, pouring the residue into water, followed by extraction with a water-miscible solvent and distilling off the solvent from the extract.
  • the product can, if desired, be further purified by various well known techniques, such as recrystallisation, reprecipitation, or the various chromatography techniques, notably column chromatography or preparative thin layer chromatography.
  • the therapeutic uses underlying the present invention arise from a number of novel observations which are documented in detail in the Examples below. To understand the rationale of the invention, it is useful to summarise the observations and to explain how they predict the therapeutic activities claimed in this invention across the range of active agents defined above.
  • Smilagenin, epismilagenin, sarsaspogenin and episarsasapogenin restore the loss of muscarinic acetylcholine receptors and adrenoceptors in cells expressing such receptors in vitro. These results demonstrate that these compounds restore towards normal cellular receptor loss (Example 1). .
  • Sarsaspogenin, episarsasapogenin cathylate and smilagenin reduce the number of wrong responses in a cognitive ability test in vivo in aged rats, which correlates with an increase in muscarinic acetylcholine receptor density in the brains of aged rats following treatment with the compounds tested. These results demonstrate that the compounds reverse neuroimpairment in vivo (Example 9).
  • Episarsasapogenin cathylate, sarsasapogenin cathylate, episarsasapogenin and epismilagenin reduce the number of wrong responses in a cognitive ability test in vivo in young rats exposed to neurotoxic agents (ibotenic acid and amyloid D, and increase in muscarinic acetylcholine receptor density in the brains. These results demonstrate that the compounds reverse neuroimpairment in vivo (Example 10). Smilagenin and sarsasapogenin improve survival and motor-sensory neurodegeneration and neuroimpairment in a mouse model of amyotrophic lateral sclerosis (ALS) and Charcot- Marie-Tooth disease (Example 11).
  • ALS amyotrophic lateral sclerosis
  • Example 11 Charcot- Marie-Tooth disease
  • the compounds have been found to slow or reverse certain aspects of neuronal degeneration. These include the reversal of adverse changes in the cell body, atrophy of neuronal extensions (neurites), reduction in release of neurotrophic factors such as neurotrophins (e.g. BDNF, NGF, NT-3, NT4/5), TGF- ⁇ super-family neurotrophic factors (e.g. GDNF) and neurokines (e.g. CNTF, LIF), and neuronal toxicity or death (apoptosis).
  • neurotrophins e.g. BDNF, NGF, NT-3, NT4/5
  • TGF- ⁇ super-family neurotrophic factors e.g. GDNF
  • neurokines e.g. CNTF, LIF
  • apoptosis neuronal toxicity or death
  • the compounds have also been found to slow or reverse decreases in cholinergic and dopaminergic function, for example, decreases in muscarinic acetylcholine and dopamine receptor density. Furthermore, we have found that the neuroprotective and the reversal of receptor loss effects are actively regulated effects, in which past deterioration is reversed towards the normal or young state with protection against continued deterioration. Still further, we have found that the reversal of apoptotic effect of the compounds appears to be regulated in the non-neoplastic domain of cell life, and does not appear likely to trigger neoplasia.
  • the above data indicate activity against the disease states already listed in this application. Furthermore, the above data indicates a likely absence of severe or life- threatening side effects such as cancer.
  • the active agents are typically non-oestrogenic.
  • side group substituents in a wide range of forms, can be present in a complex carbon skeleton without substantially adversely affecting the pharmacological activity of the structure, particularly when the side groups are small in comparison with the overall size of the molecule.
  • one physiological effect of the active agents is the ability to increase the synthesis, or release of, or to reduce the rate of degradation of, neurotrophic factors such as brain derived neurotrophic factor and/or nerve growth factor or their receptors.
  • neurotrophic factors such as brain derived neurotrophic factor and/or nerve growth factor or their receptors.
  • These effects on growth factors might be due to an effect of the compound on a cytosolic or nuclear receptor, or the binding of a compound to a promoter region with a consequent effect directly on the rate of production of mRNA for the growth factor, or as a consequence of increasing the production of another material factor.
  • the compounds appear to regulate receptors. For example, some of these compounds have been found to prevent or reverse the loss of muscarinic acetylcholine or dopamine receptors in the brain. It is believed that the compounds function by rectifying a deficiency in receptor number or function or turnover.
  • Figure 1 shows the effect of epismilagenin acetate on m3 and ⁇ 2 adrenoceptor density at day 5 in a CHO- ⁇ 2/m3 co-transfected cell line;
  • Figure 2 shows the effects of sarsasapogenin, episarsasapogenin cathylate and smilagenin on glutamate induced neurodegeneration in rat primary cortical neurons;
  • Figure 3 shows the effects of sarsasapogenin, episarsasapogenin cathylate and smilagenin on the learning ability and memory of aged rats;
  • Figure 4 shows the effects of sarsasapogenin, episarsasapogenin cathylate and smilagenin on muscarinic receptor number;
  • Figure 5 shows the survival profile of SOD-1 mice following oral administration of smilagenin
  • Figure 6 shows the survival profile of pmn mice following oral administration of sarsasapogenin.
  • epismilagenin cathylate sarsasapogenin cathylate, episarsasapogenin cathylate, episarsasapogenin succinate, epismilagenin acetate and sarsasapogenin on the expression of muscarinic acetylcholine receptor (m) in CHO cells or ⁇ 2 and m3 receptors in CHO cells transfected with either a vector for the m receptor or co-transfected with the vector for ⁇ 2 and m3 receptors were investigated.
  • m muscarinic acetylcholine receptor
  • Table 1 Effect of epismilagenin cathylate, sarsasapogenin cathylate, episarsasapogenin cathylate, episarsasapogenin succinate and sarsasapogenin on restoration of m acetylcholine receptor density.
  • the objective of this study was to examine the effects of sarsasapogenin, episarsasapogenin cathylate, episarsasapogenin, epismilagenin and smilagenin on the survival of rat primary cortical neurones exposed to glutamate, which is known to induce neurodegeneration.
  • Rat cortical neurones were cultured for 10 days; at day 10 the medium was changed to a serum-free defined medium. On day 12, 24 hours before glutamate exposure, cultures were washed a d medium was replaced with fresh medium containing positive .control ( ⁇ - oestradiol)j test compounds (sarsasapogenin, episarsasapogenin - cathylate, episarsasapogenin, epismilagenin and smilagenin) or vehicle control (DMSO, 0.25%) or diosgenin as negative control.
  • LDH lactate dehydrogenase
  • the objective of this study was to examine the anti-apoptotic effect of smilagenin on the caspase-3 activity, a marker of apoptosis, in rat primary cortical cultures exposed to glutamate
  • Rat cortical neurones were cultured for 6 days. At day 6, glutamate (100 microM, 10 min) was added. Then the cultures were washed and medium was replaced with fresh medium containing smilagenin or vehicle control (DMSO, 0.25%) for 6 h. After 6 h treatment, apoptosis was evaluated by measuring caspase 3 activity. Caspase 3 activity was detected by the cleavage of p-nitroaniline from a colorimetric caspase-3 substrate, acetyl-Asp-Glu- Val-Asp p-nitroanilide. p-Nitroalanine has a high absorbance at 405 nM. Relative caspase- 3 activity was measured as optical density.
  • Neurodegenerative disorders are characterised by a progressive loss of neurones and degradation of neuronal processes (neurites).
  • Agents that induce neurite outgrowth may promote the formation of new connections between neurones and ameliorate the symptoms of neurodegenerative conditions (Katzman et al, Faseb J., 5, 278-286, 1991).
  • MPP + a metabolite of l-methyl-4-phenyl-l,2,3,6- tetrahydropyridine (MPTP) mimics the degeneration of nigrostriatal dopaminergic neurones observed in neurodegenerative disorders such as Parkinson's disease (Mytinlineou et al, Science, 225, 529-531, 1984).
  • biochemical changes induced by this toxin include decreased levels of dopamine and its metabolites in the substantia nigra pars compacta and in the caudate nucleus (Burns et al, Proc Natl Acad Sci U.S.A., 80, 4546- 4550, 1983) and a reduction in dopamine uptake in nigrostriatal synaptosomal preparations (Heikkila et al, J Neurochem., 44, 310-313, 1985).
  • Pre-treatment with smilagenin and sarsasapogenin produced a significant increase in the neuronal survival compared to neurones exposed to MPP + alone, suggesting a significant neuroprotective effect (Table 7).
  • the objective of this study was to examine the effects of sarsasapogenin and smilagenin on the survival of rat primary spinal motor neurones exposed to glutamate, which is known to induce neurodegeneration in this model of motor neurodegeneration. 17 ⁇ -oestradiol and BDNF were used as positive controls.
  • Rat motor neurones were prepared according to the method described by (Martinou et al, Neuron, 8, 737-744, 1992). On day 10, medium was removed and the cultures were exposed to glutamate (4 microM) for 10 min at 37°C in defined medium. After the glutamate exposure, cultures were washed with Dulbecco modified Eagle medium at 37°C then placed in fresh culture medium containing test compounds. After 48 h, the extent of spinal motor neurone degeneration was determined by measuring the amount of lactate dehydrogenase (LDH) released into the culture medium as above.
  • LDH lactate dehydrogenase
  • BDNF brain derived neurotrophic factor
  • acetylcholine receptor density muscarinic and nicotinic
  • a decrease in their coupling function in cortical areas Rosci., 18, 223-253, 1995
  • muscarinic acetylcholine receptor binding is significantly reduced in the hippocampus (Narang, N, Mech. Ageing Dev., 78, 221-239, 1995) and striatum of older rats (Biegon et al, Neurobiol.
  • a Y-maze apparatus was used for the learning and memory test.
  • On the floor of each arm of the Y-maze is an array of copper rods to which electric current is applied whenever needed, with adjustable voltage.
  • Each arm is 45 cm long and has a 15 W lamp at the end, which is turned on when needed.
  • each rat was trained for 7 consecutive days, as follows. For each training session, the rat was put into one arm of the Y-maze, after two minutes rest, an electrical current was applied to the copper rods and the lamp of the clockwise arm was illuminated to indicate the non-stimulation area. If the rat went into that arm, one correct response was recorded, otherwise, one wrong response was recorded.
  • This stimulation-response test was repeated 20 times each day, with a pause of 5 sec between each two consecutive tests.
  • the number of correct responses following the twenty tests on the seventh day was used to express learning ability, (the higher the number the better the learning ability).
  • the rats were then left resting for 30 days and the procedure was repeated once more.
  • the number of correct responses of the 20 tests after the 30 day rest period was used to represent the memory ability
  • Muscarinic acetylcholine receptor density in the brain was measured. Tissue was prepared as follows: brains were removed quickly after decapitation, frozen in dry ice, and transferred to a freezer. The brains were homogenised and the pellet was finally suspended in buffer.
  • the dual-site competitive ligand binding assay was used to measure muscarinic acetylcholine receptor density.
  • the compounds reverse the neuroimpairment, the decline in BDNF levels, and the decline in the muscarinic acetylcholine and dopamine receptor density that occur in aged rats.
  • Alzheimer's disease model as a model of neurodegeneration
  • Alzheimer's disease An in vivo model of Alzheimer's disease was used to model neurodegeneration.
  • the neurotoxic agents amyloid ⁇ and ibotenic acid
  • amyloid ⁇ and ibotenic acid are injected into the brain of the rat. This leads to neuronal loss, receptor loss and cognitive impairment.
  • Previous studies showed that local injection of amyloid ⁇ in the nucleus vasalis of the rat brain caused cholinergic hypofunction and behavioural impairment up to two months post surgery (Giovannelli et al., 1995: Neuroscience, 66, 781-792.).
  • the dose for each rat was amyloid ⁇ M0 (4 ⁇ g) and ibotenic acid (1 ⁇ g) in 1 ⁇ l of saline. The injection was completed in 20 min, and the needle was withdrawn 10 min later. Then the skin was sutured.
  • the 8 groups were:
  • Episarsasapogenin cathylate, sarsasapogenin cathylate, episarsasapogenin ethylsuccinate (comparison compound), episarsasapogenin, epismilagenin and diosgenin (all at a dosage of 18 mg/kg/day) were administered to animals as stable suspensions in CMC-Na (0.5%) once daily through a gastric tube.
  • the control and the model group were given the same volume of CMC-Na (0.5%) once daily.
  • the drugs and vehicles were given for a period of two months, starting 20 days before operation.
  • Muscarinic acetylcholine receptor density was assessed.
  • the brain samples were homogenised, centrifuged, and the pellet of centrifugation at 27000xg was re-homogenised and used for measurement.
  • the concentration of 3 H-QNB was chosen at the saturation range. After incubation and separation, the bound portion was measured by liquid scintillation counter.
  • Step-Through Test learning and memory. The effect of test compounds on learning and memory was assessed using the step-through test.
  • the experiment is carried out for each rat on two consecutive days. The first day is for training; when the rat is adapted in the box for the first 3 min, then put in the light room, with its back toward the hole, and the copper rods of the dark room are charged for 5 min. The second day is for testing, when the number of crosses in 5 min are recorded. Improvements in memory are signalled by a reduction in the number of crosses.
  • the muscarinic acetylcholine receptor density in the neurodegeneration model brains was significantly lower than control.
  • Episarsasapogenin cathylate, sarsasapogenin cathylate, episarsasapogenin and epismilagenin produced a significant elevation in brain muscarinic acetylcholine receptor density, whereas diosgenin and episarsasapogenin ethylsuccinate did not significantly change the muscarinic acetylcholine receptor density.
  • the compounds of this invention act to normalise receptor number, i.e. they tend to restore receptor number to normal levels when given to animals in which the receptor level is depressed.
  • ALS Amyotrophic lateral sclerosis
  • SOD-1 Cu Zn superoxide dismutase
  • Animal models of this disease include the SOD-1 transgenic mice over-expressing SOD-1 gene and the progressive motor neuropathy (pmn, a model of Charcot-Marie-Tooth) mice.
  • SOD superoxide dismutase
  • pmn mouse
  • ALS amyofrophic lateral sclerosis
  • Charcot-Marie-Tooth disease two models of amyofrophic lateral sclerosis (ALS) and Charcot-Marie-Tooth disease.

Abstract

The invention discloses therapeutic methods and uses of certain steroidal sapogenins, related compounds and derivatives thereof, in the treatment of non-cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration or receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment.

