EP1841460A2 - Formulations therapeutiques pour le traitement de maladies associees au beta-amyloide - Google Patents

Formulations therapeutiques pour le traitement de maladies associees au beta-amyloide

Info

Publication number
EP1841460A2
EP1841460A2 EP05858504A EP05858504A EP1841460A2 EP 1841460 A2 EP1841460 A2 EP 1841460A2 EP 05858504 A EP05858504 A EP 05858504A EP 05858504 A EP05858504 A EP 05858504A EP 1841460 A2 EP1841460 A2 EP 1841460A2
Authority
EP
European Patent Office
Prior art keywords
agent
foregoing
amyloid
pharmaceutical composition
disease
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP05858504A
Other languages
German (de)
English (en)
Inventor
Francine Gervais
Francesco Bellini
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bellus Health International Ltd
Original Assignee
Bellus Health International Ltd
Neurochem International Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/871,514 external-priority patent/US7414076B2/en
Priority claimed from US10/871,512 external-priority patent/US20050038000A1/en
Priority claimed from US10/871,549 external-priority patent/US20050142191A1/en
Priority claimed from US10/871,537 external-priority patent/US20050031651A1/en
Priority claimed from US10/871,365 external-priority patent/US7244764B2/en
Priority claimed from US10/871,543 external-priority patent/US7253306B2/en
Application filed by Bellus Health International Ltd, Neurochem International Ltd filed Critical Bellus Health International Ltd
Publication of EP1841460A2 publication Critical patent/EP1841460A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/27Esters, e.g. nitroglycerine, selenocyanates of carbamic or thiocarbamic acids, meprobamate, carbachol, neostigmine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • Alzheimer's disease is a devastating disease of the brain that results in progressive memory loss leading to dementia, physical disability, and death over a relatively long period of time. With the aging populations in developed countries, the number of Alzheimer's patients is reaching epidemic proportions.
  • a main constituent of these amyloid plaques is the amyloid- ⁇ peptide (A ⁇ ), a 39-43 amino-acid protein that is produced through cleavage of the ⁇ -amyloid precursor protein (APP).
  • a ⁇ amyloid- ⁇ peptide
  • APP ⁇ -amyloid precursor protein
  • a ⁇ Alzheimer's disease
  • APP amyloid precursor protein
  • ER endoplasmic reticulum
  • Golgi apparatus or the endosomal-lysosomal pathway, and most is normally secreted as a 40 (“A ⁇ l-40") or 42 (“A ⁇ l-42”) amino acid peptide (Selkoe, Annu. Rev. Cell Biol. 10, 373-403 (1994)).
  • a role for A ⁇ as a primary cause for Alzheimer's disease is supported by the presence of extracellular amyloid ⁇ peptide ("A ⁇ ") deposits in senile plaques of Alzheimer's disease (“Alzheimer's disease”), the increased production of A ⁇ in cells harboring mutant Alzheimer's disease associated genes, e.g., amyloid precursor protein, presenilin I and presenilin II; and the toxicity of extracellular soluble (oligomeric) or fibrillar A ⁇ to cells in culture. See, e.g., Gervais, Eur. Biopharm. Review, 40-42 (Autumn 2001); May,
  • DDT 6, 459-62 (2001). Although symptomatic treatments exist for Alzheimer's disease, this disease cannot be prevented or cured at this time.
  • Alzheimer's disease is characterized by diffuse and neuritic plaques, cerebral angiopathy, and neurofibrillary tangles.
  • Plaque and blood vessel amyloid is believed to be formed by the deposition of insoluble A ⁇ amyloid protein, which may be described as diffuse or fibrillary. Both soluble oligomeric A ⁇ and fibrillar A ⁇ are also believed to be neurotoxic and inflammatory.
  • Amyloid fibrils, once deposited, can become toxic to the surrounding cells. For example, the A ⁇ fibrils organized as senile plaques have been shown to be associated with dead neuronal cells and microgliosis in patients with Alzheimer's disease.
  • a ⁇ peptide When tested in vitro, A ⁇ peptide was shown to be capable of triggering an activation process of microglia (brain macrophages), which would explain the presence of microgliosis and brain inflammation found in the brain of patients with Alzheimer's disease.
  • microglia brain macrophages
  • a ⁇ peptide was shown to be capable of triggering an activation process of microglia (brain macrophages), which would explain the presence of microgliosis and brain inflammation found in the brain of patients with Alzheimer's disease.
  • the present invention provides a method of concomitant therapeutic treatment of a subject.
  • the method generally includes administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- 5 ⁇ disease such that activities of daily living otherwise impaired by said amyloid- ⁇ disease are improved or stabilized.
  • the pharmaceutical composition includes a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent prevents or treats amyloid- ⁇ related disease, and said second agent is a therapeutic drug or nutritive supplement.
  • the present invention provides a method of preventing or treating Alzheimer's disease that includes concomitantly administering to a subject in need thereof an effective amount of a first agent that is efficacious in preventing or treating Alzheimer's disease in said subject and a second agent, wherein said first agent comprises 3-amino-l- ⁇ ropanesulfonic acid or a pharmaceutically acceptable salt thereof.
  • the present invention provides a method of preventing or treating Mild Cognitive Impairment that includes concomitantly administering to a subject in need thereof an effective amount of a first agent that is efficacious in preventing or treating Mild Cognitive Impairment in said subject and a second agent, wherein said first agent comprises 3-amino-l-propanesulfonic acid or a o pharmaceutically acceptable salt thereof.
  • the present invention provides a method of preventing or treating comprising concomitantly administering to a subject in need thereof an effective amount of a first agent that is efficacious in preventing or treating Mild Cognitive Impairment in said subject and a second agent, wherein said first agent 5 comprises 3-amino- 1 -propanesulfonic acid or a pharmaceutically acceptable salt thereof.
  • the present invention provides a method of preventing or treating Mild Cognitive Impairment that includes concomitantly administering to a subject in need thereof an effective amount of a first agent that is efficacious in preventing or treating Mild Cognitive Impairment in said subject and a o second agent, wherein said first agent is 3-amino-l-propanesulfonic acid.
  • the present invention provides a method of preventing or treating comprising concomitantly administering to a subject in need thereof an effective amount of a first agent that is efficacious in preventing or treating Mild Cognitive Impairment in said subject and a second agent, wherein said first agent is 5 3-amino-l-propanesulfonic acid.
  • the second agent can be a cholinesterase inhibitor, a statin, or memantine.
  • Figures 1-13 and 5-68 depict formulae and compounds of the invention
  • Figure 14 is a graph illustrating that a test drug given concomitantly with AChE inhibitors is able to maintain and even improve the cognitive function of patients.
  • Amyloidosis or "amyloid disease” refers to a pathological condition characterized by the presence of amyloid fibers.
  • Amyloid is a generic term referring to a group of diverse but specific protein deposits (intracellular or extracellular) which are seen in a number of different diseases. Though diverse in their occurrence, all amyloid o deposits have common morphologic properties, stain with specific dyes ⁇ e.g., Congo red), and have a characteristic red-green birefringent appearance in polarized light after staining. They also share common ultrastructural features and common X-ray diffraction and infrared spectra.
  • amyloid— ⁇ diseases includes those diseases, conditions, pathologies, and other abnormalities of the structure or function of the brain, 5 including components thereof, in which the causative agent is amyloid.
  • the area of the brain affected in an amyloid- ⁇ disease may be the stroma including the vasculature or the parenchyma including functional or anatomical regions, or neurons themselves.
  • a subject need not have received a definitive diagnosis of a specifically recognized amyloid- ⁇ disease.
  • o Local deposition of amyloid is common in the brain, particularly in elderly individuals.
  • the most frequent type of amyloid in the brain is composed primarily of A ⁇ peptide fibrils, resulting in dementia associated with sporadic (non-hereditary) Alzheimer's disease. In fact, the incidence of sporadic Alzheimer's disease greatly exceeds forms shown to be hereditary. Nevertheless, fibril peptides forming plaques are 5 very similar in both types .
  • APP is expressed and constitutively catabolized in most cells.
  • the dominant catabolic pathway appears to be cleavage of APP within the A ⁇ sequence by an enzyme provisionally termed ⁇ -secretase, leading to release of a soluble ectodomain fragment known as APPs ⁇ .
  • APP can also be o cleaved by enzymes known as ⁇ - and ⁇ -secretase at the N- and C-termini of the A ⁇ , respectively, followed by release of A ⁇ into the extracellular space.
  • a ⁇ 40 is the predominant form produced, 5-7% of total A ⁇ exists as A ⁇ 42 (Cappai et al, Int. J. Biochem. Cell Biol. 31. 885-89 (1999)).
  • a ⁇ peptide 5 appears to dramatically alter its biochemical/biophysical properties. Specifically, the additional two amino acids at the C-terminus of A ⁇ 42 are very hydrophobic, presumably increasing the propensity of A ⁇ 42 to aggregate. For example, Jarrett, et al. demonstrated that A ⁇ 42 aggregates very rapidly in vitro compared to A ⁇ 40, suggesting that the longer form of A ⁇ may be the important pathological proteins that are involved in the initial o seeding of the neuritic plaques in Alzheimer's disease (Jarrett, et al, Biochemistry 32, 4693-97 (1993); Jarrett, et al., Ann. NY. Acad. ScI 695, 144-48 (1993)).
  • Presenilin-1 Presenilin-1
  • Presenilin-2 Presenilin-2
  • ApoE apolipoprotein E gene
  • ApoE is a cholesterol transport protein
  • several groups have observed a correlation between the risk of developing Alzheimer's disease and circulating levels of cholesterol (Mahley, Science 240, 622-30 (1998); Saunders, et al, Neurology 43, 1467-72 (1993); Corder, et al, Science 261, 921-23 (1993); Jarvik, et al., Ann. NY. Acad. ScL 826, 128-46 (1997)).
  • cholesterol loading increases the production of A ⁇ protein (Simons, et al, Proc. Nat 'I Acad. Sd.
  • Amyloid- ⁇ peptide is a 39-43 amino acid peptide derived by proteolysis 5 from a large protein known as Beta Amyloid Precursor Protein (" ⁇ APP"). Mutations in ⁇ APP result in familial forms of Alzheimer's disease, Down's syndrome, cerebral amyloid angiopathy, and senile dementia, characterized by cerebral deposition of plaques composed of A ⁇ fibrils and other components, which are described in further detail below.
  • APP Amyloid Precursor Protein
  • Alzheimer's disease 717 is proximate to the site of gamma-secretase cleavage of APP in its processing to A ⁇ , and positions 670/671 are proximate to the site of ⁇ -secretase cleavage. Mutations at any of these residues may result in Alzheimer's disease, presumably by causing an increase in the amount of the 42/43 amino acid form of A ⁇ generated from APP.
  • the 5 familial form of Alzheimer's disease represents only 10% of the subject population. Most occurrences of Alzheimer's disease are sporadic cases where APP and A ⁇ do not possess any mutation.
  • a ⁇ peptides of various lengths are well known in the art. Such peptides can be made according to methods known in the art, or extracted o from the brain according to known methods ⁇ e.g., Glenner and Wong, Biochem.
  • ⁇ amyloid As used herein, the terms “ ⁇ amyloid,” “amyloid- ⁇ ,” and the like refer to 5 amyloid ⁇ proteins or peptides, amyloid ⁇ precursor proteins or peptides, intermediates, and modifications and fragments thereof, unless otherwise specifically indicated, hi particular, “A ⁇ ” refers to any peptide produced by proteolytic processing of the APP gene product, especially peptides which are associated with amyloid pathologies, including A ⁇ l-39, A ⁇ l-40, A ⁇ l-41, A ⁇ l-42, and A ⁇ l-43.
  • a ⁇ l-42 maybe referred to herein as “A ⁇ (l-42)” or simply as “A ⁇ 42” 5 or “A ⁇ 42 “ (and likewise for any other amyloid peptides discussed herein).
  • ⁇ amyloid As used herein, the terms “ ⁇ amyloid,” “amyloid- ⁇ ,” and “A ⁇ ” are synonymous.
  • the term “amyloid” refers to amyloidogenic proteins, peptides, or fragments thereof which can be soluble (e.g., monomeric or oligomeric) or insoluble (e.g., having fibrillary structure or in amyloid plaque). See, e.g., MP Lambert, et at, i o Proc. Nat 7 Acad. ScL USA 95, 6448-53 (1998).
  • amyloid- ⁇ is a peptide having 39-43 amino-acids
  • amyloid- ⁇ is an amyloidogenic peptide produced from ⁇ APP.
  • the amyloid- ⁇ diseases that are the subject of the present invention include age-related cognitive decline, early Alzheimer's disease as seen in Mild Cognitive Impairment 15 ("MCI"), vascular dementia, or Alzheimer's disease (“AD”), which may be sporadic (non-hereditary) Alzheimer's disease or familial (hereditary) Alzheimer's disease.
  • MCI Mild Cognitive Impairment 15
  • AD Alzheimer's disease
  • the amyloid- ⁇ disease may also be cerebral amyloid angiopathy ("CAA”) or hereditary cerebral hemorrhage.
  • the amyloid- ⁇ disease may be senile dementia, Down's syndrome, inclusion body myositis (“IBM”), or age-related macular degeneration
  • the present invention relates to the use of certain compounds, denoted a "first agent,” representative examples of which include substituted and unsubstituted alkanesulfonic acids, in combination with a second agent that is biologically active for the treatment or prevention of amyloid- ⁇ diseases, including Alzheimer's disease and 25 cerebral amyloid angiopathy.
  • first agent representative examples of which include substituted and unsubstituted alkanesulfonic acids
  • a second agent that is biologically active for the treatment or prevention of amyloid- ⁇ diseases, including Alzheimer's disease and 25 cerebral amyloid angiopathy.
  • the invention also relates to pharmaceutical compositions for the prevention or treatment of such diseases and methods of preparing and using these compositions.
  • the invention pertains to pharmaceutical compositions and methods of use thereof for the treatment of amyloid- ⁇ diseases.
  • the pharmaceutical compositions are selected from the group consisting of:
  • the pharmaceutical composition also comprises a second agent that is an active pharmaceutical ingredient; that is, the second agent is therapeutic and its function is beyond that of an inactive ingredient, such as a pharmaceutical carrier (or vehicle), 35 preservative, diluent, or buffer.
  • the second agent may be useful in treating or preventing an amyloid- ⁇ disease or another neurological disease.
  • the first and second agents may exert their biological effects by similar or unrelated mechanisms of action; or either one or both of the first and second agents may exert their biological effects by a multiplicity of mechanisms of action.
  • a pharmaceutical composition may also comprise a third compound, or even more yet, wherein the third (and fourth, etc.) compound has 5 the same characteristics of a second agent.
  • compositions described herein may have the first and second, third, or additional agents in the same pharmaceutically acceptable carrier or in a different pharmaceutically acceptable carrier for each described embodiment. It further should be understood that the first, second, third and additional o agent may be administered simultaneously or sequentially within described embodiments. Alternatively, a first and second agent may be administered simultaneously, and a third or additional agent may be administered before or after the first two agents.
  • a first agent in combination with a 5 second agent includes co-administration of a first agent and a second agent, which for example may be dissolved or intermixed in the same pharmaceutically acceptable carrier, or administration of a first agent, followed by the second agent, or administration of the second agent, followed by the first agent.
  • the present invention therefore, relates to methods of combination therapeutic treatment and combination pharmaceutical 0 compositions.
  • concomitant as in the phrase “concomitant therapeutic treatment” includes administering an agent in the presence of a second agent.
  • a concomitant therapeutic treatment method includes methods in which the first, second, third, or additional agents are co-administered.
  • a concomitant therapeutic treatment method also 5 includes methods in which the first or additional agents are administered in the presence of a second or additional agents, wherein the second or additional agents, for example, may have been previously administered.
  • a concomitant therapeutic treatment method may be executed step-wise by different actors.
  • one actor may administer to a subject a first agent and a second actor may administer to the subject a second agent, o and the administering steps may be executed at the same time, or nearly the same time, or at distant times, so long as the first agent (and additional agents) are after administration in the presence of the second agent (and additional agents).
  • the actor and the subject may be the same entity (e.g., human).
  • the combination of agents used within the methods and pharmaceutical 5 compositions described herein may have a therapeutic additive or synergistic effect on the condition(s) or disease(s) targeted for treatment.
  • the combination of agents used within the methods or pharmaceutical compositions described herein also may reduce a detrimental effect associated with at least one of the agents when administered alone or without the other agent(s) of the particular pharmaceutical composition.
  • the toxicity of side effects of one agent may be attenuated by another agent of the 5 composition, thus allowing a higher dosage, improving patient compliance, and improving therapeutic outcome.
  • Physicians may achieve the clinical benefits of previously recognized drugs while using lower dosage levels, thus minimizing adverse side effects.
  • the additive or synergistic effects, benefits, and advantages of the compositions apply to classes of therapeutic agents, either structural or functional o classes, or to individual compounds themselves.
  • the present methods and compositions relate to the treatment of amyloid— ⁇ diseases and conditions.
  • the various diseases and conditions involve several biological processes that produce the clinically recognized disease or condition.
  • the inventors believe that targeting more than one of these 5 biological processes simultaneously by the concomitant methods described herein enhances the therapeutic benefits of the individual agents. For example, potentiating the activity of the acetylcholine secreted by the remaining cholinergic neurons by administering cholinesterase inhibitors, while at the same time preventing further neuronal loss by enhancing clearance of A ⁇ from the brain, is clearly desirable o compared to the use of only one individual treatment. Because the therapeutic targets outlined herein are independent, yet interconnected, it is desirable to act on more than one target at the same time.
  • two agents administered simultaneously and acting on different targets may act synergistically to modify or ameliorate disease progression or symptoms. Accordingly, one embodiment of the invention is concomitant therapy with 5 a pharmaceutical composition described herein.
  • the combination of the first agent of the invention with a second (therapeutic) agent produces an enhanced therapeutic profile, for example, a profile that is greater than the sum of the benefits of the treatment with each agent independently.
  • Alzheimer's disease patients often suffer from secondary conditions o such as depression, delusions and psychosis, or sleep disturbance. From the point of view of ease of manufacture, patient compliance, and ease of administration, it is advantageous to combine multiple medicines that the Alzheimer's patient self- administers into one combined medicament. Because of cognitive impairment, patient compliance among Alzheimer's disease patients is very low, and therefore the methods 5 and pharmaceutical compositions of the present invention are especially advantageously applied to the treatment of this subject population because this combination of medicines is less likely to result in forgotten doses and may produce greater compliance. Combination of the compounds of the invention ⁇ i.e., the first agents of the compositions discussed below) with other palliative medications, which may be for diseases other than Alzheimer's, is another beneficial application of the present invention.
  • the pharmaceutical compositions disclosed herein prevent or inhibit amyloid protein assembly into insoluble fibrils which, in vivo, are deposited in various organs, or it reverses or favors deposition in subjects already having deposits
  • the compound may also prevent the amyloid protein, in its soluble, oligomeric form or in its fibrillar form, from binding or adhering to a cell surface and causing cell damage or toxicity.
  • the composition may block amyloid-induced cellular toxicity or microglial activation.
  • the compound may block amyloid-induced neurotoxicity.
  • the pharmaceutical compositions of the invention maybe administered therapeutically or prophylactically to treat diseases associated with amyloid- ⁇ fibril formation, aggregation or deposition.
  • the pharmaceutical compositions of the invention may act to ameliorate the course of an amyloid- ⁇ related disease using any of the following mechanisms (this list is meant to be illustrative and not limiting): slowing the rate of amyloid- ⁇ fibril formation or deposition; lessening the degree of amyloid- ⁇ deposition; inhibiting, reducing, or preventing amyloid- ⁇ fibril formation; inhibiting neurodegeneration or cellular toxicity induced by amyloid- ⁇ ; inhibiting amyloid- ⁇ induced inflammation; or enhancing the clearance of amyloid- ⁇ from the brain.
  • the invention pertains to a pharmaceutical composition for the treatment of an amyloid- ⁇ disease comprising a first agent and a second agent in a pharmaceutically acceptable carrier, where the first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and the second agent is a therapeutic drug or nutritive supplement.
  • the invention includes a pharmaceutical composition for the treatment of an amyloid- ⁇ disease comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein the first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and the second agent is a therapeutic drug or nutritive supplement, such that cognitive function is stabilized or further deterioration in cognitive function is prevented, slowed, or stopped.
  • the invention is a pharmaceutical composition for the treatment of an amyloid- ⁇ disease comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein the first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and the second agent is a therapeutic drug or nutritive supplement, such that activities of daily living otherwise impaired by an amyloid- ⁇ disease are improved or stabilized.
  • the invention is a pharmaceutical composition for the treatment of an amyloid- ⁇ disease comprising a first agent and a second agent in a 5 pharmaceutically acceptable carrier, wherein the first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and the second agent is a therapeutic drug or nutritive supplement, such that the pharmaceutical composition inhibits an interaction between an amyloidogenic protein and a glycoprotein or proteoglycan constituent of a basement membrane to thereby prevent or inhibit amyloid o deposition.
  • compositions for the treatment of an amyloid— ⁇ disease comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein the first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and the second agent 5 is a therapeutic drug or nutritive supplement, such that the concentration of amyloid- ⁇ or tau in the CSF of the subject changes versus an untreated subject.
  • a pharmaceutical composition for the treatment of an amyloid- ⁇ disease is also within the scope of the invention in which the composition has a first agent and at least two second agents in a pharmaceutically acceptable carrier, wherein the first agent o prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and each of the second agent is a therapeutic drug or nutritive supplement.
  • compositions for the treatment of an amyloid- ⁇ disease comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein the first agent binds amyloid- ⁇ ; and the second agent is a therapeutic 5 drug or nutritive supplement; such that amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity in the subject is prevented or inhibited.
  • Another example of the invention is a pharmaceutical composition for the treatment of an amyloid- ⁇ disease comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein the first agent binds amyloid- ⁇ ; and the o second agent is a therapeutic drug or nutritive supplement; such that cognitive function is stabilized or further deterioration in cognitive function is prevented, slowed, or stopped in the subject.
  • the invention pertains to a pharmaceutical composition for the treatment of an amyloid- ⁇ disease comprising a first agent and a second agent in a 5 pharmaceutically acceptable carrier, wherein the first agent binds amyloid- ⁇ ; and the second agent is a therapeutic drag or nutritive supplement; such that activities of daily living otherwise impaired by the amyloid- ⁇ disease are improved or stabilized in the subject.
  • the invention also includes a pharmaceutical composition for the treatment of an 5 amyloid- ⁇ disease comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein the first agent binds amyloid- ⁇ ; and the second agent is a therapeutic drag or nutritive supplement; such that the pharmaceutical composition inhibits an interaction between an amyloidogenic protein and a glycoprotein or proteoglycan constituent of a basement membrane to thereby prevent or inhibit amyloid o deposition in the subject.
  • the invention may be a pharmaceutical composition for the treatment of an amyloid- ⁇ disease comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein the first agent binds amyloid- ⁇ ; and the second agent is a therapeutic drug or nutritive supplement; such that the concentration of 5 amyloid- ⁇ or tau in the CSF of the subject changes versus an untreated subject.
  • the invention is a pharmaceutical composition for the treatment of an amyloid- ⁇ disease comprising a first agent and at least two second agents in a pharmaceutically acceptable carrier, wherein the first agent binds amyloid- ⁇ ; and the second agent is a therapeutic drag or nutritive supplement; such that amyloid- ⁇ o fibril formation, neurodegeneration, or cellular toxicity in the subject is prevented or inhibited.
  • the invention also relates to methods of making pharmaceutical compositions for use in the therapeutic and prophylactic methods described herein.
  • the first agent and the second agent are supplied as a pharmaceutical product, and they may be packaged in 5 separate containers for sale or delivery to the consumer.
  • the first agent and the second agent may be dissolved in a liquid pharmaceutically acceptable carrier, or they may be provided in a solid formulation, for example, as a homogenous mixture in a capsule or pill.
  • the pharmaceutical compositions may further comprise a pharmaceutically acceptable acid, base, buffering agent, inorganic salt, solvent, or preservative.
  • the pharmaceutical compositions of the invention may also include a compound that increases the cerebral bioavailability of either the first agent or the second agent.
  • the invention also relates to the use of a first agent and a second agent in the preparation of a pharmaceutical composition for the treatment or prevention of an amyloid- ⁇ disease comprising a first agent and a second agent in a pharmaceutically 5 acceptable carrier, wherein the first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and the second agent is a therapeutic drug or nutritive supplement.
  • the term "container” includes any receptacle for holding the therapeutic formulation.
  • the container is the packaging that 5 contains the formulation.
  • the container is not the packaging that contains the formulation, i.e., the container is a receptacle, such as a box or vial that contains the packaged formulation or unpackaged formulation and the instructions for use of the formulation.
  • packaging techniques are well known in the art. It should be understood that the instructions for use of the therapeutic formulation may be o contained on the packaging containing the therapeutic formulation, and as such the instructions form an increased functional relationship to the packaged product.
  • compositions of the invention may be effective in controlling amyloid- ⁇ deposition either following their entry into the brain (following penetration of the blood brain barrier) or from the periphery.
  • a 5 compound of a pharmaceutical composition may alter the equilibrium of A ⁇ between the brain and the plasma so as to favor the exit of A ⁇ from the brain.
  • An increase in the exit of A ⁇ from the brain would result in a decrease in A ⁇ brain concentration and therefore favor a decrease in A ⁇ deposition.
  • compounds that penetrate the brain could control deposition by acting directly on brain A ⁇ e.g., by maintaining it in a non- o fibrillar form or favoring its clearance from the brain, or protecting brain cells from the detrimental effect of A ⁇ .
  • These compounds could also prevent A ⁇ in the brain from interacting with a cell surface and therefore prevent neurotoxicity or inflammation.
  • the pharmaceutical compositions of the invention contain a first agent that prevents or inhibits ⁇ -amyloid fibril formation, either in the brain or other 5 organ of interest (acting locally) or throughout the entire body (acting systemically).
  • a first agent that prevents or inhibits ⁇ -amyloid fibril formation, either in the brain or other 5 organ of interest (acting locally) or throughout the entire body (acting systemically).
  • the first agent as described herein may inhibit or reduce an interaction between amyloid- ⁇ and a cell surface constituent, for example, a glycosaminoglycan or proteoglycan constituent of a basement membrane, and that inhibiting or reducing this interaction is primarily o responsible for the observed neuroprotective effects.
  • the first agent may also prevent an amyloid- ⁇ peptide from binding or adhering to a cell surface, a process which is known to cause cell damage or toxicity.
  • the first agent may block amyloid-induced cellular toxicity or microglial activation or amyloid-induced neurotoxicity, or inhibit amyloid- ⁇ induced inflammation.
  • the first agent may also 5 reduce the rate or amount of ⁇ -amyloid aggregation, fibril formation, or deposition, or the first agent lessens the degree of amyloid- ⁇ deposition.
  • the first agent may also inhibit, reduce, or prevent amyloid- ⁇ fibril formation.
  • the first agent may enhance the clearance of amyloid- ⁇ from the brain; or the first agent may favorably alter the equilibrium of amyloid- ⁇ between the 5 brain and the plasma to decrease the amount of amyloid- ⁇ in the brain.
  • the first agent may lower the levels of amyloid ⁇ peptides, e.g., both A ⁇ 40 and A ⁇ 42 in the CSF and the plasma, or the first agent may lower the levels of amyloid ⁇ peptides, e.g., A ⁇ 40 and A ⁇ 42 in the CSF and increase it in the plasma.
  • the first agent prevents or treats amyloid- ⁇ diseases, such as for example Alzheimer's disease.
  • the first agent may reverse or favor deposition of amyloid in a subject having amyloid deposits, or the first agent may favor plaque clearance or slow deposition in a subject having amyloid deposits.
  • the first agent decreases the amyloid- ⁇ concentration in the brain of a subject versus an untreated 5 subject, and the first agent penetrates into the brain, that is, it crosses the blood-brain barrier ("BBB") where is exerts it biological effect. Therefore, the first agent may ⁇ maintain soluble amyloid in a non-fibrillar form.
  • BBB blood-brain barrier
  • the first agent may increase the rate of clearance of soluble amyloid from the brain of a subject versus an untreated subject.
  • the invention also includes a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent prevents or inhibits 5 amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and said second agent is a therapeutic drug or nutritive supplement.
  • a method of concomitant therapeutic treatment of a subject comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ o disease, said pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and said second agent is a therapeutic drug or nutritive supplement, such that cognitive function is stabilized or further deterioration in cognitive function is prevented, slowed, or stopped.
  • the invention is a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent prevents or inhibits amyloid— ⁇ fibril formation, neurodegeneration, or cellular toxicity; and said second 5 agent is a therapeutic drug or nutritive supplement, such that activities of daily living otherwise impaired by said amyloid- ⁇ disease are improved or stabilized.
  • the invention is a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid— ⁇ o disease, said pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and said second agent is a therapeutic drug or nutritive supplement, such that said pharmaceutical composition inhibits an interaction between an amyloidogenic protein and a 5 glycoprotein or proteoglycan constituent of a basement membrane to thereby prevent or inhibit amyloid deposition.
  • the invention may be a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an o amyloid- ⁇ disease, said pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent prevents or inhibits amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and said second agent is a therapeutic drug or nutritive supplement, such that the concentration of amyloid- ⁇ or tau in the CSF of said subject changes versus an untreated subject.
  • the invention also pertains to a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said pharmaceutical composition comprising a first agent and at least two second agents in a pharmaceutically acceptable carrier, wherein said first agent prevents or inhibits o amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity; and each of said second agent is a therapeutic drug or nutritive supplement.
  • the invention is a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said 5 pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent binds amyloid- ⁇ ; and said second agent is a therapeutic drug or nutritive supplement; such that amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity in said subject is prevented or inhibited.
  • the invention is a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent binds amyloid- ⁇ ; and said second agent is a therapeutic drug or nutritive supplement; such that cognitive function o is stabilized or further deterioration in cognitive function is prevented, slowed, or stopped in said subject.
  • Another method of concomitant therapeutic treatment of a subject of the invention comprises administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said 5 pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent binds amyloid- ⁇ ; and said second agent is a therapeutic drug or nutritive supplement; such that activities of daily living otherwise impaired by said amyloid- ⁇ disease are improved or stabilized in said subject.
  • the invention pertains to a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent binds amyloid- ⁇ ; and said 5 second agent is a therapeutic drug or nutritive supplement; such that said pharmaceutical composition inhibits an interaction between an amyloidogenic protein and a glycoprotein or proteoglycan constituent of a basement membrane to thereby prevent or inhibit amyloid deposition in said subject.
  • a further example of the invention is a method of concomitant therapeutic o treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said pharmaceutical composition comprising a first agent and a second agent in a pharmaceutically acceptable carrier, wherein said first agent binds amyloid- ⁇ ; and said second agent is a therapeutic drug or nutritive supplement; such that the concentration of 5 amyloid- ⁇ or tau in the CSF of said subject changes versus an untreated subject.
  • the invention is a method of concomitant therapeutic treatment of a subject, comprising administering to a subject in need thereof an effective amount of a pharmaceutical composition for treating or preventing an amyloid- ⁇ disease, said pharmaceutical composition comprising a first agent and at least two second agents in a pharmaceutically acceptable carrier, wherein said first agent binds amyloid- ⁇ ; and said second agent is a therapeutic drug or nutritive supplement; such that amyloid- ⁇ fibril formation, neurodegeneration, or cellular toxicity in said subject is prevented or inhibited.
  • combination therapy or “therapeutic combination” means the administration of two or more "first agents,” e.g., compounds represented by
  • Such administration includes coadministration of these therapeutic agents in a substantially simultaneous manner, such as in a single tablet or capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each therapeutic agent. Also, such administration includes use of each type of therapeutic agent in a sequential manner.
  • a potential advantage of the combination therapy disclosed herein may be a reduction in the required amount of an individual therapeutic compound or the overall total amount of therapeutic compounds that are effective in treating the condition.
  • the side effects of the individual compounds can be reduced as compared to a monotherapy, which can improve subject compliance.
  • therapeutic agents can be selected to provide a broader range of complimentary effects or complimentary modes of action.
  • “combination therapy” also includes simultaneous co-administration of a first agent (e.g., an alkanesulfonic acid) and a second agent; and the term also includes methods comprising the steps of administration of the first agent, followed by the second agent, or treatment and administration of the second agent, followed by administration of the first agent.
  • a first agent e.g., an alkanesulfonic acid
  • the therapeutic value of the first agent is enhanced or maximized by co-administration with a second agent.
  • the second agent may facilitate and/or enhance the activity or efficacy of the first agent by the same and/or complementary mechanisms of action.
  • the first agent may prevent or inhibit amyloid-B fibril formation and the second agent may have a complementary effect of altering the biodistribution and/or the equilibrium amount of the aggregation forms for amyloid-B.
  • the second agent may alleviate one 5 or more symptoms of the diseases addressed herein, e.g., it may enhance cognitive function or memory, or treat depression.
  • Some general examples of compounds that may be used as a second agent according to the invention include neurotransmission enhancers; psychotherapeutic drugs; acetylcholineesterase inhibitors; calcium channel blockers; biogenic amines; o benzodiazepine tranquilizers; acetylcholine synthesis, storage, or release enhancers; acetylcholine postsynaptic receptor agonists; monoamine oxidase-A or -B inhibitors; iV-methyl-D-aspartate glutamate receptor antagonists; nonsteroidal anti-inflammatory drugs; antioxidants; and serotonergic receptor antagonists.
  • Additional examples of compounds that may be used as a second agent 5 according to the invention include agents that enhance acetylcholine synthesis, storage, or release, such as phosphatidylcholine, 4-aminopyridine, bifemelane, 3,4-diaminopyridine, choline, vesamicol, secoverine, bifemelane, tetraphenylurea, and nicotinamide; postsynaptic receptor agonists, such as arecoline, oxotremorine, bethanechol, ethyl nipecotate, and levacecarnine; iV-methyl-D-aspartate glutamate o receptor antagonists, such as milacemide and memantine; specific monoamine oxidase A inhibitors, such as moclobemide; monoamine oxidase B inhibitors, such as selegiline; thiamine and sulbutiamine; D-cycloserine; anfacine;
  • the amyloid- ⁇ peptide is a peptide having 39-43 amino-acids, which is an amyloidogenic peptide produced from ⁇ APP.
  • amyloid- ⁇ disease may be Mild Cognitive Impairment; vascular dementia; o Alzheimer's disease, including sporadic (non-hereditary) Alzheimer's disease and familial (hereditary) Alzheimer's disease; cerebral amyloid angiopathy or hereditary cerebral hemorrhage; senile dementia; Down's syndrome; inclusion body myositis; or age-related macular degeneration.
  • the method is used to treat Alzheimer's disease ⁇ e.g., 5 sporadic or familial Alzheimer's disease).
  • the method can also be used prophylactically or therapeutically to treat other clinical occurrences of amyloid- ⁇ deposition, such as in Down's syndrome individuals and in subjects with cerebral amyloid angiopathy ("CAA”) or hereditary cerebral hemorrhage.
