CA2312920C - Prevention and treatment of amyloidogenic disease - Google Patents

Prevention and treatment of amyloidogenic disease Download PDF

Info

Publication number
CA2312920C
CA2312920C CA2312920A CA2312920A CA2312920C CA 2312920 C CA2312920 C CA 2312920C CA 2312920 A CA2312920 A CA 2312920A CA 2312920 A CA2312920 A CA 2312920A CA 2312920 C CA2312920 C CA 2312920C
Authority
CA
Canada
Prior art keywords
beta
pharmaceutical composition
immunogenic
fragment
peptide
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.)
Expired - Lifetime
Application number
CA2312920A
Other languages
French (fr)
Other versions
CA2312920A1 (en
Inventor
Dale B. Schenk
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.)
Janssen Sciences Ireland UC
Original Assignee
Janssen Alzheimer Immunotherapy
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26748211&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2312920(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Janssen Alzheimer Immunotherapy filed Critical Janssen Alzheimer Immunotherapy
Publication of CA2312920A1 publication Critical patent/CA2312920A1/en
Application granted granted Critical
Publication of CA2312920C publication Critical patent/CA2312920C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/739Lipopolysaccharides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0007Nervous system antigens; Prions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/646Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the entire peptide or protein drug conjugate elicits an immune response, e.g. conjugate vaccines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • 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
    • A61P25/28Drugs 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55577Saponins; Quil A; QS21; ISCOMS
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6037Bacterial toxins, e.g. diphteria toxoid [DT], tetanus toxoid [TT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/605MHC molecules or ligands thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7023Transdermal patches and similar drug-containing composite devices, e.g. cataplasms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Abstract

The invention provides compositions and methods for treatment of amyloidogenic diseases. Such methods entail administering an agent that induces a beneficial immune response against an amyloid deposit in the patient. The methods are particularly useful for prophylactic and therapeutic treatment of Alzheimer's disease. In such methods, a suitable agent is A.beta. peptide or an antibody thereto.

Description

= PREVENTION AND TREATMENT OF
AMYLOIDOGENIC DISEASE
TECHNICAL FIELD
The invention resides in the technical fields of immunology and medicine.
BACKGROUND
Alzheimer's disease (AD) is a progressive disease resulting in senile dementia. See generally Selkoe, TINS 16, 403-409 (1993); Hardy et al., WO 92/13069; Selkoe, J. Neuropathol.
Exp. Neurol. 53, 438-447 (1994); Duff et al., Nature 373, 476-477 (1995); Games et al., Nature 373, 523 (1995). Broadly speaking the disease falls into two categories: late onset, which occurs in old age (65 + years) and early onset, which develops well before the senile period, i.e, between 35 and 60 years. In both types of disease, the pathology is the same but the abnormalities tend to be more severe and widespread in cases beginning at an earlier age. The disease is characterized by two types of lesions in the brain, senile plaques and neurofibrillary tangles. Senile plaques are areas of disorganized neuropil up to 150 pm across with extracellular amyloid deposits at the center visible by microscopic analysis of sections of brain tissue.
Neurofibrillary tangles are intracellular deposits of tau protein consisting of two filaments twisted about each other in pairs.
2 The principal constituent of the plaques is a peptide termed AO or O-amyloid peptide. AO peptide is an internal fragment of 39-43 amino acids of a precursor protein termed amyloid precursor protein (APP). Several mutations within the APP protein have been correlated with the presence of Alzheimer's disease. See, e.g., Goate et al., Nature 349, 704) (1991) (valine717 to isoleucine); Chartier Harlan et al.
Nature 353, 844 (1991)) (valine717 to glycine); Murrell et al., Science 254, 97 (1991) (valine717 to phenylalanine);
Mullan et al., Nature Genet. 1, 345 (1992) (a double mutation changing lysine595-methionine596 to asparagine595-leucine596).
Such mutations are thought to cause Alzheimer's disease by increased or altered processing of APP to AO, particularly processing of APP to increased amounts of the long form of AO
(i.e., AO1-42 and A131-43). Mutations in other genes, such as the presenilin genes, PS1 and PS2, are thought indirectly to affect processing of APP to generate increased amounts of long form AO (see Hardy, TINS 20, 154 (1997)). These observations indicate that AO, and particularly its long form, is a causative element in Alzheimer's disease.
McMichael, EP 526,511, proposes administration of homeopathic dosages (less than or equal to 10-2 mg/day) of AO
to patients with preestablished AD. In a typical human with about 5 liters of plasma, even the upper limit of this dosage would be expected to generate a concentration of no more than 2 pg/ml. The normal concentration of AO in human plasma is typically in the range of 50-200 pg/ml (Seubert et al., Nature 359, 325-327 (1992)). Because EP 526,511's proposed dosage would barely alter the level of endogenous circulating AO and because EP 526,511 does not recommend use of an adjuvant, it seems implausible that any therapeutic benefit would result.
By contrast, the present invention is directed inter alia to treatment of Alzheimer's and other amyloidogenic diseases by administration of AO or other immunogen to a patient under conditions that generate a beneficial immune response in the patient. The invention thus fulfills a longstanding need for therapeutic regimes for preventing or ameliorating the neuropathology of Alzheimer's disease.
3 SUMMARY OF THE CLAIMED INVENTION
In one aspect, the invention provides methods of preventing or treating a disease characterized by amyloid deposition in a patient. Such methods entail inducing an immune response against a peptide component of an amyloid deposit in the patient. Such induction can be active by administration of an immunogen or passive by administration of an antibody or an active fragment or derivative of the antibody. In some patients, the amyloid deposit is aggregated AO peptide and the disease is Alzheimer's disease. In some methods, the patient is asymptomatic. In some methods, the patient is under 50 years of age. In some methods, the patient has inherited risk factors indicating susceptibility to Alzheimer's disease. Such risk factors include variant alleles in presenilin gene PS1 or PS2 and variant forms of APP. In other methods, the patient has no known risk factors for Alzheimer's disease.
For treatment of patients suffering from Alzheimer's disease, one treatment regime entails administering a dose of AO peptide to the patient to induce the immune response. In some methods, the AO peptide is administered with an adjuvant that enhances the immune response to the AO peptide. In some methods, the adjuvant is alum. In some methods, the adjuvant is MPL. The dose of AO peptide administered to the patient is typically at least 1 or 10 Ag, if administered with adjuvant, and at least 50 Ag if administered without adjuvant. In some methods, the dose is at least 100 pig.
In some methods, the AO peptide is A01-42. In some methods, the AO peptide is administered in aggregated form.
In other methods, the AO peptide is administered in dissociated form.
In some methods, the therapeutic agent is an effective dose of a nucleic acid encoding AO or an active fragment or derivative thereof. The nucleic acid encoding AO
or fragment thereof is expressed in the patient to produce AO
or the active fragment thereof, which induces the immune response. In some such methods, the nucleic acid is administered through the skin, optionally via a patch. In some methods, a therapeutic agent is identified by screening a
4 library of compounds to identify a compound reactive with antibodies to AO, and administering the compound to the patient to induce the immune response.
In some methods, the immune response is directed to aggregated AO peptide without being directed to dissociated AO
peptide. For example, the immune response can comprise antibodies that bind to aggregated AO peptide without binding to dissociated AO peptide. In some methods, the immune response comprises T-cells that bind to AO complexed with MCH1 or MHCII on CD8 or CD4 cells. In other methods, the immune response is induced by administering an antibody to AO to the patient. In some methods, the immune response is induced by removing T-cells from the patient, contacting the T-cells with AO peptide under conditions in which the T-cells are primed, and replacing the T-cells in the patient.
The therapeutic agent is typically administered orally, intranasally, intradermally, subcutaneously, intramuscularly, topically or intravenously. In some methods, the patient is monitored followed administration to assess the immune response. If the monitoring indicates a reduction of the immune response over time, the patient can be given one or more further doses of the agent.
In another aspect, the invention provides pharmaceutical compositions comprising AO and an excipient suitable for oral and other routes of administration. The invention also provides pharmaceutical compositions comprising an agent effective to induce an immunogenic response against AO in a patient, and a pharmaceutically acceptable adjuvant. In some such compositions, the agent is AO or an active fragment thereof. In some compositions, the adjuvant comprises alum.
In some compositions, the adjuvant comprises an oil-in-water emulsion. In some compositions, the AO or active fragment is a component of a polylactide polyglycolide copolymer (PLPG) or other particle. The invention further provides compositions comprising AO or an active fragment linked to a conjugate molecule that promotes delivery of AO to the bloodstream of a patient and/or promotes an immune response against AO. For example, the conjugate can serve to promote an immune response against AO. In some compositions, the conjugate is cholera toxin. In some compositions, the conjugate is an immunoglobulin. In some compositions, the conjugate is attenuated diphtheria toxin CRM 197 (Gupta, Vaccine 15, 1341-3
5 (1997).
The invention also provides pharmaceutical compositions comprising an agent effect to induce an immunogenic response against AO in a patient with the proviso that the composition is free of Complete Freund's adjuvant. The invention also provides compositions comprising a viral vector encoding AO or a an active fragment thereof effective to induce an immune response against AO. Suitable viral vectors include herpes, adenovirus, adenoassociated virus, a retrovirus, sindbis, semiliki forest virus, vaccinia or avian pox.
The invention further provides methods of preventing or treating Alzheimer's disease. In such methods, an effective dose of AO peptide is administered to a patient. The invention further provides for the use of AO, or an antibody thereto, in the manufacture of a medicament for prevention or treatment of Alzheimer's disease.
In another aspect, the invention provides methods of assessing efficacy of an Alzheimer's treatment method in a patient. In these methods, a baseline amount of antibody specific for AO peptide is determined in a tissue sample from the patient before treatment with an agent. An amount of antibody specific for AO peptide in the tissue sample from the patient after treatment with the agent is compared to the baseline amount of AO peptide-specific antibody. An amount of AO peptide-specific antibody measured after the treatment that is significantly greater than the baseline amount of AO
peptide-specific antibody indicates a positive treatment outcome.
In others methods of assessing efficacy of an Alzheimer's treatment method in a patient, a baseline amount of antibody specific for AO peptide in a tissue sample from a patient before treatment with an agent is determined. An amount of antibody specific for AO peptide in the tissue sample from the subject after treatment with the agent is compared to the
6 baseline amount of AO peptide-specific antibody. A reduction or lack of significant difference between the amount of AO
peptide-specific antibody measured after the treatment compared to the baseline amount of AO peptide-specific antibody indicates a negative treatment outcome.
In other methods of assessing efficacy of an Alzheimer's disease treatment method in a patient a control amount of antibody specific for AO peptide is determined in tissue samples from a control population. An amount of antibody specific for AO peptide in a tissue sample from the patient after administering an agent is compared to the control amount of AO peptide-specific antibody. An amount of AO
peptide-specific antibody measured after the treatment that is significantly greater than the control amount of AO
peptide-specific antibody indicates a positive treatment outcome.
In other methods of assessing efficacy of an Alzheimer's treatment method in a patient, a control amount of antibody specific for AO peptide in tissues samples from a control =
population is determined. An amount of antibody specific for AO peptide in a tissue sample from the patient after administering an agent is compared to the control amount of AO
peptide-specific antibody. A lack of significant difference between the amount of AO peptide-specific antibody measured after beginning said treatment compared to the control amount of.A0 peptide-specific antibody indicates a negative treatment outcome.
Other methods of monitoring Alzheimer's disease or susceptibility thereto in a patient, comprise detecting an immune response against AO peptide in a sample from the patient. In some such methods, the patient is being administered an agent effective to treat or prevent Alzheimer's disease, and the level of the response determines the future treatment regime of the patient.
In other methods of assessing efficacy of an Alzheimer's treatment method in a patient a value for an amount of antibody specific for AO peptide in tissue sample from a patient who has been treated with an agent is determined. The
7 value is compared with a control value determined from a population of patient experiencing amelioriation of, or freedom from, symptoms of Alzheimer's disease due to treatment with the agent. A value in the patient at least equal to the control value indicates a positive response to treatment.
The invention further provides diagnostic kits for performing the above methods. Such kits typically include a reagent that specifically binds to antibodies to AO or which stimulates proliferation of T-cells reactive with AO.
BRIEF DESCRIPTION OF THE FIGURES
Fig. 1: Antibody titer after injection of transgenic mice with A01-42.
Fig. 2: Amyloid burden in the hippocampus. The percentage of the area of the hippocampal region occupied by amyloid plaques, defined by reactivity with the AO-specific mA0 3D6, was determined by computer-assisted quantitative image analysis of immunoreacted brain sections. The values for individual mice are shown sorted by treatment group. The horizontal line for each grouping indicates the median value of the distribution.
Fig 3: Neuritic dystrophy in the hippocampus. The percentage of the area of the hippocampal region occupied by dystrophic neurites, defined by their reactivity with the human APP-specific mA0 8E5, was determined by quantitative computer-assisted image analysis of immunoreacted brain sections. The values for individual mice are shown for the AN1792-treated group and the PBS-treated control group. The horizontal line for each grouping indicates the median value of the distribution.
Fig. 4: Astrocytosis in the retrosplenial cortex. The percentage of the area of the cortical region occupied by glial fibrillary acidic protein (GFAP)-positive astrocytes was determined by quantitative computer-assisted image analysis of immunoreacted brain sections. The values for individual mice are shown sorted by treatment group and median group values are indicated by horizontal lines.
8 5: Geometric mean antibody titers to A01-42 following immunization with a range of eight doses of AN1792 containing 0.14, 0.4, 1.2, 2.7, 11, 22, 100, or 300 Ag.
Fig. 6: Kinetics of antibody response to AN1792 immunization. Titers are expressed as geometric means of values for the 5 animals in each group.
Fig. 7: Quantitative image analysis of the cortical amyloid burden in PBS- and AN1792-treated mice.
Fig. 8: Quantitative image analysis of the neuritic plaque burden in PBS- and AN1792-treated mice.
Fig. 9: Quantitative image analysis of the percent of the retrosplenial cortex occupied by astrocytosis in PBS- and AN1792-treated mice.
Fig. 10: Lymphocyte Proiation Assay on spleen cells from AN1792-treated (upper panel) or PBS-treated (lower Fig. 11: Amyloid burden in the cortex was determined by quantitative image analysis of immunoreacted brain sections for mice immunized with the AO peptide conjugates A01-5, A01-12, and A013-28; the full length AO aggregates AN1792 (AO1-42) and AN1528 (A01-40) and the PBS-treated control group.
Fig. 12: Geometric mean titers of AO-specific antibody for groups of mice immunized with AO or APP derivatives combined with Freund's adjuvant.
Fig. 13: Geometric mean titers of A$-specific antibody for groups of guinea pigs immunized with AN1792, or a palmitoylated derivative thereof, combined with various adjuvants.
Fig: 14: AO levels in the cortex of 12-month old PDAPP
mice treated with AN1792 or AN1528 with different adjuvants.
DEFINITIONS
The term "substantial identity" means that two peptide seduences, when optimally aligned, such as by the programs GAP
or BESTFIT using default dap weights, share at least 65
9 percent sequence identity, preferably at least 80 or 90 percent sequence identity, more preferably at least 95 percent sequence identity or more (e.g., 99 percent sequence identity or higher). Preferably, residue positions which are not identical differ by conservative amino acid substitutions.
For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared.
When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.
Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith &
Waterman, Adv. App/. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol.
48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Nat'l. Acad. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally Ausubel et al., supra). One example of algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described in Altschul et al., J. Mol. Biol. 215:403-410 (1990). Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). Typically, default program parameters can be used to perform the sequence comparison, although customized parameters can also be used. For amino acid sequences, the BLASTP program uses as defaults a wordlength (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff & Henikoff, Proc. Natl.
Acad. Sci. USA 89, 10915 (1989)) For purposes of classifying amino acids substitutions as conservative or nonconservative, amino acids are grouped as follows: Group I (hydrophobic sidechains): norleucine, met, ala, val, leu, ile; Group II (neutral hydrophilic side 5 chains): cys, ser, thr; Group III (acidic side chains): asp, glu; Group IV (basic side chains): asn, gin, his, lys, arg;
Group V (residues influencing chain orientation): gly, pro;
and Group VI (aromatic side chains): trp, tyr, phe.
Conservative substitutions involve substitutions between amino
10 acids in the same class. Non-conservative substitutions constitute exchanging a member of one of these classes for a member of another.
Therapeutic agents of the invention are typically substantially pure. This means that an agent is typically at least about 50% w/w (weight/weight) purity, as well as being substantially free from interfering proteins and contaminants.
Sometimes the agents are at least about 80% w/w and, more preferably at least 90 or about 95% w/w purity. However, .using conventional protein purification techniques, homogeneous peptides of at least 99% w/w can be obtained.
Specific binding between two entities means an affinity of at least 106, 107, 108' 109 M-1, or 1010 M-1. Affinities greater than 108 M-1 are preferred.
The term "antibody" is used to include intact antibodies and binding fragments thereof. Typically, fragments compete with the intact antibody from which they were derived for specific binding to an antigen. Optionally, antibodies or binding fragments thereof, can be chemically conjugated to, or expressed as, fusion proteins with other proteins.
APP699, APP781, and APP77 refer, respectively, to the 695, 751, and 770 amino acid residue long polypeptides encoded by the human APP gene. See Kang et al., Nature 325, 773 (1987);
Ponte et al., Nature 331, 525 (1988); and Kitaguchi et al., Nature 331, 530 (1988). Amino acids within the human amyloid precursor protein (APP) are assigned numbers according to the sequence of the APP770 isoform. Terms such as A039, A040, A041, A042 and A043 refer to an AO peptide containing amino acid residues 1-39, 1-40, 1-41, 1-42 and 1-43.
11 The term "epitope" or "antigenic determinant" refers to a site on an antigen to which B and/or T cells respond. B-cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, Glenn E. Morris, Ed.
(1996). Antibodies that recognize the same epitope can be identified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen. T-cells recognize continuous epitopes of about nine amino acids for CD8 cells or about 13-15 amino acids for CD4 cells. T cells that recognize the epitope can be identified by in vitro assays that measure antigen-dependent proliferation, as determined by 3H-thymidine incorporation by primed T cells in response to an epitope (Burke et al., J.
Inf. Dis. 170, 1110-19 (1994)), by antigen-dependent killing (cytotoxic T lymphocyte assay, Tigges et al., J. Immunol. 156, 3901-3910) or by cytokine secretion.
The term "immunological" or "immune" response is the development of a beneficial humoral (antibody mediated) and/or a cellular (mediated by antigen-specific T cells or their secretion products) response directed against an amyloid peptide in a recipient patient. Such a response can be an active response induced by administration of immunogen or a passive response induced by administration of antibody or primed T-cells. A cellular immune response is elicited by the presentation of polypeptide epitopes in association with Class I or Class II MHC molecules to activate antigen-specific CD4+
T helper cells and/or CD8+ cytotoxic T cells. The response may also involve activation of monocytes, macrophages, NK
12 cells, basophils, dendritic cells, astrocytes, microglia cells, eosinophils or other components of innate immunity.
The presence of a cell-mediated immunological response can be determined by proliferation assays (CD4 T cells) or CTL
(cytotoxic T lymphocyte) assays (see Burke, supra; Tigges, supra). The relative contributions of humoral and cellular responses to the protective or therapeutic effect of an immunogen can be distinguished by separately isolating IgG and T-cells from an immunized syngeneic animal and measuring protective or therapeutic effect in a second subject.
An "immunogenic agent" or "immunogen" is capable of inducing an immunological response against itself on administration to a patient, optionally in conjunction with an adjuvant.
The term "naked polynucleotide" refers to a polynucleotide not complexed with colloidal materials. Naked polynucleotides are sometimes cloned in a plasmid vector.
The term "adjuvant" refers to a compound that when administered in conjunction with an antigen augments the immune response to the antigen, but when administered alone does not generate an immune response to the antigen.
Adjuvants can augment an immune response by several mechanisms including lymphocyte recruitment, stimulation of B and/or T
cells, and stimulation of macrophages.
The term "patient" includes human and other mammalian sujpjects that receive either prophylactic or therapeutic treatment.
Disaggregated or monomeric Aa means soluble, monomeric peptide units of AO. One method to prepare monomeric AI is to dissolve lyophilized peptide in neat DMSO with sonication.
The resulting solution is centrifuged to remove any nonsoluble particulates. Aggregated Afl is a mixture of oligomers in which the monomeric units are held together by noncovalent bonds.
Compositions or methods "comprising" one or more recited elements may include other elements not specifically recited.
For example, a composition that comprises PO peptide
13 encompasses both an isolated TO peptide and All peptide as a component of a larger polypeptide sequence.
DETAILED DESCRIPTION
I. General The invention provides pharmaceutical compositions and methods for prophylactic and therapeutic treatment of diseases characterized by accumulation of amyloid deposits. Amyloid deposits comprise a peptide aggregated to an insoluble mass.
The nature of the peptide varies in different diseases but in most cases, the aggregate has a 0-pleated sheet structure and stains with Congo Red dye. Diseases characterized by amyloid deposits include Alzheimer's disease (AD), both late and early onset. In both diseases, the amyloid deposit comprises a peptide termed AO, which accumulates in the brain of affected individuals. Examples of some other diseases characterized by amyloid deposits are SAA amyloidosis, hereditary Icelandic syndrome, multiple myeloma, and spongiform encephalopathies, including mad cow disease, Creutzfeldt Jakob disease, sheep scrapie, and mink spongiform encephalopathy (see Weissmann et al., Curr. Opin. Neurobiol. 7, 695-700 (1997); Smits et al., Veterinary Quarterly 19, 101-105 (1997); Nathanson et al., Am.
J. Epidemiol. 145, 959-969 (1997)). The peptides forming the aggregates in these diseases are serum amyloid A, cystantin C, IgG kappa light chain respectively for the first three, and pr.ion protein for the others.
II. Therapeutic Agents 1. Alzheimer's Disease Therapeutic agents for use in the present invention induce an immune response against A0 peptide. These agents include A0 peptide itself and variants thereof, analogs and mimetics of A0 peptide that induce and/or crossreact with antibodies to A0 peptide, and antibodies or T-cells reactive with A0 peptide. Induction of an immune response can be active as when an immunogen is administered to induce antibodies or T-cells reactive with A0 in a patient, or passive, as when an antibody is administered that itself binds to A0 in patient.
14 AO, also known as 0-amyloid peptide, or A4 peptide (see US
4,666,829; Glenner & Wong, Biochem. Biophys. Res. Commun. 120, 1131 (1984)), is a peptide of 39-43 amino acids, which is the principal component of characteristic plaques of Alzheimer's disease. All is generated by processing of a larger protein APP by two enzymes, termed 0 and 7 secretases (see Hardy, TINS
20, 154 (1997)). Known mutations in APP associated with Alzheimer's disease occur proximate to the site of 0 or 7 secretase, or within AO. For example, position 717 is proximate to the site of 7-secretase cleavage of APP in its processing to All, and positions 670/671 are proximate to the site of 0-secretase cleavage. It is believed that the mutations cause AD disease by interacting with the cleavage reactions by which 0 is formed so as to increase the amount of the 42/43 amino acid form of All generated.
AO has the unusual property that it can fix and activate both classical and alternate complement cascades. In particular, it binds to Clq and ultimately to C3bi. This association facilitates binding to macrophages leading to activation of B cells. In addition, C3bi breaks down further and then binds to CR2 on B cells in a T cell dependent manner leading to a 10,000 increase in activation of these cells.
This mechanism causes AO to generate an immune response in excess of that of other antigens.
The therapeutic agent used in the claimed methods can be any of the naturally occurring forms of AO peptide, and particularly the human forms (i.e., A039, 040, A041, A042 or A043). The sequences of these peptides and their relationship to the APP precursor are illustrated by Fig. 1 of Hardy et al., TINS 20, 155-158 (1997). For example, A042 has the sequence:
H2N-Asp-Ala-Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr-Glu-Val-His-His-Gln-Lys-Leu-Val-Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser-Asn-Lys-Gly-Ala-Ile-Ile-Gly-Leu-Met-Val-Gly-Gly-Val-Val-IIe-Ala-OH.
A041, A040 and A039 differ from A042 by the omission of Ala, Ala-Ile, and Ala-Ile-Val respectively from the C-terminal end.
A043 differs from A042 by the presence of a threonine residue at the C-terminus. The therapeutic agent can also be an active fragment or analog of a natural AO peptide that contains an epitope that induces a similar protective or therapeutic immune response on administration to a human.
Immunogenic fragments typically have a sequence of at least 3, 5, 6, 10 or 20 contiguous amino acids from a natural Peptide.
Immunogenic fragments include A01-5, 1-6, 1-12, 13-28, 17-28, 25-25, 35-40 and 35-42. Fragments from the N-terminal half of AO are preferred in some methods. Analogs include allelic, 10 species and induced variants. Analogs typically differ from naturally occurring peptides at one or a few positions, often by virtue of conservative substitutions. Analogs typically exhibit at least 80 or 90k sequence identity with natural peptides. Some analogs also include unnatural amino acids or
15 modifications of N or C terminal amino acids. Examples of unnatural amino acids are a,a-disubstituted amino acids, N-alkyl amino acids, lactic acid, 4-hydroxyproline, 7-carboxyglutamate, E-N,N,N-trimethyllysine, E-N-acetyllysine, 0-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, w-N-methylarginine.
Fragments and analogs can be screened for prophylactic or therapeutic efficacy in transgenic animal models as described below.
AO, its fragments, analogs and other amyloidogenic peptides can be synthesized by solid phase peptide synthesis or recombinant expression, or can be obtained from natural sources. Automatic peptide synthesizers are commercially available from numerous suppliers, such as Applied Biosystems, Foster City, California. Recombinant expression can be in bacteria, such as E. coli, yeast, insect cells or mammalian cells. Procedures for recombinant expression are described by Sambrook et al., Molecular Cloning: A Laboratory Manual (C.S.H.P. Press, NY 2d ed., 1989). Some forms of AO peptide are also available commercially (e.g., American Peptides Company, Inc., Sunnyvale, CA and California Peptide Research, Inc. Napa, CA).
Therapeutic agents also include longer polypeptides that include, for example, an AO peptide, active fragment or analog _
16 together with other amino acids. For example, A0 peptide can be present as intact APP protein or a segment thereof, such as the C-100 fragment that begins at the N-terminus of A0 and continues to the end of APP. Such polypeptides can be screened for prophylactic or therapeutic efficacy in animal models as described below. The A0 peptide, analog, active fragment or other polypeptide can be administered in associated form (i.e., as an amyloid peptide) or in dissociated form. Therapeutic agents also include multimers of monomeric immunogenic agents.
In a further variation, an immunogenic peptide, such as AO, can be presented as a viral or bacterial vaccine. A nucleic acid encoding the immunogenic peptide is incorporated into a genome or episome of the virus or bacteria. Optionally, the nucleic acid is incorporated in such a manner that the immunogenic peptide is expressed as a secreted protein or as a fusion protein with an outersurf ace protein of a virus or a transmembrane protein of a bacteria so that the peptide is displayed. Viruses or bacteria used in such methods should be nonpathogenic or attenuated. Suitable viruses include adenovirus, HSV, vaccinia and fowl pox. Fusion of an immunogenic peptide to HBsAg of HBV is particularly suitable.
Therapeutic agents also include peptides and other compounds that do not necessarily have a significant amino acid sequence similarity with A0 but nevertheless serve as mimetics of A0 and induce a similar immune response. For example, any peptides and proteins forming 0-pleated sheets can be screened for suitability. Anti-idiotypic antibodies against monoclonal antibodies to A0 or other amyloidogenic peptides can also be used. Such anti-Id antibodies mimic the antigen and generate an immune response to it (see Essential Immunology (Roit ed., Blackwell Scientific Publications, Palo Alto, 6th ed.), p.
181).
Random libraries of peptides or other compounds can also be screened for suitability. Combinatorial libraries can be produced for many types of compounds that can be synthesized in a step-by-step fashion. Such compounds include polypeptides, beta-turn mimetics, polysaccharides,
17 phospholipids, hormones, prostaglandins, steroids, aromatic compounds, heterocyclic compounds, benzodiazepines, oligomeric N-substituted glycines and oligocarbamates. Large combinatorial libraries of the compounds can be constructed by the encoded synthetic libraries (ESL) method described in Affymax, WO 95/12608, Affymax, WO 93/06121, Columbia University, WO 94/08051, Pharmacopeia, WO 95/35503 and Scripps, WO 95/30642. Peptide libraries can also be generated by phage display methods. See, e.g., Devlin, WO
91/18980.
Combinatorial libraries and other compounds are initially screened for suitability by determining their capacity to bind to antibodies or lymphocytes (B or T) known to be specific for AO or other amyloidogenic peptides. For example, initial screens can be performed with any polyclonal sera or monoclonal antibody to AO or other amyloidogenic peptide.
Compounds identified by such screens are then further analyzed for capacity to induce antibodies or reactive lymphocytes to =
AO or other amyloidogenic peptide. For example, multiple dilutions of sera can be tested on microtiter plates that have been precoated with AO peptide and a standard ELISA can be performed to test for reactive antibodies to AO. Compounds can then be tested for prophylactic and therapeutic efficacy in transgenic animals predisposed to an amyloidogenic disease, as. described in the Examples. Such animals include, for example, mice bearing a 717 mutation of APP described by Games et al., supra, and mice bearing a Swedish mutation of APP such as described by McConlogue et al., US 5,612,486 and Hsiao et al., Science 274, 99 (1996); Staufenbiel et al., Proc. Natl.
Acad. Sci. USA 94, 13287-13292 (1997); Sturchler-Pierrat et al., Proc. Natl. Acad. Sci. USA 94, 13287-13292 (1997);
Borchelt et al., Neuron 19, 939-945 (1997)). The same screening approach can be used on other potential agents such as fragments of AO, analogs of AO and longer peptides including AO, described above.
Therapeutic agents of the invention also include antibodies that specifically bind to AO. Such antibodies can be .
18 monoclonal or polyclonal. Some such antibodies bind specifically to the aggregated form of AO without binding to the dissociated form. Some bind specifically to the dissociated form without binding to the aggregated form. Some bind to both aggregated and dissociated forms. The production of non-human monoclonal antibodies, e.g., murine or rat, can be accomplished by, for example, immunizing the animal with AO. See Harlow & Lane, Antibodies, A Laboratory Manual (CSHP
NY, 1988). Such an immunogen can be obtained from a natural source, by peptides synthesis or by recombinant expression.
Humanized forms of mouse antibodies can be generated by linking the CDR regions of non-human antibodies to human constant regions by recombinant DNA techniques. See Queen et al., Proc. Natl. Acad. Sci. USA 86, 10029-10033 (1989) and WO
90/07861.
Human antibodies can be obtained using phage-display methods. See, e.g., Dower et al., WO 91/17271; McCafferty et al., WO 92/01047. In these methods, libraries of phage are produced in which members display different antibodies on their outersurfaces. Antibodies are usually displayed as Fv or Fab fragments. Phage displaying antibodies with a desired specificity are selected by affinity enrichment to All, or fragments thereof. Human antibodies against AO can also be produced from non-human transgenic mammals having transgenes encoding at least a segment of the human immunoglobulin locus and an inactivated endogenous immunoglobulin locus. See, e.g., Lonberg et al., W093/12227 (1993); Kucherlapati, WO
91/10741 (1991). Human antibodies can be selected by competitive binding experiments, or otherwise, to have the same epitope specificity as a particular mouse antibody. Such antibodies are particularly likely to share the useful functional properties of the mouse antibodies.
Human polyclonal antibodies can also be provided in the form of serum from humans immunized with an immunogenic agent.
Optionally, such polyclonal antibodies can be concentrated by
19 affinity purification using AO or other amyloid peptide as an affinity reagent.
Human or humanized antibodies can be designed to have IgG, IgD, IgA and IgE constant region, and any isotype, including IgG1, IgG2, IgG3 and IgG4. Antibodies can be expressed as tetramers containing two light and two heavy chains, as separate heavy chains, light chains, as Fab, Fab' F(ab1)2, and Fv, or as single chain antibodies in which heavy and light chain variable domains are linked through a spacer.
Therapeutic agents for use in the present methods also include T-cells that bind to Afl peptide. For example, T-cells can be activated against AO peptide by expressing a human MHC
class I gene and a human 0-2-microglobulin gene from an insect cell line, whereby an empty complex is formed on the surface of the cells and can bind to AO peptide. T-cells contacted with the cell line become specifically activated against the peptide. See Peterson et al., US 5,314,813. Insect cell lines expressing an MHC class II antigen can similarly be used to activate CD4 T cells.
2. Other Diseases The same or analogous principles determine production of therapeutic agents for treatment of other amyloidogenic diseases. In general, the agents noted above for use in treatment of Alzheimer's disease can also be used for treatment early onset Alzheimer's disease associated with Down's syndrome. In mad cow disease, prion peptide, active fragments, and analogs, and antibodies to prion peptide are used in place of Afl peptide, active fragments, analogs and antibodies to AO peptide in treatment of Alzheimer's disease.
In treatment of multiple myeloma, IgG light chain and analogs and antibodies thereto are used, and so forth in other diseases.
3. Carrier Proteins Some agents for inducing an immune response contain the appropriate epitope for inducing an immune response against amyloid deposits but are too small to be immunogenic. In this situation, a peptide immunogen can be linked to a suitable carrier to help elicit an immune response. Suitable carriers include serum albumins, keyhole limpet hemocyanin, immunoglobulin molecules, thyroglobulin, ovalbumin, tetanus 5 toxoid, or a toxoid from other pathogenic bacteria, such as diphtheria, E. coil, cholera, or H. pylori, or an attenuated toxin derivative. Other carriers for stimulating or enhancing an immune response include cytokines such as IL-1, IL-1 a and 0 peptides, IL-2, TINF, IL-10, GM-CSF, and chemokines, such as 10 M1Pla and 0 and RANTES. Immunogenic agents can also be linked to peptides that enhance transport across tissues, as described in O'Mahony, WO 97/17613 and WO 97/17614.
Immunogenic agents can be linked to carriers by chemical crosslinking. Techniques for linking an immunogen to a 15 carrier include the formation of disulfide linkages using N-succinimidy1-3-(2-pyridyl-thio) propionate (SPDP) and succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC) (if the peptide lacks a sulfhydryl group, this can be provided by addition of a cysteine residue). These reagents
20 create a disulfide linkage between themselves and peptide cysteine resides on one protein and an amide linkage through the e-amino on a lysine, or other free amino group in other amino acids. A variety of such disulfide/amide-forming agents are described by Immun. Rev. 62, 185 (1982). Other bifunctional coupling agents form a thioether rather than a disulfide linkage. Many of these thio-ether-forming agents are commercially available and include reactive esters of 6-maleimidocaproic acid, 2-bromoacetic acid, and 2-iodoacetic acid, 4-(N-maleimido-methyl)cyclohexane-l-carboxylic acid.
The carboxyl groups can be activated by combining them with succinimide or 1-hydroxyl-2-nitro-4-sulfonic acid, sodium salt.
Immunogenic peptides can also be expressed as fusion proteins with carriers. The immunogenic peptide can be linked at the amino terminus, the carboxyl terminus, or internally to the carrier. Optionally, multiple repeats of the immunogenic peptide can be present in the fusion protein.
21 4. Nucleic Acid Encoding Immunogens Immune responses against amyloid deposits can also be induced by administration of nucleic acids encoding PO peptide or other peptide immunogens. Such nucleic acids can be DNA or RNA. A nucleic acid segment encoding the immunogen is typically linked to regulatory elements, such as a promoter and enhancer, that allow expression of the DNA segment in the intended target cells of a patient. For expression in blood cells, as is desirable for induction of an immune response, promoter and enhancer elements from light or heavy chain immunoglobulin genes or the CMV major intermediate early promoter and enhancer are suitable to direct expression. The linked regulatory elements and coding sequences are often cloned into a vector.
A number of viral vector systems are available including retroviral systems (see, e.g., Lawrie and Tumin, Cur. Opin.
Genet. Develop. 3, 102-109 (1993)); adenoviral vectors (see, e.g., Bett et al., J. Virol. 67, 5911 (1993)); adeno-associated virus vectors (see, e.g., Zhou et al., J. Exp. Med.
179, 1867 (1994)), viral vectors from the pox family including vaccinia virus and the avian pox viruses, viral vectors from the alpha virus genus such as those derived from Sindbis and Semliki Forest Viruses (see, e.g., Dubensky et al., J. Viral.
70, 508-519 (1996)), and papillomaviruses (Ohe et al., Human Gene Therapy 6, 325-333 (1995); Woo et al., WO 94/12629 and Xi.ao & Brandsma, Nucleic Acids. Res. 24, 2630-2622 (1996)).
DNA encoding an immunogen, or a vector containing the same, can be packaged into liposomes. Suitable lipids and related analogs are described by US 5,208,036, 5,264,618, 5,279,833 and 5,283,185. Vectors and DNA encoding an immunogen can also be adsorbed to or associated with particulate carriers, examples of which include polymethyl methacrylate polymers and polylactides and poly(lactide-co-glycolides), see, e.g., McGee et al., J. Micro Encap. (1996).
Gene therapy vectors or naked DNA can be delivered in vivo by administration to an individual patient, typically by systemic administration (e.g., intravenous, intraperitoneal, nasal, gastric, intradermal, intramuscular, subdermal, or
22 intracranial infusion) or topical application (see e.g., US 5,399,346). DNA can also be administered using a gene gun. See Xiao & Brandsma, supra. The DNA encoding an immunogen is precipitated onto the surface of microscopic metal beads. The microprojectiles are accelerated with a shock wave or expanding helium gas, and penetrate tissues to a depth of several cell layers. For example, The AccelTm Gene Delivery Device manufactured by Agacetus, Inc. Middleton WI is suitable. Alternatively, naked DNA can pass through skin into the blood stream simply by spotting the DNA onto skin with chemical or mechanical irritation (see WO 95/05853).
In a further variation, vectors encoding immunogens can be delivered to cells ex vivo, such as cells explanted from an individual patient (e.g., lymphocytes, bone marrow aspirates, tissue biopsy) or universal donor hematopoietic stem cells, followed by reimplantation of the cells into a patient, usually after selection for cells which have incorporated the vector.
III. Patients Amenable to Treatment Patients amenable to treatment include individuals at risk of disease but not showing symptoms, as well as patients presently showing symptoms. In the case of Alzheimer's disease, virtually anyone is at risk of suffering from Alzheimer's disease if he or she lives long enough.
Therefore, the present methods can be administered prophylactically to the general population without any assessment of the risk of the subject patient. The present methods are especially useful for individuals who do have a known genetic risk of Alzheimer's disease. Such individuals include those having relatives who have experienced this disease, and those whose risk is determined by analysis of genetic or biochemical markers. Genetic markers of risk toward Alzheimer's disease include mutations in the APP gene, particularly mutations at position 717 and positions 670 and 671 referred to as the Hardy and Swedish mutations respectively (see Hardy, TINS, supra). Other markers of risk are mutations in the vresenilin genes, PS1 and PS2, and ApoB4, _
23 =
family history of AD, hypercholesterolemia or atherosclerosis.
Individuals presently suffering from Alzheimer's disease can be recognized from characteristic dementia, as well as the presence of risk factors described above. In addition, a number of diagnostic tests are available for identifying individuals who have AD. These include measurement of CSF tau and A042 levels. Elevated tau and decreased A042 levels signify the presence of AD. Individuals suffering from Alzheimer's disease can also be diagnosed by MMSE or ADRDA
criteria as discussed in the Examples section.
In asymptomatic patients, treatment can begin at any age (e.g., 10, 20, 30). Usually, however, it is not necessary to begin treatment until a patient reaches 40, 50, 60 or 70.
Treatment typically entails multiple dosages over a period of time. Treatment can be monitored by assaying antibody, or activated T-cell or B-cell responses to the therapeutic agent (e.g., AO peptide) over time. If the response falls, a booster dosage is indicated. In the case of potential Down's syndrome patients, treatment can begin antenatally by administering therapeutic agent to the mother or shortly after birth.
IV. Treatment Regimes In prophylactic applications, pharmaceutical compositions or medicants are administered to a patient susceptible to, or otherwise at risk of, a particular disease in an amount sufficient to eliminate or reduce the risk or delay the outset of the disease. In therapeutic applications, compositions or medicants are administered to a patient suspected of, or already suffering from such a disease in an amount sufficient to cure, or at least partially arrest, the symptoms of the disease and its complications. An amount adequate to accomplish this is defined as a therapeutically- or pharmaceutically-effective dose. In both prophylactic and therapeutic regimes, agents are usually administered in several dosages until a sufficient immune response has been achieved. Typically, the immune response is monitored and
24 repeated dosages are given if the immune response starts to fade.
Effective doses of the compositions of the present invention, for the treatment of the above described conditions vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is human or an animal, other medications administered, and whether treatment is prophylactic or therapeutic. Usually, the patient is a human, but in some diseases, such as mad cow disease, the patient can be a nonhuman mammal, such as a bovine. Treatment dosages need to be titrated to optimize safety and efficacy. The amount of immunogen depends on whether adjuvant is also administered, with higher dosages being required in the absence of adjuvant. The amount of an immunogen for administration sometimes varies from 1 Ag-500 Ag per patient and more usually from 5-500 Ag per injection for human administration. Occasionally, a higher dose of 1-2 mg per injection is used. Typically about 10, 20, 50 or 100 Ag is used for each human injection. The timing of injections can vary significantly from once a day, to once a year, to once a decade. On any given day that a dosage of immunogen is given, the dosage is greater than 1 g/patient and usually greater than 10 Ag/ patient if adjuvant is also administered, and greater than 10 g/patient and usually greater than 100 pig/patient in the absence of adjuvant. A typical regimen consists of an immunization followed by booster injections at 6 weekly intervals. Another regimen consists of an immunization followed by booster injections 1, 2 and 12 months later. Another regimen entails an injection every two months for life. Alternatively, booster injections can be on an irregular basis as indicated by monitoring of immune response.
For passive immunization with an antibody, the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg of the host body weight. Doses for nucleic acids encoding immunogens range from about 10 ng to 1 g, 100 ng to 100 mg, 1 Ag to 10 mg, or 30-300 Ag DNA per patient. Doses _ for infectious viral vectors vary from 10-109, or more, virions per dose.
Agents for inducing an immune response can be administered by parenteral, topical, intravenous, oral, subcutaneous, 5 intraperitoneal, intranasal or intramuscular means for prophylactic and/or therapeutic treatment. The most typical route of administration is subcutaneous although others can be equally effective. The next most common is intramuscular injection. This type of injection is most typically performed 10 in the arm or leg muscles. Intravenous injections as well as intraperitoneal injections, intraarterial, intracranial, or intradermal injections are also effective in generating an immune response. In some methods, agents are injected directly into a particular tissue where deposits have 15 accumulated.
Agents of the invention can optionally be administered in combination with other agents that are at least partly effective in treatment of amyloidogenic disease. In the case of Alzheimer's and Down's syndrome, in which amyloid deposits 20 occur in the brain, agents of the invention can also be administered in conjunction with other agents that increase passage of the agents of the invention across the blood-brain barrier.
Immunogenic agents of the invention, such as peptides, are
25 sometimes administered in combination with an adjuvant. A
variety of adjuvants can be used in combination with a peptide, such as AO, to elicit an immune response. Preferred adjuvants augment the intrinsic response to an immunogen without causing conformational changes in the immunogen that affect the qualitative form of the response. Preferred adjuvants include alum, 3 De-O-acylated monophosphoryl lipid A
(MPL) (see GB 2220211). QS21 is a triterpene glycoside or saponin isolated from the bark of the Quillaja Saponaria Molina tree found in South America (see Kensil et al., in Vaccine Design: The Subunit and Ajuvant Approach (eds. Powell & Newman, Plenum Press, NY, 1995); US Patent No. 5,057,540).
Other adjuvants are oil in water emulsions (such as squalene or peanut oil), optionally in combination with immune
26 stimulants, such as monophosphoryl lipid A (see Stoute et al., N. Engl. J. Med. 336, 86-91 (1997)). Another adjuvant is CpG
,(Bioworld Today, Nov. 15, 1998). Alternatively, AO can be coupled to an adjuvant. For example, a lipopeptide version of AO can be prepared by coupling palmitic acid or other lipids directly to the N-terminus of AO as described for hepatitis B
antigen vaccination (Livingston, J. Immunol. 159, 1383-1392 (1997)). However, such coupling should not substantially change the conformation of AO so as to affect the nature of the immune response-thereto. Adjuvants can be administered as a component of a therapeutic composition with an active agent or can be administered separately, before, concurrently with, or after administration of the therapeutic agent.
A preferred class of adjuvants is aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate. Such adjuvants can be used with or without other specific immunostimulating agents such as MPL or 3-DMP, QS21, polymeric or monomeric amino acids such as polyglutamic acid or polylysine. Another class of adjuvants is oil-in-water emulsion formulations. Such adjuvants can be used with or without other specific immunostimulating agents such as muramyl peptides (e.g., N-acetylmuramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1'-2'dipaimitoy1-sn-g1ycero-3-hy4roxyphosphoryloxy)-ethylamine (MTP-PE), N-acetylglucsaminyl-N-acetylmuramyl-L-Al-D-isoglu-L-Ala-dipalmitoxy propylamide (DTP-DPP) theramideTm), or other bacterial cell wall components. Oil-in-water emulsions include (a) MF59 (WO 90/14837), containing 5% Squalene, 0.5%
Twee n 80, and 0.5% Span 85 (optionally containing various amounts of MTP-PE) formulated into submicron particles using a microfluidizer such as Model 110Y microfluidizer (Microfluidics, Newton MA), (b) SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic-blocked polymer LI21, and thr-MDP, either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) Ribim adjuvant system (RAS), (Ribi Immunochem, Hamilton, MT) *Trademark
27 containing 296 squalene, 0.296 Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorylipid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton (CWS), preferably MPL + CWS (Detoxi).
Another class of preferred adjuvants is saponin adjuvants, such as Stimulon7m (QS21, Aquila, Worcester, MA) or particles generated therefrom such as ISCOMs (immunostimulating complexes) and ISCOMATRIX. Other adjuvants include Complete Freund's Adjuvant (CFA) and Incomplete Freund's Adjuvant (IFA). Other adjuvants include cytokines, such as interleukins (IL-].,IL-2, and IL-12), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF).
An adjuvant can be administered with an immunogen as a single composition, or can be administered before, concurrent with or after administration of the immunogen. Immunogen and adjuvant can be packaged and supplied in the same vial or can be packaged in separate vials and mixed before use. Immunogen and adjuvant are typically packaged with a label indicating the intended therapeutic application. If immunogen and adjuvant are packaged separately, the packaging typically includes instructions for mixing before use. The choice of an adjuvant and/or carrier depends on the stability of the vaccine containing the adjuvant, the route of administration, the dosing schedule, the efficacy of the adjuvant for the species being vaccinated, and, in humans, a pharmaceutically acceptable adjuvant is one that has been approved or is approvable for human administration by pertinent regulatory bodies. For example, Complete Freund's adjuvant is not suitable for human administration. Alum, MPL and QS21 are preferred. Optionally, two or more different adjuvants can be used simultaneously. Preferred combinations include alum with MPL, alum with QS21, MPL with QS21, and alum, QS21 and MPL
together. Also, Incomplete Freund's ajuvant can be used (Chang et al., Advanced Drug Delivery Reviews 32, 173-186 (1998)), optionally in combination with any of alum, QS21, and MPL and all combinations thereof.
Agents of the invention are often administered as pharmaceutical compositions comprising an active therapeutic
28 agent, i.e., and a variety of other pharmaceutically acceptable components. See Remington's Pharmaceutical Science (15th ed., Mack Publishing Company, Easton, Pennsylvania, 1980). The preferred form depends on the intended mode of administration and therapeutic application. The compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration. The diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution. In addition, the pharmaceutical composition or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, nonimmunogenic stabilizers and the like. However, some reagents suitable for administration to animals, such as Complete Freund's adjuvant are not typically included in compositions for human use.
Pharmaceutical compositions can also include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids and copolymers (such as latex functionalized sepharose, agarose, cellulose, and the like), polymeric amino acids, amino acid copolymers, and lipid aggregates (such as oil droplets or liposomes). Additionally, these carriers can function as immunostimulating agents (i.e., adjuvants).
For parenteral administration, agents of the invention can be administered as injectable dosages of a solution or suspension of the substance in a physiologically acceptable diluent with a pharmaceutical carrier which can be a sterile liquid such as water oils, saline, glycerol, or ethanol.
Additionally, auxiliary substances, such as wetting or emulsifying agents, surfactants, pH buffering substances and the like can be present in compositions. Other components of pharmaceutical compositions are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, and mineral oil. In general, glycols such as
29 propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions.
Typically, compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared. The preparation also can be emulsified or encapsulated in liposomes or micro particles such as polylactide, polyglycolide, or copolymer for enhanced adjuvant effect, as discussed above (see Langer, Science 249, 1527 (1990) and Hanes, Advanced Drug Delivery Reviews 28, 97-119 (1997). The agents of this invention can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner as to permit a sustained or pulsatile release of the active ingredient.
Additional formulations suitable for other modes of administration include oral, intranasal, and pulmonary formulations, suppositories, and transdermal applications.
For suppositories, binders and carriers include, for example, polyalkylene glycols or triglycerides; such suppositories can be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, preferably 1%-2%. Oral formulations include excipients, such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, and magnesium carbonate. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and contain 10%-9596 of active ingredient, preferably 25%-70%.
Topical application can result in transdermal or intradermal delivery. Topical administration can be facilitated by co-administration of the agent with cholera toxin or detoxified derivatives or subunits thereof or other similar bacterial toxins (See Glenn et al., Nature 391, 851 (1998)). Co-administration can be achieved by using the components as a mixture or as linked molecules obtained by chemical crosslinking or expression as a fusion protein.
Alternatively, transdermal delivery can be achieved using a skin path or using transferosomes (Paul et al., Eur. J.