Description

THERAPEUTIC METHODS AND USES OF SAPOGENINS AND THEIR
DERIVATIVES
Field of the Invention
The present invention relates to therapeutic methods and uses of sapogenins, related compounds and their derivatives.
The uses of the sapogenins, related compounds and their derivatives are in the treatment of non-cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor- sensory neurodegeneration, or receptor dysfunction or loss. In a further aspect, the invention relates to compositions for use in such treatments.
Background of the Invention
Cognitive dysfunction is a characteristic of dementia conditions and syndromes, such as Alzheimer's disease (AD), senile dementia of the Alzheimer's type (SDAT), Lewy body dementia and vascular dementia. A lesser degree of cognitive dysfunction is also a characteristic of certain non-dementia conditions and syndromes, such as mild cognitive impairment (MCI), age-associated memory impairment (AAMI), autism and neuroimpairment.
Non-cognitive neurodegeneration (i.e. neurodegeneration in the absence of cognitive dysfunction), non-cognitive neuromuscular degeneration (i.e. neuromuscular degeneration in the absence of cognitive dysfunction) and motor-sensory neurodegeneration are characteristic of conditions and syndromes such as Parkinson's disease, muscular dystrophy including facioscapulohumeral muscular dystrophy (FSH), Duchenne muscular dystrophy, Becker muscular dystrophy and Bruce's muscular dystrophy, Fuchs' dystrophy, myotonic dystrophy, corneal dystrophy, reflex sympathetic dystrophy syndrome (RSDSA), neurovascular dystrophy, myasthenia gravis, Lambert Eaton disease, Huntington's disease, amyotrophic lateral sclerosis (ALS) and multiple sclerosis.
Receptor dysfunction or loss - particularly dysfunction or loss of nicotinic and/or muscarinic acetylcholine receptors and/or dopamine receptors and/or adrenoceptors - is a characteristic of some or all of the above conditions and syndromes. Receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment is also a characteristic of conditions and syndromes such as postural hypotension, chronic fatigue syndrome, asthma, susceptibility to heart failure and macular degeneration.
The above conditions and syndromes are grave and growing problems in all societies where, because of an increase in life expectancy and control of adventitious disease, the demographic profile is increasingly extending towards a more aged population. Agents which can treat, or help in the management or prevention of such disorders, are urgently required.
DE-A-4303214, the disclosure of which is incorporated herein by reference, suggests the use of saponins and sapogenins in the treatment of viral diseases, but with no data that would allow one skilled in the art to select a particular group of compounds for any particular viral disease.
WO-A-99/16786, the disclosure of which is incorporated herein by reference, suggests the use of certain saponins and sapogenins in the treatment of dementia.
WO-A-99/48482, WO-A-99/48507, WO-A-01/23406, WO-A-01/23407, WO-A-01/23408, the disclosures of which are incorporated herein by reference, relate to the use of certain saponins, sapogenins and derivatives thereof in the treatment of cognitive dysfunction and allied conditions.
Chinese Patent Application No. CN-A- 1096031, the disclosure of which is incorporated herein by reference, suggests a bioactivity of the spirostane sapogenin, sarsasapogenin, in the two-way regulation of β-adrenergic and M-cholinergic receptors. No specific pharmaceutical activity is suggested. However, in "Synthesis and Applications of Isotopically Labelled Compounds", 1998, pages 315-320, Yi et al describe the use of sarsasapogenin in the treatment of senile dementia.
There exist a number of so-called "spectrum" disorders, in which a wide range of combinations of symptoms, in a wide range of relative severities, present themselves. The severity of each symptom and the particular combination of symptoms will vary from individual to individual and according to the stage of progression of the disorder. In the cases of Parkinson's disease, myasthenia gravis, Lambert Eaton disease, postural hypotension and chronic fatigue syndrome, for example, cognitive dysfunction is not a primary symptom, although it may be present as one of a number of possible secondary symptoms. Moreover, these conditions are not viral diseases or dementias. Many of these disorders are so-called "spectrum" disorders, Therefore, in many instances, a treatment for cognitive dysfunction (e.g. dementia) is not necessary.
The present invention is based upon our finding that certain sapogenins and their derivatives, including saponins, have a surprising disease-modifying activity against non- cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration as well as against receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment, and thus actively prevent and reverse the conditions. This finding enables improved treatment of certain non-viral, spectrum and non-spectrum, disorders in which cognitive dysfunction is not a primary symptom.
Brief Description of the Invention
According to one aspect of the present invention, there is provided the use of the active agents (as herein defined) in the treatment or prevention of, or in the preparation of compositions (e.g. pharmaceutical compositions, foodstuffs, food supplements and beverages) for the treatment or prevention of, (i) non-cognitive neurodegeneration, (ii) non- cognitive neuromuscular degeneration, (iii) motor-sensory neurodegeneration (iv) receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment, in human and non-human animals suffering therefrom or susceptible thereto.
The expression "active agents" refers to compounds of the following general formulae I, II and HI, sapogenin derivatives as defined below by reference to derivatives bearing at least one defined X radical substituent, sugar-substituted derivatives of such compounds as defined below, all their stereoisomers and racemic mixtures, all their pharmaceutically acceptable pro-drugs and salts, and all mixtures and combinations thereof:
Formula I:
Figure imgf000005_0001
(I)
wherein in the general formula (I):
- Ri, R2, R3, 4, R5. Re, R7. R8. Rio, R13, Ri8, R19, R20, R21. R22, R23. R24. R26, R27. R28. R29, R3o, R31, R32, are, independently of each other, either H, OH, =O, halo atom, (Me-S-), (Me- SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl or absent or OR where R = alkyl or acyl group;
- R9, Ru, Ri2,Ri4, R.5, Ri6, Rπ, R25, R33 can be either a H, OH, halo atom, (Me-S-), (Me-SO- ), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl or absent or OR where R = alkyl or acyl group; represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group;
Formula II:
Figure imgf000005_0002
(π) wherein in the general formula (II):
- Ri, R2, R3, 4) R5. Rδ> R?) Re. Rio. R13, Ris, R19, R20, R21, R22. R23, R24, R_6, 27, 28, R29, R30, R31, R32, R34, are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent;
- R9, R11, Rπ, R14, R15, Ri6. Rπ. R25, R33, R35 can be either a H, OH, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group;
Formula III:
Figure imgf000006_0001
(HI)
wherein in the general formula (HI): - Ri, R2, R3, R4, R5, Re, R7, Rs, Rio, R13, R14, Ris, R19, R20, R21, R22, R23, R24, R26, R27, 28,
R29, R30, R31, R32, R33, R34, R35j R36, R37 are, independently of each other, either H, OH, =O, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent;
-R9, R„, R12, R15) Ri6, Rπ, R25 can be either H, OH, halo atom, (Me-S-), (Me-SO-), (Me- SO2), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation;
sapogenin derivatives, particularly but not exclusively steroidal spirostane sapogenin derivatives, bearing at least one X radical substituent, wherein X is chosen from the group consisting of :
- halo atom,
- (Me-S-), (Me-SO-), (Me-SO2-),
- N3-, NH2-, MeSO2NH-, and
- alkyl; and
derivative forms of any of the above compounds, in which the carbon atom at the 3-position (i.e. the carbon to which R3 is attached, or the corresponding position in the sapogenin derivatives defined above without a formula) or, in the case of Formulae II and III, the 3- position carbon atom, the 26-position (i.e. the carbon to which R34 is attached) or each of the carbon atoms at the 3- and 26-positions, carries an O-sugar moiety wherein the sugar group is a mono-, di- or tri-saccharide.
Sugar-carrying derivative forms of sapogenin active agents are generally referred to in the art as saponins. The expression "carbohydrate" used herein includes particularly such sugar groups.
The active agents used in the present invention are preferably the non-estrogenic steroidal sapogenins, saponins, and derivatives thereof within the terms of the above definition, including all physiologically acceptable pro-drugs and salts thereof.
The active agents may be naturally occurring or non-naturally occurring. Non-naturally occurring active agents may suitably be prepared by modification of side groups and/or side atoms of naturally occurring compounds, as described below and as known in the art.
The invention also provides corresponding methods for the treatment of human and non- human animals, and compositions containing the active agents for use in the said treatment methods.
The active agents of the invention may, if desired, be co-administered with one or more additional active agent, for example one or more agent selected from, but not limited to, cholinesterase inhibitors, dopamine agonists (e.g. L-dopa), COMT inhibitors, MAO-B inhibitors, anti-cholinergics, acetylcholine agonists, serotonin agonists, AMPA receptor agonists, GABA receptor agonists, NMDA receptor agonists, β-adrenoceptor agonists, digoxin, dobutamine, anti-inflammatories, neurotrophic factors, statins, adenosine A2a receptor antagonists, aldose reductase inhibitors, immunomodulators, cannabinoid agonists, interferon β or tricyclic anti-depressants.
The active agents may be applied therapeutically or prophylactically to human and non- human animals suffering from, or susceptible to, conditions and diseases that are characterised by non-cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration, or receptor dysfunction or loss. Examples of such conditions and diseases are provided below.
I
The present invention therefore also provides the use of the active agents (as herein defined) in the treatment or prevention of, or in the preparation of compositions (e.g. pharmaceutical compositions, foodstuffs, food supplements and beverages) for the treatment or prevention of, one or more of said conditions and diseases in human and non- human animals suffering therefrom or susceptible thereto.
Detailed Description of the Invention
Examples of Active Agents
The following classes of active agents are particularly mentioned:
1. Compounds of the above general formula I, wherein:
- Ri, R2, R3, Rt, R5, Re, R7, R8, Rio, R13, Ris, R19, R20, R21 , R22, R23, R24, R26, R27, 285 R29, R30, R31, R32 are, independently of each other, either H, OH, =O, halo atom, (Me-S-), (Me- SO-), (Me-SO2-), N3-, NH2-. MeSO2NH-, alkyl or absent or OR where R = alkyl or acyl group; - R9, Rπ, R12, Ri4, Ris, Ri6, Rπ, R25, R33 can be either a H, OH, halo atom, (Me-S-), (Me-SO- ), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl or absent or OR where R = alkyl or acyl group; represents an optional double bond, wherein in addition to the above
- either R33 or Rι = alkyl group, and the stereochemistry of R25 is in the β orientation; 2. Compounds of the above general formula I, wherein:
- Ri, 2, R3, R4, R5, Re, 7, R8, Rio, R13, is, R19, R2o, R21, R22, R23, R2 , 26, 27, R28, R29, R30, R31, R32 are, independently of each other, either H, OH, =O, halo atom, (Me-S-), (Me- SO-), (Me-SO2), N3-, NH2-, MeSO2NH-, alkyl or absent or OR where R = alkyl or acyl group;
- R , Rl2, Rl5, Rl6, Rl7 = H,
- Rπ, R14, R25, R33 can be either a H, OH, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl, or absent or OR where R = alkyl or acyl group; represents an optional double bond wherein in addition to the above
- either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation;
3. Compounds of the above general formula I, wherein: -Rι=
Figure imgf000009_0001
R21= R∑2= R23=
Figure imgf000009_0002
R33 = H,
- either R33 or R14 = CH3 represents a single bond,
- the methyl group at C25 may be either in the R or S configuration - the stereochemistry of R25 is in the β orientation and wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of : - halo atom,
- (Me-S-), (Me-SO-), (Me-SO2-), and
- N3-, NH2-, MeSO2NH-
- alkyl;
4. Compounds of the above general formula I, wherein: Ris = R19 = R20 ~
Figure imgf000009_0003
-Rι4= R33 = CH3, represents a single bond, - the stereochemistry of R25 is in the β orientation and wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =O, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of :
- halo atom, - (Me-S-), (Me-SO-), (Me-SO2-), and
- N3-, NH2-, MeSO2NH-
- alkyl;
5. Compounds of the above general formula H", wherein - Ri, R2, R3, R4, R5, Re, R7, R8, Rio, π, Ris, R19, R20, R21, R22, R23, R24, 26, R27, R28, R29, R30, R31, R32, R34 are, independently of each other, either H, OH, =O, halo atom, (Me-S-), (Me-SO-), (Me-SO2), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent;
- R9, Ru, Rι2, R14, R15, R16, Rπ, R25, R33, R35 can be either a H, OH, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or Ri4 = alkyl group, and the stereochemistry of R25 is in the β orientation;
6. Compounds of the above general formula II and carbohydrate derivatives thereof, wherein:
- Ri, R2, R3, R4, R5, R~, 7, Re, Rio, Rπ, Ris, R19, R20, R21, R22, R23, R24, 26, R27, R28, R29, R30, R31, R32 are, independently of each other, either H, OH, =O, halo atom, (Me-S-), (Me- SO-), (Me-SO2), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent;
- R9, Rι2, R15, R1 , π = H,
- R3 = either H, OH, =O, and OR where R = alkyl, acyl or carbohydrate and - R, 1, Ri4, R25, R33, R35 can be either H, OH, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation; 7. Compounds of the above general formula H" and carbohydrate derivatives thereof, wherem:
-Rl=
Figure imgf000011_0001
Rl6 = Rl7 = Rl8 = Rl = 20 =
Figure imgf000011_0002
-Rι4 = CH3,
- R34= -OH or -OR where R = alkyl, acyl or carbohydrate and R35 = H or is absent represents an optional double bond, and
- the methyl group at C25 may be either in the R or S configuration and and the stereochemistry of R25 is in the β orientation wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of : - halo atom,
- (Me-S-), (Me-SO-), (Me-S02-), and
- N3-, NH2-, MeSO2NH-
- alkyl;
8. Compounds of the above general formula II and carbohydrate derivatives thereof, wherein: R17 = Ris = R19 = R20 =
Figure imgf000011_0003
-R]4 = R33 = CH3, - R34= -OH or -OR where R = alkyl, acyl or carbohydrate and
R35 = H or is absent represents an optional double bond, and the stereochemistry of R25 is in the β orientation, and wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of :
- halo atom,
- (Me-S-), (Me-SO-), (Me-SO2-), and
Figure imgf000011_0004
- alkyl; 9. Compounds of the above general formula HI, wherein:
- Ri, R2, R3, R , R5, Rg, R7, R8, Rio, R13, R14, Ris, R19, R20, R21, R22, R23, R24, R26, R27, 28, R29, R30, R31, R32, R33, R34, R35, R36, R37 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-S02-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent;
-R9, R11, R12, R15, Ri6, R17, R25 can be either H, OH, halo atom, (Me-S-), (Me-SO-), (Me- S02), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation;
10. Compounds of the above general formula in, wherein: - Ri, R2, R3, R4, R5, R$, R7, R8, Rio, R13, R14, Ris, R19, R20, R21, R22, R23, R24, 26, R27, 28, R29j R30, R31, R32, R33, R35, R36, R37 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent;
- R9, Rι2, R15, Ri6, Rπ = H, - R34 = H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-S02-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl, acyl or carbohydrate, or absent;
- Rπ, R25, can be either H, OH, halo atom, (Me-S-), (Me-SO-), (Me-S02-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation;
11. Compounds of the above general formula m and carbohydrate derivatives thereof, wherein:
Figure imgf000012_0001
-Rl4 = CH3,
- R34= -OH or -OR where R = alkyl, acyl or carbohydrate and R35 = H or is absent
R37 = H, -OH or =0 R36 = H or -OH represents a single bond, and
- the methyl group at C25 may be either in the R or S configuration and the stereochemistry of R25 is in the β orientation, wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of :
- halo atom, - (Me-S-), (Me-SO-), (Me-SO2-), and - N3-. NH2-, MeSO2NH-
- alkyl;
12. Compounds of the above general formula HI and carbohydrate derivatives thereof, wherein:
Figure imgf000013_0001
- R34= -OH or -OR where R = alkyl, acyl or carbohydrate and R35 = H or is absent
R37 = H, -OH or =0
R36= H or OH represents a single bond, and
- the methyl group at C25 may be either in the R or S configuration and the stereochemistry of R25 is in the β orientation wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of : - halo atom,
- (Me-S-), (Me-SO-), (Me-SO2-), and
- N3-, NH2-, MeS02NH-
- alkyl; 13. Substituted sapogenins, particularly but not exclusively steroidal spirostane sapogenins, wherein at least one OH-group of the sapogenin is substituted with X, chosen from the group consisting of :
- halo atom, - (Me-S-), (Me-SO-), (Me-S02-),
- N3-, NH2-, MeS02NH-, and
- alkyl;
14. Sapogenins defined above, particularly but not exclusively steroidal spirostane sapogenins, wherein in the definition of X the halo atom is a fluoro atom;
15. Substituted sapogenins selected from :
(3β-fluoro-5β, 20α,22α,25R-spirostane), (3,3-difluoro-5β, 20α,22α,25R-spirostane), (3α- methylsulphonylamino-5β,20α.22α,25R-spirostane), (3α-azido-5β, 20α,22α,25R-spiro- stane), (3α-amino-5β,20α,22α,25R-spirostane), and their stereoisomers and racemic mixtures, their pharmaceutically acceptable pro-drugs and salts;
16. Substituted sapogenins wherein the parent sapogenin which is then substituted with at least one X radical as defined above is selected from sarsasapogenin, episarsasapogenin, smilagenin, epismilagenin, and anzurogenin-D;
17. Compounds of the general formula la :