  • CAA cerebral amyloid angiopathy
  • Cerebral amyloid angiopathy refers to the specific deposition of amyloid fibrils in the walls of leptomingeal and cortical arteries, arterioles and in 5 capillaries and veins. It is commonly associated with Alzheimer's disease, Down's syndrome and normal aging, as well as with a variety of familial conditions related to stroke or dementia ⁇ see Frangione, et al, Amyloid: J. Protein Folding Disord. 8, Suppl. 1, 36-42 (2001)). CAA can occur sporadically or be hereditary. Multiple mutation sites in either A ⁇ or the APP gene have been identified and are clinically o associated with either dementia or cerebral hemorrhage.
  • CAA disorders include, but are not limited to, hereditary cerebral hemorrhage with amyloidosis of Icelandic type (HCHWA-I); the Dutch variant of HCHWA (HCHWA-D; a mutation in A ⁇ ); the Flemish mutation of A ⁇ ; the Arctic mutation of A ⁇ ; the Italian mutation of A ⁇ ; the Iowa mutation of A ⁇ ; familial British dementia; and familial Danish dementia. 5 Additionally, abnormal accumulation of APP and of amyloid- ⁇ protein in muscle fibers has been implicated in the pathology of sporadic inclusion body myositis (“IBM”) (Askanas, et al., Proc. Natl. Acad.
  • IBM sporadic inclusion body myositis
  • the compounds of the invention can be used prophylactically or therapeutically in the o treatment of disorders in which amyloid- ⁇ protein is abnormally deposited at non- neurological locations, such as treatment of IBM by delivery of the compounds to muscle fibers.
  • a ⁇ is associated with abnormal extracellular deposits, known as drusen, that accumulate along the basal surface of the retinal 5 pigmented epithelium in individuals with age-related macular degeneration (ARMD).
  • drusen abnormal extracellular deposits
  • APVMLD is a cause of irreversible vision loss in older individuals. It is believed that A ⁇ deposition could be an important component of the local inflammatory events that contribute to atrophy of the retinal pigmented epithelium, drusen biogenesis, and the pathogenesis of ARMD (Johnson, et al., Proc. Natl. Acad. ScI USA 99(18), 11830-5 o (2002)). Therefore, the invention also relates to the treatment or prevention of age- related macular degeneration.
  • the invention pertains to pharmaceutical compositions and methods of use thereof for the treatment of amyloid- ⁇ diseases.
  • the pharmaceutical compositions comprise a first agent that, e.g., prevents or inhibits amyloid- ⁇ fibril formation, 5 neurodegeneration, or cellular toxicity.
  • the pharmaceutical composition also comprises a second agent that is an active pharmaceutical ingredient; that is, the second agent is therapeutic and its function is beyond that of an inactive ingredient, such as a pharmaceutical carrier, preservative, diluent, or buffer.
  • the second agent may be useful in treating or preventing an amyloid- ⁇ disease or another neurological disease.
  • the first and second agents may exert their biological effects by similar or unrelated mechanisms 5 of action; or either one or both of the first and second agents may exert their biological effects by a multiplicity of mechanisms of action.
  • the invention pertains to pharmaceutical compositions and methods of use thereof for the treatment of amyloid diseases, amyloid- ⁇ diseases, or Alzheimer's Disease.
  • the pharmaceutical composition comprises a first o agent that modulates a first biological process and/or exerts a first biological effect, and a second agent that modulates a second biological process and/or exerts a second biological effect.
  • the first agent and the second agent may modulate different biological processes in the pathogenesis of the disease and/or exert different biological effects and/or act on different targets.
  • the first agent may be therapeutically 5 useful in the treatment of Alzheimer's disease
  • the second agent may be therapeutically useful in the treatment of CAA, dementia, ALS, or Parkinson's Disease.
  • the first agent and the second agent may have different binding affinities or specificities for peptides, proteins, or enzymes involved in the pathogenesis of the amyloid disease (e.g., Alzheimer's disease).
  • the first agent and the second o agent when simultaneously present in a subject, act synergistically to reduce, inhibit, or ameliorate the symptoms or pathogenesis of Alzheimer's disease.
  • a pharmaceutical composition may also comprise a third compound, or even more yet, wherein the third (and fourth, etc.) compound has the same characteristics of a second agent.
  • the "second agent” is selected in accord with the following therapeutic 5 principles.
  • the pathology of Alzheimer's disease includes a number of characteristic components, including but not limited to ⁇ -amyloid deposits, such as diffuse plaques and senile plaques; cytoskeletal pathology, such as hyperphosphorylated tau and paired o helical filaments; cholinergic degeneration, such as basal cholinergic neuronal loss and reduced ChAT in cortex and septum; inflammation, such as gliosis; and cognitive and behavioral dysfunction, such as cognitive loss, apathy and aggression.
  • ⁇ -amyloid deposits such as diffuse plaques and senile plaques
  • cytoskeletal pathology such as hyperphosphorylated tau and paired o helical filaments
  • cholinergic degeneration such as basal cholinergic neuronal loss and reduced ChAT in cortex and septum
  • inflammation such as gliosis
  • cognitive and behavioral dysfunction such as cognitive loss, apathy and aggression.
  • Alzheimer's disease remains confined to symptomatic interventions such as treatment with enhancers of cognitive function, e.g., acetylcholinesterase, acetyl/ butyrylcholinesterase inhibitors, or NMDA receptor antagonists.
  • enhancers of cognitive function e.g., acetylcholinesterase, acetyl/ butyrylcholinesterase inhibitors, or NMDA receptor antagonists.
  • ⁇ and gamma- secretase enzymes that generate A ⁇ from APP inhibiting the ⁇ and gamma- secretase enzymes that generate A ⁇ from APP; preventing oligomerization or fibrillogenesis of A ⁇ or enhancing its clearance from the brain, e.g. by active or passive immunization with A ⁇ , by administering anti-fibrillogenic small molecule compounds or peptides, or by metal chelation; blocking or inhibiting inflammation and neurodegeneration induced by A ⁇ ; reducing the formation of phosphorylated Tau protein in the neurofibrillary tangles; and modulating cholesterol homeostasis.
  • a range of compounds with antioxidant, neuroprotective or neurotrophic properties are contemplated for treating Alzheimer's disease; numerous approaches such as these and others are intended to be within the scope of the invention. See, e.g., J. Hardy, et al, Science 297, 353-56 (2002).
  • Pathways involved in neurodegeneration or apoptosis are also targets for therapeutic intervention.
  • the phosphorylation of poly Q ataxin by the Akt kinase is required for neurodegeneration, suggesting that the Akt kinase could be a target.
  • Tau is found in neurofibrillary tangles hyperphosphorylated and inhibition of the kinases involved in its phosphorylation, e.g., GSK-3, is also a target. See, e.g., WO 96/35,126.
  • Many strategies for targeting amyloid include, inter alia, preventing oxidative damage with anti-oxidants (e.g., melatonin, curcumin); inhibiting amyloid formation or deposition with anti-aggregation agents ⁇ e.g., peptides, metal chelators, glycosaminoglycan mimetics); altering APP metabolism ⁇ e.g., with wortmannin or secretase inhibitors); shifting the equilibrium between levels of amyloidogenic peptides in the periphery and the central nervous system ⁇ e.g., with antibodies, vaccines, gelsolin, GMl, IGF-I) and decreasing microglial activation leading to inflammation (e.g., Fc, TGF ⁇ l).
  • anti-oxidants e.g., melatonin, curcumin
  • anti-aggregation agents e.g., peptides, metal chelators, glycosaminoglycan mimetics
  • altering APP metabolism ⁇ e.g.
  • Therapeutic treatment strategies may employ anti-f ⁇ brillogenic agents.
  • a therapeutic agent may bind to A ⁇ to prevent or inhibit its fibril formation.
  • the 16-21 region of the A ⁇ peptide, KLVFFA is responsible for the ⁇ - sheet formation and the intermolecular interactions of A ⁇ during fibrillogenesis. Peptides from this region have been extensively tested for their antifibrillogenic activity (Tjernberg LO, et al, J. Biol. Chem. 272, 12601-05 (1997); Findeis, et al, Biochemistry 38, 6791-6800 (1999); Findeis, et al, Amyloid, 231-41 (Dec 2001)).
  • Agents, including non-peptidic agents, of the invention may be used as an anti-fibrillogenic agent in this way.
  • the non-amyloidogenic pathway may be regulated through phosphorylation processes. Alteration of PKC levels and activity is one of the most consistent findings in Alzheimer's disease brain tissue. In addition, altered signal transduction mechanisms, particularly PKC, are found consistently in peripheral tissues from Alzheimer's disease subjects suggesting that these changes are not secondary to neuronal loss and may be directly involved in Alzheimer's disease pathogenesis. Altered APP metabolism is a key event in the amyloid cascade hypothesis.
  • One or more kinases are principally responsible for initiating the hyperphosphorylation of tau in vivo that leads to its apparent dissociation from microtubules and aggregation into insoluble paired helical filaments. Hyperphosphorylation of tau may underlie tangle formation in Alzheimer's disease. Calpain is responsible for cleavage of p35 and treating cells with A ⁇ aggregates can trigger p35 activation and the subsequent cdk5-mediated phosphorylation of tau and perhaps other cytoplasmic substrates. D. Selkoe, Physiol. Rev. 81(2), 741-66 (2001).
  • Suitable agents for use in the invention may target any of these biological processes.
  • one drug may target more than one therapeutic approach.
  • butyryl cholinesterase inhibitors which inhibit the activity of the cholinesterase enzyme are also associated with A ⁇ (Darvesh, et al., Cell. MoI. Neurobiol. 21, 285-96 (2001)).
  • Phenserine an acetylcholinesterase inhibitor, may inhibit both the activity of the acetylcholinesterase enzyme and the processing or translation of the APP mRNA.
  • Cholesterol-lowering drugs such as statins, e.g. avorstatin, could increase processing of amyloid precursor protein by alpha-secretase, leading to the decreased production of A ⁇ peptides.
  • Nonsteroidal anti-inflammatory drugs such as ibuprofen, flurbiprofen, indomethacin, and sulindac sulphide, could selectively inhibit the production of the A ⁇ 42 peptide, in addition to inhibiting the 5 inflammation induced by A ⁇ .
  • the method relates to a method for treating or preventing an amyloid- ⁇ related o disease by administering at least two agents, each of which is a compound that exerts a therapeutic effect when so administered and is useful in treating or preventing a neurological or psychological condition or disease.
  • the first compound of the invention may be an alkanesulfonic acid that is useful for treating or preventing an amyloid- ⁇ related disease.
  • the second compound is therapeutic, i.e., its 5 function is beyond that of an inactive ingredient, such as a pharmaceutical carrier, preservative, diluent, or buffer.
  • the second compound may be useful in treating or preventing an amyloid- ⁇ related disease or another neurological disease.
  • the second compound may also be useful in diminishing specific symptoms which are characteristic of Alzheimer's disease ⁇ e.g. memory loss, anxiety, etc.
  • the first and second compounds o may exert their biological effects by similar or unrelated mechanisms of action; or either one or both of the first and second compounds may exert their biological effects by a multiplicity of mechanisms of action.
  • a third compound, or even more yet, may likewise be used in a method of the invention, wherein the third (and fourth, etc.) compound has the same characteristics of a second compound.
  • pharmaceutical compositions of the invention are formulated to be orally administered to a subject.
  • the first agent and said second agent may be simultaneously administered.
  • the first agent and the second agent may modulate different biological processes in the pathogenesis of Alzheimer's disease.
  • the first agent and the second agent may act on different targets.
  • the first o agent may be therapeutically useful in the treatment of Alzheimer's disease
  • the second agent may be therapeutically useful in the treatment of CAA.
  • the first agent and the second agent may have different binding affinities or specificities for peptides, proteins, or enzymes involved in the pathogenesis of Alzheimer's disease.
  • the first agent and the second agent when simultaneously present in a subject, act synergistically 5 to reduce, inhibit, or ameliorate the symptoms or pathogenesis of Alzheimer's disease.
  • subject includes living organisms in which amyloidosis can occur, or which are susceptible to amyloid diseases, e.g., Alzheimer's disease.
  • compositions of the present invention to a subject to be treated can be carried out using known procedures, at dosages and for periods of time effective to modulate amyloid aggregation or amyloid-induced neurotoxicity in the subject as further described herein.
  • An effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the amount of amyloid already deposited at the clinical site in the subject, the age, sex, and weight of the subject, and the ability of the therapeutic compound to modulate amyloid aggregation in the subject. Dosage regimens can be adjusted to provide the optimum therapeutic response.
  • the subject is a human.
  • the subject may be a human over 40 years old, or a human over 50 years old, or a human over 60 years old, or even a human over 70 years old.
  • the subject may be a female human, including a postmenopausal female human, who may be on hormone (estrogen) replacement therapy.
  • the subject may also be a male human.
  • a subject may be a human at risk for Alzheimer's disease, e.g. , being over the age of 40 or having a predisposition for Alzheimer's disease.
  • Alzheimer's disease predisposing factors identified or proposed in the scientific literature include, among others, a genotype predisposing a subject to Alzheimer's disease; environmental factors predisposing a subject to Alzheimer's disease; past history of infection by viral and bacterial agents predisposing a subject to Alzheimer's disease; and vascular factors predisposing a subject to Alzheimer's disease.
  • a subject may also have one or more risk factors for cardiovascular disease (e.g., atherosclerosis of the coronary arteries, angina pectoris, and myocardial infarction) or cerebrovascular disease (e.g., atherosclerosis of the intracranial or extracranial arteries, stroke, syncope, and transient ischemic attacks), such as hypercholesterolemia, hypertension, diabetes, cigarette smoking, familial or previous history of coronary artery disease, cerebrovascular disease, and cardiovascular disease.
  • cardiovascular disease e.g., atherosclerosis of the coronary arteries, angina pectoris, and myocardial infarction
  • cerebrovascular disease e.g., atherosclerosis of the intracranial or extracranial arteries, stroke, syncope, and transient ischemic attacks
  • hypercholesterolemia typically is defined as a serum total cholesterol concentration of greater than about 5.2 mmol/L (about 200 mg/dL).
  • genotypes are believed to predispose a subject to Alzheimer's disease. These include the genotypes such as presenilin-1, presenilin-2, and amyloid precursor protein (APP) missense mutations associated with familial Alzheimer's disease, and ⁇ -2-macro globulin and LRP-I genotypes, which are thought to increase the risk of acquiring sporadic (late-onset) Alzheimer's disease.
  • genotypes such as presenilin-1, presenilin-2, and amyloid precursor protein (APP) missense mutations associated with familial Alzheimer's disease
  • APP amyloid precursor protein
  • Another genetic risk factor for the development of Alzheimer's disease are variants of ApoE, the gene that encodes apolipoprotein E (particularly the apoE4 genotype), a constituent of the low-density lipoprotein particle. WJ Strittmatter, et al, Annu. Rev. Neurosci. 19, 53-77 (1996).
  • the molecular mechanisms by which the various ApoE alleles alter the likelihood of developing Alzheimer's disease are unknown, however the role of ApoE in cholesterol metabolism is consistent with the growing body of evidence linking cholesterol metabolism to Alzheimer's disease. For example, chronic use of cholesterol-lowering drugs such as statins has recently been associated with a lower incidence of Alzheimer's disease, and cholesterol-lowering drugs have been shown to reduce pathology in APP transgenic mice.
  • Alzheimer's disease also encompasses any other predisposing factors not listed above or as yet identified and includes an increased risk for Alzheimer's disease caused by head injury, medications, diet, or lifestyle.
  • the subject is in need of treatment by the methods of the invention, and is selected for treatment based on this need.
  • a subject in need of treatment is art-recognized, and includes subjects that have been identified as having a disease or disorder related to amyloid-deposition or amyloidosis, has a symptom of such a disease or disorder, or is at risk of such a disease or disorder, and would be expected, based on diagnosis, e.g., medical diagnosis, to benefit from treatment ⁇ e.g., curing, healing, preventing, alleviating, relieving, altering, remedying, ameliorating, improving, or affecting the disease or disorder, the symptom of the disease or disorder, or reducing the risk of the disease or disorder).
  • the methods of the present invention can be used for one or more of the following: to prevent, to treat Alzheimer's disease, or ameliorate symptoms of Alzheimer ' s disease, to regulate production of or levels of amyloid ⁇ (A ⁇ ) peptides or regulate the amount of ApoE isoform 4 in the bloodstream or brain of a subject.
  • the human carries one or more mutations in the genes that encode /3-amyloid precursor protein, presem ' lin-1 or presenilin-2.
  • the human carries the Apolipoprotein e4 gene.
  • the human has a family history of Alzheimer's Disease or dementia illness.
  • the human has trisomy 21 (Down's Syndrome).
  • the subject has a normal or low serum total blood cholesterol level
  • the serum total blood cholesterol level is less than about 200 mg/dL, or less than about 180, and it can range from about 150 to about 200 mg/dL.
  • the total LDL cholesterol level is less than about 100 mg/dL, or less than about 90 mg/dL and can range from about 30 to about 100 mg/dL.
  • Methods of measuring serum total blood cholesterol and total LDL cholesterol are well known to those skilled in the art and for example include those disclosed in WO 99/38498 at p.11, incorporated by reference herein. Methods of determining levels of other sterols in serum are disclosed in H. Gylling, et ah, "Serum Sterols During Stanol Ester Feeding in a Mildly Hypercholesterolemic Population", J Lipid Res. 40: 593-600 (1999).
  • the subject has an elevated serum total blood cholesterol level, hi another embodiment, the serum total cholesterol level is at least about 200 mg/dL, or at least about 220 mg/dL and can range from about 200 to about 1000 mg/dL.
  • the subject has an elevated total LDL cholesterol level, hi another embodiment, the total LDL cholesterol level is greater than about 100 mg/dL, or even greater than about 110 mg/dL and can range from about 100 to about 1000 mg/dL.
  • the human is at least about 40 years of age.
  • the human is at least about 60 years of age.
  • the human is at least about 70 years of age.
  • the human is between about 60 and 100 years of age.
  • the subject is shown to be at risk by a diagnostic brain imaging technique, for example, that measures brain activity, plaque deposition, or brain atrophy.
  • the subject exhibits no symptoms of Alzheimer's
  • the subject is a human who is at least 40 years of age and exhibits no symptoms of Alzheimer's Disease. In another embodiment, the subject is a human who is at least 40 years of age and exhibits one or more symptoms of Alzheimer's Disease.
  • the levels of amyloid ⁇ peptides in a subject's brain or blood can be reduced from levels prior to treatment from about 10 to about 100 percent, or even about 50 to about 100 percent.
  • the subject can have an elevated level of amyloid 5 A ⁇ 40 and A ⁇ 42 peptide in the blood and CSF prior to treatment, according to the present methods, of greater than about 10 pg/mL, or greater than about 20 pg/mL, or greater than about 35 pg/mL, or even greater than about 40 pg/mL.
  • the elevated level of amyloid A ⁇ 42 peptide can range from about 30 pg/mL to about 200 pg/mL, or even to about 500 pg/mL.
  • the measurable levels of amyloid ⁇ peptide in the CSF may decrease slightly from elevated levels present before onset of the disease. This effect is attributed to increased deposition, i.e., trapping of A ⁇ peptide in the brain instead of normal clearance from the brain into the CSF.
  • the subject can have an elevated level of amyloid 5 A ⁇ 4 o peptide in the blood and CSF prior to treatment, according to the present methods, of greater than about 5 pg A ⁇ 42 /mL or greater than about 50 pg A ⁇ 40 /mL, or greater than about 400 pg/mL.
  • the elevated level of amyloid A ⁇ 40 peptide can range from about 200 pg/mL to about 800 pg/mL, to even about 1000 pg/mL.
  • the subject can have an elevated level of amyloid A ⁇ 42 o peptide in the CSF prior to treatment, according to the present methods, of greater than about 5 pg/mL, or greater than about 10 pg/mL, or greater than about 200 pg/mL, or greater than about 500 pg/mL.
  • the level of amyloid ⁇ peptide can range from about 10 pg/mL to about 1,000 pg/mL, or even about 100 pg/mL to about 1,000 pg/mL.
  • the subject can have an elevated level of amyloid A ⁇ 40 peptide in the CSF prior to treatment according to the present methods of greater than about 10 pg/mL, or greater than about 50 pg/mL, or even greater than about 100 pg/mL.
  • the level of amyloid ⁇ peptide can range from about 10 pg/mL to about 1,000 pg/mL.
  • the amount of amyloid ⁇ peptide in the brain or blood of a subj ect can be evaluated by enzyme-linked immunosorbent assay ("ELISA") or quantitative immunoblotting test methods or by quantitative SELDI-TOF which are well known to those skilled in the art, such as is disclosed by Zhang, et al, J.
  • the amount of total ApoE in the bloodstream or brain of a subject can be reduced from levels prior to treatment by about 5 to about 75 percent, or, in another embodiment, by about 5 to about 50 percent.
  • the amount of total ApoE can be measured in a manner well known to one skilled in the art, for example using an ELISA test kit such as Apo-Tek ApoE test kit that is available from Organon Teknica. o
  • the methods of the invention may be applied as a therapy for a subject having
  • Alzheimer's disease or a dementia may be applied as a prophylaxis against Alzheimer's disease or dementia for subject with such a predisposition, as in a subject, e.g., with a genomic mutation in the APP gene, the ApoE gene, or a presenilin gene.
  • the subject may have (or may be predisposed to developing 5 or may be suspected of having) vascular dementia, or senile dementia, or Mild Cognitive Impairment.
  • the subject may have another amyloid- ⁇ related disease such as cerebral amyloid angiopathy, or the subject may have amyloid deposits, especially amyloid- ⁇ amyloid deposits in the subject's brain.
  • the essential features of a dementia are multiple cognitive deficits that include memory impairment and at least one of the following: aphasia, apraxia, agnosia, or a disturbance in executive functioning (the ability to think abstractly and to plan, initiate, sequence, monitor, and stop complex behavior).
  • aphasia the ability to think abstractly and to plan, initiate, sequence, monitor, and stop complex behavior.
  • agnosia the ability to think abstractly and to plan, initiate, sequence, monitor, and stop complex behavior.
  • the order of onset and relative prominence of the cognitive disturbances and associated symptoms vary with the 5 specific type of dementia, as discussed in the following.
  • Memory impairment is generally a prominent early symptom. Individuals with dementia have difficulty learning new material and may lose valuables, such as wallets and keys, or forget food cooking on the stove. In more severe dementia, individuals also forget previously learned material, including the names of loved ones. Individuals with o dementia may have difficulty with spatial tasks, such as navigating around the house or in the immediate neighborhood (where difficulties with memory are unlikely to play a role). Poor judgment and poor insight are common as well. Individuals may exhibit little or no awareness of memory loss or other cognitive abnormalities. They may make unrealistic assessments of their abilities and make plans that are not congruent with their deficits and prognosis ⁇ e.g., planning to start a new business). They may underestimate the risks involved in activities ⁇ e.g., driving).
  • the cognitive deficits In order to make a diagnosis of dementia, the cognitive deficits must be sufficiently severe to cause impairment in occupational or social functioning and must 5 represent a decline from a previous level of functioning.
  • the nature and degree of impairment are variable and often depend on the particular social setting of the individual. For example, Mild Cognitive Impairment may significantly impair an individual's ability to perform a complex job but not a less demanding one.
  • Cognitive or degenerative brain disorders are characterized clinically by o progressive loss of memory, cognition, reasoning, judgment and emotional stability that gradually leads to profound mental deterioration and ultimately death. It is generally believed that the disease begins a number of years before it manifests itself in the mild cognitive changes that are the early signs of Alzheimer's disease. "Dementia of the Alzheimer's Type” begins gradually, and is usually diagnosed after other specific causes 5 have been ruled out.
  • Diagnostic criteria for Dementia of the Alzheimer's Type include the development of multiple cognitive deficits manifested by both memory impairment (anterograde or retrograde, i.e., impaired ability to learn new information or to recall previously learned information); and one or more of the following cognitive disturbances: aphasia (language disturbance), apraxia (impaired ability to carry out o motor activities despite intact motor function), agnosia (failure to recognize or identify objects despite intact sensory function), disturbance in executive functioning ⁇ i.e., planning, organizing, sequencing, and abstracting); where these cognitive deficits each cause significant impairment in social or occupational functioning and represent a significant decline from a previous level of functioning.
  • the course is characterized by 5 gradual onset and continuing cognitive decline, and the cognitive deficits are not due to another condition that causes progressive deficits in memory and cognition ⁇ e.g., cerebrovascular disease, brain tumor, hypothyroidism, vitamin B or folic acid deficiency, niacin deficiency, hypercalcemia, neurosyphilis, HTV infection, or chemical exposure).
  • the cognitive disturbance may be accompanied by a behavioral disturbance, o such as wandering, aggression, or agitation, or a psychological disturbance, such as depression or psychosis. See “Diagnostic and Statistical Manual of Mental Disorders," 4 th Ed., Text Revision, by American Psychiatric Association (2000).
  • NINCDS-ADRDA Alzheimer's Disease and Related Disorders Association 5
  • NINCDS-ADRDA National Institute of Neurological and Communicative Disorders and Stroke- Alzheimer's Disease and the Alzheimer's Disease and Related Disorders Association 5
  • ADAS-cog Alzheimer's Disease Assessment Scale-cognitive subscale
  • Alzheimer's Disease is the prototype of a cortical degenerative disease.
  • a major component of the presenting symptoms is usually subjective complaints of memory difficulty, language impairment, dyspraxia, at which point diagnosis is primarily based on exclusion of other possible etiologies for dementia.
  • No features of the physical examination or laboratory evaluation are pathognomonic for dementia of the Alzheimer's type.
  • Some studies have apparently discriminated patients with dementia of the Alzheimer's type from patients with dementia of other etiologies and from normal controls by using techniques such as EEG, MRI, and SPECT, but these studies have been difficult to replicate consistently, and at present, brain-imaging studies are best used to exclude other identifiable causes.
  • Alzheimer's disease is characterized pathologically by generalized atrophy of the cerebral cortex and by neurofibrillary tangles, neuritic (amyloid) plaques, and granulovacuolar degeneration. Although plaques and tangles may be detected in the brains of the elderly without Alzheimer's disease, they are more numerous in patients with dementia. Controversy remains whether brains with plaques from individuals without Alzheimer's diseasewere "normal variations" or early pathological signs of incipient disease. A definitive diagnosis ultimately requires both the characteristic dementia in life and the characteristic pathology after death.
  • Brain degeneration as measured by in vivo imaging techniques such as MRI has not been found to correlate closely with the state of clinical disease.
  • the final common clinical picture is of a bedridden patient, wholly dependent on others for all basic functions, even for turning in bed. Nutrition can often be provided only by nasogastric or gastrointestinal tubes.
  • the study of the pathobiology of Alzheimer's disease has identified at least four chromosomal loci associated with familial cases; the degeneration of central neurochemical systems, especially basal forebrain structures related to acetylcholine- mediated neurotransmission; factors associated with the formation of plaques and tangles; and exogenous (e.g., infectious and toxic) processes that may contribute to the development of sporadic cases.
  • amyloid itself is a normal brain product, excessive amounts of oligomeric or fibrillar forms of A ⁇ maybe neurotoxic.
  • Alzheimer's disease advanced age and a family history of the disease are the greatest important risk factor.
  • a family history of Down's syndrome or of 5 hematological malignancies such as leukemia, myelolymphoma, or Hodgkin's disease, is also associated with an increased risk for Alzheimer's disease.
  • other risk factors identified tentatively in recent years include female sex, a past history of head trauma, and lower education.
  • Vascular dementias are highly associated with the risk factors for cerebrovascular disease. Those factors include hypertension o (especially with systolic pressures greater than 160 mmHg), cardiac disease, transient ischemic attacks, diabetes mellitus, carotid Sons, and sickle cell disease.
  • Obesity, a sedentary lifestyle, tobacco use, alcohol consumption, and elevated serum cholesterol and lipid levels may also be risk factors for cerebrovascular disease.
  • a general physical examination is a routine component of the workup for 5 Alzheimer's disease. It may reveal evidence of systemic disease causing brain dysfunction, such as an enlarged liver and hepatic encephalopathy, or it may demonstrate systemic disease related to particular CNS processes.
  • Focal neurological findings such as asymmetrical hyperreflexia or weakness, are seen more often in vascular than in degenerative diseases. Frontal release signs and primitive reflexes, o while suggesting pathology in the frontal lobe, are present in many disorders and often point to a greater extent of progression.
  • the first step in the diagnosis of Alzheimer's disease is to exclude delirium, which may be distinguished from dementia by its cardinal feature: disturbance of consciousness.
  • Level of consciousness or arousal should be determined to be stable before a diagnosis of Alzheimer's diseasecan be made with 5 confidence. It should also be distinguished from focal or specific cognitive impairments, such as those seen in aphasic or amnestic patients. Mood disorders can present with cognitive symptoms, particularly in the dementia of depression or pseudodementia. A history of a mood disorder or a current disturbance in neurovegetative function indicates the possibility of a major depressive disorder. o The course and prognosis of a dementia syndrome vary with its cause.
  • Alzheimer's disease does not necessarily equal progressive deterioration, although many of the pathobio logical processes underlying dementia are degenerative. The rate of progression may vary within families or from individual to individual. Age at onset is an important feature of Alzheimer's disease, the most common cause of dementia in the 5 United States. Onset usually occurs after age 60 years and the prevalence increases exponentially with each successive decade, although cases have been reported in patients as young as 30 years. Familial forms of dementia of the Alzheimer's type appear to have an earlier age at onset. Cerebrovascular disease, the second most common cause of Alzheimer's disease, is associated with an earlier age at onset overall.
  • the dementias can be distinguished to some extent by their course, especially earlier in the disease process.
  • Degenerative dementias are insidious in onset and gradually progressive.
  • vascular dementias may follow a stepwise pattern, in which new deficits appear abruptly and are associated with new vascular events, but the vascular dementias also often have an insidious onset and a slow but steadily progressive course.
  • the first step in the treatment of dementia is verification of the diagnosis.
  • Preventive pharmacological agents include antihypertensive, anticoagulant, or antiplatelet agents.
  • ACE angiotensin- converting enzyme
  • diuretics have not been linked to the exaggeration of cognitive impairment and are thought to lower blood pressure without affecting cerebral blood flow (cerebral blood flow is presumed to correlate with cognitive function). Surgical removal of carotid plaques may prevent subsequent vascular events in carefully selected patients.
  • neurotransmitters including acetylcholine, dopamine, norepinephrine, GABA, and serotonin, and several neuropeptides, including somatostatin and substance P, are decreased in dementia.
  • Multiple neuropharmacological strategies have been devised in the hope of replenishing the deficient neurotransmitters.
  • Replacement therapy for acetylcholine has been the most common and widely publicized strategy.
  • acetylcholine precursors e.g., choline salycilate (ArthropanTM, Purdue Pharma, L.P., Stamford, Connecticut) and lecithin from polyenylphosphatidylcholine (PhoscholTM, Nutrasal LLC, Oxford, Connecticut); cholinergic agonists, e.g., arecoline (methyl iV-methyltetrahydronicotinate, and cholinesterase inhibitors, such as described herein.
  • cholinergic agonists e.g., arecoline (methyl iV-methyltetrahydronicotinate, and cholinesterase inhibitors, such as described herein.
  • Selegiline (EldeprylTM, Somerset, Tampa, Florida), a monoamine oxidase (“MAO") type B (“MAO-B”) inhibitor, slows the progression of Parkinson's disease, presumably by limiting endogenous generation of destructive oxidative products, and the same effect may be used therapeutically in the treatment of Alzheimer's patients.
  • Similar antioxidant treatments are being used experimentally with other dementias, including Huntington's disease and vascular dementia.
  • Naloxone (Narcan) an opiate antagonist, is thought to have possible application in vascular dementia based on animal studies in which it was demonstrated to decrease the sequelae of cerebral ischemia. See, e.g., C.Stowe, et al., Ann. Pharmacother. 27, 447-48 (1993).
  • Nerve growth factor is 5 being studied as a means of promoting neural regeneration or sprouting.
  • treatment of a subject includes the application or administration of a composition of the invention to a subject, or application or administration of a composition of the invention to a cell or tissue from a subject, who has a amyloid- ⁇ related disease or condition, has a symptom of such a disease or o condition, or is at risk of (or susceptible to) such a disease or condition, with the purpose of curing, healing, alleviating, relieving, altering, remedying, ameliorating, improving, or affecting the disease or condition, the symptom of the disease or condition, or the risk of (or susceptibility to) the disease or condition.
  • treating refers to any indicia of success in the treatment or amelioration of an injury, pathology or condition, 5 including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the injury, pathology or condition more tolerable to the subject; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; improving a subject's physical or mental well-being; or, in some situations, preventing the onset of dementia.
  • the treatment or amelioration of o symptoms can be based on objective or subjective parameters; including the results of a physical examination or a psychiatric evaluation.
  • the methods of the invention successfully treat a subject's dementia by slowing the rate of or extent of cognitive decline.
  • the invention relates to a method for preventing or inhibiting amyloid 5 production in a subject.
  • a method for preventing or inhibiting amyloid 5 production in a subject comprises administering to a subject a therapeutically effective amount of a pharmaceutical composition of the invention capable of reducing the concentration of A ⁇ , such that amyloid production or accumulation is prevented or inhibited.
  • the invention relates to a method where at least the first o compound is for preventing, reducing, or inhibiting amyloid production in a subject.
  • a method comprises administering to a subject a therapeutically effective amount of a pharmaceutical composition capable of inhibiting A ⁇ accumulation, such that A ⁇ amyloidosis is prevented, reduced, or inhibited.
  • “Inhibition" of amyloid deposition includes preventing or stopping of amyloid 5 formation, e.g., fibrillogenesis, clearance of soluble A ⁇ from brain, inhibiting or slowing down of further amyloid deposition in a subject with amyloidosis, e.g., already having amyloid deposits, and reducing or reversing amyloid fibrillogenesis or deposits in a subject with ongoing amyloidosis. Inhibition of amyloid deposition is determined relative to an untreated subject, or relative to the treated subject prior to treatment, or, e.g., determined by clinically measurable improvement, e.g., or in the case of a subject with brain amyloidosis, e.g.
  • an Alzheimer's or cerebral amyloid angiopathy subject stabilization of cognitive function or prevention of a further decrease in cognitive function (i.e., preventing, slowing, or stopping disease progression), or improvement of parameters such as the concentration of A ⁇ or tau in the CSF.
  • Modulation of amyloid deposition includes both inhibition, as defined above, and enhancement of amyloid deposition or fibril formation.
  • modulating is intended, therefore, to encompass prevention or stopping of amyloid formation or accumulation, inhibition or slowing down of further amyloid aggregation in a subject with ongoing amyloidosis, e.g., already having amyloid aggregates, and reducing or reversing of amyloid aggregates in a subject with ongoing amyloidosis; and enhancing amyloid deposition, e.g., increasing the rate or amount of amyloid deposition in vivo or in vitro.
  • Amyloid-enhancing compounds may be useful in animal models of amyloidosis, for example, to make possible the development of amyloid deposits in animals in a shorter period of time or to increase amyloid deposits over a selected period of time.
  • Amyloid-enhancing compounds may be useful in screening assays for compounds which inhibit amyloidosis in vivo, for example, in animal models, cellular assays and in vitro assays for amyloidosis. Such compounds maybe used, for example, to provide faster or more sensitive assays for compounds.
  • amyloid enhancing compounds may also be administered for therapeutic purposes, e.g., to enhance the deposition of amyloid in the lumen rather than the wall of cerebral blood vessels to prevent CAA. Modulation of amyloid aggregation is determined relative to an untreated subject or relative to the treated subject prior to treatment.
  • the method is used to treat Alzheimer's disease (e.g., sporadic or familial Alzheimer's disease).
  • the method may also be used prophylactically or therapeutically to treat other clinical occurrences of amyloid- ⁇ deposition, such as in Down's syndrome individuals and in subjects with hereditary or sporadic cerebral amyloid angiopathy ("CAA"), which lead to cerebral hemorrhage (or hemorrhagic stroke).
  • CAA hereditary or sporadic cerebral amyloid angiopathy
  • the compounds of the invention can be used prophylactically or therapeutically in the treatment of disorders in which amyloid- ⁇ protein is abnormally deposited at non- neurological locations, such as treatment of IBM by delivery of the compounds to muscle fibers.
  • a ⁇ is associated with abnormal extracellular deposits, known as drusen, that accumulate along the basal surface of the retinal pigmented epithelium in individuals with age-related macular degeneration ("ARMD").
