AU2005295617A1 - Neuroprotective spirostenol pharmaceutical compositions - Google Patents

Neuroprotective spirostenol pharmaceutical compositions Download PDF

Info

Publication number
AU2005295617A1
AU2005295617A1 AU2005295617A AU2005295617A AU2005295617A1 AU 2005295617 A1 AU2005295617 A1 AU 2005295617A1 AU 2005295617 A AU2005295617 A AU 2005295617A AU 2005295617 A AU2005295617 A AU 2005295617A AU 2005295617 A1 AU2005295617 A1 AU 2005295617A1
Authority
AU
Australia
Prior art keywords
cells
hydroxycholesterol
alkyl
neuronal
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2005295617A
Inventor
Janet Greeson
Laurent Lecanu
Vassilios Papadopoulos
Gary Teper
Zhi-Xing Yao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Georgetown University
Samaritan Pharmaceuticals Inc
Original Assignee
Georgetown University
Samaritan Pharmaceuticals Inc
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 US11/031,538 external-priority patent/US20060009433A1/en
Application filed by Georgetown University, Samaritan Pharmaceuticals Inc filed Critical Georgetown University
Publication of AU2005295617A1 publication Critical patent/AU2005295617A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • A61K31/585Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin containing lactone rings, e.g. oxandrolone, bufalin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/59Compounds containing 9, 10- seco- cyclopenta[a]hydrophenanthrene ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Epidemiology (AREA)
  • Virology (AREA)
  • Psychology (AREA)
  • Pain & Pain Management (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Biotechnology (AREA)
  • Communicable Diseases (AREA)
  • Molecular Biology (AREA)
  • Cardiology (AREA)
  • Psychiatry (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Steroid Compounds (AREA)

