CA2370311C - Prevention and treatment of amyloidogenic disease - Google Patents

Prevention and treatment of amyloidogenic disease Download PDF

Info

Publication number
CA2370311C
CA2370311C CA2370311A CA2370311A CA2370311C CA 2370311 C CA2370311 C CA 2370311C CA 2370311 A CA2370311 A CA 2370311A CA 2370311 A CA2370311 A CA 2370311A CA 2370311 C CA2370311 C CA 2370311C
Authority
CA
Canada
Prior art keywords
antibody
peptide
antibodies
pbs
beta
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
CA2370311A
Other languages
French (fr)
Other versions
CA2370311A1 (en
Inventor
Dale B. Schenk
Frederique Bard
Nicki J. Vasquez
Theodore Yednock
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=23254226&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2370311(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 CA2370311A1 publication Critical patent/CA2370311A1/en
Application granted granted Critical
Publication of CA2370311C publication Critical patent/CA2370311C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • 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
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • 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
    • 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
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention provides improved agents and methods for treatment of diseases associated with amyloid deposits of A.beta. in the brain of a patient. Such methods entail administering agents that induce a beneficial immunogenic response against the amyloid deposit. The methods are useful for prophylactic and therapeutic treatment of Alzheimer's disease. Preferred agents including N-terminal fragments of A.beta. and antibodies binding to the same.

Description

PREVENTION AND TREATMENT OF AMYLOIDOGENIC DISEASE
TECHNICAL FIELD
The invention resides in the technical fields of immunology and medicine.
BACKGROUND OF THE INVENTION
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, I NeuropathoL Exp. NeuroL 53, 438-447 (1994); Duff et al., Nature 373, 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 at least two types of lesions in the brain, senile plaques and neurofibrillary tangles.
Senile plaques are areas of disorganized neuropil up to 150 IIM across with extracellular amyloid deposits at the center visible by microscopic analysis of sections of brain tissue.
Neurofibrillary tangles are intracellular deposits of microtubule associated tau protein consisting of two filaments twisted about each other in pairs.
The principal constituent of the plaques is a peptide termed Af3 or 13-amyloid peptide. A13 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 diseRsr. 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 Af3, particularly processing of APP to increased amounts of the long form of AP (i.e., A131-42 and Af31-43). Mutations in other genes, such as the presenilin genes, PSI and PS2, are thought indirectly to affect processing of APP to generate increased amounts of long form Ap (see Hardy, TINS 20, 154 (1997)). These observations indicate that Af3, 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 1(12 mg/day) of AP 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 Ap 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 AP and because EP 526,511 does not recommend use of an adjuvant, as an immunostimulant, 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 fragments of Af3 , or antibody to certain epitopes within AP 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 and, in some patients, the cognitive impairment associated with Alzheimer's disease.
This application is related to W099/27944, filed November 30, 1998, USSN 60/067,740, filed December 2, 1997, USSN 60/080,970, filed April 7, 1998, and USSN 09/201,430, filed November 30, 1998, SUMMARY OF THE CLAIMED INVENTION
In one aspect, the invention provides methods of preventing or treating a disease associated with amyloid deposits of AP in the brain of a patient. Such diseases include Alzheimer's disease, Down's syndrome and cognitive impairment. The latter can occur with or without other characteristics of an amyloidogenic disease. Some methods of the invention entail administering an effective dosage of an antibody that specifically binds to a component of an amyloid deposit to the patient. Such methods are particularly useful for preventing or treating Alzheimer's disease in human patients. Some methods entail administering an effective dosage of an antibody that binds to Aft Some methods entail administering an effective dosage of an antibody that specifically binds to an epitope within residues 1-10 of AP. In some methods, the antibody specifically binds to an epitope within residues 1-6 of AP. In some methods, the antibody specifically binds to an epitope within residues 1-5 of AP. In some methods, the antibody specifically binds to an epitope within residues 1-7 of AP. In some methods, the antibody specifically binds to an epitope within residues 3-7 of AP. In some methods, the antibody specifically binds to an epitope within residues 1-3 of AP. In some methods, the antibody specifically binds to an epitope within residues 1-4 of AP. In some methods, the antibody binds to an epitope comprising a free N-terminal residue of AP. In some methods, the antibody binds to an epitope within residues of 1-10 of AB wherein residue 1 and/or residue 7 of AP is aspartic acid. In some methods, the antibody specifically binds to Ap peptide without binding to full-length amyloid precursor protein (APP). In some methods, the isotype of the antibody is human IgGl.
In some methods, the antibody binds to an amyloid deposit in the patient and induces a clearing response against the amyloid deposit. For example, such a clearing response can be effected by Fc receptor mediated phagocytosis.
The methods can be used on both asymptomatic patients and those currently showing symptoms of disease. The antibody used in such methods can be a human, humanized, chimeric or nonhuman antibody and can be monoclonal or polyclonal. In some methods, the antibody is prepared from a human immunized with AP
peptide, which human can be the patient to be treated with antibody.
In some methods, the antibody is administered with a pharmaceutical carrier as a pharmaceutical composition. In some methods, antibody is administered at a dosage of 0.0001 to 100 mg/kg, preferably, at least 1 mg/kg body weight antibody. In some methods, the antibody is administered in multiple dosages over a prolonged period, for example, of at least six months. In some methods, the antibody is administered as a sustained release composition. The antibody can be administered, for example, intraperitoneally, orally, subcutaneously, intracranially, intramuscularly, topically, intranasally or intravenously.
In some methods, the antibody is administered by administering a polynucleotide encoding at least one antibody chain to the patient. The polynucleotide is expressed to produce the antibody chain in the patient. Optionally, the polynucleotide encodes heavy and light chains of the antibody. The polynucleotide is expressed to produce the heavy and light chains in the patient. In some methods, the patient is monitored for level of administered antibody in the blood of the patient.
In another aspect, the invention provides methods of preventing or treating a disease associated with amyloid deposits of AP in the brain of patient. For example, the methods can be used to treat Alzheimer's disease or Down's syndrome or cognitive impairment. Such methods entail administering fragments of AP or analogs thereof eliciting an immunogenic response against certain epitopes within Aft Some methods entail administering to a patient an effective dosage of a polypeptide comprising an N-terminal segment of at least residues 1-5 of AP, the first residue of AP being the N-terminal residue of the polypeptide, wherein the polypeptide is free of a C-terminal segment of AP. Some methods entail administering to a patient an effective dosage of a polypeptide comprising an N-terminal segment of AP, the segment beginning at residue 1-3 of AP and ending at residues 7-11 of AP. Some methods entail administering to a patient an effective dosage of an agent that induces an immunogenic response against an N-terminal segment of AP, the segment beginning at residue 1-3 of AP and ending at residues 7-11 of AP without inducing an immunogenic response against an epitope within residues 12-43 of AP43.
In some of the above methods, the N-terminal segment of AP is linked at its C-terminus to a heterologous polypeptide. In some of the above methods, the N-terminal segment of AP is linked at its N-terminus to a heterologous polypeptide. In some of the above methods, the N-terminal segment of AP is linked at its N and C termini to first and second heterologous polypeptides. In some of the above methods, the N-terminal segment of AP is linked at its N terminus to a heterologous polypeptide, and at its C-terminus to at least one additional copy of the N-terminal segment. In some of the above methods, the heterologous polypeptide and thereby a B-cell response against the N-terminal segment. In some of the above methods, the polypeptide further comprises at least one additional copy of the N-terminal segment. In some of the above methods, the polypeptide comprises from N-terminus to C-terminus, the N-terminal segment of AP, a plurality of additional copies of the N-terminal segment, and the heterologous amino acid segment. In some of the above methods, the N-terminal segment consists of AP1-7. In some of the above methods, the N-terminal segment consists of A133-7.
In some methods, the fragment is free of at least the 5 C-terminal amino acids in Ap43. In some methods, the fragment comprises up to 10 contiguous amino acids from AP. Fragments are typically administered at greater than 10 micrograms per dose per patient.
In some methods, the fragment is administered with an adjuvant that enhances the immune response to the AP peptide. The adjuvant and fragment can be administered in either order or together as a composition. The adjuvant can be, for example, aluminum hydroxide, aluminum phosphate, MPLTM, QS-21 (StimulonTM) or incomplete Freund's adjuvant.
The invention further provides pharmaceutical compositions comprising fragments of AP or other agents eliciting immunogenic response to the same epitopes of AP, such as described above, and an adjuvant. The invention also provides pharmaceutical compositions comprising any of the antibodies described above and a pharmaceutically acceptable carrier.
In another aspect, the invention provides methods of screening an antibody for activity in treating a disease associated with deposits of AP in the brain of a patient (e.g., Alzheimer's disease). Such methods entail contacting the antibody with a polypeptide comprising at least five contiguous amino acids of an N-terminal segment of AP beginning at a residue between 1 and 3 of AI3, the polypeptide being free of a C-terminal segment of Ap. One then determines whether the antibody specifically binds to the polypeptide, specific binding providing an indication that the antibody has activity in treating the disease.
In another aspect, the invention provides methods of screening an antibody for activity in clearing an antigen-associated biological entity. Such methods entail combining the antigen-associated biological entity and the antibody and phagocytic cells bearing Fe receptors in a medium. The amount of the antigen-associated biological entity remaining in the medium is then monitored. A reduction in the amount of the antigen-associated biological entity indicates the antibody has clearing activity against the antigen-associated biological entity. The antigen can be provided as a tissue sample or in isolated form. For example, the antigen can be provided as a tissue sample from the brain of an Alzheimer's disease patient or a mammal animal having Alzheimer's pathology.
Other tissue samples against which antibodies can be tested for clearing activity include cancerous tissue samples, virally infected tissue samples, tissue samples comprising inflammatory cells, nonmalignant abnormal cell growths, or tissue samples comprising an abnormal extracellular matrix.
In another aspect, the invention provides methods of detecting an amyloid deposit in a patient. Such methods entail administering to the patient an antibody that specifically binds to an epitope within amino acids 1-10 of AP, and detecting presence of the antibody in the brain of the patient. In some methods, the antibody binds to an epitope within residues 4-10 of AP. In some methods, the antibody is labelled with a paramagnetic label and detected by nuclear magnetic resonance tomography.
The invention further provides diagnostic kits suitable for use in the above methods. Such a kit comprises an antibody that specifically binds to an epitope with residues 1-10 of A. Some kits bear a label describing use of the antibody for in vivo diagnosis or monitoring of Alzheimer's disease.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1: Antibody titer after injection of transgenic mice with AP1-42.
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 AP-specific monoclonal antibody 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 monoclonal 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.
Fig. 5: Geometric mean antibody titers to AP1-42 following immunization with a range of eight doses of AN1792 containing 0.14, 0.4, 1.2, 3.7, 11,33, 100, or 300g.
Fig. 6: Kinetics of antibody response to AN1792 immunization. Titers are expressed as geometric means of values for the 6 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 Proliferation Assay on spleen cells from AN1792-treated (upper panel) or PBS-treated (lower panel).
Fig. 11: Total AP levels in the cortex. A scatterplot of individual Ap profiles in mice immunized with AP or APP derivatives combined with Freund' adjuvant.
Fig. 12: Amyloid burden in the cortex was determined by quantitative image analysis of immunoreacted brain sections for mice immunized with the A13 peptide conjugates A131-5, Ap1-12, and A1313-28; the full length AP aggregates AN1792 (A131-42) and AN1528 (A131-40) and the PBS-treated control group.
Fig. 13: Geometric mean titers of AP-specific antibody for groups of mice immunized with AP or APP derivatives combined with Freund's adjuvant.
Fig. 14: Geometric mean titers of AP-specific antibody for groups of guinea pigs immunized with AN1792, or a palmitoylated derivative thereof, combined with various adjuvants.
Fig: 15(A-E): Ap levels in the cortex of 12-month old PDAPP mice treated with AN1792 or AN1528 with different adjuvants.
Fig. 16: Mean titer of mice treated with polyclonal antibody to Af3.
Fig. 17: Mean titer of mice treated with monoclonal antibody 10D5 to A13.
Fig. 18: Mean titer of mice treated with monoclonal antibody 2F12 to AP.
Fig. 19: Epitope Map: Restricted N¨terminal Response. Day 175 serum from cynomolgus monkeys was tested by ELISA against a series of 10¨mer overlapping CA 02370311 2002-05-13 , peptides (SEQ ID Nos:1-41) covering the complete AN1792 sequence. Animal nuniber F10920M shows a representative N-terminal restricted response to the peptide DAEFRHDSGY (SEQ ID NO:9) which covers amino acids 1-10 of the AN1792 peptide which was used as immunizing antigen.
Fig. 20: Epitope Map: Non¨restricted N--terminal response. Day 175 serum from cynomolgus monkeys was tested by ELISA against a series of 10-mer overlapping peptides (SEQ ID Nos:1-41) covering the complete An1792 sequence.
Animal number F10975F shows a representative non-restricted N-terminal response.
Reactivity is seen against the two peptides N-terminal and one peptide C-terminal to the peptide DAEFRHDSGY (SEQ ID NO:9) which covers amino acids 1-10 of the AN1792 peptide.
DEFINITIONS
The term "substantial identity" means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 65 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. Appl. 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 ScL
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 etal., 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 chains): cys, ser, thr;
Group III (acidic side chains): asp, glu; Group IV (basic side chains): asn, gln, 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 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 from undesired contaminant. 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" or "immunoglobulin" 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 fragment including separate heavy chains, light chains Fab, Fab' F(ab')2, Fabc, and Fv.
Fragments are produced by recombinant DNA techniques, or by enzymatic or chemical separation of intact immunoglobulins. The term "antibody" also includes one or more immunoglobulin chains that are chemically conjugated to, or expressed as, fusion proteins with other proteins. The term "antibody" also includes bispecific antibody. A bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79:315-321 (1990); Kostelny etal., 1 Immunol. 148, 1547-1553 (1992).
App695, App751, 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 A1339, Ar340, A1341, A1342 and Af343 refer to an A13 peptide containing amino acid residues 1-39, 1-40, 1-41, 1-42 and 1-43.
An "antigen" is an entity to which an antibody specifically binds.
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 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., I Inf Dis. 170, 1110-19 (1994)), by antigen-dependent killing (cytotoxic T lymphocyte assay, Tigges et al., I 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 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 antibodies 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 mammal, 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 subjects that receive either prophylactic or therapeutic treatment.
Disaggregated or monomeric AP means soluble, monomeric peptide units of A. One method to prepare monomeric AP is to dissolve lyophilized peptide in neat DMSO with sonication. The resulting solution is centrifuged to remove any insoluble particulates. Aggregated AP is a mixture of oligomers in which the monomeric units are held together by noncovalent bonds.
Competition between antibodies is determined by an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen, such as AP. Numerous types of competitive binding assays are known, for example: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition assay (see Stahli et al., Methods in Enzymology 9:242-253 (1983)); solid phase direct biotin-avidin EIA
(see Kirkland et al., J. Immunol. 137:3614-3619 (1986)); solid phase direct labeled assay, solid phase direct labeled sandwich assay (see Harlow and Lane, "Antibodies, A
Laboratory Manual," Cold Spring Harbor Press (1988)); solid phase direct label RIA
using 1-125 label (see Morel et al., Molec. Immunol. 25(1):7-15 (1988)); solid phase direct biotin-avidin EIA (Cheung et al., Virology 176:546-552 (1990)); and direct labeled RIA (Moldenhauer et al., Scand. J. Immunol. 32:77-82 (1990)). Typically, such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabelled test immunoglobulin and a labelled reference immunoglobulin.
Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test immunoglobulin. Usually the test immunoglobulin is present in excess. Antibodies identified by competition assay (competing antibodies) include antibodies binding to the same epitope as the reference antibody and antibodies binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference antibody for steric hindrance to occur.
Usually, when a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to a common antigen by at least 50 or 75%.
Compositions or methods "comprising" one or more recited elements may include other elements not specifically recited. For example, a composition that comprises Ap peptide encompasses both an isolated AP peptide and AP peptide as a component of a larger polypeptide sequence.
DETAILED DESCRIPTION
I. General Several amyloidogenic diseases and conditions are characterized by presence of deposits of AP peptide aggregated to an insoluble mass in the brain of a patient. Such diseases include Alzheimer's disease, Down's syndrome and cognitive impairment. The latter is a symptom of Alzheimer's disease and Down's syndrome but can also without other characteristics of either of these diseases. For example, mild cognitive impairment or age-associated memory loss occurs in some patient who have not yet developed, or may never develop full Alzheimer's disease. Mild cognitive impairment can be defined by score on the Mini-Mental State Exam in accordance with convention.
Such diseases are characterized by aggregates of A13 that have a 13-pleated sheet structure and stain with Congo Red dye. The basic approach of preventing or treating Alzheimer's disease or other amyloidogenic diseases by generating an immunogenic response to a component of the amyloid deposit in a patient is described in WO 99127944. The present application reiterates and confirms the efficacy of the basic approach. The present application is, however, principally directed to improved reagents and methods. These improvements are premised, in part, on the present inventors having localized the preferred epitopes within AP against which an immunogenic response should be directed. The identification of preferred epitopes within AP results in agents and methods having increased efficacy, reduced potential for side effects, and/or greater ease of manufacture, formulation and administration.
Therapeutic Agents An immunogenic response can be active, as when an immunogen is administered to induce antibodies reactive with AP in a patient, or passive, as when an antibody is administered that itself binds to AP in a patient.
1. Agents Inducing Active Immune Response Therapeutic agents induce an immunogenic response specifically directed to certain epitopes within Ap peptides. Preferred agents are the Af3 peptide itself and segments thereof. Variants of such segments, analogs and mimetics of natural Ap peptide that induce and/or crossreact with antibodies to the preferred epitopes of AP
peptide can also be used.
AP, also known as P-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. Af3 is generated by processing of a larger protein APP by two enzymes, termed 13 and y secretases (see Hardy, TINS 20, 154 (1997)). Known mutations in APP associated with Alzheimer's disease occur proximate to the site of (3 or y secretase, or within Af3. For example, position 717 is proximate to the site of y-secretase cleavage of APP in its processing to AO, and positions 670/671 are proximate to the site of p-secretase cleavage. It is believed that the mutations cause AD by interacting with the =

cleavage reactions by which Al) is formed so as to increase the amount of the anlino acid form of Al) generated.
Al) 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 Al) to generate an immune response in excess of that of other antigens.
Al) has several natural occurring forms. The human forms of Al) are referred to as AP39, AP40, AP41, Ap42 and Af343. 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, Ap42 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-11e-Ala-OH,(SEQ ID NO:42).
A1341, AP40 and A1339 differ from A(342 by the omission of Ala, Ala-lie, and Ala-Ile-Val respectively from the C-terminal end. Af343 differs from AP42 by the presence of a threonine residue at the C-terminus.
Immunogenic fragments of Al) are advantageous relative to the intact molecule in the present methods for several reasons. First, because only certain epitopes within Al) induce a useful immunogenic response for treatment of Alzheimer's disease, an equal dosage of mass of a fragment containing such epitopes provides a greater molar concentration of the useful immunogenic epitopes than a dosage of intact Al).
Second, certain immunogenic fragments of Al) generate an immunogenic response against amyloid deposits without generating a significant immunogenic response against APP
protein from which Al) derives. Third, fragments of Al) are simpler to manufacture than intact Al) due to their shorter size. Fourth, fragments of Al) do not aggregate in the same manner as intact Al), simplifying preparation of pharmaceutical compositions and administration thereof.
Some immunogenic fragments of Al) have a sequence of at least 2, 3, 5, 6, 10 or 20 contiguous amino acids from a natural peptide. Some immunogenic fragments have no more than 10,9, 8, 7, 5 or 3 contiguous residues from Al). Fragments from the N-terminal half of Ap are preferred. Preferred immunogenic fragments include AP1-5, 1-6, 1-7, 1-10, 3-7, 1-3, and 1-4. The designation A31-5 for example, indicates a fragment including residues 1-5 of Ap and lacking other residues of AP. Fragments beginning at residues 1-3 of AP and ending at residues 7-11 of AP are particularly preferred. The fragment AP1-12 can also be used but is less preferred. In some methods, the fragment is an N-terminal fragment other than A131-10. Other less preferred fragments include A3I3-28, 17-28, 1-28, 25-35, 35-40 and 35-42. These fragments require screening for activity in clearing or preventing amyloid deposits as described in the Examples before use. Fragments lacking at least one, and sometimes at least 5 or 10 C-terminal amino acid present in a naturally occurring forms of AP are used in some methods. For example, a fragment lacking 5 amino acids from the C-terminal end of Ap43 includes the first 38 amino acids from the N-terminal end of AP. Other components of amyloid plaques, for example, synuclein, and epitopic fragments thereof can also be used to induce an immunogenic response.
Unless otherwise indicated, reference to AP includes the natural human amino acid sequences indicated above as well as analogs including allelic, species and induced variants. Analogs typically differ from naturally occurring peptides at one, two or a few positions, often by virtue of conservative substitutions. Analogs typically exhibit at least 80 or 90% sequence identity with natural peptides. Some analogs also include unnatural amino acids or modifications of N or C terminal amino acids at a one, two or a few positions. For example, the natural aspartic acid residue at position 1 and/or 7 of AP
can be replaced with iso-aspartic acid. Examples of unnatural amino acids are D-amino acids, a, a-disubstituted amino acids, N-alkyl amino acids, lactic acid, 4-hydroxyproline, y-carboxyglutamate, E -N,N,N-trimethyllysine, E -N-acetylly sine, 0-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, co-N-methylarginine, and isoaspartic acid. Fragments and analogs can be screened for prophylactic or therapeutic efficacy in transgenic animal models in comparison with untreated or placebo controls as described below.
Af3, its fragments, and analogs 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 A13 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 active fragment of AP peptide, together with other amino acids.
For example, preferred agents include fusion proteins comprising a segment of AP
fused to a heterologous amino acid sequence that induces a helper T-cell response against the heterologous amino acid sequence and thereby a B-cell response against the AP
segment.
Such polypeptides can be screened for prophylactic or therapeutic efficacy in animal models in comparison with untreated or placebo controls as described below.
The AP
peptide, analog, active fragment or other polypeptide can be administered in associated or multimeric form or in dissociated form Therapeutic agents also include multimers of monomeric immunogenic agents.
In a further variation, an immunogenic peptide, such as a fragment of AP, can be presented by a virus or a bacteria as part of an immunogenic composition. 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 outer surface 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, Venezuelan equine encephalitis virus and other alpha viruses, vesicular stomatitis virus, and other rhabdo viruses, vaccinia and fowl pox. Suitable bacteria include Salmonella and Shigella.
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 AP but nevertheless serve as mimetics of AP and induce a similar immune response. For example, any peptides and proteins forming 13-pleated sheets can be screened for suitability. Anti-idiotypic antibodies against monoclonal antibodies to AP 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).

Agents other than Af3 peptides should induce an immunogenic response against one or more of the preferred segments of Af3 listed above (e.g., 1-10, 1-7, 1-3, and 3-7).
Preferably, such agents induce an immunogenic response that is specifically directed to one of these segments without being directed to other segments of A.
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, 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, W091/18980.
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 Ap or other amyloidogenic peptides. For example, initial screens can be performed with any polyclonal sera or monoclonal antibody to Af3 or a fragment thereof. Compounds can then be screened for binding to a specific epitope within Ai3 (e.g., 1-10, 1-7, 1-3, 1-4, 1-5 and 3-7). Compounds can be tested by the same procedures described for mapping antibody epitope specificities. Compounds identified by such screens are then further analyzed for capacity to induce antibodies or reactive lymphocytes to AP or fragments thereof. For example, multiple dilutions of sera can be tested on microtiter plates that have been precoated with Af3 or a fragment thereof and a standard ELISA can be performed to test for reactive antibodies to Ai3 or the fragment.
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 670/671 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 at, 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-(1997)). The same screening approach can be used on other potential agents analogs of Ap and longer peptides including fragments of AP, described above.
2. Agents Inducing Passive Immune Response Therapeutic agents of the invention also include antibodies that specifically bind to AP or other component of amyloid plaques. Such antibodies can be monoclonal or polyclonal. Some such antibodies bind specifically to the aggregated form of AP 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. Some such antibodies bind to a naturally occurring short form of AP
(i.e., Ap39, 40 or 41) without binding to a naturally occurring long form of AP (i.e., A342 and A343). Some antibodies bind to a long form without binding to a short form. Some antibodies bind to AP without binding to full-length amyloid precursor protein.
Antibodies used in therapeutic methods usually have an intact constant region or at least sufficient of the constant region to interact with an Fc receptor. Human isotype IgG1 is preferred because of it having highest affinity of human isotypes for the FcRI
receptor on phagocytic cells. Bispecific Fab fragments can also be used, in which one arm of the antibody has specificity for AP, and the other for an Fc receptor. Some antibodies bind to AP with a binding affinity greater than or equal to about 106, 107, 108, 109, or 1010 M-1.
Polyclonal sera typically contain mixed populations of antibodies binding to several epitopes along the length of AP. However, polyclonal sera can be specific to a particular segment of AP, such as AP 1-10. Monoclonal antibodies bind to a specific epitope within AP that can be a conformational or nonconformational epitope.
Prophylactic and therapeutic efficacy of antibodies can be tested using the transgenic animal model procedures described in the Examples. Preferred monoclonal antibodies bind to an epitope within residues 1-10 of Af3 (with the first N terminal residue of natural Ap designated 1). Some preferred monoclonal antibodies bind to an epitope within amino acids 1-5, and some to an epitope within 5-10. Some preferred antibodies bind to epitopes within amino acids 1-3, 1-4, 1-5, 1-6, 1-7 or 3-7. Some preferred antibodies bind to an epitope starting at resides 1-3 and ending at residues 7-11 of A. Less preferred antibodies include those binding to epitopes with residues 10-15, 15-20, 25-30, 10-20, 20, 30, or 10-25 of A. It is recommended that such antibodies be screened for activity in the mouse model described in the Examples before use. For example, it has been found that certain antibodies to epitopes within residues 10-18, 16-24, 18-21 and 33-42 lack activity. In some methods, multiple monoclonal antibodies having binding specificities to different epitopes are used. Such antibodies can be administered sequentially or simultaneously. Antibodies to amyloid components other than AP can also be used. For example, antibodies can be directed to the amyloid associated protein synuclein.
When an antibody is said to bind to an epitope within specified residues, such as AP 1-5 for example, what is meant is that the antibody specifically binds to a polypeptide containing the specified residues (i.e., AP 1-5 in this an example). Such an antibody does not necessarily contact every residue within AP 1-5. Nor does every single amino acid substitution or deletion with in AP1-5 necessarily significantly affect binding affinity. Epitope specificity of an antibody can be determined, for example, by forming a phage display library in which different members display different subsequences of Aft The phage display library is then selected for members specifically binding to an antibody under test. A family of sequences is isolated. Typically, such a family contains a common core sequence, and varying lengths of flanking sequences in different members.
The shortest core sequence showing specific binding to the antibody defines the epitope bound by the antibody. Antibodies can also be tested for epitope specificity in a competition assay with an antibody whose epitope specificity has already been determined. For example, antibodies that compete with the 3D6 antibody for binding to AP bind to the same or similar epitope as 3D6, i.e., within residues AP 1-5.
Likewise antibodies that compete with the 10D5 antibody bind to the same or similar epitope, i.e, within residues AP 3-6. Screening antibodies for epitope specificity is a useful predictor of therapeutic efficacy. For example, an antibody determined to bind to an epitope within residues 1-7 of AP is likely to be effective in preventing and treating Alzheimer's disease.
Monoclonal or polyclonal antibodies that specifically bind to a preferred segment of AP without binding to other regions of AP have a number of advantages relative to monoclonal antibodies binding to other regions or polyclonal sera to intact Aft First, for equal mass dosages, dosages of antibodies that specifically bind to preferred segments contain a higher molar dosage of antibodies effective in clearing amyloid plaques. Second, antibodies specifically binding to preferred segments can induce a clearing response against amyloid deposits without inducing a clearing response against intact APP polypeptide, thereby reducing the potential for side effects.

i. General Characteristics of Immunoglobulins The basic antibody structural unit is known to comprise a tetramer of subunits. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa) and one "heavy" chain (about 50-70 kDa). The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
Light chains are classified as either kappa or lambda_ Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's isotype as IgG, IgM, IgA, IgD and IgE, respectively. Within light and heavy chains, the variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D" region of about 10 more amino acids. (See generally, Fundamental Immunology (Paul, W., ed., 2nd ed. Raven Press, N.Y., 1989), Ch.
7..
The variable regions of each light/heavy chain pair form the antibody binding site. Thus, an intact antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same. The chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. The CDRs from the two chains of each pair are aligned by the framework regions, enabling binding to a specific epitope. From N-terminal to C-terminal, both light and heavy chains comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is in accordance with the definitions of Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD, 1987 and 1991), or Chothia & Lesk, J. MoL Biol. 196:901-917 (1987); Chothia et al., Nature 342:878-883 (1989).
ii. Production of Nonhuman Antibodies The production of non-human monoclonal antibodies, e.g., murine, guinea pig, primate, rabbit or rat, can be accomplished by, for example, immunizing the animal with AP. A longer polypeptide comprising A13 or an immunogenic fragment of Af3 or anti-idiotypic antibodies to an antibody to Af3 can also be used. See Harlow &
Lane, Antibodies, A Laboratory Manual (CSHP NY, 1988).

