WO2002087552A2 - Androgen-mediated neuroprotection and uses thereof - Google Patents

Androgen-mediated neuroprotection and uses thereof Download PDF

Info

Publication number
WO2002087552A2
WO2002087552A2 PCT/CA2002/000668 CA0200668W WO02087552A2 WO 2002087552 A2 WO2002087552 A2 WO 2002087552A2 CA 0200668 W CA0200668 W CA 0200668W WO 02087552 A2 WO02087552 A2 WO 02087552A2
Authority
WO
WIPO (PCT)
Prior art keywords
androgen
testosterone
disease
cell
neural
Prior art date
Application number
PCT/CA2002/000668
Other languages
French (fr)
Other versions
WO2002087552A3 (en
Inventor
Andrea Leblanc
Morrie Gelfand
Mark Trifiro
Original Assignee
Mcgill University
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 CA002345864A external-priority patent/CA2345864A1/en
Application filed by Mcgill University filed Critical Mcgill University
Priority to AU2002257430A priority Critical patent/AU2002257430A1/en
Publication of WO2002087552A2 publication Critical patent/WO2002087552A2/en
Publication of WO2002087552A3 publication Critical patent/WO2002087552A3/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
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/568Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone
    • 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
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/568Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone
    • A61K31/569Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone substituted in position 17 alpha, e.g. ethisterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system

