WO2023146838A1 - Méthodes de traitement ou de prévention d'une maladie neurodégénérative à l'aide d'une association d'aminostérols et d'un composé d'insuline - Google Patents

Méthodes de traitement ou de prévention d'une maladie neurodégénérative à l'aide d'une association d'aminostérols et d'un composé d'insuline Download PDF

Info

Publication number
WO2023146838A1
WO2023146838A1 PCT/US2023/011403 US2023011403W WO2023146838A1 WO 2023146838 A1 WO2023146838 A1 WO 2023146838A1 US 2023011403 W US2023011403 W US 2023011403W WO 2023146838 A1 WO2023146838 A1 WO 2023146838A1
Authority
WO
WIPO (PCT)
Prior art keywords
insulin
aminosterol
derivative
ent
subject
Prior art date
Application number
PCT/US2023/011403
Other languages
English (en)
Inventor
Denise Barbut
Michael Zasloff
Original Assignee
Enterin, 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
Application filed by Enterin, Inc. filed Critical Enterin, Inc.
Publication of WO2023146838A1 publication Critical patent/WO2023146838A1/fr

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/132Amines having two or more amino groups, e.g. spermidine, putrescine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1754Insulin-like growth factor binding proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/30Insulin-like growth factors, i.e. somatomedins, e.g. IGF-1, IGF-2