. Such an immunogen can be obtained from a natural source, by peptide synthesis or by recombinant expression. Optionally, the immunogen can be administered fused or otherwise complexed with a carrier protein, as described below.
Optionally, the immunogen can be administered with an adjuvant. Several types of adjuvant can be used as described below. Complete Freund's adjuvant followed by incomplete adjuvant is preferred for immunization of laboratory animals. Rabbits or guinea pigs are typically used for making polyclonal antibodies. Mice are typically used for making monoclonal antibodies. Antibodies are screened for specific binding to AP. Optionally, antibodies are further screened for binding to a specific region of A. The latter screening can be accomplished by determining binding of an antibody to a collection of deletion mutants of an AP peptide and determining which deletion mutants bind to the antibody.
Binding can be assessed, for example, by Western blot or ELISA. The smallest fragment to show specific binding to the antibody defines the epitope of the antibody.
Alternatively, epitope specificity can be determined by a competition assay is which a test and reference antibody compete for binding to A. If the test and reference antibodies compete, then they bind to the same epitope or epitopes sufficiently proximal that binding of one antibody interferes with binding of the other. The preferred isotype for such antibodies is mouse isotype IgG2a or equivalent isotype in other species. Mouse isotype IgG2a is the equivalent of human isotype IgG I.
iii. Chimeric and Humanized Antibodies Chimeric and humanized antibodies have the same or similar binding specificity and affinity as a mouse or other nonhuman antibody that provides the starting material for construction of a chimeric or humanized antibody. Chimeric antibodies are antibodies whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin gene segments belonging to different species.
For example, the variable (V) segments of the genes from a mouse monoclonal antibody may be joined to human constant (C) segments, such as IgG1 and IgG4. Human isotype IgG I
is preferred. A typical chimeric antibody is thus a hybrid protein consisting of the V or antigen-binding domain from a mouse antibody and the C or effector domain from a human antibody.
Humanized antibodies have variable region framework residues substantially from a human antibody (termed an acceptor antibody) and complementarity determining regions substantially from a mouse-antibody, (referred to as the donor immunoglobulin). See , Queen et al., Proc. NatL Acad Sci. USA 86:10029-10033 (1989) and WO 90/07861, US 5,693,762, US 5,693,761, US 5,585,089, US 5,530,101 and Winter, US 5,225,539. The constant region(s), if present, are also substantially or entirely from a human immunoglobulin. The human variable domains are usually chosen from human antibodies whose framework sequences exhibit a high degree of sequence identity with the murine variable region domains from which the CDRs were derived.
The heavy and light chain variable region framework residues can be derived from the same or different human antibody sequences. The human antibody sequences can be the sequences of naturally occurring human antibodies or can be consensus sequences of several human antibodies. See Carter et al., WO 92122653. Certain amino acids from the human variable region framework residues are selected for substitution based on their possible influence on CDR conformation and/or binding to antigen.
Investigation of such possible influences is by modeling, examination of the characteristics of the amino acids at particular locations, or empirical observation of the effects of substitution or mutagenesis of particular amino acids.
For example, when an amino acid differs between a murine variable region framework residue and a selected human variable region framework residue, the human framework amino acid should usually be substituted by the equivalent framework amino acid from the mouse antibody when it is reasonably expected that the amino acid:
(1) noncovalently binds antigen directly, (2) is adjacent to a CDR region, (3) otherwise interacts with a CDR region (e.g. is within about 6 A of a CDR region), or (4) participates in the VL-VH interface.
Other candidates for substitution are acceptor human framework amino acids that are unusual for a human immunoglobulin at that position. These amino acids can be substituted with amino acids from the equivalent position of the mouse donor antibody or from the equivalent positions of more typical human immunoglobulins. Other candidates for substitution are acceptor human framework amino acids that are unusual for a human immunoglobulin at that position.. The variable region frameworks of humanized immunog/obulins usually show at least 85% sequence identity to a human variable region framework sequence or consensus of such sequences.

iv. Human Antibodies Human antibodies against AP are provided by a variety of techniques described below. Some human antibodies are selected by competitive binding experiments, or otherwise, to have the same epitope specificity as a particular mouse antibody, such as one of the mouse monoclonals described in Example XI. Human antibodies can also be screened for a particular epitope specificity by using only a fragment of AP as the immunogen, and/or by screening antibodies against a collection of deletion mutants of A. Human antibodies preferably have isotype specificity human IgGI.
(1) Trioma Methodology The basic approach and an exemplary cell fusion partner, SPAZ-4, for use in this approach have been described by Oestberg et al., Hybridama 2:361-367 (1983);
Oestberg, U.S. Patent No. 4,634,664; and Engleman et al., US Patent 4,634,666.
The antibody-producing cell lines obtained by this method are called triomas, because they are descended from three cells--two human and one mouse. Initially, a mouse myeloma line is fused with a human B-lymphocyte to obtain a non-antibody-producing xenogeneic hybrid cell, such as the SPAZ-4 cell line described by Oestberg, supra. The xenogeneic cell is then fused with an immunized human B-lymphocyte to obtain an antibody-producing trioma cell line. Triomas have been found to produce antibody more stably than ordinary hybridomas made from human cells.
The immunized B-lymphocytes are obtained from the blood, spleen, lymph nodes or bone marrow of a human donor. If antibodies against a specific antigen or epitope are desired, it is preferable to use that antigen or epitope thereof for immunization. Immunization can be either in vivo or in vitro. For in vivo immunization, B cells are typically isolated from a human immunized with AP, a fragment thereof, larger polypeptide containing AP or fragment, or an anti-idiotypic antibody to an antibody to AP. In some methods, B cells are isolated from the same patient who is ultimately to be administered antibody therapy. For in vitro immunization, B-lymphocytes are typically exposed to antigen for a period of 7-14 days in a media such as RPMI-1640 (see Engleman, supra) supplemented with 10% human plasma.

The immunized B-lymphocytes are fused to a xenogeneic hybrid cell such as SPAZ-4 by well known methods. For example, the cells are treated with 40-50%
polyethylene glycol of MW 1000-4000, at about 37 degrees C, for about 5-10 min. Cells are separated from the fusion mixture and propagated in media selective for the desired hybrids (e.g., HAT or AH). Clones secreting antibodies having the required binding specificity are identified by assaying the trioma culture medium for the ability to bind to AP or a fragment thereof. Thomas producing human antibodies having the desired specificity are subcloned by the limiting dilution technique and grown in vitro in culture medium. The trioma cell lines obtained are then tested for the ability to bind A13 or a fragment thereof.
Although triomas are genetically stable they do not produce antibodies at very high levels. Expression levels can be increased by cloning antibody genes from the trioma into one or more expression vectors, and transforming the vector into standard mammalian, bacterial or yeast cell lines.
(2) Transgenic Non-Human Mammals Human antibodies against Af3 can also be produced from non-human transgenic mammals having transgenes encoding at least a segment of the human immunoglobulin locus. Usually, the endogenous immunoglobulin locus of such transgenic mammals is functionally inactivated. Preferably, the segment of the human immunoglobulin locus includes unrearranged sequences of heavy and light chain components. Both inactivation of endogenous immunoglobulin genes and introduction of exogenous immunoglobulin genes can be achieved by targeted homologous recombination, or by introduction of YAC chromosomes. The transgenic mammals resulting from this process are capable of functionally rearranging the immunoglobulin component sequences, and expressing a repertoire of antibodies of various isotypes encoded by human immunoglobulin genes, without expressing endogenous immunoglobulin genes. The production and properties of mammals having these properties are described in detail by, e.g., Lonberg et al., W093/12227 (1993); US
5,877,397, US 5,874,299, US 5,814,318, US 5,789,650, US 5,770,429, US
5,661,016, US
5,633,425, US 5,625,126, US 5,569,825, US 5,545,806, Nature 148, 1547-1553 (1994), Nature Biotechnology 14,826 (1996), Kucherlapati, WO 91/10741 (1991).
Transgenic mice are particularly suitable. Anti-A13 antibodies are obtained by immunizing a transgenic nonhuman mammal, such as described by Lonberg or Kucherlapati, supra, with Ar3 or a fragment thereof. Monoclonal antibodies are prepared by, e.g., fusing B-cells from such mammals to suitable myeloma cell lines using conventional Kohler-Milstein technology.
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 affinity purification using Af3 or other amyloid peptide as an affinity reagent.
(3) Phage Display Methods A further approach for obtaining human anti-Af3 antibodies is to screen a DNA library from human B cells according to the general protocol outlined by Huse et al., Science 246:1275-1281 (1989). As described for trioma methodology, such B
cells can be obtained from a human immunized with Af3, fragments, longer polypeptides containing Af3 or fragments or anti-idiotypic antibodies. Optionally, such B
cells are obtained from a patient who is ultimately to receive antibody treatment.
Antibodies binding to Af3 or a fragment thereof are selected. Sequences encoding such antibodies (or a binding fragments) are then cloned and amplified. The protocol described by Huse is rendered more efficient in combination with phage-display technology. See, e.g., Dower et al., WO 91/17271 and McCafferty et al., WO 92/01047, US 5,877,218, US
5,871,907, US 5,858,657, US 5,837,242, US 5,733,743 and US 5,565,332. In these methods, libraries of phage are produced in which members display different antibodies on their outer surfaces. Antibodies are usually displayed as Br or Fab fragments. Phage displaying antibodies with a desired specificity are selected by affinity enrichment to an Af3 peptide or fragment thereof.
In a variation of the phage-display method, human antibodies having the binding specificity of a selected murine antibody can be produced. See Winter, WO
92/20791. In this method, either the heavy or light chain variable region of the selected murine antibody is used as a starting material. If, for example, a light chain variable region is selected as the starting material, a phage library is constructed in which members display the same light chain variable region (i.e., the murine starting material) and a different heavy chain variable region. The heavy chain variable regions are obtained from a library of rearranged human heavy chain variable regions. A
phage showing strong specific binding for AP (e.g., at least 108 and preferably at least 109 M-1) is selected. The human heavy chain variable region from this phage then serves as a starting material for constructing a further phage library. In this library, each phage displays the same heavy chain variable region (i.e., the region identified from the first display library) and a different light chain variable region. The light chain variable regions are obtained from a library of rearranged human variable light chain regions.
Again, phage showing strong specific binding for Mare selected. These phage display the variable regions of completely human anti-A(3 antibodies. These antibodies usually have the same or similar epitope specificity as the murine starting material.
v. Selection of Constant Region The heavy and light chain variable regions of chimeric, humanized, or human antibodies can be linked to at least a portion of a human constant region. The choice of constant region depends, in part, whether antibody-dependent complement and/or cellular mediated toxicity is desired. For example, isotopes IgG1 and IgG3 have complement activity and isotypes IgG2 and IgG4 do not. Choice of isotype can also affect passage of antibody into the brain. Human isotype IgG1 is preferred.
Light chain constant regions can be lambda or kappa. Antibodies can be expressed as tetramers containing two light and two heavy chains, as separate heavy chains, light chains, as Fab, Fab' F(ab')2, and Fv, or as single chain antibodies in which heavy and light chain variable domains are linked through a spacer.
vi. Expression of Recombinant Antibodies Chimeric, humanized and human antibodies are typically produced by recombinant expression. Recombinant polynucleotide constructs typically include an expression control sequence operably linked to the coding sequences of antibody chains, including naturally-associated or heterologous promoter regions. Preferably, the expression control sequences are eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and the collection and purification of the crossreacting antibodies.

These expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA.
Commonly, expression vectors contain selection markers, e.g., ampicillin-resistance or hygromycin-resistance, to permit detection of those cells transformed with the desired DNA
sequences.
E. coil is one prokaryotic host particularly useful for cloning the DNA
sequences of the present invention. Microbes, such as yeast are also useful for expression. Saccharomyces is a preferred yeast host, with suitable vectors having expression control sequences, an origin of replication, termination sequences and the like as desired. Typical promoters include 3-phosphoglycerate kinase and other glycolytic enzymes. Inducible yeast promoters include, among others, promoters from alcohol dehydrogenase, isocytochrome C, and enzymes responsible for maltose and galactose utilization.
Mammalian cells are a preferred host for expressing nucleotide segments encoding immunoglobulins or fragments thereof See Winnacker, From Genes to Clones, (VCH Publishers, NY, 1987). A number of suitable host cell lines capable of secreting intact heterologous proteins have been developed in the art, and include CHO
cell lines, various COS cell lines, HeLa cells, L cells and myeloma cell lines.
Preferably, the cells are nonhuman. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer (Queen et al., Immunol. Rev. 89:49 (1986)), and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Preferred expression control sequences are promoters derived from endogenous genes, cytomegalovirus, SV40, adenovirus, bovine papillomavirus, and the like. See Co et al., I Immunol. 148:1149 (1992).
Alternatively, antibody coding sequences can be incorporated in transgenes for introduction into the genome of a transgenic animal and subsequent expression in the milk of the transgenic animal (see, e.g., US 5,741,957, US
5,304,489, US 5,849,992). Suitable transgenes include coding sequences for light and/or heavy chains in operable linkage with a promoter and enhancer from a mammary gland specific gene, such as casein or beta lactoglobulin.
The vectors containing the DNA segments of interest can be transferred into the host cell by well-known methods, depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment, electroporation, lipofection, biolistics or viral-lased transfection can be used for other cellular hosts. Other methods used to transform mammalian cells include the use of polybrene, protoplast fusion, liposomes, electroporation, and microinjection (see generally, Sambrook et al., supra).
For production of transgenic animals, transgenes can be microinjected into fertilized oocytes, or can be incorporated into the genome of embryonic stem cells, and the nuclei of such cells transferred into enucleated oocytes.
Once expressed, antibodies can be purified according to standard procedures of the art, including HPLC purification, column chromatography, gel electrophoresis and the like (see generally, Scopes, Protein Purification (Springer-Verlag, NY, 1982)).
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, irnmunoglobulirt molecules, thyroglobulin, ovalbumin, tetanus toxoid, or a toxoid from other pathogenic bacteria, such as diphtheria, E.
coil, cholera, or H. pylori, or an attenuated toxin derivative. Other carriers include T-cell epitopes that bind to multiple MHC alleles, e.g., at least 75% of all human MHC alleles.
Such carriers are sometimes known in the art as "universal T-cell epitopes." Examples of universal T-cell epitopes include:
Influenza Hemagluttinin: HA307-319 PKYVKQNTLICLAT (SEQ ID NO:43) PADRE (common residues bolded) AKXYAAVVTLKAAAJ (SEQ ID NO:44) Malaria CS: T3 epitope EKKIAKMEKASSVFNV (SEQ ID NO:45) Hepatitis B surface antigen: HBsAg19.28 FFLLTRILT (SEQ ID NO:46) Heat Shock Protein 65: hsp65153_171 DQSIGDLIAEAMDKVGNEG (SEQ ID NO:47) bacille Calmette-Guerin QVHFQPLPPAVVKL (SEQ ID NO:48) Tetanus toxoid: TT830444 QYIKANSKFIGITEL (SEQ ID NO:49) Tetanus toxoid: TT947.967 FNNFTVSFWLRVPKVSASHLE (SEQ ID NO:50) HIV gp120 TI: KQIINMWQEVGKAMYA (SEQ ID NO:51) Other carriers for stimulating or enhancing an immune response include cytokines such as IL-1, IL-1 a and 13 peptides, IL-2, yINF, IL-10, GM-CSF, and chemokines, such as MIPla and 13 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 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 create a disulfide linkage between themselves and peptide cysteine resides on one protein and an amide linkage through the s -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-1 -carboxylic acid. The carboxyl groups can be activated by combining them with succinimide or 1-hydroxy1-2-nitro-4-sulfonic acid, sodium salt.
Immunogenic peptides can also be expressed as fusion proteins with carriers (i.e., heterologous peptides). The immunogenic peptide can be linked at its amino terminus, its carboxyl terminus, or both to a carrier. Optionally, multiple repeats of the immunogenic peptide can be present in the fusion protein. Optionally, an immunogenic peptide can be linked to multiple copies of a heterologous peptide, for example, at both the N and C termini of the peptide. Some carrier peptides serve to induce a helper T-cell response against the carrier peptide. The induced helper T-cells in turn induce a B-cell response against the immunogenic peptide linked to the carrier peptide.
Some agents of the invention comprise a fusion protein in which an N-terminal fragment of A13 is linked at its C-terminus to a carrier peptide. In such agents, the N-terminal residue of the fragment of AP constitutes the N-terminal residue of the fusion protein. Accordingly, such fusion proteins are effective in inducing antibodies that bind to an epitope that requires the N-terminal residue of A13 to be in free form. Some agents of the invention comprises a plurality of repeats of an N-terminal segment of AP
linked at the C-terminus to one or more copy of a carrier peptide. The N-terminal fragment of AP incorporated into such fusion proteins sometimes begins at AI31-3 and ends at A37-11. A131-7, A131-3, 1-4, 1-5, and 3-7 are preferred N-terminal fragment of AP. Some fusion proteins comprise different N-terminal segments of Ap in tandem. For example, a fusion protein can comprise A1-7 followed by A131-3 followed by a heterologous peptide.
In some fusion proteins, an N-terminal segment of AP is fused at its N-terminal end to a heterologous carrier peptide. The same variety of N-terminal segments of AP can be used as with C-terminal fusions. Some fusion proteins comprise a heterologous peptide linked to the N-terminus of an N-terminal segment of AP, which is in turn linked to one or more additional N-terminal segments of AP in tandem.
Some examples of fusion proteins suitable for use in the invention are shown below. Some of these fusion proteins comprise segments of AP linked to tetanus toxoid epitopes such as described in US 5,196,512, EP 378,881 and EP 427,347.
Some fusion proteins comprises segments of AP linked to carrier peptides described in US
5,736,142. Some heterologous peptides are universal T-cell epitopes. In some methods, the agent for administration is simply a single fusion protein with an AP
segment linked to a heterologous segment in linear configuration. In some methods, the agent is multimer of fusion proteins represented by the formula 2', in which x is an integer from 1-5. Preferably x is 1, 2 or 3, with 2 being most preferred. When x is two, such a multimer has four fusion proteins linked in a preferred configuration referred to as MAP4 (see US 5,229,490). Epitopes of AP are underlined.
The MAP4 configuration is shown below, where branched structures are produced by initiating peptide synthesis at both the N terminal and side chain amines of lysine. Depending upon the number of times lysine is incorporated into the sequence and allowed to branch, the resulting structure will present multiple N termini. In this example, four identical N termini have been produced on the branched lysine-containing core. Such multiplicity greatly enhances the responsiveness of cognate B
cells.

Peptide KGG =
Peptide KA
Peptide KGG
Peptide AN90549 (A13 1-7/Tetanus toxoid 830-844 in a MAP4 configuration):
DAEFRHDQYIKANSKFIGITEL (SEQ ID NO:52) AN90550 (A[3 1-7/Tetanus toxoid 947-967 in a MAP4 configuration):
DAEFREDFNNFTVSFWLRVPKVSASHLE (SEQ ID NO:53) AN90542 (Aft 1-7/Tetanus toxoid 830-844 + 947-967 in a linear configuration):
DAEFRHDQYIKANSKFIGITELFNNFTVSFWLRVPKVSASHLE (SEQ ID NO:54) AN90576: (Ag. 3-9)/Tetanus toxoid 830-844 in a MAP4 configuration):
EFRHDSGQYIKANSKFIGITELµ (SEQ ID NO:55) Peptide described in US 5,736,142 (all in linear configurations):
AN90562 (AD 1-7/peptide) AKXVAAWTLKAAADAEFRHD' (SEQ ID NO:56) AN90543 (A[31-7 x 3/ peptide): DAEFRHDDAEFRHDDAEFRIiDAKXVAAWTLKAAN
(SEQ ID NO:57) Other examples of fusion proteins (immunogenic epitope of AP bolded) include AKXVAAWTLKAAA-DAEFRHD-DAEFRED-DAEFRIID
(SEQ ID NO:58) DAEFRHD-AICXVAAWTLKAAA (SEQ ID NO:59) DAEFRHD4SQAVHAAHAE1NEAGK (SEQ ID NO:60) FRHDSGY-ISQAVHAAHAEINEAGR (SEQ ID NO:61) EFRHDSG-ISQAVHAAHAEINEAGR (SEQ ID NO:62) PKYVKQNTLKLAT-DAEFRHD-DAEFRHD-DAEFRH13 (SEQ ID NO :63) DAEFRHD-PKYVKQNTLICLAT-DAEFRHD (SEQ ID NO:64) DAEFRHD-DAEFFUID-DAEFRHD-PKYVKQNTLICLAT
(SEQ ID NO:65) =
=
DAEFRBD-DAEFRHD-PKYVKQNTLKLAT (SEQ ID NO:66) DAEFRHD-PKYVKQNTLKLAT-EKKIAKMEICASSVFNV-. QYIKANSKFIGITEL-FNNFTVSFWLRVPKVSASHLE-DAEFRHII
(SEQ ID NO:67) DAEFRHD-DAEFRHD-DAEFRHD-QYIKANSKFIGITEL-FNNFTVSFWLRVPKVSASHLE
(SEQ ID NO:68) DAEFRHD-QYIKANSKFIGITELCFNNFTVSFWLRVPICVS46_15FEZ
(SEQ ID NO:69) DAEFRHD-QYIKANSKFIGITELCFNNFTVSFWLRVPKVSASHLE-DAEFRHIY
(SEQ ID NO:70) DAEFRHD -QYIKANSKFIGITEL (SEQ
ID NO:52) on a 2 branched resin peptide Lys-Gly-Cys peptide EQVTNVGGAISQAVHAAHAEINEAGR (SEQ ID NO:71) (Synuclein fusion protein in MAP-4 configuration) The same or similar carrier proteins and methods of linkage can be used for generating immunogens to be used in generation of antibodies against AP
for use in passive immunization. For example, Ap or a fragment linked to a carrier can be administered to a laboratory animal in the production of monoclonal antibodies to Afl.
4. Nucleic Acid Encoding Therapeutic Agents Immune responses against amyloid deposits can also be induced by administration of nucleic acids encoding segments of Ap peptide, and fragments thereof, other peptide immunogens, or antibodies and their component chains used for passive immunization. Such nucleic acids can be DNA or RNA. A nucleic acid segment encoding an 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. For administration of double-chain antibodies, the two chains can be cloned in the same or separate vectors.

