AU2003251418A1 - Amidine derivatives for treating amyloidosis - Google Patents

Amidine derivatives for treating amyloidosis Download PDF

Info

Publication number
AU2003251418A1
AU2003251418A1 AU2003251418A AU2003251418A AU2003251418A1 AU 2003251418 A1 AU2003251418 A1 AU 2003251418A1 AU 2003251418 A AU2003251418 A AU 2003251418A AU 2003251418 A AU2003251418 A AU 2003251418A AU 2003251418 A1 AU2003251418 A1 AU 2003251418A1
Authority
AU
Australia
Prior art keywords
group
amyloid
pct
groups
compounds
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2003251418A
Inventor
David Boykin
Robert J. Chalifour
Xianqi Kong
Wenshuo Lu
Richard R. Tidwell
Xinfu Wu
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.)
University of North Carolina at Chapel Hill
Georgia State University Research Foundation Inc
Original Assignee
University of North Carolina at Chapel Hill
University of North Carolina System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of North Carolina at Chapel Hill, University of North Carolina System filed Critical University of North Carolina at Chapel Hill
Publication of AU2003251418A1 publication Critical patent/AU2003251418A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/155Amidines (), e.g. guanidine (H2N—C(=NH)—NH2), isourea (N=C(OH)—NH2), isothiourea (—N=C(SH)—NH2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/343Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide condensed with a carbocyclic ring, e.g. coumaran, bufuralol, befunolol, clobenfurol, amiodarone
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • 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
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • 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
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/08Antibacterial agents for leprosy
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C257/00Compounds containing carboxyl groups, the doubly-bound oxygen atom of a carboxyl group being replaced by a doubly-bound nitrogen atom, this nitrogen atom not being further bound to an oxygen atom, e.g. imino-ethers, amidines
    • C07C257/10Compounds containing carboxyl groups, the doubly-bound oxygen atom of a carboxyl group being replaced by a doubly-bound nitrogen atom, this nitrogen atom not being further bound to an oxygen atom, e.g. imino-ethers, amidines with replacement of the other oxygen atom of the carboxyl group by nitrogen atoms, e.g. amidines
    • C07C257/18Compounds containing carboxyl groups, the doubly-bound oxygen atom of a carboxyl group being replaced by a doubly-bound nitrogen atom, this nitrogen atom not being further bound to an oxygen atom, e.g. imino-ethers, amidines with replacement of the other oxygen atom of the carboxyl group by nitrogen atoms, e.g. amidines having carbon atoms of amidino groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • C07D235/30Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/32Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Cardiology (AREA)
  • Oncology (AREA)
  • Pulmonology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Communicable Diseases (AREA)
  • Dermatology (AREA)
  • Hematology (AREA)
  • Hospice & Palliative Care (AREA)
  • Urology & Nephrology (AREA)
  • Immunology (AREA)
  • Rheumatology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pain & Pain Management (AREA)
  • Obesity (AREA)
  • Psychiatry (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Vascular Medicine (AREA)