Immunol. 25, 3521-24 (1995); Cevc et al., Biochem. Biophys.
Acta 1368, 201-15 (1998)).
V. Methods of Diagnosis The invention provides methods of detecting an immune 5 response against AO peptide in a patient suffering from or susceptible to Alzheimer's disease. The methods are particularly useful for monitoring a course of treatment being administered to a patient. The methods can be used to monitor both therapeutic treatment on symptomatic patients and 10 prophylactic treatment on asymptomatic patients.
Some methods entail determining a baseline value of an immune response in a patient before administering a dosage of agent, and comparing this with a value for the immune response after treatment. A significant increase (i.e., greater than 15 the typical margin of experimental error in repeat measurements of the same sample, expressed as one standard deviation from the mean of such measurments) in value of the immune response signals a positive treatment outcome (i.e., that administration of the agent has achieved or augmented an 20 immune response). If the value for immune response does not change significantly, or decreases, a negative treatment outcome is indicated. In general, patients undergoing an initial course of treatment with an agent are expected to show an increase in immune response with successive dosages, which 25 eventually reaches a plateau. Administration of agent is generally continued while the immune response is increasing.
Attainment of the plateau is an indicator that the administered of treatment can be discontinued or reduced in dosage or frequency.
30 In other methods, a control value (i.e., a mean and standard deviation) of immune response is determined for a control population. Typically the individuals in the control population have not received prior treatment. Measured values of immune response in a patient after administering a therapeutic agent are then compared with the control value. A
significant increase relative to the control value (e.g., greater than one standard deviation from the mean) signals a _
31 positive treatment outcome. A lack of significant increase or a decrease signals a negative treatment outcome.
Administration of agent is generally continued while the immune response is increasing relative to the control value.
As before, attainment of a plateau relative to control values in an indicator that the administration of treatment can be discontinued or reduced in dosage or frequency.
In other methods, a control value of immune response (e.g., a mean and standard deviation) is determined from a control population of individuals who have undergone treatment with a therapeutic agent and whose immune responses have plateaued in response to treatment. Measured values of immune response in a patient are compared with the control value. If the measured level in a patient is not significantly different (e.g., more than one standard deviation) from the control value, treatment can be discontinued. If the level in a patient is significantly below the control value, continued administration of agent is warranted. If the level in the patient persists below the control value, then a change in treatment regime, for example, use of a different adjuvant may be indicated.
In other methods, a patient who is not presently receiving treatment but has undergone a previous course of treatment is monitored for immune response to determine whether a resumption of treatment is required. The measured value of immune response in the patient can be compared with a value of immune response previously achieved in the patient after a previous course of treatment. A significant decrease relative to the previous measurement (i.e., greater than a typical margin of error in repeat measurements of the same sample) is an indication that treatment can be resumed. Alternatively, the value measured in patient can be compared with a control value (mean plus standard deviation) determined in population of patients after undergoing a course of treatment.
Alternatively, the measured value in a patient can be compared with a control value in populations of prophylactically treated patients who remain free of symptoms of disease, or populations of therapeutically treated patients who show _ _ _
32 amelioration of disease characteristics. In all of these cases, a significant decrease relative to the control level (i.e., more than a standard deviation) is an indicator that treatment should be resumed in a patient.
The tissue sample for analysis is typically blood, plasma, serum, mucus or cerebral spinal fluid from the patient. The sample is analyzed for indicia of an immune response to any form of AO peptide, typically A042. The immune response can be determined from the presence of, e.g., antibodies or T-cells that specifically bind to AO peptide. ELISA methods of detecting antibodies specific to AO are described in the Examples section. Methods of detecting reactive T-cells have been described above (see Definitions).
The invention further provides diagnostic kits for performing the diagnostic methods described above. Typically, such kits contain an agent that specifically binds to antibodies to AO or reacts with T-cells specific for AO. The kit can also include a label. For detection of antibodies to AO, the label is typically in the form of labelled anti-idiotypic antibodies. For detection of antibodies, the agent can be supplied prebound to a solid phase, such as to the wells of a microtiter dish. For detection of reactive T-cells, the label can be supplied as 3H-thymidine to measure a proliferative response. Kits also typically contain labelling providing directions for use of the kit. The labelling may also include a chart or other correspondence regime correlating levels of measured label with levels of antibodies to AO or T-cells reactive with AO. The term labelling refers to any written or recorded material that is attached to, or otherwise accompanies a kit at any time during its manufacture, transport, sale or use. For example, the term labelling encompasses advertising leaflets and brochures, packaging materials, instructions, audio or video cassettes, computer discs, as well as writing imprinted directly on kits.
EXAMPLES
I. Prophylactic Efficacy of AO Against AD
33 These examples describe administration of A042 peptide to transgenic mice overexpressing APP with a mutation at position 717 (APP717v-=F ) that predisposes them to develop Alzheimer's-like neuropathology. Production and characteristics of these mice (PDAPP mice) is described in Games et al., Nature, supra.
These animals, in their heterozygote form, begin to deposit A0 at six months of age forward. By fifteen months of age they exhibit levels of AO deposition equivalent to that seen in Alzheimer's disease. PDAPP mice were injected with aggregated A042 (aggregated AR ) or phosphate buffered saline.
Aggregated A042 was chosen because of its ability to induce antibodies to multiple epitopes of AO.
34 A. Methods 1. Source of Mice Thirty PDAPP heterogenic female mice were randomly divided into the following groups: 10 mice to be injected with aggregated A042 (one died in transit), 5 mice to be injected with PBS/adjuvant or PBS, and 10 uninjected controls. Five mice were injected with serum amyloid protein (SAP).
2. Preparation of Immunogens Preparation of aggregated A042: two milligrams of A1342 (US
Peptides Inc, lot K-42-12) was dissolved in 0.9 ml water and made up to 1 ml by adding 0.1 ml 10 x PBS. This was vortexed and allowed to incubate overnight 37 C, under which conditions the peptide aggregated. Any unused AB was stored as a dry lyophilized powder at -20 C until the next injection.
3. Preparation of Injections 100 Ag of aggregated AB
42 in PBS per mouse was emulsified 1:1 with Complete Freund's adjuvant (CFA) in a final volume of 400 Al emulsion for the first immunization, followed by a boost of the same amount of immunogen in Incomplete Freund's adjuvant (IFA) at 2 weeks. Two additional doses in IFA were given at monthly intervals. The subsequent immunizations were done at monthly intervals in 500 Al of PBS. Injections were delivered intraperitoneally (i.p.).
PBS injections followed the same schedule and mice were injected with a 1:1 mix of PBS/ Adjuvant at 400 Al per mouse, or 500 Al of PBS per mouse. SAP injections likewise followed the same schedule using a dose of 100 Ag per injection.
4. Titration of Mouse Bleeds, Tissue Preparation and Immunohistochemistry The above methods are described infra in General Materials and Methods.
B. Results PDAPP mice were injected with either aggregated AB

(aggregated AB42), SAP peptides, or phosphate buffered saline.

W099)27944 A group of PDAPP mice were also left as uninjected, positive controls. The titers of the mice to aggregated A042 were monitored every other month from the fourth boost until the mice were one year of age. Mice were sacrificed at 13 months.
5 At all time points examined, eight of the nine aggregated A42-mice developed a high antibody titer, which remained high throughout the series of injections (titers greater than 1/10000). The ninth mouse had a low, but measurable titer of approximately 1/1000 (Figure 1, Table 1). SAPP-injected mice 10 had titers of 1:1,000 to 1:30,000 for this immunogen with only a single mice exceeding 1:10,0000.
The PBS-treated mice were titered against aggregated A042 at six, ten and twelve months. At a 1/100 dilution the PBS
mice when titered against aggregated A042 only exceeded 4 15 times background at one data point, otherwise, they were less than 4 times background at all time points (Table 1). The SAP-specific response was negligible at these time points with all titers less than 300.
Seven out of the nine mice in the aggregated A31-42 group 20 had no detectable amyloid in their brains. In contrast, brain tissue from mice in the SAP and PBS groups contained numerous 3D6-positive amyloid deposits in the hippocampus, as well as in the frontal and cingulate cortices. The pattern of deposition was similar to that of untreated controls, with 25 characteristic involvement of vulnerable subregions, such as the outer molecular layer of the hippocampal dentate gyrus.
One mouse from the AO 1-42-injected group had a greatly reduced amyloid burden, confined to the hippocampus. An isolated plague was identified in another Afl 1-42-treated 30 mouse.
Quantitative image analyses of the amyloid burden in the hippocampus verified the dramatic reduction achieved in the AN1792-treated animals (Fig. 2). The median values of the amyloid burden for the PBS group (2.22W), and for the
35 untreated control group (2.65%) were significantly greater than for those immunized with AN1792 (0.00%, p=0.0005). In contrast, the median value for the group immunized with SAP
peptides (SAPP) was 5.74%. Brain tissue from the untreated,
36 control mice contained numerous AO amyloid deposits visualized with the AO-specific monoclonal antibody (mAb) 3D6 in the hippocampus, as well as in the retrosplenial cortex. A
similar pattern of amyloid deposition was also seen in mice immunized with SAPP or PBS (Fig. 2). In addition, in these latter three groups there was a characteristic involvement of vulnerable subregions of the brain classically seen in AD, such as the outer molecular layer of the hippocampal dentate gyrus, in all three of these groups.
The brains that contained no AO deposits were also devoid of neuritic plaques that are typically visualized in PDAPP
mice with the human APP antibody 8E5. All of brains from the remaining groups (SAP-injected, PBS and uninjected mice) had numerous neuritic plaques typical of untreated PDAPP mice. A
small number of neuritic plaques were present in one mouse treated with AN1792, and a single cluster of dystrophic neurites was found in a second mouse treated with AN1792.
Image analyses of the hippocampus, and shown in Fig. 3, demonstrated the virtual elimination of dystrophic neurites in AN1792-treated mice (median 0.00%) compared to the PBS
recipients (median 0.2896, p = 0.0005).
Astrocytosis characteristic of plaque-associated inflammation was also absent in the brains of the AO1-42 injected group. The brains from the mice in the other groups contained abundant and clustered GFAP-positive astrocytes typical of AO plaque-associated gliosis. A subset of the GFAP-reacted slides were counter-stained with Thioflavin S to localize the AO deposits. The GFAP-positive astrocytes were associated with AO plaques in the SAP, PBS and untreated controls. No such association was found in the plaque-negative AO1-42 treated mice, while minimal plaque-associated gliosis was identified in one mouse treated with AN1792.
Image analyses, shown in Fig. 4 for the retrosplenial cortex, verified that the reduction in astrocytosis was significant with a median value of 1.56% for those treated with AN1792 versus median values greater than 6% for groups immunized with SAP peptides, PBS or untreated (p=0.0017)
37 Evidence from a subset of the AO 1-42- and PBS-injected mice indicated plaque-associated MHC II immunoreactivity was absent in the A431-42 injected mice, consistent with lack of an AO-related inflammatory response.
Sections of the mouse brains were also reacted with a mAO
specific for MAC-1, a cell surface protein. MAC-1 (CD11b) is an integrin family member and exists as a heterodimer with CD18. The CD11b/CD18 complex is present on monocytes, macrophages, neutrophils and natural killer cells (Mak and Simard). The resident MAC-1-reactive cell type in the brain is likely to be microglia based on similar phenotypic morphology in MAC-1 immunoreacted sections. Plaque-associated MAC-1 labeling was lower in the brains of mice treated with AN1792 compared to the PBS control group, a finding consistent with the lack of an AO-induced inflammatory response.
C. Conclusion The lack of AO plaques and reactive neuronal and gliotic changes in the brains of the A01-42-injected mice indicate that no or extremely little amyloid was deposited in their brains, and pathological consequences, such as gnosis and neuritic pathology, were absent. PDAPP mice treated with AO1-42 show essentially the same lack of pathology as control nontransgenic mice. Therefore, A131-42 injections are highly effective in the prevention of deposition or clearance of human AO from brain tissue, and elimination of subsequent neuronal and inflammatory degenerative changes. Thus, administration of AO peptide has therapeutic benefit in prevention of AD.
II. Dose Response Study Groups of five-week old, female Swiss Webster mice (N=6 per group) were immunized with 300, 100, 33, 11, 3.7, 1.2, 0.4, or 0.13 ug of AO formulated in CFA/IFA administered intraperitoneally. Three doses were given at biweekly intervals followed by a fourth dose one month later. The first dose was emulsified with CFA and the remaining doses were emulsified with IFA. Animals were bled 4-7 days
38 following each immunization starting after the second dose for measurement of antibody titers. Animals in a subset of three groups, those immunized with 11, 33, or 300 Ag of antigen, were additionally bled at approximately monthly intervals for four months following the fourth immunization to monitor the decay of the antibody response across a range of vaccine doses. These animals received a final fifth immunization at seven months after study initiation. They were sacrificed one week later to measure antibody responses to AN1792 and to perform toxicological analyses.
A declining dose response was observed from 300 to 3.7 Ag with no response at the two lowest doses. Mean antibody titers are about 1:1000 after 3 doses and about 1:10,000 after 4 doses of 11-300 Ag of antigen (see Fig. 5).
Antibody titers rose dramatically for all but the lowest dose group following the third immunization with increases in GMTs ranging from 5- to 25-fold. Low antibody responses were then detectable for even the 0.4 Ag recipients. The 1.2 and 3.7 Ag groups had comparable titers with GMTs of about 1000 and the highest four doses clustered together with GMTs of about 25,000, with the exception of the 33 Ag dose group with a lower GMT of 3000. Following the fourth immunization, the titer increase was more modest for most groups. There was a clear dose response across the lower antigen dose groups from 0.14 Ag to 11 Ag ranging from no detectable antibody for recipients of 0.14 Ag to a GMT of 36,000 for recipients of 11 Ag. Again, titers for the four highest dose groups of 11 to 300 Ag clustered together. Thus following two immunizations, the antibody titer was dependent on the antigen dose across the broad range from 0.4 to 300 Ag. By the third immunization, titers of the highest four doses were all comparable and they remained at a plateau after an additional immunization.
One month following the fourth immunization, titers were 2-to 3-fold higher in the 300 Ag group than those measured from blood drawn five days following the immunization (Fig. 6).
This observation suggests that the peak anamnestic antibody response occurred later than 5 days post-immunization. A more
39 modest (50%) increase was seen at this time in the 33 pg group. In the 300 pg dose group at two months following the last dose, GMTs declined steeply by about 70%. After another month, the decline was less steep at 45% (100 pg) and about 14% for the 33 and 11 pg doses. Thus, the rate of decline in circulating antibody titers following cessation of immunization appears to be biphasic with a steep decline the first month following peak response followed by a more modest rate of decrease thereafter.
The antibody titers and the kinetics of the response of these Swiss Webster mice are similar to those of young heterozygous PDAPP transgenic mice immunized in a parallel manner. Dosages effective to induce an immune response in humans are typically similar to dosages effective in mice.
III. Screen For Theraveutic Efficacy Against Established AD
This assay is designed to test immunogenic agents for activity in arresting or reversing neuropathological characteristics of AD in aged animals. Immunizations with 42 amino acid long Ali (AN1792) were begun at a timepoint when amyloid plaques are already present in the brains of the PDAPP
mice.
Over the timecourse used in this study, untreated PDAPP
mice develop a number of neurodegenerative changes that resemble those found in AD (Games et al., supra and Johnson-Wood et al., Proc. Natl. Acad. Sci. USA 94, 1550-1555 (1997)).
The deposition of Afl into amyloid plaques is associated with a degenerative neuronal response consisting of aberrant axonal and dendritic elements, called dystrophic neurites. Amyloid deposits that are surrounded by and contain dystrophic neurites called neuritic plaques. In both AD and the PDAPP
mouse, dystrophic neurites have a distinctive globular structure, are immunoreactive with a panel of antibodies recognizing APP and cytoskeletal components, and display complex subcellular degenerative changes at the ultrastructural level. These characteristics allow for disease-relevant, selective and reproducible measurements of neuritic plaque formation in the PDAPP brains. The dystrophic WO 99t27944 neuronal component of PDAPP neuritic plaques is easily visualized with an antibody specific for human APP (mAfl 8E5), and is readily measurable by computer-assisted image analysis.
Therefore, in addition to measuring the effects of .AN1792 on 5 amyloid plaque formation, we monitored the effects of this treatment on the development of neuritic dystrophy.
Astrocytes and microglia are non-neuronal cells that respond to and reflect the degree of neuronal injury. GFAP-positive astrocytes and MHC II-positive microglia are commonly 10 observed in AD, and their activation increases with the severity of the disease. Therefore, we also monitored the development of reactive astrocytosis and microgliosis in the AN1792-treated mice.
A. Materials and Methods 15 Forty-eight, heterozygous female PDAPP mice, 11 to 11.5 months of age, obtained from Charles River, were randomly divided into two groups: 24 mice to be immunized with 100 Ag of AN1792 and 24 mice to be immunized with PBS, each combined with Freund's adjuvant. The AN1792 and PBS groups were again 20 divided when they reached -15 months of age. At 15 months of age approximately half of each group of the AN1792- and PBS-treated animals were euthanized (n=10 and 9, respectively), the remainder continued to receive immunizations until termination at -18 months (n=9 and 12, respectively). A total 25 of.8 animals (5 AN1792, 3 PBS) died during the study. In addition to the immunized animals, one-year old (n=10), 15-month old (n=10) and 18-month old (n=10) untreated PDAPP mice were included for comparison in the ELISAs to measure a0 and APP levels in the brain; the one-year old animals were also 30 included in the immunohistochemical analyses.
Methodology was as in Example 1 unless otherwise indicated.
US Peptides lot 12 and California Peptides lot ME0339 of AN1792 were used to prepare the antigen for the six immunizations administered prior to the 15-month timepoint.
35 California Peptides lots ME0339 and ME0439 were used for the three additional immunizations administered between 15 and 18 months.