  • ARMD is a cause of irreversible vision loss in older individuals. It is believed that A ⁇ deposition could be an important component of the local inflammatory events that contribute to atrophy of the retinal pigmented epithelium, drusen biogenesis, and the pathogenesis of ARMD (Johnson, et al., Proc. Natl. Acad. Sd. USA 99(18), 11830-5 (2002)).
  • the present invention therefore relates to the use of a first agent, e.g., an alkanesulfonic acid compound, in the prevention or treatment of amyloid- ⁇ related diseases, including, inter alia, Alzheimer's disease, cerebral amyloid angiopathy, inclusion body myositis, Down's syndrome, Mild Cognitive Impairment, and macular degeneration, in combination with a second therapeutic agent.
  • a first agent e.g., an alkanesulfonic acid compound
  • the invention relates to methods employing and compositions including substituted or unsubstituted alkylsulfonic acids that are substituted or unsubstituted straight- chain alkylsulfonic acids, substituted or unsubstituted cycloalkylsulfonic acids, and substituted or unsubstituted branched- chain alkylsulfonic acids.
  • alkylsulfonic acid as used herein is to be interpreted as being synonymous with the term “alkanesulfonic acid.”
  • the subject has mild cognitive impairment (MCI), which is a condition characterized by a state of mild but measurable impairment in thinking skills, but is not necessarily associated with the presence of dementia. MCI frequently, but not necessarily, precedes Alzheimer's disease. It is a diagnosis that has most often been associated with mild memory problems, but it can also be characterized by mild impairments in other thinking skills, such as language or planning skills. However, in general, an individual with MCI will have more significant memory lapses than would be expected for someone of their age or educational background. As the condition progresses, a physician may change the diagnosis to Mild-to-Moderate Cognition Impairment, as is well understood in this art.
  • MCI mild cognitive impairment
  • the pharmaceutical compositions disclosed herein prevent or inhibit amyloid protein assembly into insoluble fibrils which, in vivo, are deposited in various organs, or it reverses or favors deposition in subjects already having deposits.
  • the agent may also prevent the amyloid protein, in its soluble, oligomeric form or in its fibrillar form, from binding or adhering to a cell surface and causing cell damage or toxicity, hi yet another embodiment, the agent may block amyloid-induced cellular toxicity or microglial activation, hi another embodiment, the 5 agent may block amyloid-induced neurotoxicity.
  • the pharmaceutical compositions of the invention may be administered therapeutically or prophylactically to treat diseases associated with amyloid- ⁇ fibril formation, aggregation or deposition.
  • the pharmaceutical compositions of the invention may act to ameliorate the course of an amyloid- ⁇ related disease using any of the o following mechanisms (this list is meant to be illustrative and not limiting): slowing the rate of amyloid- ⁇ fibril formation or deposition; lessening the degree of amyloid- ⁇ deposition; inhibiting, reducing, or preventing amyloid- ⁇ fibril formation; inhibiting neurodegeneration or cellular toxicity induced by amyloid- ⁇ ; inhibiting amyloid- ⁇ induced inflammation; or enhancing the clearance of amyloid- ⁇ from the brain ;or 5 enhancing the catabolism or degradation of amyloid- ⁇ ; or lowering the levels of amyloid- ⁇ in the CSF; or modulating the levels of amyloid- ⁇ in the plasma.
  • Another way of decreasing A ⁇ could be that these compounds act on secretases so that A
  • compositions of the invention may be effective in controlling o amyloid- ⁇ deposition either following their entry into the brain (following penetration of the blood brain barrier) or from the periphery.
  • an agent of a pharmaceutical composition may alter the equilibrium of A ⁇ between the brain and the plasma so as to favor the exit of A ⁇ from the brain.
  • An increase in the exit of A ⁇ from the brain would result in a decrease in A ⁇ brain concentration and therefore 5 favor a decrease in A ⁇ deposition.
  • agents that penetrate the brain could control deposition by acting directly on brain A ⁇ , e.g., by maintaining it in a non- fibrillar form or favoring its clearance from the brain or enhancing its degradation rate in the brain or in the peripheral organs.
  • compositions of the invention may be administered therapeutically or prophylactically to treat diseases associated with amyloid- ⁇ fibril formation, aggregation, or deposition.
  • the compositions of the invention may act to ameliorate the course of an amyloid- ⁇ related disease by a variety of mechanisms, hi one embodiment, the pharmaceutical compositions disclosed herein prevent or inhibit amyloid protein 5 assembly into insoluble fibrils which, in vivo, are deposited in various organs, or it favors plaque clearance or slows deposition in subjects already having deposits.
  • the pharmaceutical compositions may also prevent the amyloid protein, in its soluble, oligomeric form or in its fibrillar form, from binding or adhering to a cell surface and causing cell damage or toxicity.
  • the pharmaceutical compositions may block amyloid toxicity.
  • the 5 agent may act be slowing the rate of amyloid- ⁇ fibril formation or deposition.
  • the agent may lessen the degree of amyloid- ⁇ deposition.
  • Still other examples include inhibiting, reducing, or preventing amyloid- ⁇ fibril formation; inhibiting neurodegeneration or cellular toxicity induced by amyloid- ⁇ ; inhibiting amyloid- ⁇ induced inflammation; or enhancing the clearance of amyloid- ⁇ from the o brain brain or enhancing its degradation rate in the brain or in the peripheral organs.
  • At least one of the therapeutic agents of the invention maybe effective in controlling amyloid- ⁇ deposition either following their entry into the brain (following penetration of the blood brain barrier) or from the periphery.
  • an agent may alter the equilibrium of A ⁇ between the brain and the plasma so 5 as to favor the exit of A ⁇ from the brain.
  • An increase in the exit of A ⁇ from the brain would result in a decrease in A ⁇ brain concentration and therefore favor a decrease in A ⁇ deposition.
  • agents that penetrate the brain could control deposition by acting directly on brain A ⁇ , e.g., by maintaining it in a non-fibrillar form or favoring its clearance from the brain, or by favoring catabolism or acting on secretase so that A ⁇ o production is reduced.
  • the invention relates to pharmaceutical compositions comprising two agents, each of which exerts a therapeutic effect when administered to a subject in need thereof, and is useful in treating or preventing a neurological disease.
  • the first agent of a pharmaceutical composition of the invention is selected from substituted and 5 unsubstituted alkanesulfonic acids and alkanesulfuric acids that are useful for treating or preventing an amyloid- ⁇ related disease.
  • the second agent is therapeutic, i.e., its function is beyond that of an inactive ingredient, such as a pharmaceutical carrier, preservative, diluent, or buffer.
  • the second agent may be useful in treating or preventing an amyloid— ⁇ related disease or another neurological disease.
  • the first and o second agents may exert their biological effects by similar or unrelated mechanisms of action; or either one or both of the first and second agents may exert their biological effects by a multiplicity of mechanisms of action.
  • a pharmaceutical composition may also comprise a third agent, or even more yet, wherein the third (and fourth, etc.) agent has the same characteristics of a second agent. 5
  • the invention also relates to packaged pharmaceutical products containing two agents, each of which exerts a therapeutic effect when administered to a subject in need thereof, and is useful in treating or preventing a neurological disease.
  • the first agent of a pharmaceutical composition of the invention may be selected from substituted and unsubstituted alkanesulfonic acids and alkanesulfuric acids that are useful for treating or preventing an amyloid— ⁇ related disease.
  • the second agent is therapeutic, i.e., its 5 function is beyond that of an inactive ingredient, such as a pharmaceutical carrier, preservative, diluent, or buffer.
  • the second agent may be useful in treating or preventing an amyloid- ⁇ related disease or another neurological disease.
  • the agents may exert their biological effects by similar or unrelated mechanisms of action; or either one or more than one of the agents may exert their biological effects by a multiplicity of o mechanisms.
  • a pharmaceutical composition may also comprise a third agent, or even more agents yet, wherein the third (and fourth, etc.) agent has the same characteristics of a second agent.
  • the individual agents may be packaged in separate containers for sale or delivery to the consumer.
  • the agents of the invention may be supplied in a solution with an appropriate solvent or in a solvent-free form (e.g., s lyophilized). Additional components may include acids, bases, buffering agents, inorganic salts, solvents, antioxidants, preservatives, or metal chelators.
  • the additional kit components are present as pure compositions, or as aqueous or organic solutions that incorporate one or more additional kit components. Any or all of the kit components optionally further comprise buffers.
  • the present invention also includes packaged pharmaceutical products containing a first agent in combination with (e.g., intermixed with) a second agent.
  • the invention also includes a pharmaceutical product comprising a first agent packaged with instructions for using the first agent in the presence of a second agent or instructions for use of the first agent in a method of the invention.
  • the invention also includes a 5 pharmaceutical product comprising a second or additional agents packaged with instructions for using the second or additional agents in the presence of a first agent or instructions for use of the second or additional agents in a method of the invention.
  • the packaged pharmaceutical product may contain at least one of the agents and the product may be promoted for use with a second agent.
  • This invention also relates to a pharmaceutical composition for the treatment of a condition selected from mental retardation, developmental disorders, disruptive behavioral disorders, organic mental disorders (including dementia and psychoactive substance induced organic mental disorders), psychoactive substance abuse disorders, mood disorders, anxiety disorders, somatoform disorders, dissociative disorders, 5 attention deficit disorder, schizophrenia and personality disorders in a human, comprising an acetylcholine esterase inhibitor.
  • the second agent may also be a neurotransmitter-release enhancer, i.e., they possess the ability to enhance or stimulate the release of neurotransmitters such as acetylcholine, dopamine, and serotonin in humans.
  • Such conditions include Alzheimer's disease, age associated memory impairment and Mild Cognitive Impairment, and Parkinson's disease. They also include mental retardation, developmental disorders, disruptive behavioral disorders, organic mental disorders (including dementia and psychoactive substance induced organic mental disorders), psychoactive substance abuse disorders, mood disorders, anxiety disorders, somatoform disorders, dissociative disorders, attention deficit disorder, schizophrenia, and personality disorders.
  • the invention is also ideally suited for the treatment of familiar or hereditary forms of Alzheimer's disease because, for example, therapeutic prophylactic pharmaceutical treatment could be initiated earlier in a patient's life.
  • therapeutic prophylactic pharmaceutical treatment could be initiated earlier in a patient's life.
  • all therapeutic regimens that are commercially available treat only the symptoms of Alzheimer's disease, as explained elsewhere herein.
  • the present invention provides methods and compositions, however, that treat the underlying etiology of the disease itself, and therefore may be used in a prophylactic manner.
  • the biological processes that give rise to Alzheimer's disease may occur in a person for some time before clinically observable symptoms arise. Ordinarily, according to current medical science, such a period in a person's life would be undetectable and treatment with current medicines would be useless.
  • compositions of the present invention may delay the onset of symptoms.
  • the invention pertains to any novel chemical agent described herein. That is, the invention relates to novel agents, and novel methods of their use as described herein, including those coumpounds that may be within the scope of the Formulae disclosed herein, and which are not disclosed in the cited Patents and Patent Applications.
  • an alkanesulfonic acid compound may be combined with a second agent that is also useful in the treatment of Alzheimer's disease.
  • the second agent may be any therapeutic drug.
  • a "therapeutic drug” is a drug or medicine administered for legitimate or medically- approved, therapeutic or diagnostic, purpose. Therapeutic drugs may be available over-the-counter or by prescription.
  • therapeutic drugs include an adrenergic, anti-adrenergic, anti-androgen, anti-anginal, anti-anxiety, anticonvulsant, antidepressant, anti-epileptic, antihyperlipidemic, antihyperlipoproteinemic, 5 antihypertensive, anti-inflammatory, antiobessional, antiparkinsonian, antipsychotic, adrenocortical steroids, adrenocortical suppressant, aldosterone antagonists, amino acids, anabolic steroids, analeptic agents; androgens; blood glucose regulators, cardioprotectants; cardiovascular agents; cholinergic agonist and antagonists, cholinesterase deactivators or inhibitors, cognition adjuvants and enhancers, o dopaminergic agents, enzyme inhibitors, estrogen and related steroid hormones, free oxygen radical scavengers, GABA agonists, glutamate antagonists, hormones, antihypocholesterolemic agents, hypolipidemic agents,
  • the invention pertains to a pharmaceutical composition
  • a pharmaceutical composition comprising an alkanesulfonic acid and a second agent that is useful in the treatment or prevention of Alzheimer's disease.
  • the second agent may be curative, i.e., modulate the causative agents of Alzheimer's disease, or it may be palliative, i.e., alleviate the symptoms of the 5 disease, e.g. , by enhancing memory or improving cognitive function.
  • the second agent may be a drug that is useful in the treatment of Alzheimer's disease itself, or it may be used to treat a condition associated with Alzheimer's disease, e.g. a secondary condition, or it may be a drug commonly prescribed to Alzheimer's disease subjects.
  • a pharmaceutical composition of the invention may comprise a second agent that is specific for any one of the biological processes giving rise to the clinical presentation of Alzheimer's disease.
  • the invention is not to be considered bound by any particular theory of the etiology of Alzheimer's disease, and so the second agent according to the pharmaceutical compositions of the 5 invention may be any agent that either by itself or in combination with an alkanesulfonic acid is empirically observed to be efficacious in the treatment or prevention of Alzheimer's disease.
  • a summary of the biological processes believed to give rise to Alzheimer's disease itself or the symptoms thereof is useful inasmuch as any one of these biological processes may be modulated by a second agent in a pharmaceutical composition of the invention.
  • the language "in combination with" a second agent or treatment includes 5 co-administration of an alkanesulfonic acid, administration of an alkanesulfonic acid first, followed by the second, or treatment and administration of the second agent first, followed by an alkanesulfonic acid.
  • the condition associated with Alzheimer's disease may be a symptom characteristic of Alzheimer's disease, for example, hypothyroidism, cerebrovascular or o cardiovascular disease, memory loss, anxiety, or a behavioral dysfunction (e.g., apathy, aggression, or incontinence); a psychological condition or a neurological condition.
  • a symptom characteristic of Alzheimer's disease for example, hypothyroidism, cerebrovascular or o cardiovascular disease, memory loss, anxiety, or a behavioral dysfunction (e.g., apathy, aggression, or incontinence); a psychological condition or a neurological condition.
  • the neurological condition may be Huntington's disease, amyotrophic lateral sclerosis, acquired immunodeficiency, Parkinson's disease, aphasia, apraxia, agnosia, Pick disease, dementia with Lewy bodies, altered muscle tone, seizures, sensory loss, visual 5 field deficits, incoordination, gait disturbance, transient ischemic attack or stroke, transient alertness, attention deficit, frequent falls, syncope, neuroleptic sensitivity, normal pressure hydrocephalus, subdural hematoma, brain tumor, posttraumatic brain injury, or posthypoxic damage.
  • the psychological condition is depression, delusions, illusions, hallucinations, sexual disorders, weight loss, psychosis, a sleep disturbance o such as insomnia, behavioral disinhibition, poor insight, suicidal ideation, depressed mood or irritability, anhedonia, social withdrawal, or excessive guilt.
  • a sleep disturbance o such as insomnia, behavioral disinhibition, poor insight, suicidal ideation, depressed mood or irritability, anhedonia, social withdrawal, or excessive guilt.
  • the second agent i.e., a therapeutic drug may be a psychotropic medication, antidepressant (a selective serotonin reuptake inhibitor, atypical antidepressant), antipsychotic, appetite stimulants, or another drug used to treat a condition associated 5 with Alzheimer's disease, or a a nutritive supplement that is a precursor of acetylcholine (lecithin or choline), Ginkgo biloba, acetyl-L-carnitine, idebenone, propentofylline, or a xanthine derivative.
  • antidepressant a selective serotonin reuptake inhibitor, atypical antidepressant
  • antipsychotic appetite stimulants
  • another drug used to treat a condition associated 5 with Alzheimer's disease or a a nutritive supplement that is a precursor of acetylcholine (lecithin or choline), Ginkgo biloba, acetyl-L-carnitine, idebenone, prop
  • Antidepressants include selective serotonin reuptake inhibitors such as citalopram (Celexa); escitalopram (Lexapro); fluoxetine (ProzacTM); alprazolam o (XanaxTM), triptolene, fluvoxamine (LuvoxTM); paroxetine (PaxilTM); sertraline
  • Zero-oftTM mixed norepinephrine/dopamine reuptake inhibitors such as bupropion (WellbutrinTM); drugs with mixed serotonin effects such as nefazodone (SerzoneTM) and trazodone (DesyrelTM); mixed serotonin/norepinephrine reuptake inhibitors venlafaxine (EffexorTM); monoamine oxidase inhibitors including phenelzine (NardilTM) and 5 tranylcypromine (ParnateTM); and tetracyclic antidepressants such as maprotiline, mirtazapine (RemeronTM), amitriptyline (ElavilTM), amoxapine, clomipramine (AnafranilTM), desipramine (NorpraminTM), doxepin (SinequanTM), imipramine (TofranilTM), nortriptyline (AventylTM, PamelorTM), pro
  • Antidepressants tricyclic and selective serotonin reuptake inhibitors; fluoxetine (ProzacTM); sertraline (ZoloftTM); paroxetine (PaxilTM); citalopram (CelexaTM); nortriptyline; moclobemide; miratazepine; NardilTM; ParnateTM; ManerixTM; TofranilTM; ElavilTM; SinequanTM; SurmontilTM; AnafranilTM; NorpramineTM; AventylTM; EffexorTM; SerzoneTM; WelbutrinTM; DesyrelTM; and RemeronTM.
  • Antipyschotics include aripiprazole (AbilifyTM), clozapine (ClozarilTM), olanzapine (ZyprexaTM), quetiapine (SeroquelTM), risperidone (RisperdalTM), and ziprasidone (GeodonTM).
  • Antipsychotics conventional and atypical; olanzapine (ZyprexaTM); quetiapine (SeroquelTM); haloperidol (HaldolTM); risperidone (RisperidalTM); zuclopenhixol (ClopixolTM); ziprazidone; thioridazine (MellarilTM, Sandoz Pharmaceutical Corp., now Novertis, Basel, Switzerland); clozapine (ClozarilTM); olanzapine; and lithium.
  • drugs that may be a second agent include: cholinesterase inhibitors: huperzine A; antidepressants: venlafaxine, desipramine, nefazodone, trazodone, citalopram, escitalopram, nortriptyline, paroxetine; anti-agitation/mood- stabilizing agents, or anti-epileptics for convulsions: carbamazepine, gabapentin, primidone, phenytoin, clonazepam, valproic acid; neuroleptics: ziprasidone, haloperidol, risperidone, olanzapine, quetiapine; anti-inflammatory/immunomodulating drugs: colchicine, dapsone, meloxicam, nimesulide, flurbiprofen, cyclophosphamide, methotrexate, ⁇ -interferon, gamma-
  • Suitable second agents further include therapeutic drugs that are useful in the o treatment of Parkinson's Disease or its symptoms, which second agents include: (a) cabidopa and levodopa compositions; (b) dopamine agonists such as bromocriptine mesylate (ParlodelTM), pergolide mesylate (PermaxmTM), (+)-4-propyl-9- hydroxynaphthoxazine, and apomorphine; (c) anticholinergic medications such as benztropine mesylate (CogentinTM), and biperiden; (d) antihistamines such as 5 orphenadrine citrate (NorflexTM, NorgesicTM); (e) tricyclic antidepressants such as amitriptyline HCl (ElavilTM), amitriptyline HCl/perphenazine combinations (EtrafonTM), amitriptyline and chlordiazepoxide combinations (LimbitrolTM), nortriptyline HC
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of Alzheimer's Disease or its symptoms, which second agents include: (a) vasodilators or other nootropic direct brain metabolic enhancer drugs such as idebenone, propentophylline, pentoxifylline, citicoline, ebiratide, vinpocetine (CavintonTM), bromvincamine, cyclandelate, isoxsuprene, nafronyl, papaverine, suloctidil, vinburnine, vincamine, vindeburnol, naloxone, ethyl 5-isopropyloxy-4-methyl- ⁇ -carboline-3- 5 carboxylate, N'-methyl- ⁇ -carboline-3-carboxamide, methyl 6,7-dimethoxy-4-ethyl- ⁇ - carboline-3-carboxylate, ethyl 5-methoxy-4-ethyl- ⁇ -carboline-3-carboxylate, ifenprodil
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of aging or its symptoms, which second agents include: (a) monoamine oxidase B inhibitors such as selegiline; (b) acetylcholinesterase inhibitors such as physostigmine (Antilirium InjectableTM), heptylphysostigmine, tacrine (CognexTM), optionally with phosphatidylcholine co-agent; (+/-)-9-amino-l,2,3,4-tetrahydroacridin-l- ol, velnacrine maleate (MentaneTM), methanesulfonyl fluoride, phenylmethylsulfonyl fluoride, huperzine A, huperzine B, edrophonium chloride, galanthamine, and miotine; (c) angiotensin-converting enzyme inhibitors such as captopril (CapotenTM), captopril in combination with hydro
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of tinnitus (nerve deafness) or its symptoms, which second agents include: antidepressants or antianxiety medications such as amitriptyline (ElavilTM), amitriptyline/perplienazine combinations (EtrafonTM), alprazolam (XanaxTM), and triptolene; anticonvulsants such as primidone (MysolineTM), phenytoin (DilantinTM), and carbamazepine (TegQretolTM); lidocaine (XylocaineTM); tocainide; flecinide; 5 nicotinamide; aminooxyacetic acid; praxilene; aniracetam; piracetam; 13-cis-retinoic acid; and 13-trans-retinoic acid.
  • antidepressants or antianxiety medications such as amitriptyline (ElavilTM), amitriptyline/perplienazine combinations (E
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of multiple sclerosis or its symptoms, which second agents include: (a) azathioprine (ImuranTM); (b) copolymer- 1 (random polymer of L-alanine, L-glutamic o acid, L-lysine and L-tyrosine, ratio of 6.0:1.9:4.7:1.0, of molecular weight between two second agents.
  • second agents include: (a) azathioprine (ImuranTM); (b) copolymer- 1 (random polymer of L-alanine, L-glutamic o acid, L-lysine and L-tyrosine, ratio of 6.0:1.9:4.7:1.0, of molecular weight between
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of amyotrophic lateral sclerosis or its symptoms, which second agents include: thyrotropin releasing factor (Relefact TRH); serine; L-threonine; N-methyl-D-aspartate o glutamate receptor antagonists such as milacemide, trihexyphenidyl (ArtaneTM), ethopropazine (ParidolTM), procyclidine (KemadrinTM), diphenliydramine (BenadrylTM), dizocilpine (NeurogardTM), amantadine (SymmetrelTM), and memantine (NamendaTM).
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of Huntington's disease or its symptoms, which second agents include: (a) N- 5 methyl-D-aspartate glutamate receptor antagonists such as milacemide, trihexyphenidyl (ArtaneTM), ethopropazine (ParidolTM), procyclidine (KemadrinTM), diphenhydramine (BenadrylTM), dizocilpine (NeurogardTM), amantadine (SymmetrelTM), and memantine (NamendaTM); (b) agents which may enhance acetylcholine synthesis, storage or release such as phosphatidylcholine, 3,4-diaminopyridine, choline chloride, and choline o bitartrate; and (c) postsynaptic receptor agonists such as arecoline.
  • N- 5 methyl-D-aspartate glutamate receptor antagonists such as milacemide, trihexy
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of olivopontocerebellar atrophy or its symptoms, which second agents include: N-methyl-D-aspartate glutamate receptor antagonists such as milacemide, trihexyphenidyl (ArtaneTM), ethopropazine (ParidolTM), procyclidine (KemadrinTM), diphenhydramine (BenadrylTM), dizocilpine (NeurogardTM), amantadine (SymmetrelTM), and memantine (NamendaTM).
  • N-methyl-D-aspartate glutamate receptor antagonists such as milacemide, trihexyphenidyl (ArtaneTM), ethopropazine (ParidolTM), procyclidine (KemadrinTM), diphenhydramine (BenadrylTM), dizocilpine (NeurogardTM), amantadine (SymmetrelTM), and meman
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of alcoholic polyneuropathy or its symptoms, which second agents include: tiapride, physostigmine, optionally with phosphatidylcholine co-agent, piracetam, and cyclandelate.
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of hereditary motor and sensory neuropathies or its symptoms, which second agents include 3,4-diaminopyridine.
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of urinary incontinence resulting from Alzheimer's senile dementia, demyelinating diseases such as multiple sclerosis, peripheral nerve lesions, diabetes mellitus and alcoholic polyneuropathy or its symptoms, which second agents include: (a) cholinergics such as bethanechol (UrecholineTM), alone or in combination with prazosin; (b) anti-cholinergics such as hyoscyamine sulfate, atropine sulfate, propantheline (Pro-BanthineTM), oxybutynin (DitropanTM), and dicyclomine (BentylTM); (c) ⁇ -adrenergics such as ephedrine and phenylpropanolamine; (d) tricyclic agents such as imiprarnine (TofranilTM) and doxepin (AdapinTM); (e) flavoxate (UripasTM); (f
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of gastroesophageal reflux disease, hypoperistalsis and/or delayed gastric emptying or its symptoms, which second agents include: metoclopramide (ReglanTM), cisapride (PrepulsidTM), famotidine (PepcidTM), cimetidine (TagametTM), ranitidine (ZantacTM), omeprazole (PrilosecTM), and galanthamine.
  • metoclopramide (ReglanTM
  • cisapride PrepulsidTM
  • famotidine PepcidTM
  • cimetidine TagametTM
  • ranitidine ZantacTM
  • omeprazole omeprazole
  • Suitable second agents further include therapeutic drugs that are useful in the treatment of symptomology related to onset and development of atherosclerosis or its symptoms, which second agents include: (a) angiotensin-converting enzyme inhibitor free radical scavenging agents possessing sulfhydryl groups such as captopril (CapotenTM), captopril in combination with hydrochlorothiazide (CapozideTM), epi- captopril, alacepril, pivalopril, and rentiapril; (b) fibric acid derivative anti- hyperlipidemia agents such as gemfibrozil (LopidTM), clofibrate (Atromid-STM), 5 bezaf ⁇ brate, and fenofibrate; (c) metformin; (d) nicotinic acid (NicolarTM); (e) natural hydroscopic non-digestible edible plant carbohydrate polymers such as guar gum; (f) 3- hydroxy-3-methylglutaryl-CoA reductase
  • ⁇ -adrenergic agonists such as clonidine (CatapresTM), clonidine TTS (CatapresTM), guanabenz (WytensinTM), guanfacine (TenexTM) and methyldopa (AldometTM); peripherally acting adrenergic antagonists such as guanadrel (HylorelTM), guanethidine (IsmelinTM), whole root Rauwolfia alkaloids (RaudixinTM) and reserpine (SerpasilTM); ⁇ -adrenergic antagonists such as prazosin (MinipressTM), prazosin/polythiazide combination (MinizideTM), terazosin (HytrinTM) and doxazos
  • compositions of certain alkanesulfur-oxides including alkanesulfonic acids and alkanesulfuric acids, and more particularly including, for example, 3-amino- 1-propanesulfonic acid and certain salts thereof have been shown to be useful in the treatment of amyloid- ⁇ related diseases, including Alzheimer's disease and cerebral amyloid angiopathy. See WO 96/28187, WO 01/85093, and U.S. Pat. No. 5,840,294.
  • the anionic group of the composition is believed to inhibit an interaction between an amyloidogenic protein and a glycosaminoglycan (GAG) or proteoglycan constituent of a basement membrane to thus inhibit amyloid deposition.
  • GAG glycosaminoglycan
  • an amyloidogenic protein ⁇ e.g., serum amyloid A protein or ⁇ -amyloid precursor protein ( ⁇ -APP)
  • the solid support is washed extensively to remove unbound 5 material.
  • the binding of the basement membrane constituent (e.g., HSPG) to the amyloidogenic protein (e.g., ⁇ -APP) is then measured using an antibody directed against the basement membrane constituent that is conjugated to a detectable substance (e.g., an enzyme, such as alkaline phosphatase) by detecting the detectable substance.
  • a detectable substance e.g., an enzyme, such as alkaline phosphatase
  • a compound which inhibits an interaction between an amyloidogenic protein and a o glycoprotein or proteoglycan constituent of a basement membrane will reduce the amount of substance detected (e.g., will inhibit the amount of enzyme activity detected).
  • a therapeutic compound of the invention may interact with a binding site for a basement membrane glycoprotein or proteoglycan in an amyloidogenic protein and thereby inhibit the binding of the amyloidogenic protein to the basement membrane constituent.
  • Basement membrane glycoproteins and proteoglycans include laminin, collagen type IV, flbronectin and heparan sulfate proteoglycan (HSPG), perlecan, and agrin.
  • the therapeutic compound inhibits an interaction between an amyloidogenic protein and HSPG. Consensus binding site motifs for HSPG in amyloidogenic proteins have been described (see, e.g., Cardin and Weintraub, o Arteriosclerosis 9, 21-32 (1989)).
  • the method also relates to a method for treating or preventing an amyloid- ⁇ related disease by administering at least two agents, each of which exerts a therapeutic effect when so administered and is useful in treating or preventing a neurological disease.
  • the first agent of the invention is selected from alkanesulfonic acids that are 5 useful for treating or preventing an amyloid- ⁇ related disease.
  • the second agent is therapeutic, i.e., its function is beyond that of an inactive ingredient, such as a pharmaceutical carrier, preservative, diluent, or buffer.
  • the second agent maybe useful in treating or preventing an amyloid- ⁇ related disease or another neurological disease.
  • the second agent may also be useful in diminishing specific symptoms which are 0 characteristic of Alzheimer's disease (e.g., memory loss, anxiety, etc.).
  • the first and second agents may exert their biological effects by similar or unrelated mechanisms of action; or either one or both of the first and second agents may exert their biological effects by a multiplicity of mechanisms of action.
  • a third agent, or even more yet, may likewise be used in a method of the invention, wherein the third (and fourth, etc.) agent 5 has the same characteristics of a second agent.
  • the invention relates to a method of treating or preventing an amyloid- ⁇ related disease in a subject (for example, a human) comprising administering to the subject a therapeutic amount of a alkanesulfonic acid, such that amyloid fibril formation or deposition, neurodegeneration, or cellular toxicity is reduced or inhibited.
  • the invention in another embodiment, relates to a method of treating or preventing an amyloid- ⁇ related in a subject (for example, a human) comprising administering to the subject a therapeutic amount of an alkanesulfonic acid, such that cognitive function is stabilized or further deterioration in cognitive function is prevented, slowed, or stopped in subjects with brain amyloidosis, e.g., Alzheimer's disease or cerebral amyloid angiopathy, hi another embodiment, the invention relates to a method of treating or preventing an amyloid- ⁇ disease in a subject (for example, human) comprising administering to the subject a therapeutic amount of a alkanesulfonic acid, such that activities of daily living are improved or stabilized in subjects with brain amyloidosis, e.g., Alzheimer's disease.
  • the "first agent” according to the invention may be an alkanesulfonic acid or an alkanolsulfuric acid.
  • alkanesulfonic acid includes substituted or unsubstituted alkanesulfonic acids, and substituted or unsubstituted lower alkanesulfonic acids.
  • Amino-substituted compounds are especially noteworthy and the invention pertains to substituted- or unsubstituted-amino-substituted alkanesulfonic acids, and substituted- or unsubstituted-amino-substituted lower alkanesulfonic acids, and example of which is 3-amino-l-propanesulfonic acid.
  • compositions of the invention are therefore directed to a first agent that is a substituted or unsubstituted alkanesulfonic acid, substituted or unsubstituted alkanesulfuric acid (also known as an alkanol sulfuric acid), substituted or unsubstituted alkylthiosulfonic acid, substituted or unsubstituted alkylthiosulfuric acid, or an ester or amide thereof, including pharmaceutically acceptable salts thereof.
  • the invention relates to a first agent that is a substituted or unsubstituted alkanesulfonic acid, or an ester or amide thereof, including pharmaceutically acceptable salts thereof
  • the invention pertains to a first agent that is a substituted or unsubstituted lower alkanesulfonic acid, or an ester or amide thereof, including pharmaceutically acceptable salts thereof.
  • the invention includes a first agent that is a (substituted- or unsubstituted- amino)-substituted alkanesulfonic acid, or an ester or amide thereof, including pharmaceutically acceptable salts thereof.
  • the first agent is a (substituted- or unsubstituted-amino)-substituted lower alkanesulfonic acid, or an ester or amide thereof, including pharmaceutically acceptable salts thereof.
  • alkyl groups include saturated hydrocarbons having one or more carbon atoms, including straight-chain alkyl groups ⁇ e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, etc.), cyclic alkyl groups (or “cycloalkyl” or “alicyclic” or “carbocyclic” groups) (e.g., cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, etc.), branched-chain alkyl groups (isopropyl, tert-bntyl, sec-butyl, isobutyl, etc.), and alkyl-substituted alkyl groups (e.g., alkyl-substituted cycloalkyl groups and cycloal
  • aliphatic group includes organic moieties characterized by straight or branched-chains, typically having between 1 and 22 carbon atoms. In complex structures, the chains may be branched, bridged, or cross-linked. Aliphatic groups include alkyl groups, alkenyl groups, and alkynyl groups.
  • the invention relates to substituted or unsubstituted alkanesulfonic acids that are substituted or unsubstituted straight-chain alkanesulfonic acids, substituted or unsubstituted cycloalkanesulfonic acids, and substituted or unsubstituted branched-chain alkanesulfonic acids.
  • the structures of some of the compounds of this invention include stereo genie carbon atoms. It is to be understood that isomers arising from such asymmetry (e.g., all enantiomers and diastereomers) are included within the scope of this invention unless indicated otherwise. That is, unless otherwise stipulated, any chiral carbon center may be of either (R)- or ( ⁇ -stereochemistry. Such isomers can be obtained in substantially pure form by classical separation techniques and by stereochemically-controlled synthesis, hi addition, the compounds of the present invention may exist in unsolvated as well as solvated forms with acceptable solvents such as water, THF, etlianoL and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
  • solvate represents an aggregate that comprises one or more molecules of a compound, with one or more molecules of a pharmaceutical solvent, such as water, ethanol, and the like.
  • a pharmaceutical solvent such as water, ethanol, and the like.
  • a straight-chain or branched-chain alkyl group may have 20 or fewer carbon atoms in its backbone, e.g., C 1 -C 20 for straight-chain or C 3 -C 20 for branched-chain, and more, for example, 18 or fewer.
  • example cycloalkyl groups have from 4-10 carbon atoms in their ring structure, or 4-7 carbon atoms in the ring structure.
  • lower alkyl refers to alkyl groups having from 1 to 6 carbons in the chain, and to cycloalkyl groups having from 3 to 6 carbons in the ring structure. Unless the number of carbons is otherwise specified, “lower” as in “lower alkyl,” means that the moiety has at least one and less than about 8 carbon atoms.
  • a straight-chain or branched-chain lower alkyl group has 6 or fewer carbon atoms in its backbone (e.g., C 1 -C 6 for straight-chain, C 3 -C 6 for branched-chain), and more preferably 4 or fewer, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, and tert-butyl.
  • cycloalkyl groups may have from 3-8 carbon atoms in their ring structure, for example, 5 or 6 carbons in the ring structure.
  • C 1 -Q as in "Ci-C 6 alkyl" means alkyl groups containing 1 to 6 carbon atoms.
  • alkyl includes both "unsubstituted alkyls" and “substituted alkyls,” the latter of which refers to alkyl groups having substituents replacing one or more hydrogens on one or more carbons of the hydrocarbon backbone.
  • substituents may include, for example, alkenyl, alkynyl, halogeno, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, s
  • arylalkyl is an alkyl group substituted with an aryl group (e.g., phenyhnethyl (i.e., benzyl)).
  • alkylaryl moiety is an aryl group substituted with an alkyl group (e.g., j>-methylphenyl (i.e.,p-toly ⁇ )).
  • rc-alkyl means a straight-chain (i.e., unbranched) unsubstituted alkyl group.
  • An “alkyleiie” group is a divalent analog of the corresponding alkyl group.
  • alkenyl and alkynyl refer to unsaturated aliphatic groups analogous to alkyls, but which contain at least one double or triple carbon-carbon bond respectively. Suitable alkenyl and alkynyl groups include groups having 2 to about 12 carbon atoms, preferably from 2 to about 6 carbon atoms.
  • aromatic group or "aryl group” includes unsaturated and aromatic cyclic hydrocarbons as well as unsaturated and aromatic heterocycles containing one or more rings.
  • Aryl groups may also be fused or bridged with alicyclic or heterocyclic rings that are not aromatic so as to form a polycycle (e.g., tetralin).
  • An "arylene” group is a divalent analog of an aryl group.
  • Aryl groups can also be fused or bridged with alicyclic or heterocyclic rings which are not aromatic so as to form a polycycle (e.g., tetralin).
  • heterocyclic group includes closed ring structures analogous to carbocyclic groups in which one or more of the carbon atoms in the ring is an element other than carbon, for example, nitrogen, sulfur, or oxygen. Heterocyclic groups may be saturated or unsaturated. Additionally, heterocyclic groups (such as pyrrolyl, pyridyl, isoquinolyl, quinolyl, purinyl, and furyl) may have aromatic character, in which case they may be referred to as “heteroaryl” or “heteroaromatic” groups. Unless otherwise stipulated, aryl and heterocyclic (including heteroaryl) groups may also be substituted at one or more constituent atoms.
  • heteroaromatic and heteroalicyclic groups may have 1 to 3 separate or fused rings with 3 to about 8 members per ring and one or more N, O, or S heteroatoms.
  • lieteroatom includes atoms of any element other than carbon or hydrogen, preferred examples of which include nitrogen, oxygen, sulfur, and phosphorus.
  • Heterocyclic groups may be saturated or unsaturated or aromatic.
  • heterocycles include, but are not limited to, acridinyl; azocinyl; benzimidazolyl; benzofuranyl; benzothiofuranyl; benzothiophenyl; benzoxazolyl; benzthiazolyl; benztriazolyl; benztetrazolyl; benzisoxazolyl; benzisothiazolyl; benzimidazolinyl; carbazolyl; 4aH-carbazoryl; carbolinyl; chromanyl; chromenyl; cinnolinyl; decahydroquinolinyl; 2H,6H-l,5,2-dithiazinyl; dihydrofuro[2,3-b]tetrahydrofuran; furanyl; furazanyl; imidazolidinyl; imidazolinyl; imidazolyl; lH-indazolyl; indolenyl; indolinyl
  • Preferred heterocycles include, but are not limited to, pyridinyl; furanyl; thienyl; pyrrolyl; pyrazolyl; pyrrolidinyl; imidazolyl; indolyl; benzimidazolyl; lH-indazolyl; oxazolidinyl; benzotriazolyl; benzisoxazolyl; oxindolyl; benzoxazolinyl; and isatinoyl groups. Also included are fused ring and spiro compounds containing, for example, the above heterocycles.
  • a common hydrocarbon aryl group is a phenyl group having one ring.