Description

WO 2006/044665 PCT/US2005/037023 NEUROPROTECTIVE SPIROSTENOL PHARMACEUTICAL COMPOSITIONS 5 Related Applications Data This application is a continuation-in-part of U.S. Patent Application Serial No. 10/389,189, filed March 14, 2003, and U.S. Patent Application Serial No. 10/663,619, filed September 16, 2003, which claim priority to U.S. 10 Provisional Patent Application No. 60/364,140, filed March 15, 2002, U.S. Provisional Patent Application No. 60/319,846, filed January 9, 2003, U.S. Provisional Patent Application No. 60/618,696, filed October 14, 2004 and U.S. Patent Application Serial No. 11/031,538, filed January 7, 2005. These applications are all incorporated herein by reference. 15 Background Nerve cell death (degeneration) can cause potentially devastating and irreversible effects for an individual and may occur for example, as a result of stroke, heart attack or other brain or spinal chord ischemia or trauma. Additionally, neurodegenerative disorders that involve nerve cell death include 20 Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, Down's Syndrome, stroke, traumatic brain injury, cerebral ischemia, cerebral hypoxia, seizures, brain infectious conditions, spinal cord concussion/section and Korsakoff's disease. Alzheimer's Disease (AD) is a progressive neurodegenerative disorder 25 characterized clinically by progressive loss of intellectual function. AD affects about 10% of the population who are beyond the age 65. It attacks 19% of individuals 75 to 85 years old, and 45% of individuals over age 85. AD, is the fourth leading cause of death in adults, behind heart disease, cancer, and stroke. AD accounts for about 75% of senile dementia. This central nervous system 30 disorder is marked by a variety of symptoms such as degeneration of neurons, development of amyloid plaques, neurofibrillary tangles, declination of acetylcholine, and atrophy of cerebral cortex. Patients with AD suffer loss of short-term memory initially followed by a decline in cognitive function and finally a loss of the ability to care for themselves. The cost of caring for WO 2006/044665 PCT/US2005/037023 patients, including diagnosis, nursing, at-home care, and lost wages is estimated at between about $80 billion and $90 billion per year. The early onset familial Alzheimer's disease (<5% of the Alzheimer cases) is caused by a mutation either of the APP gene, or of the PS1 and PS2 5 genes. The origins of the late onset sporadic form of the Alzheimer's disease (>95% of the Alzheimer cases) remains unknown even though some predisposition factors like a mutation of the mitochondrial pseudogenes C01 and C02, or like the allele s4 of the ApoE gene have been discovered. In addition several theories have been proposed to explain the origin of the disease, 10 including the amyloidergic origin, oxidative stress, calcium homeostasis disruption, mitochondrial dysfunction/metabolic decline and excitatory amino acid toxicity. The drastic impairment of function associated with AD is caused by the presence of neuritic plaques in the neocortex and hippocampus, the loss of 15 presynaptic markers of cholinergic neurons, and the loss of cholinergic neurons. Neuritic plaques are composed of degenerating axons and nerve terminals, often surrounding an amyloid core and usually containing reactive glial elements. Another characteristic pathologic feature of Alzheimer's Disease is the neurofibrillary tangle, which is an intraneuronal mass, which corresponds to an 20 accumulation of abnormally phosphorylated tau protein polymerized into fibrillar structures termed paired helical filaments. In addition, the neurofibrillary tangle also contains highly phosphorylated neurofilament proteins. Although there has been significant progress in unfolding the 25 pathophysiologic mechanisms of the disease, the cause of AD is still poorly understood. There are several suspected causes, such as genetic predisposition (PS-1, PS-2, APP, apoE, C01, C02 gene mutations), neurotransmitter defects (acetylcholine deficiency), inflammation, metabolic decline, free radical stress, or excitatory amino acid toxicity. 30 Several compounds are currently under clinical studies for the treatment of AD according to the current understanding of its pathogenesis. Among these drugs notably are acetylcholine esterase (AchE) inhibitors. Recently, two AchE inhibitors, tacrine and donepezil, have received regulatory approval for AD treatment. While tacrine provides a moderate beneficial effect on deterioration of WO 2006/044665 PCT/US2005/037023 cognition, it suffers some adverse effects as it causes increases in serum hepatic enzymes. It thus would be highly desirable to have new neuroprotective agents, particularly agents to limit the extent or otherwise treat nerve cell death 5 (degeneration) such as may occur with stroke, heart attack or brain or spinal cord trauma, or to treat neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, Down's Syndrome and Korsakoffs disease, or any of the other conditions disclosed hereinabove. 10 Alzheimer's disease is characterized by the accumulation of a 39-43 amino acid peptide termed the f-amyloid protein or AO, in a fibrillar form, existing as extracellular amyloid plaques and interneuronal deposits, and as amyloid within the walls of cerebral blood vessels. Fibrillar AO amyloid deposition in Alzheimer's disease is believed to be detrimental to the patient and 15 eventually leads to toxicity and neuronal cell death, characteristic hallmarks of AD. Accumulating evidence implicates amyloid as a major causative factor of AD pathogenesis. Trimers and tetramers belong to the amyloid-derived diffusible ligands (ADDLs), which are non fibrillar oligomers ranging approximately from 13 to 20 108 kD (Klein, Neurochem. Int., 41, 345-352 (2002)), with potent neurotoxic properties at concentration as low as 5-10 nM (Lambert et al., Proc. Natl. Acad. Sci. USA, 95, 6448-6453 (1998); Dahlgren et al., J. Biol. Chem., 277(35), 32046-32053 (2002)). A recent report described the ADDLs as baring the neurotoxic properties of Ap. Klein, Neurochem. Int., 41, 345-352 (2002). 25 A variety of other human diseases also demonstrate amyloid deposition and usually involve systemic organs (i.e., organs or tissues lying outside the central nervous system), with the amyloid accumulation leading to organ dysfunction or failure. In AD and "systemic" amyloid diseases, there is currently no cure or effective treatment, and the patient usually dies within 3 to 10 years 30 from disease onset. Much work in AD has been accomplished, but little is conventionally known about compounds or agents for therapeutic regimes to arrest amyloid formation, deposition, accumulation and/or persistence that occurs in AD and other amyloidoses.
WO 2006/044665 PCT/US2005/037023 New compounds or agents for therapeutic regimes to arrest or reverse amyloid formation, deposition, accumulation and/or persistence that occurs in AD and other amyloidoses are therefore needed. Consequently, it would be greatly beneficial if new therapies could be 5 designed based on identified existing compounds, rationally modified compounds and/or de novo designed compounds which are active as A3 functional inhibitors. Furthermore, repairing the brain damage caused by AD and other neuropathologies by replacing neuronal losses and restoring the associated 10 functions by employing methodologies collectively known as "stem cell therapy" is extremely promising. The ability to differentiate stem cells into neurons can provide a treatment for neurodegenerative diseases and stroke. Stem cells differentiated into dopaminergic neurons have already been successfully used to treat patients suffering from Parkinson's disease (T. Barberi 15 et al., Nature Biotechnology, 21, 1200 (2003)). However, in diseasesor conditions in which the neuronal loss could be much more important, like Alzheimer's disease (AD), brain stroke or traumatic brain injury, the transplantation of differentiated stem cells, although critical, might not be enough to compensate the brain damages and to restore the hampered functions. 20 For a maximum recovery, the transplantation might have to be associated to a stimulation of neurogenesis in situ. In the human adult brain, the neural stem cells are found in the sub-ventricular zone (SVZ) and in the dentate gyrus of the hippocampus (P.S. Eriksson et al., Nature Medicine, 4, 1313 (1998); V. Silani et al., Lancet, 364, 200 (2004)) and the ability to pharmacologically induce their 25 differentiation through the neuronal pathway would constitute an important step towards the neuronal replacement therapy. Many small molecules like retinoic acid or cyclopamine, have been used to induce the neuronal differentiation of NSC in vitro, but their use in vivo is extremely difficult because of their high toxicity. Dexamethasone, fluoxetine or geldanamycin have potentially 30 dangerous side effects (S. Ding et al., Nature Biotech., 22, 833 (2004)). Thus, a continuing need exists for methods to slow, halt or reverse the progression of neurological disorders, or to ameliorate the symptoms thereof.
A
WO 2006/044665 PCT/US2005/037023 Summary of the Invention The present invention is directed to methods, kits, combinations, and compositions for treating, preventing or reducing the risk of developing a disorder or disease related to, or the symptoms associated with, neurotoxicity in 5 a subject, particularly to beta-amyloid-induced neurotoxicity. The compounds of the present invention some of which contain a common spirost-5-en-3-ol structure, and having the structure of formulas (I), (II) or (III) are disclosed below. The present invention is directed to a method of treating a condition or 10 disorder where treatment with a neurotoxicity inhibiting agents or stem cell differentiating agents of formula (I), (II) or (III) is indicated, the method comprises administration of a composition of the present invention to a subject in need thereof. More specifically, the subject invention provides a method for inhibiting the neurotoxic effects of Af3 formation or persistence of brain fi 15 amyloid deposits in a patient, the method comprising administering to the patient a therapeutically effective amount of a compound of formula (I), (I) or (III). In one aspect, the invention provides a method for promoting, maintaining or enhancing in a patient one or more of the mental or cognitive qualities selected from the group of mental or cognitive qualities associated with 20 fl-amyloid formation consisting of memory, concentration, and short term memory, the method comprising administering to the patient a therapeutically effective amount of a compound of formula (I), (II) or (III). In another aspect, the invention provides a method for reducing in a patient one or more of the mental or cognitive effects associated with fl-amyloid 25 formation selected from the group of mental or cognitive effects associated with #-amyloid formation consisting of cognitive or memory decline and mental decline, the method comprising administering to the patient a therapeutically effective amount of a compound of formula (I), (II) or (III). In yet another aspect, the invention provides a method for treating in a 30 patient mental states associated with fl-amyloid formation or persistence, the method comprising administering to the patient a therapeutically effective amount of a compound of formula (I), (II) or (III). In still another aspect the invention provides a method for treating a patient having a neurological disease or disorder selected from the group WO 2006/044665 PCT/US2005/037023 consisting of global and focal ischemic and hemorrhagic stroke, traumatic brain injury, spinal cord injury (e.g., recision or section), hypoxia-induced or ischemia-induced nerve cell damage, nerve cell damage caused by cardiac arrest or neonatal distress, epilepsy, anxiety, diabetes mellitus, multiple sclerosis, 5 phantom limb pain, causalgia, neuralgias, herpes zoster, spinal cord lesions, hyperalgesia, allodynia, AD, Huntington's disease, Korsakoff's disease and Parkinson's disease, or any of the other conditions disclosed hereinabove, wherein said treatment comprises administering to the patient a therapeutically effective amount of a compound of formula (I), (II) or (III). 10 In a further aspect, the invention provides a method for treating a disease characterized by f-amyloid deposits referred as amyloidosis, in the heart,, spleen, kidney, adrenal cortex, or liver of a patient comprising administering to the patient a therapeutically effective amount of a compound of formula (I), (II) or (III). 15 In a further aspect, the present invention provides a method for inducing the differentiation of mammalian neuronal precursor cells, such as adult or embryonic stem cells, into neuronal cells, or astrocytal cells, such as neurons, and/or astrocytes by contacting said neuronal precursor cells, in vitro or in vivo, with an effective amount of a compound of formula (I), (II) or (III). For 20 example, multipotent adult progenitor cells (MAPCs) can be obtained by methods available to the art. The cells are either differentiated in vitro to form neuronal cells, which are then administered to a target site, or are directly administered to the target site of an afflicted mammal, with or followed by a differentiating-inducing amount of a compound of formula (I), (II) or (III). The 25 stem cells then differentiate in vivo to regenerate or replace damaged tissue. In a still further aspect, the invention provides a method of identifying a compound having binding affinity to 0-amyloid comprising screening a database of known chemical compounds for structural homology to 22R hydroxycholesterol; ranking the compounds in the database based on the degree 30 of homology to 22R-hydroxycholesterol, extracting from the database compounds having the highest structural homology to 22R-hydroxycholesterol; ranking the extracted compounds according to in vitro binding to f-amyloid; and selecting the compound having the highest in vitro affinity.
WO 2006/044665 PCT/US2005/037023 In another aspect, the invention provides novel compounds which inhibit the formation of ADDLs, such as p-amyloid trimers and tetramers, by binding to As and forming stable nontoxic polymers. In still another aspect, the invention provides a method of designing a 5 compound having binding affinity to 0-amyloid comprising mapping 22R hydroxycholesterol into two or more separate building blocks; designing a new compound by modifying one or more blocks of 22R-hydroxycholesterol, ranking the designed compound according to in vitro binding to -amyloid; and selecting the compound having the highest in vitro binding affinity. 10 In a further aspect, the invention provides a method of designing a compound having binding affinity to #-amyloid comprising mapping -amyloid, constructing on a computer screen a compound that complements the structure of -amyloid or a fragment thereof; ranking the designed compound according to in vitro binding to f-amyloid; and selecting the compound having the highest in 15 vitro binding affinity. In yet another aspect, the invention provides a method of detection and quantification of AO in biological fluid comprising obtaining a sample fluid; incubating the fluid with a detectably-labeled, e.g., radiolabeled, compound of formula (I), (II) or (III); optionally in the presence of increasing concentrations 20 of unlabeled compound; separating samples from the incubation fluid and transferring the samples to a membrane; exposing the membrane to label sensitive screen; and analyzing the contents of the membrane by imaging, such as phospho-imaging, to detect the presence of AO or quantifying the amount of AO present in the biological fluid. 25 In still another aspect, the invention provides a method of diagnosing AD in a subject comprising obtaining a sample fluid from the brain of the subject; incubating the fluid with a detectably-labeled compound of formula (I), (II) or (III); optionally in the presence of increasing concentrations of unlabeled compound; separating samples from the incubation fluid and transferring the 30 samples to a membrane; exposing the membrane to label-sensitive screen; and analyzing the contents of the membrane by imaging to detect the presence of AO or quantifying the amount of AO present in the biological fluid. Accordingly, a principal aspect of this invention relates to a pharmaceutical composition for treating a disorder related to a beta-amyloid '7 WO 2006/044665 PCT/US2005/037023 induced neurotoxicity or a neurodegenerative disorder in a subject. This composition includes an effective amount of a compound of formula (I), (II) or (III), and a pharmaceutically acceptable carrier. Also within the scope of this invention is the use of a compound of formula (I), (II) or (III) for the 5 manufacture of a medicament to be used in treating one of such disorders. Treatment of these conditions is accomplished by administering to a subject a therapeutically effective amount of a compound or composition of the present invention. The details of one or more embodiments of the invention are set forth in 10 the accompanying description below. Other features, objects, and advantages of the invention will be apparent from the description and claims. Brief Description of the Drawings The figures illustrate some of the compounds of the invention, methods 15 for identifying those compounds and results of in vitro and in vivo biological test demonstrating the activity of illustrative compounds according to the invention. Fig. 1 illustrates several of the structures of the chemical structure of 22R-hydroxycholesterol (SP222) and naturally occurring derivatives. 20 Fig. 2 is a chart describing 22R-hydroxycholesterol levels in AD and control brain specimens. Fig. 3A is a line graph depicting the effect of increasing concentrations of 22R-hydroxycholesterol on rat PC12 neuronal cell viability in the absence or presence of increasing concentration of A0 1
.
42 . 25 Fig. 3B is a line graph depicting the effect of increasing concentrations of cholesterol on rat PC12 neuronal cell viability in the absence or presence of increasing concentration of A01..
42 . Fig. 3C is a line graph depicting the effect of increasing concentrations of pregnenolone on rat PC12 neuronal cell viability in the absence or presence of 30 increasing concentration of A#1- 42 . Fig. 3D is a line graph depicting the effect of increasing concentrations of 17ca-hydroxypregnenolone on rat PCl2 neuronal cell viability in the absence or presence of increasing concentration of A01.
42 .
Q
WO 2006/044665 PCT/US2005/037023 Fig. 3E is a line graph depicting the effect of increasing concentrations of DHEA on rat PC12 neuronal cell viability in the absence or presence of increasing concentration of A#l 142 . Fig. 3F is a line graph depicting the effect of increasing concentrations of 5 22S-hydroxycholesterol on rat PC12 neuronal cell viability in the absence or presence of increasing concentration of A#1p42. Fig. 4 is a line graph depicting the effect of 22R-hydroxycholesterol on differentiated human NT2N neuron viability determined in absence or presence of A#l 14 2 . 10 Fig. 5A is a line graph depicting the effect of 22R-hydroxycholesterol and DHEA on A#l 1 4 2 -induced toxicity on rat PC12 neuronal cells. Fig. 5B is a line graph depicting the effect of 22R-hydroxycholesterol and DHEA on A# 25
-
35 -induced toxicity on rat PC12 neuronal cells. Fig. 5C is a line graph depicting the effect of 22R-hydroxycholesterol 15 and DHEA on A# 1 42 -induced toxicity on human NT2 cells. Fig. 5D is a line graph depicting the effect of 22R-hydroxycholesterol and DHEA on Afl25- 35 -induced toxicity on human NT2 cells. Fig. 6A is a coomassie blue gel depicting the effect of 22R hydroxycholesterol on Afl aggregation. 20 Fig. 6B is an immunoblot analysis of the coomassie blue stained gel of Fig. 6A depicting the effect of 22R-hydroxycholesterol on Afl aggregation. Fig. 7A is an immunoblot analysis identifying A3 1
-
4 2 -22R hydroxycholesterol binding and binding site by CPBBA. Fig. 7B is an immunoblot analysis identifying Afl 14 2 by a polyclonal 25 rabbit anti--amyloid peptide antiserum on the blot shown in Fig. 7A. Fig. 7C is an immunoblot analysis identifying the 22R hydroxycholesterol binding site on A0. Fig. 7D is a computational 22R-hydroxycholesterol docking simulation to Afl 42 . 30 Fig. 7E is a computational 22R-hydroxycholesterol docking simulation to Afl1740. Fig. 7F is a computational 22R-hydroxycholesterol docking simulation to A01740.
Q
WO 2006/044665 PCT/US2005/037023 Fig. 7G is a computational 22R-hydroxycholesterol docking simulation to A0 1
..
42 . Fig. 7H is a computational 22R-hydroxycholesterol docking simulation to A017.40. 5 Fig. 71 is an amino acid sequence (SEQ ID NO:1) of the localization of the 22R-hydroxycholesterol binding site in A03p4 2 . Fig. 8 is a bar graph illustrating that three days' exposure of PC12 cells to increasing concentrations of A0 resulted in dose-dependent cell death. Figs. 9A to 9P are a series of bar graphs illustrating the effect increasing 10 concentrations of 22R-hydroxycholesterol (SP222) and derivatives on rat PC12 neuronal cell viability in the absence or presence of 0.1 pIM of A0 1
-
42 . Figs. 10 A to 1OP are a series of bar graphs illustrating the effect increasing concentrations of 22R-hydroxycholesterol (SP222) and derivatives on rat PC12 neuronal cell viability in the absence or presence of 1.0 ptM of A1- 42 . 15 Figs. 11 A to 1iP are a series of bar graphs illustrating the effect increasing concentrations of 22R-hydroxycholesterol (SP222) and derivatives on rat PC12 neuronal cell viability in the absence or presence of 10.0 AM of A0 1
-
42 . Fig. 12A is a bar graph showing that A0 exposure induces a dose-related decrease of the membrane potential-assessing luminescence. 20 Fig. 12B is a bar graph showing the effect of 22R-hydroxycholesterol (SP222) and derivatives against 0.1 pLM Af-induced neurotoxicity. Fig. 12C is a bar graph showing the effect of 22R-hydroxycholesterol (SP222) and derivatives against 1.0 ptM Ao-induced neurotoxicity. Fig. 12D is a bar graph showing the effect of 22R-hydroxycholesterol 25 (SP222) and derivatives against 10.0 AM Ao-induced neurotoxicity. Fig. 13A is a bar graph showing that AB decreased in a dose-dependent manner ATP production by PC12 cells in the presence of 0.1, 1.0 and 10.0 ptM Ao-induced neurotoxicity. Fig. 13B is a bar graph showing the effect of 22R-hydroxycholesterol 30 (SP222) and derivatives on ATP in the presence of 0.1 [M Af-induced neurotoxicity. Fig. 13C is a bar graph showing the effect of 22R-hydroxycholesterol (SP222) and derivatives on ATP in the presence of 1.0 tM A-induced neurotoxicity.
WO 2006/044665 PCT/US2005/037023 Fig. 13D is a bar graph showing the effect of 22R-hydroxycholesterol (SP222) and derivatives on ATP in the presence of 10.0 /gM A-induced neurotoxicity. Fig. 14 is a line graph showing trypan blue uptake by cells in the 5 presence of AO alone; AO + SP233 30 1 M; and AO + SP233 50 [M. Fig. 15 is a line graph showing the effect of increasing concentrations of SP233 on 0.1, 1.0, and 10.0 giM AO-induced neurotoxicity on rat PC12 neuronal cell. Fig. 16 is a line graph illustrating the effect of SP233 on MA-10 Leydig 10 cell steroid formation. Fig. 17 is a bar graph identifying Af-SP binding and binding site by CPBBA. Fig. 18 is a computational docking simulation depicting the binding energy frequencies of 22R-hydroxycholesterol (SP222) and SP233 to A0 1
.
42 . 15 Fig. 19 is a computational docking simulation depicting the probabilities of 22R-hydroxycholesterol (SP222) and SP233 binding to A01.
42 . Fig. 20A is an immunoblot analysis of AO polymerization and ADDL formation in increasing concentrations of SP233 (1, 10, 100 pM) after 24 hours incubation in cell culture medium. 20 Fig. 20B is a bar graph depicting AO monomers identified by the immunoblot analysis of Fig. 20A. Fig. 20C is a bar graph depicting AO trimers identified by the immunoblot analysis of Fig. 20A. Fig. 20D is a bar graph depicting AO tetramers identified by the 25 immunoblot analysis of Fig. 20A. Fig. 20E is a line graph depicting AO polymer and ADDLs (the sum of trimers and tetramers) formation in the immunoblot analysis of Fig. 20A. Fig. 20F is an immunoblot analysis of AO polymerization and ADDL formation in increasing concentrations of SP233 (1, 10, 100 pM) after 72 hours 30 incubation in cell culture medium. Fig. 20G is a bar graph depicting AO trimers identified by the immunoblot analysis of Fig. 20F. Fig. 20H is a bar graph depicting AO tetramers identified by the immunoblot analysis of Fig. 20F. 1 1 WO 2006/044665 PCT/US2005/037023 Fig. 201 is a bar graph depicting AO tetramers identified by the immunoblot analysis of Fig. 20F. Fig. 20J is a line graph depicting AO polymer and ADDLs (the sum of trimers and tetramers) formation in the immunoblot analysis of Fig. 20F. 5 Fig. 21. Effect of 22R-hydroxycholesterol on the differentiation of NT2 cells. Upper panel (a-d): phase-contrast photomicrographs showing morphologic changes in NT2 cells treated with 25pM 22R-hydroxycholesterol for 3, 6 and 12 days (lower panel, (a) is control) compared to untreated control cells (upper panel). Lower panel (e-h): flow cytometric analyses indicating differentiation of 10 NT2 cells toward the neuronal phenotype (NT2N) in response to treatment with 25p.M 22R-hydroxycholesterol for 3, 6 and 12 days, compared to untreated control cells. Fig. 22. Effects of steroids on the differentiation, viability and proliferation of NT2 cells. Phase-contrast photomicrographs: (a) comparison of 15 morphologic changes in NT2 cells treated for 6 days with 25pM 22S hydroxycholesterol (middle panel) or 22R-hydroxycholesterol (lower panel), and untreated control (upper panel); (b) comparison of morphologic changes in NT2 cells treated with 25pM cholesterol (upper-middle panel), progesterone (lower middle panel) or DHEA (lower panel) for 6 days, and untreated control (upper 20 panel); (c) analyses of the effects of 3-day treatments with 22R hydroxycholesterol, 22S-hydroxycholesterol, pregnenolone or progesterone on the viability and proliferation of NT2 cells using LDH (upper panel), MTT (middle panel) and BrdU (lower panel) assays. Significance: *p<0.0 5 , **p<0.01 and ***p<0.001 compared to untreated (control) values. Results shown are 25 representative of 3 independent experiments. Fig. 23. Effects of 22R-hydroxycholesterol treatment on the expression of neurofilament proteins by NT2 cells. Upper panel: Immunocytochemical staining showing the expression of neurofilament proteins NF70 (a), NF145 (b) and NF200 (c) in NT2 cells treated with 25pM 22R-hydroxycholesterol for 6 30 days versus untreated controls. Results shown are representative of 4 independent experiments; magnification, 40X. Lower panel: Immunoblot analyses (using specific antisera) and quantitative image analyses of NF70 (d), NF 145 (e), and NF200 (f) in NT2 cells that were treated for 6 days with increasing concentrations of 22R-hydroxycholesterol versus untreated controls; 11, WO 2006/044665 PCT/US2005/037023 GAPDH (glyceraldehyde-3-phosphate dehydrogenase) was used to normalize the amount of protein loaded. Significance: **p<0.01 and ***p<0.001 compared to untreated controls. Results shown are representative of 3 independent experiments. 5 Fig. 24. Possible mechanism of 22R-hydroxycholesterol-induced differentiation of NT2 cells. (a-c) Effect of 22R-hydroxycholesterol on the expression of GFRc receptor proteins in NT2 cells. Upper panel: Immunocytochemical staining showing the expression of GFRa.l (a), GFRa2 (b) and GFRc3 (c) in NT2 cells after treatment with 25 yLM 22R 10 hydroxycholesterol for 6 days versus controls. Results shown are representative of 4 independent experiments (magnification, 40X). Lower panel: Immunoblot analyses (using specific antisera) and quantitative image analyses of GFRal (a), GFRa2 (b) and GFRa3 (c) in NT2 cells treated for 6 days with 25 yIM 22R hydroxycholesterol versus controls; GAPDH was used to normalize the amount 15 of protein loaded. (d) Uptake of 3 H-22R-hydroxycholesterol by NT2 cells during their differentiation to the neuronal (NT2N) phenotype (upper panel) and interaction of 3 H-22R-hydroxycholesterol and NT2N cell proteins (lower panel). Significance: *p<0.05, and ***p<0.001 compared to untreated controls. Results shown are representative of 3 independent experiments. 20 Fig. 25. Mouse embryonic teratocarcinoma P19 cells, PC12 rat pheochromocytoma cells and NT2 human neuronal cells were cultured on 13 mm diameter glass cover-slip. When cells reached 70% confluence, the medium was replaced fresh medium containing 90 ptM SP-224. P19 and PC12 cells were then incubated for 2 days before SP224 was washed out and replaced by 25 standard medium. The culture medium was changed every 2 days for 5 days before cells were fixed for immunocytochemistry. The NT2 cells were treated for 5 days and washed-out for 10 days. Under these experimental conditions, the three types of cells displayed a significant sprouting (Panels 1B, D, F) and the most dramatic effect was observed with the P19 cells (Panel D). Interestingly, 30 the sprouting started during the washing out period, whereas no cell shape modification was observed during the treatment period. Meantime, growth arrest of the treated-cells was observed.
WO 2006/044665 PCT/US2005/037023 Fig. 26. No neuronal marker was observed on the control except for the synaptophysin for which the staining was, however, very weak (Panel C). The SP-224 exposure induced a strong expression of the different neuronal markers studied, pII tubulin (Panel A), synaptophysin (Panel B), MAP2 (Panel C) and 5 ChAT (Panel D). The migrating neuroblasts marker DCX was also strongly expressed in SP-224-exposed P19 cells (Panel E). Panel A even shows pIII tubulin immunostaining on axons-like formation which length is significantly greater than the 100 pim scale bar. Fig. 27. P19 cells were treated for 2 days and washed-out for 30 days 10 before being immunolabeled for neuronal markers. The network of axons and dendrites has dramatically extended as shown by the PII tubulin staining (Panel A). The importance of the synaptophysin labeling (Panel B) shows that the newly formed neurons established synaptic connections. Fig. 28. The expression of the glial markers GFAP and CNPase, 15 reflecting the ability of SP-224 to differentiate P19 cells in astrocytes and oligodendrocytes has been studied in order to assess an eventual specificity of the differentiation process. Using the protocol 2 days with 90 M SP-224 followed by 5 days of culture in SP-224 free medium, SP-224 induced also a differentiation of the P19 into oligodendrocytes (Panel B) whereas no positive 20 GFAP signal was detected (Panel D). Both markers GFAP and CNPase were not expressed in controlled P19 cells. Fig. 29. Male Long-Evans rats weighing 300-325 g were implanted with an osmotic micropump which outlet was implanted into the left cerebral ventricle following the coordinates D = 3.4 mm, L = 1.4 mm and AP = 0.92 mm 25 caudal to bregma. The tank of the osmotic pump was implanted in a subcutaneous pocket in the midscapular area of the back of the rat. SP-224 at 375 p.M solubilized in polypropylene glycol/glycerol/distilled water (50/25/25) was perfused by i.c.v. route at 5 p1 per hour for 2 weeks. Rats were sacrificed 3 weeks after the end of the brain infusion. The immunohistochemistry revealed 30 important BrdU areas showing that SP-224 was able to induce a self-renewal of the neural stem cells in the SVZ (Panels C-G). In addition some BrdU positive cells were detected at a significant distance of the SVZ (white arrows) suggestion that certain cells might have entered a migration process. SP-224 1 WO 2006/044665 PCT/US2005/037023 infusion also induced the expression of the early neuronal marker DCX in the SVZ cells, showing that as in vitro, SP-224 was able to push the neural stem cells present in that area toward the neuronal differentiation (Panels D-H). 5 Detailed Description of the Invention While the present invention may be embodied in many different forms, several specific embodiments are discussed herein with the understanding that the present disclosure is to be considered only as an exemplification of the principles of the invention, and it is not intended to limit the invention to the 10 embodiments illustrated. Abbreviations used herein are as follows: 5-cholesten-3,22R-diol, (22R hydroxycholesterol); 5-cholesten-3p, 22S-diol, (22S-hydroxycholesterol); 5 cholesten-3p-ol (cholesterol); 5-androsten-3p-ol-17-one or dehydroepiandrosterone (DHEA); 5-pregnen-3p,17c-diol-20-one (17Ua 15 hydroxypregnenolone); 5-pregnen-3p-ol-20-one (pregnenolone); Ntera2/D1 teratocarcinoma cells (NT2); differentiated human NT2 neurons (NT2N); P amyloid peptide, (Ap); Alzheimer's disease, (AD); cholesterol-protein binding blot assay (CPBBA); retinoic acid (RA). One aspect of this invention relates to a method of treating a disorder 20 related to neurotoxicity, particularly AD, comprising administering to a subject in need thereof at least one compound of formula (I), (II) and/or (III). The formula of compound (I) is as follows:
R
12
R
13
R
1 7
R
1 0
R
8
R
9
R
14
R
15
R
3
R
4
R
6 (I) 25 wherein each of R 1 , R 2 , R 4 , R 7 , R1, R 1 2 , R 15 , and R 16 , independently, is hydrogen, (C 1 -Cs)alkyl, hydroxy, amino, carboxyl, oxo, sulfonic acid, or (C 1 Cs)alkyl that is optionally inserted with -NH-, -N((C 1 -Cs)alkyl)-, -0-, -S-, -SO-,
-SO
2 -, -0-S02-, -S0 2 -0-, -C(O)-, -C(O)-O-, -0-C(O)-, -C(O)-NR'-, or -NR' C(O)-, wherein R' is H or (C 1
-C
8 )alkyl; R 3 is hydroxy, (C1-C 6 )alkylCO2-, 1 r, WO 2006/044665 PCT/US2005/037023
HO
2
C(CH
2
)
2
CO
2 -, toluene-4-sulfonyloxy, or benzoyloxy; each of R 6 , R 8 , R 9 , Rio, R 13 and R 14 , independently, is hydrogen, (C 1 -Cs)alkyl, hydroxyl(C1
C
8 )alkyl, (C 1 -Cs)alkoxy, or hydroxy; and R1 7 is CH(CH 3
)CH(OH)(CH
2
)
2 CH(CH3)2 or 5 -CH(CH 3
)CH(OC(=O)CH
3
)(CH
2
)
2 CH(CH3)CH2N(C(=O)CH3)2; or a pharmaceutically acceptable salt thereof. Preferably R 1 , R 2 , R 4 , R 6 , R 7 , Rs, R 9 , Ru, R 1 2 , R 14 and R 1 5 are H. Preferably, Rio and R 13 are CH 3 ; and preferably, R 16 is H or acetoxy. The structure of formula (II) is as follows:
CH
3
R
1 9
R
1 2 R 1 3 R20 RR11 O
R
1 R1 0
R
8
R
9
R
1 4
R
1 R3 Ry 10 R 4
R
6 (II) wherein each of R 1 , R 2 , R 4 , R 7 , RI, R 12 , and R 15 , independently, is hydrogen, (C 1
-C
4 ) alkyl, hydroxy, amino, carboxyl, oxo, sulfonic acid, or (C 1 C 6 )alkyl that is optionally inserted with -NH-, -N((C 1
-C
4 )alkyl)-, -0-, -S-, -SO-, -S02-, -0-SO 2 -, -S0 2 -0-, -C(O)-, -C(O)-O-, -O-C(O)-, -C(O)-NR'-, or -NR' 15 C(O)- wherein R' is H or (C 1
-C
8 )alkyl; R 3 is hydroxy, (C 1
-C
6 )alkylCO 2 -,
HO
2
C(CH
2
)
2
CO
2 -, toluene-4-sulfonyloxy, or benzoyloxy; each of R 6 , R 8 , R 9 , Rio, R 13 and R 14 , independently, is hydrogen, (C 1
-C
4 )alkyl, hydroxyl(C1 Cs)alkyl, (C 1 -Cs)alkoxy, or hydroxy; R 19 is OH or (C 1
-C
2 )alkoxy; and R 20 is butyl 3-substituted by methyl amidomethyl, or a pharmaceutically acceptable 20 salt thereof. Preferably, R 1 , R 2 , R 4 , R 6 , R 7 , R 8 , R 9 , R 11 , R 1 2 , R 14 and R 15 are H. Preferably, RIO and RI 3 are CH 3 . Compounds of formula (III) are shown below: CH3 X CHs R12 eR13 R R1 R0 R2 |,0R R15 ,4 R9 R14 R1 R3 Ry R4 R(III) 1A: WO 2006/044665 PCT/US2005/037023 wherein each of R 1 , R 2 , R 4 , R 7 , RI, R 12 , and R 15 , independently, is hydrogen, (C 1
-C
8 )alkyl, hydroxy, amino, carboxyl, oxo, sulfonic acid, or (C 1 C 8 )alkyl that is optionally inserted with -NH-, -N((C 1
-C
8 )alkyl)-, -0-, -S-, -SO-,
-SO
2 -, -O-SO 2 -, -S02-0-, -C(O)-, -C(O)-O-, -O-C(O)-, -C(O)-NR'-, or -NR' 5 C(O)-, wherein R' is H or (C 1
-C
8 )alkyl; R3 is hydroxy, (C1-C 6 )alkylCO 2 -,
HO
2
C(CH
2
)
2 CO2-, toluene-4-sulfonyloxy, or benzoyloxy; each of R 6 , R 8 , R 9 , Rio, R 13 and R 14 , independently, is hydrogen, (C 1
-C
8 )alkyl, hydroxyl (C 1 Cs)alkyl, (C 1 -Cs)alkoxy, or hydroxy; and X is 0, N(H), N(Ac), N(toluene-4 sulfonyloxy), or a pharmaceutically acceptable salt thereof. 10 RIO and R 13 are preferably CH 3 In compounds of formula (III), R 1 , R 2 , R 4 , R 6 , R 7 , R 8 , R 9 , Ra, R 12 , R 14 and R 15 are preferably H. R 1 , R 2 or R 12 preferably are H or OH. R3 may also be OR 23 wherein R 23 is a removable hydroxy-protecting group such as tosyl, mesyl, trialkylsilyl, THP, EtO(Et), benzyl, benzyl 15 oxycarbony and the like. True (C 8