Figure imgf000014_0001
(la)
wherein the group R is selected from hydrogen; alkylcarbonyl; alkoxycarbonyl; alkyl- carbamoyl; or arylcarbonyl; or sulpho (H03S); phosphono ((HO)2P(0)-); or a mono-, di- or tri-saccharide; wherein any alkyl group is optionally substituted with aryl, amino, mono- or di-alkyl-amino, a carboxylic acid residue (-COOH), or any combination thereof;
18. Derivative forms of the above compounds as defined as items 1 to 17, in which the 3-position carbon atom or, in the case of Formulae H and HI, the 3-position carbon atom, the 26-position carbon atom or each of the carbon atoms at the 3- and 26-positions, carries an O-sugar moiety wherein the sugar group is a mono-, di- or tri-saccharide, for example a mono aldose or ketose having 5 or 6 carbon atoms, preferably in the cyclised furanose or pyranose form, either as the α or β anomer and having D or L optical isomerism, or any di- and tri-oligosaccharide combination thereof; acylated forms of sugar residues are also to be included within the term "sugar"; examples of suitable sugars include glucose, mannose, fructose, galactose, maltose, cellobiose, sucrose, rhamnose, xylose, arabinose, fucose, quinovose, apiose, lactose, galactose-glucose, glucose-arabinose, fucose-glucose, rhamnose-glucose, glucose-glucose-glucose, glucose-rhamnose, mannose-glucose, glucose- (rhamnose)-glucose, glucose-(rhamnose)-rhamnose, glucose-(glucose)-glucose, galactose- (rhamnose)-galactose and acylated (e.g. acetylated) derivatives thereof.
In the above compound definitions:
Optional amino, mono-alkyl-amino and di-alkyl-amino substituents of alkyl groups, where present, are preferably a mono-substituent at the α position of the alkyl group.
Optional -COOH substituents of alkyl groups, where present, may be at the terminal or any other position of the alkyl group.
"Alkyl" means an aliphatic hydrocarbon group which may be straight or branched having about 1 to about 20 carbon atoms in the chain. Preferred alkyl groups have 1 to about 12 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl are attached to a linear alkyl chain. "Lower alkyl" means about 1 to about 4 carbon atoms in the chain which may be straight or branched. Exemplary alkyl groups include methyl, ethyl, n-propyl, t-propyl, M-butyl, t-butyl, _?-butyl, n-pentyl, 3-pentyl.
"Aryl" means any group comprising an aromatic ring or system of fused rings, and preferably contains up to 12 carbon atoms. An exemplary aryl group is the phenyl group. An aryl group may optionally be mono- or poly-substituted, for example by substituents independently selected from halo (e.g. chloro or bromo), alkyl, cycloalkyl, hydroxy, alkoxy, amino, nitro, acylamino, carboxy and alkoxycarbonyl.
"Carboxylic acid residue" means the group -COOH.
"Acyl" means an H-CO- or Alkyl-CO- group wherein the alkyl group is as defined below. Preferred acyls contain a lower alkyl. Exemplary acyl groups include formyl. acetyl, propanoyl, 2-methylpropanoyl, butanoyl and palmitoyl;
"Optionally substituted" means that the said group may be substituted with one or more substituents, which may be the same or different, preferably one or more substituents which individually have a size which is small (e.g. less than about 20% of the largest molecular dimension) in relation to the parent group being substituted; suitable substituents include halo (e.g. chloro or bromo), alkyl, cycloalkyl, hydroxy, alkoxy, amino, acylamino, aryl, aroylamino, carboxy, alkoxycarbonyl, aralkoxycarbonyl, heteroaralkoxycarbonyl, and optionally substituted carbamoyl, preferably subject to the size limitation set out above ;
"Pharmaceutically acceptable" means it is, within the scope of sound medical and veterinary judgement, suitable for use in contact with the cells of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. "Pharmaceutically acceptable prodrugs" means those prodrugs of the compounds which are, within the scope of sound medical and veterinary judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds. The term "prodrug" means compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. Functional groups which may be rapidly transformed, by metabolic cleavage, in vivo form a class of groups reactive with the carboxyl group. Because of the ease with which the metabolically cleavable groups of the compounds are cleaved in vivo, the compounds bearing such groups act as pro-drugs. A thorough discussion of prodrugs is provided in the following: Design of Prodrugs, H. Bundgaard, ed., Elsevier, 1985; Methods in Enzymology, K. Widder et al, Ed., Academic Press, 42, p.309-396, 1985; A Textbook of Drug Design and Development, Krogsgaard-Larsen and H. Bundgaard, ed., Chapter 5; Design and Applications of Prodrugs p.113-191, 1991; Advanced Drug Delivery Reviews, H. Bundgard, 8, p.1-38, 1992; Journal of Pharmaceutical Sciences, 77, p. 285, 1988; Chem. Pharm. Bull., N. Nakeya et al, 32, p. 692, 1984; Pro-drugs as Novel Delivery Systems, T. Higuchi and V. Stella, Vol. 14 of the A.C.S. Symposium Series, and Bioreversible Carriers in Drug Design, Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, 1987, which are incorporated herein by reference;
"Pharmaceutically acceptable salts" means the relatively non-toxic, inorganic and organic acid addition salts, and base addition salts, of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds. In particular, acid addition salts can be prepared by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. See, for example S. M. Berge, et al., Pharmaceutical Salts, J. Pharm. Sci., 66: p.l- 19 (1977) which is incorporated herein by reference. Base addition salts can also be prepared by separately reacting the purified compound in its acid form with a suitable organic or inorganic base and isolating the salt thus formed. Base addition salts include pharmaceutically acceptable metal and amine salts. Examples of suitable acid addition salts are those formed with acids selected from hydrochloric, sulphuric, phosphoric and nitric acids. Examples of suitable base addition salts are those formed with bases selected from sodium hydroxide, potassium hydroxide and ammonium hydroxide.
A particularly preferred class of active agents are the compounds of the general formula la.
In some of the compounds of formula la, the C25 methyl group is in the S configuration; these compounds of the invention are sarsasapogenin and episarsasapogenin or derivatives thereof. In other compounds of formula la, the C25 methyl group is in the R configuration; these compounds of the invention are smilagenin and epismilagenin or derivatives thereof.
In the above formula la, -OR may, for example, be selected from the following (unless excluded by proviso):, hydroxy, cathylatc (ethoxycarbonyloxy), acetate, succinate, cinnamate, ferulate, propionate, butyrate, valerate, isovalerate, caproate, isocaproate, diethylacetate, octanoate, decanoate, laurate, myristate, palmitate, stearate, benzoate, phenylacetate, phenylpropionate, cinnamate, p-nitrobenzoyloxy, 3,5-dinitrobenzoyloxy, p- chlorobenzoyloxy, 2,4-dichlorobenzoyloxy, p-bromobenzoyloxy, m-bromobenzoyloxy. p- methoxybenzoyloxy, phthalyl, glycinate, alaninate, valinate, phenylalaninate, isoleucinate, methioninate, argininate, asparaginate, aspartate, cysteinate, glutamate, histidinate, lysinate, prolinate, serinate, threoninate, tryptophanate, tyrosinate, fumarate or maleate. Of the compounds of general formula la and their pharmaceutically acceptable salts, particularly preferred are the following compounds: sarsasapogenin sarsasapogenin cathylate sarsasapogenin acetate sarsasapogenin succinate and pharmaceutically acceptable salts thereof sarsasapogenin glycinate and pharmaceutically acceptable salts thereof sarsasapogenin alaninate and pharmaceutically acceptable salts thereof sarsasapogenin valinate and pharmaceutically acceptable salts thereof sarsasapogenin phenylalaninate and pharmaceutically acceptable salts thereof sarsasapogenin isoleucinate and pharmaceutically acceptable salts thereof sarsasapogenin methioninate and pharmaceutically acceptable salts thereof episarsasapogenin episarsasapogenin cathylate episarsasapogenin acetate episarsasapogenin succinate and pharmaceutically acceptable salts thereof episarsasapogenin glycinate and pharmaceutically acceptable salts thereof episarsasapogenin alaninate and pharmaceutically acceptable salts thereof episarsasapogenin valinate and pharmaceutically acceptable salts thereof episarsasapogenin phenylalaninate and pharmaceutically acceptable salts thereof episarsasapogenin isoleucinate and pharmaceutically acceptable salts thereof episarsasapogenin methioninate and pharmaceutically acceptable salts thereof smilagenin smilagenin cathylate smilagenin acetate smilagenin succinate and pharmaceutically acceptable salts thereof smilagenin glycinate and pharmaceutically acceptable salts thereof smilagenin alaninate and pharmaceutically acceptable salts thereof smilagenin valinate and pharmaceutically acceptable salts thereof smilagenin phenylalaninate and pharmaceutically acceptable salts thereof smilagenin isoleucinate and pharmaceutically acceptable salts thereof smilagenin methioninate and pharmaceutically acceptable salts thereof epismilagenin epismilagenin cathylate epismilagenin acetate epismilagenin succinate and pharmaceutically acceptable salts thereof epismilagenin glycinate and pharmaceutically acceptable salts thereof epismilagenin alaninate and pharmaceutically acceptable salts thereof epismilagenin valinate and pharmaceutically acceptable salts thereof epismilagenin phenylalaninate and pharmaceutically acceptable salts thereof epismilagenin isoleucinate and pharmaceutically acceptable salts thereof epismilagenin methioninate and pharmaceutically acceptable salts thereof.
Of the saponin (R = sugar) compounds of general formula la, particularly preferred are the following compounds: sarsasapogenin, episarsasapogenin, smilagenin and epismilagenin in which, in each case, the 3-position carbon atom carries an O-sugar moiety wherein the sugar group is selected from glucose, mannose, fructose, galactose, maltose, cellobiose, sucrose, rhamnose, xylose, arabinose, fucose, quinovose, apiose, lactose, galactose-glucose, glucose-arabinose, fucose-glucose, rhamnose-glucose, glucose-glucose-glucose, glucose- rhamnose, mannose-glucose, glucose-(rhamnose)-glucose, glucose-(rhamnose)-rhamnose, glucose-(glucose)-glucose, galactose-(rhamnose)-galactose and acylated (e.g. acetylated) derivatives thereof.
Further examples of suitable active agents include 16,22-epoxycoprostan-3β-ol, smilagenone, coprosterol and pharmaceutically acceptable pro-drugs and salts thereof.
Compositions and Uses
The present invention thus enables and provides a method for treating or preventing non- cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration or receptor dysfunction or loss in the absence of cognitive, neural or neuromuscular impairment (particularly but not exclusively in relation to the specific disease states mentioned above) in a human or non-human animal in need thereof, which comprises administering to the said human or non-human animal an effective dosage of an active agent (as defined herein) or a pharmaceutically acceptable salt thereof.
The active agent may be administered in the form of a composition comprising the active agent and any suitable additional component. The composition may, for example, be a pharmaceutical composition (medicament), a foodstuff, food supplement or beverage. Such a composition may contain a mixture of the specified compounds, and/or of their pharmaceutically acceptable salts. According to a further aspect of the present invention, there is provided a composition having activity against, and for use in treating, non-cognitive neurodegeneration, non- cognitive neuromuscular degeneration, motor-sensory neurodegeneration or receptor dysfunction or loss in the absence of cognitive, neural or neuromuscular impairment in a human or non-human animal, which comprises an effective amount of a compound of the active agent.
The term "pharmaceutical composition" in the context of this invention means a composition comprising an active agent and comprising additionally pharmaceutically acceptable carriers, diluents, adjuvants, excipients, or vehicles, such as preserving agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, sweetening agents, flavoring agents, perfuming agents, antibacterial agents, antifungal agents, lubricating agents and dispensing agents, depending on the nature of the mode of administration and dosage forms.
The terms "foodstuff, "food supplement" and "beverage" used herein have the normal meanings for those terms, and are not restricted to pharmaceutical preparations.
The dosage of the active agent will vary widely, depending on the severity of the symptoms to be treated or prevented. The selection of appropriate dosages is within the ability of one of ordinary skill in this art, without undue burden. The dosage of the active agent may, for example, be greater than about 0.1 mg/kg body weight, for example greater than about 0.3 mg/kg body weight, preferably administered once per day. More typically, the dosage will be between about 1 and about 25 mg/kg, e.g. between about 1 and about 10 mg/kg, preferably administered once per day. For human use, the dosage may conveniently be between about 70 and about 700 mg per day.
"Pharmaceutically acceptable dosage forms" means dosage forms of the compounds or compositions of the invention, and includes, for example, tablets, dragees, powders, elixirs, syrups, liquid preparations, including suspensions, sprays, inhalants, tablets, lozenges, emulsions, solutions, granules, capsules and suppositories, as well as liquid preparations for injections, including liposome preparations. Techniques and formulations generally may be found in Remington, Pharmaceutical Sciences, Mack Publishing Co., Easton, PA, latest edition. In general, reference herein to the presence of one of a specified group of compounds includes within its scope the presence of a mixture of two or more of such compounds.
This invention provides for the treatment of (i) non-cognitive neurodegeneration, (ii) non- cognitive neuromuscular degeneration, (iii) motor-sensory neurodegeneration, or (iv) receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment, in a human or non-human animal subject suffering from, or susceptible to, any of: Parkinson's disease, postencephalitic Parkinsonism, depression, schizophrenia, muscular dystrophy including facioscapulohumeral muscular dystrophy (FSH), Duchenne muscular dystrophy, Becker muscular dystrophy and Brace's muscular dystrophy, Fuchs' dystrophy, myotonic dystrophy, corneal dystrophy, reflex sympathetic dystrophy syndrome (RSDSA), neurovascular dystrophy, myasthenia gravis, Lambert Eaton disease, Huntington's disease, motor neurone diseases including amyotrophic lateral sclerosis (ALS), multiple sclerosis, postural hypotension, traumatic neurodegeneration e.g. following stroke or following an accident (for example, traumatic head injury or spinal cord injury), Batten's disease, Cockayne syndrome, Down syndrome, corticobasal ganglionic degeneration, multiple system atrophy, cerebral atrophy, olivopontocerebellar atrophy, dentatorubral atrophy, pallidoluysian atrophy, spinobulbar atrophy, optic neuritis, sclerosing pan-encephalitis (SSPE), attention deficit disorder, post-viral encephalitis, post-poliomyelitis syndrome, Fahr's syndrome, Joubert syndrome, Guillain-Barre syndrome, lissencephaly, Moyamoya disease, neuronal migration disorders, autistic syndrome, polyglutamine disease, Niemann- Pick disease, progressive multifocal leukoencephalopathy, pseudotumor cerebri, Refsum disease, Zellweger syndrome, supranuclear palsy, Friedreich's ataxia, spinocerebellar ataxia type 2, Rhett syndrome, Shy-Drager syndrome, tuberous sclerosis, Pick's disease, chronic fatigue syndrome, neuropathies including hereditary neuropathy, diabetic neuropathy and mitotic neuropathy, prion-based neurodegeneration, including Creutzfeldt-Jakob disease (CJD), variant CJD, new variant CJD, bovine spongiform encephalopathy (BSE), GSS, FFI, kuru and Alper's syndrome, Joseph's disease, acute disseminated encephalomyelitis, arachnoiditis, vascular lesions of the central nervous system, loss of extremity neuronal function, Charcot-Marie-Tooth disease, susceptibility to heart failure, asthma, and macular degeneration.
The invention therefore includes methods of treating or preventing the above diseases and conditions in a human or non-human animal suffering therefrom or susceptible thereto, which comprises administering to the said human or non-human animal an effective amount of an active agent as defined herein, as well as uses of the active agents in the preparation of compositions for said treatment or prevention.
In the cases of Parkinson's disease, postencephalitic Parkinsonism, postural hypotension, autistic syndrome, chronic fatigue syndrome, myasthenia gravis, and Lambert Eaton disease, as well as any other conditions within the range of disease states to which the present invention relates which are disclosed in, or obvious from, the treatments made available or disclosed in the prior art acknowledged above, the present invention may be subject to the proviso that either symptoms of cognitive dysfunction are absent, or any symptoms of cognitive dysfunction presented by a subject to be treated are secondary or ancillary to the symptoms of non-cognitive neurodegeneration, non-cognitive neuromuscular degeneration, motor-sensory neurodegeneration or receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment.
Preparation of Compounds for Use in the Invention
Smilagenin, epismilagenin and sarsasapogenin are commercially available materials. Suppliers include, for example, Sigma Aldrich, Research Plus Inc. and Steraloids Inc. Preparative methods for these materials are also to be found in the literature (e.g. a preparation of episarsasapogenin is given in JACS p.5225 (1959)). Episarsasapogenin pan be prepared by reduction of sarsasapogenone using a metal hydride reducing agent. Sarsasapogenone can be prepared using the method of Lajis et al, Steroids, 1993, 58, 387-- 389.