Definitions

  • the present application relates generally to methods of treatment and/or prevention using a combination of at least one aminosterol compound and at least one insulin compound.
  • Squalamine is the most abundant member of a larger aminosterol family comprising at least 12 related compounds (Rao et al., 2000). squalamine
  • Aminosterol 1436 is an aminosterol isolated from the dogfish shark, which is structurally related to squalamine (U.S. Patent No. 5,840,936; Rao, Shinnar et al. 2000). It is also known as MSI-1436, trodusquemine and produlestan.
  • Aminosterols also have potential therapeutic use in the areas of neurodegenerative disease.
  • Age-related neurodegeneration is a significant unsolved problem and challenge. The number of people over 60 years is expected to rise from 841 million in 2013 to more than 2 billion in 2050 (United Nations. World population ageing 2013. United Nations Department of Economic and Social Affairs Population Division; Available online at: http://www.un.org/en/development/desa/population/publications/pdf/ageing/WorldPopulationAg eingReport2013.pdf).
  • AD Alzheimer’s Disease
  • PD Parkinson’s Disease
  • aminosterol compounds for the treatment of neurodegenerative conditions such as Alzheimer’s disease and Parkinson’s disease was previously described. See e.g., WO 2019/241503, US 2020-0038412, US 2018-0319837, and US 2020-0038413. Diseases such as Alzheimer’s disease (AD) are prevalent and place great burden on the healthcare system.
  • AD Alzheimer’s disease
  • aminosterol 1436 in delaying the maturation process of mice was previously described. See US 2020-0038412 and US 2018-0319837.
  • the present disclosure is directed to the discovery that a combination of an aminosterol and an insulin compound is effective in treating various conditions.
  • a method of treating, preventing, and/or delaying the progression and/or onset of neurodegeneration in a subject in need comprising administering to the subject a therapeutically effective amount of a combination of: (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or any combination thereof.
  • the aminosterol and insulin compound can be administered (i) concomitantly, (ii) as an admixture, (iii) separately and simultaneously or concurrently, or (iv) separately and sequentially.
  • the functional equivalent of insulin comprises an insulin-like growth factor (IGF).
  • IGF insulin-like growth factor
  • the IGF comprises IGF-1, IGF-2, or a combination thereof.
  • the IGF comprises an IGF-binding protein (IGFBP), optionally bound thereto.
  • IGF-binding protein comprises one or more of IGFBP1, IGFBP2, IGFBP3, IGFBP4, IGFBP5, IGFBP6, and IGFBP7.
  • the insulin derivative comprises lispro, prandial insulin, aspart, glulisine, novolin, velosulin, isophane (neutral protamine hagedorn), insulin glargine, detemir, degludec, humulin, novolog, or a combination thereof.
  • the neurodegeneration is age-related.
  • the neurodegeneration is correlated with one or more conditions or diseases selected from the group consisting of age-related dementia, Alzheimer’s disease, Parkinson’s disease, Lewy Body dementia, frontotemporal dementia, vascular dementia, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), multiple system atrophy (MSA), progressive supranuclear palsy (PSP), olivo-ponto-cerebellar degeneration, peripheral sensory neuropathy, cerebral palsy, and age related cognitive decline without a specific diagnosis from the group above.
  • age-related dementia Alzheimer’s disease, Parkinson’s disease, Lewy Body dementia, frontotemporal dementia, vascular dementia, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), multiple system atrophy (MSA), progressive supranuclear palsy (PSP), olivo-ponto-cerebellar degeneration, peripheral sensory neuropathy, cerebral palsy, and age related cognitive decline without a specific diagnosis from the group above
  • the progression or onset of the neurodegeneration is slowed, halted, or reversed over a defined time period following administration of (a) and (b). In some embodiments, the neurodegeneration is positively impacted by administration of (a) and (b).
  • the positive impact and/or progression of neurodegeneration is measured quantitatively or qualitatively by one or more techniques selected from the group consisting of electroencephalogram (EEG), neuroimaging, functional MRI, structural MRI, diffusion tensor imaging (DTI), [18F]fluorodeoxy glucose (FDG) PET, agents that label amyloid, [18F]F-dopa PET, radiotracer imaging, volumetric analysis of regional tissue loss, specific imaging markers of abnormal protein deposition, multimodal imaging, and biomarker analysis.
  • EEG electroencephalogram
  • neuroimaging functional MRI
  • structural MRI structural MRI
  • DTI diffusion tensor imaging
  • FDG fluorodeoxy glucose
  • radiotracer imaging volumetric analysis of regional tissue loss, specific imaging markers of abnormal protein deposition, multimodal imaging, and biomarker analysis.
  • the progression or onset of neurodegeneration is slowed, halted, or reversed by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • a method of reversibly slowing the growth, maturation, and/or aging of subject, and/or extending the potential lifespan of the subject comprising administering to the subject a therapeutically effective amount of an aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the method comprises additionally administering insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin.
  • the aminosterol has the formula: wherein:
  • R 1 is H or D
  • R 2 is H or D; provided that all of R 1 are H, all of R 2 are H, or all of R 1 and R 2 are H, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the method is directed to retarding the aging process of a subject. In some embodiments, the method is directed to extending the potential lifespan of a subject. In some embodiments, the method is directed to delaying maturation and/or slowing growth of a subject.
  • the aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin are administered during a critical “developmental window” of the subject. In some embodiments, the critical “developmental window” is prior to the onset of maturity of the subject.
  • the delayed maturation and/or slowed growth is measured by height and/or weight, as compared to a subject the same age and sex, who is not administered the aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof; and/or (b) the subject administered the aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, has delayed growth and/or maturation, as measured by height and/or weight, as compared to a subject the same age and sex and who is not administered aminosterol, by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%; and/or (c) the delayed maturation is measured by a delay in skeletal maturation; (d) the subject administered the aminosterol,
  • characteristics of aging impacted are selected from the group consisting of muscle endurance, coordination, social behavior and cognitive ability.
  • the method : (a) improves impaired muscle endurance, as compared to a subject not administered aminosterol, which is the same sex and age, by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%; and/or (b) improves impaired coordination, as compared to a subject not administered aminosterol, which is the same sex and age, by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%; and/or (c) improves impaired muscle endurance, as compared to
  • a method of treating, preventing, or delaying the onset of age- related diseases, conditions or health problems in a subject comprising administering to the subject a therapeutically effective amount of an aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, wherein the aminosterol has the formula: wherein:
  • R 1 is H or D
  • R 2 is H or D; provided that all of R 1 are H, all of R 2 are H, or all of R 1 and R 2 are H, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the age-related disease, condition, or health problem is selected from the group consisting of atherosclerosis and cardiovascular disease, cancer, arthritis, cataracts, osteoporosis, diabetes, hypertension, Alzheimer’s disease, arthritis, and osteoporosis.
  • the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and/or (b) the insulin, an immunologically active insulin fragment, a derivative thereof, or a functional equivalent thereof, are administered in combination with at least one additional active agent to achieve either an additive or synergistic effect.
  • the additional active agent is administered via a method selected from the group consisting of (a) concomitantly; (b) as an admixture; (c) separately and simultaneously or concurrently; and (d) separately and sequentially.
  • the neurodegeneration comprises: (a) Parkinson’s disease (PD) and the additional active agent comprises a drug selected from the group consisting of levodopa optionally combined with a dopa decarboxylase inhibitor such as carbidopa and benserazide, or combined with a COMT inhibitor such as tolcapone and entacapone; dopamine agonists such as bromocriptine, pergolide, pramipexole, ropinirole, piribedil, cabergoline, apomorphine, lisuride, and rotigotine; MAO-B inhibitors such as selegiline and rasagiline; amantadine; anticholinergics; clozapine; cholinesterase inhibitors for dementia; and modafinil for daytime sleepiness; (b) Alzheimer’s disease (AD) and the additional active agent comprises a drug selected from the group consisting of glutamate; antipsychotic drugs; huperzin
  • the aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof is of pharmaceutically acceptable grade.
  • the aminosterol is in the form of a phosphate.
  • the subject is a common pet, such as a dog or cat; or (b) the subject is a livestock, such as a horse, cattle, goat, sheep, pig or any farm animal. In some embodiments, the subject is a human.
  • the aminosterol or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and the insulin or fragment thereof, derivative thereof, or functional equivalent thereof are administered via a method selected from the group consisting of (a) concomitantly; (b) as an admixture; (c) separately and simultaneously or concurrently; and (d) separately and sequentially.
  • the aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and the insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin are administered via any pharmaceutically acceptable method.
  • the aminosterol or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and (b) the insulin or the fragment thereof, derivative thereof, or functional equivalent thereof are administered from a method independently selected from the group consisting of oral, nasal, pulmonary, inhalation, sublingual, buccal, rectal, vaginal, intravenous, intra-arterial, intradermal, intraperitoneal, intrathecal, intramuscular, epidural, intracerebral, intracerebroventricular, transdermal, and any combination thereof.
  • the insulin or the fragment thereof, derivative thereof, or functional equivalent thereof is administered via an insulin pump.
  • the method of administration for each of (a) and/or (b) is independently selected from nasal administration, oral administration, or a combination thereof.
  • the aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof is administered intranasally.
  • the insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin is administered intranasally.
  • the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof
  • the insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin are formulated into the same dosage form or separate dosage forms, each independently selected from the group consisting of liquid dispersions, gels, aerosols, lyophilized formulations, tablets, and capsules.
  • the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and (b) the insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin are formulated into the same dosage form or separate dosage forms, each independently selected from the group consisting of controlled release formulations, fast melt formulations, delayed release formulations, extended release formulations, pulsatile release formulations, and mixed immediate release and controlled release formulations.
  • the amount of the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof administered to the subject comprises: (a) about 0.1 to about 20 mg/kg body weight of the subject; (b) about 0.1 to about 15 mg/kg body weight of the subject; (c) about 0.1 to about 10 mg/kg body weight of the subject; (d) about 0.1 to about 5 mg/kg body weight of the subject; or (e) about 0.1 to about 2.5 mg/kg body weight of the subject.
  • the amount of the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof administered to the subject comprises: (a) about 0.001 to about 500 mg/day; (b) about 0.001 to about 250 mg/day; (c) about 0.001 to about 125 mg/day; (d) about 0.001 to about 50 mg/day; (e) about 0.001 to about 25 mg/day; (f) about 0.001 to about 10 mg/day; (g) about 0.001 to about 6 mg/day; (h) about 0.001 to about 4 mg/day; or (i) about 0.001 to about 2 mg/day.
  • the amount of insulin, an immunologically active insulin fragment, a derivative thereof, or a functional equivalent thereof can range from about 0.1 to about 2 units/kg/day.
  • the method of administration of the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof comprises oral administration and wherein the therapeutically effective amount comprises: (a) about 1 to about 300 mg/day; or (b) about 25 to about 500 mg/day.
  • the method of administration of the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof comprises intranasal administration and wherein the therapeutically effective amount comprises 0.001 mg/day up to about 6 mg/day.
  • administration of: (a) the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and/or (b) insulin, an immunologically active insulin fragment, a derivative thereof, or a functional equivalent thereof comprises administration of (a) and/or (b) on an empty stomach, optionally within two hours of the subject waking.
  • no food is consumed by the subject after about 60 to about 90 minutes from administration of the aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • aminosterol has the formula:
  • Compound III (ENT-03), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the aminosterol has the formula:
  • the aminosterol has the formula of Compound III-N:
  • the aminosterol has the formula:
  • Compound III-d4 (ENT-03 -d4), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the C25 configuration is R. In some embodiments, the C25 configuration is S.
  • the aminosterol has the formula:
  • Compound III (ENT-03-d3), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the C25 configuration is R. In some embodiments, the C25 configuration is S.
  • the aminosterol has the formula:
  • Compound IV (A5 ENT-03), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the C25 configuration is R. In some embodiments, the C25 configuration is S.
  • the aminosterol has the formula:
  • the aminosterol has the formula:
  • Compound V (A4 ENT-03), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the C25 configuration is R. In some embodiments, the C25 configuration is S.
  • the aminosterol has the formula:
  • the aminosterol is selected from the group consisting of the following, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof:
  • the insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin is selected from the group consisting of: insulin, inhaled insulin, synthetic insulin, human insulin, synthetic human insulin, porcine insulin, bovine insulin, shark insulin, rapid acting insulin analogs, long acting insulin analogs, intermediate-acting insulin analogs, mixed insulin, mixed insulin analogs, aspart insulin, glulisine insulin, lispro (lyspro) insulin, detemir insulin, glargine insulin, degludec insulin, NPH (Neutral Protamine Hagedorn) insulin, NPA insulin, insulin aspart protamine, and any combination thereof.
  • Figure 1 shows the accumulation of aminosterol 1436 within the centers of the brain that control growth, maturation, and senescence following intravenous administration to a rat via a peripheral vein, or injected directly into the 3 rd ventricle of the brain.
  • Figure 2 shows weight (g) (y axis) vs age (days) (x axis) for three groups of mice administered 10 mg/kg or 5 mg/kg aminosterol 1436 (MSI-1436), and a control group. While all animals reached the mature weight of about 40 grams, the control animals reached maturity at 120 days, and the animals that received 5 and 10 mg/kg of aminosterol 1436 reached maturity at 150 and 255 days, respectively.
  • Figure 3 shows a graph of weight (g) (y axis) vs time for animals given either vehicle or 10 mg/kg (/./?.) of aminosterol 1436 every 3 days for two doses, for a total of 20 mg/kg over a 6 day period. Animals were then weighed and body length measured once weekly for a period of 40 days. At Day 0 animals in the control group had a starting weight (g) of 16 g, while animals in the aminosterol 1436 group had a weight of 12 g. At day 40, the control group had a weight of 24 g, or an increase of 50%. In contrast, at Day 40 the aminosterol 1436 group had a weight of 11 g, or a decrease of 8.3%.
  • Figure 4 shows a crystal structure of compound R-(24).
  • Figures 5A and 5B show percent weight loss over time in mice administered ENT-02 (MSI-1436) (circles) or ENT-03 (Compound III; squares).
  • Fig. 5C shows intraperitoneal administration of ENT-03 to C57bl/6 male mice once weekly over 6 weeks caused a dose dependent weight loss.
  • Figure 6 shows the results of administration of ENT-03 (Compound III) and ENT-02 (MSI-1436) on growing mice. While both compounds affected weight gain, ENT-02 had a more profound effect, having suppressed growth as well as having induced consumption of body fat. In contrast, the animals treated with ENT-03 continued normal growth, although they “slimmed down”, suggesting that ENT-03 re-established a new optimal body weight “set point.”
  • Figure 7 shows images of a mucosal layer of the stomach showing a reduced mucosal layer in the 78 week old stomach (Fig. 7B) vs. the younger 20 week old stomach (Fig. 7A).
  • Figure 8 shows ICso curves for PTP1B inhibition by three aminosterols tested ENT-02 (MSI-1436), ENT-03 (Compound III), and D-1436; Fig. 8A) and a control PTP1B inhibitor (Fig. 8B) according to Example 1.
  • Figures 9 A-H show that PTP1B inhibition by ENT-03 ameliorates the cognitive impairment of hAPP-J20 (Figs. 9A-9D) and PS 19 (Figs. 9E-9H) mice.
  • Figures 10A-10C show volcano plots showing significance (as negative logio- transformed FDR-adjusted p-values) against the magnitude (log2-transformed fold change) of differentially expressed genes in the following three contrasts: (Fig. 10A) aged mice compared to young mice, (Fig. 10C) ENT-03 -treated aged mice compared to vehicle-treated aged mice, (Fig. 10B) ENT-03 -treated young mice compared to vehicle-treated young mice. Genes identified as having different levels between groups are represented as red (up-regulated) or blue (down-regulated) dots, and non-significant genes are represented as black dots. The horizontal red lines represent the applied p-value threshold.
  • Figures 11 A and 1 IB show chromatograms for the LC/MS/MS analysis of brain extract from elderly humans for ENT-03.
  • Fig. 11 A is the chromatogram for the brain extract and Fig.
  • IB is a quality control sample of synthetic ENT-03.
  • Figures 12A and 12B show chromatograms for the LC/MS/MS analysis of mouse pup brain extract (Fig. 12A) and liver extract (Fig. 12B).
  • Figure 13 shows approximate concentrations of ENT-03 measured in the brain and liver of neonatal mice over the first 3 weeks of life.
  • Figure 14 shows gene expression profiles of the jejunum (Figs. 14A-14D) or ileum (Figs. 14D-14F) of young and aged mice treated with ENT-03 or control.
  • Figure 15 shows a set of heat maps investigating the overlap of differentially expressed genes between pairs of contrasts.
  • the values in the plot represent the number of intersecting differentially expressed genes (adjusted p-value ⁇ 0.05) between specific pairs of contrasts.
  • the colors of the squares represent the Jaccard index (a quotient of the intersection and the union of genes) for the contrasts on the x- and y-axis: (Fig. 15 A) differentially expressed genes in both directions, (Fig. 15B) up-regulated genes, (Fig. 15C) down-regulated genes, and (Fig.
  • Figure 16 shows the results on mouse weight gain following administration of ENT-03 as compared to administration of a deuterated form of ENT-03, ENT-03D3.
  • Figures 17A and 17B show representative chromatograms of ENT-03 in 4 day old mouse brain extracts.
  • Figure 17A MRM 619.6/545.5, upper tracing: endogenous ENT-03 in extract; MRM 623.6/549.5 lower tracing: extract + 2.2 ng ENT-03-t/v/gram brain tissue;
  • Figure 17B MRM 619.6/474.5, upper tracing: endogenous ENT-03 in extract; MRM 623.6/478.5 lower tracing: extract +2.2 ng ENT-03 -t/v/gram brain tissue.
  • Figure 19 shows a Venn diagram of transcripts down-regulated in ageing and up- regulated by ENT-03. A plot showing the numbers of overlapping and non-overlapping differentially expressed genes between the two sets of transcripts that were down-regulated in old versus young mice and up-regulated upon treatment compared to control. Numbers of features are shown from treatment with ENT-02 (MSI-1436) and ENT-03.
  • Figure 20 shows a scatter plot comparing significant genes in ENT-02 (MSI-1436) vs control (young) against ENT-03 vs untreated (young). Genes are represented by points. The color of the point indicates which set the gene is assigned to. For each gene the log2(fold change) in the ENT-02 (MSI-1436) vs control (young) contrast (y-axis) and the log2(fold change) in the ENT-03 vs untreated (young) contrast (x-axis) are shown.
  • Figure 21 shows an upset plot of significant genes according to Example 12.
  • the leftmost bar chart shows the size of each set used as input.
  • the top bar chart shows the exclusive size of each set (z.e., each gene is only counted once in this bar chart).
  • the dot-plot in the centre shows the sets interacting in each case.
  • FIG. 22A A Venn diagram of overlapping genes in MSI-1436 (aminosterol 1436) vs control (young) against ENT-03 vs untreated (young) — all vs all
  • Fig. 22B shows a Venn diagram of overlapping genes in MSI-1436 vs control (young) against ENT- 03 vs untreated (young) — up vs up.
  • Fig. 22C shows a Venn diagram of overlapping genes in MSI-1436 vs control (young) against ENT-03 vs untreated (young) — down vs down.
  • Fig. 22A A Venn diagram of overlapping genes in MSI-1436 (aminosterol 1436) vs control (young) against ENT-03 vs untreated (young) — all vs all
  • Fig. 22B shows a Venn diagram of overlapping genes in MSI-1436 vs control (young) against ENT- 03 v
  • FIG. 22D shows a Venn diagram of overlapping genes in MSI-1436 vs control (young) against ENT-03 vs untreated (young) — up vs down.
  • Fig. 22E shows a Venn diagram of overlapping genes in MSI- 1436 vs control (young) against ENT-03 vs untreated (young) — down vs up.
  • Figure 23 shows a scatter plot comparing significant genes in MSI-1436 ( aminosterol 1436) vs control (old) against ENT-03 vs untreated (old). Genes are represented by points. The colour of the point indicates which set the gene is assigned to. For each gene the log2(fold change) in the MSI-1436 vs control (old) contrast (y-axis) and the log2(fold change) in the ENT- 03 vs untreated (old) contrast (x-axis) are shown.
  • Figure 24 shows an Upset plot of significant genes according to Example 12.
  • the leftmost bar chart shows the size of each set used as input.
  • the top bar chart shows the exclusive size of each set (z.e., each gene is only counted once in this bar chart).
  • the dot-plot in the centre shows the sets interacting in each case.
  • FIG. 25A shows a Venn diagram of overlapping genes in ENT-02 (MSI-1436) vs control (old) against ENT-03 vs untreated (old) — all vs all.
  • Fig. 25B shows a Venn diagram of overlapping genes in ENT-02 (MSI-1436) vs control (old) against ENT-03 vs untreated (old) — up vs up.
  • Fig. 25C shows a Venn diagram of overlapping genes in ENT-02 (MSI-1436) vs control (old) against ENT-03 vs untreated (old) — down vs down.
  • Fig. 25A shows a Venn diagram of overlapping genes in ENT-02 (MSI-1436) vs control (old) against ENT-03 vs untreated (old) — up vs down.
  • Fig. 25A shows a Venn diagram of overlapping genes in ENT-02 (MSI-1436) vs control (
  • 25D shows a Venn diagram of overlapping genes in ENT-02 (MSI-1436) vs control (old) against ENT-03 vs untreated (old) — up vs down.
  • Fig. 25E shows a Venn diagram of overlapping genes in ENT-02 (MSI-1436) vs control (old) against ENT-03 vs untreated (old) — down vs up.
  • Figure 26 shows a scatter plot comparing significant genes in Old vs young (control) against Old vs young (untreated). Genes are represented by points. The colour of the point indicates which set the gene is assigned to. For each gene the log2(fold change) in the Old vs young (control) contrast (y-axis) and the log2(fold change) in the Old vs young (untreated) contrast (x-axis) are shown.
  • Figure 27 shows an Upset plot of significant genes according to Example 12. A plot showing the interaction between sets of up- and down-regulated genes. The leftmost bar chart shows the size of each set used as input. The top bar chart shows the exclusive size of each set (z.e., each gene is only counted once in this bar chart). The dot-plot in the centre shows the sets interacting in each case.
  • FIG. 28A shows a Venn diagram of overlapping genes in Old vs young (control) against Old vs young (untreated) — all vs all.
  • Fig. 28B shows a Venn diagram of overlapping genes in Old vs young (control) against Old vs young (untreated) — up vs up.
  • Fig. 28C shows a Venn diagram of overlapping genes in Old vs young (control) against Old vs young (untreated) — down vs down.
  • Fig. 28D shows a Venn diagram of overlapping genes in Old vs young (control) against Old vs young (untreated) — up vs down.
  • Fig. 28E shows a Venn diagram of overlapping genes in Old vs young (control) against Old vs young (untreated) — down vs up.
  • Figure 29 shows a scatter plot comparing significant genes in Old vs young (ENT-02; MSI-1436) against Old vs young (ENT-03). Genes are represented by points. The colour of the point indicates which set the gene is assigned to. For each gene the log2(fold change) in the Old vs young (ENT-02; MSI- 1436) contrast (y-axis) and the log2(fold change) in the Old vs young (ENT-03) contrast (x-axis) are shown.
  • Figure 30 shows an Upset plot of significant genes according to Example 12. A plot showing the interaction between sets of up- and down-regulated genes. The leftmost bar chart shows the size of each set used as input. The top bar chart shows the exclusive size of each set (z.e., each gene is only counted once in this bar chart). The dot-plot in the centre shows the sets interacting in each case.
  • Figures 31 A-27E Fig. 31 A shows a Venn diagram of overlapping genes in Old vs young (MSI- 1436) against Old vs young (ENT-03) — all vs all. Fig.
  • 3 IB shows a Venn diagram of overlapping genes in Old vs young (MSI- 1436) against Old vs young (ENT-03) — up vs up.
  • Fig. 31C shows a Venn diagram of overlapping genes in Old vs young (MSI- 1436) against Old vs young (ENT-03) — down vs down.
  • Fig. 3 ID shows a Venn diagram of overlapping genes in Old vs young (MSI- 1436) against Old vs young (ENT-03) — up vs down.
  • Fig. 3 IE shows a Venn diagram of overlapping genes in Old vs young (MSI- 1436) against Old vs young (ENT-03) — down vs up.