Definitions

  • the invention relates to the prevention, treatment and study of neural disease and more particularly relates to androgen-mediated neuroprotection and its use for the prevention, treatment and study of neural disease.
  • estrogen reverses the behavioral and biochemical changes in ovariectomized rats (Simpkins et al . 1997) and enhances neuritic outgrowth and survival (Woolley and McEwen 1993, 1994; McEwen and Woolley 1994; Brinton et al . 1997; Woolley et al . 1997; McEwen et al . 1999).
  • Estrogen acts through genomic transactivation and non- genomic pathways (reviewed by Woolley 1999) .
  • Genomic events include up-regulation of brain-derived neurotrophic factor, nerve growth factor (NGF) , epidermal growth factor (Birge 1996) and of Bcl-2 proteins (Dubai et al . 1999; Pike 1999).
  • Estrogen also modulates p53 activity and cell fate (Wade et al . 1999).
  • Non-genomic events involve signal transduction, and it has been shown that estrogen activates the mitogen-activated protein kinase cascade in the cerebral cortex (Singh et al . 1999; 2000b; Toran-Allerand 2000a; 2000b) .
  • NGF nerve growth factor
  • Birge epidermal growth factor
  • Estrogen also modulates p53 activity and cell fate (Wade et al . 1999).
  • Non-genomic events involve signal transduction, and it has been shown that estrogen activates the mitogen-activated protein kinase cascade in the cerebral cortex (Singh et al . 1999; 2000b;
  • estrogen decreases the amount of amyloid- ⁇ peptide produced in neurons (Jaffe et al . 1994; Xu et al . 1998)
  • estrogen acts as an antioxidant, although it is unlikely that physiological levels of estrogen will have antioxidant activity (Behl et al . 1997; Moosmann and Behl 1999) .
  • An object of the invention is to provide methods and compounds for the prevention and/or treatment of neural or neurodegenerative disease or dementia. Accordingly, the invention provides a method of preventing or treating a neural disease in an animal, said method comprising administering an androgen or androgen-related compound to said animal.
  • the animal is a mammal, in a further embodiment, a human.
  • the animal is male.
  • the animal is female.
  • the neural disease is selected from the group consisting of Alzheimer disease, Parkinson's disease, amyotropic lateral sclerosis (AL ⁇ ), cerebellar degeneration, ischemia (stroke), traumatic injuries, prion diseases (e.g. Creutzfeldt-Jakob disease) , Huntington disease, frontal lobe dementia, vascular dementia, infection related dementia (e.g. HIV), head injury, hereditary cerebral amyloidogenesis, Down's Syndrome and cerebral hemorrhage.
  • a ⁇ amyotropic lateral sclerosis
  • stroke ischemia
  • prion diseases e.g. Creutzfeldt-Jakob disease
  • Huntington disease frontal lobe dementia
  • vascular dementia e.g. HIV
  • head injury e.g. HIV
  • hereditary cerebral amyloidogenesis e.g. HIV
  • the neural disease is an A ⁇ - mediated neural disease.
  • the androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone.
  • the androgen is testosterone.
  • the androgen-related compound is a testosterone ester, in a further embodiment, the testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate.
  • the invention further provides a composition for preventing or treating a neural disease in an animal, said composition comprising an androgen or androgen- related compound in admixture with a pharmaceutically suitable carrier.
  • the invention further provides a use of an androgen or androgen-related compound or the above-mentioned composition for preventing or treating a neural disease in an animal.
  • the invention further provides a use of an androgen or androgen-related compound for preparation of a medicament for preventing or treating a neural disease in an animal.
  • the invention further provides a commercial package comprising an androgen or androgen-related compound or the above-mentioned composition together with instructions for the prevention or treatment of a neural disease.
  • the neural disease is selected from the group consisting of Alzheimer disease, Parkinson's disease, amyotropic lateral sclerosis (ALS) , cerebellar degeneration, ischemia (stroke), traumatic injuries, prion diseases (e.g. Creutzfeldt-Jakob disease) , Huntington disease, frontal lobe dementia, vascular dementia, infection related dementia (e.g. HIV), head injury, hereditary cerebral amyloidogenesis, Down's
  • the androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone.
  • the androgen is testosterone.
  • the androgen-related compound is a testosterone ester, in a further embodiment, the testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate.
  • the invention further provides a method for preventing or inhibiting cell death, comprising treating a cell with an androgen or androgen-related compound.
  • the cell is a neural cell.
  • the cell is a mammalian cell, in a further embodiment a human cell.
  • the androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone.
  • the androgen is testosterone.
  • the androgen-related compound is a testosterone ester, in a further embodiment, the testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate.
  • the invention further provides a method of identifying or characterizing a test compound for the prevention and/or treatment of neural or neurodegenerative disease, said method comprising: contacting said test compound with a cell comprising an androgen receptor; measuring a test level of androgen-associated activity; comparing said test level of androgen- associated activity with a corresponding control level of androgen-associated activity in a corresponding cell which was not exposed to said test compound; wherein a difference between said test level and said control level indicates that the test compound may be used for prevention and/or treatment of neural or neurodegenerative disease.
  • the above-mentioned cell is a neural cell.
  • the above-mentioned androgen-associated activity is an inhibition or prevention of cell death or apoptosis.
  • the invention further provides a method of identifying and/or characterizing a mechanism and/or component associated with androgen-mediated effects on cell death, said method comprising: assessing an alteration in a cell death- as-sociated phenotype between an androgen treated cell and a corresponding untreated cell; and comparing said alteration with a corresponding alteration assessed in corresponding androgen treated versus untreated mutant cells; wherein differences in said alteration are used to identify and/or characterize a mechanism and/or component associated with androgen-mediated effects on cell death.
  • Figure 1 Chemical structures of androgen, estrogens and flutamide.
  • Figure 2 Androgens offer neuroprotection against serum deprivation-mediated apoptosis.
  • **p ⁇ 0.005 indicate the significance of the difference between serum deprived neurons in the absence and in the presence of hormone.
  • FIG. 3 Mibolerone protects against serum deprivation- mediated neuronal apoptosis. Serum deprived neurons were treated with 4 nM epitestosterone, methyltestosterone or testosterone in the presence or absence of 3 nM mibolerone for 96 h. Data represents the mean and SEM from three independent neuronal preparations. *p ⁇ 0.05, **p ⁇ 0.005 indicate the significance of the difference between serum deprived neurons in the absence and in the presence of hormone.
  • Aromatase inhibitor, 4-androsten-4-OL-3, 17- dione, does not inhibit testosterone neuroprotection. Serum deprived neurons were treated in the absence or presence of 4 nM testosterone and 5 and 50 ng/mL aromatase inhibitor, 4-androsten-4-OL-3, 17-dione (Al), and kept in culture for 96 h. Neuronal apoptosis was measured as described and expressed relative to control serum deprived neurons (arbitrarily placed at 100%) .
  • FIG. 5 Flutamide prevents testosterone-mediated neuroprotection. Serum-deprived neurons treated with
  • Figure 6 Neuroprotection by steroid hormones against A ⁇ - 2 -mediated toxicity of human neurons in primary culture. Results show mean and SEM of three independent experiments (200 cells/experiment) .
  • Figure 7 Further results demonstrating estrogen- and androgen-mediated protection of human neurons against
  • BSA- ⁇ E2 conjugated-17- ⁇ -estradiol conjugated-17- ⁇ -estradiol conjugated-17- ⁇ -estradiol
  • Figure 8 Effect of hormone pre-incubation on hormone- mediated protection of human neurons against
  • the neurons were pre-treated with either hormone or hormone and antagonist (tamoxifen [TXM] for estrogens and flutamide [Flut] for androgens) for one hour prior to
  • Described herein is an assessment of the role of physiological concentrations of androgens on serum deprivation-mediated apoptosis and on intracellular amyloid beta peptide-mediated neurotoxicity of human primary CNS neuron cultures.
  • Applicants have determined that androgens, such as testosterone, protect neurons against cell death or apoptosis, such as that induced by serum deprivation, by acting through androgen receptors. Applicants have further determined that androgens, such as testosterone,
  • amyloid beta peptide e.g. A ⁇ _ 42
  • estrogens e.g. 17- ⁇ -
  • estradiol protect neurons from intracellular amyloid
  • beta peptide e.g. A ⁇ i_ 2
  • a ⁇ i_ 2 beta peptide
  • Alzheimer's amyloid- ⁇ peptide in primary rat cortical neurons (Tirassa et al . 1997; Gouras et al . 2000). A preponderance in dementia among females has been suggested (Molsa et al . 1982) .
  • Testosterone propionate prevents developmental neuronal loss in the medial preoptic nucleus of males or sex-reversed female rats (Dodson and Gorski 1993) .
  • Androgens can increase the volume, neuron number and synapses of developing rat superior cervical ganglion
  • estrogens e.g. 17- ⁇ -estradiol
  • neuroprotection is also conferred when the androgen is added prior to exposure of cells to amyloid beta peptide, thus further demonstrating a prophylactic use. Neuroprotective effects of both occur at physiological concentrations. Therefore, applicants conclude that neurons are as responsive to androgens as estrogens with respect to neuronal survival.
  • Applicants demonstrate herein that physiological levels of androgen, such as testosterone, protect against cell death or apoptosis, such as serum deprivation- mediated neuronal apoptosis, through interaction with androgen receptors.
  • Applicants have confirmed the presence of androgen receptors in the human neuron cultures studied.
  • Applicants have shown that the non- aromatizable form ' of androgen, mibolerone, induces neuroprotection similar to testosterone.
  • Mibolerone is a highly specific synthetic androgen that binds the androgen receptor with 100-fold higher affinity than the natural androgen testosterone (Wilson and French 1976; Traish et al . 1986; Turcotte et al . 1988; Markiewicz and Gurpide 1997).
  • aromatase inhibitor 4-androsten-4-OL-3, 17-dione
  • 4-androsten-4-OL-3 17-dione
  • the neuroprotective effect of testosterone is blocked by the pure synthetic anti-androgen, flutamide (Brogden and
  • estrogens e.g. 17- ⁇ -estradiol
  • intracellular amyloid beta peptide e.g. A ⁇ i_ 42
  • tamoxifen a compound that inhibits the neuroprotective effect of estrogens.
  • the neuroprotective effect of testosterone is 100% up to 48 h after serum deprivation. Thereafter, there is increasing neuronal apoptosis, even in the presence of testosterone, although the levels are generally 60% lower than in absence of hormone. Since the media was changed every 48 h, turnover of testosterone cannot be responsible for the less protective effect. It is more likely that cumulative insult caused by continuous serum deprivation is responsible for the inability of testosterone to neuroprotect completely in time. As shown in other systems, inhibiting cell death with one compound may provide a certain degree of treatment of neurodegenerative diseases and combination therapies including both cell death inhibitors and pro-survival factors may be used to completely suppress neuronal cell death.
  • Epitestosterone is aromatized into 17- ⁇ -estradiol
  • the transcriptionally inactive 17- ⁇ - estradiol is also neuroprotective (Green et al . 1997).
  • 17- ⁇ -estradiol mediates neuroprotection through signal transduction rather than through a genomic pathway.
  • methyltestosterone and mibolerone are not as neuroprotective as testosterone indicates that the structure of the steroid may be very important in mediating the neuroprotective effect of androgens.
  • Comparison of the chemical structure of the three compounds shows that mibolerone and methyltestosterone share a 17-methyl group that is absent in testosterone. It is envisioned that this methyl group accounts for the lesser neuropotency of methyltestosterone and mibolerone.
  • androgens are nuclear receptor proteins that can activate gene transcription or act through signal transduction. The neuroprotective effect of estrogen is known to act through the estrogen receptor and to activate both genomic and non-genomic pathways of neuronal protection (Woolley 1999) . Through the genomic pathway, estrogen upregulates Bcl-2 levels (Dubai et al .
  • An advantage of the present invention is the use of primary neural cultures rather than neuronal cell lines, which eliminates the above mentioned differences and reflects more accurately the processes associated with neural cells in vivo .
  • androgen receptors are expressed in the temporal, frontal and hippocampal regions of the brain (Puy et al . 1995; Finley and Kritzer 1999) . Androgen receptors are selectively localized to neuronal subtypes, and immunoreactivity appears specific to pyramidal neurons in primate prefrontal cortex (Finley and Kritzer 1999) . The number of androgen receptors does not differ in male and female rat or monkey brains (Clancy et al . 1992). The neuroprotective effect of testosterone through androgen receptors observed in human neurons in culture and described herein shall also occur in the human brain.
  • Applicant's findings demonstrate that androgens can help in the treatment of Alzheimer disease in a manner similar to estrogen-replacement therapy in women. Symptoms associated with the decreasing levels of androgen in both men and women are alleviated by hormone replacement therapy. The content of androgens in women's HRT should be taken into consideration in epidemiologic studies on the effect of HRT against Alzheimer disease. Androgens thus provide an effective treatment for aging animals and of neuroprotection against neural disease, for example, Alzheimer disease.
  • such an animal is a mammal.
  • such animals are human.
  • such an animal is male.
  • such an animal is female.
  • the invention relates to androgens and androgen- related compounds and their use in conferring protection against cell death or apoptosis.
  • a cell is a neural cell, in which case the androgens and androgen-related compounds confer neuroprotection, i.e.
  • the invention provides a method for preventing or inhibiting cell death, comprising treating or contacting a cell with, or exposing a cell to, an androgen or androgen-related compound.
  • the cell is a neural cell.
  • the cell is a mammalian cell, in a further embodiment, a human cell.
  • the above-mentioned neural cell death or apoptosis, neurodegeneration or neurotoxicity is caused by the presence of a level of amyloid beta peptide or an amyloid beta peptide-like compound, i.e. is amyloid beta peptide- or amyloid beta peptide-like compound-mediated.
  • a level of amyloid beta peptide or an amyloid beta peptide-like compound i.e. is amyloid beta peptide- or amyloid beta peptide-like compound-mediated.
  • the amyloid beta for example the A ⁇ _ 42 peptide recognize the C-terminus of this molecule, there is actually variability with respect to the N-terminal residue or N-terminal limit of the peptides detected as such (see Naslund, et al . 2000). Accordingly, in certain embodiments, the amyloid beta
  • peptide is of the structure A ⁇ n . 42 , where "n" defines the N-terminus of the peptide.
  • amyloid is of the structure A ⁇ n . 42 , where "n" defines the N-terminus of the peptide.
  • the amyloid is of the structure A ⁇ n . 42 , where "n" defines the N-terminus of the peptide.
  • the amyloid is of the structure A ⁇ n . 42 , where "n" defines the N-terminus of the peptide.
  • beta peptide is A ⁇ - 2
  • the amyloid beta peptide is another peptide which is derived from the amyloid precursor protein.
  • amyloid beta peptides have been described in for example Selkoe (1998) . A higher than normal intracellular level of amyloid beta
  • peptide 1-42 (or in some cases by A ⁇ n - 42 , based on the antibodies used as noted above) in neural tissue has been shown to be associated with neural disease and dementia, such as Alzheimer disease (D'Andrea et al . 2001; Gouras et al . 2000; Naslund et al . 2000) . Applicants further describe that estrogens (e.g. 17-
  • amyloid beta peptide e.g. A ⁇ i_ 42
  • neurotoxicity a phenomenon that has not been described prior to Applicants' work described herein. Such neuroprotection is shown to be blocked by tamoxifen, indicating that it is mediated through estrogen receptors.
  • Amyloid beta peptide-like compounds refers to any compound (such as a homologous or related peptide) which possesses similar structure, features and/or activity with an amyloid beta peptide. Such a feature may be an association with a neural disease or dementia, such as Alzheimer disease. Such activity may be the ability to elicit neurotoxicity.
  • “Androgen-related” compounds refer to any compounds which are related to an androgen by having similar structure and/or activity (i.e. an androgen-associated activity) .
  • “Androgen-associated activity” in this case refers to any activity associated with an androgen, including binding to an androgen receptor or a fragment, homolog or variant thereof (which retains androgen binding activity) , or affecting an androgen-mediated process.
  • Such processes may include androgen-mediated signaling pathways or androgen-mediated gene expression.
  • gene expression may be measured by detection of the corresponding RNA or protein, or via the use of a suitable reporter construct comprising a transcriptional regulatory element (s) normally associated with such a gene whose expression is androgen-mediated, operably- linked to a reporter gene.
  • a first nucleic acid sequence is "operably-linked" with a second nucleic acid sequence when the first nucleic acid sequence is placed in a functional relationship with the second nucleic acid sequence.
  • a promoter is operably-linked to a coding sequence if the promoter affects the transcription or expression of the coding sequences.
  • operably-linked DNA sequences are contiguous and, where necessary to join two protein coding regions, in reading frame.
  • enhancers generally function when separated from the promoters by several kilobases and intronic sequences may be of variable lengths, some polynucleotide elements may be operably-linked but not contiguous.
  • Transcriptional regulatory element is a generic term that refers to DNA sequences, such as initiation and termination signals, enhancers, and promoters, splicing signals, polyadenylation signals which induce or control transcription of protein coding sequences with which they are operably-linked.
  • the expression of such a reporter gene may be measured on the transcriptional or translational level, e.g. by the amount of RNA or protein produced.
  • RNA may be detected by for example Northern analysis or by the reverse transcriptase-polymerase chain reaction (RT-PCR) method (see for example Sambrook et al (1989) Molecular Cloning: A Laboratory Manual (second edition) , Cold Spring Harbor Laboratory Press, Cold
  • Protein levels may be detected either directly using affinity reagents (e.g. an antibody or fragment thereof [for methods, see for example Harlow, E. and Lane, D (1988) Antibodies : A
  • Suitable reporter genes include but are not limited to chloramphenicol acetyltransferase, beta-D galactosidase, luciferase, or green fluorescent protein.
  • such an androgen-mediated process is neuroprotection, as determined by for example the inhibition or prevention of neural cell death or apoptosis as described in the examples below.
  • Cell death or apoptosis may be measured by a number of methods known in the art (see for example Apoptosis Techniques and
  • Binding activity may be measured by contacting a test compound with an androgen receptor or fragment, homolog or variant thereof which retains binding activity. Using appropriate detection means (e.g. radiolabelling, fluorescence, reporter enzymes, ligand- binding partner systems [e.g. biotin- (strept) avidin] ) and control samples, the amount and affinity of binding of a test compound may be determined.
  • detection means e.g. radiolabelling, fluorescence, reporter enzymes, ligand- binding partner systems [e.g. biotin- (strept) avidin]
  • Androgen-related compounds also comprise compounds which share a similar structure or property (ies) with an androgen or a known androgen-related compound.
  • androgens include but are not limited to testosterone, testosterone enanthate, methyltestosterone, epitestosterone, and mibolerone.