Also, as starting materials, unsubstituted saponins and sapogenins may occur naturally in a range of plant species, notably plants of the genus Smilax, Asparagus, Anemarrhena, Yucca or Agave. Where smilagenin or sarsasapogenin is used in accordance with this invention, it may be in the form of a plant extract, or dry powdered plant material, derived from a plant . of the genus Smilax, Asparagus, Anemarrhena, Yucca or Agave.
Methods for preparing the active agents are well known to one of ordinary skill in this art. Examples are shown, for example, in WO-A-02/079221 (Examples 5 to 16 therein, which describe the preparation of sarsasapogenin cathylate, episarsasapogenin cathylate, episarsasapogenin succinate, epismilagenin cathylate, episarsasapogenin glycinate hydrochloride, sarsasapogenin glycinate hydrochloride, epismilagenin glycinate hydrochloride, epismilagenin L-alaninate hydrochloride, epismilagenin L-valinate hydrochloride, epismilagenin L-isoleucinate hydrochloride, epismilagenin L- phenylalaninate hydrochloride and epismilagenin L-methioninate hydrochloride). The compounds of formula la, other than those with R = H, can be prepared using conventional techniques from compounds in which R = H.
The preferred reaction is a nucleophilic substitution reaction, in which a compound having OH at the 3-position is reacted with a compound of formula
L-R,
in which R is selected from alkylcarbonyl; alkoxycarbonyl; alkylcarbamoyl; or arylcarbonyl; or wherein any alkyl group is optionally substituted with aryl, amino, mono- alkyl-amino, di-alkyl-amino, a carboxylic acid residue (-COOH), or any combination thereof; and L is a leaving group, under conditions suitable for nucleophilic substitution.
The compound L-R may, for example, be a carboxylic acid or, if appropriate, an anhydride, or an acyl halide (e.g. an acyl chloride). For example, where R is a cathylate (ethoxycarbonyl) moiety, the compound L-R may suitably be ethyl chloroformate.
The reaction is suitably performed in a base such as pyridine, optionally in the presence of an acid such as hydrochloric acid.
The reaction details for nucleophilic substitution reactions are well known. See, for example, RC Larock, in Comprehensive Organic Transformations, VCH publishers, 1989.
Dihydrosarsasapogenin may be made using the method described in Marker and Rohrmann (1939), Sterols LIJI; The structure of the side chain of sarsasapogenin, J. Am. Chem. Soc. 61, pp 846-851. 16,22-Epoxycoprostan-3β-ol may be made using the method described in Scheer et al, (1955), The C-25 isomerism of smilagenin and sarsasapogenin: J. Am. Chem. Soc. 77, pp 641-646.
In the reactions described herein it may be necessary to protect reactive functional groups, for example hydroxy, carboxy or amino groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice. For examples, see TW Green and PGM Wuts, in "Protective Groups in Organic Chemistry", John Wiley & Sons, 1991; JFW McOmie in "Protective Groups in Organic Chemistry", Plenum Press, 1973. For protecting amino substituents in compounds of formula L-R wherein R is amino-substituted, it is preferred to use an alkoxycarbonyl protecting group, whereby the amino function is present as an alkoxycarbonylamino group (preferably t.-butoxycarbonylamino) during the synthetic steps, until deprotection in acid conditions in a dry solvent.
The compound thus prepared may be recovered from the reaction mixture by conventional means. For example, the compound may be recovered by distilling off the solvent from the reaction mixture or, if necessary after distilling off the solvent from the reaction mixture, pouring the residue into water, followed by extraction with a water-miscible solvent and distilling off the solvent from the extract. Additionally, the product can, if desired, be further purified by various well known techniques, such as recrystallisation, reprecipitation, or the various chromatography techniques, notably column chromatography or preparative thin layer chromatography.
Discussion of the Basis for the Activity
The therapeutic uses underlying the present invention arise from a number of novel observations which are documented in detail in the Examples below. To understand the rationale of the invention, it is useful to summarise the observations and to explain how they predict the therapeutic activities claimed in this invention across the range of active agents defined above.
Smilagenin, epismilagenin, sarsaspogenin and episarsasapogenin restore the loss of muscarinic acetylcholine receptors and adrenoceptors in cells expressing such receptors in vitro. These results demonstrate that these compounds restore towards normal cellular receptor loss (Example 1). .
Sarsasapogenin, episarsasapogenin cathylate, episarsasapogenin, epismilagenin and smilagenin prevent chemically induced neurodegeneration in rat cortical neurones in vitro These results demonstrate that these compounds are neuroprotective and prevent neurodegeneration and neuroimpairment in vitro (Example 2).
Sarsasapogenin, smilagenin, 16,22-epoxycoprostan-30-ol, smilagenone, smilagenin glycinate hydrochloride and coprosterol reverse the chemically induced neurodegeneration in rat cortical neurones in vitro. These results demonstrate that the compounds reverse sensory neurodegeneration and neuroimpairment in vitro (Example 3).
Smilagenin reverses the chemical induced apoptosis in neurones, .demonstrating that this compound is anti-apoptotic and neuroprotective in vitro (Example 4).
Smilagenin and sarsasapogenin increase the neurite outgrowth (neurite number and neurite branching) in rat cortical neurones in vitro, demonstrating their neurotrophic effects in vitro (Example 5).
Smilagenin and sarsasapogenin prevent and reverse neurotoxin-induced neurodegeneration (neurotoxin 1 -methyl -4-phenylpyridinium (MPP4) in mesenchephalic dopaminergic neurones in vitro. These results demonstrate that these compounds prevent and reverse neurodegeneration and neuroimpairment in vitro (Examples 6 and 7).
Sarsasapogenin and smilagenin reverse the chemically induced neurodegeneration in rat spinal motor neurones in vitro. These results demonstrate that the compounds reverse neurodegeneration and neuroimpairment of motor neurones in vitro (Example 8).
Sarsaspogenin, episarsasapogenin cathylate and smilagenin reduce the number of wrong responses in a cognitive ability test in vivo in aged rats, which correlates with an increase in muscarinic acetylcholine receptor density in the brains of aged rats following treatment with the compounds tested. These results demonstrate that the compounds reverse neuroimpairment in vivo (Example 9).
Smilagenin and sarsasapogenin reverse the decline of muscarinic acetylcholine and dopamine receptors and the decline in brain derived neurotrophic factor (BDNF) in aged animals. These results demonstrate that the compounds reverse motor-sensory neurodegeneration and neuroimpairment and are neurotrophic in vivo (Example 9).
Episarsasapogenin cathylate, sarsasapogenin cathylate, episarsasapogenin and epismilagenin, reduce the number of wrong responses in a cognitive ability test in vivo in young rats exposed to neurotoxic agents (ibotenic acid and amyloid D, and increase in muscarinic acetylcholine receptor density in the brains. These results demonstrate that the compounds reverse neuroimpairment in vivo (Example 10). Smilagenin and sarsasapogenin improve survival and motor-sensory neurodegeneration and neuroimpairment in a mouse model of amyotrophic lateral sclerosis (ALS) and Charcot- Marie-Tooth disease (Example 11).
In summary, the compounds have been found to slow or reverse certain aspects of neuronal degeneration. These include the reversal of adverse changes in the cell body, atrophy of neuronal extensions (neurites), reduction in release of neurotrophic factors such as neurotrophins (e.g. BDNF, NGF, NT-3, NT4/5), TGF-β super-family neurotrophic factors (e.g. GDNF) and neurokines (e.g. CNTF, LIF), and neuronal toxicity or death (apoptosis). The compounds are strongly neuroprotective, stimulative of neurite outgrowth, and preventive of neurotoxicity. The compounds have also been found to slow or reverse decreases in cholinergic and dopaminergic function, for example, decreases in muscarinic acetylcholine and dopamine receptor density. Furthermore, we have found that the neuroprotective and the reversal of receptor loss effects are actively regulated effects, in which past deterioration is reversed towards the normal or young state with protection against continued deterioration. Still further, we have found that the reversal of apoptotic effect of the compounds appears to be regulated in the non-neoplastic domain of cell life, and does not appear likely to trigger neoplasia.
The above data, taken together, indicate activity against the disease states already listed in this application. Furthermore, the above data indicates a likely absence of severe or life- threatening side effects such as cancer. The active agents are typically non-oestrogenic.
The prior art acknowledged above shows the sound basis of prediction for extension of the above observations and sound predictions of therapeutic activity to the related chemical structures and derivatives embraced by the term "active agents" in the present invention.
It is well known in the art and in pharmacology that sugar, ester and other groupings at suitable locations on steroid molecules, particularly at the 3- and/or 26-positions, can be easily cleaved off by in vivo hydrolysis, and the same effects would be expected to be observed at other carbon atoms of the molecules. Further, it is well known in the art and in pharmacology that salts, free acids and free bases within the terms of the expression "active agents" as used herein are readily convertible in vivo between each other according to the pH of the body fluid in which the active agent is present. Further, it is well known that side group substituents, in a wide range of forms, can be present in a complex carbon skeleton without substantially adversely affecting the pharmacological activity of the structure, particularly when the side groups are small in comparison with the overall size of the molecule.
For all these reasons, the claims of beneficial pharmacological activity made in the present application are seen to be reasonable and based on sound and credible prediction from the test data assembled and presented herein.
Without wishing to be bound by theory, it is believed that one physiological effect of the active agents is the ability to increase the synthesis, or release of, or to reduce the rate of degradation of, neurotrophic factors such as brain derived neurotrophic factor and/or nerve growth factor or their receptors. These effects on growth factors might be due to an effect of the compound on a cytosolic or nuclear receptor, or the binding of a compound to a promoter region with a consequent effect directly on the rate of production of mRNA for the growth factor, or as a consequence of increasing the production of another material factor.
In addition, the compounds appear to regulate receptors. For example, some of these compounds have been found to prevent or reverse the loss of muscarinic acetylcholine or dopamine receptors in the brain. It is believed that the compounds function by rectifying a deficiency in receptor number or function or turnover.
Brief Description of the Drawings
In order to illustrate the invention further by way of non-limiting example, reference will now be made to the accompanying drawings and to the Examples which follow.
In the drawings:
Figure 1 shows the effect of epismilagenin acetate on m3 and β2 adrenoceptor density at day 5 in a CHO-β2/m3 co-transfected cell line;
Figure 2 shows the effects of sarsasapogenin, episarsasapogenin cathylate and smilagenin on glutamate induced neurodegeneration in rat primary cortical neurons;
Figure 3 shows the effects of sarsasapogenin, episarsasapogenin cathylate and smilagenin on the learning ability and memory of aged rats; Figure 4 shows the effects of sarsasapogenin, episarsasapogenin cathylate and smilagenin on muscarinic receptor number;
Figure 5 shows the survival profile of SOD-1 mice following oral administration of smilagenin; and
Figure 6 shows the survival profile of pmn mice following oral administration of sarsasapogenin.
Detailed Description of the Drawings and Examples
EXAMPLE 1
Restoration of receptor loss in vitro
The effects of epismilagenin cathylate, sarsasapogenin cathylate, episarsasapogenin cathylate, episarsasapogenin succinate, epismilagenin acetate and sarsasapogenin on the expression of muscarinic acetylcholine receptor (m) in CHO cells or β2 and m3 receptors in CHO cells transfected with either a vector for the m receptor or co-transfected with the vector for β2 and m3 receptors were investigated.
The results are illustrated in Table 1 below and in Figure 1 of the drawings. Over the culturing period of CHO cells transfected with a vector for the m receptor, treatment with epismilagenin cathylate, sarsasapogenin cathylate, episarsasapogenin cathylate, episarsasapogenin succinate and sarsasapogenin each prevents the decrease in m receptor number. Over the culturing period of the CHO cells co-transfected with the vector for β2 and m3 receptors, the density of the m3 receptor did not alter; whereas the density of the β2 adrenoceptors decreased. Incubation with epismilagenin acetate (Figure 1) did not significantly alter the density of m3 receptors; but significantly prevented the decrease in β2 adrenoceptors .
Table 1 - Effect of epismilagenin cathylate, sarsasapogenin cathylate, episarsasapogenin cathylate, episarsasapogenin succinate and sarsasapogenin on restoration of m acetylcholine receptor density.
Figure imgf000029_0001
Thus, the experiments indicate that each of epismilagenin cathylate, sarsasapogenin cathylate, episarsasapogenin cathylate, episarsasapogenin succinate, epismilagenin acetate and sarsasapogenin were able to prevent decline in receptor number with time and also tend to restore receptor number to normal levels when given to cells in which the receptor level is depressed.
EXAMPLE 2
Neuroprotective effect of sarsasapogenin, episarsasapogenin cathylate, episarsasapogenin, epismilagenin and smilagenin in neurones
The objective of this study was to examine the effects of sarsasapogenin, episarsasapogenin cathylate, episarsasapogenin, epismilagenin and smilagenin on the survival of rat primary cortical neurones exposed to glutamate, which is known to induce neurodegeneration.
Rat cortical neurones were cultured for 10 days; at day 10 the medium was changed to a serum-free defined medium. On day 12, 24 hours before glutamate exposure, cultures were washed a d medium was replaced with fresh medium containing positive .control (β- oestradiol)j test compounds (sarsasapogenin, episarsasapogenin - cathylate, episarsasapogenin, epismilagenin and smilagenin) or vehicle control (DMSO, 0.25%) or diosgenin as negative control.
On day 13, cultures were exposed to glutamate. After the incubation period, the cultures were washed with and placed in fresh medium, supplemented with relevant compounds or vehicle to evaluate their protective effects, 24 h after glutamate exposure. Neuronal cell survival was evaluated by measuring lactate dehydrogenase (LDH) activity released in the media 24 h after test compound treatment or glutamate + test compound exposure, using the CytoTox 96 non-radioactive kit and quantitated by measuring wavelength absorbance at 450 nm.
Following exposure of rat primary cortical cultures with glutamate, there was a significant degeneration of cortical neurones, 24 h post-treatment, demonstrated by an increase in lactate dehydrogenase release into the culture medium.
In primary cortical cultures pre-treated with the compounds for 24 h, there was also a significant reduction in the glutamate-induced neurodegeneration (Figure 2; Table 2).
Table 2 - Effect of sarsasapogenin, episarsasapogenin cathylate, episarsasapogenin, epismilagenin and smilagenin on prevention of glutamate-induced neurodegeneration
Figure imgf000030_0001
Sarsasapogenin, episarsasapogenin cathylate, episarsasapogenin, epismilagenin and smilagenin all displayed significant neuroprotective effects against glutamate-induced neurodegeneration in rat primary cortical neurones in vitro. EXAMPLE 3
Reversal of neurodegeration by sarsasapogenin, smilagenin, 16,22-epoxycoprostan-3β- ol, smilagenone, smilagenin glycinate hydrochloride and coprosterol in neurones
As above mentioned, exposure of rat primary cortical cultures to glutamate (100 μM; 10 min) caused an increase in lactate dehydrogenase (LDH) activity measured after 24 h, indicating a significant neurodegeneration. Treatment with 17β-oestradiol after glutamate exposure produced a significant decrease in the LDH activity compared to neurones exposed to glutamate, suggesting a significant neuroprotective effect. Similarly, treatment with sarsasapogenin, smilagenin, 16,22-epoxycoprostan-3β-ol, smilagenone, smilagenin glycinate hydrochloride and coprosterol produced a significant decrease in the LDH activity compared to neurones exposed to glutamate, suggesting a significant neuroprotective effect (Table 3).
Table 3 - Effect of different compounds on cortical neurones previously exposed to glutamate
Figure imgf000031_0001
Figure imgf000032_0001
In conclusion, in rat primary cortical neurones sarsasapogenin, smilagenin, 16,22- epoxycoprostan-3β-ol, smilagenone, smilagenin glycinate hydrochloride and coprosterol reversed the neurodegeneration induced by glutamate, suggesting a therapeutic potential in neurodegenerative disorders.
EXAMPLE 4
Anti-apoptotic effect of smilagenin in neurones
The objective of this study was to examine the anti-apoptotic effect of smilagenin on the caspase-3 activity, a marker of apoptosis, in rat primary cortical cultures exposed to glutamate
Primary cultures of cortical neurones