-C
22 ) fatty acid esters of compounds in which
C
3 is hydroxy-substituted are also within the invention, wherein the alkoxy chain can comprise 1-2 CH=CH units and/or 1-2 OH epoxy or methyl substituents. Preferred stereoisomers are 3S, as well as 1OR and 13S, and are also 20S, 22S and 25S wherein the carbon skeleton is numbered in accord with spirosten 20 3-ol numbering. Thus, a preferred compound of formula (III) is (22S,25S) (20S)-spirost-5-en-3p-yl hexanoate (SP233). Note that the carbon atoms shown in formula (I), (II) or (III) are saturated with hydrogen unless otherwise indicated. Unless defined otherwise, each of the term "alkyl," the prefix "alk" (as in 25 alkoxy), and the suffix "-alkyl" (as in hydroxyalkyl) refers to a C 1
-
8 hydrocarbon chain, linear (e.g., butyl) or branched (e.g., isobutyl). Alkylene, alkenylene, and alkynylene refer to divalent C 1
.
8 alkyl (e.g., ethylene), alkene, and alkyne radicals, respectively. The term "alkyl" includes cycloakyl, (cycloalkyl)alkyl and alkyl(cycloalkyl)alkyl. The term "alkenyl" likewise includes alkyl 30 comprising 1-2 CH=CH units, as well as the corresponding cycloalkenyl moieties. Specifically, (C 1
-C
8 )alkyl can be methyl, ethyl, propyl, isopropyl, butyl, iso-butyl, sec-butyl, pentyl, 3-pentyl, beptyl or octyl hexyl; (C 3
-C
8 )cycloalkyl can be monocyclic, bicyclic or tricyclic and includes cyclopropyl, cyclobutyl, 1'7 WO 2006/044665 PCT/US2005/037023 cyclopentyl, cyclohexyl, bicyclo[2.2.2] octanyl or norbornyl, as well as various terpene and terpenoid structures. (C 3 -C)cycloalkyl(Cl-C2)alkyl includes the foregoing cycloalkyl and can be cyclopropyhnethyl, cyclobutylmethyl, cyclopentylmethyl, cyclohexylmethyl, 2-cyclopropylethyl, 2-cyclobutylethyl, 2 5 cyclopentylethyl, or 2-cyclohexylethyl. Heterocycloalkyl wherein the cycloalkyl ring system is monocyclic, bicyclic or tricyclic and optionally comprises 1-2 S, non-peroxide 0 or N(R') as well as 4-5 ring carbon atoms; such as morpholinyl, piperidinyl, piperazinyl, indanyl, 1,3-dithian-2-yl, and the like. Any cycloalkyl ring system optionally includes 1-3 double bonds or epoxy moieties and 10 optionally is substituted with 1-3 OH, (C 1
-C
6 )alkanoyloxy, (CO), (C 1
-C
6 )alkyl or
(C
2
-C
6 )alkynyl. (C 1
-C
8 )alkoxy can include methoxy, ethoxy, propoxy, isopropoxy, butoxy, iso-butoxy, sec-butoxy, pentoxy, 3-pentoxy, or hexyloxy;
(C
2
-C
6 )alkenyl can be vinyl, allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-hexenyl, 2-hexenyl, 15 3-hexenyl, 4-hexenyl, or 5-hexenyl; hydroxy(C1-Cs) alkyl or hydroxy(C1
C
8 )alkoxyl can be alkyl substituted with 1 or 2 OH groups, such as alkyl substituted with 1 or 2 OH groups such as hydroxymethyl, 1-hydroxyethyl, 2 hydroxyethyl, 1-hydroxypropyl, 2-hydroxypropyl, 3-hydroxypropyl, 1 hydroxybutyl, 4-hydroxybutyl, 3,4-dihydroxybutyl, 1-hydroxypentyl, 5 20 hydroxypentyl, 1-hydroxyhexyl, or 6-hydroxyhexyl; (C 1
-C
6 )alkylCO 2 - can be acetoxy, propanoyloxy, butanoyloxy, isobutanoyloxy, pentanoyloxy, or hexanoyloxy. Shown below in Table 1 are several compounds of formula (I), (II) and (III) described above that can be used to practice this invention: 25 10 WO 2006/044665 PCT/US2005/037023 Table 1. Denomination Chemical Name Origin SP222 22R-hydroxycholesterol Mammalian SP223 (20)-26-acetylamino-(224)-hydroxyfurost-5-en-34-ra sp. (asteraceae) yl acetate SP224 (20a)-254-methyl-(22R,26)-azacyclofurost-5-en- 3 4- Solanum asperunz ol (solanaceae) SP225 (204)-26-acetylamino-(224)-methoxyfurost-5-en- 3 a- Gynura sp. (asteraceae) yl acetate SP226 (20)-25 -methyl-N-acetyl-(22R,26)-azacyclofurost- Solanum asperum 5-en-3 -ol (solanaceae) SP227 (22R,254)-(20x)-spirost-5-en-(2a,3 )-diol Gynurajaponica (asteraceae) SP228 (204)-26-acetylamino-(224)-ethoxyfurost-5-en- 3 4-yl Gynura sp. (asteraceae) acetate SP229 (20a)-254-methyl-N-paratoluenesulfonyl-(22R, 2 6 )- Solanum aviculare azacyclofurost-5-en-34-yl paratoluenesulfonate (solanaceae) SP230 (22R,254)-(20ac)-(14a,20a)-spirost-5-en-(3p,12p)- Gynurajaponica diol (asteraceae) SP231 (22R,25S)-(204)-spirost-5-en-34-ol Gynurajaponica (asteraceae) SP232 (22R,254)-(20a)-spirost-5-en-3 -yl benzoate Gynura sp. (asteraceae) SP233 (22S,25S)-(20S)-spirost-5-en-3 -yl hexanoate Gynura sp. (asteraceae) SP234 (22R,254)-(20Oc)-spirost-5-en-(14,34)-diol Gynurajapoica (asteraceae) SP235 (22R,25S)-(20a)-spirost-5-en-3p-ol Gynurajaponica (asteraceae) SP236 (22R,25S)-(20a)-spirost-5-en-3 -yl succinate Gynura sp. (asteraceae) SP237 26-diacetylamino-(22 )-acetoxy-(16 )-acetoxy- Acllya heterosexuals cholest-5-en-yl acetate (saprolegniaceae) SP238 (20a)-25S-methyl-N-acetyl-(22S,26)-azacyclofurost- Solanur asperum 5-en-3 -yl propanoate (solanaceae) Other 22R-hydroxycholesterol analogs useful in the present invention can 5 be identified through structure-based database searching. Two approaches may be followed. One approach is based on the structure of 22R-hydroxycholesterol. 22R-hydroxycholesterol is subdivided into several building blocks, the database G j WO 2006/044665 PCT/US2005/037023 is searched for compounds that include one or more of the building blocks of 22R-hydroxycholesterol. A refined search based on the results presented in this application may be formulated such that the 22R hydroxy functionality of the 22R-hydroxycholesterol is conserved. Compounds having structural similarity 5 to 22R-hydroxycholesterol are extracted from the database and tested in vitro for their binding affinity to A0. The compounds with the highest binding affinity are selected for further in vivo studies. The second approach is based on the structure of A0. Briefly, in (receptor) structure-based 3D-database searching, the 3D structure of the target molecule AO is determined through NMR analysis, 10 then large chemical databases containing the 3D structures of hundreds of thousands of structurally diverse synthetic compounds and natural products are searched through computerized molecular docking to identify small molecules that can interact effectively with A0. In forming a template 3-D structure of AO, each atom of the backbone of 15 the AO is assigned a position according to a starting conformation, the positions for the atoms of the side chains are assigned according to the internal coordinates of minimum energy for each side chain. The template structure thus obtained is refined by minimizing the internal energy of the template. Based on the refined structure of AO, a host-guest complex is formed by disposing a compound from a 20 compound database around A0. The structure of the host-guest complex is defined by the position occupied by each atom in the complex in a three dimensional referential. A geometry-fit group is formed by selecting the compounds which can be disposed in the target binding site without significant unfavorable overlap with 25 the atoms of the A0. For each compound in the geometry fit group, a predicted binding affinity to the receptor site of AO is determined by minimizing an energy function describing the interactions between the atoms of the compound and those of A0. The minimization of the energy function is conducted by changing the position of the compound such that a guest-host complex structure 30 corresponding to a minimum of the energy function is obtained. The compounds having the most favorable energy interaction with the atoms of the binding site are identified for optional further processing, for example through display and visual inspection of compound AO complexes to identify the most promising compound candidates.
WO 2006/044665 PCT/US2005/037023 The displayed complexes are visually examined to form a group of candidate compounds for in vitro testing. For example, the complexes are inspected for visual determination of the quality of docking or fitting of the compound into the receptor site(s) or pocket(s) of Afl. Visual inspection 5 provides an effective basis for identifying compounds for in vitro testing. After putative binding compounds have been identified, the ability of such compounds to specifically bind to AO is confirmed in vitro and/or in vivo. In another aspect, the present invention provides novel compounds which are rationally designed to bind to A0. Rational design of the novel compounds is 10 based on information relating to the binding site of Afl. The structures of AO and a lead compound is analyzed such that compound structures having possible activity in binding to the binding site of AO are formulated. The structure of the lead compounds is divided into design blocks, the modification of which is probed for influence on the interactions between the 15 lead compound and the binding site of A0. Compounds having different design block combinations are then synthesized and their activity in relation to the identified mechanism is tested. Such tests are conducted in vitro and/or in vivo, in the same manner described above. The information obtained through such tests is then incorporated in a new cycle of rational drug design. The design 20 synthesis-testing cycle is repeated until a lead compound having the desired properties is identified. The lead compound is then clinically tested. In another aspect, the present invention provides novel compounds which inhibit the formation of ADDLs, such as trimers and tetramers by binding to As and forming stable nontoxic oligomers or polymers. 25 The term "treat" or "treatment" as used herein refers to any treatment of a disorder or disease associated with a disease or disorder related to neurotoxicity, or beta-amyloid-induced neurotoxicity, in a subject, and includes, but is not limited to, preventing the disorder or disease from occurring in a subject who may be predisposed to the disorder or disease, but has not yet been diagnosed as 30 having the disorder or disease; inhibiting the disorder or disease, for example, arresting the development of the disorder or disease; relieving the disorder or disease, for example, causing regression of the disorder or disease; or relieving the condition caused by the disease or disorder, for example, stopping the symptoms of the disease or disorder. As used herein, "neurodegenerative ,' I WO 2006/044665 PCT/US2005/037023 disorder" or "neuropathology" is intended to encompass all disorders stated above. The term "prevent" or "prevention," in relation to a disease or disorder related to neurotoxicity, or beta-amyloid-induced neurotoxicity, in a subject, 5 means no disease or disorder development if none had occurred, or no further disorder or disease development if there had already been development of the disorder or disease, or no symptoms to logically observable signs of the disease. Stem cells are characterized by a capacity to self-renew and to generate progeny capable of differentiating into multiple yet distinct cell lineages. 10 Although stem cells derived from early embryos can differentiate into all somatic cell types, those derived from adult tissues are thought to produce only the cell lineages characteristic of the tissues wherein they reside. See, e.g., Evans et al., Nature, 292, 154 (1981); Martin, PNAS USA, 78, 7634 (1981), Pedersen, Reprod. Fetil. Dev., 16, 543 (1994). For example, hematopoietic stem 15 cells resident in bone marrow give rise to only blood elements (Morrison et al., Annu. Rev. Cell. Dev. Biol., 11, 35 (1994)). Stem cells also have been identified in the gut, gonads, skin, and brain of adults (Hull et al., Development, 106, 619 (1989); Morrison et al., Cell, 88, 287 (1997)). Neural stem cells have been proposed as useful vectors for treating diseases of the central nervous system 20 such as neurodegenerative disorders, but their lack of accessibility limits their utility. Recently, several reports demonstrated that some stem cells may have greater plasticity than previously envisioned. Eglitis and Mezey reported that a few donor cells from bone marrow transfused into immunodeficient mice were 25 recovered as macroglia in the host brain (Eglitis et al., PNAS USA, 94, 4080 (1997)). However, the nature of the marrow cells that engrafted in brain was not determined. Azizi et al., PNAS USA, 95, 3908 (1998) infused rat brains with human marrow stromal cells (MSCs) that are capable of expansion, self-renewal, and differentiation into multiple mesenchymal cell lineages, as demonstrated by 30 Owen, J. Cell. Sci. Suppl., 10, 63 (1988) and Aubin, J. Cell. Biochem. Suppl., 30, 73 (1998). The human MSCs migrated in brain in a manner similar to paraventricular astrocytes, but whether the cells differentiated in neural cell types was not defined. In related experiments, Bjornson et al., Science, 238, 534 (1999), showed that neural stem cells differentiated into myeloid and lymphoid WO 2006/044665 PCT/US2005/037023 cell lineages after transplantation into the hematopoietic system of irradiated hosts. Together, these findings suggest that it may be possible to reconstitute a tissue by using stein cells from a separate dermal origin. To determine whether mesenchymal stem cells can adopt neural cell 5 fates, Kopen et al., PNAS USA, 96,,10711 (1999), injected a purified population of murine MSCs into the lateral ventricles of neonatal mice and examined the fate of these cells by immunohistochemistry. By 12 days postinjection, MSCs migrated throughout the forebrain and cerebellum without disruption to the host brain architecture. Some MSCs within the striaturn and the molecular layer of 10 the hippocampus expressed glial fibrillary acidic protein and, therefore, had differentiated into mature astrocytes. MSCs also populated neuron rich regions including the Islands of Calleja, the olfactory bulb, and the internal granular layer of the cerebellum. A large number of MSCs also were found within the external granular layer of the cerebellum. In addition, neurofilament positive 15 donor cells were found within the reticular fonnation of the brain stem, suggesting that MSCs also may have differentiated into neurons. See also, "Bone Marrow Cells as a Source of Neurons for Brain and Spinal Cord Repair," Sanchez-Rames et al., U.S. Pat. No. 6,528,245; Woodbury et al., J. Neurosci. Res, 61, 364 (2000); Hofstetter et al., PNAS, 99, 2199 (2002). Therefore, 20 MSCs are capable of producing differentiated progeny of a different dermal origin after implantation into neonatal mouse brains. These results suggest that native or transgenic stein cells are potentially useful as vectors for treating a variety of central nervous system disorders, and agents that would enhance their ability to differentiate into neuronal cells in vitro or in vivo would be of value. 25 Thus, another aspect of the method provides a method for inducing the differentiation of mammalian, e.g., human, neuronal precursor cells into neuronal cells or astrocytal cells, including mature neurons, by contacting the neuronal precursor cells, in vitro or in vivo, with an effective amount of a compound of formula (I), (II) and/or (III). Thus, the present compounds can be 30 used to pre-condition or to begin differentiation of neuronal precursor cells, such as neuronal stem cells, mesenchymal stem cells, marrow stromal cells (MSCs), multipotent adult stem cells, or embryonic stem cells, in vitro. Once committed to the neuronal lineage, the stem cells can be administered to a target site, as in the CNS, of a patient afflicted with a neurodegenerative disorder. In another WO 2006/044665 PCT/US2005/037023 approach, stem cells and an effective differentiating amount of a compound of formula (I), (II) or (III) can be administered concomitantly, e.g., together or in close sequence, to the target site of the afflicted patient. Of course, direct administration of the present compounds to the patient can cause stimulation and 5 differentiation of the patient's reserve of neuronal precursor cells, including uncommitted adult stem cells, into committed neural precursor cells or into mature, functional neuronal cells in vivo. Stimulation of such neural growth or repair is expected to lead to effective treatments for neurodegenerative disorders. "MAPC" or multipotent adult progenitor cell, refers to a cell that is 10 derived from non-embryonic tissue but which can give rise to cell lineages of all three germ layers (i.e., endoderm, mesoderm, and ectoderm) upon differentiation. This cell has been extensively characterized in U.S. Patent application Serial Nos. 10/048,757 and 10/467,963, the contents of which are incorporated by reference for the description of MAPCs and the isolation 15 thereof, especially from human tissue. MAPCs have been extensively characterized with respect to cell surface marker expression, and are negative for cell surface expression of GlyA, CD44, CD45 and HLA. Other references which disclose pluripotent or multipotent lineage committed or undifferentiated cells that are generally referred to as "stem cells" 20 include U.S. Pat. Nos. 6,090,625; 5,827,735 (mesenchymal stem cells), 6,200,806; 5,843,780 and published U.S. applications 20030008392 (embryonic stem cells); 20030013910 (pluripotent non-embryonic stem cells); 20030073234 (clonal human embryonic stem cell line); 20030161817 (totipotent stem cells from umbilical cord matrix); 20020142457; 20021060509 and 20020164794. 25 In addition, stem cells can be genetically altered, so that they comprise additional copies of one or more genes encoding proteins of interest, or over express an endogenous gene. A genetically altered stem cell may contain DNA encoding enzymes that increase the synthesis of an amino acid such as L-DOPA, under the control of a promoter that directs strong expression of the recombinant 30 protein. Schwartz et al. used transgenic BSCs expressing L-DOPA to treat the brains of mice lesioned with 6-hydroxydopamine. See, Human Gene Therapy, 10, 2539 (1999). Alternatively, the cell may express a gene that can be regulated by an inducible promoter or other control mechanism where conditions WO 2006/044665 PCT/US2005/037023 necessitate highly controlled regulation or timing of the expression of an enzyme. Stem cells useful in the present method can be genetically modified to contain isolated heterologous DNA by introducing isolated heterologous DNA 5 or RNA into the cell by a variety of methods known to those of skill in the art. These methods are generally grouped into four major categories: (1) viral transfer, including the use of DNA or RNA viral vectors, such as retroviruses (including lentiviruses), Simian virus 40 (SV40), adenovirus, Sindbis virus, and bovine papillomavirus, for example; (2) chemical transfer, including calcium 10 phosphate transfection and DEAE dextran transfection methods; (3) membrane fusion transfer, using DNA-loaded membranous vesicles such as liposomes, red blood cell ghosts, and protoplasts, for example; and (4) physical transfer techniques, such as microinjection, electroporation, or direct "naked" DNA transfer. 15 Stem cells or their committed progeny can be administered with the compounds of the invention via localized injection, including by catheter administration, systemic injection, intraperitoneal injection, intracranial or intra spinal injection, intramuscular, intrahepatic, parenteral administration, intraarterial injection, injection into the lateral cerebral ventricles or 20 intraplacental injection. Injection can be directed to multiple sites, such as the sites of trauma or other injury. An important issue concerning the therapeutic use of stem cells is the quantity of cells necessary to achieve an optimal effect. In current human studies of autologous mononuclear bone marrow cells, empirical doses ranging 25 from 1 to 4 x 107 cells have been used with encouraging results. However, different scenarios may require optimization of the amount of cells injected into a tissue of interest. For example, see U.S. Pat. No. 6,767,531 which discloses techniques for bone marrow replacement using autologous stem cell transplantation. Thus, the quantity of cells to be administered will vary for the 30 subject being treated. In a preferred embodiment, between 10 4 to 108, more preferably 105 to 107 , and most preferably, 3 x 107 stem cells of the invention and optionally, 50 to 500 ptg/kg per day of a cytokine can be administered to a human subject. However, the precise determination of what would be considered an effective dose may be based on factors individual to each subject, WO 2006/044665 PCT/US2005/037023 including their size, age, target organ, and amount of time since the unwanted accumulation of substrate began. Therefore, dosages can be readily ascertained by those skilled in the art from this disclosure and the knowledge in the art. It is noted that human subjects are treated generally longer than the mice 5 or other experimental animals, such that treatment has a length proportional to the length of the disease process and effectiveness. The doses may be single doses or multiple doses over a period of several days. Thus, one of skill in the art can scale up from animal experiments, e.g., rats, mice, canines and the like, to humans, by techniques from this disclosure and documents cited herein and the 10 knowledge in the art. The treatment generally has a length proportional to the length of the disease process and drug effectiveness and the subject being treated. An effective amount of stem cells and/or an efficacious compound of the invention can be formulated with a pharmaceutically acceptable carrier to form a 15 pharmaceutical composition before being administered for treatment of a disease related to neurotoxicity. "An effective amount" or "pharmacologically effective amount" refers to the amount of the compound which is required to confer therapeutic effect on the treated subject. The interrelationship of dosages for animals and humans (based on milligrams per square meter of body surface) is 20 described by Freireich et al., Cancer Chemother. Rep., 5_, 219 (1966). Body surface area may be approximately detennined from height and weight of the patient. See, e.g., Scientific Tables, Geigy Pharmaceuticals, Ardley, New York, 1970, 537. Effective doses can be based on in vitro concentrations of the present compounds found to be effective to inhibit the toxicity of AP at concentrations 25 corresponding to known concentrations of As in human AD patients, as fully taught below. Doses of the present compounds useful to stimulate a model neuronal cell line are disclosed below. Effective doses will also vary, as recognized by those skilled in the art, depending on the route of administration, the excipient usage, and the optional co-administration with other therapeutic 30 agents. Toxicity and therapeutic efficacy of the active ingredients can be determined by standard pharmaceutical procedures, e.g., for determining LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and WO 2006/044665 PCT/US2005/037023 therapeutic effects is the therapeutic index and it can be expressed as the ratio
LD
5 0
/ED
50 . Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of 5 affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects. Included in the methods, kits, combinations and pharmaceutical compositions of the present invention are the crystalline forms (e.g., polymorphs), enantiomeric forms, isomeric forms and tautomers of the described 10 compounds and the pharmaceutically-acceptable salts thereof. Illustrative pharmaceutically acceptable salts are prepared from formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, stearic, salicylic, p-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), 15 methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, toluenesulfonic, 2-hydroxyethanesulfonic, sulfanilic, cyclohexylaminosulfonic, algenic, b hydroxybutyric, galactaric and galacturonic acids. The term "prodrug" refers to a drug or compound (active moiety) that elicits the pharmacological action results from conversion by metabolic 20 processes within the body. Prodrugs are generally considered drug precursors that, following administration to a subject and subsequent absorption, are converted to an active or a more active species via some process, such as a metabolic process. Other products from the conversion process are easily disposed of by the body. Prodrugs generally have a chemical group present on 25 the prodrug which renders it less active and/or confers solubility or some other property to the drug, such as an ester or acyl group. Once the chemical group has been cleaved from the prodrug the more active drug is generated. Prodrugs may be designed as reversible drug derivatives and utilized as modifiers to enhance drug transport to site-specific tissues. The design of prodrugs to date 30 has been to increase the effective water solubility of the therapeutic compound for targeting to regions where water is the principal solvent. For example, Fedorak, et al., Am. J. Physiol., 269, G210-218 (1995), describe dexamethasone beta -D-glucuronide. McLoed, et al., Gastroenterol., 106, 405-413 (1994), describe dexamethasone-succinate-dextrans. Hochhaus, et al., Biomed. Chrom., WO 2006/044665 PCT/US2005/037023 6, 283-286 (1992), describe dexamethasone-21-sulphobenzoate sodium and dexamethasone-21-isonicotinate. Additionally, J. Larsen and H. Bundgaard, Int. J. Pharmaceutics, 37, 87 (1987) describe the evaluation of N-acylsulfonamides as potential prodrug derivatives. J. Larsen et al., Int. J. Pharmaceutics, 47, 103 5 (1988) describe the evaluation of N-methylsulfonamides as potential prodrug derivatives. Prodrugs are also described in, for example, Sinkula et al., J. Pharm. Sci., 64, 181-210 (1975). Prodrugs are also useful as synthetic intermediates in the preparation of other compounds of formulas (I), (I), or (III), by synthetic interconversions known to the art. For example, see, I.T. Harrison, 10 Compendium of Organic Synthetic Methods, Wiley-Interscience (1971), for methods useful to interconvert spirostenol substituents. The term "derivative" refers to a compound that is produced from another compound of similar structure by the replacement of substitution of one atom, molecule or group by another. For example, a hydrogen atom of a compound 15 may be substituted by alkyl, acyl, amino, etc., to produce a derivative of that compound. "Plasma concentration" refers to the concentration of a substance in blood plasma or blood serum. "Drug absorption" or "absorption" refers to the process of movement 20 from the site of administration of a drug toward the systemic circulation, for example, into the bloodstream of a subject. "Bioavailability" refers to the extent to which an active moiety (drug or metabolite) is absorbed into the general circulation and becomes available at the site of drug action in the body. "Metabolism" refers to the process of chemical 25 transformations of drugs in the body. "Phannacodynamics" refers to the factors which determine the biologic response observed relative to the concentration of drug at a site of action. "Pharmacokinetics" refers to the factors which determine the attainment and maintenance of the appropriate concentration of drug at a site of action. 30 "Plasma half-life" refers to the time required for the plasma drug concentration to decrease by 50% from its maximum concentration. The use of the term "about" in the present disclosure means "approximately," and encompasses variations in parameters that would arise during practice of the relevant art. Illustratively, the use of the term "about" WO 2006/044665 PCT/US2005/037023 indicates that dosages outside the cited ranges may also be effective and safe, and such dosages are also encompassed by the scope of the present claims. The term "measurable serum concentration" means the serum concentration (typically measured in mg, tg, or ng of therapeutic agent per ml, 5 dl, or 1 of blood seriun) of a therapeutic agent absorbed into the bloodstream after administration. The term "phannaceutically acceptable" is used adjectivally herein to mean that the modified noun is appropriate for use in a phannaceutical product. Pharmaceutically acceptable salts include metallic ions and organic ions. More 10 preferred metallic ions include, but are not limited to appropriate alkali metal (Group Ia) salts, alkaline earth metal (Group Ila) salts and other physiological acceptable metal ions. Exemplary ions include aluminum, calcium, lithium, magnesium, potassium, sodium and zinc in their usual valences. Preferred organic ions include protonated tertiary amines and quaternary ammonium 15 cations, including in part, trimethylamine, diethylamine, NN' dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. Exemplary pharmaceutically acceptable acids include without limitation hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, methanesulfonic acid, acetic 20 acid, formic acid, tartaric acid, maleic acid, malic acid, citric acid, isocitric acid, succinic acid, lactic acid, gluconic acid, glucuronic acid, pyruvic acid oxalacetic acid, fumaric acid, propionic acid, aspartic acid, glutamic acid, benzoic acid, and the like. The compositions of the present invention are usually administered in the 25 form of pharmaceutical compositions. These compositions can be administered by any appropriate route including, but not limited to, oral, nasogastric, rectal, transdermal, parenteral (for example, subcutaneous, intramuscular, intravenous, intramedullary and intradermal injections, or infusion techniques administration), intranasal, transmucosal, implantation, vaginal, topical, buccal, 30 and sublingual. Such preparations may routinely contain buffering agents, preservatives, penetration enhancers, compatible carriers and other therapeutic or non-therapeutic ingredients. The present invention also includes methods employing a pharmaceutical composition that contains the composition of the present invention associated 90 WO 2006/044665 PCT/US2005/037023 with pharmaceutically acceptable carriers or excipients. As used herein, the terms "pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipients" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and 5 the like. The use of such media and agents for ingestible substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the compositions, its use is contemplated. Supplementary active ingredients can also be incorporated into the compositions. In making the compositions of the present invention, the compositions(s) can be mixed with a 10 pharmaceutically acceptable excipient, diluted by the excipient or enclosed within such a carrier, which can be in the fonn of a capsule, sachet, or other container. The carrier materials that can be employed in making the composition of the present invention are any of those commonly used excipients in phannaceutics and should be selected on the basis of compatibility with the 15 active drug and the release profile properties of the desired dosage form. Illustratively, pharmaceutical excipients are chosen below as examples: (a) Binders such as acacia, alginic acid and salts thereof, cellulose . derivatives, methylcellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, magnesium aluminum silicate, polyethylene glycol, gums, polysaccharide acids, 20 bentonites, hydroxypropyl methylcellulose, gelatin, polyvinylpyrrolidone, polyvinylpyrrolidone/vinyl acetate copolymer, crospovidone, povidone, polymethacrylates, hydroxypropylmethylcellulose, hydroxypropylcellulose, starch, pregelatinized starch, ethylcellulose, tragacanth, dextrin, microcrystalline cellulose, sucrose, or glucose, and the like. 25 (b) Disintegration agents such as starches, pregelatinized corn starch, pregelatinized starch, celluloses, cross-linked carboxymethylcellulose, sodium starch glycolate, crospovidone, cross-linked polyvinylpyrrolidone, croscarmellose sodium, microcrystalline cellulose, a calcium, a sodium alginate complex, clays, alginates, gums, or sodium starch glycolate, and any 30 disintegration agents used in tablet preparations. (c) Filling agents such as lactose, calcium carbonate, calcium phosphate, dibasic calcium phosphate, calcium sulfate, microcrystalline cellulose, cellulose powder, dextrose, dextrates, dextran, starches, pregelatinized starch, sucrose, WO 2006/044665 PCT/US2005/037023 xylitol, lactitol, mannitol, sorbitol, sodium chloride, polyethylene glycol, and the like. (d) Surfactants such as sodium lauryl sulfate, sorbitan monooleate, polyoxyethylene sorbitan monooleate, polysorbates, polaxomers, bile salts, 5 glyceryl monostearate, Pluronic T M line (BASF), and the like. (e) Solubilizer such as citric acid, succinic acid, fumaric acid, malic acid, tartaric acid, maleic acid, glutaric acid sodium bicarbonate and sodium carbonate and the like. (f) Stabilizers such as any antioxidation agents, buffers, or acids, and the 10 like, can also be utilized. (g) Lubricants such as magnesium stearate, calcium hydroxide, talc, sodium stearyl fumarate, hydrogenated vegetable oil, stearic acid, glyceryl behapate, magnesium, calcium and sodium stearates, stearic acid, talc, waxes, Stearowet, boric acid, sodium benzoate, sodium acetate, sodium chloride, DL 15 leucine, polyethylene glycols, sodium oleate, or sodium lauryl sulfate, and the like. (h) Wetting agents such as oleic acid, glyceryl monostearate, sorbitan monooleate, sorbitan monolaurate, triethanolamine oleate, polyoxyethylene sorbitan monooleate, polyoxyethylene sorbitan monolaurate, sodium oleate, or 20 sodium lauryl sulfate, and the like. (i) Diluents such lactose, starch, mannitol, sorbitol, dextrose, microcrystalline cellulose, dibasic calcium phosphate, sucrose-based diluents, confectioner's sugar, monobasic calcium sulfate monohydrate, calcium sulfate dihydrate, calcium lactate trihydrate, dextrates, inositol, hydrolyzed cereal solids, 25 amylose, powdered cellulose, calcium carbonate, glycine, or bentonite, and the like. (j) Anti-adherents or glidants such as talc, corn starch, DL-leucine, sodium lauryl sulfate, and magnesium, calcium, or sodium stearates, and the like. 30 (k) Pharmaceutically compatible carrier comprises acacia, gelatin, colloidal silicon dioxide, calcium glycerophosphate, calcium lactate, maltodextrin, glycerine, magnesium silicate, sodium caseinate, soy lecithin, sodium chloride, tricalcium phosphate, dipotassium phosphate, sodium stearoyl WO 2006/044665 PCT/US2005/037023 lactylate, carrageenan, monoglyceride, diglyceride, or pregelatinized starch, and the like. Additionally, drug formulations are discussed in, for example, Remington's The Science and Practice of Pharmacy (2000). Another discussion 5 of drug formulations can be found in Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980. The tablets or granules comprising the inventive compositions may be film coated or enteric-coated. Besides being useful for human treatment, the present invention is also 10 useful for other subjects including veterinary animals, reptiles, birds, exotic animals and farm animals, including mammals, rodents, and the like. Mammal includes a primate, for example, a monkey, or a lemur, a horse, a dog, a pig, or a cat. A rodent includes a rat, a mouse, a squirrel, or a guinea pig. The pharmaceutical compositions of the present invention are useful 15 where administration of an inhibitor of neurotoxicity is indicated. It has been found that these compositions are particularly effective in the treatment of senile cognitive impairment and/or dementia (for example, AD). For treatment of a neurodegenerative disorder, compositions of the invention can be used to provide a dose of a compound of the present invention 20 in an amount sufficient to elicit a therapeutic response, e.g., reduction of Af induced cytoxicity, for example a dose of about 5 ng to about 1000 mg, or about 100 ng to about 600 mg, or about 1 mg to about 500 mg, or about 20 mg to about 400 mg. Typically a dosage effective amount will range from about 0.0001 mg/kg to 1500 mg/kg, more preferably 1 to 1000 mg/kg, more preferably from 25 about 1 to 150 mg/kg of body weight, and most preferably about 50 to 100 mg/kg of body weight. A dose can be administered in one to about four doses per day, or in as many doses per day to elicit a therapeutic effect. Illustratively, a dosage unit of a composition of the present invention can typically contain, for example, about 5 ng, 50 ng 100 ng, 500 ng, 1 mg, 10 mg, 20 mg, 40 mg, 80 mg, 30 100 mg, 125 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 450 mg, 500 mg, 550 mg, 600 mg, 700 mg, 800 mg, 900 mg, or 1000 mg of a compound of the present invention. The dosage form can be selected to accommodate the desired frequency of administration used to achieve the specified dosage. The amount of the unit dosage form of the composition that is administered and the WO 2006/044665 PCT/US2005/037023 dosage regimen for treating the condition or disorder depends on a variety of factors, including, the age, weight, sex and medical condition, of the subject, the severity of the condition or disorder, the route and frequency of administration, and this can vary widely, as is well known. 