For immunizations, 100 Ag of AN1792 in 200 Al PBS or PBS
alone was emulsified 1:1 (vol:vol) with Complete Freund's adjuvant (CFA) or Incomplete Freund's adjuvant (IFA) or PBS in a final volume of 400 Al. The first immunization was delivered with CFA as adjuvant, the next four doses were given with IFA and the final four doses with PBS alone without added adjuvant. A total of nine immunizations were given over the seven-month period on a two-week schedule for the first three doses followed by a four-week interval for the remaining injections. The four-month treatment group, euthanized at 15 months of age, received only the first 6 immunizations.
B. Results 1.. Effects of AN1792 Treatment on Amyloid Burden The results of AN1792 treatment on cortical amyloid burden determined by quantitative image analysis are shown in Fig. 7.
The median value of cortical amyloid burden was 0.28% in a group of untreated 12-month old PDAPP mice, a value representative of the plaque load in mice at the study's initiation. At 18 months, the amyloid burden increased over 17-fold to 4.87% in PBS-treated mice, while AN1792-treated mice had a greatly reduced amyloid burden of only 0.01%, notably less than the 12-month untreated and both the 15- and 18-month PBS-treated groups. The amyloid burden was significantly reduced in the AN1792 recipients at both 15 (96%
reduction; p=0.003) and 18 (>99% reduction; p=0.0002) months.
Typically, cortical amyloid deposition in PDAPP mice initiates in the frontal and retrosplenial cortices (RSC) and progresses in a ventral-lateral direction to involve the temporal and entorhinal cortices (EC). Little or no amyloid was found in the EC of 12 month-old mice, the approximate age at which AN1792 was first administered. After 4 months of AN1792 treatment, amyloid deposition was greatly diminished in the RSC, and the progressive involvement of the EC was entirely eliminated by AN1792 treatment. The latter observation showed that AN1792 completely halted the progression of amyloid that would normally invade the temporal and ventral cortices, as well as arrested or possibly reversed deposition in the RSC.
The profound effects of AN1792 treatment on developing cortical amyloid burden in the PDAPP mice are further demonstrated by the 18-month group, which had been treated for seven months. A near complete absence of cortical amyloid was found in the AN1792-treated mouse, with a total lack of diffuse plaques, as well as a reduction in compacted deposits.
2. AN1792 Treatment-associated Cellular and Morphological Changes A population of AO-positive cells was found in brain regions that typically contain amyloid deposits. Remarkably, in several brains from AN1792 recipients, very few or no extracellular cortical amyloid plaques were found. Most of the AO immunoreactivity appeared to be contained within cells with large lobular or clumped soma. Phenotypically, these cells resembled activated microglia or monocytes. They were immunoreactive with antibodies recognizing ligands expressed by activated monocytes and microglia (MHC II and CD11b) and were occasionally associated with the wall or lumen of blood vessels. Comparison of near-adjacent sections labeled with AO
and MHC II-specific antibodies revealed that similar patterns of these cells were recognized by both classes of antibodies.
Detailed examination of the AN1792-treated brains revealed that the MHC II-positive cells were restricted to the vicinity of the limited amyloid remaining in these animals. Under the fixation conditions employed, the cells were not immunoreactive with antibodies that recognize T cell (CD3, CD3e) or B cell (CD45RA, CD45RB) ligands or leukocyte common antigen (CD45), but were reactive with an antibody recognizing leukosialin (CD43) which cross-reacts with monocytes. No such cells were found in any of the PBS-treated mice.
PDAPP mice invariably develop heavy amyloid deposition in the outer molecular layer of the hippocampal dentate gyrus.
The deposition forms a distinct streak within the perforant pathway, a subregion that classically contains amyloid plaques in AD. The characteristic appearance of these deposits in PBS-treated mice resembled that previously characterized in untreated PDAPP mice. The amyloid deposition consisted of both diffuse and compacted plaques in a continuous band. In contrast, in a number of brains from AN1792-treated mice this pattern was drastically altered. The hippocampal amyloid deposition no longer contained diffuse amyloid, and the banded pattern was completely disrupted. Instead, a number of unusual punctate structures were present that are reactive with anti-AO antibodies, several of which appeared to be amyloid-containing cells.
MHC II-positive cells were frequently observed in the vicinity of extracellular amyloid in AN1792-treated animals.
The pattern of association of AO-positive cells with amyloid was very similar in several brains from AN1792-treated mice.
The distribution of these monocytic cells was restricted to the proximity of the deposited amyloid and was entirely absent from other brain regions devoid of AO plaques.
Quantitative image analysis of MEC II and MAC I-labeled sections revealed a trend towards increased immunoreactivity in the RSC and hippocampus of AN1792-treated mice compared to the PBS group which reached significance with the measure of MAC 1 reactivity in hippocampus.
These results are indicative of active, cell-mediated removal of amyloid in plaque-bearing brain regions.
3. AN1792 Effects on AO Levels: ELISA Determinations . (a) Cortical Levels In untreated PDAPP mice, the median level of total AO in the cortex at 12 months was 1,600 ng/g, which increased to 8,700 ng/g by 15 months (Table 2). At 18 months the value was 22,000 ng/g, an increase of over 10-fold during the time course of the experiment. PBS-treated animals had 8,600 ng/g total AO at 15 months which increased to 19,000 ng/g at 18 months. In contrast, AN1792-treated animals had 81% less total AO at 15 months (1,600 ng/g) than the PBS-immunized group. Significantly less (p=0.0001) total AO (5,200 ng/g) was found at 18 months when the AN1792 and PBS groups were compared (Table 2), representing a 72% reduction in the AO
that would otherwise be present. Similar results were obtained when cortical levels of A042 were compared, namely that the AN1792-treated group contained much less A042, but in this case the differences between the AN1792 and PBS groups were significant at both 15 months (p=0.04) and 18 months (p=0.0001, Table 2).
Table 2: Median AO Levels (ng/g) in Cortex Age TotalAti A42 Op Total A13 AM 40 Total AO42 00 12 1,600 1,300 OW
15 8,700 8,300 OW 8,600 7,200 (9) 1,600 1,300 OW
18 22,200 18,500 OW
19,000 15,900 (12) 5,200' 000- (9) *p= 0.1:412 ** p = 0.0001 (b) Hippocampal Levels In untreated PDAPP mice, median hippocampal levels of total AO at twelve months of age were 15,000 ng/g which increased to 51,000 ng/g at 15 months and further to 81,000 ng/g at 18 months (Table 3). Similarly, PBS immunized mice showed values of 40,000 ng/g and 65,000 ng/g at 15 months and 18 months, respectively. AN1792 immunized animals exhibited less total AO, specifically 25,000 ng/g and 51,000 ng/g at the respective 15-month and 18-month timepoints. The 18-month AN1792-treated group value was significantly lower than that of the PBS treated group (p= 0.0105; Table 3). Measurement of A042 gave the same pattern of results, namely that levels in the AN1792-treated group were significantly lower than in the PBS group (39,000 ng/g vs. 57,000 ng/g, respectively;
p=0.0022) at the 18-month evaluation (Table 3).

Table 3: Median Aft Levels (ng/g) in Rippocampus Age Total AO AO42 (n) Total AO A942 (n) Total AO42 (n) 12 15,5()0 11,100 (10) 5 15 51,500 44,400 (10) 40,100 35,700 (9) 24,500 22,100 (10) 18 80,800 64,200 (10) 65,400 57,100 (12) 50,900 38,900- (9) * p = 0.0105 ** p = 0.0022 (c) Cerebellar Levels In 12-month untreated PDAPP mice, the median cerebellar level of total AO was 15 ng/g (Table 4). At 15 months, this 10 median increased to 28 ng/g and by 18 months had risen to 35 ng/g. PBS-treated animals displayed median total AO values of 21 ng/g at 15 months and 43 ng/g at 18 months. AN1792-treated animals were found to have 22 ng/g total AO at 15 months and significantly less (p=0.002) total AO at 18 months (25 ng/g) 15 than the corresponding PBS group (Table 4).
Table 4: Median AO Levels (ng/g) in Cerebellum Age (months) Total All (n) Total AO (n) Total AO (n) 12 15.6 (10) 15 27.7 (10) 20.8 (9) 21.7 (10) 20 18 35.0 (10) 43.1 (12) 24.8 (9) = * p = 0.0018 4. Effects of AN1792 Treatment on APP Levels APP-a and the full-length APP molecule both contain all or part of the AO sequence and thus could be potentially impacted by the generation of an AN1792-directed immune response. In 25 studies to date, a slight increase in APP levels has been noted as neuropathology increases in the PDAPP mouse. In the cortex, levels of either APP-a/FL (full length) or APP-a were essentially unchanged by treatment with the exception that APP-a was reduced by 19% at the 18-month timepoint in the 30 AN1792-treated vs. the PBS-treated group. The 18-month AN1792-treated APP values were not significantly different from values of the 12-month and 15-month untreated and 15-month PBS groups. In all cases the APP values remained within the ranges that are normally found in PDAPP mice.
5. Effects of AN1792 Treatment on Neurodegenerative and Gliotic Pathology Neuritic plaque burden was significantly reduced in the frontal cortex of AN1792-treated mice compared to the PBS
group at both 15 (84%; p=0.03) and 18 (55%; p=0.01) months of age (Fig. 8). The median value of the neuritic plaque burden increased from 0.32% to 0.49% in the PBS group between 15 and 18 months of age. This contrasted with the greatly reduced development of neuritic plaques in the AN1792 group, with median neuritic plaque burden values of 0.05% and 0.22%, in the 15 and 18 month groups, respectively.
Immunizations with AN1792 seemed well tolerated and reactive astrocytosis was also significantly reduced in the RSC of AN1792-treated mice when compared to the PBS group at both 15 (56%; p=0.011).and 18 (39%; p=0.028) months of age (Fig. 9). Median values of the percent of astrocytosis in the PBS group increased between 15 and 18 months from 4.26% to 5.21%. AN1792-treatment suppressed the development of astrocytosis at both time points to 1.89% and 3.2%, respectively. This suggests the neuropil was not being damaged by the clearance process.
6. Antibody Responses As described above, eleven-month old, heterozygous PDAPP
mice (N=24) received a series of 5 immunizations of 100 Ag of AN1792 emulsified with Freund's adjuvant and administered intraperitoneally at weeks 0, 2, 4, 8, and 12, and a sixth immunization with PBS alone (no Freund's adjuvant) at week 16.
As a negative control, a parallel set of 24 age-matched transgenic mice received immunizations of PBS emulsified with the same adjuvants and delivered on the same schedule.
Animals were bled within three to seven days following each immunization starting after the second dose. Antibody responses to AN1792 were measured by ELISA. Geometric mean titers (GMT) for the animals that were immunized with AN1792 were approximately 1,900, 7,600, and 45,000 following the second, third and last (sixth) doses respectively. No An-specific antibody was measured in control animals following the sixth immunization.
Approximately one-half of the animals were treated for an additional three months, receiving immunizations at about 20, 24 and 27 weeks. Each of these doses was delivered in PBS
vehicle alone without Freund's adjuvant. Mean antibody titers remained unchanged over this time period. In fact, antibody titers appeared to remain stable from the fourth to the eighth bleed corresponding to a period covering the fifth to the ninth injections.
To determine if the An-specific antibodies elicited by immunization that were detected in the sera of AN1792-treated mice were also associated with deposited brain amyloid, a subset of sections from the AN1792- and PBS-treated mice were reacted with an antibody specific for mouse IgG. In contrast to the PBS group, AO plaques in AN1792-treated brains were coated with endogenous IgG. This difference between the two groups was seen in both 15-and 18-month groups. Particularly striking was the lack of labeling in the PBS group, despite the presence of a heavy amyloid burden in these mice. These results show that immunization with a synthetic An protein generates antibodies that recognize and bind in vivo to the An in amyloid plaques.
7. Cellular-Mediated Immune Responses Spleens were removed from nine AN1792-immunized and 12 PBS-immunized 18-month old PDAPP mice 7 days after the ninth immunization. Splenocytes were isolated and cultured for 72 h in the presence of A040, An42, or An40-1 (reverse order protein). The mitogen Con A served as a positive control.
Optimum responses were obtained with >1.7 M protein. Cells from all nine AN1792-treated animals proliferated in response to either A01-40 or A01-42 protein, with equal levels of incorporation for both proteins (Fig. 10, Upper Panel). There was no response to the A040-1 reverse protein. Cells from control animals did not respond to any of the AO proteins (Fig. 10, Lower Panel).
C. Conclusion The results of this study show that AN1792 immunization of PDAPP mice possessing existing amyloid deposits slows and prevents progressive amyloid deposition and retard consequential neuropathological changes in the aged PDAPP
mouse brain. Immunizations with AN1792 essentially halted amyloid developing in structures that would normally succumb to amyloidosis. Thus, administration of AO peptide has therapeutic benefit in the treatment of AD.
IV. Screen of AO Fragments 100 PDAPP mice age 9-11 months are immunized with 9 different regions of APP and Afi to determine which epitopes convey the response. The 9 different immunogens and one control are injected i.p. as described above. The immunogens include four human AO peptide conjugates 1-12, 13-28, 32-42, 1-5, all coupled to sheep anti-mouse IgG via a cystine link;
an APP polypeptide aa 592-695, aggregated human AO 1-40, and aggregated human AO 25-35, and aggregated rodent A1342.
Aggregated Afi42 and PBS are used as controls. Ten mice are used per treatment group. Titers are monitored as above and mice are euthanized at the end of 4 months of injections.
Histochemistry, Afi levels, and toxicology are determined post mortem.
A. Materials and Methods 1. Preparation of Immunogens Preparation of coupled AO peptides: four human AO peptide conjugates (amino acid residues 1-5, 1-12, 13-28, and 33-42, each conjugated to sheep anti-mouse IgG) were prepared by coupling through an artificial cysteine added to the AO
peptide using the crosslinking reagent sulfo-EMCS. The AO
peptide derivatives were synthesized with the following final amino acid sequences. In each case, the location of the inserted cysteine residue is indicated by underlining. The _ pciriuD20/2s3so A/313-28 peptide derivative also had two glycine residues added prior to the carboxyl terminal cysteine as indicated.
A01-12 peptide NH2-DAEFRHDSGYEVC COOH
AO1-5 peptide NH2-DAEFRC COON
A033-42 peptide NH2-C-amino-heotanoic acid-GLMVGGVVIA COON
A013-28 peptide Ac-NH-HHQKLVPFAEDVGSNKGGC-COOH
To orepare for the coupling reaction, ten mg of sheep anti-mouse IgG (Jackson ImmunoResearch Laboratories) was dialyzed overnight against 10 mM sodium borate buffer, pH 8.5.
The dialyzed antibody was then concentrated to a volume of 2 mL using an Amicon Centriprep tube. Ten mg sulfo-EMCS
[N (e-maleimidocuproyloxy) succinimide) (Molecular Sciences Co.) was dissolved in one mL deionized water. A 40-fold molar excess of sulfo-EMCS was added dropwise with stirring to the sheep anti-mouse IgG and then the solution was stirred for an additional ten min. The activated sheep anti-mouse IgG was purified and buffer exchanged by passage over a 10 mL gel filtration column (Pierce Presto* Column, obtained from Pierce Chemicals) equilibrated with 0.1 M NaPO4, 5 mM EDTA, pH 6.5.
Antibody containing fractions, identified by absorbance at 280 nm, were pooled and diluted to a concentration of approximately 1 mg/mL, using 1.4 mg per OD as the extinction coefficient. A 40-fold molar excess of AO peptide was dissolved in 20 mL of 10 mM NaPO4, pH 8.0, with the exception of the A033-42 peptide for which 10 mg was first dissolved in 0.5 mL of DMSO and then diluted to 20 mL with the 10 mM NaPO4 buffer. The peptide solutions were each added to 10 mL of activated sheep anti-mouse IgG and rocked at room temperature for 4 hr. The resulting conjugates were concentrated to a final volume of less than 10 mL using an Amicon Centriprep tube and then dialyzed against PBS to buffer exchange the buffer and remove free peptide. The conjugates were passed through 0.22 'u-pore size filters for sterilization and then aliquoted into fractions of 1 mg and stored frozen at -20 C.
The concentrations of the conjugates were determined using the BCA protein assay (Pierce Chemicals) with horse IgG for the *Trademark standard curve. Conjugation was documented by the molecular weight increase of the conjugated peptides relative to that of the activated sheep anti-mouse IgG. The AB 1-5 sheep anti-mouse conjugate was a pool of two conjugations, the rest were 5 from a single preparation.
2. Preparation of aggregated AB peptides Human 1-40 (AN1528; California Peptides Inc., Lot ME0541), human 1-42 (AN1792; California Peptides Inc., Lots ME0339 and ME0439), human 25-35, and rodent 1-42 (California Peptides 10 Inc., Lot ME0218) peptides were freshly solubilized for the preparation of each set of injections from lyophilized powders that had been stored desiccated at -20 C. For this purpose, two mg of peptide were added to 0.9 ml of deionized water and the mixture was vortexed to generate a relatively uniform 15 solution or suspension. Of the four, AN1528 was the only peptide soluble at this step. A 100 Al aliquot of 10X PBS (1X
PBS: 0.15 M NaC1, 0.01 M sodium phosphate, pH 7.5) was then added at which point AN1528 began to precipitate. The suspension was vortexed again and incubated overnight at 37 C
20 for use the next day.
Preparation of the pBx6 protein: An expression plasmid encoding pBx6, a fusion protein consisting of the 100-amino acid bacteriophage MS-2 polymerase N-terminal leader sequence followed by amino acids 592-695 of APP (BAPP) was constructed 25 as described by Oltersdorf et al., J. Biol. Chem. 265, 4492-4497 (1990). The plasmid was transfected into E. coli and the protein was expressed after induction of the promoter. The bacteria were lysed in 8M urea and pBx6 was partially purified by preparative SDS PAGE. Fractions containing pBx6 were 30 identified by Western blot using a rabbit anti-pBx6 polyclonal antibody, pooled, concentrated using an Amicon Centriprep tube and dialysed against PBS. The purity of the preparation, estimated by Coomassie Blue stained SDS PAGE, was approximately 5 to 10%.
*Trademark B. Results and Discussion 1. Study Design One hundred male and female, nine- to eleven-month old heterozygous PDAPP transgenic mice were obtained from Charles River Laboratory and Taconic Laboratory. The mice were sorted into ten groups to be immunized with different regions of AO
or APP combined with Freund's adjuvant. Animals were distributed to match the gender, age, parentage and source of the animals within the groups as closely as possible. The immunogens included four AO peptides derived from the human sequence, 1-5, 1-12, 13-28, and 33-42, each conjugated to sheep anti-mouse IgG; four aggregated AO peptides, human 1-40 (AN1528), human 1-42 (AN1792), human 25-35, and rodent 1-42;
and a fusion polypeptide, designated as pBx6, containing APP
amino acid residues 592-695. A tenth group was immunized with PBS combined with adjuvant as a control.
For each immunization, 100 Ag of each AO peptide in 200 Al PBS or 200 Ag of the APP derivative pBx6 in the same volume of PBS or PBS alone was emulsified 1:1 (vol:vol) with Complete Freund's adjuvant (CFA) in a final volume of 400 Al for the first immunization, followed by a boost of the same amount of immunogen in Incomplete Freund's adjuvant (IFA) for the subsequent four doses and with PBS for the final dose.
Immunizations were delivered intraperitoneally on a biweekly schedule for the first three doses, then on a monthly schedule thereafter. Animals were bled four to seven days following each immunization starting after the second dose for the measurement of antibody titers. Animals were euthanized approximately one week after the final dose.
2. AO and APP Levels in the Brain Following about four months of immunization with the various AO peptides or the APP derivative, brains were removed from saline-perfused animals. One hemisphere was prepared for immunohistochemical analysis and the second was used for the quantitation of AO and APP levels. To measure the concentrations of various forms of beta amyloid peptide and amyloid precursor protein, the hemisphere was dissected and _ homogenates of the hippocampal, cortical, and cerebellar regions were prepared in 5 M guanidine. These were diluted and the level of amyloid or APP was quantitated by comparison to a series of dilutions of standards of AO peptide or APP of known concentrations in an ELISA format.
The median concentration of total AO for the control group immunized with PBS was 5.8-fold higher in the hippocampus than in the cortex (median of 24,318 ng/g hippocampal tissue compared to 4,221 ng/g for the cortex). The median level in the cerebellum of the control group (23.4 ng/g tissue) was about 1,000-fold lower than in the hippocampus. These levels are similar to those that we have previously reported for heterozygous PDAPP transgenic mice of this age (Johnson-Woods et al., 1997, supra).
For the cortex, a subset of treatment groups had median total AO and A31-42 levels which differed significantly from those of the control grout) (p < 0.05), those animals receiving AN1792, rodent Al-42 or the Al-5 peptide conjugate.
The median levels of total N3 were reduced by 75%, 79% and 61%, respectively, compared to the control for these treatment groups. There were no discernable correlations between AO-specific antibody titers and AO levels in the cortical region of the brain for any of the groups.
In the hippocampus, the median reduction of total AO
associated with AN1792 treatment (46%, p = 0.0543) was not as great as that observed in the cortex (75%, p = 0.0021).
However, the magnitude of the reduction was far greater in the hippocampus than in the cortex, a net reduction of 11,186 ng/g tissue in the hippocampus versus 3,171 ng/g tissue in the cortex. For groups of animals receiving rodent A01-42 or Al-5, the median total AO levels were reduced by 36% and 26%, respectively. However, given the small group sizes and the high variability of the amyloid peptide levels from animal to animal within both groups, these reductions were not significant. When the levels of Al-42 were measured in the hippocampus, none of the treatment-induced reductions reached significance. Thus, due to the smaller AO burden in the cortex, changes in this region are a more sensitive indicator of treatment effects. The changes in AO levels measured by ELISA in the cortex are similar, but not identical, to the results from the immunohistochemical analysis (see below).
Total AS was also measured in the cerebellum, a region typically unaffected in the AD pathology. None of the median AO concentrations of any of the groups immunized with the various AS peptides or the APP derivative differed from that of the control group in this region of the brain. This result suggests that non-pathological levels of AS are unaffected by treatment.
APP concentration was also determined by ELISA in the cortex and cerebellum from treated and control mice. Two different APP assays were utilized. The first, designated APP-a/FL, recognizes both APP-alpha (a, the secreted form of APP which has been cleaved within the AS sequence), and full-length forms (FL) of APP, while the second recognizes only APP-a. In contrast to the treatment-associated diminution of AS in a subset of treatment groups, the levels of APP were unchanged in all of the treated compared to the control animals. These results indicate that the immunizations with AS peptides are not depleting APP; rather the treatment effect is specific to A.
In summary, total AS and A$1-42 levels were significantly reduced in the cortex by treatment with AN1792, rodent AS1-42 or A$1-5 conjugate. In the hippocarmous, total AS was significantly reduced only by AN1792 treatment. No other treatment-associated changes in AS or APP levels in the hippocampal, cortical or cerebellar regions were significant.
2. Histochemical Analyses Brains from a subset of six groups were prepared for immunohistochemical analysis, three groups immunized with the AO peptide conjugates A01-5, A01-12, and Ali13-28; two groups immunized with the full length AS aggregates AN1792 and AN1528 and the PBS-treated control group. The results of image analyses of the amyloid burden in brain sections from these groups are shown in Fig. 11. There were significant reductions of amyloid burden in the cortical regions of three VA:099/77944 of the treatment groups versus control animals. The greatest reduction of amyloid burden was observed in the group receiving AN1792 where the mean value was reduced by 97% (p 0.001). Significant reductions were also observed for those animals treated with AN1528 (9596, p = 0.005) and the A131-5 peptide conjugate (67%, p = 0.02).
The results obtained by quantitation of total AO or AO1-42 by ELISA and amyloid burden by image analysis differ to some extent. Treatment with AN1528 had a significant impact on the level of cortical amyloid burden when measured by quantitative image analysis but not on the concentration of total AO in the same region when measured by ELISA. The difference between these two results is likely to be due to the specificities of the assays. Image analysis measures only insoluble AO
aggregated into plaques. In contrast, the ELISA measures all forms of AO, both soluble and insoluble, monomeric and aggregated. Since the disease pathology is thought to be associated with the insoluble plaque-associated form of AO, the image analysis technique may have more sensitivity to reveal treatment effects. However since the ELISA is a more rapid and easier assay, it is very useful for screening purposes. Moreover it may reveal that the treatment-associated reduction of AO is greater for plaque-associated than total AO
To determine if the AO-specific antibodies elicited by immunization in the treated animals reacted with deposited brain amyloid, a subset of the sections from the treated animals and the control mice were reacted with an antibody specific for mouse IgG. In contrast to the PBS group, AO-containing plaques were coated with endogenous IgG for animals immunized with the AO peptide conjugates A31-5, A31-12, and A313-28; and the full length AO aggregates AN1792 and AN1528.
Brains from animals immunized with the other AO peptides or the APP peptide pBx6 were not analyzed by this assay.
3. Measurement of Antibody Titers Mice were bled four to seven days following each immunization starting after the second immunization, for a =
total of five bleeds. Antibody titers were measured as A01-42-binding antibody using a sandwich ELISA with plastic multi-well plates coated with A31-42. As shown in Fig. 12, peak antibody titers were elicited following the fourth dose for 5 those four vaccines which elicited the highest titers of AN1792-specific antibodies: AN1792 (peak GMT: 94,647), AN1528 (peak GMT: 88,231), Al-l2 conjugate (peak GMT: 47,216)and rodent AO1-42 (peak GMT: 10,766). Titers for these groups declined somewhat following the fifth and sixth doses. For 10 the remaining five immunogens, peak titers were reached following the fifth or the sixth dose and these were of much lower magnitude than those of the four highest titer groups:
AO1-5 conjugate (peak GMT: 2,356), pBx6 (peak GMT: 1,986), AO13-28 conjugate (peak GMT: 1,183), AO33-42 conjugate (peak 15 GMT: 658), A025-35 (peak GMT: 125). Antibody titers were also measured against the homologous peptides using the same ELISA
sandwich format for a subset of the immunogens, those groups immunized with Al-5, AO13-28, A25-35, AO33-42 or rodent Ag1-42. These titers were about the same as those measured 20 against 101-42 except for the rodent 1141-42 immunogen in which case antibody titers against the homologous immunogen were about two-fold higher. The magnitude of the AN1792-specific antibody titer of individual animals or the mean values of treatment groups did not correlate with efficacy measured as 25 the reduction of AO in the cortex.
4. Lvmphoproliferative Responses AO-dependent lymphoproliferation was measured using spleen cells harvested approximately one week following the final, sixth, immunization. Freshly harvested cells, 10 per well, 30 were cultured for 5 days in the presence of A31-40 at a concentration of 5 11M for stimulation. Cells from a subset of seven of the ten groups were also cultured in the presence of the reverse peptide, 1040-1. As a positive control, additional cells were cultured with the T cell mitogen, PHA, 35 and, as a negative control, cells were cultured without added peptide.

= 56 Lymphocytes from a majority of the animals proliferated in response to PHA. There were no significant responses to the A040-1 reverse peptide. Cells from animals immunized with the larger aggregated AO peptides, AN1792, rodent AO1-42 and AN1528 proliferated robustly when stimulated with A11-40 with the highest cpm in the recipients of AN1792. One animal in each of the groups immunized with A131-12 conjugate, AO13-28 conjugate and 1025-35 proliferated in response to AO1-40. The remaining groups receiving AO1-5 conjugate, AO33-42 conjugate pBx6 or PBS had no animals with an AO-stimulated response.
These results are summarized in Table 5 below.
Table 5 Immunogen Conjugate AO Amino Acids Responders AO1-5 yes 5-mer 0/7 A31-12 yes 12-mer 1/8 A1313-28 yes 16-mer 1/9 AO25-35 11-mer 1/9 A033-42 yes 10-mer 0/10 A31-40 40-mer 5/8 AO1-42 42-mer 9/9 r A01-42 42-mer 8/8 pBx6 0/8 PBS 0-mer 0/8 These results show that AN1792 and AN1528 stimulate strong T cell responses, most likely of the CD4+ phenotype. The absence of an AO-specific T cell response in animals immunized with A31-5 is not surprising since peptide epitopes recognized by CD4+ T cells are usually about 15 amino acids in length, although shorter peptides can sometimes function with less efficiency. Thus the majority of helper T cell epitopes for the four conjugate peptides are likely to reside in the IgG
conjugate partner, not in the AO region. This hypothesis is supported by the very low incidence of proliferative responses for animals in each of these treatment groups. Since the A431-5 conjugate was effective at significantly reducing the level _ of AO in the brain, in the apparent absence of AO-specific T
cells, =the key effector immune response induced by immunization with this peptide appears to be antibody.
Lack of T-cell and low antibody response from fusion peptide pBx6, encompassing APP amino acids 592-695 including all of the AO residues may be due to the poor immunogenicity of this particular preparation. The poor immunogenicity of the A025-35 aggregate is likely due to the peptide being too small to be likely to contain a good T cell epitope to help the induction of an antibody response. If this peptide were conjugated to a carrier protein, it would probably be more immunogenic.
V. Preparation of Polyclonal Antibodies for Passive Protection 20 non-transgenic mice are immunized with AO or other immunogen, optionally plus adjuvant, and are euthanized at 4-5 months. Blood is collected from immunized mice. Optionally, IgG is separated from other blood components. Antibody specific for the immunogen may be partially purified by affinity chromatography. An average of about 0.5-1 mg of immunogen-specific antibody is obtained per mouse, giving a total of 5-10 mg.
VI. Passive Immunization with Antibodies to AB
Groups of 7-9 month old PDAPP mice each are injected with 0.5 mg in PBS of polyclonal anti-A0 or specific anti-AO
monoclonals as shown below. All antibody preparations are purified to have low endotoxin levels. Monoclonals can be prepared against a fragment by injecting the fragment or longer form of AO into a mouse, preparing hybridomas and screening the hybridomas for an antibody that specifically binds to a desired fragment of AO without binding to other nonoverlapping fragments of AO.
Table 6 NNW.' _________________________________________________________________ Antibody Epitope Mouse polyclonal Anti-Aggregated AO42 anti-human A042 _ Mice are injected ip as needed over a 4 month period to maintain a circulating antibody concentration measured by ELISA titer of greater than 1/1000 defined by ELISA to AO42 or other immunogen. Titers are monitored as above and mice are euthanized at the end of 4 months of injections.
Histochemistry, AO levels and toxicology are performed post mortem. Ten mice are used per group.
VII. Comparison of Different Adjuvants This examples compares CFA, alum, an oil-in water emulsion and MPL for capacity to stimulate an immune response.
_ PCr/US98/25386 A. Materials and Methods 1. Study Design One hundred female Hartley strain six-week old guinea pigs, obtained from Elm Hill, were sorted into ten groups to be immunized with AN1792 or a palmitoylated derivative thereof combined with various adjuvants. Seven groups received injections of AN1792 (33 g unless otherwise specified) combined with a) PBS, b) Freund's adjuvant, c) MPL, d) squalene, e) MPL/squalene f) low dose alum, or g) high dose alum (300 g AN1792). Two groups received injections of a palmitoylated derivative of AN1792 (33 g) combined with a) PBS or b) squalene. A final, tenth group received PBS alone without antigen or additional adjuvant. For the group receiving Freund's adjuvant, the first dose was emulsified with CFA and the remaining four doses with IFA. Antigen was administered at a dose of 33 Ag for all groups except the high dose alum group, which received 300 Ag of .AN1792. Injections were administered intraperitoneally for CFA/IFA and intramuscularly in the hind limb quadriceps alternately on the right and left side for all other groups. The first three doses were given on a biweekly schedule followed by two doses at a monthly interval). Blood was drawn six to seven days following each immunization, starting after the second dose, for measurement of antibody titers.
2. Preparation of Immunocrens Two mg A042 (California Peptide, Lot ME0339) was added to 0.9 ml of deionized water and the mixture was vortexed to generate a relatively uniform suspension. A 100 1 aliquot of 10X PBS (1X PBS, 0.15 M NaC1, 0.01 M sodium phosphate, pH 7.5) was added. The suspension was vortexed again and incubated overnight at 37 C for use the next day. Unused A01-42 was stored with desiccant as a lyophilized powder at -20 C.