Description

WO 03/103598 PCT/US03/17992 AMIDINE DERIVATIVES FOR TREATING AMYLOIDOSIS RELATED APPLICATION This application claims the priority of U.S. Provisional Patent Application No. 5 60/3 87,001, filed June 7, 2002, which is incorporated herein by reference. BACKGROUND OF THE INVENTION 10 Amyloidosis 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 deposits have common morphologic properties, stain with specific dyes (e.g., Congo red), and have 15 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-related diseases can either be restricted to one organ or spread to 20 several organs. The first instance is referred to as "localized amyloidosis" while the second is referred to as "systemic amyloidosis." Some amyloidotic diseases can be idiopathic, but most of these diseases appear as a complication of a previously existing disorder. For example, primary 25 amyloidosis can appear without any other pathology or can follow plasma cell dyscrasia or multiple myeloma. 1 WO 03/103598 PCT/US03/17992 Secondary amyloidosis is usually seen associated with chronic infection (such as tuberculosis) or chronic inflammation (such as rheumatoid arthritis). A familial form of secondary amyloidosis is also seen in Familial Mediterranean Fever (FMF). This familial type of amyloidosis, as one of the other types of familial amyloidosis, is genetically -5 inherited and is found in specific population groups. In these two types of amyloidosis, deposits are found in several organs and are thus considered systemic amyloid diseases. Another type of systemic amyloidosis is found in long-term hemodialysis patients. In each of these cases, a different amnyloidogenic protein is involved in amyloid deposition. "Localized amyloidoses" are those that tend to involve a single organ system. 10 Different amyloids are also characterized by the type-of protein present-in the deposit. For example, neurodegenerative diseases such as scrapie, bovine spongiform encephalitis, Creutzfeldt-Jakob disease, and the like are characterized by the appearance and accumulation of a protease-resistant fonn of a prion protein (referred to as AScr or PrP-27) in the central nervous system. Similarly, Alzheimet's disease, another neurodegenerative 15 disorder, is characterized by neuritic plaques and neurofibrillary tangles. In this case, the plaque and blood vessel amyloid is formed by the-deposition of fibrillary As amyloid protein. Other diseases such as adult-onset diabetes (Type II diabetes) are characterized by the localized accumulation of amyloid in the pancreas. Once these amyloids have formed, there is no known, widely accepted therapy or 20 treatment which significantly dissolves amyloid deposits in situ. Each amyloidogenic protein has the ability to organize into p-sheets and to form insoluble fibrils which may be deposited extracellularly or intracellularly.. Each amyloidogenic protein, although different in amino acid sequence, has the same property of forming fibrils and binding to other elements such as proteoglycan, amyloid P and 25 complement component. Moreover, each amyloidogenic protein has amino acid sequences which, although different, will show similarities such as regions with the ability to bind to the glycosaminoglycan (GAG) portion ofproteoglycan (referred to as the GAG binding site) as well as other regions which will promote p-sheet formation. 2 WO 03/103598 PCT/US03/17992 In specific cases, amyloidotic fibrils, once deposited, can become toxic to the surrounding cells. For example, the AP fibrils organized as senile plaques have been shown to be associated with dead neuronal cells and microgliosis in patients with Alzheimer's disease. When tested in vitro, A[s peptide was shown to be capable of triggering an 5 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. In another type of amnyloidosis seen in patients with Type II diabetes, the amyloidogenic protein IAPP has been shown to induce p-islet cell toxicity in vitro.- Hence, appearance of IAPP fibrils in the pancreas of Type II diabetic patients contributes to the loss 10 of the p islet cells (Langerhans) and organ dysfunction. People suffering from Alzheimer's disease develop a progressive dementia in adulthood, accompanied by three main structural changes in the brain: diffuse loss of neurons in multiple parts of the brain; accumulation of intracellular protein deposits termed neurofibrillary tangles; and accumulation of extracellular protein deposits termed amyloid 15 or senile plaques,. surrounded by misshapen nerve terminals (dystrophic neurites). A main constituent of these amyloid plaques is the amyloid-p peptide (Ap), a 39-43 amino-acid protein that is produced through cleavage of the P-amyloid precursor protein (APP). Although symptomatic treatments exist for Alzheimer's disease, this disease cannot be prevented or cured at this time. 20 - SUMMARY OF THE INVENTION The present invention relates to the use of amidine compounds in the treatment of amyloid-related diseases. hi particular, the invention relates to a method of treating or preventing an amyloid-related disease in a subject comprising administering to the subject a 25 therapeutic amount of an amidine compound. Among the compounds for use in the invention are those according to the following Formula, such that, when administered, amyloid fibril formation, neuro degeneration, or cellular toxicity is reduced or inhibited: 3 WO 03/103598 PCT/US03/17992 R a-N N--Ra 2 \ Y 1 A Y 2 Rb1-N N-Rb 2 ~' c2 R R q (Formula X) BRIEF DESCRIPTION OF DRAWINGS 5 FIG..1 -Effect of pentamidine-type compounds on Ap(l-40) assembly determined by ThT assay. FIG. 2- Effect of pentamidine-like compounds on Ap(1-40) assembly determined by ThT assay. FIG. 3 -Effect of amidine-type compounds on Ap(1-40) assembly determined by ThT LO assay. FIG. 4 -Effect of pentamidine-type compounds on IAPP assembly determined by ThT assay. DETAILED DESCRIPTION OF THE INVENTION .5 The present invention relates to the use of amidine compounds in the treatment of amyloid-related diseases. Amyloid-Related Diseases AA (reactive) Amyloidosis Generally, AA amyloidosis is a manifestation of a number of diseases that provoke 0 a sustained acute phase response. Such diseases include chronic inflammatory disorders, ' chronic local or systemic microbial infections, and malignant neoplasms. 4 WO 03/103598 PCT/US03/17992 AA fibrils are generally composed of 8,000 Dalton fragments (AA peptide or protein) formed by proteolytic cleavage of serum amyloid A protein (ApoSAA), a circulating apolipoprotein which once secreted is complexed with HDL and which is synthesized in hepatocytes in response to such cytokines as IL-1, L-6 and TNF. 5 Deposition can be widespread in the body, with a preference for parenchymal organs. The spleen is usually a deposition site, and the kidneys may also be affected. Deposition is also common in the heart and gastrointestinal tract. AA amyloid diseases include, but are not limited to inflammatory diseases, such as rheumatoid arthritis, juvenile chronic arthritis, ankylosing spondylitis, psoriasis, psoriatic 10 arthropathy, Reiter's syndrome, Adult Still's disease, Behoet's syndrome, and Crohn's disease. AA deposits are also produced as a result of chronic microbial infections, such as leprosy, tuberculosis, bronchiectasis, decubitus ulcers,-cbronic pyelonephritis, osteomyelitis, and Whipple's disease. Certain malignant neoplasms can also result in AA fibril amyloid deposits. These include such conditions as Hodgkin's lymphoma, renal carcinoma, 15 carcinomas. of gut, lung and urogenital tract, basal cell carcinoma, and hairy cell leukemia. AL Amyloidoses AL amyloid deposition is generally associated with almost any dyscrasia of the B lymphocyte lineage, ranging from malignancy of plasma cells (multiple myeloma) to benign monoclonal gammopathy. At times, the presence of amyloid deposits may be a primary Z0 indicator of the underlying dyscrasia. Fibrils of AL amyloid deposits are composed ofmonoclonal immunoglobulin light chains or fragments thereof. More specifically, the fragments are derived from the N terminal region of the light chain (kappa or lambda) and contain all or part of the variable (VL) domain thereof. Deposits generally occur in the mesenchymal tissues, causing t5 peripheral and autonomic neuropathy, carpal tunnel syndrome, macroglossia, restrictive cardiomyopathy, arthropathy of large joints, immune dyscrasias, myelomas, as well as occult dyscrasias. However, it should be noted that almost any tissue, particularly visceral organs such as the heart, may be involved., 5 WO 03/103598 PCT/US03/17992 Hereditary Systemic Amyloidoses There are many forms of hereditary systemic amyloidoses. Although they are relatively rare conditions, adult onset of symptoms and their inheritance patterns (usually autosomal dominant) lead to persistence of such disorders in the general population. 5 Generally, the syndronies are attributable to point mutations in the precursor protein leading to production of variant amyloidogenic peptides or proteins. Table 1 summarizes the fibril* composition of exemplary forms of these disorders. Table 1 Fibril Peptide/Protein Genetic variant Clinical Syndrome Transthyretin and fragments Met3O, many others Familial amyloid (ATTR) polyneuropathy (FAP), (Mainly peripheral nerves) Transthyretin and fragments Thr45, Ala6O, Ser84, Metl 11, Cardiac involvement (ATTR) Ile122 predominant without neuropathy N-terminal fragment of Arg26 'Familial amyloid Apolipoprotein Al (apoAI) polyneuropathy (FAP), (mainly peripheral nerves) N-terminal fragment of Arg26, Arg5O, Arg 60, others Ostertag-type, non-neuropathic Apoliproprotein Al (AapoAI) (predominantly visceral involvement) Lysozyme (Alys) Thr56, His67 Ostertag-type, non-neuropathic (predominantly visceral involvement) Fibrogen V chain fragment Leu554, Val 526 Cranial neuropathy with lattic' corneal dystrophy Gelsolin fragment (Agel) Asn187, Tyr187 Cranial neuropathy with lattice corneal dystrophy Cystatin C fragment Glu68 Hereditary cerebral hemorrhage (cerebral amyloid angiopathy) Icelandic type p-amyloid protein (Ap) derived Gln693 Hereditary cerebral hemorrhage from Amyloid Precursor Protein (cerebral amyloid angiopathy) (APP) Dutchtype p-amyloid protein (Ap) derived Ile717, Phe717, Gly717 Familial Alzheimer's Disease from Amyloid Precursor Protein (APP) p-amyloid protein (AP) derived Asn670, Leu671 Familial Dementia- probably from Amyloid Precursor Protein Alzheimer's Disease (APP) Prion Protein (PrP) derived from Leu102, Va1167, Asn178, Familial Creutzfeldt-Jakob Prp precursor protein Lys200 disease; Gerstmann-Straussler 51-91 insert Scheinker syndrome (hereditary spongiform encephalopathies; prion diseases) AA derived from Serum Familial Mediterranean fever, ayloid A protein (ApoSAA) predominant renal involvement (autosomal recessive) AA derived from Serum Muckle-Well's syndrome, anyloid A protein (ApoSAA) Inepbropathy, deafness, urticaria, 6 WO 03/103598 PCT/US03/17992 limb pain Unknown Cardiomyopathy with persistent atrial standstill Unknown Cutaneous deposits (bullous, paular, pustulodermal) Data derived froni Tan SY, Pepys MB. Amyloidosis. Histopathology, 25(5), 403-414 (Nov 1994). The data provided in Table 1 are exemplary and are not intended to limit the scope of the invention. For example, more than 40 separate point mutations in the transthyretin 5 gene have been described, all of which give rise to clinically similar forms of familial amyloid polyneuropathy. Transthyretin (TTR) is a 14 iloDalton protein that is also sometimes referred to as prealbumin. It is produced by the liver and choroid plexus, and it functions in transporting thyroid honnones and vitamin A. At least 50 variant forms of the protein, each 10 characterized by a single amino acid change, are responsible for various forms of familial amyloid polyneuropathy. For example, substitution of praline for leucine at position 55 results in a particularly progressive form of neuropathy; substitution of methionine for leucine at position 111 resulted in a severe cardiopathy in Danish patients. Amyloid deposits isolated from heart tissue of patients with systemic amyloidosis 15 have revealed that the deposits are composed of a heterogeneous mixture of TTR and fragments thereof, collectively referred to as ATTR, the full length sequences of which have been characterized. ATTR fibril components can be extracted from such plaques and their structure and sequence determined according to the methods Imown in the art (e.g., Gustavsson, A., et al., Laboratory Invest. 73: 703-708, 1995; Kametani, F., et al., Biochem. ZO Biophys. Res. Common. 125: 622-628, 1984; Pras, M., et al., PNAS 80: 539-42, 1983). Persons having point niutations in the molecule apolipoprotein Al (e.g., Gly->Arg26; Trp -> Arg5O; Leu -+ Arg6O) exhibit a form of amyloidosis ("Ostertag type") characterized by deposits of the protein apolipoprotein AI or fragments thereof (AApoAI). These patients have low levels of high density lipoprotein (HDL) and present with a !5 peripheral neuropathy or renal failure. 7 WO 03/103598 PCT/US03/17992 A mutation in the alpha chain of the enzyme lysozyme (e.g., Ile->Thr56 or Asp-+His57) is the basis of another form of bstertag-type non-neuropathic hereditary amyloid reported in English families. Here, fibrils of the mutant lysozyme protein (Alys) are deposited, and patients generally exhibit impaired renal function. This protein, unlike 5 most of the fibril-forning'proteins described herein, is usually present in whole (unfragmented) form (Benson, M.D., et al. CIBA Fdn. Symp. 199: 104-131, 1996). Q-amyloid peptide (AP) is a 39-43 amino acid peptide derived by proteolysis from a large protein known as Beta Amyloid Precursor protein (pAPP). Mutations in @APP result in familial forms of Alzheimer's disease, Down's syndrome'or senile dementia, 10 characterized by cerebral deposition of plaques composed of AP fibrils and other components, which are described in further detail below. Known mutations in APP associated with Alzheimer's disease occur proximate to the cleavage sites of P or gamma secretase, or within Ap. For example, position 717 is proximate to the site of gamma secretase cleavage of APP in its processing to As, and positions 670/671 are proximate to [5 the site of p-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 AD generated from APP. The structure and sequence of As peptides of various lengths are well known in the art. Such peptides can be made according to methods known in the art (e.g., Glenner and !0 Wong, Biochem Biophys. Res. Comm. 129: 885-890, 1984; Glenner and Wong, Biochem Biophys. Res. Comm. 122: 113 1-1135, 1984). In addition, various forms of the peptides are commercially available. As used herein, the term "p amyloid" or "amyloid-p" refer to amyloid P proteins or peptides, amyloid s precursor proteins or peptides, intermediates, and modifications and ,5 fragments thereof, unless otherwise specifically indicated. In particular, "Ap" refers to any peptide produced by proteolytic processing of the APP gene product, especially peptides which are associated with amyloid pathologies, including A01-3 9 , Ap 1
.
4 o, A013 4 1 , Ap1.
42 , and Ap1I43. 8 WO 03/103598 PCT/US03/17992 For convenience of nomenclature, "Api-42" maybe referred to herein as "AP(1-42) or simply as "Ap 42 " or "Ap42" (and likewise for any other amyloid peptides discussed herein). As used herein, the terms "P amyloid," "amyloid-p," and "Ap" are synonymous. Unless otherwise specified, the term "amyloid" refers to amyloidogenic proteins, 5 peptides, or fragments thereof which can be soluble (e.g.,.monomeric or oligomeric) or insoluble (e.g., having fibrillary structure or in amyloid plaque). Gelsolin is a calcium binding protein that binds to fragments and actin filaments. Mutations at position 187 (e.g., Asp->Asn; Asp->Tyr) of the protein result in a form of hereditary systemic amyloidosis, usually found in patients from Finland, as well as persons 10 of Dutch or Japanese origin. In afflicted individuals, fibrils formed from gelsolin fragments (Agel), usually consist of amino acids 173-243 (68 kDa carboxyterminal fragment) and are deposited in blood vessels and basement membranes, resulting in corneal dystrophy and cranial neuropathy which progresses to peripheral neuropathy, dystrophic skin changes and deposition in other organs. (Kangas, H., et al. Human Mol. Genet. 5(9): 1237-1243, 1996). 5 Other mutated proteins, such as mutant alpha chain of fibrinogen (AfibA) and mutant cystatin C (Acys) also form fibrils and produce characteristic hereditary disorders. AfibA fibrils form deposits characteristic of a nonneuropathic hereditary amyloid with renal disease; Acys deposits are characteristic of a hereditary cerebral amyloid angiopathy reported in Iceland (Isselbacher, Harrison's Principles of Internal Medicine, McGraw-Hill, 0 San Francisco, 1995; Benson, et al.). In at least some cases, patients with cerebral amyloid angiopathy (CAA) have been shown to have amyloid fibrils containing a non-mutant form of cystatin C in conjunction with amyloid beta protein (Nagai, A., et al. Molec. Chem. Neuropathol. 33: 63-78, 1998). Certain forms of prion disease are now considered to be heritable, accounting for up 5 to -15% of cases, which were previously thought to be predominantly infectious in nature. (Baldwin, et al., in Research Advances inAlzheiner's Disease and.Related Disorders, John Wiley and Sons, New York, 1995). In such prion disorders, patients develop plaques composed of abnormal isoforms of the normal prion protein (PrPS). 9 WO 03/103598 PCT/US03/17992 A predominant mutant isoform, PrPS", also referred to as. AScr, differs from the normal cellular protein in its resistance to protease degradation, insolubility after detergent extraction, deposition in secondary lysosomes, post-translational synthesis, and high p pleated sheet content. Genetic linkage has been established for at least five mutations 5 resulting in Creutzfeldt-lacob disease (CJD), Gerstmann-Straussler-Scheinker syndrome (GSS), and fatal familial insomnia (FFI). (Baldwin, supra) Methods for extracting fibril peptides from scrapie fibrils, determining sequences and making such peptides are known in the art (e.g., Beekes, M., et al, J. Gen. Virol. 76: 2567-76, 1995). For example, one form of GSS has been linked to i PrP mutation at codon 102, L0 while telencephalic GSS segregates with a mutation at codon 117. Mutations at codons 198 and 217 result in a form of GSS in which neuritic plaques characteristic of Alzheimer's disease contain PrP instead of As peptide. Certain forms of familial CJD have been associated with mutations at codons 200 and 210; mutations at codons 129 and 178 have been found in both familial CJD and FFI. (Baldwin, supra). .5 Senile Systemic Amyloidosis Amyloid deposition, either systemic or focal, increases with age. For example, fibrils of wild type transthyretin (TTR) are commonly found in the heart tissue of elderly individuals. These may be asymptomatic, clinically silent, or may result in heart failure. Asymptomatic fibrillar focal deposits may also occur in the brain (AP), corpora amylacea of 0 the prostate (Ap 2 microglobulin), joints and seminal vesicles. Cerebral Amyloidosis 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 AP peptide fibrils, resulting in dementia or sporadic (non-hereditary) Alzheimer's disease. In 5 fact, the incidence of sporadic Alzheimer's disease greatly exceeds forms shown to be hereditary. Fibril peptides forming these plaques are very similar to those described above, with reference to hereditary forms of Alzheimer's disease (AD). 10 WO 03/103598 PCT/US03/17992 Cerebral amyloid angiopathy (CAA) refers to the specific deposition of amyloid fibrils in the walls of leptomingeal and cortical arteries, arterioles and in 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 5 Frangione et al., Amyloid: .Protein Folding Disord. 8, Suppl. 1, 36-42 (2001)). CAA can occur sporadically or be hereditary. Multiple mutation sites in either AP or the APP gene have been identified and are clinically associated with either dementia or cerebral hemorrhage. Exemplary CAA disorders include, but are not limited to, hereditary cerebral hemorrhage with amyloidosis of Icelandic type (HCHWA-I); the Dutch variant of HCHWA [0 (HCHWA-D; a mutation in AP); the Flemish mutation of AP; the Arctic mutation of Ap; the Italian mutation of Ap; the Iowa mutation of Ap; familial British dementia; and familial Danish dementia. Dialysis-related Amyloidosis Plaques composed of P2 microglobulin (Ap 2 M) fibrils commonly develop in patients .5 receiving long term hemodialysis or peritoneal dialysis. p2 microglobulin is a 11.8 IdloDalton polypeptide and is the light chain of Class I MHC antigens, which are present on all nucleated cells. Under normal circumstances, it is continuously shed from cell membranes and is normally filtered by the kidney. Failure of clearance, such as in the case of impaired renal function, leads to deposition in the kidney and other sites (primarily in .0 collagen-rich tissues of the joints). Unlike other fibril proteins, Ap 2 M molecules are generally present in unfragmented form in the fibrils. (Benson, supra). Islet Amyloid Polypeptide and Diabetes Islet hyalinosis (amyloid deposition) was first described over a century ago as the presence of fibrous protein aggregates in the pancreas of patients with severe hyperglycemia 5 (Opie, EL., JExp. Med. 5: 397-428, 1990). Today, islet amyloid, composed predominantly of islet amyloid polypeptide (IAPP), or amylin, is a characteristic histopathological marker in over 90% of all cases of Type II diabetes (also known as Non-Insulin Dependent Diabetes, or NIDDM). These fibrillar accumulations result from the aggregation of the islet, amyloid polypeptide (IAPP) or'amylin, which is a 37 amino acid peptide, derived from a o larger precursor peptide, called pro-IAPP. 11 WO 03/103598 PCT/US03/17992 IAPP co-localizes and is co-secreted with insulin in response to p-cell secretagogues. This pathological feature is not associated with insulin-dependent (Type I) diabetes and is a unifying characteristic for the heterogeneous clinical phenotypes diagnosed as NIDDM (Type II diabetes). 5 Longitudinal studies in cats and immunocytochemical investigations in monkeys have shown that a progressive increase in islet amyloid is associated with a dramatic decrease in the population of insulin-secreting p-cells and increased severity of the disease. More recently, transgenic studies have strengthened the relationship between IAPP plaque formation and P-cell dysfunction, indicating that amyloid deposition is a principal factor in 10 Type-II. diabetes. IPP has also been shown to induce p-islet cell toxicity in vitro, indicating that appearance of IAPP fibrils in the pancreas of Type II or Type I diabetic patients (post transplantation) could contribute to the loss of the P islet cells (Langerhans) and organ dysfunction. In patients with Type-II diabetes, the accumulation of pancreatic IAPP leads t5 to a buildup of IAPP-amyloid as insoluble fibrous deposits which eventually replace the insulin-producing P cells of the islet resulting in p cell depletion and failure (Westermark, P., Grimelius, L., Acta Path. Microbiol. Scand., sect. A. 81: 291-300, 1973; de Koning, EJP., et al., Diabetologia 36: 378-384, 1993; and Lorenzo, A., et al., Nature 368: 756-760, 1994). 