Rat cortical neurones were cultured for 6 days. At day 6, glutamate (100 microM, 10 min) was added. Then the cultures were washed and medium was replaced with fresh medium containing smilagenin or vehicle control (DMSO, 0.25%) for 6 h. After 6 h treatment, apoptosis was evaluated by measuring caspase 3 activity. Caspase 3 activity was detected by the cleavage of p-nitroaniline from a colorimetric caspase-3 substrate, acetyl-Asp-Glu- Val-Asp p-nitroanilide. p-Nitroalanine has a high absorbance at 405 nM. Relative caspase- 3 activity was measured as optical density. In addition the relative activity of caspase-3 was standardised to protein concentration of the sample, which was also measured as an optical density (Du et al, J Neurochem., 69, 1382-1388, 1997; Sawada et al, Faseb J., 14, 1202-1214, 2000).
Smilagenin reverses the increase in glutamate induced caspase 3 activity in rat primary cortical neurones, demonstrating the anti-apoptotic effect of smilagenin (Table 4) Table 4 - Effect of smilagenin on glutamate induced caspase 3 activity in cortical neurones
Figure imgf000033_0001
EXAMPLE 5
Neurodegenerative disorders are characterised by a progressive loss of neurones and degradation of neuronal processes (neurites). Agents that induce neurite outgrowth may promote the formation of new connections between neurones and ameliorate the symptoms of neurodegenerative conditions (Katzman et al, Faseb J., 5, 278-286, 1991).
Exposure to 17β-oestradiol (0.3, 3, 30 pM) significantly increased the length of existing neurites in rat primary cortical neurones (Table 5). Exposure to 17β-oestradiol (3, 30 pM) significantly increased the percentage of neurones displaying neurites in rat primary cortical neurones (Table 6). Exposure to smilagenin and sarsasapogenin (0.3, 3, 30 pM) significantly increased the length of existing neurites and the percentage of neurones displaying neurites in rat primary cortical neurones (Tables 5 and 6).
In conclusion, smilagenin and sarsasapogenin have neurotrophic effects in vitro.
Table 5 .- Effect of 17β-oestradioI, smilagenin and sarsasapogenin on neurite length measured, using optical micrometry
Figure imgf000033_0002
Figure imgf000034_0001
Table 6 - Effect of 17β-oestradiol, smilagenin and sarsasapogenin on the number of neurones displaying neurites
Figure imgf000034_0002
EXAMPLE 6
Smilagenin and sarsasapogenin prevent the neurodegeneration caused by exposure to the neurotoxin, l-methyl-4-phenylpyridinium (MPP"j) in rat mesencephalic dopaminergic neurones; a model of Parkinson's disease in vitro.
Damage caused by the neurotoxin, MPP+, a metabolite of l-methyl-4-phenyl-l,2,3,6- tetrahydropyridine (MPTP) mimics the degeneration of nigrostriatal dopaminergic neurones observed in neurodegenerative disorders such as Parkinson's disease (Mytinlineou et al, Science, 225, 529-531, 1984). The most prominent biochemical changes induced by this toxin include decreased levels of dopamine and its metabolites in the substantia nigra pars compacta and in the caudate nucleus (Burns et al, Proc Natl Acad Sci U.S.A., 80, 4546- 4550, 1983) and a reduction in dopamine uptake in nigrostriatal synaptosomal preparations (Heikkila et al, J Neurochem., 44, 310-313, 1985).
Pre-treatment of dopaminergic neurones with smilagenin and sarsasapogenin significantly reduced the neuronal death following exposure of the dopaminergic specific neurotoxin MPP+ (2 μM) when compared with MPP+ alone. Glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF), molecules that are involved in neuronal growth, were used as positive controls. Pre-treatment with smilagenin and sarsasapogenin produced a significant increase in the neuronal survival compared to neurones exposed to MPP+ alone, suggesting a significant neuroprotective effect (Table 7).
Table 7 - Effect of pre-treatment with BDNF and GDNF, smilagenin and sarsasapogenin on dopaminergic neurones after MPP+ (2 μM) exposure
Figure imgf000035_0001
In this model of Parkinson's disease in vitro, pre-treatment with smilagenin and sarsasapogenin significantly prevented the neuronal degeneration following exposure to the dopaminergic specific neurotoxin, MPP+, demonstrating a neuroprotective effect.
EXAMPLE 7
Smilagenin and sarsasapogenin also reverse the neurodegeneration caused by exposure to the neurotoxin l-methyl-4-phenylpyridinium (MPP+) in rat mesencephalic dopaminergic neurones; a model of Parkinson's disease in vitro.
Treatment of dopaminergic neurones with smilagenin and sarsasapogenin significantly reduced the neuronal death following exposure to the dopaminergic specific neurotoxin MPP+ (2 μM) when compared with MPP+ alone. Glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF), molecules that are involved in neuronal growth, and 17β-oestradiol were used as positive controls. Treatment with smilagenin and sarsasapogenin produced a significant increase in the neuronal survival compared to neurones exposed to MPP+ alone (Table 8). Table 8- Effect of treatment with BDNF and GDNF, smilagenin, sarsasapogenin and 17β-oestradiol on dopaminergic neurones after MPP+ (2 uM) exposure
Figure imgf000036_0001
Exposure to MPP+ caused not only a significant decrease in dopaminergic number but also in the percentage of neurites. This study shows that smilagenin and sarsasapogenin significantly increased the number of neurites of the neurones in vitro Table 9. These results demonstrate that the compounds reverse motor neurodegeration.
Table 9 - Effect of smilagenin and sarsasapogenin on percentage of neurites in dopaminergic neurones after MPP+ (2 uM) exposure
Figure imgf000036_0002
EXAMPLE 8
Neuroprotective effect of sarsasapogenin and smilagenin in spinal motor neurones
The objective of this study was to examine the effects of sarsasapogenin and smilagenin on the survival of rat primary spinal motor neurones exposed to glutamate, which is known to induce neurodegeneration in this model of motor neurodegeneration. 17β-oestradiol and BDNF were used as positive controls.
Primary cultures of spinal motor neurones
Rat motor neurones were prepared according to the method described by (Martinou et al, Neuron, 8, 737-744, 1992). On day 10, medium was removed and the cultures were exposed to glutamate (4 microM) for 10 min at 37°C in defined medium. After the glutamate exposure, cultures were washed with Dulbecco modified Eagle medium at 37°C then placed in fresh culture medium containing test compounds. After 48 h, the extent of spinal motor neurone degeneration was determined by measuring the amount of lactate dehydrogenase (LDH) released into the culture medium as above.
Results
Following exposure of glutamate, there was a significant degeneration of rat primary spinal motor neurones, 48 h post-treatment, demonstrated by an increase in lactate dehydrogenase release into the culture medium.
In primary rat primary spinal motor neurones treated with sarsasapogenin or smilagenin for 48 h, there was a significant reduction in the glutamate-induced neurodegeneration (Table 10).
Table 10 - Effect of sarsasapogenin and smilagenin on glutamate-induced neurodegeneration in spinal motor neurones
Figure imgf000037_0001
Sarsasapogenin and smilagenin reversed the glutamate-induced neurodegeneration in rat spinal motor neurones in this in vitro model of motor neurodegeneration EXAMPLE 9
In the second half of life (in humans from the age of 40 onwards) the density of neurones in the brain decreases (Selkoe, D J, Sci. Am. 267, 134-142, 1992). The alterations in cortical function may be due to a reduction in the number of neurones, their interconnections, a decrease in neurotrophins such as brain derived neurotrophic factor (BDNF; Bothwell, M, Functional interactions of neurotrophins and neurotrophin receptors, Annu. Rev. Neurosci., 18, 223-253, 1995), a decrease in acetylcholine receptor density (muscarinic and nicotinic) and/or a decrease in their coupling function in cortical areas (Rinne et al, Brain Res., 336, 19-25, 1985; Selkoe, D J, Sci. Am. 267, 134-142, 1992). Furthermore during ageing, muscarinic acetylcholine receptor binding is significantly reduced in the hippocampus (Narang, N, Mech. Ageing Dev., 78, 221-239, 1995) and striatum of older rats (Biegon et al, Neurobiol. Aging., 10, 305-310, 1989) and humans (Rinne et al, Brain Res., 336, 19-25, 1985). In addition, in Alzheimer's disease, the decline in cholinergic activity is associated with amyloid β plaque deposition (von der Kammer et al, Biochem. Soc. Symp. 131-140, 2001). Other neurodegenerative disorders, such as Parkinson's disease, show a characteristic decline in dopaminergic activity (Drukarch et al, Expert. Opin. Investig. Drugs, 10, 1855-1868, 2001).
Oral administration of sarsasapogenin, episarsasapogenin cathylate or smilagenin to aged rats (Sprague-Dawley rats 20 month old), for a two or three month period reverses the impairment in learning and memory ability, the decline in muscarinic acetylcholine and dopamine receptors and the decline in the neurotrophin BDNF, alterations that are characteristic of the ageing process.
Aged Sprague-Dawley rats aged were divided into different groups, one control and groups treated for 2-3 months with either sarsasapogenin, episarsasapogenin cathylate or smilagenin (18 mg kg"* day'l, n = 10). A control group (n=14) of untreated young rats was also included in the study. The daily dose of drug was mixed in a minimum amount of food and was administered every morning separately to each rat.
A Y-maze apparatus was used for the learning and memory test. On the floor of each arm of the Y-maze is an array of copper rods to which electric current is applied whenever needed, with adjustable voltage. Each arm is 45 cm long and has a 15 W lamp at the end, which is turned on when needed. After 3 months drug administration, each rat was trained for 7 consecutive days, as follows. For each training session, the rat was put into one arm of the Y-maze, after two minutes rest, an electrical current was applied to the copper rods and the lamp of the clockwise arm was illuminated to indicate the non-stimulation area. If the rat went into that arm, one correct response was recorded, otherwise, one wrong response was recorded. This stimulation-response test was repeated 20 times each day, with a pause of 5 sec between each two consecutive tests. The number of correct responses following the twenty tests on the seventh day was used to express learning ability, (the higher the number the better the learning ability). The rats were then left resting for 30 days and the procedure was repeated once more. The number of correct responses of the 20 tests after the 30 day rest period was used to represent the memory ability
Muscarinic acetylcholine receptor density in the brain was measured. Tissue was prepared as follows: brains were removed quickly after decapitation, frozen in dry ice, and transferred to a freezer. The brains were homogenised and the pellet was finally suspended in buffer.
The dual-site competitive ligand binding assay was used to measure muscarinic acetylcholine receptor density.
The results are shown in Figures 3 and 4 of the drawings. The Y-maze experiments revealed that both the learning ability and memory are impaired in aged rats. Sarsasapogenin, episarsasapogenin cathylate and smilagenin restored the learning and memory ability following administration in aged rats. Muscarinic acetylcholine receptor density was markedly reduced in aged rats. Sarsasapogenin, episarsasapogenin cathylate and smilagenin significantly restored the muscarinic acetylcholine receptor density.
Young rats showed a significantly higher dopamine (D) 1 and 2 receptor density (157.5 ± 33.2; 200.6 ± 50.9 fmol/mg protein, respectively) compared to aged rats (129.2 ± 36.8; 153.8 ± 40.5 fmol/mg protein, Oι and D2, respectively). By contrast, smilagenin and sarsasapogenin treatment in aged rats for 3 months restored the Oι and D2 receptor density (smilagenin 177 ± 10.9; 217 ± 45.7 fmol/mg protein; sarsasapogenin 172.0 ± 44.0; 206.4 ± 60.5 respectively).
Young rats showed significantly higher BDNF levels (1.647 ± 0.277 ng/g tissue) compared to aged rats (1.205 ± 0.219 ng/g tissue). By contrast, smilagenin and sarsasapogenin treatment in aged rats for 3 months, partially restored BDNF levels (1.342 ± 0.07; 1.410 ±
0.232 ng/g tissue, respectively). Thus, the compounds reverse the neuroimpairment, the decline in BDNF levels, and the decline in the muscarinic acetylcholine and dopamine receptor density that occur in aged rats.
EXAMPLE 10
Alzheimer's disease model as a model of neurodegeneration
An in vivo model of Alzheimer's disease was used to model neurodegeneration. In this model the neurotoxic agents (amyloid β and ibotenic acid) are injected into the brain of the rat. This leads to neuronal loss, receptor loss and cognitive impairment. Previous studies showed that local injection of amyloid β in the nucleus vasalis of the rat brain caused cholinergic hypofunction and behavioural impairment up to two months post surgery (Giovannelli et al., 1995: Neuroscience, 66, 781-792.). In addition the co-injection of amyloid β with a small amount of ibotenic acid into the rat hippocampus synergistically produces neuronal loss with infiltration of glial cells not only adjacent but also far from the injected site (Morimoto et al., 1998: Neuroscience, 84, 479-487).
Our studies used the method of Morimoto (Morimoto et al., 1998: Neuroscience, 84, 479- 487) with some modifications (unilateral instead of bilateral injection). Three months old, Sprague Dawley rats, were randomly divided into different groups. Injection of amyloid βt. 40 and ibotenic acid (both from Sigma) was accomplished by means of a stereotaxic instrument (Stoelting Co.) and the coordinates were AP= -0.5mm (right to medial line), L= -2.8mm (backward from bregma), H= -7.0mm (ventral to dura). The dose for each rat was amyloid βM0 (4 μg) and ibotenic acid (1 μg) in 1 μl of saline. The injection was completed in 20 min, and the needle was withdrawn 10 min later. Then the skin was sutured.
The 8 groups were:
Operated control injected with normal saline (control) Model (control injected with amyloid β + ibotenic acid) Model + Episarsasapogenin cathylate (18 mg/kg/day)*
Model + Sarsasapogenin cathylate (18 mg/kg/day)* Model + Episarsasapogenin ethylsuccinate (18 mg/kg/day) (comparison) Model + Episarsasapogenin (18 mg/kg/day)*
Model + Epismilagenin (18 mg/kg/day)*
Model + Diosgenin (i.e. negative control, 18 mg/kg/day)
* Compounds in accordance with the present invention
Episarsasapogenin cathylate, sarsasapogenin cathylate, episarsasapogenin ethylsuccinate (comparison compound), episarsasapogenin, epismilagenin and diosgenin (all at a dosage of 18 mg/kg/day) were administered to animals as stable suspensions in CMC-Na (0.5%) once daily through a gastric tube. The control and the model group were given the same volume of CMC-Na (0.5%) once daily. The drugs and vehicles were given for a period of two months, starting 20 days before operation.
Muscarinic acetylcholine receptor density was assessed. The brain samples were homogenised, centrifuged, and the pellet of centrifugation at 27000xg was re-homogenised and used for measurement. The concentration of 3H-QNB was chosen at the saturation range. After incubation and separation, the bound portion was measured by liquid scintillation counter.
Step-Through Test: learning and memory. The effect of test compounds on learning and memory was assessed using the step-through test. A 60 x 15 x 15 cm box, divided into 2 equally sized rooms, one dark room with copper rod base, which was electrically charged (40 V ac) when in use, while the other was a light room but not electrically charged. Between the two rooms there is an opening (hole) for the rat to go through. The experiment is carried out for each rat on two consecutive days. The first day is for training; when the rat is adapted in the box for the first 3 min, then put in the light room, with its back toward the hole, and the copper rods of the dark room are charged for 5 min. The second day is for testing, when the number of crosses in 5 min are recorded. Improvements in memory are signalled by a reduction in the number of crosses.
The muscarinic acetylcholine receptor density in the neurodegeneration model brains was significantly lower than control. Episarsasapogenin cathylate, sarsasapogenin cathylate, episarsasapogenin and epismilagenin produced a significant elevation in brain muscarinic acetylcholine receptor density, whereas diosgenin and episarsasapogenin ethylsuccinate did not significantly change the muscarinic acetylcholine receptor density. Thus the experiments indicate that the compounds of this invention act to normalise receptor number, i.e. they tend to restore receptor number to normal levels when given to animals in which the receptor level is depressed.
The number of wrong responses (error number) in 5 min was significantly higher in the neurodegeneration model group than the confrol group, indicating an impairment of memory (see Table 11). Epismilagenin, episarsasapogenin cathylate, episarsasapogenin and sarsasapogenin cathylate each significantly decreased the number of wrong responses, whereas diosgenin and episarsasapogenin ethylsuccinate were both ineffective in decreasing the number of wrong responses.
Table 11
Figure imgf000042_0001
Statistical analysis using unpaired Student t test. * denotes p<0.05
EXAMPLE 11
Amyotrophic lateral sclerosis (ALS) is a progressive fatal neurodegenerative disorder that causes motor neurone degeneration, skeletal atrophy, paralysis and death. The cause of this disease is heterogeneous: mutations in the Cu Zn superoxide dismutase (SOD-1) gene is responsible for some forms of human ALS. Animal models of this disease include the SOD-1 transgenic mice over-expressing SOD-1 gene and the progressive motor neuropathy (pmn, a model of Charcot-Marie-Tooth) mice. Smilagenin and sarsasapogenin increase the lifespan and improve the behavioural deficits of the superoxide dismutase (SOD) mouse (Figure 5) and pmn mouse (Figure 6), two models of amyofrophic lateral sclerosis (ALS) and Charcot-Marie-Tooth disease.
The foregoing broadly describes the present invention without limitation. Variations and modifications as will be readily apparent to those of ordinary skill in this art are intended to be within the scope of this application and any subsequent patent(s).