W099)27944 A palmitoylated derivative of AN1792 was prepared by coupling palmitic anhydride, dissolved in dimethyl formamide, to the amino terminal residue of AN1792 prior to removal of the nascent peptide from the resin by treatment with 5 hydrofluoric acid.
To prepare vaccine doses with Complete Freund's adjuvant (CFA) (group 2), 33 Ag of AN1792 in 200 Al PBS was emulsified 1:1 (vol:vol) with CFA in a final volume of 400 Al for the first immunization. For subsequent immunizations, the antigen 10 was similarly emulsified with Incomplete Freund's adjuvant (IFA).
To prepare vaccine doses with MPL for groups 5 and 8, lyophilized powder (Ribi ImmunoChem Research, Inc., Hamilton, MT) was added to 0.2% aqueous triethylamine to a final 15 concentration of 1 mg/ml and vortexed. The mixture was heated to 65 to 70 C for 30 sec to create a slightly opaque uniform suspension of micelles. The solution was freshly prepared for each set of injections. For each injection in group 5, 33 yg of AN1792 in 16.5 Al PBS, 50 Ag of MPL (50 Al) and 162 Al of 20 PBS were mixed in a borosilicate tube immediately before use.
To prepare vaccine doses with the low oil-in-water emulsion, AN1792 in PBS was added to 5% squalene, 0.5% Tween 80, 0.5% Span 85 in PBS to reach a final single dose concentration of 33 Ag AN1792 in 250 Al (group 6). The mixture 25 was emulsified by passing through a two-chambered hand-held device 15 to 20 times until the emulsion droplets appeared to be about equal in diameter to a 1.0 Am diameter standard latex bead when viewed under a microscope. The resulting suspension was opalescent, milky white. The emulsions were freshly 30 prepared for each series of injections. For group 8, MPL in 0.2% triethylamine was added at a concentration of 50 Ag per dose to the squalene and detergent mixture for emulsification as noted above. For the palmitoyl derivative (group 7), 33 Ag per dose of palmitoyl-NH-A01-42 was added to squalene and 35 vortexed. Tween 80 and Span 85 were then added with _ vortexing. This mixture was added to PBS to reach final concentrations of 5% squalene, 0.5% Tween 80, 0.5% Span 85 and the mixture was emulsified as noted above.
To prepare vaccine doses with alum (groups 9 and 10), AN1792 in PBS was added to Alhydrogel (aluminum hydroxide gel, Accurate, Westbury, NY) to reach concentrations of 33 Ag (low dose, group 9) or 300 Ag (high dose, group 10) AN1792 per 5 mg of alum in a final dose volume of 250 Al. The suspension was gently mixed for 4 hr at RT.
3. Measurement of Antibody Titers Guinea pigs were bled six to seven days following immunization starting after the second immunization for a total of four bleeds. Antibody titers against A042 were measured by ELISA as described in General Materials and Methods.
4. Tissue Preparation . After about 14 weeks, all guinea pigs were administered CO2. Cerebrospinal fluid was collected and the brains were removed and three brain regions (hippocampus, cortex and cerebellum) were dissected and used to measure the concentration of total A43 protein using ELISA.
*Trademark E. Results 1. Antibody Responses There was a wide range in the potency of the various adjuvants when measured as the antibody response to AN1792 following immunization. As shown in Fig. 13, when AN1792 was administered in PBS, no antibody was detected following two or three immunizations and negligible responses were detected following the fourth and fifth doses with geometric mean titers (GMTs) of only about 45. The o/w emulsion induced modest titers following the third dose (GMT 255) that were maintained following the fourth dose (GMT 301) and fell with the final dose (GMT 54). There was a clear antigen dose response for AN1792 bound to alum with 300 pg being more immunogenic at all time points than 33 gig. At the peak of the antibody response, following the fourth immunization, the difference between the two doses was 43% with GMTs of about 1940 (33 g) and 3400 (300 pig) . The antibody response to 33 Ag AN1792 plus MPL was very similar to that generated with almost a ten-fold higher dose of antigen (300 jig) bound to alum. The addition of MPL to an o/w emulsion decreased the potency of the vaccine relative to that with MPL as the sole adjuvant by as much as 75%. A palmitoylated derivative of AN1792 was completely non-immunogenic when administered in PBS
and gave modest titers when presented in an o/w emulsion with GMTs of 340 and 105 for the third and fourth bleeds. The highest antibody titers were generated with Freund's adjuvant with a peak GMT of about 87,000, a value almost 30-fold greater than the GMTs of the next two most potent vaccines, MPL and high dose AN1792/alum.
The most promising adjuvants identified in this study are MPL and alum. Of these two, MPL appears preferable because a 10-fold lower antigen dose was required to generate the same antibody response as obtained with alum. The response can be increased by increasing the dose of antigen and /or adjuvant and by optimizing the immunization schedule. The o/w emulcrion was a very weak adjuvant for AN1792 and adding an o/w emulsion to MPL adjuvant diminished the intrinsic adjuvant activity of MPL alone.
2. AO Levels In The Brain At about 14 weeks the guinea pigs were deeply anesthetized, the cerebrospinal fluid (CSF) was drawn and brains were excised from animals in a subset of the groups, those immunized with Freund's adjuvant (group 2), MPL (group 5), alum with a high dose, 300 jig, of AN1792 (group 10) and the PBS immunized control group (group 3). To measure the level of AO peptide, one hemisphere was dissected and homogenates of the hippocampal, cortical, and cerebellar regions were prepared in 5 M guanidine. These were diluted and quantitated by comparison to a series of dilutions of AO
standard protein of known concentrations in an ELISA format.
The levels of AO protein in the hippocampus, the cortex and the cerebellum were very similar for all four groups despite the wide range of antibody responses to AO elicited by these vaccines. Mean AO levels of about 25 ng/g tissue were measured in the hippocampus, 21 ng/g in the cortex, and 12 ng/g in the cerebellum. Thus, the presence of a high circulating antibody titer to AO for almost three months in some of these animals did not alter the total AO levels in their brains. The levels of AO in the CSF were also quite similar between the groups. The lack of large effect of AN1792 immunization on endogenous AO indicates that the immune response is focused on pathological formations of AO
VIII. Immune Response to Different Adjuvants in Mice Six-week old female Swiss Webster mice were used for this study with 10-13 animals per group. Immunizations were given 14,099/27!M4 on days 0, 14, 28, 60, 90 and 20 administered subcutaneously in a dose volume of 200 Al. PBS was used as the buffer for all formulations. Animals were bleed seven days following each immunization starting after the second dose for analysis of antibody titers by ELISA. The treatment regime of each group is summarized in Table 7.
=

Table 7 Experimental Design of Betabloc Study 010 ¨Group N a Adjuvant b Dose Antigen Dose (14g) 1 10 MPL , 12.5 g AN1792 33 5 2 10 MPL 25 g AN1792 33 3 10 MPL 50 g AN1792 33 4 13 MPL 125 ilg , AN1792 33 5 13 MPL 50 g AN1792 , 6 13 MPL 50g AN1528 33 10 7 10 PBS AN1792 , 33 8 10 PBS none 9 10 Squalene 5% AN 1792 33 emulsified , 10 10 Squalene 5% - AN1792 33 admixed 11 10 Alum 2 mg AN1792 33 _ 15 12 13 MPL + Alum 50 g/2 mg AN1792 33 13 10 QS21 5 ,g AN1792 33 14 10 QS21 10 g AN1792 33 15 10 QS21 25 g AN1792 33 16 13 QS21 25 ,g AN1792 _ 20 17 13 QS21 25 g AN1528 33 18 13 QS21 + MPL 25 g/50 g , AN1792 33 19 13 QS21 + Alum 25 g/2 mg AN1792 33 Footnotes:
a Number of mice in each group at the initiation of the experiment.
25 h The adjuvants are noted. The buffer for all these formulations was PBS. For group 8, there was no adjuvant and no antigen.
The ELISA titers of antibodies against A/342 in each group are shown in Table 8 below.

Table 8.
Geometric Mean Antibody Titers Week of Bleed Treatment Group 2.9 5.0 8.7 12.9 16.7 7 37 395 484 972 , 2149 14 , 104 541 3247 1609 19 , 63 243 554 299 441 The table shows that the highest titers were obtained for groups 4, 5 and 18, in which the adjuvants were 125 itg MPL, 50 Ag MPL and QS21 plus MPL.
IX. Therapeutic Efficacy of Different Adluvants A therapeutic efficacy study was conducted in PDAPP
transgenic mice with a set of adjuvants suitable for use in humans to determine their ability to potentiate immune responses to AO and to induce the immune-mediated clearance of amyloid deposits in the brain.
_ =

One hundred eighty male and female, 7.5- to 8.5-month old heterozygous PDAPP transgenic mice were obtained from Charles River Laboratories. The mice were sorted into nine groups containing 15 to 23 animals per group to be immunized with AN1792 or AN1528 combined with various adjuvants. Animals were distributed to match the gender, age, and parentage of the animals within the groups as closely as possible. The adjuvants included alum, MPL, and QS21, each combined with both antigens, and Freund's adjuvant (FA) combined with only AN1792. An additional group was immunized with AN1792 formulated in PBS buffer plus the preservative thimerosal without adjuvant. A ninth group was immunized with PBS alone as a negative control.
Preparation of aggregated AO peptides: human 01-40 (AN1528; California Peptides Inc., Napa, CA; Lot ME0541) and human 01-42 (AN1792; California Peptides Inc., Lot ME0439) peptides were freshly solubilized for the preparation of each set of injections from lyophilized powders that had been stored desiccated at -20 C. For this purpose, two mg of peptide were added to 0.9 ml of deionized water and the mixture was vortexed to generate a relatively uniform solution or suspension. AN1528 was soluble at this step, in contrast to AN1792. A 100 Al aliquot of 10X PBS (1X PBS: 0.15 M NaC1, 0.01 M sodium phosphate, pH 7.5) was then added at which point AN1528 began to precipitate. The suspensions were vortexed again and incubated overnight at 37 C for use the next day.
To prepare vaccine doses with alum (Groups 1 and 5), AO
peptide in PBS was added to Alhydrogel (two percent aqueous aluminum hydroxide gel, Sargeant, Inc., Clifton, NJ) to reach concentrations of 100 Ag AO peptide per 1 mg of alum. 10X PBS
was added to a final dose volume of 200 Al in 1X PBS. The suspension was then gently mixed for approximately 4 hr at RT
prior to injection.
To prepare vaccine doses for with MPL (Groups 2 and 6), lyophilized powder (Ribi ImmunoChem Research, Inc., Hamilton, MT; Lot 67039-E0896B) was added to 0.2k aqueous triethylamine to a final concentration of 1 mg/ml and vortexed. The mixture was heated to 65 to 70 C for 30 sec to create a slightly opaque uniform suspension of micelles. The solution was stored at 4 C. For each set of injections, 100 Ag of peptide per dose in 50 yl PBS, 50 yg of MPL per dose (50 Al) and 100 yl of PBS per dose were mixed in a borosilicate tube immediately before use.
To prepare vaccine doses with QS21 (Groups 3 and 7), lyophilized powder (Aquila, Framingham, MA; Lot A7018R) was added to PBS, pH 6.6-6.7 to a final concentration of 1 mg/ml and vortexed. The solution was stored at -20 C. For each set of injections, 100 Mg of peptide per dose in 50 Ml PBS, 25 Mg of QS21 per dose in 25 Ml PBS and 125 yl of PBS per dose were mixed in a borosilicate tube immediately before use.
To prepare vaccine doses with Freund's Adjuvant (Group 4), 100 Mg of AN1792 in 200 Ml PBS was emulsified 1:1 (vol:vol) with Complete Freund's Adjuvant (CFA) in a final volume of 400 yl for the first immunization. For subsequent immunizations, the antigen was similarly emulsified with Incomplete Freund's Adjuvant (IFA). For the vaccines containing the adjuvants alum, MPL or QS21, 100 yg per dose of AN1792 or AN1528 was combined with alum (1 mg per dose) or MPL (50 Mg per dose) or QS21 (25 yg per dose) in a final volume of 200 yl PBS and delivered by subcutaneous inoculation on the back between the shoulder blades. For the group receiving FA, 100 Mg of AN1792 was emulsified 1:1 (vol:vol) with Complete Freund's adjuvant (CFA) in a final volume of 400 Ml and delivered intraperitoneally for the first immunization, followed by a boost of the same amount of immunogen in Incomplete Freund's adjuvant (IFA) for the subsequent five doses. For the group receiving AN1792 without adjuvant, 10 Mg AN1792 was combined with 5 Mg thimerosal in a final volume of 50 Ml PBS and delivered subcutaneously. The ninth, control group received only 200 yl PBS delivered subcutaneously. Immunizations were given on a biweekly schedule for the first three doses, then on a monthly schedule thereafter on days 0, 16, 28, 56, 85 and 112. Animals were bled six to seven days following each immunization starting after the second dose for the measurement of antibody titers. Animals were euthanized approximately one week after the final dose. Outcomes were measured by ELISA assay of AO and APP levels in brain and by immunohistochemical evaluation of the presence of amyloid plaques in brain sections. In addition, AO-specific antibody titers, and AO-dependent proliferative and cytokine responses were determined.
Table 9 shows that the highest antibody titers to AO1-42 were elicited with FA and AN1792, titers which peaked following the fourth immunization (peak GMT: 75,386) and then declined by after the final, sixth immunization. The peak mean titer elicited by MPL with AN1792 was 62% lower than that generated with FA (peak GMT: 28,867) and was also reached early in the immunization scheme, after 3 doses, followed by a decline to 2896- of the peak value after the sixth immunization.
The peak mean titer generated with QS21 combined with AN1792 (GMT: 1,511) was about 5-fold lower than obtained with MPL.
In addition, the kinetics of the response were slower, since an additional immunization was required to reach the peak response. Titers generated by alum-bound AN1792 were marginally greater than those obtained with QS21 and the .
response kinetics were more rapid. For AN1792 delivered in PBS with thimerosal the frequency and size of titers were barely greater than that for PBS alone. The peak titers generated with MPL and AN1528 (peak GMT 3099) were about 9-fold lower than those with AN1792. Alum-bound AN1528 was very poorly immunogenic with low titers generated in only some of the animals. No antibody responses were observed in the control animals immunized with PBS alone.
*Trademark , _ , Table 9 Geometric Mean Antibody Titers' Week of Bleed Treatment 3.3 5.0 9.0 13.0 17.0 5 Alum/ 102 1,081 2,366 1,083 572 AN1792 (12/21)b (17/20) (21/21) (19/21) (18/21) MPL/ 6241 - 28,867 - 1,1242 5,665 8,204 AN1792 (21/21) (21/21) (21/21) (20/20) (20/20) QS21/ _ 30 227 327 ' 1,511 1,188 10 AN1792 (1/20) (10/19) (10/19) (17/18) (14/18) CFA/ 10,076 61,279 75,386 ' 41,628 30,574 AN1792 (15/15) (15/15) (15/15) (15/15) (15/15) Alum/ 25 33 39 37 31 AN1528 (0/21) (1/21) (3/20) (1/20) (2/20) 15 MPL/ s 184 2,591 1,633 1,156 3,099 AN1528 (15/21) (20/21) (21/21) (20/20) (20/20) QS21/ 29 221 51 ' 820 2,994 AN1528 (1/22) (13/22) (4/22) (20/22) (21/22) PBS plus 25 . 33 ' 39 ' 37 47 20 Thimerosal (0/16) (2/16) (4/16) (3/16) (4/16) _ PBS ' 25 25 25 25 25 (0/16) (0/16) (0/15) (0/12) (0/16) Footnotes:
' Geometric mean antibody titers measured against A01-42 ' Number of responders per group 25 The results of AN1792 or AN1592 treatment with various adjuvants, or thimerosal on cortical amyloid burden in 12-month old mice determined by ELISA are shown in Fig. 14. In PBS control PDAPP mice, the median level of total AO in the cortex at 12 months was 1,817 ng/g. Notably reduced levels of Afl were observed in mice treated with AN1792 plus CFA/IFA, AN1792 plus alum, AN1792 plus MPL and QS21 plus AN1792. The reduction reached statistical significance (p<0.05) only for AN1792 plus CFA/IFA. However, as shown in Examples I and III, the effects of immunization in reducing AO levels become substantially greater in 15 month and 18 month old mice.
Thus, it is expected that at least the AN1792 plus alum, AN1792 plus MPL and AN1792 plus QS21 compositions will achieve statistical significance in treatment of older mice. By contrast, the AN1792 plus the preservative thimerosal showed a median level of AO about the same as that in the PBS treated mice. Similar results were obtained when cortical levels of A042 were compared. The median level of A042 in PBS controls was 1624 ng/g. Notably reduced median levels of 403, 1149, 620 and 714 were observed in the mice treated with AN1792 plus CFA/IFA, AN1792 plus alum, AN1792 plus MPL and AN1792 plus QS21 respectively, with the reduction achieving statistical significance (p=0.05) for the AN1792 CFA/IFA treatment group.
The median level in the AN1792 thimerosal treated mice was 1619 ng/g 042.
X. Toxicity Analysis Tissues were collected for histopathologic examination at the termination of studies described in Examples 2, 3 and 7.
In addition, hematology and clinical chemistry were performed on terminal blood samples from Examples 3 and 7. Most of the major organs were evaluated, including brain, pulmonary, lymphoid, gastrointestinal, liver, kidney, adrenal and gonads.
Although sporadic lesions were observed in the study animals, there were no obvious differences, either in tissues affected or lesion severity, between AN1792 treated and untreated animals. There were no unique histopathological lesions noted in AN-1782-immunized animals compared to PBS-treated or untreated animals. There were also no differences in the clinical chemistry profile between adjuvant groups and the PBS
treated animals in Example 7. Although there were significant increases in several of the hematology parameters between animals treated with ANI792 and Freund's adjuvant in Example 7 relative to PBS treated animals, these type of effects are expected from Freund's adjuvant treatment and the accompanying peritonitis and do not indicate any adverse effects from AN1792 treatment. Although not part of the toxicological evaluation, PDAPP mouse brain pathology was extensively examined as part of the efficacy endpoints. No sign of treatment related adverse effect on brain morphology was noted in any of the studies. These results indicate that AN1792 treatment is well tolerated and at least substantially free of side effects.
XI. Prevention and Treatment of Subjects A single-dose phase I trial is performed to determine safety. A therapeutic agent is administered in increasing dosages to different patients starting from about 0.01 the level of presumed efficacy, and increasing by a factor of three until a level of about 10 times the effective mouse dosage is reached.
A phase II trial is performed to determine therapeutic efficacy. Patients with early to mid Alzheimer's Disease defined using Alzheimer's disease and Related Disorders Association (ADRDA) criteria for probable AD are selected.
Suitable patients score in the 12-26 range on the Mini-Mental State Exam (MMSE). Other selection criteria are that patients are likely to survive the duration of the study and lack complicating issues such as use of concomitant medications that may interfere. Baseline evaluations of patient function are made using classic psychometric measures, such as the MMSE, and the ADAS, which is a comprehensive scale for evaluating patients with Alzheimer's Disease status and function. These psychometric scales provide a measure of progression of the Alzheimer's condition. Suitable qualitative life scales can also be used to monitor treatment.
Disease progression can also be monitored by MRI. Blood profiles of patients can also be monitored including assays of immunogen-specific antibodies and T-cells responses.
Following baseline measures, patients begin receiving treatment. They are randomized and treated with either therapeutic agent or placebo in a blinded fashion. Patients are monitored at least every six months. Efficacy is determined by a significant reduction in progression of a treatment group relative to a placebo group.
A second phase II trial is performed to evaluate conversion of patients from non-Alzheimer's Disease early memory loss, sometimes referred to as age-associated memory impairment (AAMI), to probable Alzheimer's disease as defined as by ADRDA
criteria. Patients with high risk for conversion to Alzheimer's Disease are selected from a non-clinical population by screening reference populations for early signs of memory loss or other difficulties associated with pre-Alzheimer's symptomatology, a family history of Alzheimer's Disease, genetic risk factors, age, sex, and other features found to predict high-risk for Alzheimer's Disease. Baseline scores on suitable metrics including the MMSE and the ADAS
together with other metrics designed to evaluate a more normal population are collected. These patient populations are divided into suitable groups with placebo comparison against dosing alternatives with the agent. These patient populations are followed at intervals of about six months, and the endpoint for each patient is whether or not he or she converts to probable Alzheimer's Disease as defined by ADRDA criteria at the end of the observation.

XII. General Materials and Methods 1. Measurement of Antibody Titers Mice were bled by making a small nick in the tail vein and collecting about 200 Al of blood into a microfuge tube.
Guinea pigs were bled by first shaving the back hock area and then using an 18 gauge needle to nick the metatarsal vein and collecting the blood into microfuge tubes. Blood was allowed to clot for one hr at room temperature (RT), vortexed, then centrifuged at 14,000 x g for 10 min to separate the clot from the serum. Serum was then transferred to a clean microfuge tube and stored at 4 C until titered.
Antibody titers were measured by ELISA. 96-well microtiter plates (Costar EIA plates) were coated with 100 Al of a solution containing either 10 Ag/ml either A042 or SAPP or other antigens as noted in each of the individual reports in Well Coating Buffer (0.1 M sodium phosphate, pH 8.5, 0.1 6-sodium azide) and held overnight at RT. The wells were aspirated and sera were added to the wells starting at a 1/100 dilution in Specimen Diluent (0.014 M sodium phosphate, pH
7.4, 0.15 M NaC1, 0.6Pr, bovine serum albumin, 0.05%.
thimerosal). Seven serial dilutions of the samples were made directly in the plates in three-fold steps to reach a final dilution of 1/218,700. The dilutions were incubated in the coated-plate wells for one hr at RT. The plates were then washed four times with PBS containing 0.05':1 Tween 20. The second antibody, a goat anti-mouse Ig conjugated to horseradish peroxidase (obtained from Boehringer Mannheim), was added to the wells as 100 Al of a 1/3000 dilution in Specimen Diluent and incubated for one hr at RT. Plates were again washed four times in PBS, Tween 20. To develop the chromogen, 100 Al of Slow TMB (3,3',5,5'-tetramethyl benzidine obtained from Pierce Chemicals) was added to each well and incubated for 15 min at RT. The reaction was stopped by the addition of 25 Al of 2 M H2SO4. The color intensity was then read on a Molecular Devices Vmax at (450 nm - 650 nm).
*Trademark Titers were defined as the reciprocal of the dilution of serum giving one half the maximum OD. Maximal OD was generally taken from an initial 1/100 dilution, except in cases with very high titers, in which case a higher initial 5 dilution was necessary to establish the maximal OD. If the 50% point fell between two dilutions, a linear extrapolation was made to calculate the final titer. To calculate geometric mean antibody titers, titers less than 100 were arbitrarily assigned a titer value of 25.
10 2. Lymphocyte proliferation assay Mice were anesthetized with isoflurane. Spleens were removed and rinsed twice with 5 ml PBS containing 10% heat-inactivated fetal bovine serum (PBS-FES) and then homogenized in a 50 A Centricon unit (Dako A/S, Denmark) in 1.5 ml PBS-FES
15 for 10 sec at 100 rpm in a Medimachine (Dako) followed by filtration through a 100 pore size nylon mesh. Splenocytes were washed once with 15 ml PBS-FBS, then pelleted by centrifugation at 200 x g for 5 min. Red blood cells were lysed by resuspending the pellet in 5 mL buffer containing 20 0.15 M NH4C1, 1 M KHCO3, 0.1 M NaEDTA, pH 7.4 for five min at RT. Leukocytes were then washed as above. Freshly isolated spleen cells (105 cells per well) were cultured in triplicate sets in 96-well U-bottomed tissue culture-treated microtiter plates (Corning, Cambridge, MA) in RPMI 1640 medium (JRH
25 Biosciences, Lenexa, KS) supplemented with 2.05 mM L
glutamine, 1% Penicillin/Streptomycin, and 10% heat-inactivated FBS, for 96 hr at 37 C. Various Afl peptides, A01-16, A01-40, Afl1-42 or A040-1 reverse sequence protein were also added at doses ranging from 5 AM to 0.18 AM in four 30 steps. Cells in control wells were cultured with Concanavalin A (Con A) (Sigma, cat. # C-5275, at 1 Ag/m1) without added protein. Cells were pulsed for the final 24 hr with 3H-thymidine (1 ACi/well obtained from Amersham Corp., Arlington Heights IL). Cells were then harvested onto UniFilter plates *Trademark and counted in a Top Count Microplate Scintillation Counter (Packard Instruments, Downers Grove, IL). Results are expressed as counts per minute (cpm) of radioactivity incorporated into insoluble macromolecules.
4. Brain Tissue Preparation After euthanasia, the brains were removed and one hemisphere was prepared for immunohistochemical analysis, while three brain regions (hippocampus, cortex and cerebellum) were dissected from the other hemisphere and used to measure the concentration of various AO proteins and APP forms using specific ELISAs (Johnson-Wood et al., supra).
Tissues destined for ELISAs were homogenized in 10 volumes of ice-cold guanidine buffer (5.0 M guanidine-HC1, 50 mM Tris-HC1, pH 8.0). The homogenates were mixed by gentle agitation using an Adams Nutator (Fisher) for three to four hr at RT, then stored at -20 C prior to quantitation of AO and APP.
Previous experiments had shown that the analytes were stable under this storage condition, and that synthetic AO protein (Bachem) could be quantitatively recovered when spiked into homogenates of control brain tissue from mouse littermates (Johnson-Wood et al., supra).
5. Measurement of AO Levels The brain homogenates were diluted 1:10 with ice cold Casein Diluent (0.25% casein, PBS, 0.05% sodium azide, 20 Ag/m1 aprotinin, 5 mM EDTA pH 8.0, 10 Ag/m1 leupeptin) and then centrifuged at 16,000 x g for 20 min at 4 C. The synthetic AO protein standards (1-42 amino acids) and the APP
standards were prepared to include 0.5 M guanidine and 0.1%
bovine serum albumin (BSA) in the final composition. The *Trademark "total" AO sandwich ELISA utilizes monoclonal antibody (mAO) 266, specific for amino acids 13-28 of AO (Seubert, et al.), as the capture antibody, and biotinylated mA0 3D6, specific for amino acids 1-5 of AO (Johnson-Wood, et al), as the reporter antibody. The 3D6 mAO does not recognize secreted APP or full-length APP, but detects only AO species with an amino-terminal aspartic acid. This assay has a lower limit of sensitivity of -50 pg/ml (11 pM) and shows no cross-reactivity to the endogenous murine AO protein at concentrations up to 1 ng/ml (Johnson-Wood et al., supra).
The 01-42 specific sandwich ELISA employs mA0 21F12, specific for amino acids 33-42 of AO (Johnson-Wood, et al.), as the capture antibody. Biotinylated mA0 3D6 is also the reporter antibody in this assay which has a lower limit of sensitivity of about 125 pg/ml (28 pM, Johnson-Wood et al.).
For the AO ELISAs, 100 Al of either mAO 266 (at 10 Ag/ml) or mA0 21F12 at (5 Ag/ml) was coated into the wells of 96-well immunoassay plates (Costar) by overnight incubation at RT.
The solution was removed by aspiration and the wells were blocked by the addition of 200 Al of 0.25t human serum albumin in PBS buffer for at least 1 hr at RT. Blocking solution was removed and the plates were stored desiccated at 4 C until used. The plates were rehydrated with Wash Buffer [Tris-buffered saline (0.15 M NaC1, 0.01 M Tris-HC1, pH 7.5), plus 0Ø5t Tween 20] prior to use. The samples and standards were added in triplicate aliquots of 100 Al per well and then incubated overnight at 4 C. The plates' were washed at least three times with Wash Buffer between each step of the assay.
The biotinylated mAO 3D6, diluted to 0.5 jig/ml in Casein Assay Buffer (0.25t casein, PBS, 0.05t Tween 20, pH 7.4), was added and incubated in the wells for 1 hr at RT. An avidin-horseradish peroxidase conjugate, (Avidin-HRP obtained from Vector, Burlingame, CA), diluted 1:4000 in Casein Assay Buffer, was added to the wells for 1 hr at RT. The colorimetric substrate, Slow TMB-ELISA (Pierce), was added and allowed to react for 15 minutes at RT, after which the enzymatic reaction was stopped by the addition of 25 Al 2 N