  • Two-ring hydrocarbon aryl groups include naphthyl, indenyl, benzocyclooctenyl, benzocycloheptenyl, pentalenyl, and azulenyl groups, as well as the partially hydrogenated analogs thereof such as indanyl and tetrahydronaphthyl.
  • Exemplary three- ring hydrocarbon aryl groups include acephthylenyl, fluorenyl, phenalenyl, phenanthrenyl, and anthracenyl groups.
  • Aryl groups also include heteromonocyclic aryl groups, i.e., single-ring heteroaryl groups, such as thienyl, furyl, pyranyl, pyrrolyl, imidazolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, and pyridazinyl groups; and oxidized analogs thereof such as pyridonyl, oxazolonyl, pyrazolonyl, isoxazolonyl, and thiazolonyl groups.
  • heteromonocyclic aryl groups i.e., single-ring heteroaryl groups, such as thienyl, furyl, pyranyl, pyrrolyl, imidazolyl, pyrazolyl, pyridinyl, pyrazinyl, pyrimidinyl, and pyridazinyl groups
  • oxidized analogs thereof such as pyridonyl,
  • the corresponding hydrogenated (i.e., non-aromatic) heteromonocylic groups include pyrrolidinyl, pyrrolinyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperidyl and piperidino, piperazinyl, and morpholino and niorphormyl groups.
  • Aryl groups also include fused two-ring heteroaryls such as indolyl, isoindolyl, indolizinyl, indazolyl, quinolinyl, isoquinolinyl, phthalazinyl, quinoxalinyl, quinazolinyl, cinnolinyl, chromenyl, isochromenyl, benzothienyl, benzimidazolyl, benzothiazolyl, purinyl, quinolizinyl, isoquinolonyl, quinolonyl, naphthyridinyl, and pteridinyl groups, as well as the partially hydrogenated analogs such as chromanyl, isochromanyl, indolinyl, isoindolinyl, and tetrahydroindolyl groups.
  • heteroaryls such as indolyl, isoindolyl, indolizinyl, indazolyl,
  • Aryl groups also include fused three-ring groups such as phenoxathiinyl, carbazolyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and dibenzofuranyl groups.
  • each Ar group may be selected from the group consisting of substituted or unsubstituted phenyl, pyrrolyl, furyl, thienyl, thiazolyl, isothiaozolyl, imidazolyl, triazolyl, tetrazolyl, pyrazolyl, oxazolyl, isooxazolyl, pyridinyl, pyrazinyl, pyridazinyl, and pyrimidinyl groups.
  • phenyl substituted or unsubstituted phenyl, 1 -naphthyl, 2-naphthyl, biphenyl, 1 -pyrrolyl, 2- pyrrolyl, 3 -pyrrolyl, 3 -pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4- oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5- thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2- pyrimidyl, 4-pyrimidyl, 5 -benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1- isoquinolyl
  • amine refers to an unsubstituted or substituted moiety of the formula -NR a R b , in which R a and R b are each independently hydrogen, alkyl, aryl, or heterocyclyl, or R a and R b , taken together with the nitrogen atom to which they are attached, form a cyclic moiety having from 3 to 8 atoms in the ring.
  • amino includes cyclic amino moieties such as piperidinyl or pyrrolidinyl groups, unless otherwise stated.
  • alkylamino as used herein 5 means an alkyl group having an amino group attached thereto.
  • Suitable alkylamino groups include groups having 1 to about 12 carbon atoms, for example, 1 to about 6 carbon atoms.
  • amino includes compounds or moieties in which a nitrogen atom is covalently bonded to at least one carbon or heteroatom.
  • dialkylamino includes groups wherein the nitrogen atom is bound to at least two alkyl o groups.
  • arylamino and diarylamino include groups wherein the nitrogen is bound to at least one or two aryl groups, respectively.
  • alkylarylamino refers to an amino group which is bound to at least one alkyl group and at least one aryl group.
  • alkaminoalkyl refers to an alkyl, alkenyl, or alkynyl group substituted with an alkylamino group.
  • amide or “aminocarbonyl” includes compounds or 5 moieties which contain a nitrogen atom which is bound to the carbon of a carbonyl or a thiocarbonyl group.
  • alkylthio refers to an alkyl group, having a sulfhydryl group attached thereto. Suitable alkylthio groups include groups having 1 to about 12 carbon atoms, preferably from 1 to about 6 carbon atoms. o
  • alkylcarboxyl as used herein means an alkyl group having a carboxyl group attached thereto.
  • alkoxy as used herein means an alkyl group having an oxygen atom attached thereto.
  • Representative alkoxy groups include groups having 1 to about 12 carbon atoms, preferably 1 to about 6 carbon atoms, e.g., methoxy, ethoxy, propoxy, 5 tert-butoxy and the like.
  • Examples of alkoxy groups include methoxy, ethoxy, isopropyloxy, propoxy, butoxy, and pentoxy groups.
  • the alkoxy groups can be substituted with groups such as alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, o dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate,
  • halogen substituted alkoxy groups include, but are not limited to, fluoromethoxy, difluoromethoxy, trifluoromethoxy, chloromethoxy, dichloromethoxy, trichloromethoxy, etc., as well as perhalogenated alkyloxy groups.
  • acylarnino includes moieties wherein an amino moiety is bonded to an acyl group.
  • the acylamino group includes alkylcarbonylamino, 5 arylcarbonylamino, carbamoyl and ureido groups.
  • alkoxyalkyl examples include alkyl groups, as described above, which further include oxygen, nitrogen or sulfur atoms replacing one or more carbons of the hydrocarbon backbone.
  • carbonyl or “carboxy” includes compounds and moieties which o contain a carbon connected with a double bond to an oxygen atom.
  • moieties which contain a carbonyl include aldehydes, ketones, carboxylic acids, amides, esters, anhydrides, etc.
  • ether or “ethereal” includes compounds or moieties which contain an oxygen bonded to two carbon atoms.
  • an ether or ethereal group includes 5 "alkoxyalkyl” which refers to an alkyl, alkenyl, or alkynyl group substituted with an alkoxy group.
  • nitro means -NO 2 ;
  • thiol means SH;
  • hydroxyl or “hydroxy” means -OH.
  • acyl refers to a carbonyl group that is attached through its carbon atom to a hydrogen (i.e., a formyl), an aliphatic group (e.g., acetyl), an aromatic group (e.g., benzoyl), and the like.
  • substituted acyl includes acyl groups where one or more of the hydrogen atoms on one or more carbon atoms are replaced by, for example, an alkyl group, alkynyl group, halogen, hydroxyl, alkylcarbonyloxy, 5 arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, o carbamoyl and ureido), imin
  • a "sulfonic acid” or “sulfonate” group is a -SO 3 H or -SO 3 ' X + group bonded to a carbon atom, where X + is a cationic counter ion group.
  • a "sulfonic acid” 5 compound has a -SO 3 H or -SO 3 -X + group bonded to a carbon atom, where X + is a cationic group.
  • a “sulfate” as used herein is a -OSO 3 H or -OSO 3 -X + group (which may also be represented as -SO 4 H or -SO 4 -X + ) bonded to a carbon atom, and a "sulfuric acid" compound has a -SO 3 H or -OSO 3 -X + group bonded to a carbon atom, where X + is a cationic group.
  • a suitable cationic group may be a 5 hydrogen atom. In certain cases, the cationic group may actually be another group on the therapeutic compound that is positively charged at physiological pH, for example an amino group.
  • a "counter ion” is required to maintain electroneutrality, and is pharmaceutically acceptable in the compositions of the invention.
  • anionic counter ions include halide, triflate, sulfate, nitrate, hydroxide, carbonate, bicarbonate, o acetate, phosphate, oxalate, cyanide, alkylcarboxylate, N-hydroxysuccinimide , iV-hydroxybenzotriazole, alkoxide, thioalkoxide, alkane sulfonyloxy, lialogenated alkane sulfonyloxy, arylsulfonyloxy, bisulfate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, or lactobionate.
  • the chemical moieties of the compounds of the invention may be "substituted or unsubstituted.”
  • substituted means that the moiety has o substituents placed on the moiety other than hydrogen (/. e. , in most cases, replacing a hydrogen), which allow the molecule to perform its intended function.
  • substituents include moieties selected from straight or branched alkyl (e.g.,Ci-C 5 ), cycloalkyl (e.g., C 3 -C 8 ), amino groups (including -NH 2 ), -SO 3 H, -OSO 3 H, -CN, -NO 2 , halogen (e.g., -F, -Cl, -Br, or -I), -CH 2 OCH 3 , -OCH 3 , -SH, -SCH 3 , -OH, and -CO 2 H, 5 alkoxy (preferably C 1 -C 6 ), thioalkyl (preferably C 1 -C 6 ), alkenyl (preferably C 2 -C 6 ), alkynyl (preferably C 2 -C 6 ), heterocyclic, carbocyclic, aryl (e.g., phenyl), aryloxy (e.g., phenoxy), aralkyl (e.g.
  • R'R 0-3 (substituted or unsubstituted phenyl), (CR'R") 0-3 (C 3 -C 8 cycloalkyl), (CR'R") 0-3 CO 2 R' (e.g., -CO 2 H), and (CR'R") 0-3 OR' groups, wherein R' and R" are each independently hydrogen, a C 1 -C 5 alkyl, C 2 -C 5 alkenyl, C 2 -C 5 alkynyl, or aryl group; or the side chain of any naturally occurring amino acid.
  • a substituent may be selected from straight or branched alkyl (preferably C 1 -C 5 ), cycloalkyl (preferably C 3 -C 8 ), alkoxy (preferably C 1 -C 6 ), thioalkyl (preferably C 1 -C 6 ), alkenyl (preferably C 2 -C 6 ), alkynyl (preferably C 2 -C 6 ), heterocyclic, carbocyclic, aryl (e.g., phenyl), aryloxy (e.g., phenoxy), aralkyl (e.g., benzyl), aryloxyalkyl (e.g., phenyloxyalkyl), arylacetamidoyl, alkylaryl, heteroaralkyl, alkylcarbonyl and arylcarbonyl or other such acyl group, heteroarylcarbonyl, or heteroaryl group, (CR'R")o-ioNR'R" (e.g., -
  • (CR'R")o -10 substituted or unsubstituted phenyl), (CR'R") O -io(C 3 -C 8 cycloalkyl), (CR'R")o -10 C0 2 R' (e.g., -CO 2 H), or (CR'R")o -10 OR 5 group, or the side chain of any naturally occurring amino acid; wherein R 5 and R 55 are each independently hydrogen, a C 1 -C 5 alkyl, C 2 -C 5 alkenyl, C 2 -C 5 alkynyl, or aryl group, or R 5 and R 5 ' taken together are a benzylidene group or a -(CH 2 ) 2 O(CH 2 ) 2 - group.
  • substituted 55 is meant to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • the permissible substituents can be one or more.
  • substitution 55 or “substituted with 55 includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • substituted 55 is meant to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • a "substituent" may be, selected from the group consisting of, for example, halogeno, trifluoromethyl, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkylcarbonyloxy, arylcarbonyloxy, C 1 -C 6 alkoxycarbonyloxy, aryloxycarbonyloxy, C 1 -C 6 alkylcarbonyl, 5 C 1 -C 6 alkoxycarbonyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylthio, arylthio, heterocyclyl, aralkyl, and aryl (including heteroaryl) groups.
  • alkanesulfonic acids have the following structure as depicted in Formula I (see the Drawings, attached hereto) where Y is either an amino group (having the formula -NR a R b ) or a sulfonic acid group (having the formula o -SO 3 " X + ), n is an integer from 1 to 5, and X is hydrogen or a cationic group
  • alkanesulfonic acids include the those depicted within Formula Ha, Formula lib, Formula Hc, and Formula lid (see Figures).
  • One embodiment of the invention is the use of 3-amino-l-propanesulfonic acid and pharmaceutically acceptable salts thereof as a first agent of the pharmaceutical 5 compositions described herein and the methods of using them.
  • an “agent,” as in a “first agent” or a “second agent” is generally intended to describe a chemical compound of suitable purity for use in a pharmaceutical preparation.
  • the agent is a "small molecule,” that is, a compound that that is not itself the product of gene transcription or translation (e.g., protein, RNA, or DNA) and has a o low molecular weight, e.g. , less than about 2500.
  • the agent may be a biological product, such as an antibody or an immunogenic peptide.
  • alkanesulfonic acids may be prepared by the methods illustrated in the general reaction schemes as, for example, described in U.S. Pat. Nos.
  • agents of the present 5 invention may be readily prepared in accordance with the synthesis schemes and protocols described herein, as illustrated in the specific procedures provided. However, those skilled in the art will recognize that other synthetic pathways for forming the agents of this invention may be used, and that the following is provided merely by way of example, and is not limiting to the present invention. See, e.g., "Comprehensive o Organic Transformations," 2 nd Ed., by R.C.
  • alkenes can include either the E- or Z- geometry, where appropriate, hi 5 addition, the compounds of the present invention may exist in unsolvated as well as solvated forms with acceptable solvents such as water, THF, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
  • the invention pertains, at least in part to a pharmaceutical composition having a first agent that is a compound of Formula I- A (see the Drawings sheets attached hereto), wherein R 1 is a substituted or unsubstituted cycloalkyl, aryl, arylcycloalkyl, bicyclic or tricyclic ring, a bicyclic or tricyclic fused ring group, or a substituted or unsubstituted C 2 -C 10 alkyl group; R is selected from a group consisting o hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, and benzoimidazolyl; Y is SO 3 -X + , OSO 3 -X + , or SSO 3 ⁇ X + ; X + is hydrogen, a compound of Formula I-
  • the invention pertains, at least in part a pharmaceutical composition having a first agent that is a compound of Formula II- A (see the Drawings sheets attached hereto) wherein R 1 is a substituted or unsubstituted cyclic, bicyclic, tricyclic, or benzoheterocyclic group or a substituted or unsubstituted C 2 -C 10 alkyl o group; R 2 is hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, benzoimidazolyl, or linked to R 1 to form a heterocycle; Y is SO 3 -X + , OSO 3 -X + , or SSO 3 -X + ; X + is hydrogen, a cationic group, or an ester forming moiety; m is 0 or 1;
  • the invention pertains, at least in part to a pharmaceutical composition having a first agent that is a compound of Formula III- A (see the Drawings sheets attached hereto) wherein A is nitrogen or oxygen; R 11 is hydrogen, salt-forming cation, ester forming group, — (CH 2 ) X — Q, or when A is nitrogen, A and R 1 * taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof; Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl; x is 0, 1, 2, 3, or 4; n is 0, 1 ,2 ,3, 4, 5, 6, 7, 8, 9, or 10; R 3 , R 3a , R 4 , R 4a , R 5 , R 5a , R 5 , R 6a , R 7 and R 7a are each independently hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl,
  • the invention pertains at least in part to a pharmaceutical composition having a first agent that is a compound of Formula TV (see the attached Drawings) wherein: A is nitrogen or oxygen; R 11 is hydrogen, salt- forming cation, ester forming group, -(CH 2 ) X -Q, or when A is nitrogen, A and R 11 taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof; Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl; x is O 5 1, 2, 3, or 4; n is 0, 1 ,2 ,3, 4, 5, 6, 7, 8, 9, or 10; R 4 , R 4a , R 5 , R 5a , R 6 , R 6a , R 7 , and R 7a are each independently hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl,
  • the invention includes a pharmaceutical composition having a first agent that is a compound of Formula V-A (see the attached Drawings) wherein A is nitrogen or oxygen; R 11 is hydrogen, salt-forming cation, ester forming group, — (CH 2 ) X — Q, or when A is nitrogen, A and R ⁇ taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof; Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl; x is 0, 1, 2, 3, or 4; n is 0, 1 ,2 ,3, 4, 5, 6, 7, 8, 9, or 10; aa is a natural or unnatural amino acid residue; m is 0, 1, 2, or 3; R 14 is hydrogen or protecting group; R 15 is hydrogen, alkyl or aryl, and pharmaceutically acceptable salts and prodrugs thereof.
  • A is nitrogen or oxygen
  • R 11 is hydrogen, salt-forming cation, ester forming group,
  • the invention includes a pharmaceutical composition having a first agent that is a compound of the Formula VI-A (see the attached Drawings) wherein n is 1, 2 3 3, 4, 5, 6, 7, 8, 9, or 10; A is oxygen or nitrogen; R 11 is hydrogen, salt- forming cation, ester forming group, — (CH 2 ) X — Q, or when A is nitrogen, A and R 11 taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof; Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl; x is 0, 1, 2, 3, or 4; R 19 is hydrogen, alkyl or aryl; Y 1 is oxygen, sulfur, or nitrogen; Y 2 is carbon, nitrogen, or oxygen; R 20 is hydrogen, alkyl, amino, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl,
  • the invention includes a pharmaceutical composition having a first agent that is a compound of Formula VII-A (see the attached Drawings) wherein: n is 2, 3, or 4; A is oxygen or nitrogen; R 11 is hydrogen, salt-forming cation, ester forming group, -(CH 2 ) X -Q, or when A is nitrogen, A and R 11 taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof; Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl; x is 0, 1, 2, 3, or 4; G is a direct bond or oxygen, nitrogen, or sulfur; z is 0, 1, 2, 3, 4, or 5; m is 0 or 1; R 24 is selected from a group consisting hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, aroyl, alkylcarbonyl, aminoalkylcarbonyl, cycloalky
  • m is 0, 1, or 2.
  • n is 0, 1, or 2.
  • R 3 is aryl, e.g., heteroaryl or phenyl, hi yet another embodiment, Z is S(O) 2 .
  • the compound of the invention is of the Formula II-B (see the attached Drawings) wherein: each R 4 is independently selected from the group consisting of hydrogen, halogen, hydroxyl, thiol, amino, amidino, cyano, nitro, alkyl, o aryl, carbocyclic or heterocyclic; J is absent, oxygen, nitrogen, sulfur, or a divalent link- moiety comsisting of, without limiting to, lower alkylene, alkylenyloxy, alkylenylamino, alkylenylthio, alkylenyloxyalkyl, alkylenylamonialkyl, alkylenylthioalkyl, alkenyl, alkenyloxy, alkenylamino, or alkenylthio; and q is 1, 2, 3, 4, or 5, and pharmaceutically acceptable salts, esters and prodrugs thereof.
  • R 4 is aryl, e.g., substituted or unsubstituted phenyl, hi another embodiment, R 4 is halogen (e.g., chlorine, fluorine, bromine, or iodine), hi yet another embodiment, R 4 is alkyl, e.g., methyl, ethyl, propyl, butyl, pentyl, trifluoromethyl, etc.
  • J is absent or oxygen
  • m is 1 or n is 1.
  • the compound can be R- or iS-isomer.
  • the compound may be selected from the group consisting of those compounds depicted in either Table X or Table Y (see the attached Drawings) and pharmaceutically acceptable salts, prodrugs, and esters thereof.
  • the compound is selected from the group consisting 5 of those compounds depicted in either Table Z-I or Table Z-2 (see the attached Drawings) and pharmaceutically acceptable salts, prodrugs, and esters thereof.
  • the compound of the invention is of the Formula III-B (see the attached Drawings) wherein: X is oxygen or nitrogen; m and n are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10; q is 1, 2, 3, 4, or 5; R 1 is hydrogen, metal ion, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, or a moiety together with X 5 to form a natural or unnatural amino acid residue, or — (CH 2 ) p — Y; Y is hydrogen or a heterocyclic moiety selected from the group consisting of thiazolyl, triazolyl, tetrazolyl, amidino, imidazolyl, benzothiazo
  • the compound of the invention may be a compound of Formula IV-B (see the attached Drawings).
  • m is 0.
  • Examples of compounds of the invention include those compounds depicted in o Table W (see the attached Drawings) and pharmaceutically acceptable salts, esters, and prodrugs thereof.
  • the invention pertains to compounds of Formula V-B (see the attached Drawings) wherein: R 6 is a substituted or unsubstituted heterocyclic moiety.
  • m is 0 or 1.
  • n is 0 or 1.
  • R 6 is thiazolyl, oxazoylyl, pyrazolyl, indolyl, pyridinyl, thiazinyl, thiophenyl, benzothiophenyl, dihydroimidazolyl, dihydrothiazolyl, oxazolidinyl, thiazolidinyl, tetrahydropyrimidinyl, or oxazinyl.
  • Z is S(O) 2 .
  • the invention pertains to the following compounds o depicted in Table V (see the attached Drawings) and pharmaceutically acceptable salts, esters, and prodrugs thereof.
  • the invention pertains to compounds of Formula I-C:
  • R 1 is a substituted or unsubstituted cycloalkyl, heterocyclic, aryl, arylcycloalkyl, bicyclic or tricyclic ring, a bicyclic or tricyclic fused ring group, or a substituted or unsubstituted C 2 -Ci O alkyl group;
  • R 2 is selected from a group consisting of hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, and benzoimidazolyl;
  • Y is SO 3 -X + , OSOfX + , or SSOfX + ;
  • X + is hydrogen, a cationic group, or ester-forming group; and each of L 1 and L 2 is independently a substituted or unsubstituted Ci-C 5 alkyl group or absent, or a pharmaceutically acceptable salt thereof, provided that when R 1 is alkyl, L 1 is absent.
  • the invention pertains to compounds of Formula H-C:
  • R 1 is a substituted or unsubstituted cyclic, bicyclic, tricyclic, or benzoheterocyclic group or a substituted or unsubstituted C 2 -Ci O alkyl group;
  • R 2 is hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, benzoimidazolyl, or linked to R L to form a heterocycle;
  • Y is SOfX + , OSOfX + , or SSOfX + ;
  • X + is hydrogen, a cationic group, or an ester forming moiety; m is 0 or 1 ; n is 1, 2, 3, or 4;
  • L is substituted or unsubstituted Ci-C 3 alkyl group or absent, or a pharmaceutically acceptable salt thereof, provided that when R 1 is alkyl, L is absent.
  • R 2 is hydrogen.
  • R 1 is straight chain alkyl, for example, ethyl, n-pentyl, n-heptyl, or n-octyl.
  • R 1 is t-butyl.
  • R 1 is C 7 -C 10 bicycloalkyl or tricycloalkyl, such as, for example, tricyelo[3.3.1.0 3;7 ]decyl (or adamantyl), bicyclo[2.1.2]heptyl, or indolyl.
  • R 1 is tetrahydronaphthyl.
  • L 2 is -(CH 2 ) 3 -.
  • L 2 is -
  • L 2 is -(CH 2 ) 2 -.
  • L 2 is substituted alkyl, e.g., -CH 2 -(CHOH)-CH 2 -.
  • L 1 is CH 2 CH 2 or absent.
  • R 1 is branched alkyl, e.g., t-butyl. In another embodiment, R 1 is adamanyl. In another embodiment, R 1 is cyclic alkyl, e.g., cyclopropyl, cyclohexyl, cycloheptyl, cyclo-octyl, etc. The cycloalkyl moieties may be substituted further, e.g., with additional alkyl groups or other groups which allow the molecule to perform its intended function. In another embodiment, R 1 is alkyl substituted with a propargyl moiety (e.g., HC ⁇ €-). Bi another embodiment, R 1 is cyclohexyl substituted withone or more methyl or propargyl groups.
  • L 1 is a C]-C 2 alkyl linker group (e.g., -CH(CH 3 )- or - (CH 2 ) 2 -/.
  • R 1 is phenyl.
  • R 1 is substituted with a methoxy group.
  • L 1 is C 3 , e.g., -(CH 2 ) S - or C(CH 3 ) 2 -.
  • the ester group is a methyl , ethyl, propyl, butyl, cyclohexyl, or benzyl ester. In other embodiments, the ester group may be propenyl.
  • L 1 is substituted with a carboxylate group.
  • R 1 is substituted with a subsituted amido group, wherein the amido group is substituted with an alkyl, e.g., methyl, ethyl, propyl, butyl, pentyl, or hexyl group.
  • the amido group is substituted with an alkyl (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, cyclohexyl, bezyl or aryl group.
  • the amido group is substituted with a -CH(CH 2 )2 group.
  • R 1 itself may be substituted with a phenyl or may be branched or straight chain alkyl.
  • R 1 may also be substituted with a thioether moiety.
  • thioemers examples include S-Me, S-Et, etc.
  • the alkyl R 1 moiety is substituted with both an aryl or a thioether moiety and an amido moiety.
  • the alkyl R ! moiety may be substituted with both a thioether and a carboxylate moiety.
  • alkyl R 1 groups are substituted with hydroxyl.
  • R 1 groups, e.g., alkyl R 1 groups may also be substituted with both thioether and hydroxyl groups.
  • R 1 groups, e.g., alkyl R 1 groups are substituted with cyano groups.
  • R 1 groups including -CN moieties include -C(CH 3 ) 2 CN, cyclohexyl substituted with one or more eyano groups, etc.
  • alkyl R 1 groups are substituted with aryl groups.
  • the aryl groups may be substituted phenyl, for example.
  • the substituted phenyl may be substituted with one or more substituents such as hydroxy, cyano and alkoxy.
  • alkyl R 1 groups are substituted with terrazolyl or substituted or unsubstituted benzyl.
  • L 1 is -C(CH 3 ) 2 ⁇ (CH 2 )-. In another embodiment, L 1 is - (C(CH 3 ) 2 -CHOH-. In yet another embodiment, L 1 is -(C(CH 3 ) 2 CH(OMe)-.
  • R ! is substituted or unsubstituted phenyl. In a further embodiment, R 1 is /? ⁇ r ⁇ -s ⁇ bstituted phenyl. Examples of substitutuents include but are not limited to fluorine, chlorine, bromine, iodine, methyl, t-butyl, alkoxy, methoxy, etc. In other embodiment, R 1 is substituted at the meta position.
  • substituents include methoxy, chloro, methyl, t-butyl, fluoro, alkyl, alkoxy, iodo, trifluoroalkyl, methoxy, etc.
  • R 1 is phenyl substituted in the ortho position, with similar substituents.
  • L 1 comprises a cycloalkyl moiety, e.g., cyclopentyl.
  • L 1 comprises an alkyenyl group and, optionally, a substituted aryl group, with substittuents similar to those described about.
  • R 1 is cyclopropyl or cyclohexyl.
  • the cyclopropyl or cyclohexyl group is subsituted with an ether group or an alkyl group.
  • the ether group is a benzyl ether group.
  • R 1 is alkyl, it is substituted with groups such as phenyl, or hydroxy.
  • the invention pertains to compounds of Formula III-C:
  • A is nitrogen or oxygen
  • R ⁇ is hydrogen, salt-forming cation, ester forming group, — (CH 2 ) X — Q, or when A is nitrogen, A and R 11 taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof;
  • Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl; x is 0, 1, 2, 3, or 4; n is 0, 1 ,2 ,3, 4, 5, 6, 7, 8, 9, or 10;
  • R 3 , R 3a , R 4 , R 4a , R 5 , R 5a , R 6 , R 6a , R 7 and R 7a are each independently hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, cyano, halogen, amino, tetrazolyl, or two R groups on adjacent ring atoms taken together with the ring atoms form a double bond, provided that one of R 3 , R 3a , R 4 , R 4a , R 5 , R 5a , R 6 , R 6a , R 7 and R 7a is a moiety of the Formula HIa-C:
  • m is O, 1, 2, 3, or 4;
  • R ⁇ , R B , R c , R D , and R E are independently selected from a group of hydrogen, halogen, hydroxy!, alkyl, alkoxyl, halogenated alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, cyano, thiazolyl, triazolyl, imidazolyl, tetrazolyl, benzothiazolyl, and benzoimidazolyl; and pharmaceutically acceptable salts and esters thereof, provided that said compound is not 3-(4-phenyl-l, 2, 3, 6-tetrahydro-l-pyridyl)-l-propanesulfonic acid.
  • n is 2, 3 or 4.
  • R 11 is a salt-forming cation.
  • salt forming cations include pharmaceutically acceptable salts described herein as well as lithium, sodium, potassium, magnesium, calcium, barium, zinc, iron, and ammonium.
  • R 11 is an ester-forming group.
  • An ester-forming group include groups when bound form an ester. Examples of such groups include substituted or unsubstituted alkyl, aryl, alkenyl, alkynyl, or cycloalkyl.
  • A is oxygen.
  • R and R are taken together with the carbon atoms to which they are attached to form a double bond
  • R A , R B , R c , R D , and R E are each hydrogen.
  • R , R B , R D , and R E are each hydrogen and R c is a halogen, such as fluorine, chlorine, iodine, or bromine.
  • R 3 or R 5a is a moiety of Formula IHa-C.
  • R 4 , R D 5 R 6 , and R 7 are each hydrogen.
  • R 4a , R 5a , R 6a , and R 7a are each hydrogen.
  • R 3a is hydroxyl, cyano, acyl, or hydroxyl.
  • R 1 ' and A taken together are a natural or ; unnatural amino acid residue or a pharmaceutically acceptable salt or ester thereof.
  • amino acid residues include esters and salts of phenylalanine and leucine.
  • n 0, 1, or 3.
  • A is nitrogen or oxygen
  • R 11 is hydrogen, salt- forming cation, ester forming group, — (CH 2 ) X — Q 5 or when A is nitrogen, A and R 11 taken together may be the residue of a natural or unnatural 5 amino acid or a salt or ester thereof;
  • Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl;
  • x is O, 1, 2, 3, or 4;
  • n is O, 1 ,2 ,3, 4, 5, 6, 7, 8, 9, or 10;
  • R 8 , R 9 , R 10 , R ⁇ , and R 12 are independently selected from a group of hydrogen, halogen, hydroxyl, alkyl, alkoxyl, halogenated alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, cyano, thiazolyl, triazolyl, imidazolyl, tetrazolyl, benzothiazolyl, and benzoimidazolyl, and pharmaceutically acceptable salts and esters thereof.
  • R 1 ' is a salt-forming cation.
  • R 11 is an ester- forming group.
  • An ester-forming group include groups when bound form an ester. Examples of such groups include substituted or unsubstituted alkyl, aryl, alkenyl, alkynyl, or cycloalkyl.
  • A is oxygen.
  • n is 2, 3, or
  • R 4 , R 5 , R 6 and R 7 are each hydrogen.
  • R 4a , R 5a , R 6a , and R 7a also may be hydrogen.
  • Examples of R 8 , R 9 , R 10 , R 11 , and R 12 include hydrogen.
  • R 8 , R 9 , R n , R 12 are each hydrogen, and R 10 is a halogen, (e.g., fluorine, chlorine, bromine, or iodine), nitro, or alkyl (e.g., methyl, ethyl, butyl), o
  • A-R 11 maybe the residue of an amino acid, e.g., a phenyl alanine residue.
  • R 9 , R 10 , R 11 and R 12 are each hydrogen, and R 8 is not hydrogen, e.g., halogen, e.g., fluorine, bromine, chlorine, or iodine.
  • the invention pertains to compounds of Formula V-C: R 15 O
  • A is nitrogen or oxygen
  • R 11 is hydrogen, salt-forming cation, ester forming group, — (CH 2 ) X — Q, or when A is nitrogen, A and R 1 ' taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof;
  • 0 Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl;
  • x is O, 1, 2, 3, or 4;
  • n is 0, 1 ,2 ,3, 4, 5, 6, 7, 8, 9, or 10;
  • aa is a natural or unnatural amino acid residue; 5 m is 0, 1, 2, or 3;
  • R 14 is hydrogen or protecting group
  • R 15 is hydrogen, alkyl or aryl, and pharmaceutically acceptable salts and prodrugs thereof.
  • R 11 is a salt-forming cation.
  • salt forming o cations include pharmaceutically acceptable salts described herein as well as lithium, sodium, potassium, magnesium, calcium, barium, zinc, iron, and ammonium
  • R 11 is an ester-forming group.
  • An ester-forming group includes groups when bound form an ester. Examples of such groups include substituted or unsubstituted alkyl, aryl, alkenyl, alkynyl, or cycloalkyl.
  • A is 5 oxygen.
  • n is 2, 3 or 4.
  • ra is 0.
  • A-R 11 is a residue of a natural amino acid, or a sail or ester thereof. Examples of amino acid residues, include, but are not limited, to leucine or phenylalanine residues, and pharmaceutically acceptable salts and esters thereof. 5 Examples of possible esters include methyl, ethyl, and t-butyl.
  • m is 1.
  • aa include natural and unnatural amino acid residues such as phenylalanine, glycine, and leucine.
  • (aa) ra is a residue of phe-phe; and pharmaceutically acceptable salts or an appropriate protecting group, o
  • R 15 is hydrogen or substituted alkyl, e.g., arylalkyl.
  • unnatural amino acid refers to any derivative of a natural amino acid including D forms, and a- and j ⁇ -amino acid derivatives. It is noted that certain amino acids, e.g., hydroxyproline, that are classified as a non-natural amino acid herein, may be found in nature within a certain organism or a particular protein. Amino acids with s many different protecting groups appropriate for immediate use in the solid phase synthesis of peptides are commercially available.
  • non-natural amino acids and amino acid derivatives may be used according to the invention (common abbreviations in parentheses): /3-alanine (/3-ALA), ⁇ -ammobutyric acid (GABA), o 2-aminobutyi ⁇ c acid (2- Abu), ⁇ ,j8-dehydro-2-aminobutyiic acid (8-AU),
  • ACPC 1-aminocyclopropane-l-carboxylic acid
  • Amisobutyric acid Aib
  • 2-amino- lhiazoline-4-carboxylic acid 5-aminovaleric acid (5-Ava)
  • 6-aminohexanoic acid (6-Ahx)
  • 8-aminooctanoic acid (8-Aoc)
  • 11-aminoundecanoic acid 11 -Aim
  • 12-aminododecanoic acid (12-Ado
  • 4-amino-3-hydroxy-6-methylheptanoic acid (Statine, Sta)
  • aminooxyacetic acid Aoa
  • 2-aminotetraline-2-carboxylic acid ATC
  • the invention pertains, at least in part, to compounds of Formula VI-C:
  • n is i, 2, 3, 4, 5, 6, 7, 8, 9, or 10;
  • A is oxygen or nitrogen
  • R 1 ' is hydrogen, salt-forming cation, ester forming group, -(CH 2 ) X -Q, or o when A is nitrogen, A and R u taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof;
  • Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl; x is 0, 1, 2, 3, or 4; 5 R 19 is hydrogen, alkyl or aryl;
  • Y 1 is oxygen, sulfur, or nitrogen
  • Y 2 is carbon, nitrogen, or oxygen
  • R 20 is hydrogen, alkyl, amino, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, thiazolyl, triazolyl, tetrazolyl, imidazolyl, benzothiazolyl, or o benzoimidazolyl;
  • R 21 is hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, thiazolyl, triazolyl, tetrazolyl, imidazolyl, benzothiazolyl, benzoimidazolyl, or absent if Y 2 is oxygen;
  • R 22 is hydrogen, alkyl, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, 5 arylalkyl, thiazolyl, triazolyl, tetrazolyl, imidazolyl, benzothiazolyl, benzoimidazolyl; or R 22 is hydrogen, hydroxyl, alkoxy or aryloxy if Y 1 is nitrogen; or R 22 is absent if Y 1 is oxygen or sulfur;or R 22 and R 2 ' may be linked to form a cyclic moiety if Y 1 is nitrogen;
  • R is hydrogen, alkyl, amino, mercaptoalkyl, alkenyl, alkynyl, cycloalkyl, aryl, arylalkyl, thiazolyl, triazolyl, tetrazolyl, imidazolyl, benzothiazolyl, or o benzoimidazolyl, or absent if Y 2 is nitrogen or oxygen; or pharmaceutically acceptable salts thereof.
  • R 11 is a salt-forming cation.
  • salt forming cations include pharmaceutically acceptable salts described herein as well as lithium, sodium, potassium, magnesium, calcium, barium, zinc, iron, and ammonium.
  • the salt is a sodium salt.
  • A is oxygen.
  • Y 1 is oxygen or sulfur, and R 22 is absent.
  • Y 2 is oxygen and R 21 is absent.
  • R 20 include benzyl, aryl (e.g., phenyl), allcyl, cycloalkyl (e.g., adamantyl), etc.
  • Y 2 is nitrogen and R 21 is hydrogen.
  • R 2! is benzyl.
  • R 20 and R 21 are linked to form a pyridyl ring.
  • Y 1 is sulfur.
  • the invention pertains to compounds of Formula VII-C:
  • n 2, 3, or 4;
  • A is oxygen or nitrogen;
  • R 11 is hydrogen, salt-forming cation, ester forming group, — (CH 2 ) X — Q, or when
  • A is nitrogen, A and R 1 ' taken together may be the residue of a natural or unnatural amino acid or a salt or ester thereof;
  • Q is hydrogen, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, or benzoimidazolyl; x is 0, 1, 2, 3, or 4;
  • G is a direct bond or oxygen, nitrogen, or sulfur; z is O, 1, 2, 3, 4, or 5; m is 0 or 1 ;
  • R 24 is selected from a group consisting of hydrogen, alkyl, mercaptoalkyl, allcenyl, alkynyl, aroyl, alkylcarbonyl, aminoalkylcarbonyl, cycloalkyl, aryl, arylalkyl, thiazolyl, triazolyl, imidazolyl, benzothiazolyl, and benzoimidazolyl; each R 25 is independently selected from hydrogen, halogen, cyano, hydroxyl, alkoxy, thiol, amino, nitro, alkyl, aryl, carbocyclic, or heterocyclic, and pharmaceutically acceptable salts thereof.
  • R 11 is hydrogen.
  • A is oxygen.
  • n may be 3 and m may be 1.
  • R 24 is hydrogen or benzyl.
  • z is 0, 2, or 3.
  • R 25 is hydroxyl or alkoxy, e.g., methoxy, ethoxy, etc.
  • two or more R 25 substituents can be linked to form a fused ring (e.g., to form a methylendioxyphenyl moiety).
  • the invention pertains to both salt forms and acid/base forms of the compounds of the invention.
  • the invention pertains not only to the particular salt forms of compounds shown herein as salts, but also the invention includes other pharmaceutically acceptable salts, and the acid and/or base form of the compound.
  • the invention also pertains to salt forms of compounds shown herein.
  • Exemplary compounds of the invention are shown in the Figures and Tables attached hereto. Intended to be part of this invention are the exemplary compounds and selected groups and subsets thereof for any of the formulas recited herein, e.g., Formula I-C through VlI-C, provided in the two U.S. Patent Applications filed June 18, 2004, both entitled “Methods and Compositions for Treating Amyloid Related Diseases"
  • WO 00/64420, WO 97/023458 and WO 96/28187 does not pertain to methods of using the compounds described in WO 00/64420, WO 97/023458 and WO 96/28187 for the treatment of diseases or disorders described therein.
  • the invention pertains to methods of using the compounds described in WO 00/64420, WO 97/023458 and WO 96/28187 for methods described in this application, which are not described in WO 00/64420, WO 97/023458 and WO 96/28187.
  • WO 00/64420, WO 97/023458 and WO 96/28187 are incorporated by reference herein in their entirety.
  • the invention relates to the compounds in Table 2 (see attached Drawings). Additionally or alternatively the invention relates to the compounds in Table 2 A (see attached Drawings). In one embodiment the invention does not pertain to the compounds in Table 2A and/or Figures 15 through 32. In one embodiment, the invention does pertain to the compounds in Table 2A and/or Figures 15 through 32.
  • Blockers of sodium or calcium ion channel activity are well known in the art and can be used as the A moiety in the compounds and methods of the present invention.
  • any compound that opens potassium or chloride ion channels can be used as the A moiety in the compounds and methods of the present invention.
  • Antagonists of NMDA receptors and augmenters of endogenous GABA inhibition are also known to one of skill in the art and can be used in the methods and compounds of the invention.
  • 2,3-quinoxalinediones are reported to have NMDA receptor antagonistic activity ⁇ see, e.g., U.S. Pat. No. 5,721,234).
  • Exemplary calcium and zinc chelators include moieties known in the art for chelation of divalent cations, including (in addition to those mentioned supra) ethylenediaminetetraacetic acid (EDTA), ethylene glycol 5 bis(beta-aminoethyl ether)-N,N,N',./V'-tetraacetic acid, and the like.
  • EDTA ethylenediaminetetraacetic acid
  • ethylene glycol 5 bis(beta-aminoethyl ether)-N,N,N',./V'-tetraacetic acid and the like.
  • Exemplary iron chelators include enterobactin, pyridoxal isonicotinyl hydrazones, N,N'-bis(2-hydroxybenzoyl)-ethylenediamine-iV,i ⁇ r '-diacetic acid (HBED), 1-substituted- 2-alkyl-3-hydroxy-4-pyridones, including l-(2'-carboxyethyl)-2-methyl-3-hydroxy- 4-pyridone, and other moieties known in the art to chelate iron.
  • enterobactin enterobactin
  • pyridoxal isonicotinyl hydrazones N,N'-bis(2-hydroxybenzoyl)-ethylenediamine-iV,i ⁇ r '-diacetic acid (HBED)
  • HBED N,N'-bis(2-hydroxybenzoyl)-ethylenediamine-iV,i ⁇ r '-diacetic acid
  • N ⁇ -substituted arginine analogs especially of the L configuration
  • L-N7-nitro-arginine a specific inhibitor of cerebral NO synthase
  • L-N ⁇ -amino-arginine and L-N ⁇ -alkyl- argmines
  • an ester for example, the methyl ester
  • Exemplary antioxidants include ascorbic acid, tocopherols including alpha-tocopherol, and the like.
  • the invention relates to pharmaceutical compositions comprising a first agent, e.g., an alkanesulfonic acid, and another drug that targets secondary symptoms of Alzheimer's disease, such as behavioral and emotional difficulties.
  • a first agent e.g., an alkanesulfonic acid
  • another drug that targets secondary symptoms of Alzheimer's disease, such as behavioral and emotional difficulties.
  • some approved medications exist that appear to improve memory and cognition, but do not address the underlying pathology, as discussed more o fully elsewhere herein.
  • Alzheimer's Disease is associated with degeneration of cholinergic neurons in 5 the basal forebrain that play a fundamental role in cognitive functions, including memory. Alzheimer's disease patients exhibit a marked reduction in acetylcholinesterase activity and choline uptake. Becker, et al., Drug Development Research 12, 163-95 (1988). It has been observed that A ⁇ .can inhibit acetylcholine release via its effects on, for example, choline transport, post-synaptic events and o nicotinic acetylcholine receptors ⁇ c ⁇ , c&, /34) (Wang et al., J. Biol. Chem., vol. 275, pp.
  • an alkylsulfonic acid could thus normalize acetylcholine esterase (AChE) efflux levels (e.g. from the hippocampus) which are usually decreased in the presence of A ⁇ in the brain.
  • AChE acetylcholine esterase
  • AChE has been found to be associated with senile plaques and has been shown to interact with A/3, promote amyloid fibril formation and increase its toxicity (De Ferrari et al., Biochemistry 40: 10447-10457, 2001).
  • the present invention is related to increasing levels of acetylcholine by the administration of an inhibitor of choline esterase ⁇ e.g., acetylcholinesterase or butyrylcholinesterase), and a first agent alkyl sulfonic acid which acts synergistically to treat an amyloid, (e.g., amyloid-/?) disease.
  • choline esterase e.g., acetylcholinesterase or butyrylcholinesterase
  • a first agent alkyl sulfonic acid which acts synergistically to treat an amyloid, (e.g., amyloid-/?) disease.
  • Cholinergic neurons make up a major neuronal system of the central and peripheral nervous systems.
  • Cholinergic neurons produce the neurotransmitter acetylcholine.
  • acetylcholine is a neurotransmitter and is released by cholinergic neurons in, among other places, the hippocampus and frontal cortex of the brain.
  • the hippocampal area of the brain, particularly those areas where acetylcholine is released, is believed to have functions associated with cognition, learning, and memory. Degenerative diseases with symptoms such as loss of cognition, learning, and memory, have been linked to a loss in cholinergic neurons.
  • Cholinergic dysfunction characterized by marked degeneration of cholinergic innervation in the basal forebrain, and reduction of choline acetyltransferase, acetylcholinesterase, and the nicotinic and muscarinic receptors are known to be very early features of Alzheimer's disease.