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., I Virol. 67, 5911 (1993)); adeno-associated virus vectors (see, e.g., Zhou et al., I 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., I Virol. 70, 508-519 (1996)), Venezuelan equine encephalitis virus (see US
5,643,576) and rhabdoviruses, such as vesicular stomatitis virus (see WO
96/34625)and papillomaviruses (Ohe et al., Human Gene Therapy 6, 325-333 (1995); Woo et al., WO
94/12629 and Xiao & 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., I 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 intracranial infusion) or topical application (see e.g., US 5,399,346). Such vectors can further include facilitating agents such as bupivacine (US 5,593,970). 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. Screening Antibodies for Clearing Activity The invention provides methods of screening an antibody for activity in clearing an amyloid deposit or any other antigen, or associated biological entity, for which clearing activity is desired. To screen for activity against an amyloid deposit, a tissue sample from a brain of a patient with Alzheimer's disease or an animal model having characteristic Alzheimer's pathology is contacted with phagocytic cells bearing an Fc receptor, such as microglial cells, and the antibody under test in a medium in vitro.
The pagocytic cells can be a primary culture or a cell line, such as BV-2, C8-B4, or THP-1. In some methods, the components are combined on a microscope slide to facilitate microscopic monitoring. In some methods, multiple reactions are performed in parallel in the wells of a microtiter dish. In such a format, a separate miniature microscope slide can be mounted in the separate wells, or a nonmicroscopic detection format, such as ELISA detection of AP can be used. Preferably, a series of measurements is made of the amount of amyloid deposit in the in vitro reaction mixture, starting from a baseline value before the reaction has proceeded, and one or more test values during the reaction. The antigen can be detected by staining, for example, with a fluorescently labelled antibody to AP or other component of amyloid plaques. The antibody used for staining may or may not be the same as the antibody being tested for clearing activity. A
reduction relative to baseline during the reaction of the amyloid deposits indicates that the antibody under test has clearing activity. Such antibodies are likely to be useful in preventing or treating Alzheimer's and other amyloidogenic diseases.
Analogous methods can be used to screen antibodies for activity in clearing other types of biological entities. The assay can be used to detect clearing activity against virtually any kind of biological entity. Typically, the biological entity has some role in human or animal disease. The biological entity can be provided as a tissue sample or in isolated form. If provided as a tissue sample, the tissue sample is preferably unfixed to allow ready access to components of the tissue sample and to avoid perturbing the conformation of the components incidental to fixing. Examples of tissue samples that can be tested in this assay include cancerous tissue, precancerous tissue, tissue containing benign growths such as warts or moles, tissue infected with pathogenic microorganisms, tissue infiltrated with inflammatory cells, tissue bearing pathological matrices between cells (e.g., fibrinous pericarditis), tissue bearing aberrant antigens, and scar tissue. Examples of isolated biological entities that can be used include AP, viral antigens or viruses, proteoglycans, antigens of other pathogenic microorganisms, tumor antigens, and adhesion molecules. Such antigens can be obtained from natural sources, recombinant expression or chemical synthesis, among other means. The tissue sample or isolated biological entity is contacted with phagocytic cells bearing Fc receptors, such as monocytes or microglial cells, and an antibody to be tested in a medium. The antibody can be directed to the biological entity under test or to an antigen associated with the entity In the latter situation, the object is to test whether the biological entity is vicariously phagocytosed with the antigen. Usually, although not necessarily, the antibody and biological entity (sometimes with an associated antigen) are contacted with each other before adding the phagocytic cells. The concentration of the biological entity and/or the associated antigen, if present, remaining in the medium is then monitored. A
reduction in the amount or concentration of antigen or the associated biological entity in the medium indicates the antibody has a clearing response against the antigen and/or associated biological entity in conjunction with the phagocytic cells (see, e.g., Example 14).
IV. 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 the need for 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 presenilin genes, PS1 and PS2, and ApoE4, 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 A1342 levels. Elevated tau and decreased A342 levels signify the presence of AD. Individuals suffering from Alzheimer's disease can also be diagnosed by 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., AP 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.
V. TREATMENT REGIMES
In prophylactic applications, pharmaceutical compositions or medicaments are administered to a patient susceptible to, or otherwise at risk of, Alzheimer's disease in an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the outset of the disease, including biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development 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 (biochemical, histologic and/or behavioral), including its complications and intermediate pathological phenotypes in development of the disease. In some methods, administration of agent reduces or eliminates myocognitive impairment in patients that have not yet developed characteristic Alzheimer's pathology. An amount adequate to accomplish therapeutic or prophylactic treatment is defined as a therapeutically- or prophylactically-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 repeated dosages are given if the immune response starts to wane.
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 nonhuman mammals including transgenic mammals can also be treated. 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 1-500 j_ig per patient and more usually from 5-500 g per injection for human administration.
Occasionally, a higher dose of 1-2 mg per injection is used. Typically about 10, 20, 50 or 100 ps is used for each human injection. The mass of immunogen also depends on the mass ratio of immunogenic epitope within the immunogen to the mass of immunogen as a whole.
Typically, 10-3to 10-5 micromoles of immunogenic epitope are used for microgram of immunogen. 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 lig/ patient if adjuvant is also administered, and greater than 10 g/patient and usually greater than 100 g/patient in the absence of adjuvant. A typical regimen consists of an immunization followed by booster injections at time intervals, such as 6 week 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. For example dosages can be 1 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg. An exemplary treatment regime entails administration once per every two weeks or once a month or once every 3 to 6 months. In some methods, two or more monoclonal antibodies with different binding specificities are administered simultaneously, in which case the dosage of each antibody administered falls within the ranges indicated. Antibody is usually administered on multiple occasions.
Intervals between single dosages can be weekly, monthly or yearly. Intervals can also be irregular as indicated by measuring blood levels of antibody to AP in the patient. In some methods, dosage is adjusted to achieve a plasma antibody concentration of 1-1000 ug/ml and in some methods 25 ¨ 300 ug/ml. Alternatively, antibody can be administered as a sustained release formulation, in which case less frequent administration is required.

Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, human antibodies show the longest half life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patent can be administered a prophylactic regime.
Doses for nucleic acids encoding immunogens range from about 10 ng to 1 g, 100 ng to 100 mg, 1 jag to 10 mg, or 30-3001.1g DNA per patient. Doses for infectious viral vectors vary from 10-100, or more, virions per dose.
Agents for inducing an immune response can be administered by parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intracranial, intraperitoneal, intranasal or intramuscular means for prophylactic and/or therapeutic treatment. The most typical route of administration of an immunogenic agent is subcutaneous although other routes can be equally effective. The next most common route is intramuscular injection. This type of injection is most typically performed in the arm or leg muscles. In some methods, agents are injected directly into a particular tissue where deposits have accumulated, for example intracranial injection.
Intramuscular injection on intravenous infusion are preferred for administration of antibody. In some methods, particular therapeutic antibodies are injected directly into the cranium. In some methods, antibodies are administered as a sustained release composition or device, such as a MedipadTM device.
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 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 sometimes administered in combination with an adjuvant. A variety of adjuvants can be used in combination with a peptide, such as AP, 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 aluminum hydroxide and aluminum phosphate, 3 De-0-acylated monophosphoryl lipid A (MPLTm) (see GB 2220211 (RIBI ImmunoChem Research Inc., Hamilton, Montana, now part of Corixa). StimulonTm QS-2I 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 Adjuvant Approach (eds. Powell & Newman, Plenum Press, NY, 1995); US Patent No.
5,057,540),(Aquila BioPharmaceuticals, Framingham, MA). Other adjuvants are oil in water emulsions (such as squalene or peanut oil), optionally in combination with immune stimulants, such as monophosphoryl lipid A (see Stoute et at, N EngL J. Med.
336, 86-91 (1997)). Another adjuvant is CpG ( WO 98/40100). Alternatively, AP can be coupled to an adjuvant. However, such coupling should not substantially change the conformation of Ap 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, QS-21, 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 inununostimulating 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-acetylmurarnyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1'-Tdipalmitoyl-sn-glycero-3-hydroxyphosphoryloxy)-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%
TweeTrig0, , and 0.5% SpanTm-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% Squalene, 0.4% Tween 80, 5%

pluronic-blocked polymer L121, and thr-MDP, either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) RibiTM adjuvant system (RAS), (Ribi ImmunoChem, Hamilton, MT) containing 2% squalene, 0.2%
Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphoryllipid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton (CWS), preferably MPL + CWS (DetoxTm). Another class of preferred adjuvants is saponin adjuvants, such as StimulonTM (QS-21, Aquila, Framingham, 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-1, 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. Irnmunogen 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 immunogenic formulation 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 QS-21 are preferred. Optionally, two or more different adjuvants can be used simultaneously. Preferred combinations include alum with MPL, alum with QS-21, MPL
with QS-21, and alum, QS-21 and MPL together. Also, Incomplete Freund's adjuvant can be used (Chang etal., Advanced Drug Delivery Reviews 32, 173-186 (1998)), optionally in combination with any of alum, QS-21, and MPL and all combinations thereof Agents of the invention are often administered as pharmaceutical compositions comprising an active therapeutic 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.
Pharmaceutical compositions can also include large, slowly metabolized macromolecules such as proteins, polysaccharides such as chitosan, polylactic acids, polyglycolic acids and copolymers (such as latex functionalized sepharose(TM), 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 that 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 propylene glycol or polyethylene glycol are preferred liquid carriers, particularly for injectable solutions. Antibodies 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 release of the active ingredient. An exemplary composition comprises monoclonal antibody at 5 mg/mL, formulated in aqueous buffer consisting of 50 mM L-histidine, 150 mM NaCl, adjusted to pH 6.0 with HC1.
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%-95% 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 crosslinlcing or expression as a fusion protein.
Alternatively, transdermal delivery can be achieved using a skin path or using transferosomes (Paul etal., Eur. J. Immunol. 25, 3521-24 (1995); Cevc et al., Biochem. Biophys. Acta 1368, 201-15 (1998)).
VI. Methods of Diagnosis The invention provides methods of detecting an immune response against A13 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 prophylactic treatment on asymptomatic patients. The methods are useful for monitoring both active immunization (e.g., antibody produced in response to administration of immunogen) and passive immunization (e.g., measuring level of administered antibody).

1. Active Immunization 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 the typical margin of experimental error in repeat measurements of the same sample, expressed as one standard deviation from the mean of such measurements) in value of the immune response signals a positive treatment outcome (i.e., that administration of the agent has achieved or augmented an 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 immunogenic agent are expected to show an increase in immune response with successive dosages, which 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.
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 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 a patient can be compared with a control value (mean plus standard deviation) determined in a 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 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, mucous or cerebrospinal fluid from the patient. The sample is analyzed for indication of an immune response to any form of AP peptide, typically Af342. The immune response can be determined from the presence of, e.g., antibodies or T-cells that specifically bind to AP peptide. ELISA methods of detecting antibodies specific to AP are described in the Examples section. Methods of detecting reactive T-cells have been described above (see Definitions). In some methods, the immune response is determined using a clearing assay, such as described in Section III above. In such methods, a tissue sample from a patient being tested is contacted with amyloid deposits (e.g., from a PDAPP
mouse) and phagocytic cells bearing Fe receptors. Subsequent clearing of the amyloid deposit is then monitored. The existence and extent of clearing response provides an indication of the existence and level of antibodies effective to clear AP in the tissue sample of the patient under test.

2. Passive Immunization In general, the procedures for monitoring passive immunization are similar to those for monitoring active immunization described above. However, the antibody profile following passive immunization typically shows an immediate peak in antibody concentration followed by an exponential decay. Without a further dosage, the decay approaches pretreatment levels within a period of days to months depending on the half-life of the antibody administered. For example the half-life of some human antibodies is of the order of 20 days.
In some methods, a baseline measurement of antibody to AP in the patient is made before administration, a second measurement is made soon thereafter to determine the peak antibody level, and one or more further measurements are made at intervals to monitor decay of antibody levels. When the level of antibody has declined to baseline or a predetermined percentage of the peak less baseline (e.g., 50%, 25% or 10%), administration of a further dosage of antibody is administered. In some methods, peak or subsequent measured levels less background are compared with reference levels previously determined to constitute a beneficial prophylactic or therapeutic treatment regime in other patients. If the measured antibody level is significantly less than a reference level (e.g., less than the mean minus one standard deviation of the reference value in population of patients benefiting from treatment) administration of an additional dosage of antibody is indicated.
3. Diagnostic Kits 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 A. The kit can also include a label. For detection of antibodies to AP, 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. Kits also typically contain labeling providing directions for use of the kit. The labeling may also include a chart or other correspondence regime correlating levels of measured label with levels of antibodies to All The term labeling 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 labeling encompasses advertising leaflets and brochures, packaging materials, instructions, audio or video cassettes, computer discs, as well as writing imprinted directly on kits.
The invention also provides diagnostic kits for performing in vivo imaging. Such kits typically contain an antibody binding to an epitope of Ap, preferably within residues 1-10. Preferably, the antibody is labelled or a secondary labeling reagent is included in the kit. Preferably, the kit is labelled with instructions for performing an in vivo imaging assay.
VII. In Vivo Imaging The invention provides methods of in vivo imaging amyloid deposits in a patient. Such methods are useful to diagnose or confirm diagnosis of Alzheimer's disease, or susceptibility thereto. For example, the methods can be used on a patient presenting with symptoms of dementia. If the patient has abnormal amyloid deposits, then the patient is likely suffering from Alzheimer's disease. The methods can also be used on asymptomatic patients. Presence of abnormal deposits of amyloid indicates susceptibility to future symptomatic disease. The methods are also useful for monitoring disease progression and/or response to treatment in patients who have been previously diagnosed with Alzheimer's disease.
The methods work by administering a reagent, such as antibody, that binds to Ap to the patient, and then detecting the agent after it has bound.
Preferred antibodies bind to AP deposits in a patient without binding to full length APP
polypeptide.
Antibodies binding to an epitope of AP within amino acids 1-10 are particularly preferred. In some methods, the antibody binds to an epitope within amino acids 7-10 of A. Such antibodies typically bind without inducing a substantial clearing response. In other methods, the antibody binds to an epitope within amino acids 1-7 of AP.
Such antibodies typically bind and induce a clearing response to Af3. However, the clearing response can be avoided by using antibody fragments lacking a full length constant region, such as Fabs. In some methods, the same antibody can serve as both a treatment and diagnostic reagent. In general, antibodies binding to epitopes C-terminal of residue 10 of Ado not show as strong signal as antibodies binding to epitopes within residues 1-10, presumably because the C-terminal epitopes are inaccessible in amyloid deposits.
Accordingly, such antibodies are less preferred.

Diagnostic reagents can be administered by intravenous injection into the body of the patient, or directly into the brain by intracranial injection or by drilling a hole through the skull. The dosage of reagent should be within the same ranges as for treatment methods. Typically, the reagent is labelled, although in some methods, the primary reagent with affinity for AP is unlabelled and a secondary labeling agent is used to bind to the primary reagent. The choice of label depends on the means of detection.
For example, a fluorescent label is suitable for optical detection. Use of paramagnetic labels is suitable for tomographic detection without surgical intervention.
Radioactive labels can also be detected using PET or SPECT.
Diagnosis is performed by comparing the number, size and/or intensity of labelled loci to corresponding base line values. The base line values can represent the mean levels in a population of undiseased individuals. Base line values can also represent previous levels determined in the same patient. For example, base line values can be determined in a patient before beginning treatment, and measured values thereafter compared with the base line values. A decrease in values relative to base line signals a positive response to treatment.
EXAMPLES
I. PROPHYLACTIC EFFICACY OF AP AGAINST AD
These examples describe administration of A1342 peptide to transgenic mice overexpressing APP with a mutation at position 717 (APP7i7v-4F) 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 AP at six months of age forward. By fifteen months of age they exhibit levels of Ali deposition equivalent to that seen in Alzheimer's disease.
PDAPP mice were injected with aggregated A1342 (aggregated A1342) or phosphate buffered saline. Aggregated A342 was chosen because of its ability to induce antibodies to multiple epitopes of AP.
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 Ap42 (one died in transit), mice to be injected with PBS/adjuvant or PBS, and 10 uninjected controls. Five mice were injected with peptides derived from the sequence of serum amyloid protein (SAP).
2. Preparation of Immunogens 5 Preparation of aggregated A[342: 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 A13 was stored as a dry lyophilized powder at -20 C until the next injection.
3. Preparation of Injections For each injection, 1001.1g of aggregated A1342 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 tl 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 4001,t1 per mouse, or 500 Ill of PBS per mouse.
SAP
injections likewise followed the same schedule using a dose of 100 lag 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 A1342 (aggregated A1342), SAP peptides, or phosphate buffered saline. A group of PDAPP mice were also left as uninjected, positive controls. The titers of the mice to aggregated A1342 were monitored every other month from the fourth boost until the mice were one year of age.
Mice were sacrificed at 13 months. At all time points examined, eight of the nine aggregated A1342 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 had titers of 1:1,000 to 1:30,000 for this immunogen with only a single mouse exceeding 1:10,0000.
The PBS-treated mice were titered against aggregated Ap42 at six, ten and twelve months. At a 1/100 dilution the PBS mice, when titered against aggregated AP42, only exceeded 4 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 AP1-42 treated group had no detectable amyloid in their brains. In contrast, brain tissue from mice in the SAP and PBS groups contained numerous 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 characteristic involvement of vulnerable subregions, such as the outer molecular layer of the hippocampal dentate gyrus. One mouse from the AP 1-42-injected group had a greatly reduced amyloid burden, confined to the hippocampus. An isolated plaque was identified in another AP 1-42-treated mouse.
Quantitative image analyses of the amyloid burden in the hippocampus verified the dramatic reduction achieved in the AP42(AN1792)-treated animals (Fig. 2).
The median values of the amyloid burden for the PBS group (2.22%), and for the 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, control mice contained numerous AP amyloid deposits visualized with the AP-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 AP 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 0.28%, p = 0.0005).
Astrocytosis characteristic of plaque-associated inflammation was also absent in the brains of the A131-42 injected group. The brains from the mice in the other groups contained abundant and clustered GFAP-positive astrocytes typical of AP
plaque-associated gliosis. A subset of the GFAP-reacted slides were counter-stained with Thioflavin S to localize the AP deposits. The GFAP-positive astrocytes were associated with AP plaques in the SAP, PBS and untreated controls. No such association was found in the plaque-negative AP1-42 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) Evidence from a subset of the Af31-42- and PBS-injected mice indicated plaque-associated MHC II immunoreactivity was absent in the A131-42 injected mice, consistent with lack of an AP-related inflammatory response.
Sections of the mouse brains were also reacted with a mAb specific with a monoclonal antibody 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 AP-induced inflammatory response.
C. CONCLUSION
The lack of Af3 plaques and reactive neuronal and gliotic changes in the brains of the Ap1-42-injected mice indicate that no or extremely little amyloid was deposited in their brains, and pathological consequences, such as gliosis and neuritic pathology, were absent. PDAPP mice treated with A131-42 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 AP from brain tissue, and elimination of subsequent neuronal and inflammatory degenerative changes.
Thus, administration of AP peptide can have both preventative and therapeutic benefit in prevention of AD.
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 AP
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 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 jig 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 doses of immunogenic formulations. 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 jig 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 pz 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 jig recipients.
The 1.2 and 3.7 fig 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 jig 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 1.1g to 11 jig ranging from no detectable antibody for recipients of 0.14 jig to a GMT of 36,000 for recipients of 11 jig. Again, titers for the four highest dose groups of 11 to 300 g clustered together. Thus following two immunizations, the antibody titer was dependent on the antigen dose across the broad range from 0.4 to 300 g. 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 jig 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 modest (50%) increase was seen at this time in the 33 g group. In the 300 g 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 g) and about 14% for the 33 and 11 g 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 THERAPEUTIC EFFICACY AGAINST ESTABLISHED AD
This assay is designed to test immunogenic agents for activity in arresting or reversing neuropathologic characteristics of AD in aged animals.
Immunizations with 42 amino acid long Ar3 (AN1792) were begun at a time point when amyloid plaques are already present in the brains of the PDAPP mice.
Over the time course 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 AP 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 neuronal component of PDAPP neuritic plaques is easily visualized with an antibody specific for human APP (monoclonal antibody 8E5), and is readily measurable by computer-assisted image analysis. Therefore, in addition to measuring the effects of AN1792 on 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 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 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 lAg of AN1792 and 24 mice to be immunized with PBS, each combined with Freund's adjuvant. The AN1792 and PBS groups were again 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 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 AP and APP levels in the brain; the one-year old animals were also 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 time point.
California Peptides lots ME0339 and ME0439 were used for the three additional immunizations administered between 15 and 18 months.
For immunizations, 100 lag of AN1792 in 2001.11 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 4001.11. 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, B. Results 1. Effects of AN1792 Treatment on Amyloid Burden The results of AN1792 treatment on cortical amyloid burden determined Typically, cortical amyloid deposition in PDAPP mice initiates in the frontal and retrosplenial cortices (RSC) and progresses in a ventral-lateral direction to 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 30 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 An-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 A13 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 A13 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 gyms. 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-A13 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 AP-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 Ar3 plaques.

Confocal microscopy of MHCII- and AP-labelled sections revealed that plaque material was contained within many of the monocytic cells.
Quantitative image analysis of MHC 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 clearance of amyloid in plaque-bearing brain regions.
3.AN1792 Effects on AP Levels: ELISA Determinations (a) Cortical Levels In untreated PDAPP mice, the median level of total AP 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 Ap at 15 months which increased to 19,000 ng/g at 18 months. In contrast, AN1792-treated animals had 81%
less total AP at 15 months (1,600 ng/g) than the PBS-immunized group.
Significantly less (p=0.0001) total AP (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 AP that would otherwise be present. Similar results were obtained when cortical levels of A1342 were compared, namely that the AN1792-treated group contained much less Ap42, 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 Ali Levels (ng/g) in Cortex Age Total A1342 (n) Total A1342 (n) Total A1342 (n) 12 1,600 1,300 (10) 15 8,700 8,300 (10) 8,600 7,200 (9) 1,600 1,300* (10) 18 22,200 18,500 (10) 19,000 15,900 (12) 5,200** 4,000** (9) * p = 0.0412 ** p = 0.0001 (b) Hippocampal Levels In untreated PDAPP mice, median hippocampal levels of total Ap 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 AP, 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 AP42 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=0.002) at the 18-month evaluation (Table 3).
Table 3: Median AP Levels (ng/g) in Hippocampus Age Total AP42 (n) Total A1342 (n) Total Ap42 (n) 12 15,500 11,100 (10) 51,500 44,400 (10) 40,100 35,70 (9) 24,50 22,100 (10) 80_g00 64 700 (10) 65,400 57,10 (17) _50,90 38,900" (9) * p 0.0105 ** p = 0.0022 (c) Cerebellar Levels In 12-month untreated PDAPP mice, the median cerebellar level of total AP was 15 ng/g (Table 4). At 15 months, this median increased to 28 ng/g and by 18 months had risen to 35 ng/g. PBS-treated animals displayed median total AP
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 Ap at 15 months and significantly less (p=0.002) total AP
at 18 months (25 ng/g) than the corresponding PBS group (Table 4).
Table 4: Median Ap Levels (ng/g) in Cerebellum Age Total A13 (n) Total Ap (n) Total Ap (n) 12 15.6 (10) 15 27.7 (10) 20.8 (9) 21.7 (10) 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 A13 sequence and thus could be potentially impacted by the generation of an AN1792-directed immune response. In 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 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 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 i_tg 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 Ap-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 AP-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, AP
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 AP protein generates antibodies that recognize and bind in vivo to the AP 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 AP40, A1342, or A1340-1 (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 AI31-40 or A31-42 protein, with equal levels of incorporation for both proteins (Fig. 10, Upper Panel). There was no response to the AP40-1 reverse protein. Cells from control animals did not respond to any of the Ap 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 neuropathologic 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 AP peptide has therapeutic benefit in the treatment of AD.
IV. SCREEN OF AP FRAGMENTS
100 PDAPP mice age 9-11 months were immunized with 9 different regions of APP and A13 to determine which epitopes convey the efficacious response.
The 9 different immunogens and one control are injected i.p. as described above. The immunogens include four human AP peptide conjugates 1-12, 13-28, 32-42, 1-5, all coupled to sheep anti-mouse IgG via a cystine link; an APP polypeptide amino acids 592-695, aggregated human AP 1-40, and aggregated human AP 25-35, and aggregated rodent A1342. Aggregated A1342 and PBS were used as positive and negative controls, respectively. Ten mice were used per treatment group. Titers were monitored as above and mice were euthanized at the end of 4 months of injections. Histochemistry, AP
levels, and toxicology analysis was determined post mortem.
A. Materials and Methods 1. Preparation of Immunogens Preparation of coupled AP peptides: four human AP 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 AP
peptide using the crosslinking reagent sulfo-EMCS. The AP 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 A13-28 peptide derivative also had two glycine residues added prior to the carboxyl terminal cysteine as indicated.

A131-12 peptide NH2-DAEFREIDSGYEVC-COOH (SEQ ID NO:72) A31-5 peptide NH2-DAEFRC-COOH (SEQ ID NO:73) AP33-42 peptide N112-C-amino-heptanoic acid-GLMVGGVVIA-COOH
(SEQ ID NO:74) A013-28 peptide Ac-NH-111-1QKLVFFAEDVGSNKGGC-COOH
(SEQ ID NO:75) To prepare for the coupling reaction, ten mg of sheep anti-mouse IgG
(Jackson ImmunoResearch Laboratories) was dialyzed overnight against 10 niM
sodium borate buffer, pH 8.5. The dialyzed antibody was then concentrated to a volume of 2 mL
using an AmicoTri Centriprelitabe. Ten mg sulfo-EMCS
[N (e-maleimidocuproyloxy) succinirnide] (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 Presg Column, obtained from Pierce Chemicals) equilibrated with 0.1 M NaPO4, 5 niM 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 A.13 peptide was dissolved in 20 mL of 10 inM 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 inM
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 pm-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 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 AP 1-5 sheep anti-mouse conjugate was a pool of two conjugations, the rest were from a single preparation.

2. Preparation of aggregated AP 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 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 solution or suspension. Of the four, AN1528 was the only peptide soluble at this step. A

aliquot of 10X PBS (1X PBS: 0.15 M NaCl, 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 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 (APP) was constructed 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 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%.
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 AP 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 Af3 peptides derived from the human sequence, 1-5, 1-12, 13-28, and 33-42, each conjugated to sheep anti-mouse IgG; four aggregated Ap 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 g of each AP peptide in 200 I PBS or 200 g 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 1 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. AP and APP Levels in the Brain Following about four months of immunization with the various Af3 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 AP 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 AP peptide or APP of known concentrations in an ELISA format.
The median concentration of total AP 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 AP and AP1-42 levels which differed significantly from those of the control group (p <
0.05), those animals receiving AN1792, rodent A13 1-42 or the AP1-5 peptide conjugate as shown in Fig. 11. The median levels of total AP were reduced by 75%, 79% and 61%, respectively, compared to the control for these treatment groups. There were no discernable correlations between AP-specific antibody titers and Ap levels in the cortical region of the brain for any of the groups.
In the hippocampus, the median reduction of total Ap 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 AP1-42 or Ap1-5, the median total AP 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 AP1-42 were measured in the hippocampus, none of the treatment-induced reductions reached significance. Thus, due to the smaller AP burden in the cortex, changes in this region are a more sensitive indicator of treatment effects. The changes in AP levels measured by ELISA in the cortex are similar, but not identical, to the results from the immunohistochemical analysis (see below).
Total iv was also measured in the cerebellum, a region typically minimally affected with AD pathology. None of the median Ap concentrations of any of the groups immunized with the various AP 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 AP 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 AP sequence), and full-length forms (FL) of APP, while the second recognizes only APP-a. In contrast to the treatment-associated diminution of Ap 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 AP peptides are not depleting APP; rather the treatment effect is specific to AP.
In summary, total AP and A13 1-42 levels were significantly reduced in the cortex by treatment with AN1792, rodent Af31-42 or A131-5 conjugate. In the hippocampus, total AP was significantly reduced only by AN1792 treatment. No other treatment-associated changes in AP 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 AP peptide conjugates A131-5, A131-12, and A1313-28; two groups immunized with the full length AP
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.
12. There were significant reductions of amyloid burden in the cortical regions of three 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 (95%, p = 0.005) and the AP1-5 peptide conjugate (67%, p = 0.02).
The results obtained by quantitation of total AP or A131-42 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 AP 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 AP
aggregated into plaques. In contrast, the ELISA measures all forms of A13, both soluble and insoluble, monomeric and aggregated. Since the disease pathology is thought to be associated with the insoluble plaque-associated form of Ap, 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 AP is greater for plaque-associated than total Ap.
To determine if the Ap-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, AP-containing plaques were coated with endogenous IgG for animals immunized with the A13 peptide conjugates Ar31-5, A[31-12, and 28; and the full length AP aggregates AN1792 and AN1528. Brains from animals immunized with the other AP 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 AP1-42-binding antibody using a sandwich ELISA with plastic multi-well plates coated with Af31-42. As shown in Fig. 13, peak antibody titers were elicited following the fourth dose for those four immunogenic formulations which elicited the highest titers of AN1792-specific antibodies: AN1792 (peak GMT: 94,647), AN1528 (peak GMT:
88,231), AP1-12 conjugate (peak GMT: 47,216)and rodent A131-42 (peak GMT:
10,766).
Titers for these groups declined somewhat following the fifth and sixth doses.
For 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:
A[31-5 conjugate (peak GMT: 2,356), pBx6 (peak GMT: 1,986), A[313-28 conjugate (peak GMT: 1,183), AI333-42 conjugate (peak GMT: 658), A1325-35 (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 A131-5, AI313-28, A1325-35, A333-42 or rodent A131-42. These titers were about the same as those measured against Af31-42 except for the rodent AI31-42 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 the reduction of AP in the cortex.
4. Lymphoproliferative Responses A3-dependent lymphoproliferation was measured using spleen cells harvested approximately one week following the final, sixth, immunization.
Freshly harvested cells, 105 per well, were cultured for 5 days in the presence of A131-40 at a concentration of 5 [tM for stimulation. Cells from a subset of seven of the ten groups were also cultured in the presence of the reverse peptide, A[340-1. As a positive control, additional cells were cultured with the T cell mitogen, PHA, and, as a negative control, cells were cultured without added peptide.