  • Figure 32 Heat maps of overlaps between contrasts: A plot showing the number of overlapping selected genes between the contrasts performed. Note that the numbers on the diagonal represent the total number of selected genes found for each contrast. The colours of the squares represent the Jaccard index (the intersection over the union) for the contrasts on the x- axis with those on the y-axis.
  • Fig. 32A Heat map of overlaps of up- and down-regulated (y- axis) vs. up- and down-regulated (x-axis) selected genes for each contrast.
  • Fig. 32B Heat map of overlaps of up-regulated (y-axis) vs. up-regulated (x-axis) selected genes for each contrast.
  • Fig. 32A Heat map of overlaps of up- and down-regulated (y- axis) vs. up- and down-regulated (x-axis) selected genes for each contrast.
  • Fig. 32B Heat map of overlaps of up-regulated (y-axis) vs. up-regulated (
  • FIG. 32C Heat map of overlaps of down-regulated (y-axis) vs. down-regulated (x-axis) selected genes for each contrast.
  • Fig. 32D Heat map of overlaps of up-regulated (y-axis) vs. down- regulated (x-axis) selected genes for each contrast.
  • Figure 33 shows HPLC overlay of ENT-037?, ENT-03S, and ENT-03 (Injection volume: 1 pL).
  • Figure 34 shows a picture depicting the elimination of neuroinflammation following intranasal administration of ENT-03 to PS 19 mice, which are engineered to produce large amounts of tau-protein and develop a very aggressive form of neurodegeneration/ Alzheimer’s disease.
  • Figure 35A and 35B show improved performance by the PS19 mice following ENT-03 treatment in a Morris water maze, which is a test of spatial learning for rodents that relies on distal cues to navigate from start locations around the perimeter of an open swimming arena to locate a submerged escape platform. Spatial learning is assessed across repeated trials and reference memory is determined by preference for the platform area when the platform is absent. Trial-dependent, latent and discrimination learning can be assessed using modifications of the basic protocol. Fig.
  • 35A shows the number of training days on the X axis vs escape latency in seconds (e.g., time it takes to find the platform) for four groups of tested animals: wild type (WT) vehicle, WT ENT-03, PS19 vehicle, and PS19 ENT-03. Escape latency significantly declined for the PS19 ENT-03 group as compared to the PS19 untreated group.
  • Fig. 35B shows platform area (Y axis) vs each of the four groups of animals: WT vehicle, WT ENT-03, PS19 vehicle, and PS 19 ENT-03.
  • Figure 36 shows localization to the arcuate nucleus, proximity to NPY producing cells, and action via pstat3 stimulation in the arcuate and subventricular zone (which is the neurogenic zone).
  • the four groups of tested animals are shown on the X axis, e.g., WT vehicle, WT ENT- 03, PS19 vehicle, and PS19 ENT-03.
  • Measurement of P-STAT3, NPY, P-STAT3/NPY, and P- STAT3/NPY/DAPI is shown on the X axis.
  • Figure 37 shows a graph of ENT-03 concentration in brain of mice vs age, with the X axis showing days after birth and Y axis showing the quantity of ENT-03. As body weight increases (open triangle), the amount of brain ENT-03 sharply decreases.
  • Figure 38 shows a graph of time (days) vs body weight for ENT-03 treated animals and vehicle-treated animals.
  • ENT-03 treated animals exhibited significantly slower weight gain, and overall less weight gain, indicating an ENT-03 impact on metabolicy function.
  • FIG 39 shows the results of a glucose tolerance test (GTT), which measures normalized blood glucose (%) vs minutes for WT vehicle treated animals (6) and WT ENT-03 treated animals (8).
  • GTT glucose tolerance test
  • This invention relates to methods of treating and/or preventing neurodegeneration to a subject in need.
  • the neurodegeneration may be age-related, and/or may be correlated with a condition such as age-related dementia, Alzheimer’s disease, Parkinson’s disease, Lewy Body dementia, frontotemporal dementia, vascular dementia, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), multiple system atrophy (MSA), progressive supranuclear palsy (PSP)), olivo-ponto-cerebellar degeneration, or age related cognitive decline without a specific diagnosis from the group above.
  • age-related dementia Alzheimer’s disease, Parkinson’s disease, Lewy Body dementia, frontotemporal dementia, vascular dementia, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), multiple system atrophy (MSA), progressive supranuclear palsy (PSP)
  • olivo-ponto-cerebellar degeneration or age related cognitive decline without a specific diagnosis from the group above.
  • the method comprises administering a therapeutically effective amount of a combination of: (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof.
  • the present invention is also directed to methods of delaying maturation, retarding the aging process, and/or increasing the potential lifespan of subject, which can be an animal or human.
  • the invention is also directed to methods of preventing, treating, and/or delaying onset of age-related diseases or conditions in a subject.
  • the methods comprise administering to the subject an aminosterol, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin.
  • Aminosterols such as ENT-03 particularly when combined with insulin or an insulin analog, are expected to be useful in treating neurodegenerative diseases such as for example Alzheimer’s disease and Parkinson’s disease, as well as other neurodegenerative diseases described herein.
  • neurodegenerative diseases such as for example Alzheimer’s disease and Parkinson’s disease, as well as other neurodegenerative diseases described herein.
  • 85% of AD patients have insulin resistance or Type 2 diabetes, and insulin is known to be important in nerve cell and blood vessel health.
  • neuroinflammation is secondary to the metabolic abnormalities associated with neurodegenerative diseases.
  • ENT-03 is expected to be useful in treating neurodegenerative diseases, particularly when combined with insulin or an insulin analog, as aminosterols such as ENT-03 (i) are a centrally active inhibitor of phosphatases, including PTP1B, (ii) reverse insulin resistance in the brain, (iii) promote dose dependent weight loss, (iv) eliminate inflammation in the brain of AD models, and (v) reverse memory loss and normalize behavior in animal models of AD and increase lifespan by more than 50%. See Figs. 34 and 35 and the corresponding examples. Fig. 34 shows elimination of hypothalamic elimination, and elimination of hippocampal inflammation has also been demonstrated (data not shown). Fig. 36 shows localization to the arcuate nucleus, proximity to NPY producing cells, and action via pstat3 stimulation in the arcuate and subventricular zone (which is the neurogenic zone). See also Figs. 37, 38, and 39.
  • FIG 39 shows the results of a glucose tolerance test (GTT), which measures normalized blood glucose (%) vs minutes for WT vehicle treated animals (6) and WT ENT-03 treated animals (8).
  • GTT glucose tolerance test
  • the figure shows that ENT-03 treated animlas has a much quicker attainment of normalized blood glucose.
  • Figure 38 shows a graph of time (days) vs body weight for ENT-03 treated animals and vehicle-treated animals.
  • ENT-03 treated animals exhibited significantly slower weight gain, and overall less weight gain, indicating an ENT-03 impact on metabolicy function.
  • the present invention is based on the discovery of the unexpected and unprecedented activity of a combination of an aminosterol, including but not limited to Compound III (ENT- 03), and insulin in effectively treating mice induced to form high levels of tau protein and develop Alzheimer’s disease. See Example 1. It was observed that the diseased mice treated with this regimen experienced increase in life-span relative to untreated mice, and improvements in mobility and exploratory behavior which rendered the treated mice indistinguishable from healthy mice in these behaviors.
  • an aminosterol including but not limited to Compound III (ENT- 03)
  • insulin in effectively treating mice induced to form high levels of tau protein and develop Alzheimer’s disease. See Example 1. It was observed that the diseased mice treated with this regimen experienced increase in life-span relative to untreated mice, and improvements in mobility and exploratory behavior which rendered the treated mice indistinguishable from healthy mice in these behaviors.
  • ENT-03 treated PS19 mice showed the elimination of neuroinflammation following intranasal administration of ENT-03. This is highly surprising, as PS 19 mice are engineered to produce large amounts of tau-protein and develop a very aggressive form of neurodegeneration/ Alzheimer’s disease. The elimination of neuroinflammation was correlated with an improvement in spatial learning for the treated mice. This is shown in Figs. 35 A and 35B, which show the results for wild type and PS 19 mice following a Morris water maze test, which is a test of spatial learning for rodents that relies on distal cues to navigate from start locations around the perimeter of an open swimming arena to locate a submerged escape platform. The time it took to find the platform significantly declined for the PS19 ENT-03 group as compared to the PS 19 untreated group.
  • an aminosterol such as Compound III (ENT-03) promotes effects against a neurodegenerative disease such as AD is not yet known. However, without being bound by theory, it is theorized that the effect may be similar to that of aminosterol 1436 ’which has effects on the hypothalamus within the brain of the animal. As seen in Fig. 1, when radioactive aminosterol 1436 is administered to a rat intravenously via a peripheral vein (IV), or injected directly into the 3 rd ventricle of the brain (ICV), the compound accumulates within the centers of the brain that control growth, maturation and senescence.
  • IV peripheral vein
  • IMV 3 rd ventricle of the brain
  • the hypothalamus is a portion of the brain that contains a number of small nuclei with a variety of functions. One of the most important functions of the hypothalamus is to link the nervous system to the endocrine system via the pituitary gland. The hypothalamus is responsible for the regulation of certain metabolic processes and other activities of the autonomic nervous system.
  • Example 9 also details data supporting the use of ENT-03 (Compound III) in treating neurological diseases such as Alzheimer’s disease.
  • PTP1B dependent mechanisms have been utilized for reversal of memory impairment and normalization of behavior and reduction in neuronal loss in beta amyloid and tau mouse models of Alzheimer’s disease (Rieke, Cruz et al. 2020).
  • Other studies have shown reduction in the toxicity of beta amyloid aggregates by ENT- 02 in vitro and in a C. elegans model of Alzheimer’s disease (Limbocker, Chia et al. 2019).
  • ENT-02 (MSI- 1436) reverses several conditions (in mice) that are associated with ageing, such as metabolic syndrome, Alzheimer’s disease, atherosclerosis, cancer and a reduced capacity for regenerative repair.
  • the data in Example 6 demonstrates that ENT-03 can treat Alzheimer’s disease in murine models, which are acceptable animal models for human Alzheimer’s disease.
  • the Morris water maze was used to test the effect of ENT-03 (Compound III) on spatial learning and memory deficits in 2 mouse models of familial Alzheimer’s disease, hAPP-J20 mice that express a double mutant of the human amyloid precursor protein (Mucke et al., 2000), and PS19 mice that express the P301S mutant of the human microtubule associated protein tau (Yoshiyama et al., 2007).
  • ENT-03 Compound III
  • livestock that can be treated with the methods of the invention include but are not limited to goats, sheep, horses, rabbits, cattle, chickens and other poultry, pigs, camel, alpaca, llama, etc.
  • zoo animals that can be treated with the methods of the invention include but are not limited to elephants, lions, tigers, giraffes, etc.
  • pets that can be treated with methods of the invention include but are not limited to dogs, cats, pigs, ferrets, rabbits, rodents (e.g., gerbils, hamsters, chinchillas, rats, and guinea pigs), and avian pets (e.g., parrots, passerines, and fowl).
  • Methods of slowing maturation and aging, and extending potential lifespan are useful for example, in animal husbandry, to extend the potential lifespan of, for example, livestock animals such as horses, cattle, sheep, pigs and goats.
  • livestock animals such as horses, cattle, sheep, pigs and goats.
  • the methods of the invention can also be used to slow the growth of common pets such as dogs or cats, maintaining them in a smaller, younger state for a longer period of time than would normally occur.
  • methods of delaying aging could result in extending the potential lifespan of an animal such as a pet.
  • the methods of the invention can be administered to animals or humans either prior to maturity or after maturity.
  • the methods of the invention administered to a subject prior to maturity can result in slowed growth, slowed maturation, as well as other results described herein.
  • the methods of the invention administered to a subject after maturity can result in (1) delayed aging; (2) treating, preventing, or delaying onset of age-related diseases and/or conditions; and (3) extending potential lifespan, as well as other results described herein.
  • the invention can be used to slow the maturation and aging process in subjects, and additionally extend potential lifespan.
  • a subject can be treated with a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03) or a derivative or salt thereof prior to maturity and would thereby grow more slowly than an untreated subject, potentially resulting in an extended lifespan, barring unforeseen consequences such as infection, accidents, or organic disease.
  • the method can also comprise administering a combination of at least one aminosterol and an insulin compound described herein.
  • the invention can also be used to slow the aging process.
  • the subject would be treated at maturity and beyond with a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03) or a derivative or salt thereof, which can result in a slowed aging process for the subject as compared to an untreated subject, resulting in the treated subject remaining more youthful for a longer period of time. Characteristics of aging that may be impacted by the methods of the invention are described herein.
  • the method can also comprise administering a combination of at least one aminosterol and an insulin compound described herein.
  • the method comprises administering to a subject, such as an animal or human, a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03) or a derivative or salt thereof for a desirable period of time.
  • the age-related disease, condition, or health problem can be, for example, selected from the group consisting of atherosclerosis and cardiovascular disease, cancer, arthritis, cataracts, osteoporosis, diabetes, hypertension, Alzheimer’s disease, arthritis, and osteoporosis.
  • the method can also comprise administering a combination of at least one aminosterol and an insulin compound described herein.
  • Compound III is found in subdural hematoma fluid in humans and in mouse pup brains.
  • Compound III-7? (Compound III having a C25 stereo center of configuration R) may synthesized in the brain through the condensation of spermine and Compound I, as shown below.
  • Compound I is believed to arise from the metabolism of 27-hydroxycholesterol (also called (25R)26-dihydroxycholesterol), a biologically active oxysterol released into the circulation from many tissues, including vascular endothelium and macrophages (Griffiths et al., 2019; Bjorkhem et al., 2002; Javitt et al., 2002). Within the brain, 27-hydroxycholesterol undergoes successive metabolism by CYP27A1, CYP7B1, and HSD-3B7, possibly in that sequence (Meaney et al., 2007). The synthesis of Compound III-7?
  • the first step is likely catalyzed by a brain steroid 5 alpha-reductase.
  • the missing biosynthetic link is the enzyme that couples the spermine to the bile acid.
  • ENT-03 is the preferred compound, although any derivative or salt thereof that improves the pharmacological characteristics of Compound III can be used in the methods of the invention.
  • a derivative of Compound III may have one or more chemical modifications which do not modify, or drastically diminish, or may improve, the activity of Compound III. Such “activity” may include pharmacological targets and affinity therefore, including changes in affinities for different subtypes of a particular receptor target.
  • Such modifications are introduced to influence metabolism, ease of administration, biodistribution, or any combination thereof.
  • examples of such variants or derivatives include, but are not limited to, (1) substitutions of the sulfate or carboxylic acid by a sulfonate, sulfate, phosphate, carboxylate, or other anionic moiety chosen to circumvent metabolic removal of the sulfate moiety and oxidation of the cholesterol side chain; (2) replacement of a hydroxyl group by a non-metabolizable polar substituent, such as a fluorine atom, to prevent its metabolic oxidation or conjugation; and (3) substitution of various ring hydrogen atoms to prevent oxidative or reductive metabolism of the steroid ring system.
  • Other derivatives include replacement of one or more hydrogens of the aminosterol with deuterium or the unsaturation of any one or more C-C single bonds of the aminosterol.
  • the pharmaceutical composition can comprise one or more pharmaceutically acceptable carriers or excipients.
  • the methods of the invention can employ a formulation of Compound III as an insoluble salt of phosphate, polyphosphate, hydrochloride or an organic phosphate ester.
  • Compound III is shown below:
  • the aminosterol has the formula: wherein: R 1 is H or D; and
  • R 2 is H or D; provided that all of R 1 are H, all of R 2 are H, or all of R 1 and R 2 are H, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the aminosterol has the formula:
  • Compound III (ENT-03), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the aminosterol has the formula:
  • the aminosterol has the formula of Compound III-N:
  • the aminosterol has the formula:
  • Compound III (ENT-03 -d4), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the aminosterol has the formula:
  • Compound III (ENT-03-d3), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the aminosterol has the formula:
  • Compound IV (A5 ENT-03), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the aminosterol has the formula:
  • the aminosterol has the formula:
  • Compound V (A4 ENT-03), or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof.
  • the aminosterol has the formula:
  • Aminosterol 1436 or its derivatives or salts thereof can be administered via any pharmaceutically acceptable method.
  • the pharmaceutical composition in the methods of the invention can be administered intravenously, intradermally, subcutaneously, orally, rectally, sublingually, intrathecally, intranasally, or by inhalation.
  • Pharmaceutical compositions appropriate for each of the specific routes are utilized.
  • exemplary aminosterols include but are not limited to: Compound B, Compound F,
  • Insulin is a peptide hormone produced in the pancreas by the islets of Langerhans, which regulates the amount of glucose in the blood. Insulin is produced in the pancreas and the Brockmann body (in some fish), and released when any of several stimuli are detected. Insulin is produced commercially for use in medicine using recombinant technology. Human insulin is mainly produced either in E. coli or Saccharomyces cerevisiae and purified for medicinal use. (Baeshen et al. 2014). Insulin functions to maintain normal blood glucose levels by facilitating cellular glucose uptake, regulating carbohydrate, lipid and protein metabolism and promoting cell division and growth through its mitogenic effects.
  • Insulin is an anabolic hormone that promotes glucose uptake, glycogenesis, lipogenesis, and protein synthesis of skeletal muscle and fat tissue through the tyrosine kinase receptor pathway.
  • the most important hormone that the pancreas produces is insulin.
  • Insulin is released by the 'beta cells' in the islets of Langerhans in response to food. Its role is to lower glucose levels in the bloodstream and promote the storage of glucose in fat, muscle, liver and other body tissues.
  • the function of insulin is to promote the uptake of glucose by muscle cells that use it for energy and by fat cells that store it as triglycerides, or fats, and by liver cells. It does this by upregulating GLUT4 in muscle, fat, and liver cells.
  • AD neurodegenerative diseases
  • metabolic disease e.g., diabetes
  • neurodegenerative disorders e.g., neurodegenerative disorders
  • insulin Various forms of insulin including, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin are within the scope of the present invention.
  • an insulin “fragment” may include a small part derived, cut off, or broken from a larger peptide, polypeptide or protein, which retains the desired biological activity of the larger peptide, polypeptide or protein, for example, any biological activity (i.e., immunologically) known in the art to belong to insulin.
  • the terms “functional equivalent” or “functionally equivalent” to described different types of insulin are used interchangeably herein to refer to proteins, peptides, or polypeptides having similar or identical activity (i.e., immunologically) known in the art to belong to wild-type or native insulin.
  • the functional equivalent of insulin comprises an insulin-like growth factor (IGF).
  • the IGF comprises IGF-1, IGF-2, or a combination thereof.
  • the IGF comprises an IGF-binding protein (IGFBP), optionally bound thereto.
  • the IGF-binding protein comprises one or more of IGFBP1, IGFBP2, IGFBP3, IGFBP4, IGFBP5, IGFBP6, and IGFBP7.
  • Insulin-like growth factors (IGFs) are proteins with high sequence similarity to insulin. The similarity may comprise at least 80% sequence identity, at least 90% sequence identity, or at least 100% sequence identity to insulin.
  • “Derivatives” of insulin are used herein to refer to sequences with substantial identity to a reference insulin sequence (i.e., wild type human insulin). A skilled artisan can produce polypeptide variants having single or multiple amino acid substitutions, deletions, additions or replacements.
  • variants may include inter alia: (a) variants in which one or more amino acid residues are substituted with conservative or non-conservative amino acids; (b) variants in which one or more amino acids are added; (c) variants in which at least one amino acid includes a substituent group; (d) variants in which amino acid residues from one species are substituted for the corresponding residue in another species, either at conserved or non-conserved positions; and (d) variants in which a target protein is fused with another peptide or polypeptide such as a fusion partner, a protein tag or other chemical moiety, that may confer useful properties to the target protein, such as, for example, an epitope for an antibody.
  • a target protein is fused with another peptide or polypeptide such as a fusion partner, a protein tag or other chemical moiety, that may confer useful properties to the target protein, such as, for example, an epitope for an antibody.
  • the insulin derivative comprises a commercially available or known insulin derivative, for example, lispro, prandial insulin, aspart, glulisine, novolin, velosulin, isophane (neutral protamine hagedorn), insulin glargine, detemir, degludec, humulin, novolog, or a combination thereof.
  • the methods of the invention are administered to a subject, including a human, during a “developmental window” in the life of the subject.
  • a developmental window for example, administration of the method during a developmental window of the animal can result in reversible slowing of the growth rate and maturation process of the animal.
  • the “developmental window” is from birth to the animal or human reaches maturity as evidenced by ceased growth. In mice, this window extends from weaning to just prior to maturity, e.g., about 4-5 months of age. Comparable windows for other animals would correspond to the periods of growth specific for those animals, as described below.
  • this invention relates to methods of treating and/or preventing neurodegeneration to a subject in need, including but not limited to age-related neurodegeneration.
  • the neurodegeneration may also be correlated with age-related dementia, Alzheimer’s disease, Parkinson’s disease, Lewy Body dementia, frontotemporal dementia, vascular dementia, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), multiple system atrophy (MSA), progressive supranuclear palsy (PSP)), olivo-ponto-cerebellar degeneration, or age related cognitive decline without a specific diagnosis from the group above.
  • age-related dementia Alzheimer’s disease, Parkinson’s disease, Lewy Body dementia, frontotemporal dementia, vascular dementia, amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), multiple system atrophy (MSA), progressive supranuclear palsy (PSP)), olivo-ponto-cerebellar degeneration, or age related cognitive decline without a specific diagnosis from the group above.
  • the method comprises comprising administering a pharmaceutical composition comprising a therapeutically effective amount of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof, to the subject.
  • a pharmaceutical composition comprising a therapeutically effective amount of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof, to the subject.
  • the subject can be an animal or human.
  • the aminosterol is administered intranasally, although any pharmaceutically acceptable delivery method can be used, as detailed herein, including but not limited to oral administration.
  • the insulin compound can be administered using any pharmaceutically acceptable method.
  • Examples of neurodegenerative diseases that can be treated with methods of the invention include but are not limited to Parkinson’s disease (PD), Alzheimer’s disease (AD), Lewy body dementia (LBD), Multiple sclerosis (MS), multiple system atrophy (MSA), Progressive supranuclear palsy, Olivo-ponto-cerebellar degeneration, Amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD), frontotemporal dementia (FTD), vascular dementia, Friedreich’s ataxia and/or a related symptom in a subject in need is provided, comprising administering to the subject a therapeutically effective amount of a combination of: (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof.
  • PD Parkinson’s disease
  • AD Alzheimer’s disease
  • LDD Lewy body dementia
  • MS
  • administering results in improvement of one or more symptoms of the neurodegenerative disease or on one or more clinically accepted scoring metrics, by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • the improvement can be measured using any clinically recognized tool or assessment.
  • the period of time over which the progression or onset of a neurodegenerative disorder discussed above is measured can be for example, one or more months or one or more years, e.g., about 6 months, about 1 year, about 18 months, about 2 years, about 36 months, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, or about 20 years, or any amount of months or years in between the values of about 6 months to about 20 years or more.
  • a neurodegenerative disorder may be positively impacted by administering a pharmaceutical composition comprising a therapeutically effective amount of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof.
  • a “positive impact” includes for example slowing advancement of the condition, improving symptoms, etc.