H2SO4. The reaction product was quantified using a Molecular Devices Vmax measuring the difference in absorbance at 450 nm and 650 nm.
6. Measurement of APP Levels Two different APP assays were utilized. The first, designated APP-a/FL, recognizes both APP-alpha (a) and full-length (FL) forms of APP. The second assay is specific for APP-a. The APP-a/FL assay recognizes secreted APP including the first 12 amino acids of AO. Since the reporter antibody (2H3) is not specific to the a-clip-site, occurring between amino acids 612-613 of APP695 (Esch et al., Science 248, 1122-1124 (1990)); this assay also recognizes full length APP (APP-FL). Preliminary experiments using immobilized APP antibodies to the cytoplasmic tail of APP-FL to deplete brain homogenates of APP-FL suggest that approximately 30-40% of the APP-a/FL
APP is FL (data not shown). The capture antibody for both the APP-a/FL and APP-a assays is mA0 8E5, raised against amino acids 444 to 592 of the APP695 form (Games et al., supra).
The reporter mAfl for the APP-a/FL assay is mA0 2H3, specific for amino acids 597-608 of APP695 (Johnson-Wood et al., supra) and the reporter antibody for the APP-a assay is a biotinylated derivative of mA0 16H9, raised to amino acids 605 to 611 of APP. The lower limit of sensitivity of the APP-a/FL
assay is about 11 ng/m1 (150 pM) (Johnson-Wood et al.) and that of the APP-a specific assay is 22 ng/ml (0.3 nM). For both APP assays, mA0 8E5 was coated onto the wells of 96-well EIA plates as described above for mA0 266. Purified, recombinant secreted APP-a was used as the reference standard for the APP-a assay and the APP-a/FL assay (Esch et al., supra). The brain homogenate samples in 5 M guanidine were diluted 1:10 in ELISA Specimen Diluent (0.014 M phosphate buffer, pH 7.4, 0.6% bovine serum albumin, 0.05% thimerosal, 0.5 M NaC1, 0.1% NP40). They were then diluted 1:4 in Specimen Diluent containing 0.5 M guanidine. Diluted homogenates were then centrifuged at 16,000 x g for 15 seconds at RT. The APP standards and samples were added to the plate in duplicate aliquots and incubated for 1.5 hr at RT. The biotinylated reporter antibody 2H3 or 16H9 was incubated with samples for 1 hr at RT. Streptavidin-alkaline phosphatase (Boehringer Mannheim), diluted 1:1000 in specimen diluent, was incubated in the wells for I hr at RT. The fluorescent substrate 4-methyl-umbellipheryl-phosphate was added for a 30-*
min RT incubation and the plates were read on a Cytofluor tm 2350 fluorimeter (Millipore) at 365 nm excitation and 450 nm emission.
7. Immunohistochemistry Brains were fixed for three days at 4 C in 4t paraformaldehyde in PBS and then stored from one to seven days at 4 C in It paraformaldehyde, PBS until sectioned. Forty-micron-thick coronal sections were cut on a vibratome at RT
and stored in cryoprotectant (30% glycerol, 30% ethylene glycol in phosphate buffer) at -20 C prior to immunohistochemical processing. For each brain, six sections at the level of the dorsal hippocampus, each separated by consecutive 240 Am intervals, were incubated overnight with one of the following antibodies: (1) a biotinylated anti-A13 (mA0, 3D6, specific for human AO) diluted to a concentration of 2 Ag/ml in PBS and 1% horse serum; or (2) a biotinylated mAfl specific for human APP, 8E5, diluted to a concentration of 3 flg/m1 in PBS and 1.0%- horse serum; or (3) a mA0 specific for glial fibrillary acidic protein (GFAP; Sigma Chemical Co.) diluted 1:500 with 0.25%- Triton X-100 and 1% horse serum, in Tris-buffered saline, pH 7.4 (TBS); or (4) a mA0 specific for CD11b, MAC-1 antigen, (Chemicon International) diluted 1:100 with 0.25%- Triton X-100 and 1% rabbit serum in TBS; or (5) a mA0 specific for MHC II antigen, (Pharmingen) diluted 1:100 with 0.25% Triton X-100 and 1% rabbit serum in TBS; or (6) a rat mA0 specific for CD 43 (Pharmingen) diluted 1:100 with 1%
* Trademark rabbit serum in PBS or (7) a rat mA0 specific for CD 45RA
(Pharmingen) diluted 1:100 with 1% rabbit serum in PBS; or (8) a rat monoclonal AO specific for CD 45R3 (Pharmingen) diluted 1:100 with 1% rabbit serum in PBS; or (9) a rat monoclonal AO
5 specific for CD 45 (Pharmingen) diluted 1:100 with 1% rabbit serum in PBS; or (10) a biotinylated polyclonal hamster AO
specific for CD3e (Pharmingen) diluted 1:100 with 1% rabbit serum in PBS or (11) a rat mA0 specific for CD3 (Serotec) diluted 1:200 with 1% rabbit serum in PBS; or with (12) a 10 solution of PBS lacking a primary antibody containing 1%
normal horse serum.
Sections reacted with antibody solutions listed in 1,2 and 6-12 above were pretreated with 1.0% Triton X-100, 0.4%
hydrogen peroxide in PBS for 20 min at RT to block endogenous 15 peroxidase. They were next incubated overnight at 4 C with Primary antibody. Sections reacted with 3D6 or 8E5 or CD3e mAOs were then reacted for one hr at RT with a horseradish peroxidase-avidin-biotin-complex with kit components "A" and "B" diluted 1:75 in PBS (Vector Elit: Standard Kit, Vector 20 Labs, Burlingame, CA.). Sections reacted with antibodies specific for CD 45RA, CD 45RB, CD 45, CD3 and the PBS solution devoid of primary antibody were incubated for 1 hour at RT
with biotinylated anti-rat IgG (Vector) diluted 1:75 in PBS or biotinylated anti-mouse IgG (Vector) diluted 1:75 in PBS, 25 respectively. Sections were then reacted for one hr at RT
with a horseradish peroxidase-avidin-biotin-complex with kit components "A" and "B" diluted 1:75 in PBS (Vector Elite Standard Kit, Vector Labs, Burlingame, CA.).
Sections were developed in 0.01% hydrogen peroxide, 0.05%
30 3,3'-diaminobenzidine (DAB) at RT. Sections destined for incubation with the GFAP-, MAC-1- AND MHC II-specific antibodies were pretreated with 0.6% hydrogen peroxide at RT
to block endogenous peroxidase then incubated overnight with the primary antibody at 4 C. Sections reacted with the GFAP
35 antibody were incubated for 1 hr at RT with biotinylated anti-mouse IgG made in horse (Vector Laboratories; Vectastairi Elite * Trademark ABC Kit) diluted 1:200 with TBS. The sections were next reacted for one hr with an avidin-biotin-peroxidase complex (Vector Laboratories; Vectastain Elite ABC Kit) diluted 1:1000 with TBS. Sections incubated with the MAC-1-or MHC II-specific mA0 as the primary antibody were subsequently reacted for 1 hr at RT with biotinylated anti-rat IgG made in rabbit diluted 1:200 with TBS, followed by incubation for one hr with avidin-biotin-peroxidase complex diluted 1:1000 with TBS.
Sections incubated with GFAP-, MAC-1- and MEC II-specific antibodies were then visualized by treatment at RT with 0.05%
DAB, 0.01% hydrogen peroxide, 0.04% nickel chloride, TBS for 4 and 11 min, respectively.
Immunolabeled sections were mounted on glass slides (VWR, Superfrost slides), air dried overnight, dipped in Propar (Anatech) and overlaid with coverslips using Permount (Fisher) as the mounting medium.
To counterstain AO plaques, a subset of the GFAP-positive sections were mounted on Superfros slides and incubated in aqueous 1% Thioflavin S (Sigma) for 7 min following immunohistochemical processing. Sections were then dehydrated and cleared in Propar, then overlaid with coverslips mounted with Permount.
8. Image Analysis A Videometric 150 Image Analysis System (041:1cor, Inc., Gaithersburg, MD) linked to a Nikon Microphot-FX microscope through a CCD video camera and a Sony Trinitron monitor was used for quantification of the immunoreactive slides. The image of the section was stored in a video buffer and a color-and saturation-based threshold was determined to select and calculate the total pixel area occupied by the immunolabeled structures. For each section, the hipnocampus was manually outlined and the total pixel area occupied by the hipnocampus * Trademark was calculated. The percent amyloid burden was measured as:
(the fraction of the hippocampal area containing AO deposits immunoreactive with mA0 3D6) x 100. Similarly, the percent neuritic burden was measured as: (the fraction of the hippocampal area containing dystrophic neurites reactive with =
mA0 8E5) x100. The C-Imaging System (Compix, Inc., Cranberry Township, PA) operating the Simple 32 Software Application program was linked to a Nikon Microphot-FX microscope through an Optronics camera and used to quantitate the percentage of the retrospenial cortex occupied by GFAP-positive astrocytes and MAC-1-and MHC II-positive microglia. The image of the immunoreacted section was stored in a video buffer and a monochrome-based threshold was determined to select and calculate the total pixel area occupied by immunolabeled cells. For each section, the retrosplenial cortex (RSC) was manually outlined and the total pixel area occupied by the RSC
was calculated. The percent astrocytosis was defined as: (the fraction of RSC occupied by GFAP-reactive astrocytes) X 100.
Similarly, percent microgliosis was defined as: (the fraction of the RSC occupied by MAC-1- or MHC II-reactive microglia) X
100. For all image analyses, six sections at the level of the dorsal hippocampus, each separated by consecutive 240 Am intervals, were quantitated for each animal. In all cases, the treatment status of the animals was unknown to the observer.
Although the foregoing invention has been described in detail for purposes of clarity of understanding, it will be obvious that certain modifications may be practiced within the scope of the appended claims.
* Trademark !
, I

S
= -q %a .p.
I
_ TITER AT 50% MAXIMAL O.D.
Aggreated AB Infected mice n , Age of PDAPP mouse 100 mouse 101 mouse 102 _mouse 103 mouse 104 mouse 105 mouse 106 , mouse 107 mouse 108 i =

1000 15000 50000, 80000 100000 k.., I 6, 15000 65000 30000 55000 300 15500 15005 50000 66656 Co4 1.., IN
8 20000 55000 55000 50000 400 1,5000 15555 50000 6-6-6-66 .
w_ 15 40000 20000, 60000 50000 900 1E500 w 70000 25000 20000 =
=
=
______________________________________________________________________ .
. .
CO
o Co e.pi Co4 I
.

PBS Injected mice on both Immunogem = _ at 1/100 , . . _ I:
_____ Age of PDAPP mouse 113 mouse 114 mouse115 r_nouse116 mouse117 n < 4x _____________________________________________________ 6 bkg < 4x bkg __ < 4x bkg __ < 4x __ bkg < 4x bkg 13 _ , ' 10 5 x.pkg <4x bkg <4x bkg .< 4x bkg __ < 4x bkg mo 12< 4x bkg <4x bkg <4x bkg ,< 4x bkg <4x bkg oo ti..
w cc a.

!

Claims (125)

CLAIMS:
1. A pharmaceutical composition comprising an immunogenic agent comprising A.beta.
or an immunogenic fragment thereof effective to induce an immunogenic response comprising antibodies against A.beta. in a patient, and a pharmaceutically acceptable adjuvant, wherein the adjuvant enhances the immune response to A.beta..
2. The pharmaceutical composition of claim 1, wherein the adjuvant comprises alum.
3. The pharmaceutical composition of claim 1, wherein the adjuvant comprises 3 De-O-acylated monophosphoryl lipid A (MPL.TM.).
4. The pharmaceutical composition of claim 1, wherein the adjuvant comprises Stimulon.TM. QS-21.
5. The pharmaceutical composition of claim 1, wherein the adjuvant comprises M-CSF.
6. The pharmaceutical composition of claim 1, further comprising GM-CSF.
7. The pharmaceutical composition of any one of claims 1 - 6, wherein the agent is encapsulated within a particle.
8. The pharmaceutical composition of claim 7, wherein the particle is a polylactide polyglycolide copolymer (PLPG) particle.
9. The pharmaceutical composition of any one of claims 1-8, wherein the agent is A.beta..
10. The pharmaceutical composition of claim 9, wherein A.beta. is A.beta.43.
11. The pharmaceutical composition of claim 9, wherein A.beta. is A.beta.42.
12. The pharmaceutical composition of claim 9, wherein A.beta. is A.beta.41.
13. The pharmaceutical composition of claim 9, wherein A.beta. is A.beta.40.
14. The pharmaceutical composition of claim 9, wherein A.beta. is A.beta.39.
15. The pharmaceutical composition of any one of claims 9-14, comprising at least µg of A.beta..
16. The pharmaceutical composition of any one of claims 9-14, comprising at least µg of A.beta..
17. The pharmaceutical composition of any one of claims 9-14, comprising at least 50 µg of A.beta..
18. The pharmaceutical composition of any one of claims 9-14, comprising at least 100 µg of A.beta..
19. The pharmaceutical composition of any one of claims 1-8, wherein the agent is an immunogenic fragment of A.beta..
20. The pharmaceutical composition of claim 19, wherein the immunogenic A.beta.
fragment is from the N-terminal half of A.beta..
21. The pharmaceutical composition of claim 20, wherein the immunogenic A.beta.
fragment is A.beta.1-5.
22. The pharmaceutical composition of claim 20, wherein the immunogenic A.beta.
fragment is A.beta.1-6.
23. The pharmaceutical composition of any one of claims 19-22, comprising at least µg of the immunogenic A.beta. fragment.
24. The pharmaceutical composition of any one of claims 19-22, comprising at least µg of the immunogenic A.beta. fragment.
25. The pharmaceutical composition of any one of claims 19-22, comprising at least 50 µg of the immunogenic A.beta. fragment.
26. The pharmaceutical composition of any one of claims 19-22, comprising at least 100 µg of the immunogenic A.beta. fragment.
27. A use of an immunogenic agent comprising A.beta. or an immunogenic fragment thereof effective to induce an immune response comprising antibodies to A.beta. in the manufacture of a medicament for treating or preventing a disease characterized by amyloid deposit in a patient by means of the antibodies.
28. The use of claim 27, wherein the medicament further comprises a pharmaceutically acceptable adjuvant that enhances the immune response to A.beta..
29. The use of claim 28, wherein the adjuvant comprises alum.
30. The use of claim 28, wherein the adjuvant comprises 3 De-O-acylated monophosphoryl lipid A (MPL.TM.).
31. The use of claim 28, wherein the adjuvant comprises Stimulon .TM. QS-21.
32. The use of claim 28, wherein the adjuvant comprises M-CSF.
33. The use of claim 27 or 28, wherein the medicament further comprises GM-CSF.
34. A use of an immunogenic agent comprising A.beta., or a fragment thereof, effective to induce an immune response comprising antibodies to A.beta., for treating or preventing a disease characterized by amyloid deposit in a patient.
35. The use of any one of claims 27-34, wherein the agent is encapsulated within a particle.
36. The use of claim 35, wherein the particle is a polylactide polyglycolide copolymer (PLPG) particle.
37. The use of any one of claims 27-36, wherein the agent is A.beta..
38. The use of claim 37, wherein A.beta. is A.beta.43.
39. The use of claim 37, wherein A.beta. is A.beta.42.
40. The use of claim 37, wherein A.beta. is A.beta.41.
41. The use of claim 37, wherein A.beta. is A.beta.40.
42. The use of claim 37, wherein A.beta. is A.beta.39.
43. The use of any one of claims 37-42, wherein the A.beta. comprises at least 10 µg.
44. The use of any one of claims 37-42, wherein the A.beta. comprises at least 20 µg.
45. The use of any one of claims 37-42, wherein the A.beta. comprises at least 50 µg.
46. The use of any one of claims 37-42, wherein the A.beta. comprises at least 100 µg.
47. The use of any one of claims 27-36, wherein the agent is an immunogenic fragment of A.beta..
48. The use of claim 47, wherein the immunogenic A.beta. fragment is from the N-terminal half of A.beta..
49. The use of claim 48, wherein the immunogenic A.beta. fragment is A.beta.1-5.
50. The use of claim 48, wherein the immunogenic A.beta. fragment is A.beta.1-6.
51. The use of any one of claims 48-50, wherein the immunogenic A.beta.
fragment comprises at least 10 µg.
52. The use of any one of claims 47-50, wherein the immunogenic A.beta.
fragment comprises at least 20 µg.
53. The use of any one of claims 47-50, wherein the immunogenic A.beta.
fragment comprises at least 50 µg.
54. The use of any one of claims 47-50, wherein the immunogenic A.beta.
fragment comprises at least 100 µg.
55. A pharmaceutical composition comprising an immunogenic agent comprising A.beta.
or an immunogenic fragment thereof linked to a carrier molecule to form a conjugate, and a pharmaceutically acceptable carrier.
56. The composition of claim 55, wherein the carrier molecule is cholera toxin.
57. The composition of claim 55, wherein the carrier molecule is an immunoglobulin.
58. The composition of claim 55, wherein the carrier molecule is a diphtheria toxin.
59. The composition of claim 58, wherein the diphtheria toxin is CRM 197.
60. The composition of claim 55, wherein the carrier molecule is a tetanus toxoid.
61. The pharmaceutical composition of any one of claims 55-60, further comprising an adjuvant.
62. The pharmaceutical composition of claim 61, wherein the adjuvant comprises alum.
63. The pharmaceutical composition of claim 61, wherein the adjuvant comprises 3 De-O-acylated monophosphoryl lipid A (MPL.TM.).
64. The pharmaceutical composition of claim 61, wherein the adjuvant comprises Stimulon.TM. QS-21.
65. The pharmaceutical composition of claim 61, wherein the adjuvant comprises M-CSF.
66. The pharmaceutical composition of any one of claims 55-60, further comprising GM-CSF.
67. The pharmaceutical composition of any one of claims 55-66, wherein the agent is A.beta..
68. The pharmaceutical composition of claim 67, wherein A.beta. is A.beta.43.
69. The pharmaceutical composition of claim 67, wherein A.beta. is A.beta.42.
70. The pharmaceutical composition of claim 67, wherein A.beta. is A.beta.41.
71. The pharmaceutical composition of claim 67, wherein A.beta. is A.beta.40.
72. The pharmaceutical composition of claim 67, wherein A.beta. is A.beta.39.
73. The pharmaceutical composition of any one of claims 67-72, comprising at least µg of A.beta..
74. The pharmaceutical composition of any one of claims 67-72, comprising at least µg of A.beta..
75. The pharmaceutical composition of any one of claims 67-72, comprising at least 50 µg of A.beta..
76. The pharmaceutical composition of any one of claims 67-72, comprising at least 100 µg of A.beta..
77. The pharmaceutical composition of any one of claims 55-66, wherein the agent is an immunogenic fragment of A.beta..
78. The pharmaceutical composition of claim 77, wherein the immunogenic A.beta.
fragment is from the N-terminal half of A.beta..
79. The pharmaceutical composition of claim 78, wherein the immunogenic A.beta.
fragment is A.beta.1-5.
80. The pharmaceutical composition of claim 78, wherein the immunogenic A.beta.
fragment is A.beta.1-6.
81. The pharmaceutical composition of any one of claims 77-80, comprising at least µg of the immunogenic A.beta. fragment.
82. The pharmaceutical composition of any one of claims 77-80, comprising at least µg of the immunogenic A.beta. fragment.
83. The pharmaceutical composition of any one of claims 77-80, comprising at least 50 µg of the immunogenic A.beta. fragment.
84. The pharmaceutical composition of any one of claims 77-80, comprising at least 100 µg of the immunogenic A.beta. fragment.
85. A use of A.beta. or an immunogenic fragment thereof linked to a carrier molecule to form a conjugate in the manufacture of a medicament for treating or preventing a disease characterized by an amyloid deposit in a patient.
86. A use of A.beta. or an immunogenic fragment thereof linked to a carrier molecule to form a conjugate for treating or preventing a disease characterized by an amyloid deposit in a patient.
87. The use of claim 85 or 86, wherein the carrier molecule is cholera toxin.
88. The use of claim 85 or 86, wherein the carrier molecule is an immunoglobulin.
89. The use of claim 85 or 86, wherein the carrier molecule is a diphtheria toxin.
90. The use of claim 89, wherein the diphtheria toxin is CRM 197.
91. The use of claim 85 or 86, wherein the carrier molecule is a tetanus toxoid.
92. The use of claim 85, wherein the medicament further comprises an adjuvant.
93. The use of claim 92, wherein the adjuvant comprises alum.
94. The use of claim 92, wherein the adjuvant comprises 3 De-O-acylated monophosphoryl lipid A (MPL.TM.).
95. The use of claim 92, wherein the adjuvant comprises Stimulon.TM. QS-21.
96. The use of claim 92 wherein the adjuvant comprises M-CSF.
97. The use of claim 92, wherein the medicament further comprises GM-CSF.
98. The use of any one of claims 92-97, wherein the medicament comprises A.beta..
99. The use of claim 98, wherein A.beta. is A.beta.43.
100. The use of claim 98, wherein A.beta. is A.beta.42.
101. The use of claim 98, wherein A.beta. is A.beta.41.
102. The use of claim 98, wherein A.beta. is A.beta.40.
103. The use of claim 98, wherein A.beta. is A.beta.39.
104. The use of any one of claims 98-103, wherein A.beta. comprises at least 10 µg.
105. The use of any one of claims 98-103, wherein A.beta. comprises at least 20 µg.
106. The use of any one of claims 98-103, wherein A.beta. comprises at least 50 µg.
107. The use of any one of claims 98-103, wherein A.beta. comprises at least 100 µg.
108. The use of any one of claims 85-97, wherein the conjugate comprises an immunogenic fragment of A.beta..
109. The use of claim 108, wherein the immunogenic A.beta. fragment is from the N-terminal half of A.beta..
110. The use of claim 109, wherein the immunogenic A.beta. fragment is A.beta.1-5.
111. The use of claim 109 wherein the immunogenic A.beta. fragment is A.beta.1-6.
112. The use of any one of claims 108-111, wherein the immunogenic A.beta.
fragment comprises at least 10 µg.
113. The use of any one of claims 108-111, wherein the immunogenic A.beta.
fragment comprises at least 20 µg.
114. The use of any one of claims 108-111, wherein the immunogenic A.beta.
fragment comprises at least 50 µg.
115. The use of any one of claims 108-111, wherein the immunogenic A.beta.
fragment comprises at least 100 µg.
116. The use of any one of claims 85 or 87-92, wherein the medicament further comprises a pharmaceutically acceptable carrier.
117. A use of a viral vector encoding A.beta. or an immunogenic fragment thereof effective to induce an immune response comprising antibodies against A.beta. in the manufacture of a medicament for treating or preventing a disease characterized by an amyloid deposit in a patient.
118. A use of a viral vector encoding A.beta. or an immunogenic fragment thereof effective to induce an immune response comprising antibodies against A.beta. for treating or preventing a disease characterized by an amyloid deposit in a patient.
119. The use of claim 117 or 118, wherein the viral vector is herpes, adenovirus, adenoassociated virus, a retrovirus, sindbis, semliki forest virus, vaccinia or avian pox.
120. A method of assessing treatment outcome of an Alzheimer's treatment method in a patient, comprising:

determining a baseline amount of antibody specific for A.beta. peptide in a sample from the patient before treatment with an agent capable of inducing antibodies specific for A.beta. peptide; and comparing an amount of antibody specific for A.beta. peptide in a sample from the patient after treatment with the agent to the baseline amount of A.beta.
peptide-specific antibody, wherein an amount of A.beta. peptide-specific antibody measured after the treatment that is significantly greater than the baseline amount of A.beta. peptide-specific antibody indicates a positive treatment outcome, and wherein a reduction or lack of significant difference between the amount of A.beta. peptide-specific antibody measured after the treatment compared to the baseline amount of A.beta. peptide-specific antibody indicates a negative treatment outcome.
121. The method of claim 120, wherein the amounts of antibody are measured as antibody titers.
122. The method of claim 121, wherein the amounts of antibody are measured by an ELISA assay
123. A method of assessing treatment outcome of an Alzheimer's treatment method in a patient administered an agent capable of inducing an antibody specific for A.beta. peptide, said method comprising the steps of:
determining a control amount of antibody specific for A.beta. peptide in samples from a control population; and comparing an amount of antibody specific for A.beta. peptide in a sample from the patient administered the agent to the control amount of A.beta. peptide-specific antibody, wherein an amount of A.beta. peptide-specific antibody measured after the treatment that is significantly greater than the control amount of A.beta. peptide-specific antibody indicates a positive treatment outcome, and wherein a lack of significant difference between the amount of A.beta. peptide-specific antibody measured after beginning said treatment compared to the control amount of A13 peptide-specific antibody indicates a negative treatment outcome.
124. The method of claim 123, wherein the treatment outcome determines the future treatment regime of the patient.
125. A method of assessing treatment outcome of an Alzheimer's treatment method in a patient, comprising:

determining a value for an amount of antibody in a sample from the patient who has been treated with an agent capable of inducing an antibody specific for A.beta. peptide, wherein the antibody is specific for A.beta. peptide; and comparing the value with a control value determined from a population of patients experiencing amelioration of, or freedom from, symptoms of Alzheimer's disease due to treatment with the agent, wherein a value in the patient at least equal to the control value indicates a positive response to treatment.
CA2312920A 1997-12-02 1998-11-30 Prevention and treatment of amyloidogenic disease Expired - Lifetime CA2312920C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US6774097P 1997-12-02 1997-12-02
US60/067,740 1997-12-02
US8097098P 1998-04-07 1998-04-07
US60/080,970 1998-04-07
PCT/US1998/025386 WO1999027944A1 (en) 1997-12-02 1998-11-30 Prevention and treatment of amyloidogenic disease

Publications (2)

Publication Number Publication Date
CA2312920A1 CA2312920A1 (en) 1999-06-10
CA2312920C true CA2312920C (en) 2016-07-12

Family

ID=26748211

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2312920A Expired - Lifetime CA2312920C (en) 1997-12-02 1998-11-30 Prevention and treatment of amyloidogenic disease

Country Status (38)

Country Link
US (26) US20040171815A1 (en)
EP (5) EP2305282B1 (en)
JP (4) JP4677094B2 (en)
KR (3) KR20120135915A (en)
CN (1) CN100374151C (en)
AR (1) AR020050A1 (en)
AT (4) ATE493149T1 (en)
AU (1) AU1706199A (en)
BG (1) BG104562A (en)
BR (1) BR9815357A (en)
CA (1) CA2312920C (en)
CO (1) CO4980846A1 (en)
CY (3) CY1108331T1 (en)
CZ (1) CZ303137B6 (en)
DE (7) DE06075704T1 (en)
DK (4) DK2305282T3 (en)
EA (3) EA007218B1 (en)
EE (1) EE05377B1 (en)
ES (4) ES2428740T3 (en)
GE (2) GEP20053715B (en)
HK (2) HK1095265A1 (en)
HR (2) HRP20000443B1 (en)
HU (2) HU228061B1 (en)
ID (1) ID25504A (en)
IL (5) IL136252A0 (en)
IS (1) IS5500A (en)
MY (1) MY134906A (en)
NO (3) NO328865B1 (en)
NZ (1) NZ504569A (en)
PE (1) PE20001383A1 (en)
PL (2) PL202706B1 (en)
PT (4) PT1033996E (en)
SA (1) SA99191269B1 (en)
SG (1) SG111083A1 (en)
SI (4) SI1033996T1 (en)
TR (1) TR200001608T2 (en)
TW (1) TWI239847B (en)
WO (1) WO1999027944A1 (en)