Claims

1. Use of one or more active agent selected from:
A. compounds of Formula I:
Figure imgf000044_0001
(I)
wherein in the general formula (I):
- R) R2, R3, R4, R5, Re, R7, R8, Rio, R13, Ris, R19, R20, R21, R22, R23, R24, R26, R27, 285 R29, R30, R , R32 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me- SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl or absent or OR where R = alkyl or acyl group;
- R9, R11, R12, Ri4, Ris, Ri6, Rπ, R25. R33 can be either a H, OH, halo atom, (Me-S-), (Me-SO- ), (Me-S02-), N3-, NH2-, MeS02NH-, alkyl or absent or OR where R = alkyl or acyl group; lllli represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group;
B. compounds of Formula Et:
Figure imgf000045_0001
(H) wherein in the general formula (H):
- Ri, 2, R3, R , R5, e, R7, R8> Rio, R13, is, R19, R20, R21, R22, R23, R24, R26, R27, R28, R29, R30, R31, R32, R34 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent;
- R9, R11, Rπ, R14, R15, Riβ, Rπ, R25, R33, R35 can be either a H, OH, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond. wherein in addition to the above
- either R33 or R14 = alkyl group;
C. compounds of Formula JH:
Figure imgf000045_0002
(HI) wherein in the general formula (HI):
- Ri, R2, R3, R , R5, Re, R7, R8, Rio, Rπ, ι4, Ris, Ri9, R20, R21, R22, R23, R24, R26, R27, R28, R29, R30, R31, R32, R33, R34, R35. R36. R37 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-S02-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent;
-R9, Rπ, R12, Ris, Riβ, Rπ, R25 can be either H, OH, halo atom, (Me-S-), (Me-SO-), (Me- SO2), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation;
D. sapogenin derivatives bearing at least one X radical substituent, wherein X is chosen from the group consisting of :
- halo atom,
- (Me-S-), (Me-SO-), (Me-S02-),
- N3-, NH2-, MeS02NH-, and
- alkyl; and
E. derivative forms of any of the above compounds, in which the carbon atom at the 3- position or, in the case of Formulae II and HI, the 3-position carbon atom, the 26-position or each of the carbon atoms at the 3- and 26-positions, carries an O-sugar moiety wherein the sugar group is a mono-, di- or tri-saccharide;
all their stereoisomers and racemic mixtures, all their pharmaceutically acceptable pro- drugs and salts, and all mixtures and combinations thereof
in the treatment or prevention of, or in the preparation of compositions for the treatment or prevention of, (i) non-cognitive neurodegeneration, (ii) non-cognitive neuromuscular degeneration, (iii) motor-sensory neurodegeneration, or (iv) receptor dysfunction or loss in the absence of cognitive, neural and neuromuscular impairment, in human and non-human animals suffering therefrom or susceptible thereto.
2. A use according to claim 1, wherein the active agent, or at least one of the active agents, is selected from: a. Compounds of the above general formula I, wherein:
- Ri, 2, R3, 4, 5> R«, R7, Rs, Rio, R13, is, R19, R20, R21, R22, R23, R2 , R26, R27, 28, R29, R30, R31, R32 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me- SO-), (Me-S02-), N3-, NH2-, MeSO2NH-, alkyl or absent or OR where R = alkyl or acyl group;
- R9, Rπ, Ri2, Ri4, Ris, RIO, R17, R25, R33 can be either a H, OH, halo atom, (Me-S-), (Me-SO- ), (Me-SO2-), N3-, NH2-, MeS02NH-, alkyl or absent or OR where R = alkyl or acyl group; represents an optional double bond, wherein in addition to the above
- either R33 or R = alkyl group, and the stereochemistry of R25 is in the β orientation;
b. Compounds of the above general formula I, wherein: - Ri, R2, R3, R4, R5, Re, R7, Rs, Rio, Rπ, Ris, R19, R20, R21, R22, R23, R24, R26, R27, R28, R29, R30, R31, R32 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me- SO-), (Me-S02), N3-, NH2-, MeS02NH-, alkyl or absent or OR where R = alkyl or acyl group;
- R9, Rl2, Rl5, Rl6, Rl7 = H, - Rn, R14, R25, R33 can be either a H, OH, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeS02NH-, alkyl or absent or OR where R = alkyl or acyl group; represents an optional double bond wherein in addition to the above
- either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation;
c. Compounds of the above general formula I, wherein:
-Rι=
Figure imgf000047_0001
Ri6 = π = Ris = R1 = R20 =
Figure imgf000047_0002
- either R33 or R,4 = CH3 represents a single bond,
- the methyl group at C25 may be either in the R or S configuration
- the stereochemistry of R25 is in the β orientation and wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =O, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of :
- halo atom,
- (Me-S-), (Me-SO-), (Me-SO2-), and - N3-, NH2-, MeSO2NH- - alkyl;
d. Compounds of the above general formula I, wherein:
Figure imgf000048_0001
R20 = 24= R25"
Figure imgf000048_0003
j2 = H,
Figure imgf000048_0002
represents a single bond,
- the stereochemistry of R25 is in the β orientation and wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of :
- halo atom,
- (Me-S-), (Me-SO-), (Me-S02-), and - N3-, NH2-, MeS02NH- - alkyl;
e. Compounds of the above general formula π, wherein
" Rl, R2, R3, t, R5, Rδ, R7, R8, RlO, Rl3, Rl8, Rl9, R20, R2I, R22, 23, R24, R26, 27, R28, R29,
R30, R31, R32, R34 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-SO2), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent;
- R9, Rπ, Rπ, R14, R15, RIO, Rπ, R25, R33, R35 can be either a H, OH, halo atom, (Me-S-), (Me-SO-), (Me-S02-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent; -^ represents an optional double bond, wherein in addition to the above
- either R33 or R]4 = alkyl group, and the stereochemistry of R25 is in the β orientation;
f. Compounds of the above general formula II or carbohydrate derivatives thereof, wherein: - R1; R2, R3, R-t, R5, Re, R7, Rs, Rio, R13, Ris, R19, R20, R21, R22, R23, R24, 26, R27, R28, R29, R30, R31, R32 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me- SO-), (Me-SO2), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent; - R9, R12, R15, Riβ, R17 = H,
- R34= either H, OH, =0, and OR where R = alkyl, acyl or carbohydrate and
- Rπ, Ri4, R25, R33, R35 can be either H, OH, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or Ri4 = alkyl group, and the stereochemistry of R25 is in the β orientation;
g. Compounds of the above general formula II or carbohydrate derivatives thereof, wherein:
-Rι=
Figure imgf000049_0001
Ris = R19 = R20 =
R∑l= R22=
Figure imgf000049_0002
R31= R32= R33 = H,
-R] = CH3,
- R3 = -OH or -OR where R = alkyl, acyl or carbohydrate and R35 = H or is absent represents an optional double bond, and
- the methyl group at C25 may be either in the R or S configuration and and the stereochemistry of R2s is in the β orientation wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =O, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of :
- halo atom,
- (Me-S-), (Me-SO-), (Me-SO2-), and - N3-, NH2-, MeSO2NH-
- alkyl;
h. Compounds of the above general formula II or carbohydrate derivatives thereof, wherein:
Figure imgf000049_0003
Figure imgf000050_0001
- R34= -OH or -OR where R = alkyl, acyl or carbohydrate and R35 = H or is absent represents an optional double bond, and the stereochemistry of R25 is in the β orientation and wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of : - halo atom,
- (Me-S-), (Me-SO-), (Me-S02-), and - N3-, NH2-, MeSO2NH-
- alkyl;
i. Compounds of the above general formula HI, wherein:
- Ri, R2, R3, R , Rs, Re, R7, Rs, Rio, R13, R14, Ris, R19, R20, R21, R22, R23, R24, R26, R27, 28, R29, R30, R31, R32, R33, R34, R35, R36, R37 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-S02-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl or acyl group, or absent; -R9, Rπ, R12, R15, Riβ, Rπ, R25 can be either H, OH, halo atom, (Me-S-), (Me-SO-), (Me- SO;,), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above
- either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation;
j . ■ Compounds of the above general formula HI or carbohydrate derivatives thereof, wherein:
- Ri, R2, R3, R , R5, Re, R7, Rs, Rio, R13, R14, Ris, R19, R2o, R21, R22, R23, R24, R26, R27, R28, R29ι R30, R31, R32, R33, R35, R36, R37 are, independently of each other, either H, OH, =0, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent;
Figure imgf000050_0002
- R34 = H, OH, =O, halo atom, (Me-S-), (Me-SO-), (Me-SO2-), N3-, NH2-, MeSO2NH-, alkyl, OR where R = alkyl, acyl or carbohydrate, or absent; - Rn, R2s, can be either H, OH, halo atom, (Me-S-), (Me-SO-), (Me-S02-), N3-, NH2-, MeS02NH-, alkyl, OR where R = alkyl or acyl group, or absent; represents an optional double bond, wherein in addition to the above - either R33 or R14 = alkyl group, and the stereochemistry of R25 is in the β orientation;
k. Compounds of the above general formula III, wherein:
-Rι=
Figure imgf000051_0001
R20 = R32~ R33 = H,
Figure imgf000051_0002
- R34= -OH or -OR where R = alkyl, acyl or carbohydrate and R35 = H or is absent
R37 = H, -OH or =0 R36= H or -OH represents a single bond, and
- the methyl group at C25 may be either in the R or S configuration and the stereochemistry of R25 is in the β orientation wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of :
- halo atom,
- (Me-S-), (Me-SO-), (Me-S02-), and - N3-, NH2-, MeSO2NH-
- alkyl;
1. Compounds of the above general formula IH or carbohydrate derivatives thereof, wherein:
Figure imgf000051_0003
Ris = R19 = R20 =
Figure imgf000051_0004
-Rι4= R33 = CH3,
- -OH or -OR where R = alkyl, acyl or carbohydrate and R35 = H or is absent R37 = H, -OH or =0 R36 = H or -OH represents a single bond, and
- the methyl group at C25 may be either in the R or S configuration and the stereochemistry of R25 is in the β orientation wherein in addition to the above at least one of R3 or R23 is a X radical, the possible remaining substituent being H, OH, =0, and OR where R = alkyl or acyl group or absent, and X is chosen from the group consisting of :
- halo atom,
- (Me-S-), (Me-SO-), (Me-S02-), and - N3-, NH2-, MeSO2NH-
- alkyl;
m. Substituted sapogenins wherein at least one OH-group of the sapogenin is substituted with X, chosen from the group consisting of : - halo atom,
- (Me-S-), (Me-SO-), (Me-S02-),
- N3-, NH2-, MeSO2NH-, and
- alkyl;
n. Sapogenins defined above wherein in the definition of X the halo atom is a fluoro atom;
0. Substituted sapogenins selected from :
(3β-fluoro-5β, 20α,22α,25R-spirostane), (3,3-difluoro-5β, 20α,22α,25R-spirostane), (3α- methylsulphonylamino-5β,20α,22α,25R-spirostane), (3α-azido-5β, 20α,22α,25R-spiro- stane), (3α-amino-5β,20α,22α,25R-spirostane), and their stereoisomers and racemic mixtures, their pharmaceutically acceptable pro-drugs and salts;
p. Substituted sapogenins wherein the parent sapogenin which is then substituted with at least one X radical as defined above is selected from sarsasapogenin, episarsasapogenin, smilagenin, epismilagenin, and anzurogenin-D; Compounds of the general formula la:
Figure imgf000053_0001
(la)
wherein the group R is selected from hydrogen; alkylcarbonyl; alkoxycarbonyl; alkyl- carbamoyl; or arylcarbonyl; or sulpho (H03S); phosphono ((HO)2P(0)-); or a mono-, di- or tri-saccharide; wherein any alkyl group is optionally substituted with aryl, amino, mono- or di-alkyl-amino, a carboxylic acid residue (-COOH), or any combination thereof; and
r. Derivative forms of the above compounds as defined as items a to q, in which the
3-position carbon atom or, in the case of Formulae II and in, the 3-position carbon atom, the 26-position carbon atom or each of the carbon atoms at the 3- and 26-positions, carries an O-sugar moiety wherein the sugar group is a mono-, di- or tri-saccharide, and acylated derivatives thereof.
3. A use according to claim 1 or claim 2, wherein the active agent, or at least one of the active agents, is selected from compounds of the general formula la.
4. A use according to any one of the preceding claims, wherein the active agent, or at least one of the active agents, is selected from: sarsasapogenin sarsasapogenin cathylate sarsasapogenin acetate sarsasapogenin succinate and pharmaceutically acceptable salts thereof sarsasapogenin glycinate and pharmaceutically acceptable salts thereof sarsasapogenin alaninate and pharmaceutically acceptable salts thereof sarsasapogenin valinate and pharmaceutically acceptable salts thereof sarsasapogenin phenylalaninate and pharmaceutically acceptable salts thereof sarsasapogenin isoleucinate and pharmaceutically acceptable salts thereof sarsasapogenin methioninate and pharmaceutically acceptable salts thereof episarsasapogenin episarsasapogenin cathylate episarsasapogenin acetate episarsasapogenin succinate and pharmaceutically acceptable salts thereof episarsasapogenin glycinate and pharmaceutically acceptable salts thereof episarsasapogenin alaninate and pharmaceutically acceptable salts thereof episarsasapogenin valinate and pharmaceutically acceptable salts thereof episarsasapogenin phenylalaninate and pharmaceutically acceptable salts thereof episarsasapogenin isoleucinate and pharmaceutically acceptable salts thereof episarsasapogenin methioninate and pharmaceutically acceptable salts thereof smilagenin smilagenin cathylate smilagenin acetate smilagenin succinate and pharmaceutically acceptable salts thereof smilagenin glycinate and pharmaceutically acceptable salts thereof smilagenin alaninate and pharmaceutically acceptable salts thereof smilagenin valinate and pharmaceutically acceptable salts thereof smilagenin phenylalaninate and pharmaceutically acceptable salts thereof smilagenin isoleucinate and pharmaceutically acceptable salts thereof smilagenin methioninate and pharmaceutically acceptable salts thereof epismilagenin epismilagenin cathylate epismilagenin acetate epismilagenin succinate and pharmaceutically acceptable salts thereof epismilagenin glycinate and pharmaceutically acceptable salts thereof epismilagenin alaninate and pharmaceutically acceptable salts thereof epismilagenin valinate and pharmaceutically acceptable salts thereof epismilagenin phenylalaninate and pharmaceutically acceptable salts thereof epismilagenin isoleucinate and pharmaceutically acceptable salts thereof epismilagenin methioninate and pharmaceutically acceptable salts thereof.
saponin derivatives of sarsasapogenin, episarsasapogenin, smilagenin and epismilagenin in which, in each case, the 3-position carbon atom carries an O-sugar moiety wherein the sugar group is selected from glucose, mannose, fructose, galactose, maltose, cellobiose, sucrose, rhamnose, xylose, arabinose, fucose, quinovose, apiose, lactose, galactose-glucose, glucose-arabinose, fucose-glucose, rhamnose-glucose, glucose-glucose-glucose, glucose- rhamnose, mannose-glucose, glucose-(rhamnose)-glucose, glucose-(rhamnose)-rhamnose, glucose-(glucose)-glucose, galactose-(rhamnose)-galactose and acylated derivatives thereof;
16,22-epoxycoprostan-3β-ol, smilagenone, coprosterol, and pharmaceutically acceptable pro-drugs and salts thereof.
5. A use according to any one of the preceding claims, wherein the active agent is present in a composition selected from pharmaceutical compositions, foodstuffs, food supplements and beverages.
6. A use according to any one of the preceding claims, wherein the active agent is present with one or more additional active agent.
7. A use according to claim 6, wherein the one or more additional active agent is selected from, but not limited, to cholinesterase inhibitors, dopamine agonists, COMT inhibitors, MAO-B inhibitors, anti-cholinergics, acetylcholine agonists, serotonin agonists, AMPA receptor agonists, GABA receptor agonists, NMDA receptor agonists, β- adrenceptor agonists, digoxin, dobutamine, anti-inflammatories, neurotrophic factors, statins, adenosine A2a receptor antagonists, aldose reductase inhibitors, immunomodulators, cannabinoid agonists, interferon β or tricyclic anti-depressants.
8. A use according to any one of the preceding claims, wherein the human or non- human animal is suffering from, or is susceptible to. any of: Parkinson's disease, postencephalitic Parkinsonism, depression, schizophrenia, muscular dystrophy including facioscapulohumeral muscular dystrophy (FSH), Duchenne muscular dystrophy, Becker muscular dystrophy and Bruce's muscular dystrophy, Fuchs' dysfrophy, myotonic dystrophy, corneal dystrophy, reflex sympathetic dystrophy syndrome (RSDSA), neurovascular dystrophy, myasthenia gravis, Lambert Eaton disease, Huntington's disease, motor neurone diseases including amyofrophic lateral sclerosis (ALS), multiple sclerosis, postural hypotension, traumatic neurodegeneration e.g. following stroke or following an accident (for example, traumatic head injury or spinal cord injury), Batten's disease, Cockayne syndrome, Down syndrome, corticobasal ganglionic degeneration, multiple system atrophy, cerebral atrophy, olivopontocerebellar atrophy, dentatorubral atrophy, pallidoluysian atrophy, spinobulbar atrophy, optic neuritis, sclerosing pan-encephalitis (SSPE), attention deficit disorder, post-viral encephalitis, post-poliomyelitis syndrome, Fahr's syndrome, Joubert syndrome, Guillain-Barre syndrome, lissencephaly, Moyamoya disease, neuronal migration disorders, autistic syndrome, polyglutamine disease, Niemann- Pick disease, progressive multifocal leukoencephalopathy, pseudotumor cerebri, Refsum disease, Zellweger syndrome, supranuclear palsy, Friedreich's ataxia, spinocerebellar ataxia type 2, Rhett syndrome, Shy-Drager syndrome, tuberous sclerosis, Pick's disease, chronic fatigue syndrome, neuropathies including hereditary neuropathy, diabetic neuropathy and mitotic neuropathy, prion-based neurodegeneration, including Creutzfeldt- Jakob disease (CJD), variant CJD, new variant CJD, bovine spongiform encephalopathy (BSE), GSS, FFI, kuru and Alper's syndrome, Joseph's disease, acute disseminated encephalomyelitis, arachnoiditis, vascular lesions of the central nervous system, loss of extremity neuronal function, Charcot-Marie-Tooth disease, susceptibility to heart failure, asthma, and macular degeneration.
PCT/GB2003/001380 2002-03-27 2003-03-27 Theraputic methods and uses of sapogenins and their derivatives WO2003082893A2 (en)