12 WO 03/103598 PCT/US03/17992 Diseases caused by the death or malfunctioning of a particular type or types of cells can be treated by transplanting into the patient healthy cells of the relevant type of cell. This approach has been used for Type I diabetes patients. Often pancreatic islet cells are cultured in vitro prior to transplantation to increase their numbers, to allow them to recover 5 after the isolation procedure or to reduce their immunogenicity. However, in many instances islet cell transplantation is unsuccessful, due to death of the transplanted cells. One reason for this poor success rate is IAPP, which can fonn fibrils and become toxic to the cells in vitro. In addition, IAPP fibrils are likely to continue to grow after the cells are transplanted and cause death or dysfunction of the cells. This may occur even when the 10 cells are from ahealthy donor and the patient receiving the transplant does not have a disease that is characterized by the presence of fibrils. For example, compounds of the present invention may also be used in preparing tissues or cells for transplantation according to the methods described in International Patent Application (PCT) number WO 01/03,680. Hormone-derived Amyloidoses t5 Endocrine organs may harbor amyloid deposits, particularly in aged individuals. Hormone-secreting tumors may also contain hormone-derived amyloid plaques, the fibrils of which are made up of polypeptide hormones such as calcitonin (medullary carcinoma of the thyroid), islet amyloid polypeptide (amylin; occurring in most patients with Type II diabetes), and atrial natriuretic peptide (isolated atrial amyloidosis). Sequences and !0 structures of these proteins are well known in the art. Miscellaneous Anyloidoses There are a variety of other forms of amyloid disease that are normally manifest as localized deposits of amyloid. In general, these diseases are probably the result of the localized production or lack of catabolism of specific fibril precursors or a predisposition of .5 a particular tissue (such as the joint) for fibril deposition. Examples of such idiopathic deposition include nodular AL amyloid, cutaneous amyloid, endocrine amyloid, and tumor related amyloid. 13 WO 03/103598 PCT/US03/17992 The compounds of the invention may be administered therapeutically or prophylactically to treat diseases associated with amyloid-p fibril formation, aggregation or deposition. The compounds of the invention may act to ameliorate the course of an amyloid p related disease using any of the following mechanisms (this list is meant to be illustrative 5 and not limiting): slowing the rate of amyloid-P fibril formation or deposition; lessening the degree of amyloid-P deposition; inhibiting, reducing, or preventing amyloid-p fibril formation; inhibiting neurodegeneration or cellular toxicity induced by amyloid-p; inhibiting amyloid-p induced inflammation; or enhancing the clearance of amyloid-p from the brain. 10 Compounds of the invention may be effective in controlling amyloid-p deposition either following their entry into the brain (following penetration of the blood brain barrier) or from the periphery. When acting from the periphery, a compound may alter the equilibrium of AP between the brain and the plasma so as to favor the exit of As from the brain. An increase in the exit of Ap from the brain would result in a decrease in AP brain 15 concentration and therefore favor a decrease in As deposition. Alternatively, compounds that penetrate the brain could control deposition by acting directly on brain AP, e.g., by maintaining it in.a non-fibrillar form or favoring its clearance from the brain. In a preferred embodiment, the method is used to treat Alzheimer's disease (e.g., sporadic or familial AD). The method can also be used prophylactically or therapeutically ,0 to treat other clinical occurrences of amyloid-p deposition, such as in Down's syndrome individuals and in patients with cerebral amyloid angiopathy ("CAA") or hereditary cerebral hemorrhage. Additionally, abnormal accumulation of APP and of amyloid-P protein in muscle fibers has been implicated in the pathology of sporadic inclusion body myositis (IBM) 5 (Askanas, V., et aL. (1996) Prod. Nat. Acad. Sci. USA 93: 1314-1319; Askanas, V. et al (1995) Current Opinion in Rheumatology 7: 486-496). Accordingly, the compounds of the invention can be used prophylactically or therapeutically in the treatment of disorders in which amyloid-beta protein is abnormally deposited at non-neurological locations, such as treatment of ]M by delivery of the compounds to muscle fibers. 14 WO 03/103598 PCT/US03/17992 The present invention therefore relates to the use of amidine compounds in the prevention or treatment 'of amyloid-related diseases, including, inter alia, Alzheimer's disease, cerebral amyloid angiopathy, inclusion body myositis, Down's syndrome, and type II diabetes. 5 Preferred compounds of the invention have at least two amidine moieties (preferably arylaridines, more preferably benzamidines). In one particular embodiment, the present invention relates to the novel use of amidine compounds in the prevention or treatment of amyloid-related diseases, such as those disclosed in U.S. Patent Nos. 5,428,051, 4,963,589, 5,202,320, 5,935,982, 5,521,189, 10 5,686,456, 5,627,184, 5,622,955, 5,606,058, 5,668,167, 5,667,975, 6,025,398, 6,214,883, 5,817,687, 5,792,782, 5,939,440, 6,017,941, 5,972,969, 6,046,226, 6,294,565 (B1), 6,156,779, 6,326,395, 6,008,247, 6,127,554, 6,172,104, 4,940,723, 5,594,138, 5,602,172, 5,206,236, 5,843,980, 4,933,347, 5,668,166, 5,817,686, 5,723,495, 4,619,942, 5,792,782, 5,639,755, 5,643,935, and 5,578,631, each of which are hereby incorporated herein by [5 reference in their entirety. In another embodiment, the invention relates to a method of treating or preventing an amyloid-related disease in a subject (preferably a human) comprising administering to the subject a therapeutic amount of a compound according to the following Formula, such that amyloid fibril-formation or deposition, neurodegeneration, or cellular toxicity is O reduced or inhibited. In another embodiment, the invention relates to a method of treating or preventing an amyloid-related disease in a subject (preferably a human) comprising administering to the subject a therapeutic amount of a compound according to the following Formula, such that cognitive function is stabilized or further deterioration in cognitive function is prevented, slowed, or stopped in patients with brain amyloidosis, e.g., .5 Alzheimer's disease or cerebral amyloid angiopathy: Ra'-N N-Ra2 b
Y
1 A Y 2 / bb9 R -N N-R Im q (Formula X) 15' WO 03/103598 PCT/US03/17992 wherein each of Rai, RbI, R"1, Ra2, Rb 2 , and Ro 2 is independently a hydrogen, a Z group, or Rai and RbI or R and Rb 2 are both taken together along with the nitrogen atoms to which they are bound to form a ring structure; each of Y' and Y 2 is independently a direct bond or a linking moiety; 5 m and q are each independently an integer selected from zero to five inclusive, such that 15m+q 5, or in another embodiment, 2 5m+qs5, or in another embodiment 1nm+qs10, or in another embodiment, 2 im+qs10; and The A group is a carrier moiety selected from substituted or unsubstituted aliphatic and aromatic groups, and combinations thereof; preferably such that the Y' and Y 2 moieties 10 -are bonded to an aromatic group. The A group preferably is a divalent group (i.e., m+q=2) such as an alkylene group (L e., -(CH2)k-- and substituted analogs thereof (including groups in which a -CH 2 - moiety is substituted by an oxygen atom), where k is I to 12 (preferably 6 to 9, more preferably 7 to 9), an alkenylene group (preferably 2 to 12 carbon atoms, more preferably 6 to 9 carbon 15 atoms, including groups with more than one double bond), an alkynylene group (preferably 2 to 12 carbon atoms, more preferably 6 to 9 carbon atoms, including groups with more than one triple bond), an alkoxyalkylene group, an ailcylaminoalkylene group, a thioalkoxyalkylene group, an arylenedialkylene group, a heteroarylenedialkylene group, an arylene group, a heteroarylene group, an oligoethereal group such as an 0. oligo(alkyleneoxide) group, or an arylene-di(oligoalkyleneoxide) group, each of which may. be substituted (with a Z group as defined below, e.g., a hydroxyalkylene group) or unsubstituted. The A group also includes the corresponding moieties of the Formulae I- IV herein. 16 WO 03/103598 PCT/US03/17992 In preferred aspects of the invention, the invention relates to a method of treating or preventing an amyloid-related disease in a subject (preferably a human) comprising administering to the subject a therapeutic amount of a compound according to one of the following Formulae, such that amyloid fibril formation or deposition, neurodegeneration, or 5 cellular toxicity is reduced or inhibited. In another embodiment, the invention relates to a method of treating or preventing an amyloid-related disease in a subject (preferably a human) comprising administering to the subject a therapeutic amount of a compound according to one of the following Formulae, such that cognitive function is stabilized or further deterioration in cognitive function is prevented, slowed, or stopped in patients with 10 brain amyloidosis, e.g., Alzheimer's disease or cerebral amyloid angiopathy: (R 1 ) (R2 Ra1-N N-Ra2 Rb-N X 1 ' X 2 NR RC1M Ro 2 (Formula 1) Ra1-N R \ 1 R N1__N 1-R )n .5 (Formula II) Ra1 Ra 2 RbIK I (RI) (R2) P/ N N N In 0N)a RN ,R mM 2 22 2N 3 4 (Formula III) 17 WO 03/103598 PCT/US03/17992 NRaK NR RK m M ( N / Rq (Rl
(R
2 )p (Formula ) ba N 2 N r \RR21N In -~ N V q 5 (Formula Nb) R'* N-A-N R .0whereinW',Re',R W, R, , R , ,Y,and Y areas defined herein, and Ais as defined above; WO 03/103598 PCT/US03/17992 each of R1 and R 2 is independently a hydrogen or a Z group, or two adjacent or proximate R 1 and R 2 groups, along with the corresponding X 1 and X 2 groups, if present (e.g., in Formula 11), taken together with the ring (e.g., phenyl ring) to which they ate bound foma fused ring structure, e.g., an aromatic or heteroaromatic (e.g., benzofuran) structure, 5 or a cycloalkyl or heterocylic structure; each of R 3 and R 4 is independently selected from the group consisting of hydrogen, substituted or unsubstituted straight or branched alkyl (preferably C 1 -Cs), cycloalIkyl (preferably C 3 -Cs), carbocyclic, aryl (e.g., phenyl), heterocyclic, and heteroaryl; each of R1* and R 2 * is independently selected from the group consisting of 10 substituted or unsubstituted straight or branched alkyl, cycloalkyl, heterocyclic, aryl (including phenyl), and heteroaryl; each of X and X 2 is independently a direct bond, or an oxygen, a NR' group (where R' is hydrogen (i.e., NH), a Cr-C 5 alkyl, C 2 rC 5 alkenyl, C-Cs alkynyl, or aryl group), a sulfonamide group (i.e., NHSO 2 or SO 2 NH), a carbonyl, amide (i.e., NHCO or CONE), a 15 Cr-C 5 alkylene group (e.g., -CH 2 -), C 2 -Cs alkenylene group (e.g., E or Z-CH=CH-),
C
2 rCS alkyhylene group, or a sulfur atom, or combinations thereof (e.g., -OCH 2 -,
-CH
2 0-, E or Z -OCH=CH- or -CH=CHO-); 19 WO 03/103598 PCT/US03/17992 M is a divalent group such as an alkylene group, i.e., -(CH2)1-- and substituted analogs thereof (including groups in which a -CHr2- moiety is substituted by an oxygen atom), where k is 1 to 12 (preferably 5 to 10, more preferably 6 to 9, most preferably 7 to 8), an alkenylene group (preferably 2 to 12 carbon atoms, more preferably 6 to 9 carbon 5 atoms, including groups with more than one double bond), an alkynylene group (preferably 2 to 12 carbon atoms, more preferably 6 to 9 carbon atoms, including groups with more than one triple bond), an ailcoxyalkylene group, an alkylaminoalkylene group, a thicalkoxyalkylene group, an arylenedialkylene group, an alkylenediarylene group, a heteroarylenedialkylene group, an arylene group, a heteroarylene group, an oligoethereal 10 group such as an oligo(alkyleneoxide) group, or an arylene-di(oligoalkyleneoxide) group, each of which may be substituted (with, for example, a Z group as defined herein, e.g., a hydroxyalkylene group such as -(CH 2
)
0
-
6
(CHOH)(CH
2
)
0
-
6 -; or other such substituted moieties, e.g., -(CH2)0- 6
(CHZ)(CH
2 )0- 6 -, including -(CH 2 )0-6(CHCO2aIkyl)(CH 2 )0- 6 -) or 15 unsubstituted; Z is a substituted or uansubstituted moiety selected from straight or branched alkyl (preferably C-C 5 ), cycloalkyl (preferably C3-Cs), alkoxy (preferably CrC 6 ), thioalkyl (preferably C-C 6 ), alkenyl (preferably C-C 6 ), alkynyl (preferably C2-C6), heterocyclic, carbocyclic, aryl (e.g., phenyl), aryloxy (e.g., phenoxy), aralkyl (e.g., benzyl),'aryloxyalkyl !0 (e.g., phenyloxyalkyl), arylacetatnidoyl, alkylaryl, heteroaralky, alkylcarbonyl and arylcarbonyl or other such acyl group, heteroarylcarbonyl, or heteroaryl group, (CR'R")o..NR'R' (e.g., -NH2), (CR'R")o.3CN (e.g., -CN), NO2, halogen (e.g., F, Cl, Br, or 1), (CR'R")o- 3 C(halogen)3 (e.g., -CF 3 ), (CR'R")D_ 3 CH(halogen)2, (CR'R")- 3 CH2(halogen), (CR'R)o-3CONR'R",, (CR'R"')o_3(CNH-)NR'R"', (CR'R")o-3S(0)1-2NR'R", 5 (CR'R")o-3CHO, (CR'R")o_30(CR'R")o..3H, (CR'R")o-3S(O)o 3R' (e.g., -SO3H1), (CR'R")o-30(CR'R")o- 3 H (e.g., -CH20CH 3 and -OCH3), (CR'R")o-3S(CR'R")o_ 3 H (e.g., -SH and-SCH 3 ), (CR'R")-30H (e.g., -OH), (CR'R")o-3COR', (CR'R")o- 3 (substituted or unsubstituted phenyl), (CR'R")..3(C-C 8 cycloalkyl), (CR'R")o- 3 CO2R' (e.g., -CO2H), or (CR'R")o..30R' group, or the side chain of any naturally 0 occurring amino acid; 20 WO 03/103598 PCT/US03/17992 in another embodiment, Z is a substituted or unsubstituted moiety selected from straight or branched alkyl (preferably C-C 5 ), cycloalkyl (preferably C 3 -Cs), alkoxy (preferably C-C 6 ), thioalkyl (preferably CrC), alkenyl (preferably C 2
-C
6 ), alkynyl (preferably d 2
-C
6 ), heterocyclic, carbocyclic, aryl (e.g., phenyl), aryloxy (e.g., phenoxy), 5, 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")c..oNR'R" (e.g., -NH 2 ), (CR'R")o-ioCN (e.g., -CN), NO 2 , halogen (e.g., F, C1, Br, or I), (CR'R")oicC(halogen) 3 (e.g., -CF3), (CR'R")o... 10CH(halogen) 2 , (CR'R") 0
_
10
CH
2 (halogen), (CR'R")o-ioCONR'R", (CR'R")o. 10. lo(CNH)NR'R", (CR'R")og1oS(O)j.-2NR'R"', (CR'R")o-joCHO, (CR'R")o-joO(CR')o...1H, (CR'R")o 0 oS(O)o- 3 R' (e.g., -SO'3H), (CR'R")o...
1 oO(CR'R")o-oH (e.g., -CH 2 0CH 3 and -OCH 3 ), (CR'R")ogoS(CR'R")o.3H (e.g., -SH and -SCH 3 ), (CR'R")ooOH (e.g., OH), (CR'R")oioCOR', (CR'R")o..o(substituted or unsubstituted phenyl), 15 (CR'R")o-io(C 3
-C
8 cycloalkyl), (CR'R")o 1 oCO 2 R' (e.g., -C0211), or (CR'R")o.oOR' group, or the side chain of any naturally occurring amino acid; wherein R' and R" are each independently hydrogen, a Cr-C 5 alkyl,
C
2 -Cs alkenyl, C2-Cs alkynyl, or aryl group, or R' and R" taken together are a benzylidene group or a -(CH 2
)
2 0(CH 2
)
2 - group; 20 m and q are each independently an integer selected from zero to five inclusive; in Formula I, m and q are each independently an integer selected from zero to four inclusive, and n and p are each independently an integer selected from zero to four inclusive, such that m+ns5 and p+qs5, wherein either m or q is at least one; and preferably m and q are one; !5 in Formula II, m is an integer selected from one to six inclusive, and n is an integer selected from zero to five inclusive, such that m+ns6; in Formula III, m, n, p, and q are each independently an integer selected from zero to . three inclusive, m+ns4, p+q 4, and m+q>1 (preferably m=q=1); 21 WO 03/103598 PCT/US03/17992 in Formula IV and IVb, m and n are each independently an integer selected from zero to three inclusive, p and q are each independently an integer selected from zero to four inclusive, m+ns4, p+q 5, and m+q1 (preferably m=q=1); and pharmaceutically acceptable salts thereof. 5 The chemical structures herein are drawn according to the conventional standards known in the art. Thus, where an atom, such as a carbon atom, as drawn appears to have an unsatisfied valency, then that valency is assumed to be satisfied by a hydrogen atom even though that hydrogen atom is not necessarily explicitly drawn. In an alternate embodiment, the invention relates to novel compounds, and novel 10 methods of their use as described herein, which are within the scope of the Formulae disclosed herein, and which are not disclosed in the above-referenced U.S. Patents, The groups Ral, RbI, RI, R, R12, and R 2 in the above Formulae are preferably a hydrogen, or a substituted or unsubstituted Cr-Cs alkyl or Cr-Cs alkoxy group or a hydroxy group. Preferred RaI and R groups are hydrogen, hydroxyl, alkyloxy groups (especially [5 lower alkyloxy groups, e.g. methoxy), aryloxy, acyloxy, and aroyloxy (i.e., R-(C=O)-O-, wherein R is aliphatic or aromatic). The phrase Ra and Rb both taken together along with the nitrogen atoms to which they are bound to form a ring structure" means that the two Ra and Rb groups are a moiety which joins the two nitrogen atoms in a heterocycle, such as the following ring structures: Zr N R4,wherein ris an integer from zero to 4 inclusive, NZr N R, wherein r is an integer from zero to 6 inclusive, 22 WO 03/103598 PCT/US03/17992 Zr N or R ,wherein r is an integer from zero to 4 inclusive. In another embodiment of the invention, for example, in compounds of Formula II, R and Rbl or R2 and Rb 2 are both taken together along with the nitrogen atoms to which they are bound to form a ring structure which is a nonaromatic ring, or an alicyclic ring, or a 5 monocyclic ring, or anon-fused ring. In some embodiments of Formula I, e.g., Ra, R I, WR , R2, R 2, and R2 are preferably a hydrogen, or a substituted or unsubstituted Ci-Cg alkyl group, wherein the alkyl substituent is any member of the group Z defined above, but not an aryl (e.g., phenyl) or alkyl group. Likewise, in certain embodiments of Formula II, R 1 is a moiety selected 10 from the Z group defined above other than an substituted aryl (e.g., phenyl) or heteroaryl group. The groups R 1 and R 2 are preferably a hydrogen, a substituted or unsubstituted Cr
C
8 alkyl group, a substituted or unsubstituted C 2 -Cs alkenyl group, a halogen (particularly bromine), a substituted or unsubstituted aryl or heteroaryl group, a substituted or .5 unsubstituted amino group, a nitro group, or a substituted or unsubstituted Cr-C 8 alkoxy group (particularly methoxy). Each Y group may be a direct bond, or a "linking moiety" (or "linking group") which is a group that is covalently bound to at least two other moieties and may be, for example, a single divalent atom or an oligomethylene group. A linking moiety which is a :0 linear chain of carbon atoms maybe optionally substituted or unsaturated. 23 WO 03/103598 PCT/US03/17992 Preferably a linking moiety is relatively small compared to the rest of the molecule, and more preferably less than about 250 molecular weight, and even more preferably less than about 75 molecular weight. Especially preferred linking moieties are -(CH 2 )n (wherein n is 1, 2, or 3), -NR'- (where R' is hydrogen, a Cr-C 5 alkyl, C 2
-C
5 alkenyl, C 2
-C
5 5 alkynyl, or aryl group), -S-, -O-,-NH-CHr 2 -- , and -CH=CH- (both E and Z configurations), or combinations thereof. The linking moiety may also be (CRR"), CROR(CRR%, CRSH(CRRY)n, CRNRRX(CRRZ)., (CRR").O(CRRY)., wherein each n is independently either 0, 1, 2, or 3, and R!, R7, RX, RI, and Rz are each independently hydrogen, a substituted or unsubstituted C-C 5 branched or straight chain 10 alkyl or alkoxy, C2-C 5 branched or straight chain alkenyl, aryloxycarbonyl, arylaminocarbonyl, arylalkyl, acyl, aryl, or C 3
-C
8 ring group. "Inhibition" of amyloid deposition includes preventing or stopping of amyloid formation, e.g., fibrillogenesis, inhibiting or slowing down of further amyloid deposition in a subject with amyloidosis, e.g., already having amyloid deposits, and reducing or reversing 1S 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 in pancreatic function in a diabetic patient, or in the case of a patient with brain amyloidosis, e.g., an Alzheimer's or cerebral amyloid angiopathy patient, stabilization of cognitive 20 function or prevention of a further decrease in cognitive function (i.e., preventing, slowing, or stopping disease progression). The term "alkyl" includes saturated aliphatic groups, including straight-chain alkyl groups (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, etc.), branched-chain alkyl groups (isopropyl, tert-butyl, isobutyl, etc.), cycloalkyl (alicyclic) 25 groups (cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, etc.), alkyl, substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups. Unless otherwise specified, the term alkyl further includes alkyl groups, which can further include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone. 24 WO 03/103598 PCT/US03/17992 In certain embodiments, a straight chain or branched chain alkyl has 6 or fewer carbon atoms in its backbone (e.g., C-C 6 for straight chain, C 3
-C
6 for branched chain), and more preferably 4 or fewer. Likewise, preferred cycloalkyls have from 3-8 carbon atoms in their ring structure, and more preferably have 5 or 6 carbons in the ring structure. The term 5 C 1
-C
6 includes alkyl groups containing'1 to 6 carbon atoms. An "alkylene" group is a divalent moiety derived from the corresponding alkyl group. Moreover, unless otherwise specified the term alkyl includes both "unsubstituted alkyls" and "substituted alkyls," the latter of which refers to alkyl moieties having substituents replacing one or more hydrogens on one or more carbons of the hydrocarbon 10 backbone. Such substituents can include, for.example, alkenyl, alkynyl, halogen, 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 5 alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. Cycloalkyls-may be further substituted, e.g., with the substituents described above. :0 An "arylalkyl" moiety is an alkyl group substituted with an aryl (e.g., phenylmethyl (i.e., benzyl)). An "alkylaryr' moiety is ati aryl group substituted with an alkyl group (e.g., p-methylphenyl (i.e., p-tolyl)). The term "n-alky1" means a straight chain (i.e., unbranched) unsubstituted alkyl group. The term "alkenyl"includes unsaturated aliphatic groups analogous in length and 5 possible substitution to the alkyls described above, but that contain at least one double bond. For example, the term "alkenyl" includes straight-chain alkenyl groups (e.g., ethylenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl, etc.), branched chain alkenyl groups, cycloalkenyl (alicyclic) groups (cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl, etc.), alkyl or alkenyl substituted cycloalkenyl D groups, and cycloalkyl or cycloalkenyl substituted alkenyl groups. The term alkenyl may further include alkenyl groups which include oxygen, nitrogen, sulftr or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone. 25 WO 03/103598 PCT/US03/17992 In certain embodiments, a straight chain or branched chain alkenyl group has 6 or fewer carbon atoms in its backbone (e.g., C 2
-C
6 for straight chain, C 3
-C