5 In one embodiment of the present invention, the composition is administered to a subject in an effective amount, that is, the composition is administered in an amount that achieves a therapeutically effective dose of a compound of the present invention in the blood serum of a subject for a period of time to elicit a desired therapeutic effect. Illustratively, in a fasting adult human 10 (fasting for generally at least 10 hours) the composition is administered to achieve a therapeutically effective dose of a compound of the present invention in the blood serum of a subject from about 5 minutes after administration of the composition. In another embodiment of the present invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood 15 serum of a subject at about 10 minutes from the time of administration of the composition to the subject. In another embodiment of the present invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood serum of a subject at about 20 minutes from the time of administration of the composition to the subject. In yet another embodiment of 20 the present invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood serum of a subject at about 30 minutes from the time of administration of the composition to the subject. In still another embodiment of the present invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood serum of a subject at 25 about 40 minutes from the time of administration of the composition to the subject. In one embodiment of the present invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood serum of a subject at about 20 minutes to about 12 hours from the time of administration of the composition to the subject. In another embodiment of the present 30 invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood serum of a subject at about 20 minutes to about 6 hours from the time of administration of the composition to the subject. In yet another embodiment of the present invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood serum of WO 2006/044665 PCT/US2005/037023 a subject at about 20 minutes to about 2 hours from the time of administration of the composition to the subject. In still another embodiment of the present invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood serum of a subject at about 40 minutes to 5 about 2 hours from the time of administration of the composition to the subject. And in yet another embodiment of the present invention, a therapeutically effective dose of the compound of the present invention is achieved in the blood serum of a subject at about 40 minutes to about 1 hour from the time of administration of the composition to the subject. 10 In one embodiment of the present invention, a composition of the present invention is administered at a dose suitable to provide a blood serum concentration with a half maximum dose of a compound of the present invention. Illustratively, a blood serum concentration of about 0.01 to about 1000 nM, or about 0.1 to about 750 nM, or about 1 to about 500 nM, or about 20 15 to about 1000 nM, or about 100 to about 500 nM, or about 200 to about 400 nM is achieved in a subject after administration of a composition of the present invention. Contemplated compositions of the present invention provide a therapeutic effect as compound of the present invention medications over an 20 interval of about 5 minutes to about 24 hours after administration, enabling once a-day or twice-a-day administration if desired. In one embodiment of the present invention, the composition is administered at a dose suitable to provide an average blood serum concentration with a half maximum dose of a compound of the present invention of at least about 1 pg/ml, or at least about 5 ytg/ml, or at 25 least about 10 pg/ml, or at least about 50 pzg/ml, or at least about 100 gg/ml, or at least about 500 sg/ml, or at least about 1000 pg/ml in a subject about 10, 20, 30, or 40 minutes after administration of the composition to the subject. The amount of therapeutic agent necessary to elicit a therapeutic effect can be experimentally determined based on, for example, the absorption rate of 30 the agent into the blood serum, the bioavailability of the agent, and the potency for treating the disorder. It is understood, however, that specific dose levels of the therapeutic agents of the present invention for any particular subject depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, and diet of the subject WO 2006/044665 PCT/US2005/037023 (including, for example, whether the subject is in a fasting or fed state), the time of administration, the rate of excretion, the drug combination, and the severity of the particular disorder being treated and form of administration. Treatment dosages generally may be titrated to optimize safety and efficacy. Typically, 5 dosage-effect relationships from in vitro and/or in vivo tests initially can provide useful guidance on the proper doses for subject administration. Studies in animal models generally may be used for guidance regarding effective dosages for treatment of gastrointestinal disorders or diseases in accordance with the present invention. In terms of treatment protocols, it should be appreciated that 10 the dosage to be administered will depend on several factors, including the particular agent that is administered, the route administered, the condition of the particular subject, etc. Generally speaking, one will desire to administer an amount of the compound that is effective to achieve a serum level commensurate with the concentrations found to be effective in vitro for a period of time 15 effective to elicit a therapeutic effect. Thus, where a compound is found to demonstrate in vitro activity at, for example, a half-maximum effective dose of 200 nM, one will desire to administer an amount of the drug that is effective to provide about a half-maximum effective dose of 200 nM concentration in vivo for a period of time that elicits a desired therapeutic effect, for example, treating 20 a disorder related to high beta-amyloid-induced neurotoxicity and other indicators as are selected as appropriate measures by those skilled in the art. Determination of these parameters is well within the skill of the art. These considerations are well known in the art and are described in standard textbooks. In order to measure and determine the effective amount of a compound 25 of the present invention to be delivered to a subject, serum compound of the present invention concentrations can be measured using standard assay techniques. Contemplated compositions of the present invention provide a therapeutic effect over an interval of about 30 minutes to about 24 hours after 30 administration to a subject. In one embodiment compositions provide such therapeutic effect in about 30 minutes. In another embodiment compositions provide therapeutic effect over about 24 hours, enabling once-a-day administration to improve patient compliance.
WO 2006/044665 PCT/US2005/037023 The present methods, kits, and compositions can also be used in combination ("combination therapy") with another pharmaceutical agent that is indicated for treating or preventing a neurodegenerative disorder, such as, for example, acetylcholinesterase inhibitors (i.e., galantamine, donezepil 5 hydrochloride). When used in conjunction with the present invention, that is, in combination therapy, an additive or synergistic effect may be achieved such that many if not all of unwanted side effects can be reduced or eliminated. The reduced side effect profile of these drugs is generally attributed to, for example, the reduced dosage necessary to achieve a therapeutic effect with the 10 administered combination. The phrase "combination therapy" embraces the administration of a composition of the present invention in conjunction with another pharmaceutical agent that is indicated for treating or preventing a neurodegenerative disorder in a subject, as part of a specific treatment regimen intended to provide a beneficial 15 effect from the co-action of these therapeutic agents for the treatment of a neurodegenerative disorder. The beneficial effect of the combination includes, but is not limited to, pharmacokinetic or pharmacodynamic co-action resulting from the combination of therapeutic agents. Administration of these therapeutic agents in combination typically is carried out over a defined time period (usually 20 substantially simultaneously, minutes, hours, days, weeks, months or years depending upon the combination selected). "Combination therapy" generally is not intended to encompass the administration of two or more of these therapeutic agents as part of separate monotherapy regimens that incidentally and arbitrarily result in the combinations of the present invention. "Combination therapy" is 25 intended to embrace administration of these therapeutic agents in a sequential manner, that is, where each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering 30 to the subject a single tablet or capsule having a fixed ratio of each therapeutic agent or in multiple, single capsules, or tablets for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route. The composition of the present invention can be administered orally or nasogastric, while the other therapeutic WO 2006/044665 PCT/US2005/037023 agent of the combination can be administered by any appropriate route for that particular agent, including, but not limited to, an oral route, a percutaneous route, an intravenous route, an intramuscular route, or by direct absorption through mucous membrane tissues. For example, the composition of the present 5 invention is administered orally or nasogastric and the therapeutic agent of the combination may be administered orally, or percutaneously. The sequence in which the therapeutic agents are administered is not narrowly critical. "Combination therapy" also can embrace the administration of the therapeutic agents as described above in further combination with other biologically active 10 ingredients, such as, but not limited to, an analgesic, for example, and with non drug therapies, such as, but not limited to, surgery. The therapeutic compounds which make up the combination therapy may be a combined dosage form or in separate dosage forms intended for substantially simultaneous administration. The therapeutic compounds that 15 make up the combination therapy may also be administered sequentially, with either therapeutic compound being administered by a regimen calling for two step administration. Thus, a regimen may call for sequential administration of the therapeutic compounds with spaced-apart administration of the separate, active agents. The time period between the multiple administration steps may 20 range from, for example, a few minutes to several hours to days, depending upon the properties of each therapeutic compound such as potency, solubility, bioavailability, plasma half-life and kinetic profile of the therapeutic compound, as well as depending upon the effect of food ingestion and the age and condition of the subject. Circadian variation of the target molecule concentration may also 25 determine the optimal dose interval. The therapeutic compounds of the combined therapy whether administered simultaneously, substantially simultaneously, or sequentially, may involve a regimen calling for administration of one therapeutic compound by oral route and another therapeutic compound by an oral route, a percutaneous route, an intravenous 30 route, an intramuscular route, or by direct absorption through mucous membrane tissues, for example. Whether the therapeutic compounds of the combined therapy are administered orally, by inhalation spray, rectally, topically, buccally, sublingually, or parenterally (for example, subcutaneous, intramuscular, intravenous and intradennal injections), separately or together, each such WO 2006/044665 PCT/US2005/037023 therapeutic compound will be contained in a suitable pharmaceutical formulation of pharmaceutically-acceptable excipients, diluents or other formulations components. For oral administration, the pharmaceutical composition can contain a 5 desired amount of a compound of formula (I), (I) or (III) and be in the form of, for example, a tablet, a hard or soft capsule, a lozenge, a cachet, a troche, a dispensable powder, granules, a suspension, an elixir, a liquid, or any other form reasonably adapted for oral administration. Illustratively, such a pharmaceutical composition can be made in the form of a discrete dosage unit containing a 10 predetermined amount of the active compound such as a tablet or a capsule. Such oral dosage forms can further comprise, for example, buffering agents. Tablets, pills and the like additionally can be prepared with enteric coatings. Phannaceutical compositions suitable for buccal or sublingual administration include, for example, lozenges comprising the active compound 15 in a flavored base, such as sucrose, and acacia or tragacanth, and pastilles comprising the active compound in an inert base such as gelatin and glycerin or sucrose and acacia. Liquid dosage forms for oral administration can include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert 20 diluents commonly used in the art, such as water. Such compositions can also comprise, for example, wetting agents, emulsifying and suspending agents, and sweetening, flavoring, and perfuming agents. Examples of suitable liquid dosage forms include, but are not limited, aqueous solutions comprising the active compound and beta-cyclodextrin or a 25 water soluble derivative of beta-cyclodextrin such as sulfobutyl ether beta cyclodextrin; heptakis-2,6-di-0-methyl-beta-cyclodextrin; hydroxypropyl-beta cyclodextrin; and dimethyl-beta-cyclodextrin. The pharmaceutical compositions of the present invention can also be administered by injection (intravenous, intramuscular, subcutaneous). Such 30 injectable compositions can employ, for example, saline, dextrose, or water as a suitable carrier material. The pH value of the composition can be adjusted, if necessary, with suitable acid, base, or buffer. Suitable bulking, dispersing, wetting or suspending agents, including mannitol and polyethylene glycol (such as PEG 400), can also be included in the composition. A suitable parenteral WO 2006/044665 PCT/US2005/037023 composition can also include an active compound lyophilized in injection vials. Aqueous solutions can be added to dissolve the composition prior to injection. The pharmaceutical compositions can be administered in the form of a suppository or the like. Such rectal formulations preferably contain the active 5 compound in a total amount of, for example, about 0.075 to about 75% w/w, or about 0.2 to about 40% w/w, or about 0.4 to about 15% w/w. Carrier materials such as cocoa butter, theobroma oil, and other oil and polyethylene glycol suppository bases can be used in such compositions. Other carrier materials such as coatings (for example, hydroxypropyl methylcellulose film coating) and 10 disintegrants (for example, croscarmellose sodium and cross-linked povidone) can also be employed if desired. The subject compounds may be free or entrapped in microcapsules, in colloidal drug delivery systems such as liposomes, microemulsions, and macroemulsions. 15 These pharmaceutical compositions can be prepared by any suitable method of pharmaceutics, which includes the step of bringing into association active compound of the present invention and a carrier material or carriers materials. In general, the compositions are uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and 20 then, if necessary, shaping the product. For example, a tablet can be prepared by compressing or molding a powder or granules of the compound, optionally with one or more accessory ingredients. Compressed tablets can be prepared by compressing, in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binding agent, lubricant, inert 25 diluent and/or surface active/dispersing agent(s). Molded tablets can be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid diluent. Tablets of the present invention can also be coated with a conventional coating material such as OpadryTM White YS-1-18027A (or another color) and 30 the weight fraction of the coating can be about 3% of the total weight of the coated tablet. The compositions of the present invention can be formulated so as to provide quick, sustained or delayed release of the compositions after administration to the patient by employing procedures known in the art.
WO 2006/044665 PCT/US2005/037023 When the excipient serves as a diluent, it can be a solid, semi-solid or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, chewable tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, 5 emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), soft and hard gelatin capsules and sterile packaged powders. In one embodiment of the present invention, the manufacturing processes may employ one or a combination of methods including: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, (5) wet 10 granulation, or (6) fusion. Lachman et al., The Theory and Practice of Industrial Pharmacy (1986). In another embodiment of the present invention, solid compositions, such as tablets, are prepared by mixing a therapeutic agent of the present invention with a pharmaceutical excipient to form a solid preformulation composition 15 containing a homogeneous mixture of the therapeutic agent and the excipient. When referring to these preformulation compositions(s) as homogeneous, it is meant that the therapeutic agent is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms, such as tablets, pills and capsules. This solid preformulation is 20 then subdivided into unit dosage forms of the type described herein. Compressed tablets are solid dosage forms prepared by compacting a formulation containing an active ingredient and excipients selected to aid the processing and improve the properties of the product. The term "compressed tablet" generally refers to a plain, uncoated tablet for oral ingestion, prepared by 25 a single compression or by pre-compaction tapping followed by a final compression. The tablets or pills of the present invention may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer 30 dosage component, the latter being in the form of an envelope over the former. A variety of materials can be used for such enteric layers or coatings, including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate. dn WO 2006/044665 PCT/US2005/037023 Use of a long-term sustained release implant may be suitable for treatment of neurodegenerative disorders in patients who need continuous administration of the compositions of the present invention. "Long-term" release, as used herein, means that the implant is constructed and arranged to 5 deliver therapeutic levels of the active ingredients for at least 30 days, and preferably 60 days. Long-term sustained release implants are well known to those of ordinary skill in the art and include some of the release systems described above. In another embodiment of the present invention, the compound for 10 treating a neurodegenerative disorder comes in the form of a kit or package containing one or more of the therapeutic compounds of the present invention. These therapeutic compounds of the present invention can be packaged in the form of a kit or package in which hourly, daily, weekly, or monthly (or other periodic) dosages are arranged for proper sequential or simultaneous 15 administration. The present invention further provides a kit or package containing a plurality of separately-packaged dosage units, adapted for successive daily administration, each dosage unit comprising at least one of the therapeutic compounds of the present invention. This drug delivery system can be used to facilitate administering any of the various embodiments of the 20 therapeutic compounds of the present invention. In one embodiment, the system contains a plurality of dosages to be administered daily or weekly. The kit or package can also contain the agents utilized in combination therapy to facilitate proper administration of the dosage forms. The kits or packages also contain a set of instructions for the subject. 25 The invention will be further described by reference to the following detailed examples, wherein A01 -42 and AO peptide fragments were purchased from American Peptide Co. (Sunnyvale, CA). Polyclonal rabbit anti--amyloid peptide (cat. no. 71-5800) was obtained from Zymed Laboratories (San Francisco, CA). [22- 3 H]R-hydroxycholesterol (sp. act. 20 Ci/mmol) was 30 synthesized by American Radiolabeled Chemical (St Louis, MO). Cholesterol, 22R-hydroxycholesterol, 22S-hydroxycholesterol, pregnenolone, 17a hydroxypregnenolone, progesterone, dehydroepiandrosterone (DHEA) and 3 amino-9-ethylcarbazole (AEC) were purchased from Sigma-Aldrich (St. Louis, MO). Cell culture supplies were purchased from GIBCO (Grand Island, NY), Al WO 2006/044665 PCT/US2005/037023 and cell culture plasticware was from Coming (Coning, NY). Electrophoresis reagents and materials were supplied from Bio-Rad (Richmond, CA). All other chemicals used were of analytical grade and were obtained from various commercial sources. 5 Example I Tissue samples All human tissue samples were obtained from the Harvard Brain Tissue Resource Center (Belmont, MA). Samples for steroid measurements were either 10 snap frozen or passively frozen in liquid nitrogen. Brain hippocampus and frontal cortex samples were obtained from 19 patients, 12 AD (6 men and 6 women) and 7 age-matched control patients (4 men and 3 women). AD patients were classified by the Harvard Tissue Resource Center as having " severe AD." Mean age for all patients was 74.6+7.2 years for AD patients and 73.4+10.5 15 years for control. Mean post-mortem interval was 10.2 hours for AD patients and 14.7 hours for control. Protocols for the use of human tissue were approved by the Georgetown University Internal Review Board. Purification and measurement of 22R-hydroxycholesterol Samples were extracted and purified by reverse phase HPLC as 20 previously described. Brown et al., J. Neurochem. 74, 847-859 (2000). Fractions containing 22R-hydroxycholesterol were collected (retention time of 22R-hydroxycholesterol=55minutes) and levels of 22R-hydroxycholesterol were determined using the cholesterol oxidase assay. Gamble et al., J. Lipid Res. 19, 1068-1070 (1978). 25 Cell culture, cellular toxicity & viability assays Rat PC12 cells were cultured as previously described. Yao et al., Brain Res. 889, 181-190 (2001). Human NT2 precursor (Ntera2/D1 teratocarcinoma) cells were obtained from Stratagene (La Jolla, CA) and cultured following the instructions of the supplier. Differentiated human NT2 neurons (NT2N) were 30 obtained after treatment of the NT2 precursor cells with retinoic acid. Andrews, Dev. Biol., 103, 285-293 (1984). AP was dissolved in media and used either in the aggregated (left overnight at 4'C) or soluble (containing oligomers such as dimers and tetramers) forms examined by electrophoresis as previously described. Yao et al., Brain Res. (2001). Cellular toxicity for As and AP
A')
WO 2006/044665 PCT/US2005/037023 fragments was assayed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay (Trevigen, Gaithersburg, MD) as previously described. Id. Cell viability was measured using the trypan blue exclusion method as previously described. Id. In brief, for these studies, cells were treated 5 for 72 hours with steroids in the presence or absence of increasing concentrations of Ap. At the end of the incubation, the cells were washed three times with PBS and incubated for 15 minutes with 0.1 % trypan blue stain solution at room temperature. After washing three times with PBS, 0.1 N NaOH was added to the cells and trypan blue staining was quantified using the Victor 10 quantitative detection spectrophotometer (EGG-Wallac, Gaithersburg, MD) at 450 nm. Cell protein levels were determined in the same samples by the method of Bradford (Bradford, Anal. Biochem., 72, 248-254 (1976)), where coomassie blue staining is detected at 590 nm. Cholesterol-protein binding blot assay (CPBBA) 15 Purified Ap1 2 protein (50pM) or various AP fragments (50 [M) and 3H 22R-hydroxycholesterol were incubated either alone or in the presence of increasing concentrations of unlabeled 22R-hydroxycholesterol in 20 pl volume for 24 hours at 37'C. At the end of the incubation time, samples were separated by 1.5% agarose (Type I-B) gel electrophoresis and transferred to nitrocellulose 20 membrane (Schleicher & Schuell, Keene, NH) in 10XSSC buffer. The membrane was exposed to tritium-sensitive screen and analyzed by phosphoimaging using the Cyclone Storage phosphor system (Packard BioScience, Meridien, CT). Image-densitometric analysis was performed using the OptiQuant software (Packard). This method allows for the separation, 25 visualization and identification of AP complexes, which have incorporated radiolabeled cholesterol (Yao, Z. & Papadopoulos, V., manuscript submitted) and 22R-hydroxycholesterol under native conditions. Low molecular weight unincorporated 22i-hydroxycholesterol is separated and eliminated during electrophoresis. 30 AP aggregation assay Purified AP1-42 protein (50 mM) in cell culture media was incubated either alone or in the presence of increasing concentrations of 22R hydroxycholesterol for 24 hours at 37*C. At the end of the incubation, proteins were separated by SDS-PAGE on 4-20% gradient acrylamide-bis-acrylamide gel Aq WO 2006/044665 PCT/US2005/037023 at 125V for 2 hours. Proteins were visualized by coomassie blue staining. AP species were identified by immunoblot analysis. Yao, Z. et al., Brain Res. (2001). Immunoblot analysis 5 The membrane with the 22R-hydroxycholesterol-Ap peptide complexes was then used to examine As levels. Membranes were blocked by incubating the nitrocellulose in 5% milk and treated for immunodetection of AP using ECL reagents (Amersham-Pharmacia, Piscataway, NJ). Li et al., Proc. Natl. Acad. Sci. USA, 98, 1267-1272 (2001). Anti-Ap antibody and secondary antibodies 10 were used at 0.2 tg/ml and 1:5000 dilution, respectively. Peptide modeling and 22R-hydroxycholesterol docking Computer docking of 22R-hydroxycholesterol with Ap 17 .40 and Ap 25
-
35 was accomplished using a As structure generated from the solution structure of Ap 1
.
4 oMet(O) (MMDB Id: 7993 PDB Id: 1BA) resulting from data generated by 15 CD and NMR spectroscopy. Watson et al., Biochemistry, 3, 12700 (1998). The Met(O) SME 35 residue was replaced by Met retaining the adjacent backbone dihedral angles and the coordinates for residues 17-40 extracted. The 22R hydroxycholesterol structure was developed using the Alchemy 2000 program (Tripos, St. Louis, MO). The docking was accomplished using Monte Carlo 20 simulated annealing (Li et al., Proc. Natl. Acad. Sci. USA, (2001)) and implemented in modified versions of Autogrid/Autodock. Morris et al., J. Comput. Chem. 19, 1639-1662 (1998). The conformation of minimum energy of approximately 109 conformations was evaluated. Five sessions consisting of 100 runs, each starting at a random initial relative location and orientation of the 25 ligand with the target were executed. Each run was comprised of 100 annealing cycles using about 2 x 104 improvement steps. The total computation time using the modified program was about 15 minutes using a 1.7 GHz, 1GB RAM PC. Statistics Statistical analysis was performed by one-way analysis of variance 30 (ANOVA) and unpaired Student's t test using the INSTAT 3.00 package (GraphPad, San Diego, CA).
AA
WO 2006/044665 PCT/US2005/037023 Results As depicted in Fig. 2, endogenous 22R-hydroxycholesterol levels in human brain were measured by the cholesterol oxidase assay after HPLC purification. Data presented is means ± SEM for duplicate measurements from 5 12 AD and 7 age-matched control samples. Fig. 2 shows that levels of 22R hydroxycholesterol in hippocampus of AD patient's brain specimens were decreased by 60% (p=0.04) compared to age-matched controls. 22R hydroxycholesterol levels were also decreased by 50% in frontal cortex of AD patient's brain specimens compared to age-matched controls, although in a non 10 significant manner. PC12 cells were treated for 24 hours with the indicated concentrations of Ap 14 2 in the absence or presence of increasing concentrations of 22R hydroxycholesterol (Fig. 3A), cholesterol (Fig. 3B), pregnenolone (Fig. 3C) or 17a-hydroxypregnenolone (Fig. 3D), DHEA (Fig. 3E) or 22S 15 hydroxycholesterol (Fig. 3F). Results shown are means -SD (n=6-12). The ability of 22R-hydroxycholesterol to rescue rat PC12 neuronal cells from Ap induced cytotoxicity was examined using the mitochondrial diaphorase assay MTT. ApI- 42 induced a dose-dependent neurotoxicity that reached 26% 20 (p<0.001) and 40% (p<0.001) cell death in the presence of 5.0 and 50 pLM AP, respectively (Fig. 3A). Increasing concentrations of 22R-hydroxycholesterol did not affect PC 12 cell viability, although a non-significant improvement was seen in the presence of 10 and 100 ptM of 22R-hydroxycholesterol (Fig. 3A). 22R hydroxycholesterol was able to rescue all the cells from 25 tM Ap-induced 25 cytotoxicity (p<0.001) and to rescue 50% (p<0.01) of the cells dying in the presence of 50 ptM AP (Fig. 3A). Interestingly, 22R-hydroxycholesterol was effective only when present at the same time with Ap. Pretreatment of PC12 cells with 22R-hydroxycholesterol followed by treatment with AP failed to offer any protection to the cells (data not shown). 30 The neuroprotective effect of 22R-hydroxycholesterol could not be replicated using either its precursor cholesterol (Fig. 3B) or its metabolite pregnenolone (Fig. 3C). In contrast, both cholesterol and pregnenolone alone were toxic to the cells. Moreover, the presence of cholesterol accentuated the WO 2006/044665 PCT/US2005/037023 toxic effect of low concentrations of Ap. 17a-hydroxypregnenolone alone was also toxic to the cells (Fig. 3D). 100 tM DHEA had a positive effect on cell viability. The same concentration of DHEA protected against the 5 RM (p<0.001), but not 50 [tM, Ap-induced cytotoxicity (Fig. 3E). The effect of 22R 5 hydroxycholesterol was stereospecific because 22S-hydroxycholesterol not only failed to protect against the AP1-induced neurotoxicity, but at a 100 pM concentration was neurotoxic (Fig. 3F). It should be noted that, in the presented studies, aggregated AP (left overnight at 4*C) was used. In separate experiments, soluble AP (containing 10 oligomers) was directly added to PC12 cells and found to be toxic (data not shown). 22R-hydroxycholesterol also protected against the As oligomer-induced toxicity (not shown). The neuroprotective effect of 22R-hydroxycholesterol was not restricted to PC12 cells but was replicated on differentiated human NT2N neurons (Fig. 4). 15 Differentiated human NT2N neurons were treated for 72 hours with 25 ptM Ap. 42 in the presence or absence of 22R-hydroxycholesteol. 25 ptM As inhibit by 50% (p<0.001) human neuron viability, while 1 and 10 piM 22R hydroxycholesterol protected by 50% (p<0.01) and 100% (p<0.001), respectively, against the Ap-induced toxicity (Fig. 4). To assess whether 22R 20 hydroxycholesterol rescues human NT2 cells against other toxic insults, NT2 cells were treated for three days with 5 mM glutamate in the presence or absence of 1 to 50 piM 22R-hydroxycholesterol. Glutamate induced a 30% decrease in cell viability, determined using the MTT assay and the presence of 22R hydroxycholesterol failed to protect the cells (data not shown). 25 The results obtained from using MTT assay were further confinned with the trypan blue dye exclusion assay. PC12 cells were treated for 72 hours with increasing concentrations of A 142 (Fig. 5A) or Ap 25 -3 5 (Fig. 5B) in the presence or absence of 100 pM 22R-hydroxycholesterol or DHEA. NT2 cells were treated for 72 hours with increasing concentrations of A 142 (Fig. 5C) or AP 25
-
35 30 (Fig. 5D) in the presence or absence of 25 gM 22R-hydroxycholesterol or DHEA. Levels of viability were measured using the trypan blue assay as described under Materials and Methods. Results are expressed as fold trypan blue stained cells per total cell protein over control untreated cells. Results Ar.
WO 2006/044665 PCT/US2005/037023 shown are means ± SD (n=6-12). Figs. 5A and 5C show that 22R hydroxycholesterol rescued both the rat PC12 (Fig. 5A) and human NT2 (Fig. 5C) cells from Ap 142 -induced cell death. In contrast, DHEA only protected the rat PC12 cells from AP 1 42 -induced cell death but not NT2 cells (Figs. 5A and 5 5C). Neither 22R-hydroxycholesterol nor DHEA could rescue the PC12 and NT2 cells from the AP 25
.
35 -induced cell death (Figs. 5B and 5D). The ability of 22R-hydroxycholesterol to alter AP aggregation was also examined. Purified AP 142 protein (50 ptM) in cell culture media was incubated either alone or in the presence of increasing concentrations of the 22R 10 hydroxycholesterol for 24 hours at 37 0 C. At the end of the incubation proteins were separated by SDS-PAGE and visualized by coomassie blue (Fig. 6A). As species formed were identified by immunoblotting using an anti-AD polyclonal antiserum (Fig. 6B). AP aggregation can be seen on the top of the gel and it is absent in control-media lane. Figs. 6A and 6B show that 22R-hydroxycholesterol 15 did not affect As aggregation identified by immunoblot analysis (Fig. 6B) of the coomassie blue stained gels (Fig. 6A). A 100 kDa band recognized by the As polyclonal antiserum used in all samples, including control-media, probably reflects non-specific binding of the antiserum. The mechanism of action of 22R-hydroxycholesterol was then examined. 20 Considering that 22R-hydroxycholesterol was neuroprotective only when in presence of AP, the direct interaction between 22R-hydroxycholesterol and AP was explored with a novel method, the CPBBA method. Co-incubation of radiolabeled 22R-hydroxycholesterol together with Api42 for 24 hours at 37'C demonstrated the presence of a high molecular weight radiolabeled band (Fig. 25 7A) recognized by an antibody specific to AP (Fig. 7B). The specificity of the radiolabeling of Api- 42 by 22R-hydroxycholesterol was demonstrated by competition studies using unlabeled 22R-hydroxycholesterol (Fig. 7A). In these studies, 50 and 200 piM 22R-hydroxycholesterol inhibited by 50 and 90%, respectively, the binding of radiolabeled 22R-hydroxycholesterol to 50 pM Ap1 30 42, as indicated by image analysis of the radiolabeled AP142 (Fig. 7A). Equal loading of Ap1-42 in the incubation reactions and in CPBBA was assessed by immunoblot analysis of the radiolabeled Ap 14 2 (Fig. 7B). It should be noted that, despite the decreased radiolabeling of Api 4 2 observed in the presence of A'7 WO 2006/044665 PCT/US2005/037023 50-200 pM 22R-hydroxycholesterol, there were no differences in the amount of Ap 1
-
42 present in each lane. These data demonstrate that, under native conditions, 22R-hydroxycholesterol binds to AP. Using CPBBA and various As synthetic peptides, the 22R-hydroxycholesterol-binding site in As was mapped 5 to amino acids 17-40 of AP (Fig. 7C and 7E). Interestingly peptide As25-35, which maintained its neurotoxicity in the presence of 22R-hydroxycholesterol (Fig. 7B and 7D), did not bind 22R-hydroxycholesterol (Fig. 7C). These data were further confirmed using computational docking simulations. The docking results show that Ap 17
.
4 o forms a pocket where 22R-hydroxycholesterol could 10 dock (Fig. 7D). The pocket formed by amino acids G 29
A
3 0
I
31 captures the C 27 -29 atoms of 22R-hydroxycholesterol. The orientation R, versus S, is permissive for 22R-hydroxycholesterol docking. Similar studies using Ap25-35 indicated that, despite the presence of some of the amino acids present in the 19-36 area, the docking energy of Ap 2 5 35 for 22R-hydroxycholesterol (-6.05 10 kcal/mol) is high 15 relative to Ap1 7 -o (-8.6939 kcal/mol) and to ApiA2 (-9.6960 kcal/mol), suggesting that this steroid does not bind to Ap 2 5
-