Lymphocytes from a majority of the animals proliferated in response to PHA. There were no significant responses to the A1340-1 reverse peptide. Cells from animals immunized with the larger aggregated Ap peptides, AN1792, rodent A131-42 and AN1528 proliferated robustly when stimulated with A131-40 with the highest cpm in the recipients of AN1792. One animal in each of the groups immunized with Af31-12 conjugate, A313-28 conjugate and A1325-35 proliferated in response to A131-40.
The remaining groups receiving Af31-5 conjugate, A1333-42 conjugate pBx6 or PBS
had no animals with an AP-stimulated response. These results are summarized in Table 5 below.
Table 5 Immunogen Conjugate AP Amino Acids Responders Af31-5 Yes 5-mer 0/7 A[31-12 Yes I2-mer 1/8 A[313-28 Yes 16-mer 1/9 A1325-35 11-mer 1/9 A[333-42 Yes 10-mer 0/10 A[31-40 40-mer 5/8 A[31-42 42-mer 9/9 r AP1-42 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 AP-specific T
cell response in animals immunized with AP1-5 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 AP region. This hypothesis is supported by the very low incidence of proliferative responses for animals in each of these treatment groups. Since the Af31-5 conjugate was effective at significantly reducing the level of AP in the brain, in the apparent absence of AP-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 AP residues may be due to the poor immunogenicity of this particular preparation. The poor immunogenicity of the AP25-35 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 125 non-transgenic mice were immunized with 1001Ig A131-42, plus CFA/IFA adjuvant, and euthanized at 4-5 months. Blood was collected from immunized mice. IgG was 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 60-120 mg.
VI. Passive Immunization with Antibodies to AP
Groups of 7-9 month old PDAPP mice each were injected with 0.5 mg in PBS of polyclonal anti-AP or specific anti-AP monoclonals as shown below. All antibody preparations were purified to have low endotoxin levels. Monoclonals can be prepared against a fragment by injecting the fragment or longer form of AI3 into a mouse, preparing hybridomas and screening the hybridomas for an antibody that specifically binds to a desired fragment of AP without binding to other nonoverlapping fragments of AP.

Table 6 Antibody Epitope 10D5 Af3 1-12 266 Af3 13-28 Mouse polyclonal Anti-Aggregated Af342 anti-human Af342 Mice were injected ip as needed over a 4 month period to maintain a circulating antibody concentration measured by ELISA titer of greater than defined by ELISA to Af342 or other immunogen. Titers were monitored as above and mice were euthanized at the end of 6 months of injections. Histochemistry, Af3 levels and toxicology were performed post mortem. Ten mice were used per group.
Additional studies of passive immunization are described in Examples XI and XII below.
VII. Comparison of Different Adjuvants This example compares CFA, alum, an oil-in water emulsion and MPL for capacity to stimulate an immune response.
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 p.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 t.tg AN1792). Two groups received injections of a palmitoylated derivative of AN1792 (33 !Az) combined with a) PBS orb) 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 jig for all groups except the high dose alum group, which received 300 lig 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 Immunogens Two mg A1342 (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 100111 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 A131-42 was stored with desiccant as a lyophilized powder at -20 C.
A palmitoylated derivative of AN1792 was prepared by coupling palmitic anhydride, dissolved in dimethyl formamide, to the amino terminal residue of prior to removal of the nascent peptide from the resin by treatment with hydrofluoric acid.
To prepare formulation doses with Complete Freund's adjuvant (CFA) (group 2), 33 lig of AN1792 in 200 p.1 PBS was emulsified 1:1 (vol:vol) with CFA in a final volume of 400 [11 for the first immunization. For subsequent immunizations, the antigen was similarly emulsified with Incomplete Freund's adjuvant (IFA).
To prepare formulation 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 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 tig of AN1792 in 16.5 1 PBS, 50 idg of MPL (50 .1) and 162 1 of PBS were mixed in a borosilicate tube immediately before use.
To prepare formulation 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 g AN1792 in 250 pi (group 6). The mixture 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 p.m diameter standard latex bead when viewed under a microscope. The resulting suspension was opalescent, milky white. The emulsions were freshly prepared for each series of injections. For group 8, MPL in 0.2% triethylamine was added at a concentration of 50 fig per dose to the squalene and detergent mixture for emulsification as noted above. For the palmitoyl derivative (group 7), 33 fig per dose of palmitoyl-NH-A1-42 was added to squalene and 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, formulation doses with alum (groups 9 and 10), AN1792 in TM
PBS was added to Alhydrogel (aluminum hydroxide gel, Accurate, Westbury, NY) to reach concentrations of 33 jig (low dose, group 9) or 300 jig (high dose, group 10) AN1792 per 5 mg of alum in a final dose volume of 250 I. 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 Af342 were measured by ELISA as described in General Materials and Methods.
4. Tissue Preparation After about 14 weeks, all guinea pigs were euthanized by administering 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 Af3 protein using ELISA.
B. 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. 14, 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 3001,tg being more immunogenic at all time points than 33 g. At the peak of the antibody response, following the fourth immunization, the difference between the two doses was 43% with GMTs of about (33 g) and 3400 (300 g). The antibody response to 33 lig AN1792 plus MPL was very similar to that generated with almost a ten-fold higher dose of antigen (300 g) bound to alum. The addition of MPL to an o/w emulsion decreased the potency of the formulations 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 formulations, 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 emulsion 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. AP 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 fig, of AN1792 (group 10) and the PBS immunized control group (group 3). To measure the level of A13 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 A13 standard protein of known concentrations in an ELISA format. The levels of A13 protein in the hippocampus, the cortex and the cerebellum were very similar for all four groups despite the wide range of antibody responses to AP elicited by these formulations.
Mean AP
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 AP for almost three months in some of these animals did not alter the total Ap levels in their brains. The levels of AP in the CSF were also quite similar between the groups.
The lack of large effect of AN1792 immunization on endogenous Af3 indicates that the immune response is focused on pathological formations of A.
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 on days 0, 14, 28, 60, 90 and administered subcutaneously in a dose volume of 200 1. 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 Group Na Adjuvant Dose Antigen Dose (1-1g) 1 10 MPL 12.5 g AN1792 33 2 10 MPL 25 pig AN1792 33 3 10 MPL 50 g AN I 792 33 4 13 MPL 125 jig AN1792 33 13 MPL 50 g AN1792 150 6 13 MPL 50 ps AN1528 33 8 10 PBS None 9 10 Squalene 5% AN1792 33 emulsified 10 Squalene 5% AN1792 33 admixed 11 10 Alum 2 mg AN I 792 33 12 13 MPL + Alum 50 g/2 mg AN1792 33 13 10 QS-21 5 g AN1792 33 14 10 QS-21 10 g AN I 792 33 18 13 QS-21 + MPL 25 g/50 g AN1792 33 19 13 QS-21 + 25 g/2 mg AN1792 33 Alum Footnotes:
a Number of mice in each group at the initiation of the experiment.
b The adjuvants are noted. The buffer for all these formulations was PBS. For group 8, 5 there was no adjuvant and no antigen.

The ELISA titers of antibodies against AP42 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 The table shows that the highest titers were obtained for groups 4, 5 and 18, in which the 5 adjuvants were 125 ug MPL, 50 ug MPL and QS-21 plus MPL.
IX. Therapeutic Efficacy of Different Adjuvants 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 AP and to induce the immune-mediated clearance of amyloid 10 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 - _ AN 1792 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 QS-21, each combined with both antigens, and Freund's adjuvant (FA) combined with only AN1792. An additional group . = _ 5 was immunized with AN1792 formulated in PBS buffer plus the preservative thimerosarA
without adjuvant. A ninth group was immunized with PBS alone as a negative control.
Preparation of aggregated Al3 peptides: human A131-40 (AN1528;
California Peptides Inc., Napa, CA; Lot ME0541) and human AIM-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 nil 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 1 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 formulation doses with alum (Groups 1 and 5), A13 peptide in PBS was added to Alhydrogel (two percent aqueous aluminum hydroxide gel, Sargeant, Inc., Clifton, NJ) to reach concentrations of 100 jig AI3 peptide per 1 mg of alum. IOX
PBS was added to a final dose volume of 200 I in IX PBS. The suspension was then gently mixed for approximately 4 hr at RT prior to injection.
To prepare formulation doses for with MPL (Groups 2 and 6), lyophilized powder (Ribi ImmunoChem Research, Inc., Hamilton, MT; Lot 67039-E0896B) was added to 0.2% 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 jig of peptide per dose in 50 pl PBS, 50 jig of MPL per dose (50 p.1) and 100 Ill of PBS
per dose were mixed in a borosilicate tube immediately before use.
To prepare formulation doses with QS-21 (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 jig of peptide per dose in 50 1 PBS, 25 lug of QS-21 per dose in 25 I PBS and 125 I of PBS per dose were mixed in a borosilicate tube immediately before use.
To prepare formulation doses with Freund's Adjuvant (Group 4), 100 tig of AN1792 in 200 1 PBS was emulsified 1:1 (vol:vol) with Complete Freund's Adjuvant (CFA) in a final volume of 400 1 for the first immunization. For subsequent immunizations, the antigen was similarly emulsified with Incomplete Freund's Adjuvant (IFA). For the formulations containing the adjuvants alum, MPL or QS-21, 100 lig per dose of AN1792 or AN1528 was combined with alum (1 mg per dose) or MPL (50 g per dose) or QS-21 (25 lig per dose) in a final volume of 200 !A PBS and delivered by subcutaneous inoculation on the back between the shoulder blades. For the group receiving FA, 100 g of AN1792 was emulsified 1:1 (vol:vol) with Complete Freund's adjuvant (CFA) in a final volume of 400 I 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 without adjuvant, 10 g AN1792 was combined with 5 g thimerosal in a final volume of 50 .1 PBS and delivered subcutaneously. The ninth, control group received only 200 IA
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 AP and APP levels in brain and by immunohistochemical evaluation of the presence of amyloid plaques in brain sections. In addition, AP-specific antibody titers, and AP-dependent proliferative and cytokine responses were determined.
Table 9 shows that the highest antibody titers to AP1-42 were elicited with FA and AN1792, titers which peaked following the fourth immunization (peak GMT:
75,386) and then declined by 59% 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 28% of the peak value after the sixth immunization.
The peak mean titer generated with QS-21 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 QS-21 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.

Table 9 Geometric Mean Antibody Titers' Week of Bleed Treatment 3.3 5.0 9.0 13.0 17.0 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) QS-21/ 30 227 327 1,511 1,188 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) MPL/ 184 2,591 1,653 1,156 3,099 AN1528 (15/21) (20/21) (21/21) (20/20) (20/20) QS-21/ 29 221 51 820 2,994 AN1528 (1/22) (13/22) (4/22) (20/22) (21/22) PBS plus . 25 33 39 37 47 Thimerosal (0/16) (2/16) (4/16) (3/16) (4/16) (0/16) (0/16) (0/15) (0/12) (0/16) Footnotes:
a Geometric mean antibody titers measured against A131-42 b Number of responders per group The results of AN1792 or AN1528 treatment with various adjuvants, or thimerosal on cortical amyloid burden in 12-month old mice determined by ELISA
are shown in Fig. 15. In PBS control PDAPP mice, the median level of total A13 in the cortex at 12 months was 1,817 ng/g. Notably reduced levels of A13 were observed in mice treated with AN1792 plus CFA/IFA, AN1792 plus alum, AN1792 plus MPL and QS-21 plus AN1792. The reduction reached statistical significance (p<0.05) only for plus CFA/IFA. However, as shown in Examples I and III, the effects of immunization in reducing A13 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 plus QS-21 compositions will achieve statistical significance in treatment of older mice.
By contrast, the AN1792 plus the preservative thimerosal showed a median level of A13 about the same as that in the PBS treated mice. Similar results were obtained when cortical levels of A1342 were compared. The median level of A1342 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 QS-21 respectively, with the reduction achieving statistical significance (p=0.05) for the AN1792 CFA/IFA treatment group. The median level in the thimerosal treated mice was 1619 ng/g A1342.
A further therapeutic adjuvant/immunogen efficacy study was performed in 9 - 10.5 month old male and female heterozygous PDAPP transgenic mice. The duration of the study was 25 weeks with 29-40 animals per treatment group;
therefore the animals were 15 ¨ 16.5 months old at termination.. The treatment groups are identified in Table 10 below.
Adjuvant Immunogen Dilution Buffer Administration Group 1: MPL-SE AN1792-GCS (75 g) PBS SC
(250 [II) Group 2: ISA 51 AN1792-GCS (75 ag) PBS IP
(400 I) Group 3: QS21 AN1792-GCS (75 g) PBS SC
(250 1) Group 4: QS21 abbrev. AN1792-GCS (75 g) PBS SC
(250 1) Group 5: PBS SC
(250 1) Table 10 abbreviations: MAP ¨ multi-antigenic peptide; TT ¨ tetanus toxoid t-cell epitope (830-844); SQ ¨ subcutaneous; IP ¨ intraperitoneally; PBS ¨ phosphate, buffered saline; ISA-51 is a commercially available adjuvant similar to IFA;
GCS is a glycine/citrate/sucrose formulation, MPL-SE is MPL in a stabilized water and oil emulsion.
The immunization schedule was identical for all of the treatment groups except for Group 3, the QS21/AN1792 abbreviated schedule group. The mice were injected on weeks 0, 2, 4, 8, 12, 16, 20, 24, with bleeds on weeks 3, 5, 9, 13, 17, 21 and 25. Groups 1, 2, received eight injections and Group 3 received four injections during the 25-week period of the study. Group 4, the QS21/AN1792 abbreviated schedule, received injections on weeks 0, 2, 4, and 8 only. This group was not injected for the remainder of the study, although they were bled on the same bleed schedule as the rest of the study to follow titer decay. Groups 3 and 5, QS21/AN1792 and PBS respectively, served as the positive and negative controls for this study.
The titers were determined by the anti-AB antibody titer assay.
Group 1, the MPL-SE/AN1792 group, raised a peak geometric mean titer (GMT) of 17,100 at 9 weeks falling to a GMT of 10,000 at 25 weeks. Initially, the MPL-SE titers rose at a somewhat higher rate than the QS21/AN1792 control group (Group 4).
Group 2, the ISA 51/AN1792 group, produced high titers throughout the study reaching a GMT of over 100,000 for the last 9 weeks of the study.
Group 3, the QS21/AN1792 control group, reached its peak titer at 17 weeks with a GMT of 16,000. The titer then fell over the next 8 weeks to finish with a GMT of 8,700. One animal in this group failed to raise a titer over the entire course of the experiment.
Group 4, the QS21/AN1792 abbreviated injection schedule group, reached a peak titer of 7,300 at 13 weeks, five weeks after its final injection. The titer then fell to a GMT of 2,100 at the final bleed (25 weeks). As in the control group, one animal failed to raise a detectable titer, while another animal lost all titer by the end of the decay period.
Group 5, the PBS alone group, had no titers.
To evaluate the cortical AB levels, total AB and AB1-42 were measured by ELISA.
Briefly, one brain hemisphere was dissected for cortical, hippocampal, and cerebellar tissue followed by homogenization in 5M guanidine buffer and assayed for brain AB.
The cortical total AB and AB42 results are similar. A Mann-Whitney statistical analysis was performed to determine significance between the groups with a p value of 0.05 indicating a significant change in AB.

All treatment groups significantly lowered total AB levels as compared to the PBS control group (see Table 11). The MPL-SE/AN1792 group, showed the greatest change in AB, and it is significantly better than the other treatment groups.
The QS21/AN1792 abbreviated group, was similar in its overall change of AB to the control group that received all eight injections. The AB levels in the ISA

group, were similarly lowered compared to the CFA/IFA:MAP(AB1-7) group.
Table 11 Cortical AB levels PBS MPL-SE ISA QS-21 QS-21 (4) MEDIAN 7,335 1,236 3,026 2,389 2,996 (ng/g tissue) RANGE 550 ¨ 18,358 70 ¨ 3,977 23 ¨ 9,777 210 ¨ 11,167 24 ¨ 16,834 (ng/g tissue) p value ---- <0.0001 <0.0001 <0.0001 <0.0001 In conclusion, MPL-SE, ISA-51 and QS21 adjuvants combined with AN1792 are effective in inducing a sufficient immune response significantly to retard AB
deposition in the cortex.
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-1528-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 AN1792 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 treatment is well tolerated and at least substantially free of side effects.
XI. Therapeutic Treatment with Anti-A(3 antibodies This examples tests the capacity of various monoclonal and polyclonal antibodies to AP to inhibit accumulation of AP in the brain of heterozygotic transgenic mice.
1. Study Design Sixty male and female, heterozygous PDAPP transgenic mice, 8.5 to 10.5 months of age were obtained from Charles River Laboratory. The mice were sorted into six groups to be treated with various antibodies directed to A. Animals were distributed to match the gender, age, parentage and source of the animals within the groups as closely as possible. As shown in Table 10, the antibodies included four murine AP-specific monoclonal antibodies, 2H3 (directed to AP residues 1-12), 10D5 (directed to Af3 residues 1-16), 266 (directed to AP residues 13-28 and binds to monomeric but not to aggregated AN1792), 21F12 (directed to AP residues 33-42). A fifth group was treated with an AP-specific polyclonal antibody fraction (raised by immunization with aggregated AN1792). The negative control group received the diluent, PBS, alone without antibody.
The monoclonal antibodies were injected at a dose of about 10 mg/kg (assuming that the mice weighed 50 g). Injections were administered intraperitoneally every seven days on average to maintain anti-AP titers above 1000. Although lower titers were measured for mAb 266 since it does not bind well to the aggregated AN1792 used as the capture antigen in the assay, the same dosing schedule was maintained for this group. The group receiving monoclonal antibody 2H3 was discontinued within the first three weeks since the antibody was cleared too rapidly in vivo. Animals were bled prior to each dosing for the measurement of antibody titers. Treatment was continued over a six-month period for a total of 196 days. Animals were euthanized one week after the final dose.
Table 12 EXPERIMENTAL DESIGN
TreatmentNa Treatment Antibody Antibody Group Isotype Antibody Specificity 1 9 none NAb NA
(PBS alone) 2 10 Polyclonal A131-42 mixed 3 0 mAbc 2H3 A131-12 IgG1 4 8 mAb 10D5 A131-16 IgG1 5 6 mAb 266 A1313-28 IgG1 6 8 mAb 21F12 I A1333-42 IgG2a Footnotes a. Number of mice in group at termination of the experiment. All groups started with 10 animals per group.
b. NA: not applicable c. mAb: monoclonal antibody 2. Materials and Methods a. Preparation of the Antibodies The anti-AP polyclonal antibody was prepared from blood collected from two groups of animals. The first group consisted of 100 female Swiss Webster mice, 6 to 8 weeks of age. They were immunized on days 0, 15, and 29 with 100 lig of combined with CFA/IFA. A fourth injection was given on day 36 with one-half the dose of AN1792. Animals were exsanguinated upon sacrifice at day 42, serum was prepared and the sera were pooled to create a total of 64 ml. The second group consisted of 24 female mice isogenic with the PDAPP mice but nontransgenic for the human APP
gene, 6 to 9 weeks of age. They were immunized on days 0, 14, 28 and 56 with 100 [tg of AN1792 combined with CFA/IFA. These animals were also exsanguinated upon sacrifice at day 63, serum was prepared and pooled for a total of 14 ml. The two lots of sera were pooled. The antibody fraction was purified using two sequential rounds of precipitation with 50% saturated ammonium sulfate. The final precipitate was dialyzed against PBS and tested for endotoxin. The level of endotoxin was less than 1 EU/mg.
The anti-AP monoclonal antibodies were prepared from ascites fluid. The fluid was first delipidated by the addition of concentrated sodium dextran sulfate to ice-cold ascites fluid by stirring on ice to a reach a final concentration of 0.238%.
Concentrated CaCl2 was then added with stirring to reach a final concentration of 64mM.
This solution was centrifuged at 10,000 x g and the pellet was discarded. The supernatant was stirred on ice with an equal volume of saturated ammonium sulfate added dropwise.
The solution was centrifuged again at 10,000 x g and the supernatant was discarded. The pellet was resuspended and dialyzed against 20 mM Tris-HC1 , 0.4 M NaC1, pH
7.5. This fraction was applied to a Pharmacia FPLC Sepharose Q Column and eluted with a reverse gradient from 0.4 M to 0.275 M NaC1 in 20 mM Tris-HC1, pH 7.5.
The antibody peak was identified by absorbance at 280 nm and appropriate fractions were pooled. The purified antibody preparation was characterized by measuring the protein concentration using the BCA method and the purity using SDS-PAGE.
The pool was also tested for endotoxin. The level of endotoxin was less than 1 EU/mg. titers, titers less than 100 were arbitrarily assigned a titer value of 25.
3. AP and APP Levels in the Brain:
Following about six months of treatment with the various anti-AP antibody preparations, brains were removed from the animals following saline perfusion.
One hemisphere was prepared for immunohistochemical analysis and the second was used for the quantitation of AP and APP levels. To measure the concentrations of various forms of beta amyloid peptide and amyloid precursor protein (APP), the hemisphere was dissected and homogenates of the hippocampal, cortical, and cerebellar regions were prepared in 5M guanidine. These were serially diluted and the level of amyloid peptide or APP was quantitated by comparison to a series of dilutions of standards of AP peptide or APP of known concentrations in an ELISA format.
The levels of total AP and of AP1-42 measured by ELISA in homogenates of the cortex, and the hippocampus and the level of total AP in the cerebellum are shown in Tables 11, 12, and 13, respectively. The median concentration of total AP
for the control group, inoculated with PBS, was 3.6-fold higher in the hippocampus than in the cortex (median of 63,389 ng/g hippocampal tissue compared to 17,818 ng/g for the cortex). The median level in the cerebellum of the control group (30.6 ng/g tissue) was more than 2,000-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-Wood et al., 1997).
For the cortex, one treatment group had a median AP level, measured as A131-42, which differed significantly from that of the control group (p <
0.05), those animals receiving the polyclonal anti-AP antibody as shown in Table 13. The median level of AP1-42 was reduced by 65%, compared to the control for this treatment group.
The median levels of A131-42 were also significantly reduced by 55% compared to the control in one additional treatment group, those animals dosed with the mAb 10D5 (p =
0.0433).

Table 13 CORTEX
c' cz, --..
--.1 t4 Treatment 1 Na Medians Means Group ;
Total AP = A[342 Total A13 A1342 :
:
' LISA valueb P value' 1 % Change ELISA value 1 P
value i % Change !
ELISA value ELISA value PBS ' 9 17818 ' NAd NA 1 13802 ' NA 1 NA
16150+/-7456e 12621+/-5738 n , iv -..1 Polyclonal anti--.3 6160 A10055 -65 4892 0.0071 -65 5912+/-4492 4454+/-3347 H
- _ --, _. 0. + -I-____________________________________________________________________________ H
mAb 10D5 8 7915 0.1019 -56 6214 0.0433 -55 9695+/-6929 6943+/-3351 "

. _ H
mAb 2666 .-9144 1 I 0.1255 -49 8481 0.1255 -39 L 9204+/-9293 7489+/-6921 1 H
H
i .
I
H
mAb 2IF12 : 8 15158 1 0.2898 1 -15 . 13578 : 0.7003 -2 12481+/-7082 11005+/-6324 c7, 1 .
Footnotes:
a. Number of animals per group at the end of the experiment b. ng/g tissue 5 c. Mann Whitney analysis d. NA: not applicable 00 n e. Standard Deviation 1-3 --..
.o.
oo ,..
c) In the hippocampus, the median percent reduction of total AP associated with treatment with polyclonal anti-AP antibody (50%, p = 0.0055) was not as great as that observed in the cortex (65%) (Table 14). However, the absolute magnitude of the reduction was almost 3-fold greater in the hippocampus than in the cortex, a net reduction of 31,683 ng/g tissue in the hippocampus versus 11,658 ng/g tissue in the cortex. When measured as the level of the more amyloidogenic form of AP, A131-42, rather than as total A13, the reduction achieved with the polyclonal antibody was significant (p =
0.0025).
The median levels in groups treated with the mAbs 10D5 and 266 were reduced by 33%
and 21%, respectively.

Table 14 HIPPOCAMPUS

o -I
N
GO

Treatment : Na Medians 1 Means Group Total Ar. ! A1342 Total A13 A1342 ELISA 1 P % ELISA i P 1 %
ELISA value ELISA value valueb value' Change , value value Change n PBS 9 63389 NAd NA 54429 NA I NA
58351+/-13308e 52801+/-14701 i I.) Polyclonal 1 10 31706 I 0.0055 -50 1 27127 I 0.0025 -50 30058+/-22454 24853+/-18262 L.., co anti-A[342 ' L.., H
, I.) mAb 10D5 i 8 46779 0.0675 -26 1 36290 1 0.0543 -33 '44581+/-18632 36465+/-17146 0 I
1, , , H
mAb 266 16 48689 0.0990 -23 1 43034 i 0.0990 1 -21 1 36419+/-27304 32919+/-25372 H
I
I
H
mAb 21F12 ! 851563 0.7728 .
! -19 ! 47961 : 0.8099 1 -12 1 57327+/-28927 50305+/-23927 0, i 1 Footnotes:
a. Number of animals per group at the end of the experiment b. ng/g tissue c. Mann Whitney analysis .0 n d. NA: not applicable e. Standard Deviation , .1.