  • a subject which can be an animal or human, a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof.
  • an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • Examples of age-related diseases that can be treated, prevented, or onset can be delayed include, but are not limited to, atherosclerosis and cardiovascular disease, cancer, arthritis, cataracts, osteoporosis, type 2 diabetes, and hypertension.
  • atherosclerosis and cardiovascular disease cancer
  • arthritis cataracts
  • osteoporosis type 2 diabetes
  • hypertension hypertension.
  • the incidence of all of these diseases increases rapidly with aging (increases exponentially with age, in the case of cancer). Of the roughly 150,000 people who die each day across the globe, about two thirds — 100,000 per day — die of age-related causes. In industrialized countries, the proportion is higher, reaching 90%.
  • Age related health conditions include but are not limited to arthritis, heart disease, atherosclerosis, Cardiovascular disease (CVD), osteoporosis, cataracts, Hypertension (HTN or HT), and diabetes. These age-related health conditions may be positively impacted by administering a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol disclosed herein, or a derivative or salt thereof.
  • a “positive impact” includes for example slowing advancement of the condition, improving symptoms, etc.
  • the progression or onset of the age related health condition is slowed or prevented over a defined time period as measured by a medically- recognized technique.
  • the progression or onset of the age related health condition can be slowed by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • the invention is directed to methods of delaying growth and/or maturation of a subject, which can be a human or animal, comprising administering a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof to the subject.
  • the method can additionally comprise administering an insulin compound.
  • Administration of the pharmaceutical composition comprising an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof can be limited to a brief period during development of a subject, sufficient to slow the rate of growth.
  • administration of the pharmaceutical composition comprising an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) can be as a maintenance protocol.
  • mice with various pituitary mutations that result in small body size live among the longest of any strain of mouse (Blagosklonny, 2013).
  • Mice that grow slowly during the first 2-3 months of age outlive mice that grow more rapidly during that early period of life (Miller et al., 2002).
  • Suppression of the growth hormone/IGF-1 hormonal axis is believed to be, in part, involved in the delay in early growth (Vanhooren and Libert, 2013) though how a delay in early growth translates into longevity is unknown.
  • the extent of the growth and/or maturation delay can be controlled, with a greater delay occurring with greater doses and longer duration of administration of aminosterol.
  • the growth rate of a subject which can be an animal or human, can be slowed in a measured fashion from a few % to over 50% as compared to that of an untreated subject, which is the same sex and age.
  • a subject treated at an early age with a dose that reduced growth to 50% normal would take twice as long to reach the size of an untreated subject.
  • the treated sibling would resemble a 2.5 month old and not reach maturity until 10 months of age. It is theorized that the treated subject with slowed growth will simultaneously age more slowly than the untreated subject and is anticipated to live longer.
  • administering delays growth, as measured by height and/or weight, as compared to a subject who is not administered the same aminosterol or a derivative or salt thereof, which is the same sex and age.
  • the delay can be for example about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • the delay in growth can be measured over any time period, and will vary depending upon the subject being treated. See e.g., Fig. 2.
  • Maturation can be measured in a variety of ways. In humans, maturation is simply the process of children growing and obtaining adult stature. Females tend to mature sooner than boys. Maturation as the process from early childhood, to adolescence and then to full adult stature. Childhood is generally regarded as the time until which one reaches adolescence. The start of adolescence begins with the onset of puberty where hormonal and physical changes begin to occur. Initially, rapid changes begin to occur with increases in height, weight, stature and the development of secondary sex characteristics (Lloyd, R. S., and Oliver, J. L. Strength and Conditioning for Young Athletes: Science and Application. Routledge, 2014.
  • administering delays maturation, as measured by skeletal maturation, as compared to a subject who is not administered an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof, which is the same sex and age.
  • the delay can be for example about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • the delay in maturation can be measured over any time period, and will vary depending upon the subject being treated. For example, a mouse having a typical maturation period of 40 days will have a delay in maturation upon administration of a method according to the invention over a typical period of about 150-300 days, depending upon the dose of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof administered.
  • an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • Bone age is the degree of maturation of a child’s bones. As a person grows from fetal life through childhood, puberty, and finishes growth as a young adult, the bones of the skeleton change in size and shape. These changes can be seen by x-ray.
  • the “bone age” of a child is the average age at which children reach this stage of bone maturation. See “Bone age,” Wikipedia (https://en.wikipedia.org/wiki/Bone_age
  • the long bones are those that grow primarily by elongation at an epiphysis at one end of the growing bone.
  • the long bones include the femurs, tibias, and fibulas of the lower limb, the humeri, radii, and ulnas of the upper limb (arm + forearm), and the phalanges of the fingers and toes.
  • the epiphyses become calcified and appear on the x-rays, as do the carpal and tarsal bones of the hands and feet, separated on the x-rays by a layer of invisible cartilage where most of the growth is occurring.
  • the invention is directed to methods of retarding the aging process of a subject, which can be an animal or human, comprising administering a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof to the subject.
  • a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof to the subject.
  • the composition can be administered either prior to or after maturity of the subject.
  • the method can also comprise administration of at least one insulin compound described herein.
  • Characteristics of aging that may be impacted by administration of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof include, but are not limited to, aspects of aging impacted by the hypothalamus. It has been theorized that the endocrine function of the hypothalamus essentially controls the aging process. T. Hicklin (2017). The hypothalamus is known to regulate important processes including growth, development, reproduction and metabolism. It has also been shown that the hypothalamus regulates aging throughout the body. Hypothalamic stem cells appear to exert their anti-aging effects by releasing molecules called microRNAs (miRNAs). Zhang et al. (2017); Zhang et al. (2013).
  • miRNAs microRNAs
  • Delays in aging can be measured by tissue analysis and behavioral testing to assess changes in a subject’s age-impaired muscle endurance, coordination, social behavior and cognitive ability.
  • Muscular endurance which is the ability to use muscles for extended periods of time at less than their full strength, can be measured using a variety of methods. For example, ACSM (2000) recommends the partial curl-up test to measure endurance of the abdominal muscles and the push-up test to assess endurance of the upper body. Coordination is evaluated by testing the patient’s ability to perform rapidly alternating and point-to-point movements correctly.
  • Cognitive ability can be measured using cognitive ability tests. Cognitive ability tests assess abilities involved in thinking (e.g., reasoning, perception, memory, verbal and mathematical ability, and problem solving).
  • Examples of cognitive ability tests include but are not limited to the Cognitive Abilities Test (CogAT), Wechsler Adult Intelligence Scale for adults and the Wechsler Intelligence Scale for Children for school-age test-takers, the Stanford-Binet Intelligence Scales, Woodcock-Johnson Tests of Cognitive Abilities, the Kaufman Assessment Battery for Children, the Cognitive Assessment System, and the Differential Ability Scales.
  • CogAT Cognitive Abilities Test
  • Wechsler Adult Intelligence Scale for adults and the Wechsler Intelligence Scale for Children for school-age test-takers
  • Stanford-Binet Intelligence Scales Woodcock-Johnson Tests of Cognitive Abilities
  • the Kaufman Assessment Battery for Children the Cognitive Assessment System
  • Differential Ability Scales Differential Ability Scales.
  • administering improves impaired muscle endurance, and/or improves impaired coordination, and/or improves impaired cognitive ability, as compared to an untreated subject, which is the same sex and age, by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%.
  • an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • a derivative or salt thereof improves impaired muscle endurance, and/or improves impaired coordination, and/or improves impaired cognitive ability, as compared to an untreated subject, which is the same sex and age, by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 9
  • One embodiment of the invention is directed to methods of extending the potential lifespan of a subject, which can be an animal or human.
  • a subject has not yet reached maturity, and in another aspect the subject to be treated has reached maturity.
  • the methods comprise administering a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof to the subject.
  • the method can also comprise administration of at least one insulin compound.
  • Lifespan and life expectancy are not synonymous. Life expectancy is defined statistically as the mean number of years remaining for an individual or a group of people at a given age. Id. Life expectancy increases with age as the individual survives the higher mortality rates associated with childhood. Life expectancy is an average for all people in the population — including those who die shortly after birth, those who die in early adulthood (e.g. childbirth, war), and those who live unimpeded until old age. Lifespan is an individual-specific concept — maximum lifespan is therefore an upper bound rather than an average.
  • an increased lifespan is defined as a lifespan greater than life expectancy.
  • a dog administered a composition of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) according to the invention is projected to live longer than a dog having the same life expectancy but which is not treated with a method according to the invention.
  • Life expectancies for different animals, breeds of animals, humans in various countries, etc. are all readily available.
  • a subject treated with a pharmaceutical composition comprising a therapeutically effective amount of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) formulation has an increased lifespan, as compared to a control, of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, or about 50%.
  • a “control” is defined as an animal which is the same sex, same age, and same type/breed. For a human, a “control” refers to the same sex, same age, same socioeconomic background, and same geographic residence.
  • compositions may be used for administration of aminosterols or derivatives or salts thereof.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Any pharmaceutically acceptable dosage form may be employed in the methods of the invention.
  • the composition can be formulated into a dosage form selected from the group consisting of liquid dispersions, gels, aerosols, lyophilized formulations, tablets, capsules, or an intranasal formulations utilizing a powder or liquid.
  • the aminosterol or derivatives or salts thereof may be incorporated into a dosage form selected from the group consisting of controlled release formulations, fast melt formulations, delayed release formulations, extended release formulations, pulsatile release formulations, and mixed immediate release and controlled release formulations.
  • the dosage form may comprise a combination of the forgoing formulation options (e.g., a controlled release tablet).
  • An exemplary dosage form is a nasal spray.
  • a nasal spray is designed to deliver drug to the upper nasal cavity, and can be a liquid or powder formulation, and in a dosage form such as an aerosol, liquid spray, or powder.
  • Another exemplary dosage form is an orally administered dosage form, such as a tablet or capsule.
  • These dosage forms can be formulated by any method known in the art. Such methods include the step of bringing into association the aminosterol or derivatives or salts thereof with the carrier that constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • a nasal spray comprising a dry powder, liquid suspension, liquid emulsion, or other suitable nasal dosage form.
  • an oral dosage form is a liquid, capsule, or tablet designed to disintegrate in either the stomach, upper small intestine, or more distal portions of the intestine with a dissolution rate appropriate to achieve the intended therapeutic benefit.
  • Formulations or compositions of the invention may be packaged together with, or included in a kit along with instructions or a package insert.
  • Such instructions or package inserts may address recommended storage conditions, such as time, temperature and light, taking into account the shelf-life of the aminosterol or derivatives or salts thereof.
  • Such instructions or package inserts may also address the particular advantages of the aminosterol or derivatives or salts thereof, such as the ease of storage for formulations that may require use in the field, outside of controlled hospital, clinic or office conditions.
  • composition comprising an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or derivatives or salts thereof will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual patient, the method of administration, the scheduling of administration, and other factors known to practitioners.
  • the “effective amount” for purposes herein is thus determined by such considerations.
  • the dosage of an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • a derivative or salt thereof is selected from the group consisting of 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, or 150 mg/kg.
  • the dosage of an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • a derivative or salt thereof is selected from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25,
  • an effective oral dose generally falls between about 10 mg to about 400 mg, or about 50 mg to about 350 mg, or about 100 mg to about 300 mg, or about 100 mg to about 200 mg.
  • an effective dose may be about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, about 200, about 205, about 210, about 215, about 220, about 225, about 230, about 235, about 240, about 245, about 250, about 255, about 260, about 265, about 270, about 275, about 280, about 285, about 290, about 295,
  • Dosing period The pharmaceutical composition comprising an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof can be administered for any suitable period of time, including as a maintenance dose for a prolonged period of time. Dosing can be done on an as needed basis using any pharmaceutically acceptable dosing regimen. For example, dosing can be once or twice daily, once every other day, once every three days, once every four days, once every five days, once every six days, once a week, or divided over multiple time periods during a given day (e.g., twice daily). The dosing schedule may include administration during the morning, midday, or during the evening, or a combination thereof.
  • an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • Dosing can be done on an as needed basis using any pharmaceutically acceptable dosing regimen. For example, dosing can be once or twice daily, once every other day, once every three days, once every four days, once every five days, once
  • the composition can be administered: (1) as a single dose, or as multiple doses over a period of time; (2) at a maintenance dose for an indefinite period of time; (3) once, twice or multiple times; (4) daily, every other day, every 3 days, weekly, or monthly; (5) for a period of time such as 1, 2, 3, or 4 weeks, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months, 1 year, 1.5 years, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 10.5, 11, 11.5, 12,
  • Exemplary dosing regimens include, but are not limited to: Initiating with a “low” initial daily dose, and gradually increasing the daily dose until a dose is reached that elicits evidence of a measurable impact, e.g., slowed growth rate (e.g., height and weight), improved age-related conditions (e.g., muscle endurance, coordination, social behavior and cognitive ability), or other indicia of desirable effects.
  • a “low” dose is from about 10 to about 100 mg per person, and the final effective daily dose may be between about 25 to about 1000 mg/person.
  • Another exemplary dosing regimen includes: Initiating with a “high” initial dose, and reducing the subsequent daily dosing to that required to elicit a desirable response, with the “high” daily dose being between about 50 to about 1000 mg/person, and the subsequent lower daily oral dose being between about 25 to about 500 mg/person.
  • Yet another exemplary dosing regimen includes periodic dosing, where an effective dose can be delivered once every about 1, about 2, about 3, about 4, about 5, about 6 days, or once weekly, with the initial dose determined to be capable of delaying maturation, retarding the aging process, and/or extending the potential lifespan of a subject, which can be an animal or human.
  • the first or initial “large” dose of an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • a derivative or salt thereof can be selected from the group consisting of about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, about 300, about 325, about 350, about 375, about 400, about 425, about 450, about 475, about 500, about 525, about 550, about 575, about 600, about 625, about 650, about 675, about 700, about 725, about 750, about 775, about 800, about 825, about 850, about 875, about 900, about 925, about 950, about 975, about 1000, about 1025, about 1050, about 1075, about 1100, about 1125, about 1150, about 1175, about 1200, about 1225, about 1250, about
  • the second smaller dose of an aminosterol is less than the first or initial dose and can be selected from the group consisting of about, 10, about 25, about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, about 300, about 325, about 350, about 375, about 400, about 425, about 450, about 475, about 500, about 525, about 550, about 575, about 600, about 625, about 650, about 675, about 700, about 725, about 750, about 775, about 800, about 825, about 850, about 875, about 900, about 925, about 950, about 975, or about 1000 mg.
  • an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • a derivative or salt thereof is less than the first or initial dose and can be selected from the group consisting of about, 10, about 25, about 50, about 75, about 100, about 125, about 150, about 175, about 200, about
  • the periodic aminosterol or a derivative or salt thereof dosage can be selected from the group consisting of about 10, about 25, about 50, about 75, about 100, about 125, about 150, about 175, about 200, about 225, about 250, about 275, about 300, about 325, about 350, about 375, about 400, about 425, about 450, about 475, about 500, about 525, about 550, about 575, about 600, about 625, about 650, about 675, about 700, about 725, about 750, about 775, about 800, about 825, about 850, about 875, about 900, about 925, about 950, about 975, and about 1000 mg.
  • Any pharmaceutical used for therapeutic administration can be sterile. Sterility is readily accomplished by for example filtration through sterile filtration membranes (e.g., 0.2 micron membranes). Any pharmaceutically acceptable sterility method can be used in the compositions of the invention.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more pharmaceutical compositions useful in the disclosed methods of treatment.
  • the kits may include, for instance, containers filled with an appropriate amount of a pharmaceutical composition, either as a powder, to be dissolved, or as a sterile solution, in addition to the aminosterol or a derivative or salt thereof.
  • Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the aminosterol or a derivative or salt thereof may be employed in conjunction with other therapeutic compounds.
  • compositions according to the invention may also comprise one or more binding agents, filling agents, lubricating agents, suspending agents, sweeteners, flavoring agents, preservatives, buffers, wetting agents, disintegrants, effervescent agents, and other excipients.
  • excipients are known in the art.
  • filling agents include lactose monohydrate, lactose anhydrous, and various starches
  • binding agents include various celluloses and cross-linked polyvinylpyrrolidone, microcrystalline cellulose, such as Avicel® PHI 01 and Avicel® PHI 02, microcrystalline cellulose, and silicified microcrystalline cellulose (ProSolv SMCCTM).
  • Suitable lubricants may include colloidal silicon dioxide, such as Aerosil® 200, talc, stearic acid, magnesium stearate, calcium stearate, and silica gel.
  • colloidal silicon dioxide such as Aerosil® 200, talc, stearic acid, magnesium stearate, calcium stearate, and silica gel.
  • sweeteners may include any natural or artificial sweetener, such as sucrose, xylitol, sodium saccharin, cyclamate, aspartame, and acesulfame.
  • sweeteners may include any natural or artificial sweetener, such as sucrose, xylitol, sodium saccharin, cyclamate, aspartame, and acesulfame.
  • flavoring agents are Magnasweet® (trademark of MAFCO), bubble gum flavor, and fruit flavors, and the like.
  • preservatives examples include potassium sorbate, methylparaben, propylparaben, benzoic acid and its salts, other esters of parahydroxybenzoic acid such as butylparaben, alcohols such as ethyl or benzyl alcohol, phenolic compounds such as phenol, or quaternary compounds such as benzalkonium chloride.
  • Suitable diluents include pharmaceutically acceptable inert fillers, such as microcrystalline cellulose, lactose, dibasic calcium phosphate, saccharides, and/or mixtures of any of the foregoing.
  • diluents include microcrystalline cellulose, such as Avicel® PHI 01 and Avicel® PHI 02; lactose such as lactose monohydrate, lactose anhydrous, and Pharmatose® DCL21; dibasic calcium phosphate such as Emcompress®; mannitol; starch; sorbitol; sucrose; and glucose.
  • Suitable disintegrants include lightly crosslinked polyvinyl pyrrolidone, corn starch, potato starch, maize starch, and modified starches, croscarmellose sodium, cross-povidone, sodium starch glycolate, and mixtures thereof.
  • effervescent agents include effervescent couples such as an organic acid and a carbonate or bicarbonate.
  • Suitable organic acids include, for example, citric, tartaric, malic, fumaric, adipic, succinic, and alginic acids and anhydrides and acid salts.
  • Suitable carbonates and bicarbonates include, for example, sodium carbonate, sodium bicarbonate, potassium carbonate, potassium bicarbonate, magnesium carbonate, sodium glycine carbonate, L-lysine carbonate, and arginine carbonate.
  • sodium bicarbonate component of the effervescent couple may be present.
  • Optimal oral dosing appears to be on an empty stomach.
  • An aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • a derivative or salt thereof for example, is expected to bind tightly to foodstuff, and be unavailable to interact with the intestinal epithelium. Only as the food material is digested is the aminosterol or a derivative or salt thereof freed.
  • an aminosterol i.e., ENT-03 or another aminosterol disclosed herein
  • a derivative or salt thereof is administered in combination with at least one additional active agent to achieve either an additive or synergistic effect.
  • the additional active agent can be administered via a method selected from the group consisting of (a) concomitantly; (b) as an admixture; (c) separately and simultaneously or concurrently; or (d) separately and sequentially.
  • the additional active agent is a different aminosterol from that administered in primary method.
  • the method of the invention comprises administering a first aminosterol which is an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a salt or derivative thereof intranasally and administering a second aminosterol, different from the first aminosterol or a salt or derivative thereof, orally.
  • a first aminosterol which is an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a salt or derivative thereof intranasally and administering a second aminosterol, different from the first aminosterol or a salt or derivative thereof, orally.
  • each dose of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof is taken on an empty stomach, optionally within about two hours of the subject waking.
  • no food is taken or consumed after about 60 to about 90 minutes of taking the dose of an aminosterol (i.e., ENT-03 or another aminosterol disclosed herein) or a derivative or salt thereof.
  • the aminosterol or a derivative or salt thereof can be a pharmaceutically acceptable grade of at least one aminosterol or a derivative or salt thereof.
  • the subject can be a human.
  • the subject to be treated according to the methods of the invention can be a member of a patient population at risk for being diagnosed with neurodegenerati on .
  • the invention encompasses a method of treating, preventing and/or slowing the onset or neurodegeneration and/or a related symptom in a subject in need.
  • the neurodegeneration is correlated with abnormal a- synuclein (aS) pathology and/or dopaminergic dysfunction.
  • AS a- synuclein
  • the method comprises (a) determining a dose of an aminosterol or a salt or derivative thereof for the subject, wherein the dose of the aminosterol or a salt or derivative thereof is determined based on the effectiveness of the dose in improving or resolving a neurodegeneration symptom being evaluated, (b) followed by administering the dose to the subject for a period of time, wherein the method comprises (i) identifying a neurodegeneration symptom to be evaluated; (ii) identifying a starting dose of the aminosterol or a salt or derivative thereof for the subject; and (iii) administering an escalating dose of the aminosterol or a salt or derivative thereof to the subject over a period of time until an effective dose for the neurodegeneration symptom being evaluated is identified, wherein the effective dose is the aminosterol dose where improvement or resolution of the neurodegeneration symptom is observed, and fixing the dose at that level for that particular neurodegeneration symptom in that particular subject.