Families Citing this family (292)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU1072897A (en) 1995-12-12 1997-07-03 Karolinska Innovations Ab Peptide binding the klvff-sequence of amyloid beta
US8173127B2 (en) * 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US6703015B1 (en) 1999-09-03 2004-03-09 Ramot At Tel-Aviv University Ltd. Filamentous bacteriophage displaying an β-amyloid epitope
US7588766B1 (en) * 2000-05-26 2009-09-15 Elan Pharma International Limited Treatment of amyloidogenic disease
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US6761888B1 (en) * 2000-05-26 2004-07-13 Neuralab Limited Passive immunization treatment of Alzheimer's disease
US6905686B1 (en) 1997-12-02 2005-06-14 Neuralab Limited Active immunization for treatment of alzheimer's disease
US6787523B1 (en) * 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US7179892B2 (en) * 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
TWI239847B (en) * 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7964192B1 (en) * 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US20080050367A1 (en) 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
US20050059802A1 (en) * 1998-04-07 2005-03-17 Neuralab Ltd Prevention and treatment of amyloidogenic disease
WO1999060024A1 (en) * 1998-05-21 1999-11-25 The University Of Tennessee Research Corporation Methods for amyloid removal using anti-amyloid antibodies
US20030147882A1 (en) 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US7060670B1 (en) 1999-05-05 2006-06-13 Neurochem (International) Limited Stereoselective antifibrillogenic peptides and peptidomimetics thereof
PL352312A1 (en) * 1999-05-13 2003-08-11 American Cyanamid Co Adiuvant associated preparations
AU2008203784B2 (en) * 1999-05-28 2012-01-19 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
PE20010212A1 (en) * 1999-06-01 2001-02-22 Neuralab Ltd COMPOSITIONS OF THE A-BETA PEPTIDE AND PROCESSES TO PRODUCE THEM
UA81216C2 (en) * 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
AU2005202644B2 (en) * 1999-06-01 2009-03-05 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
PT1409654E (en) 1999-06-16 2008-11-25 Boston Biomedical Res Inst Immunological control of beta-amyloid levels in vivo
JP2003509020A (en) * 1999-09-03 2003-03-11 ラモット・ユニバーシティ・オーソリティ・フォー・アプライド・リサーチ・アンド・インダストリアル・ディベロップメント・リミテッド Pharmaceuticals, compositions, and uses thereof useful for diagnosis, treatment, and prevention of plaque forming diseases
US7384640B1 (en) 1999-09-30 2008-06-10 Wyeth Holdings Corporation Mutant cholera holotoxin as an adjuvant
US20020094335A1 (en) * 1999-11-29 2002-07-18 Robert Chalifour Vaccine for the prevention and treatment of alzheimer's and amyloid related diseases
US20070135337A2 (en) * 1999-11-29 2007-06-14 Neurochem (International) Limited Vaccine for the Prevention and Treatment of Alzheimer's and Amyloid Related Diseases
JP4804690B2 (en) * 1999-12-08 2011-11-02 インテレクト・ニューロサイエンシズ・インコーポレーテッド Chimeric peptide as immunogen, antibody thereto, and immunization using chimeric peptide or antibody
EP1752472A3 (en) * 1999-12-08 2007-04-25 Intellect Neurosciences, Inc. Chimeric amyloid beta peptides
KR100879810B1 (en) * 2000-02-21 2009-01-22 하. 룬드벡 아크티에셀스카브 Novel method for down-regulation of amyloid
HUP0300067A3 (en) * 2000-02-21 2010-03-29 Lundbeck & Co As H Novel method for down-regulation of amyloid
IL151378A0 (en) * 2000-02-24 2003-04-10 Univ Washington Humanized antibodies that sequester amyloid beta peptide
WO2001077167A2 (en) * 2000-04-05 2001-10-18 University Of Tennessee Research Corporation Methods of investigating, diagnosing, and treating amyloidosis
US6713450B2 (en) 2000-05-22 2004-03-30 New York University Synthetic immunogenic but non-amyloidogenic peptides homologous to amyloid β for induction of an immune response to amyloid β and amyloid deposits
JP2003516929A (en) * 2000-06-01 2003-05-20 ニユーララブ・リミテツド Prevention and treatment of amyloidogenic diseases
US7371920B2 (en) 2000-06-20 2008-05-13 The Governing Council Of The University Of Toronto Transgenic mouse model of neurodegenerative disorders
DE60118913T2 (en) 2000-06-20 2007-01-25 The Governing Council Of The University Of Toronto, Toronto Transgenic animal model for neurodegenerative diseases
JP5008244B2 (en) 2000-06-23 2012-08-22 ワイス・ホールディングズ・コーポレイション Assembly of wild-type and chimeric influenza virus-like particles (VLPs)
NZ523428A (en) 2000-06-28 2008-03-28 Prana Biotechnology Ltd Neurotoxic oligomers
CA2414772C (en) * 2000-07-07 2011-06-28 Jan Naslund Prevention and treatment of alzheimer's disease
AU2007200047B2 (en) * 2000-07-07 2009-11-26 Bioarctic Neuroscience Ab Prevention and treatment of Alzheimer's disease
PT1317479E (en) * 2000-09-06 2009-10-29 Univ Pasteur Methods and compositions for diseases associated with amyloidosis
US20040038302A1 (en) * 2000-09-19 2004-02-26 Roger Nitsch Methods and compounds for treating brain amyloidosis
IL139308A0 (en) * 2000-10-26 2001-11-25 Marikovsky Moshe Peptides from amyloid precursor protein which inhibit tumor growth and metastasis
CA2429000A1 (en) 2000-11-10 2002-05-16 Wyeth Holdings Corporation Adjuvant combination formulations
DE60141976D1 (en) 2000-11-29 2010-06-10 Univ Rochester HELPERVIRUS FREE HERPESVIRUS AMPLIFICATE PARTNERS AND ITS USES
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
PE20020574A1 (en) * 2000-12-06 2002-07-02 Wyeth Corp HUMANIZED ANTIBODIES THAT RECOGNIZE THE AMYLOID PEPTIDE BETA
DE60144198D1 (en) 2000-12-28 2011-04-21 Wyeth Llc RECOMBINANT PROTECTION PROTEIN FROM STREPTOCOCCUS PNEUMONIAE
US7264810B2 (en) 2001-01-19 2007-09-04 Cytos Biotechnology Ag Molecular antigen array
US6815175B2 (en) 2001-03-16 2004-11-09 Cornell Research Foundation, Inc. Anti-amyloid peptide antibody based diagnosis and treatment of a neurological disease or disorder
GB0109297D0 (en) 2001-04-12 2001-05-30 Glaxosmithkline Biolog Sa Vaccine
ES2312569T3 (en) 2001-04-30 2009-03-01 Eli Lilly And Company HUMANIZED ANTIBODIES.
EP2165714B1 (en) 2001-04-30 2013-10-23 Eli Lilly And Company Humanized antibodies recognizing the beta-amyloid peptide
US8092791B2 (en) 2001-05-23 2012-01-10 University Of Rochester Method of producing herpes simplex virus amplicons, resulting amplicons, and their use
US6906169B2 (en) 2001-05-25 2005-06-14 United Biomedical, Inc. Immunogenic peptide composition comprising measles virus Fprotein Thelper cell epitope (MUFThl-16) and N-terminus of β-amyloid peptide
CN1977971A (en) 2001-06-07 2007-06-13 惠氏控股有限公司 Mutant forms of cholera holotoxin as an adjuvant
US7332174B2 (en) 2001-06-07 2008-02-19 Wyeth Holdings Corporation Mutant forms of cholera holotoxin as an adjuvant
US7829087B2 (en) 2001-07-09 2010-11-09 Elan Pharmaceuticals, Inc. Methods of treating cognitive impairment
CA2453403C (en) 2001-07-09 2013-10-08 Elan Pharmaceuticals, Inc. Methods of inhibiting amyloid toxicity
WO2003016467A2 (en) * 2001-08-17 2003-02-27 Eli Lilly And Company Use of antibodies having high affinity for soluble ass to treat conditions and diseases related to ass
US7771722B2 (en) 2001-08-17 2010-08-10 Eli Lilly And Company Assay method for alzheimer's disease
WO2003016466A2 (en) * 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
EP1519740A4 (en) * 2001-08-17 2005-11-09 Lilly Co Eli Rapid improvement of cognition in conditions related to a-beta
ES2295401T3 (en) * 2001-08-17 2008-04-16 Washington University TEST METHOD FOR ALZHEIMER'S DISEASE.
MY144532A (en) 2001-08-20 2011-09-30 Lundbeck & Co As H Novel method for down-regulation of amyloid
US20030082191A1 (en) 2001-08-29 2003-05-01 Poduslo Joseph F. Treatment for central nervous system disorders
MX339524B (en) 2001-10-11 2016-05-30 Wyeth Corp Novel immunogenic compositions for the prevention and treatment of meningococcal disease.
EP1572894B1 (en) * 2001-11-21 2016-04-13 New York University Synthetic immunogenic but non-deposit-forming polypeptides and peptides homologous to amyloid beta, prion protein, amylin, alpha synuclein, or polyglutamine repeats for induction of an immune response thereto
US20040052928A1 (en) 2002-09-06 2004-03-18 Ehud Gazit Peptides and methods using same for diagnosing and treating amyloid-associated diseases
AR038568A1 (en) 2002-02-20 2005-01-19 Hoffmann La Roche ANTI-A BETA ANTIBODIES AND ITS USE
US20040001848A1 (en) * 2002-03-01 2004-01-01 Szu-Yi Chou Method of producing disease-specific antigens
EP1480666B1 (en) * 2002-03-05 2012-06-13 Ramot at Tel-Aviv University Ltd. Immunizing composition and method for inducing an immune response against the beta-secretase cleavage site of amyloid precursor protein
MY139983A (en) * 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
CA2493119A1 (en) * 2002-07-17 2004-01-22 Mindset Biopharmaceuticals Usa Inc. Peptides and methods of screening immunogenic peptide vaccines against alzheimer's disease
MXPA05000819A (en) 2002-07-19 2005-08-29 Cytos Biotechnology Ag Vaccine compositions containing amyloid beta1-6 antigen arrays.
AU2003256789A1 (en) 2002-08-13 2004-02-25 U.S. Department Of Veterans Affairs Method of detecting and preventing alzheimer's disease, particularly at prodromal and early stages
US7785608B2 (en) 2002-08-30 2010-08-31 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
CA2498407A1 (en) * 2002-09-12 2004-03-25 The Regents Of The University Of California Immunogens and corresponding antibodies specific for high molecular weight aggregation intermediates common to amyloids formed from proteins of differing sequence
WO2010011999A2 (en) 2008-07-25 2010-01-28 The Regents Of The University Of California Methods and compositions for eliciting an amyloid-selective immune response
BR0315157A (en) * 2002-10-09 2005-08-09 Rinat Neuroscience Corp Methods of treating alzheimer's disease by employing antibodies directed against amyloid beta peptide and compositions thereof
US8697082B2 (en) 2002-11-01 2014-04-15 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US20080014194A1 (en) 2003-10-31 2008-01-17 Elan Pharmaceuticals, Inc. Prevention and Treatment of Synucleinopathic and Amyloidogenic Disease
US8506959B2 (en) * 2002-11-01 2013-08-13 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US9034337B2 (en) 2003-10-31 2015-05-19 Prothena Biosciences Limited Treatment and delay of outset of synucleinopathic and amyloidogenic disease
TW200509968A (en) * 2002-11-01 2005-03-16 Elan Pharm Inc Prevention and treatment of synucleinopathic disease
CN100450551C (en) * 2002-11-29 2009-01-14 中国医学科学院基础医学研究所 Recombinated adeno-associated virus genes vaccine for prevention and cure of Alzheimer disease and use thereof
US20070010435A1 (en) 2002-12-19 2007-01-11 New York University Method for treating amyloid disease
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
CA2513722A1 (en) * 2003-02-01 2004-08-19 Neuralab Limited Active immunization to generate antibodies to soluble a-beta
ATE468886T1 (en) 2003-02-10 2010-06-15 Applied Molecular Evolution ABETA-BINDING MOLECULES
JP5036302B2 (en) * 2003-02-10 2012-09-26 ティオー − ビービービー ホールディング ベスローテン フェンノートシャップ Nucleic acids differentially expressed at the blood-brain barrier under inflammatory conditions
US8663650B2 (en) 2003-02-21 2014-03-04 Ac Immune Sa Methods and compositions comprising supramolecular constructs
WO2004083251A2 (en) * 2003-03-17 2004-09-30 Wyeth Holdings Corporation Mutant cholera holotoxin as an adjuvant and an antigen carrier protein
KR100546066B1 (en) * 2003-03-21 2006-01-26 한국생명공학연구원 The transformed plant cell expressing tandem repeats of ?-amyloid gene and plant produced by the same
US7632816B2 (en) * 2003-03-28 2009-12-15 New York University Treatment of Alzheimer amyloid deposition
US7732162B2 (en) 2003-05-05 2010-06-08 Probiodrug Ag Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
ES2246178B1 (en) * 2003-05-08 2007-03-01 Universidad De Zaragoza. USE OF ANTIBODIES FOR THE TREATMENT OF AMYLOID DISEASES.
US7358331B2 (en) * 2003-05-19 2008-04-15 Elan Pharmaceuticals, Inc. Truncated fragments of alpha-synuclein in Lewy body disease
TWI374893B (en) 2003-05-30 2012-10-21 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
JP4888876B2 (en) * 2003-06-13 2012-02-29 田平 武 Recombinant adeno-associated virus vector for the treatment of Alzheimer's disease
KR101215821B1 (en) 2003-06-30 2012-12-28 텔 아비브 유니버시티 퓨쳐 테크놀로지 디벨롭먼트 엘.피. Peptides antibodies directed thereagainst and methods using same for diagnosing and treating amyloid-associated diseases
WO2005014041A2 (en) * 2003-07-24 2005-02-17 Novartis Ag Use of an amyloid beta dna vaccine for the treatment and/or prevention of amyloid diseases
US7807171B2 (en) 2003-07-25 2010-10-05 Ac Immune Sa Therapeutic vaccine targeted against P-glycoprotein 170 for inhibiting multidrug resistance in the treatment of cancers
GB0321615D0 (en) 2003-09-15 2003-10-15 Glaxo Group Ltd Improvements in vaccination
US20070264280A1 (en) * 2003-11-07 2007-11-15 Federoff Howard J Compositions and Methods for Treating Neurological Diseases
US7674599B2 (en) 2003-11-08 2010-03-09 Elan Pharmaceuticals, Inc. Methods of using antibodies to detect alpha-synuclein in fluid samples
EP2336147B1 (en) 2003-12-17 2014-04-30 Janssen Alzheimer Immunotherapy A beta immunogenic peptide carrier conjugates and methods of producing same
US20050225165A1 (en) * 2004-04-13 2005-10-13 Naik Sanjeev M Brake by-wire control system
ATE419008T1 (en) 2004-04-26 2009-01-15 Innate Pharma ADJUVANT COMPOSITION AND METHOD OF USE THEREOF
GB0410220D0 (en) 2004-05-07 2004-06-09 Kirkham Lea Ann Mutant pneumolysin proteins
SE0401601D0 (en) 2004-06-21 2004-06-21 Bioarctic Neuroscience Ab Protofibril specific antibodies and uses thereof
CN103143011A (en) * 2004-06-25 2013-06-12 魁北克益得生物医学公司 Compositions and methods for treating neurological disorders
WO2006014638A2 (en) * 2004-07-19 2006-02-09 The General Hospital Corporation ANTIBODIES TO CROSS-LINKED AMYLOID β OLIGOMERS
KR20070040824A (en) 2004-07-30 2007-04-17 리나트 뉴로사이언스 코퍼레이션 Antibodies directed against amyloid-beta peptide and methods using same
EP1787998A4 (en) 2004-08-11 2008-08-27 Mitsubishi Chem Corp Antibody and utilization of the same
US7807804B2 (en) * 2004-10-05 2010-10-05 Wyeth Llc Methods and compositions for improving recombinant protein production
WO2006044666A2 (en) * 2004-10-15 2006-04-27 Northeastern University Detection of disease associated proteolysis by direct mass spectrometry analysis of low molecular weight peptides
GB0424563D0 (en) 2004-11-05 2004-12-08 Novartis Ag Organic compounds
AR051800A1 (en) * 2004-12-15 2007-02-07 Wyeth Corp BETA ANTIBODIES USED TO IMPROVE COGNITION
AR052051A1 (en) 2004-12-15 2007-02-28 Neuralab Ltd AB HUMANIZED ANTIBODIES USED TO IMPROVE COGNITION
EP1853299A4 (en) * 2005-01-14 2009-11-11 Univ California Compositions and methods for inhibiting drusen formation and for diagnosing or treating drusen-related disorders
EP1841455A1 (en) 2005-01-24 2007-10-10 Amgen Inc. Humanized anti-amyloid antibody
GT200600031A (en) 2005-01-28 2006-08-29 ANTI-BETA ANTIBODY FORMULATION
DOP2006000022A (en) * 2005-01-28 2006-08-15 Wyeth Corp STABILIZED LIQUID FORMULATIONS OF POLYPTIDES
US20090170798A1 (en) * 2005-04-20 2009-07-02 Japan As Represented By President Of National Center For Geriatrics And Gerontology Highly safe intranasally administrable gene vaccines for treating alzheimer's disease
CN101228272A (en) * 2005-04-20 2008-07-23 生物载体株式会社 Highly safe intranasally administrable gene vaccines for treating alzheimer's disease
UY29504A1 (en) 2005-04-29 2006-10-31 Rinat Neuroscience Corp DIRECTED ANTIBODIES AGAINST BETA AMYLOID PEPTIDE AND METHODS USING THE SAME.
WO2006119352A2 (en) * 2005-05-03 2006-11-09 University Of South Florida Method of treating cognitive decline and synaptic loss related to alzheimer's disease
EP1879613B1 (en) 2005-05-05 2011-11-30 Merck Sharp & Dohme Corp. Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
WO2006126682A1 (en) * 2005-05-27 2006-11-30 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Vaccine for prevention/treatment of alzheimer disease
CA2611815A1 (en) 2005-06-17 2006-12-28 Elan Pharma International Limited Methods of purifying anti a beta antibodies
CN104618085A (en) 2005-07-10 2015-05-13 适应性频谱和信号校正股份有限公司 Adaptive margin and band control method and device
WO2007056401A1 (en) * 2005-11-09 2007-05-18 Merck & Co., Inc. Method for identifying modulators of osbp useful for treating alzheimer's disease
CA2632218A1 (en) * 2005-11-28 2007-10-11 Zymogenetics, Inc. Il-21 receptor antagonists
DK1963369T3 (en) 2005-11-28 2013-06-03 Zymogenetics Inc IL-21 Antagonists
PL1954718T3 (en) 2005-11-30 2015-04-30 Abbvie Inc Anti-a globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
WO2007064972A2 (en) 2005-11-30 2007-06-07 Abbott Laboratories Monoclonal antibodies against amyloid beta protein and uses thereof
WO2007067512A2 (en) * 2005-12-08 2007-06-14 Merck & Co., Inc. Method for identifying modulators of adprh useful for treating alzheimer's disease
SI1960428T1 (en) 2005-12-12 2011-10-28 Hoffmann La Roche Antibodies against amyloid beta with glycosylation in the variable region
RU2015111675A (en) 2005-12-12 2015-08-10 Ац Иммуне Са SPECIFIC IN RESPECT TO AMYLOID BETA (A BETA) 1-42 MONOCLONAL ANTIBODIES WITH THERAPEUTIC PROPERTIES
CA2638775A1 (en) * 2006-02-22 2007-08-30 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Peptide vaccine for inducing production of anti-amyloid-.beta.-peptide antibody
KR101413615B1 (en) 2006-03-23 2014-07-18 바이오악틱 뉴로사이언스 에이비 Improved antibody selective for human protofibrils and the use thereof
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US7479550B2 (en) 2006-06-02 2009-01-20 The Board Of Regents Of The University Of Texas System Amyloid β gene vaccines
SG173385A1 (en) * 2006-07-14 2011-08-29 Ac Immune S A Ch Humanized antibody against amyloid beta
WO2008021296A2 (en) * 2006-08-14 2008-02-21 Thymon, L.L.C. Compositions and methods for the treatment and prophylaxis of alzheimer's disease
US7744890B2 (en) * 2006-10-12 2010-06-29 Wyeth Llc Methods and compositions with reduced opalescence
WO2008070284A2 (en) * 2006-10-16 2008-06-12 Johnnie B. Byrd, Sr. Alzheimer's Center And Research Institute Amyloid beta peptides and methods of uses thereof
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
US20080214987A1 (en) 2006-12-22 2008-09-04 Nanomed Devices, Inc. Microdevice And Method For Transdermal Delivery And Sampling Of Active Substances
AR064642A1 (en) 2006-12-22 2009-04-15 Wyeth Corp POLINUCLEOTIDE VECTOR THAT INCLUDES IT RECOMBINATING CELL THAT UNDERSTANDS THE VECTOR POLYPEPTIDE, ANTIBODY, COMPOSITION THAT UNDERSTANDS THE POLINUCLEOTIDE, VECTOR, RECOMBINATING CELL POLYPEPTIDE OR ANTIBODY, USE OF THE COMPOSITION AND A COMPOSITION AND A METHOD
DK2436696T3 (en) 2007-01-05 2017-09-04 Univ Zurich Anti-beta-amyloid antibody and uses thereof
IL199534A (en) 2007-01-05 2013-01-31 Univ Zuerich Isolated human antibody which is capable of selectively recognizing a neoepitope of a disorder-associated protein, a polynucleotide encoding the antibody, a vector comprising the polynucleotide, a host cell comprising the polynucleotide or the vector, a composition comprising the antibody and methods and uses associated therewith
DK2104682T3 (en) 2007-01-11 2017-01-16 Michael Bacher DIAGNOSIS AND TREATMENT OF ALZHEIMER'S AND OTHER DEMENTIA DISEASES
SI2842967T1 (en) 2007-01-18 2017-01-31 Eli Lilly And Company Pegylated amyloid beta fab
US8147833B2 (en) 2007-02-23 2012-04-03 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
EP2118300B1 (en) 2007-02-23 2015-05-27 Prothena Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
EP2124952A2 (en) 2007-02-27 2009-12-02 Abbott GmbH & Co. KG Method for the treatment of amyloidoses
US7618944B2 (en) * 2007-03-01 2009-11-17 Intezyne Technologies, Inc. Encapsulated amyloid-beta peptides
EP2117540A1 (en) 2007-03-01 2009-11-18 Probiodrug AG New use of glutaminyl cyclase inhibitors
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
WO2008131298A2 (en) * 2007-04-18 2008-10-30 Elan Pharma International Limited Prevention and treatment of cerebral amyloid angiopathy
DK2142514T3 (en) 2007-04-18 2015-03-23 Probiodrug Ag Thiourea derivatives as glutaminyl cyclase inhibitors
EP2012122A1 (en) * 2007-07-06 2009-01-07 Medigene AG Mutated parvovirus structural proteins as vaccines
JP2010528583A (en) * 2007-06-11 2010-08-26 エーシー イミューン ソシエテ アノニム Humanized antibody against amyloid β
JP2010530744A (en) * 2007-06-12 2010-09-16 エーシー イミューン ソシエテ アノニム Humanized antibody against amyloid β
US8613923B2 (en) 2007-06-12 2013-12-24 Ac Immune S.A. Monoclonal antibody
US8048420B2 (en) 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
DK2173378T3 (en) 2007-06-27 2014-05-12 Admune Therapeutics Llc COMPLEXES OF IL-15 AND IL-15RALFA AND APPLICATIONS THEREOF
JP5889529B2 (en) 2007-07-27 2016-03-22 ヤンセン・サイエンシズ・アイルランド・ユーシー Treatment of amyloidogenic diseases
CN101827613A (en) 2007-09-27 2010-09-08 免疫疫苗技术有限公司 Use of liposomes in a carrier comprising a continuous hydrophobic phase for delivery of polynucleotides in vivo
ES2445590T3 (en) 2007-10-05 2014-03-04 Genentech, Inc. Use of anti-beta amyloid antibody in eye diseases
CN101998863B (en) * 2007-10-05 2015-09-16 基因技术公司 The purposes of anti-amyloid beta antibody in oculopathy
ES2612788T3 (en) * 2007-10-05 2017-05-18 Genentech, Inc. Methods and compositions for the diagnosis and treatment of amyloidosis
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
EP2224000B1 (en) * 2007-10-29 2020-05-13 TAO Health Life Pharma Co., Ltd. Antibody and use thereof
CA2910933C (en) 2007-12-07 2017-04-11 Zymogenetics, Inc. Anti-human il-21 monoclonal antibodies
AU2008343855B2 (en) 2007-12-21 2013-08-15 Amgen Inc. Anti-amyloid antibodies and uses thereof
US9345753B2 (en) * 2008-01-16 2016-05-24 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Vaccine for alzheimer's disease
WO2009146523A1 (en) * 2008-06-05 2009-12-10 Immunovaccine Technologies Inc. Compositions comprising liposomes, an antigen, a polynucleotide and a carrier comprising a continuous phase of a hydrophobic substance
WO2010028246A2 (en) * 2008-09-05 2010-03-11 Id Biomedical Corporation Of Quebec Novel compositions and adjuvants
WO2010034072A1 (en) * 2008-09-26 2010-04-01 The University Of Melbourne Alzheimer's disease biomarkers
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
JPWO2010050585A1 (en) * 2008-10-31 2012-03-29 ディナベック株式会社 Alzheimer's disease treatment vector
JP2012508174A (en) 2008-11-05 2012-04-05 ワイス・エルエルシー Multi-component immunogenic composition for preventing β-hemolytic streptococci (BHS) disease
CN102317316B (en) 2008-12-19 2014-08-13 帕尼玛制药股份公司 Human anti-alpha-synuclein autoantibodies
US8614297B2 (en) * 2008-12-22 2013-12-24 Hoffmann-La Roche Inc. Anti-idiotype antibody against an antibody against the amyloid β peptide
US9925282B2 (en) 2009-01-29 2018-03-27 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
AU2010221418B2 (en) 2009-03-02 2015-06-04 Dignity Health Diagnostic devices and methods of use
FR2945538B1 (en) 2009-05-12 2014-12-26 Sanofi Aventis HUMANIZED ANTIBODIES SPECIFIC TO THE PROTOFIBRILLARY FORM OF THE BETA-AMYLOID PEPTIDE.
CA2769822C (en) 2009-08-13 2019-02-19 The Johns Hopkins University Methods of modulating immune function
EP2475428B1 (en) 2009-09-11 2015-07-01 Probiodrug AG Heterocylcic derivatives as inhibitors of glutaminyl cyclase
US9757398B2 (en) 2010-02-20 2017-09-12 Euroespes Biotecnnologia, S.L. Prevention and treatment of alzheimer's disease by amyloid beta peptide and sphin-gosine-1-phosphate
WO2011106885A1 (en) * 2010-03-03 2011-09-09 The University Of British Columbia Oligomer-specific amyloid beta epitope and antibodies
JP6026284B2 (en) 2010-03-03 2016-11-16 プロビオドルグ エージー Inhibitors of glutaminyl cyclase
WO2011107507A1 (en) 2010-03-03 2011-09-09 Boehringer Ingelheim International Gmbh Biparatopic abeta binding polypeptides
MX2012010470A (en) 2010-03-10 2012-10-09 Probiodrug Ag Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5).
WO2011130377A2 (en) 2010-04-15 2011-10-20 Abbott Laboratories Amyloid-beta binding proteins
JP5945532B2 (en) 2010-04-21 2016-07-05 プロビオドルグ エージー Benzimidazole derivatives as inhibitors of glutaminyl cyclase
WO2011151076A2 (en) 2010-06-04 2011-12-08 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts, Universitätsmedizin MONOCLONAL ANTIBODIES TARGETING Αβ OLIGOMERS
NZ606357A (en) 2010-07-30 2015-05-29 Genentech Inc Safe and functional humanized anti beta-amyloid antibody
EP3527220A1 (en) 2010-08-12 2019-08-21 AC Immune S.A. Vaccine engineering
JP6147665B2 (en) 2010-08-14 2017-06-14 アッヴィ・インコーポレイテッド Amyloid beta-binding protein
EP2608805B1 (en) 2010-08-23 2017-07-05 Wyeth LLC STABLE FORMULATIONS OF NEISSERIA MENINGITIDIS rLP2086 ANTIGENS
EP2613806B2 (en) 2010-09-10 2022-10-26 Wyeth LLC Non-lipidated variants of neisseria meningitidis orf2086 antigens
AU2011315920B2 (en) 2010-10-15 2016-04-28 The Board Of Regents Of The University Of Texas System Antibodies that bind amyloid oligomers
TW201223561A (en) 2010-10-26 2012-06-16 Ac Immune Sa Preparation of an antigenic construct
WO2012066549A1 (en) 2010-11-15 2012-05-24 Ramot At Tel-Aviv University Ltd. Dipeptide analogs for treating conditions associated with amyloid fibril formation
WO2012123563A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Benz imidazole derivatives as inhibitors of glutaminyl cyclase
RU2473133C2 (en) * 2011-03-30 2013-01-20 Государственное образовательное учреждение высшего профессионального образования "Северо-Осетинская государственная медицинская академия" Министерства здравоохранения и социального развития РФ Method for prevention of systemic amyloidosis and its nephropathic form in experimental animals
ES2602794T3 (en) 2011-03-31 2017-02-22 Pfizer Inc Novel bicyclic pyridinones
AU2012265320B2 (en) 2011-06-01 2016-11-03 Xiamen Innovax Biotech Co. Ltd. Fusion protein comprising diphtheria toxin non-toxic mutant CMR197 or fragment thereof
WO2012172449A1 (en) 2011-06-13 2012-12-20 Pfizer Inc. Lactams as beta secretase inhibitors
DK2723379T3 (en) 2011-06-23 2018-10-15 Biogen Int Neuroscience Gmbh ANTI-ALPHA SYNUCLEIN BINDING MOLECULES
US9926353B2 (en) 2011-07-19 2018-03-27 New York University Immunotherapeutic modulation of amyloidogenic disease using non-fibrillogenic, non-amyloidogenic polymerized proteins and peptides
US8906382B2 (en) 2011-07-19 2014-12-09 New York University Method for treating amyloid disease
CN102895659B (en) * 2011-07-29 2014-10-29 复旦大学 Composite vaccine for Alzheimer's disease prevention and treatment, and preparation method thereof
US20150065449A1 (en) 2011-08-12 2015-03-05 Florida State University Research Foundation, Inc. Treating Amyloidoses With A Vitamin B12 Composition Including Melatonin, Resveratrol, and EGCG
US20150065448A1 (en) * 2011-08-12 2015-03-05 The Florida State University Research Foundation Inc. Methods of treating amyloidoses with vitamin b12 and diagnostic test for detecting the presence of amyloid-beta peptides
ES2605565T3 (en) 2011-08-31 2017-03-15 Pfizer Inc Hexahydropyran [3,4-D] [1,3] thiazin-2-amine compounds
DK2579042T3 (en) 2011-10-04 2014-07-21 Affiris Ag Method for detecting Aß-specific antibodies in a biological sample
CA2853100A1 (en) * 2011-10-24 2013-05-02 Intellect Neurosciences, Inc. Compositions and methods for treatment of proteinopathies
JP2012102131A (en) * 2012-01-05 2012-05-31 Elan Pharma Internatl Ltd Prophylaxis and treatment for amyloidogenic disease
SA115360586B1 (en) 2012-03-09 2017-04-12 فايزر انك Neisseria meningitidis compositions and methods thereof
US8986710B2 (en) 2012-03-09 2015-03-24 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
ES2585262T3 (en) 2012-05-04 2016-10-04 Pfizer Inc Hexahydropyran [3,4-d] [1,3] thiazin-2-amine heterocyclic compounds substituted as inhibitors of PPA, BACE1 and BACE2
CA2878054C (en) 2012-06-29 2018-09-11 Pfizer Inc. Novel 4-(substituted-amino)-7h-pyrrolo[2,3-d]pyrimidines as lrrk2 inhibitors
JP2015529239A (en) 2012-09-20 2015-10-05 ファイザー・インク Alkyl-substituted hexahydropyrano [3,4-d] [1,3] thiazin-2-amine compounds
UA110688C2 (en) 2012-09-21 2016-01-25 Пфайзер Інк. Bicyclic pirydynony
US10058530B2 (en) 2012-10-25 2018-08-28 The General Hospital Corporation Combination therapies for the treatment of Alzheimer's disease and related disorders
EP3563849A3 (en) 2012-10-25 2020-02-12 The General Hospital Corporation Combination therapies for the treatment of alzheimer's disease and related disorders
US9414752B2 (en) 2012-11-09 2016-08-16 Elwha Llc Embolism deflector
EP2931731A1 (en) 2012-12-11 2015-10-21 Pfizer Inc. Hexahydropyrano [3,4-d][1,3]thiazin-2-amine compounds as inhibitors of bace1
WO2014097038A1 (en) 2012-12-19 2014-06-26 Pfizer Inc. CARBOCYCLIC- AND HETEROCYCLIC-SUBSTITUTED HEXAHYDROPYRANO[3,4-d][1,3]THIAZIN-2-AMINE COMPOUNDS
AU2014203873A1 (en) 2013-01-07 2015-07-30 Biomedical Research Models, Inc. Therapeutic vaccines for treating herpes simplex virus type 2 infections
CA2897678A1 (en) 2013-02-13 2014-08-21 Pfizer Inc. Heteroaryl-substituted hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
US9233981B1 (en) 2013-02-15 2016-01-12 Pfizer Inc. Substituted phenyl hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
EA201591360A1 (en) 2013-02-19 2016-03-31 Пфайзер Инк. AZABENZIMADAZLES AS INHIBITORS INHIBITORS PDE4 FOR THE TREATMENT OF THE CNS AND OTHER DISORDERS
EP2964665B1 (en) 2013-03-08 2018-08-01 Pfizer Inc Immunogenic fusion polypeptides
WO2014159614A1 (en) * 2013-03-14 2014-10-02 Flow Pharma, Inc. Adjuvant and antigen particle formulation
US9102752B2 (en) 2013-03-15 2015-08-11 United Biomedical, Inc. Peptide vaccine for prevention and immunotherapy of dementia of the Alzheimer's type
EP2787347A1 (en) 2013-04-03 2014-10-08 Affiris AG Method for detecting Aß-specific antibodies in a biological sample
US10525005B2 (en) 2013-05-23 2020-01-07 The General Hospital Corporation Cromolyn compositions and methods thereof
US10195257B2 (en) 2013-07-28 2019-02-05 Qantu Therapeutics, Inc. Vaccine formulations comprising quillaja desacylsaponins and beta amyloid peptides or tau protein to induce a Th2 immune response
EP3041502A2 (en) 2013-09-08 2016-07-13 Pfizer Inc. Neisseria meningitidis compositions and methods thereof
JP6425717B2 (en) 2013-10-04 2018-11-21 ファイザー・インク Novel bicyclic pyridinone as a gamma secretase modulator
JP2016534063A (en) 2013-10-22 2016-11-04 ザ ジェネラル ホスピタル コーポレイション Cromolyn derivatives and related imaging and treatment methods
WO2015092592A1 (en) 2013-12-17 2015-06-25 Pfizer Inc. Novel 3,4-disubstituted-1h-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7h-pyrrolo[2,3-c]pyridazines as lrrk2 inhibitors
MX369074B (en) 2014-04-01 2019-10-28 Pfizer Chromene and 1,1 a,2,7b-tetrahydrocyclopropa[c]chromene pyridopyrazinediones as gamma-secretase modulators.
CA2944971C (en) 2014-04-10 2019-05-07 Pfizer Inc. 2-amino-6-methyl-4,4a,5,6-tetrahydropyrano[3,4-d][1,3]thiazin-8a(8h)-yl-1,3-thiazol-4-yl amides
WO2015165961A1 (en) 2014-04-29 2015-11-05 Affiris Ag Treatment and prevention of alzheimer's disease (ad)
EP3137097B1 (en) * 2014-04-29 2023-06-21 ADvantage Therapeutics, Inc. Treatment and prevention of alzheimer's disease (ad)
PT3137094T (en) 2014-04-29 2023-03-09 Advantage Therapeutics Inc Treatment and prevention of alzheimer's disease (ad)
PT3137093T (en) 2014-04-29 2017-12-19 Affiris Ag Treatment and prevention of alzheimer's disease (ad)
WO2015165971A1 (en) * 2014-04-29 2015-11-05 Affiris Ag Treatment and prevention of alzheimer's disease (ad)
EP3157951B1 (en) * 2014-06-17 2020-05-13 Academia Sinica Humanized anti-ige antibodies that crosslink cd23 on b lymphocytes but do not sensitize mast cells
MA53887A (en) 2014-07-10 2021-10-27 Bioarctic Ab ENHANCED PROTOFIBRILL A-BETA BINDING ANTIBODY
WO2016012896A1 (en) 2014-07-24 2016-01-28 Pfizer Inc. Pyrazolopyrimidine compounds
CN106795165B (en) 2014-08-06 2019-09-10 辉瑞公司 Imidazopyridazine compounds
MA41115A (en) 2014-12-02 2017-10-10 Biogen Int Neuroscience Gmbh ALZHEIMER'S DISEASE TREATMENT PROCESS
DK3253755T3 (en) 2015-02-03 2020-09-28 Pfizer Novel cyclopropabenzofuranyl-pyridopyrazine diones
BR112017017460A2 (en) 2015-02-19 2018-04-10 Pfizer Inc. neisseria meningitidis compositions and methods thereof
BR112017026191B1 (en) 2015-06-17 2023-10-10 Pfizer Inc TRICYCLIC COMPOUNDS INHIBITING PHOSPHODIESTERASE, THEIR PHARMACEUTICAL COMPOSITION AND USE THEREOF
CR20170562A (en) 2015-06-24 2018-02-01 Hoffmann La Roche TRANSFERRINE ANTI-RECEIVER ANTIBODIES WITH DESIGNATED AFFINITY.
MX2018003215A (en) 2015-09-14 2018-06-08 Pfizer Novel imidazo [4,5-c] quinoline and imidazo [4,5-c][1,5] naphthyridine derivatives as lrrk2 inhibitors.
JP2018531923A (en) 2015-09-24 2018-11-01 ファイザー・インク N- [2- (2-Amino-6,6-disubstituted-4,4a, 5,6-tetrahydropyrano [3,4-d] [1,3] thiazin-8a (8H) -yl)- 1,3-thiazol-4-yl] amide
EP3353182A1 (en) 2015-09-24 2018-08-01 Pfizer Inc Tetrahydropyrano[3,4-d][1,3]oxazin derivatives and their use as bace inhibitors
EP3353174A1 (en) 2015-09-24 2018-08-01 Pfizer Inc N-[2-(3-amino-2,5-dimethyl-1,1-dioxido-5,6-dihydro-2h-1,2,4-thiadiazin-5-yl)-1,3-thiazol-4-yl]amides useful as bace inhibitors
AR106189A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche BIESPECTIFIC ANTIBODIES AGAINST HUMAN A-b AND THE HUMAN TRANSFERRINE RECEIVER AND METHODS OF USE
CA2997801A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific anti-human cd20/human transferrin receptor antibodies and methods of use
AU2016353553B2 (en) * 2015-11-09 2022-01-20 The University Of British Columbia Amyloid beta epitopes and antibodies thereto
JP7065516B2 (en) 2015-11-09 2022-05-12 ザ・ユニバーシティ・オブ・ブリティッシュ・コロンビア N-terminal epitope of amyloid beta and conformational selective antibody against it
AU2016354688B2 (en) * 2015-11-09 2021-12-16 The University Of British Columbia Epitopes in Amyloid beta mid-region and conformationally-selective antibodies thereto
BR112018015191B1 (en) 2016-02-23 2023-10-17 Pfizer Inc 6,7-DI-HYDRO-5H-PYRAZOLO[5,1-B][1,3]OXAZINE-2-CARBOXAMIDE COMPOUNDS, THEIR USE AND PHARMACEUTICAL COMPOSITION COMPRISING THEM
DK3478679T3 (en) 2016-07-01 2021-06-21 Pfizer 5,7-dihydro-pyrrolo-pyridine derivatives for the treatment of neurological and neurodegenerative diseases
CN109922800B (en) 2016-08-31 2023-06-13 通用医疗公司 Macrophage/microglial cell in neuroinflammation related to neurodegenerative diseases
US20180125920A1 (en) 2016-11-09 2018-05-10 The University Of British Columbia Methods for preventing and treating A-beta oligomer-associated and/or -induced diseases and conditions
SG11201906519RA (en) 2017-01-31 2019-08-27 Pfizer Neisseria meningitidis compositions and methods thereof
CA3056030A1 (en) 2017-03-10 2018-09-13 Pfizer Inc. Novel imidazo[4,5-c]quinoline derivatives as lrrk2 inhibitors
ES2910083T3 (en) 2017-03-10 2022-05-11 Pfizer Substituted cyclic imidazo[4,5-c]quinoline derivatives
RU2678987C2 (en) 2017-04-28 2019-02-05 Общество с ограниченной ответственностью "Научно-производственная фирма ВЕРТА" Peptide for treatment of alzheimer's disease
CN110997693A (en) 2017-06-07 2020-04-10 阿德克斯公司 Tau aggregation inhibitors
CN110944998B (en) 2017-06-22 2022-08-16 辉瑞大药厂 Dihydro-pyrrolo-pyridine derivatives
US11209638B2 (en) * 2017-07-05 2021-12-28 West Virginia University Systems and methods for supporting and positioning body tissue samples in a microscope
AU2017423862A1 (en) 2017-07-20 2020-02-06 Aztherapies, Inc. Powdered formulations of cromolyn sodium and ibuprofen
EP3668886A2 (en) 2017-08-18 2020-06-24 Adrx, Inc. Tau aggregation peptide inhibitors
HUE061510T2 (en) 2017-08-22 2023-07-28 Biogen Ma Inc Pharmaceutical compositions containing anti-beta amyloid antibodies
ES2812698T3 (en) 2017-09-29 2021-03-18 Probiodrug Ag Glutaminyl cyclase inhibitors
FI3768669T3 (en) 2018-03-23 2023-04-26 Pfizer Piperazine azaspiro derivaves
AU2019299347A1 (en) 2018-07-02 2021-01-21 Aztherapies, Inc. Powdered formulations of cromolyn sodium and alpha-lactose