Priority Applications (18)

Application Number Priority Date Filing Date Title
BR0308788-3A BR0308788A (en) 2002-03-27 2003-03-27 Therapeutic processes and uses of sapogenins and their derivatives
MXPA04009321A MXPA04009321A (en) 2002-03-27 2003-03-27 Theraputic methods and uses of sapogenins and their derivatives.
EP03722713A EP1490071B1 (en) 2002-03-27 2003-03-27 Therapeutic uses of sapogenins
IL16416103A IL164161A0 (en) 2002-03-27 2003-03-27 Therapeutic methods and uses of sapogenins and their derivatives
JP2003580357A JP4573157B2 (en) 2002-03-27 2003-03-27 Method of treatment and use of sapogenin and its derivatives
KR1020047014567A KR101130212B1 (en) 2002-03-27 2003-03-27 Theraputic methods and uses of SAPOGENIN and their derivatives
CA2477787A CA2477787C (en) 2002-03-27 2003-03-27 Therapeutic methods and uses of sapogenins and their derivatives
NZ535093A NZ535093A (en) 2002-03-27 2003-03-27 Therapeutic methods and uses of sapogenins and their derivatives
AU2003229877A AU2003229877B8 (en) 2002-03-27 2003-03-27 Theraputic methods and uses of sapogenins and their derivatives
CN038071886A CN1642558B (en) 2002-03-27 2003-03-27 Uses of sapogenins and their derivatives
US10/507,000 US20050130948A1 (en) 2002-03-27 2003-03-27 Therapeutic methods and uses of sapogenins and their derivatives
DE60326437T DE60326437D1 (en) 2002-03-27 2003-03-27 THERAPEUTIC USE OF SAPOGENINES
KR1020127008441A KR20120128596A (en) 2002-03-27 2003-03-27 Theraputic methods and uses of sapogenin and their derivatives
NO20044468A NO20044468L (en) 2002-03-27 2004-10-20 Use of sapogenins and hence in the manufacture of drugs
HR20041009A HRP20041009A2 (en) 2002-03-27 2004-10-27 Theraputic methods and uses of sapogenins and their derivatives
HK04110420.5A HK1067546A1 (en) 2002-03-27 2004-12-31 Therapeutic uses of sapogenins
AU2008207565A AU2008207565A1 (en) 2002-03-27 2008-08-27 Therapeutic methods and uses of sapogenins and their derivatives
US13/085,225 US20110190249A1 (en) 2002-03-27 2011-04-12 Therapeutic methods and uses of sapogenins and their derivatives

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
ARP020101170 2002-03-27
ARP020101170A AR033079A1 (en) 2001-03-28 2002-03-27 SAPOGENINE DERIVATIVES, SYNTHESIS AND USE, AND METHODS BASED ON USE
US36817802P 2002-03-28 2002-03-28
US60/368,178 2002-03-28
GBPCT/GB02/01578 2002-03-28
PCT/GB2002/001578 WO2002079221A2 (en) 2001-03-28 2002-03-28 Sapogenin derivatives, their synthesis and use

Publications (3)

Publication Number Publication Date
WO2003082893A2 true WO2003082893A2 (en) 2003-10-09
WO2003082893A8 WO2003082893A8 (en) 2003-12-31
WO2003082893A3 WO2003082893A3 (en) 2004-04-15

Family

ID=30001210

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2003/001380 WO2003082893A2 (en) 2002-03-27 2003-03-27 Theraputic methods and uses of sapogenins and their derivatives

Country Status (10)

Country Link
KR (1) KR20100093621A (en)
CN (1) CN1642558B (en)
AT (1) ATE424211T1 (en)
IL (1) IL164161A0 (en)
NZ (2) NZ547897A (en)
PE (1) PE20040306A1 (en)
PL (1) PL213697B1 (en)
RU (1) RU2332999C2 (en)
TW (1) TWI329016B (en)
WO (1) WO2003082893A2 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2868700A1 (en) * 2004-04-09 2005-10-14 Michel Coisy USE OF DOPAMINE OR ITS BIOLOGICAL PRECURSORS AGAINST ALGONURODYSTRPHY.
WO2007049932A1 (en) 2005-10-28 2007-05-03 Sun-Yeou Kim Extract of the family dioscoreaceae and composition for preventing or treating peripheral neuropathy comprising the same
JP2008519020A (en) * 2004-11-05 2008-06-05 フィトファーム・パブリック・リミテッド・カンパニー Polymorphic form of salsasapogenin
WO2010084356A1 (en) 2009-01-24 2010-07-29 Phytopharm Plc Treatment of neurotrophic factor mediated disorders
EP2402362A2 (en) 2004-04-28 2012-01-04 Phytopharm PLC Crystalline forms of smilagenin
WO2012010896A1 (en) 2010-07-20 2012-01-26 Phytopharm Plc Treatment of l-dopa, dopamine agonist and/or dopamine enhancer induced disorders
WO2013097835A1 (en) * 2011-12-27 2013-07-04 Centro De Investigacion Y Desarrollo De Los Medicamentos (Cidem) Spirosteroidal systems having neuroactive, anti-inflammatory effects
WO2013149580A1 (en) * 2012-04-03 2013-10-10 Chiming Che Timosaponin compounds
CN109988218A (en) * 2017-12-29 2019-07-09 中国科学院上海药物研究所 A kind of sarsasapogenin derivative and its preparation method and application
EP3572085A1 (en) * 2018-05-25 2019-11-27 Neuro-Sys Synergestic combination composition comprising a steroidal saponin, a first polyphenolic compound and optionnaly a second polyphenolic compound
US10696712B2 (en) 2015-07-06 2020-06-30 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US10723758B2 (en) 2014-06-18 2020-07-28 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
EP3564253A4 (en) * 2016-12-30 2020-07-29 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Antidepressant compound and preparation method and application thereof
US10752653B2 (en) 2016-05-06 2020-08-25 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US10759828B2 (en) 2011-09-08 2020-09-01 Sage Therapeutics, Inc. Neuroactive steroids, compositions, and uses thereof
US10765685B2 (en) 2015-07-06 2020-09-08 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US10781231B2 (en) 2016-07-07 2020-09-22 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11104701B2 (en) 2013-03-13 2021-08-31 Sage Therapeutics, Inc. Neuroactive steroids and methods of use thereof
US11111266B2 (en) 2016-10-18 2021-09-07 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11117924B2 (en) 2015-07-06 2021-09-14 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11149054B2 (en) 2016-10-18 2021-10-19 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11149056B2 (en) 2016-09-30 2021-10-19 Sage Therapeutics, Inc. C7 substituted oxysterols and methods of use thereof
US11884697B2 (en) 2016-04-01 2024-01-30 Sage Therapeutics, Inc. Oxysterols and methods of use thereof