35 in agreement with the CPBBA data. Discussion The levels of 22R-hydroxycholesterol were found to be lower in AD 20 patient's brain specimens compared to age-matched controls. Levels of 22R hydroxycholesteral were significantly decreased in hippocampus, a structure in the limbic system of the brain that is critical to cognitive functions, as learning and memory, and is affected in AD. The physicological function of AP is to control cholesterol transport (Yao et al., FASEB, 16,1677-1679 ( . Based 25 on this finding, the decrease of 22R-hydroxycholesterol might be due to the overproduction of As in AD patient's brain (Roher et al., J. Biol. Chem., 286, 3072-3083 (1993); Younkin, J. Physiol., 92, 289-292 (1998)) that blocks cholesterol trafficking or decreases cholesterol uptake by the cells, thus affecting the availability of the substrate cholesterol for neurosteroid formation resulting 30 in decreased synthesis of 22R-hydroxycholesterol in AD patient's brain. Alternatively, increased de novo synthesis of pregnenolone and DHEA from cholesterol in AD brain specimens will also exhaust the available intermediate 22R-hydroxycholesterol in AD. The presence of increased levels of A O WO 2006/044665 PCT/US2005/037023 pregnenolone and DHEA in AD hippocampus (Brown et al., Neurobiology of Aging, 24, 57-65 (2003)), is induced by AP (Brown, et al., J. Neurochem., 74, 847-859 (2000)). It is also possible that both events, Ap-induced decrease in cholesterol trafficking and increase in cholesterol metabolism might occur in AD 5 and lead to decreased 22R-hydroxycholesterol levels. For these studies, a well-established rat PC12 neuronal cell model was used. However, the neuroprotective effect of 22R-hydroxycholesterol was not restricted to rodent neurons but it was also seen in human NT2 and NT2N neuronal cells. NT2 cells is a clonal line of human teratocarcinoma cells and 10 NT2N, derived from NT2 cells, are post-mitotic, terminally differentiated neurons that possess cell surface markers consistent with neurons of the central nervous system. Andrews, Dev. Biol. (1984). 22R-hydroxycholesterol was found to protect both rat and human neurons from Ap-induced toxicity in a dose dependent manner with IC 50 s of 10 and 3 pM for PC12 and NT2T cells, 15 respectively. Treatment of the cells with 22R-hydroxycholesterol offered full protection against AP used at 25 pM concentration and 50% neuroprotection against the peptide used at 50 pM. Using the MTT assay, which measures the formation of blue formazan, in addition to the effect of 22R-hydroxycholesterol, the neuroprotective 20 properties of various steroids involved in the metabolism of cholesterol was examined. From the steroids tested on Ap-induced PC12 neurotoxicity, all were toxic except for 22R-hydroxycholesterol and DHEA. The neuroprotective effect of DHEA on rodent neurons is in agreement with previous studies. Kimonides et al., Proc. Natl. Acad. Sci. USA, 95, 1852-1857 (1998); Cardounel et al., Proc. 25 Soc. Exp. Biol. Med., 222, 145-149 (2000). However, in contrast to 22R hydroxycholesterol, DHEA had no effect on Ap-induced human NT2 cell death, suggesting that the effect of 22R-hydroxycholesterol is not species specific, probably because this steroid interacts directly with Ap. The precursor of 22R hydroxycholesterol, cholesterol, was found to be neurotoxic. However, the 30 presence of an hydroxyl group at carbon 22(R) not only relieves the toxic effect of cholesterol but also protects against Ap-induced neurotoxicity. The specificity of the effect of 22R-hydroxycholesterol is further evidenced by the observation Ao WO 2006/044665 PCT/US2005/037023 that its enantiomer 22S-hydroxycholesterol is inactive and at high concentrations neurotoxic. The direct interaction between 22R-hydroxycholesterol and AP was shown using a novel assay, the CPBBA. This assay allows for the study and 5 visualization of the direct interaction, under native conditions, between the radiolabeled steroid and AP, or AP peptide fragments. Radiolabeled 22R hydroxycholesterol binds As and the unlabeled 22R-hydroxycholesterol displaces the bound steroid. CPBBA indicated that 22R-hydroxycholesterol binds to ApI- 42 and Ap1 7 .40, but barely interacts with API.
4 o. Mass spectrometric 10 analysis of purified amyloid plaques revealed that Ap1i 42 is the principal component of amyloid deposits, therefore, Ap 14 2 is believed to be the main culprit in the pathogenesis of AD. Roher et al., J. Biol. Chem. (1993); Younkin, Physiol. (1998). The shorter AP form of 40 amino acids is believed to have no pathologic effect (Brown et al., J. Neurochem. (2000)) and is less abundant in 15 AD brain (Roher et al., J. Biol. Chem. (1993); Younkin, J. Physiol. (1998)). Computational modeling simulations based on the reported structure of As indicated that amino acids 19-36 capture capture the side chain of 22R hydroxycholesterol when the hydroxyl group has the R orientation. Interestingly, the peptide A3 2 5
-
35 _hat is known for its toxic effects (Schubert et al., Proc. Natl. 20 Acad. Sci. USA, 92, 1989-1993 (1995)) retained its neurotoxic property even in presence of 22R-hydroxycholesterol. Computational modeling simulations and CPBBA failed to show an interaction between 22R-hydroxycholesterol and peptide Apj 2 s 35 , suggesting that it is the three dimensional conformation of Api. 42 and Ap1740 that confers the ability of amino acids 19-36 to interact with 22R 25 hydroxycholesterol rather than the primary amino acid sequence. 22R-hydroxycholesterol binding to amino acids 17-40 of Ap- 4 2 leads to the protection/rescuing of both rodent and human neuronal cells from the ApI 4 2 induced cytotoxicity and cell death. The exact mechanism by which 22R hydroxycholesterol acts to block the neurotoxic effect of As is not known. 30 However, the data presented herein indicated that it does not affect AP polymerization. Binding of 22R-hydroxycholesterol to AIp42 might either change the conformation of the AP monomer or polymer, thus rendering it inactive, or prohibit As from interacting with the cell or activating intracellular WO 2006/044665 PCT/US2005/037023 mechanism mediating its toxic effect. Thus, the low levels of 22R hydroxycholesterol in AD patient's brain compared to age-matched controls, in addition to the increased production of A 1-42 in AD brains, results in decreased/lost ability of the brain to fight against the Ap1.4 2 -induced 5 neurotoxicity. This might be particularly true for presenilin 1-liked familial Alzheimer's disease (FAD) patients, who have the highest levels of Ap 1
-
42 . Borchelt et al., Neuron, 17, 1005-1013 (1996). Example II 10 Materials Al1 42 peptide was purchased from American Peptide Co. (Sunnyvale, CA). 22R-hydroxycholesterol (SP222) was purchased from Sigma (St Louis, MO). [22- 3 H]R-hydroxycholesterol (sp. act. 20 Ci/mmol) was synthesized by American Radiolabeled Chemical (St Louis, MO). The 22R-hydroxycholesterol 15 derivatives (SP223-238) were purchased from Interbioscreen (Moscow, Russia). Cells culture supplies were purchased form GIBCO (Grand Island, NY) and cell culture plasticware was from Corning (Corning, NY) and Packard BioSciences Co. (Meriden, CT). In silico screening for 22R-hydroxycholesterol derivatives 20 The Interbioscreen Database of naturally occurring entities was screened for compounds containing the 22R-hydroxycholesterol structure using the ISIS software (Information Systems, Inc., San Leandro, CA). The structure of the selected and tested 22R-hydroxycholesterol (SP222) and derivatives (SP223 238) are shown in Fig.1 and the denomination, chemical name and origin for 25 each of these compounds is shown in Table 1. Cell culture and treatments PC12 cells (rat pheochromocytoma neurons) from ATCC (Manassas, VA) were cultured at 37 *C and 5% CO 2 in RPMI 1640 medium devoid of glutamine and supplemented with 10% fetal bovine serum and 5% horse serum. 30 Yao Z, Drieu K and Papadopoulos V., The Gingko biloba extract EGb 761 rescues PC12 neuronal cells from #-amyloid-induced cell death by inhibiting the formation of -amyloid-derived diffusible neurotoxic ligands, Brain Res, 889, 181-190 (2001). Cells were seeded in 96-well plates (8 x 10 4 cells/well). After an overnight period of incubation, increasing concentrations of aggregated AO -1; WO 2006/044665 PCT/US2005/037023 (0.1, 1 and 10 pM) were added to the cells in the presence or absence of the indicated concentrations of the SP compounds to be tested. After 72-hours incubation time various parameters, markers of cell viability, were determined. Mouse MA-10 tumor Leydig cells were maintained at 37'C in DMEM/Ham's 5 F12 (Biofluids, Rockville, MD) medium supplemented with 5% heat-inactivated fetal calf serum and 2.5% horse serum in 5% CO 2 . Cells were plated on 96-well plates at the density of 2.5x1 04 cells/well for overnight. The cells were stimulated with the indicated concentrations of the various SP compounds in 0.2 ml/well serum-free medium for 2 hours. The culture medium was collected and 10 tested for progesterone production by radioimmunoassay. MTT cytotoxicity assay The cellular toxicity of AO was assessed using the 3-(4,5 dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay (Trevigen, Gaithersburg, MD). Briefly, 10 pl of the MTT solution were added to the cells 15 cultured in 100 [l medium. After an incubation period of 4 hours, 100 tl of detergent were added and cells were incubated overnight at 37*C. Formazan blue formation was quantified at 600 nm and 690 nm using the Victor quantitative detection spectrophotometer (EGG-Wallac, Gaithersburg, MD) and the results expressed as (D0 600 - D0 690 ). Although the MTT assay has been widely used to 20 assess cytotoxicity in neuronal cells treated with AO it has been suggested that the results obtained in the presence of various steroids might reflect the A# dependent vesicle recycling leading to increased MTT formazan exocytosis and loss. Liu Y and Schubert D, Steroid hormones block amyloid fibril-induced 3 (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) formazan 25 exocytosis: relationship to neurotoxicity, J. Neurochem., 19, 1639-1662 (1998). For that reason, additional cytotoxicity and cell viability assays were used. Trypan blue cell viability measurement Cell viability was measured using the trypan blue exclusion method as we previously described. Yao et al., The Gingko biloba extract EGb 761 rescues 30 PC12 neuronal cells from -amyloid-induced cell death by inhibiting the formation of #-amyloid-derived diffusible neurotoxic ligands, Brain Res., 889, 181-190 (2001). In brief, cells were treated for 72 hours with SP compounds in the presence or absence of increasing concentrations of A0. At the end of the incubation, cells were washed three times with PBS and incubated for 15 WO 2006/044665 PCT/US2005/037023 minutes with 0.1 % trypan blue stain solution at room temperature. After washing three times with PBS, 0.1 N NaOH was added to the cells and trypan blue was quantified using the Victor quantitative detection spectrophotometer at 450 nm. 5 Measurement of membrane potential Cells viability was also assessed using the luminescence-based kit CytoLiteTM (Packard BioScience Co.) according to the recommendations of the manufacturer. Briefly, cells were cultured and treated in 96-well plates and after 72-hours incubation time, 25 pl of Activator solution was added to the cells 10 followed by 150 gl of Amplifier solution. Luminescence was measured on a TopCount NXTTM counter (Packard BioSciences Co.) following a 5 minute precount delay. Determination of cellular ATP levels Cellular ATP concentrations were measured using the ATPLite-MTM 15 luminescence assay (Packard BioSciences Co.). For this assay, cells were cultured on black 96-well ViewPlateTM and the ATP concentrations were measured on a TopCount NXTTM counter (Packard BioSciences Co.) following the recommendations of the manufacturer. Radioimmunoassay 20 Progesterone production by MA- 10 cells was measured by radioimmunoassay using anti-progesterone antisera (ICN, Costa Mesa, CA), following the conditions recommended by the manufacturer. The progesterone production was normalized by the amount of protein in each well. Radioinununoassay data was analyzed using the MultiCalc software (EG&G 25 Wallac, Gaithersburg, MD). 22R-hydroxycholesterol-protein binding blot assay (CPBBA) Purified AP (50ptM) and 3 H-22R-hydroxycholesterol were incubated either alone or in the presence of 100 pM of unlabeled 22R-hydroxycholesterol (SP-222) or the various 22R-hydroxycholesterol derivatives in 20 l volume for 30 8 or 24 hours at 37'C. At the end of the incubation time, samples were separated by 1.5% agarose (Type I-B) gel electrophoresis under native conditions and transferred to nitrocellulose membrane (Schleicher&Schuell, Keene, NH) in 10XSSC buffer. The membrane was exposed to tritium-sensitive screen and analyzed by phosphoimaging using the Cyclone Storage phosphor system WO 2006/044665 PCT/US2005/037023 (Packard BioScience). Image-densitometric analysis was performed using the OptiQuant software (Packard BioScience). This method allows for the separation, visualization and identification of AB complexes, which have incorporated radiolabeled cholesterol (Yao Z. and Papadopoulos-V., Function of 5 p-amyloid in cholesterol transport: a lead to neurotoxicity, FASEB J., 16,1677 1679 (2002)), and 22R-hydroxycholesterol (Yao et al., J. Neurochem., 83, 1110 1119 (2,02)), or 22R-hydroxycholesterol derivatives under native conditions. Low molecular weight unincorporated 22R-hydroxycholesterol and derivatives are separated and eliminated during electrophoresis. 10 Peptide modeling and docking simulations Computer docking of 22R-hydroxycholesterol and 16 of its derivatives with Ao1.
42 was accomplished using an AO structure initialized by the solution structure of A# 1
.
4 OMet(O) (MMDB Id: 7993 PDB Id: IBA) resulting from data generated by CD and NMR spectroscopy. Watson A.A., Fairlie D.P. and Craik 15 D.J., Solution structure of methionine-oxidized amyloid beta-peptide (1-40). Does oxidation affect conformational switching?, Biochem., 37, 12700-12706 (1998). The Met(O) SME 35 residue was replaced by Met retaining the adjacent backbone dihedral angles and the 141 and A42 residues appended. The energy of the structure was then minimized using the Alchemy 2000 program (Tripos, 20 St. Louis, MO). The 22R-hydroxycholesterol derivative structures were also generated using Alchemy 2000. Molecular docking was accomplished using Monte Carlo simulated annealing as previously described. Li et al., Cholesterol binding at the cholesterol recognition/ interaction amino acid consensus (CRAC) of the peripheral-type benzodiazepine receptor and inhibition of steroidogenesis 25 by an HIV TAT-CRAC peptide, Proc. Natl. Acad. Sci. USA, 98, 1267-1272 (2001), implemented in modified versions of Autogrid/Autodock. Morris et al., Distributed automated docking of flexible ligands to proteins: parallel applications of AutoDock 2.4, J. Comput. Chem., 19, 1639-1662 (1998). For each of the compounds/AO pairs approximately 108 conformations were 30 evaluated to obtain the selected one of minimum energy. Three sessions consisting of 100 runs, each starting at a random initial relative location and orientation of the ligand with respect to the target were executed. Each run was comprised of 100 annealing cycles using about 2 x 10 4 improvement steps. The WO 2006/044665 PCT/US2005/037023 average computation time for each ligand/target pair was about 22 hours using a 1.7 GHz, 1 GB RAM PC. Statistical analysis Statistical analysis was performed by one-way analysis of variance 5 (ANOVA) and unpaired Student's t test using the INSTAT 3.00 (GraphPad, San Diego, CA). Results Three days exposure of PC12 cells to increasing concentrations of AO resulted in dose-dependent cell death (Fig. 8), reaching a maximum of 50% of 10 the cells, in agreement with our previous data. Yao et al., J Neurochem., 8, 1110-1119 (2002); and Yao et al., The Gingko biloba extract EGb 761 rescues PC12 neuronal cells from f-amyloid-induced cell death by inhibiting the formation of 0-amyloid-derived diffusible neurotoxic ligands, Brain Res., 889, 181-190 (2001). To stay close to the concentrations of AO present in AD brain, 15 0.1-10 pM concentrations of AO were used. The compounds tested for their neuroprotective properties were examined at 30 and 50 AM concentrations (Figs. 9-15). Figs. 9-11 show the effect of the lead compound 22R-hydroxycholesterol (SP222) and the compounds containing the 22R-hydroxycholesterol structure 20 (SP223-238) on Af-induced neurotoxicity determined using the MTT assay, a measurement of the NADPH diaphorase activity. Figs. 9-11 show the effects of these compounds on 0.1, 1.0 and 10.0 pM Af-induced neurotoxicity, respectively, expressed as a percentage of inhibition of the NADPH diaphorase activity. The 100% inhibition level corresponds to the decrease of the blue 25 formazan formation induced by AO administered alone. SP222 protects PC12 cells against AO 0.1 pM and 1 pM but provides a limited neuroprotection against AO given at 10 pM. It should be noted that a big variability was observed for the effect of SP-222 on high concentrations of AO, depending on the passage of the cells used. SP228, SP229, SP233, SP235, 30 SP236, SP237 and SP238 displayed neuroprotective activity against AO 0.1 pM but only SP233, SP235, SP236 and SP238 exerted a significantly more robust effect than SP222 (Figs. 9A-9P). SP233, SP236 and SP238 maintained their neuroprotective properties against 1 AM AO-induced toxicity (Figs. 10A-10P) WO 2006/044665 PCT/US2005/037023 but only SP233 and SP238 kept this property in the presence of 10 LM AO (Figs. 11A-11P). Results obtained with the MTT assay were confirmed using the membrane potential-assessing Cytolite assay for SP222, SP233, SP235, SP236 5 and SP238. Fig. 12A shows that A0 exposure induces a dose-related decrease of the membrane potential-assessing luminescence. Although SP222 protected against 0.1 pM A0 (Fig. 12B), it failed to do so against the two highest concentrations of A0 (Figs. 12C and 12D). The various SP compounds used displayed a significantly better neuroprotective effect compared to SP222 as 10 shown by the increase in measured luminescence. The neuroprotective effect of SP233 and SP238 against 10 ptM A0 seen using the MTT assay (Fig. 11) was replicated by the raise of the signal under the same conditions (Fig. 12D). ATP levels, an index of mitochondrial function, were measured in PC12 cells treated with increasing concentrations of A0 in the presence or absence of 15 the SP222-SP238 compounds (Figs. 13A-13D). A0 decreased in a dose dependent manner ATP production by PC12 cells; 18%, 22% and 25% decrease in ATP levels measured in the presence of 0.1, 1.0 and 10 ytM A0, respectively (p<0.001 by ANOVA; Fig. 13A). From the compounds tested only SP233 and SP236 were able to reverse the 0.1 and 1.0 stM A-induced decrease in ATP 20 levels (Fig. 13B and 13C). No beneficial effect of the SP compounds on ATP synthesis was seen in the presence of 10 ptM A0. Trypan blue uptake by the cells was the fourth test used to assess the impact of the promising SP233 compound on A1-induced toxicity (Fig. 14). As expected, 0.1, 1 and 10 yM A-induced a dose-dependent (33%, 36% and 97%, 25 respectively; p<0.001 by ANOVA) increase in trypan blue uptake by PC12 cells. SP233 at 30 and 50 ptM inhibited the A#-induced cell death (p<0.001 by ANOVA). Fig. 15 shows that the neuroprotective effect of SP233 is dose dependent and it is maintained in the presence of all three concentrations of A0, although its efficacy decreases in presence of high, supra-physiopathological, A0 30 concentrations. One of the reasons in identifying 22R-hydroxycholesterol derivatives is the need of biologically active (neuroprotective) compounds that cannot be metabolized by P450scc to pregnenolone and then to tissue-specific final steroid products. To assess the metabolism of these compounds by steroidogenic cells WO 2006/044665 PCT/US2005/037023 we examined their ability to fonn steroids in MA- 10 mouse tumor Leydig cells, a well-characterized steroidogenic cell model where 22R-hydroxycholesterol is an excellent P450scc substrate and can produce large amounts of steroids. Li et al., cholesterol binding at the cholesterol recognition/ interaction amino acid 5 consensus (CRAC) of the peripheral-type benzodiazepine receptor and inhibition of steroidogenesis by an HIV TAT-CRAC peptide, Proc. Natl. Acad. Sci. USA, 98, 1267-1272 (2001). Fig. 16 shows that in contrast to SP222, SP233 could not be metabolized to final steroid products. The direct interaction of 22R-hydroxycholesterol derivatives to AO was 10 shown in displacement studies performed against the radiolabeled 22R hydroxycholesterol/A 3 complex (Fig. 17). Co-incubation of radiolabeled 22R hydroxycholesterol together with AO for 24 hours at 37'C demonstrated the presence of a high molecular weight radiolabeled band (Fig. 17) recognized by an antibody specific to AO (Yao et al., 2002 and data not shown). The 15 specificity of radiolabeling of AO by 22R-hydroxycholesterol was demonstrated by competition studies using unlabeled 22R-hydroxycholesterol (Fig. 17) where 100 gM SP222 displaced by 80% radiolabeled SP222 compound bound to AO. From the SP compounds tested, SP237, SP238, SP226, SP227 and SP233 displaced radiolabeled 22R-hydroxycholesterol binding to AO by 46, 44, 65, 38 20 and 35%, respectively (Fig. 17). These data were further confirmed using computational docking simulations with A0. The docking results show that AoL 42 forms a pocket in the 19-36 amino acids area where 22R-hydroxycholesterol binds, in agreement with our previous data. Yao et al., J. Neurochem., 83, 1110-1119 (2002). The 25 docking energy for the various compounds tested placed in order of minimal energy required for binding to AO was: (-10.34 kcal/mol) SP2 2 9<SP232<SP224<SP237<SP222<SP233<SP228<SP223<SP230<SP234<S P225<SP238<SP236<SP226<SP235<SP231<SP227 (-8.35 kcal/mol). Figs. 18 and 19 compare the binding characteristics of SP222 with SP233. This is an 30 analysis of 100 docking runs with each of the compounds. The data shows that about 23% of the time SP233 docks with energy of -7.0 to -7.5 Kcal/mol while SP222 docks about 25% of the time with only 5.5 to 6.0 kcal/mol. The probability of SP233 having a stronger (more negative) docking energy is significantly greater than that for SP222. Almost 100% of the time SP233 binds WO 2006/044665 PCT/US2005/037023 with less than -6.0 kcal/mol while the equivalent number for SP222 is only about -4.0 kcal/mol. Analysis of the distribution of the binding energy frequencies indicates a bimodal profile suggesting the presence of two binding sites in A0. For SP233 peaks might be present at both -7 to-7.5 and -8 to -8.5 kcal/mol 5 whereas with SP222 the peaks seem to be at -5.5 to -6.0 and -4.0 to -4.5 kcal/mol. Discussion Using this assay, some of the compounds tested, namely SP233, SP235, SP236 and SP238, exhibited neuroprotective activity even when PC12 cells were 10 exposed to concentrations as high as 10 pM A0. Interestingly, these compounds were more efficacious to the reference 22R-hydroxycholesterol (SP222) molecule. A late event in the mechanism of action of AO is the direct or indirect disruption of the mitochondrial respiratory chain, leading to a decrease in ATP 15 production that alone could lead to cell death. SP222, SP235, and SP238 compounds, which were able to rescue the PC12 cells from AO-induced toxicity, did not block the A-induced changes in ATP synthesis. Although such an apparent discrepancy remains to be explained it is possible that the MTT assay (mitochondrial diaphorase activity) and ATP synthesis do not reflect the status of 20 the same part of the respiratory chain. In contrast, SP233 and SP236 blocked, although in part, the Af-induced decrease in ATP production. The ability of SP233 to preserve ATP stocks could explain the potent neuroprotective effect of this compound, which was further confirmed by the trypan blue uptake cell viability assay. It should be noted that SP233 was found to be not only the most 25 efficacious in all assays used but also the most potent, offering neuroprotection in vitro against AO at concentrations as low as 10 pM. The studies presented herein were performed using 0.1, 1.0 and 10 pM A0 1
.
42 . These concentrations are supra-physiopathological since the concentrations of Al 1
.
42 present in cerebrospinal fluid of AD patients and 30 controls range from 500 1000 ng/l (0.1-0.2 nM). Even if Ao1.42 might be present in AD brain at 10 times higher concentration, the estimated pathophysiological concentrations of Al 1
-
42 would be in the range of 1-2 nM, which is 100-10,000 times less than the concentrations used in Applicants' experiments. With these WO 2006/044665 PCT/US2005/037023 considerations in mind it is clear that the 75% protection offered by SP233 against 0.1 giM A0 is pharmacologically relevant. Unlike 22R-hydroxycholesterol, its bioactive derivative SP233 was unable to induce steroid formation. 5 The neuroprotective property of the SP compounds may follow a structure/activity relationship (SAR). SP231 and SP235 are stereoisomers of diosgenin (Fig. 1), but only SP235 is protective against A-induced neurotoxicity. The stereochemistry of the SP235 is C3R, C10R, C13S, C20S, C22S, C25S, a motif shared by SP233 and SP236 (Fig. 1). SP compounds 10 exhibiting high neuroprotective activity and being active in the presence of high concentrations of A0 contained an ester, preferably a fatty acid or a fatty acid like structure, on C3. Indeed, SP235 that possesses an unsubstituted hydroxyl group in C3 offers limited neuroprotection acting only against 0.1 ytM A0. In contrast, SP236 that is the succinic ester at C3 of SP235 is active against higher 15 A0 concentrations and SP233, which is a hexanoic ester at C3 of SP235 is the most potent compound. The finding that SP238 was able to protect PC12 cells against Ag-induced toxicity, although it had no effect on maintaining ATP levels, further supports this hypothesis because its derivative without any side chain on C3 (SP226) did not offer neuroprotection. The finding that benzoic 20 acid substitution, present on SP232, was not effective in neuroprotection suggested that the presence of an aliphatic chain at this level is more relevant that an aromatic structure. Although these data are indicative of a SAR and highlights the importance of the presence of a fatty acid chain at C3. The ability of SP222 derivatives to offer neuroprotection by binding and 25 inactivating Ap 142 was examined. SP compounds exhibiting neuroprotective properties against Ag-induced cell death displace radiolabeled 22R hydroxycholesterol bound to the amyloid peptide. Computational docking simulations were used to further characterize the SP-A0 interaction. The studies revealed that two binding sites might be present 30 on A0 for the bioactive SP compounds. One binding site seems to be more specific for 22R-hydroxycholesterol (SP222), whereas the second binding site displays higher affinity for compounds such as SP233 and SP236. Although SP226 is shown to bind to this second binding site too, the calculated binding energy for this compound is much lower than the energy displayed by the r% WO 2006/044665 PCT/US2005/037023 neuroprotective SP molecules. A subsequent computational docking simulation study indicated that the binding energies of SP222 and SP233 follow a bimodal distribution, a finding that strongly supports the presence of two binding sites on Afl. Further calculation of binding energies indicated that SP222 has less 5 affinity for the second binding site compared to SP233 and suggests that the presence of the ester chain might be responsible for the ability of SP233 to bind to both sites on A3. Based on these observations, occupancy of the AO second binding site might be required for a sustained inactivation of the amyloid peptide. 10 Other mechanisms not related to a direct inactivation of AO could also contribute to the neuroprotective activity of SP233. A possible modulation of the steroid receptor family cannot be excluded although little is known about the binding of spirostenols on nuclear receptors. It has been shown that A0 inhibits the fusion of GLUT3-containing vesicles leading to the disruption of 15 mitochondrial homeostasis and, thus to neuronal death. On the other hand, the glucose absorption is enhanced in normal and streptozotocin-induced diabetic mice by spirostenol derivatives extracted from Polygonati rhizome. Taken together, these results suggest that restoration of glucose transport inside the cell might be a protective mechanism activated by the spirostenol SP233. 20 Example III Materials and Methods Immunoblot analysis of As polymerization and amyloid derived diffusible ligand (ADDL) formation 25 Increasing concentration of Ap (0.1, 1 and 10pM) were incubated in PC12 culture medium for 24 hours and 72 hours at 37'C under 5% CO 2 with or without increasing concentrations of SP233 (1, 10 and 100 pM). At the end of the incubation time, samples were separated by 4-20% Tris-Glycine gel electrophoresis (Invitrogen) under native conditions at 125V for 2 hours and 30 transferred to nitrocellulose membrane (HybondTM ECLm, Amersham Pharmacia Biotech) at 130A for 30 minutes. Non-specific adsorption of the antibodies was blocked by incubating the nitrocellulose in 5% milk. The blots were treated for immunodetection of Ap species using a polyclonal antibody to Ap that recognizes a 30 amino acid peptide of the As protein (Zymed WO 2006/044665 PCT/US2005/037023 Laboratories, San Francisco, CA). Membranes were incubated in primary antibody for 1 hour at room temperature at a dilution of 1:2,000. Then, membranes were incubated in the secondary antibody at a dilution of 1:1,000 for 1.5 hours at room temperature. The blots were visualized using the ECLIm 5 Western Blotting Analysis System (Amersham Biosciences). Image densitometric analysis was performed using the OptiQuant software (Packard BioScience). This method allows for the separation, visualization and identification of AP complexes, polymers and ADDLs. Results 10 Electrophoresis were performed after incubating increasing concentrations of AP with increasing concentrations of SP233 in order to characterize the oligomers formed by AP and the interference induced by SP233 in that formation. FIGs. 20A and 20F show the immunoblot analysis performed after a 24 hour (FIGs. 20A) and 72 hour (FIGs. 20F) incubation period. Under 15 our experimental conditions, no monomer species were detectable at any time in the presence of 0.1 tM AP and no trimer or tetramer (ADDLs) were detectable at 24 hours and 72 hours with AP at 0.1 and 1 pM. SP233 decreases in a dose dependent manner the amount of the monomeric species quantified at 24 hours (FIGs. 20B) and 72 hours (FIGs. 20G) ion the presence of 1 and 10 pM 20 concentrations of Ap. This dose-effect relationship is also observed against the trimeric and tetrameric ADDLs at 24 hours (FIGs. 20C, 20D) and 72 hours (FIGs. 20H, 20I) when SP233 is co-incubated with 10 pM Ap. Conversely, SP233 induces a small dose-dependent increase of the polymeric species amount (FIGs. 20E, 20J) suggesting that SP233 binds the AP and inhibit the formation of 25 the neurotoxic ADDLs by forming stable heavy complexes with the peptide. Discussion Trimers and tetramers belong to the amyloid-derived diffusible ligands (ADDLs), which are non fibrillar oligomers ranging approximately from 13 to 108 kD (Klein, Neurochem. Int., 41, 345-352 (2002)), with potent neurotoxic 30 properties at concentration as low as 5-10 nM (Lambert et al., Proc. Natl. Acad. Sci. USA, 95, 6448-6453 (1998); Dahlgren et al., J. Biol. Chem., 277(35), 32046-32053 (2002)). A recent report described the ADDLs as baring the neurotoxic properties of Ap. Klein, Neurochem. Int., 41, 345-352 (2002).
WO 2006/044665 PCT/US2005/037023 SP233 decreased in a dose-dependent manner the formation of the trimers and tetramers after 24 hours or 72 hours incubation, accounting for its neuroprotective effect. Moreover, SP233 decreased the amount of monomers available for ADDLs formation. The dose-dependent decrease of the ADDL 5 amount by SP233 was accompanied by a dose-dependent increase of high weight polymers aggregation, which suggests that the 22R-hydroxycholesterol and the SP233 inactivate AP by binding to it and forming stable non-toxic polymers. Example IV 10 Differentiation of Neuronal Precursor Cells Using 22R-Hydroxycholesterol This example demonstrates 22R-hydroxycholesterol inbits the proliferation of NT2 cells and induces them to differentiate into "neuron-like" and "astrocyte-like" cells. During 22R-hydroxycholesterol-induced differentiation of NT2 cells, the expression of high molecular weight 15 neurofilament protein NF 200 was increased, the expression of low molecular weight neurofilament protein NF 70 was decreased, and the protein expression of GFRa2 was increased. These effects of 22R-hydroxycholesterol were stereospecific because its enantiomer 22S-hydroxycholesterol or other steroids did not induce NT2 cell differentiation. 20 Cell culture, cell viability (cytotoxicity), cell proliferation and flow cytometry Human NT2 precursor (Ntera2/D 1 teratocarcinoma) cells were obtained from Stratagene (La Jolla, CA, USA) and cultured following the instructions of the supplier. Effects of 22R-hydroxycholesterol on cell viability (an inverse 25 measure of cytotoxicity) and cell proliferation were determined using the 3-(4,5 dimethylthiazol-2-yl)-2,5-diphelnyl tetrazolium bromide (MTT) assay (Trevigen, Gaithersburg, MD, USA), lactate dehydrogenase release (LDH) assay (Boehringer Mannheim; Indianapolis, IN, USA) and 5-bromo-2'-deoxyuridine (BrdU) ELISA (Boehringer Manneim; Indianapolis, IN, USA) as previously 30 described (Yao et al., Brain Res., 889, 181 (2001)) or following company protocol. For flow cytometry, cells were grown in 10-cm dishes for 24 hours and treated with various concentrations of 22R-hydroxycholesterol for 0 (control)) 3, 6 and 12 days. After trypsinization, the cells were collected, suspended (1-2 X 106 cells in 0.1 ml citrate/DMSO solution), stained with a WO 2006/044665 PCT/US2005/037023 solution containing propidium iodide (50 [tg/ml) and RNase (10 tg/ml), and subjected to flow cytometric analysis in an FACS Scan flow cytometer (Becton Dickinson, Menlo Park, CA, USA) equipped with a Modfit software program (Vindelov et al., Cytometry, 3, 332 (1983)). 5 Immunocytochemistry and immunoblot analyses For experiments on 70-, 145- and 200-kDa neurofilament proteins and GFRa receptors, NT2 cells were cultured in 8-well chamber slides and treated with 22R-hydroxycholesterol at the concentration of 25 pM for 6 days. Cells were fixed with newly prepared 3.7% formaldehyde for 15 minutes and blocked 10 by blocking reagent (Zymed Laboratories, San Francisco, CA, USA). Immunostaining was performed using rabbit anti-NF200, anti-NF145 and anti NF70 (all at 1:200), and goat anti-GFRd, anti-GFRa2 and anti-GFRa3 (all at 1:100); Santa Cruz Biotechnology (Santa Cruz, CA, USA). All antibodies were used at 200 [tg/ml. After overnight incubation at 4'C, the immunoreactivity was 15 detected using horseradish peroxidase-conjugated anti-rabbit or anti-mouse IgG for 1 hour at room temperature and visualized using AEC. For immunoblots, NT2 cells were cultured in 6-well plates and treated for 6 days with various concentrations of 22R-hydroxycholesterol. Cells were then washed with phosphate-buffered saline (PBS) and lysed with loading 20 buffer. Proteins were resolved by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) on 4-20% gradient acrylamide-bis-acrylamide gel at 125 V for 2 hours and electrotransferred onto nitrocellulose membranes. After blocking with 5% nonfat milk, the membranes were subjected to immunoblot analysis by incubation overnight with rabbit anti-NF200, anti-NF145 and anti 25 NF70 (all at 1:4000) (Chemicon International, Temecula, CA, USA) and goat anti-GFRcl, anti-GFRa2 and anti-GFRa3 (all at 1:500) (200 [tg/ml; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), goat anti-rabbit or rabbit anti-goat IgG-horseradish peroxidase being used as the secondary antibody (1:5000) (Zymed Laboratories, San Francisco, CA, USA). Protein bands were visualized 30 using ECL reagents (Amersham Pharmacia Biotech, Piscataway, NJ, USA). Image-densitometric analyses of the immunoreactive protein bands were performed using OptiQuant-image analysis software (Packard BioScience, WO 2006/044665 PCT/US2005/037023 Meriden, CT, USA). Glyceraldehyde-3-phosphate dehydrogenase (GAPPH) served as an internal standard. 3 H-22R-hydroxycholesterol uptake assay To determine 22R-hydroxycholesterol uptake, NT2 cells (2 x 10 5 5 cells/well) were incubated in 24-well plates for 0, 1, 3 and 6 days in media containing 1 ml of 10% fetal bovine serum in the presence of 0.1 pLCi 3 H-22R hydroxycholesterol. Following incubation, the cells were washed with PBS and lysed in 1 ml of 0.1 N NaOH. Radioactivity was measured by liquid scintillation spectrometry. Protein levels were quantified by the dye-binding assay of 10 Bradford (Bradford, Anal. Biochem., 2, 242 (1976)) using bovine serum albumin as standard. Cholesterol-protein binding blot assay (CPBBA) NT2 cell protein (4 ptg) and 3 H-22R-hydroxycholesterol (0.1 pCi) were incubated either alone or in the presence of increasing concentrations of 15 unlabeled 22R-hydroxycholesterol in a 20-pl volume for 3 hours at 37 0 C. After incubation, samples were separated by 1.0% agarose (Type I-B) gel electrophoresis and transferred to a nitrocellulose membrane (Schleicher & SchuelI, Keene, NH, USA) in 10 x saline-sodium citrate (SSC) buffer. The membrane was exposed to a tritium-sensitive screen and analyzed by 20 phosphoimaging using the Cyclone Storage Phosphor System (Packard BioScience, Meridien, CT, USA). Image-densitometric analyses were performed using the OptiQuant software (Packard). This method allows for the separation, visualization and identification of 22R-hydroxycholesterol-protein complexes which have incorporated radiolabeled cholesterol (Yao et al., FASEB, 16, 677 25 (2002)) and 22R-hydroxycholesterol (Yao et al., J. Neurochem., 83., 1110 (2002)) under native conditions. Low molecular weight unincorporated 22R hydroxycholesterol is separated and eliminated during electrophoresis. Statistics Statistical analysis was performed by one-way analysis of variance 30 (ANOVA) and unpaired Student's t test using the INSTAT 3.00 package (GraphPad, San Diego, CA, USA). RESULTS Phase-contrast photomicrographs showed that treatment of NT2 cells with 25 [tM 22R-hydroxycholesterol for 3, 6 and 12 days induced morphologic