  • Other neurotransmitter systems such as glutamatergic, serotonergic, and dopaminergic, are also disrupted in Alzheimer's disease, but at a later stage of the disease.
  • the present invention also relates to the combination use of a nicotinic acetylcholine receptor agonist or muscarinic agonist that results in cognition enhancement.
  • Nicotinic acetylcholine receptor agonists improve cognitive function in Alzheimer's patients. Wilson, et al., Pharmacol. Biochem. Behavior 51, 509-14 (1995); Arneric, et al., Alzheimer Disease Assoc. Disorders 9(suppl. 2), 50-61 (1995); Buccafusco, et al., Behav. Pharmacol. 10, 681-90 (1999).
  • Muscarinic and nicotinic agonists have been reported to enhance cognitive tasks in animal models and in humans. Schwarz, et al, J. Pharmacol. Experim. Theraput. 291, 812-22 (1999); Veroff, et al,
  • the number of cholinergic neurons innervating the hippocampus typically decreases, and the progressive loss of these cholinergic neurons mirrors the loss in memory and cognitive function in these subjects.
  • Acetylcholine is synthesized by choline acetyltransferase ("ChAT"). Once released by the neuron, it is degraded by cholinesterases, e.g., acetylcholinesterase ("AChE").
  • choline esterase inhibitors are known. Certain cholinesterase inhibitors are approved for use in treatments for improving memory and learning in Alzheimer's subjects.
  • Tacrine (CognexTM, Warner-Lambert Co., now Pfizer, New York, New York) was the first approved cholinesterase inhibitor but is rarely used because of negative side effects like stomach and liver problems.
  • Donepezil (AriceptTM, Eisai Co, Ltd) is more selective for acetylcholinesterase and shows fewer side effects than tacrine.
  • Rivastigmine targets a specific subtype of acetylcholinesterase that is present at high concentrations in the brains of Alzheimer's subjects.
  • Galanthamine (ReminylTM, Janssen Pharmaceutica Products, LP) has a dual mode of action in the brain; in addition to working as an acetylcholinesterase inhibitor, galanthamine also appears to exert action on the nicotinic acetylcholine receptors in the brain.
  • These cholinesterase inhibitors may be acetyl-cholinesterase or butyryl- cholinesterase inhibitors or both. Another example is phenserine (currently in advanced clinical trials in the United States).
  • phenserine may inhibit ⁇ -APP production by a separate and distinct mechanism of action at the level of the mRNA level.
  • AIT-082 also in advanced clinical trials.
  • the degradation pathway of AchE may also be inhibited by inhibitors such as physostigmine (SynaptonTM, or (Antilirium InjectableTM, Forrest Laboratories, New York, New York)), quilostigmine, tolserine, thiatolserine, cymserine, thiacymserine, neostigmine, eseroline, zifrosilone, mestinon, huperzine A and icopezil. Phenserine, an acetylcholinesterase inhibitor, is in development (Axonyx, New
  • Phenserine which has been shown to increase memory and learning in the laboratory animals, works through two mechanisms: it inhibits the degradation of the neurotransmitter acetylcholine in the brains of animals, and it inhibits the production of a toxic form of the ⁇ -amyloid protein in the brain that is thought to be a cause of the death of brain cells in Alzheimer's disease. Unlike other acetylcholinesterase inhibitors that simply suppress the activity of the enzyme, Phenserine's dual mechanism of action suggests that it not only has the potential to improve memory and cognition but also to slow the progression of the disease.
  • Phenserine Compared to currently marketed drugs for Alzheimer's, Phenserine is more brain-targeted versus the rest of the body and is more rapidly cleared from the blood. In preclinical studies, Phenserine demonstrated a brain-to-blood ratio of 10:1. These properties of Phenserine could potentially maximize the therapeutic effects of the drug in the brain and reduce side effects by clearing the drug from the blood quickly. Since undesirable side effects and drug interactions often arise due to the presence of drugs in the body for an extended period, Phenserine's rapid disappearance from the blood suggests that it will represent a more tolerable treatment option to existing therapies.
  • Phenserine is rapidly cleared from the body, the drug remains bound to the acetylcholinesterase enzyme in the brain allowing it to have a long duration of therapeutic action.
  • Substituted phenserines and phenylcarbamates of eseroline, noreseroline, and benzylnoreseroline are also specific inhibitors of acetylcholinesterase See, e.g., U.S. Pat. Nos. 5,171,750; 5,378,723; 5,409,948; 5,998,460; 5,948,763; 6,410,747; 6,462,171; and 6,495,700; as well as WO 93/06105.
  • Suitable cholinesterase inhibitors include galanthamine derivatives available from Janssen, metrifonate available from Bayer Corp., ipidacrine available from Nikken Chemicals Co. Ltd., TAK-147, T-82 available from SS Pharmaceutical Co. Ltd., methanesulfonyl fluoride, CHF-2819, phenserine, physostigmine available from Forest Laboratories, Inc., huperzine, cymserine available from Axonyx Inc., tolserine available from National Institutes of Health, ER-127528 available from Eisai Co. Ltd., and combinations thereof.
  • the present invention relates to a method for maintaining or preventing a decrease in the levels of acetylcholine in the frontal cortex or hippocampus regions of the brain in mammals comprising administering to a mammal in need thereof, an effective amount of a first agent, e.g., an alkanesulfonic acid or a pharmaceutically acceptable salt thereof, and optionally a choline esterase inhibitor.
  • a first agent e.g., an alkanesulfonic acid or a pharmaceutically acceptable salt thereof, and optionally a choline esterase inhibitor.
  • the present invention relates to a method for inhibiting conditions or detrimental effects caused by a deficiency of choline acetyltransferase or acetylcholine in the frontal cortex or hippocampus regions of the brain in mammals comprising administering to a mammal in need thereof, an effective amount of a first agent, e.g., an alkanesulfonic acid, or a pharmaceutically acceptable salt thereof, and optionally a choline esterase inhibitor.
  • a first agent e.g., an alkanesulfonic acid, or a pharmaceutically acceptable salt thereof, and optionally a choline esterase inhibitor.
  • the present invention relates to a pharmaceutical formulation 5 comprising a first agent, e.g., an alkanesulfonic acid or a pharmaceutically acceptable salt thereof, and optionally a choline esterase inhibitor; and a pharmaceutical carrier, diluent, or excipient.
  • a first agent e.g., an alkanesulfonic acid or a pharmaceutically acceptable salt thereof, and optionally a choline esterase inhibitor
  • a pharmaceutical carrier diluent, or excipient.
  • Another embodiment of the present invention is where the condition caused by a decrease of choline acetyltransferase or acetylcholine in the frontal cortex or o hippocampus regions of the brain is Alzheimer's disease.
  • the term "effective amount” means an amount of a first agent, e.g., an alkanesulfonic acid, that is capable of maintaining brain cell ability to produce stable levels of acetylcholine in the brain, such as in the hippocampus and frontal cortex regions, or inhibiting conditions or detrimental effects caused by a decrease of 5 acetylcholine in mammals.
  • a first agent e.g., an alkanesulfonic acid
  • an alkanesulfonic acid or other such first agent is co-administered with an AChE inhibitor the term "effective amount” also means an amount of such an agent capable of inhibiting AChE.
  • an inhibitor of AChE may be represented as "AChEi.”
  • the term “inhibiting” in the context of inhibiting conditions or o detrimental effects caused by a deficiency of CIiAT or acetylcholine in the frontal cortex or hippocampus regions of the brain includes its generally accepted meaning, i.e., prohibiting, restraining, alleviating, ameliorating, slowing, stopping, or reversing the progression or severity of a decrease in ChAT and acetylcholine and the pathological sequelae, i.e., symptoms, resulting from that event.
  • up-regulating ChAT refers to increasing the enzymatic activity of
  • ChAT i.e., promoting the conversion of choline to acetylcholine. This promotion would include an increase in the efficiency or rate of reaction of ChAT and choline or an increase in the amount of ChAT present at the site of action. This increase in the amount of enzyme present may be due to gene regulation or another synthetic step in the o enzyme's formation or a decrease in the enzyme's de-activation and metabolism.
  • a ⁇ can inhibit the efflux of acetylcholine from neurons upon new stimulation, and in addition that exogenous A/3 may inhibit high affinity choline uptake.
  • acetylcholine efflux levels e.g. from hippocampus
  • A/3 may act in several different ways to 5 exert these effects, such as acting at the choline transporter, modulating post synaptic events, or acting on neuronal acetyl cholinesterase receptors (e.g., nAChr ( ⁇ 7, o2/34).
  • a first agent of the invention e.g., an alkanesulfonic acid, may act similarly to normalize acetylcholine levels by binding to A ⁇ .
  • an alkanesulfonic acid may thus prevent A ⁇ from inhibiting the efflux of acetylcholine, thereby leading to an increase in the amount of acetylcholine at the synapse. It is likely therefore that an alkanesulfonic acid and an acetylcholinesterase inhibitor will act synergistically to ameliorate cholinergic neurotransmission, as both agents act to potentiate the levels of acetylcholine.
  • Ester Neurosciences Herzlia Pituach, Israel
  • antisense drug (ENlOl) for the treatment of myasthenia gravis, demonstrated for the first time effective and safe use of an orally-administered antisense therapy for a neurological condition that lessened the severity of symptoms of myasthenia gravis , with no cholinergic symptoms nor significant adverse events based on balancing cholinergic transmission via controlled modulation of the company's novel target, a stress-response variant of acetylcholinesterase .
  • AChE is an enzyme that degrades the neurotransmitter acetylcholine.
  • ENlOl selectively inhibits the production of the target at the critical stage of its biosynthesis, thereby allowing an effective treatment, while minimizing side effects and substantially improving upon the short-duration palliative relief currently observed with conventional inhibitors.
  • ENlOl is the lead compound in Ester's disease- modifying platform technology for the pre-expression control of a specific variant of the AChE protein, which is applicable to a wide range of neurological disorders.
  • Useful muscarinic receptor agonists include cevimeline, PD-151832 available from Pfizer Inc., YM-796 available from Yamanouchi Pharmaceutical Inc., and P-58 available from Phytopharm pic.
  • Suitable acetylcholine release stimulators include minaprine, and montirelin available from Grunenthal GmbH, T-588 available from Toyama Chemical Co. Ltd., XE-991.
  • Useful choline uptake stimulators include MKC-
  • Suitable nicotinic cholinergic receptor agonists include altinicline available from SEBIA Neurosciences, Inc., SB3-1553A, ABT-089 (U.S. Pat. No. 5,278,176, Abbott Laboratories), nicotine patch, GRS-21, and TC-2403.
  • tacrine became the first agent approved specifically for the treatment of cognitive symptoms of Alzheimer's disease.
  • Tacrine is a reversible cholinesterase inhibitor and is thought to work by increasing the availability of intrasynaptic acetylcholine in the brains of Alzheimer's disease patients. The medication may also have other actions.
  • Donepezil another reversible cholinesterase inhibitor, is now available for the treatment of Alzheimer's disease.
  • Another example of a second agent 5 is xanomeline, which is a muscarinic selective ml and m4 (muscarinic) acetylcholine receptor agonist and shows moderate improvement in cognitive performance, greater efficacy in decreasing psychotic symptoms, and agitation.
  • N.C.Bodick, et al. "Effects of xanomeline, a selective muscarinic receptor agonist, on cognitive function and behavioral symptoms in Alzheimer disease.” Arch. Neurol. 54, 465-73 (1997).
  • the o second agent may also be an ergot alkaloid or a vinca alkaloid, such as HydergineTM (Sandoz Pharmaceutical Corp., now Novartis, Basel, Switzerland) and nicergolin; or it may be a nootropic, such as piracetam, oxiracetam, pramiracetam, and aniracetam; which have cholinergic and dopaminergic properties as well as effects on protein processing.
  • HydergineTM Sandoz Pharmaceutical Corp., now Novartis, Basel, Switzerland
  • nicergolin or it may be a nootropic, such as piracetam, oxiracetam, pramiracetam, and aniracetam; which have cholinergic and dopaminergic properties as well as effects on protein processing.
  • B.Saletu, et al. "Nicergoline in senile dementia of Alzheimer type and s multi-infarct dementia: a double-blind, placebo-controlled, clinical and EEG/ERP mapping
  • the second agent maybe a carbamate derivative of physostigmine, such as eptastigmine, which is an inhibitor of acetylcholinesterase.
  • physostigmine such as eptastigmine
  • NMDA JV-methyl- 5 D-Aspartate
  • An increased level of one or more glutamate-related compounds is associated with many neurodegenerative disorders and neurodegeneration associated with long term disease states such as Huntington's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS, which is also known as motor neuron disease), Parkinson's disease and acquired immunodeficiency (AIDS).
  • Excitatory amino o acid receptor antagonists that block NMDA receptors are recognized for usefulness in the treatment of disorders.
  • NMDA receptors are intimately involved in the phenomenon of excitotoxicity, which maybe a critical determinant of outcome of several neurological disorders.
  • Disorders known to be responsive to blockade of the NMDA receptor include acute cerebral ischemia (stroke or cerebral trauma, for example), muscular spasm, 5 convulsive disorders, neuropathic pain and anxiety, and may be a significant causal factor in chronic neurodegenerative disorders such as Parkinson's disease, amyotrophic lateral sclerosis (ALS), Alzheimer's disease and Huntington's disease.
  • NMDA receptor-mediated injury e.g., that injury resulting from stimulation of the NMDA receptor by glutamate or other excitatory 5 amino acids or structurally similar compounds may be used in one embodiment of the invention.
  • NMDA receptor antagonists Some examples of NMDA receptor antagonists are known and commercially available. Memantine (EbixaTM or AxuraTM or NamendaTM recently available in the U.S. from Merz Pharmaceuticals, Frankfurt am Main, Germany), which operates by yet o another mechanism, appears to prevent or reduce the brain damage caused by Alzheimer's disease by blocking NMDA receptors in the brain. See U.S. Pat. No. 5,614,560. Memantine (l-amino-3,5-dimethyl adamantine) reduces neuronal damage by blocking NMDA receptor-operated channel activation by excitatory amino acids (such as glutamate-related compounds) at concentrations that are readily 5 obtainable in human subjects taking the drug (Wesemann, et ah, J.
  • excitatory amino acids such as glutamate-related compounds
  • Neural Transmission (Supp.) 16, 143 (1980)). Others include ethopropazine (ParidolTM), diphenhydramine (BenadrylTM), dizocilpine (NeurogaurdTM), and amantadine (SymmetrelTM).
  • NMDA antagonists have NMDA antagonist activity without causing hypofunction because of activity at some other type of receptor.
  • NMDA antagonists such as PCP and MK-801, because of activity at the additional neuronal receptors.
  • Such drugs, and the receptors they interact with in addition to NMDA receptors include the following: Ibogaine, which also suppresses excitatory activity at sigma receptors and which may also be active at serotonin 5 receptors, and Eliprodil, which also increases inhibitory activity at sigma receptors; Certain anti-cholinergic drugs such as procyclidine, trihexyphenydyl, and biperiden, which also suppress excitatory activity at muscarinic acetylcholine receptors; and Certain quinoxalinediones, including NBQX, ACEA 1021, and ACEA 1031, which are discussed below, and which suppress activity at non-NMDA receptors ⁇ i.e., kainic acid o receptors and AMPA receptors), in addition to NMDA receptors: As described in
  • NMDA antagonists 5 offer good candidates both for treating Alzheimer's disease, and for additional developmental research to identify analogs having adjusted balances in their dual or multiple receptor binding affinities.
  • the antiparkinsonian agents procyclidine, trihexyphenydyl, and biperiden all have affinities for muscarinic receptors that are several times higher than for NMDA receptors.
  • Further examples of NMDA receptor antagonists include those in U.S. Pat.
  • No. 4,906,779 which discloses disubstituted guanidines, e.g., N,N '-di-m-tolyl guanidine, N ⁇ V'-di-o-ethylphenyl 5 guanidine, iV,iV " '-di-m-ethylphenyl guanidine, and ⁇ N'-di-o-iodophenyl-guanidine; U.S. Pat. No. 5,498,610, which discloses 5-(l-hydroxy-2-piperidino)-propyl- 2(lH,3H)-indolone analogs.
  • a muscarinic agonist may also be used in this invention.
  • a class of styryl amidine derivatives which are antagonists of the human NMDA receptor, are selective for those containing the NR2B subunit, and may, in some embodiments, be o used in the invention.
  • Suitable NMDA receptor antagonists also include ipenoxazone, which is available from Nippon Chemiphar Co. Ltd.
  • agonist refers to a molecule which, when interacting with a biologically active molecule, causes a change (e.g., enhancement) in the 5 biologically active molecule, or which positively modulates the activity of the biologically active molecule.
  • An agonist interacts with a receptor and initiates a physiological or a pharmacological response characteristic of that receptor.
  • agonists include, but are not limited to proteins, nucleic acids, carbohydrates, lipids or any other molecules which bind or interact with biologically active molecules.
  • Antagonist refers to a molecule which, when interacting with a biologically active molecule, blocks or negatively modulates the biological activity of the biologically active molecule. Antagonists oppose the receptor- associated responses normally induced by other bioactive agents (i.e., agonists). Antagonists and inhibitors include, but are not limited to, proteins, nucleic acids, 5 carbohydrates, lipids or any other molecules that bind or interact with biologically active molecules. Inhibitors and antagonists may effect the biology of entire cells, organs, or organisms (e.g., an inhibitor that slows or prevents neuronal degeneration and death).
  • Estrogen replacement decreases the risk of Alzheimer's disease in postmenopausal women, delays the age of onset, and perhaps slows the decline.
  • Estrogenic agent include estrogen, lasofoxifene, droloxifene, tamoxifen, and raloxifene (EvistaTM, Eli Lilly, Indianapolis, Indiana).
  • compositions or therapeutic combinations that further comprise hormone replacement agents and compositions.
  • Useful hormone agents and compositions include androgens, estrogens, progestins, their pharmaceutically acceptable salts and derivatives. Combinations of these agents and compositions also are useful.
  • estrogens include, but are not limited to, androgen and estrogen combinations such as the combination of esterified estrogens (sodium estrone sulfate and sodium equilin sulfate) and methyltestosterone available from Solvay Pharmaceuticals, Inc., Marietta, GA., as EstratestTM; the blend of nine synthetic estrogenic substances including sodium estrone sulfate, sodium equilin sulfate, sodium 17 ⁇ -dihydroequilin sulfate, sodium 17ce-estradiol sulfate, sodium 17/3-dihydroequilin sulfate, sodium 17 ⁇ -dihydroequilenin sulfate, sodium 17/3-dihydroequilenin sulfate, sodium equilenin sulfate and sodium 17/?-estradiol sulfate; available from Duramed Pharmaceuticals, Inc., Cincinnati, Ohio, as CenestinTM; ethinyl
  • EstinylTM esterified estrogen combinations such as sodium estrone sulfate and sodium equilin sulfate; available from Solvay as EstratabTM and from Monarch Pharmaceuticals, Bristol, Tenn., as MenestTM; estropipate, available from Pharmacia & Upjohn, Peapack, NJ., as OgenTM and from Women First Health Care, Inc., San Diego, Calif., as Ortho-EstTM; and conjugated estrogens (17 ⁇ - dihydroequilin, 17 ⁇ -estradiol, and 17/3-dihydroequilin), available from Wyeth, Philadelphia, Pa., as PremarinTM.
  • Another estrogen example is disclosed in U.S. Pat. No. 6,610,706.
  • Progestins and estrogens may also be administered as combinations including estradiol and norethindrone, available from Pharmacia & Upjohn, Peapack, NJ., as
  • AygestinTM from Ortho-McNeil under the tradename MicronorTM, and from Watson as NOR-QDTM; norgestrel; available from Wyeth as OvretteTM; micronized progesterone, from Solvay as PrometriurnTM; and medroxyprogesterone acetate; available from Pharmacia & Upjohn under the tradename Pro veraTM.
  • Nonsteroidal anti-inflammatory drugs appear to be associated with a lower likelihood of developing Alzheimer's disease. Anti-inflammatory drugs are believed to interfere with aspects of the microglial, astrocytic, and cytokine responses that occur in Alzheimer's disease.
  • NSAIDs including ibuprofen, naproxen, sulindac, o and indomethacin, have been shown to be selective A ⁇ 42-lowering agents. A subset of NSAIDs lower amyloidogenic A ⁇ 42 independently of cyclooxygenase activity. S.Weggen, et at, "A subset of NSAIDs lower amyloidogenic A ⁇ 42 independently of cyclooxygenase activity.” Nature 414, 212-16 (2001).
  • NSAIDs lower A ⁇ 42 have not been established, the effect is independent of cyclooxygenase inhibition, which is the primary anti-inflammatory target of these compounds.
  • NSADDs do not appear to change the total level of A ⁇ produced but shift cleavage from A ⁇ 42 to a less toxic shorter 38-amino acid A ⁇ peptide (A ⁇ 38), which suggests that they interact with ⁇ -secretase.
  • One class of developmental compounds are inhibitors of PDE4, which act as anti-inflammatory drug in mice. These anti- inflamatory agent, e.g., rolipram, appear to block the microglial inflammatory response, and may have toxic side effects, but newer analogs without such properties are in development.
  • Memory Pharmaceutical's MEM 1414 is a PDE4 inhibitor currently in testing for Alzheimer's disease.
  • Suitable anti-inflammatory agents include COX-2 inhibitors (such as VioxxTM and CelebrexTM), cytokine inhibitors (such as thalidomide disclosed in WO 95/04533 and dexanabinol) complement inhibitors, leukotriene receptor antagonists and combinations thereof.
  • COX-2 inhibitors such as VioxxTM and CelebrexTM
  • cytokine inhibitors such as thalidomide disclosed in WO 95/04533 and dexanabinol
  • complement inhibitors such as leukotriene receptor antagonists and combinations thereof.
  • Examples include acetic acid aerivatives sulindac (ClinorilTM, Merck & Co., Inc., Rahway, New Jersey), indomethacin (IndocinTM, Merck & Co., Inc., Rahway, New Jersey); etodolac (LodineTM, Wyeth, Madison, New Jersey), nabumetone (RelafenTM, GlaxoSmithKline, Middlesex, England), tolmetin sodium (TolectinTM, McNeil Pharmaceuticals, Spring House, Pennsylvania); anthranilic acid derivatives: meclofenamate sodium (MeclomenTM, Pfizer, New York, New York), mefenamic acid (PonstelTM, Pfizer, New York, New York); enolic acid derivatives: piroxicam (FeldeneTM, Pfizer, New York, New York), mobic (meloxicam); phenylacetic acid derivatives: arthrotec (diclofenac/misoprosto
  • Cyclosporins a class of drugs best known as immunosuppressants, were discovered to have a new use as the most effective neuroprotectants across the spectrum of neurological disease models when they cross the blood-brain barrier. Cyclosporins protect the brain's mitochondria and prevent neuron death due to traumatic brain and spinal cord injury, stroke, Alzheimer's, 5 Parkinson's, Huntington's diseases and amyotrophic lateral sclerosis (ALS) animal models.
  • ALS amyotrophic lateral sclerosis
  • the CNS As a lipid rich organ, the CNS is particularly susceptible to effects of lipid peroxidation in modulating cellular signaling pathways, cell dysfunction, and cell death o in the nervous system.
  • lipid peroxidation in modulating cellular signaling pathways, cell dysfunction, and cell death o in the nervous system.
  • oxidative stress In Alzheimer's disease, emerging evidence provides strong support for a role for oxidative stress in neurodegeneration, as multiple indices of oxidative stress have been observed, including protein oxidation, decreased polyunsaturated fatty acids, mitochondrial and nuclear DNA damage.
  • Free radicals ⁇ e.g., superoxide radicals
  • phagocytes are used by phagocytes to kill bacteria 5 and to oxidatively destroy foreign matter. Ordinarily excess superoxide is quenched by superoxide dismutase, however if oxidative stress causes the overproduction of radicals, or if the production of the superoxide exceeds the capacity of superoxide dismutase, then unintended oxidative damage may occur.
  • B. Halliwell Acta Neurol. Scand. 126, 23-33 (1989).
  • protein oxidation, DNA oxidation, and lipid o peroxidation are greater than in age-matched controls.
  • S.S. Pitchumoni et al, N. Engl. J. Med. 46(12), 1566-72 (1998).
  • antioxidant is any substance capable of protecting against the damages of oxidative stress caused by reactive oxygen species such as free radicals. Antioxidants are generally desined so that they may be oxidized over other materials. In addition to 5 superoxide dismutase, catalase and glutathione peroxidase react with hydrogen peroxide and convert it to water and diatomic oxygen. Other antioxidants include vitamin E ( ⁇ -tocopherol), vitamin C (ascorbic acid), vitamin A (retinoic acid), co-enzyme Q, and selegiline.
  • Vitamin E and its derivitatives e.g., ⁇ -tocopherol quenches a free radical by o donating a hydrogen atom thereby producing a tocopheroxy radical, which scavenges yet another peroxyl radical to produce ⁇ -tocopherol quinone, a stable compound.
  • vitamin E is lipophilic and therefore soluble in the central nervous system and able to localize in a cell membrane thus preventing lipid peroxidation.
  • Suitable Vitamin E derivatives include but are not limited to ⁇ -tocopherol , ⁇ -tocopherol , ⁇ -tocopheraol, ⁇ -tocopheral, ⁇ -tocopherol, ⁇ 3 -tocopherol, ⁇ 2 -tocopherol and ⁇ -tocopherol, and pharmaceutically acceptable ester derivatives thereof, e.g., the corresponding acetate, succinate and nicotinate forms.
  • Additional antioxidants include citric acid in its various forms, including its administration as a combination of potassium citrate monohydrate and citric acid monohydrate.
  • agents that supplement the chain-breaking antioxidant property of Vitamin E can be co-adminstered, e.g., ubiquinol, seleno-amino acids and sulfhydryl compounds (e.g., glutathione, sulfhydryl proteins, cysteine and methionine).
  • ubiquinol e.g., seleno-amino acids
  • sulfhydryl compounds e.g., glutathione, sulfhydryl proteins, cysteine and methionine.
  • second or futher agents are butylated hydroxytoluene, butylated hydroxyanisole, propyl gallate, dodecylgallate, tert-butylhydroquinone, dihydrolipoic acid, prostaglandin B 1 oligomers (also known as polymeric 15-keto prostaglandin B or PGB x ), 2-aminomethyl-4-tert-butyl-6-iodophenol, 2-aminomethyl-4-tert-butyl-6- propionylphenol, 2,6-di-tert-butyl-4-[2'-thenoyl]phenol, N,N'-diphenyl-p- phenylenediarm ' ne, ethoxyquin, probucol, ebselen, 5-[[3,5-bis(l,l-dimethylethyl)-4- hydroxyphen-yl]methylene]-3-(dimethylamino )-4-thiazo
  • Additional antioxidants and free radical trapping substances that can be coadministered in accordance with the invention include plant (e.g., vegetable) active ingredients.
  • This category includes dimethyl sulfoxide, parthenolide, lycopene, daidzin, genistein, quercetin, morin, curcumin, apigenin, sesamol, chlorogenic acid, fisetin, ellagic acid, quillaia saponin, capsaicin, ginsenoside, silymarin, kaempferol, ginkgetin, bilobetin, isoginkgetin, isorhamnetin, herbimycin, rutin, bromelain, levendustin A, and erbstatin.
  • Vitamin E lacks negative medication interaction and may be used in combination with other Alzheimer's disease therapies.
  • C.Behl, et al "Vitamin E protects nerve cells from beta-amyloid protein toxicity.” Biochem. Biophys. Res. Commun. 186, 944-50 (1992); M.Sano, et al, "Rationale and design of a multicenter study of selegiline and a-tocopherol in the treatment of Alzheimer disease using novel clinical outcomes.” Alzheimer Dis. Assoc. Disord. 10, 132-40 (1996); H.Kappus, et al, "Tolerance and safety of vitamin E: a toxicological position report.” Free Radic. Biol. Med. 13, 55-74 (1992)).
  • Selegiline inhibits monoamine oxidase, which may convert certain protoxins into toxins. L.S. Schneider, J. CHn. Psychiatry 57, 30-36 (1996). Selegiline and other monoamine oxidase type B inhibitors may protect neurons from oxidative damage while not interfering with the action of type A inhibitors, which metabolize serotonin and norepinephrine. Selegiline also inhibits oxidative deamination of dopamine, which prevents the formation of free radicals and subsequent neuronal damage. M. Sano, et al, Alzheimer Dis. Assoc. Disord. 10, 132-140 (1996). Selegiline, through its anti- oxidative and neuroprotective properties may slow progression of Alzheimer's disease. Selegiline effect on catecholamine metabolism may also contribute to the efficacy of selegiline in delaying the progression of Alzheimer's disease in patients with moderate impairment.
  • antioxidants include free radical scavengers (Egb-761, yuyu Industrial, CP 1-21, dexanabinol and iron chelators, which prevent iron from reacting to form hydroxy! radicals.
  • Desferoxamine prevents radical damage in vivo, and clinical trial shows that it may slow the progression of Alzheimer's disease.
  • HCT- 1026 NO-flurbiprofen
  • NicOx SA Sophia Antipolis, France
  • compositions or therapeutic combinations that further comprise at least one (one or more) activators for peroxisome proliferator-activated receptors (“PPAR”).
  • the activators act as agonists for the peroxisome proliferator-activated receptors.
  • Three subtypes of PPAR have been identified, and these are designated as peroxisome proliferator-activated receptor alpha (“PP ARa”), peroxisome proliferator-activated receptor gamma (“PPAR ⁇ ”) and peroxisome proliferator-activated receptor delta (“PPAR ⁇ ,” which is also known as 'TPAR ⁇ " or "NUCl”).
  • PPARs are usually activated by fatty acids and similar derivatives.
  • PPAR ⁇ has been identified as being useful in increasing high density lipoprotein (HDL) levels in humans. See, e.g., WO 97/28149, which describes PPAR agonists that are 5 useful for raising high density lipoprotein (HDL) plasma levels in mammals.
  • PP ARa activator compounds are useful for, among other things, lowering triglycerides, moderately lowering LDL levels and increasing HDL levels.
  • Useful examples of PP ARa activators include fibrates.
  • PPAR ⁇ In contrast to PP ARa, the function of PPAR ⁇ is not well understood. Although o PPAR ⁇ is ubiquitously expressed the brain, adipose tissue and skin have higher levels of relative rnRNA expression (J.M. Peters, et al.,Mol Cell. Biol. 20, 5119-28 (2000)). The expression profile of PPAR ⁇ suggests that it may be involved in brain functions. G. Xing, et al., Biochem. Biophys. Res. Commun. 217, 1015-25 (1995). Furthermore, PPAR ⁇ may be implicated in reverse cholesterol transport, W.R. Oliver, et al., Proc. 5 Natl Acad. Sci. USA 98, 5306-11 (2001).
  • PPAR ⁇ agonists examples include valproic Acid (Lampen, etal, Tox. Appl. Pharmacol. 160, 238-49 (1999)), GW501516 (W.R. Oliver, et al, Proc. Nat'lAcad. ScL USA 98, 5306-11 (2001)), L-165041, L-165461, L-783483, and L-796449 (Berger, et al., J. Biol. Chem. 214, 6718-25 (1999)).
  • Routine experimentation may be performed to determine if a composition affects o the release of A ⁇ from at least one cell in vivo.
  • a suitable assay involves SM-4 cells, which are stably transfected with Swedish mutant amyloid Precursor Protein, and then treated with a PP ARa or PPAR ⁇ agonist, such as pirinixic acid, or derivative thereof. After treatment, the media is collected and assayed for A ⁇ 40 or A ⁇ 42 .
  • a statistically significant decrease (p ⁇ 0.05) in A ⁇ 40 or A ⁇ 42 concentration in the media compared to 5 appropriate control(s) indicates that the treatment inhibited or prevented A ⁇ 4 o or A ⁇ 42 production or release from the cells. If a compound decreases A ⁇ 42 production or release by a statistically significant amount relative to control (absence of the compound or presence of vehicle) it is considered to be an A ⁇ 42 -modulating agent according to the invention.
  • An exemplary PPAR agonist is pirinixic acid, which has been shown to induce a 5 decrease in A ⁇ 42 production or release from SM-4 cells in a concentration-dependent manner.
  • Pirinixic acid has been identified as a hypolipidemic agent, see, U.S. Pat. No. 3,814,761, which characterized it and related compounds as anti-lipidemic agents.
  • Fibrates are known to act as cholesterol-lowering agents but they generally are not known to reduce A ⁇ 42 production or release. For example, it has been reported that when SM-4 cells were treated with clofibrate and the culture media was collected in order to assay A ⁇ 42 levels, clofibrate 5 was found to increase A ⁇ 42 extracellular levels at a concentration range of 50-500 ⁇ M. Similar results were found with 5,8,11,14-eicosatetraynoic acid ("ETYA”) at 20-50 ⁇ M concentrations.
  • EYA 5,8,11,14-eicosatetraynoic acid
  • Patent Application Publication No. 2003/0013699 which describes novel heterocycles designed to prevent, treat, or ameliorate symptoms of Alzheimer's Disease, regulating production or levels of amyloid- ⁇ peptides in the bloodstream or brain.
  • fibric acid derivatives include 5 clofibrate (such as ethyl 2-(p-chlorophenoxy)-2-methylpropionate, for example
  • Atromid-STM capsules which are commercially available from Wyeth, Madison, New Jersey
  • gemfibrozil such as 5-(2,5-dimethylphenoxy)-2,2-dimethylpentanoic acid, for example, LopidTM tablets, which are commercially available from Pfizer, New York, New York
  • ciprofibrate C.A.S. Registry No. 52214-84-3, see, U.S. Pat. No. 3,948,973, o which describes the synthesis of such halocyclopropyl substitutedphenoxyalkanoic acids and esters
  • bezafibrate C.A.S. Registry No. 41859-67-0, see, U.S. Pat. No.
  • fenofibrate such as TricorTM micronized fenofibrate (2-[4-(4-chlorobenzoyl)-phenoxy]-2-methylpropanoic acid, 1-methylethyl ester), which is available from Abbott Laboratories, Abbott Park, Illinois, or LipanthylTM micronized fenofibrate, available from Laboratoire Founier, Chenove, France).
  • PP ARa activators include suitable fluorophenyl compounds as disclosed in U.S. Pat. No. 6,028,109, which describes the use of agnosts of PP ARa for the manufacture of a medicament for the treatment of obesity and the methods of treating obesity; certain substituted phenylpropionic compounds as disclosed in WO 00/75103, which describes novel substituted phenylpropionic acid derivatives capable of binding as a ligand to PP ARa to thereby activate the receptor and thus show a potent effect of lowering blood lipid; and PP ARa activator compounds as disclosed in WO 98/43081, which describes methods and compositions for treating a host having a gastrointestinal disease by administering to the host a composition containing a pharmaceutically effective amount of a modulator of a PPAR.
  • Non-limiting examples of suitable PPAR ⁇ activators include derivatives of glitazones or thiazolidinediones, such as, troglitazone (such as RezulinTM 5[[4-[(3,4-dihydro-6-hydroxy-2,5,7,8-tetramethyl- 2H-l-benzopyran-2-yl) methoxy]phenyl]methyl]-2,4-thiazolidinedione) commercially available from Pfizer, New York, New York); rosiglitazone (such as AvandiaTM rosiglitazone maleate 5-[[4-[2-(methyl-2-pyridinylamino)ethoxy]phenyl]methyl]-2,4- thiazolidinedione, (Z)-2-butenedioate) available from GlaxoSmithKline, Middlesex, England) and pioglitazone (such as ActosTM pioglitazone hydrochloride (5 - [ [4- [2-(
  • thiazolidinediones include ciglitazone, englitazone, darglitazone and BRL 49653, see, WO 98/05331, which relates such compounds for the prevention and treatment of type 2 diabetes and cardiovascular disease; PPAR ⁇ activator compounds disclosed in WO 00/76488, which describes methods for delaying or preventing the onset of Type 1 diabetes; and PPAR ⁇ activator compounds disclosed in U.S. Pat. No.-5,994,554, which describes a method for determining whether a compound does or does not interact directly with PPAR ⁇ using radiolabeling.
  • PPAR ⁇ activator compounds include certain acetylphenols, see, e.g., U.S. Pat. No. 5,859,051, which describes acetylphenol and its analogues for use as antiobesity and antidiabetic compounds; certain quinoline phenyl compounds as disclosed in WO 99/20275, which describes the use of such compounds for mediating the activity of PPAR receptors with such compounds; aryl compounds as disclosed by WO 99/38845, which describes such compounds for use as PPAR ⁇ modulators for the treatment for conditions such as type 2 diabetes and obesity; certain 1,4-disubstituted phenyl compounds as disclosed in WO 00/63161, which describes such compounds as highly selective agonists for the PPAR receptor or prodrugs of agonists for the PPAR ⁇ receptor, and therefore useful in the treatment of type 2 diabetes; certain aryl compounds as disclosed in WO 01/00579, which describes such compounds as modulators of PPAR ⁇ activity which are useful in pharmaceutical composition
  • PPAR ⁇ compounds are useful for, among other things, lowering triglyceride levels or raising HDL levels.
  • PPAR ⁇ activators include suitable thiazole and oxazole derivatives, such as C.A.S. Registry No.
  • WO 01/00603 which describes the use of pamoic acid or one of its derivatives for 5 the preparation of a medicament for the treatment of diseases characterized by deposits of amyloid aggregates; suitable non- ⁇ -oxidizable fatty acid analogues); certain fluoro, chloro or thio phenoxy phenylacetic acids; see, e.g., WO 97/28149, which describes such compounds as useful for raising high density lipoprotein (HDL) plasma levels in mammals and for preventing, halting or slowing the progresssion of atherosclerotic o cardiovascular diseases and related conditions; see, e.g., U.S. Pat. No.
  • HDL high density lipoprotein
  • Non-limiting examples include certain substituted aryl compounds as disclosed in U.S. Pat. No.-6,248,781, which describes the ability of such compounds in o the treatment or prevention of conditions mediated by nuclear receptors, in particular
  • PPAR PPAR
  • WO 00/23416 WO 00/23415; WO 00/23425; WO 00/23445; WO 00/23451; and WO 00/63153, all of which describe compounds that may be utilized in the treatment of conditions mediated by PP ARa or PPAR ⁇ activator compounds, such as diabetes and obesity.
  • PP ARa or PPAR ⁇ activator compounds 5 include activator compounds as disclosed in WO 97/25042, which describes the use of a pharmaceutically effective amount of an agonist of PP ARa and PPAR ⁇ for the treatment or prophylaxis of Syndrome X; activator compounds as disclosed in WO 00/63190, which describes novel compounds that may be utilized in the treatment or prevention of conditions mediated by nuclear receptors, in particular PPAR; activator compounds as disclosed in WO 01/21181, which describes novel drugs efficacious against diseases in association with glycometabolism and lipid metabolibsm by inhibiting or promoting 5 PP ARa or PPAR ⁇ ; biaryl-oxa(thia)zole compounds as disclosed in WO 01/16120, in which modulators of PPARs are useful in the treatment of type 2 diabetes and of cardiovascular diseases; compounds as disclosed in WO 00/63196, which describes compounds that are useful in the treatment of conditions mediated by nuclear receptors, in particular Retinoid
  • Pat. No. 6,166,049 which describes a method comprising the administration of 5 PP ARa and PPAR ⁇ ; oxazole compounds as disclosed in WO 01/17994, which describes chemical modification of a phosphorous-based PPAR agonist; and dithiolane compounds as disclosed in WO 01/25225, and WO 01/25226, which describes methods for synthesizing novel dithiolane derivatives with high affinity for PP ARa or PPAR ⁇ .
  • PPAR activator compounds include substituted benzylthiazolidine- 0 2,4-dione compounds as disclosed in WO 01/14349, WO 01/14350, and WO 01/04351, all of which show how such a compound, as a ligand of human PPAR, enhances the transcriptional activity of the receptor and effects the lowering of blood sugar level and lipid level; mercaptocarboxylic compounds as disclosed in WO 00/50392, which demonstrates how such compounds exhibit excellent antihyperglycemic and PPAR- 5 activating effects; ascofuranone compounds as disclosed in WO 00/53563, which demonstrates how such compounds are usable in preventing or treating diabetes, chronic inflammation, digestive cancers, etc.; carboxylic compounds as disclosed in WO 99/46232, which have and effect of regulating PPARs; compounds as disclosed in WO 99/12534, which describes aromatic compounds that exhibit control effects against PPAR; benzene compounds as disclosed in WO 99/15520, which describes compounds that exhibit