6 for branched chain). Likewise, cycloalkenyl groups may have from 3-8 carbon atoms in their ring structure, and more preferably have 5 or 6 carbons in the ring structure. The term C 2
-C
6 5 includes alkenyl groups containing 2 to 6 carbon atoms. An "alkenylene" group is a divalent moiety derived from the corresponding alkenyl group. Moreover, unless otherwise specified the term alkenyl includes both "unsubstituted alkenyls" and "substituted alkenyls," the latter of which refers to alkenyl moieties having substituents replacing one or more hydrogens on one or more carbons of the hydrocarbon 10 backbone. Such substituents can include, for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate (and lower alkyl esters thereof), alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including .5 alkyl amino, dialkylanino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. 0 The term "alkynyl" includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one triple bond. For example, the term "alkynyl" includes straight-chain alkynyl groups (e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl, etc.), branched-chain alkynyl groups, and cycloalkyl or cycloalkenyl substituted alkynyl groups. 5 Unless specified otherwise, the term alkynyl further includes alkynyl groups which include oxygen, nitrogen, sulfur or phosphorous atoms replacing one or more carbons of the hydrocarbon backbone. In certain embodiments, a straight chain or branched chain alkynyl group has 6 or fewer carbon atoms in its backbone (e.g., C-C 6 for straight chain, C-Ce for branched chain). The term C 2 rC 6 includes alkynyl groups containing 2 to 6 carbon atoms. ) An "alkynylene" group is a divalent moiety derived from the corresponding alkynyl group. 26 WO 03/103598 PCT/US03/17992 Moreover, unless otherwise specified the term alkynyl includes both "unsubstituted alkynyls" and "substituted alkynyls," the latter of which refers to alkynyl moieties having substituents replacing one or more hydrogens on one or more carbons of the hydrocarbon backbone. 5 Such substituents can include, for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbony, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylanino, 10 diarylamino, and alkylarylamino), acylarmino (including alkylcarbonylamino, arylcarbonylanino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, aryltbio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. .5 Unless the number of carbons is otherwise specified, "lower alkyl" as used herein means an alkyl group, as defined above, but having from one to five carbon atoms in its backbone structure. "Lower alkeny1" and "lower alkynyr'have chain lengths of, for example, 2-5 carbon atoms. The term "acy1" refers to a carbonyl group that is attached through its carbon atom ,0 to a hydrogen (i.e., a formyl), an aliphatic group (e.g., acetyl), an aromatic group (e.g., benzoyl), and the like. The term "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, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, 5 -akoxycarbonyl, 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), anidino, imino, sulfhydryl, alkylthio, aryltbio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, 0 sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. 27 WO 03/103598 PCT/US03/17992 Th6 term "acylamino" includes moieties wherein an amino moiety is bonded to an acyl group. For example, the acylamino group includes alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido groups. The terms "alkoxyalkyl", "alkylaminoalkyl" and "thioalkoxyalkyV include alkyl 5 groups, as described above, which further include oxygen, nitrogen or sulfur atoms replacing one or more carbons of the hydrocarbon backbone. The terms "alkoxy" or "alkyloxy" include substituted and unsubstituted alkyl, alkenyl, and alkynyl groups covalently linked to an oxygen atom. Examples of alkoxy groups include methoxy, ethoxy, isopropyloxy, propoxy, butoxy, and pentoxy groups. 10 Examples of substituted ailcoxy groups include halogenated alkoxy groups. The alkoxy groups can be substituted with groups such as alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylatee, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbony, alkylthiocarbonyl, alkoxyl, phosphate, 5 phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diar-ylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), anidino, imino, sulfhydryl, alkyltbio, arylthio, thiocarboxylate, sulfates, alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic 0 moieties. Examples ofhalogen substituted alkoxy groups include, but are not limited to, fluoromethoxy, difluoromethoxy, trifluoromethoxy, chloromethoxy, dichloromethoxy, trichloromethoxy, etc., as well as perhal6genated alkyloxy groups. The term "amine" or "amino" includes compounds or moieties in which a nitrogen atom is covalently bonded to at least one carbon or heteroatom; 5 The term "alkylamino" includes groups wherein the nitrogen is bound to at least one alkyl group. The term "dialkylamino" includes groups. wherein the nitrogen atom is bound to at least two alkyl groups. The term "arylamino" and "diarylamino" include groups wherein the nitrogen is bound to at least one or two aryl groups, respectively. 28 WO 03/103598 PCT/US03/17992 The term "alkylarylanino" refers to an amino group which is bound to at least one alkyl group and at least one aryl group. The term "alkaminoalkyl" refers to an alkyl, aikenyl, or alkynyl group substituted with an alkylamino group. 5 The term aided" or "aminocarbonyl" includes compounds or moieties which contain a nitrogen atom which is bound to the carbon of a carbonyl or a thiocarbonyl group. The term "carbonyl" or "carboxy" includes compounds and moieties which contain a carbon connected with a double bond to an oxygen atom. Examples of moieties which contain a carbonyl include aldehydes, ketones, carboxylic acids, aides, esters, anhydrides, 10 etc. The term "ether" or "ethereal" includes compounds or moieties which contain an oxygen bonded to two carbon atoms. For example, an ether or ethereal group includes "alkoxyalkyl" which refers to an alkyl, alkenyl, or alkynyl group substituted with an alkoxy group. 15 The term "hydroxy" or "hydroxyl" includes the groups -OH or -O- (with an appropriate counter ion). The term "halogen" includes fluorine, bromirie, chlorine, iodine, etc. The term "perhalogenated" generally refers to a moiety wherein all hydrogens are replaced by halogen atoms. t0 Arylenedialkylene or arylenedialkyl groups include those groups which have an arylene group to which are bound two other alkylene groups, which may be the same or different, and which two alkylene groups are in trn bound to other moieties. Examples of arylenedialkylene or arylenedialkyl groups include the following: Rh h (CR2)f(
(CR
2 )f (CR2)g
(CR
2 )g or ,and 29 WO 03/103598 PCT/US03/17992
(CR
2 )f (CR(CR2) (CR)9 ((CR2)f / R2)g
(CR
2 ) (C 2f -,or wherein each R group is independently a hydrogen (preferred) or is selected from the group Z defined above, and 1:f58, 1Sgs8, Osb 4. 5 Alkylenediarylene groups include groups which have an alkylene (or cycloalkylene) group to which are bound two other arylene groups, which may be the same or different, and which two alkylene groups are in turn bound to other moieties. Examples of alkylenediarylene groups include the following: Rh R
(CR
2 )f
(CR
2 )
(CR
2 )g Y-C and Rh R
(CR
2 )f \ (C 3
-C
6 CYCO (C alkyl) \ / wherein each R group is independently a hydrogen (preferred) or is selected from the. group Z defined above, 15y510 (preferably ly 4 ), lf-58, 1:g58, 0hs4, and 0:is4. Heteroarylenedialkylene or heteroarylenedialkyl groups include those groups which have a heteroarylene group to which are bound two other alkylene groups, which may be 5 the same or different, and which two alkylene groups are in turn bound to other moieties. Examples of heteroarylenedialkylene or heteroarylenedialkyl groups include the following: 30 WO 03/103598 PCT/US03/17992 Rh R
(CR
2 )f
(CR
2 )g X , wherein Oh53, and 0si:3, and X =NR' (wherein R' is hydrogen, a C 1 -Cs alkyl, C 2
-C
5 alkenyl, C 2 Cs alkynyl, or aryl group), 0, or S, 1sf!s8, 15gs8,
CR
2 )f X ICR2)g C2f(CR2)g or X ,)wherein hOb2, and X NI 5 (wherein R' is hydrogen, a Cr-C 5 alkyl, C 2 rC 5 alkenyl, C 2 rC 5 alkynyl, or aryl group), 0, or S, 1sf<8, 1sgs8, Rh \( ARh R C 2 )g (CR 2 ) N ( 2)O or , wherein Osbs3, 1f 8, 1sgs8,or Rh N R N (CR 2 )g CR 2 )f N or , wherein Ob<2, wherein each R group is independently a hydrogen (preferred) or is selected from the group tO Z defined above, 1:5f8, 1:gs8, and h and i are as indicated. An arylene group is an aromatic group which is capable of being connected covalently to other substituents through at least two positions, including the following examples: Rh Rh Rh
I'~
31 WO 03/103598 PCT/US03/17992 ) or wherein each R group is independently a hydrogen (preferred) or is selected from the group Z defined above, and Osi4; for example: OH 5 A heteroarylene group is a heteroaromatic group which is capable of being connected covalently to other substituents through at least two positions, including the following examples: Rh R X wherein 0h-3, and Osis3, and X NR' (wherein R' is hydrogen, a Cr-CS alkyl, G 2
-C
5 alkenyl, C 2
-C
5 alkynyl, or aryl group), 0, or S, h Rh 10 X or X ,wherein 0<h-2, and X=NR' (wherein R' is hydrogen, a CI-Cs alkyl, C 2
-C
5 alkenyl, C 2 -CS alIkynyl, or aryl group), 0, or S, Rh Rh N or N wherein Ohs3, or Rh N R N or N , wherein 0:h:2, 32 WO 03/103598 PCT/US03/17992 wherein each R group is independently a hydrogen (preferred) or is selected from the group Z defined above, and hand i are as indicated; for example, the following groups; N ,,or 5 Likewise, the invention relates to the following heteroarylene groups h - (C 2 ) (CR2) ,wherein X=NR' (whereinR' is hydrogen, a C-Cs alkyl, C-CS alkenyl, C-C 5 alkynyl, or aryl group), 0, or S; 0 fS8, 0:5g8; and each R group is independently a hydrogen (preferred) or is selected from the group Z defamed above. .0 In general, the term "aryl" includes groups, including 5- and 6-membered single ring aromatic groups that may include from zero to four heteroatoms, for example, groups derived from benzene, pyrrole, furan, thiophene, thiazole, isothiaozole, imidazole, triazole, tetrazole, pyrazole, oxazole, isooxazole, pyridine, pyrazine, pyridazine, and pyrimidine, and the like. 5 Furthermore, the term "aryl" includes multicyclic aryl groups, e.g., groups derived from tricyclic, bicyclic, e.g., naphthalene, benzoxazole, benzodioxazole, benzothiazole, benzoimidazole, benzothiophene, methylenedioxyphenyl, quinoline, isoquinoline, napthyridine, indole, benzofuran, purine, benzofuran, deazapurine, or indolizine. Those aryl groups having heteroatoms in the ring structure may also be referred to as 0 "aryl heterocycles," "heterocycles," "heteroaryls" or "heteroaromatics". 33 WO 03/103598 PCT/US03/17992 An aromatic ring can be substituted at one or more ring positions with such. substituents as described above, as for example, halogen, hydroxyl, alkyl (e.g. tolyl), alkoxy, alkylcarbonyloxy, arylcarbonyloxy, ailcoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, alkylaminoacarbonyl, arylalkyl aminocarbonyl, 5 alkenylaminocarbonyl, alkylcarbonyl, arylcarbonyl, arylalkylcarbonyl, alkenylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylthiocarbonyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including allkylcarbonylamino, aryloarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sulfates, 10 alkylsulfinyl, sulfonato, sulfamoyl, sulfonamido, nitro, trifluoromethyl, cyano, azido, heterocyclyl, alkylaryl, or an aromatic or heteroaromatic moiety. 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). The term "heterocyclic" or "heterocycle" includes heteroaryls as well as any ring L5 formed which incorporate a heteroatom or an atom which is not carbon. The ring may be saturated or unsaturated and may contain one or more double bonds. Examples of preferred heterocyclic groups include pyridyl, furanyl, thiophenyl, morpholinyl, and indolyl groups. The term "heteroatom" includes atoms of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, sulfur and phosphorus. 10 -An "arylene" group is a divalent moiety derived from an aryl group. An oligoethereal group, such as an oligo(alkyleneoxide) group, includes polyethyleneglycol (PEG) and short chain analogs thereof including
-[(CR
2 )O]t(CR2)r--, wherein 1:t56 and 15s56, and each R group is independently a hydrogen (preferred) or is selected from the group Z defined above. :5 An arylene-di(oligoalkyleneoxide) group is an aryl group which has two oligoalkyleneoxide groups bound to it which in turn are bound to other moieties, and include the following examples: [(CR2)sO]t(CR 2 )s-Aryl--[(CR 2 )sO]t(CR 2 ), 34 WO 03/103598 PCT/US03/17992 wherein "Aryl" is an arylene moiety, 1t56, 05s56, and each R group is independently a hydrogen (preferred) or is selected from the group Z defined above. Preferred arylene di(oligoalkylensoxide) groups include: Rh
[(CR
2 ),O]t(CR 2 )s
[(CR
2 )sO]t(CR 2 )s Rh
['R
2 )sO]t(CR 2 )S
[(CR
2 )sO~t(CR 2 )s 5 ,or Rh
[(CR
2 )sO]t(CR 2 )s [(CR2)sO]t(CR2)s wherein 15t56, Os56, 0!i 4, and each R group is independently a hydrogen (preferred) or is selected from the group Z defined above. 35 WO 03/103598 PCT/US03/17992 The term "substituted" means that the moiety has substituents placed on the moiety other than hydrogen which allow the molecule to perform its intended function. Examples of substituents include moieties selected from straight or branched alkyl (preferably C1-Cs), cycloalkyl (preferably C3-Cs), alkoxy (preferably C 1 -C), thioalkyl (preferably C 1
-C
6 ), 5 alkenyl (preferably C 2
-C
6 ), ailcynyl (preferably C 2
-C
6 ), heterocyclic, carbocyclio, 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.
3 NR'R" (e.g., -NH 2 ), (CR'R")o- 3 CN (e.g., -- CN), NO 2 , halogen (e.g., F, Cl, Br, or 10 1), (CR'R")o- 3 C(halogen) 3 (e.g., -CF 3 ), (CR'R")o...
3 CH(halogen) 2 , (CR'R")o- 3
CH
2 (halogen), (CR'R")o-3CONR'R", (CR'R")o- 3 (CNF)NR'R", (CR'R")o.3S(O)1.-2NR'R"', (CR'R"')o-3CHO, (CR'R")o..30(CR'R")o-3HL (CR'R")o- 3 S(O)o...
3 R' (e.g., -S0 3 H), (CR'R")o.
3 0(CR'R")o- 3 H (e.g., -CH 2 0CH 3 and OCH 3 ), (CR'R")o- 3 S(CR'R")o- 3 H (e.g., -SH and -SCH 3 ), (CR'R")o-30H (e.g., -OH), 15 (CR'R")o-3COR', (CR'R")o- 3 (substituted or unsubstituted phenyl), (CR'R")o- 3
(C
3
-C
8 cycloalkyl), (CR'R") 0 -3CO 2 R' (e.g., -CO 2 H), or (CR'R")o- 3 0R' group, or the side chain of any naturally occurring amino acid; wherein R' and R" are each independently hydrogen, a C 1
-C
5 alkyl, 02-CS alkenyl, C2C5 alkynyl, or aryl group, or R' and R"taken together are a benzylidene group or a -(CH2)2O(CH2)r group. Preferably, 20 substitutions enhance the ability of tlie compounds of the invention to perform its intended function, e.g., inhibit formation of amyloid deposits. In compounds of the invention, it is preferred that m=1 and that n=-0, 1, or 2. In compounds of Formula I, preferably p=O, 1, or 2, and q=1. It is especially preferred that molecules according to Formula I are symmetric, thus R=R2, Rel=R 2 , R1=R 2 , m=q, 25 n=p, and Y'=Y 2 . Likewise, it is preferred that R 1
=R
2 , and X 1
=X
2 in molecules of Formula I. 36 WO 03/103598 PCT/US03/17992 One group of preferred compounds of the invention are those of Formula Ia: Ra1 Ra N 2 N Rbi Rb RN N" 1. 2 (FormulaIa) wherein M is R R :h1 Rh2 x 5 wherein, in a preferred aspect, R and Rbl together, or R2 and Rb 2 together, represent a C2 to C 3 alkylene; Rdl and Ro2 are H; Rhl is H; and Rhl is OCH 3 or O(C 6
H
4 )R, wherein R is H or lower-alkyl, and X is 0, NR' (wherein R' is hydrogen, a C 1
-C
5 alkyl, C 2
-C
5 alkenyl, C2 C5 alkynyl, or aryl group), or S. In another group of preferred compounds of Formula Ia, Ra1 and Rl together, or Ra 10 and R2 together, represent a C2 linear, saturated alkylene; R** and R2 are -(lower alkyl) OH; and Rhl and Rl 2 are each H. The "lower alkyl" group of Rel and R are preferably ethylene. In yet another group of preferred compounds of Formula Ia, Ra and Rl together, or R and Rb 2 together, represent a C 4 alkylene; Rdl and Rd2 are H (preferred), lower alkyl, 15 cycloalkyl, aryl, hydroxyalkyl, aminoalkyl or alkylaminoalkyl; Rbl and R 2 are independently selected from the group consisting of H (preferred), lower alkyl, halogen, alkoxy, aryloxy, or arylalkoxy. In still yet another group of preferred compounds of Fdrmula Ia, Ra, R 2 , Re and Rbl are H; Rdl and Rz are isopropyl or -(CH 2 )3N(CH3)2; and RhI and R are H. 20 In a further group of preferred compounds of Formula Ia, Ra and Rbl together, or R and Rb2 together, represent a phenylene group which is optionally substituted with up to three -CONHRMNRR! groups where Rd is lower alkyl and R* and R! are each independently selected from the group consisting of H or lower alkyl; and R"d, R , Rht, and Rd are H. An especially preferred compound of Formula Ia has RbI, R', RW, R*, Rb 2 , and Re 2 25 being H, and RaI and R! groups being hydroxy or methoxy. 37 WO 03/103598 PCT/US03/17992 Another group of preferred compounds are those of Fonnula Ib: alN a 2 N N-R RbI-N M N-Rb2 R R1R 2 R0 2 (Formula Ib) wherein M is Rhl Rh 2 5 wherein X is 0, NR' (wherein R' is hydrogen, a C 1
-C
5 alkyl, C 2
-C
5 alkenyl, C 2 -Cs alkynyl, or aryl group), or S; Rhl and R"2 are each independently selected from the group consisting of H, loweralkyl, aryl, alkylaryl,- aminoalkyl, aminoaryl, halogen, alkoxy, aryloxy, or oxyarylalkyl; R 1 and R 2 are each independently selected from the group consisting of H, loweralkyl, alkoxy, alkylaryl, aryl, aryloxy, aminoalkyl, aninoaryl, or halogen; and each 10 Ra, R2, R 1 , and R" 2 group is independently selected from the group consisting of H, loweralkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, alkylamiinoalkyl, cycloalkyl, aryl, hydroxy, or alkylaryl; or Rai and Rbl together, or R2 and R2 together, represent C 2
-C
10 alkyl, hydroxyalkyl, or alkylene; and each R and Re 2 group is independently H, hydroxy, loweralkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, alkylamino, alkylaminoalkyl, 15 cycloalkyl, hydroxycycloalkyl, alkoxycycloalkyl, aryl, or alkylaryl. 38 WO 03/103598 PCT/US03/17992 Another group of preferred compounds are those of Formula Ic: R R2 Ral-N N-Ra 2 11 R 1-N N-R \R0 Ro2 (Formula Ic) wherein M is hl h 2 X 5 wherein X is S, 0, or NR' (wherein R' is hydrogen, a C 1 -Cs alkyl, C 2
-C
5 ailcenyl, C 2
-C
5 alkynyl, or aryl group); Rbl, Rb 2 , R", and R 2 are each independently selected from the group consisting of H, loweralkyl, alkoxy, alkoxyalkyl, cycloalkyl, aryl, hydroxyalkyl, aminoalkyl or alkylaminoalkyl; R' and R2 are H, lower alkyl, alkoxy, alkoxyalkyl, hydroxyalkyl, cycloalkyl, aryl, aminoalkyl, alkylaminoalkyl or halogen; R' and R'V are 10 OY, or R' and R ' together, or R and R2 together represent
R
5 + wherein R 5 is YO, N Y is H or lower alkyl; each of X1 and X 2 are -(CH 2 )-, where n is an integer from 0 to 2; and 15 Rh' and Rh2 are each independently selected from the group consisting of H, lower aIkyl, halogen, alkoxy, aryloxy, or oxyarylalkyl. Yet another group of preferred compounds are those ofFormula Ic, wherein M is
-(CH
2 )r where n is an integer from 2 to 16 (or 2 to 12, or 2 to 10); each of X and X 2 is 0, NH, or S; Ra, R!2, R', and Rb 2 are H; or R and RbI together, or R2 and Rb 2 together 20 represent -(CH 2 )m-, wherein m is 2, 3, or 4; each of R' and R 2 are H, OCH 3 , NO 2 or NH 2 ; RdI and R 2 are-H, CH 3 or CH 2
CH
3 .In another embodiment, when X is 0 or S, both R 1 and R"' cannot be H; and when X 2 is 0 or S, both R 2 and Rd 2 cannot be H. 39 WO 03/103598 PCT/US03/17992 Another group of preferred compounds are those of Formula Id: al Ra2 NN R1 RR 2 IN R' (Formula Id) wherein each Rai, V 2 ' Rb, and Rb 2 are independently selected from the group consisting of H, loweralkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, cycloalkyl, aryl, or 5 alkylaryl; or two R 1 and RL together, or R2 and R" 2 together represent C 2 -Cio alkylene; R* and Re 2 are independently H, hydroxy, loweralkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, cycloalkyl, aryl, or alkylaryl; and R' is H, loweralkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, cycloalkyl, aryl, or alkylaryl. Another group of preferred compounds are those of Formula le: Ra-N N-Ra2 Rbi-N X1'M'X2 N fN-Rb 2 10 \RI Re 2 (Formula Ie) wherein M is an alkylene group (e.g., C 2 to C 16 ), and X' and X 2 are oxygen. In another group of preferred compounds of Formula le, Ra1 and Rbl together, or R2 and Rb 2 together, represent a C 2 linear, saturated alkylene; R"' and Re2 are H. 40 WO 03/103598 PCT/US03/17992 Another group of preferred compounds of the invention are those of Formula Ha: E-Y' (R )n Y9 (Formula Ila) wherein E is Ra1-N N N RWi--N N -N bi DI \R or 'Rc or R" 5 wherein Y', Y 2 , Z, and R' are as defined above; n is 0 -4; Y 2 is preferably 0, NH, S, a substituted or unsubstituted methylene group, or a direct bond; Z may be a hydrogen atom, or Z is preferably alkyl, aryl, alkoxy, aryloxy, hydroxy, a substituted or unsubstituted amino, nitro, sulfo, or halogen group; Ra', RbI, and RV 1 are independently hydrogen, lower alkyl, aromatic, hydroxyl, or alkoxy; and B is a direct bond or a substituted or unsubstituted 10 alkylene group containing from I to 16 carbon atoms, or a biphenylene group, or a combination biphenylene-alkylene group, the group -[(CH 2 ).O]m(CH 2 )a-where mis I to 6 and n is 2 to 6, or a heterocyclic group. Compounds of Formula lIb are also within the invention: NH R
H
2 N I (CH2) O (Formula llb) 15 wherein n= 2, 3, 4, 5, 6, 7, 8, 9, or 10; and R =hydrogen, hydroxy, halogen, phenyl, biphenyl, naphthyl, alkoxy, carboxy, alkoxycarbonyl, aryloxycarbonyl, or aryloxy. 41 WO 03/103598 PCT/US03/17992 Another group of preferred compounds are of Formula I]Ia: Ra-N 2NRa 2 RG- N M R b a H H (Formula Ma) wherein M is R hl Rh x 5 wherein X is S, 0, or NR' (wherein R' is hydrogen, a C 1
-C
5 alkyl, C 2
-C
5 alkenyl, C 2
-C
5 alkynyl, or aryl group); Ra, Ra2, Rbl, and Rb 2 are each independently selected from the group consisting of H, lower alkyl, alkoxyalkyl, cycloalkyl, aryl, alkylaryl, hydroxyalkyl, aminoalkyl, or alkylaminoalkyl; or Ra' and Rbl together, or R! 2 and R 2 together represent a
C
2 to Cio alkyl, hydroxyalkyl, or alkylene; or Ra' and RbI together, or Ra2 and Rb 2 together 10 are: (R )n -. wherein n is a number from I to 3, and R ' is H or -CONBIR"NR 5 Ri 6 , wherein R" is lower 'alkyl and R 5 and R ' are each independently selected from the group consisting of H and lower alkyl; and R and Re are H, hydroxy, lower alkyl, cycloalkyl, aryl, alkylaryl, 15 alkoxyalkyl, hydroxycycloalkyl, alkoxycycloalkoxy, hydroxyalkyl, aminoalkyl or alkylaminoalkyl; and RhS and R" are.each independently selected from the group consisting of H, lower alkyl, halogen, aryl, arylalkyl, aminoalkyl, aminoaryl, alkoxy, aryloxy, or oxyarylalkyl. 42 WO 03/103598 PCT/US03/17992 Yet another group of preferred compounds are of Formula IIlb: Ra1N N R Rb M b2 N M N H H (Formula IIb) wherein each pair of R with RbI and R 2 with Rb 2 together represent -(CH 2 )m- wherein m is from two to four; R* and Re 2 are independently H or loweralky; and M, which may be 5 substituted with a lower alkyl group, is selected from the group consisting of -CH=CH-CH 2 CH 2 -, -CH 2