K-A
WO 2006/044665 PCT/US2005/037023 changes (Fig. 1, upper panel). A dramatic change in morphology of the cells was evident after 6 days of 22R-hydroxycholesterol treatment (Fig. 21c). Flow cytometric analyses showed further that 22R-hydroxycholesterol influenced the proliferation of NT2 cells (Fig. 21, lower panel). The data clearly show that the 5 percentage of cells in the Go/G 1 phase was increased from 35.32% to 50.34% and that the percentage of cells in the G 2 /M phase was decreased from 34.43% to 15.90% with 12 days of treatment. These effects of 22R-hydroxycholesterol on NT2 cell morphology (differentiation) could not be replicated by incubating the cells for 6 days in the 10 presence of 25 pM concentrations of its enantiomer 22S-hydroxycholesterol (Fig. 22a), its precursor (cholesterol), or its metabolites progesterone and DHEA (Fig. 22b). Interestingly, the effect of 22R-hydroxycholesterol of inducing differentiation of NT2 cells was found not only to be stereospecific, but also restricted to these NT2 cells since such an effect was not found using PC12 cells, 15 MDA-MB-231 cells or the human glioma cell line U87 (data not shown). During CNS development in vivo, cells actively participate in two opposing processes: proliferation and death (Ross, Trends Neurosci., 19, 62 (1996)). In NT2 cells, early programmed cell death has been associated with the induction of differentiation by RA, it occurs mainly in undifferentiated cells, and 20 it coincides with first detection of the neuronal phenotype (NT2N) (Guilemain et al., J. Neurosci. Res., 71, 38 (2003)). In the present study, the effects of 22R hydroxycholesterol, 22S-hydroxycholesterol, pregnenolone and progesterone on NT2 cell survival and proliferation were determined using LDH, MTI formazan exocytosis and BrdU assays. Results obtained with the LDH (Fig. 22c, upper 25 panel) and MTT (Fig. 22c, middle panel) assays showed that all of these steroids were cytotoxic at high concentrations (25-50 pM). At the 50 pM concentration, all four steroids significantly inhibited NT2 cell proliferation, whereas at the 25 iM concentration cell proliferation was significantly inhibited only by 22R hydroxycholesterol (p<0.05) and 22S-hydroxycholesterol (p<0.01) (Fig. 22c, 30 bottom panel). These data indicated that apoptotic cell death of undifferentiated NT2 cells also occurred during 22R-hydroxycholesterol induced differentiation. Considering that increases in the expression of neurofilament proteins can be detected in NT2 cells after three days of RA-induced differentiation (Pleasure et al., J. Neurosci. Res., 35, 585 (1993)), the effect of 6 days of WO 2006/044665 PCT/US2005/037023 treatment with 22R-hydroxycholesterol on the expression of neurofilament proteins NF70, NF145 and NF 200 in NT2 cells was examined by imunocytochemical (Fig. 23, upper panel) and immunoblot (Fig. 23, lower panel) analyses. Results showed that NF70 protein expression in the cells 5 decreased progressively in the presence of increasing concentrations of 22R hydroxycholesterol. Immunoblot assays showed that NF70 protein level decreased from 83% to 18% of the control level by increasing the concentration of 22R-hydroxycholesterol from 1 FLM to 50 ptM, although this change was not reflected in the immunocytochemical data due to the low level of NF70 protein 10 expression by the cells. 22R-Hydroxycholesterol also influenced NF145 expression, decreasing it to 56% of control levels at a concentration of 25 pM, although changes were not significant with treatment at lower concentrations, except for the increase that was apparent at 5 pLM (Fig. 23e). 22R Hydroxycholesterol (5-25 piM) treatment led to marked increases in NF200 15 protein expression (p<0.001), this increase being slightly less pronounced at 25 pM than at the 5 ptM concentration; 180% versus 210% of the control value (Fig. 23t). To explore the mechanism of action of 22R-hydroxycholesterol, its effect on the expression of glial cell line-derived neurotrophic factor receptor proteins 20 were examined using immunocytochemistry (Fig. 24a, band c, upper panel) and imunoblot analysis (Fig. 24a, band c, lower panel). Results showed. that the presence of 25 LM 22R-hydroxycholesterol had no significant effect on the expression of GFRal (Fig. 24a). In contrast, 22R-hydroxycholesterol (25 ptM) strongly influenced GFRa2 protein expression, increasing it by 45% over the 25 control level with 6 days of treatment (Fig. 24b, lower panel). Treatment with 22R-hydroxycholesterol (25 pLM) also influenced GFRa3 protein expression in NT2 cells, decreasing it by about 16% (Fig. 24c, lower panel). To determine whether 22R-hydroxycholesterol enters into NT2 cells during differentiation, its uptake by the cells after 1, 3 and 6 days of culture was 30 measured. Assays based on 3 H-22R-hydroxycholesterol uptake showed that it was taken up by NT2 cells after 1 day in culture (Fig. 24d, upper panel). CPBBA was used to study the direct interaction between NT2 cell total protein and 22R hydroxycholesterol. The generated pattern showed that after 3 hours incubation WO 2006/044665 PCT/US2005/037023 at 37'C the intensity of radiolabeling of protein with 3 H-22R-hydroxycholesterol increased in presence of increasing amounts of protein and decreased in the presence of increasing concentrations of unlabeled 22R-hydroxycholesterol (Fig. 24d, lower panel). 5 DISCUSSION Although undifferentiated NT2 cells are derived from a germ cell tumor and do not represent completely authentic models for studying neuronal differentiation, they do appear to represent a committed human neuronal precursor cell line that retains some stem cell characteristics and which is 10 capable only of terminal differentiation into neurons (Pleasure et al., Neurosci. Res, 35, 585 (1993)). Therefore, the transformation of ***NT2 cells to NT2N cells provides a unique model system for studies of human neurons (Ibid.; Pleasure et al., J. Neurosci., 12, 1802 (1992)). NT2 cells differentiate extensively in vitro when exposed to RA, a process that is marked by the appearance of 15 several morphologically distinct cell types and by changes in cell surface phenotype (Ibid.; Andrews, Dev. Biol., 103, 285 (1984)). Among the differentiated cells, the neurons form clusters interconnected by extended networks of axon bundles and express neurofilament proteins and other neuronal markers (Ibid.). This commitment of NT2 cells to the stable 20 neuronal phenotype (NT2N) following RA treatment has been shown to be irreversible, as determined by the lack of mitotic activity or phenotypic reversion over a 2-month period in culture. With regard to time course, it has been shown that islands of NT2 cells show increased expression of neurofilament proteins after three days of treatment with RA, and by 10-14 days the morphology of 25 these cells begins to resemble that of neurons. On this basis, and upon considering that NT2 cells have been extensively used to study RA-induced differentiation of progenitor cells into both neurons and astrocytes (Bani Yaghoub et al., Exp. Neurol., 156 16 (1999); Neuroreport, 10 3843 (1999); Sandhu et al., J. Neurosci. Res., 68, 604 (2002)), this model was used to examine 30 the effects of22R-hydroxycholesterol on cellular proliferation and differentiation. These results demonstrate that treatment of NT2 cells with 25 M 22R hydroxycholesterol induces their differentiation into "neuron-like" or "astrocyte like" cells. With regard to cellular morphology, the phase-contrast -' 17 WO 2006/044665 PCT/US2005/037023 photomicrographs provided in Fig. 21 show clearly that treatment of NT2 cells with 25 [tM 22R-hydroxycholesterol for periods of 3, 6 and 12 days induced morphologic changes, these changes being evident after 6 days of treatment. Flow cytometric analyses showed further that these morphological changes 5 induced by 22R-hydroxycholesterol coincided with an effect on proliferation of the NT2 cells. The percentage of cells in the Go/G 1 phase of the cell cycle was increased and the percentage of cells in the in the G 2 /M phase was decreased after 12 days of treatment with the steroid (Fig. 21, lower panel). This finding is of interest in light of other recent results which indicate that, like treatment with 10 22R-hydroxycholesterol, treatment of NT2 cells with hexamethylene bisacetamide (HMBA; the prototype compound of a group of hybrid polar compounds that induce differentiation in various transformed cells) also led to an accumulation of the cells in the G 1 phase and terminal differentiation (Baldassare et al., Oncogene, 21, 1739 (1999)). This HMBA-induced growth arrest and 15 differentiation of NT2 cells involved increased expression of the cyclin dependent kinase inhibitor p27, enhanced association of p27 with cyclin E/CDK2 complexes, and suppression of kinase activity associated with cyclin E/CDK2. These effects of both 22R-hydroxycholesterol and HBMA indicate that they can act by influencing molecular mechanisms involved in coordinating cell 20 cycle regulation and neuronal differentiation. More specifically, these results indicate that 22R-hydroxycholesterol may serve as a developmentally-controlled endogenous molecular signal involved in regulating cell division; i.e., it may be involved in determining whether cells continue to proliferate or arrest growth and proceed to differentiation and 25 programmed death. This view is strengthened upon considering that the effect of 22R-hydroxycholesterol of inducing differentiation of NT2 cells was stereospecific, not observed with its precursor (cholesterol) or its metabolites progesterone and DHEA (Fig. 2b), and not found using PC12 cells, MDA-MB 231 cells or the human glioma cell line U87. 30 During CNS development in vivo, cells actively participate in two opposing processes: proliferation and death (Ross, Trends Neurosci., 19, 62 (1996)). In NT2 cells, early programmed cell death (apoptosis) has been associated with the induction of differentiation by RA, it occurs mainly in undifferentiated cells, and it coincides with first detection of the neuronal WO 2006/044665 PCT/US2005/037023 phenotye (NT2N), i.e., significant apoptotic cell death could be quantified during the third and fourth days of the RA treatment using annexin V labeling as a marker of apoptosis (Guillemain et al., J. Neurosci. Res., 71, 38 (2003)). In the present study, the effects of 22R-hydroxycholesterol, 22S-hydroxycholesterol, 5 pregnenolone and progesterone on NT2 cell survival and proliferation using LDH, MTT formazan exocytosis and BrdU assays was determined (see Fig. 22). Results obtained with the LDH and MTT assays showed that all of these steroids were cytotoxic at high concentrations (25-50 [pM). At the 50 piM concentration, all four steroids significantly inibited NT2 cell proliferation, whereas at the 25 10 pM concentration cell proliferation was significantly inhibited only by 22R hydroxycholesterol and 22S-hydroxycholesterol. Thus, apoptotic cell death of undifferentiated NT2 cells also occurred during 22R-hydroxycholesterol-induced differentiation. It might also be noted that 22R-hydroxycholesterol affected the viability of undifferentiated NT2 cells only at high concentration (>10 pM) and 15 had no effect on differentiated NT2N neuronal cells, as determined by MTT assay. Regarding the effects of 22R-hydroxycholesterol treatment on the expression of neurofilament proteins by NT2 cells, treatment with 22R hydroxycholesterol for 6 days led to changes in the expression of NF70, NF145 20 and NF200 (see Fig. 23). Immunoblot assays showed that NF70 protein level was decreased from 83% to 18% of the control level by increasing the concentration of 22R-hydroxycholesterol from 1 FLM to 50 pM. 22R Hydroxycholesterol also influenced NF145 expression, decreasing it to 56% of control levels at a concentration of 25 [tM, and led to highly significant increases 25 in NF200 protein expression over the concentration range of 5-25 p.M. As neurofilaments are the major cytoskeletal element in nerve axons and dendrites, as their expression is both tissue-specific and developmentally regulated, and as they are normally restricted to neurons, these findings provide convincing support for an effect of 22R-hydroxycholesterol of inducing neuronal 30 differentiation of NT2 cells. This view is further substantiated by previous studies which have shown that RA-induced differentiation of NT2 cells into neurons involves the induction of neurofilament proteins. For example, Lee et al., J. Neurosci., 6, 514 (1986) have shown that neurofilament proteins NF195, WO 2006/044665 PCT/US2005/037023 NF170, and NF70 could be detected in a few NT2 cells with a non-neuronal morphology as early as two days after exposure to RA, that the number of neurofilament-positive cells increased with time following exposure to RA, and that after two weeks many of the cells expressing neurofilaments exhibited a 5 neuronal morphology. Also, in the study of Webb et al, J. Neuroimmunol., 11 67 (1986), NT2 cell cultures cells exhibited a "neuron-like" morphology and expressed NF55 and NF210 three weeks after initiating RA treatment. These findings are also of interest upon considering that neurofilament proteins accumulate in neurons in many neurodegenerative diseases, including AD. 10 In an effort to clarify the mechanism underlying 22R-hydroxycholesterol induced neuronal differentiation, its effect on the expression of GFRa receptor proteins in NT2 cells was examined using immunocytochemistry and immunoblot analyses (Fig. 24). It was found that 6 days of treatment with 25 [tM 22R-hydroxycholesterol increased GFRa2 protein expression by 45% over the 15 control level, and decreased GFR3 protein expression by about 16%. It is known that members of the GDNF family and the GDNF-family ligands are crucial for the development and maintenance of distinct sets of central and peripheral neurons, the ligands acting by activating intracellular signaling pathways though their receptor tyrosine kinases (Airaksinen et al., Nat. Rev. 20 Neurosci., 3, 383 (2002)). With further regard to these findings, members of the GDNF family (GDNF, neurturin, artemin and persephin) are critical regulators of neurodevelopment and support the survival of midbrain dopaminergic and pineal motor neurons Sanchez et al., Nature, 382, 70 (1996); Widenfalk et al., J 25 Neurosci., 17, 8506 (1997)). GDNF family ligands bind to their specific GFRc receptors and activate Ret transmembrane receptor tyrosine kinase (Airaksinen et al., Mol. Cell. Neurosci., 13, 313 (1999); Airaksinen (2002)). Recent studies have shown that GDNF induces proliferative inhibition of NT2 cells through RET-mediated up-regulation of the cyclin-dependent kinase inhibitor p 2 7 kiP, 30 (Baldassarre et al., Oncogene, 21, 1739 (2002)), which plays a key role in NT2 cell survival and differentiation (Baldassarre et al., Oncogene, 18, 6241 (1999)). In general, these findings, which indicated that treatment with 22R hydroxycholesterol induced changes in the expression of GRFcc2 and GRFa3 WO 2006/044665 PCT/US2005/037023 may be taken to indicate that exposure of NT2 cells to 22R-hydroxycholesterol not only influenced mechanisms involved in the regulation of neuronal development, but also those involved in the formation of "astrocyte-like" and possibly "oligodendrocyte-like" cells. In this connection, Bani-Yaghoub et al., 5 Neuroreport, 10, 3843 (1999) have reported that NT2 cells can differentiate into astrocytes, and concluded that NT2 cells, like embryonal stem cells, can give rise to both neuronal and astrocytic lineages. Also, Sandhu et al., J. Neurosci. Res., 68, 604 (2002) found that NT2 cells can be differentiated into functional astrocytes by a 4-week treatment with RA, differentiation being accompanied by 10 decreased cell proliferation and cell-cycle arrest, as measured by flow cytometry, immunostaining for Ki67 and incorporation of BrdU. Example IV also shows that 3 H-22R-hydroxycholesterol is taken up by NT2 cells after one day in culture, and that after 3 hours of incubation at 37'C the intensity of radiolabeling of protein with 3 H-22R-hydroxycholesterol 15 increased in the presence of increasing amounts of protein and decreased in the presence of increasing concentrations of unlabeled 22R-hydroxycholesterol (Fig. 24). These results indicate that a physicochemical interaction between 22R hydroxycholesterol and cell protein contributes to its effect of inducing differentiation of NT2 cells. 20 The results presented herein gain added significance upon considering previous results which have shown that neurosteroids, such as pregnenolone and DHEA, may play a physiological role in the brain by preserving and/or enhancing cognitive function (Valee et al., PNAS USA, 94, 14865 (1997)) and by improving memory in aged animals (Roberts, The Biological Role of 25 Dehydroepiandrosterone, Kalima et al., eds., de Gruyter (1990) at pages 13-42). Pregnenolone and DHEA accumulate independently of their supply by peripheral endocrine organs (Baulieu et al., J. Steroid Biochem. Mol. Biol., 37, 395 (1990), and appear to act as neuromodulators (Paul et al., FASEB J., 6, 2311 (1992)). Glial cells can convert cholesterol to pregnenolone. 30 In vitro studies have shown that oligodendrocytes, a glioma cell line, and Schwann cells express the cytochrome P450 cholesterol side-chain-cleavage enzyme which is involved in the conversion of cholesterol to pregnenolone (Jung-Testas et al., Endocrinol., 125, 2083 (1989); Papadopoulos et at., PNAS USA, 89, 5113 (1992); Akwa et al., C. R. Acad. Sci. III (France), 316, 410 '7 1 WO 2006/044665 PCT/US2005/037023 (1993)). It has also been well described that 22R-hydroxycholesterol is one of the three hydroxylated intermediates formed during this enzymatic cleavage reaction (Dixon et al., Biochem. Biophys. Res. Commun., 49, 161 (1970); Hall, Anal. N.Y. Acad. Sci., 458, 203 (1985)). It is also noteworthy that the levels of 5 22R-hydroxycholesterol were found to be lower in brain specimens of AD patients compared to age-matched controls. Levels of 22R-hydroxycholesterol were significantly decreased in the hippocampus, a structure of the limbic system of the brain that is critical to maintaining cognitive functions, such as learning and memory, and which is affected in AD (Yao et al., J. Neurochem., 10 83, 1110 (2002)). Although the exact mechanism underlying 22R hydroxycholesterol-induced differentiation of NT2 cells to the NT2N phenotye remains unclear, even with the results presented herein, Example 4 indicates that 22R-hydroxycholesterol activates some unknown protein(s) by binding to, or interacting with this protein(s), increases GFRa2 protein expression and 15 activates the GFRca-Ret pathway, and thereby inhibits cell proliferation and induces differentiation. Example IV Use of Spirostenol to Induce Neuronal Differentiation 20 A. Materials and Methods 1. Materials (20a)-254-methyl-(22R,26)-azacyclofurost-5-en-34-ol (SP-224) was purchased from Interbioscreen (Moscow, Russia). Cells culture supplies were purchased from GIBCO (Grand Island, NY) and cell culture plasticware was 25 from Coming (Coming, NY). Mice embryonic teratocarcinoma P19 cells, PC12 rat pheochromocytoma cells and NT2 human neuroblastoma cells were purchased from ATCC (Manassas, VA). For the in vivo studies, male Long Evans rats weighing 300-325 g were purchased from Charles-River and housed in the Department of Comparative Medicine at Georgetown University. The 30 osmotic micropumps (2ML2) delivering 5.0 ptl per hour for 14 consecutive days were purchased from DURECT Corp. (Cupertino, CA). Rabbit anti-GAP43, guinea pig anti-doublecortin (DCX), mouse anti-pIII tubulin and rabbit anti-choline-acetyl transferase (ChAT) antibodies were purchased from Chemicon (Chemicon International, Temecula, CA), mouse anti- WO 2006/044665 PCT/US2005/037023 CNPase, rabbit anti-GFAP and mouse anti-MAP2 antibodies were purchased from Abcam (Cambridge, MA), rabbit anti-synaptophysin antibody was purchased from Zymed Laboratories (San Francisco, CA) and the mouse anti bromo-desoxyuridine (BrdU) antibody was purchased from Neomarkers (Lab 5 Vision Corp., Fremont, CA). Texas red-, rhodamine-, rhodamine (TRITC)- and FITC-conjugated antibodies were purchased from Jackson Immunoresearch Laboratories (West Grove, PA). The DAPI and ethidium bromide nuclear counterstain dye were obtained from Molecular Probes (Eugene, OR). 10 2. Cell culture and treatment Mouse embryonic teratocarcinoma P19 cells were cultured at 37'C in 95% C02 in Alpha Minimum Essential medium with ribonucleosides and deoxyribonucleosides containing bovine calf serum (7.5%) and fetal bovine serum (2.5%) on 13 mm diameter glass cover-slip. PC12 cells (rat 15 pheochromocytoma neurons) were cultured at 37*C and 5% CO 2 in RPMI 1640 medium devoid of glutamine and supplemented with 10% fetal bovine serum and 5% horse serum on 13 nun diameter glass cover-slip. When cells reached 70% confluence, the medium was replaced fresh medium containing 90 pM SP 224. P19 and PC12 cells were then incubated for 2 days before SP224 was 20 washed out and replaced by standard medium. The culture medium was changed every 2 days for 5 days or every 2 days for 30 days before cells were fixed for immunocytochemistry. The NT2 cells were treated for 5 days and washed-out for 10 days. 3. In vivo study 25 Male Long-Evans rats weighing 300-325 g (3-4 months old) were housed following a natural day-night cycle with food and water ad libitum. Prior to surgery, rats were anesthetized with equithesin (3 ml/kg, i.p.) and then placed on a stereotaxic frame. Using an electrode micromanipulator, the outlet of an . osmotic micropump was implanted into the left cerebral ventricle following the 30 coordinates D = 3.4 mm, L = 1.4 mm and AP = 0.92 mm caudal to bregma. The tank of the osmotic pump was implanted in a subcutaneous pocket in the midscapular area of the back of the rat. After surgery, rats were placed on a heating blanket for recovery. During the whole procedure, the body temperature was monitored and kept stable at 37" C. SP-224 at 375 p.M solubilized in '-71) WO 2006/044665 PCT/US2005/037023 polypropylene glycol/glycerol/distilled water (50/25/25) was perfused by i.c.v. route at 5 pl per hour for 2 weeks. Rats were sacrificed 3 weeks after the end of the brain infusion by intracardiac perfusion, first with a washing solution (NaC1 8 g/l, dextrose 4 g/l, sucrose 8 g/l, calcium chloride 0.23 g/l, sodium cacodylate 5 anhydrous 0.25 g/l, in deionized water) and then with fixative cacodylate buffer (sucrose 40 g/l, paraformaldehyde 40 g/l, sodium cacodylate anhydrous 10.72 g/l, in deionized water). Brains were stored in fixative cacodylate buffer for an additional week before being embedded in paraffin. To study the neural stem cells proliferation rats were daily injected with a BrdU solution at 100 mg/kg. 10 The first injection took place the day following the surgery and the last injection was performed the day previous to the euthanasia. 4. Immunocyto- and immunohistochemistry P19 cells were fixed with methanol at -20'C for 15 minutes before being permeabilized for 30 minutes at room temperature with 1X PBS pH=7.4 15 containing 0.1% Triton 1OOX. Cell were incubated for 24 hours at 4*C with antibodies raised against GAP43 (1/1000), DCX (1/3000), PIII Tubulin (1/500), synaptophysin (1/200), MAP2 (1/500), ChAT (1/2000), GFAP (1/1000) or CNPase (1/250). Positive immunostaining was revealed by Texas red-, rhodamine-, rhodamine (TRITC)- or FITC-conjugated antibodies incubated at 20 room temperature for 2 hours (1/200). Cells nucleus was counterstained using DAPI or ethidium bromide. Brains were embedded in a paraffin block immediately after excision and cut into 20-tm thick slices. These brain slices were processed with different primary antibodies; anti-BrdU (1/100) and anti-DCX (1/3000) were incubated 24 25 hours at 4'C and the signal was revealed with FITC- or rhodamine-conjugated secondary antibody (1/200). B. Results 1. Induction of neurites-like sprouting in human undifferenciated neuronal NT2 cells, in mouse embryonic teratocarcinoma P19 cells and in mouse 30 pheochromocytoma PC12 cells. The above-referenced cell lines were used because they have been extensively used as a model of neuronal differentiation (Houldsworth et al., 2002; Chou et al., 2003; Das et al., 2004). To induce the sprouting, cells were plated in 6-well plates and treated for 2 days (P19) or 5 days (PC 12 and NT2) '7 A WO 2006/044665 PCT/US2005/037023 with 90 pM SP-224 before being washed with fresh medium and cultured for 5 (P19) or 10 (PC 12 and NT2) more days. Under our experimental conditions the three types of cells displayed a significant sprouting (Fig. 25B, D, F) and the most spectacular effect was observed with the P19 cells (Fig. 25D). 5 Interestingly, the sprouting started during the washing out period whereas no cell shape modification was observed during the treatment period. Meantime, we observed a growth arrest of the treated-cells. The control cells were treated with a SP-224 free medium for 2 days before being washed out under the same conditions as the SP-224-treated cells. The control cells kept growing even after 10 the confluence was reached without displaying any sprouting of any kind (Fig. 25A, C, E). 2. Identification of neuronal and non-neuronal markers in the differentiated P19 cells The ability of SP-224 to induce the neuronal differentiation of the P19 15 cells was assessed as follows. P19 cells were treated for 2 days followed by a 5 days wash out period (Fig. 26). The neuronal markers expression was also studied after 28 days (Fig. 27) in order to in order to verify whether the expression observed at 5 days was temporary or permanent. No neuronal marker was observed on the control except for the synaptophysin for which the staining 20 was, however, very weak (Fig. 26C). The SP-224 exposure induced a strong expression of the different neuronal markers studied, II tubulin (Fig. 26A), synaptophysin (Fig. 26B), MAP2 (Fig. 26C) and ChAT (Fig. 26D). The migrating neuroblasts marker DCX was also strongly expressed in SP-224 exposed P19 cells (Fig. 26E). The Fig. 26A even shows pIII tubulin 25 immunostaining on axons-like formation which length is significantly greater than the 100 tm scale bar. After 28 days of wash out the neuronal differentiation is even more important. The network of axons and dendrites has dramatically extended as shown by the PII tubulin staining (Fig. 27A). The importance of the synaptophysin labeling (Fig. 27B) shows that the newly 30 formed neurons established synaptic connections. The expression of the glial markers GFAP and CNPase, reflecting the ability of SP-224 to differentiate P19 cells in astrocytes and oligodendrocytes, has been studied in order to assess an eventual specificity of the differentiation process. Using the protocol -- 2 days with 90 pM SP-224 followed by 5 days of WO 2006/044665 PCT/US2005/037023 culture in SP-224 free medium-- the SP-224 induced also a differentiation of the P19 into oligodendrocytes (Fig. 28B) whereas no positive GFAP signal was detected (Fig. 28D). Both markers GFAP and CNPase were not expressed in controlled P19 cells. 5 3. In vivo characterization of a neural stem cells proliferation and identification of the early neuronal marker DCX This experiment was conducted to validate the results obtained in vitro and to assess the ability of the SP-224 to induce a neuronal differentiation of the neural stem cells present in the sub-ventricular zone (SVZ) of the rat brain. The 10 SP-224 was administered in a solution at 375 pM by intra-cerebroventricular route for 2 weeks, corresponding to a total quantity of 530 nM of SP-224 per rat. To simulate the wash out period of the in vitro protocol, rats were left in husbandry for another 3 weeks period before the brains were removed. The immunohistochemistry revealed an important BrdU showing that SP-224 was 15 able to induce a self-renewal of the neural stem cells in the SVZ (Fig. 29C-G). In addition some BrdU positive cells were detected at a significant distance of the SVZ (white arrows) suggestion that certain cells might have entered a migration process. SP-224 infusion also induced the expression of the early neuronal marker DCX in the SVZ cells, showing that as in vitro, SP-224 was 20 able to push the neural stem cells present in that area toward the neuronal differentiation (Fig. 29D-H). C. Discussion It is believed that neural stem cell therapy can be used to replace damaged neurons to treat several neurodegenerative diseases and conditions like 25 Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, traumatic brain injury, stroke and peripheral nerve injury (T.L. Limke et al., Stem Cell Res., 12, 615 (2003); D.J. Watson et al., J. Neuropathol. Exp. Neurol., 62, 368 (2003); S. Chiba et al., J. Neurol. Sci., 219, 107 (2004); V. Silani et al., Lancet, 364, 200 (2004); Y. Tai et al., Curr. Opin. Pharmacol., 4, 98 (2004); M. Tohill et 30 al., Biotechol. Appl. Biochem., 40, 17 (2004)). The two most promising strategies are the transplantation of stem cells differentiated in vitro and the induction of the proliferation and differentiation of endogenous stem cells (0. Lindvall et al., Nature Medicine, 10, S42 (2004)). However, the low number of neural precursors available can limit the repair capability of an adult brain and WO 2006/044665 PCT/US2005/037023 therefore a combination of both strategies, transplantation and in situ differentiation, appears to be the therapeutic approach the most likely to succeed. Many small molecules like retinoic acid or cyclopamine have been used as tools to induce the neuronal differentiation of NSC in vitro, but their use in vivo is 5 extremely difficult because of their toxicity. Dexamethasone, fluoxetine or geldanamycin have potentially dangerous side effects (S. Ding et al., Nature Biotech., 22, 833 (2004)). In accord with the present invention, the naturally occurring furostenol, (20a)-25 -methyl-(22R,26)-azacyclofurost-5-en-3 -ol (SP 224) was obtained and employed to induce neurogenesis in vitro and in vivo in 10 rat brain. SP-224 induced neurites-like structure formation in P19, NT2 and PC12 cells after 2-5 days of treatment followed by 5-10 days of wash-out showing the first evidence of a possible differentiation of these stem cells. Interestingly, SP 224 exerted an effect only if a period of wash-out was included in the treatment 15 protocol. Indeed, we did not observe any neurites processes when the "therapeutic pressure" was not removed after 2 days suggesting a "hit and run" mechanism of action. This structural modification was accompanied in differentiated P19 by the appearance of the specific neuronal markers p111 tubulin, and MAP2. The II-tubulin immunostaining allowed visualization 20 showing that the length reached by the axons formed after 5 days of wash-out was approximatively 500 tM. Importantly, SP-224 also induced the expression of synaptophysin showing that the newly formed neurons were able to establish synapses. The formation of synapses, indispensable for the integration of newly formed 25 neurons to a pre-existing network, kept evolving after the end of the 5 days wash-out period as showed after 1 month of growth suggesting that the effect of SP-224 was not transient. In addition, the differentiating P19 cells showed a cholinergic phenotype as they displayed a strong ChAT signal. The fact that SP 224 was able to push the P19 cells toward a cholinergic phenotype is of interest 30 as it presents this compound as an ideal agent to pre-treat human neural stem cells before being re-implanted into the hippocampus of a patient suffering from AD. The neuronal differentiation was confirmed by the expression of doublecortin, a marker which has been demonstrated to be expressed specifically WO 2006/044665 PCT/US2005/037023 in migrating immature neurons (F. Francis et al., J. Neuron., 23, 247 (1999)). These results suggest that not only is SP-224 capable of inducing in vitro neuronal differentiation of neural stein cells and promoting the formation of synapse and communication network, but it also triggers the migration of the 5 differentiating neurons toward areas where they perform their nonnal function. SP-224 did not trigger the differentiation of the P19 cells into astrocytes and the expression of the oligodendrocyte-specific marker CNPase was extremely low, suggesting that the neuronal differentiation pathway is specifically activated in vitro by SP-224. 10 The two main genninal area of the adult mammalian brain are the sub ventricular zone (SVZ) and the dentate gyrus of the hippocampus (B.J. Chiasson et al., J. Neurosci., 19, 4462 (1999); Alvarez-Buylla et al., Brain Res. Bull., 57, 571 (2002)). The continuous perfusion of SP-224 inside the left ventricle for 2 weeks induced a dramatic increase of the BrdU by the cells of the SVZ in rat 15 brain. These data showed that SP-224 was able to induce the proliferation of the NSC present in the SVZ. Moreover, the BrdU immunostaining co-localized with the early marker of neuronal differentiation DCX, therefore confirming in vivo the differentiating properties of SP-224 observed in vitro. The BrdU staining has also been detected close to the SVZ, but inside the striatum, 20 suggesting some migrating cells. It has to be noted that the effect of SP-224 was displayed without any apparent toxic effect on the brain tissue. In conclusion, SP-224 appears to be a promising small molecule to be used for stem cell therapy. It is believed that other compounds of formula (I), (II) or (III) will exhibit similar bioactivity, since these compounds were selected 25 to mimic the protein binding affinity of 22R-hydroxycholesterol. A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Accordingly, other 30 embodiments are within the scope of the following claims, and as various changes can be made to the above compositions, formulations, combinations, and methods without departing from the scope of the invention, it is intended that all matter contained in the above description be interpreted as illustrative 170 WO 2006/044665 PCT/US2005/037023 and not in a limiting sense. All patent documents and references cited herein are incorporated by reference.