  • androgen-related compounds include but are not limited to those compounds produced by the modification of an androgen.
  • Such compounds include, for example, certain testosterone esters (e.g. testosterone enanthate; testosterone propionate) , which, upon hydrolysis of the ester, yield active testosterone.
  • the above mentioned compound could be a peptide, a polypeptide, a fragment of a polypeptide, an organic natural molecule, or a synthetic molecule. Such compounds may have agonistic or antagonistic activity.
  • Such compounds may further include pro-drugs that are metabolized in vivo to produce a compound as described above.
  • pro-drugs include the testosterone esters noted above.
  • the invention therefore provides methods of preventing or treating a neural or neurodegenerative disease or a dementia in an animal comprising administering an androgen or an androgen- related compound to an animal.
  • an animal is a mammal.
  • the animal is a human.
  • the animal is of the male sex.
  • the animal is female.
  • the neural or neurodegenerative disease or dementia is selected from the group consisting of
  • Alzheimer disease Parkinson's disease, amyotropic lateral sclerosis (ALS) , cerebellar degeneration, ischemia (stroke), traumatic injuries, prion diseases
  • neural or neurodegenerative disease or dementia may further comprise any condition characterized by or associated with a level of an amyloid beta peptide or an amyloid beta peptide-like compound in neural tissue, including, but not limited to, Alzheimer disease.
  • Alzheimer disease e.g. Creutzfeldt-Jakob disease
  • Huntington disease e.g. Creutzfeldt-Jakob disease
  • frontal lobe dementia e.g. vascular dementia
  • vascular dementia e.g. HIV
  • infection related dementia e.g. HIV
  • head injury e.g. HIV
  • the above mentioned neural or neurodegenerative disease or dementia may further comprise any condition characterized by or associated with a level of an amyloid beta peptide or an amyloid beta peptide-like compound in neural tissue, including, but not limited to, Alzheimer disease.
  • such a peptide is A ⁇ _ 42 .
  • such conditions include hereditary cerebral amyloidogenesis, where amyloid deposit results in the rupture of blood vessels in the brain (resulting in early death by hemorrhage), and Down's Syndrome, in which the presence of intracellular amyloid peptides precedes extracellular amyloid deposition (Gyure et al . 2001).
  • the invention further provides a composition for the prevention and/or treatment of a neural or neurodegenerative disease comprising an androgen or androgen-related compound in admixture with a pharmaceutically acceptable carrier.
  • the invention further provides a use of an androgen or androgen-related compound or the above-mentioned composition for the prevention and/or treatment of a neural or neurodegenerative disease.
  • the invention further provides a use of an androgen or androgen-related compound for preparation of a medicament for the prevention and/or treatment of a neural or neurodegenerative disease.
  • the invention further provides commercial packages comprising an androgen or an androgen-related compound or the above-mentioned composition together with instructions for the prevention and/or treatment of a neural or neurodegenerative disease.
  • the androgens include but are not limited to testosterone, mibolerone, methyltestosterone and epitestosterone. In an embodiment, the androgen is testosterone.
  • an androgen or androgen- related compound may be used therapeutically in formulations or medicaments to prevent or treat a neural or neurodegenerative disease.
  • the invention provides corresponding methods of medical treatment, in which a therapeutic dose of an androgen or androgen-related compound is administered in a pharmacologically acceptable formulation.
  • the invention also provides therapeutic compositions comprising an androgen or androgen-related compound and a pharmacologically acceptable excipient or carrier.
  • the therapeutic composition may be soluble in an aqueous solution at a physiologically acceptable pH.
  • the androgens or androgen-related compounds of the invention are lipid-soluble or soluble in polar solvents (e.g. ethanol) , which, once dissolved, may be diluted in aqueous solution.
  • compositions containing (comprising) an androgen or an androgen-related compound.
  • compositions include an androgen or androgen-related compound in a therapeutically or prophylactically effective amount sufficient to inhibit neural or neurodegenerative disease, and a pharmaceutically acceptable carrier.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as reduction of neural or neurodegenerative disease progression.
  • a therapeutically effective amount of androgen or androgen-related compound may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects.
  • prophylactically effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as preventing or inhibiting neural or neurodegenerative disease onset or progression.
  • a prophylactically effective amount can be determined as described above for the therapeutically effective amount.
  • specific dosage regimens may be adjusted over time according to the individual need and the professional judgement of the person administering or supervising the administration of the compositions.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the carrier is suitable for parenteral administration.
  • the carrier can be suitable for intravenous, intraperitoneal, intramuscular, sublingual or oral administration.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated.
  • Supplementary active compounds can also be incorporated into the compositions.
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, monostearate salts and gelatin.
  • the androgen can be administered in a time release formulation, for example in a composition which includes a slow release polymer.
  • the active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, polylactic acid and polylactic, polyglycolic copolymers (PLG) . Many methods for the preparation of such formulations are patented or generally known to those skilled in the art.
  • Sterile injectable solutions can be prepared by incorporating the active compound (e.g. androgen or androgen-related compound) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • an androgen may be formulated with one or more additional compounds that enhance the solubility of the androgen.
  • compositions of the present invention comprising an androgen or androgen-related compound, may be provided in containers or commercial packages which further comprise instructions for use of the androgen for the prevention and/or treatment of a neural or neurodegenerative disease.
  • a further aspect the invention is a method of preventing or treating a neural or neurodegenerative disease comprising administering a prophylactically or therapeutically effective amount of an androgen or androgen-related compound and an estrogen or estrogen-related compound to an animal.
  • the invention further provides a use of an androgen or androgen-related compound and an estrogen or estrogen-related compound for the prevention or treatment of a neural or neurodegenerative disease, as well as for the preparation of a medicament for the prevention or treatment of a neural or neurodegenerative disease.
  • the invention further provides a composition comprising an androgen or androgen-related compound and an estrogen or estrogen- related compound in admixture with a pharmaceutically acceptable excipient or carrier.
  • the invention further provides a commercial package comprising an androgen or androgen-related compound and an estrogen or estrogen- related compound or the above-mentioned composition together with instructions for preventing or treating a neural or neurodegenerative disease.
  • the invention further provides screening methods for identifying and characterizing compounds for the prevention and/or treatment of neural or neurodegenerative disease.
  • Such compounds may include the androgen-related compounds as described above.
  • the invention further provides a method for identifying or characterizing a test compound for the prevention and/or treatment of neural or neurodegenerative disease, said method comprising: contacting said test compound with a cell comprising an androgen receptor; measuring a test level of androgen- associated activity; comparing said test level of androgen-associated activity with a corresponding control level of androgen- associated activity in a corresponding cell which was not exposed to said test compound.
  • test level may indicate that the test compound may be used for prevention and/or treatment of a neural or neurodegenerative disease.
  • the just noted difference may in various embodiments represent an increase or a decrease, depending on the activity measured.
  • the cell is a neural cell.
  • the androgen-associated activity is neuroprotection or the inhibition or prevention of cell death or apoptosis.
  • an increase in neuroprotection or a decrease in cell death or apoptosis as a result of treatment with the test compound indicates that the test compound can be used for the prevention and/or treatment of neural or neurodegenerative disease.
  • the above-mentioned cell comprising an androgen receptor is a cell which comprises endogenous levels or expression of an androgen receptor.
  • the cell may also comprise an appropriate host cell comprising an exogenously introduced source of androgen receptor.
  • a host cell may be prepared by the introduction of a nucleic acid encoding an androgen receptor into the host cell and providing conditions for the expression of an androgen receptor.
  • a nucleic acid is DNA.
  • host cells may be prokaryotic or eukaryotic, bacterial, yeast, amphibian or mammalian. In an embodiment, such host cells are human.
  • a homolog, variant and/or fragment of an androgen receptor which retains activity may also be used in the methods of the invention.
  • Homologs include protein sequences which are substantially identical to the amino acid sequence of an androgen receptor, sharing significant structural and functional homology with an androgen receptor.
  • Variants include, but are not limited to, proteins or peptides which differ from an androgen receptor by any modifications, and/or amino acid substitutions, deletions or additions. Modifications can occur anywhere including the polypeptide backbone, (i.e. the amino acid sequence) , the amino acid side chains and the amino or carboxy termini. Such substitutions, deletions or additions may involve one or more amino acids.
  • Fragments include a fragment or a portion of an androgen receptor or a fragment or a portion of a homolog or variant of an androgen receptor.
  • the above-mentioned method may be employed either with a single test compound or a plurality or library (e.g. a combinatorial library) of test compounds. In the latter case, synergistic effects provided by combinations of compounds may also be identified and characterized.
  • the above-mentioned compounds may be used for prevention and/or treatment of neural or neurodegenerative disease or may be used as lead compounds for the development and testing of additional compounds having improved specificity, efficacy and/or pharmacological (e.g. pharmacokinetic) properties.
  • the above-mentioned method may further be used to identify agonists or antagonists of androgen receptors.
  • the above- mentioned test compound may be selected from a group of compounds having similar structure to an androgen or and androgen-related compound and/or is known to have androgen receptor binding activity.
  • the above-mentioned method has a greater probability of identifying a compound for prevention and/or treatment of neural or neurodegenerative disease.
  • one or a plurality of the steps of the screening/testing methods of the invention may be automated.
  • the invention further provides systems and methods for the identification and characterization of such mechanisms.
  • Such systems and methods may comprise a comparison of androgen treated cells versus corresponding untreated cells, with respect to cell death.
  • such cells are neural cells.
  • Such methods may be utilized for screening, wherein mutations resulting in cell death-associated phenotypes which are androgen dependent may be identified, and subsequently the mutation may be identified at the genotypic level, giving rise to the identification of a gene involved in androgen-associated cell death mechanisms.
  • the amount or nature of neuroprotection typically conferred by an androgen is different in a certain cell, that cell harbors an alteration, such as a mutation, which gives rise to this difference.
  • a mutation is thus associated with a gene involved in a mechanism of androgen-mediated protection.
  • the above noted mutation renders the cell less responsive or unresponsive to androgen-mediated inhibition or prevention of cell death.
  • the invention further provides a method of identifying and/or characterizing mechanisms and/or components associated with androgen-mediated effects on cell death, said method comprising: assessing an alteration in a cell death- associated phenotype between an androgen or androgen- related compound-treated cell and a corresponding untreated cell comparing said alteration with a corresponding alteration assessed in corresponding androgen treated versus untreated mutant cells; wherein differences in said alteration are used to identify and/or characterize mechanisms and/or components associated with androgen-mediated effects on cell death.
  • the androgen is mibolerone, methyltestosterone or epitestosterone.
  • the androgen is testosterone.
  • the above-mentioned cell is a neural cell.
  • the just noted difference in said alteration is a reduction or elimination of responsiveness to androgen-mediated inhibition and/or prevention of cell death.
  • Example 1 Demonstration of androgen-mediated and estrogen-mediated neuroprotection against serum- deprivation-mediated apoptosis of human primary neurons.
  • Estrogen is hypothesized to play an important role against Alzheimer's disease in women (Birge 1996). Women receiving HRT are less susceptible to Alzheimer's disease
  • methyltestosterone did not significantly inhibit apoptosis at 24 h, but did show a 20% reduction in apoptosis between 48 and 96 h of treatment (p ⁇ 0.05).
  • the anti-androgen, epitestosterone also had no statistically significant effect at 24 h but reduced apoptosis by 20-40% from 48 to 96 h of treatment (p > 0.05).
  • ⁇ -estradiol (p ⁇ 0.05) were also neuroprotective from 24 to 96 h of treatment.
  • the neuroprotective effect of mibolerone is not as strong as that of testosterone.
  • methyltestosterone also shows neuroprotection at a lower level than in testosterone.
  • Comparison of the chemical structure of these compounds indicates that the presence of the 17-methyl group decreases the neuropotency of androgens.
  • the addition of mibolerone to testosterone, epitestosterone or methyltestosterone does not increase neuroprotection in cells treated with hormone in absence of mibolerone suggesting that both mibolerone and natural androgens act through the same receptor.
  • the affinity of mibolerone is at least 100- fold higher than testosterone, epitestosterone or methyltestosterone (Wilson and French 1976; Turcotte et al . , 1988).
  • the slow neuroprotective effect of methyltestosterone which is only observed after 48 h of treatment, indicates that metabolites of methyltestosterone may be produced over time and promote neuroprotection through both androgen receptor dependent (antagonized by flutamide) and androgen receptor independent (not antagonized by flutamide) mechanisms.
  • flutamide could not inhibit the endogenous anti-androgen, epitestosterone-mediated neuroprotection indicating that ⁇ the neuroprotection of epitestosterone is independent of androgen receptors.
  • Human fetal brain tissue (12-16 weeks) was obtained in accordance with the guidelines established by the Medical Research Council and approved by the Institutional Review Board of McGill University. Neurons were isolated and cultured as previously described (LeBlanc 1995) . To summarize, brain tissue was minced in phosphate buffered saline and dissociated with 0.25% trypsin (Gibco-BRL, Rockville, MD, USA) . The cells were subsequently treated with 10% serum and 0.1 mg/ml deoxyribonuclease I (Roche Molecular Biochemical, Indianapolis, IN, USA) and the resulting homogenate
  • the neurons were plated at 3 x 10 6 cells/mL on poly-L-lysine
  • ACLARTM 33C; 5 mm; Allied Chemical Corp. coverslips and cultured in vitro for 10 days.
  • the media contains phenol-free minimal essential media in Earle's balanced salt solution containing 0.225% sodium bicarbonate, 1 mM sodium pyruvate, 2 mM L-glutamine, 0.1% dextrose, I x antibiotic
  • Pen-Strep All products from Gibco-BRL
  • 5% decomplemented fetal bovine serum HyClone, Logan. UT
  • the neurons were serum-deprived in the absence or
  • the media was changed every 48 h. Hormones were dissolved in various stock concentrations in 100% ethanol and added to the media to give final concentrations of 2 and 4 nM or the indicated dose with equivalent amounts of ethanol. Control serum-deprived neurons receive the equivalent amount of ethanol. Similarly, mibolerone, flutamide and aromatase inhibitor were dissolved in ethanol and added to the media to give final concentrations of 3 nM mibolerone (DuPont NEN, Boston.
  • coverslips were fixed with 4% paraformaldehyde, 4% sucrose in phosphate buffered solution (Harlow and Lane 1988).
  • Androgen receptors were identified by incubating 6 nM [ 3 H] mibolerone (DuPont NEN, Boston, MA. USA; Spec. Act. 85 Ci/mmol) with 6 x 10 6 neurons to measure total binding. Non-specific binding was assessed by competing the binding of [ 3 H] -mibolerone with a 200-fold excess cold mibolerone as previously described (Kaufman et al . 1993). Specific binding was determined by subtracting non- specific from total binding and dividing by the protein concentration as determined by the Lowry assay (Lowry et al . 1951) .
  • Example 7 A ⁇ - 42 -mediated neurotoxicity is protected by steroid hormones.
  • a toxic dose of A ⁇ x _ 2 and Dextran Texas Red fluorescent dye marker was microinjected into neurons and neurons were immediately treated with varying doses of 17- ⁇ -estradiol, transcriptionally inactive 17- ⁇ -estradiol analogue, testosterone enanthate, methyltestosterone and epitestosterone.
  • Microinjected neurons were incubated for 24 hours, fixed and submitted to TUNEL analysis to assess cell death by A ⁇ _ 42 . The experiment was done on 100 microinjected cells/well x 2 wells/experiments in three independent neuron preparations.
  • a ⁇ _ 42 induces TUNEL-positive cell death in 60-70% of neurons within 24 hours of injection.
  • Post-hoc analysis with Dunnett's test shows that a physiological concentration of 2 nM of 17- ⁇ -estradiol and methyltestosterone prevent A ⁇ - 42 mediated neuronal cell death by 50% (p ⁇ 0.01).
  • Testosterone is less but still neuroprotective (p ⁇ 0.05 for 2 and 4 nM and p ⁇ 0.01 at 10 nM) . In contrast, epitestosterone and 17- ⁇ -estradiol do not protect against A ⁇ _ 42 -mediated apoptosis.
  • Figure 7 illustrates further results demonstrating that estrogen and androgens protect human neurons against
  • Neurons were microinjected with a lethal dose of A ⁇ - 42 and
  • estradiol (BSA- ⁇ E2) .
  • Neuronal cell death was assessed by TUNEL 24 hours after treatment. % cell death represents the number of TUNEL positive neurons over total neurons
  • Each sample represents 200 microinjected cells from each of a minimum of three experiments done on different neuron preparations.
  • Figure 8 illustrates further results obtained similarly to those described for figure 7, but in this case the neurons were pre-incubated with 10 nM hormones
  • Figure 9 illustrates results demonstrating that estrogens and androgens protect through their respective receptors.
  • the neurons were pre-treated with either hormone or hormone and antagonist (tamoxifen for estrogens and flutamide for androgens) for one hour and
  • Alzheimer amyloid ⁇ precursor protein J. Biol . Chem . 269, 13065-13068. Jorm A. F., Korten A. E. and Henderson A. S. (1987) The Prevalence of dementia: a quantitative integration of the literature. Acta Psychia tr Scand. 76, 465- 479. Kaufman M. , Pinsky L., Trifiro M. , Lumbroso R. ,
  • amyloid ⁇ -peptide in serum deprived human primary neuron cultures possible involvement of apoptosis.
  • Singh S. M. Gauthier S. and Labrie F. (2000b) Androgen receptor antagonists (antiandrogens) : structure- activity relationships. Curr. Med. Chem . 1 , 211-247. Singh M., Setalo G., Guan X., Frail D. E. and Toran- Allerand D. (2000a) Estrogen-induced activation of the mitogen-activated protein kinase cascade in the
  • Toran-Allerand C. D. (2000b) Novel sites and mechanisms of oestrogen action in the brain. Novartis Found.
  • Alzheimer's disease - Beta-amyloid resurrected Alzheimer's disease - Beta-amyloid resurrected.