.4 Total A13 was also measured in the cerebellum (Table 15). Those groups dosed with the polyclonal anti-A and the 266 antibody showed significant reductions of the levels of total A13 (43% and 46%, p = 0.0033 and p = 0.0184, respectively) and that group treated with 10D5 had a near significant reduction (29%, p = 0.0675).

Table 15 CEREBELLUM
Treatment Na Medians Means Group =
Total AP Total AP
ELISA P % ELISA value =
value b value Change PBS 9 30.64 NAd NA = 40.00+/-31.89e --t- 1-=
Polyclonal 10 17.61 0.0033 -43 18.15+/-4.36 anti-A1342 t-mAb 10D5 : 8 21.68 0.0675 -29 27.29+/-19.43 mAb 266 6 ; 16.59 0.0184 -46 19.59+/-6.59 mAb 21F12 8 . 29.80 >0.9999 -3 32.88+/-9.90 Footnotes:
a. Number of animals per group at the end of the experiment b. ng/g tissue c. Mann Whitney analysis d. NA: not applicable e. Standard Deviation APP concentration was also determined by ELISA in the cortex and cerebellum from antibody-treated and control, PBS-treated 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 A13 sequence), and full-length forms (FL) of APP, while the second recognizes only APP-a. In contrast to the treatment-associated diminution of AP in a subset of treatment groups, the levels of APP were virtually unchanged in all of the treated compared to the control animals. These results indicate that the immunizations with AP antibodies deplete AP without depleting APP.

In summary, AP levels were significantly reduced in the cortex, hippocampus and cerebellum in animals treated with the polyclonal antibody raised against AN1792. To a lesser extent monoclonal antibodies to the amino terminal region of A131-42, specifically amino acids 1-16 and 13-28 also showed significant treatment effects.
4. Histochemical Analyses:
The morphology of AB-immunoreactive plaques in subsets of brains from mice in the PBS, polyclonal AB42, 21F12, 266 and 10D5 treatment groups was qualitatively compared to that of previous studies in which standard immunization procedures with A1342 were followed.
The largest alteration in both the extent and appearance of amyloid plaques occurred in the animals immunized with the polyclonal AB42 antibody. The reduction of amyloid load, eroded plaque morphology and cell-associated A13 immunoreactivity closely resembled effects produced by the standard immunization procedure. These observations support the ELISA results in which significant reductions in both total A13 and A1342 were = achieved by administration of the polyclonal AB42 antibody.
In similar qualitative evaluations, amyloid plaques in the 10D5 group were also reduced in number and appearance, with some evidence of cell-associated immunoreactivity. Relative to control-treated animals, the polyclonal Ig fraction against AB
and one of the monoclonal antibodies (10D5) reduced plaque burden by 93% and 81%, respectively (p<0.005). 21F12 appeared to have a relatively modest effect on plaque burden.
Micrographs of brain after treatment with pabABI-42 show diffuse deposits and absence of many of the larger compacted plaques in the pabA131_42 treated group relative to control treated animals.
5. Measurement of Antibody Titers:
A subset of three randomly chosen mice from each group were bled just prior to each intraperitoneal inoculation, for a total of 30 bleeds. Antibody titers were measured as Af31-42-binding antibody using a sandwich ELISA with plastic multi-well plates coated with A131-42 as described in detail in the General Materials and Methods. Mean titers for each bleed are shown in Figures 16-18 for the polyclonal antibody and the monoclonals 10D5 and 21F12, respectively. Titers averaged about 1000 over this time period for the polyclonal antibody preparation and were slightly above this level for the 10D5- and 21F12-treated animals.
6. Lymphoproliferative Responses:
AP-dependent lymphoproliferation was measured using spleen cells harvested eight days following the final antibody infusion. Freshly harvested cells, 105 per well, were cultured for 5 days in the presence of A131-40 at a concentration of 5 [tM for stimulation. As a positive control, additional cells were cultured with the T cell mitogen, PHA, and, as a negative control, cells were cultured without added peptide.
Splenocytes from aged PDAPP mice passively immunized with various anti-AP antibodies were stimulated in vitro with AN1792 and proliferative and cytokine responses were measured. The purpose of these assays was to determine if passive immunization facilitated antigen presentation, and thus priming of T cell responses specific for AN1792.
No AN1792-specific proliferative or cytokine responses were observed in mice passively immunized with the anti-AP antibodies.
XII: FURTHER STUDY OF PASSIVE IMMUNIZATION
In a second study, treatment with 10D5 was repeated and two additional anti-AB antibodies were tested, monoclonals 3D6 (A131_5) and 16C11 (AB33-42).
Control groups received either PBS or an irrelevant isotype-matched antibody (TM2a). The mice were older (11.5-12 month old heterozygotes) than in the previous study, otherwise the experimental design was the same. Once again, after six months of treatment, 10D5 reduced plaque burden by greater than 80% relative to either the PBS or isotype-matched antibody controls (p=0.003). One of the other antibodies against AB, 3D6, was equally effective, producing an 86% reduction (p=0.003). In contrast, the third antibody against the peptide, 16C11, failed to have any effect on plaque burden. Similar findings were obtained with AB42 ELISA
measurements. These results demonstrate that an antibody response against AB
peptide, in the absence of T cell immunity, is sufficient to decrease amyloid deposition in PDAPP mice, but that not all anti-AB antibodies are efficacious. Antibodies directed to epitopes comprising amino acids 1-5 or 3-7 of AP are particularly efficacious.
In summary, we have shown that passively administered antibodies against AB
reduced the extent of plaque deposition in a mouse model of Alzheimer's disease. When held at modest serum concentrations (25-70 g/ml), the antibodies gained access to the CNS at levels sufficient to decorate B-amyloid plaques. Antibody entry into the CNS
was not due to abnormal leakage of the blood-brain barrier since there was no increase in vascular permeability as measured by Evans Blue in PDAPP mice. In addition, the concentration of antibody in the brain parenchyma of aged PDAPP mice was the same as in non-transgenic mice, representing 0.1% of the antibody concentration in serum (regardless of isotype).
XIII: MONITORING OF ANTIBODY BINDING
To determine whether antibodies against AB could be acting directly within the CNS, brains taken from saline-perfused mice at the end of the Example XII, were examined for the presence of the peripherally-administered antibodies. Unfixed cryostat brain sections were exposed to a fluorescent reagent against mouse immunoglobulin (goat anti-mouse IgG-Cy3). Plaques within brains of the 10D5 and 3D6 groups were strongly decorated with antibody, while there was no staining in the 16C11 group. To reveal the full extent of plaque deposition, serial sections of each brain were first immunoreacted with an anti-AB antibody, and then with the secondary reagent. 10D5 and 3D6, following peripheral administration, gained access to most plaques within the CNS. The plaque burden was greatly reduced in these treatment groups compared to the 16C11 group. These data indicate that peripherally administered antibodies can enter the CNS where they can directly trigger amyloid clearance. It is likely that 16C11 also had access to the plaques but was unable to bind.

XIV: EX VIVO SCREENING ASSAY FOR ACTIVITY OF AN ANTIBODY AGAINST
AMYLOID DEPOSITS
To examine the effect of antibodies on plaque clearance, we established an ex vivo assay in which primary microglial cells were cultured with unfixed cryostat sections of either PDAPP mouse or human AD brains. Microglial cells were obtained from the cerebral cortices of neonate DBA/2N mice (1-3 days). The cortices were mechanically dissociated in HBSS- (Hanks' Balanced Salt Solution, Sigma) with 50 lig/m1DNase I (Sigma).
The dissociated cells were filtered with a 1001.tm cell strainer (Falcon), and centrifuged at 1000 rpm for 5 minutes. The pellet was resuspended in growth medium (high glucose DMEM, 10%FBS, 25ng/m1 rmGM-CSF), and the cells were plated at a density of 2 brains per T-75 plastic culture flask. After 7-9 days, the flasks were rotated on an orbital shaker at 200 rpm for 2h at 37 C. The cell suspension was centrifuged at 1000rpm and resuspended in the assay medium.
10-1.tm cryostat sections of PDAPP mouse or human AD brains (post-mortem interval < 3hr) were thaw mounted onto poly-lysine coated round glass coverslips and placed in wells of 24-well tissue culture plates. The coverslips were washed twice with assay medium consisting of H-SFM (Hybridoma-serum free medium, Gibco BRL) with 1%
FBS, glutamine, penicillin/streptomycin, and 5ng/m1rmGM-CSF (R&D). Control or anti-AB
antibodies were added at a 2x concentration (5 jig/m1 final) for 1 hour. The microglial cells were then seeded at a density of 0.8x 106 cells/ml assay medium. The cultures were maintained in a humidified incubator (37 C, 5%CO2) for 24hr or more. At the end of the incubation, the cultures were fixed with 4% paraformaldehyde and permeabilized with 0.1%
Triton-X100. The sections were stained with biotinylated 3D6 followed by a streptavidin /
Cy3 conjugate (Jackson ImmunoResearch). The exogenous microglial cells were visualized by a nuclear stain (DAPI). The cultures were observed with an inverted fluorescent microscope (Nikon, TE300) and photomicrographs were taken with a SPOT digital camera using SPOT software (Diagnostic instruments). For Western blot analysis, the cultures were extracted in 8M urea, diluted 1:1 in reducing tricine sample buffer and loaded onto a 16%
tricine gel (Novex). After transfer onto immobilon, blots were exposed to 5 jig/ml of the pabAB42 followed by an HRP-conjugated anti-mouse antibody, and developed with ECL
(Amersham) When the assay was performed with PDAPP brain sections in the presence of 16C11 (one of the antibodies against AB that was not efficacious in vivo), B-amyloid plaques remained intact and no phagocytosis was observed. In contrast, when adjacent sections were cultured in the presence of 10D5, the amyloid deposits were largely gone and the microglial cells showed numerous phagocytic vesicles containing A13. Identical results were obtained with AD brain sections; 10D5 induced phagocytosis of AD plaques, while 16C11 was ineffective. In addition, the assay provided comparable results when performed with either mouse or human microglial cells, and with mouse, rabbit, or primate antibodies against A13.
Table 16 shows whether binding and/or phagocytosis was obtained for several different antibody binding specificities. It can be seen that antibodies binding to epitopes within an 1-7 both bind and clear amyloid deposits, whereas antibodies binding to epitopes within amino acids 4-10 bind without clearing amyloid deposits. Antibodies binding to epitopes C-terminal to residue 10 neither bind nor clear amyloid deposits.

Table 16: Analysis of Epitope Specificity Antibody Staining Phagocytosis epitope isotype N-Term mab 3D6 1-5 IgG2b + +
10D5 3-6 IgG1 + +
22C8 3-7 IgG2a + +
6E10 5-10 IgG1 + -14A8 4-10 rat IgG1 + -18G11 10-18 rat IgG1- -266 16-24 IgG1- -22D12 18-21 IgG2b- -C-Term 2G3 -40 IgG1- -16C11 -40/-42 IgG1 - -21F12 -42 IgG2a - -Immune serum rabbit (CFA) 1-6 + +
mouse (CFA) 3-7 + +
mouse (QS-21) 3-7 + +
monkey (Q5-21) 1-5 + +
mouse (MAP1-7) + +
Table 17 shows results obtained with several antibodies against AB, comparing their abilities to induce phagocytosis in the ex vivo assay and to reduce in vivo plaque burden in passive transfer studies. Although 16C11 and 21F12 bound to aggregated synthetic AB peptide with high avidity, these antibodies were unable to react with B-amyloid plaques in unfixed brain sections, could not trigger phagocytosis in the ex vivo assay, and were not efficacious in vivo. 10D5, 3D6, and the polyclonal antibody against AB were active by all three measures. The 22C8 antibody binds more strongly to an analog form of natural A13 in which aspartic acid at positions 1 and 7 is replaced with iso-aspartic acid. These results show that efficacy in vivo is due to direct antibody mediated clearance of the plaques within the CNS, and that the ex vivo assay is predictive of in vivo efficacy.
The same assay has been used to test clearing of an antibody against a fragment of synuclein referred to as NAC. Synuclein has been shown to be an amyloid plaque-associated protein. An antibody to NAC was contacted with a brain tissue sample containing amyloid plaques, an microglial cells, as before. Rabbit serum was used as a control. Subsequent monitoring showed a marked reduction in the number and size of plaques indicative of clearing activity of the antibody.
Table 17 The ex vivo assay as predictor of in vivo efficacy.
Antibody Isotype Avidity for Binding to Ex vivo In vivo aggregated fl-amyloid efficacy efficacy AB (pM) plaques monoclonal 3D6 IgG2b 470 + + +
10D5 IgG1 43 + + +
16C11 IgG1 90 - -21F12 IgG2a 500 - -TM2a IgG1 - - - -polyclonal 1-42 mix 600 + + +
Confocal microscopy was used to confirm that AB was internalized during the course of the ex vivo assay. In the presence of control antibodies, the exogenous microglial cells remained in a confocal plane above the tissue, there were no phagocytic vesicles containing A13, and the plaques remained intact within the section. In the presence of 10D5, nearly all plaque material was contained in vesicles within the exogenous microglial cells.
To determine the fate of the internalized peptide, 10D5 treated cultures were extracted with 8M urea at various time-points, and examined by Western blot analysis. At the one hour time point, when no phagocytosis had yet occurred, reaction with a polyclonal antibody against AB
revealed a strong 4 kD band (corresponding to the AB peptide). AB
immunoreactivity decreased at day 1 and was absent by day 3. Thus, antibody-mediated phagocytosis of AB
leads to its degradation.
To determine if phagocytosis in the ex vivo assay was Fc-mediated, F(ab')2 fragments of the anti-AB antibody 3D6 were prepared. Although the F(ab')2 fragments retained their full ability to react with plaques, they were unable to trigger phagocytosis by microglial cells. In addition, phagocytosis with the whole antibody could be blocked by a reagent against murine Fc receptors (anti-CD16/32). These data indicate that in vivo clearance of AB occurs through Fe-receptor mediated phagocytosis.
XV: PASSAGE OF ANTIBODIES THROUGH BLOOD BRAIN BARRIER
This example determines the concentration of antibody delivered to the brain following intravenous injection into a peripheral tissue of either normal or PDAPP mice.
PDAPP or control normal mice were perfused with 0.9% NaCl. Brain regions (hippocampus or cortex) were dissected and rapidly frozen. Brain were homogenized in 0.1%
triton +
protease inhibitors. Immunoglobulin was detected in the extracts by ELISA.
Fab'2 Goat Anti-mouse IgG were coated onto an RIA plate as capture reagent. The serum or the brain extracts were incubated for 1 hr. The isotypes were detected with anti-mouse IgGI-HRP or IgG2a-HRP or IgG2b-HRP (Caltag). Antibodies, regardless of isotype, were present in the CNS at a concentration that is 1:1000 that found in the blood. For example, when the concentration of IgG1 was three times that of IgG2a in the blood, it was three times IgG2a in the brain as well, both being present at 0.1% of their respective levels in the blood. This result was observed in both transgenic and nontransgenic mice - so the PDAPP
does not have a uniquely leak blood brain barrier.
XVI: THERAPEUTIC EFFICACY OF AN AB PEPTIDE IN MAP CONFIGURATION
A therapeutic adjuvant/immunogen efficacy study was performed in 9 - 10.5 month old male and female heterozygous PDAPP transgenic mice to test the efficacy of a fusion protein comprising AB1-7 in tetrameric MAP configuration as described above. The duration of the study was 25 weeks with 29 ¨ 40 animals per treatment group;
therefore the animals were 15 ¨ 16.5 months old at termination. The methodology used in this study is the same as that in the therapeutic study of different adjuvants in Example VIII
above. . The treatment groups are identified in Table 18 below.

Table 18 Adjuvant Immunogen Dilution Buffer Administration Group 1: CFA/IFA MAP(AB 1-7:TT) (100 g) PBS IP (400 pi) Group 2: QS21 AN1792-GCS (75 jig) PBS SC (250 I) Group 3: PBS SC (250 I) Table abbreviations: MAP ¨ multi-antigenic peptide; TT ¨ tetanus toxoid t-cell epitope (830-844); SC ¨ subcutaneous; IP ¨ intraperitoneally; PBS ¨ phosphate buffered saline;
GCS is a glycine/citrate/sucrose formulation.
The immunization schedule was identical for all of the treatment groups. The mice were injected on weeks 0, 2, 4, 8, 12, 16, 20, 24, with bleeds on week 3, 5, 9, 13, 17, 21 and 25. Groups 1, 2, 3, 4, and 6 received eight injections Groups 2 and 3, QS21/AN1792 and PBS respectively, served as the positive and negative controls for this study.
The titers were determined by the anti-AB antibody titer assay.
Group 1, CFA/IFA:MAP(AB1-7:TT) group, had low titer levels. The peak GMT reached was only 1,200 at 13 weeks, falling to a GMT of 600 by week 25.
There were 3 of the 30 mice that did not raise any titer and another 7 mice that did not exceed a titer of 400 by the end of the study.
Group 2, the QS21/AN1792 control group, reached its peak titer at 17 weeks with a GMT of 16,000. The titer then fell over the next 8 weeks to finish with a GMT of 8,700. One animal in this group failed to raise a titer over the entire course of the experiment.
Group 3, the PBS alone group, had no titers.
Both treatment groups showed a significant lowing in cortical AB levels as compared to the PBS control group (see Table 19). The CFA/IFA:MAP(AB1-7) group, significantly lowered AB as compared to the PBS control group in spite of the relatively low titers of anti-AB antibodies.

Table 19 Cortical AB levels MEDIAN 7,335 3,692 2,389 (ng/g tissue) RANGE 550 ¨ 18,358 240 ¨ 10,782 210 ¨ 11,167 (ng/g tissue) p value 0.0003 <0.0001 In conclusion, the AB 1-7MAP immunogen is effective in inducing a sufficient immune response significantly to retard AB deposition in the cortex.
XVII. EPITOPE MAPPING OF IMMUNOGENIC RESPONSE TO A13 IN
MONKEYS
This example analyzes the response of a primate to immunization with AN1792 (i.e., A131-42). Eleven groups of monkeys (4/sex/group) were immunized with AN1792 (75 or 300 jig/dose) in combination with QS-21 adjuvant (50 or 100 lag/dose) or 5%
sterile dextrose in water (D5W, control group). All animals received IM
injections on one of three injection schedules as shown in Table 20 for a total of 4, 5 or 8 doses.
Serum samples (from 4 monkeys/sex/group) collected on Day 175 of the study and CSF samples (from 3 monkeys/sex/group) collected on Day 176 of the study (at the 6 month necropsy) were evaluated for their ability to bind to A131-40 peptide and APP.

CA 02370311 2002-05-13 e-=
Table 20: Group Assignments and Dose Levels Group Schedule' # Monkeys AN1792 Dose QS-21 Dose Dose Route -No. (M/F) ().g/dose) (t.ig/dose) 4 b 1 4/4 0 0 IM
' 2 1 , 4/4 Vehicle' 50 1M
3 1 , 4/4 , Vehicle 100 IM

=

a. Schedule 1, Dose Days I, 15, 29, 57, 85, 113, 141, 169; Schedule 2, Dose Days 1, 29, 57, 113, 169; Schedule 3, Dose Days 1,43, 85, 169 5 b. D5W injection control group c. Vehicle consists of the glycine/citrate/sucrose buffer which is the excipient for AN1792.
The exact array of linear peptides recognized by the antibodies in the serum samples from animals immunized with AN1792 was determined by an ELISA that measured 10 the binding of these antibodies to overlapping peptides that covered the entire Af31-42 sequence. Biotinylated peptides with partial sequences of AN1792 were obtained from Chiron Technologies as 10 amino acid peptides with an overlap of 9 residues and a step of one residue per peptide (synthesis No. 5366, No. 5331 and No. 5814). The first 32 peptides (from the eight amino acid position upstream of the N-terminal of AN1792 down to the twenty-fourth amino acid of AN1792) are biotinylated on the C-terminal with a linker of GGK. The last 10 peptides (repeating the thirty-second peptide from, the previous series) are biotinylated on the N-terminal with a linker consisting of :EGEG (SEQ ID
NO:76)).
The lyophilized biotinylated peptides were dissolved at a concentration of 5 mM in DMSO. These peptide stocks were diluted to 51.1,M in TTBS (0.05% Tween 20, 25 mM Tris HC1, 137 mM NaC1, 5.1 mM KC1, pH=7.5). 100 [1.1 aliquots of this 5 I.LM
solution were added in duplicate to streptavidin pre-coated 96-well plates (Pierce).
Plates were incubated for one hour at room temperature, then washed four times with TTBS. Serum samples were diluted in specimen diluent without azide to normalize titers, and 100 pi was added per well. These plates were incubated one hour at room temperature and then washed four times with TTBS. H'RP-conjugated goat anti-human antibody (Jackson ImmunoResearch) was diluted 1:10,000 in specimen diluent without azide and 100 41 was added per well. The plates were again incubated and washed. To develop the color reaction, TMB (Pierce), was added at 100 41 per well and incubated for 15 min prior to the addition of 30 41 of 2 N H2SO4 to stop the reaction.
The optical density was measured at 450 nm on a VmaXmor Spectramjcolorimetric plate reader.
Immunization with AN1792 resulted in the production of antibodies in 100%
of the animals in all of the dose groups by Day 175. Mean titers in the groups ranged from 14596 - 56084. There was a trend for titers to be higher within an immunization schedule in the presence of higher antigen and/or higher adjuvant concentration, but no statistically significant differences could be demonstrated due to the high variability in individual animal responses to the immunizations.
Sera which were positive for antibodies to AN1792 were also positive for antibodies to AP1-40. Mean titers in the groups ranged from 36867 - 165991, and as for anti-AN1792 titers, showed no statistically significant differences between groups at Day 175.
Binding to AN1792 showed a highly positive correlation (Spearman r = 0.8671) with binding ' to Ap1-40.
Of the 48 monkeys immunized on various schedules with AN1792, 33 yielded CSF samples of adequate volume and quality for analysis. Thirty-two (97%) of these monkeys had positive titers to AN1792. Titers ranged from 2-246, with a mean of 49.44 21.34. CSF anti-AN1792 levels were 0.18 0.11% of what was measured in the serum and demonstrated a highly positive correlation (Spearman r = 0.7840) with serum titers. No differences were seen across groups or between sexes in the percentage of antibody in the CSF. The level of antibody in the CSF is consistent with the passive transfer of peripherally generated antibody across the blood-brain-barrier into the central nervous system.
Testing of a subset of anti-AN1792 positive CSF samples demonstrated that, like the antibody in serum samples, antibody in the CSF cross-reacts with A131-40. Titers to AI31-40 showed a high correlation (Spearman r = 0.9634) to their respective AN1792 titers.
Testing of a subset of CSF samples with the highest titers to AN1792 showed no binding to APP, as for the serum antibodies.
When sera from Day 175 was tested against a series of overlapping 10-mer peptides, antibodies from all of the monkeys bound to the peptide whose sequence covered amino acids 1-10 of the AN1792 peptide (amino acids 653-672 of APP). In some animals, this was the only peptide to which binding could be measured (see Fig. 19).
In other animals, other reactivities could be measured, but in all cases the reactivity to the N-terminal peptide sequence was the predominant one. The additional reactivities fell into two groups. First and most common, was the binding to peptides centering around the N-terminal 1-10 AN1792 peptide (Figure 20). Binding of this type was directed to the peptides covering amino acids -1-8, -1-9, and 2-11 of the AN1792 peptide.
These reactivities, combined with that to the 1-10 peptide, represent the overwhelming majority of reactivity in all animals. Epitope mapping of individual animals over time indicates that the antibody reactivity to the 1-10 peptide proceeds the spread to the adjacent peptides. This demonstrates a strong biasing of the immune response to the N-terminus of the AN1792 peptide with its free terminal aspartic acid residue. The second minor detectable activity in some animals was binding to peptides located C-terminally to the major area and centered around peptides covering amino acids 7-16,11-20 and 16-25 of the peptide. These reactivities were seen in only 10-30% of the monkeys.
Variability in response between different animals (e.g., whether amino acids 1-10 were the exclusive or predominant reactive epitope) did not correlate with antigen/adjuvant dose, dosing schedule, or antibody titer, and is probably a reflection of each individual animal's genetic make-up.
XVIII. PREVENTION AND TREATMENT OF HUMAN SUBJECTS
A single-dose phase I trial is performed to determine safety in humans. 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) or mild cognitive impairment (MCI), 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.
XIX. 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 p.1 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 ETA plates) were coated with 100 p.1 of a solution containing either 10 Ag/m1 either Af342 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% 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, 0.6% 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% Tween 20. The second antibody, a goat anti-mouse Ig conjugated to horseradish peroxidase (obtained from Boehringer Mannheim), was added to the wells as 100111 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 I 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 I
of 2 M
H2SO4. The color intensity was then read on a Molecular Devices Vmax at (450 nm - 650 nm).
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 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.
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-FBS) and TM
- then homogenized in a 50 Centricon unit (Dako A/S, Denmark) in 1.5 ml PBS-FBS for 10 -rm sec at 100 rpm in a Medimachine (Dalco) followed by filtration through a 100 micron 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 0.15 M NH4CI, 1 M K1iCO3, 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 (J'RH Biosciences, Lenexa, KS) supplemented with 2.05 rnM L glutamine, 1% Penicillin/Streptomycin, and 10%
heat-inactivated FBS, for 96 hr at 37 C. Various AP peptides, AP1-16, Ap1-40, A31-42 or AP40-1 reverse sequence protein were also added at doses ranging from 5 to 0.18 micromolar in four steps. Cells in control wells were cultured with Concanavalin A (Con A) (Sigma, cat. #
C-5275, at 1 microgram/ml) without added protein. Cells were pulsed for the final 24 hr with 311-thymidine (1 p.Ci/well obtained from Amersham Corp., Arlington Heights IL). Cells TM TM, = _ were then harvested onto UniFilter plates and counted in a Top CounMicroplate 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 AP 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 TM
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 AP and APP. Previous experiments had shown that the analytes were stable under this storage condition, and that synthetic AP
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 Ap Levels The brain homogenates were diluted 1:10 with ice cold Casein Diluent (0.25%
casein, PBS, 0.05% sodium azide, 20 p.g/m1 aprotinin, 5 rnM EDTA pH 8.0, 10 g/m1 leupeptin) and then centrifuged at 16,000 x g for 20 min at 4 C. The synthetic AP 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 "total" Af3 sandwich ELISA utilizes monoclonal antibody monoclonal antibody 266, specific for amino acids 13-28 of AP (Seubert, et al.), as the capture antibody, and biotinylated monoclonal antibody 3D6, specific for amino acids 1-5 of Al3 (Johnson-Wood, et al), as the reporter antibody. The 3D6 monoclonal antibody does not recognize secreted APP or full-length APP, but detects only Ap species with an amino-terminal aspartic acid. This assay has a lower limit of sensitivity of-.5O ng/ml (11M) and shows no cross-reactivity to the endogenous murine AP protein at concentrations up to 1 ng/ml (Johnson-Wood et al., supra).
The Ap1-42 specific sandwich ELISA employs mAP 21F12, specific for amino acids 33-42 of AP (Johnson-Wood, et al.), as the capture antibody.
Biotinylated mAP
3D6 is also the reporter antibody in this assay which has a lower limit of sensitivity of about 125 fig/m1 (28 [tM, Johnson-Wood et al.). For the AP ELISAs, 100 11.1 of either mAp 266 (at 10 g/ml) or mAP 21F12 at (5 pig/m1) 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 1.t1 of 0.25% 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 0.05% Tween 20] prior to use. The samples and standards were added in triplicate aliquots of 100 .1 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 mAP 3D6, diluted to 0.5 1.tg/m1 in Casein Assay Buffer (0.25% casein, PBS, 0.05% 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 IA 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 AP. 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, (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-oc/FL and APP-a assays is mAb 8E5, raised against amino acids 444 to 592 of the APP695 form (Games et al., supra). The reporter mAb for the APP-cc/FL assay is mAb 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 mAb 16H9, raised to amino acids 605 to 611 of APP. The lower limit of sensitivity of the APP-aFL assay is about 11 ngiml (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, mAb 8E5 was coated onto the wells of 96-well EIA plates as described above for mAb 266. Purified, recombinant secreted APP-a was used as the reference standard for the APP-a assay and the APP-cc/FL
assay (Esch et at, 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 16119 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 1 hr at RT.
The fluorescent substrate 4-methyl-umbellipheryl-phosphate was added for a 30-mM RT
incubation and the plates were read on a CytofluOrTM 2350 fluorimeter (Millipore) at 365 rim excitation and 450 run emission.
7. Immunohistochemistry Brains were fixed for three days at 40C in 4% paraformaldehyde in PBS and then stored from one to seven days at 4 C in 1% parafonnaldehyde, 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 tim intervals, were incubated overnight with one of the following antibodies: (1) a biotinylated anti-Afl (mAb, 3D6, specific for human Af3) diluted to a concentration of 2 p.g/m1 in PBS and 1% horse serum;
or (2) a biotinylated mAb specific for human APP, 8E5, diluted to a concentration of 3 jig/m1 in PBS