-CH=CH-CH
2 -, and -CH=CH-CH=CH-. Another group of preferred compounds are those of Formula IIlc: 1Ra Rb2 I 1 C N' N C3 ~ c4 N (Formula I) wherein R' and R 2 are independently H or -CONHIR 5
NR
6
R
7 , wherein R is lower alkyl, R 6 10 and R 7 are each independently selected from the group consisting of H and lower alkyl; R!', Ra 2 , RbI, and Rb 2 are independently selected from the group consisting of H, lower alkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, cycloalkyl, aryl, or alkylaryl, or R and RbI together, or R!2 and R 2 together represent C 2
-C
10 alkylene; R"d and R 2 are independently H, hydroxy, lower alkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, 15 alkylaminoalkyl, cycloalkyl, aryl, or alkylaryl; R' 3 and Re are independently H, hydroxy, loweralkyl, alkoxyalkyl, hydroxyalkyl, aminoalkyl, alkylaminoalkyl, cycloalkyl, aryl, or alkylaryl; and R' is H, loweralkyl, alkoxyalkyl, hydroxyallcyl, aminoalkyl, alkylaninoalkyl, cycloalkyl, aryl, alkylaryl, or halogen. In another embodiment, the present invention relates to pharmaceutical 20 compositions comprising compounds according to any of the Formulae herein for the treatment of an amyloid-related disease, as well as methods of manufacturing such pharmaceutical compositions. 43 WO 03/103598 PCT/US03/17992 The compounds of the invention can be formulated to ensure proper distribution in vivo. For example, the blood-brain barrier (BBB) excludes many highly hydrophilic compounds. To ensure that the more hydrophilic therapeutic compounds of the invention cross the BBB, they can be formulated, for example, in liposomes. For methods of 5 manufacturing liposomes, see, e.g., U.S. Patent 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"), thus providing targeted drug delivery (see, e.g., V. V. Ranade (1989) J. Clin. Phaimacol. 29:685). Exemplary targeting moieties include folate or biotin (see, e.g., U.S. Patent No. 10 5,416,016 to Low et al.); mannosides (Umezawa et al. (1988) Biochem. Biophys. Res. Common. 153:1038); antibodies (P. G. Bloeman et al. (1995) FEBS Lett. 357:140; M. Owais et al. (1995) Antimicrob. Agents Chemother. 39:180); surfactant protein A receptor (Briscoe et al. (1995) Am. J. Physiol. 1233:134); gpl20 (Schreier et al. (1994) J. Biol. Chem. 269:9090); see also K. Keinanen; M. L. Laukkanen (1994) FEBS Lett. 346:123; J. . [5 Killion; L J. Fidler (1994) Inmunomethods 4:273. In a preferred embodiment, the therapeutic compounds of the invention are formulated in liposomes; in a more preferred embodiment, the liposomes include a targeting moiety. To ensure that compounds of the invention cross the BBB, they may be coupled to a BBB transport vector (for review of BBB transport vectors and mechanisms, see Bickel, et ;0 al., Adv. Drug Delivery Reviews, vol. 46, pp. 247-279, 2001). Exemplary transport vectors include cationized albumin or the OX26 monoclonal antibody to the transferrin receptor; these proteins undergo absorptive-mediated and receptor-mediated transcytosis through the BBB, respectively. 44 WO 03/103598 PCT/US03/17992 Examples of other BBB transport vectors that target receptor-mediated transport systems into the brain include factors such as insulin, insulin-like growth factors (IGF-I, IGF-I), angiotensin II, atrial and brain natriuretic peptide (ANP, BNP), interleukin I (IL-1) and transferrin. Monoclonal antibodies to the receptors which bind these factors may also 5 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 dynorphin analogue B-2078 and the ACTH analogue ebiratide can also cross the brain via absorptive-mediated transcytosis 10 and are potential transport vectors. Other BBB transport vectors target systems for transporting nutrients into the brain. Examples of such BBB transport vectors include hexose moieties, e.g. glucose, monocarboxylic acids, e.g. lactic acid, neutral amino acids, e.g. phenylalanine, amines, e.g. choline, basic amino acids, e.g. arginine, nucleosides, e.g. adenosine, purine bases, e.g. 15- adenine, and thyroid hormone, e.g. triiodothyridine. Antibodies to the extracellular domain of nutrient transporters can also be used as transport vectors. Other possible vectors include angiotensin II and ANP, which may be involved in regulating BBB permeability. In some cases, the bond linking the therapeutic compound to the transport vector may be cleaved following transport into the brain in order to liberate the biologically active 20 compound. Exemplary linkers include disulfide bonds, ester-based linkages, thioether linkages, aide bonds, acid-labile linkages, and Schiff base linkages. Avidin/biotin linkers, in which avidin is covalently coupled to the BBB drag transport vector, may also be used. Avidin itself may be a drug transport vector. To administer the therapeutic compound by other than parenteral administration, it 25 may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. For example, the therapeutic compound may be administered to a subject in an appropriate carrier, for example, liposomes, or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Liposomes include water-in-oil-in-water CGF emulsions as well as conventional 30 liposomes (Strejan et aL., (1984) J. Neuroimmunol. 7:27). 45 WO 03/103598 PCT/US03/17992 The therapeutic compound may also be administered parenterally, intraperitoneally, intraspinally, or intracerebrally. Dispersions can be prepared in glycerol, liquid 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 5 microorganisms. Pharmaceutical 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. In all cases, the composition must be sterile and must be fluid to the extent that easy syringability exists. It must be stable 10 under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. 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 .5 maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, tliimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, sodium 0 chloride, or polyalcohols such as mannitol and sorbitol, in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin. Sterile injectable solutions can be prepared by incorporating the therapeutic 5 compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the therapeutic compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient (i.e., the therapeutic compound) plus any additional desired ingredient from a previously sterile-filtered solution thereof. 46 WO 03/103598 PCT/US03/17992 The therapeutic compound- can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The therapeutic compound 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. For oral therapeutic administration, the 5 therapeutic compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. The percentage of the therapeutic compound in the compositions and preparations. may, of course, be varied. The amount of the therapeutic compound in such therapeutically useful compositions is such that a suitable dosage will be obtained. 0 It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and unifority of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical .5 vehicle. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such a therapeutic compound for the treatment of amyloid deposition in subjects. ?10 The present invention therefore includes pharmaceutical formulations comprising the compounds of the Formulae described herein, including pharmaceutically acceptable salts thereof, in pharmaceutically acceptable carriers for aerosol, oral and parenteral administration. Also, the present invention includes such compounds, or salts thereof, which have been lyophilized and which may be reconstituted to form pharmaceutically 25. acceptable formulations for administration, as by intravenous, intramuscular, or subcutaneous injection. Administration may also be intradermal or transdermal. In accordance with the present invention, a compound 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, 30 suspension or emulsion. Alternatively, the compounds or salts may also be administered by inhalation, intravenously or intramuscularly as a liposomal suspension. 47 WO 03/103598 PCT/US03/17992 Pharmaceutical formulations are also provided which are suitable for administration as an aerosol, by inhalation. These formulations comprise a solution or suspension of the desired compound of any Formula herein, or a salt thereof, or a plurality of solid particles of the compound or salt. The desired formulation may be placed in a small chamber and 5 nebulized. Nebulizationmaybe accomplished by compressed air or by ultrasonic energy to form a plurality of liquid droplets or solid particles comprising the compounds 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 compound of any Formula described herein, or a salt thereof, in any appropriate mamer known in the art, 10 such as by micronization. Most preferably, the size of the solid particles or droplets will be from about 1 to about 2 microns. In this respect, commercial nebulizers are available to achieve this purpose. Preferably, when the pharmaceutical formulation suitable for administration as an aerosol is in the form of a liquid, the formulation will comprise a water-soluble compound t5 of any Formula 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. Active compounds are administered at a therapeutically effective dosage sufficient ,0 to inhibit amyloid deposition in a subject. A "therapeutically effective" dosage preferably inhibits amyloid deposition by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still -more preferably by at least about 80% relative to untreated subjects. In the case of an Alzheimer's patient, a therapeuticallyly effective" dosage stabilizes cognitive function or prevents a further decrease in cognitive 5 function (i.e., preventing, slowing, or stopping disease progression). 48 WO 03/103598 PCT/US03/17992 The ability of a compound to inhibit amyloid deposition can be evaluated in an 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 As deposition is seen. Likewise, the ability of a compound to prevent or 5 reduce cognitive impairment in a model system may be indicative of efficacy in humans. Alternatively, the ability of a compound can be evaluated by examining the ability of the compound to inhibit amyloid fibril formation in vit-o, e.g., using a fibrillogenesis assay such as that described herein, including a ThT, CD, or EM assay. Also the binding of a compound to amyloid fibrils may be measured using a MS assay as described herein. .0 The present invention is also related to prodrugs of the compounds of the Fonnulae disclosed herein. Prodrugs are compounds which are converted in vivo to active forms (see, e.g., R.B. Silverman, 1992, "The Organic Chemistry of Drug Design and Drug Action," Academic Press, Chp. 8). Prodrugs can be used to alter the biodistribution (e.g., to allow compounds which would not typically enter the reactive site of the protease) or the 5 pharmacokinetics for a particular compound. For example, a carboxylic acid group, can be esterified, e.g., with a methyl group or an ethyl group to yield an ester. When 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 3 intermediate compound which subsequently decomposes to yield the active compound. The prodrug moieties may be metabolized in vivo by esterases or by other mechanisms to carboxylic acids. Examples of prodrugs and their uses are well known in the art (See, e.g., Berge et al. (1977) "Phannaceutical Salts", J. Pharm. Sci. 66:1-19). The prodrugs can be prepared in i situ during the final isolation and purification of the compounds, or by separately reacting the purified compound 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. 49 WO 03/103598 PCT/US03/17992 Examples of cleavable carboxylic acid prodrag moieties include substituted and unsubstituted, branched or unbranched lower alkyl ester moieties, (e.g., ethyl esters, 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), 5 acylamino lower alkyl esters, acyloxy lower alkyl esters (e.g., pivaloyloxymethyl ester), aryl esters (phenyl ester), aryl-loiver alkyl esters (e.g., benzyl ester), substituted (e.g., with methyl, halo, or methoxy substituents) aryl and aryl-lower alkyl esters, amides, lower-alkyl amides, dilower alkyl aides, and hydroxy aides. It will be noted that the structures of some of the compounds of this invention 10 include stereogenic carbon atoms. It is to be understood accordingly that the isomers arising from such asymmetry (e.g., all snantiomers and diastereomers) are included within the scope of this invention unless indicated otherwise. That is, unless otherwise stipulated, any chiral carbon center maybe of either (R)- or (S)-stereochemistry. Such isomers can be obtained in substantially pure form by classical separation techniques and by L5 stereochemically controlled synthesis. Furthermore, alkenes can include either the E- or Z geometry, where appropriate. Certain embodiments of the present compounds can contain a basic functional group, such as amino or alkylamino, and are, thus, capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable acids. The term "pharmaceutically ,0 acceptable salts" in this respect, refers to the relatively non-toxic, inorganic and organic acid addition salts of compounds of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds of the invention, or by separately reacting a purified compound of the invention in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. .5 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, funarate, succinate, tartrate, napthylate, mesylate, glucoheptonate, lactobionate, 2-hydroxyethylsulfonate, and laurylsulphonate salts and the like. (See, e.g., Berge et al. (1977) "Pharnaceutical Salts", J~ 0 Pharm. Sci. 66:1-19). 50 WO 03/103598 PCT/US03/17992 In other cases, the compounds of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases. The term "pharmaceutically acceptable salts" in these instances refers to the relatively non-toxic, inorganic and organic base addition salts 5 of compounds of the present invention. These salts can likewise be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound 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 acceptable organic 10 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 ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like. 15 Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments and methods described herein. Such equivalents are intended to be encompassed by the scope of the following claims. All patents, patent applications, and literature references cited herein are hereby expressly incorporated by reference in their entirety. This invention is further 20 -illustrated by the following examples which should not be construed as limiting. EXAMPLES 51 WO 03/103598 PCT/US03/17992 The synthesis of amidine compounds of the invention is described in U.S. Patent Nos. 5,428,051, 4,963,589, 5,202,320, 5,935,982, 5,521,189, 5,686,456, 5,627,184, 5,622,955, 5,606,058, 5,668,167, 5,667,975, 6,025,398, 6,214,883, 5,817,687, 5,792,782, 5,939,440, 6,017,941, 5,972,969, 6,046,226, 6,294,565 (B1), 6,156,779, 6,326,395, 5 6,008,247, 6,127,554, 6,172,104, 4,940,723, 5,206,236, 5,843,980, 4,933,347, 5,668,166, 5,817,686, 5,723,495,4,619,942, 5,792,782, 5,639,755, 5,643,935, 5,602,172, 5,594,138, and 5,578,631. Many of the compounds may also be purchased from Sigma-Aldrich Co. (Milwaukee, USA). The compounds may also be synthesized according to art-recognized techniques. 10 Test compounds were purchased from commercial sources or synthesized and screened by thioflavin T fluorescent assay ("ThT assay"). Alternatively, one could screen test compounds by circular -dichroism ("CD"), electron microscopy ("EM"), or mass spectroscopy ("MS") assays. The MS assay gives data on the ability of compounds to bind to an amyloid protein, whereas the ThT, EM, and CD assays give data on inhibition of 15 fibrillogenesis. The thioflavin T fluorescent assay for fibrillogenesis is based on the principle that the fluorescent dye, thioflavin T, binds specifically to fibrillar, but not to unaggregate As peptide (LeVine III, H., 1993, Protein Science 2:404-410). Upon binding, thioflavin T develops a characteristic fluorescence (Naiki, H., et al., 1996, Lab. Invest. 74: 374-383) 20 which can be easily detected. The dye is believed to interact with the stacked cross-p pleated sheets, the common structural motif of all arnyloid (LeVine III, H., 1995, Amyloid: Int. J. Exp. Clin Invest. 2:1.6). Thioflavin T is widely used to assay the effect of compounds on fibrillogenesis of As peptide and other amyloid proteins (Bronfnan, P.C., et al., 1995, Neuroscience Left. 218:201-203). In this assay, test compounds are incubated with a 25 solution of AP (1-40) (20 pffM) or IAPP (10 pM) containing 5 FM Thioflavin T, in 0.02M Tris/0.02M acetate/0.15M NaC1/0.005% azide/pH 7.40 at 37*C in sealed 384 well microplates. Readings (ex 430 nm/em'485nm) are taken at various time intervals with a microplate fluorescence reader. An increase in fluorescence signifies the appearance of amyloid or intermediates in the production of amyloid, as illustrated in the Figures (in 30 general, a compound which inhibits fibrillogenesis produces lower fluorescence in the assay because the fluorescence of ThT is greater when bound to fibrils). 52 WO 03/103598 PCT/US03/17992 Protocol: AP peptide: AP (1-40) 95% purity (American Peptide Company, Inc, Sunnyvale, Cal. USA) is disaggregated in trifluoroacetic acid and filtered through a 0.02 pLM filter, (Whatman Anotop 25 plus, 0.02 pm, Catalogue no. 6809 4102) in hexafluoroisopropanol (HFIP). Solutions of Ap (1-40) or IA.PP at 600 pm in HFIP are 5 stored at -80C. Assay mixture: The mixture is prepared as two solutions which are combined upon addition to the 384 well microplate (Corning Costar cat. 3705). i) Solution A consists of test compounds in 0.02M Tris/0.02M acetate/0.15M NaCI/0.01 % azide at pH 7.40 or buffer alone (control), ii) Solution B consists of AP (1-40) 40 pM or IAPP 20 pM, Thioflavin T 10 mM in 10 0.02M Tris/0.02M acetate/0.15M NaC1 at pH 7.40. This solution is prepared by drying the Ap peptide under nitrogen and then resuspending this in 0.04M Tris base with 15 minutes sonication. An equal volume of 0.04M acetic acid containing 0.3 M NaCl is added and the solution is adjusted to 7.40i0.02. A small volume of 20 mM Thioflavin T is added to the solution to give a final 5 gM concentration of Thioflavin T. 15 iii) The microplate is loaded with 40 ptL of solution A followed by 40 pL of solution B which gives a final 20 [.M Ap (1-40) or 10 FM IAPP, 5 gM Thioflavin T, and a given concentration of test compound in 0.02M Tris/0.02M acetate/0.15M NaC/0.005% azide, pH 7
.
40 . The plate is sealed and loaded into the microplate fluorescence reader. Fluorescence 20 measurement data analysis: The HTS-7000 Bio Assay Reader, Perkin Elmer, is used to perform kinetic runs of about 1 day. Readings were taken at 15 minute intervalswith one minute shaking before each read. Bandpass filters used were: excitation 430 nm, emission 485 mm. Similarly, in the electron microscopy ("EM") assay, each sample was sonicated for 1 25 min to disrupt large clumps before testing. The sample (5-gL aliquot) was placed on freshly cleaved mica and allowed to air dry. The mica was placed in a Balzers High-vacuum, Freeze-Etch Unit (model 301), shadowed with platinum (30" angle), and coated with a carbon film. The replica was removed from the mica by flotation and transferred onto a 300-mesh copper grid. Samples were examined using a Joel 2000 FX transmission electron 30 microscope. 53 WO 03/103598 PCT/US03/17992 In the circular dichroism ("CD") assay, samples were transferred to 0.1-cm path length quartz cuvettes and CD scans were taken using a Jasco J-715 spectropolarimeter. Readings were taken at 37 *C, between 190 and 240 nm, with a resolution of 0.1 nm and a bandwidth of 1 nm. 5 And in the mass spectroscopy ("MS") assay, samples were prepared as aqueous solutions containing 20% ethanol, 200 IM of a test compound and 20 sM of solubilized Ap40. The pH value of each sample was adjusted to 7.4 (i0.2) by addition of 0.1% aqueous sodium hydroxide. The solutions were then analyzed by electrospray ionization mass spectroscopy using a Waters ZQ 4000 mass spectrometer. Samples were introduced 10 by direct infusion at a flow-rate of 25 L/nin within 2 hr. after sample preparation. The source temperature was kept at 70 *C and the cone voltage was 20 V for all the analysis. Data were processed using Masslynx 3.5 software. The MS assay gives data on the ability of compounds to bind to Ap, whereas the ThT, EM and CD assays give data on inhibition of fibrillogenesis. 15 Some selected compounds of the present invention are presented in Table 2 below. Although particular salts ate depicted (such as the hydrochloride), the free base and other pharmaceutically acceptable salts are within the present invention. Table 2 Structures and Activities of Some Compounds of the Invention in Soluble AP 20 Assays Code ThT MS Structure C eTiT CD EM M No. Assay Assay HN H\ 1 + + H 50 2HCl 1 + + + + H4O250;2 1 + + + [HOCH2CH 2
SO
3 H]2. 54 WO 03/103598 PCT/US03/17992 SrcueCode ThT C EMMS StructureNo. Assay C M Assay NH
C-NH
2
(HCI)
2 2
NH
2 0
C-NH
2 HOI 3
Q-NH
2 NH HN 0S 1 SO HCI 4
H
2 N HNNH 2 H 2 N NH N.2HC1 H * NE H22-0-0.2 Ha 3<-~ / NH 2 .6 ± + + + BN NH
H
2 0 0 -\\-f 9N2 + + + + 112 -e NH 2 1 + 0211CNH 2H + + I + BN NI 55 WO 03/103598 PCT/US03/17992 Structure Code Ta CD EM Asay
NH
H 2HC .12 + + + + -, \ /NH, Ht' - NH
H
2 0 0 2HC1 13 + + + + O O2HQ0 14 + + H H O O 2H 15 + + H H O O2Q 16 + + - + H H O17 + - + H -H O O2HQ 18 + + - + HH <N 19 + + + H H O O -J ] 20 nd - + H H Br Br H NH 2\ 1925 + + nd Br Br [N 0
N
2 1 0 1 2 pr nd H Br Br H BH 25601 24 + + EN \. NH 2 NNH 25 +I + rid H 56 WO 03/103598 PCT/US03I17992 Structure Code Thff CD EM Asa No. AssayAsa H2 O - 2HC 26. nd \/NH N\O~~o'N 2H 27 pr nd H /-]H H NH NH20 NH
C-NH
2 29 00H 3 /(HEd) 2 30 N-4u 31. + + HN32 + ON- O H3 33 + 34- +- + H H HN -36 + + -37 + - + 38 + - + EN O~~COOH 39 + + 57 WO 03/103598 PCT/US03I17992 Structure CoeTTCD EM Asa NAssayAsa BN NH
H
2 41 + + 2 NH 4 C1 Ha 01-,COH42 + Br/ 43 + * HN HBr. / -- "r 44 +
H
2 N Br Ha C j-I-'zXNo 45 +
H
2 N HN\ NH, HCI 46 li- - d "NN 2HCI HN NNH NH 2 47
NH
2 'N48 + HN NH HCI' HC' HN JNH, HNNH 49 00 50 HN YNH HN YNH
NH
2 CF3COzH CF3CO 2 H NH 2 51 HCHI HCI HN H2MN NH HN"NH 2 NH HCI
NH
2 52 58 WO 03/103598 PCT/US03/17992 Structure Node Assa CD EM S NH HcI O O NH2 No. AsayAsa NQ- 0c HNtNNH SN54 H2NNNIz54
H
2 N H 2 NH 2HHCI H2N H H N NH 2 55 NH 0F 2 00 2 H CF 3
CO
2 H NH - J w 56 57 + NI 5I I
NH
2 r NH 2 I H 2 59-I
N
0 I HH NH2 0INN H NN~H H
-
60 + - + N YIC 0 'YNH 59 WO 03/103598 PCT/US03/17992 Structure Code ThT CD EM MS StructureNo. Assay D Assay HN - O O H 61 + + + H H HN NH 62 + + NH 06 H. H NkN63' + H
H')
NH?
NH
2 HN -~\ /~ NH 6 4+ NN CN \I \ N~ 65 - +. + NN -I H H I-N -& - IC 42 66 +
H
2 . H
H
2 N HN N- 67 0 60 WO 03/103598 PCT/US03/17992 Structure Code ThT CD EM MS StutueNo. Assay Assay 68
H
2 N H NH 2 HN 69 -0 H2 70 + + +
-
H + N 71 H NI HH
H
2
NH
2 HN NH 73 N/ H H
H
2 N~ /~ ' 74 + + -NH, HN HN NH H, - r- NH, 7 6 + + HIM-PNH NH HN
H
2 N NH 77 61 WO 03/103598 PCT/US03/17992 Code' ThT MS Structure No Assay CD EM Assay
H
2 N HN -/ ac)A~ 78 + +