Abstract

Androgens, such as testosterone, methyltestosterone, and epitestosterone, are disclosed herein as being neuroprotective, in that they inhibit or prevent cell death or apoptosis of neural cells. A neuroprotective effect of androgens on primary cultures of human neurons with respect to serum deprivation-mediated apoptosis and amyloid beta peptide mediated neurotoxicity is described. The invention thus provides methods and uses of androgens as well as compositions and commercial packages comprising androgens for the prevention and/or treatment of neural or neurodegenerative disease and for the prevention and/or inhibition of cell death. The invention further provides methods of identifying and screening compounds for the prevention and/or treatment of neural or neurodegenerative disease.

Description

ANDROGEN-MEDIATED NEUROPROTECTION AND USES THEREOF
FIELD OF THE INVENTION
The invention relates to the prevention, treatment and study of neural disease and more particularly relates to androgen-mediated neuroprotection and its use for the prevention, treatment and study of neural disease.
BACKGROUND OF THE INVENTION
Epidemiological studies have shown that decreasing levels of estrogen are a risk factor for Alzheimer' s disease and hormone replacement therapy (HRT) offers protection against Alzheimer's disease (Paganini-Hill
1996; Schneider et al . 1996; Tang et al . 1996; Schneider et al . 1997). In animal models and cell cultures, estrogen reverses the behavioral and biochemical changes in ovariectomized rats (Simpkins et al . 1997) and enhances neuritic outgrowth and survival (Woolley and McEwen 1993, 1994; McEwen and Woolley 1994; Brinton et al . 1997; Woolley et al . 1997; McEwen et al . 1999). Estrogen acts through genomic transactivation and non- genomic pathways (reviewed by Woolley 1999) . Genomic events include up-regulation of brain-derived neurotrophic factor, nerve growth factor (NGF) , epidermal growth factor (Birge 1996) and of Bcl-2 proteins (Dubai et al . 1999; Pike 1999). Estrogen also modulates p53 activity and cell fate (Wade et al . 1999). Non-genomic events involve signal transduction, and it has been shown that estrogen activates the mitogen-activated protein kinase cascade in the cerebral cortex (Singh et al . 1999; 2000b; Toran-Allerand 2000a; 2000b) . In addition,
estrogen decreases the amount of amyloid-β peptide produced in neurons (Jaffe et al . 1994; Xu et al . 1998)
and can protect against amyloid-β peptide-mediated neurotoxicity (Goodman and Mattson 1996; Behl et al . 1997). Others propose that estrogen acts as an antioxidant, although it is unlikely that physiological levels of estrogen will have antioxidant activity (Behl et al . 1997; Moosmann and Behl 1999) .
There is therefore a need to characterize such systems and identify new compounds useful for the prevention, treatment and study of neural disease.
SUMMARY OF THE INVENTION
An object of the invention is to provide methods and compounds for the prevention and/or treatment of neural or neurodegenerative disease or dementia. Accordingly, the invention provides a method of preventing or treating a neural disease in an animal, said method comprising administering an androgen or androgen-related compound to said animal. In an embodiment, the animal is a mammal, in a further embodiment, a human. In an embodiment, the animal is male. In an embodiment, the animal is female.
In certain embodiments, the neural disease is selected from the group consisting of Alzheimer disease, Parkinson's disease, amyotropic lateral sclerosis (ALΞ), cerebellar degeneration, ischemia (stroke), traumatic injuries, prion diseases (e.g. Creutzfeldt-Jakob disease) , Huntington disease, frontal lobe dementia, vascular dementia, infection related dementia (e.g. HIV), head injury, hereditary cerebral amyloidogenesis, Down's Syndrome and cerebral hemorrhage.
In an embodiment, the neural disease is an Aβ- mediated neural disease.
In certain embodiments, the androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone. In an embodiment, the androgen is testosterone. In an embodiment, the androgen-related compound is a testosterone ester, in a further embodiment, the testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate. The invention further provides a composition for preventing or treating a neural disease in an animal, said composition comprising an androgen or androgen- related compound in admixture with a pharmaceutically suitable carrier.
The invention further provides a use of an androgen or androgen-related compound or the above-mentioned composition for preventing or treating a neural disease in an animal.
The invention further provides a use of an androgen or androgen-related compound for preparation of a medicament for preventing or treating a neural disease in an animal.
The invention further provides a commercial package comprising an androgen or androgen-related compound or the above-mentioned composition together with instructions for the prevention or treatment of a neural disease.
In certain embodiments, the neural disease is selected from the group consisting of Alzheimer disease, Parkinson's disease, amyotropic lateral sclerosis (ALS) , cerebellar degeneration, ischemia (stroke), traumatic injuries, prion diseases (e.g. Creutzfeldt-Jakob disease) , Huntington disease, frontal lobe dementia, vascular dementia, infection related dementia (e.g. HIV), head injury, hereditary cerebral amyloidogenesis, Down's
Syndrome and cerebral hemorrhage. In embodiments, the androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone. In an embodiment, the androgen is testosterone. In an embodiment, the androgen-related compound is a testosterone ester, in a further embodiment, the testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate.
The invention further provides a method for preventing or inhibiting cell death, comprising treating a cell with an androgen or androgen-related compound. In an embodiment, the cell is a neural cell. In an embodiment, the cell is a mammalian cell, in a further embodiment a human cell. In certain embodiments, the androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone. In an embodiment, the androgen is testosterone. In an embodiment, the androgen-related compound is a testosterone ester, in a further embodiment, the testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate.
The invention further provides a method of identifying or characterizing a test compound for the prevention and/or treatment of neural or neurodegenerative disease, said method comprising: contacting said test compound with a cell comprising an androgen receptor; measuring a test level of androgen-associated activity; comparing said test level of androgen- associated activity with a corresponding control level of androgen-associated activity in a corresponding cell which was not exposed to said test compound; wherein a difference between said test level and said control level indicates that the test compound may be used for prevention and/or treatment of neural or neurodegenerative disease. In an embodiment the above-mentioned cell is a neural cell. In an embodiment the above-mentioned androgen-associated activity is an inhibition or prevention of cell death or apoptosis.
The invention further provides a method of identifying and/or characterizing a mechanism and/or component associated with androgen-mediated effects on cell death, said method comprising: assessing an alteration in a cell death- as-sociated phenotype between an androgen treated cell and a corresponding untreated cell; and comparing said alteration with a corresponding alteration assessed in corresponding androgen treated versus untreated mutant cells; wherein differences in said alteration are used to identify and/or characterize a mechanism and/or component associated with androgen-mediated effects on cell death.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1: Chemical structures of androgen, estrogens and flutamide.
Figure 2: Androgens offer neuroprotection against serum deprivation-mediated apoptosis. (a) Time study of neuroprotection by 4 nM testosterone, epitestosterone or
methyltestosterone and 2 nm 17-β-estradiol or 17-α- estradiol treatment at 24, 48, 72 and 96 h of serum deprivation. The level of apoptosis in hormone treated neurons is expressed as percentage neuronal cell death detected by propidium iodide and TUNEL staining. Data represents the mean and SEM of experiments of 10 independent neuronal cultures for all except methyltestosterone (n= 5) . (b) Dose response effect of each hormone on neuronal protection. Data represents the mean and SEM of eight independent experiments. *p <
0.05, **p < 0.005 indicate the significance of the difference between serum deprived neurons in the absence and in the presence of hormone.
Figure 3: Mibolerone protects against serum deprivation- mediated neuronal apoptosis. Serum deprived neurons were treated with 4 nM epitestosterone, methyltestosterone or testosterone in the presence or absence of 3 nM mibolerone for 96 h. Data represents the mean and SEM from three independent neuronal preparations. *p < 0.05, **p < 0.005 indicate the significance of the difference between serum deprived neurons in the absence and in the presence of hormone.
Figure 4: Aromatase inhibitor, 4-androsten-4-OL-3, 17- dione, does not inhibit testosterone neuroprotection. Serum deprived neurons were treated in the absence or presence of 4 nM testosterone and 5 and 50 ng/mL aromatase inhibitor, 4-androsten-4-OL-3, 17-dione (Al), and kept in culture for 96 h. Neuronal apoptosis was measured as described and expressed relative to control serum deprived neurons (arbitrarily placed at 100%) .
Data represents mean and SEM of three independent experiments. *p < 0.01, **p < 0.005 indicate the significance of the difference between serum deprived neurons in the absence and in the presence of hormone/drug.
Figure 5: Flutamide prevents testosterone-mediated neuroprotection. Serum-deprived neurons treated with
4 nM androgens in the absence or presence of 2 and 20 μM flutamide for 96 h. Data represents the mean and SEM of four independent experiments. *p < 0.05, **p < 0.002 indicates the significance of the difference between neurons that are serum deprived and those treated with hormone. Comparison of hormone and flutamide treated cells were made to serum deprivation in the presence of flutamide.
Figure 6: Neuroprotection by steroid hormones against Aβι-2-mediated toxicity of human neurons in primary culture. Results show mean and SEM of three independent experiments (200 cells/experiment) . Figure 7: Further results demonstrating estrogen- and androgen-mediated protection of human neurons against
intracellular Aβι_42-mediated toxicity in primary cultures.
17-β-estradiol (βE2) , 17-α-estradiol (αE2), testosterone enanthate (Test), epitestosterone (Epi-Test) , methyl testosterone (Methyl-test) or cell impermeable BSA
conjugated-17-β-estradiol (BSA-βE2) were used. Neuronal cell death was assessed by TUNEL 24 hours after treatment. % cell death represents the number of TUNEL positive neurons over total neurons (stained with DAPI) . Each sample represents 200 microinjected cells from each of a minimum of three experiments done on different
neuron preparations. *p<0.01 relative to Aβι_42- microinjected cells in absence of hormones.
Figure 8 : Effect of hormone pre-incubation on hormone- mediated protection of human neurons against
intracellular Aβι_42-mediated toxicity in primary cultures. Neurons were pre-incubated with 10 nM hormones for 1 hour
prior to microinjection with Aβι-42. Treatment with hormones was continued for 24 hours before measuring cell death as above. Figure 9: Effect of hormone antagonists on hormone- mediated protection of human neurons against
intracellular Aβι-42-mediated toxicity in primary cultures.
The neurons were pre-treated with either hormone or hormone and antagonist (tamoxifen [TXM] for estrogens and flutamide [Flut] for androgens) for one hour prior to
microinjection with Aβι_42. * p<0.01.
DETAILED DESCRIPTION OF THE INVENTION
Described herein is an assessment of the role of physiological concentrations of androgens on serum deprivation-mediated apoptosis and on intracellular amyloid beta peptide-mediated neurotoxicity of human primary CNS neuron cultures.
Applicants have determined that androgens, such as testosterone, protect neurons against cell death or apoptosis, such as that induced by serum deprivation, by acting through androgen receptors. Applicants have further determined that androgens, such as testosterone,
protect neurons from amyloid beta peptide (e.g. Aβι_42)-
mediated neurotoxicity, and that estrogens (e.g. 17-β-
estradiol) protect neurons from intracellular amyloid
beta peptide (e.g. Aβi_2) -mediated neurotoxicity. Prior to applicant's studies, much attention has
been given to the role of 17-β-estradiol against Alzheimer's disease. With regard to the aging CNS in males, men in their sixties are usually less prone to Alzheimer's disease than women of the same age (Molsa et al . 1982; Jorm et al . 1987). Androgens eventually decrease with age (Flood et al . 1995; Vermeulen 1991). Testosterone replacement therapy improves depression, and verbal and spatial memory in aging men (Sternbach 1998) . At the molecular level, testosterone is shown to increase NGF and p75-nerve growth factor receptor and to decrease
Alzheimer's amyloid-β peptide in primary rat cortical neurons (Tirassa et al . 1997; Gouras et al . 2000). A preponderance in dementia among females has been suggested (Molsa et al . 1982) .
In women, androgens are also present (although at much lower levels) and decrease with age (Rako 1998) . Decreasing androgen levels are associated with a number of post-menopausal conditions such as osteoporosis, depression, reduced muscle and bone mass and increased visceral fat (Davis 1999) . Evaluation of testosterone in neuronal cell lines failed to reveal a role (Green et al 1997) .
Testosterone propionate prevents developmental neuronal loss in the medial preoptic nucleus of males or sex-reversed female rats (Dodson and Gorski 1993) .
Androgens can increase the volume, neuron number and synapses of developing rat superior cervical ganglion
(Wright et al . 1991) . In aging, there is little evidence that androgens regulate neuronal survival. However, testosterone deficiency in males is associated with conditions that indicate CNS neuronal dysfunction such as depression, anxiety and memory loss (Sternbach 1998) .
Furthermore, replacement therapy significantly improves these symptoms.
Applicants have demonstrated that androgens offer as much neuroprotection against growth factor deprivation mediated neuronal apoptosis of CNS differentiated human
neurons as 17-β-estradiol . Applicants have further demonstrated that androgens, (e.g. testosterone) further offer as much neuroprotection against amyloid beta
peptide (e.g. Aβι-2) -mediated neurotoxicity as 17-β- estradiol. Applicants have further demonstrated that
estrogens (e.g. 17-β-estradiol) offer neuroprotection
against intracellular amyloid beta peptide (e.g. Aβι_42)- mediated neurotoxicity. Such neuroprotection is also conferred when the androgen is added prior to exposure of cells to amyloid beta peptide, thus further demonstrating a prophylactic use. Neuroprotective effects of both occur at physiological concentrations. Therefore, applicants conclude that neurons are as responsive to androgens as estrogens with respect to neuronal survival.
Applicants demonstrate herein that physiological levels of androgen, such as testosterone, protect against cell death or apoptosis, such as serum deprivation- mediated neuronal apoptosis, through interaction with androgen receptors. Applicants have confirmed the presence of androgen receptors in the human neuron cultures studied. Applicants have shown that the non- aromatizable form' of androgen, mibolerone, induces neuroprotection similar to testosterone. Mibolerone is a highly specific synthetic androgen that binds the androgen receptor with 100-fold higher affinity than the natural androgen testosterone (Wilson and French 1976; Traish et al . 1986; Turcotte et al . 1988; Markiewicz and Gurpide 1997). Furthermore, aromatase inhibitor, 4-androsten-4-OL-3, 17-dione, does not block testosterone-mediated neuroprotection. However, the neuroprotective effect of testosterone is blocked by the pure synthetic anti-androgen, flutamide (Brogden and
Chrisp 1991; Simard et al . 1986; Namer 1988; Labrie 1993; Markiewicz and Gurpide 1997; Singh et al . 2000a). Therefore, the neuroprotective effect of testosterone is mediated through androgen receptors. It is further shown herein that the neuroprotective
effect of estrogens (e.g. 17-β-estradiol) against
intracellular amyloid beta peptide (e.g. Aβi_42) -mediated neurotoxicity may be blocked by the antagonist tamoxifen, indicating that the neuroprotective effect of estrogens is mediated through estrogen receptors.
The neuroprotective effect of testosterone is 100% up to 48 h after serum deprivation. Thereafter, there is increasing neuronal apoptosis, even in the presence of testosterone, although the levels are generally 60% lower than in absence of hormone. Since the media was changed every 48 h, turnover of testosterone cannot be responsible for the less protective effect. It is more likely that cumulative insult caused by continuous serum deprivation is responsible for the inability of testosterone to neuroprotect completely in time. As shown in other systems, inhibiting cell death with one compound may provide a certain degree of treatment of neurodegenerative diseases and combination therapies including both cell death inhibitors and pro-survival factors may be used to completely suppress neuronal cell death.
The neuroprotective effect of the weak endogenous antiandrogen, epitestosterone (Nuck and Lucky 1987; Starka et al . 1989; Starka et al . 1991) is surprising. This is clearly not the case for the other anti-androgen, flutamide. Furthermore, flutamide did not antagonize the epitestosterone effect, indicating that epitestosterone, unlike mibolerone, testosterone and methyltestosterone, cannot act through the androgen receptor.
Epitestosterone is aromatized into 17-α-estradiol
(Finkelstein et al . 1981). Given that the neuroprotective effect of epitestosterone occurs only after 48 h of treatment, it is possible that the neuroprotective effect is mediated through aromatization
into 17-α-estradiol . As shown by others, and by
applicants herein, the transcriptionally inactive 17-α- estradiol is also neuroprotective (Green et al . 1997).
It is proposed that 17-α-estradiol mediates neuroprotection through signal transduction rather than through a genomic pathway.