Family Cites Families (433)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US589566A (en) * 1897-09-07 meevten
US6096318A (en) 1973-05-07 2000-08-01 The Ohio State University Antigenically modified HCG polypeptides
JPS57212347A (en) * 1981-06-25 1982-12-27 Nissan Motor Co Ltd Air-fuel ratio control system
US4902506A (en) 1983-07-05 1990-02-20 The University Of Rochester Immunogenic conjugates
GB8308235D0 (en) * 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5158769A (en) * 1984-03-07 1992-10-27 New York Blood Center, Inc. Pre-S gene coded peptide hepatitis B immunogens, vaccines, diagnostics, and synthetic lipid vesicle carriers
US5417986A (en) * 1984-03-16 1995-05-23 The United States Of America As Represented By The Secretary Of The Army Vaccines against diseases caused by enteropathogenic organisms using antigens encapsulated within biodegradable-biocompatible microspheres
US5208036A (en) 1985-01-07 1993-05-04 Syntex (U.S.A.) Inc. N-(ω, (ω-1)-dialkyloxy)- and N-(ω, (ω-1)-dialkenyloxy)-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4666829A (en) 1985-05-15 1987-05-19 University Of California Polypeptide marker for Alzheimer's disease and its use for diagnosis
IT1190383B (en) * 1985-08-01 1988-02-16 Eniricerche Spa PEPTIDOMINETIC SUBSTANCES WITH HYPOTHENSIVE ACTION
US5618920A (en) * 1985-11-01 1997-04-08 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
EP0247091B1 (en) 1985-11-01 1993-09-29 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
US4713366A (en) * 1985-12-04 1987-12-15 The Ohio State University Research Foundation Antigenic modification of polypeptides
US5096706A (en) * 1986-03-25 1992-03-17 National Research Development Corporation Antigen-based treatment for adiposity
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US6548640B1 (en) * 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US5278049A (en) * 1986-06-03 1994-01-11 Incyte Pharmaceuticals, Inc. Recombinant molecule encoding human protease nexin
US5231170A (en) * 1986-08-27 1993-07-27 Paul Averback Antibodies to dense microspheres
US5220013A (en) * 1986-11-17 1993-06-15 Scios Nova Inc. DNA sequence useful for the detection of Alzheimer's disease
US5223482A (en) 1986-11-17 1993-06-29 Scios Nova Inc. Recombinant Alzheimer's protease inhibitory amyloid protein and method of use
US5187153A (en) 1986-11-17 1993-02-16 Scios Nova Inc. Methods of treatment using Alzheimer's amyloid polypeptide derivatives
US4879213A (en) 1986-12-05 1989-11-07 Scripps Clinic And Research Foundation Synthetic polypeptides and antibodies related to Epstein-Barr virus early antigen-diffuse
US4818397A (en) * 1986-12-22 1989-04-04 Sundstrand Corporation Shut-off valve seal
DE3702789A1 (en) 1987-01-30 1988-08-18 Bayer Ag PROCUREMENT PROTEIN OF APC POLYPEPTIDE, FOR CODING DNA AND DIAGNOSTIC USE OF DNA AND PROTEIN
US4912206A (en) * 1987-02-26 1990-03-27 The United States Of America As Represented By The Department Of Health And Human Services CDNA clone encoding brain amyloid of alzheimer's disease
DE3883899T3 (en) * 1987-03-18 1999-04-22 Sb2 Inc CHANGED ANTIBODIES.
US4883666A (en) 1987-04-29 1989-11-28 Massachusetts Institute Of Technology Controlled drug delivery system for treatment of neural disorders
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US5245015A (en) 1991-04-26 1993-09-14 Tanox Biosystems, Inc. Monoclonal antibodies which neutralize HIV-1 through reaction with a conformational epitope in vitro
US5583112A (en) 1987-05-29 1996-12-10 Cambridge Biotech Corporation Saponin-antigen conjugates and the use thereof
US5849298A (en) * 1987-06-24 1998-12-15 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of bovine myelin
US5869054A (en) * 1987-06-24 1999-02-09 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of autoantigens
US5645820A (en) * 1987-06-24 1997-07-08 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
US5571500A (en) * 1987-06-24 1996-11-05 Autoimmune, Inc. Treatment of autoimmune diseases through administration by inhalation of autoantigens
US5571499A (en) * 1987-06-24 1996-11-05 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
ATE130762T1 (en) 1987-06-24 1995-12-15 Autoimmune Inc TREATMENT OF AUTOIMMUNE DISEASES BY ORAL ADMINISTRATION OF AUTOANTIGENS.
US5641474A (en) * 1987-06-24 1997-06-24 Autoimmune, Inc. Prevention of autoimmune diseases by aerosol administration of autoantigens
US4912208A (en) * 1987-06-29 1990-03-27 Abbott Laboratories Fluorophores for encapsulation into liposomes
US5004697A (en) * 1987-08-17 1991-04-02 Univ. Of Ca Cationized antibodies for delivery through the blood-brain barrier
US4966753A (en) 1987-08-18 1990-10-30 Molecular Rx, Inc. Immunotherapeutic methods and compositions employing antigens characteristic of malignant neoplasms
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
CA1339014C (en) 1987-10-08 1997-03-25 Ronald E. Majocha Antibodies to a4 amyloid peptide
US5231000A (en) * 1987-10-08 1993-07-27 The Mclean Hospital Antibodies to A4 amyloid peptide
AU2728588A (en) 1987-10-23 1989-05-23 Genetics Institute Inc. Composition and method for treating cancers characterized by over-expression of the c-fms proto-oncogene
US5089603A (en) * 1989-06-21 1992-02-18 Tanox Biosystems, Inc. Antigenic epitopes present on membrane-bound but not secreted iga
AU3056289A (en) 1988-01-13 1989-08-11 Mclean Hospital Corporation, The Genetic constructs containing the alzheimer brain amyloid gene
US4912094B1 (en) 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
US5576184A (en) * 1988-09-06 1996-11-19 Xoma Corporation Production of chimeric mouse-human antibodies with specificity to human tumor antigens
US5098706A (en) * 1988-11-01 1992-03-24 The Regents Of The University Of California Juvenile hormone esterase for insect control
WO1990005142A1 (en) 1988-11-10 1990-05-17 Imperial Cancer Research Technology Ltd. Polypeptides
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
IL162181A (en) * 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5227159A (en) * 1989-01-31 1993-07-13 Miller Richard A Anti-idiotype antibodies reactive with shared idiotopes expressed by B cell lymphomas and autoantibodies
US5262332A (en) 1989-04-05 1993-11-16 Brigham And Women's Hospital Diagnostic method for Alzheimer's disease: examination of non-neural tissue
AU5525090A (en) 1989-04-14 1990-11-16 Research Foundation For Mental Hygiene, Inc. Monoclonal antibody to amyloid peptide
AU5439790A (en) 1989-04-14 1990-11-16 Research Foundation For Mental Hygiene, Inc. Cerebrovascular amyloid protein-specific monoclonal antibody sv17-6e10
CA2017507C (en) * 1989-05-25 1996-11-12 Gary Van Nest Adjuvant formulation comprising a submicron oil droplet emulsion
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
DE69033487T2 (en) 1989-12-20 2000-06-29 Autoimmune Inc TREATING AUTOIMMUNE DISEASES BY ADMINISTRATING AUTOANTIGENS IN THE FORM OF AEROSOL
GB8928874D0 (en) * 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US5859205A (en) * 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
EP1690935A3 (en) 1990-01-12 2008-07-30 Abgenix, Inc. Generation of xenogeneic antibodies
AU651097B2 (en) 1990-03-02 1994-07-14 Autoimmune, Inc. Enhancement of the down-regulation of autoimmune diseases by oral administration of autoantigens
GB9005705D0 (en) 1990-03-14 1990-05-09 Health Lab Service Board Particle manipulation
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
EP0451700A1 (en) 1990-04-10 1991-10-16 Miles Inc. Recombinant APP minigenes for expression in transgenic mice as models for Alzheimers's disease
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
CA2079880A1 (en) 1990-04-24 1991-10-25 William E. Van Nostrand Purification, detection and methods of use of protease nexin-2
US5753624A (en) * 1990-04-27 1998-05-19 Milkhaus Laboratory, Inc. Materials and methods for treatment of plaquing disease
WO1991016819A1 (en) * 1990-04-27 1991-11-14 Molecular Rx., Inc. Method and composition for treatment of central nervous systems disease states associated with abnormal amyloid beta protein
GB9009548D0 (en) 1990-04-27 1990-06-20 Celltech Ltd Chimeric antibody and method
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
ES2109945T3 (en) 1990-06-01 1998-02-01 Chiron Corp COMPOSITIONS AND PROCEDURES FOR IDENTIFYING BIOLOGICALLY ACTIVE MOLECULES.
US5914109A (en) * 1990-06-15 1999-06-22 New York University Heterohybridomas producing human monoclonal antibodies to HIV-1
JPH06507782A (en) 1990-06-15 1994-09-08 サイオス ノバ インコーポレイテッド Non-human recombinant mammals exhibiting amyloid formation symptoms of Alzheimer's disease
WO1991019795A1 (en) 1990-06-19 1991-12-26 Immuvax Nonpathogenic variant virus
GB9014932D0 (en) 1990-07-05 1990-08-22 Celltech Ltd Recombinant dna product and method
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
KR970005049B1 (en) * 1990-07-16 1997-04-11 더 리젠츠 오브 더 유니벌시티 오브 캘리포니아 Vaculovirus expression vector and retinoblastoma poly peptide
US5780587A (en) * 1990-08-24 1998-07-14 President And Fellows Of Harvard College Compounds and methods for inhibiting β-protein filament formation and neurotoxicity
NZ239643A (en) 1990-09-17 1996-05-28 North American Vaccine Inc Vaccine containing bacterial polysaccharide protein conjugate and adjuvant (c-nd-che-a-co-b-r) with a long chain alkyl group.
US5702906A (en) 1990-09-25 1997-12-30 Genentech, Inc. Antibodies to neurotrophic factor-4 (NT-4)
CA2092823A1 (en) 1990-09-28 1992-03-29 Barry D. Greenberg Transgenic animals with alzheimer's amyloid precursor gene
EP0553244B8 (en) * 1990-10-05 2005-06-08 Celldex Therapeutics, Inc. Targeted immunostimulation with bispecific reagents
CA2092905C (en) 1990-10-15 2002-01-08 Howard L. Weiner Treatment of autoimmune diseases by oral administration of autoantigens
GB9023352D0 (en) 1990-10-26 1990-12-05 Lynxvale Ltd Vaccinia vectors,vaccinia genes and expression products thereof
EP0568575B2 (en) 1991-01-21 2010-11-03 Elan Pharmaceuticals, Inc. Test and model for alzheimer's disease
HU214453B (en) 1991-03-01 1998-03-30 Merial Method for improving the organoleptic qualities of meat
DE4107857A1 (en) * 1991-03-12 1992-09-17 Thomae Gmbh Dr K CYCLIC UREA DERIVATIVES, MEDICAMENTS CONTAINING SUCH COMPOUNDS AND METHOD FOR THE PRODUCTION THEREOF
US5192753A (en) * 1991-04-23 1993-03-09 Mcgeer Patrick L Anti-rheumatoid arthritic drugs in the treatment of dementia
WO1992019267A1 (en) 1991-05-08 1992-11-12 Schweiz. Serum- & Impfinstitut Bern Immunostimulating and immunopotentiating reconstituted influenza virosomes and vaccines containing them
WO1994004679A1 (en) * 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
EP0940468A1 (en) * 1991-06-14 1999-09-08 Genentech, Inc. Humanized antibody variable domain
US5672805A (en) 1991-07-18 1997-09-30 The Regents Of The University Of California Transgenic mice expressing the neurotoxic C-terminus of β-amyloid precursor protein
US5434050A (en) * 1991-08-13 1995-07-18 Regents Of The University Of Minnesota Labelled β-amyloid peptide and methods of screening for Alzheimer's disease
US5283185A (en) 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
ATE148889T1 (en) 1991-09-18 1997-02-15 Affymax Tech Nv METHOD FOR SYNTHESIS OF VARIOUS COLLECTIONS OF OLIGOMERS
US5837268A (en) 1991-10-16 1998-11-17 University Of Saskatchewan GnRH-leukotoxin chimeras
CA2124967C (en) 1991-12-17 2008-04-08 Nils Lonberg Transgenic non-human animals capable of producing heterologous antibodies
DK0620849T3 (en) 1992-01-07 2003-10-20 Elan Pharm Inc Transgenic animal models for Alzheimer's disease
US5679348A (en) * 1992-02-03 1997-10-21 Cedars-Sinai Medical Center Immunotherapy for recurrent HSV infections
DE69333107T2 (en) * 1992-02-11 2004-01-29 Jackson H M Found Military Med DUAL SUPPORT FOR IMMUNOGENIC CONSTRUCTS
US5314813A (en) 1992-02-19 1994-05-24 Scripps Research Institute Drosophila cell lines expressing genes encoding MHC class I antigens and B2-microglobulin and capable of assembling empty complexes and methods of making said cell lines
CA2117492C (en) 1992-02-28 2009-04-07 Howard L. Weiner Bystander suppression of autoimmune diseases
US5714350A (en) * 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
EP0561087B1 (en) 1992-03-20 1999-08-04 N.V. Innogenetics S.A. Mutated form of the beta-amyloid precursor protein gene
US5441870A (en) * 1992-04-15 1995-08-15 Athena Neurosciences, Inc. Methods for monitoring cellular processing of β-amyloid precursor protein
US5604102A (en) 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
US5851787A (en) * 1992-04-20 1998-12-22 The General Hospital Corporation Nucleic acid encoding amyloid precursor-like protein and uses thereof
WO1993022332A2 (en) 1992-04-24 1993-11-11 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
GB9209118D0 (en) 1992-04-28 1992-06-10 Sb 120 Amsterdam Bv Vaccine compositions
DE69327534T2 (en) * 1992-06-18 2000-06-08 Harvard College VACCINE AGAINST DIPHTHERIETOXIN
DK0761231T3 (en) * 1992-06-25 2000-05-08 Smithkline Beecham Biolog Vaccine containing adjuvants
US5837672A (en) * 1992-07-10 1998-11-17 Athena Neurosciences, Inc. Methods and compositions for the detection of soluble β-amyloid peptide
US6610493B1 (en) 1993-06-17 2003-08-26 Brigham And Women's Hospital Screening compounds for the ability to alter the production of amyloid-β peptide
US5766846A (en) * 1992-07-10 1998-06-16 Athena Neurosciences Methods of screening for compounds which inhibit soluble β-amyloid peptide production
WO1994001772A1 (en) 1992-07-13 1994-01-20 The Children's Medical Center Corporation SCREEN FOR ALZHEIMER'S DISEASE THERAPEUTICS BASED ON β-AMYLOID PRODUCTION
IL102687A (en) 1992-07-30 1997-06-10 Yeda Res & Dev Conjugates of poorly immunogenic antigens and synthetic pepide carriers and vaccines comprising them
AU4719393A (en) 1992-07-31 1994-03-03 Medeva Holdings B.V. Expression of recombinant fusion proteins in attenuated bacteria
AU667578B2 (en) 1992-08-27 1996-03-28 Deakin Research Limited Retro-, inverso-, and retro-inverso synthetic peptide analogues
FI96267C (en) 1992-09-16 1996-06-10 Formit Foodprocessing Ab Oy Roller and machine for peeling or shaping potatoes and the like
US5958883A (en) * 1992-09-23 1999-09-28 Board Of Regents Of The University Of Washington Office Of Technology Animal models of human amyloidoses
DE69311048T2 (en) * 1992-09-29 1997-12-11 Nippon Telegraph & Telephone Multi / demultiplexer with grating of grouped waveguides and returned optical paths
KR100310939B1 (en) 1992-10-01 2002-06-20 추후제출 Complex Combination Chemistry Library Encrypted with Tags
AU5358494A (en) 1992-10-13 1994-05-09 Duke University Method of inhibiting binding of amyloid precursor protein to beta-amyloid protein
ES2203620T3 (en) 1992-10-26 2004-04-16 Elan Pharmaceuticals, Inc. PROCEDURES FOR THE IDENTIFICATION OF INHIBITORS OF THE PRODUCTION OF THE BETA-AMILOID PEPTIDE.
US5605811A (en) * 1992-10-26 1997-02-25 Athena Neurosciences, Inc. Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
US5972336A (en) * 1992-11-03 1999-10-26 Oravax Merieux Co. Urease-based vaccine against helicobacter infection
US5733647A (en) * 1992-11-05 1998-03-31 Polymer Innovations, Inc. Insole
US6210671B1 (en) * 1992-12-01 2001-04-03 Protein Design Labs, Inc. Humanized antibodies reactive with L-selectin
WO1994012629A1 (en) 1992-12-02 1994-06-09 Baylor College Of Medicine Episomal vectors for gene therapy
DK0680338T3 (en) * 1993-01-22 2003-07-21 Sloan Kettering Inst Cancer Ganglioside KLH conjugate vaccines with QS-21
DE69432629T3 (en) 1993-01-25 2008-01-17 Takeda Pharmaceutical Co. Ltd. ANTIBODIES AGAINST BETA AMYLOID OR DERIVATIVES AND ITS USE
US5955317A (en) * 1993-01-25 1999-09-21 Takeda Chemical Industries, Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5750106A (en) * 1993-01-28 1998-05-12 Novartis Ag Human monoclonal antibodies to cytomegalovirus
US5358708A (en) 1993-01-29 1994-10-25 Schering Corporation Stabilization of protein formulations
US5989565A (en) * 1993-01-29 1999-11-23 University Of Pittsburgh Elution and identification of T cell epitopes from viable cells
US5472693A (en) 1993-02-16 1995-12-05 The Dow Chemical Company Family of anti-carcinoembryonic antigen chimeric antibodies
CA2115811A1 (en) * 1993-02-17 1994-08-18 Claus Krebber A method for in vivo selection of ligand-binding proteins
CA2115900A1 (en) 1993-02-22 1994-08-23 Gerald W. Becker Pharmaceutical screens and antibodies
EP0689551B1 (en) * 1993-03-17 2008-02-20 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Immunogenic chimeras comprising nucleic acid sequences encoding endoplasmic reticulum signal sequence peptides and at least one other peptide, and their uses in vaccines and disease treatments
JP3880063B2 (en) * 1993-03-18 2007-02-14 シティミューン サイエンシーズ,インコーポレイテッド Compositions and methods for reducing the toxicity of biologically active agents
ES2109685T5 (en) * 1993-03-23 2005-09-01 Smithkline Beecham Biologicals S.A. COMPOSITIONS FOR VACCINES CONTAINING MONOFOSFORIL-LIPIDO TO 3-O-DISABLED.
CA2119090A1 (en) * 1993-03-26 1994-09-27 Wayne R. Gombotz Compositions for controlled release of biologically active tgf-.beta.
IT1270939B (en) 1993-05-11 1997-05-26 Angeletti P Ist Richerche Bio PROCEDURE FOR THE PREPARATION OF IMMUNOGEN AND DIAGNOSTIC REAGENTS, AND IMMUNOGEN AND DIAGNOSTIC REAGENTS SO OBTAINABLE.
DE69426077T3 (en) * 1993-05-25 2004-09-02 Wyeth Holdings Corp. ADJUVANTS FOR VACCINE AGAINST THE RESPIRATORY SYNCITIAL VIRUS
WO1994028412A1 (en) 1993-05-28 1994-12-08 The Miriam Hospital Composition and method for in vivo imaging of amyloid deposits
DE69429723T2 (en) 1993-06-04 2002-09-26 Whitehead Biomedical Inst STRESS PROTEINS AND THEIR USE
US5464823A (en) * 1993-07-20 1995-11-07 The Regents Of The University Of California Mammalian antibiotic peptides
CA2168459C (en) 1993-07-30 2002-10-01 Mohammed Anjam Khan Fusion proteins containing the c-terminal of tetanus toxin linked to a heterologous protein
EP0664814A1 (en) 1993-08-18 1995-08-02 MorphoSys AG Lipopolysaccharide-binding and neutralizing peptides
AU705889B2 (en) 1993-08-26 1999-06-03 Regents Of The University Of California, The Method, compositions and devices for administration of naked polynucleotides which encode antigens and immunostimulatory peptides
DK96493D0 (en) 1993-08-26 1993-08-26 Mouritsen Og Elsner Aps PROCEDURE FOR INDUCING ANTIBODY RESPONSE TO SELF-PROTEINS AND AUTOVACCINE PROCESSED BY THE PROCEDURE
AU707083B2 (en) 1993-08-26 1999-07-01 Bavarian Nordic Inc. Inducing antibody response against self-proteins with the aid of foreign T-cell epitopes
FR2709247B1 (en) * 1993-08-27 1995-09-29 Martin Jean Raymond Device for anchoring spinal instrumentation on a vertebra.
WO1995006407A1 (en) 1993-08-30 1995-03-09 The Regents Of The University Of California Novel component of amyloid in alzheimer's disease and methods for use of same
CN1105728C (en) 1993-09-07 2003-04-16 史密丝克莱恩比彻姆公司 Recombinant IL4 antibodies useful in treatment of IL4 mediated disorders
US5652334A (en) * 1993-09-08 1997-07-29 City Of Hope Method for design of substances that enhance memory and improve the quality of life
US5385887A (en) * 1993-09-10 1995-01-31 Genetics Institute, Inc. Formulations for delivery of osteogenic proteins
PT735893E (en) * 1993-09-14 2009-03-06 Pharmexa Inc Pan dr-binding peptides for enhancement of the immune response
US5415584A (en) 1993-09-21 1995-05-16 Tomco2 Equipment Company Particle blast cleaning apparatus
US5470951A (en) * 1993-09-29 1995-11-28 City Of Hope Peptides for antagonizing the effects of amyloid βprotein
US5858981A (en) * 1993-09-30 1999-01-12 University Of Pennsylvania Method of inhibiting phagocytosis
CA2174501A1 (en) 1993-10-20 1995-04-27 Warren J. Strittmatter Method of binding material to the .beta.-amyloid peptide
ES2182850T3 (en) 1993-10-22 2003-03-16 Genentech Inc PROCEDURES AND COMPOSITIONS FOR MICROENCAPSULATION OF ASSISTANTS.
NZ276860A (en) 1993-11-02 1997-09-22 Affymax Tech Nv Apparatus and its use for synthesising diverse molecular products on substrates
US5744368A (en) * 1993-11-04 1998-04-28 Research Foundation Of State University Of New York Methods for the detection of soluble amyloid β-protein (βAP) or soluble transthyretin (TTR)
US5827690A (en) 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
GB9326253D0 (en) 1993-12-23 1994-02-23 Smithkline Beecham Biolog Vaccines
US5434170A (en) * 1993-12-23 1995-07-18 Andrulis Pharmaceuticals Corp. Method for treating neurocognitive disorders
US5877399A (en) * 1994-01-27 1999-03-02 Johns Hopkins University Transgenic mice expressing APP-Swedish mutation develop progressive neurologic disease
AU1909695A (en) * 1994-01-27 1995-08-15 Regents Of The University Of Minnesota Transgenic non-human mammals with progressive neurologic disease
JPH09511492A (en) * 1994-02-03 1997-11-18 ザ ピコワー インスティテュート フォア メディカル リサーチ Compositions and methods for advanced glycosylation end product mediated modulation of amyloidosis
AUPM411994A0 (en) 1994-02-25 1994-03-24 Deakin Research Limited Epitopes
KR100366831B1 (en) 1994-02-28 2003-03-06 스미또모 가가꾸 고오교오 가부시끼가이샤 Polyolefin Resin Compositions and Resin Films
US5795954A (en) 1994-03-04 1998-08-18 Genentech, Inc. Factor VIIa inhibitors from Kunitz domain proteins
US6270757B1 (en) 1994-04-21 2001-08-07 Genetics Institute, Inc. Formulations for IL-11
US6372716B1 (en) * 1994-04-26 2002-04-16 Genetics Institute, Inc. Formulations for factor IX
CA2189634A1 (en) 1994-05-06 1995-11-16 John J. Baldwin Combinatorial dihydrobenzopyran library
ATE202940T1 (en) 1994-05-25 2001-07-15 John Mcmichael MEANS AND METHODS FOR TREATING PLAQUE DISEASES
US5622701A (en) * 1994-06-14 1997-04-22 Protein Design Labs, Inc. Cross-reacting monoclonal antibodies specific for E- and P-selectin
US5663046A (en) 1994-06-22 1997-09-02 Pharmacopeia, Inc. Synthesis of combinatorial libraries
US5798100A (en) * 1994-07-06 1998-08-25 Immunomedics, Inc. Multi-stage cascade boosting vaccine
US6417178B1 (en) * 1994-07-19 2002-07-09 University Of Pittsburgh Amyloid binding nitrogen-linked compounds for the antemortem diagnosis of alzheimer's disease, in vivo imaging and prevention of amyloid deposits
NZ290089A (en) 1994-07-27 1999-05-28 Queensland Inst Med Res Recombinant polyepitope cytotoxic t lymphocyte (ctl) vaccines
WO1996008565A2 (en) 1994-09-16 1996-03-21 Cancer Research Fund Of Contra Costa RECOMBINANT PEPTIDES DERIVED FROM THE Mc3 ANTI-BA46 ANTIBODY, METHODS OF USE THEREOF, AND METHODS OF HUMANIZING ANTIBODY PEPTIDES
ATE178104T1 (en) * 1994-10-14 1999-04-15 Oriental Sangyo Co Ltd ELECTROLYTIC DEVICE FOR PRODUCING CARBON DIOXIDE
NL9401735A (en) * 1994-10-19 1996-06-03 Crown Gear Bv Gear transmission of a cylindrical pinion with a crown wheel, the crown wheel used in this gear transmission, a method according to which the crown wheel can be made and a tool with which the crown wheel can be made.
US5872005A (en) * 1994-11-03 1999-02-16 Cell Genesys Inc. Packaging cell lines for adeno-associated viral vectors
JPH08137927A (en) * 1994-11-07 1996-05-31 Hitachi Ltd Method for displaying arrangement/wiring of parts
US6114133A (en) * 1994-11-14 2000-09-05 Elan Pharmaceuticals, Inc. Methods for aiding in the diagnosis of Alzheimer's disease by measuring amyloid-β peptide (x-≧41)
US5589154A (en) 1994-11-22 1996-12-31 Rutgers, The State University Of New Jersey Methods for the prevention or treatment of vascular hemorrhaging and Alzheimer's disease
US5688651A (en) * 1994-12-16 1997-11-18 Ramot University Authority For Applied Research And Development Ltd. Prevention of protein aggregation
AU695129B2 (en) * 1995-02-06 1998-08-06 Genetics Institute, Llc Formulations for IL-12
US5786180A (en) 1995-02-14 1998-07-28 Bayer Corporation Monoclonal antibody 369.2B specific for β A4 peptide
US5624937A (en) * 1995-03-02 1997-04-29 Eli Lilly And Company Chemical compounds as inhibitors of amyloid beta protein production
US5854215A (en) * 1995-03-14 1998-12-29 Praecis Pharmaceuticals Incorporated Modulators of β-amyloid peptide aggregation
US5854204A (en) * 1995-03-14 1998-12-29 Praecis Pharmaceuticals, Inc. Aβ peptides that modulate β-amyloid aggregation
US6303567B1 (en) * 1995-03-14 2001-10-16 Praecis Pharmaceuticals, Inc . Modulators of β-amyloid peptide aggregation comprising D-amino acids
US5817626A (en) * 1995-03-14 1998-10-06 Praecis Pharmaceuticals Incorporated Modulators of beta-amyloid peptide aggregation
CA2215162A1 (en) * 1995-03-23 1996-09-26 Cantab Pharmaceuticals Research Limited Vectors for gene delivery
US5869046A (en) * 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
UA56132C2 (en) 1995-04-25 2003-05-15 Смітклайн Бічем Байолоджікалс С.А. Vaccine composition (variants), method for stabilizing qs21 providing resistance against hydrolysis (variants), method for manufacturing vaccine
DE69633175T2 (en) 1995-05-23 2005-08-11 Morphosys Ag MULTIMETER PROTEINS
WO1996039176A1 (en) 1995-06-05 1996-12-12 Brigham & Women's Hospital USE OF ORAL TOLERANCE TO SUPPRESS BOTH Th1 AND Th2 IMMUNE RESPONSES AND TO SUPPRESS ANTIBODY PRODUCTION
JP2001517065A (en) 1995-06-07 2001-10-02 アセナ ニューロサイエンシーズ,インコーポレイテッド Methods for identifying therapeutic agents for Alzheimer's disease using transgenic animal models
US5948763A (en) 1995-06-07 1999-09-07 New York University Peptides and pharmaceutical compositions thereof for treatment of disorders or diseases associated with abnormal protein folding into amyloid or amyloid-like deposits
IT1276680B1 (en) * 1995-06-08 1997-11-03 Oris Spa SYNTHESIS PROCESS OF 1-PYRIDYNIOPROPAN-3-SULPHONATE
US5910427A (en) 1995-06-22 1999-06-08 La Jolla Institute For Allergy And Immunology Antigen non-specific glycosylation inhibiting factor derivatives
US5780361A (en) * 1995-06-23 1998-07-14 Nec Corporation Salicide process for selectively forming a monocobalt disilicide film on a silicon region
DE69619894T2 (en) * 1995-06-30 2002-11-14 American Cyanamid Co Stable macrolides and macrolide vaccine compositions
JP4073955B2 (en) * 1995-07-07 2008-04-09 ダーウィン モレキュラー コーポレイション Chromosome 1 gene and gene product associated with Alzheimer's disease
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6685940B2 (en) 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
AUPN443995A0 (en) 1995-07-27 1995-08-17 Csl Limited Papillomavirus polyprotein
DE69621940T2 (en) 1995-08-18 2003-01-16 Morphosys Ag PROTEIN - / (POLY) PEPTIDE LIBRARIES
WO1997006809A1 (en) * 1995-08-21 1997-02-27 Cytrx Corporation Compositions and methods for growth promotion
AU707484B2 (en) 1995-09-14 1999-07-08 Regents Of The University Of California, The Antibodies specific for native PrPsc
US5664823A (en) * 1995-09-18 1997-09-09 Ford Global Technologies, Inc. Instrument panel brace
US5731284A (en) 1995-09-28 1998-03-24 Amgen Inc. Method for treating Alzheimer's disease using glial line-derived neurotrophic factor (GDNF) protein product
EP0854919A1 (en) 1995-10-10 1998-07-29 Novartis AG Melanoma-associated protein
US5985242A (en) * 1995-10-27 1999-11-16 Praecis Pharmaceuticals, Inc. Modulators of β-amyloid peptide aggregation comprising D-amino acids
US5750361A (en) * 1995-11-02 1998-05-12 The Regents Of The University Of California Formation and use of prion protein (PRP) complexes
ES2203722T3 (en) 1995-11-10 2004-04-16 Elan Corporation, Plc PEPTIDES THAT FACILITATE TRANSPORTATION THROUGH FABRICS AND METHODS OF IDENTIFYING AND USING THEM.
WO1997018855A1 (en) 1995-11-21 1997-05-29 Eduard Naumovich Lerner Device for enhanced delivery of biologically active substances and compounds in an organism
AU1072897A (en) 1995-12-12 1997-07-03 Karolinska Innovations Ab Peptide binding the klvff-sequence of amyloid beta
JPH09178743A (en) 1995-12-27 1997-07-11 Oriental Yeast Co Ltd Determinationof soluble app
US6015662A (en) * 1996-01-23 2000-01-18 Abbott Laboratories Reagents for use as calibrators and controls
ZA97452B (en) * 1996-01-25 1997-08-15 Trinity College Dublin Streptococcus equi vaccine.
US5770700A (en) * 1996-01-25 1998-06-23 Genetics Institute, Inc. Liquid factor IX formulations
WO1997026913A1 (en) 1996-01-26 1997-07-31 The Trustees Of Columbia University In The City Of New York A POLYPEPTIDE FROM LUNG EXTRACT WHICH BINDS AMYLOID-β PEPTIDE
GB9602294D0 (en) * 1996-02-05 1996-04-03 Zeneca Ltd Heterocyclic compounds
US6096313A (en) 1996-02-09 2000-08-01 Ludwig Institute For Cancer Research Compositions containing immunogenic molecules and granulocyte-macrophage colony stimulating factor, as an adjuvant
WO1997032017A1 (en) 1996-02-26 1997-09-04 Morphosys Gesellschaft Für Proteinoptimierung Mbh Novel method for the identification of nucleic acid sequences encoding two or more interacting (poly)peptides
US6150091A (en) * 1996-03-06 2000-11-21 Baylor College Of Medicine Direct molecular diagnosis of Friedreich ataxia
US5870587A (en) * 1996-03-20 1999-02-09 International Business Machines Corporation Information-handling system, method, and article of manufacture including a mechanism for providing an improved application binary interface
DE69731357T2 (en) 1996-03-23 2006-02-02 The Research Foundation For Microbial Diseases Of Osaka University, Suita FUNCTIONAL ANTIGEN FRAGMENT OF TETANUS TOXIN AND TETANUS VACCINE
BR9708401A (en) * 1996-03-29 2000-01-04 Univ Otago Para-smallpox Virus Vectors
WO1997036601A1 (en) * 1996-04-03 1997-10-09 Anergen, Inc. Cyclic peptide vaccines for treatment and prevention of diabetes
AU735733B2 (en) * 1996-04-19 2001-07-12 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Antigenically reactive regions of the hepatitis A virus polyprotein
US6284533B1 (en) * 1996-05-01 2001-09-04 Avant Immunotherapeutics, Inc. Plasmid-based vaccine for treating atherosclerosis
JP2000516452A (en) 1996-07-16 2000-12-12 プリュックテュン,アンドレアス Immunoglobulin superfamily domains and fragments with increased solubility
CA2262006A1 (en) 1996-07-26 1998-02-05 Sloan-Kettering Institute For Cancer Research Method and reagents for genetic immunization
US7147851B1 (en) 1996-08-15 2006-12-12 Millennium Pharmaceuticals, Inc. Humanized immunoglobulin reactive with α4β7 integrin
CA2183901A1 (en) 1996-08-22 1998-02-23 Johanna E. Bergmann Targets for therapy and diagnosis of alzheimer's disease and down syndrome in humans
GB9617616D0 (en) 1996-08-22 1996-10-02 Osteometer Biotech As Assaying protein fragments in body fluids
PT929574E (en) 1996-08-27 2005-11-30 Praecis Pharm Inc MODULATORS OF BETA-AMYLOID PEPTIDES COMPREHENDING D-AMINOACIDES
US6057367A (en) * 1996-08-30 2000-05-02 Duke University Manipulating nitrosative stress to kill pathologic microbes, pathologic helminths and pathologically proliferating cells or to upregulate nitrosative stress defenses
US6797495B2 (en) * 1996-11-05 2004-09-28 The Regents Of The University Of California Somatic cells with ablated PrP gene and methods of use
US6022859A (en) * 1996-11-15 2000-02-08 Wisconsin Alumni Research Foundation Inhibitors of β-amyloid toxicity
WO1998022120A1 (en) 1996-11-19 1998-05-28 The Wistar Institute Of Anatomy & Biology Diagnostic and therapeutic reagents for alzheimer's disease
US6962984B2 (en) 1996-12-05 2005-11-08 Nihon University IgA nephropathy-related DNA
US20030068316A1 (en) * 1997-02-05 2003-04-10 Klein William L. Anti-ADDL antibodies and uses thereof
US6218506B1 (en) 1997-02-05 2001-04-17 Northwestern University Amyloid β protein (globular assembly and uses thereof)
EA199900758A1 (en) 1997-03-03 2000-04-24 Бёрингер Ингельхайм Фармасьютиклс, Инк. SMALL MOLECULES USEFUL AT TREATMENT OF INFLAMMATORY DISEASES
US6277375B1 (en) * 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US5798102A (en) 1997-03-04 1998-08-25 Milkhaus Laboratory, Inc. Treatment of cardiomyopathy
US6611610B1 (en) * 1997-04-02 2003-08-26 Gentex Corporation Vehicle lamp control
US6057098A (en) * 1997-04-04 2000-05-02 Biosite Diagnostics, Inc. Polyvalent display libraries
NZ337765A (en) 1997-04-09 2001-09-28 Mindset Biopharmaceuticals Usa Recombinant antibodies having specificity for beta-amyloid N-terminus and C-terminus and use in treating Alzheimer's Disease
US20020086847A1 (en) * 1997-04-09 2002-07-04 Mindset Biopharmaceuticals (Usa) Recombinant antibodies specific for beta-amyloid ends, DNA encoding and methods of use thereof
US8173127B2 (en) 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
EE9900461A (en) 1997-04-15 2000-06-15 Farmaceutisk Laboratorium Ferring A/S Modified TNFα molecules, DNA encoding them, and vaccines containing these TNFα molecules and DNA
US6787319B2 (en) * 1997-04-16 2004-09-07 American Home Products Corp. β-amyloid peptide-binding proteins and polynucleotides encoding the same
US5858205A (en) * 1997-05-13 1999-01-12 Uop Llc Multizone catalytic reforming process
WO1998052581A1 (en) * 1997-05-20 1998-11-26 Ottawa Civic Hospital Loeb Research Institute Vectors and methods for immunization or therapeutic protocols
WO1998056418A1 (en) 1997-06-13 1998-12-17 Genentech, Inc. Stabilized antibody formulation
WO1999000150A2 (en) 1997-06-27 1999-01-07 Regents Of The University Of California Drug targeting of a peptide radiopharmaceutical through the primate blood-brain barrier in vivo with a monoclonal antibody to the human insulin receptor
IT1293511B1 (en) 1997-07-30 1999-03-01 Gentili Ist Spa MONOCLONAL CATALYTIC ANTIBODIES WITH PROTEASIC ACTIVITY FOR THE SELECTIVE LYSIS OF THE PROTEIN COMPONENT OF PLATES AND RELATED AGGREGATES
WO1999006545A2 (en) 1997-08-01 1999-02-11 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Composition and method for the detection of diseases associated with amyloid-like fibril or protein aggregate formation
EP1005569A2 (en) 1997-08-01 2000-06-07 MorphoSys AG Novel method and phage for the identification of nucleic acid sequences encoding members of a multimeric (poly)peptide complex
ES2500490T3 (en) 1997-08-29 2014-09-30 Antigenics Inc. Compositions comprising adjuvant QS-21 and polysorbate or cyclodextrin as excipient
US6175057B1 (en) * 1997-10-08 2001-01-16 The Regents Of The University Of California Transgenic mouse model of alzheimer's disease and cerebral amyloid angiopathy
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US6913745B1 (en) * 1997-12-02 2005-07-05 Neuralab Limited Passive immunization of Alzheimer's disease
US6905686B1 (en) * 1997-12-02 2005-06-14 Neuralab Limited Active immunization for treatment of alzheimer's disease
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US6923964B1 (en) 1997-12-02 2005-08-02 Neuralab Limited Active immunization of AScr for prion disorders
US7588766B1 (en) * 2000-05-26 2009-09-15 Elan Pharma International Limited Treatment of amyloidogenic disease
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US6787523B1 (en) * 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6761888B1 (en) 2000-05-26 2004-07-13 Neuralab Limited Passive immunization treatment of Alzheimer's disease
US6710226B1 (en) * 1997-12-02 2004-03-23 Neuralab Limited Transgenic mouse assay to determine the effect of Aβ antibodies and Aβ Fragments on alzheimer's disease characteristics
US6750324B1 (en) * 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US20080050367A1 (en) * 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
BR9815345A (en) 1997-12-03 2000-11-21 Fujisawa Pharmaceutical Co Soft composition of pelleted drug, inhaler using it and method for its manufacture
JP2002511385A (en) 1997-12-03 2002-04-16 ニューララブ リミテッド Method for suppressing β-amyloid-related changes in Alzheimer's disease
FR2777015B3 (en) 1998-02-23 2000-09-15 Financ De Biotechnologie METHOD AND MEANS FOR OBTAINING CELLULAR AND ANIMAL MODELS OF NEURODEGENERATIVE DISEASES
US6528624B1 (en) * 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6194551B1 (en) * 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US20050059591A1 (en) * 1998-04-07 2005-03-17 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050059802A1 (en) * 1998-04-07 2005-03-17 Neuralab Ltd Prevention and treatment of amyloidogenic disease
CA2327505A1 (en) * 1998-04-28 1999-11-04 Smithkline Beecham Corporation Monoclonal antibodies with reduced immunogenicity
NO314086B1 (en) 1998-05-08 2003-01-27 Gemvax As Peptides and pharmaceutical compositions containing them, nucleic acid sequences encoding such peptides, plasmids and virus vectors encompassing such DNA sequences and their use for the preparation of pharmaceutical preparations for
JP2003532618A (en) 1998-05-19 2003-11-05 イエダ リサーチ アンド デベロプメント カンパニイ リミテッド Cells, nervous system-specific antigens and their uses
US20030147882A1 (en) * 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
WO1999060024A1 (en) 1998-05-21 1999-11-25 The University Of Tennessee Research Corporation Methods for amyloid removal using anti-amyloid antibodies
US6432710B1 (en) 1998-05-22 2002-08-13 Isolagen Technologies, Inc. Compositions for regenerating tissue that has deteriorated, and methods for using such compositions
US6710228B1 (en) * 1998-05-29 2004-03-23 Mycogen Corporation Cotton cells, plants, and seeds genetically engineered to express insecticidal and fungicidal chitin binding proteins (lectins)
US6727349B1 (en) * 1998-07-23 2004-04-27 Millennium Pharmaceuticals, Inc. Recombinant anti-CCR2 antibodies and methods of use therefor
PT1117421E (en) 1998-10-05 2004-11-30 Pharmexa A S NEW METHODS FOR THERAPEUTIC VACCINATION
WO2000023082A1 (en) * 1998-10-19 2000-04-27 Yeda Research And Development Co. Ltd. Treatment of systemic lupus erythematosus by down-regulating the autoimmune response to autoantigens
US7112661B1 (en) 1998-10-30 2006-09-26 The Research Foundation Of State University Of New York Variable heavy chain and variable light chain regions of antibodies to human platelet glycoprotein Ib alpha
GB2348203B (en) 1998-11-04 2002-06-19 Imp College Innovations Ltd Solube beta-forms of prion proteins, methods of preparation and use
EP1148891B1 (en) 1999-01-19 2004-03-17 PHARMACIA &amp; UPJOHN COMPANY Method of packaging an oxidation sensitive medicinal substance
EP1146898A1 (en) 1999-01-22 2001-10-24 Matthew John During Vaccine-mediated treatment of neurological disorders
US7629311B2 (en) 1999-02-24 2009-12-08 Edward Lewis Tobinick Methods to facilitate transmission of large molecules across the blood-brain, blood-eye, and blood-nerve barriers
US7282570B2 (en) * 1999-04-20 2007-10-16 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
CA2388092A1 (en) 1999-05-05 2000-11-16 Neurochem, Inc. Stereoselective antifibrillogenic peptides and peptidomimetics thereof
US6787637B1 (en) * 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
PE20010212A1 (en) 1999-06-01 2001-02-22 Neuralab Ltd COMPOSITIONS OF THE A-BETA PEPTIDE AND PROCESSES TO PRODUCE THEM
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
PT1409654E (en) * 1999-06-16 2008-11-25 Boston Biomedical Res Inst Immunological control of beta-amyloid levels in vivo
EP1192462A1 (en) 1999-07-01 2002-04-03 Scios Inc. Prevention and treatment of amyloid-associated disorders
IL147599A0 (en) 1999-07-15 2002-08-14 Genetics Inst Formulations for il-11
WO2001010900A2 (en) 1999-08-04 2001-02-15 University Of Southern California Globular assembly of amyloid beta protein and uses thereof
JP2003509020A (en) 1999-09-03 2003-03-11 ラモット・ユニバーシティ・オーソリティ・フォー・アプライド・リサーチ・アンド・インダストリアル・ディベロップメント・リミテッド Pharmaceuticals, compositions, and uses thereof useful for diagnosis, treatment, and prevention of plaque forming diseases
US6294171B2 (en) * 1999-09-14 2001-09-25 Milkhaus Laboratory, Inc. Methods for treating disease states comprising administration of low levels of antibodies
US6824780B1 (en) 1999-10-29 2004-11-30 Genentech, Inc. Anti-tumor antibody compositions and methods of use
CN1433321A (en) 1999-11-29 2003-07-30 尼奥切姆公司 Vaccine for prevention and treatment of Alzhermer's and amyloid related diseases
US20020094335A1 (en) * 1999-11-29 2002-07-18 Robert Chalifour Vaccine for the prevention and treatment of alzheimer's and amyloid related diseases
JP4804690B2 (en) 1999-12-08 2011-11-02 インテレクト・ニューロサイエンシズ・インコーポレーテッド Chimeric peptide as immunogen, antibody thereto, and immunization using chimeric peptide or antibody
US6399314B1 (en) * 1999-12-29 2002-06-04 American Cyanamid Company Methods of detection of amyloidogenic proteins
KR100879810B1 (en) 2000-02-21 2009-01-22 하. 룬드벡 아크티에셀스카브 Novel method for down-regulation of amyloid
HUP0300067A3 (en) 2000-02-21 2010-03-29 Lundbeck & Co As H Novel method for down-regulation of amyloid
IL151378A0 (en) 2000-02-24 2003-04-10 Univ Washington Humanized antibodies that sequester amyloid beta peptide
US6294221B1 (en) * 2000-03-08 2001-09-25 E. I. Du Pont De Nemours And Company Process for spray-coating with frequent changes between aqueous and non-aqueous coating agents inside a spray-coating chamber
US6548840B1 (en) * 2000-04-03 2003-04-15 Hrl Laboratories, Llc Monolithic temperature compensation scheme for field effect transistor integrated circuits
WO2001077167A2 (en) 2000-04-05 2001-10-18 University Of Tennessee Research Corporation Methods of investigating, diagnosing, and treating amyloidosis
DE60134499D1 (en) 2000-04-13 2008-07-31 Corixa Corp IMMUNOSTIMULATING COMBINATIONS CONTAINING AMINO ALKYL GLUCOSAMINE IDEPHOSPHATE AND QS-21
US6713450B2 (en) * 2000-05-22 2004-03-30 New York University Synthetic immunogenic but non-amyloidogenic peptides homologous to amyloid β for induction of an immune response to amyloid β and amyloid deposits
CA2414772C (en) * 2000-07-07 2011-06-28 Jan Naslund Prevention and treatment of alzheimer's disease
US20020009445A1 (en) * 2000-07-12 2002-01-24 Yansheng Du Human beta-amyloid antibody and use thereof for treatment of alzheimer's disease
EP1172378A1 (en) 2000-07-12 2002-01-16 Richard Dr. Dodel Human beta-amyloid antibody and use thereof for treatment of alzheimer's disease
US20030092145A1 (en) * 2000-08-24 2003-05-15 Vic Jira Viral vaccine composition, process, and methods of use
US7199102B2 (en) * 2000-08-24 2007-04-03 The Regents Of The University Of California Orally administered peptides synergize statin activity
PT1317479E (en) 2000-09-06 2009-10-29 Univ Pasteur Methods and compositions for diseases associated with amyloidosis
IT1319277B1 (en) 2000-10-24 2003-09-26 Chiesi Farma Spa MELTING PROTEINS USEFUL FOR ALZHEIMER'S MILK IMMUNIZATION TREATMENT.
IL139308A0 (en) 2000-10-26 2001-11-25 Marikovsky Moshe Peptides from amyloid precursor protein which inhibit tumor growth and metastasis
US7668586B2 (en) * 2000-11-02 2010-02-23 Cornell Research Foundation, Inc. In vivo multiphoton diagnostic detection and imaging of a neurodegenerative disease
WO2002042462A2 (en) * 2000-11-27 2002-05-30 Praecis Pharmaceuticals Inc. Therapeutic agents and methods of use thereof for treating an amyloidogenic disease
PE20020574A1 (en) * 2000-12-06 2002-07-02 Wyeth Corp HUMANIZED ANTIBODIES THAT RECOGNIZE THE AMYLOID PEPTIDE BETA
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
WO2002060920A2 (en) * 2000-12-27 2002-08-08 Board Of Regents, University Of Texas System Prion isomers, methods of making, methods of using, and compositions and products comprising prion isomers
US20020160394A1 (en) * 2001-01-24 2002-10-31 Bayer Corporation Regulation of transthyretin to treat obesity
DE60121729T2 (en) * 2001-04-19 2007-11-29 Dr. Hermann Schätzl Prion protein dimers for vaccinations
EP2165714B1 (en) 2001-04-30 2013-10-23 Eli Lilly And Company Humanized antibodies recognizing the beta-amyloid peptide
ES2312569T3 (en) 2001-04-30 2009-03-01 Eli Lilly And Company HUMANIZED ANTIBODIES.
US6524624B1 (en) * 2001-05-16 2003-02-25 Alcide Corporation Two-part disinfecting systems and compositions and methods related thereto
US6906169B2 (en) 2001-05-25 2005-06-14 United Biomedical, Inc. Immunogenic peptide composition comprising measles virus Fprotein Thelper cell epitope (MUFThl-16) and N-terminus of β-amyloid peptide
GB0113179D0 (en) 2001-05-31 2001-07-25 Novartis Ag Organic compounds
US6888849B2 (en) * 2001-06-15 2005-05-03 Lucent Technologies Inc. Method for evaluating capacity utilization of a terminus in a communication system
WO2003000714A2 (en) 2001-06-22 2003-01-03 Panacea Pharmaceuticals, Inc. Compositions and methods for preventing protein aggregation in neurodegenerative diseases
JP4317010B2 (en) 2001-07-25 2009-08-19 ピーディーエル バイオファーマ,インコーポレイティド Stable lyophilized pharmaceutical formulation of IgG antibody
US20030135035A1 (en) 2001-08-09 2003-07-17 Mark Shannon Human ZZAP1 protein
EP1519740A4 (en) 2001-08-17 2005-11-09 Lilly Co Eli Rapid improvement of cognition in conditions related to a-beta
WO2003016467A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company Use of antibodies having high affinity for soluble ass to treat conditions and diseases related to ass
WO2003016466A2 (en) 2001-08-17 2003-02-27 Eli Lilly And Company ANTI-Aβ ANTIBODIES
US20030082191A1 (en) 2001-08-29 2003-05-01 Poduslo Joseph F. Treatment for central nervous system disorders
JP4411068B2 (en) 2001-09-10 2010-02-10 アンチキャンサー インコーポレーテッド Improved resolution of tumor metastasis
US6736142B2 (en) * 2001-09-13 2004-05-18 Gines Sanchez Gomez Protective tube and harness
US6907297B2 (en) 2001-09-28 2005-06-14 Ethicon, Inc. Expandable intracardiac return electrode and method of use
US6597198B2 (en) * 2001-10-05 2003-07-22 Intel Corporation Current mode bidirectional port with data channel used for synchronization
GB0124446D0 (en) * 2001-10-11 2001-12-05 Short Brothers Ltd Aircraft propulsive power unit
US7781413B2 (en) 2001-10-31 2010-08-24 Board Of Regents, The University Of Texas System SEMA3B inhibits tumor growth and induces apoptosis in cancer cells
CA2466034C (en) 2001-11-08 2012-12-18 Protein Design Labs, Inc. Stable aqueous pharmaceutical formulations of daclizumab antibodies
EP1572894B1 (en) * 2001-11-21 2016-04-13 New York University Synthetic immunogenic but non-deposit-forming polypeptides and peptides homologous to amyloid beta, prion protein, amylin, alpha synuclein, or polyglutamine repeats for induction of an immune response thereto
WO2003051374A2 (en) 2001-12-17 2003-06-26 New York State Office Of Mental Health SEQUESTRATION OF Aβ IN THE PERIPHERY IN THE ABSENCE OF IMMUNOMODULATING AGENT AS A THERAPEUTIC APPROACH FOR THE TREATMENT OR PREVENTION OF BETA-AMYLOID RELATED DISEASES
AR038568A1 (en) 2002-02-20 2005-01-19 Hoffmann La Roche ANTI-A BETA ANTIBODIES AND ITS USE
AU2003224624B2 (en) 2002-02-21 2008-08-28 Duke University Reagents and treatment methods for autoimmune diseases
US6688651B2 (en) * 2002-02-21 2004-02-10 Dmt Co., Ltd. Device for locking cap nut for coupling
MY139983A (en) * 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
CA2390700C (en) * 2002-06-11 2004-09-07 Henry Tsang Illuminated soap bar with sound
GB0213437D0 (en) 2002-06-12 2002-07-24 Univ Cranfield Temporary tattoos: A novel vehicle for skin testing
US6892535B2 (en) * 2002-06-14 2005-05-17 Volvo Construction Equipment Holding Sweden Ab Hydraulic circuit for boom cylinder combination having float function
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
MXPA05000819A (en) 2002-07-19 2005-08-29 Cytos Biotechnology Ag Vaccine compositions containing amyloid beta1-6 antigen arrays.
AU2003250102B2 (en) 2002-07-24 2009-09-17 Innogenetics N.V. Fragments of beta-amyloid as targets for vaccination against alzheimer disease
EP1545582A4 (en) 2002-10-01 2008-09-17 Univ Northwestern Amyloid beta-derived diffusible ligands (addls), addl-surrogates, addl-binding molecules, and uses thereof
US20040080762A1 (en) * 2002-10-25 2004-04-29 Xerox Corporation Half-tone based enhancement of black
US20060019850A1 (en) * 2002-10-31 2006-01-26 Korzenski Michael B Removal of particle contamination on a patterned silicon/silicon dioxide using dense fluid/chemical formulations
FR2846667B1 (en) 2002-11-06 2004-12-31 Pasteur Institut VARIABLE FRAGMENTS OF SINGLE-CHAIN CAMELIDE ANTIBODIES DIRECTED AGAINST BETA-AMYLOID PEPTIDE 1-42 AND THEIR APPLICATIONS FOR DIAGNOSIS AND TREATMENT OF NEUROAGREGATIVE DISEASES
AU2003293543A1 (en) 2002-12-13 2004-07-09 Abgenix, Inc. System and method for stabilizing antibodies with histidine
US6787129B1 (en) 2003-01-13 2004-09-07 Zenitech Llc Castor polyester as gloss agents in anionic systems
CA2513722A1 (en) 2003-02-01 2004-08-19 Neuralab Limited Active immunization to generate antibodies to soluble a-beta
ATE468886T1 (en) 2003-02-10 2010-06-15 Applied Molecular Evolution ABETA-BINDING MOLECULES
ZA200507757B (en) 2003-04-04 2007-01-31 Genentech Inc High concentration antibody and protein formulations
EP1480041A1 (en) 2003-05-22 2004-11-24 Innogenetics N.V. Method for the prediction, diagnosis and differential diagnosis of Alzheimer's disease
TWI374893B (en) * 2003-05-30 2012-10-21 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
US20060182321A1 (en) 2003-07-07 2006-08-17 Agency For Science, Technology And Research Method and apparatus for extracting third ventricle information
WO2005014041A2 (en) 2003-07-24 2005-02-17 Novartis Ag Use of an amyloid beta dna vaccine for the treatment and/or prevention of amyloid diseases
US20060233788A1 (en) 2003-09-05 2006-10-19 Heiman Mark L Anti-ghrelin antibodies
CA2445743A1 (en) 2003-10-08 2005-04-08 The University Of British Columbia Methods for modulating neuronal responses
WO2005035753A1 (en) 2003-10-10 2005-04-21 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
HUE026000T2 (en) 2003-12-17 2016-04-28 Wyeth Llc Immunogenic peptide carrier conjugates and methods of producing same
EP2336147B1 (en) 2003-12-17 2014-04-30 Janssen Alzheimer Immunotherapy A beta immunogenic peptide carrier conjugates and methods of producing same
US20050214222A1 (en) 2004-02-13 2005-09-29 Mckinnon Stuart J In vivo imaging of amyloid plaques in glaucoma using intravenous injectable dyes
ES2367576T3 (en) 2004-03-15 2011-11-04 Janssen Pharmaceutica Nv NEW COMPOUNDS AS MODULATORS OF OPIOID RECEPTORS.
JP2008505115A (en) 2004-07-02 2008-02-21 ユニバーシティー オブ ピッツバーグ Amyloid imaging as a surrogate marker for the effectiveness of anti-amyloid therapy
KR20070040824A (en) * 2004-07-30 2007-04-17 리나트 뉴로사이언스 코퍼레이션 Antibodies directed against amyloid-beta peptide and methods using same
US7328838B2 (en) * 2004-09-09 2008-02-12 Igt Counterfeit cashless instrument detection methods and systems
CN101035564A (en) 2004-09-10 2007-09-12 惠氏公司 Humanized anti-5T4 antibodies and anti-5T4 antibody/calicheamicin conjugates
US7807804B2 (en) 2004-10-05 2010-10-05 Wyeth Llc Methods and compositions for improving recombinant protein production
WO2006047670A2 (en) 2004-10-26 2006-05-04 Wyeth Methods for assessing antibodies to neurodegenerative disease-associated antigens
US20060026911A1 (en) * 2004-11-18 2006-02-09 Sutton Adam F Footer track with moisture vent
EP1838854B1 (en) 2004-12-15 2012-10-31 Janssen Alzheimer Immunotherapy Antibodies that recognize Beta Amyloid Peptide
US20060240486A1 (en) 2004-12-15 2006-10-26 Johnson-Wood Kelly L Immunoprecipitation-based assay for predicting in vivo efficacy of beta-amyloid antibodies
AR052051A1 (en) 2004-12-15 2007-02-28 Neuralab Ltd AB HUMANIZED ANTIBODIES USED TO IMPROVE COGNITION
AR051800A1 (en) 2004-12-15 2007-02-07 Wyeth Corp BETA ANTIBODIES USED TO IMPROVE COGNITION
US20060153772A1 (en) 2004-12-15 2006-07-13 Wyeth Contextual fear conditioning for predicting immunotherapeutic efficacy
DOP2006000022A (en) 2005-01-28 2006-08-15 Wyeth Corp STABILIZED LIQUID FORMULATIONS OF POLYPTIDES
GT200600031A (en) 2005-01-28 2006-08-29 ANTI-BETA ANTIBODY FORMULATION
EP1879613B1 (en) 2005-05-05 2011-11-30 Merck Sharp & Dohme Corp. Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
CA2611815A1 (en) * 2005-06-17 2006-12-28 Elan Pharma International Limited Methods of purifying anti a beta antibodies
CN101238124A (en) * 2005-07-18 2008-08-06 默克公司 Spiropiperidine beta-secretase inhibitors for the treatment of Alzheimer's disease
JP2009516654A (en) * 2005-11-10 2009-04-23 ロスキャンプ リサーチ, エルエルシー Regulation of angiogenesis by A-beta peptide
AU2007233831B2 (en) 2006-03-30 2013-02-14 Glaxo Group Limited Antibodies against amyloid-beta peptide
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2010044803A1 (en) 2008-10-17 2010-04-22 Elan Pharma International Limited Treatment of amyloidogenic diseases
SG173385A1 (en) 2006-07-14 2011-08-29 Ac Immune S A Ch Humanized antibody against amyloid beta
CA2680762A1 (en) 2007-03-12 2008-09-25 National Institute Of Radiological Sciences Pet visualization of amyloid-associated neuroinflammation in the brain
WO2008131298A2 (en) 2007-04-18 2008-10-30 Elan Pharma International Limited Prevention and treatment of cerebral amyloid angiopathy
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
JP5889529B2 (en) 2007-07-27 2016-03-22 ヤンセン・サイエンシズ・アイルランド・ユーシー Treatment of amyloidogenic diseases
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
CA2710984C (en) * 2007-12-28 2018-05-29 Elan Pharmaceuticals, Inc. Treatment and prophylaxis of amyloidosis
TR201816335T4 (en) 2008-09-18 2018-11-21 Cedars Sinai Medical Center Optical method for the detection of Alzheimer's disease.
EP2538982A4 (en) 2010-02-25 2016-02-17 Janssen Alzheimer Immunotherap Pet monitoring of ab-directed immunotherapy
EP2560681A4 (en) 2010-04-22 2013-09-25 Janssen Alzheimer Immunotherap Use of tau to monitor immunotherapy