NH
2 HN OH 079 +
H
2 N 81NHZ HN NH NH 2 N 8 HN -~ N N ' N,0 80. + H H
NH
2 HN NN 8 81+ SN+ 62 HNH HH N H2NH HN Z H 84 + H 0 0H N N 8
H
2 -- H 2 H NNH 86 + o H IH ~ /P 62 WO 03/103598 PCT/US03I17992 Str ucture Code Thff CD 'EM MS No. Assay Assay N 87 + N 89 1 H 'H H H H0 H H > jc M),- 91 + N0 1 N 92 + + + H H HN- 0 a/ NH HN
NH
2 H - I -~-,)- 94 + HH HOO\/ NOH 9' + 63 WO 03/103598 PCT/US03/17992 Structure No.e Assa CD EM Asa HOI HCI HCI CD .sEM Asay 0 0 HIO H,0 -H -H96 + N N-H
NH
2
NH
2 HN. N NH 10+ HN H H NH H Nllr<.N \/ NH 98 + *0 0 HN - NH 99 + Yee
NH
2
NH
2 0 HN - NH 100 + HNT, -,NH SN HN N - OH+ NN 7 102 +
NH
2 NH KN I ' ~ / \~ NH HN -~N N N1032 NH NH 64 WO 03/103598 PCT/US03I17992 Code ThiT MS Structure No. Assay CD EM Assay
N
HN NN 104 + HH
N
2 H
N
HNN 105+
NH
2 H N N HN NH 106 + NH NH N N 107
NH
2 NH 2 "NN HN K '. NH 103 +
NH
2 NH 2 HN ~- NH 109 + NH 2 NH, H
\/NH
2 I .. H 110+ /NH FIN NH, 65 WO 03/103598 PCT/US03/17992 Structure Code Thi CD EM MS StrctreNo. Assay Assay 0aN HN / \111+
H
2 N NH H NH 11 +N, 4 N + HN N HN H/113 + + HNH NH 2 HN - NH 114 SNH 2 H N I \' 1 N Y 111 HN NH, ' H 11 -+
H
2 S11
NH
2 H - NH' + + 'N2N H NH 117 + + ,NN HNa HN0 NH 118
H
2 N NH 2
H
2 N+ 66 WO 03/103598 PCT/US03/17992 Structure Code ThT CD EM S StrutureNo. Assay EM Assay H H
H
2
NH
2 120 + + H2N' 1* 0n 0 121 + + HN NH
H
2 N NH HN NH, N NH 12+ + I-' . HN 122 - + +
NH
2 F N 123 + + -H NHN fNH 124, + +
NH
2 N7 125 + + N HH N HH 'P NH 2 127 + H N
NH
2 67 WO 03/103598 PCT/US03/17992 Structure Code TT CD EM As Str NH2ay H2NAN S HN O 128 9 + + HNI'N NH 2
H
2 N -NH HN\~ \ /\ 0
H
2 N NH
NH
2 HN HHNH 130
^NH
2 NH
H
2 HN HN/\ 131
NH
2 N
H
2 N
CF
3
CO
2 H NH H CF 3 COZH NH HN 132 Br Br IIi HM -C NH N 1 133
CH
3 O NHOCH3 HN NH 134 + - + 0 \/0]135 HN NH 68 WO 03/103598 PCT/US03/17992 Structure Code ThT CD EM MS StrutOr No. Assay Assay OH OH 136+ HN H . H C 2 COC1 N2HCCIC1zH 2
NH
2 NH' CFCOH CF 3 CO2H N'H
H
2 N O
NH
2 139 HO'N CFc0 2 H CF 3
CO
2 H N' OH
H
2 N
NH
2 140 HO'N CF 3 2 H CF 3
C
2 H N OH H2N C2 CFC2N NH2i
H
2 N 0 NH 2 HON CF 3 CO 2 H CF 3 C0 2 H N OzH 0 0 / 142
H
2 N CF3CO 2 H CF 3
CO
2 H /NH 2 H HN H NH
H
2 NN NH 2 14 I IO N I _, _14 CFsCO 2 H I H H I o 0 CF3CO 2 H HN NH 3 CI N. 144
NO
2 In each indicated assay, "+"P= active; "-"=inactive; "pr"= promoting; "nd" or blank entry not determined. 69 WO 03/103598 PCT/US03/17992 The following compounds in Table 3 may also be employed according to the methods described herein. Table 3 Additional Exemplary Compounds for Use in The Methods of The Invention 5 HN NH 2HC
H
2 N / N " 2HCI NN 2HCi N2HC1 H. 7NH NH HN - NH \/ ~ o'"' ON 0 ' I12HC1
H
2 N / NH 2 2C 0,-"," J 2H 1 7, o--c -2HC1 HH2N \H2H ' ~>-' \/ +C+~ \ /~-(~ 2HCI ~S\/ +C 0\/SK/~2HCI H24 N N0 WO 03/103598 PCT/US03/17992 N )-N~\, O*CH 2 ) C-O \ / - HN~ 2HCI H H -H
H
2 N Cz- CH2 ff- C : D -2HCI H H H H N N \/ a-(- CH- 2 ) C-0 HN 2HCI N N H H N HH22 H N C -NC O-JCH 2 ) e-0 C-/N- 2HCI N N N~ C=- O-(CH 2 ),F jO /c=- 2HCI c N
NH
2 N -N H NH H ML O-(CH 2 ),F-o \ / -K . 2HCI
H
2 N
NH
2 HN HH 2 -- HH N ~N-C~ /0-(CH 2 ),i-O \/CNK .2HC
H
2 N
NH
2 HNH -- HH N 7 S-CC &/O-(CH 2 )~- \ / C&S_< 2HCI
H
2 N
NH
2 HN H H2H2 H71 WO 03/103598 PCT/US03I17992 NH 00 NH S-NH_ NH-S-f / \ 2.HCl
NH
2 0 0 NH 2 NH 0 0 NH II~~~ I / \2HCI
NH
2 - NH 2 C N 0 0N S-NH NH- / \ NH - 0 - NH NH0 0 N ~ C-NH 0 ~~ N-S 2 HCI NH - -0 NH NH .0 0 NH
NH
2 - NH 2 ' [NH 0 0 NH C-NH NH-C2 C NH H N 00N - 1 -- H ~ H- '- NH HCI N;- 0 0C-2 N ~NH '- "' NH""2C 72 WO 03/103598 PCT/US03I17992 NH / -S-NH / N HC
NH
2 -H- 2i HC o -0 NH 2 NH 0 0 N II NH /~N- 2 SN/ 2HCI NH, -0 NH 2 NH 0 / \ II0 NH
N
2 - NH-S/ 2 HCI o 1 -0
NH
2 No 0N S-N /1 _,,N N0 2 HCI NHN -s"I I - -/< o0 NH o 0 -a 11is, < N 2 HCI NH - 00 - NH 9 NH / 0 / N
NH
2
-
H- 2 HCI - NH 2 0H 0 - NH 73 WO 03/103598 PCT/US03/17992 NH 0 .0 NH
NH
2 -O OHNH 2 -NH NH-0 C NH' 0 NH
NHOO
2 NH-C 2 HCI -H NH 2 N / 0 N /H2j22HHC NHH N 0 0 NHN HN NH [00 100 3 Carbon chain length (n) H H 15 74 WO 03/103598 PCT/US03/17992 IAPP fibrillogenesis 100 S 80 - - - - -- - - 60 - - - - -200 uM 2 40- 0100 uM 20 0 1 2 3 4 5 6 7 Carbon chain length (n) The present invention also relates to novel compounds and the synthesis thereof. Accordingly, the following examples are presented to illustrate how some of those 5 compounds may be prepared. General Aspects Chemicals were purchased from Aldrich. Analytical thin-layer chromatography (TLC) was performed on silica gel 60 F 25 4 plastic-backed plates. Solvents were reagent 10 grade unless otherwise specified., The 1H (500 MHz) and "C (125 MHz) were recorded on a Varian Inova 500. The chemical shifts are reported on the 8 scales in parts per million (ppm). The infra-red (IR) spectra were carried out on a Perkin-Elmer Spectra One spectrometer (neat compound on NaCi plate). 15 1,4-bis(4-anidinoanilino)butane H2 N NH2 Step 1: 1,4-bis(4-cyanoanilino)butane NC F + H2N NH2 Et 3 N, DMSO N 44 -ef.. A mixture of 4-fluorobenzonitrile (3 g, 0.025 mol), 1, 4-diaminobutane(0.6 g, 0.006 20 mol), triethylamine (5 mL) and DMSO (16 mL) was heated at 150 "C with stirring for 3h. The mixture was then poured into iced water (250 mL) and the precipitate was collected by filtration. Recrystallization of the crude product (0.58 g) from DMSO/II 2 O (6:1) gave the product as a light yellow solid, 0.48 g, yield 27.6%. 75 WO 03/103598 PCT/US03/17992 Step 2: 1,4-bis(4-anidinoanilino)butane LHCl(gas)' NN Nc--(GX.N .NH H N EtOR/dioxane 74H~ 2. NH 3 NEtOH H 2 N NH 2 A mixture of 1,4-bis(4-cyanoanilino)butane (0.44 g, 1.52 mmol) in ethanol (30 mL) 5 and dioxane (10 mL) was cooled to 0 *C and saturated with HCI gas. The resulting mixture was stirred at room temperature until the IR indicated the disappearance of the nitrile absorbance peak at -2200 cm. Diethyl ether (100 mL) was added, and the formed precipitate was collected and washed with diethyl ether. The solid thus obtained was then placed into a 50-mL round bottom flask. Ethanolic ammonia (2 M, 30 mL) was slowly 10 added via syringe. The resulting mixture was refluxed for 3h, and then was cooled to room temperature. Diethyl ether (100 mL) was added to induce precipitation. The precipitate thus formed was collected, washed with ether, and recrystallized from H20 to give 0.50 g of product, yield 99%. 15 Linear dibenzamidine and diimidazolino compounds H2 0O 0 H 2HC - 0 2HC1
H
2 N -0 NH 2 N H H n=-3-10 n4-~10 Step 1: ax, w-bis(4-cyanophenoxy)alkanes Sodium (1.2 g, 0.05 mol) was cut into small pieces and slowly added to a stirred solution of dry ethanol (40 mL). After complete dissolution of the sodium, 4-cyanophenol 20 (6 g, 0.05 mol) was added and followed by the dropwise addition of 1,4-dibromobutane (5.4 g, 0.025 mol). The resulting mixture was stirred at reflux for 1-2 days and then cooled to room temperature. The white solid formed in the reaction was collected by vacuum filtration, washed with water and dried under vacuum. The obtained product, 1,4-bis(4 cyanophenoxy)butane (7.18 g, 98% yield), was used directly for the next step without 25 purification. Analogous compounds with n = 3, 5, 6, 7, 8, 9, and 10 were prepared and yields ranged from 70 - 95%. The 1H and "C NMR of the compounds were consistent with the structures. Step 2: Dibenzamidines and diinidazolino compounds 76 WO 03/103598 PCT/US03/17992 A mixture of a, co-bis(4-cyanophenoxy)alkane (3.42 mmol), dioxane (15 mL) and ethanol (40 mL) was cooled to 0 *C. Dry HCl gas was bubbled through the mixture until saturation. The mixture was stirred at room temperature until the IR nitrile absorbance at 2200 cm' subsided. 5 Diethyl ether (100 mL) was then added and a white precipitate was formed. The precipitate was collected by vacuum filtration, washed with diethyl ether, and placed into a 50-mL round bottom flask. Ethanolic ammonia solution (2 M, 30 mL; in the preparation of dibenzamidines) or ethylenediamine in MeOH (1.5 M, 30 mL; in the preparation of diimidazolino compounds) was added slowly via syringe. The resulting mixture was stirred 10 at reflux for 3h. After the mixture was cooled to room temperature, diethyl ether (100 mL) was added. The white precipitate that formed was collected and washed with diethyl ether. The solid was then recrystallized with HCI (2 N) giving the desired product. Dibenzamidine compounds with n= 3 -10 were prepared and yields ranged from 60 - 85%. Diimidazolino compounds with n=4 -10 were prepared and yields ranged from 50 -92%. 15 1-( 4 -amidino)phenoxy-8-bromooctane, hydrobronide HN HBr. H2HO Step 1: 1-(4-cyano)phenoxy-8-bromooetane In a 100-mL round-bottom flask were placed 4-cyanophenol (2.38 g, 20 mmol), 20 K( 2
CO
3 (anhydrous, 25 mmol) and DMF (50 mL). The mixture was stirred at room temperature for 30 min. When the mixture became cloudy, 8-bromooctanol (20 mmol) was added dropwise via syringe. The mixture was then refluxed for 5h, cooled to room temperature, and poured into iced water (200 mL). White precipitate was formed and . collected by vacuum filtration. The pure product (4.1 g, 88.7% yield) was obtained as a 25 white solid after silica gel flash column chromatography (eluent: 20 - 40% ethyl acetate in hexane). Step 2: 8 -(4-anidinophenoxy)octanol The corresponding amidine compounds were obtained by serial treatments with saturated ethanolic hydrochloride solution and ethanolic ammonia analogously as described 30 above. Step 3: 1-(4-amidinophenoxy)-8-bromooctane, hydrobromide 77 WO 03/103598 PCT/US03/17992 In a 50-mL round-bottom flask were placed 8-(4-amidinophenoxy)octanol (2.14 g, 6.8mmol) and dichloromethane (30 mL). The mixture was cooled to 0 "C, and PBr (3.4 mmol, 0.5 eq.) was added dropwise via syringe. Then the mixture was stirred overnight at room temperature. The white solid starting material gradually dissolved and turned into a 5 yellow oil phase immiscible with the dichloromethane. Upon completion of the reaction, water was added to quench the reaction, and the dichloromethane was evaporated under reduced pressure to give a white solid as crude product. Pure product (white solid, 780 mg, 31% yield) was obtained after silica gel' flash column chromatography (eluent CHC1 3 /MeOHIAcOH 9415/1) and subsequent recrystallization from HBr/CH 3 CN (2 N). 10 9-(4-amidinophenoxy)nonanoic acid, hydrochloride B1N H 2N H O ,- COOH Step 1: 9-(4-cyanophenoxy)nonanol In a 100-mL round-bottom flask, 4-cyanophenol (2.38 g,. 20 nmol) and K 2 C0 3 15 (anhydrous, 25 mmol) were mixed in DMF (50 mL). The mixture was stirred at room temperature for 30 min. When the mixture became cloudy, 9-bromononanol (20 mnol) was added dropwise via syringe. The mixture was then refluxed for Sh, cooled to room temperature, and poured into iced water (200 mL). The white precipitate that formed was collected by vacuum filtration. The pure product (4.8 g, 98 % yield) was obtained as a white 20 solid after silica gel flash column chromatography (eluent: 20 - 40% ethyl acetate in hexane). Step 2: 9-(4-cyanophenoxy)nonanoic acid To a solution of 9-(4-cyanophenoxy)nonanol (2.5 g, 10.2 mmol) in DMF (50 mL), PDC (19 g, 61 nmol, 6 eq.) was added. The mixture was stirred at 50 "C overnight, then 25 cooled to room temperature, and poured into iced water (150 mL), The mixture was extracted with ethyl acetate (4 x 50 mL). The combined organic layers were Washed with brine and dried over sodium sulfate. Purification via silica gel flash column chromatography (eluent 25 -50 % ethyl acetate in hexane) gave product as a white solid, 1.65 g, 62% yield. Step 3: 9-(4-cyanophenoxy)nonanoic acid, ethyl ester 78 WO 03/103598 PCT/US03/17992 In a 100-mL round-bottom flask, thionyl chloride (0.88 mL, 12 mmol) was added to anhydrous ethanol (50 mL). The mixture was stirred for 10 min, then 9-(4 cyanophenoxy)nonanoic acid (1.65 g, 6.02 mmol) was added in one portion. The reaction was monitored by TLC. Upon completion of the reaction,. ethanol was removed under 5 reduced pressure. Ether (100 mL) and saturated sodium bicarbonate solution (100 mL) was added. The organic phase was separated and dried over sodium sulfate. The product (1.6 g, 87.7 % yield) was obtained as a white solid after evaporation of the solvent. Step 4: 9-(4-amidinophenoxy)nonanoic acid hydrochloride 9-(4-cyanophenoxy)nonanoic acid ethyl ester (1.6 g, 5.28 mmol) was dissolved in a 10 mixture of ethanol and dioxane (50/10 mL) in a sealed 100-mL round-bottom flask. The mixture was saturated with HC (g) at 0 'C and stirred at room temperature until IR showed the disappearance of the nitrile absorbance at 2200 cm 1 . Ethanol/dioxane was then removed under reduced pressure, and ether (100 mL) was added to induce precipitation. The precipitate was collected and immediately placed into a dry 100-mL flask. Ethanolic 15 ammonia (2 M, 40 mL) was added via syringe. The mixture was refluxed for 3 h, followed by removal of the solvent and addition of ether to induce precipitation. The solid that formed was collected and recrystalllized from HCl (2 N). Final product was obtained as a colorless needle crystal, 0.56 g, 32.3 %. 1H NMR (500 MHz, DMSO-d 6 ): 11.96 (s, 1H1), 9.16 (s, 2H), 8.85 (s, 2H), 7.80 (d, 2H, J= 8.5 Hz), 7.13 (d,2H, J=8.5 Hz), 4.06 (t, 2H, J= . 20 6.5 Hz), 2.18 (t, 2H, J = 7.5 H1z), 1.73-1.70 (m, 211), 1.49-1.46 (m, 2H), 1.40-1.38 (m, 2H-), 1.30-1.27 (m, 6H); "C NMR (125 MIHz, DMSO-d6): 174.46, 164.71, 163.08, 130.15, 119.23, 114.74, 68.08, 33.66, 28.66, 28.57, 28.47, 28.40, 25.34, 24.46. Some SubstitutedPentamidines 25HC 25 -H2N7 OR R22~ Step 1: 1,5-Bis(4-cyano-2-methoxyphenoxy)pentane NC OH+Br Br NaOEOOII NC NC< -0 0l 7CN R2 R2 Rj 79- WO 03/103598 PCT/US03/17992 Sodium (0.3 g, 0.014 mo1) was cut into small pieces and slowly added to a stirred solution of dry ethanol (30 iL). After complete dissolution of the sodium, 4-hydroxy-3 methoxybenzonitrile (2 g, 0.013 mol) was added and followed by the dropwise addition of 1, 5-dibromopentane (0.9 mL, 0.007 mol). The resulting mixture was stirred at reflux for 2 5 days, and then cooled to room temperature. The light brown precipitate in the mixture was collected, washed with water and dried under vacuum. The product obtained (1.45 g, 73%) was used directly for the next step without purification. The 'H and "C NMR of the compounds were consistent with the structures. Step 2: Corresponding Pentamidines LO A mixture of substituted 1,5-bis(4-cyanophenoxy)pentane (in this example, R 1 = methoxy and R 2 = hydrogen) (1.8 g, 4.91 mmol), dioxane (15 mL) and ethanol (50 mL) was cooled to 0 "C. Dry HCI gas was bubbled through the mixture until saturation. The mixture was stirred at room temperature until lR showed the disappearance of the nitrile absorbance at 2200 cm m . Then diethyl ether (100 mL) was added and the white precipitate that formed 15 was collected by vacuum filtration and washed with diethyl ether. The white solid obtained was placed into a 50-mL round-bottom flask and ammonia ethanol solution (2 M, 30 mL) was added slowly via syringe. The resulting mixture was stirred at reflux for 3h. After the mixture was cooled to room temperature, diethyl ether (100 mL) was added and a white precipitate formed. The precipitate was collected and washed 20 with diethyl ether. The solid was then recrystallized from 2 N HCI giving the desired product (0.92 g, 40% yield). In like manner, the corresponding compound with R bromine and R 2 = bromine was synthesized in 53% yield. Compound # 139 NCCN 1) Na 2
CO
3 , NH 2 OH.HCI H20, EtOH, A 2) Preparative HPLC I NH 2 25 HO CF3CO 2 H CF 2
CO
2 H OH 80 WO 03/103598 PCT/US03/17992 A mixture of 1,5-bis(4-cyanophenoxy)pentane (153 mg, 0.5 mmol), sodium carbonate (180 mg, 1.7 mmol) and hydroxylamine hydrochloride (278 mg, 4 mmol) in 80 % ethanol (10 mL) was heated at reflux for 2h, The mixture was cooled to room temperature. Some solid precipitated and was removed by filtration. The filtrate was concentrated to 5 dryness under reduced pressure. The crude product was purified by preparative RP--HPLC (Vydac C18, 215 nm, 50 mL/min, 0 % to 90 % MeCN in H20 containing 0.1 % TFA) and lyophilized to give a white solid, 127.2 mg, 42%. The heptane and nonane anolags were prepared in the same way. .0 Compound # 55 0 NCI NMF NC CN 0 0 1) HCI, 1,4-dioxane, EtOH H 2 NH2 2) (NH 4 )C0 3 , ELOH e o 3) Preparaive HPLC NH CF 3
CO
2 H CFaCO 2 H NH Step 1: To a cold solution (0 *C) of 1,5-diaminopentane (0.35 mL, 3 mmol) and triethylamine (0.98 mL, 7 mmol) in DME (10 mL) was added 4-cyanobenzoyl chloride (1 g, 6 mmnol). The mixture was stirred overnight at room temperature, and then diluted with [5 water. The beige solid that precipitated was collected by filtration and dried in vacuo, giving the corresponding amid, 1 g, 92 %. Step 2: A suspension of the 1,5-bis-(4-cyanobenzanido)pentane (465 mg, 1.3 mmol), in a mixture of absolute ethanol (25 mL) and 1,4-dioxane (20 mL), was cooled to 0 *C, saturated with dry HC1, and the resulting mixture was stirred for 60 hours at room 20 temperature. The solvent was evaporated under reduced pressure. A brownish solid was obtained. A mixture of the solid and ammonium carbonate (2.5 g, 25 mmol) in ethanol (25 mL) was stirred overnight at room temperature. A small amount of activated charcoal was added, then the mixture was filtered over Celite. The solvent was evaporated under reduced pressure. The crude product was purified by preparative RP-HPLC (Vydac C18, 215 nm, 50 25 mL/min, 0 % to 90 % MeCN in H20 containing 0.1 % TFA) and lyophilized to give the title compound as a white solid, 410 mg, 51%. The heptane and nonane analogs were prepared in the same way. . 81 WO 03/103598 PCT/US03/17992 Compound # 54 Hya OH+
K
2 0O 3 0 DMF 0 0 Step 1: A mixture of4-hydroxybenzaldehyde (2.7 g, 22 mmol), 1,5-dibromopentane (1.35 mL, 10 mmol) and potassium carbonate (5.2 g) in dry DMF (25 mL) was heated at 5 100 "C with an oil bath for 5 hours. The mixture was cooled to room temperature, then water (100 mL) was added. The solid that formed was collected by filtration, rinsed with water and dried in vacuo. The desired bis-aldehyde was obtained as a brownish solid, 2.8 g, 89%. 0 0~ N.N H H + '(I-Pro)2- CN NC O o CN oI KZC03 N'N 0 0 THF to Step 2: Diisopropyl (cyanomethyl)phosphonate (0.86 mL, 4.2 mmol) was added to suspension of sodium hydride (4.4 mmol) in THF at (0 *C).The mixture was stirred at room temperature for 1 hour. A solution of the bis-aldehyde (2 mmol) in-TEF was added. The mixture was stirred at room temperature for 2h, then diluted with ethyl acetate, washed subsequently with water, saturated sodium bicarbonate, brine and dried over magnesium [.5 sulfate. The solvent was evaporated under reduced pressure. The crude solid was washed with a mixture of ethyl acetate and hexane (1 to 10, 10 mL) the dried in vacuo to afford the bis-nitrile, 0.51 g, 71 % yield. 0 o 1) HCI, EtOH H 2 N H NC ON 2) NH3, MtcH R NH HCI HCI NH 82 WO 03/103598 PCT/US03/17992 Step 3: A suspension of the bis-nitrile (0.48 g, 1.34 mmol) in ethanol (20 mL) was saturated with HCl at 0 "C. The mixture was stirred at room temperature for 3 days. The solvent was evaporated under reduced pressure. The solid was then dissolved in 2 N NH 3 in ethanol (20 mL) and the mixture was heated at reflux 'for 2h. The mixture was cooled to 5 room-temperature and the solvent was evaporated under reduced pressure. The resulting solid was dried in vacuo, then recrystallized from 2 N HC with the addition of a few drops of ethanol. The solid was collected by filtration, rinsed with water and dried overnight in vacuo, giving the title compound as a light yellow solid, 0.44 g, 71 %. Compound # 137 OH + NC CN PN DMF .0 CN O00C Step 1: A mixture of 4-hydroxybenzylcyanide (2.56 g, 19.2 mmol), 1,7 dibromoheptane (1.49 mL, 8.7 mmol), potassium carbonate (11 g) in DMF (30 mL) was heated with an oil bath at 100 "C for 3 hours. The mixture was cooled to room temperature and diluted with water (150 mL). A solid precipitated. The solid was collected by filtration 15 and rinsed with water. It was then dissolved in ethyl acetate, washed subsequently with 10% NaOH (3 x 20 mL), brine (30 mL) and dried over magnesium sulfate. The solvent was evaporated under reduced pressure. The resulting solid was dried in vacuo to give the 1,7 bis(4-cyanomethylphenoxy)heptane as a tan solid, 2.58 g, 82 %. NC OO CN ,.- I) HCI, MtOH, 0 1,4-dloxane 2) NH 3 , EtOH, A HOI NH 2 ' o H2H 20 83 WO 03/103598 PCT/US03/17992 Step 2: A solution of 1,5-bis( 4 -cyanomethylphenoxy)heptane (750 mg, 5.07 nol) in a mixture of 1,4-dioxane (10 mL) and absolute ethanol (10 mL) was saturated with HC1 at 0 "C. The mixture was then stirred at room temperature for the 3 days. The solvent was evaporated under reduced pressure and the residue was dried in vacuo, The residue was 5 'dissolved in 2 N ammonia in ethanol (20 mL) and the mixture was heated at reflux for 3h. The solvent was evaporated under reduced pressure. The crude solid was recrystallized from 2 N HC / acetone. The crystals were collected and dried in vacuo. The title compound was obtained as an off-white solid, 655.3 mg, 60 %. 10 Compound # 51, CN CN 1) BH3:THF, A 2) HCI, MeOH NH 2 HCI HCI H2 Step 1: A solution ofborane:tetrahydrofuran complex (10 mL, 10 mmol) was added to a solution of the bis-nitrile (510 mg, 1.53 mmol) at 0 "C. The mixture was then heated at reflux for 18 hours, then cooled with an ice bath. The excess of reagent was quenched by 15 the slow addition of methanol (10 mL). The resulting mixture was heated at reflux for 15 minutes, then the solvent was removed under reduced pressure. The residue was coevapotated 3 times with methanol, then suspended in mixture of methanol (20 iL) and concentrated HCI (6 rhiL). The mixture was heated at reflux for 1.5 hour. The mixture was then reduce to about 5 mL under reduced pressure. A fine white solid had formed. The 20 mixture was diluted with ethanol and cooled to -10 "C. The solid was collected by filtration, rinsed with cold ethanol and dried overnight in vacuo. The 1,5-bis (4-(2 aminoethyl)phenoxy)pentane dihydrochloride was obtained as a fine white powder, 564.6 mg, 89%. 'N NBoc + ON
NH
2 HCI HCI H 2 N NHBc NBoc O Hunig's base HN B NH THF, dichloromethane NHBCc BocHN 84 WO 03/103598 PCT/US03/17992 Step 2: NN'-bis(tert-butoxycarbonyl)-H-pyrazole-1carboxamidine (0.78 g, 2.5 mmol) was added to a suspension of the 1,5-bis(4-(2-aminoethyl)phenoxy)pentane dihydrochloride (470 mg, 1.13 mmol) and Hunig's base (0.435 mL) in a mixture of THF (5 mL) and dichloromethane (20 mL). The mixture was stirred at room temperature for 2 days. 5 Excess reagent was quenched with 1,2-ethylenediamine. The mixture was diluted with 'hloroform, washed subsequently with 1 N HCI, saturated sodium carbonatebrine, and dried over magnesium sulfate. The solvent was removed under reduced pressure. The crude product was purified by flash chromatography on silica gel (0.5 % to 1% McOH in CHC1 3 ) giving a white foamy solid 246.5 mg, 26 %. SNNH HN oo HCI' HN B. NH 1,4-dioxane HN-H HCI HCI H NH 10 NHBOc BocHN NH 2 H2N Step 3: A solution of 4 M HC1 in 1,4-dioxane (5 mL) was added to a solution of the protected bis guanidino compound (246 mg, 0.297 mmol) in 1,4-dioxane (10 mL). The mixture was stirred at room temperature for one day. The solvent was evaporated under reduced pressure. The product was dissolved in water, then the aqueous soultion was 15 lyophilized, giving the title compound as a white solid, 146.4 mg, 99 %. 85