and 1.0% horse serum; or (3) a mAb 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 mAb specific for CD1 1 b, MAC-1 antigen, (Chemicon International) diluted 1:100 with 0.25% Triton X-100 and 1% rabbit serum in TBS; or (5) a mAb 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 mAb specific for CD 43 (Pharmingen) diluted 1:100 with 1% rabbit serum in PBS or (7) a rat mAb specific for CD 45RA (Pharmingen) diluted 1:100 with 1% rabbit serum in PBS; or (8) a rat monoclonal A.13 specific for (Pharmingen) diluted 1:100 with 1% rabbit serum in PBS; or (9) a rat monoclonal AP
specific for CD 45 (Pharmingen) diluted 1:100 with 1% rabbit serum in PBS; or (10) a biotinylated polyclonal hamster AP specific for CD3e (Pharmingen) diluted 1:100 with 1%
rabbit serum in PBS or (11) a rat mAb specific for CD3 (Serotec) diluted 1:200 with 1%
rabbit serum in PBS; or with (12) a 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 peroxidase. They were next incubated overnight at 4 C with primary antibody. Sections reacted with 3D6 or 8E5 or CD3e mAbs were then reacted for one hr at RT with a horseradish peroxidase-avidin-biotin-complex with kit components "A"
and "B"
TM, diluted 1:75 in PBS (Vector Elite Standard Kit, Vector 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, 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% 3,3%
diaminobenzidine (DAB) at RT. Sections destined for incubation with the GFAP-, 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 antibody were incubated for 1 hr at RT with biotinylated TM-anti-mouse IgG made in horse (Vector Laboratories; Vectastain Elite 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 monoclonal antibody 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 MHC 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 TM
slides), air dried overnight, dipped in Propar (Anatech) and overlaid with coverslips using TM -Permount (Fisher) as the mounting medium.
To counterstain Ap plaques, a subset of the GFAP-positive sections were TM
mounted on Superfrost 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 Perrnount.
8. Image Analysis TM
A Videometrie 150 Image Analysis System (Oncor, Inc., Gaithersburg, MD) TM
linked to a Nikon Microphot-FX microscope through a CCD video camera and a Sony TrinitronTmmonitor 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 hippocampus was manually outlined and the total pixel area occupied by the hippocampus was calculated. The percent arnyloid burden was measured as:
(the fraction of the hippocampal area containing Afl deposits imrnunoreactive with mAb 3D6) x 100. Similarly, the percent neuritic burden was measured as: (the fraction of the hippocampal area containing dystrophic neurites reactive with monoclonal antibody 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 TM
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 rnicroglia. The image of the imrnunoreacted 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 microg,lia) X
100. For all image analyses, six sections at the level of the dorsal hippocampus, each separated by consecutive 240 pm 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.

_ _ _ From the foregoing it will be apparent that the invention provides for a number of uses. For example, the invention provides for the use of any of the antibodies to Af3 described above in the treatment, prophylaxis or diagnosis of amyloidogenic disease, or in the manufacture of a medicament or diagnostic composition for use in the same. Likewise, the invention provides for the use of any of the epitopic fragments of AP
described above for the treatment or prophylaxis of amyloidogenic disease or in the manufacture of a medicament for use in the same.

--..
' ....1 "

o . , TITER AT 50% MAXIMAL 0Ø
-Aggreated AB Injected mice Age ol PDAPP mouse 100 mouse 101 mouse 102 mouse 103 mouse 104 mouse 105 mouse 106 mouse 107 mouse 108 6 15000, 65000 30000 55000 300 15000 18000 50000 siiiiiiii n 10 40000 20000 60000, 50000 900 15000 50000 20000, 4-6-600 0 I.) 70000 25000 20000 u.) ---I
1-, W
H
H
IV

.
H
I
H
.
H
.
I
H

PBS Injected mice on both Immunogen 5 .
at 1/100 -.
Pt Age of PDAPP mouse 113 mouse 114 mouse115 mouse 116 mouse 117 el _____ ,-3 6 < 4x bkg , < 4x bkg < 4x bkg < 4x bk_g < 4x bkg c) 5 x bkg < 4x bkg < 4x bkg <
4x bkg < 4x bkg o o 12 < 4x bkg < 4x bkg < 4x bkg < 4x bkq < 4x bkq .
_ =
4, ,-, o SEQUENCE LISTING
- <110> Neuralab Limited <120> Prevention and Treatment of Amyloidogenic Disease <130> 08-893080CA
<140> Not Yet Known <141> 2000-05-26 <150> US 09/322,289 <151> 1999-05-28 <160> 77 <170> PatentIn Ver. 2.1 <210> 1 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 1 Glu Glu Ile Ser Glu Val Lys Met Asp Ala <210> 2 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 2 Glu Ile Ser Glu Val Lys Met Asp Ala Glu <210> 3 <211> 10 <212> PRT
<213> Artificial Sequence <220>

<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 3 Ile Ser Glu Val Lys Met Asp Ala Glu Phe <210> 4 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 4 Ser Glu Val Lys Met Asp Ala Glu Phe Arg <210> 5 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 5 Glu Val Lys Met Asp Ala Glu Phe Arg His <210> 6 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 6 Val Lys Met Asp Ala Glu Phe Arg His Asp <210> 7 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 7 Lys Met Asp Ala Glu Phe Arg His Asp Ser <210> 8 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 8 Met Asp Ala Glu Phe Arg His Asp Ser Gly <210> 9 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 9 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr <210> 10 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 10 Ala Glu Phe Arg His Asp Ser Gly Tyr Glu <210> 11 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 11 Glu Phe Arg His Asp Ser Gly Tyr Glu Val <210> 12 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 12 Phe Arg His Asp Ser Gly Tyr Glu Val His <210> 13 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 13 Arg His Asp Ser Gly Tyr Glu Val His His <210> 14 <211> 10 <212> PRT
<213> Artificial Sequence <220>

<.223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 14 His Asp Ser Gly Tyr Glu Val His His Gin <210> 15 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 15 Asp Ser Gly Tyr Glu Val His His Gin Lys <210> 16 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 16 Ser Gly Tyr Glu Val His His Gin Lys Leu <210> 17 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 17 Gly Tyr Glu Val His His Gln Lys Leu Val <210> 18 <,211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 18 Tyr Glu Val His His Gin Lys Leu Val Phe <210> 19 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 19 Glu Val His His Gin Lys Leu Val Phe Phe <210> 20 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 20 Val His His Gin Lys Leu Val Phe Phe Ala <210> 21 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 21 His His Gin Lys Leu Val Phe Phe Ala Glu <210> 22 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 22 His Gin Lys Leu Val Phe Phe Ala Glu Asp <210> 23 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 23 Gin Lys Leu Val Phe Phe Ala Glu Asp Val <210> 24 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 24 Lys Leu Val Phe Phe Ala Glu Asp Val Gly <210> 25 <211> 10 <212> PRT
<213> Artificial Sequence <220>

<,223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 25 Leu Val Phe Phe Ala Glu Asp Val Gly Ser <210> 26 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 26 Val Phe Phe Ala Glu Asp Val Gly Ser Asn <210> 27 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 27 Phe Phe Ala Glu Asp Val Gly Ser Asn Lys <210> 28 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 28 Phe Ala Glu Asp Val Gly Ser Asn Lys Gly <210> 29 F

<211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 29 Ala Glu Asp Val Gly Ser Asn Lys Gly Ala <210> 30 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 30 Glu Asp Val Gly Ser Asn Lys Gly Ala Ile <210> 31 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 31 Asp Val Gly Ser Asn Lys Gly Ala Ile Ile <210> 32 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 32 Val Gly Ser Asn Lys Gly Ala Ile Ile Gly <210> 33 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 33 Gly Ser Asn Lys Gly Ala Ile Ile Gly Leu <210> 34 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 34 Ser Asn Lys Gly Ala Ile Ile Gly Leu Met <210> 35 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 35 Asn Lys Gly Ala Ile Ile Gly Leu Met Val <210> 36 <211> 10 <212> PRT
<213> Artificial Sequence <220>

<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 36 Lys Gly Ala Ile Ile Gly Leu Met Val Gly <210> 37 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 37 Gly Ala Ile Ile Gly Leu Met Val Gly Gly <210> 38 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 38 Ala Ile Ile Gly Leu Met Val Gly Gly Val <210> 39 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 39 Ile Ile Gly Leu Met Val Gly Gly Val Val <210> 40 211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 40 Ile Gly Leu Met Val Gly Gly Val Val Ile <210> 41 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:10-mer peptide from AN1792 sequence (human Abeta42, beta-amyloid peptide) <400> 41 Gly Leu Met Val Gly Gly Val Val Ile Ala <210> 42 <211> 42 <212> PRT
<213> Homo sapiens <220>
<223> human Abeta42 beta-amyloid peptide <400> 42 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu Val His His Gin 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 Ile Ala <210> 43 <211> 13 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:influenza hemagglutinin HA-307-319 universal T-cell epitope <400> 43 - Pro Lys Tyr Val Lys Gin Asn Thr Leu Lys Leu Ala Thr <210> 44 <211> 13 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:PADRE universal T-cell epitope <220>
<221> MOD_RES
<222> (3) <223> Xaa = any amino acid <400> 44 Ala Lys Xaa Val Ala Ala Trp Thr Leu Lys Ala Ala Ala <210> 45 <211> 16 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:malaria CS, T3 epitope universal T-cell epitope <400> 45 Glu Lys Lys Ile Ala Lys Met Glu Lys Ala Ser Ser Val Phe Asn Val <210> 46 <211> 10 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:hepatitis B
surface antigen HBsAg-19-28 universal T-cell epitope <400> 46 Phe Phe Leu Leu Thr Arg Ile Leu Thr Ile 5210> 47 <211> 19 <212> PRT
= <213> Artificial Sequence <220>
<223> Description of Artificial Sequence:heat shock protein 65 hsp65-153-171 universal T-cell epitope <400> 47 Asp Gin Ser Ile Gly Asp Leu Ile Ala Glu Ala Met Asp Lys Val Gly Asn Glu Gly <210> 48 <211> 14 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:bacille Calmette-Guerin universal T-cell epitope <400> 48 Gln Val His Phe Gin Pro Leu Pro Pro Ala Val Val Lys Leu <210> 49 <211> 15 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:tetanus toxoid TT-830-844 universal T-cell epitope <400> 49 Gin Tyr Ile Lys Ala Asn Ser Lys Phe Ile Gly Ile Thr Glu Leu <210> 50 <211> 21 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:tetanus toxoid TT-947-967 universal T-cell epitope <400> 50 phe Asn Asn Phe Thr Val Ser Phe Trp Leu Arg Val Pro Lys Val Ser . Ala Ser His Leu Glu <210> 51 <211> 16 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:HIV gp120 Ti universal T-cell epitope <400> 51 Lys Gin Ile Ile Asn Met Trp Gin Glu Val Gly Lys Ala Met Tyr Ala <210> 52 <211> 22 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AN 90549 Abeta 1-7/tetanus toxoid 830-844 <400> 52 Asp Ala Glu Phe Arg His Asp Gin Tyr Ile Lys Ala Asn Ser Lys Phe Ile Gly Ile Thr Glu Leu <210> 53 <211> 28 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AN 90550 Abeta 1-7/tetanus toxoid 947-967 <400> 53 Asp Ala Glu Phe Arg His Asp Phe Asn Asn Phe Thr Val Ser Phe Trp Leu Arg Val Pro Lys Val Ser Ala Ser His Leu Glu 210> 54 <211> 43 <212> PRT
- <213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AN90542 Abeta 1-7/tetanus toxoid 830-844 + 947-967 <400> 54 Asp Ala Glu Phe Arg His Asp Gin Tyr Ile Lys Ala Asn Ser Lys Phe Ile Gly Ile Thr Glu Leu Phe Asn Asn Phe Thr Val Ser Phe Trp Leu Arg Val Pro Lys Val Ser Ala Ser His Leu Glu <210> 55 <211> 22 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AN 90576 Abeta 3-9/tetanus toxoid 830-844 <400> 55 Glu Phe Arg His Asp Ser Gly Gin Tyr Ile Lys Ala Asn Ser Lys Phe Ile Gly Ile Thr Glu Leu <210> 56 <211> 20 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AN90562 Abeta 1-7/peptide <220>
<221> MOD_RES
<222> (1)..(20) <223> Xaa = any amino acid <400> 56 Ala Lys Xaa Val Ala Ala Trp Thr Leu Lys Ala Ala Ala Asp Ala Glu ?he Arg His Asp <210> 57 <211> 34 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:AN90543 Abeta 1-7 x 3/peptide <220>
<221> MOD_RES
<222> (1)..(34) <223> Xaa = any amino acid <400> 57 Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Ala Lys Xaa Val Ala Ala Trp Thr Leu Lys Ala Ala Ala <210> 58 <211> 34 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <220>
<221> MOD_RES
<222> (3) <223> Xaa = any amino acid <400> 58 Ala Lys Xaa Val Ala Ala Trp Thr Leu Lys Ala Ala Ala Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp <210> 59 211> 20 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <220>
<221> MOD_RES
<222> (10) <223> Xaa = any amino acid <400> 59 Asp Ala Glu Phe Arg His Asp Ala Lys Xaa Val Ala Ala Trp Thr Leu Lys Ala Ala Ala <210> 60 <211> 24 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 60 Asp Ala Glu Phe Arg His Asp Ile Ser Gln Ala Val His Ala Ala His Ala Glu Ile Asn Glu Ala Gly Arg <210> 61 <211> 24 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 61 Phe Arg His Asp Ser Gly Tyr Ile Ser Gln Ala Val His Ala Ala His Ala Glu Ile Asn Glu Ala Gly Arg 210> 62 <211> 24 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 62 Glu Phe Arg His Asp Ser Gly Ile Ser Gin Ala Val His Ala Ala His Ala Glu Ile Asn Glu Ala Gly Arg <210> 63 <211> 34 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 63 Pro Lys Tyr Val Lys Gin Asn Thr Leu Lys Leu Ala Thr Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp <210> 64 <211> 27 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 64 Asp Ala Glu Phe Arg His Asp Pro Lys Tyr Val Lys Gln Asn Thr Leu Lys Leu Ala Thr Asp Ala Glu Phe Arg His Asp <210> 65 , 211> 34 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 65 Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Pro Lys Tyr Val Lys Gin Asn Thr Leu Lys Leu Ala Thr <210> 66 <211> 27 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 66 Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Pro Lys Tyr Val Lys Gln Asn Thr Leu Lys Leu Ala Thr <210> 67 <211> 79 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 67 Asp Ala Glu Phe Arg His Asp Pro Lys Tyr Val Lys Gin Asn Thr Leu Lys Leu Ala Thr Glu Lys Lys Ile Ala Lys Met Glu Lys Ala Ser Ser Val Phe Asn Val Gin Tyr Ile Lys Ala Asn Ser Lys Phe Ile Gly Ile Thr Glu Leu Phe Asn Asn Phe Thr Val Ser Phe Trp Leu Arg Val Pro = Lys Val Ser Ala Ser His Leu Glu Asp Ala Glu Phe Arg His Asp <210> 68 <211> 57 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 68 Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Asp Ala Glu Phe Arg His Asp Gin Tyr Ile Lys Ala Asn Ser Lys Phe Ile Gly Ile Thr Glu Leu Phe Asn Asn Phe Thr Val Ser Phe Trp Leu Arg Val Pro Lys Val Ser Ala Ser His Leu Glu <210> 69 <211> 44 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 69 Asp Ala Glu Phe Arg His Asp Gin Tyr Ile Lys Ala Asn Ser Lys Phe Ile Gly Ile Thr Glu Leu Cys Phe Asn Asn Phe Thr Val Ser Phe Trp Leu Arg Val Pro Lys Val Ser Ala Ser His Leu Glu <210> 70 <211> 51 <212> PRT
<213> Artificial Sequence 220>
<223> Description of Artificial Sequence:fusion protein with Abeta epitope <400> 70 Asp Ala Glu Phe Arg His Asp Gin Tyr Ile Lys Ala Asn Ser Lys Phe Ile Gly Ile Thr Glu Leu Cys Phe Asn Asn Phe Thr Val Ser Phe Trp Leu Arg Val Pro Lys Val Ser Ala Ser His Leu Glu Asp Ala Glu Phe Arg His Asp <210> 71 <211> 26 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:synuclein fusion protein <400> 71 Glu Gin Val Thr Asn Val Gly Gly Ala Ile Ser Gin Ala Val His Ala Ala His Ala Glu Ile Asn Glu Ala Gly Arg <210> 72 <211> 13 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Abeta 1-12 peptide with inserted Cys residue <400> 72 Asp Ala Glu Phe Arg His Asp Ser Gly Tyr Glu Val Cys <210> 73 <211> 6 <212> PRT
<213> Artificial Sequence <220>

,223> Description of Artificial Sequence:Abeta 1-5 peptide with inserted Cys residue . <400> 73 Asp Ala Glu Phe Arg Cys <210> 74 <211> 12 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Abeta 33-42 peptide with inserted Cys residue <220>
<221> MOD_RES
<222> (2) <223> Xaa = amino-heptanoic acid <400> 74 Cys Xaa Gly Leu Met Val Gly Gly Val Val Ile Ala <210> 75 <211> 19 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Abeta 13-28 peptide with two Gly residues added and inserted Cys residue <220>
<221> MOD_RES
<222> (1) <223> Xaa = N-acetyl His <400> 75 Xaa His Gin Lys Leu Val Phe Phe Ala Glu Asp Val Gly Ser Asn Lys Gly Gly Cys <210> 76 <211> 4 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:linker = <400> 76 Glu Gly Glu Gly <210> 77 <211> 7 <212> PRT
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:N-terminal segment of Abeta <400> 77 Asp Ala Glu Phe Arg His Asp

Claims (17)

WHAT IS CLAIMED IS:
1 . A pharmaceutical composition comprising a chimeric antibody that specifically binds to an epitope within residues 1-7 of A.beta., where the isotype of the antibody is human IgG1, and a pharmaceutically acceptable carrier.
2. The pharmaceutical composition according to claim 1, wherein the antibody specifically binds to an epitope within:
residues 1-6 of A.beta.;
residues 1-5 of A.beta.;
residues 3-6 of A.beta.; or residues 3-7 of A.beta..
3. The pharmaceutical composition according to claim 1 or 2, wherein the antibody specifically binds to an epitope comprising a free N-terminal residue of A.beta..
4. The pharmaceutical composition according to any one of claims 1 to 3, wherein the antibody binds to an epitope within residues of 1-7 of A.beta.
wherein residue 1 and/or residue 7 of A.beta. is iso-aspartic acid.
5. The pharmaceutical composition according to any one of claims 1 to 4, wherein the antibody is a polyclonal antibody or a monoclonal antibody.
6. The pharmaceutical composition according to any one of claims 1 to 5, wherein the antibody comprises two copies of the same pair of light and heavy chains.
7. The pharmaceutical composition according to any one of claims 1 to 6, wherein the antibody specifically binds to A.beta. peptide without binding to full length amyloid precursor protein (APP).
8. The pharmaceutical composition according to any one of claims 1 to 7 wherein the pharmaceutical composition additionally comprises at least one other antibody that binds to a different epitope of A.beta..
9. The pharmaceutical composition of any one of claims 1 to 8, wherein the carrier is a physiologically acceptable diluent for parenteral administration.
10. The composition of any one of claims 1 to 9, which is adapted to be administered intrA.beta.eritoneally, orally, intranasally, subcutaneously, intramuscularly, topically or intravenously.
11. The composition of any one of claims 1 to 10, which is adapted to be administered in multiple dosages over a period of at least six months.
12. The composition of any one of claims 1 to 11, which is adapted to be administered as a sustained release composition.
13. The composition according to any one of claims 1 to 12, wherein the composition is for use in preventing or treating a disease associated with amyloid deposits of A.beta. in the brain of a patient.
14. A use of the composition as defined in any one of claims 1 to 13 in the manufacture of a medicament for preventing or treating a disease associated with amyloid deposits of A.beta. in the brain of a patient.
15. A use of the composition as defined in any one of claims 1 to 13 for preventing or treating a disease associated with amyloid deposits of A.beta.
in the brain of a patient.
16. The use of claim 14 or 15, wherein the composition is for use at a dosage of antibody at least 1 mg/kg body weight of the patient.
17. The use of claim 16, wherein the dosage of antibody is at least 10 mg/kg body weight of the patient.
CA2370311A 1999-05-28 2000-05-26 Prevention and treatment of amyloidogenic disease Expired - Lifetime CA2370311C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/322,289 US7964192B1 (en) 1997-12-02 1999-05-28 Prevention and treatment of amyloidgenic disease
US09/322,289 1999-05-28
PCT/US2000/014810 WO2000072880A2 (en) 1999-05-28 2000-05-26 Prevention and treatment of amyloidogenic disease

Publications (2)

Publication Number Publication Date
CA2370311A1 CA2370311A1 (en) 2000-12-07
CA2370311C true CA2370311C (en) 2014-12-02

Family

ID=23254226

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2370311A Expired - Lifetime CA2370311C (en) 1999-05-28 2000-05-26 Prevention and treatment of amyloidogenic disease

Country Status (38)

Country Link
US (6) US7964192B1 (en)
EP (3) EP2108376B1 (en)
JP (2) JP5580504B2 (en)
KR (2) KR100961485B1 (en)
CN (2) CN1814287A (en)
AR (1) AR024074A1 (en)
AT (1) ATE434444T1 (en)
AU (1) AU5303100A (en)
BG (2) BG65882B1 (en)
BR (1) BR0011000A (en)
CA (1) CA2370311C (en)
CL (1) CL2010000018A1 (en)
CO (1) CO5170495A1 (en)
CZ (1) CZ304876B6 (en)
DE (2) DE10084643T1 (en)
EA (2) EA006630B1 (en)
EE (1) EE05671B1 (en)
ES (1) ES2624908T3 (en)
GB (2) GB2368794B (en)
HK (1) HK1045116B (en)
HR (2) HRP20010877B1 (en)
HU (1) HU230550B1 (en)
IL (2) IL146435A0 (en)
IS (1) IS6154A (en)
MX (1) MXPA01011621A (en)
MY (1) MY132784A (en)
NO (3) NO337301B1 (en)
NZ (1) NZ515403A (en)
PE (1) PE20010191A1 (en)
PL (1) PL200458B1 (en)
SG (3) SG140456A1 (en)
SK (1) SK287513B6 (en)
TR (2) TR200202231T2 (en)
TW (1) TWI263503B (en)
UA (1) UA81215C2 (en)
UY (1) UY26182A1 (en)
WO (1) WO2000072880A2 (en)
ZA (1) ZA200109487B (en)

Families Citing this family (173)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0866805A1 (en) 1995-12-12 1998-09-30 Karolinska Innovations AB PEPTIDE BINDING THE KLVFF-SEQUENCE OF AMYLOID $g(b)
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
US6743427B1 (en) 1997-12-02 2004-06-01 Neuralab Limited Prevention and treatment of 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
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
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20080050367A1 (en) 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
US20030147882A1 (en) 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
US6919075B1 (en) * 1999-09-03 2005-07-19 Ramot At Tel Aviv University Ltd. Bacteriophage displaying aβ epitopes and method of use
US20020094335A1 (en) * 1999-11-29 2002-07-18 Robert Chalifour Vaccine for the prevention and treatment of alzheimer's and amyloid related diseases
WO2001042306A2 (en) * 1999-12-08 2001-06-14 Mindset Biopharmaceuticals (Usa), Inc. Chimeric peptides (short b-cell epitope joined to a t-cell epitope) and their use for immunization
EP1752472A3 (en) * 1999-12-08 2007-04-25 Intellect Neurosciences, Inc. Chimeric amyloid beta peptides
CZ20022748A3 (en) 2000-02-21 2004-03-17 Pharmexa A/S Novel method for controlling amyloid content
ME00183B (en) * 2000-02-21 2011-02-10 Pharmexa As Novel method for down-regulation of amyloid
AU4178601A (en) 2000-02-24 2001-09-03 Univ Washington Humanized antibodies that sequester abeta peptide
IL152625A0 (en) * 2000-05-22 2003-06-24 Univ New York SYNTHETIC IMMUNOGENIC BUT NON-AMYLOIDOGENIC PEPTIDES HOMOLOGOUS TO AMYLOID beta FOR INDUCTION OF AN IMMUNE RESPONSE TO AMYLOID beta AND AMYLOID DEPOSITS
CA2414772C (en) 2000-07-07 2011-06-28 Jan Naslund Prevention and treatment of alzheimer's disease
AU1225702A (en) * 2000-09-19 2002-04-02 Evotec Neurosciences Gmbh Methods and compounds for treating brain amyloidosis
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
TWI255272B (en) * 2000-12-06 2006-05-21 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
CA2438832A1 (en) 2001-02-23 2002-09-06 Elan Pharmaceuticals, Inc. Transgenic knockouts of bace-1
WO2002088306A2 (en) * 2001-04-30 2002-11-07 Eli Lilly And Company Humanized antibodies
DE60229051D1 (en) 2001-04-30 2008-11-06 Lilly Co Eli HUMANIZED ANTIBODIES
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
CA2451998A1 (en) * 2001-08-17 2003-02-27 Eli Lilly And Company Anti-a.beta. antibodies
ES2391905T3 (en) * 2001-08-17 2012-11-30 Washington University Test method for Alzheimer's disease
CA2457145C (en) * 2001-08-17 2010-12-21 Washington University Assay method for alzheimer's disease
US20060073149A1 (en) * 2001-08-17 2006-04-06 Bales Kelly R Rapid improvement of cognition in condition related to abeta
JP2005500389A (en) * 2001-08-17 2005-01-06 イーライ・リリー・アンド・カンパニー Use of antibodies with high affinity for soluble Aβ to treat Aβ-related conditions and diseases
MY144532A (en) * 2001-08-20 2011-09-30 Lundbeck & Co As H Novel method for down-regulation of amyloid
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
US20070128191A1 (en) * 2001-12-26 2007-06-07 Universidad De Zaragoza Polyclonal antibodies, preparation method thereof and use of same
AR038568A1 (en) 2002-02-20 2005-01-19 Hoffmann La Roche ANTI-A BETA ANTIBODIES AND ITS USE
IL163812A0 (en) 2002-03-05 2005-12-18 Univ Ramot Immunizing composition and method for inducing an immune response against the -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
AU2003226356A1 (en) * 2002-04-12 2003-10-27 Ramot At Tel Aviv University Ltd. Prevention of brain inflammation as a result of induced autoimmune response
SG173214A1 (en) * 2002-04-19 2011-08-29 Univ Toronto Immunological methods and compositions for the treatment of alzhemier's disease
AU2003223474B2 (en) * 2002-04-25 2008-09-04 Eli Lilly And Company Method for treating anxiety and mood disorders in older subjects
WO2004013172A2 (en) * 2002-07-24 2004-02-12 Innogenetics N.V. Fragments of beta-amyloid as targets for vaccination against alzheimer disease
US9535076B2 (en) * 2002-09-12 2017-01-03 The Regents Of The University Of California Methods and compositions for eliciting an amyloid-selective immune response
JP2006519762A (en) * 2002-10-09 2006-08-31 ライナット ニューロサイエンス コーポレイション Methods for treating Alzheimer's disease using antibodies against amyloid β peptide and compositions thereof
TW200509968A (en) 2002-11-01 2005-03-16 Elan Pharm Inc Prevention and treatment of synucleinopathic disease
US20080014194A1 (en) 2003-10-31 2008-01-17 Elan Pharmaceuticals, Inc. 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
US8506959B2 (en) 2002-11-01 2013-08-13 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US8697082B2 (en) 2002-11-01 2014-04-15 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
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
AT413945B (en) * 2003-01-14 2006-07-15 Mattner Frank Dr Use of a compound having a binding capacity to an antibody specific for the natural N-terminal AB42 sequence, for preparing a vaccine for preventing and treating Alzheimer's 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
KR20050118669A (en) * 2003-02-01 2005-12-19 뉴랄랩 리미티드 Active immunization to generate antibodies to soluble a-beta
WO2004071408A2 (en) 2003-02-10 2004-08-26 Applied Molecular Evolution, Inc. Aβ BINDING MOLECULES
US8663650B2 (en) 2003-02-21 2014-03-04 Ac Immune Sa Methods and compositions comprising supramolecular constructs
CA2519511A1 (en) * 2003-03-17 2004-09-30 Wyeth Holdings Corporation Mutant cholera holotoxin as an adjuvant and an antigen carrier protein
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
EP1480041A1 (en) * 2003-05-22 2004-11-24 Innogenetics N.V. Method for the prediction, diagnosis and differential diagnosis of Alzheimer's disease
TWI306458B (en) 2003-05-30 2009-02-21 Elan Pharma Int Ltd 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
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
WO2005047860A2 (en) 2003-11-08 2005-05-26 Elan Pharmaceuticals, Inc. Antibodies to alpha-synuclein
EP2479184A3 (en) * 2003-12-17 2013-09-04 Janssen Alzheimer Immunotherapy Beta immunogenic peptide carrier conjugates and methods of producing the same
SG149013A1 (en) 2003-12-17 2009-01-29 Wyeth Corp Immunogenic peptide carrier conjugates and methods of producing same
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
AT500483B1 (en) * 2004-07-13 2006-01-15 Mattner Frank Dr Kit for prevention or treatment of Alzheimer's disease comprises means for inducing sequestration of amyloid beta in plasma and apheresis apparatus which exhibits an amyloid beta precursor protein receptor
US20060024667A1 (en) * 2004-07-29 2006-02-02 Karen Manucharyan Compositions and methods for Alzheimer's disease
EA016357B1 (en) 2004-07-30 2012-04-30 Ринат Ньюросайенс Корп. Antibodies directed against amyloid-beta peptide and methods using same
MX2007001679A (en) 2004-08-09 2007-05-23 Elan Pharm Inc Prevention and treatment of synucleinopathic and amyloidogenic disease.
EP1787998A4 (en) * 2004-08-11 2008-08-27 Mitsubishi Chem Corp Antibody and utilization of the same
EP1797119A2 (en) * 2004-08-11 2007-06-20 Wisconsin Alumni Research Foundation Method of reducing the effects of alpha beta and compositions therefore
US7300773B2 (en) 2004-08-27 2007-11-27 Wyeth Research Ireland Limited Production of TNFR-Ig
US7294484B2 (en) 2004-08-27 2007-11-13 Wyeth Research Ireland Limited Production of polypeptides
TWI374935B (en) 2004-08-27 2012-10-21 Pfizer Ireland Pharmaceuticals Production of α-abeta
WO2006042158A2 (en) * 2004-10-05 2006-04-20 Wyeth Methods and compositions for improving recombinant protein production
AR051800A1 (en) * 2004-12-15 2007-02-07 Wyeth Corp BETA ANTIBODIES USED TO IMPROVE COGNITION
WO2006066089A1 (en) 2004-12-15 2006-06-22 Neuralab Limited Humanized amyloid beta antibodies for use in improving cognition
GT200600031A (en) * 2005-01-28 2006-08-29 ANTI-BETA ANTIBODY FORMULATION
WO2006113347A2 (en) * 2005-04-13 2006-10-26 The University Of Tennessee Research Foundation Diagnostic and therapeutic potential of immune globulin intravenous (igiv) products
MY148086A (en) 2005-04-29 2013-02-28 Rinat Neuroscience Corp Antibodies directed against amyloid-beta peptide and methods using same
CN101171031B (en) * 2005-05-05 2011-09-28 默沙东公司 Peptide conjugate compositions and methods for the prevention and treatment of alzheimer's disease
EP2388274A1 (en) * 2005-06-17 2011-11-23 Janssen Alzheimer Immunotherapy Methods of purifying anti A Beta antibodies
PT1976877E (en) 2005-11-30 2014-04-29 Abbvie Inc Monoclonal antibodies against amyloid beta protein and uses thereof
SI2289909T1 (en) * 2005-11-30 2015-03-31 Abbvie Inc. Screening method, process for purifying of non-diffusible a-beta oligomers, selective antibodies against said non-diffusible a-beta oligomers and a process for manufacturing of said antibodies
EP1954718B1 (en) 2005-11-30 2014-09-03 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
CA2632822C (en) 2005-12-12 2018-08-28 Ruth Greferath A beta 1-42 specific monoclonal antibodies with therapeutic properties
UA99097C2 (en) 2005-12-12 2012-07-25 Ф. Хоффманн-Ля Рош Аг Antibodies against amyloid beta 4 with glycosylated in the variable region
WO2007090126A2 (en) * 2006-01-30 2007-08-09 Invitrogen Corporation Compositions and methods for detecting and quantifying toxic substances in disease states
CN101384279B (en) * 2006-02-24 2012-02-22 奇斯药制品公司 Anti-amyloid immunogenic compositions, methods and uses
EP2325209A3 (en) 2006-03-23 2011-08-03 BioArtic Neuroscience AB Improved protofibril selective antibodies and the use thereof
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
JP2009538924A (en) * 2006-06-01 2009-11-12 エラン ファーマシューティカルズ,インコーポレイテッド A neuroactive fragment of APP
SI2046833T1 (en) * 2006-07-14 2013-12-31 Ac Immune S.A. Humanized antibody against amyloid beta
EP3988566A1 (en) * 2006-07-14 2022-04-27 AC Immune SA Humanized antibody against amyloid beta
WO2008011348A2 (en) 2006-07-14 2008-01-24 Ac Immune S.A. Humanized antibody against amyloid beta
HUE033466T2 (en) * 2006-10-02 2017-12-28 Ac Immune Sa Humanized antibody against amyloid beta
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
EA017611B1 (en) * 2007-01-05 2013-01-30 Юнивэсэти Оф Цюрих Method for preparing a disorder-associated protein specific binding molecule or an antibody or a binding fragment or at least one immunoglobulin chain, binding said protein (variants), a binding molecule, an antibody, an immunoglobulin chain or a binding fragment thereof (variants), an antigen, a polynucleotide, vector and a host-cell, a composition comprising them and a kit and a method of diagnosing or treating neurological disorders by using said subjects (variants)
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
KR101160385B1 (en) 2007-01-18 2012-07-10 일라이 릴리 앤드 캄파니 PEGYLATED Aß FAB
US8147833B2 (en) * 2007-02-23 2012-04-03 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
CA2678963C (en) 2007-02-23 2018-05-01 Elan Pharmaceuticals, Inc. Prevention and treatment of synucleinopathic and amyloidogenic disease
EP3067066B1 (en) 2007-02-23 2019-03-27 Prothena Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
JP5930573B2 (en) 2007-03-01 2016-06-15 プロビオドルグ エージー New use of glutaminyl cyclase inhibitors
DK2142514T3 (en) 2007-04-18 2015-03-23 Probiodrug Ag Thiourea derivatives as glutaminyl cyclase inhibitors
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
CA2685372A1 (en) 2007-05-02 2008-11-13 F. Hoffmann-La Roche Ag Method for stabilizing a protein
US8048420B2 (en) 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
US8613923B2 (en) 2007-06-12 2013-12-24 Ac Immune S.A. Monoclonal antibody
EP2182983B1 (en) * 2007-07-27 2014-05-21 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases with humanised anti-abeta antibodies
EP2020602A1 (en) * 2007-08-02 2009-02-04 Araclon Biotech High sensitivty immunoassays and kits for the determination of peptides and proteins of biological interest
WO2009048539A2 (en) 2007-10-05 2009-04-16 Genentech, Inc. Monoclonal antibody
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
PT2237803E (en) 2007-12-28 2015-10-16 Prothena Biosciences Ltd Treatment and prophylaxis of amyloidosis
EP2106802A1 (en) * 2008-04-02 2009-10-07 SIGMA-TAU Industrie Farmaceutiche Riunite S.p.A. Modified peptides as synthetic vaccines in amyloid-associated disease
AT506819B1 (en) * 2008-06-12 2011-06-15 Affiris Forschungs Und Entwicklungs Gmbh VACCINE FOR THE TREATMENT OF ALZHEIMER DISEASE
AT509611B1 (en) * 2008-06-12 2012-04-15 Affiris Forschungs-Und Entwicklungs Gmbh VACCINE
EP2145898A1 (en) * 2008-07-15 2010-01-20 CHIESI FARMACEUTICI S.p.A. Anti-amyloid immunogenic compositions, methods and uses
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
EP3521309A1 (en) 2008-12-19 2019-08-07 Biogen International Neuroscience GmbH Human anti-alpha-synuclein antibodies
US8614297B2 (en) 2008-12-22 2013-12-24 Hoffmann-La Roche Inc. Anti-idiotype antibody against an antibody against the amyloid β peptide
AU2010221418B2 (en) 2009-03-02 2015-06-04 Dignity Health Diagnostic devices and methods of use
CA2772488C (en) 2009-09-11 2018-04-17 Probiodrug Ag Heterocyclic derivatives as inhibitors of glutaminyl cyclase
JP6026284B2 (en) 2010-03-03 2016-11-16 プロビオドルグ エージー Inhibitors of glutaminyl cyclase
BR112012022102A2 (en) 2010-03-03 2017-01-10 Boehringer Ingelheim Int a-beta binding polypeptides.
US8269019B2 (en) 2010-03-10 2012-09-18 Probiodrug Ag Inhibitors
JP2013523182A (en) 2010-04-15 2013-06-17 アボット・ラボラトリーズ Amyloid beta-binding protein
EP2560953B1 (en) 2010-04-21 2016-01-06 Probiodrug AG Inhibitors of glutaminyl cyclase
RU2607368C2 (en) 2010-07-30 2017-01-10 Ац Иммуне С.А. Safe and functional humanized antibodies
US9289488B2 (en) 2010-08-12 2016-03-22 Ac Immune Sa Vaccine engineering
EP2603524A1 (en) 2010-08-14 2013-06-19 AbbVie Inc. Amyloid-beta binding proteins
US9687447B2 (en) 2010-10-26 2017-06-27 Ac Immune S.A. Method for preparing liposome-based constructs
US11231426B2 (en) 2010-11-29 2022-01-25 Inven2 As Methods and compositions for monitoring phagocytic activity
US8530670B2 (en) 2011-03-16 2013-09-10 Probiodrug Ag Inhibitors
TR201815563T4 (en) 2011-06-23 2018-11-21 Biogen Int Neuroscience Gmbh Anti-alpha synuclein binding molecules.
RU2639509C2 (en) 2011-06-27 2017-12-21 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. Micro-rna-biomarkers indicating alzheimer's disease
WO2013012811A2 (en) 2011-07-19 2013-01-24 New York University Immunotherapeutic modulation of amyloidogenic disease using non-fibrillogenic, non-amyloidogenic polymerized proteins and peptides
WO2013013056A1 (en) 2011-07-19 2013-01-24 New York University Method for treating amyloid disease
GB201113570D0 (en) 2011-08-05 2011-09-21 Glaxosmithkline Biolog Sa Vaccine
ES2937409T3 (en) * 2011-10-28 2023-03-28 Prothena Biosciences Ltd Humanized antibodies that recognize alpha-synuclein
JP2015504674A (en) 2012-01-11 2015-02-16 アリゾナ ボード オブ リージェンツ ア ボディ コーポレート オブ ザ ステイト オブ アリゾナ アクティング フォー アンド オン ビハーフ オブ アリゾナ ステイト ユニバーシティーArizona Board Of Regents, A Body Corporate Of The State Of Arizona Acting For And On Behalf Of Arizona State University Bispecific antibody fragments of neurological disease proteins and methods of use
MX366972B (en) * 2012-01-20 2019-08-01 Ananth Annapragada Methods and compositions for objectively characterizing medical images.
AU2013287119A1 (en) * 2012-07-03 2015-01-22 Janssen Alzheimer Immunotherapy C-terminal and central epitope a-beta antibodies
EP2892353A4 (en) * 2012-09-10 2016-10-26 Yeda Res & Dev Individualized immunomodulation therapy for neurodegenerative disorders, cns injury and age-related dementia
EA035943B1 (en) 2013-03-13 2020-09-03 Протена Биосайенсес Лимитед Antibody binding to human tau protein
US9102752B2 (en) 2013-03-15 2015-08-11 United Biomedical, Inc. Peptide vaccine for prevention and immunotherapy of dementia of the Alzheimer's type
EP2873679A1 (en) 2013-11-13 2015-05-20 F.Hoffmann-La Roche Ag Camelid single-domain antibody directed against amyloid bêta and methods for producing conjugates thereof
EP2873680A1 (en) 2013-11-13 2015-05-20 F.Hoffmann-La Roche Ag Oligopeptide and methods for producing conjugates thereof
WO2015165961A1 (en) 2014-04-29 2015-11-05 Affiris Ag Treatment and prevention of alzheimer's disease (ad)
CN104122400B (en) * 2014-07-01 2016-03-02 上海依科赛生物制品有限公司 A kind of human beta-amyloid detection kit and uses thereof
AU2015286824A1 (en) * 2014-07-07 2017-02-09 AbbVie Deutschland GmbH & Co. KG Immunogenic products based on mutein amyloid SS (ASS) amino acid sequences and uses thereof
SI3166970T1 (en) 2014-07-10 2021-09-30 Bioarctic Ab Improved a-beta protofibril binding antibodies
MA41115A (en) 2014-12-02 2017-10-10 Biogen Int Neuroscience Gmbh ALZHEIMER'S DISEASE TREATMENT PROCESS
LT3313879T (en) 2015-06-24 2022-03-25 F. Hoffmann-La Roche Ag Anti-transferrin receptor antibodies with tailored affinity
NZ741067A (en) 2015-10-02 2023-07-28 Hoffmann La Roche Bispecific anti-human cd20/human transferrin receptor antibodies and methods of use
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
CA3004498A1 (en) * 2015-11-09 2017-05-18 Neil R. Cashman Amyloid beta epitopes and antibodies thereto
CN108350052A (en) * 2015-11-09 2018-07-31 英属哥伦比亚大学 Epitope in amyloid beta intermediate region and its conformation antibodies selective
CA3004482A1 (en) 2015-11-09 2017-05-18 Neil R. Cashman N-terminal epitopes in amyloid beta and conformationally-selective antibodies thereto
US10188749B2 (en) 2016-04-14 2019-01-29 Fred Hutchinson Cancer Research Center Compositions and methods to program therapeutic cells using targeted nucleic acid nanocarriers
CA3022515A1 (en) 2016-05-02 2017-11-09 Prothena Biosciences Limited Antibodies recognizing tau
US10752679B2 (en) 2016-05-02 2020-08-25 Prothena Biosciences Limited Tau immunotherapy
US10906964B2 (en) 2016-05-02 2021-02-02 Prothena Biosciences Limited Antibodies recognizing tau
CN110352252A (en) * 2016-09-22 2019-10-18 威廉马歇莱思大学 The molecular hybridization probe for capturing and analyzing for complex sequence
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
CN110881274A (en) 2017-05-02 2020-03-13 普罗塞纳生物科学有限公司 Antibodies recognizing TAU
CA3073066A1 (en) 2017-08-22 2019-02-28 Biogen Ma Inc. Pharmaceutical compositions containing anti-beta amyloid antibodies
ES2812698T3 (en) 2017-09-29 2021-03-18 Probiodrug Ag Glutaminyl cyclase inhibitors
PE20212324A1 (en) 2019-03-03 2021-12-14 Prothena Biosciences Ltd ANTIBODIES THAT RECOGNIZE TAU

Family Cites Families (364)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE878999A (en) * 1979-09-26 1980-03-26 Herstal Sa COMPOSITE GUN AND METHOD FOR THE PRODUCTION THEREOF
US4902506A (en) 1983-07-05 1990-02-20 The University Of Rochester Immunogenic conjugates
CH652145A5 (en) 1982-01-22 1985-10-31 Sandoz Ag METHOD FOR IN VITRO PRODUCTION OF HYBRID OMEN WHAT human monoclonal antibodies GENERATE.
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4634666A (en) 1984-01-06 1987-01-06 The Board Of Trustees Of The Leland Stanford Junior University Human-murine hybridoma fusion partner
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
US5618920A (en) 1985-11-01 1997-04-08 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
EP0279582A3 (en) 1987-02-17 1989-10-18 Pharming B.V. Dna sequences to target proteins to the mammary gland for efficient secretion
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
EP0307434B2 (en) 1987-03-18 1998-07-29 Scotgen Biopharmaceuticals, Inc. Altered antibodies
JPS63245689A (en) * 1987-03-31 1988-10-12 Suntory Ltd Human amyloid-related protein monoclonal antibody
US4883666A (en) * 1987-04-29 1989-11-28 Massachusetts Institute Of Technology Controlled drug delivery system for treatment of neural disorders
US5229490A (en) 1987-05-06 1993-07-20 The Rockefeller University Multiple antigen peptide system
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5583112A (en) 1987-05-29 1996-12-10 Cambridge Biotech Corporation Saponin-antigen conjugates and the use thereof
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
US5869054A (en) 1987-06-24 1999-02-09 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of autoantigens
US5849298A (en) 1987-06-24 1998-12-15 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of bovine myelin
US5571499A (en) 1987-06-24 1996-11-05 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
US5641474A (en) 1987-06-24 1997-06-24 Autoimmune, Inc. Prevention of autoimmune diseases by aerosol 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
US5004697A (en) 1987-08-17 1991-04-02 Univ. Of Ca Cationized antibodies for delivery through the blood-brain barrier
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
US5231000A (en) * 1987-10-08 1993-07-27 The Mclean Hospital Antibodies to A4 amyloid peptide
US5089603A (en) 1989-06-21 1992-02-18 Tanox Biosystems, Inc. Antigenic epitopes present on membrane-bound but not secreted iga
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
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
ES2055785T3 (en) 1989-01-17 1994-09-01 Eniricerche Spa SYNTHETIC PEPTIDES AND THEIR USE AS UNIVERSAL VEHICLES FOR THE PREPARATION OF IMMUNOGENOUS CONJUGATES SUITABLE FOR THE DEVELOPMENT OF SYNTHETIC VACCINES.
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
HU212924B (en) 1989-05-25 1996-12-30 Chiron Corp 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
IT1237764B (en) 1989-11-10 1993-06-17 Eniricerche Spa SYNTHETIC PEPTIDES USEFUL AS UNIVERSAL CARRIERS FOR THE PREPARATION OF IMMUNOGENIC CONJUGATES AND THEIR USE FOR THE DEVELOPMENT OF SYNTHETIC VACCINES.
US5633076A (en) 1989-12-01 1997-05-27 Pharming Bv Method of producing a transgenic bovine or transgenic bovine embryo
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
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
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
ATE153534T1 (en) 1990-04-27 1997-06-15 John Mcmichael METHOD AND COMPOSITION FOR TREATING CNS DISEASES CAUSED BY ABNORMAL BETA-AMYLOID PROTEIN
GB9009548D0 (en) * 1990-04-27 1990-06-20 Celltech Ltd Chimeric antibody and method
US5753624A (en) 1990-04-27 1998-05-19 Milkhaus Laboratory, Inc. Materials and methods for treatment of plaquing disease
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
EP0600866B1 (en) 1990-06-01 1997-12-03 Chiron Corporation Compositions and methods for identifying biologically active molecules
US5593970A (en) 1990-06-11 1997-01-14 Biochem Pharma Inc. Heterocyclic anthracycline analogs
CA2085127C (en) 1990-06-15 2002-12-10 Barbara Cordell Transgenic non-human mammal displaying the amyloid-forming pathology 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
CA2109602C (en) 1990-07-10 2002-10-01 Gregory P. Winter Methods for producing members of specific binding pairs
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
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
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
ES2246502T3 (en) 1990-08-29 2006-02-16 Genpharm International, Inc. TRANSGENIC NON-HUMAN ANIMALS ABLE TO PRODUCE HETEROLOGICAL ANTIBODIES.
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
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)
ATE175118T1 (en) 1990-10-05 1999-01-15 Medarex Inc TARGETED IMMUNOSTIMULATION WITH BISPECIFIC SUBSTANCES
GB9023352D0 (en) 1990-10-26 1990-12-05 Lynxvale Ltd Vaccinia vectors,vaccinia genes and expression products thereof
US5877015A (en) 1991-01-21 1999-03-02 Imperial College Of Science, Technology Of Medicine APP770 mutant in alzheimer's disease
EP0501882B1 (en) 1991-03-01 2000-07-12 Merial Method of anti LHRH immunoneutralization of non-castrated male animals and peptide therefore
US5192753A (en) 1991-04-23 1993-03-09 Mcgeer Patrick L Anti-rheumatoid arthritic drugs in the treatment of dementia
JPH06500128A (en) 1991-05-08 1994-01-06 シュバイツ・ゼルム―・ウント・インプフィンスティテュート・ベルン Immune stimulating and immunoenhancing reconstituted influenza virosomes and vaccines containing the same
US5858657A (en) 1992-05-15 1999-01-12 Medical Research Council Methods for producing members of specific binding pairs
DE69230142T2 (en) 1991-05-15 2000-03-09 Cambridge Antibody Tech METHOD FOR PRODUCING SPECIFIC BINDING PAIRS
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
LU91067I2 (en) 1991-06-14 2004-04-02 Genentech Inc Trastuzumab and its variants and immunochemical derivatives including immotoxins
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
DK0604552T3 (en) 1991-09-18 1997-08-04 Affymax Tech Nv Process for the synthesis of different assemblies of oligomers
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5837268A (en) 1991-10-16 1998-11-17 University Of Saskatchewan GnRH-leukotoxin chimeras
JPH07503132A (en) 1991-12-17 1995-04-06 ジェンファーム インターナショナル,インコーポレイティド Transgenic non-human animals capable of producing xenoantibodies
US5679348A (en) 1992-02-03 1997-10-21 Cedars-Sinai Medical Center Immunotherapy for recurrent HSV infections
EP1958646A1 (en) 1992-02-11 2008-08-20 Henry M. Jackson Foundation For The Advancement Of Military Medicine Dual carrier immunogenic construct
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5604102A (en) * 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
US5441870A (en) 1992-04-15 1995-08-15 Athena Neurosciences, Inc. Methods for monitoring cellular processing of β-amyloid precursor protein
US5851787A (en) 1992-04-20 1998-12-22 The General Hospital Corporation Nucleic acid encoding amyloid precursor-like protein and uses thereof
CA2118508A1 (en) 1992-04-24 1993-11-11 Elizabeth S. Ward Recombinant production of immunoglobulin-like domains in prokaryotic cells
WO1993025210A1 (en) 1992-06-18 1993-12-23 President And Fellows Of Harvard College Diphtheria toxin vaccines
SG49909A1 (en) 1992-06-25 1998-06-15 Smithkline Beecham Biolog Vaccine composition 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
IL102687A (en) 1992-07-30 1997-06-10 Yeda Res & Dev Conjugates of poorly immunogenic antigens and synthetic pepide carriers and vaccines comprising them
HUT71860A (en) 1992-08-27 1996-02-28 Deakin Res Ltd Retro-, inverso-, and retro-inverso synthetic peptide analogues
US5958883A (en) 1992-09-23 1999-09-28 Board Of Regents Of The University Of Washington Office Of Technology Animal models of human amyloidoses
PT665897E (en) 1992-10-01 2003-11-28 Trustees Of Columbia U In The COMPLEX COMBINATION CHEMICAL LIBRARIES ENCODED WITH LABELS
AU5358494A (en) 1992-10-13 1994-05-09 Duke University Method of inhibiting binding of amyloid precursor protein to beta-amyloid protein
US5605811A (en) 1992-10-26 1997-02-25 Athena Neurosciences, Inc. Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
EP1298436B1 (en) 1992-10-26 2010-07-14 Elan Pharmaceuticals, Inc. Methods for the identification of Beta-amyloid peptide (BAP) release inhibitor compounds
US5972336A (en) 1992-11-03 1999-10-26 Oravax Merieux Co. Urease-based vaccine against helicobacter infection
US6210671B1 (en) 1992-12-01 2001-04-03 Protein Design Labs, Inc. Humanized antibodies reactive with L-selectin
US5674703A (en) 1992-12-02 1997-10-07 Woo; Savio L. C. Episomal vector systems and related methods
JP3720353B2 (en) 1992-12-04 2005-11-24 メディカル リサーチ カウンシル Multivalent and multispecific binding proteins, their production and use
ATE372783T1 (en) 1993-01-22 2007-09-15 Sloan Kettering Inst Cancer GANGLIOSIDE-KLH CONJUGATE VACCINES WITH QS-21 FOR DELAYING RECURRENCE OF MELANOMA
EP0683234B2 (en) * 1993-01-25 2007-06-06 Takeda Chemical Industries, Ltd. Antibody against beta-amyloid or their derivative and use thereof
US5955317A (en) 1993-01-25 1999-09-21 Takeda Chemical Industries, Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5358708A (en) 1993-01-29 1994-10-25 Schering Corporation Stabilization of protein formulations
US5472693A (en) 1993-02-16 1995-12-05 The Dow Chemical Company Family of anti-carcinoembryonic antigen chimeric antibodies
DE614989T1 (en) 1993-02-17 1995-09-28 Morphosys Proteinoptimierung Method for in vivo selection of ligand binding proteins.
CA2115900A1 (en) * 1993-02-22 1994-08-23 Gerald W. Becker Pharmaceutical screens and antibodies
DE69435075T2 (en) 1993-03-17 2009-02-19 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services IMMUNOGENEOUS CHIMERAL COMPRISING NUCLEIC ACID SEQUENCES, THE ENDOPLASMATIC RETICULAR SIGNAL SEPARATIVE PEPTIDE AND AT LEAST ONE OTHER PEPTIDE, THEIR USE IN VACCINES, AND THE TREATMENT OF DISEASES
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
CN1087176C (en) 1993-03-23 2002-07-10 史密斯克莱·比奇曼生物公司 Vaccine compositions containing 3-O deacylated monophosphoryl lipid A
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.
EP0705109B2 (en) 1993-05-25 2004-01-02 American Cyanamid Company Adjuvants for vaccines against respiratory syncytial virus
WO1994028412A1 (en) 1993-05-28 1994-12-08 The Miriam Hospital Composition and method for in vivo imaging of amyloid deposits
ATE212378T1 (en) 1993-06-04 2002-02-15 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
EP0664814A1 (en) 1993-08-18 1995-08-02 MorphoSys AG Lipopolysaccharide-binding and neutralizing peptides
CA2169635C (en) 1993-08-26 2002-11-12 Dennis A. Carson Method, compositions and devices for administration of naked polynucleotides which encode biologically active 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
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
ES2236693T3 (en) 1993-09-07 2005-07-16 Smithkline Beecham Corporation RECOMBINANT ANTIBODIES AGAINST IL4 USEFUL IN THE TREATMENT OF CONDITIONS AFTER IL4.
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
DE69435171D1 (en) 1993-09-14 2009-01-08 Pharmexa Inc PAN DR BINDEPROTEINS FOR INCREASING THE IMMUNE RESPONSE
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
AU7979094A (en) 1993-10-22 1995-05-08 Genentech Inc. Methods and compositions for microencapsulation of adjuvants
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
US5914349A (en) 1994-01-10 1999-06-22 Teva Pharmaceutical Industries, Ltd. Compositions containing and methods of using 1-aminoindan and derivatives thereof and process for preparing optically active 1-aminoindan derivatives
EP0742831B1 (en) 1994-01-27 2004-09-29 The Regents Of The University Of Minnesota Transgenic non-human mammals with progressive neurologic disease
US5877399A (en) 1994-01-27 1999-03-02 Johns Hopkins University Transgenic mice expressing APP-Swedish mutation develop progressive neurologic disease
WO1995020979A1 (en) 1994-02-03 1995-08-10 The Picower Institute For Medical Research Compositions and methods for advanced glycosylation endproduct-mediated modulation of amyloidosis
AUPM411994A0 (en) 1994-02-25 1994-03-24 Deakin Research Limited Epitopes
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
JP3064013B2 (en) * 1994-05-25 2000-07-12 ジョン マクマイケル, Methods and materials for the treatment of plaque forming disorders
US5505947A (en) 1994-05-27 1996-04-09 The University Of North Carolina At Chapel Hill Attenuating mutations in Venezuelan Equine Encephalitis virus
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
WO1996003144A1 (en) 1994-07-27 1996-02-08 The Council Of The Queensland Institute Of Medical Research Polyepitope 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
US5872005A (en) 1994-11-03 1999-02-16 Cell Genesys Inc. Packaging cell lines for adeno-associated viral vectors
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
EP0820299B1 (en) 1995-02-06 2002-04-24 Genetics Institute, Inc. 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
ES2175083T3 (en) 1995-03-14 2002-11-16 Praecis Pharm Inc AMULOID AGGREGATION MODULATORS.
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
EP0871755A1 (en) 1995-03-23 1998-10-21 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
US7153510B1 (en) 1995-05-04 2006-12-26 Yale University Recombinant vesiculoviruses and their uses
CA2222055A1 (en) 1995-05-23 1996-11-28 Morphosys Gesellschaft Fur Proteinoptimierung Mbh Multimeric proteins
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
US5910427A (en) 1995-06-22 1999-06-08 La Jolla Institute For Allergy And Immunology Antigen non-specific glycosylation inhibiting factor derivatives
PT750907E (en) 1995-06-30 2002-08-30 American Cyanamid Co METHOD FOR THEIR USE AND PROCESS FOR THEIR PREPARATION
ATE305039T1 (en) 1995-07-07 2005-10-15 Darwin Molecular Corp GENE LOCALIZED ON CHROMOSOME 1 WHICH GENES PRODUCT IS ASSOCIATED WITH ALZHEIMER'S DISEASE.
AUPN443995A0 (en) 1995-07-27 1995-08-17 Csl Limited Papillomavirus polyprotein
US6685940B2 (en) 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
ATE219517T1 (en) 1995-08-18 2002-07-15 Morphosys Ag PROTEIN/(POLY)PEPTIDE LIBRARIES
US5824322A (en) 1995-08-21 1998-10-20 Cytrx Corporation Compositions and methods for growth promotion
DE69632056T2 (en) 1995-09-14 2004-12-30 The Regents Of The University Of California, Oakland FOR NATIVE PRP-SC SPECIFIC ANTIBODIES
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
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
EP0859959B1 (en) 1995-11-10 2003-08-06 ELAN CORPORATION, Plc Peptides which enhance transport across tissues and methods of identifying and using the same
WO1997018855A1 (en) 1995-11-21 1997-05-29 Eduard Naumovich Lerner Device for enhanced delivery of biologically active substances and compounds in an organism
EP0866805A1 (en) 1995-12-12 1998-09-30 Karolinska Innovations AB PEPTIDE BINDING THE KLVFF-SEQUENCE OF AMYLOID $g(b)
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
US5770700A (en) 1996-01-25 1998-06-23 Genetics Institute, Inc. Liquid factor IX formulations
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
JP2000505306A (en) 1996-02-26 2000-05-09 モルフォシス・アクチェンゲゼルシャフト Novel method for identifying 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
HUP9902438A3 (en) 1996-03-29 2000-03-28 Bayer Ag Parapoxvirus vectors
EP0954324A4 (en) 1996-04-03 2000-11-02 Anergen Inc Cyclic peptide vaccines for treatment and prevention of diabetes
WO1997040147A1 (en) 1996-04-19 1997-10-30 The Government Of The United States Of America, Represented By The Secretary Of The Department Of Health And Human Services 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
EP0938506B1 (en) 1996-07-16 2003-11-05 Plückthun, Andreas, Prof. Dr. 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
EP1586584A1 (en) 1996-08-27 2005-10-19 Praecis Pharmaceuticals Incorporated Modulators of beta-amyloid peptide aggregation comprising D-amino acids
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)
CA2278547A1 (en) 1997-03-03 1998-09-11 Boehringer Ingelheim Pharmaceuticals, Inc. Small molecules useful in the treatment of inflammatory disease
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
DK1005368T3 (en) 1997-03-10 2010-01-04 Ottawa Hospital Res Inst Use of nucleic acids containing non-methylated CpG dinucleotide in combination with alum as adjuvants
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
US6787319B2 (en) 1997-04-16 2004-09-07 American Home Products Corp. β-amyloid peptide-binding proteins and polynucleotides encoding the same
AU7690898A (en) 1997-05-20 1998-12-11 Ottawa Civic Hospital Loeb Research Institute Vectors and methods for immunization or therapeutic protocols
AU740284B2 (en) 1997-06-13 2001-11-01 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
JP2002501721A (en) 1997-08-01 2002-01-22 モルフォシス・アクチェンゲゼルシャフト Novel method and phage for identifying nucleic acid sequences encoding members of multimeric (poly) peptide complexes
DE69842048D1 (en) 1997-08-01 2011-01-27 Max Planck Gesellschaft COMPOSITION AND METHOD FOR DETECTING DISEASES ASSOCIATED WITH THE FORMATION OF AMYLOID-SIMILAR FIBRILLES OR PROTEIN AGGREGATES
ATE435661T1 (en) 1997-08-29 2009-07-15 Antigenics Inc COMPOSITIONS CONTAINING ADJUVANT QS-21 WITH POLYSORBATE OR CYCLODEXTRIN AS AID
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
US20010016053A1 (en) * 1997-10-10 2001-08-23 Monte A. Dickson Multi-spectral imaging sensor
US6923964B1 (en) 1997-12-02 2005-08-02 Neuralab Limited Active immunization of AScr for prion disorders
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
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
US6761888B1 (en) 2000-05-26 2004-07-13 Neuralab Limited Passive immunization treatment of Alzheimer's disease
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
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
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US6743427B1 (en) * 1997-12-02 2004-06-01 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6913745B1 (en) 1997-12-02 2005-07-05 Neuralab Limited Passive immunization of Alzheimer's disease
DE1033998T1 (en) 1997-12-03 2001-05-23 Brigham And Women S Hospital B METHOD FOR SUPPRESSING BETA AMYLOID-RELATED CHANGES IN ALZHEIMER
FR2777015B3 (en) 1998-02-23 2000-09-15 Financ De Biotechnologie METHOD AND MEANS FOR OBTAINING CELLULAR AND ANIMAL MODELS OF NEURODEGENERATIVE DISEASES
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US20050059802A1 (en) 1998-04-07 2005-03-17 Neuralab Ltd Prevention and treatment of amyloidogenic disease
US20050059591A1 (en) 1998-04-07 2005-03-17 Neuralab Limited Prevention and treatment of amyloidogenic disease
ES2230848T3 (en) 1998-04-28 2005-05-01 Smithkline Beecham Corporation MONOCLONAL ANTIBODIES WITH REDUCED IMMUNOGENICITY.
US20030147882A1 (en) 1998-05-21 2003-08-07 Alan Solomon 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
US6727349B1 (en) 1998-07-23 2004-04-27 Millennium Pharmaceuticals, Inc. Recombinant anti-CCR2 antibodies and methods of use therefor
US6126967A (en) * 1998-09-03 2000-10-03 Ascent Pediatrics Extended release acetaminophen particles
HUP0103976A3 (en) 1998-10-05 2008-04-28 Pharmexa As Novel 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
CN101362518A (en) 1999-01-19 2009-02-11 法玛西雅厄普约翰美国公司 Gamma-irradiation sterilized polyethylene packaging
JP2002535289A (en) 1999-01-22 2002-10-22 ドウアリング,マシユー・ジヨン 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
IL146009A0 (en) 1999-05-05 2002-07-25 Neurochem Inc Stereoselective antifibrillogenic peptides and peptidomimetics thereof
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
AR024558A1 (en) 1999-06-01 2002-10-16 Neuralab Ltd COMPOSITIONS OF THE A-BETA PEPTIDE AND PROCESSES TO PRODUCE THE SAME
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
EP1409654B1 (en) 1999-06-16 2008-08-20 Boston Biomedical Research Institute Immunological control of beta-amyloid levels in vivo
IL147599A0 (en) 1999-07-15 2002-08-14 Genetics Inst Formulations for il-11
AU6524500A (en) 1999-08-04 2001-03-05 Northwestern University Amyloid beta protein (globular assembly and uses thereof)
US6919075B1 (en) 1999-09-03 2005-07-19 Ramot At Tel Aviv University Ltd. Bacteriophage displaying aβ epitopes and method of use
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
KR20080059676A (en) 1999-11-29 2008-06-30 뉴로겜 인터내셔널 리미티드 Vaccine for the prevention and treatment of alzheimer'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
WO2001042306A2 (en) 1999-12-08 2001-06-14 Mindset Biopharmaceuticals (Usa), Inc. Chimeric peptides (short b-cell epitope joined to a t-cell epitope) and their use for immunization
US6399314B1 (en) 1999-12-29 2002-06-04 American Cyanamid Company Methods of detection of amyloidogenic proteins
ME00183B (en) 2000-02-21 2011-02-10 Pharmexa As Novel method for down-regulation of amyloid
CZ20022748A3 (en) 2000-02-21 2004-03-17 Pharmexa A/S Novel method for controlling amyloid content
AU4178601A (en) 2000-02-24 2001-09-03 Univ Washington Humanized antibodies that sequester abeta peptide
CA2404237C (en) 2000-04-05 2010-01-26 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
KR100360985B1 (en) 2000-04-26 2002-11-18 주식회사 동진쎄미켐 Resist stripper composition
IL152625A0 (en) 2000-05-22 2003-06-24 Univ New York SYNTHETIC IMMUNOGENIC BUT NON-AMYLOIDOGENIC PEPTIDES HOMOLOGOUS TO AMYLOID beta FOR INDUCTION OF AN IMMUNE RESPONSE TO AMYLOID beta AND AMYLOID DEPOSITS
CA2414772C (en) 2000-07-07 2011-06-28 Jan Naslund Prevention and 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
US20020009445A1 (en) 2000-07-12 2002-01-24 Yansheng Du 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
ATE441663T1 (en) 2000-09-06 2009-09-15 Aventis Pharma Sa METHODS AND COMPOSITIONS FOR AMYLOIDOSIS-RELATED DISEASES
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
DE60128138T2 (en) 2000-11-02 2008-01-03 Cornell Research Foundation, Inc. IN VIVO MULTIPHOTON DIAGNOSTIC DETECTION AND IMAGE DISPLAY OF A NEURODEEGENERATIVE DISEASE
JP2004534511A (en) 2000-11-27 2004-11-18 プラエシス ファーマシューティカルズ インク. Therapeutic agents for treating amyloidogenic diseases and uses thereof
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
TWI255272B (en) 2000-12-06 2006-05-21 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
AU2002246734A1 (en) 2000-12-27 2002-08-12 University Of Texas Health Science Center Houston Prion isomers, methods of making, methods of using, and compositions and products comprising prion isomers
FI20002897A (en) * 2000-12-29 2002-06-30 Nokia Corp Electronic device and means to insure a detachable unit in a functional position
WO2002059621A2 (en) 2001-01-24 2002-08-01 Bayer Corporation Regulation of transthyretin to treat obesity
EP1251138B1 (en) 2001-04-19 2006-07-26 Hermann Dr. Schätzl Prion protein dimers useful for vaccination
DE60229051D1 (en) 2001-04-30 2008-11-06 Lilly Co Eli HUMANIZED ANTIBODIES
WO2002088306A2 (en) 2001-04-30 2002-11-07 Eli Lilly And Company Humanized antibodies
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
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
JP2005500389A (en) 2001-08-17 2005-01-06 イーライ・リリー・アンド・カンパニー Use of antibodies with high affinity for soluble Aβ to treat Aβ-related conditions and diseases
US20060073149A1 (en) 2001-08-17 2006-04-06 Bales Kelly R Rapid improvement of cognition in condition related to abeta
CA2451998A1 (en) 2001-08-17 2003-02-27 Eli Lilly And Company Anti-a.beta. antibodies
US20030082191A1 (en) 2001-08-29 2003-05-01 Poduslo Joseph F. Treatment for central nervous system disorders
US7002711B2 (en) * 2001-08-31 2006-02-21 Kabushiki Kaisha Toshiba Image reading device and method
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
DK1441589T3 (en) 2001-11-08 2012-08-06 Abbott Biotherapeutics Corp Stable, liquid, pharmaceutical composition of IgG 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
US20050227941A1 (en) 2001-12-17 2005-10-13 Karen Duff Sequestration of ass 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
DE60334364D1 (en) 2002-02-21 2010-11-11 Univ California TREATMENT PROCEDURE USING ANTI-CD22 ANTIBODIES
MY139983A (en) 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
DE60336902D1 (en) 2002-07-19 2011-06-09 Cytos Biotechnology Ag VACCINE COMPOSITIONS CONTAINING AMYLOID BETA 1-6 ANTIGENARRAYS
WO2004013172A2 (en) 2002-07-24 2004-02-12 Innogenetics N.V. Fragments of beta-amyloid as targets for vaccination against alzheimer disease
AU2003279728B2 (en) 2002-10-01 2007-09-27 Northwestern University Amyloid beta-derived diffusible ligands (addls), addl-surrogates, addl-binding molecules, and uses thereof
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
US20040191243A1 (en) 2002-12-13 2004-09-30 Bei Chen 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
KR20050118669A (en) 2003-02-01 2005-12-19 뉴랄랩 리미티드 Active immunization to generate antibodies to soluble a-beta
WO2004071408A2 (en) 2003-02-10 2004-08-26 Applied Molecular Evolution, Inc. Aβ BINDING MOLECULES
PT1610820E (en) 2003-04-04 2010-12-16 Novartis Ag 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
TWI306458B (en) 2003-05-30 2009-02-21 Elan Pharma Int Ltd 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
WO2005026211A2 (en) 2003-09-05 2005-03-24 Eli Lilly And Company Anti-ghrelin antibodies
CA2445743A1 (en) 2003-10-08 2005-04-08 The University Of British Columbia Methods for modulating neuronal responses
EP2479184A3 (en) 2003-12-17 2013-09-04 Janssen Alzheimer Immunotherapy Beta immunogenic peptide carrier conjugates and methods of producing the same
SG149013A1 (en) 2003-12-17 2009-01-29 Wyeth Corp Immunogenic peptide carrier conjugates and methods of producing same
EA016357B1 (en) 2004-07-30 2012-04-30 Ринат Ньюросайенс Корп. Antibodies directed against amyloid-beta peptide and methods using same
WO2006042158A2 (en) 2004-10-05 2006-04-20 Wyeth Methods and compositions for improving recombinant protein production
US20060160161A1 (en) 2004-10-26 2006-07-20 Elan Pharmaceuticals, Inc. Methods for assessing antibodies to neurodegenerative disease-associated antigens
WO2006066089A1 (en) 2004-12-15 2006-06-22 Neuralab Limited Humanized amyloid beta antibodies for use in improving cognition
AR051800A1 (en) 2004-12-15 2007-02-07 Wyeth Corp BETA ANTIBODIES USED TO IMPROVE COGNITION
TW200636066A (en) 2004-12-15 2006-10-16 Elan Pharm Inc Humanized antibodies that recognize beta amyloid peptide
WO2006066118A2 (en) 2004-12-15 2006-06-22 Neuralab Limited Contextual fear test for predicting efficacy of alzheimer immunotherapeutic treatment
US20060240486A1 (en) 2004-12-15 2006-10-26 Johnson-Wood Kelly L Immunoprecipitation-based assay for predicting in vivo efficacy of beta-amyloid antibodies
KR20070107079A (en) 2005-01-28 2007-11-06 와이어쓰 Stabilized liquid polypeptide formulations
GT200600031A (en) 2005-01-28 2006-08-29 ANTI-BETA ANTIBODY FORMULATION
EP2388274A1 (en) 2005-06-17 2011-11-23 Janssen Alzheimer Immunotherapy Methods of purifying anti A Beta antibodies
FR2887558B1 (en) 2005-06-28 2007-08-17 Aubert & Duval Soc Par Actions MARTENSITIC STAINLESS STEEL COMPOSITION, PROCESS FOR MANUFACTURING A MECHANICAL PART THEREFROM, AND PIECE THUS OBTAINED
AU2006270084B2 (en) 2005-07-18 2011-08-25 Merck Sharp & Dohme Corp. Spiropiperidine beta-secretase inhibitors for the treatment of Alzheimer's disease
WO2010044803A1 (en) 2008-10-17 2010-04-22 Elan Pharma International Limited Treatment of amyloidogenic diseases
KR20100016661A (en) 2007-04-18 2010-02-12 얀센 알츠하이머 이뮤노테라피 Prevention and treatment of cerebral amyloid angiopathy
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
PT2237803E (en) * 2007-12-28 2015-10-16 Prothena Biosciences Ltd Treatment and prophylaxis of amyloidosis

Also Published As

Publication number Publication date
EP2305302A2 (en) 2011-04-06
CL2010000018A1 (en) 2010-06-04
BG65882B1 (en) 2010-04-30
TWI263503B (en) 2006-10-11
NO337301B1 (en) 2016-03-07
EE05671B1 (en) 2013-08-15
IL146435A0 (en) 2002-07-25
SG140456A1 (en) 2008-03-28
WO2000072880A3 (en) 2001-05-31
CZ20013824A3 (en) 2002-11-13
GB2368794A (en) 2002-05-15
EA200101251A1 (en) 2002-06-27
DE60042451D1 (en) 2009-08-06
HUP0201250A2 (en) 2002-08-28
GB0130969D0 (en) 2002-02-13
NZ515403A (en) 2004-05-28
HK1045116B (en) 2009-12-31
NO20160043A1 (en) 2002-01-25
ATE434444T1 (en) 2009-07-15
HU230550B1 (en) 2017-02-28
CN1359301B (en) 2012-10-10
PL200458B1 (en) 2009-01-30
MXPA01011621A (en) 2005-06-22
CO5170495A1 (en) 2002-06-27
US20090297511A1 (en) 2009-12-03
IS6154A (en) 2001-11-09
NO20100264L (en) 2002-01-25
US20110306756A1 (en) 2011-12-15
AR024074A1 (en) 2002-09-04
SG143032A1 (en) 2008-06-27
ZA200109487B (en) 2003-02-17
EP2305302A3 (en) 2012-01-04
SK16982001A3 (en) 2002-11-06
AU5303100A (en) 2000-12-18
EA006630B1 (en) 2006-02-24
EP1185298B1 (en) 2009-06-24
JP5039432B2 (en) 2012-10-03
IL187526A (en) 2013-08-29
BG110407A (en) 2009-11-30
US20040219146A1 (en) 2004-11-04
HRP20010877B1 (en) 2011-02-28
US7964192B1 (en) 2011-06-21
IL187526A0 (en) 2008-03-20
CN1359301A (en) 2002-07-17
BG66083B1 (en) 2011-03-31
HUP0201250A3 (en) 2005-06-28
JP5580504B2 (en) 2014-08-27
EE200100626A (en) 2003-02-17
SG147273A1 (en) 2008-11-28
BG106241A (en) 2002-08-30
BR0011000A (en) 2002-02-19
CZ304876B6 (en) 2014-12-29
SK287513B6 (en) 2010-12-07
EA021614B1 (en) 2015-07-30
KR101058482B1 (en) 2011-08-24
HK1045116A1 (en) 2002-11-15
JP2003517461A (en) 2003-05-27
EP2108376A3 (en) 2012-04-18
GB2368794B (en) 2004-10-20
PE20010191A1 (en) 2001-02-14
KR20090068386A (en) 2009-06-26
DE10084643T1 (en) 2002-07-11
UA81215C2 (en) 2007-12-25
TR200202231T2 (en) 2002-11-21
EP2305302B1 (en) 2017-12-20
KR20020038585A (en) 2002-05-23
NO20015773L (en) 2002-01-25
KR100961485B1 (en) 2010-06-08
US20140341911A1 (en) 2014-11-20
TR200103447T2 (en) 2002-04-22
MY132784A (en) 2007-10-31
NO20015773D0 (en) 2001-11-27
EP2108376A2 (en) 2009-10-14
JP2007269804A (en) 2007-10-18
PL357182A1 (en) 2004-07-26
EP2108376B1 (en) 2017-03-01
ES2624908T3 (en) 2017-07-18
WO2000072880A2 (en) 2000-12-07
HRP20090510A2 (en) 2009-12-31
CN1814287A (en) 2006-08-09
HRP20010877A2 (en) 2005-04-30
EP1185298A2 (en) 2002-03-13
EA200501560A1 (en) 2006-04-28
CA2370311A1 (en) 2000-12-07
NO340768B1 (en) 2017-06-19
UY26182A1 (en) 2000-12-29
US20040265308A1 (en) 2004-12-30
GB0417096D0 (en) 2004-09-01

Similar Documents

Publication Publication Date Title
CA2370311C (en) Prevention and treatment of amyloidogenic disease
US6761888B1 (en) Passive immunization treatment of Alzheimer&#39;s disease
US6750324B1 (en) Humanized and chimeric N-terminal amyloid beta-antibodies
US8034348B2 (en) Prevention and treatment of amyloidogenic disease
US6743427B1 (en) Prevention and treatment of amyloidogenic disease
US6787637B1 (en) N-Terminal amyloid-β antibodies
US6710226B1 (en) Transgenic mouse assay to determine the effect of Aβ antibodies and Aβ Fragments on alzheimer&#39;s disease characteristics
US20050059591A1 (en) Prevention and treatment of amyloidogenic disease
US20050059802A1 (en) Prevention and treatment of amyloidogenic disease
US7588766B1 (en) Treatment of amyloidogenic disease
AU2008203784B2 (en) Prevention and treatment of amyloidogenic disease
AU2011265382A1 (en) Prevention and treatment of amyloidogenic disease
AU2005200811A1 (en) Prevention and treatment of amyloidogenic disease
NZ570086A (en) Prevention and treatment of amyloidogenic disease

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20200526