  • starting dosages of the aminosterol or a salt or derivative thereof for oral administration can range, for example, from about 1 mg up to about 175 mg/day, or any amount in-between these two values.
  • An exemplary starting dosage is 25 mg/day.
  • the composition is administered orally and the dosage is escalated in about 25 mg increments.
  • the composition is administered orally and the dose of aminosterol or a salt or derivative thereof for the subject following dose escalation is fixed at a range of from about 1 mg up to about 500 mg/day, or any amount in-between these two values.
  • the dose of the aminosterol or a salt or derivative thereof for the subject following escalation is fixed at a dose of about 1, about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, about 200, about 205, about 210, about 215, about 220, about 225, about 230, about 235, about 240, about 245, about 250, about 255, about 260, about 265, about 270, about 275, about 280, about 285, about 290, about 295, about 300, about 305, about 310, about 315, about 320, about 325, about
  • the composition is administered intranasally (IN) and the starting dosage of the aminosterol or a salt or derivative thereof ranges from about 0.001 mg to about 3 mg/day, or any amount in-between these two values.
  • the starting dosage for IN administration, prior to dose escalation can be, for example, about 0.001, about 0.005, about 0.01, about 0.02, about 0.03, about 0.05, about 0.06, about 0.07, about 0.08, about 0.09, about 0.1, about 0.15, about 0.2, about 0.25, about 0.3, about 0.35, about 0.4, about 0.45, about 0.5, about 0.55, about 0.6, about 0.65, about 0.7, about 0.75, about 0.8, about 0.85, about 0.9, about 1.0, about 1.1, about 1.25, about 1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.75, about 1.8, about 1.9, about 2.0, about 2.1, about 2.25, about 2.3, about 2.4,
  • the composition is administered intranasally and the dosage of the aminosterol or a salt or derivative thereof is escalated in increments of about 0.01, about 0.05, about 0.1, about 0.2, about 0.25, about 0.3, about 0.35, about 0.4, about 0.45, about 0.5, about 0.55, about 0.6, about 0.65, about 0.7, about 0.75, about 0.8, about 0.85, about 0.9, about 0.95, about 1, about 1.1, about 1.2, about 1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.8, about 1.9, or about 2 mg.
  • the composition is administered intranasally and the dose of the aminosterol or a salt or derivative thereof for the subject following escalation is fixed at a range of from about 0.001 mg up to about 6 mg/day, or any amount in-between these two values.
  • the composition is administered intranasally and the dose of the aminosterol or a salt or derivative thereof for the subject following dose escalation is a dose which is sub therapeutic when given orally or by injection.
  • the aminosterol or a salt or derivative thereof is formulated for intranasal administration in a composition which is a dry powder nasal spray or liquid nasal spray.
  • the dosage of the aminosterol or a salt or derivative thereof is escalated every about 3 to about 5 days.
  • the dose of the aminosterol or a salt or derivative thereof is escalated about Ix/week, about 2x/week, about every other week, or about Ix/month.
  • the dose of the aminosterol or a salt or derivative thereof is escalated every about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, or about 14 days.
  • the fixed dose of the aminosterol or a salt or derivative thereof is given once per day, every other day, once per week, twice per week, three times per week, four times per week, five times per week, six times per week, every other week, or every few days.
  • the fixed dose of the aminosterol or a salt or derivative thereof can be administered for a first defined period of time of administration, followed by a cessation of administration for a second defined period of time, followed by resuming administration upon recurrence of SZ or a symptom of SZ.
  • the fixed dose can be incrementally reduced after the fixed dose of the aminosterol or a salt or derivative thereof has been administered to the subject for a period of time.
  • the fixed dose is varied plus or minus a defined amount to enable a modest reduction or increase in the fixed dose.
  • the fixed dose can be increased or decreased by about 1%, about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, or about 20%.
  • a “fixed aminosterol dose,” also referred to herein as a “fixed escalated aminosterol dose,” which will be therapeutically effective is determined for each patient by establishing a starting dose of an aminosterol composition and a threshold for improvement of a particular symptom which is used as a tool or marker for evaluating the effectiveness of the aminosterol dosage. Following determining a starting aminosterol dosage for a particular patient, the aminosterol dose is then progressively escalated by a consistent amount over consistent time intervals until the desired improvement is achieved; this aminosterol dosage is the “fixed escalated aminosterol dosage” for that particular patient for that particular symptom. In exemplary embodiments, an orally administered aminosterol dose is escalated every about 3 to about 5 days by about 25 mg until the desired improvement is reached.
  • Symptoms evaluated, along with tools for measuring symptom improvement may be specifically described below, including but not limited to constipation, hallucinations, sleep disturbances (e.g. REM disturbed sleep or circadian rhythm dysfunction), cognitive impairment, depression, or alpha-synuclein aggregation.
  • sleep disturbances e.g. REM disturbed sleep or circadian rhythm dysfunction
  • cognitive impairment e.g. depression, or alpha-synuclein aggregation.
  • This therapeutically effective “fixed dose” is then maintained throughout treatment and/or prevention. Thus, even if the patient goes “off drug” and ceases taking the aminosterol composition, the same “fixed dose” is taken with no ramp up period following re-initiation of aminosterol treatment. Not to be bound by theory, it is believed that the aminosterol dose is dependent on the severity of nerve damage relating to the symptom establishing the “fixed dose” threshold (e.g. for constipation, the dose may be related to the extent of nervous system damage in the patient’s gut).
  • Dose escalation When determining a “fixed aminosterol dosage” for a particular patient, a patient is started at a lower dose and then the dose is escalated until a positive result is observed for the symptom being evaluated.
  • An exemplary symptom to be evaluated can be constipation, but any symptom associated with the disease or disorder to be treated can be used as a marker for evaluating aminosterol dosage.
  • Aminosterol doses can also be de-escalated (reduced) if any given aminosterol dose induces a persistent undesirable side effect, such as diarrhea, vomiting, or nausea.
  • the starting aminosterol dose is dependent on the severity of the symptom - e.g. for a patient experiencing severe constipation, defined as less than one spontaneous bowel movement (SBM) a week, the starting oral aminosterol dose can be about 150 mg/day or greater. In contrast, for a patient having moderate constipation, e.g., defined as having more than one SBM a week, the starting oral aminosterol dose can be about 75 mg/day. Thus, as an example, a patient experiencing moderate constipation can be started at an oral aminosterol dosage of about 75 mg/day, whereas a patient experiencing severe constipation can be started at an oral aminosterol dosage of about 150 mg/day.
  • SBM spontaneous bowel movement
  • a patient experiencing moderate symptoms can be started at an oral aminosterol dosage of from about 10 mg/day to about 75 mg/day, or any amount in-between these values.
  • the starting oral aminosterol dosage for a moderate symptom can be about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 60, about 65, about 70, or about 75 mg/day.
  • the patient when the patient is experiencing severe symptoms (for the symptom being used to calculate the fixed escalated aminosterol dose), the patient can be started at an oral aminosterol dosage ranging from about 75 to about 175 mg/day, or any amount inbetween these two values.
  • the starting oral aminosterol dosage for a severe symptom can be about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150 about 155, about 160, about 165, about 170, or about 175 mg/day.
  • the starting oral aminosterol dose may be about 125 mg or about 175 mg/day; again dependent on the severity of the symptom, such as constipation.
  • Starting intranasal (IN) aminosterol dosages prior to dose escalation can be, for example, about 0.001 mg to about 3 mg/day, or any amount in-between these two values.
  • the starting aminosterol dosage for IN administration, prior to dose escalation can be, for example, about 0.001, about 0.005, about 0.01, about 0.02, about 0.03, about 0.05, about 0.06, about 0.07, about 0.08, about 0.09, about 0.1, about 0.15, about 0.2, about 0.25, about 0.3, about 0.35, about 0.4, about 0.45, about 0.5, about 0.55, about 0.6, about 0.65, about 0.7, about 0.75, about 0.8, about 0.85, about 0.9, about 1.0, about 1.1, about 1.25, about 1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.75, about 1.8, about 1.9, about 2.0, about 2.1, about 2.25, about 2.3, about 2.4, about 2.5, about 2.6
  • the aminosterol dose is given periodically as needed.
  • the aminosterol dose can be given once per day.
  • the aminosterol dose can also be given every other day, 2, 3, 4, or 5x per week, once/week, or 2x/week.
  • the aminosterol dose can be given every other week, or it can be given for a few weeks, followed by skipping a few weeks (as the effects persist following treatment), followed by restarting aminosterol treatment.
  • the dose can be escalated following any suitable time period.
  • the aminosterol dose is escalated every about 3 to about 7 days by about a defined amount until a desired improvement is reached.
  • threshold improvement can be an increase of one SBM per week or at least a total of three bowel movements per week.
  • the aminosterol dose can be escalated every about 1, about 2, about 3, about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, or about 14 days.
  • the aminosterol dose can be escalated about Ix/week, about 2x/week, about every other week, or about Ix/month.
  • the aminosterol dosage can be increased by a defined amount.
  • the dose can be escalated in increments of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, or by about 50 mg.
  • the dosage can be increased in increments of about, for example, about 0.1, about 0.2, about 0.25, about 0.3, about 0.35, about 0.4, about 0.45, about 0.5, about 0.55, about 0.6, about 0.65, about 0.7, about 0.75, about 0.8, about 0.85, about 0.9, about 0.95, about 1, about 1.1, about 1.2, about 1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.8, about 1.9, or about 2 mg.
  • neurodisease symptoms described herein include, but are not limited to, (a) at least one non-motor aspect of experiences of daily living as defined by Part I of the Unified Parkinson’s Disease Rating Scale (UPDRS), such as for example cognitive impairment, hallucinations and psychosis, depressed mood, anxious mood, apathy, features of dopamine dysregulation syndrome, sleep problems, daytime sleepiness, pain, urinary problems, constipation problems, lightheadedness on standing, and fatigue; (b) at least one motor aspect of experiences of daily living as defined by Part II of the UPDRS, such as for example, speech, saliva and drooling, chewing and swallowing, eating tasks, dressing, hygiene, handwriting, turning in bed, tremors, getting out of a bed, a car, or a deep chair
  • UPDRS Unified Parkinson’s Disease Rating Scale
  • the starting aminosterol or a salt or derivative thereof dose is higher if the neurodegeneration symptom being evaluated is severe.
  • the starting dose can be based on a baseline score of a cognitive test or tool, wherein if the baseline score correlates with an assessment of mild cognitive impairment, then the starting dose of aminosterol or a salt or derivative thereof is lower than if the baseline score correlates with an assessment of severe cognitive impairment.
  • a subject experiencing moderate or mild cognitive impairment as determined by a clinical scale or test is administered a starting oral dose of from about 10 to about 75 mg/day; or a subject experiencing severe cognitive impairment as determined by a clinical scale or test is administered a starting oral dose greater than about 75 mg/day.
  • the method results in slowing, halting, or reversing progression or onset of neurodegeneration over a defined period of time following administration of the fixed escalated dose of the aminosterol or a salt or derivative thereof, as measured by a medically- recognized technique.
  • the method of the invention can result in positively impacting the neurodegeneration, as measured by a medically-recognized technique.
  • the positive impact and/or progression of neurodegeneration, and/or improvement or resolution of the neurodegeneration symptom being evaluated may be measured quantitatively or qualitatively by one or more clinically recognized scales, tools, or techniques).
  • Such techniques include computed tomography (CT), magnetic resonance imaging (MRI), magnetic resonance spectroscopy, functional MRI (fMRI), diffusion tensor imaging, single photon emission computed tomography (SPECT), and positron emission tomography (PET).
  • the progression or onset of neurodegeneration may be slowed, halted, or reversed by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, as measured by a medically-recognized technique.
  • the fixed escalated dose of the aminosterol or a salt or derivative thereof reverses dysfunction caused by the neurodegeneration and treats, prevents, improves, and/or resolves the neurodegeneration symptom being evaluated.
  • the improvement or resolution of the neurodegeneration-related symptom can be measured using a clinically recognized scale or tool.
  • scales or tools include, for example, the Uniformed Parkinson’s Disease Scale (UPDRS), Mini Mental State Examination (MMSE), Mini Mental Parkinson (MMP), Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE), The 7-Minute Screen, Abbreviated Mental Test Score (AMTS), Cambridge Cognitive Examination (CAMCOG), Clock Drawing Test (CDT), General Practitioner Assessment of Cognition (GPCOG), Mini-Cog, Memory Impairment Screen (MIS), Montreal Cognitive Assessment (MoCA), Rowland Universal Dementia Assessment (RUD A), Self- Administered Gerocognitive Examination (SAGE), Short and Sweet Screening Instrument (SAS-SI), Short Sweet Test (SBT), St.
  • UDRS Uniformed Parkinson’s Disease Scale
  • MMSE Mini Mental State Examination
  • MMP Mini Mental Parkinson
  • IQCODE Informant Questionnaire on Cognitive Decline in the Elderly
  • IQCODE The 7-Minute Screen, Abbreviated Mental Test Score (AMTS), Cambridge Cognitive Examination (CAMCOG), Clock
  • the improvement in the neurodegeneration-related symptom is at least about 3%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or at least about 100%, as measured using a clinically recognized scale or tool.
  • the neurodegeneration correlated with abnormal aS pathology and/or dopaminergic dysfunction is related to or correlated with a neurodegenerative disease or neurological disease associated with neural cell death.
  • the neurodegenerative disease or neurological disease or related symptom associated with neural cell death is: (a) selected from the group consisting of septic shock, intracerebral bleeding, subarachnoidal hemorrhage, multi-infarct dementia, inflammatory diseases, neurotrauma, peripheral neuropathies, polyneuropathies, metabolic encephalopathies, and infections of the central nervous system; or(b) selected from the group consisting of synucleopathies, Alzheimer’s disease, Parkinson’s disease, dementia with Lewy bodies, multiple system atrophy, Huntington’s disease, multiple sclerosis, parkinsonism, amyotrophic lateral sclerosis (ALS), schizophrenia, Friedreich’s ataxia, vascular dementia, spinal muscular atrophy, frontotemporal dementia, supranuclear palsy, progressive supranu
  • the neurodegeneration correlated with abnormal aS pathology and/or dopaminergic dysfunction is related to or correlated with a psychological or behavioral disorder.
  • the psychological or behavioral disorder can be selected from the group consisting of aberrant motor and obsessive-compulsive behaviors, sleep disorders, REM sleep behavior disorder (RBD), depression, major depressive disorder, agitation, anxiety, delirium, irritability, ADHD, apathy, bipolar disorder, disinhibition, addiction, illusion and delusions, amnesia, autism,
  • the neurodegeneration correlated with abnormal aS pathology and/or dopaminergic dysfunction is related to or correlated with a cerebral ischemic disorder or a general ischemic disorder.
  • the cerebral ischemic disorder can be selected from the group consisting of cerebral microangiopathy, intrapartal cerebral ischemia, cerebral ischemia during/after cardiac arrest or resuscitation, cerebral ischemia due to intraoperative problems, cerebral ischemia during carotid surgery, chronic cerebral ischemia due to stenosis of bloodsupplying arteries to the brain, sinus thrombosis or thrombosis of cerebral veins, cerebral vessel malformations, and diabetic retinopathy; or the general ischemic disorder can be selected from the group consisting of high blood pressure, high cholesterol, myocardial infarction, cardiac insufficiency, cardiac failure, congestive heart failure, myocarditis, pericarditis, perimyocarditis, coronary heart disease, angina pectoris, congenital heart disease
  • the neurodegeneration-related symptom is selected from the group consisting of: cognitive impairment (CI) as determined by an IQ score; CI as determined by a memory or cognitive function test; decline in thinking and reasoning skills; confusion; poor motor coordination; loss of short term memory; loss of long term memory; identity confusion; impaired judgement; forgetfulness; depression; anxiety; irritability; obsessive-compulsive behavior; apathy and/or lack of motivation; emotional imbalance; problem solving ability; impaired language; impaired reasoning; impaired decision-making ability; impaired ability to concentrate; impaired communication; impaired ability to conduct routine tasks such as cooking; self-care, including feeding and dressing; constipation; neurodegeneration; sleep problem, sleep disorder, and/or sleep disturbance; hypertension; hypotension; sexual dysfunction; cardiovascular disease; cardiovascular dysfunction; difficulty with working memory; gastrointestinal (GI) disorders; attention deficit and hyperactivity disorder; seizures; urinary dysfunction; difficulty with mastication; vision problems; and muscle weakness.
  • cognitive impairment as determined by an IQ score
  • CI as determined by a memory or cognitive function test
  • the neurodegeneration-related symptom to be evaluated is cognitive impairment (CI) as determined by an IQ score or as determined by a memory or cognitive function test and wherein: (a) progression or onset of the CI is slowed, halted, or reversed over a defined period of time following administration of the fixed escalated dose of the aminosterol or a salt or derivative thereof, as measured by a medically-recognized technique; (b) the CI is positively impacted by the fixed escalated dose of the aminosterol or a salt or derivative thereof, as measured by a medically-recognized technique; (c) the CI is positively impacted by the fixed escalated dose of the aminosterol or a salt or derivative thereof, as measured by a medically- recognized technique and the positive impact on and/or progression of cognitive decline is measured quantitatively or qualitatively by one or more medically-recognized techniques selected from the group consisting of ADASCog, Mini-Mental State Exam(MMSE), Mini-cog
  • MMSE Mini
  • the neurodegeneration-related symptom to be evaluated is depression and (a) the method results in improvement in a subject’s depression, as measured by one or more clinically-recognized depression rating scale; (b) the method results in improvement in a subject’s depression, as measured by one or more clinically-recognized depression rating scale and the improvement is in one or more depression characteristics selected from the group consisting of mood, behavior, bodily functions such as eating, sleeping, energy, and sexual activity, and/or episodes of sadness or apathy; and/or (c) the method results in improvement in a subject’s depression, as measured by one or more clinically-recognized depression rating scale, and the improvement a subject experiences following treatment is about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95 or about 100%.
  • the one or more clinically-recognized depression rating scale can be selected from the group consisting of the Patient Health Questionnaire-9 (PHQ-9); the Beck Depression Inventory (BDI); Zung Self-Rating Depression Scale; Center for Epidemiologic Studies-Depression Scale (CES- D); and the Hamilton Rating Scale for Depression (HRSD).
  • PHQ-9 Patient Health Questionnaire-9
  • BDI Beck Depression Inventory
  • CES- D Center for Epidemiologic Studies-Depression Scale
  • HRSD Hamilton Rating Scale for Depression
  • the neurodegeneration-related symptom to be evaluated is constipation, and (a) treating the constipation prevents and/or delays the onset and/or progression of the neurodegeneration; (b) the fixed escalated aminosterol dose causes the subject to have a bowel movement; (c) the method results in an increase in the frequency of bowel movement in the subject; (d) the method results in an increase in the frequency of bowel movement in the subject and the increase in the frequency of bowel movement is defined as: (i) an increase in the number of bowel movements per week of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, and about 100%; and/or (ii) a percent decrease in the amount of time between each successive bowel movement selected from the group consisting of about 5%, about 10%, about 15%, about 20%, about
  • the neurodegeneration-related symptom to be evaluated is neurodegeneration correlated with neurodegeneration, and (a) treating the neurodegeneration prevents and/or delays the onset and/or progression of the neurodegeneration; (b) the method results in treating, preventing, and/or delaying the progression and/or onset of neurodegeneration in the subject; (c) progression or onset of the neurodegeneration is slowed, halted, or reversed over a defined period of time following administration of the fixed escalated dose of the aminosterol or a salt or derivative thereof, as measured by a medically-recognized technique; and/or (d) the neurodegeneration is positively impacted by the fixed escalated dose of the aminosterol or a salt or derivative thereof, as measured by a medically-recognized technique.
  • the positive impact and/or progression of neurodegeneration can be measured quantitatively or qualitatively by one or more techniques selected from the group consisting of electroencephalogram (EEG), neuroimaging, functional MRI, structural MRI, diffusion tensor imaging (DTI), [18F]fluorodeoxy glucose (FDG) PET, agents that label amyloid, [18F]F-dopa PET, radiotracer imaging, volumetric analysis of regional tissue loss, specific imaging markers of abnormal protein deposition, multimodal imaging, and biomarker analysis.
  • EEG electroencephalogram
  • neuroimaging functional MRI
  • structural MRI structural MRI
  • DTI diffusion tensor imaging
  • FDG fluorodeoxy glucose
  • radiotracer imaging volumetric analysis of regional tissue loss, specific imaging markers of abnormal protein deposition, multimodal imaging, and biomarker analysis.
  • the progression or onset of neurodegeneration can be slowed, halted, or reversed by about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 100%, as measured by a medically-recognized technique.
  • the neurodegeneration-related symptom to be evaluated is a sleep problem, sleep disorder, or sleep disturbance and (a) the sleep problem, sleep disorder, or sleep disturbance comprises a delay in sleep onset, sleep fragmentation, REM-behavior disorder, sleep-disordered breathing including snoring and apnea, day-time sleepiness, micro-sleep episodes, narcolepsy, circadian rhythm dysfunction, REM disturbed sleep, or any combination thereof; (b) the sleep problem, sleep disorder, or sleep disturbance comprises REM-behavior disorder, which comprises vivid dreams, nightmares, and acting out the dreams by speaking or screaming, or fidgeting or thrashing of arms or legs during sleep; (c) treating the sleep problem, sleep disorder, or sleep disturbance prevents or delays the onset and/or progression of the neurodegeneration; (d) the method results in a positive change in the sleeping pattern of the subject; wherein the positive change is defined as: (i) an increase in the total amount of sleep obtained
  • each defined period of time is independently selected from the group consisting of about 1 day to about 10 days, about 10 days to about 30 days, about 30 days to about 3 months, about 3 months to about 6 months, about 6 months to about 12 months, and about greater than 12 months.
  • the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof may be administered alone or in combination with one or more other therapeutic agents.
  • An example of an additional therapeutic agent is one known to treat the condition the aminosterol is being administered to treat.
  • the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof can be co-administered or combined with drugs commonly prescribed to treat PD or related symptoms, such as levodopa (usually combined with a dopa decarboxylase inhibitor or COMT inhibitor), dopamine agonists and MAO-B inhibitors.
  • drugs commonly prescribed to treat PD or related symptoms such as levodopa (usually combined with a dopa decarboxylase inhibitor or COMT inhibitor), dopamine agonists and MAO-B inhibitors.
  • Exemplary dopa decarboxylase inhibitors are carbidopa and benserazide.
  • Exemplary COMT inhibitors are tolcapone and entacapone.
  • Dopamine agonists include, for example, bromocriptine, pergolide, pramipexole, ropinirole, piribedil, cabergoline, apomorphine, lisuride, and rotigotine.
  • MAO-B inhibitors include, for example, selegiline and rasagiline.
  • Other drugs commonly used to treat PD include, for example, amantadine, anticholinergics, clozapine for psychosis, cholinesterase inhibitors for dementia, and modafinil for daytime sleepiness.
  • the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof can be co-administered or combined with drugs commonly prescribed to treat AD or related symptoms, such as glutamate, antipsychotic drugs, huperzine A, acetylcholinesterase inhibitors and NMDA receptor antagonists such as memantine (Akatinol®, Axura®, Ebixa®/Abixa®, Memox® and Namenda®).
  • drugs commonly prescribed to treat AD or related symptoms such as glutamate, antipsychotic drugs, huperzine A, acetylcholinesterase inhibitors and NMDA receptor antagonists such as memantine (Akatinol®, Axura®, Ebixa®/Abixa®, Memox® and Namenda
  • the aminosterol composition can be co-administered or combined with drugs commonly prescribed to treat diabetes mellitus or related symptoms, such as insulin (NPH insulin or synthetic insulin analogs) (e.g., Humulin®, Novolin®) and oral antihyperglycemic drugs.
  • drugs commonly prescribed to treat diabetes mellitus or related symptoms such as insulin (NPH insulin or synthetic insulin analogs) (e.g., Humulin®, Novolin®) and oral antihyperglycemic drugs.
  • Oral antihyperglycemic drugs include but are not limited to (1) biguanides such as metformin (Glucophage®); (2) Sulfonylureas such as acetohexamide, chlorpropamide (Diabinese®), glimepiride (Amaryl®), glipizide (Glucotrol®), tolazamide, Tolbutamide, and glyburide (Diabeta®, Micronase®); (3) Meglitinides such as repaglinide (Prandin®) and nateglinide (Starlix®); (4) Thiazolidinediones such as rosiglitazone (Avandia®) and pioglitazone (Actos®); (5) Alpha-glucosidase inhibitors such as acarbose (Precose®) and miglitol (Glyset®); (6) Dipeptidyl peptidase-4 inhibitors such as Sitagliptin (januvia®); (7)
  • the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof can be co-administered or combined with drugs commonly prescribed to treat Huntington’s chorea or related symptoms, such as medications prescribed to help control emotional and movement problems associated with Huntington’s chorea.
  • Such medications include, but are not limited to, (1) antipsychotic drugs, such as haloperidol and clonazepam; (2) drugs used to treat dystonia, such as acetylcholine regulating drugs (trihexyphenidyl, benztropine (Cogentin®), and procyclidine HC1); GABA-regulating drugs (diazepam (Valium®), lorazepam (Ativan®), clonazepam (Klonopin®), and baclofen (Lioresal®)); dopamine-regulators (levodopa/carbidopa (Sinemet®), bromocriptine (parlodel), reserpine, tetrabenazine) ; anticonvulsants (carbamazepine (Tegretol®) and botulinum toxin (Botox®)); and (3) drugs used to treat depression (fluoxetine, sertraline, and nortriptyline). Other drugs
  • peripheral sensory neuropathy In methods of treating, preventing, and/or slowing the onset or progression of peripheral sensory neuropathy or related symptoms associated with peripheral sensory neuropathy, the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof can be co-administered or combined with drugs commonly prescribed to treat peripheral sensory neuropathy or related symptoms.
  • Peripheral sensory neuropathy refers to damage to nerves of the peripheral nervous system, which may be caused either by diseases of or trauma to the nerve or the side-effects of systemic illness.
  • Drugs commonly used to treat this condition include, but are not limited to, neurotrophin-3, tricyclic antidepressants (e.g., amitriptyline), antiepileptic therapies (e.g., gabapentin or sodium valproate), synthetic cannabinoids (Nabilone) and inhaled cannabis, opiate derivatives, and pregabalin (Lyrica®).
  • neurotrophin-3 tricyclic antidepressants
  • antiepileptic therapies e.g., gabapentin or sodium valproate
  • synthetic cannabinoids e.g., gabapentin or sodium valproate
  • inhaled cannabis opiate derivatives
  • opiate derivatives opiate derivatives
  • pregabalin Logabalin
  • the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof can be co-administered or combined with drugs commonly prescribed to treat Amyotrophic lateral sclerosis or related symptoms, such as riluzole (Rilutek®), KNS-760704 (an enantiomer of pramipexole), olesoxime (TRO 19622), talampanel, arimoclomol, medications to help reduce fatigue, ease muscle cramps, control spasticity, reduce excess saliva and phlegm, control pain, depression, sleep disturbances, dysphagia, and constipation.
  • drugs commonly prescribed to treat Amyotrophic lateral sclerosis or related symptoms such as riluzole (Rilutek®), KNS-760704 (an enantiomer of
  • the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof can be coadministered or combined with drugs commonly prescribed to treat multiple sclerosis or related symptoms, such as corticosteroids (e.g., methylprednisolone), plasmapheresis, fmgolimod (Gilenya®), interferon beta-la (Avonex®, CinnoVex®, ReciGen® and Rebif®), interferon beta-lb (Betaseron® and Betaferon®), glatiramer acetate (Copaxone®), mitoxantrone, natalizumab (Ty
  • the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof can be co-administered or combined with drugs commonly prescribed to treat cognitive impairment, such as donepezil (Aricept®), galantamine (Razadyne®), rivastigmine (Exelon®); and stimulants such as caffeine, amphetamine (Adderall®), lisdexamfetamine (Vyvanse®), and methylphenidate (Ritalin®); NMDA antagonists such as memantine (Nameda®); supplements such as ginko biloba, L-theanine, piracetam, oxiracitam,
  • the combination of (a) an aminosterol comprising a spermine moiety, or a pharmaceutically acceptable salt, solvate, prodrug, or derivative thereof, and optionally (b) insulin, an immunologically active insulin fragment, an insulin derivative, or a functional equivalent of insulin, or a combination thereof can be co-administered or combined with drugs commonly used to treat depression.
  • SSRIs selective serotonin reuptake inhibitors
  • citalopram Celexa®, Cipramil®
  • escitalopram Lexapro®, Cipralex®
  • paroxetine Paxil®, Seroxat®
  • fluoxetine Prozac®
  • fluvoxamine Livox®, Faverin®
  • sertraline Zoloft®, Lustral®
  • indalpine Upstene®
  • zimelidine Normalud®, Zelmid®
  • serotonin-norepinephrine reuptake inhibitors such as desvenlafaxine (Pristiq®), duloxetine (Cymbalta®), levomilnacipran (Fetzima®), milnacipran (Ixel®, Savella®), venlafaxine (Effexor®)
  • serotonin modulators and stimulators SMSs
  • vilazodone Viib
  • Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially.
  • Administration “in combination” further includes the separate administration of one of the compounds or agents administered first, followed by the second.
  • the regimen selected can be administered concurrently since activation of the aminosterol induced response does not require the systemic absorption of the aminosterol into the bloodstream and thus eliminates concern over the likelihood systemic of drug-drug interactions between the aminosterol and the administered drug.
  • the phrase “therapeutically effective amount” means a dose of aminosterol, or a salt or derivative thereof that provides the specific pharmacological effect for which the compound or compounds are being administered. It is emphasized that a therapeutically effective amount will not always be effective in achieving the intended effect in a given subject, even though such dose is deemed to be a therapeutically effective amount by those of skill in the art. For convenience only, exemplary dosages are provided herein. Those skilled in the art can adjust such amounts in accordance with standard practices as needed to treat a specific subject. The therapeutically effective amount may vary based on the route of administration and dosage form, the age and weight of the subject, and/or the severity of the subject’s condition. For example one of skill in the art would understand that the therapeutically effective amount for treating a small individual may be different from the therapeutically effective amount for treating a large individual.
  • administering includes prescribing for administration, as well as actually administering, and includes physically administering by the subject being treated or by another.
  • subject or “patient” or “individual” refers to any subject, patient, or individual and the terms are used interchangeably herein.
  • the terms “subject,” “patient,” and “individual” includes mammals, and, in particular humans.
  • compositions and methods are intended to mean that the compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the elements as defined herein would not exclude other materials or steps that do not materially affect the basic and novel characteristic(s) of the claimed invention.
  • Consisting of shall mean excluding more than trace elements of other ingredients and substantial method steps. Embodiments defined by each of these transition terms are within the scope of this invention. When an embodiment is defined by one of these terms (e.g., “comprising”), it should be understood that this disclosure also includes alternative embodiments, such as “consisting essentially of’ and “consisting of’ for said embodiment.
  • treatment means any treatment of a disease or condition or associated disorder, in a patient, including:
  • Inhibiting or preventing the disease or condition that is, arresting or suppressing the development of clinical symptoms, such as neurological deficits resulting from cerebral ischemia, also included within “treatment” is provision of neuroprotection; and/or relieving the disease or condition that is, causing the regression of clinical symptoms (e.g., increasing neurological performance or reducing neurological deficits).
  • treatment encompasses “providing neuroprotection” to the subject.
  • Treatment and “providing neuroprotection” may comprise the administration of the therapeutics agent(s) or compositions disclosed herein.
  • “Pharmaceutically acceptable salt” refers to salts of a compound, which salts are suitable for pharmaceutical use and are derived from a variety of organic and inorganic counter ions well known in the art.
  • Pharmaceutically acceptable salts include, when the compound contains an acidic functionality, by way of example only, sodium, potassium, calcium, magnesium, ammonium, and tetraalkylammonium.
  • salts of organic or inorganic acids such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, and oxalate.
  • Reductive amination refers to a synthetic procedure whereby a ketone or aldehyde is reacts with an amine to form in imine or iminium which is subsequently reduced to form an amine.
  • Methods and conditions to affect reductive amination are known to those or ordinary skill in the art. See, for example, Dangerfield et al., J. Org. Chem., 2010, 75, 5470- 5477; Taibakhsh et al., Synthesis, 2011, 490-496; and Abdel-Magid et al., J. Org.
  • PS 19 mice are engineered to produce large amounts of tau-protein and develop a very aggressive form of neurodegeneration/ Alzheimer’s disease. Average lifespan in these animals is 10-11 months.
  • PS-19 animals were intranasally treated with ENT-03, which is an aminosterol comprising a spermine moiety, starting at 10 months (in late stages of disease) and dosed biweekly.
  • ENT-03 which is an aminosterol comprising a spermine moiety
  • the animals improved dramatically, becoming much faster and more agile, venturing further in exploratory behavior, indistinguishable from a young animal and from wild-type controls.
  • Neuroinflammation in the mice was eliminated. See Fig. 34.
  • improved performance was seen in a Morris water maze (MWW) test, which is a test of spatial learning for rodents. See Fig. 35.
  • the MWM has proven to be a robust and reliable test that is strongly correlated with hippocampal synaptic plasticity and NMDA receptor function.
  • 35 A shows the number of training days on the X axis vs escape latency in seconds (e.g., time it takes to find the platform) for four groups of tested animals: wild type (WT) vehicle, WT ENT-03, PS19 vehicle, and PS19 ENT-03. Escape latency significantly declined for the PS19 ENT-03 group as compared to the PS 19 untreated group.
  • Fig. 35B shows platform area (Y axis) vs each of the four groups of animals: WT vehicle, WT ENT-03, PS19 vehicle, and PS19 ENT-03.
  • Figure 36 shows localization to the arcuate nucleus, proximity to NPY producing cells, and action via pstat3 stimulation in the arcuate and subventricular zone (which is the neurogenic zone).
  • the four groups of tested animals are shown on the X axis, e.g., WT vehicle, WT ENT-03, PS19 vehicle, and PS19 ENT-03.
  • Measurement of P-STAT3, NPY, P- STAT3/NPY, and P-STAT3/NPY/DAPI is shown on the X axis.
  • P-STAT3, or phosphorylated STAT3 is the activated form of STAT3.
  • STAT3 is a transcription factor involved in many cellular functions.
  • Neuropeptide Y is one the most potent orexigenic peptides found in the brain. It stimulates food intake with a preferential effect on carbohydrate intake. It decreases latency to eat, increases motivation to eat and delays satiety by augmenting meal size.
  • PS- 19 mice and normal WT controls were subjected to genomic analysis to confirm that they were in fact the PS-19 mice exhibiting the same behavior as the wild-type controls.
  • Aminosterol 1436 was administered as an aqueous solution in water.
  • BDF1 mice are available from Charles River Laboratories. The animals were fed standard lab chow, offered ad libitum. The animals were weighed and body length measured every 5 days.
  • C57BL/6 males (12-16 grams) were administered either vehicle or 10 mg/kg (z. .) of aminosterol 1436 every 3 days for two doses, for a total of 20 mg/kg over a 6 day period.
  • the mice were about 3 weeks old, and there were 10 mice/arm.
  • the animals were weighed and their body length measured once weekly for a period of 40 days.
  • the results shown in Fig. 3 indicate that growth rates of the animals were slowed upon administration of the aminosterol comprising a spermine moiety (aminosterol 1436), which is consistent with the results shown in Fig. 2 and described in Example 2.
  • aminosterol 1436 comprising a spermine moiety
  • animals in the aminosterol 1436 group had a weight of 12 g.
  • the control group had a weight of 24 g, or an increase of 50%.
  • the aminosterol 1436 group had a weight of 11 g, or a decrease of 8.3%.
  • the animals additionally be treated with an insulin compound.
  • the insulin compound is expected to act synergistically with the aminosterol compound comprising a spermine moiety.
  • BDG-4 (370.54 g, 916.3 mmol, commercially available from Bridge Organics) was dissolved in di chloromethane (DCM, 3,600 mL) and treated with ethylene glycol (210 mL, 3.77 mol, 4.11 eq.) along with /?-toluenesulfonic acid-hydrate (36.27 mmol, 0.04 eq.). The mixture was refluxed until the reaction was complete (TLC in 70:30 hexane: acetone showed ca 3-5% residual starting material that did not change). After about 4 h reflux, the mixture was cooled, treated with of 10% potassium carbonate (150 mL), and the layers separated.
  • DCM di chloromethane
  • ethylene glycol 210 mL, 3.77 mol, 4.11 eq.
  • /?-toluenesulfonic acid-hydrate 36.27 mmol, 0.04 eq.
  • the DCM was reextracted with 800 mL of 5% sodium chloride solution and 10 mL of 10% potassium bicarbonate solution. The two aqueous layers were back-extracted with 500 mL of DCM.
  • the combined DCM extracts were dried over magnesium sulfate, filtered, washed with DCM (2 x 100 mL), and concentrated under vacuum to afford a solid, which was slurried in acetonitrile (4.8 L) containing tri ethylamine (2 mL) at 55 °C for 1 h, then 35 °C for 1 hour, and finally at 12 °C for 2 h.
  • Lithium aluminum hydride (7.44 g, 196.05 mmol, pellets) was added to anhydrous THF (435 mL) under nitrogen and stirred overnight at 20 °C to break up the pellets.
  • the suspension was cooled to 10 °C and a solution of BDG-5 (77.57 g, 172.91 mmol) was added dropwise over 160 min.
  • the mixture was stirred 1 h further, and was quenched by adding 2 mL of ethyl acetate, followed by dropwise addition of 20% aqueous potassium hydroxide solution (7.44 mL) over 10 min. The mixture became very thick, then thinned out and became more granular with additional stirring for 15 h at 20 °C.
  • BDG-7 Preparation of BDG-7
  • BDG-6 (214.55 g, 510.1 mmol) was dissolved in DCM (3.0 L) and treated with 4- dimethylaminopyridine (250.3 g, 2.048 mol) and then benzoyl chloride (180 mL, 217.98 g, 1.551 mol) dropwise. An exotherm to 31 °C was noted after about 40 mL of benzoyl chloride was added. The temperature was held to 25 °C for the remainder of the addition which took a total of 35 min. The solution was stirred overnight at 25 °C, diluted with DCM (200 mL), and treated with 10% aqueous potassium bicarbonate (2 L) generating a small amount of carbon dioxide.
  • BDG-7 (22.13 kg, 35.2 mol) was dissolved in 1 : 1 tetrahydrofuran: methanol solution (220 L) at 25 °C and treated with a solution of 50% sodium hydroxide (3.7 L, 70.4 mol, 2.0 eq.) in deionized water (11 L) and stirred for 4 h. An aliquot (0.1 mL) of the reaction mixture was partitioned between ethyl acetate (0.5 mL each) and 1 M potassium bicarbonate solution. The organic layer was analyzed by TLC (70:30 hexane: acetone) and the reaction was judged complete.
  • An aqueous solution of potassium bisulfate was prepared by mixing deionized water (23 L), 96% sulfuric acid (3.7 L, 66.6 moles), and 45% potassium hydroxide (5.7 L, 66.7 moles). The resulting solution of potassium bisulfate was added via spray ball to the reaction mixture to a final pH of 8.39 (pH meter). The mixture was vacuum-distilled to about 60 L volume; and treated with of ethyl acetate (100 L) and water (100 L). The 2-phase mixture was agitated for 10 min, and the layers separated over about 20 min. The aqueous phase was re-extracted with 37 L of ethyl acetate.
  • the total ethyl acetate layer (about 160 L) was vacuum concentrated to a volume of 80 L at 50 °C. Another 50 L of ethyl acetate was added, and the solution dried over anhydrous sodium sulfate (15 kg) by stirring overnight. The solution was filtered over Celite® 545 to remove the sodium sulfate. The filter cake and the reactor were rinsed with 2 x 37 L of ethyl acetate. The dried ethyl acetate solution was vacuum concentrated from about 225 L volume to 40 L. Acetonitrile (50 L) was added and the solution was re-distilled to remove ethyl acetate. A second and third batch of acetonitrile (50 and 100L) were added and evaporated.
  • the filter cake was dried by vacuum for several h at -20 °C, followed by letting the filter warm slowly to room temperature (about 18 °C) and then drying with warm nitrogen yielding 11.92 kg of BDG-8.
  • the mother liquors were chromatographed to isolate additional product.
  • a column of silica gel (19 kg) was packed in methylene chloride.
  • the filtrate of the product was vacuum concentrated to dryness and /i was added to the column in methylene chloride. Elution with 100 L of methylene chloride (DCM) gave the product.
  • the column was flushed with 20 L of 85: 15 methylene chloride: ethyl acetate, then 20 L of 60:40 methylene chloride: ethyl acetate, and finally back to methylene chloride (40 L).
  • the organic phase was vacuum filtered through a pad of sodium sulfate/silica gel (1 kg of each) in a sintered glass filter funnel and rinsed with hexane/di chloromethane (1/1), di chloromethane, and finally 5% methyl -t-butyl ether (MTBE) in dichloromethane.
  • the combined filtrates were evaporated in two batches in a Buchi apparatus at 40 °C. Hexane was added to each batch and evaporated again until a thick slurry formed. The solids were filtered, washed with hexane, and dried in a vacuum oven to afford 1 (4.58 kg).
  • the hydrolysis of the ketal was carried out overnight at RT, water was then added to the flask, and the majority of the acetone was removed in vacuo. The material was partitioned between ethyl acetate and water, and then the organic layer washed with brine.
  • the resulting solution was applied directly to amberchrome and eluted with a step gradient of acetonitrile in water with 0.5% TFA (10% increments 500 mL per increment) until aminosterol eluted (-60% acetonitrile). The gradient was held at this point until all of the aminosterol eluted.
  • ENT-03 was analyzed for purity via HPLC (Waters Acquity ELSD) under the following conditions: Mobile phase A: 0.1% formic acid in water; Mobile phase B: 0.1% formic acid in acetonitrile; Column: Kinetex XB-C18 (2.1 x 75 mm, 1.7pm); Gradient: 5-95%/8 min, hold 95% B/l min; 0.6 mL/min flowrate; ELSD detector; Retention time: 1.96 min and 99.9% peak area.
  • HPLC Waters Acquity ELSD
  • ENT-03 was analyzed by mass spectrometry (Waters Acquity TQD); Mobile phase A: 0.1% formic acid in water; Mobile phase B: 0.1% formic acid acetonitrile; Column: Kinetex XB-C18 (2.1 x 75 mm, 1.7 pm); Gradient: 5-95%/8 min, hold 95% B/l min; Flowrate: 0.6 ml/min; MS (ES+, M+H): Calc’d 619.55; Found: 619.31.
  • Tetrahydrofuran (THF, 10 mL) was added to 60% sodium hydride (330 mg, 8.25 mmol) in a 50 mL flask under nitrogen while cooling in an ice bath.
  • Triethyl 2- phosphonopropionate (A, 1.31 g, 5.55 mmol) was added neat by syringe over 5 min so that gas evolution was controlled. The solution was stirred another 10 min after the addition ended.
  • the substrate aldehyde 1 (2.613 g, 5.00 mmol) was dissolved in THF (4 mL) and this was added to the flask over 5 minutes, keeping the reaction temperature at 3-8°C and washing in the residue with THF (0.5 mL) twice.
  • the reaction was checked by thin layer chromatography (TLC) using 4: 1 heptane:EtOAc and Hannesians stain. Conversion was complete on the first check 5 min after the addition. At 10 minutes, a saturated NH4Q (10 mL) solution was added at once with rapid stirring. The mixture was diluted with water until the solids dissolved. The aqueous phase was extracted with ethyl acetate (2 x 5 mL), and the combined organic phases were dried over Na2SC>4 and concentrated to afford a gel (2.77 g). The TLC before and after workup shows an increase in the polar impurity spots. The crude was analyzed by LC-MS and ’H-NMR.
  • the E:Z ratio of the product was 5.0: 1.0 by the NMR shifts at 6.68 and 5.84 ppm.
  • the crude was purified by MPLC using a 25 g Biotage Snap column. The crude was loaded onto the column with DCM and the product was eluted with 0 to 35% EtOAc in hexanes over 12 column volumes. The top spot on the TLC was collected. The Rf of this spot was 0.65 in 4: 1 4: 1 heptane: EtOAc. The more polar spots present in the crude were removed by this purification to yield Compound 2 (1.99 g, 66%, E:Z ratio remained 5: 1 after column) as a white foam.
  • the aqueous layer was extracted with ethyl acetate (3 x 20 mL) and the combined organic phase was dried with Na2SC>4 and concentrated under vacuum to afford an amorphous white solid (1.488 g).
  • the crude material was purified by Biotage MPLC with a 50 g Snap Ultra column. The product was loaded with a minimal amount of DCM and eluted with a gradient of 0 to 50% EtOAc in hexanes using 20 column volumes. Peaks were collected with a 254 nm detection threshold of 20 mAU. Partial separation of (E) and (Z)-olefins was seen in the fractions. The earlier fractions were combined and analyzed by 1H-NMR.
  • the reactors were sealed and purged with 200 psi of nitrogen 6 times, then with 200 psi of hydrogen 6 times.
  • the orange catalyst solution was removed from the glovebox, the septum was fitted with a nitrogen balloon, and the catalyst solution (17.13 mL, 0.137 mmol) was injected into each purged reactor using a syringe with a 10 inch needle.
  • the ports were shut, and the reactions were pressurized with hydrogen and heated to the desired temperature and pressure with 1000 RPM stirring. Hydrogen uptake was monitored to judge reaction completion.
  • a blank of 21 mL of iPrOAc was subjected to the same program in another reactor to gauge hydrogen uptake by the solvent.
  • Bruker APEX3 software suite including SHELXTL was used for diffraction experiments including data collection and integration, and for solving, refining, displaying, and publishing of structural results.
  • a Cryostream 800 PLUS low temperature device was used. Keeping a crystal in a cold nitrogen gas stream prevents possible decay and reduces thermal motion of atoms and increases scattering power leading to better quality structures.
  • a total of 804 frames were collected. The total exposure time was 1.33 hours.
  • the frames were integrated with the Bruker SAINT software package using a narrow-frame algorithm.
  • the goodness-of-fit was 1.035.
  • the largest peak in the final difference electron density synthesis was 0.348 e'/A3 and the largest hole was -0.246 e'/A 3 with an RMS deviation of 0.051 e'/A 3 .
  • the calculated density was 1.200 g/cm3 and F(000), 1048 e'. Results confirmed assignment of the C- 25 stereochemistry as the //-configuration.
  • the absolute configuration (as shown below) was determined with single crystal diffraction data collected at 223K (see Fig. 4).
  • Triethyl phosphonopropionate 500 mg 2.0 mmol was dissolved in EtOD and catalytic sodium ethoxide was added, stirred for 2 h, and the reaction mixture stripped to exchange the majority of the acidic methylene protons for deuterium.
  • the phosphonate was added to anhydrous THF (30 mL) and treated with potassium t-butoxide (225 mg 2.0 mmol) at 0 °C for 30 min under nitrogen.
  • the aldehyde 14 1.0 g 1.9 mmol was added in one portion at 0 °C in THF ( ⁇ 5 mL) with rinses. The ice bath was removed, and the reaction mixture was allowed to run overnight at rt.
  • reaction mixture became slightly cloudy relatively quickly and is likely done in 1-2 h.
  • the reaction mixture was partitioned between hexane/ethyl acetate 50/50 (-100 mL) and water, and then washed with brine.
  • the organic was dried over Na2SC>4 and the solvent removed in vacuo.
  • the relatively clean material was chromatographed on silica gel with a hexane ethyl acetate gradient to afford Compound 15 (860 mg, 1.4 mmol, 74%).
  • ENT-03-d3 was analyzed for purity via HPLC (Agilent) under the following conditions: Mobile phase A: 0.1% formic acid in water; Mobile phase B: 0.1% formic acid in acetonitrile; Column: Kinetex XB-C18 (2.1 x 75 mm, 1.7 pm); Gradient: 5-95%/8 min, hold 95% B; 0.6 ml/min flowrate; ELSD detector; Retention time: 4.93 and 95.5% peak area.
  • ENT- 03-d3 was analyzed by LC/MS (Waters Aquity HPLC-ZQ MS); Mobile phase A: 0.1% formic acid in water; Mobile phase B: acetonitrile; Column: Waters XBridge C18 (4.6 x 50 mm, 3.5 pm). Flowrate: 1.1 mL/min. MS (ES+, M+H): Calc’d: 622.57; Found: 622.60.
  • ENT-03-d4 was analyzed for purity via HPLC (Agilent) under the following conditions: Mobile phase A: 0.1% formic acid in water; Mobile phase B: 0.1% formic acid in acetonitrile; Column: Kinetex XB- C18 (2.1 x 75 mm, 1.7 pm); Gradient: 5-95%/8 min, hold 95% B; 0.6 ml/min flowrate; ELSD detector; Retention time: 4.94 and 100% peak area.
  • ENT-03-d4 was analyzed by LC/MS (Waters Aquity HPLC-ZQ MS); Mobile phase A: 0.1% formic acid in water; Mobile phase B: acetonitrile; Column: Waters XBridge C18 (4.6 x 50 mm, 3.5 pm). Flowrate: 1.1 mL/min. MS (ES+, M+H): Cak’d: 623.58; Found: 623.31.
  • Example 5 ENT-03 (Compound III) activity as an inhibitor of protein tyrosine phosphatase IB (PTP1B)
  • ENT-02 MSI-1436
  • ENT-03 Compound III
  • D-1436 were dissolved in dimethyl sulfoxide (DMSO) to a stock concentration of 10 mM.
  • DMSO dimethyl sulfoxide
  • the compounds were tested in a 10- dose IC50 mode with 3 -fold serial dilution, in singlet, starting at 100 pM.
  • the enzyme was the human truncated form (1-321), recombinantly produced in Escherichia coli. Fluorescence was measured to monitor enzyme activity. The phosphatase activities were monitored as a timecourse measurement of the increase in fluorescence signal from fluorescent substrate, and initial linear portion of slope (signal/min) was analyzed. No compounds exhibited fluorescent background that could interfere with the assay.
  • IC50 curves were generated for the three aminosterols tested (Fig. 8A) and the control compound (Fig. 8B). Curve fits were performed when the activities at the highest concentration of compounds were less than 65%.
  • ENT-02 (MSI-1436), already known to inhibit PTP1B, exhibited an IC50 of 2.89 pM; ENT-03 (Compound III) exhibited an IC50 of 1.03 pM; D-1436 exhibited an IC50 of 2.09 pM; and the control PTP1B compound exhibited an IC50 of 2.47 pM.
  • ENT-03 (Compound III) is a potent inhibitor of PTP1B, and has potential therapeutic utility known to be associated with PTP1B inhibitors.
  • Example 6 ENT-03 (Compound III) as a weight loss agent in mice
  • ENT-02 (MSI-1436) is known to induce weight loss through a mechanism that involves certain brain circuits that control appetite.
  • Trodusquemine causes weight loss and a shift to lipid oxidation when administered systemically to mice.
  • the pharmacological target appears to lie within the hypothalamus, including the arcuate nucleus, median eminence, and the paraventricular nucleus, based on localization of radioactive Trodusquemine, and cFos activation following intraventricular administration (Ahima et al., 2002).
  • ENT-03 Compound III
  • ENT-03 administration results in weight loss with kinetics similar to that seen following administration of ENT-02.
  • Administration of both compounds resulted in a decrease in weight, with a nadir at about day 16, followed a gradual recovery to the starting weight by about day 45. While the initial decrease in weight was similar for both compounds, by about 10 days the effects diverged, with ENT-02 causing a more severe decline. By 27 days both sets of treated mice had lost about 10% of their starting body weight.
  • mice 103291 The dosing schedule used to determine the effect of orally administered squalamine and ENT-02 on the GI tracts of young and old mice was as follows. Male C57B1/6 mice, aged 20 and 78 weeks, were obtained from Jackson labs. Animals were exposed to 12 hr light dark cycles and provided Teklad standard mouse diet and water ad lib. Animals were assigned to the treatment groups shown in Table 7.
  • GI tracts of the animals were sectioned into stomach, duodenumjejunum, ileum, caecum, colon, and rectum. The tissues were then sent for histology, and the transcriptomes analyzed by RNAseq. Table 8 shows the respective mRNA amounts in young and old mouse stomach.
  • stomach transcripts (p(adj ) ⁇ 0.05): 70 decreased, and 16 increased with ageing; in the case of the jejunum, over 400 transcripts decreased, 200 increased with ageing; and for the ileum, 700 transcripts decreased, while 400 increased with ageing.
  • ageing is associated with changes in the transcriptome.
  • the “ageing” genes that ENT-03 appears to complement most significantly within the stomach are listed in Table 9B.
  • the “restored” ageing genes include those involved in tissue renewal (fibroblast growth factor 2; zinc finger protein 383; forkhead box C2); neuronal differentiation (neural cell adhesion molecule 2); immunity (toll-like receptors 9 and 12; interleukin 2 receptor, beta chain), neurotransmitter synthesis and uptake (choline and serotonin transporters), and mitochondrial respiration (cytochrome c oxidase subunit 6B2).
  • genes include those involved in tissue renewal (fibroblast growth factor 2 (Fgf2), zinc finger protein 382 (Zfp382) and forkhead box C2 (Foxc2)); in neuronal differentiation (neural cell adhesion molecule 2 (Ncam2)); in immunity (toll-like receptors 9 and 12 (Tlr9, Tlr 12), interleukin 2 receptor, beta chain (I12rb) and CD300 (Cd3001d)); in neurotransmitter synthesis and uptake (choline and serotonin transporters (Slc5a7, Slc6a4)); and in mitochondrial respiration (cytochrome c oxidase subunit 6B2 (Cox6b2)).
  • RNA-sequencing data was performed using R programming language. Transcripts with less than one read count per million reads in all samples of each tissue were removed. The raw count data for the samples were then normalised using trimmed mean of M-values normalisation and transformed with voom (Law et al., 2014), resulting in log2- transformed counts per million with associated precision weights. Normalised data provide the input for statistical hypothesis testing, in which genes that are significantly different between sample groups are identified. Statistical comparisons were performed using linear modelling, as implemented in the Bioconductor package limma (Ritchie et al., 2015).
  • Fig. 15 shows a set of heatmaps investigating the overlap of differentially expressed genes between pairs of contrasts.
  • the animals additionally be treated with an insulin compound.
  • the insulin compound is expected to act synergistically with the aminosterol compound comprising a spermine moiety.
  • the objective of this example was to determine the pharmacokinetic profile of ENT-03 (Compound III) via intravenous and oral administration in male SD Rats.
  • Animal feeding control Animals were food fasted overnight prior to dosing and fed 4 hours after administration having free access to water.
  • Dose formulation processing during dosing The dose formulation was kept stirring at room temperature and was used within 2 hours.
  • PK Sample Analyses Concentrations of ENT-03 (Compound III) in the plasma and dose samples were analyzed using a LC-MS/MS method. WinNonlin (PhoenixTM, version 6.1) or other similar software was used for pharmacokinetic calculations. The following pharmacokinetic parameters were calculated, whenever possible from the plasma concentration versus time data.
  • PO administration F, T1/2, Cmax, Tmax, AUCinf, AUCiast, MRTinf, Number of Points for Regression.
  • PTP1B dependent mechanisms have been utilized for reversal of memory impairment and normalization of behavior and reduction in neuronal loss in beta amyloid and tau mouse models of Alzheimer’s disease (Rieke, Cruz et al. 2020). Other studies have shown reduction in the toxicity of beta amyloid aggregates by trodusquemine in vitro and in a C. elegans model of Alzheimer’s disease (Limbocker, Chia et al. 2019).
  • ENT-02 (MSI- 1436) reverses several conditions (in mice) that are associated with ageing, such as metabolic syndrome, Alzheimer’s disease, atherosclerosis, cancer and a reduced capacity for regenerative repair. As we have shown in this report, ENT-03 can treat Alzheimer’s disease in murine models.
  • the Morris water maze was used to test the effect of ENT-03 on spatial learning and memory deficits in 2 mouse models of familial Alzheimer’s disease, hAPP-J20 mice that express a double mutant of the human amyloid precursor protein (Mucke et al., 2000), and PS19 mice that express the P301S mutant of the human microtubule associated protein tau (Yoshiyama et al., 2007).
  • hAPP-J20 mice expressing human APP bearing the Swedish and Indiana familial mutations B6.Cg-Zbtb20Tg(PDGFB-APPSwInd)20Lms/2Mmjax, (Mucke et al., 2000)
  • PS19 mice expressing the P301S mutation of human tau protein B6;C3-Tg(Prnp- MAPT*P301S)PS19Vle/J (Yoshiyama et al., 2007)
  • B6;C3-Tg(Prnp- MAPT*P301S)PS19Vle/J Yoshiyama et al., 2007
  • Clinical grade ENT-03 (provided by Enterin, Inc.) was administered intraperitoneally (z. .) once per week for 6 weeks at a dose of 2.5 mg/kg bodyweight starting at 4.5 months of age. The behavioral experiments were conducted 10 days after the last injection. Vehicle treated controls received sterile saline (0.9% NaCl in water).
  • mice stayed on the platform for 15 seconds of each trial before being removed to their cages. After the training period, the platform was removed from the pool and the probe trial was executed within 1 minute on the following day. On the probe day, crossings of the platform area and target quadrant were counted and swimming speed was measured using Ethovision automated video tracking software (Noldus).
  • the animals additionally be treated with an insulin compound.
  • the insulin compound is expected to act synergistically with the aminosterol compound comprising a spermine moiety.
  • 7-HOCA As the most abundant bile acid in human cerebrospinal fluid (Ogundare et al., 2010; Meaney et al., 2007; Saeed et al., 2014; Saeed et al., 2014). 7-HOCA did not appear to a be a biologically active bile acid, in that it did not activate either the FXR, LXR, or RXR/NURR1 receptors, for which bile acids and oxysterols are known ligands (Ogundare et al., 2010). Bjbrkhem has proposed that the brain metabolizes 27-hydroxycholesterol that enters from the periphery into 7-HOCA to facilitate the efflux of the oxysterol back into the circulation (Meaney et al., 2007).
  • ENT-03 in subdural hematoma fluid reflects its possible role in the development of this structure.
  • a highly vascularized sac-like “organ” develops, with one side deriving from the dura(“ outer membrane”), the other from the subarachnoid (“inner membrane”) (Yamashima et al., 2000).
  • High concentrations of numerous growth factors, including VEGF accumulate within the fluid (Edlmann et al., 2017).
  • VEGF vascular endothelial growth factor
  • Patient #1 66 yo male with 3 weeks of gait instability, headache, difficulty with fine motor movements.
  • Indications for drainage were large, symptomatic SDH. Drainage procedure was right sided craniotomy for evacuation of SDH.
  • Patient #2 75 yo male who sustained fall 3 weeks prior. Presented with 2-3 days of worsening headache, gait instability, and left lower extremity weakness. CT with bilateral subacute SDH (1.7 cm on right, 1.6 cm on left). Indications for drainage were large, symptomatic SDH. Drainage procedure was bedside twist drill hole craniostomy.
  • FIG. 11A shows the presence of ENT-03 in brain extract.
  • Fig. 1 IB shows a reference sample of synthetic ENT-03.
  • 3-oxo-bile acids comprise between 18-40% of the unconjugated bile acids within amniotic fluid during the last trimester of human fetal development (Nakagawa et al. 1990) and are present as a significant percentage of the bile acids in healthy newborn urine, gradually diminishing during the first postnatal month (Wahlen et al., 1989 and Kimura et al., 1999). Since 3-oxo-bile acids are abundant during the newborn period, Applicant focused the search for the putative polyamine- bile acid molecule in neonatal mice. Brain and liver extracts were prepared from mice between day 1 through day 24 of age using a protocol designed to capture ENT-03 based on its physical properties as follows.
  • Frozen neonatal mice were obtained from Layne Laboratories, and tissues dissected in the frozen state. Tissues were dissected in the frozen state: “pinky” (Id); “large pinky” (2-5d); “small fuzzy”(6-9d); “large fuzzy”(10-14d); “hopper” (15-18d); “small frozen”(18-24d). Tissues (0.5-7 grams) were placed into 4 volumes of methanol containing 0.12N HC1 and heated at 80°C for 5 hours. The tissues were macerated, followed by a brief centrifugation. The supernatant was collected, the volume reduced under a stream of air, and extracted with 1 volume of chloroform/1 volume of methanol. The upper phase was reduced in volume and the samples further analyzed by LC/MS/MS.
  • ENT-03 (Compound III) could be detected in brain and liver of neonatal mice (Figs. 12A and 12B, respectively).
  • Approximate concentrations of ENT-03 (Compound III) measured in the brain and liver of neonatal mice over the first 3 weeks of life are presented in Fig. 13. In both brain and liver the highest concentrations appear at birth with a gradual reduction over the following 3 weeks.
  • ENT-03 Compound III
  • Kinetex® 5pm C18 100A 50 * 2.1 mm chromatographic column Phenomenex, Torrance, CA, USA.
  • the column was maintained at 25 °C, the flow rate was 0.3 mL/min, and the injection volume was 10 pL.
  • the mobile phase (MP) consisted of A: 0.1% formic acid in water and B: 0.1% formic acid in acetonitrile (ACN, v/v).
  • the mobile phase gradient was as follows: after injection, initial conditions with MPA at 80% were held for 0.3 min, decreased to 70% in 1.7 min, to 50% in 0.5 min and held constant for 0.5 min, returning to initial conditions for another 3.5 min of reequilibration time.
  • Retention time of ENT-03 was approximately 2.3 min and total run time was 7 min.
  • a turbo ion spray interface was used as the ion source, operating in positive ion mode. Acquisition was performed in multiple reaction monitoring (MRM) mode using m/z
  • ENT-03 Compound III tissue level estimation
  • calibration curves were prepared from ENT-03 chromatographic peak areas ratios to the internal standard and using linear regression with a (1/x 2 ) weighting factor that was chosen based on goodness-of-fit criteria, including coefficient of determination (r 2 ), the back-calculated concentration of individual calibrators, and minimization of the intercept value.
  • Example 11 ENT-05 and regulatory phosphatase inhibition
  • ENT-05 was synthesized and assayed against the bank of regulatory phosphatases.
  • ENT-05 which differs from ENT-03 (Compound III) with respect to the polyamine, and the presence of a hydroxyl group on Cl 2, exhibits inhibitory activity with great specificity against the proto-oncogene PTPN11 (E76K), as shown in Tables 15A and 15B below.
  • ENT-06 differs from ENT-03 in the substitution of a spermidine for a spermine. ENT- 06 exhibits the following inhibitory activity against the human regulatory phosphatases (Tables 16A and 16B):
  • Phosphatase assays used The assays used were from Recation Biology Inc.
  • Activation peptide H2N-LN(pY)IDLDLV(dPEG8)LST(pY)ASINFQK-amide (Fortanet et al., 2016)
  • Substrate DiFMUP [6,8-difluoro-7-hydroxy-4-methylcoumarin]
  • concentration in the assay For wild type: 0.35 pM Activating peptide; 100 pM DiFMUP; for mutant: No Activating peptide; 100 pM DiFMUP Assay buffer: 60 mM HEPES (pH 7.4), 1 mM EDTA, 75 mM KC1, 75mM NaCl, 0.01% Brij-35, 5 mM DTT, and 10% DMSO (final).
  • Assay buffer 25 mM HEPES (pH 7.5), 5 mM MgCh, 0.01% Brij-35, 1 mM DTT, and 1% DMSO.
  • Assay buffer 25 mM HEPES (pH 7.5), 5 mM MgCh, 0.01% Brij-35, 1 mM DTT, and 1% DMSO.
  • PPI A, and PP1B 1 mM MnCh was added to the assay buffer.
  • the concentration of DiFMUP varied with the phosphatase chosen: 2 pM for PTPN1/PTP1B-CD; 30 pM for PP1B; 10 pM for all other phosphatases.
  • the phosphatase inhibitors were PTP1B CAS 765317-72-4 (Sigma Aldrich cat# 539741); Cantharidic acid (Santa Cruz Biotech, cat# sc-201323); SHP009 (ChemieTek, cat# CT- SHP099).
  • ENT-05 To assess the activity of ENT-05 in vivo, the compound was added to 500 ml of distilled water to a concentration of 10 pg/ml. A Xenopus leavis tadpole, premetamorphic, was placed into the solution. Over the course of about 1 hour, the tadpole gradually swelled, stopped moving, and eventually died. A second animal was placed in 500ml of water, containing 10 pg/ml ENT-05, but also 50 mM NaCl. In this case, the animal survived. The animal was then transferred to spring water without either salt or ENT-05 and remained active over the following days. No similar effect could be observed with ENT-06 or ENT-03.
  • ENT-05 has identified a phosphatase that activates the NKCC2 transporter of the channel that can be inhibited by ENT-05.
  • ENT-05 clearly has a potential utility in the treatment of hypertension, similar to the drug furosemide which inhibits renal NKCC2 by a mechanism not involving inhibition of phosphatase activity as well as intracranial hypertension (raised intracranial pressure) and intraocular hypertension (glaucoma).
  • ENT-03 a polyamine-bile acid conjugate, is expected to be metabolized through the chain of reactions utilized in the synthesis of bile acids.
  • the following molecule was synthesized ENT-03D 3 (C24D 2 , C25D1)
  • Example 14 ENT-03 stimulates the transcription of genes involved in red blood cell production and immune cell function in the spleen of aged mice
  • the spleen of the mouse plays a primary immune function (Smith et al., 2019). In the adult mouse, however, the spleen can become a red blood cell forming organ in a variety of experimental settings (Morita et al., 2011), although it normally cedes this function to the bone marrow during the first few weeks of postnatal life (Wolber et al., 2002).
  • hemoglobin a and 0 proteins, aminolevulinic acid synthase are at the top of the list (hemoglobin a and 0 proteins, aminolevulinic acid synthase) along with abundant proteins involved in the structure/function of the red blood cell (the chloride-bicarbonate exchanger, band 4.2 protein, and 2,3 biphosphoglycerate mutase). These data are compatible with the hypothesis that ENT-03 is stimulating a program in the aged mouse normally operational during the first few weeks of postnatal life.
  • the immune genes induced by ENT-03 correspond to those of the innate arm. These include IL-21, secreted by T cells and NKT cells, which has broad stimulatory effects across the breadth of the innate immune system (Spolski et al., 2014); and the IL22 binding protein (IL22ra2), secreted by numerous innate immune cells, that helps curb the action of the proinflammatory cytokine IL22 following resolution of an infection (Huber et al., 2012) reducing the probability of post-inflammatory tumorigenesis (Huber et al., 2012).
  • IL-21 secreted by T cells and NKT cells
  • IL22ra2 IL22 binding protein
  • DC-SIGN DC-SIGN
  • macrophage chemokine receptor CCR2 which plays a critical role in the normal reparative response to tissue injury (Boniakowski et al., 2018)
  • Fcamr Fc alpha/mu receptor
  • the animals additionally be treated with an insulin compound.
  • the insulin compound is expected to act synergistically with the aminosterol compound comprising a spermine moiety.
  • Example 15 Treatment induced gene expression in mouse stomach tissues, comparison of ENT-03 and ENT-02 (MSI-1436)
  • the purpose of this example is to identify transcriptional changes between young and old mice and compare the effects of ENT-02 (MSI-1436) treatments on gene expression with those of ENT-03. Mice were treated with ENT-03, ENT-02 (MSI-1436) or a vehicle control. Samples of stomach tissues from the mice were analysed by RNA-sequencing on an Illumina platform.
  • the aims of this analysis were to: identify transcriptional changes between young and old mice; determine if ENT-01 or ENT-02 (MSI-1436) treatments could reverse those age- related changes; and compare the effects of ENT-02 (MSI-1436) treatments on gene expression with those of ENT-03.
  • FIG. 20 shows a scatter plot comparing significant genes in ENT-02 (MSI-1436) vs. control (young) against ENT-03 vs untreated (young).
  • Figure 26 shows a scatter plot comparing significant genes in Old vs young (control) against Old vs young (untreated).
  • Figure 29 shows a scatter plot comparing significant genes in Old vs young (ENT-02 (MSI-1436)) against Old vs young (ENT-03).
  • Fig. 22 shows Venn diagrams of significant genes in ENT-02 (MSI-1436) vs control (young) against ENT-03 vs untreated (young). Each plot considers a different interaction of sets; either ignoring direction of perturbation, considering only up-regulated genes, considering only down-regulated genes, or examining the over- lap between those genes up-regulated in one contrast and those genes down-regulated in another.
  • the symbol U denotes the universe.
  • Fig. 23 shows a scatter plot comparing significant genes in ENT-02 (MSI-1436) vs. control (old) against ENT-03 vs untreated (old).
  • Fig. 25 shows Venn diagrams of significant genes in ENT-02 (MSI-1436) vs control (old) against ENT-03 vs untreated (old).
  • Figure 28 shows Venn diagrams of significant genes in Old vs young (control) against Old vs young (untreated).
  • Figure 31 shows Venn diagrams of significant genes in Old vs young (ENT-02 (MSI-1436)) against Old vs young (ENT-03).
  • Venn diagrams of up- and down-regulated genes Each plot considers a different interaction of sets; either ignoring direction of perturbation, considering only up-regulated genes, considering only down-regulated genes, or examining the overlap between those genes up-regulated in one contrast and those genes down-regulated in another.
  • the symbol U denotes the universe.
  • This prophetic example describes an exemplary method of retarding the aging process of a subject.
  • the method comprising administering a pharmaceutical composition comprising a therapeutically effective amount of Compound III (ENT-03) or a pharmaceutically acceptable salt or derivative thereof to the subject.
  • One or more adult human subjects can be given a suitable dosage of Compound III (ENT-03) via any pharmaceutically acceptable method, such as oral, intranasal, or injectable.
  • An exemplary daily or weekly dosage can be, for example, about 1 to about 20 mg administered intranasally daily.
  • the characteristics of aging impacted by administration of Compound III (ENT-03) or a derivative or salt thereof that can be measured include muscle endurance, coordination, social behavior and cognitive ability.
  • muscle endurance is measured for each subject prior to initial Compound III (ENT-03) dosing to establish a baseline.
  • the partial curl-up test can be used to measure endurance of the abdominal muscles and the push-up test can be used to assess endurance of the upper body.
  • the muscle endurance tests are repeated periodically to measure improvement. It is anticipated that muscle endurance will improve following Compound III (ENT-03) dosing by about 5% or more.
  • Coordination can also be evaluated for each subject prior to initial Compound III (ENT- 03) dosing to establish a baseline by testing the patient’s ability to perform rapidly alternating and point-to-point movements correctly. Following initiation of Compound III (ENT-03) dosing, the coordination tests are repeated periodically to measure improvement. It is anticipated that coordination will improve following Compound III (ENT-03) dosing by about 5% or more.
  • Cognitive ability can also be evaluated for each subject prior to initial Compound III (ENT-03) dosing to establish a baseline using a conventional cognitive ability test. Following initiation of Compound III (ENT-03) dosing, the cognitive ability test is repeated periodically to measure improvement. It is anticipated that cognitive ability will improve following Compound III (ENT-03) dosing by about 5% or more.
  • the animals additionally be treated with an insulin compound.
  • the insulin compound is expected to act synergistically with the aminosterol compound comprising a spermine moiety.
  • This prophetic example describes an exemplary method of delaying growth and/or maturation of a subject, comprising administering a pharmaceutical composition comprising a therapeutically effective amount of Compound III (ENT-03) or a pharmaceutically acceptable salt or derivative thereof to the subject.
  • One or more juvenile dogs can be given a suitable dosage of Compound III (ENT-03) via any pharmaceutically acceptable method, such as oral, intranasal, or injectable.
  • An exemplary daily or weekly dosage can be, for example, about 20 to about 160 mg/m 2 /day administered daily via any pharmaceutically acceptable route.
  • the rate of growth of each dog can be measured by recording each dog’s height and weight prior to treatment, and then periodically after initiation of treatment. At least one control dog, of the same sex and breed as the tested dogs, does not receive Compound III (ENT-03) treatment.
  • Example 3 and Figure 2 Consistent with the results described in Example 3 and Figure 2, the treated dogs are expected to show slower growth in terms of height and weight as compared to the untreated dog(s). However, the end point in terms of height and weight of both the treated and untreated dogs is expected to be the same. It is expected that administration of Compound III (ENT-03) will result in slowing growth, in terms of height and/or weight, by about 5% or more.
  • the animals additionally be treated with an insulin compound.
  • the insulin compound is expected to act synergistically with the aminosterol compound comprising a spermine moiety.
  • This prophetic example describes an exemplary method of delaying and/or preventing progression and/or onset of age-related neurodegeneration in a subject, comprising administering a pharmaceutical composition comprising a therapeutically effective amount of Compound III (ENT-03) or a pharmaceutically acceptable salt or derivative thereof to the subject.
  • composition comprising Aminosterol 1436.
  • the composition can be administered via any pharmaceutically acceptable method, such as oral, injectable, or intranasally.
  • the composition is administered daily intranasally at a dosage of about 1 to about 20 mg.
  • Neurodegeneration is evaluated prior to treatment to form a baseline, using a medically recognized technique, and then periodically following initiation of treatment. At least one control subject, of the same sex and age as the tested subjects, does not receive Compound III (ENT-03) treatment.
  • the treated subjects are expected to show slowed progression and/or onset of neurodegeneration as compared to the untreated control subject. It is expected that administration of Compound III (ENT-03) will result in slowing progression and/or onset of neurodegeneration by about 5% or more.
  • the animals additionally be treated with an insulin compound.
  • the insulin compound is expected to act synergistically with the aminosterol compound comprising a spermine moiety.
  • This prophetic example describes an exemplary method of extending the potential lifespan of a subject, which can be an animal or human.
  • mice can be given a suitable dosage of Compound III (ENT-03) via any pharmaceutically acceptable method, such as oral, intranasal, or injectable.
  • the mice can be juveniles or adults.
  • An exemplary daily or weekly dosage can be, for example, about 1 to about 10 mg/kg every 3 days administered via IPeritoneal or INasal.
  • a control group of mice, of the same sex, are not treated.
  • Fahn S ER Members of the UPDRS Development Committee. UNIFIED PARKINSON’S DISEASE RATING SCALE. Florham Park, NJ: Macmillan Health Care Information (1987).
  • Frisinia et al., “The neuropathological basis for depression in Parkinson’s disease,” Parkinsonism Relat Disord., 2009 Feb; 15(2): 144-148.
  • Kim et al. “Poststroke Induction of ?-Synuclein Mediates Ischemic Brain Damage,” J. Neurosci., 36(26):7055-65 (2016).
  • Kimura, et al. “Profile of urinary bile acids in infants and children: developmental pattern of excretion of unsaturated ketonic bile acids and 7beta-hydroxylated bile acids,” Pediatr Res S, 603-609 (1999).
  • Palma et al. “Treatment of autonomic dysfunction in Parkinson disease and other synucleinopathies”. Mov. Disord. (Review), 33(3):372-90 (March 2018).
  • Palsetia et al. “The Clock Drawing Test versus Mini-mental Status Examination as a Screening Tool for Dementia: A Clinical Comparison,” Indian J. Psychol. Med., 40: 1-10 (2018).
  • Prudente de Mello et al. "Insulin and Autophagy in Neurodegeneration,” Front. Neurosci., 73:491 (2019).
  • PIPs protein tyrosine phosphatases with lipidphosphatase activities in human disease
  • Vanhooren, V., Libert, C. “The mouse as a model organism in aging research: usefulness, pitfalls and possibilities,” Ageing Res. Rev., 12'. 8-21 (2013).
  • Wimo Wimo, et al., “The worldwide economic impact of dementia 2010,” Alzheimer ’s Dement., 9: 1-11 (2013).
  • Yamashima “The inner membrane of chronic subdural hematomas: pathology and pathophysiology,” Neurosurg Clin N Am 11, 413-424 (2000).
  • Zhao et al. “A comparative study of the amount of ?-synuclein in ischemic stroke and Parkinson’s disease,” Neurol. 5cz. 3 37(5):749-54 (2016).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Endocrinology (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Molecular Biology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Steroid Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des méthodes permettant de traiter et/ou prévenir la progression et/ou l'apparition d'une neurodégénérescence à l'aide d'un nouveau composé aminostérol et d'insuline ou de dérivés de ceux-ci. L'invention concerne également des méthodes permettant de ralentir de façon réversible la croissance et/ou le vieillissement d'un patient et/ou d'allonger la durée de vie potentielle du patient, comprenant l'administration du composé aminostérol ou de dérivés ou sels correspondants. L'invention concerne également des méthodes permettant de traiter, prévenir ou retarder l'apparition de maladies ou affections liées à l'âge, comprenant l'administration d'aminostérol ou de dérivés ou sels correspondants.
PCT/US2023/011403 2022-01-25 2023-01-24 Méthodes de traitement ou de prévention d'une maladie neurodégénérative à l'aide d'une association d'aminostérols et d'un composé d'insuline WO2023146838A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263302796P 2022-01-25 2022-01-25
US63/302,796 2022-01-25