Also Published As

Publication number Publication date
US20050163788A1 (en) 2005-07-28
US20120276116A1 (en) 2012-11-01
NO20092874L (en) 2000-07-31
US8034348B2 (en) 2011-10-11
BG104562A (en) 2001-01-31
EP2305282A2 (en) 2011-04-06
US20050249727A1 (en) 2005-11-10
HU228061B1 (en) 2012-09-28
EP1033996A4 (en) 2004-05-26
DE06075479T1 (en) 2006-11-16
NZ504569A (en) 2005-01-28
AR020050A1 (en) 2002-04-10
PL202698B1 (en) 2009-07-31
WO1999027944A1 (en) 1999-06-10
NO331425B1 (en) 2011-12-27
TWI239847B (en) 2005-09-21
GEP20043319B (en) 2004-05-10
HRP20090568A2 (en) 2010-06-30
EP1033996A1 (en) 2000-09-13
US20040170641A1 (en) 2004-09-02
US20050013815A1 (en) 2005-01-20
KR20090078366A (en) 2009-07-17
JP2002502802A (en) 2002-01-29
NO20100061L (en) 2000-07-31
EP1679080A3 (en) 2006-07-19
GEP20053715B (en) 2005-12-26
HRP20000443A2 (en) 2000-10-31
DE06075704T1 (en) 2006-12-28
KR20010032635A (en) 2001-04-25
DE69840922D1 (en) 2009-07-30
ATE399016T1 (en) 2008-07-15
DK2305282T3 (en) 2013-10-07
EA200601132A1 (en) 2007-02-27
JP2006131639A (en) 2006-05-25
WO1999027944A9 (en) 1999-09-23
HK1095265A1 (en) 2007-05-04
HRP20000443B1 (en) 2009-11-30
ES2262460T1 (en) 2006-12-01
IL205298A0 (en) 2011-07-31
CA2312920A1 (en) 1999-06-10
EE05377B1 (en) 2011-02-15
EE200000379A (en) 2001-04-16
KR100936419B1 (en) 2010-01-12
US20040228865A1 (en) 2004-11-18
SI1994937T1 (en) 2011-04-29
CZ20001706A3 (en) 2000-11-15
DK1679080T3 (en) 2009-09-28
US20060034858A1 (en) 2006-02-16
HU228493B1 (en) 2013-03-28
DE69839647D1 (en) 2008-08-07
US20040171815A1 (en) 2004-09-02
SI1679080T1 (en) 2009-12-31
HUP0100627A3 (en) 2006-03-28
EP2305282A3 (en) 2011-05-18
US20080096818A1 (en) 2008-04-24
NO20002784D0 (en) 2000-05-31
US20060029611A1 (en) 2006-02-09
US20050037026A1 (en) 2005-02-17
EA009283B1 (en) 2007-12-28
KR20120135915A (en) 2012-12-17
CY1108331T1 (en) 2014-02-12
DK1994937T3 (en) 2011-02-07
EP1679080A2 (en) 2006-07-12
JP4677094B2 (en) 2011-04-27
US20140227274A1 (en) 2014-08-14
IL193245A0 (en) 2009-09-22
PT1033996E (en) 2008-10-02
CN1281366A (en) 2001-01-24
MY134906A (en) 2007-12-31
US20050048049A1 (en) 2005-03-03
PL202706B1 (en) 2009-07-31
US20050191292A1 (en) 2005-09-01
EP1994937B1 (en) 2010-12-29
TR200001608T2 (en) 2001-07-23
US20080227718A1 (en) 2008-09-18
NO20002784L (en) 2000-07-31
SG111083A1 (en) 2005-05-30
HK1094535A1 (en) 2007-04-04
PL342649A1 (en) 2001-06-18
US20080227719A1 (en) 2008-09-18
DE1033996T1 (en) 2001-06-07
CN100374151C (en) 2008-03-12
CY1109368T1 (en) 2014-07-02
AU1706199A (en) 1999-06-16
US6946135B2 (en) 2005-09-20
IL136252A (en) 2010-11-30
KR101248711B1 (en) 2013-03-28
JP2010215651A (en) 2010-09-30
US20050031629A1 (en) 2005-02-10
EP2305282B1 (en) 2013-07-10
ATE435659T1 (en) 2009-07-15
PT2305282E (en) 2013-09-27
NO332813B1 (en) 2013-01-21
EP1994937A2 (en) 2008-11-26
US20050142132A1 (en) 2005-06-30
US20050019330A1 (en) 2005-01-27
HUP0100627A1 (en) 2001-06-28
EP1690547A1 (en) 2006-08-16
US20050191314A1 (en) 2005-09-01
EA200401473A1 (en) 2005-04-28
SI1033996T1 (en) 2009-02-28
SA99191269B1 (en) 2006-03-15
EA200000608A1 (en) 2000-12-25
PE20001383A1 (en) 2000-11-25
EP1994937A3 (en) 2008-12-24
EP1033996B1 (en) 2008-06-25
SI2305282T1 (en) 2013-10-30
ES2263408T1 (en) 2006-12-16
CY1111370T1 (en) 2015-08-05
EA007218B1 (en) 2006-08-25
US8642044B2 (en) 2014-02-04
CO4980846A1 (en) 2000-11-27
ES2310017T3 (en) 2008-12-16
JP2006077030A (en) 2006-03-23
US20090191231A1 (en) 2009-07-30
NO328865B1 (en) 2010-06-07
ATE493149T1 (en) 2011-01-15
IL136252A0 (en) 2001-05-20
JP4677334B2 (en) 2011-04-27
IS5500A (en) 2000-05-17
DK1033996T3 (en) 2008-09-22
ES2263408T3 (en) 2009-11-16
US6972127B2 (en) 2005-12-06
ATE433760T1 (en) 2009-07-15
IL191904A0 (en) 2008-12-29
US7014855B2 (en) 2006-03-21
US6982084B2 (en) 2006-01-03
US8034339B2 (en) 2011-10-11
EP1690547B1 (en) 2009-07-08
ES2428740T3 (en) 2013-11-11
CZ303137B6 (en) 2012-04-25
JP4677333B2 (en) 2011-04-27
PT1994937E (en) 2011-02-22
US20130058869A1 (en) 2013-03-07
US20040171816A1 (en) 2004-09-02
ID25504A (en) 2000-10-05
DE69840969D1 (en) 2009-08-20
US20050249725A1 (en) 2005-11-10
DE69842082D1 (en) 2011-02-10
US8535673B2 (en) 2013-09-17
US6808712B2 (en) 2004-10-26
US20050019343A1 (en) 2005-01-27
BR9815357A (en) 2000-10-24
IL191904A (en) 2014-05-28
PT1679080E (en) 2009-09-10
US20050196399A1 (en) 2005-09-08
EP1679080B1 (en) 2009-06-17

Similar Documents

Publication Publication Date Title
CA2312920C (en) Prevention and treatment of amyloidogenic disease
US6866850B2 (en) Prevention and treatment of amyloidogenic disease
AU2006202899B2 (en) Prevention and treatment of amyloidogenic disease
AU2016256668A1 (en) Prevention and treatment of amyloidogenic disease
AU2013206069A1 (en) Prevention and treatment of amyloidogenic disease

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20181130