  • the invention 5 also relates to combining separate pharmaceutical compositions in kit form. That is, the invention includes a kit wherein two separate units are combined: a pharmaceutical composition comprising at least a compound of any of the Formulae described herein and a separate pharmaceutical composition comprising at least one cholesterol biosynthesis inhibitor or lipid-lowering agent as described above. In one embodiment, 0 the kit may include directions for the administration of the separate components.
  • the kit form is particularly advantageous when the separate components must be administered in different dosage forms (e.g., oral and parenteral) or are administered at different dosage intervals.
  • compositions used in the methods of the 5 present invention can further comprise one or more AcylCo A: Cholesterol O- acyltransferase (“ACAT”) Inhibitors, which can reduce LDL and VLDL levels, coadministered with or in combination with the compound(s) of the Formulae herein discussed above.
  • ACAT is an enzyme responsible for esterifying excess intracellular cholesterol and may reduce the synthesis of VLDL, which is a product of cholesterol o esterification, and overproduction of apo B-100-containing lipoproteins.
  • Non-limiting examples of useful ACAT inhibitors include avasimibe ([[2,4,6-tris(l-methylethyl)phenyl]acetyl]sulfamic acid, 2,6-bis(l-methylethyl)phenyl ester, formerly known as CI-IOl 1), HL-004, lecimibide (DuP-128) and CL-277082 (N-(2,4-difluorophenyl)-iV-[[4-(2,2-dimethylpropyl)phenyl]methyl]-N-heptylurea). See P. Chang et al, "Current, New and Future Treatments in Dyslipidaemia and Atherosclerosis", Drugs 60(1), 55-93 (2000).
  • WO 00/28981 discloses the administration of an inhibitor of HMG CoA reductase (3-hydroxy-
  • 3-methylglutaryl CoA reductase 3-methylglutaryl CoA reductase to reduce the risk of onset of Alzheimer's disease.
  • the inhibitors used were lovastatin, pravastatin, or a combination thereof.
  • WO 00/31548 also discloses inhibitors of HMG CoA reductase, particularly statins.
  • simvastatin is a suggested inhibitor, contrasting with the results disclosed in WO 00/28981, which states that the prevalence of Alzheimer's disease in simvastatin-treated subjects was not decreased.
  • HMG-CoA 3-hydroxy-3-methylglutaryl-coenzyme A
  • reductase is the enzyme catalyzing the early rate-limiting step in cholesterol biosynthesis, i.e., conversion of HMG-CoA to mevalonate.
  • Cholesterol and triglycerides circulate in the bloodstream as part of lipoprotein complexes. These complexes may be separated by density ultracentrifugation into high (HDL), intermediate (BDL), low (LDL), and very low (VLDL) density lipoprotein fractions.
  • Triglycerides (TG) and cholesterol synthesized in the liver are incorporated into VLDLs and released into the plasma for delivery to pheripheral tissues.
  • VLDLs are transformed into DDLs and cholesterol-rich LDLs.
  • HDLs, containing apolipoprotein A are hypothesized to participate in the reverse transport of cholesterol from tissues back to the liver. Elevated levels of total cholesterol, i.e., hypercholesterolemia, low LDL- cholesterol (LDL-C), and apolipoprotein B (a membrane transport protein for LDL) promote human atherosclerosis.
  • HDL-cholesterol HDL-C
  • apolipoprotein A apolipoprotein A
  • Cardiovascular morbidity and mortality vary directly with the level of total cholesterol and LDL-C, and inversely with the level of HDL-C.
  • HMG-CoA reductase inhibitors have been shown to reduce total serum cholesterol levels, LDL-C, and apolipoprotein B, most likely by increasing the catabolism of LDL and hepatic extraction of LDL precursors, blocking enzymatic cholesterol synthesis, and simultaneously increasing HD levels.
  • These lipid lowering drugs lower serum cholesterol levels and reduce the incidence of both cardiovascular and cerebrovascular events. See, e.g., U.S. Pat. Nos.
  • Statins well known for the treatment of prevention of coronary heart disease, block a rate-limiting step in the biosynthesis of cholesterol by HMG-CoA reductase. See U.S. Pat. No. 6,465,516.
  • Alzheimer's disease is typically characterized pathologically by the presence of senile plaques and neurofibrillary tangles found at autopsy in the brains of subjects afflicted with the disease, vascular components of the disease have also been noted. These include lesions in the cerebral microcirculation and vascular deposits of A ⁇ protein, which is also a major constituent of the senile plaques found in Alzheimer's disease.
  • apoE4 and a second putative risk factor for Alzheimer's disease bind to a receptor, the lipoprotein receptor related protein, which is important for cellular uptake of cholesterol.
  • the lipoprotein receptor related protein which is important for cellular uptake of cholesterol.
  • apolipoprotein E isoform 4 (ApoE isoform 4) is a major genetic risk factor for Alzheimer's disease.
  • PCT Patent Application No. WO 95/06470 discloses administration of an HMG-CoA reductase inhibitor (statin) to regulate levels of (ApoE isoform 4) in humans to prevent and treat Alzheimer's Disease.
  • Statin HMG-CoA reductase inhibitor
  • a normal cellular 5 function of ApoE is uptake and delivery of lipids.
  • the ApoE isoform correlates with an increased risk for atherosclerosis, increased amyloid plaque deposition and increased risk of Alzheimer's disease.
  • K.Fassbender, et al "Simvastatin Strongly Reduces Levels of Alzheimer's Disease /3-amyloid peptides A ⁇ 42 and A ⁇ 40 in vitro and in vivo", Proc. Nat'lAcad. Sd. USA 98: 5856-5861 (2001).
  • PCT Patent Application WO 00/28981 o discloses at page 3 that patients possessing the ApoE isoform 4 have an increased risk for Alzheimer's disease, as well as elevated levels of cholesterol and increased risk for heart disease.
  • Levels of cholesterol in the brain are critical for synapse formation and maintenance and recent studies identify cholesterol as a limiting factor in 5 synaptogenesis. Reduced cholesterol may place a limit on plastic processes thus reducing the tendency to develop Alzheimer's disease.
  • An issue for very long axons is the ability to supply sufficient cholesterol for rapid axonal growth, especially in regeneration. Alzheimer's disease brain contains less cholesterol, and this contributes to Alzheimer's disease-related alterations in membrane composition, membrane fluidity, o and lipid bilayer structure and dynamics.
  • Statins as inhibitors of cholesterol synthesis, may reduce the prevalence of Alzheimer's disease. Long term potentiation is inhibited by cholesterol biosynthesis inhibitors and long term potentiation induction is associated with pathway-specific increases in lipid production. For example, axonal growth ceases when cholesterol synthesis is inhibited by pravastatin and could be reactivated by 5 addition of cholesterol to either cell bodies or distal axons.
  • HMG CoA reductase inhibitor refers to any compound which inhibits the bioconversion of 3-hydroxy-3-methylglutaryl coenzyme A to mevalonic acid catalyzed by the enzyme HMG CoA reductase.
  • the inhibiting effect of any such compounds can be readily determined by those skilled in the art according to standard o assays.
  • HMG CoA reductase inhibitors will be known to those skilled in the art.
  • suitable cholesterol biosynthesis inhibitors include competitive inhibitors of HMG CoA reductase, the rate-limiting step in cholesterol biosynthesis, squalene synthase inhibitors, squalene epoxidase inhibitors and mixtures thereof.
  • HMG CoA reductase inhibitors suitable for use in the invention include, but are not 5 limited to, pravastatin (for example PravacholTM which is available from Bristol Meyers Squibb) and related compounds, as disclosed in U.S. Pat. No. 4,346,227; and lovastatin and related compounds, as disclosed in U.S. Pat. Nos. 4,231,938 and 4,346,227.
  • pravastatin for example PravacholTM which is available from Bristol Meyers Squibb
  • lovastatin and related compounds as disclosed in U.S. Pat. Nos. 4,231,938 and 4,346,227.
  • lovastatin and pravastatin are used as HMG CoA reductase inhibitors in the invention.
  • Lovastatin marketed under the trade name MevacorTM, is a competitive inhibitor of HMG CoA reductase.
  • HMG CoA reductase inhibitors which may be employed in the invention 5 include atorvastatin (LipitorTM, Pfizer, New York, New York) and other HMG CoA reductase inhibitors.
  • 6-[2-(substituted-pyrrol-l-yl)alkyl]pyran-2-ones and derivatives such as disclosed in U.S. Pat. No. 4,647,576; fluvastatin (LescolTM, Novartis, Basel, Switzerland); fluindostatin (Sandoz XU-62-320); pyrazole analogs of mevalonolactone derivatives as disclosed in PCT application WO 86/03488; rivastatin and other o pyridyldihydroxyheptenoic acids as disclosed in European Patent 491226A; Searle's SC 45355 (a 3-substituted pentanedioic acid derivative) dichloroacetate; imidazole analogs of mevalonolactone, as disclosed in PCT application WO 86/07054; 3-carboxy- 2-hydroxy-propane-phosphonic acid derivatives, as disclosed in French Patent No.
  • HMG CoA reductase inhibitors include statins such as fluvastatin, simvastatin (for example ZocorTM which is available from Merck & Co.), atorvastatin, cerivastatin, CI-981 and pitavastatin (such as NK-104 of Negma Kowa of Japan), rosuvastatin; HMG CoA synthetase inhibitors, for example L-659,699 ((E 5 E)-Il- [3'R-(hydroxy-methyl)-4'-oxo-2'R-oxetanyl]-3,5,7R-tri- methyl-2,4-undecadienoic 5 acid); squalene synthesis inhibitors, for example squalestatin 1; and squalene epoxidase inhibitors, for example, NB-598 ((E)-N-ethyl-N-(6,6-dimethyl-2-hepten-4-ynyl)-3- [
  • statins include AdvicorTM (lovastatin/niacin); cerivastatin (BaycolTM, Bayer Corp., withdrawn from U.S. market); MevacorTM (lovastatin, Merck & Co., Inc., Rahway, New Jersey); rivastatin; rosuvastatin; pitavastatin; mevastatin; velostatin; and ZocorTM (simvastatin, Merck & Co., Inc., Rahway, New Jersey).
  • AdvicorTM lovastatin/niacin
  • cerivastatin BoycolTM, Bayer Corp., withdrawn from U.S. market
  • MevacorTM lovastatin, Merck & Co., Inc., Rahway, New Jersey
  • rivastatin rosuvastatin
  • pitavastatin mevastatin
  • mevastatin velostatin
  • ZocorTM imvastatin, Merck & Co., Inc., Rahway, New Jersey
  • HMG-CoA 5 reductase inhibitors include pyrazole analogs of a mevalonolactone, indene analogs of mevalonolactone, 3-carboxy-2-hydroxy-propanephosphinic acid derivatives, 6-[2-(substituted-pyrrol-l-yl)-alkyl] ⁇ yran-2-one, heterocyclic analogs of mevalonolactone including imidazole analogs, naphthyl analogs of mevalonolactone, octahydro-naphthalene derivatives, keto analogs of lovastatin, and 2,3-di-substituted pyrrole, furan, or tliiophene compounds.
  • statins in combination with substrates of nitric oxide synthase are known to facilitate transport of drugs across the blood brain barrier ("BBB").
  • BBB blood brain barrier
  • an especially useful pharmaceutical composition is a combination of a statin second agent and a nitric oxide synthase substrate second agent (e.g., L-Arg) as well as a first agent as described herein.
  • Nitric Oxide Synthase A family of enzymes called Nitric Oxide Synthase (“NOS”) form nitric oxide from L-arginine, and the nitric oxide produced is responsible for the endothelium dependent relaxation and activation of soluble guanylate cyclase, neurotransmission in the central and peripheral nervous systems, and activated macrophage cytotoxicity.
  • Nitric Oxide Synthase occurs in many distinct isoforms which include a constitutive form (cNOS) and an inducible form (iNOS). The constitutive form is present in normal endothelial cells, neurons and some other tissues.
  • nitric oxide by the constitutive form in endothelial cells is thought to play an important role in normal blood pressure regulation, prevention of endothelial dysfunction such as hyperlipodemia, arteriosclerosis, thrombosis, and restenosis.
  • the inducible form of nitric oxide synthase has been found to be present in activated macrophages and is induced in vascular smooth muscle cells, for example, by various cytokines or microbial products.
  • the conversion of precursor substrates such as L-arginine into nitric oxide is enzymatically catalyzed by NOS and the resulting by-product of the conversion of L-arginine is L-citrulline.
  • L-arginine as used herein includes all biochemical equivalents (i.e. salts, precursors, and its basic form).
  • this invention provides a method to enhance delivery of a first agent to brain tissue of an individual comprising introducing the composition into the blood stream of the individual substantially contemporaneously with a blood flow enhancing amount of L-arginine.
  • this invention provides a method to enhance delivery of a desired composition to brain tissue of an individual comprising introducing the composition into the blood stream of the individual substantially contemporaneously with a blood flow enhancing amount of L-arginine or a blood flow-enhancing amount of a non-ecNOS NO-generating system.
  • compositions used in the methods of the present invention may further comprise one or more Cholesteryl Ester Transfer Protein ("CETP") Inhibitors coadministered with or in combination with the compound(s) of the Formulae described herein.
  • CETP is responsible for the exchange or transfer of cholesteryl ester carrying HDL and triglycerides in VLDL.
  • Non-limiting examples of suitable CETP inhibitors are disclosed in PCT Patent 5 Application No. WO 00/38721 and U.S. Pat. No. 6,147,090, which are incorporated herein by reference.
  • Pancreatic cholesteryl ester hydrolase (pCEH) inhibitors such as WAY-121898 also may be coadministered with or in combination with the fibric acid derivative(s) and sterol absorption inhibitor(s) discussed above.
  • compositions used in the methods of the o present invention may further comprise probucol or derivatives thereof (such as AGI-
  • compositions used in the methods of the 5 present invention may further comprise one or more low-density lipoprotein (LDL) receptor activators, coadministered with or in combination with a compound of any Formula discussed above.
  • LDL-receptor activators include HOE-402, an imidazolidinyl-pyrimidine derivative that directly stimulates LDL receptor activity. See, M. Huettinger et al., "Hypolipidemic activity of HOE-402 is 0 Mediated by Stimulation of the LDL Receptor Pathway.” Arterioscler. Thromb. 13,
  • compositions used in the methods of the present invention can further comprise plant sterols, plant stanols or fatty acid esters of plant stanols, such as sitostanol ester used in BenecolTM margarine, which can reduce 5 cholesterol levels, coadministered with or in combination with a compound of any
  • a total daily dosage of plant sterols, plant stanols or fatty acid esters of plant stanols can range from about 0.5 to about 20 grams per day in single or 2- 4 divided doses.
  • compositions used in the methods of the o present invention can further comprise one or more antioxidants, such as probucol, tocopherol, ascorbic acid, /3-carotene and selenium, or vitamins such as vitamin B 6 or vitamin B 12 , coadministered with or in combination with a compound of any Formula herein.
  • antioxidants such as probucol, tocopherol, ascorbic acid, /3-carotene and selenium
  • vitamins such as vitamin B 6 or vitamin B 12
  • a total daily dosage of antioxidants or vitamins can range from about 0.05 to about 10 grams per day in single or 2-4 divided doses.
  • compositions used in the methods of the present invention can further comprise monocyte and macrophage inhibitors such as polyunsaturated fatty acids (PUFA), thyroid hormones including throxine analogues such as CGS-26214 (a thyroxine compound with a fluorinated ring), gene therapy and 5 use of recombinant proteins such as recombinant apo E, coadministered with or in combination with a compound of any Formula herein.
  • PUFA polyunsaturated fatty acids
  • thyroid hormones including throxine analogues such as CGS-26214 (a thyroxine compound with a fluorinated ring)
  • gene therapy a thyroxine compound with a fluorinated ring
  • recombinant proteins such as recombinant apo E, coadministered with or in combination with a compound of any Formula herein.
  • a total daily dosage of these agents can range from about 0.01 to about 1000 mg/day in single or 2-4
  • Fassbender et al disclose that use of simvastatin and lovastatin, alone or in o combination with methyl-/3-cyclodextrin, can reduce intracellular and secreted A ⁇ levels in vitro and that treatment of animals with simvastatin reduces brain and cerebrospinal fluid levels of A ⁇ in vivo.
  • U.S. Pat. No. 6,071,899 discloses compounds, which may have a general application in any disorder that involves endothelial dysfunction, such as atherosclerosis, 5 or may have a general application in any disorder that involves lipid peroxidation in conjunction with enzyme activity, including inflammatory conditions of the brain such as Alzheimer's Disease (see col. 5, lines 16-29).
  • PCT Patent Application WO 99/38498 discloses methods for preventing or treating Alzheimer's disease by administering a plasma-triglyceride level-lowering agent o (e.g. , f ⁇ brates), optionally hi combination with a cholesterol level-lowering agent such as statins, bile acid sequestrants or agents that block intestinal cholesterol absorption (e.g., /3-sitosterol, SCH 48461 ((3R,4S)-l,4-bis-(4-methoxyphenyl)-3-(3-phenylpropyl)-2- azetidi- none), CP-148,623, saponins, neomycin and ACAT inhibitors).
  • a plasma-triglyceride level-lowering agent o e.g. , f ⁇ brates
  • a cholesterol level-lowering agent such as statins, bile acid sequestrants or agents that block intestinal cholesterol absorption
  • SCH 48461 ((3R,4S)
  • Simvastatin has been used to reduce levels of /3-amyloid peptides A ⁇ 42 and A ⁇ 40 o in vitro and in vivo, for example, in guinea pigs. Wolozin, B. et al, Arch. Neurol.
  • Acyl-CoA cholesterol acyltransferase (ACAT) inhibitors have been used to decrease plasma cholesterol in various animal models including rats, guinea pigs and rabbits (Tanaka et al, J. Med. Chem. 41:2390-2410, 1998; Junquero et al, Biochem. Pharmacol. 61:97- 5 108, 2001).
  • ACAT cholesterol acyltransferase
  • ACAT inhibitors include but are not limited to Glibenclamide, CI-976 (PD128042), NTE-122, Fatty acid Anilides, F12511, Avasimibe, TS-962 (HL- 004), iV-Chlorosulfonyl isocyanate and derivatives, SR-9223i, Pyripyropenes, PD- 132301, PD-132301-2, DUP-128, YM-17E, BW447A, Alzheimer's disease 6591, CL- 277,082, Melinamide, Hydroxyphenyl Urea derivatives, R-106578, Indoline derivatives o with amide or urea moiety, 57-118, 58-035, CI-999, CI-1011, JV-alkyl-iV-[(fluoro ⁇ henox- y)benzyl]-iV'-arylureas and derivatives, SKF-99085, EAB309, iV-alkyl-i
  • Amyloid Inhibitors Anti-Amyloid Therapeutic Approaches
  • presenilins There are a number of possibilities for the function of presenilins: (1) They could be required for intracellular trafficking and sorting of APP to the ⁇ -secretase compartment, or (2) They could serve as a co- factor for ⁇ -secretase cleavage.
  • ⁇ -Secretase cleaves at the C-terminus of A ⁇ and is primarily responsible for generating the pathogenic 42-amino acid form of A ⁇ , A ⁇ 42, which forms insoluble toxic 5 fibrils and accumulates in senile plaques.
  • the normal function of APP is unknown, it is predominantly expressed in the brain and is suspected of participating in cell adhesion, synaptic growth, o and neural repair.
  • Amyloid- ⁇ forms a continuum of aggregation species: monomelic amyloid— ⁇ , soluble oligomeric amyloid- ⁇ , insoluble protofibrils, amyloid- ⁇ , diffuse amyloid, compact amyloid, and neuritic or senile amyloid, the latter two being the pathologic and diagnostic hallmarks of Alzheimer's disease.
  • amyloid— ⁇ may alter membrane potential and firing, synaptic transmission, synaptic plasticity, and learning.
  • Amyloid- ⁇ especially amyloid- ⁇ 1-42, has been shown to be neurotoxic. Accordingly, amyloid- ⁇ itself represents a significant drug target.
  • Dense microspheres or spherons may be turned into plaques when they are burst in vitro or when injected into experimental animals.
  • P. Averback J Alzheimer's Disease 1, 1-34 (1998).
  • the compound NX-D2858 (Nymox Pharmaceutical Corp., o Dorval, Quebec, Canada) blocks the transformation of spherons into senile plaques and may stop or slow the progress of Alzheimer's disease.
  • AteroidTM Hunter-Fleming,
  • mucopolysaccharides such as glycosaminoglycans having an average molecular weight equal to 2,400 Da, which 5 have been described as suitable for the treatment of Alzheimer's disease.
  • EP 1,181,024 is AteroidTM (Hunter-Fleming,) and related mucopolysaccharides, such as glycosaminoglycans having an average molecular weight equal to 2,400 Da, which 5 have been described as suitable for the treatment of Alzheimer's disease.
  • Suitable amyloid aggregation o inhibitors also include reumacon available from Conpharm AB.
  • the present invention pertains to a method for treating or preventing a disease state associated with amyloidosis, the method comprising administering to a subject a therapeutically effective amount of an agent for reducing the concentration of fibrillar or o soluble A ⁇ , such that said disease state associated with amyloidosis is treated or prevented.
  • the methods of the invention are based, at least in part, on inhibiting an interaction between an amyloidogenic protein and a constituent of basement membrane to inhibit amyloid deposition.
  • the constituent of basement membrane is a glycoprotein or proteoglycan, e.g., heparan sulfate proteoglycan.
  • a therapeutic agent used in the method of the invention may interfere with binding of a basement membrane constituent to a target binding site on an amyloidogenic protein, 5 thereby inhibiting amyloid deposition.
  • the methods of the invention involve administering to a subject a therapeutic agent which inhibits amyloid deposition.
  • "Inhibition of amyloid deposition” is intended to encompass prevention of amyloid formation, inhibition of further amyloid deposition in a subject with ongoing amyloidosis and reduction of amyloid deposits in a subject with ongoing amyloidosis. o Inhibition of amyloid deposition is determined relative to an untreated subject or relative to the treated subject prior to treatment.
  • Amyloid deposition is inhibited by inhibiting an interaction between an amyloidogenic protein and a constituent of basement membrane.
  • Basement membrane refers to an extracellular matrix comprising glycoproteins and proteoglycans, including laminin, collagen type IV, fibronectin and heparan sulfate 5 proteoglycan ("HSPG").
  • HSPG heparan sulfate 5 proteoglycan
  • amyloid deposition is inhibited by interfering with an interaction between an amyloidogenic protein and a sulfated glycosaminoglycan such as HSPG.
  • Sulfated glycosaminoglycans are known; to be present in all types of amyloids (see Snow, et al. Lab. Invest. 56, 120-23 (1987)) and amyloid deposition and HSPG deposition occur coincidentally in animal models of 0 amyloidosis (see Snow, et al. Lab. Invest. 56, 665-7
  • a therapeutic compound of the invention to inhibit an interaction between an amyloidogenic protein and a glycoprotein or proteoglycan constituent of a basement membrane may be assessed by an in vitro binding assay, such as that described in U.S. Pat. No. 5,164,295.
  • an in vitro binding assay such as that described in U.S. Pat. No. 5,164,295.
  • a therapeutic agent of the invention may interact with a binding site for a basement membrane glycoprotein or proteoglycan in an amyloidogenic protein and thereby inhibits the binding of the amyloidogenic protein to the basement membrane constituent.
  • Basement membrane glycoproteins and proteoglycans include laminin, collagen type IV, fibronectin and HSPG.
  • the therapeutic agent 5 inhibits an interaction between an amyloidogenic protein and HSPG.
  • Consensus binding site motifs for HSPG in amyloidogenic proteins have been described (see, e.g., Cardin and Weintraub, Arteriosclerosis 9, 21-32 (1989)).
  • Metal Chelators have been described (see, e.g., Cardin and Weintraub, Arteriosclerosis 9, 21-32 (1989)).
  • Zn 2+ mediates neurodegenerative processes observed in seizure, ischemia, trauma, and Alzheimers disease. Zn 2+ is observed in the extracellular plaque and degenerating neurons in Alzheimer's disease, which may contribute to neuronal degeneration in Alzheimer's disease. Oxidative damage in the neocortex associated with Alzheimer's disease may be the result of gradual build up of metal ions like zinc and copper. Copper and zinc have particularly high concentrations in the /3-amyloid plaques in the brains of Alzheimer's subjects. Both metals are essential, but normally only small amounts are required and excess metals are excreted. It is hypothesized that ⁇ -amyloid converts dissolved oxygen to hydrogen peroxide, which in turn causes cell damage.
  • Metal chelators may be used to diminish the oxidative burden.
  • clioquinol an antibiotic and bioavailable Cu/Zn chelator
  • Zinc and other divalent cations appear necessary for A ⁇ aggregation.
  • metal chelation may have some therapeutic benefit in the treatment of Alzheimer's disease, either by preventing A ⁇ aggregation or by disrupting preformed aggregates.
  • the copper-zinc chelator clioquinol may dissolve amyloid-beta deposits in postmortem brain tissue from Alzheimer's disease subjects.
  • the antibiotic clioquinol also known as 5-chloro-7-iodo- 8-hydroxyquinoline or iodochlorhydroxyquin
  • a known copper/zinc chelator is a reasonably well tolerated drug in humans and is currently in a phase II clinical trial for Alzheimer's disease.
  • T.E. Golde J Clin. Invest. 111, 11-18 (2003).
  • Clioquinol chelates copper and zinc in vitro, and reduces A ⁇ deposition in a mouse model.
  • interim results from a randomized, double-blind, placebo-controlled clinical trial in 32 subjects with Alzheimer's disease suggested that this drug slows the rate of cognitive decline in the most severely affected group.
  • Suitable copper/zinc chelators include clioquinol available from PN Gerolymatos
  • Phanquinone (4,7-phenanthroline-5,6-dione) has hitherto been used for the treatment of various disorders, such as amoebiasis. However, its use for the treatment or prevention of memory improvement has been suggested. Phanquinone has been o marketed by Ciba-Geigy as EntobexTM. Phanquinone is also a metal chelator in the same family as clioquinol. According to the present invention the use of phanquinone for the manufacture of a pharmaceutical composition for the treatment or prevention of memory impairment is provided.
  • Alzheimer's disease 5 Patients with Alzheimer's disease may also be treated for behavioral disturbances associated with progression of the disease. Use of such treatments is intended to decrease psychotic symptoms such as paranoia, delusions and hallucinations, and associated or independent agitation, screaming, combativeness or violence, and thereby increase the comfort and safety of patients. Anti-psychotics and antidepressants o can be used intermittently in patients with defined psychotic symptoms.
  • Benzodiazepines maybe used briefly and judiciously for emergency sedation but otherwise should be avoided because they can produce delirium and tend to further compromise residual cognitive capacities.
  • Lithium Esskalith
  • centrally active ⁇ -adrenergic blockers carbamazepine (TegretolTM, Ciba-Geigy Pharmaceuticals, now 5 Novartis, Basel, Switzerland)
  • valproate Depakene
  • Resperidone can also be used for psychoses associated with Alzheimer's disease.
  • Olanzabine, sertindole, and quetiapine can also be used.
  • Still other examples include trazodone; ⁇ blockers, propranolol, metoprolol and pindotol (especially for some agitated patients with o dementia).
  • trazodone When male patients display intrusive disinhibited sexual behavior, a particular problem in patients with frontal lobe dementias, medroxyprogesterone and related hormonal agents may be employed.
  • Glycosaminoglycan polysulfate (AteroidTM) can also improve depressive symptomatology in old-age dementia. Prog. Neuropsychopharmacol. Biol. Psychiatry 13, 977-81 (1989). Treatment of apathy is also considered.
  • Dopaminergic agents such as psychostimulant CD-amphelamine, methylphemidate), amantadine (SymmetrelTM, Du Pont Multi-Source Products, Wilmington, DE), bromocriptine and buproprion are helpful in the treatment of severe apathy.
  • A.E.Wallace, et al "Double-blind, placebo- 5 controlled trial of methylphenidate in older, depressed, medically ill patients.” Am. J. Psychiatry 152, 929-31 (1995).
  • SSRIs are often chosen as the initial treatment because of their better side effect profiles. Once-a-day dosing may be appropriate.
  • SSPJs include fluoxetine (ProzacTM, Pulvules, Dista, Eli Lilly, Indianapolis, Indiana), paroxetine, sertraline, o bupropion, and venlagaxin.
  • agents with prominent anticholinergic activity e.g., amitriptyline, imipramine.
  • sample dosing strategies are given here for nortriptyline.
  • MAOIs are also considered for individuals unresponsive to or unable to take other agents (tranylcypromine and 5 phenelzine are examples)
  • trazodone or Zolpidem When sleep disturbances occur without other psychiatric symptoms beyond the dementia itself, some clinicians prescribe trazodone or Zolpidem.
  • Benzodiazepines e.g., lorazepam or oxazepam
  • chloral hydrate may be used.
  • Triazolam in particular is not recommended for individuals with dementia because of its association with amnesia.
  • o Diphenhydramine which is found in most over-the-counter sleep preparations, is used by some clinicians, but its anticholinergic properties make it suboptimal for the treatment of demented patients.
  • Some individuals with dementia show disinhibited behavior, including making inappropriate jokes, neglecting personal hygiene, exhibiting undue familiarity with 5 strangers, or disregarding conventional rules of social conduct. Occasionally, they may harm others by striking out. Suicidal behavior may occur, especially in mildly impaired individuals, who are more likely to have insight into their deficits and to be capable of formulating (and carrying out) a plan of action.
  • Anxiety is fairly common, and some patients manifest "catastrophic reactions," overwhelming emotional responses to o relatively minor stressors, such as changes in routine or environment.
  • Depressed mood with or without neurovegetative changes, is quite common, as are sleep disturbances independent of depression.
  • Delusions can occur, especially those involving themes of opposition (e.g., the belief that misplaced possessions have been stolen). Misidentifications of familiar people as unfamiliar (or vice versa) frequently occur. 5 Hallucinations can occur in all sensory modalities, but visual hallucinations are most common. Some patients exhibit a peak period of agitation (or other behavioral disturbances) during the evening hours, which is sometimes referred to as "sundowning.”
  • Delirium is frequently superimposed on dementia because the underlying brain disease increases susceptibility to the effects of medications or concurrent general medical conditions. Individuals with dementia may also be especially vulnerable to psychosocial stressors (e.g., going to the hospital, bereavement), which may exacerbate their intellectual deficits and associated problems.
  • Dementia is sometimes accompanied by motor disturbances, which may include gait difficulties, slurred speech, and a variety of abnormal movements. Other neurological symptoms, such as myoclonus and seizures, may also occur.
  • Glycosaminoglycan polysulfate e.g., AteroidTM
  • AteroidTM effects upon depressive symptomatology in geriatric patients.
  • SSRIs are often chosen as the initial treatment because of their better side effect profiles. Once-a-day dosing is appropriate. Fluoxetine,. Paroxetine,. Sertraline, bupropion and venlagaxin are examples of SSRIs treatment.
  • tricyclic and heterocyclic agents theoretical reasoning and clinical experience suggest avoiding agents with prominent anticholinergic activity (e.g., amitriptyline, imipramine).
  • sample dosing strategies are given here for nortriptyline
  • Depression is common in patients with dementia. Patients with depression o should be carefully evaluated for suicide potential. Depressed mood may respond to improvements in the living situation or stimulation-oriented treatments, but patients with severe or persistent depressed mood with or without a full complement of neurovegetative signs should be treated with antidepressant medications. Although formal evaluation of the efficacy of antidepressants for demented patients is limited, 5 there is considerable clinical evidence supporting their use. The choice among agents is based on the side effect profile and the characteristics of a given patient. MAOIs are also considered for individuals unresponsive to or unable to take other agents (tranylcypromine and phenelzine are examples).
  • Sleep disorder is common in dementia (Satlin A: Sleep disorders in dementia. Psychiatr. Ann. 24, 186-90 (1994); C.C.Hoch, et al, "Sleep patterns in Alzheimer, depressed, and healthy elderly.” West J. Nurs. Res. 10, 239-56 5 (1988)) and is not always so disruptive that the risk of medication side effects is outweighed by the need for a pharmacologic trial.
  • Benzodiazepines e.g., lorazepam or oxazepam
  • chloral hydrate may in some cases be used.
  • Triazolam in particular is not recommended o for individuals with dementia because of its association with amnesia.
  • Diphenhydramine which is found in most over-the-counter sleep preparations, is used by some clinicians, but its anticholinergic properties make it suboptimal for the treatment of demented patients.
  • Nutritional supplements Vitamin B 12, homocysteine
  • a first agent is co-administered with one or more nutritional supplements to improve the safety or efficacy of treatment.
  • treatment can be optimized by provision of nutrients depleted in the subject.
  • suitable second agents include the following vitamins, some of which have an antioxidant and/or anti-inflammatory effect: retinol, vitamin A aldehyde (retinal), vitamin A acid (retinoic acid), retinyl acetate, vitamin B 1 (thiamine HCl), thiamine propyl disulfide, thiamine disulfide, thiamine disulfide 0,0-diisobutyrate, thiamine disulfide hydrochloride, thiamine disulfide phosphate, thiamine mononitrate, o thiamine 1,5-salt, thiamine phosphoric acid ester chloride, thiamine phosphoric acid ester phosphate salt, thiamine triphosphoric acid ester, vitamin B 2 (riboflavin), riboflavin tetrabutyrate, riboflavine 5'-phosphate ester monosodium salt, vitamin B 5 (pantothenic acid), pantothenic acid sodium salt, pantothenic acid calcium salt, vitamin B 6 (
  • EP 296979 Pyridine derivatives for the treatment of impairment of short-term memory are disclosed in U.S. Pat. No. 4,448,779. Choline derivatives for treating mental decline in the elderly are suggested in EP 201623. Indole or indolin derivatives for the improvement of processes involved in learning are disclosed in EP 241006, JP 6107544, 5 U.S. Pat. No. 5,494,928, WO 97/47598, and U.S. Pat. No. 4,778,812. Pilocardin derivatives for improving memory functions are disclosed in U.S. Pat. No. 4,977,176. Glycine-containing compositions for enhancing cognitive functions are disclosed in U.S. Pat. No. 5,731,349.
  • Peptide derivatives for treating mental decline and improving mental capacity are disclosed in U.S. Pat. No. 5,439,930, RU 2099078, and WO 95/15310.
  • Xanthine derivatives for the treatment of age-related memory impairment are disclosed in WO 94/19349.
  • Compounds enhancing the stimulus-induced release of neurotransmitters, especially acetylcholine, may also be used to treat memory impairment.
  • Examples are 2- benzyl-2-propyl 2-amino-2-R-acetate derivatives disclosed in EP 293351, l-(4-chlorophenyl)-2-methyl-2-propyl 2-amino-3-methyl-butanoate disclosed in GB 2205097, polycyclic hetero-aromatic derivatives disclosed in U.S. Pat. No.
  • Compounds that modulate the function of the kainate receptor may be used to improve memory, e.g., alkyl carboxy amino acids, such as (2S,4R)-4-methyl glutamic acid.
  • alkyl carboxy amino acids such as (2S,4R)-4-methyl glutamic acid.
  • Hypothalamic hypophysiotropic hormones such as somatostatin and growth- hormone releasing factor, may improve learning abilities.
  • Uronic acids improve cerebral efficiency in general, such as improvement of memory. DE 2555010.
  • WO 98/33498 discloses the use of breflate or analogous compounds thereof for the treatment of a mammal suffering from a cognitive dysfunction.
  • Breflate or analogous compounds thereof enhance the long-term potential of nerve cells.
  • Suitable monoamine reuptake inhibitors include NS-2330.
  • Suitable nootropic agents include oxiracetam available from ISF Societa Per Azioni, pramiracetam available from Warner Lambert Co., idebenone available from Takeda Chemical Inds. Ltd., anapsos available from ASAC Pharmaceuticals International, nebracetam available from Boehringer Ingelheim Corp., JTP-2942 available from Japan Tobacco Inc., fasoracetam available from Nippon Shinyaku Co.
  • a useful dopamine receptor agonist is speramine.
  • Useful AMPA receptor ligands 5 include CX-516, CX-691 available from Cortex Pharmaceuticals Inc. and combinations thereof.
  • Suitable calcium channel blockers include tamolarizine available from Nippon Chemiphar Co., Ltd., nimodipine available from Bayer AG, PD-I 76078 available from Elan Pharmaceuticals, Inc., and combinations thereof.
  • Suitable apoptosis inhibitors include acetyl-L-carnitine, CEP-1347 available from Cephalon, hie, TCH-346 available o from Novartis AG and combinations thereof.
  • a useful caspase inhibitor is pralnacasan.
  • Suitable monoamine oxidase inhibitors include moclobemide available from Roche Holding AG, selegiline, rasagiline available from Teva Pharmaceutical Inds. Ltd., SL- 25.1188, Ro-41-1049 available from Roche Holding AG, and combinations thereof.
  • a useful 5-HTla receptor agonist is AP-159 available from Asahi Kasei Corp.; a suitable 5 NGF stimulator is xaliprodene available from Sanofi-Synthelabo.
  • Suitable neuroprotective agents include citicholine, GS-1590 available from Leo Pharmaceutical Products Ltd. AJS, CPI-1189 available from Centaur Pharmaceuticals Inc., SR-57667 available from Sanofi-Synthelabo and combinations thereof.
  • Suitable H3 histamine receptor antagonists include GT-2016 and GT-2331 (both available from Gliatech, Inc.) o and combinations thereof.
  • Useful prolylendopeptidase inhibitors include ONO- 1603 available from Ono Pharmaceutical Co. Ltd., Z-321 available from Zeria Pharmaceutical Co. Ltd. and combinations thereof.
  • a useful calcium modulator includes neurocalc available from Apollo Biopharmaceuticals, Inc.
  • a suitable corticortropin releasing factor receptor 5 antagonist includes NBI- 113 available from Neurocrine Biosciences, Inc.
  • GABA modulator includes NGD 97-1 available from Neurogen Corp.
  • a suitable sigma receptor ligand is igmesine available from Pfizer Inc.
  • a useful imidazoline/alpha adrenergic receptor antagonist is efaroxan available from Reckitt & Colman PLC.
  • a suitable vasoactive intestinal peptide receptor agonist is stearyl-NIe-VIP.
  • a useful 0 benzodiazepine inverse agonist is S-8510 available from Shionogi & Co. Ltd.
  • a suitable cannabinoid receptor agonist is dronabinol available from Unimed Pharmaceuticals Inc.
  • Useful thyrotropin releasing hormone receptor agonists include taltireline available from Tanabe Seiyaku Co. Ltd.
  • a suitable 5-HT3 antagonist is GYKI-46903.
  • a useful topoisomerase II inhibitor is 5 iododoxorubicin available from Pharmacia & Upjohn AB.
  • a suitable steroid receptor agonist is GL-701 available from Leland Stanford Junior University.
  • a useful corticosteroid receptor antagonist is anticort.
  • a suitable nitric oxide modulator is GL- 701.
  • a suitable RAGE inhibitor is ALT-711 available from Alteon Inc. RAGE is a multiligand receptor of the immunoglobulin superfamily that is implicated in homeostasis and chronic disease. Bucciarelli, et ah, Cell MoI Life Sd. 59(7), 1117-28 (2002).
  • the invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one compound selected from the group consisting of D-phosphoserine and L- phosphoserine and a second therapeutic agent selected from the group consisting of antipsychotics, antidepressants, psychostimulants, and Alzheimer's disease therapeutics.
  • the second therapeutic agent is an antipsychotic o selected from the group consisting of typical antipsychotics, atypical antipsychotics, and depot antipsychotics.
  • second therapeutic agents include Chlorpromazine, Thioridazine, Mesoridazine, Fluphenazine, Perphenazine, Trifluoperazine, Thiothixene, Haloperidol (HaldolTM, McNeil Pharmaceuticals, Spring House, Pennsylvania), Loxapine, Molindone (MobanTM, Du Pont Multi-Source Products, Wilmington, DE), 5 Clozapine, Risperidone, Olanzapine, Quetiapine, Haloperidol decanoate, Fluphenazine decanoate, Fluphenazine enanthate, Amitriptyline, Amoxapine, Bupropion, Bupropion SR, Clomipramine, Desipramine, Doxepin, Fluoxetine, Fluvoxamine, Imipramine, Maprotiline, Mirtazapine, Nefazodone, Nortriptyline, Paroxetine, Phenelzine, Protriptyline, Sertraline,
  • compositions used in the methods to treat or prevent a neuropsychiatric disorder characterized by attenuated NMDA 5 neurotransmission may be Alzheimer's disease, Down's syndrome, depression, benign forgetfulness, cerebral amyloid angiopathy, vascular dementia, hemorrhagic stroke, Mild Cognitive Impairment ("MCI”), or close head injury.
  • MCI Mild Cognitive Impairment