Claims (6)

1. A method of treating or preventing an amyloid-related disease such as Alzheimer's disease, cerebral amyloid angiopathy, or type II diabetes in a subject comprising administering to said subject a therapeutic amount of an amidine compound.
2. A method of treating or preventing an amyloid-related disease in a subject comprising administering to said subject a therapeutic amount of a compound according to one of the following Formulae, such that amyloid fibril formation or deposition, neurodegeneration, or cellular toxicity is reduced or inhibited: o 0 2HCI N N H H (Formula I) -N LI~ 0 - N 0 / 2HCI H /N H (Formula II) HN CIH 3 N (Formula III) 86 WO 03/103598 PCT/US03/17992
3. A chemical compound according to the formula: N -N N O / 2HCI H H
4. A chemical compound according to the formula: N - N -N / 2HCI H N H
5. A chemical compound according to the formula: HN CIH 3 N
6. A chemical compound according to one of the following formulas: ~ ~H NH, JI K,-, 0 - H, and 87
AU2003251418A 2002-06-07 2003-06-09 Amidine derivatives for treating amyloidosis Abandoned AU2003251418A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US38700102P 2002-06-07 2002-06-07
US60/387,001 2002-06-07
PCT/US2003/017992 WO2003103598A2 (en) 2002-06-07 2003-06-09 Amidine derivatives for treating amyloidosis