Claims (34)

1. A method of inducing the differentiation of mammalian neuronal precursor cells into neuronal cells, comprising contacting said neuronal 5 precursor cells with an effective amount of a compound of formula (I): R 1 2 R 13 R 17 R 11 R16 R 1 R 10 R 8 R 9 R 1 4 R 15 R3VR R 4 R 6 (I) wherein each of R 1 , R 2 , R 4 , R 7 , Ru, R 12 , R 15 , and R 16 , independently, is hydrogen, (C 1 -C 8 )alkyl, hydroxy, amino, carboxyl, oxo, sulfonic acid, or (C 1 Cs)alkyl that is optionally inserted with -NH-, -N((C1-Cs)alkyl)-, -0-, -S-, -SO-, 10 -S02-, -O-SO 2 -, -S02-0-, -C(O)-, -C(0)-0-, -0-C(0)-, -C(O)-NR'-, or -NR' C(O)-, wherein R' is H or (C 1 -Cs)alkyl; R 3 is hydroxy, (CI-C 6 )alkylCO 2 -, HO 2 C(CH 2 ) 2 CO 2 -, toluene-4-sulfonyloxy, or benzoyloxy; each of R 6 , Rs, R 9 , Rio, R 13 , and R 1 4 , independently, is hydrogen, (CI-C 8 )alkyl, hydroxy(C1 Cs)alkyl, (CI-Cs)alkoxy, or hydroxy; and R 17 is 15 CH(CH 3 )CH(OH)(CH 2 ) 2 CH(CH 3 ) 2 or -CH(CH 3 )CH(OC(=0)CH 3 )(CH 2 ) 2 CH(CH 3 )CH 2 N(C(=O)CH3)2; or a pharmaceutically acceptable salt thereof.
2. The method of claim 1 wherein R 1 , R 2 , R 4 , R 6 , R 7 , R 8 , R 9 , Ru, 20 R 1 2 , R 14 and Ris are H.
3. The method of claim 2 wherein R 16 is H or acetoxy.
4. The method of claim 1 wherein the compound is selected from 25 the group consisting of 22R-hydroxycholesterol or 26-diacetylamino-(22 ) acetoxy-(164)-acetoxy-cholest-5-en-yl acetate.
5. A method of inducing the differentiation of mammalian neuronal precursor cells into neuronal cells, comprising treating said neuronal precursor 30 cells with an effective amount of a compound of formula (II): OA WO 2006/044665 PCT/US2005/037023 CH 3 R 19 R12 R13 ',- R20 RR11 O R 1 R 10 R 8 R 9 R 14 R 15 R3 J R7 R 4 R 6 (II) wherein each of R 1 , R 2 , R 4 , R 7 , R 11 , R 12 , and R 15 , independently, is hydrogen, (C 1 -C 4 ) alkyl, hydroxy, amino, carboxyl, oxo, sulfonic acid, or (C 1 C 6 )alkyl that is optionally inserted with -NH-, -N((CI-C 4 )alkyl)-, -0-, -S-, -SO-, 5 -SO 2 -, -0-SO 2 -, -SO 2 -0-, -C(O)-, -C(0)-0-, -O-C(O)-, -C(O)-NR'-, or -NR' C(O)- wherein R' is H or (C 1 -Cs)alkyl; R 3 is hydroxy, (C1-C 6 )alkylCO 2 -, HO 2 C(CH 2 ) 2 CO 2 -, toluene-4-sulfonyloxy, or benzoyloxy; each of R 6 , R 8 , R 9 , Rio, R 13 and R 14 , independently, is hydrogen, (C1-C 4 )alkyl, hydroxyl(C1 Cs)alkyl, (C1-Cs)alkoxy, or hydroxy; R 1 9 is OH or (C 1 -C 2 )alkoxy; and R 20 is 10 butyl 3-substituted by methyl amidomethyl, or a pharmaceutically acceptable salt thereof.
6. The method of claim 5 wherein R 1 , R 2 , R 3 , R 4 , R 6 , R 7 , R 8 , R 9 , Ru, R 1 2 , R 14 and R 15 are H. 15
7. The method of claim 5 wherein RIO and R 13 are CH 3 .
8. The method of claim 7 wherein R 1 9 is methoxy. 20
9. The method of claim 5 wherein R 3 is acetoxy or OH.
10. The method of claim 5 wherein the compound is selected from the group consisting of (204)-26-acetylamino-(224)-hydroxyfurost-5-en-34-yl acetate, (204)-26-acetylamino-(224)-methoxyfurost-5-en-3a-yl acetate, and 25 (204)-26-acetylamino-(22 )-ethoxyfurost-5-en-34-yl acetate.
11. A method of inducing the differentiation of mammalian neuronal precursor cells into neuronal cells, comprising contacting said neuronal cells with an effective amount of a compound of formula (III): 01 WO 2006/044665 PCT/US2005/037023 R12 R13 RIR11 O R 1 R10 9R8 1 1 R3 Ry R4(I) wherein each of R 1 , R 2 , R 4 , R 7 , R 11 , R 12 , and R 15 , independently, is hydrogen, (C 1 -C 8 )alkyl, hydroxy, amino, carboxyl, oxo, sulfonic acid, or (C 1 C 8 )alkyl that is optionally inserted with -NH-, -N((C 1 -C 8 )alkyl)-, -0-, -S-, -SO-, 5 -SO 2 -, -0-S02-, -S02-0-, -C(O)-, -C(O)-O-, -O-C(O)-, -C(O)-NR'-, or -NR' C(O)-, wherein R' is H or (C 1 -C 8 )alkyl; R 3 is hydroxy, (C 1 -C 6 )alkylCO 2 -, HO 2 C(CH 2 ) 2 CO 2 -, toluene-4-sulfonyloxy, or benzoyloxy; each of R 6 , R 8 , R 9 , Rio, R 13 , and R 1 4 , independently, is hydrogen, (C1-Cs)alkyl, hydroxyl(C1 C 8 )alkyl, (C 1 -C 8 )alkoxy, or hydroxy; and X is 0, N(H), N(Ac), N(toluene-4 10 sulfonyloxy), or a pharmaceutically acceptable salt thereof.
12. The method of claim 11 wherein R 3 is OH or (C 1 -C 6 )CO 2 -.
13. The method of claim 11 or 12 wherein X is 0. 15
14. The method of claim 11 or 12 wherein X is NH.
15. The method of claim 12 wherein R 10 and R 13 are CH 3 . 20
16. The method of claim 11 wherein R 1 , R 2 or R 12 are OH.
17. The method of claim 12, 13 or 15 wherein R 1 , R 2 , R 4 , R6, R 7 , R 8 , R 9 , R 11 , R 1 2 , R 14 and R 15 are H. 25
18. The method of claim 17 wherein the compound is selected from the group consisting of (20a)-254-methyl-(22R,26)-azacyclofurost-5-en-3 -ol, (204)-254-methyl-N-acetyl-(22R,26)-azacyclofurost-5-en-3 -ol, (22R,254) (20c)-spirost-5-en-(2a,34)-diol, (20a)-25 -methyl-N-paratoluenesulfonyl (22R,26)-azacyclofurost-5-en-34-y1 paratoluenesulfonate, (22R,254)-(20a) O 0 WO 2006/044665 PCT/US2005/037023 (14a,20a)-spirost-5-en-(3p,12)-diol, (22R,25S)-(20 )-spirost-5-en-3 -ol, (22R,25 )-(20a)-spirost-5-en-3p-yl benzoate, (22S,25S)-(20S)-spirost-5-en-3p yl hexanoate, (22R,25 )-(20a)-spirost-5-en-(14,3 )-diol, (22R,25S)-(20a) spirost-5-en-3 -ol, (22R,25S)-(20a)-spirost-5-en-3p-yl succinate, and (20a) 5 25S-methyl-N-acetyl-(22S,26)-azacyclofurost-5-en-3p-yl propanoate.
19. The method of claim 18 wherein the compound is (22S,25S) (20S)-spirost-5-en-3 -yl hexanoate. 10
20. The method of claim 1, 5 or 11 wherein the mammalian neuronal precursor cells are human cells.
21. The method of claim 1, 5 or 11 wherein the mammalian neuronal precursor cells are NT2 cells. 15
22. The method of claim 20 wherein the neuronal cells exhibit the NT2N phenotype.
23. The method of claim 22 wherein the neuronal cells are neurons. 20
24. The method of claim 1, 5 or 11 wherein said contacting is carried out in vitro.
25. The method of claim 1, 5 or 11 wherein said contacting is carried 25 out in vivo.
26. The method of claim 25 wherein said contacting is carried out by sequentially or simultaneously administering said precursor cells and said compound. 30
27. The method of claim 1, 5 or 11 wherein the amount is effective to increase GFRa2 protein expression. O0 WO 2006/044665 PCT/US2005/037023
28. The method of claim 1, 5 or 11 wherein the neuronal precursor cell is a stem cell.
29. The method of claim 27 wherein the stem cell is a neuronal stem 5 cell.
30. The method of claim 27 wherein the stem cell is an embryonic stem cell. 10
31. The method of claim 27 wherein the stem cell is a multipotent adult progenitor cell (MAPC).
32. The method of claim 27 wherein the stem cell is a marrow stromal cell. 15
33. A composition comprising a therapeutically effective amount of neuronal precursor cells, in combination with a differentiation-inducing amount of a compound of formula I, II or III. 20
34. The composition of claim 33 wherein the neuronal precursor cell is a stem cell. QA
AU2005295617A 2004-10-14 2005-10-14 Neuroprotective spirostenol pharmaceutical compositions Abandoned AU2005295617A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US61869604P 2004-10-14 2004-10-14
US60/618,696 2004-10-14
US11/031,538 US20060009433A1 (en) 2003-03-14 2005-01-07 Neuroprotective spirostenol pharmaceutical compositions
US11/031,538 2005-01-07
PCT/US2005/037023 WO2006044665A2 (en) 2004-10-14 2005-10-14 Neuroprotective spirostenol pharmaceutical compositions

Publications (1)

Publication Number Publication Date
AU2005295617A1 true AU2005295617A1 (en) 2006-04-27

Family

ID=36203560

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2005295617A Abandoned AU2005295617A1 (en) 2004-10-14 2005-10-14 Neuroprotective spirostenol pharmaceutical compositions

Country Status (5)

Country Link
EP (1) EP1809298A4 (en)
JP (1) JP2008516972A (en)
AU (1) AU2005295617A1 (en)
CA (1) CA2584333A1 (en)
WO (1) WO2006044665A2 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008534623A (en) * 2005-04-01 2008-08-28 サマリタン,ファーマスーティカルス,インク. Use of spirostenol for the treatment of mitochondrial disorders
GB0619860D0 (en) * 2006-10-06 2006-11-15 Birkeland Innovasjon As Treatment of insulin resistance and disorders associated therewith
US20170129915A1 (en) * 2014-04-25 2017-05-11 Resilio Company Limited Therapeutic agent for diseases associated with nerve axon dysfunction, including therapeutic agent for alzheimer's disease
WO2024024395A1 (en) * 2022-07-29 2024-02-01 国立大学法人富山大学 Pharmaceutical or food and beverage composition for prevention or treatment of optic nerve disorder

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4303214A1 (en) * 1993-02-04 1994-08-11 Wolfgang Marks Treatment of diseases of viral, viroidal or oncogenic origin by steroid saponins or their aglycones
CN1131237C (en) * 1997-09-26 2003-12-17 中国人民解放军军事医学科学院放射医学研究所 Usage of steroi saponin for preventing and curing senile dementia and new steroid saponin
KR20090009997A (en) * 1998-03-26 2009-01-23 파이토팜 피엘씨 Smilagenin and anzurogenin-d for the treatment of alzheimer's disease
GB0019290D0 (en) * 2000-08-04 2000-09-27 Symphar Sa Methods for inducing apolipoprotein E secretion
EP1496913A4 (en) * 2002-03-15 2007-10-31 Samaritan Pharmaceuticals Inc Neuroprotective spirostenol pharmaceutical compositions
AU2003231427A1 (en) * 2002-04-15 2003-10-27 Chengdu Di'ao Pharmaceutical Group Co., Ltd. Compounds and preparation methods of carboxylate and monoester succinate derivative of diosgenin

Also Published As

Publication number Publication date
WO2006044665A3 (en) 2007-07-05
WO2006044665A2 (en) 2006-04-27
JP2008516972A (en) 2008-05-22
EP1809298A2 (en) 2007-07-25
EP1809298A4 (en) 2008-06-18
CA2584333A1 (en) 2006-04-27

Similar Documents

Publication Publication Date Title
US20060009433A1 (en) Neuroprotective spirostenol pharmaceutical compositions
AU2003218180B2 (en) Neuroprotective spirostenol pharmaceutical compositions
JP7082186B2 (en) How to treat amyotrophic lateral sclerosis with pridopidin
US20100178277A1 (en) Methods and compositions for stimulating cells
EP3570940B1 (en) Pridopidine for use in the treatment of fragile x syndrome
WO2009135091A1 (en) Use of asenapine and related compounds for the treatment of neuronal or non-neuronal diseases or conditions
AU2005295617A1 (en) Neuroprotective spirostenol pharmaceutical compositions
Chen et al. Retracted: Allopregnanolone restores the tyrosine hydroxylase‐positive neurons and motor performance in a 6‐OHDA‐injected mouse model
US20230364076A1 (en) Use of n-(3-(4-(3-(diisobutylamino)propyl)piperazin-1-yl)propyl)-1h-benzo[d]imidazol-2-amine sulphate salts and solvates thereof for the treatment of motor neuron diseases and neuromuscular junction disorders
EP3628315A1 (en) Combination of acetylcholinesterase inhibitor and 5-ht4 receptor agonist as neuroprotective agent in the treatment of neurodegenerative diseases
Yao et al. 22R-Hydroxycholesterol induces differentiation of human NT2 precursor (Ntera2/D1 teratocarcinoma) cells
CN102625703A (en) Pregnenolone sulfate for the treatment of neurologic disorders
KR20210125982A (en) 2-fluorinated bile acids for the treatment of neurodegenerative diseases
RU2814327C1 (en) Use of sulphate salts of n-(3-(4-(3-(diisobutylamino)propyl)piperazin-1-yl)propyl)-1n-benzo[d]imidazol-2-amine and their solvates for treatment of motor neuron diseases and neuromuscular diseases
Dudas et al. Low molecular weight glycosaminoglycan C3 attenuates AF64A-stimulated, low-affinity nerve growth factor receptor-immunoreactive axonal varicosities in the rat septum
EP4159239A1 (en) Agent for protecting and/or regenerating neuromuscular junctions
CN101076250A (en) Neuroprotective spirostenol pharmaceutical compositions
WO2023117990A1 (en) USE OF N-(3-(4-(3-(DIISOBUTYLAMINO)PROPYL)PIPERAZIN-1-YL)PROPYL)-1H-BENZO[d]IMIDAZOL-2-AMINE SUCCINATE SALTS AND SOLVATES THEREOF FOR THE TREATMENT OF MOTOR NEURON DISEASES AND NEUROMUSCULAR JUNCTION DISORDERS
Zhang et al. Effect of Donepezil on Vascular Dementia in Rats via PI3K/AKT Pathway
Kotlyarova et al. Comparative pharmacokinetic analysis of а novel prolonged release dosage form of lithium citrate in mice
AU2001274195B2 (en) Use of riluzole or its salts for preventing and treating adrenoleukodystrophy
JP2008534623A (en) Use of spirostenol for the treatment of mitochondrial disorders

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application