  • compositions used in the methods of the o present invention can further comprise one or more bile acid sequestrants (insoluble anion exchange resins), coadministered with or in combination with a compound of any Formula herein.
  • bile acid sequestrants insoluble anion exchange resins
  • Bile acid sequestrants bind bile acids in the intestine, interrupting the enterohepatic circulation of bile acids and causing an increase in the faecal excretion of 5 steroids. Use of bile acid sequestrants is desirable because of their non-systemic mode of action. Bile acid sequestrants can lower intrahepatic cholesterol and promote the synthesis of apo B/E (LDL) receptors which bind LDL from plasma to further reduce cholesterol levels in the blood.
  • LDL apo B/E
  • Non-limiting examples of suitable bile acid sequestrants include cholestyramine (a styrene-divinylbenzene copolymer containing quaternary ammonium cationic groups 5 capable of binding bile acids, such as QuestranTM cholestyramine which are available from Bristol-Myers Squibb), colestipol (a copolymer of diethylenetriamine and l-chloro-2,3-epoxypropane, such as ColestidTM tablets which are available from Pharmacia), colesevelam hydrochloride (such as WelCholTM Tablets (poly(allylamine hydrochloride) cross-linked with epichlorohydrin and alkylated with 1-bromodecane and o (6-bromohexyl)-trimethylammonium bromide) which are available from Sankyo), water soluble derivatives such as 3,3-ioene, JV-(cycloalkyl) alkylamines and poliglusam,
  • Suitable inorganic cholesterol sequestrants include bismuth salicylate plus montniorillonite clay, aluminum hydroxide and calcium carbonate antacids.
  • compositions used in the methods of the present invention can further comprise one or more ileal bile acid transport (“IBAT”) inhibitors (or apical sodium co-dependent bile acid transport (“ASBT”) inhibitors) o coadministered with or in combination with a compound of any Formula herein.
  • IBAT ileal bile acid transport
  • ASBT apical sodium co-dependent bile acid transport
  • IBAT inhibitors can inhibit bile acid transport to reduce LDL cholesterol levels.
  • suitable IBAT inhibitors include benzothiepines such as therapeutic compounds comprising a 2,3,4,5-tetrahydro-l-benzothie- pine 1,1-dioxide structure such as are disclosed in PCT Patent Application WO 00/38727 which is incorporated herein 5 by reference.
  • the compositions used in the methods of the present invention can further comprise nicotinic acid (niacin) or derivatives thereof coadministered with or in combination with a compound of any Formula herein.
  • nicotinic acid derivative means a compound comprising a o pyridine-3-carboxylate structure or a pyrazine-2-carboxylate structure, including acid forms, salts, esters, zwitterions and tautomers, where available.
  • nicotinic acid derivatives include niceritrol, nicofuranose and acipimox (5 -methyl pyrazine-2- carboxylic acid 4-oxide). Nicotinic acid and its derivatives inhibit hepatic production of VLDL and its metabolite LDL and increases HDL and apo A-I levels.
  • compositions, therapeutic combinations or methods of the present invention can further comprise one or more obesity control medications.
  • Useful obesity control medications include, but are not limited to, drugs that reduce energy intake or suppress appetite, drugs that increase energy expenditure and nutrient-partitioning agents.
  • Suitable obesity control medications include, but are not limited to, noradrenergic agents (such as diethylpropion, mazindol, phenylpropanolamine, phentermine, phendimetrazine, phendamine tartrate, methamphetamine, phendimetrazine and tartrate); serotonergic agents (such as sibutramine, fenfluramine, dexfenfluramine, fluoxetine, fluvoxarnine and paroxtine); thermogenic agents (such as ephedrine, caffeine, theophylline, and selective /33-adrenergic agonists); an alpha-blocking agent; a kainite or AMPA receptor antagonist; a leptin-lipolysis stimulated receptor; a phosphodiesterase enzyme inhibitor; a compound having nucleotide sequences of the mahogany gene; a fibroblast growth factor- 10 polypeptide; a monoamine oxidase inhibitor (such as befloxa
  • compositions, therapeutic combinations or methods of the present invention can further comprise one or more blood modifiers.
  • blood modifiers include but are not limited to anti-coagulants (argatroban, bivalirudin, dalteparin sodium, desirudin, dicumarol, lyapolate sodium, nafamostat mesylate, phenprocoumon, tinzaparin sodium, warfarin sodium); antithrombotic (anagrelide hydrochloride, bivalirudin, cilostazol, dalteparin sodium, danaparoid sodium, dazoxiben hydrochloride, efegatran sulfate, enoxaparin sodium, fluretofen, ifetroban, ifetroban sodium, lamifiban, lotrafiban hydrochloride, napsagatran, orbofiban acetate, roxifiban acetate, sibrafiban, tinzaparin sodium, trifenagrel, abcixima
  • compositions, therapeutic combinations or methods of the present invention can further comprise one or more cardiovascular agents.
  • cardiovascular agents include but are not limited to calcium channel blockers (clentiazem maleate, amlodipine o besylate, isradipine (DynaCircTM, Reliant Pharmaceuticals, Liberty Corner, NJ), nimodipine, felodipine (PlendilTM, Merck & Co., Inc., Rahway, New Jersey), nilvadipine, nifedipine, teludipine hydrochloride, diltiazem hydrochloride ((CardizemTM or Cardizem SRTM, Aventis, France), belfosdil, verapamil hydrochloride (CalanTM or Calan SRTM, G.D.
  • adrenergic blockers 5 (fenspiride hydrochloride, labetalol hydrochloride, proroxan, alfuzosin hydrochloride, acebutolol, acebutolol hydrochloride, alprenolol hydrochloride, atenolol, bunolol hydrochloride, carteolol hydrochloride, celiprolol hydrochloride, cetamolol hydrochloride, cicloprolol hydrochloride, dexpropranolol hydrochloride, diacetolol hydrochloride, dilevalol hydrochloride, esmolol hydrochloride, exaprolol hydrochloride, o flestolol sulfate, labetalol hydrochloride, levobetaxolol hydrochloride, levobunolol hydrochlor
  • Nitric oxide is a vasodilator of the peripheral vasculature in normal tissue of the o body. Increasing generation of nitric oxide by nitric oxide synthase causes vasodilation without loss of blood pressure. The blood-pressure-independent increase in blood flow through brain tissue increases cerebral bioavailability of blood-born compositions. This increase in nitric oxide may be stimulated by administering L-arginine. As nitric oxide is increased, cerebral blood flow is consequently increased, and drugs in the blood 5 stream are carried along with the increased flow into brain tissue.
  • L-arginine may be used in the pharmaceutical compositions of the invention to enhance delivery of agents to brain tissue after introducing a pharmaceutical composition into the blood stream of the subject substantially contemporaneously with a blood flow enhancing amount of L-arginine.
  • Agents of the invention that exert their physiological effect in vivo in the brain may be more useful if they gain access to target cells in the brain.
  • Non-limiting examples of brain cells are neurons, glial cells (astrocytes, oligodendrocytes, microglia), cerebrovascular cells (muscle cells, endothelial cells), and cells that comprise the meninges.
  • the blood brain barrier typically restricts access to brain cells by acting as a physical and functional blockade that separates the brain parenchyma from the systemic circulation (see, e.g., Pardridge, et al, J. Neurovirol. 5(6), 556-69 (1999); Rubin, et al, Rev. Neurosci. 22, 11-28 (1999)). Circulating molecules are normally able to gain access to brain cells via one of two processes: lipid-mediated transport through the BBB by free diffusion, or active (or catalyzed) transport.
  • the agents of the invention may be formulated to improve distribution in vivo, for example as powdered or liquid tablet or solution for oral administration or as a nasal spray, nose drops, a gel or ointment, through a tube or catheter, by syringe, by packtail, by pledget, or by submucosal infusion.
  • the blood-brain barrier excludes many highly hydrophilic agents.
  • they may be formulated, for example, in liposomes.
  • liposomes see, e.g., U.S. Pat. Nos. 4,522,811; 5,374,548; and 5,399,331.
  • the liposomes may comprise one or more moieties which are selectively transported into specific cells or organs ("targeting moieties” or “targeting groups” or “transporting vectors”), thus providing targeted drug delivery (see, e.g., V. V. Ranade J. Clin. Pharmacol. 29, 685 (1989)).
  • the agents may be linked to targeting groups that facilitate penetration of the blood brain barrier.
  • the method of the present invention employs a naturally occurring polyamine linked to an agent that is a small molecule and is useful for inhibiting A ⁇ deposition.
  • agents of the invention may be coupled to a BBB transport vector (for review of BBB transport vectors and mechanisms, see, Bickel, et al, Adv.
  • Exemplary transport vectors include cationized albumin or the 0X26 monoclonal antibody to the transferrin receptor; these proteins undergo absorptive-mediated and receptor-mediated transcytosis through the BBB, respectively.
  • Natural cell metabolites that may be used as targeting groups include, inter alia, putrescine, spermidine, spermine, or DHA.
  • Other exemplary targeting moieties include folate or biotin (see, e.g., U.S. Pat. No. 5,416,016); mannosides (Umezawa, et al, Biochem. Biophys. Res. Commun. 153, 1038 (1988)); antibodies (P.G.
  • BBB transport vectors examples include factors such as insulin, insulin-like growth factors ("IGF-I,” and “IGF-II”), angiotensin II, atrial and brain natriuretic peptide ("ANP,” and “BNP”), interleukin I (“IL-I”) and transferrin. Monoclonal antibodies to the receptors that bind these factors may also be used as BBB transport vectors.
  • BBB transport vectors targeting mechanisms for absorptive-mediated transcytosis include cationic moieties such as cationized LDL, albumin or horseradish peroxidase coupled with polylysine, cationized albumin or cationized immunoglobulins. Small basic oligopeptides such as the dynorpliin analogue E-2078 and the ACTH analogue ebiratide may also cross the brain via absorptive-mediated transcytosis and are potential transport vectors.
  • BBB transport vectors target systems for transporting nutrients into the brain.
  • BBB transport vectors include hexose moieties, e.g. , glucose and monocarboxylic acids, e.g., lactic acid and neutral amino acids, e.g., phenylalanine and amines, e.g., choline and basic amino acids, e.g., arginine, nucleosides, e.g., adenosine and purine bases, e.g., adenine, and thyroid hormone, e.g., triiodothyridine.
  • Antibodies to the extracellular domain of nutrient transporters may also be used as transport vectors.
  • Other possible vectors include angiotensin II and ANP, which may be involved in regulating BBB permeability.
  • the bond linking the therapeutic agent to the transport vector may be cleaved following transport into the brain in order to liberate the biologically active agent.
  • exemplary linkers include disulfide bonds, ester-based linkages, thioether linkages, amide bonds, acid-labile linkages, and Schiff base linkages.
  • Avidin/biotin linkers in which avidin is covalently coupled to the BBB drug transport vector, may also be used. Avidin itself may be a drug transport vector.
  • Transcytosis including receptor-mediated transport of compositions across the blood brain barrier, may also be suitable for the agents of the invention.
  • Transferrin receptor-mediated delivery is disclosed in U.S. Pat. Nos. 5,672,683; 5,383,988;
  • Transferrin-mediated transport is also known.
  • EGF receptor-mediated delivery is disclosed in Y. Deguchi, et al, Bioconjug. Chem. 10, 32-37 (1999), and transcytosis is described in A. Cerletti, et al, J. Drug Target. 8, 435-46 (2000). Insulin fragments have also been 5 used as carriers for delivery across the blood brain barrier .
  • M. Fukuta, et al, Pharm. Res. 11. 1681-88 (1994). Delivery of agents via a conjugate of neutral avidin and cationized human albumin has also been described.
  • 5,023,252 discloses 5 the use of pharmaceutical compositions comprising a neurologically active drug and a compound for facilitating transport of the drug across the blood-brain barrier including a macrocyclic ester, diester, amide, diamide, amidine, diamidine, thioester, dithioester, thioamide, ketone or lactone.
  • U.S. Pat. No. 5,024,998 discloses parenteral solutions of aqueous-insoluble drugs with cyclodextrin derivatives.
  • U.S. Pat. No. 5,039,794 o discloses the use of a metastatic tumor-derived egress factor for facilitating the transport of compounds across the blood-brain barrier.
  • U.S. Pat. No. 5,124,146 discloses a method for delivery of therapeutic agents across the blood-brain barrier at sites of increase permeability associated with 5 brain lesions.
  • U.S. Pat. No. 5,153,179 discloses acylated glycerol and derivatives for use in a medicament for improved penetration of cell membranes.
  • U.S. Pat. No. 5,177,064 discloses the use of lipoidal phosphonate derivatives of nucleoside antiviral agents for delivery across the blood-brain barrier.
  • U.S. Pat. No. 5,254,342 discloses receptor-mediated transcytosis of the blood-brain barrier using the transferrin receptor in o combination with pharmaceutical compounds that enhance or accelerate this process.
  • U.S. Pat. No. 5,258,402 discloses treatment of epilepsy with imidate derivatives of anticonvulsive sulfamate.
  • U.S. Pat. No. 5,270,312 discloses substituted piperazines as central nervous system agents.
  • U.S. Pat. No. 5,284,876 discloses fatty acid conjugates of dopamine drugs.
  • U.S. Pat. No. 5,389,623 discloses the use of lipid dihydropyridine 5 derivatives of anti-inflammatory steroids or steroid sex hormones for delivery across the blood-brain barrier.
  • U.S. Pat. No. 5,405,834 discloses prodrug derivatives of thyrotropin releasing hormone.
  • U.S. Pat. No. 5,434,137 discloses methods for the selective opening of abnormal brain tissue capillaries using bradykinin infused into the carotid artery.
  • U.S. Pat. No. 5,442,043 discloses a peptide conjugate between a peptide having a biological 5 activity and incapable of crossing the blood-brain barrier and a peptide which exhibits no biological activity and is capable of passing the blood-brain barrier by receptor- mediated endocytosis.
  • U.S. Pat. No. 5,525,727 discloses compositions for differential uptake and retention in brain tissue comprising a conjugate o of a narcotic analgesic and agonists and antagonists thereof with a lipid form of dihydropyridine that forms a redox salt upon uptake across the blood-brain barrier that prevents partitioning back to the systemic circulation.
  • PCT Publication Number WO 91/04745 discloses transport across the blood-brain barrier using cell adhesion molecules and fragments thereof to increase the permeability of tight junctions in vascular endothelium.
  • PCT Publication Number WO 91/14438 discloses 5 the use of a modified, chimeric monoclonal antibody for facilitating transport of substances across the blood-brain barrier.
  • PCT Publication Number WO 94/01131 discloses lipidized proteins, including antibodies.
  • PCT Publication Number WO 94/03424 discloses the use of amino acid derivatives as drag conjugates for facilitating transport across the blood-brain barrier.
  • PCT Publication Number WO o 94/06450 discloses conjugates of neurologically-active drags with a dihydropyridine- type redox targeting moiety and comprising an amino acid linkage and an aliphatic residue.
  • PCT Publication Number WO 94/02178 discloses antibody-targeted liposomes for delivery across the blood-brain barrier.
  • PCT Publication Number WO 95/07092 discloses the use of drag-growth factor conjugates for delivering drags across the blood- 5 brain barrier.
  • PCT Publication Number WO 96/00537 discloses polymeric microspheres as injectable drag-delivery vehicles for delivering bioactive agents to sites within the central nervous system.
  • PCT Publication Number WO 96/04001 discloses omega- 3-fatty acid conjugates of neurologically-active drugs for brain tissue delivery.
  • PCT WO 96/22303 discloses fatty acid and glycerolipid conjugates of neurologically-active drugs for brain tissue delivery.
  • an ester, amide or hydrazide derivative of an agent of the invention for example, from the corresponding carboxylic acid and a suitable reagent.
  • a carboxylic acid- containing compound, or a reactive equivalent thereof may be reacted with a hydroxyl- containing compound, or a reactive equivalent thereof, so as to provide the corresponding ester.
  • a carboxylic acid- containing compound, or a reactive equivalent thereof may be reacted with a hydroxyl- containing compound, or a reactive equivalent thereof, so as to provide the corresponding ester.
  • the compound may also act from the periphery, causing a change in the equilibrium of the amyloid protein concentration in the two compartments i.e. (systemic vs central). In this case a compound may not be required to penetrate the brain to induce or decrease the concentration of A ⁇ in the brain (a "sink" effect)
  • the present invention is also related to prodrugs of the agents of the Formulae disclosed herein.
  • Prodrugs are agents which are converted in vivo to active forms ⁇ see, e.g., R.B. Silverman, 1992, "The Organic Chemistry of Drug Design and Drug Action,"
  • Prodrugs can be used to alter the biodistribution ⁇ e.g., to allow agents which would not typically enter the reactive site of the protease) or the pharmacokinetics for a particular agent.
  • a carboxylic acid group can be esterif ⁇ ed, e.g., with a methyl group or an ethyl group to yield an ester.
  • the ester is administered to a subject, the ester is cleaved, enzymatically or non-enzymatically, reductively, oxidatively, or hydrolytically, to reveal the anionic group.
  • An anionic group can be esterified with moieties ⁇ e.g., acyloxymethyl esters) which are cleaved to reveal an intermediate agent which subsequently decomposes to yield the active agent.
  • the prodrug moieties may be metabolized in vivo by esterases or by other mechanisms to carboxylic acids.
  • prodrugs examples are well known in the art ⁇ see, e.g., Berge, et al, "Pharmaceutical Salts", J. Pharm. ScL 66, 1-19 (1977)).
  • the prodrugs can be prepared in situ during the final isolation and purification of the agents, or by separately reacting the purified agent in its free acid form with a suitable derivatizing agent.
  • Carboxylic acids can be converted into esters via treatment with an alcohol in the presence of a catalyst.
  • cleavable carboxylic acid prodrug moieties include substituted and unsubstituted, branched or unbranched lower alkyl ester moieties, (e.g., ethyl esters, 5 propyl esters, butyl esters, pentyl esters, cyclopentyl esters, hexyl esters, cyclohexyl esters), lower alkenyl esters, dilower alkyl-amino lower-alkyl esters (e.g., dimethylaminoethyl ester), acylamino lower alkyl esters, acyloxy lower alkyl esters (e.g., pivaloyloxymethyl ester), aryl esters (phenyl ester), aryl-lower alkyl esters (e.g., benzyl ester), substituted (e.g., with methyl, halo, or methoxy substituents) aryl and o aryl
  • the present invention relates to pharmaceutical compositions comprising agents according to any of the Formulae herein for the 5 treatment of an amyloid- ⁇ related disease, as well as methods of manufacturing such pharmaceutical compositions.
  • the agents of the present invention may be prepared by the methods illustrated in the general reaction schemes as, for example, in the patents and patent applications refered to herein, or by modifications thereof, using readily available o starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants which are in themselves known, but are not mentioned here. Functional and structural equivalents of the agents described herein and which have the same general properties, wherein one or more simple variations of substituents are made which do not adversely affect the essential nature or the utility of 5 the agent.
  • the agents of the invention may be supplied in a solution with an appropriate solvent or in a solvent-free form (e.g., lyophilized).
  • the agents and buffers necessary for carrying out the methods of the invention may be packaged as a kit.
  • the kit may be commercially used according to the methods o described herein and may include instructions for use in a method of the invention.
  • Additional kit components may include acids, bases, buffering agents, inorganic salts, solvents, antioxidants, preservatives, or metal chelators.
  • the additional kit components are present as pure compositions, or as aqueous or organic solutions that incorporate one or more additional kit components. Any or all of the kit components optionally further comprise buffers.
  • the therapeutic agent may also be administered parenterally, intraperitoneally, intraspinally, or intracerebrally.
  • Dispersions can be prepared in glycerol, liquid 5 polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • the agent may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent.
  • suitable diluents include saline and aqueous buffer solutions.
  • Liposomes include water-in-oil-in- water CGF emulsions as well as conventional liposomes (Strejan et ah, J. Neuroimmunol. 7, 27 (1984)).
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the composition must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved o against the contaminating action of microorganisms such as bacteria and fungi.
  • Suitable pharmaceutically acceptable carriers include, without limitation, any non-immunogenic pharmaceutical adjuvants suitable for oral, parenteral, nasal, mucosal, transdermal, intravascular (IV), intraarterial (IA), intramuscular (IM), and subcutaneous (SC) administration routes, such as phosphate buffer saline (PBS).
  • the vehicle can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of o surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents are included, for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition.
  • Prolonged absorption of the injectable compositions can be brought about 5 by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
  • Sterile injectable solutions can be prepared by incorporating the therapeutic agent in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the therapeutic agent into a sterile vehicle 5 which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient (i.e., the therapeutic agent) plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the therapeutic agent can be orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the therapeutic agent and other ingredients may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • the therapeutic agent may be incorporated with excipients and used in the form of ingestible 5 tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • the percentage of the therapeutic agent in the compositions and preparations may, of course, be varied.
  • the amount of the therapeutic agent in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of therapeutic agent calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle.
  • the specification for the dosage unit forms of the invention are dictated by and 5 directly dependent on (a) the unique characteristics of the therapeutic agent and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such a therapeutic agent for the treatment of amyloid deposition in subjects.
  • the present invention therefore includes pharmaceutical formulations comprising o the agents of the Formulae described herein, including pharmaceutically acceptable salts thereof, in pharmaceutically acceptable carriers for aerosol, oral and parenteral administration.
  • the present invention includes such agents, or salts thereof, which have been lyophilized and which may be reconstituted to form pharmaceutically acceptable formulations for administration, as by intravenous, intramuscular, or 5 subcutaneous injection. Administration may also be intradermal or transdermal.
  • an agent of the Formulae described herein, and pharmaceutically acceptable salts thereof may be administered orally or through inhalation as a solid, or may be administered intramuscularly or intravenously as a solution, suspension or emulsion. Alternatively, the agents or salts may also be 5 administered by inhalation, intravenously or intramuscularly as a liposomal suspension.
  • compositions are also provided which are suitable for administration as an aerosol, by inhalation. These formulations comprise a solution or suspension of the desired agent of any Formula herein, or a salt thereof, or a plurality of solid particles of the agent or salt.
  • the desired formulation may be placed in a small o chamber and nebulized. Nebulization may be accomplished by compressed air or by ultrasonic energy to form a plurality of liquid droplets or solid particles comprising the agents or salts.
  • the liquid droplets or solid particles should have a particle size in the range of about 0.5 to about 5 microns.
  • the solid particles can be obtained by processing the solid agent of any Formula described herein, or a salt thereof, in any appropriate 5 manner known in the art, such as by micronization.
  • the size of the solid particles or droplets will be, for example, from about 1 to about 2 microns, hi this respect, commercial nebulizers are available to achieve this purpose.
  • a pharmaceutical formulation suitable for administration as an aerosol may be in the form of a liquid, the formulation will comprise a water-soluble agent of any Formula o described herein, or a salt thereof, in a carrier which comprises water.
  • a surfactant may be present which lowers the surface tension of the formulation sufficiently to result in the formation of droplets within the desired size range when subjected to nebulization.
  • Peroral compositions also include liquid solutions, emulsions, suspensions, and the like.
  • the pharmaceutically acceptable carriers suitable for preparation of such 5 compositions are well known in the art.
  • Typical components of carriers for syrups, elixirs, emulsions and suspensions include ethanol, glycerol, propylene glycol, polyethylene glycol, liquid sucrose, sorbitol, and water.
  • typical suspending agents include methyl cellulose, sodium carboxymethyl cellulose, tragacanth, and sodium alginate;
  • typical wetting agents include lecithin and polysorbate o 80; and typical preservatives include methyl paraben and sodium benzoate.
  • Peroral liquid compositions may also contain one or more components such as sweeteners, flavoring agents and colorants disclosed above.
  • compositions may also be coated by conventional methods, typically with pH or time-dependent coatings, such that the subject agent is released in 5 the gastrointestinal tract in the vicinity of the desired topical application, or at various times to extend the desired action.
  • dosage forms typically include, but are not limited to, one or more of cellulose acetate phthalate, polyvinylacetate phthalate, hydroxypropyl methyl cellulose phthalate, ethyl cellulose, waxes, and shellac.
  • compositions useful for attaining systemic delivery of the subject agents include sublingual, buccal and nasal dosage forms.
  • Such compositions typically 5 comprise one or more of soluble filler substances such as sucrose, sorbitol and mannitol; and binders such as acacia, macrocrystalline cellulose, carboxymethyl cellulose and hydroxypropyl methyl cellulose. Glidants, lubricants, sweeteners, colorants, antioxidants and flavoring agents disclosed above may also be included.
  • compositions of this invention can also be administered topically to a o subject, e.g., by the direct laying on or spreading of the composition on the epidermal or epithelial tissue of the subject, or transdermally via a "patch".
  • Such compositions include, for example, lotions, creams, solutions, gels and solids.
  • These topical compositions may comprise an effective amount, usually at least about 0.1%, or evan from about 1% to about 5%, of an agent of the invention.
  • Suitable carriers for topical 5 administration typically remain in place on the skin as a continuous film, and resist being removed by perspiration or immersion in water.
  • the carrier is organic in nature and capable of having dispersed or dissolved therein the therapeutic agent.
  • the carrier may include pharmaceutically acceptable emolients, emulsifiers, thickening agents, solvents and the like.
  • Active agents are administered at a therapeutically effective dosage sufficient to inhibit amyloid deposition in a subject.
  • a "therapeutically effective" dosage inhibits amyloid deposition by, for example, at least about 20%, or by at least about 40%, or even by at least about 60%, or by at least about 80% relative to untreated subjects.
  • a "therapeutically effective" dosage stabilizes 5 cognitive function or prevents a further decrease in cognitive function ⁇ i.e., preventing, slowing, or stopping disease progression).
  • the present invention accordingly provides therapeutic drugs.
  • therapeutic or “drag” is meant an agent having a beneficial ameliorative or prophylactic effect on a specific disease or condition in a living human or non-human animal.
  • active agents are administered at a therapeutically effective dosage sufficient to decrease deposition in a subject of amyloid protein, e.g., A/340 or A/342.
  • a therapeutically effective dosage inhibits amyloid deposition by, for example, at least about 15%, or by at least about 40%, or even by at least 60%, or at least by about 80% relative to untreated subjects.
  • active agents are administered at a therapeutically effective dosage sufficient to increase or enhance amyloid protein, e.g., A/340 or A/342, in the.blood of a subject
  • a therapeutically effective dosage increases the concentration by, for example, at least about 15%, or by at least about 40%, or even by at least 60%, or at least by about 80% relative to untreated subjects.
  • active agents are administered at a therapeutically effective dosage sufficient to improve ADAS-cog test scores by, e.g., at least about 1 point, at least about 2 points, at least about 3 points, at least about 4 points, at least about 5 points, at least about 10 points, at least about 12 points, at least about 15 points, or at least about 20 points relative to untreated subjects.
  • Toxicity and therapeutic efficacy of such agents can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED50, and usually a larger therapeutic index are more efficacious. While agents that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such agents to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • doses depend upon a number of factors within the ken of the ordinarily skilled physician, veterinarian, or researcher.
  • the dose(s) of the small molecule will vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable, and the effect which the practitioner desires the small molecule to have upon the nucleic acid or polypeptide of the invention.
  • Exemplary doses include milligram or microgram amounts of the small molecule per kilogram of subject or sample weight (e.g. , about 1 microgram per kilogram to about
  • appropriate doses depend upon the potency with respect to the expression or activity to be modulated. Such appropriate doses may be determined using the assays described herein.
  • a physician, - veterinarian, or researcher may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained.
  • the specific dose level for any particular animal subject will depend upon a variety of factors including the activity of the specific agent employed, the age, body weight, general health, gender, and diet of the subject, the time of administration, the route of administration, the rate of excretion, any drag combination, and the degree of expression or activity to be modulated.
  • the ability of an agent to inhibit amyloid deposition can be evaluated in an 5 animal model system that may be predictive of efficacy in inhibiting amyloid deposition in human diseases, such as a transgenic mouse expressing human APP or other relevant animal models where A ⁇ deposition is seen. Likewise, the ability of an agent to prevent or reduce cognitive impairment in a model system may be indicative of efficacy in humans. Alternatively, the ability of an agent can be evaluated by examining the ability o of the agent to inhibit amyloid fibril formation in vitro, e.g., using a fibrillogenesis assay such as that described herein, including a ThT, CD 5 or EM assay. Also the binding of an agent to amyloid fibrils may be measured using a MS assay as described herein.
  • Certain embodiments of the present agents can contain a basic functional group, 5 such as amino or alkylamino, and are, thus, capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable acids.
  • pharmaceutically acceptable salts refers to the relatively non-toxic, inorganic and organic acid addition salts of agents of the present invention. These salts can be prepared in situ during the final isolation and purification of the agents of the invention, or by separately o reacting a purified agent of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the hydrohalide (including hydrobromide and hydrochloride), sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, 5 succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate,
  • the agents of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically acceptable salts o with pharmaceutically acceptable bases.
  • pharmaceutically acceptable salts refers to the relatively non-toxic, inorganic and organic base addition salts of agents of the present invention. These salts can likewise be prepared in situ during the final isolation and purification of the agents, or by separately reacting the purified agent in its free acid form with a suitable base, such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically 5 acceptable organic primary, secondary or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylaniine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like.
  • ''Pharmaceutically acceptable salts also includes, for example, derivatives of agents modified by making acid or base salts thereof, as described further below and elsewhere in the present application.
  • pharmaceutically acceptable salts include mineral or organic acid salts of basic residues such as amines; and alkali or o organic salts of acidic residues such as carboxylic acids.
  • Pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent agent formed, for example, from non-toxic inorganic or organic acids.
  • Such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric acid; and the salts 5 prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, palmoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, and isethionic acid.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, and nitric acid
  • organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic,
  • salts may be synthesized from the parent agent which contains a basic or o acidic moiety by conventional chemical methods. Generally, such salts may be prepared by reacting the free acid or base forms of these agents with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two.
  • Test compounds were purchased from commercial sources or synthesized and screened by mass spectroscopy ("MS") assays.
  • MS assay gives data on the ability s of compounds to bind to an amyloid.
  • MS mass spectroscopy
  • the MS assay gives data on the ability of compounds to bind to soluble A ⁇ , whereas the ThT, EM and 5 CD assays give data on inhibition of fibrillogenesis.
  • the results from the assay for binding to A ⁇ are summarized in Table 2. "+++” indicates strong binding; “++” indicates moderate binding; “+” indicates weak binding; “-” indicates no detectable binding; and entries left blank were not determined.
  • test compound at 20 ⁇ M was incubated with 50 ⁇ M A ⁇ (l-40) fibers for 1 h at 37°C in Tris buffered saline (20 mM Tris, 150 mM NaCl, pH 7.4 containing 0.01 sodium azide). Following incubation, the solution was centrifuged for 20 min at 21,000 g to sediment the A ⁇ (l-40) fibers along with any bound test compound. The amount of test compound remaining in the supernatant was determined by reading the absorbance.
  • test compound bound was then calculated by comparing the amount remaining in the supernatants of incubations with A ⁇ to the amount remaining in control incubations which do not contain A ⁇ fibers.
  • Thioflavin T and Congo Red both of which are known to bind to A ⁇ fibers, are included in each assay run as positive controls.
  • test compounds were diluted to 40 ⁇ M, which is twice the concentration in the final test, and then scanned using the Hewlett Packard 8453 UV/VIS spectrophotometer to determine if the absorbance was sufficient for detection.
  • alkanesulfonic acid namely 3-amino-l-propanesulfonic acid
  • other therapeutic compounds used to diminish symptoms characteristic of Alzheimer's disease (e.g., loss of cognitive functions).
  • the examples comprise the use of an alkanesulfonic acid in combination with cognitive enhancers such as acetylcholine esterase inhibitors ("AChEi").
  • AChEi acetylcholine esterase inhibitors
  • Test alkanesulfonic acid Patients were treated with the test alkanesulfonic acid for a period of 9 months. A group of patients received the test compound alone while another group were treated with the test compound in combination with an AChEi, namely donepezil.
  • Alzheimer's patients were categorized as being “mild” or “moderate” according to their MMSE ("Mini Mental State Examination") score.
  • MMSE Minimum Mental State Examination
  • B.W.Rover, et al "Mini-mental state exam in clinical practice.” Hospital Practice 22(1 A), 99 et seq. (1987).
  • a MMSE score in the range of 19 to 26 was considered “mild,” and a score in the range 13 to 18 was considered “moderate”
  • the change in mental function of these patients was analyzed by using their ADAS-Cog scores, which were recorded periodically over a nine month period.
  • a positive change in ADAS-Cog score reflects a deterioration of cognitive function of a "mild AD" patient; stabilization is seen by a change of ⁇ 1; and a negative change shows an improvement in cognitive function.
  • the medical literature predicts that patients with Alzheimer's disease who do not receive any treatment will, on average, have a change in ADAS-cog score over nine months varying from +2.5 (for "mild” patients) to +5 (for "moderate” patients).
  • the alkanesulfonic acid used in the study is known to have an effect on the concentration of A ⁇ in the brain.
  • Test compound had a decrease of 34% in A ⁇ CSF concentration.
  • the test compound was previously shown to decrease the levels of both soluble and insoluble A ⁇ 42 in the brain of transgenic mice. Based on the mice studies o the test compound is hypothesized to favor the clearance of A ⁇ from the brain and CSF prior to its deposition. The decrease in A ⁇ CSF concentration seen in patients treated with the test compound is greater than that seen in the placebo group, where patients showed a non-significant increase of 15% in their A ⁇ CSF concentration. This result demonstrates a difference of 49% between the two groups.
  • Patients treated with the 5 triple therapy showed a decrease of 31 % while patients on AChEi and statin showed an increase of 45% in their A ⁇ 42 CSF levels. This triple therapy showed a greater effect on the change (-76%) of A ⁇ 42 CSF concentration when compared to the appropriate controls.
  • test o compound with AchEi and statin showed a much greater effect on the A ⁇ CSF concentration than test compound alone.
  • CSF was obtained from patients before and after treatment with the test compound at daily doses of 100 mg, 200 mg, or 300 mg.
  • CSF was fractionated by FPLC following treatment with formic acid, and then the A ⁇ containing fraction was 5 lyophilised.
  • the amount of A ⁇ peptide was measured using an ELISA assay
  • the test alkanesulfonic acid-containing composition was found to reduce the CSF level of A ⁇ when patients were treated with 200 or 300 mg daily doses. A majority of patients on placebo and on 100 mg daily doses showed stable A ⁇ CSF levels over a 3-month period, whereas the greatest reduction of A ⁇ occurred in patients o receiving 200 or 300 mg daily doses.
  • the presence of a drug in the cerebrospinal fluid suggests that the drug crosses the blood brain barrier to penetrate the brain.
  • the presence of the alkanesulfonic acid in the CSF was determined in patients who had received treatment for three months, hi these patients, CSF was collected five hours following dosing, and levels of alkanesulfonic acid were determined by LC-MS/MS. 5
  • the test alkanesulfonic acid was found to be present in CSF of patients in a dose- dependent manner, e.g., patients receiving 200 or 300 mg daily dosing had a greater concentration than that seen in patients with 100 mg daily dosing.
  • Cognitive function with combination therapy - alkanesulfonic acid plus an acetyl cholenesterase inhibitor Mild to moderate Alzheimer's disease patients who had been on cognition enhancers (AriceptTM or ExelonTM) were co-medicated with daily doses of test drug (300 mg alkanesulfonic acid) for six months. At the time the experiments were undertaken the peer-reviewed medical literature indicated that patients who had been on AChE inhibitors for more than twelve months would be expected to show a decline in cognitive functions of at least two to three points on the ADAS-Cog scale over a six month period.