The fact that methyltestosterone and mibolerone are not as neuroprotective as testosterone indicates that the structure of the steroid may be very important in mediating the neuroprotective effect of androgens. Comparison of the chemical structure of the three compounds shows that mibolerone and methyltestosterone share a 17-methyl group that is absent in testosterone. It is envisioned that this methyl group accounts for the lesser neuropotency of methyltestosterone and mibolerone. Like estrogen, androgens are nuclear receptor proteins that can activate gene transcription or act through signal transduction. The neuroprotective effect of estrogen is known to act through the estrogen receptor and to activate both genomic and non-genomic pathways of neuronal protection (Woolley 1999) . Through the genomic pathway, estrogen upregulates Bcl-2 levels (Dubai et al .
1999; Pike 1999) and it is envisioned that increased Bcl-
2 levels enhance neuronal protection against serum deprivation. Whether androgens regulate gene expression or activity of survival genes in a manner similar to estrogens remains to be determined but this is a likely mechanism to explain the neuroprotective nature of androgens. Applicants results in primary cultures of human neurons contrast with those observed in the estrogen responsive, human SK-N-SH, neuronal cell line (Green et al . 1997). Androgens are metabolized by these cells and can affect proliferation (Maggi et al . 1998) . However, it is not unexpected to find differences between primary neurons and neuroblastoma cell lines as these differ in many ways such as in the state of differentiation, cell growth and cell death. An advantage of the present invention is the use of primary neural cultures rather than neuronal cell lines, which eliminates the above mentioned differences and reflects more accurately the processes associated with neural cells in vivo . In vivo, androgen receptors are expressed in the temporal, frontal and hippocampal regions of the brain (Puy et al . 1995; Finley and Kritzer 1999) . Androgen receptors are selectively localized to neuronal subtypes, and immunoreactivity appears specific to pyramidal neurons in primate prefrontal cortex (Finley and Kritzer 1999) . The number of androgen receptors does not differ in male and female rat or monkey brains (Clancy et al . 1992). The neuroprotective effect of testosterone through androgen receptors observed in human neurons in culture and described herein shall also occur in the human brain.
Applicant's findings demonstrate that androgens can help in the treatment of Alzheimer disease in a manner similar to estrogen-replacement therapy in women. Symptoms associated with the decreasing levels of androgen in both men and women are alleviated by hormone replacement therapy. The content of androgens in women's HRT should be taken into consideration in epidemiologic studies on the effect of HRT against Alzheimer disease. Androgens thus provide an effective treatment for aging animals and of neuroprotection against neural disease, for example, Alzheimer disease. In an embodiment, such an animal is a mammal. In an embodiment, such animals are human. In an embodiment, such an animal is male. In an embodiment, such an animal is female.
It is envisioned that the time of treatment is to be considered in this regard, since in post-mortem human hippocampus, the amount of androgen receptors has been observed to decrease with age in the CAl region (Tohgi et al . 1995). Observations have been made with respect to HRT and the treatment of Alzheimer disease in post- menopausal women in this regard (Mulnard et al . , 2000). The invention relates to androgens and androgen- related compounds and their use in conferring protection against cell death or apoptosis. In an embodiment, such a cell is a neural cell, in which case the androgens and androgen-related compounds confer neuroprotection, i.e. the inhibition or prevention of neural cell death or apoptosis, to prevent or reduce neurodegeneration and neurotoxicity, and for the prevention and/or treatment of neural disease. Therefore, in an aspect the invention provides a method for preventing or inhibiting cell death, comprising treating or contacting a cell with, or exposing a cell to, an androgen or androgen-related compound. In an embodiment, the cell is a neural cell. In an embodiment, the cell is a mammalian cell, in a further embodiment, a human cell.
In an embodiment, the above-mentioned neural cell death or apoptosis, neurodegeneration or neurotoxicity, is caused by the presence of a level of amyloid beta peptide or an amyloid beta peptide-like compound, i.e. is amyloid beta peptide- or amyloid beta peptide-like compound-mediated. As antibodies used for detection of
for example the Aβι_42 peptide recognize the C-terminus of this molecule, there is actually variability with respect to the N-terminal residue or N-terminal limit of the peptides detected as such (see Naslund, et al . 2000). Accordingly, in certain embodiments, the amyloid beta
peptide is of the structure Aβn.42, where "n" defines the N-terminus of the peptide. In an embodiment, the amyloid
beta peptide is Aβι-2, in further embodiments, the amyloid beta peptide is another peptide which is derived from the amyloid precursor protein. Such amyloid beta peptides have been described in for example Selkoe (1998) . A higher than normal intracellular level of amyloid beta
peptide 1-42 (or in some cases by Aβn-42, based on the antibodies used as noted above) in neural tissue has been shown to be associated with neural disease and dementia, such as Alzheimer disease (D'Andrea et al . 2001; Gouras et al . 2000; Naslund et al . 2000) . Applicants further describe that estrogens (e.g. 17-
β-estradiol) offer neuroprotection against intracellular
amyloid beta peptide (e.g. Aβi_42) -mediated neurotoxicity, a phenomenon that has not been described prior to Applicants' work described herein. Such neuroprotection is shown to be blocked by tamoxifen, indicating that it is mediated through estrogen receptors.
"Amyloid beta peptide-like compounds" refers to any compound (such as a homologous or related peptide) which possesses similar structure, features and/or activity with an amyloid beta peptide. Such a feature may be an association with a neural disease or dementia, such as Alzheimer disease. Such activity may be the ability to elicit neurotoxicity. "Androgen-related" compounds refer to any compounds which are related to an androgen by having similar structure and/or activity (i.e. an androgen-associated activity) . "Androgen-associated activity" in this case refers to any activity associated with an androgen, including binding to an androgen receptor or a fragment, homolog or variant thereof (which retains androgen binding activity) , or affecting an androgen-mediated process. Such processes may include androgen-mediated signaling pathways or androgen-mediated gene expression. Such gene expression may be measured by detection of the corresponding RNA or protein, or via the use of a suitable reporter construct comprising a transcriptional regulatory element (s) normally associated with such a gene whose expression is androgen-mediated, operably- linked to a reporter gene. A first nucleic acid sequence is "operably-linked" with a second nucleic acid sequence when the first nucleic acid sequence is placed in a functional relationship with the second nucleic acid sequence. For instance, a promoter is operably-linked to a coding sequence if the promoter affects the transcription or expression of the coding sequences. Generally, operably-linked DNA sequences are contiguous and, where necessary to join two protein coding regions, in reading frame. However, since for example enhancers generally function when separated from the promoters by several kilobases and intronic sequences may be of variable lengths, some polynucleotide elements may be operably-linked but not contiguous. "Transcriptional regulatory element" is a generic term that refers to DNA sequences, such as initiation and termination signals, enhancers, and promoters, splicing signals, polyadenylation signals which induce or control transcription of protein coding sequences with which they are operably-linked. The expression of such a reporter gene may be measured on the transcriptional or translational level, e.g. by the amount of RNA or protein produced. RNA may be detected by for example Northern analysis or by the reverse transcriptase-polymerase chain reaction (RT-PCR) method (see for example Sambrook et al (1989) Molecular Cloning: A Laboratory Manual (second edition) , Cold Spring Harbor Laboratory Press, Cold
Spring Harbor, New York, USA) . Protein levels may be detected either directly using affinity reagents (e.g. an antibody or fragment thereof [for methods, see for example Harlow, E. and Lane, D (1988) Antibodies : A
Labora tory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY] ; a ligand which binds the protein) or by other properties (e.g. fluorescence in the case of green fluorescent protein) or by measurement of the protein' s activity, which may entail enzymatic activity to produce a detectable product (e.g. with altered spectroscopic properties) or a detectable phenotype (e.g. alterations in cell growth). Suitable reporter genes include but are not limited to chloramphenicol acetyltransferase, beta-D galactosidase, luciferase, or green fluorescent protein.
In an embodiment, such an androgen-mediated process is neuroprotection, as determined by for example the inhibition or prevention of neural cell death or apoptosis as described in the examples below. Cell death or apoptosis may be measured by a number of methods known in the art (see for example Apoptosis Techniques and
Protocols, edited by Judes Poirier, Humana Press, Totowa,
NJ, Vol. 29 of Neuromethods series, Series editor Alan A. Boulton, Glen B. Baker, 1997) . An example of such a method of measuring cell death is TUNEL, as described in
Example 6 below.
Binding activity may be measured by contacting a test compound with an androgen receptor or fragment, homolog or variant thereof which retains binding activity. Using appropriate detection means (e.g. radiolabelling, fluorescence, reporter enzymes, ligand- binding partner systems [e.g. biotin- (strept) avidin] ) and control samples, the amount and affinity of binding of a test compound may be determined.
Androgen-related compounds also comprise compounds which share a similar structure or property (ies) with an androgen or a known androgen-related compound. Examples of androgens include but are not limited to testosterone, testosterone enanthate, methyltestosterone, epitestosterone, and mibolerone. In embodiments, androgen-related compounds include but are not limited to those compounds produced by the modification of an androgen. Such compounds include, for example, certain testosterone esters (e.g. testosterone enanthate; testosterone propionate) , which, upon hydrolysis of the ester, yield active testosterone.
The above mentioned compound could be a peptide, a polypeptide, a fragment of a polypeptide, an organic natural molecule, or a synthetic molecule. Such compounds may have agonistic or antagonistic activity.
Such compounds may further include pro-drugs that are metabolized in vivo to produce a compound as described above. Examples include the testosterone esters noted above.
Accordingly, the invention therefore provides methods of preventing or treating a neural or neurodegenerative disease or a dementia in an animal comprising administering an androgen or an androgen- related compound to an animal. In an embodiment, such an animal is a mammal. In an embodiment, the animal is a human. In an embodiment, the animal is of the male sex. In an embodiment, the animal is female. In certain embodiments, the neural or neurodegenerative disease or dementia is selected from the group consisting of
Alzheimer disease, Parkinson's disease, amyotropic lateral sclerosis (ALS) , cerebellar degeneration, ischemia (stroke), traumatic injuries, prion diseases
(e.g. Creutzfeldt-Jakob disease), Huntington disease, frontal lobe dementia, vascular dementia, infection related dementia (e.g. HIV) and head injury. The above mentioned neural or neurodegenerative disease or dementia may further comprise any condition characterized by or associated with a level of an amyloid beta peptide or an amyloid beta peptide-like compound in neural tissue, including, but not limited to, Alzheimer disease. In
embodiments, such a peptide is Aβι_42. In further embodiments, such conditions include hereditary cerebral amyloidogenesis, where amyloid deposit results in the rupture of blood vessels in the brain (resulting in early death by hemorrhage), and Down's Syndrome, in which the presence of intracellular amyloid peptides precedes extracellular amyloid deposition (Gyure et al . 2001).
The invention further provides a composition for the prevention and/or treatment of a neural or neurodegenerative disease comprising an androgen or androgen-related compound in admixture with a pharmaceutically acceptable carrier.
The invention further provides a use of an androgen or androgen-related compound or the above-mentioned composition for the prevention and/or treatment of a neural or neurodegenerative disease.
The invention further provides a use of an androgen or androgen-related compound for preparation of a medicament for the prevention and/or treatment of a neural or neurodegenerative disease.
The invention further provides commercial packages comprising an androgen or an androgen-related compound or the above-mentioned composition together with instructions for the prevention and/or treatment of a neural or neurodegenerative disease.
In various embodiments, the androgens include but are not limited to testosterone, mibolerone, methyltestosterone and epitestosterone. In an embodiment, the androgen is testosterone.
In various embodiments, an androgen or androgen- related compound may be used therapeutically in formulations or medicaments to prevent or treat a neural or neurodegenerative disease. The invention provides corresponding methods of medical treatment, in which a therapeutic dose of an androgen or androgen-related compound is administered in a pharmacologically acceptable formulation. Accordingly, the invention also provides therapeutic compositions comprising an androgen or androgen-related compound and a pharmacologically acceptable excipient or carrier. The therapeutic composition may be soluble in an aqueous solution at a physiologically acceptable pH. In some embodiments, the androgens or androgen-related compounds of the invention are lipid-soluble or soluble in polar solvents (e.g. ethanol) , which, once dissolved, may be diluted in aqueous solution.
The invention provides pharmaceutical compositions (medicaments) containing (comprising) an androgen or an androgen-related compound. In one embodiment, such compositions include an androgen or androgen-related compound in a therapeutically or prophylactically effective amount sufficient to inhibit neural or neurodegenerative disease, and a pharmaceutically acceptable carrier.
A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as reduction of neural or neurodegenerative disease progression. A therapeutically effective amount of androgen or androgen-related compound may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual. Dosage regimens may be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as preventing or inhibiting neural or neurodegenerative disease onset or progression. A prophylactically effective amount can be determined as described above for the therapeutically effective amount.
For any particular subject, specific dosage regimens may be adjusted over time according to the individual need and the professional judgement of the person administering or supervising the administration of the compositions.
As used herein "pharmaceutically acceptable carrier" or "excipient" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. In one embodiment, the carrier is suitable for parenteral administration. Alternatively, the carrier can be suitable for intravenous, intraperitoneal, intramuscular, sublingual or oral administration. Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated.
Supplementary active compounds can also be incorporated into the compositions.
Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, monostearate salts and gelatin. Moreover, the androgen can be administered in a time release formulation, for example in a composition which includes a slow release polymer. The active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, polylactic acid and polylactic, polyglycolic copolymers (PLG) . Many methods for the preparation of such formulations are patented or generally known to those skilled in the art.
Sterile injectable solutions can be prepared by incorporating the active compound (e.g. androgen or androgen-related compound) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. In accordance with an alternative aspect of the invention, an androgen may be formulated with one or more additional compounds that enhance the solubility of the androgen.
In accordance with another aspect of the invention, therapeutic compositions of the present invention, comprising an androgen or androgen-related compound, may be provided in containers or commercial packages which further comprise instructions for use of the androgen for the prevention and/or treatment of a neural or neurodegenerative disease.
Applicants have demonstrated herein that androgens provide neuroprotective effects via different pathways and mechanisms than those conferred by estrogens. Therefore, it is envisioned that combined therapy comprising both the administration of an androgen or androgen-related compound with an estrogen or estrogen- related compound may be further effective in the prevention and/or treatment of a neural or neurodegenerative disease. Thus, in the case of women receiving hormone replacement therapy (HRT) , the additional administration of an androgen or androgen- related compound may improve the neuroprotective effects of HRT. Therefore, a further aspect the invention is a method of preventing or treating a neural or neurodegenerative disease comprising administering a prophylactically or therapeutically effective amount of an androgen or androgen-related compound and an estrogen or estrogen-related compound to an animal. The invention further provides a use of an androgen or androgen-related compound and an estrogen or estrogen-related compound for the prevention or treatment of a neural or neurodegenerative disease, as well as for the preparation of a medicament for the prevention or treatment of a neural or neurodegenerative disease. The invention further provides a composition comprising an androgen or androgen-related compound and an estrogen or estrogen- related compound in admixture with a pharmaceutically acceptable excipient or carrier. The invention further provides a commercial package comprising an androgen or androgen-related compound and an estrogen or estrogen- related compound or the above-mentioned composition together with instructions for preventing or treating a neural or neurodegenerative disease.
The invention further provides screening methods for identifying and characterizing compounds for the prevention and/or treatment of neural or neurodegenerative disease. Such compounds may include the androgen-related compounds as described above.