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6861788B2 (en) * 2003-06-03 2005-03-01 Motorola, Inc. Switchable display/mirror method and apparatus
CN103232520A (en) * 2013-05-13 2013-08-07 中国药科大学 Spirosteroid compounds and preparation method and medical application thereof
RU2635485C1 (en) * 2017-03-21 2017-11-13 Федеральное государственное автономное учреждение "Межотраслевой научно-технический комплекс "Микрохирургия глаза" имени академика С.Н. Федорова" Министерства здравоохранения Российской Федерации Method for optic nerve atrophy treatment after craniocerebral injury
CN112457362B (en) * 2019-09-06 2023-09-19 上海青东生物科技有限公司 Halogenated tetracyclic triterpene derivative and preparation and application thereof

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0162330A1 (en) * 1984-04-30 1985-11-27 Roecar Holdings (Netherlands Antilles) N.V. Use of sterolins and spiroketalins in the production of medicaments for the treatment of psoriasis
US4680289A (en) * 1985-06-05 1987-07-14 Progenics, Inc. Treatment of obesity and diabetes using sapogenins
DE4303214A1 (en) * 1993-02-04 1994-08-11 Wolfgang Marks Treatment of diseases of viral, viroidal or oncogenic origin by steroid saponins or their aglycones
EP0659762A2 (en) * 1993-12-24 1995-06-28 Mitsubishi Chemical Corporation Sialic acid derivatives
WO1998002450A2 (en) * 1996-07-11 1998-01-22 Inflazyme Pharmaceuticals Ltd. 6,7-oxygenated steroids and uses related thereto
WO1999048507A2 (en) * 1998-03-26 1999-09-30 Phytopharm Plc Steroidal saponins for treating alzheimer's disease
EP1024146A1 (en) * 1997-09-26 2000-08-02 Institute of Radiation Medicine Academy of Military Medical Sciences of the Pla The use of steroid saponin compounds to prevent senility, and novel steroid saponin compounds
WO2001023407A1 (en) * 1999-09-29 2001-04-05 Phytopharm Plc Sapogenin derivatives and their use in the treatment of cognitive dysfunction
WO2001023408A1 (en) * 1999-09-29 2001-04-05 Phytopharm Plc 5-hydroxysapogenin derivatives with anti-dementia activity
WO2001023406A1 (en) * 1999-09-29 2001-04-05 Phytopharm Plc 5-beta-sapogenin and pseudosapogenin derivatives and their use in the treatment of dementia
WO2001049703A2 (en) * 2000-01-06 2001-07-12 Phytopharm Plc Substituted sapogenins and their use
JP2002030096A (en) * 2000-07-11 2002-01-29 Kyorin Pharmaceut Co Ltd Agent for regenerating neurocyte process and method for producing the same
WO2003077869A2 (en) * 2002-03-15 2003-09-25 Samaritan Pharmaceuticals, Inc Neuroprotective spirostenol pharmaceutical compositions

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH05246866A (en) * 1992-03-06 1993-09-24 Ruibosuteii Japan:Kk Agent for stimulating cerebral metabolism and for improving cerebral function
JPH092956A (en) * 1995-06-22 1997-01-07 Mitsubishi Chem Corp Therapeutic and preventing agent for neuropathy
JPH092957A (en) * 1995-06-22 1997-01-07 Mitsubishi Chem Corp Therapeutic and preventing agent for peripheral neuropathy

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0162330A1 (en) * 1984-04-30 1985-11-27 Roecar Holdings (Netherlands Antilles) N.V. Use of sterolins and spiroketalins in the production of medicaments for the treatment of psoriasis
US4680289A (en) * 1985-06-05 1987-07-14 Progenics, Inc. Treatment of obesity and diabetes using sapogenins
DE4303214A1 (en) * 1993-02-04 1994-08-11 Wolfgang Marks Treatment of diseases of viral, viroidal or oncogenic origin by steroid saponins or their aglycones
EP0659762A2 (en) * 1993-12-24 1995-06-28 Mitsubishi Chemical Corporation Sialic acid derivatives
WO1998002450A2 (en) * 1996-07-11 1998-01-22 Inflazyme Pharmaceuticals Ltd. 6,7-oxygenated steroids and uses related thereto
EP1024146A1 (en) * 1997-09-26 2000-08-02 Institute of Radiation Medicine Academy of Military Medical Sciences of the Pla The use of steroid saponin compounds to prevent senility, and novel steroid saponin compounds
WO1999048507A2 (en) * 1998-03-26 1999-09-30 Phytopharm Plc Steroidal saponins for treating alzheimer's disease
WO2001023407A1 (en) * 1999-09-29 2001-04-05 Phytopharm Plc Sapogenin derivatives and their use in the treatment of cognitive dysfunction
WO2001023408A1 (en) * 1999-09-29 2001-04-05 Phytopharm Plc 5-hydroxysapogenin derivatives with anti-dementia activity
WO2001023406A1 (en) * 1999-09-29 2001-04-05 Phytopharm Plc 5-beta-sapogenin and pseudosapogenin derivatives and their use in the treatment of dementia
WO2001049703A2 (en) * 2000-01-06 2001-07-12 Phytopharm Plc Substituted sapogenins and their use
JP2002030096A (en) * 2000-07-11 2002-01-29 Kyorin Pharmaceut Co Ltd Agent for regenerating neurocyte process and method for producing the same
WO2003077869A2 (en) * 2002-03-15 2003-09-25 Samaritan Pharmaceuticals, Inc Neuroprotective spirostenol pharmaceutical compositions

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Section Ch, Week 199343 Derwent Publications Ltd., London, GB; Class B04, AN 1993-339641 XP002157713 & JP 05 246866 A (KATO K), 24 September 1993 (1993-09-24) *
INOUE K ET AL: "Cholestanol induces apoptosis of cerebellar neuronal cells." BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS. UNITED STATES 5 MAR 1999, vol. 256, no. 1, 5 March 1999 (1999-03-05), pages 198-203, XP001188107 ISSN: 0006-291X *
KYOUNG SOOK KIM, ET AL.: "Effects of cholestanol feeding on corneal dystrophy in mice." BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1085, 1991, pages 343-349, XP008027217 *
PATENT ABSTRACTS OF JAPAN vol. 1997, no. 05, 30 May 1997 (1997-05-30) & JP 09 002956 A (MITSUBISHI CHEM CORP), 7 January 1997 (1997-01-07) *
PATENT ABSTRACTS OF JAPAN vol. 1997, no. 05, 30 May 1997 (1997-05-30) & JP 09 002957 A (MITSUBISHI CHEM CORP), 7 January 1997 (1997-01-07) *
PATENT ABSTRACTS OF JAPAN vol. 2002, no. 05, 3 May 2002 (2002-05-03) & JP 2002 030096 A (KYORIN PHARMACEUT CO LTD), 29 January 2002 (2002-01-29) *
See also references of EP1490071A2 *
SUN, HUI ET AL: "Retinal stimulates ATP hydrolysis by purified and reconstituted ABCR, the photoreceptor-specific ATP-binding cassette transporter responsible for Stargardt disease" JOURNAL OF BIOLOGICAL CHEMISTRY (1999), 274(12), 8269-8281 , XP001165795 *

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005102299A1 (en) * 2004-04-09 2005-11-03 Michel Coisy Use of dopamine or the biological precursors thereof against algoneurodystrophy
FR2868700A1 (en) * 2004-04-09 2005-10-14 Michel Coisy USE OF DOPAMINE OR ITS BIOLOGICAL PRECURSORS AGAINST ALGONURODYSTRPHY.
EP2402362A2 (en) 2004-04-28 2012-01-04 Phytopharm PLC Crystalline forms of smilagenin
EP2479183A3 (en) * 2004-11-05 2013-08-28 Phytopharm PLC Polymorphs of sarsasapogenin
JP2008519020A (en) * 2004-11-05 2008-06-05 フィトファーム・パブリック・リミテッド・カンパニー Polymorphic form of salsasapogenin
EP2479183A2 (en) 2004-11-05 2012-07-25 Phytopharm PLC Polymorphs of sarsasapogenin
US8202554B2 (en) 2005-10-28 2012-06-19 Sun-Yeou Kim Extract of the family dioscoreaceae and composition for preventing or treating peripheral neuropathy comprising the same
JP2009513626A (en) * 2005-10-28 2009-04-02 キム・スンヨウ An extract of a plant of the genus Yamaceae, and a composition for preventing or treating peripheral neuropathy containing the same
WO2007049932A1 (en) 2005-10-28 2007-05-03 Sun-Yeou Kim Extract of the family dioscoreaceae and composition for preventing or treating peripheral neuropathy comprising the same
WO2010084356A1 (en) 2009-01-24 2010-07-29 Phytopharm Plc Treatment of neurotrophic factor mediated disorders
WO2012010896A1 (en) 2010-07-20 2012-01-26 Phytopharm Plc Treatment of l-dopa, dopamine agonist and/or dopamine enhancer induced disorders
AU2011281336B2 (en) * 2010-07-20 2015-03-05 Junaxo, Inc. Treatment of L-DOPA, dopamine agonist and/or dopamine enhancer induced disorders
US10759828B2 (en) 2011-09-08 2020-09-01 Sage Therapeutics, Inc. Neuroactive steroids, compositions, and uses thereof
WO2013097835A1 (en) * 2011-12-27 2013-07-04 Centro De Investigacion Y Desarrollo De Los Medicamentos (Cidem) Spirosteroidal systems having neuroactive, anti-inflammatory effects
WO2013149580A1 (en) * 2012-04-03 2013-10-10 Chiming Che Timosaponin compounds
US11319338B2 (en) 2012-04-03 2022-05-03 Goldporp Pharma Limited Timosaponin compounds
US11905309B2 (en) 2013-03-13 2024-02-20 Sage Therapeutics, Inc. Neuroactive steroids and methods of use thereof
US11104701B2 (en) 2013-03-13 2021-08-31 Sage Therapeutics, Inc. Neuroactive steroids and methods of use thereof
US10723758B2 (en) 2014-06-18 2020-07-28 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US10696712B2 (en) 2015-07-06 2020-06-30 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US10765685B2 (en) 2015-07-06 2020-09-08 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11117924B2 (en) 2015-07-06 2021-09-14 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11732000B2 (en) 2015-07-06 2023-08-22 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11884697B2 (en) 2016-04-01 2024-01-30 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US10752653B2 (en) 2016-05-06 2020-08-25 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11878995B2 (en) 2016-05-06 2024-01-23 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11407782B2 (en) 2016-05-06 2022-08-09 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11279730B2 (en) 2016-07-07 2022-03-22 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US10781231B2 (en) 2016-07-07 2020-09-22 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11149056B2 (en) 2016-09-30 2021-10-19 Sage Therapeutics, Inc. C7 substituted oxysterols and methods of use thereof
US11613556B2 (en) 2016-10-18 2023-03-28 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11149054B2 (en) 2016-10-18 2021-10-19 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
US11111266B2 (en) 2016-10-18 2021-09-07 Sage Therapeutics, Inc. Oxysterols and methods of use thereof
EP3564253A4 (en) * 2016-12-30 2020-07-29 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Antidepressant compound and preparation method and application thereof
US11142543B2 (en) 2016-12-30 2021-10-12 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Antidepressant compound and preparation method and application thereof
CN109988218B (en) * 2017-12-29 2022-11-25 中国科学院上海药物研究所 Sarsasapogenin derivative and preparation method and application thereof
CN109988218A (en) * 2017-12-29 2019-07-09 中国科学院上海药物研究所 A kind of sarsasapogenin derivative and its preparation method and application
EP3572085A1 (en) * 2018-05-25 2019-11-27 Neuro-Sys Synergestic combination composition comprising a steroidal saponin, a first polyphenolic compound and optionnaly a second polyphenolic compound
WO2019224388A1 (en) * 2018-05-25 2019-11-28 Neuralia Synergestic combination composition comprising a steroidal saponin, a first polyphenolic compound and a second polyphenolic compound

Also Published As

Publication number Publication date
RU2332999C2 (en) 2008-09-10
CN1642558A (en) 2005-07-20
TWI329016B (en) 2010-08-21
WO2003082893A3 (en) 2004-04-15
ATE424211T1 (en) 2009-03-15
NZ547344A (en) 2007-11-30
PE20040306A1 (en) 2004-05-29
RU2004130281A (en) 2006-01-20
PL213697B1 (en) 2013-04-30
PL372941A1 (en) 2005-08-08
TW200400042A (en) 2004-01-01
CN1642558B (en) 2012-05-30
WO2003082893A8 (en) 2003-12-31
KR20100093621A (en) 2010-08-25
IL164161A0 (en) 2005-12-18
NZ547897A (en) 2008-02-29

Similar Documents

Publication Publication Date Title
WO2003082893A2 (en) Theraputic methods and uses of sapogenins and their derivatives
US20110190249A1 (en) Therapeutic methods and uses of sapogenins and their derivatives
RU2325396C2 (en) Derivatives of 5b-sapogenin and pseudo sapogenin and application for dementia medical treatment
EP1383787B1 (en) Sapogenin derivatives, their synthesis and use
US20190002492A1 (en) Derivatives of allopregnanolone and of epiallopregnanolone and uses thereof for treating a neuropathological condition
AU2002242894A1 (en) Sapogenin derivatives, their synthesis and use
BRPI0622138A2 (en) compounds useful for treating neurodegenerative disorders
US20050130948A1 (en) Therapeutic methods and uses of sapogenins and their derivatives
CA2477787C (en) Therapeutic methods and uses of sapogenins and their derivatives
EP2111864A2 (en) Therapeutic methods and uses of sapogenins and their derivatives
KR20120128596A (en) Theraputic methods and uses of sapogenin and their derivatives
TWI357816B (en) Therapeutic methods and uses of sapogenins and the
NZ540712A (en) Steroidal sapogenin derivatives, their synthesis and their use in the treatment of cognitive dysfunction

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2003722713

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2477787

Country of ref document: CA

Ref document number: 1-2004-501326

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 535093

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 502/MUMNP/2004

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 1020047014567

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 164161

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: PA/a/2004/009321

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2003580357

Country of ref document: JP

Ref document number: 2003229877

Country of ref document: AU

Ref document number: 20038071886

Country of ref document: CN

Ref document number: 372941

Country of ref document: PL

ENP Entry into the national phase

Ref document number: 2004130281

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: P20041009A

Country of ref document: HR

WWP Wipo information: published in national office

Ref document number: 1020047014567

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2003722713

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10507000

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 12007501847

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 196300

Country of ref document: IL