Abstract

La présente invention a trait à des procédés et des compositions pharmaceutiques pour le traitement de maladies associées au bêta-amyloïde, y compris la maladie d'Alzheimer. L'invention a trait, par exemple, à un procédé de traitement thérapeutique concomitant d'un sujet, comprenant l'administration d'une quantité efficace d'un premier agent et d'un deuxième agent, ledit premier agent traitant une maladie associée au bêta-amyloïde, la dégénérescence, ou la toxicité cellulaire; et ledit deuxième agent étant un médicament thérapeutique ou un supplément nutritif.
EP05858504A 2004-06-18 2005-06-17 Formulations therapeutiques pour le traitement de maladies associees au beta-amyloide Withdrawn EP1841460A2 (fr)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US87161304A 2004-06-18 2004-06-18
US10/871,514 US7414076B2 (en) 2003-06-23 2004-06-18 Methods and compositions for treating amyloid-related diseases
US10/871,512 US20050038000A1 (en) 2003-06-23 2004-06-18 Methods and compositions for the treatment of amyloid-and epileptogenesis-associated diseases
US10/871,549 US20050142191A1 (en) 2003-06-23 2004-06-18 Pharmaceutical formulations of amyloid inhibiting compounds
US10/871,537 US20050031651A1 (en) 2002-12-24 2004-06-18 Therapeutic formulations for the treatment of beta-amyloid related diseases
US10/871,365 US7244764B2 (en) 2003-06-23 2004-06-18 Methods and compositions for treating amyloid-related diseases
US10/871,543 US7253306B2 (en) 2003-06-23 2004-06-18 Pharmaceutical drug candidates and methods for preparation thereof
PCT/IB2005/004199 WO2007049098A2 (fr) 2004-06-18 2005-06-17 Formulations therapeutiques pour le traitement de maladies associees au beta-amyloide

Publications (1)

Publication Number Publication Date
EP1841460A2 true EP1841460A2 (fr) 2007-10-10

Family

ID=37968188

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05858504A Withdrawn EP1841460A2 (fr) 2004-06-18 2005-06-17 Formulations therapeutiques pour le traitement de maladies associees au beta-amyloide

Country Status (4)

Country Link
EP (1) EP1841460A2 (fr)
JP (1) JP2008504372A (fr)
CA (1) CA2582385A1 (fr)
WO (1) WO2007049098A2 (fr)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009019534A2 (fr) 2006-10-12 2009-02-12 Bellus Health (International) Limited Procédés, composés, compositions et véhicules permettant de délivrer l'acide 3-amino-1-propane sulfonique
WO2010040232A1 (fr) * 2008-10-09 2010-04-15 Waratah Pharmaceuticals Inc. Utilisation de scyllo-inositols pour le traitement de troubles liés à la dégénérescence maculaire
EP2349988A4 (fr) * 2008-11-14 2012-05-02 Bellus Health Inc Sels nutritifs organiques, leurs procédés de préparation et leurs utilisations
WO2010096925A1 (fr) * 2009-02-26 2010-09-02 Bellus Health (International) Limited Matériau comestible complété et/ou enrichi en homotaurine, procédés de préparation et utilisations
PL3061821T3 (pl) 2009-07-22 2020-01-31 PureTech Health LLC Kompozycje do leczenia zaburzeń łagodzonych przez aktywację receptora muskarynowego
US10265311B2 (en) 2009-07-22 2019-04-23 PureTech Health LLC Methods and compositions for treatment of disorders ameliorated by muscarinic receptor activation
US10052299B2 (en) 2009-10-30 2018-08-21 Retrotope, Inc. Alleviating oxidative stress disorders with PUFA derivatives
KR102020611B1 (ko) 2011-04-26 2019-09-10 레트로토프 인코포레이티드 Pufa와 관련된 신경퇴행성 질환 및 근육 질환
EP2701698B1 (fr) 2011-04-26 2020-12-23 Retrotope, Inc. Troubles impliquant l'oxydation des acides gras polyinsaturés (pufa)
EP3730135A1 (fr) 2011-04-26 2020-10-28 Retrotope, Inc. Troubles de traitement compromis de l'énergie et déficiences mitochondriales
KR102110200B1 (ko) 2011-04-26 2020-05-13 레트로토프 인코포레이티드 산화성 망막 질환
RU2488388C1 (ru) * 2012-05-24 2013-07-27 Ооо "Валента Интеллект" Фармацевтическая композиция для профилактики и лечения психических, поведенческих, когнитивных расстройств
CN102793694B (zh) * 2012-08-14 2014-06-04 武汉华纳联合药业有限公司 3-氨基-1-丙烷磺酸及其衍生物在制备治疗脑卒中的药物中的应用
JP7048976B2 (ja) 2015-11-23 2022-04-06 レトロトップ、 インコーポレイテッド 1,4-ジエン系の部位特異的同位体標識
ITUA20161679A1 (it) * 2016-03-15 2017-09-15 Neuraxpharm Italy S P A Composizione per prevenire e trattare patologie neurodegenerative e disturbi cognitivi
KR102006777B1 (ko) 2018-01-29 2019-10-08 주식회사 종근당 에스오메프라졸 및 탄산수소나트륨을 포함하는 약제학적 제제
KR102080023B1 (ko) * 2018-01-29 2020-02-21 주식회사 종근당 에스오메프라졸 및 탄산수소나트륨을 포함하는 안정한 약제학적 조성물
IL277071B1 (en) 2018-03-08 2024-03-01 Incyte Corp Aminopyrizine diol compounds as PI3K–y inhibitors
JP7053882B2 (ja) * 2018-04-12 2022-04-12 ザ ジェイ. デビッド グラッドストーン インスティテューツ、 ア テスタメンタリー トラスト エスタブリッシュド アンダー ザ ウィル オブ ジェイ. デビッド グラッドストーン Apoe4/4に関連した障害を治療するための医薬組成物およびキット
US11046658B2 (en) 2018-07-02 2021-06-29 Incyte Corporation Aminopyrazine derivatives as PI3K-γ inhibitors
BR112021005802B1 (pt) 2018-09-28 2022-02-15 Karuna Therapeutics, Inc Composição farmacêutica oral
IL295783A (en) 2020-02-21 2022-10-01 Retrotope Inc Processes for isotopic modification of polyunsaturated fatty acids and their derivatives

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR0016652A (pt) * 1999-12-23 2002-11-19 Neurochem Inc Métodos para inibir angiopatia amilóide cerebral, para tratar um estado de doença caracterizado por angiopatia amilóide cerebral em um indivìduo, e para diagnosticar angiopatia amilóide cerebral em um indivìduo
CA2511599A1 (fr) * 2002-12-24 2004-07-15 Neurochem (International) Limited Formulations therapeutiques destinees au traitement de maladies liees au peptide beta-amyloide

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2007049098A2 *

Also Published As

Publication number Publication date
WO2007049098A3 (fr) 2007-10-04
CA2582385A1 (fr) 2005-12-18
JP2008504372A (ja) 2008-02-14
WO2007049098A2 (fr) 2007-05-03

Similar Documents

Publication Publication Date Title
US20050031651A1 (en) Therapeutic formulations for the treatment of beta-amyloid related diseases
WO2007049098A2 (fr) Formulations therapeutiques pour le traitement de maladies associees au beta-amyloide
AU2003291910B2 (en) Therapeutic formulations for the treatment of beta-amyloid related diseases
CN100571701C (zh) 治疗β淀粉样蛋白相关疾病的治疗性制品
US7244749B2 (en) Compositions and methods for lowering plasma lipoprotein(a) and risk factors of cardiovascular diseases
KR102051030B1 (ko) 소아 담즙울체성 간 질환 치료용 담즙산 재순환 억제제
ES2275007T3 (es) Uso de derivados de azetidinona sustitudos en el tratamiento de la enfermedad de alzeimer.
JP4711600B2 (ja) シトステロール血症の処置のための置換アゼチジノン化合物の使用
DE60224163T2 (de) Kombinationen von ezetimibe und aspirin zur behandlung von kardiovaskulären erkrankungen
JP2004517919A (ja) 血管状態の処置のための心臓血管薬剤とステロール吸収インヒビターとの組み合わせ
JP2008514718A (ja) 置換アゼチドノンおよびcb1アンタゴニストの組み合わせ
US6235706B1 (en) Combination therapy for reducing the risks associated with cardiovascular disease
AU2637895A (en) Compositions for treatment of chronic inflammatory diseases
PT1429756E (pt) Tratamento de xantoma com derivados de azetidinona como inibidores de absorção de esteróis
CA2265827A1 (fr) Traitement combine destine a reduire les risques de maladies cardio-vasculaires
JP2004517916A (ja) ニコチン酸およびその誘導体ならびにステロール吸収阻害剤の併用、および血管適応症の治療
CN109476686B (zh) sGC刺激剂的磷前药
WO2014087231A2 (fr) Composition pharmaceutique possédant une action neuro-protectrice, nootrope et anti-amnésique, et méthode de prévention et/ou de traitement d'états liés à une perturbation du flux sanguin dans le cerveau due à des maladies cérébrovasculaires et neurodégénératives
RU2703729C2 (ru) Комбинация и набор с анксиолитическим действием
CA2541815A1 (fr) Procedes et moyens pour moduler le metabolisme lipidique

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070111

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR LV MK YU

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/27 20060101ALI20071024BHEP

Ipc: A61P 25/28 20060101ALI20071024BHEP

Ipc: A61K 31/445 20060101ALI20071024BHEP

Ipc: A61K 45/06 20060101ALI20071024BHEP

Ipc: A61K 31/185 20060101AFI20071024BHEP

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1109857

Country of ref document: HK

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

Owner name: BELLUS HEALTH (INTERNATIONAL) LIMITED

17Q First examination report despatched

Effective date: 20080723

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

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

18D Application deemed to be withdrawn

Effective date: 20090203

RBV Designated contracting states (corrected)

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU MC NL PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1109857

Country of ref document: HK