Accordingly, the invention further provides a method for identifying or characterizing a test compound for the prevention and/or treatment of neural or neurodegenerative disease, said method comprising: contacting said test compound with a cell comprising an androgen receptor; measuring a test level of androgen- associated activity; comparing said test level of androgen-associated activity with a corresponding control level of androgen- associated activity in a corresponding cell which was not exposed to said test compound.
A difference between the test level and the control level may indicate that the test compound may be used for prevention and/or treatment of a neural or neurodegenerative disease. The just noted difference may in various embodiments represent an increase or a decrease, depending on the activity measured.
In an embodiment, the cell is a neural cell. In an embodiment, the androgen-associated activity is neuroprotection or the inhibition or prevention of cell death or apoptosis. In an embodiment, an increase in neuroprotection or a decrease in cell death or apoptosis as a result of treatment with the test compound indicates that the test compound can be used for the prevention and/or treatment of neural or neurodegenerative disease. In an embodiment, the above-mentioned cell comprising an androgen receptor is a cell which comprises endogenous levels or expression of an androgen receptor.
The cell may also comprise an appropriate host cell comprising an exogenously introduced source of androgen receptor. Such a host cell may be prepared by the introduction of a nucleic acid encoding an androgen receptor into the host cell and providing conditions for the expression of an androgen receptor. In an embodiment, such a nucleic acid is DNA. Such host cells may be prokaryotic or eukaryotic, bacterial, yeast, amphibian or mammalian. In an embodiment, such host cells are human.
A homolog, variant and/or fragment of an androgen receptor which retains activity may also be used in the methods of the invention. Homologs include protein sequences which are substantially identical to the amino acid sequence of an androgen receptor, sharing significant structural and functional homology with an androgen receptor. Variants include, but are not limited to, proteins or peptides which differ from an androgen receptor by any modifications, and/or amino acid substitutions, deletions or additions. Modifications can occur anywhere including the polypeptide backbone, (i.e. the amino acid sequence) , the amino acid side chains and the amino or carboxy termini. Such substitutions, deletions or additions may involve one or more amino acids. Fragments include a fragment or a portion of an androgen receptor or a fragment or a portion of a homolog or variant of an androgen receptor. The above-mentioned method may be employed either with a single test compound or a plurality or library (e.g. a combinatorial library) of test compounds. In the latter case, synergistic effects provided by combinations of compounds may also be identified and characterized. The above-mentioned compounds may be used for prevention and/or treatment of neural or neurodegenerative disease or may be used as lead compounds for the development and testing of additional compounds having improved specificity, efficacy and/or pharmacological (e.g. pharmacokinetic) properties. The above-mentioned method may further be used to identify agonists or antagonists of androgen receptors. In an embodiment, the above- mentioned test compound may be selected from a group of compounds having similar structure to an androgen or and androgen-related compound and/or is known to have androgen receptor binding activity. In the case of the latter embodiment, the above-mentioned method has a greater probability of identifying a compound for prevention and/or treatment of neural or neurodegenerative disease. In certain embodiments, one or a plurality of the steps of the screening/testing methods of the invention may be automated.
The reduction of cell death/apoptosis by androgen or androgen-related compound as described herein indicates the presence of various androgen-associated mechanisms involved in cell death. Therefore, the invention further provides systems and methods for the identification and characterization of such mechanisms. Such systems and methods may comprise a comparison of androgen treated cells versus corresponding untreated cells, with respect to cell death. In an embodiment, such cells are neural cells. Such methods may be utilized for screening, wherein mutations resulting in cell death-associated phenotypes which are androgen dependent may be identified, and subsequently the mutation may be identified at the genotypic level, giving rise to the identification of a gene involved in androgen-associated cell death mechanisms. For example, if the amount or nature of neuroprotection typically conferred by an androgen is different in a certain cell, that cell harbors an alteration, such as a mutation, which gives rise to this difference. Such a mutation is thus associated with a gene involved in a mechanism of androgen-mediated protection. In embodiments, the above noted mutation renders the cell less responsive or unresponsive to androgen-mediated inhibition or prevention of cell death.
Accordingly, the invention further provides a method of identifying and/or characterizing mechanisms and/or components associated with androgen-mediated effects on cell death, said method comprising: assessing an alteration in a cell death- associated phenotype between an androgen or androgen- related compound-treated cell and a corresponding untreated cell comparing said alteration with a corresponding alteration assessed in corresponding androgen treated versus untreated mutant cells; wherein differences in said alteration are used to identify and/or characterize mechanisms and/or components associated with androgen-mediated effects on cell death. In various embodiments, the androgen is mibolerone, methyltestosterone or epitestosterone. In an embodiment, the androgen is testosterone. In an embodiment the above-mentioned cell is a neural cell. In an embodiment, the just noted difference in said alteration is a reduction or elimination of responsiveness to androgen-mediated inhibition and/or prevention of cell death. Although various embodiments of the invention are disclosed herein, many adaptations and modifications may be made within the scope of the invention in accordance with the common general knowledge of those skilled in this art. Such modifications include the substitution of known equivalents for any aspect of the invention in order to achieve the same result in substantially the same way. Numeric ranges are inclusive of the numbers defining the range. In the claims, the word "comprising" is used as an open-ended term, substantially equivalent to the phrase "including, but not limited to". The following examples are illustrative of various aspects of the invention, and do not limit the broad aspects of the invention as disclosed herein.
EXAMPLES
Example 1: Demonstration of androgen-mediated and estrogen-mediated neuroprotection against serum- deprivation-mediated apoptosis of human primary neurons. Estrogen is hypothesized to play an important role against Alzheimer's disease in women (Birge 1996). Women receiving HRT are less susceptible to Alzheimer's disease
(Tang et al . 1996). As HRT often contains androgens (Gelfand 1992) and men's susceptibility to Alzheimer's disease increases with age in parallel with reduced levels of androgens (Vermeulen 1991) . Applicants have investigated the role of testosterone enanthate, methyltestosterone and epitestosterone in neuroprotection against serum deprivation in primary cultures of human neurons.
It has been shown that these human neurons undergo a protracted form of cell death with active recombinant caspases (Zhang et al . 2000). Serum deprivation also induces a protracted cell death but there is a significant amount of neuronal cell death by serum deprivation within 24 h (p < 0.005) (Fig. 2a). The addition of physiological concentrations of 4 nM testosterone enanthate to serum-deprived neurons eliminates apoptosis completely for 24 and 48 h and significantly reduces apoptosis at 72 and 96 h of treatment. In contrast, methyltestosterone did not significantly inhibit apoptosis at 24 h, but did show a 20% reduction in apoptosis between 48 and 96 h of treatment (p < 0.05). The anti-androgen, epitestosterone also had no statistically significant effect at 24 h but reduced apoptosis by 20-40% from 48 to 96 h of treatment (p > 0.05). As shown (Behl et al . 1997; Green et al . 1997; Pike 1999; Green and Simpkins 2000), 2 nM
physiological concentrations of 17-β- (p < 0.005) and 17-
α-estradiol (p < 0.05) were also neuroprotective from 24 to 96 h of treatment.
Addition of 10 times less or between 10 and 100 times more hormone to serum-deprived neurons shows that the lower 0.2 or 0.4 nM concentration are slightly more neuroprotective than the 2 and 4 nM concentration (Fig. 2b) . Increasing the levels of hormone slightly reduced
the neuroprotective effect in 17-α-estradiol and androgen treated cultures. These results indicate that the neuroprotective effect of androgens and estrogens at physiological concentrations is receptor mediated and rules out a possible antioxidant function in neuroprotection. It is possible that the higher concentrations of hormones are slightly toxic to neurons thereby reducing the neuroprotective effect. Most importantly, the significant neuroprotective effect observed with the lowest dose of hormone indicates the strong neuropotency of these hormones. Example 2: Demonstration of direct androgen-mediated protection of neurons without aromatization into estrogens
To determine if the neuroprotective role of androgens was directly through androgen receptors or through aromatization into estrogens (Balthazart and Ball 1998), applicants first determined if androgen receptors were present in the cultures studied using non- aromatizable [3H]mibolerone in a binding assay. Results described herein show that androgen receptors are present at 8 ± 2 fmoles/mg protein. The effect of the non- aromatizable androgen, mibolerone, on neuroprotection was assessed (Fig. 3) . The results herein show that similar to testosterone, mibolerone significantly protects neurons against serum deprivation even after 96 h of serum deprivation. The neuroprotective effect of mibolerone is not as strong as that of testosterone. Similarly, methyltestosterone also shows neuroprotection at a lower level than in testosterone. Comparison of the chemical structure of these compounds (Fig. 1) indicates that the presence of the 17-methyl group decreases the neuropotency of androgens. The addition of mibolerone to testosterone, epitestosterone or methyltestosterone does not increase neuroprotection in cells treated with hormone in absence of mibolerone suggesting that both mibolerone and natural androgens act through the same receptor. The affinity of mibolerone is at least 100- fold higher than testosterone, epitestosterone or methyltestosterone (Wilson and French 1976; Turcotte et al . , 1988). Therefore, mibolerone binding to the androgen receptor would compete out the other androgens as evidenced by the levels of neuroprotection consistent with a mibolerone-specific effect. Together, these results indicate that androgens can be directly neuroprotective without being aromatized to estrogen.
To confirm that testosterone is not acting by aromatization into estrogen, a cell permeable aromatase inhibitor, 4-androsten-4-OL-3, 17-dione, was added to the testosterone-treated neurons (Fig. 4). The results show that the aromatase inhibitor does not have a significant effect on neuronal cell death by serum deprivation (p > 0.8). In the presence of testosterone, 4-androsten-4-OL- 3, 17-dione, does not prevent testosterone-mediated neuronal protection (p > 0.4). These results confirm the direct action of testosterone rather than an indirect effect through aromatization into estrogens.
To determine if activation of the androgen receptor results in the neuroprotective action of testosterone, the effect of a non-steroid pure anti-androgen, flutamide (Simard et al . 1986) on testosterone-mediated neuroprotection was assessed (Fig. 5) . Flutamide alone
at either 2 or 20 μM does not have a significant effect on neuronal survival or cell death. However, flutamide significantly abolishes testosterone-mediated neuroprotection. Together with the mibolerone and aromatase inhibitor studies, these results strongly suggest that the neuroprotective function of testosterone occurs through the androgen receptor. Flutamide also significantly inhibits methyltestosterone-mediated neuroprotection. The effect is not as significant as seen with testosterone. However, methyltestosterone is also less neuroprotective than testosterone either because of the 17-methyl group as discussed later or the lower affinity of methyltestosterone for the androgen receptor (Wiita et al . 1995). Alternatively, the slow neuroprotective effect of methyltestosterone, which is only observed after 48 h of treatment, indicates that metabolites of methyltestosterone may be produced over time and promote neuroprotection through both androgen receptor dependent (antagonized by flutamide) and androgen receptor independent (not antagonized by flutamide) mechanisms.
In contrast, flutamide could not inhibit the endogenous anti-androgen, epitestosterone-mediated neuroprotection indicating that ■ the neuroprotection of epitestosterone is independent of androgen receptors.
Together, these results confirm that the neuroprotective effect of testosterone depends on an interaction with androgen receptors and can be competed out with antagonists .
Example 3: Neuronal culture
Human fetal brain tissue (12-16 weeks) was obtained in accordance with the guidelines established by the Medical Research Council and approved by the Institutional Review Board of McGill University. Neurons were isolated and cultured as previously described (LeBlanc 1995) . To summarize, brain tissue was minced in phosphate buffered saline and dissociated with 0.25% trypsin (Gibco-BRL, Rockville, MD, USA) . The cells were subsequently treated with 10% serum and 0.1 mg/ml deoxyribonuclease I (Roche Molecular Biochemical, Indianapolis, IN, USA) and the resulting homogenate
filtered through 130- and 170- μm nylon mesh. The neurons were plated at 3 x 106 cells/mL on poly-L-lysine
(Sigma Chemicals, St Louis, MO, USA) coated ACLAR™ (33C; 5 mm; Allied Chemical Corp.) coverslips and cultured in vitro for 10 days. The media contains phenol-free minimal essential media in Earle's balanced salt solution containing 0.225% sodium bicarbonate, 1 mM sodium pyruvate, 2 mM L-glutamine, 0.1% dextrose, I x antibiotic
Pen-Strep (all products from Gibco-BRL) and 5% decomplemented fetal bovine serum (HyClone, Logan. UT,
USA) . In complete serum containing media, the basal amount of testosterone is present at 9 pM and estrogen is at 18 pM.
Example 4: Neuronal treatment
The neurons were serum-deprived in the absence or
presence of 2 nM 17-α-estradiol or I7-β-estradiol, 4 nM testosterone enanthate, epitestosterone or methyltestosterone. (The concentration represents peak physiological levels in reproductive age women and men.) All hormones were purchased from Sigma (St Louis, MO, USA) except methyltestosterone obtained from USP. Testosterone enanthate was used because ester increases the duration and action of testosterone. Testosterone enanthate will not bind the androgen receptor unless the ester is hydrolyzed. The testosterone enanthate is hydrolyzed into testosterone in the neuronal cultures as evidenced by the antagonistic effect of flutamide. The chemical structure of these compounds is shown in Fig. 1. The media was changed every 48 h. Hormones were dissolved in various stock concentrations in 100% ethanol and added to the media to give final concentrations of 2 and 4 nM or the indicated dose with equivalent amounts of ethanol. Control serum-deprived neurons receive the equivalent amount of ethanol. Similarly, mibolerone, flutamide and aromatase inhibitor were dissolved in ethanol and added to the media to give final concentrations of 3 nM mibolerone (DuPont NEN, Boston.
MA, USA), 2 μM and 20 μM flutamide (Sigma, St Louis, MO, USA) , or 5 ng/mL and 50 ng/mL 4-androsten-4-OL-3, 17- dione (Sigma. St Louis, MO, USA) . At the end of the treatment, coverslips were fixed with 4% paraformaldehyde, 4% sucrose in phosphate buffered solution (Harlow and Lane 1988).
Example 5: Determination of androgen receptors
Androgen receptors were identified by incubating 6 nM [3H] mibolerone (DuPont NEN, Boston, MA. USA; Spec. Act. 85 Ci/mmol) with 6 x 106 neurons to measure total binding. Non-specific binding was assessed by competing the binding of [3H] -mibolerone with a 200-fold excess cold mibolerone as previously described (Kaufman et al . 1993). Specific binding was determined by subtracting non- specific from total binding and dividing by the protein concentration as determined by the Lowry assay (Lowry et al . 1951) .
Example 6: Determination of neuronal cell death by TUNEL
Fixed neurons were pemeabilized with 0.1% Triton X- 100 in 0.1% sodium citrate. Cell death was detected by TUNEL (TdT-mediated dUTP nick-end labeling) using the Cell Death Kit I (Roche Molecular Biochemicals,
Indianapolis, IN. USA) as described by the manufacturer. All cells were counterstained with 100 ng/mL propidium iodide (Pharmingen) to allow confirmation of the apoptotic morphology of the cells and to detect the total number of cells present under fluorescence microscopy. The percentage of neuronal cell death was determined by screening five areas of each coverslip (a minimum of 500 cells) and comparing the total number of TUNEL-positive (green fluorescence) and morphologically apoptotic cells over the total number of cells (red fluorescence) present in each sample. The neuronal cell death was confirmed in representative experiments of each assay with MTT reducing assays. Briefly, neurons were plated directly into 24-well plates and studied for MTT reduction using the Cell Proliferation Kit I (MTT) as described by the manufacturer (Roche Molecular Biochemicals, Indianapolis,
IN, USA; data not shown). A two-tailed Student's t-test for unpaired samples was used for comparison between the level of neuronal cell death in serum-deprived neurons and that in serum-deprived neurons treated with hormones, p-values of <0. 05 were used as indicative of statistical significance.
Example 7 : Aβι-42 -mediated neurotoxicity is protected by steroid hormones.
As presented herein, applicants have demonstrated that testosterone and estradiol protect neurons against serum deprivation-mediated apoptosis. In this study applicants have further tested if any of the androgens and estrogens could prevent Aβι_2 -mediated neurotoxicity.
Protocol :