Publications (1)

Publication Number Publication Date
WO2023146838A1 true WO2023146838A1 (fr) 2023-08-03

Family

ID=87472325

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/011403 WO2023146838A1 (fr) 2022-01-25 2023-01-24 Méthodes de traitement ou de prévention d'une maladie neurodégénérative à l'aide d'une association d'aminostérols et d'un composé d'insuline

Country Status (1)

Country Link
WO (1) WO2023146838A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120323214A1 (en) * 2012-05-16 2012-12-20 Totada R Shantha Alzheimer's disease treatment with multiple therapeutic agents delivered to the olfactory region through a special delivery catheter and iontophoresis
US20160222052A1 (en) * 2013-09-10 2016-08-04 Swansea University Deuterated compounds
US20200306216A1 (en) * 2019-04-01 2020-10-01 Anis Ahmad Cure for alzheimer's disease
WO2021005147A1 (fr) * 2019-07-09 2021-01-14 Genesis Pharma Sa Combinaison
WO2021025974A1 (fr) * 2019-08-02 2021-02-11 Enterin, Inc. Dérivés de squalamine humaine, compositions associées les comprenant, et procédés d'utilisation correspondants

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120323214A1 (en) * 2012-05-16 2012-12-20 Totada R Shantha Alzheimer's disease treatment with multiple therapeutic agents delivered to the olfactory region through a special delivery catheter and iontophoresis
US20160222052A1 (en) * 2013-09-10 2016-08-04 Swansea University Deuterated compounds
US20200306216A1 (en) * 2019-04-01 2020-10-01 Anis Ahmad Cure for alzheimer's disease
WO2021005147A1 (fr) * 2019-07-09 2021-01-14 Genesis Pharma Sa Combinaison
WO2021025974A1 (fr) * 2019-08-02 2021-02-11 Enterin, Inc. Dérivés de squalamine humaine, compositions associées les comprenant, et procédés d'utilisation correspondants

Similar Documents

Publication Publication Date Title
US20230123701A1 (en) Human aminosterol ent-03 compounds, related compositions comprising the same, and methods of using the same
US20220372066A1 (en) Human squalamine derivatives (ent-06), related compositions comprising the same, and method of using the same
US20190381071A1 (en) Methods and compositions for treating and/or preventing the progression and/or onset of age-related neurodegeneration
JP2011511845A (ja) アルファ7(α7)ニコチン作動薬と抗精神病薬との組合せ物
JP2005526077A (ja) 神経防御性スピロステノール医薬組成物
WO2009008891A1 (fr) Procédés d'amélioration de la fonction cognitive en utilisant des composés non peptidiques
JP2016523918A (ja) 神経変性状態の予防および治療のためのアドレナリン受容体拮抗薬
US20230086867A1 (en) Polymorphic forms of metopimazine
WO2009120700A2 (fr) Inhibition de dcps
US20210283097A1 (en) Methods of diagnosing and treating alzheimer's disease with s-equol
WO2020261230A1 (fr) Polythérapie comprenant de l'acétyl-leucine et du miglustat
US20120053220A1 (en) Novel lipoxygenase inhibitors as neuroprotective agents
JP2016538269A (ja) 筋ジストロフィーの処置のための方法
JP2017197554A (ja) アルツハイマー病を治療する方法及び医薬品
WO2023146838A1 (fr) Méthodes de traitement ou de prévention d'une maladie neurodégénérative à l'aide d'une association d'aminostérols et d'un composé d'insuline
US20200038420A1 (en) Aminosterol compositions and methods of using the same for treating depression
WO2023146842A1 (fr) Isomères r et s d'aminostérols en c25 et procédés de fabrication et d'utilisation associés
JP2002513759A (ja) 神経発達障害の処置における4−ピペリジンメタノール誘導体の使用
US20200038415A1 (en) Aminosterol compositions and methods of using the same for treating erectile dysfunction
WO2021090172A1 (fr) Compositions et méthodes de suppression et/ou de traitement de maladies neurodégénératives et/ou d'une affection clinique associée
US20200038417A1 (en) Methods and compositions for treating cognitive impairment
US20200038413A1 (en) Methods of treating parkinson's disease using aminosterol compositions
US20210315907A1 (en) Compositions and methods for treating brain-gut disorders
WO2024064897A1 (fr) Procédés de réduction de la neurodégénérescence associée à des maladies neurodégénératives
CA3235692A1 (fr) Formes modifiees d'ambroxol a usage therapeutique

Legal Events

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

Ref document number: 23747511

Country of ref document: EP

Kind code of ref document: A1