Publications (1)

Publication Number Publication Date
AU2003251418A1 true AU2003251418A1 (en) 2003-12-22

Family

ID=29736249

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2003251418A Abandoned AU2003251418A1 (en) 2002-06-07 2003-06-09 Amidine derivatives for treating amyloidosis

Country Status (6)

Country Link
EP (1) EP1572129A4 (en)
JP (1) JP2006501160A (en)
AU (1) AU2003251418A1 (en)
CA (1) CA2488493A1 (en)
NO (1) NO20040497L (en)
WO (1) WO2003103598A2 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003265967A1 (en) * 2003-09-05 2005-04-21 Georgia State University Research Foundation, Inc Novel amidine compounds for treating microbial infections
WO2005040132A1 (en) * 2003-10-24 2005-05-06 University Of North Carolina At Chapel Hill Dicationic triaryl analogs as anti-protozoan agents
WO2005079780A1 (en) * 2004-01-23 2005-09-01 Neurochem (International) Limited Amidine derivatives for treating amyloidosis
US7262223B2 (en) * 2004-01-23 2007-08-28 Neurochem (International) Limited Amidine derivatives for treating amyloidosis
CA2600570C (en) * 2005-03-14 2011-12-06 Transtech Pharma, Inc. Benzazole derivatives, compositions, and methods of use as .beta.-secretase inhibitors
AU2006201739A1 (en) * 2005-05-05 2006-11-23 The University Of North Carolina At Chapel Hill Synthesis and antiprotozoal activity of dicationic 3,5-diphenylisoxazoles
SI2247558T1 (en) * 2008-02-14 2022-07-29 Eli Lilly And Company Novel imaging agents for detecting neurological dysfunction
FR2933692B1 (en) * 2008-07-10 2022-12-02 Univ Nancy 1 Henri Poincare NEW COMPOUNDS, THEIR USE AS MEDICINES, AND METHOD FOR PREPARING THEM
CA2786891C (en) 2009-02-20 2017-08-01 Meisho.Co.,Ltd Immunopotentiating composition and process for producing same

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6635668B1 (en) * 1998-07-22 2003-10-21 The University Of North Carolina At Chapel Hill Imidazoline receptor binding compounds
BR0212078A (en) * 2001-08-31 2004-09-28 Neurochem Int Ltd Method of treating or preventing an amyloid-related disease in a patient, pharmaceutical composition, chemical compound, and use of a compound.

Also Published As

Publication number Publication date
WO2003103598A3 (en) 2006-03-09
JP2006501160A (en) 2006-01-12
EP1572129A2 (en) 2005-09-14
EP1572129A4 (en) 2007-03-28
CA2488493A1 (en) 2003-12-18
WO2003103598A2 (en) 2003-12-18
NO20040497L (en) 2004-04-14

Similar Documents

Publication Publication Date Title
US20040147531A1 (en) Amidine derivatives for treating amyloidosis
CA2373093C (en) Methods and compounds for inhibiting amyloid deposits
EP1276483B1 (en) Compositions and methods for treating amyloidosis using sulphonate derivatives
US20060199771A1 (en) Stereoselective antifibrillogenic peptides and peptidomimetics thereof
AU2003251418A1 (en) Amidine derivatives for treating amyloidosis
US7262223B2 (en) Amidine derivatives for treating amyloidosis
CA2473102A1 (en) Suppression of cytotoxic protein conformers
JP2020500183A (en) Combination therapy for the treatment of pulmonary hypertension
AU2002325117A1 (en) Amidine derivatives for treating amyloidosis
US6562821B2 (en) Aryl-N-cyanoguanidines and methods related thereto
AU2004316081A1 (en) Amidine derivatives for treating amyloidosis
AU2005203635B2 (en) Methods and compounds for inhibiting amyloid deposits
AU2005202454A1 (en) Compositions and methods for treating amyloidosis

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period