A toxic dose of Aβx_2 and Dextran Texas Red fluorescent dye marker was microinjected into neurons and neurons were immediately treated with varying doses of 17-β-estradiol, transcriptionally inactive 17-β-estradiol analogue, testosterone enanthate, methyltestosterone and epitestosterone. Microinjected neurons were incubated for 24 hours, fixed and submitted to TUNEL analysis to assess cell death by Aβι_42. The experiment was done on 100 microinjected cells/well x 2 wells/experiments in three independent neuron preparations.
As shown in Figure 6, Aβι_42 induces TUNEL-positive cell death in 60-70% of neurons within 24 hours of injection. A statistically significant difference between the various groups is observed by ANOVA analysis of the data (p<0.0001, df=15) . Post-hoc analysis with Dunnett's test shows that a physiological concentration of 2 nM of 17-β-estradiol and methyltestosterone prevent Aβι-42 mediated neuronal cell death by 50% (p<0.01).
Testosterone is less but still neuroprotective (p<0.05 for 2 and 4 nM and p<0.01 at 10 nM) . In contrast, epitestosterone and 17-β-estradiol do not protect against Aβι_42 -mediated apoptosis.
Figure 7 illustrates further results demonstrating that estrogen and androgens protect human neurons against
intracellular Aβι_42-mediated toxicity in primary cultures.
Neurons were microinjected with a lethal dose of Aβι-42 and
immediately treated with 2, 4, or 10 nM 17-β-estradiol
(βE2) , 17-α-estradiol (αE2), testosterone enanthate (Test), epitestosterone (Epi-Test), methyl testosterone
(Methyl-test) or cell impermeable BSA conjugated-7-β-
estradiol (BSA-βE2) . Neuronal cell death was assessed by TUNEL 24 hours after treatment. % cell death represents the number of TUNEL positive neurons over total neurons
(stained with DAPI) . Each sample represents 200 microinjected cells from each of a minimum of three experiments done on different neuron preparations.
*p<0.01 relative to Aβι-42-microinjected cells in absence of hormones.
Figure 8 illustrates further results obtained similarly to those described for figure 7, but in this case the neurons were pre-incubated with 10 nM hormones
for 1 hour prior to microinjection with Aβι-42. Treatment with hormones was continued for 24 hours before measuring cell death as above. These results indicate a that these hormones may be highly neuroprotective, particularly when administered prior to Aβι_42 treatment.
Figure 9 illustrates results demonstrating that estrogens and androgens protect through their respective receptors. The neurons were pre-treated with either hormone or hormone and antagonist (tamoxifen for estrogens and flutamide for androgens) for one hour and
microinjected with the Aβι_2. Incubation of 24 hours followed by cell death measures was performed as described above. * p<0.01.
Applicants therefore conclude that both physiological concentrations of androgens and estrogen can protect against intracellular Aβx_42 neurotoxicity. The role of estrogen against extracellular Aβ toxicity has previously been reported (Bonnefont et al . , 1998; Goodman et al . , 1996; Green et al . , 1996; Gridley et al . , 1997; Gridley et al . , 1998; Gursoy et al . , 2001; Keller et al . ,
1997; Kim et al . , 2001; Mook-Jung et al . , 1997; Svensson and Nordberg, 1999; Thomas et al . , 2001). However, prior to applicants' studies presented herein, it has not been shown that either hormone protects against "intracellular" Aβ-mediated neurotoxicity, nor has the effect of androgens against Aβ-mediated toxicity been shown. Further, applicants have demonstrated nearly complete protection, particularly in cases where hormone was added prior to Aβ introduction. Competition studies described herein with specific antagonists demonstrate that the protective activity of estrogens and androgens is manifested through their respective receptors.
All references cited above or in the References section below are herein incorporated by reference.
REFERENCES
Balthazart J. and Ball G. F. (1998) New Insights into the regulation and function of brain estrogen synthase (aromatase). Trends Neurosci . 21,. 243-249.
Behl C, Skutella T., Lezoualc'h F. , Post A., Widmann M. , Newton C. and Holsboer F. (1997) Neuroprotection against oxidative stress by estrogens: structure- activity relationship. Mol . . Pharmacol . 51, 535-541. Birge S. (1996) Is there a role for estrogen replacement therapy In the prevention and treatment of dementia? JAGS 44, 865-870. Bonnefant, A.B., Munoz, F.J. and Inestrosa, N.C. (1998) Estrogen protects neuronal cells from the cytotoxicity induced by acetylcholinesterase-amyloid complexes. FEBS Lett, 441, 220-224. Brinton R. D., Tran J. , Proffitt P. and Montoya M. (1997)
17 β-estradiol enhances the outgrowth and survival of neocortical neurons in culture. Neurochem . Res . 22, 1339-1351.
Brogden R. N. and Chrisp P. (1991) Flutamide. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in advanced prostatic cancer. Drugs Aging 1, 104-115. Clancy A. N., Bonsall R. W. and Micheal R. P. (1992)
Immunocytochemical labeling of androgen receptors in the brain of rat and monkey. Life Sci . 50, 409-417.
D'Andrea, M.R., R.G. Nagele, H.Y. Wang, P.A. Peterson, and D.H. Lee (2001) Evidence that neurones accumulating amyloid can undergo lysis to form amyloid plaques in Alzheimer's disease. Histopa thology. 38:120-34.
Davis S. (1999) Androgen replacement in women: a commentary. J. Clin . Endocrinol . Metabol . 84, 1886- 1891.
Dodson R. and Gorski R. A. (1993) . Testosterone propionate administration prevents the loss of neurons within the central part of the medial preoptic nucleus. J. Neurobiol . 24, 80-88.
Dubai D., Shughrue P., Wilson M., Merchenthaler I. and
Wise P. (1999) Estradiol modulates Bel' 2 in cerebral ischemia: a potential role for estrogen receptors. J. Neurosci . 19, 6385-6393. Finkelstein M., Weidenfeld J. , Ne'eman Y., Samuni A., Mizrachi Y. and Ben-Uzilio R. (1981) Comparative studies of the aromatization of testosterone and epitestosierone by human placental aromatase. Endocrinology 108, 943-947. Finley S. K. and Kritzer M. F. (1999) Immunoreactivity for intracellular androgen receptors in identified subpopulations of neurons, astrocytes and oligodendrocytes in primate prefrontal cortex. J. Neurobiol . 40, 446-457.
Flood J. F., Farr S. A., Kaiser F. E., La Regina M. and
Morley J. E. (1995) Age-related decrease of plasma testosterone in SAMPS Mice: Replacement improves age-related Impairment of learning and memory. Physiol . Behav 57. 669673.
Gelfand M. (1992) Estrogen-androgen hormone replacement therapy. In: Hormone replacement therapy. (Swartz
D. , ed. ) Williams & Wilkins, Baltimore. 221-234.
Goodman, Y., Bruce, A.J., Cheng, B. and Mattson, M.P. (1996) Estrogens attenuate and corticosterone exacerbates excitotoxicity, oxidative injury, and amyloid beta-peptide toxicity in hippocampal neurons. J Neurochem, 66, 1836-1844.
Goodman Y. and Mattson M. P. (1996) Ceramide protects hippocampal neurons against excitotoxic and oxidative insults, and amyloid-peptide toxicity. J.
Neurochem . 66, 869-872.
Gouras, G.K., J. Tsai, J. Naslund, B. Vincent, M. Edgar,
F. Checler, J.P. Greenfield, V. Haroutunian, J.D. Buxbaum, H. Xu, P. Greengard, and N.R. Relkin (2000) Intraneuronal Abeta42 accumulation in human brain.
Am J Pa thol . 156: 15-20.Gouras G. K., Xu H., Gross R. S., Greenfield J. P., Hai B., Wang R. and Greengard P. (2000) Testosterone reduces neuronal secretion of
Alhzeimer's β-amyloid peptides. Proc . Na tl . Acad. Sci . USA 97, 1202-1205. Green, P.S., Gridley, K.E. and Simpkins, J.W. (1996) Estradiol protects against beta-amyloid (25-35)- induced toxicity in SK-N-SH human neuroblastoma cells. Neurosci Lett, 218, 165-168.
Green P. S. and Simpkins J. W. (2000) Neuroprotective effects of estrogens: potential mechanisms of action. Int . J. Dev Neurosci 18, 347-358. Green P. S., Bishop J. and Simpkins J. W. (1997) 17 alpha-estradiol exerts neuroprotective effects on SK-N-SH cells. J. Neurosci 17, 511-515. Gridley, K.E., Green, P.S. and Simpkins, J.W. (1997) Low concentrations of estradiol reduce beta-amyloid (25- 35) -induced toxicity, lipid peroxidation and glucose utilization in human SK-N-SH neuroblastoma cells. Brain Res, 778, 158-165. Gridley, K.E., Green, P.S. and Simpkins, J.W. (1998) A novel, synergistic interaction between 17 beta- estradiol and glutathione in the protection of neurons against beta-amyloid 25-35-induced toxicity in vitro. Mol Pharmacol , 54, 874-880. Gursoy, E., Cardounel, A. and Kalimi, M. (2001) The environmental estrogenic compound bisphenol A exerts estrogenic effects on mouse hippocampal (HT-22) cells: neuroprotection against glutamate and amyloid beta protein toxicity. Neurochem Int, 38, 181-186. Gyure, K.A. , R. Durham, W.F. Stewart, J.E. Smialek, and J.C. Troncoso (2001) Intraneuronal Ab-amyloid precedes development of amyloid plaques in Down syndrome. Arch Pa thol Lab Med. 125:489-92. Harlow E. and Lane 0. (1988) Immunoprecipitations .
Antibodies. A laboratory manual. Cold Spring Harbor Labora tories . New York, 421-466. Jaffe A. B., Toran-Allerand C. 0., Greenyard P. and Gandy S. E. (1994) Estrogen regulates metabolism of
Alzheimer amyloid β precursor protein. J. Biol . Chem . 269, 13065-13068. Jorm A. F., Korten A. E. and Henderson A. S. (1987) The Prevalence of dementia: a quantitative integration of the literature. Acta Psychia tr Scand. 76, 465- 479. Kaufman M. , Pinsky L., Trifiro M. , Lumbroso R. ,
Sabbaghian N. and Gottlieb B. (1993) Kinetic evidence for a unique testosterone-receptor complex in 5α-reductase sufficient genital skin fibroblasts
and the effects of 5α-reductase deficiency on its formation. J. Steroid Biochem . Molec . Biol . 45, 467- 476. Keller, J.N., Germeyer, A., Begley, J.G. and Mattson, M.P. (1997) 17Beta-estradiol attenuates oxidative impairment of synaptic Na+/K+-ATPase activity, glucose transport, and glutamate transport induced by amyloid beta-peptide and iron. J Neurosci Res, 50, 522-530.
Kim, H., Bang, O.Y., Jung, M.W., Ha, S.D., Hong, H.S., Huh, K., Kim, S.U. and Mook-Jung, I. (2001) Neuroprotective effects of estrogen against beta- amyloid toxicity are mediated by estrogen receptors in cultured neuronal cells. Neurosci Lett, 302, 58- 62. Labrie F. (1993) Mechanism of action and pure antiandrogenic properties of flutamide. Cancer 72, 3816-3827. LeBlanc A. C. (1995) Increased production of 4 kDa
amyloid β-peptide in serum deprived human primary neuron cultures: possible involvement of apoptosis. J. Neurosci . 15, 7837-7846. Lowry 0. H., Rosebrough N. J., Farr A. L. and Randall R.
J. (1951) Protein replacement with the Folin phenol reagent. J. Biol . Chem . 193, 265-275.
McEwen B. S. and Woolley C. S. (1994) Estradiol and progesterone regulate neuronal structure and synaptic connectivity in adult as well as developing brain. Exp Gerontol . 29, 431-436.
McEwen B. S., Tanapat P. and Weiland N. G. (1999)
Inhibition of dendritic spine induction on hippocampal CAl pyramidal neurons by a nonsteroidal estrogen antagonist in female rats. Endocrinology
140, 1044-1047.
Maggi R., Poletti A., Casulari L.A., Pimpinelli F. , Piva
F., Zanisi M. R. and Martini L. (1998) Effects and metabolism of steroid hormones in human neuroblastoma cells. Steroids 63, 257-262.
Markiewicz L. and Gurpide E. (1997) Estrogenic and progestagenic activities of physiologic and synthetic androgens, as measured by in vitro bioassays. Meth Find Exp. Clin . Pharmacol . 19, 215-
222.
Molsa P. K.. Marttila R. J. and Rinne U. K. (1982)
Epidemiology of dementia in a Finnish population.
Acta Neurol . Scand. 65, 541-552. Mook-Jung, I., Joo, I., Sohn, S., Kwon, H.J., Huh, K. and
Jung, M.W. (1997) Estrogen blocks neurotoxic effects of beta-amyloid (1-42) and induces neurite extension on B103 cells. Neurosci Lett, 235, 101-104. Moosmann B. and Behl C. (1999) The antioxidant neuroprotective effects of estrogens and phenolic compounds are independent from their estrogenic properties. Proc. Na tl Acad. Sci USA 96, 8867-8872.
Mulnard R. A., Cotman C. W., Kawas C, van Dyck C. H., Sano M., Doody R. , Koss E, Pfeiffer E., Jin S.,
Gamst A., Grundman M., Thomas R. and Thai L. (2000)
Estrogen replacement therapy for treatment of mild to moderate Alzheimer Disease. JAMA 283, 1007-1015.
Namer M. (1988) Clinical applications of antiandrogens. J. Steroid Biochem . . 31, 719-729.
Naslund, J. , V. Haroutunian, R. Mohs, K.L. Davis, P.
Davies, P. Greengard, and J.D. Buxbaum (2000)
Correlation between elevated levels of amyloid beta- peptide in the brain and cognitive decline. Jama . 283:1571-7.
Nuck B. A. and Lucky A. W. (1987) Epitestosterone: a potential new antiandrogen. J. Invest . Derma tol . 89.
209-211.
Paganini-Hill A. (1996) Oestrogen replacement therapy and Alzheimer's disease. Br J. Obster. Gyn . 103, 80-86. Pike C. (1999) Estrogen Modulates Neuronal Bcl-x
expression and β-Amyloid-lnduced Apoptosis: Relevance to Alzheimer's Disease. J. Neurochem . 72, 1552-1563. Puy L., MacLusky N. J. , Becker L. , Karsan N., Trachtenberg J. and Brown T. J. (1995) Immunocytochemical detection of androgen receptor in human temporal cortex: characterization and application of polyclonal androgen receptor antibodies in frozen and paraffin-embedded tissues. J. Steroid Biochem . Molec . Biol . 55, 197-209. Rako S. (1998) Testosterone deficiency: a key factor in the increased cardiovascular risk to women following hysterectomy or with natural aging. J. Women 's Heal th 7, 825-829.
Schneider L. S., Farlow M. R. , Henderson V. W. and Pogoda J. M. (1996) Effects of estrogen replacement therapy on response to tacrine in patients with Alzheimers disease. Neurology 46, 1580-1584. Schneider L. S., Farlow M. R. and Pogoda J. M. (1997) Potential Role for Estrogen Replacement in the Treatment of Alzheimer's Dementia. Ant. J. Med. 103, 46S-50S. Selkoe, D.J. (1998) The cell biology of βamyloid precursor protein and presenilin in Alzheimer's
Disease. Trends in Cell Biology 8, 447-453.
Simard J. , Luthy I., Guay J. , Belanger A. and Labrie F. (1986) Characteristics of interaction of the antiandrogen fiutamide with the androgen receptor in various target tissues. Mol . Cell . Endocrinol . 44, 261-270. Simpkins J. W., Green P. S., Gridley K. E., Singh M. , de Fiebre N. C. and Rajakumar G. (1997) The role of estrogen replacement therapy in memory enhancement and the prevention of neuronal loss associated with Alzheimer's. Am . J. Med. 103, 19S-25S. Singh M., Setalo G. Jr., Guan X., Warren M. and Toran-
Allerand C. D. (1999) Estrogen-induced activation of mitogen-activated protein kinase in cerebral cortical explants: convergence of estrogen and neurotrophin signaling pathways. J. Neurosci 19, 1179-1188.
Singh S. M., Gauthier S. and Labrie F. (2000b) Androgen receptor antagonists (antiandrogens) : structure- activity relationships. Curr. Med. Chem . 1 , 211-247. Singh M., Setalo G., Guan X., Frail D. E. and Toran- Allerand D. (2000a) Estrogen-induced activation of the mitogen-activated protein kinase cascade in the
cerebral cortex of estrogen receptor-α knock out mice. J. Neurosci . . 20, 1694-1700. Starka L., Bicikova M. and Hampl R. (1989) Epitestosterone - an endogenous antiandrogen? J. Steroid Biochem . 33, 1019-1021. Starka L., Hampl R., Bicikova M., Jelinek R. and Doskocil M. (1991) Observations on the biological activity of epitestosterone. Physiol . Res . 40, 317-326. Sternbach H. (1998) Age-associated testosterone decline in men: clinical issues for psychiatry. Am . J. Psychia try 155, 1310-1318. Svensson, A.L. and Nordberg, A. (1999) Beta-estradiol attenuate amyloid beta-peptide toxicity via nicotinic receptors. Neuroreport, 10, 3485-3489.
Tang M. X., Jacobs D., Stern Y., Marder K. , Schofield P., Gurland B., Andrews H. and Mayeux R. (1996) Effect of oestrogen during menopause on risk and age of onset of Alzheimer's disease. Lancet 348, 429-432. Thomas, T., Bryant, M., Clark, L., Garces, A. and Rhodin, J. (2001) Estrogen and raloxifene activities on amyloid-beta-induced inflammatory reaction. Microvasc Res, 61, 28-39. Tirassa P., Thiblin I., Agren G., Vigneti E., Aloe L. and Stenfors C. (1997) High-dose anabolic steroids modulate concentration of nerve growth factor and expression of its low affinity receptor (p75-NGFr) in male rat brain. J. Neurosci . Res . 47, 198-207. Tohgi H., Utsugisawa K., Yamagata M. and Yoshimura M. (1995) Effects of age on messenger RNA expression of glucocorticoid, thyroid hormone, androgen. and estrogen receptors in postmortem human hippocampus.
Brain Res . 700, 245-253. Toran-Allerand C. D, (2000a) Estrogen as a treatment for Alzheimer disease. JAMA 284, 307-308.
Toran-Allerand C. D. (2000b) Novel sites and mechanisms of oestrogen action in the brain. Novartis Found.
Symp. 230, 56-69. discussion 69-73. Traish A. M., Muller R. E. and Wotiz H. H. (1986) Binding of 7 alpha 17 alpha-dimethyl-19-nortestosterone
(mibolerone) to androgen and progesterone receptors in human and animal tissues. Endocrinology 118,
1327-1333. Turcotte G., Chapdelaine A., Roberts K. 0. and Chevalier S. (1988) Androgen binding as evidenced by a whole cell assay system using cultured canine prostatic epithelial cells. J. Steroid Biochem . 29, 69-76. Vermeulen A. (1991) Androgens in the aging male. J. Clin .
Endocrinol . Metabolism . 73, 221-224. Wade S. B., Oommen P., Conner W, C, Earnest 0. J. and
Miranda R. C. (1999) Overlapping and divergent actions of estrogen and the neurotrophins on cell fate and p53-dependent signal transduction in conditionally immortalized cerebral cortical neuroblasts. J. Neurosci 19, 6994-7006.
Wiita B., Artis A., Ackerman 0. M. and Longcope C. (1995)
Binding of 17-alpha-methyltestosterone in vitro to human sex hormone binding globulin and rat ventral prostate androgen receptors. Ther. Drug Moni t . 17,
377-380.
Wilson E. M. and French F. S. (1976) Binding properties of androgen receptors. Evidence for identical receptors in rat testis, epididymis, and prostate. J. Biol . Chem . 251, 5620-5629.
Woolley C. S. (1999) Effects of estrogen in the CNS. Curr
Opin . Neurobiol . 9, 349-354.
Woolley C. S. and McEwen B. S. (1993) Roles of estradiol and progesterone in regulation of hippocampal dendritic spine density during the estrous cycle in the rat. J. Comp . Neurol . 336, 293-306.
Woolley C. S. and McEwen B. S. (1994) Estradiol regulates hippocampal dendritic spine density via an N-methyl-
D-aspartate receptor-dependent mechanism. J. Neurosci . 14, 7680-7687. Woolley C. S., Weiland N. G., McEwen B. S. and
Schwartzkroin P. A. (1997) Estradiol increases the sensitivity of hippocampal CAl pyramidal cells to
NMDA receptor-mediated synaptic input: correlation with dendritic spine density. J. Neurosci . 17, 1848-
1859.
Wright A. F. , Goedert M. and Hastie N. 0. (1991) Familial
Alzheimer's disease - Beta-amyloid resurrected.
Na ture 349, 653-654. Xu H., Gouras G., Greenfield J. , Voncent B., Nalsund J.. Mazzarelli L., Fried G., Jovanovic J. , Seeger M., Relkin N., Liao F. , Checler F., Buxbaum J., Chait B., Thinakaran G., Sisodia S., Wang R., Greengard P. and Gandy S. (1998) Estrogen reduces neuronal
generation of Alzheimer β-amyloid peptides. Na t . Med. 4, 447-451. Zhang Y., Goodyer C. and LeBlanc A. (2000) Selective and protracted apoptosis in human primary neurons microinjected with active caspase-3, 6, 7 and 8. J. Neurosci . 20, 8384-8389.

Claims

WHAT IS CLAIMED IS:
1. A method of preventing or treating a neural disease in an animal, said method comprising administering an androgen or androgen-related compound to said animal.
The method of claim 1 wherein said animal is a mammal ,
The method of claim 2, wherein said mammal is a human.
The method of claim 1, wherein said animal is male,
5. The method of claim 1, wherein said neural disease is selected from the group consisting of Alzheimer disease, Parkinson's disease, amyotropic lateral sclerosis (ALS) , cerebellar degeneration, ischemia (stroke) , traumatic injuries, prion diseases, Huntington disease, frontal lobe dementia, vascular dementia, infection related dementia, head injury, hereditary cerebral amyloidogenesis, Down's Syndrome and cerebral hemorrhage.
6. The method of claim 1, wherein said androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone.
7. The method of claim 6, wherein said androgen is testosterone.
8. The method of claim 1, wherein said androgen- related compound is a testosterone ester.
9. The method of claim 8, wherein said testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate.
10. A composition for preventing or treating a neural disease in an animal, said composition comprising an androgen or androgen-related compound in admixture with a pharmaceutically suitable carrier.
11. Use of an androgen or androgen-related compound or the composition of claim 10 for preventing or treating a neural disease in an animal.
12. The use of claim 11, wherein said neural disease is selected from the group consisting of Alzheimer disease, Parkinson's disease, amyotropic lateral sclerosis (ALS) , cerebellar degeneration, ischemia (stroke) , traumatic injuries, prion diseases, Huntington disease, frontal lobe dementia, vascular dementia, infection related dementia, head injury, hereditary cerebral amyloidogenesis, Down's Syndrome and cerebral hemorrhage.
13. The use of claim 11, wherein said androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone.
14. The use of claim 13, wherein said androgen is testosterone .
15. The use of claim 11, wherein said androgen- related compound is a testosterone ester.
16. The use of claim 15, wherein said testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate.
17. Use of an androgen or androgen-related compound for preparation of a medicament for preventing or treating a neural disease in an animal.
18. A commercial package comprising an androgen or androgen-related compound together with instructions for the prevention or treatment of a neural disease.
19. A commercial package comprising the composition of claim 10 together with instructions for the prevention or treatment of a neural disease.
20. The commercial package of claim 18, wherein said neural disease is selected from the group consisting of Alzheimer disease, Parkinson's disease, amyotropic lateral sclerosis (ALS) , cerebellar degeneration, ischemia (stroke) , traumatic injuries, prion diseases, Huntington disease, frontal lobe dementia, vascular dementia, infection related dementia, head injury, hereditary cerebral amyloidogenesis, Down's Syndrome and cerebral hemorrhage.
21. The commercial package of claim 18, wherein said androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone .
22. The commercial package of claim 21, wherein said androgen is testosterone.
23. The commercial package of claim 18, wherein said androgen-related compound is a testosterone ester.
24. The commercial package of claim 23, wherein said testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate .
25. A method for preventing or inhibiting cell death, comprising treating a cell with an androgen or androgen-related compound.
26. The method of claim 25, wherein said cell is a human cell.
27. The method of claim 25, wherein said cell is a neural cell.
28. The method of claim 25, wherein said androgen is selected from the group consisting of testosterone, mibolerone, methyltestosterone and epitestosterone.
29. The method of claim 28, wherein said androgen is testosterone.
30. The method of claim 25, wherein said androgen- related compound is a testosterone ester.
31. The method of claim 30, wherein said testosterone ester is selected from the group consisting of testosterone enanthate and testosterone propionate.
32. A method of identifying or characterizing a test compound for prevention and/or treatment of neural or neurodegenerative disease, said method comprising: contacting said test compound with a cell comprising an androgen receptor; - measuring a test level of androgen- associated activity; and comparing said test level of androgen-associated activity with a corresponding control level of androgen- associated activity in a corresponding cell which was not exposed to said test compound; wherein a difference between said test level and said control level indicates that the test compound may be used for prevention and/or treatment of neural or neurodegenerative disease.
33. The method of claim 32, wherein said cell is a neural cell.
34. The method of claim 32, wherein said androgen- associated activity is an inhibition or prevention of cell death or apoptosis.
35. A method of identifying and/or characterizing a mechanism and/or component associated with androgen- mediated effects on cell death, said method comprising: assessing a first alteration in a cell death- associated phenotype between an androgen treated cell and a corresponding untreated cell; and comparing said alteration with a corresponding second alteration assessed in corresponding androgen treated versus untreated mutant cells; wherein differences in said first and second alterations are used to identify and/or characterize a mechanism and/or component associated with androgen-mediated effects on cell death.
PCT/CA2002/000668 2001-05-01 2002-04-30 Androgen-mediated neuroprotection and uses thereof WO2002087552A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002257430A AU2002257430A1 (en) 2001-05-01 2002-04-30 Androgen-mediated neuroprotection and uses thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CA2,345,864 2001-05-01
CA002345864A CA2345864A1 (en) 2001-05-01 2001-05-01 Androgen-mediated neuroprotection and its use for prevention, treatment and study of neural disease
US29403101P 2001-05-30 2001-05-30
US60/294,031 2001-05-30
US33056701P 2001-10-25 2001-10-25
US60/330,567 2001-10-25

Publications (2)

Publication Number Publication Date
WO2002087552A2 true WO2002087552A2 (en) 2002-11-07
WO2002087552A3 WO2002087552A3 (en) 2003-10-02

Family

ID=27171527

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2002/000668 WO2002087552A2 (en) 2001-05-01 2002-04-30 Androgen-mediated neuroprotection and uses thereof

Country Status (2)

Country Link
AU (1) AU2002257430A1 (en)
WO (1) WO2002087552A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2214488A1 (en) * 2007-11-06 2010-08-11 Ellis L. Kline Compositions and methods for treating parkinson's disease and related disorders
US7807396B2 (en) 2000-02-24 2010-10-05 University Of Kentucky Research Foundation Insulin degrading enzyme assays for treatment of alzheimer's disease
WO2011073419A1 (en) * 2009-12-18 2011-06-23 Institut National De La Sante Et De La Recherche Medicale (Inserm) New steroid inhibitors of pgp for use for inhibiting multidrug resistance
US9044503B2 (en) 2004-08-27 2015-06-02 University Of Kentucky Research Foundation Amyloid peptide inactivating enzyme to treat alzheimer's disease peripherally
WO2020247407A1 (en) * 2019-06-03 2020-12-10 Aihol Corporation Hyaluronan conjugates and uses thereof
WO2022204827A1 (en) * 2021-03-24 2022-10-06 兰州大学 Application of halcinonide and derivatives thereof for preparing drug treating and/or preventing cerebrovascular disease

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4791099A (en) * 1984-10-29 1988-12-13 Chaovanee Aroonsakul Method of treatment for central nervous system diseases such as Alzheimer's's disease
US5795883A (en) * 1993-04-13 1998-08-18 Jenapharm Gmbh Androgens and anabolic agents
WO2000010536A1 (en) * 1998-08-25 2000-03-02 Columbia Laboratories (Bermuda) Limited Extended release buccal bioadhesive tablet
WO2001072307A1 (en) * 2000-03-29 2001-10-04 The J. David Gladstone Institutes Methods of treating cognitive decline disease conditions with androgens

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH06100466A (en) * 1992-02-14 1994-04-12 Tsumura & Co Controlling agent for cerebroneurocytic organ lesion with stress nature

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4791099A (en) * 1984-10-29 1988-12-13 Chaovanee Aroonsakul Method of treatment for central nervous system diseases such as Alzheimer's's disease
US5795883A (en) * 1993-04-13 1998-08-18 Jenapharm Gmbh Androgens and anabolic agents
WO2000010536A1 (en) * 1998-08-25 2000-03-02 Columbia Laboratories (Bermuda) Limited Extended release buccal bioadhesive tablet
WO2001072307A1 (en) * 2000-03-29 2001-10-04 The J. David Gladstone Institutes Methods of treating cognitive decline disease conditions with androgens

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Section Ch, Week 199419 Derwent Publications Ltd., London, GB; Class B01, AN 1994-156568 XP002232575 & JP 06 100466 A (TSUMURA & CO), 12 April 1994 (1994-04-12) *
FINKELSTEIN M ET AL: "COMPARATIVE STUDIES OF THE AROMATIZATION OF TESTOSTERONE AND EPI TESTOSTERONE BY HUMAN PLACENTAL AROMATASE" ENDOCRINOLOGY, vol. 108, no. 3, 1981, pages 943-947, XP008014327 ISSN: 0013-7227 cited in the application *
GOURAS GUNNAR K ET AL: "Testosterone reduces neuronal secretion of Alzheimer's beta-amyloid peptides." PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES, vol. 97, no. 3, 1 February 2000 (2000-02-01), pages 1202-1205, XP002232570 Feb. 1, 2000 ISSN: 0027-8424 cited in the application *
GREEN PATTIE S ET AL: "17 alpha-Estradiol exerts neuroprotective effects on SK-N-SH cells." JOURNAL OF NEUROSCIENCE, vol. 17, no. 2, 1997, pages 511-515, XP002232574 ISSN: 0270-6474 cited in the application *
HAMMOND JENNIFER ET AL: "Testosterone-mediated neuroprotection through the androgen receptor in human primary neurons." JOURNAL OF NEUROCHEMISTRY, vol. 77, no. 5, June 2001 (2001-06), pages 1319-1326, XP002232572 ISSN: 0022-3042 *
NUÑEZ J L ET AL: "Androgens reduce cell death in the developing rat visual cortex." BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH. NETHERLANDS 29 DEC 2000, vol. 125, no. 1-2, 29 December 2000 (2000-12-29), pages 83-88, XP001148402 ISSN: 0165-3806 *
PAPASOZOMENOS S: "The heat shock-induced hyperphosphorylation of tau is estrogen-independent and prevented by androgens: Implications for Alzheimer disease." PROC. NATL. ACAD. SCI., vol. 94, 1997, pages 6612-6617, XP002232571 USA *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7807396B2 (en) 2000-02-24 2010-10-05 University Of Kentucky Research Foundation Insulin degrading enzyme assays for treatment of alzheimer's disease
US9044503B2 (en) 2004-08-27 2015-06-02 University Of Kentucky Research Foundation Amyloid peptide inactivating enzyme to treat alzheimer's disease peripherally
EP2214488A1 (en) * 2007-11-06 2010-08-11 Ellis L. Kline Compositions and methods for treating parkinson's disease and related disorders
EP2214488A4 (en) * 2007-11-06 2010-11-03 Ellis L Kline Compositions and methods for treating parkinson's disease and related disorders
AU2008325211B2 (en) * 2007-11-06 2014-11-13 Signal Coordinating Therapy, Inc. Compositions and methods for treating Parkinson's disease and related disorders
AU2015200707B2 (en) * 2007-11-06 2016-11-03 Signal Coordinating Therapy, Inc. Compositions and methods for treating Parkinson's disease and related disorders
WO2011073419A1 (en) * 2009-12-18 2011-06-23 Institut National De La Sante Et De La Recherche Medicale (Inserm) New steroid inhibitors of pgp for use for inhibiting multidrug resistance
WO2020247407A1 (en) * 2019-06-03 2020-12-10 Aihol Corporation Hyaluronan conjugates and uses thereof
WO2022204827A1 (en) * 2021-03-24 2022-10-06 兰州大学 Application of halcinonide and derivatives thereof for preparing drug treating and/or preventing cerebrovascular disease

Also Published As

Publication number Publication date
WO2002087552A3 (en) 2003-10-02
AU2002257430A1 (en) 2002-11-11

Similar Documents

Publication Publication Date Title
Hammond et al. Testosterone‐mediated neuroprotection through the androgen receptor in human primary neurons
US6812213B2 (en) Steroidal sapogenins and their derivatives for treating alzheimer&#39;s disease
Grove-Strawser et al. Membrane estrogen receptors activate the metabotropic glutamate receptors mGluR5 and mGluR3 to bidirectionally regulate CREB phosphorylation in female rat striatal neurons
Keller et al. 17β‐Estradiol attenuates oxidative impairment of synaptic Na+/K+‐ATPase activity, glucose transport, and glutamate transport induced by amyloid β‐peptide and iron
Callier et al. Neuroprotective properties of 17β‐estradiol, progesterone, and raloxifene in MPTP C57Bl/6 mice
US9234038B2 (en) Compositions and method for the diagnosis, prevention and treatment of alzheimer&#39;s disease
Capela et al. The neurotoxicity of hallucinogenic amphetamines in primary cultures of hippocampal neurons
Jourdain et al. Oestrogens prevent loss of dopamine transporter (DAT) and vesicular monoamine transporter (VMAT2) in substantia nigra of 1‐methyl‐4‐phenyl‐1, 2, 3, 6‐tetrahydropyridine mice
US20110312894A1 (en) Methods of diagnosing and treating neurodegenerative diseases
WO2002087552A2 (en) Androgen-mediated neuroprotection and uses thereof
US20090062245A1 (en) Modulation of Neurodegenerative Diseases Through The Progesterone Receptor
Ba et al. The role of Ca2+ channel modulation in the neuroprotective actions of estrogen in β-amyloid protein and 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) cytotoxic models
Zhang et al. Direct binding of estradiol enhances Slack (sequence like a calcium-activated potassium channel) channels’ activity
DE60214673T2 (en) METHOD FOR SCREENING COMPOUNDS OF ACTIVITY IN THE TREATMENT OF A BONE RISK ASSOCIATED WITH OSTEOCLASTS
CA2345864A1 (en) Androgen-mediated neuroprotection and its use for prevention, treatment and study of neural disease
US20060211673A1 (en) Modulation of neurodegenerative diseases through the estrogen receptor
WO2005068501A1 (en) Methods and compositions for modulating a steroid receptor
Raymond Involvement of the Sigma-1 Receptor in Neuronal Cell Death and Alzheimer's Disease
KR20230161941A (en) Compositions and methods for identification of compounds that protect against lipofuscin cytotoxicity
US20050159352A1 (en) Use of monocarboxylate transporter protein for thyroid hormone transport
RU2242978C2 (en) Steroid sapogenin and their derivatives for treatment of alzheimer&#39;s disease
Luoma Molecular mechanisms of excitotoxicity: a mechanism for deafferentation-induced death and a mechanism underlying the neuroprotective effects of progesterone.
Sandau Glucocorticoids exacerbate hypoxia induced neuronal death in the developing rat cortex through regulation of the pro-apoptotic gene Bnip3
VENLAFAXINE DRUGS AND THE BRAIN
Alves Androgen Receptor is Expressed in Murine Choroid Plexuses and Downregulated by 5?-dihydrotestosterone in Male and Female Mice

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 NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ 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 CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE 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
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

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

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP