US20040171815A1 - Humanized antibodies that recognize beta amyloid peptide - Google Patents

Humanized antibodies that recognize beta amyloid peptide Download PDF

Info

Publication number
US20040171815A1
US20040171815A1 US10/703,713 US70371303A US2004171815A1 US 20040171815 A1 US20040171815 A1 US 20040171815A1 US 70371303 A US70371303 A US 70371303A US 2004171815 A1 US2004171815 A1 US 2004171815A1
Authority
US
United States
Prior art keywords
antibody
sequence
antibodies
humanized
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/703,713
Other languages
English (en)
Inventor
Dale Schenk
Ted Yednock
Guriq Basi
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.)
Crimagua Ltd
Janssen Sciences Ireland UC
Wyeth LLC
Original Assignee
Neuralab Ltd
Wyeth LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26748211&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20040171815(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US09/201,430 external-priority patent/US6787523B1/en
Priority claimed from US09/322,289 external-priority patent/US7964192B1/en
Priority claimed from US10/010,942 external-priority patent/US7189819B2/en
Priority claimed from US10/388,389 external-priority patent/US7179892B2/en
Priority to US10/703,713 priority Critical patent/US20040171815A1/en
Application filed by Neuralab Ltd, Wyeth LLC filed Critical Neuralab Ltd
Assigned to ELAN PHARMACEUTICALS, INC. reassignment ELAN PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SCHENK, DALE B., Yednock, Ted, BASI, GURIQ
Assigned to NEURALAB LIMITED, WYETH reassignment NEURALAB LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ELAN PHARMACEUTICALS, INC.
Publication of US20040171815A1 publication Critical patent/US20040171815A1/en
Priority to US11/516,724 priority patent/US20080050367A1/en
Priority to US11/842,042 priority patent/US9051363B2/en
Priority to US11/842,056 priority patent/US20080281082A1/en
Priority to US11/842,023 priority patent/US7893214B2/en
Assigned to WYETH, ELAN PHARMA INTERNATIONAL LIMITED reassignment WYETH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NEURALAB LIMITED
Assigned to WYETH, CRIMAGUA LIMITED reassignment WYETH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ELAN PHARMA INTERNATIONAL LIMITED
Assigned to WYETH, JANSSEN ALZHEIMER IMMUNOTHERAPY reassignment WYETH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CRIMAGUA LIMITED
Assigned to WYETH LLC reassignment WYETH LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: WYETH
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/739Lipopolysaccharides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0007Nervous system antigens; Prions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/646Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the entire peptide or protein drug conjugate elicits an immune response, e.g. conjugate vaccines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55577Saponins; Quil A; QS21; ISCOMS
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6037Bacterial toxins, e.g. diphteria toxoid [DT], tetanus toxoid [TT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/605MHC molecules or ligands thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/70Web, sheet or filament bases ; Films; Fibres of the matrix type containing drug
    • A61K9/7023Transdermal patches and similar drug-containing composite devices, e.g. cataplasms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • U.S. application Ser. No. 10/010,942 is also a continuation-in-part of U.S. application Ser. No. 09/580,015 filed May 26, 2000, which is a continuation-in-part of U.S. application Ser. No. 09/322,289, filed May 28, 1999, which is a continuation-in-part of U.S. application Ser. No. 09/201,430, filed Nov. 30, 1998, which is a an application claiming the benefit under 35 U.S.C. 119(e) of U.S. application Ser. No. 60/080,970, filed Apr. 7, 1998, each of which is incorporated herein by reference for all purposes.
  • AD Alzheimer's disease
  • TINS 16:403 (1993); Hardy et al., WO 92/13069; Selkoe, J. Neuropathol. Exp. Neurol . 53:438 (1994); Duff et al., Nature 373:476 (1995); Games et al., Nature 373:523 (1995).
  • 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.
  • 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, neurofibrillary tangles and senile plaques.
  • Neurofibrillary tangles are intracellular deposits of microtubule associated tau protein consisting of two filaments twisted about each other in pairs.
  • Senile plaques i.e., amyloid plaques
  • amyloid plaques are areas of disorganized neuropil up to 150 ⁇ m across with extracellular amyloid deposits at the center which are visible by microscopic analysis of sections of brain tissue. The accumulation of amyloid plaques within the brain is also associated with Down's syndrome and other cognitive disorders.
  • a ⁇ or ⁇ -amyloid peptide The principal constituent of the plaques is a peptide termed A ⁇ or ⁇ -amyloid peptide.
  • a ⁇ peptide is a 4-kDa internal fragment of 39-43 amino acids of a larger transmembrane glycoprotein named protein termed amyloid precursor protein (APP).
  • APP amyloid precursor protein
  • a ⁇ is primarily found in both a short form, 40 amino acids in length, and a long form, ranging from 42-43 amino acids in length.
  • Part of the hydrophobic transmembrane domain of APP is found at the carboxy end of A ⁇ , and may account for the ability of A ⁇ to aggregate into plaques, particularly in the case of the long form. Accumulation of amyloid plaques in the brain eventually leads to neuronal cell death. The physical symptoms associated with this type of neural deterioration characterize Alzheimer's disease.
  • Such mutations are thought to cause Alzheimer's disease by increased or altered processing of APP to A ⁇ , particularly processing of APP to increased amounts of the long form of A ⁇ (i.e., A ⁇ 1-42 and A ⁇ 1-43). Mutations in other genes, such as the presenilin genes, PS1 and PS2, are thought indirectly to affect processing of APP to generate increased amounts of long form A ⁇ (see Hardy, TINS 20: 154 (1997)).
  • McMichael, EP 526,511 proposes administration of homeopathic dosages (less than or equal to 10 ⁇ 2 mg/day) of A ⁇ 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 A ⁇ in human plasma is typically in the range of 50-200 pg/ml (Seubert et al., Nature 359:325 (1992)). Because EP 526,511 's proposed dosage would barely alter the level of endogenous circulating A ⁇ and because EP 526,511 does not recommend use of an adjuvant, as an immunostimulant, it seems implausible that any therapeutic benefit would result.
  • the present invention features new immunological reagents, in particular, therapeutic antibody reagents for the prevention and treatment of amyloidogenic disease (e.g., Alzheimer's disease).
  • amyloidogenic disease e.g., Alzheimer's disease
  • the invention is based, at least in part, on the identification and characterization of two monoclonal antibodies that specifically bind to A ⁇ peptide and are effective at reducing plaque burden and/or reducing the neuritic dystrophy associated with amyloidogenic disorders. Structural and functional analysis of these antibodies leads to the design of various humanized antibodies for prophylactic and/or therapeutic use.
  • the invention features humanization of the variable regions of these antibodies and, accordingly provides for humanized immunoglobulin or antibody chains, intact humanized immunoglobulins or antibodies, and functional immunoglobulin or antibody fragments, in particular, antigen binding fragments, of the featured antibodies.
  • Polypeptides comprising the complementarity determining regions of the featured monoclonal antibodies are also disclosed, as are polynlcleotide reagents, vectors and host suitable for encoding said polypeptides.
  • amyloidogenic diseases or disorders e.g., Alzheimer's disease
  • pharmaceutical compositions and kits for use in such applications are disclosed, as are pharmaceutical compositions and kits for use in such applications.
  • antibodies e.g., humanized antibodies having altered effector functions, and therapeutic uses thereof.
  • FIG. 1 depicts an alignment of the amino acid sequences of the light chain of mouse 3D6, humanized 3D6, Kabat ID 109230 and germline A19 antibodies. CDR regions are indicated by arrows. Bold italics indicate rare murine residues. Bold indicates packing (VH+VL) residues. Solid fill indicates canonical/CDR interacting residues. Asterisks indicate residues selected for backmutation in humanized 3D6, version 1.
  • FIG. 2 depicts an alignment of the amino acid sequences of the heavy chain of mouse 3D6, humanized 3D6, Kabat ID 045919 and germline VH3-23 antibodies. Annotation is the same as for FIG. 1.
  • FIG. 3 graphically depicts the A ⁇ binding properties of 3D6, chimeric 3D6 and 1D5.
  • FIG. 3A is a graph depicting binding of A ⁇ to chimeric 3D6 (PK1614) as compared to murine 3D6.
  • FIG. 3B is a graph depicting competition of biotinylated 3D6 versus unlabeled 3D6, PK1614 and 10D5 for binding to A ⁇ .
  • FIG. 4 depicts a homology model of 3D6 VH and VL, showing ⁇ -carbon backbone trace.
  • VH is shown in as a stippled line
  • VL is shown as a solid line.
  • CDR regions are indicated in ribbon form.
  • FIG. 5 graphically depicts the A ⁇ binding properties of chimeric 3D6 and humanized 3D6.
  • FIG. 5A depicts ELISA results measuring the binding of humanized 3D6v1 and chimeric 3D6 to aggregated A ⁇ .
  • FIG. 5B depicts ELISA results measuring the binding of humanized 3D6v1 and humanized 3D6v2 to aggregated A ⁇ .
  • FIG. 6 is a graph quantitating the binding of humanized 3D6 and chimeric 3D6 to A ⁇ plaques from brain sections of PDAPP mice.
  • FIG. 7 is a graph showing results of a competitive binding assay testing the ability of humanized 3D6 versions 1 and 2, chimeric 3D6, murine 3D6, and 10D5 to compete with murine 3D6 for binding to A ⁇ .
  • FIG. 8 graphically depicts of an ex vivo phagocytosis assay testing the ability of humanized 3D6v2, chimeric 3D6, and human IgG to mediate the uptake of A ⁇ by microglial cells.
  • FIG. 9 depicts an alignment of the 10D5 VL and 3D6 VL amino acid sequences. Bold indicates residues that match 10 D5 exactly.
  • FIG. 10 depicts an alignment of the 10D5 VH and 3D6 VH amino acid sequences. Bold indicates residues that match 10D5 exactly.
  • the present invention features new immunological reagents and methods for preventing or treating Alzheimer's disease or other amyloidogenic diseases.
  • the invention is based, at least in part, on the characterization of two monoclonal immunoglobulins, 3D6 and 10D5, effective at binding beta amyloid protein (A ⁇ ) (e.g., binding soluble and/or aggregated A ⁇ ), mediating phagocytosis (e.g., of aggregated A ⁇ ), reducing plaque burden and/or reducing neuritic dystrophy (e.g., in patient).
  • a ⁇ beta amyloid protein
  • mediating phagocytosis e.g., of aggregated A ⁇
  • reducing plaque burden and/or reducing neuritic dystrophy e.g., in patient.
  • the invention is further based on the determination and structural characterization of the primary and secondary structure of the variable light and heavy chains of these immunoglobulins and the identification of residues important for activity and immunogenicity.
  • Immunoglobulins are featured which include a variable light and/or variable heavy chain of the preferred monoclonal immunoglobulins described herein.
  • Preferred immunoglobulins e.g., therapeutic immunoglobulins, are featured which include a humanized variable light and/or humanized variable heavy chain.
  • Preferred variable light and/or variable heavy chains include a complementarity determining region (CDR) from the monoclonal immunoglobulin (e.g., donor immunoglobulin) and variable framework regions substantially from a human acceptor immunoglobulin.
  • CDR complementarity determining region
  • substantially from a human acceptor immunoglobulin means that the majority or key framework residues are from the human acceptor sequence, allowing however, for substitution of residues at certain positions with residues selected to improve activity of the humanized immunoglobulin (e.g., alter activity such that it more closely mimics the activity of the donor immunoglobulin) or selected to decrease the immunogenicity of the humanized immunoglobulin.
  • the invention features a humanized immunoglobulin light or heavy chain that includes 3D6 variable region complementarity determining regions (CDRs) (i.e., includes one, two or three CDRs from the light chain variable region sequence set forth as SEQ ID NO:2 or includes one, two or three CDRs from the heavy chain variable region sequence set forth as SEQ ID NO:4), and includes a variable framework region substantially from a human acceptor immunoglobulin light or heavy chain sequence, provided that at least one residue of the framework residue is backmutated to a corresponding murine residue, wherein said backmutation does not substantially affect the ability of the chain to direct A ⁇ binding.
  • CDRs 3D6 variable region complementarity determining regions
  • the invention features a humanized immunoglobulin light or heavy chain that includes 3D6 variable region complementarity determining regions (CDRS) (e.g., includes one, two or three CDRs from the light chain variable region sequence set forth as SEQ ID NO:2 and/or includes one, two or three CDRs from the heavy chain variable region sequence set forth as SEQ ID NO:4), and includes a variable framework region substantially from a human acceptor immunoglobulin light or heavy chain sequence, provided that at least one framework residue is substituted with the corresponding amino acid residue from the mouse 3D6 light or heavy chain variable region sequence, where the framework residue is selected from the group consisting of (a) a residue that non-covalently binds antigen directly; (b) a residue adjacent to a CDR; (c) a CDR-interacting residue (e.g., identified by modeling the light or heavy chain on the solved structure of a homologous known immunoglobulin chain); and (d) a residue participating in the VL-VH
  • CDRS 3D6
  • the invention features a humanized immunoglobulin light or heavy chain that includes 3D6 variable region CDRs and variable framework regions from a human acceptor immunoglobulin light or heavy chain sequence, provided that at least one framework residue is substituted with the corresponding amino acid residue from the mouse 3D6 light or heavy chain variable region sequence, where the framework residue is a residue capable of affecting light chain variable region conformation or function as identified by analysis of a three-dimensional model of the variable region, for example a residue capable of interacting with antigen, a residue proximal to the antigen binding site, a residue capable of interacting with a CDR, a residue adjacent to a CDR, a residue within 6 ⁇ of a CDR residue, a canonical residue, a vernier zone residue, an interchain packing residue, an unusual residue, or a glycoslyation site residue on the surface of the structural model.
  • the framework residue is a residue capable of affecting light chain variable region conformation or function as identified by analysis of a three-dimensional model of the variable region,
  • the invention features a humanized immunoglobulin light chain that includes 3D6 variable region CDRs (e.g., from the 3D6 light chain variable region sequence set forth as SEQ ID NO:2), and includes a human acceptor immunoglobulin variable framework region, provided that at least one framework residue selected from the group consisting of L1, L2, L36 and L46 (Kabat numbering convention) is-substituted with the corresponding amino acid residue from the mouse 3D6 light chain variable region sequence.
  • 3D6 variable region CDRs e.g., from the 3D6 light chain variable region sequence set forth as SEQ ID NO:2
  • a human acceptor immunoglobulin variable framework region provided that at least one framework residue selected from the group consisting of L1, L2, L36 and L46 (Kabat numbering convention) is-substituted with the corresponding amino acid residue from the mouse 3D6 light chain variable region sequence.
  • the invention features a humanized immunoglobulin heavy chain that includes 3D6 variable region CDRs (e.g., from the 3D6 heavy chain variable region sequence set forth as SEQ ID NO:4), and includes a human acceptor immunoglobulin variable framework region, provided that at least one framework residue selected from the group consisting of H49, H93 and H94 (Kabat numbering convention) is substituted with the corresponding amino acid residue from the mouse 3D6 heavy chain variable region sequence.
  • 3D6 variable region CDRs e.g., from the 3D6 heavy chain variable region sequence set forth as SEQ ID NO:4
  • a human acceptor immunoglobulin variable framework region provided that at least one framework residue selected from the group consisting of H49, H93 and H94 (Kabat numbering convention) is substituted with the corresponding amino acid residue from the mouse 3D6 heavy chain variable region sequence.
  • Preferred light chains include kappa II framework regions of the subtype kappa II (Kabat convention), for example, framework regions from the acceptor immunoglobulin Kabat ID 019230, Kabat ID 005131, Kabat ID 005058, Kabat ID 005057, Kabat ID 005059, Kabat ID U21040 and Kabat ID U41645.
  • Preferred heavy chains include framework regions of the subtype III (Kabat convention), for example, framework regions from the acceptor immunoglobulin Kabat ID 045919, Kabat ID 000459, Kabat ID 000553, Kabat ID 000386 and Kabat ID M23691.
  • the invention features a humanized immunoglobulin light or heavy chain that includes 10D5 variable region complementarity determining regions (CDRs) (i.e., includes one, two or three CDRs from the light chain variable region sequence set forth as SEQ ID NO: 14 or includes one, two or three CDRs from the heavy chain variable region sequence set forth as SEQ ID NO: 16), and includes a variable framework region substantially from a human acceptor immunoglobulin light or heavy chain sequence, provided that at least one residue of the framework residue is backmutated to a corresponding murine residue, wherein said backmutation does not substantially affect the ability of the chain to direct A ⁇ binding.
  • CDRs 10D5 variable region complementarity determining regions
  • the invention features a humanized immunoglobulin light or heavy chain that includes 10D5 variable region complementarity determining regions (CDRs) (e.g., includes one, two or three CDRs from the light chain variable region sequence set forth as SEQ ID NO:14 and/or includes one, two or three CDRs from the heavy chain variable region sequence set forth as SEQ ID NO: 16), and includes a variable framework region substantially from a human acceptor immunoglobulin light or heavy chain sequence, provided that at least one framework residue is substituted with the corresponding amino acid residue from the mouse 3D6 light or heavy chain variable region sequence, where the framework residue is selected from the group consisting of (a) a residue that non-covalently binds antigen directly; (b) a residue adjacent to a CDR; (c) a CDR-interacting residue (e.g., identified by modeling the light or heavy chain on the solved structure of a homologous known immunoglobulin chain); and (d) a residue participating in the VL-V
  • CDRs 10D
  • the invention features a humanized immunoglobulin light or heavy chain that includes 10D5 variable region CDRs and variable framework regions from a human acceptor immunoglobulin light or heavy chain sequence, provided that at least one framework residue is substituted with the corresponding amino acid residue from the mouse 10D5 light or heavy chain variable region sequence, where the framework residue is a residue capable of affecting light chain variable region conformation or function as identified by analysis of a three-dimensional model of the variable region, for example a residue capable of interacting with antigen, a residue proximal to the antigen binding site, a residue capable of interacting with a CDR, a residue adjacent to a CDR, a residue within 6 ⁇ of a CDR residue, a canonical residue, a vernier zone residue, an interchain packing residue, an unusual residue, or a glycoslyation site residue on the surface of the structural model.
  • the framework residue is a residue capable of affecting light chain variable region conformation or function as identified by analysis of a three-dimensional model of the variable region,
  • the invention features, in addition to the substitutions described above, a substitution of at least one rare human framework residue.
  • a rare residue can be substituted with an amino acid residue which is common for human variable chain sequences at that position.
  • a rare residue can be substituted with a corresponding amino acid residue from a homologous germline variable chain sequence (e.g., a rare light chain framework residue can be substituted with a corresponding germline residue from an A1A17, A18, A2, or A19 germline immunoglobulin sequence or a rare heavy chain framework residue can be substituted with a corresponding germline residue from a VH3-48, VH3-23, VH3-7, VH3-21 or VH3-11 germline immunoglobulin sequence.
  • the invention features a humanized immunoglobulin that includes a light chain and a heavy chain, as described above, or an antigen-binding fragment of said immunoglobulin.
  • the humanized immunoglobulin binds (e.g., specifically binds) to beta amyloid peptide (A ⁇ ) with a binding affinity of at least 10 7 M ⁇ 1 , 10 8 M ⁇ 1 , or 10 9 M ⁇ 1 .
  • the immunoglobulin or antigen binding fragment includes a heavy chain having isotype ⁇ 1.
  • the immunoglobulin or antigen binding fragment binds (e.g., specifically binds) to both soluble beta amyloid peptide (A ⁇ ) and aggregated A ⁇ .
  • the irnmunoglobulin or antigen binding fragment mediates phagocytosis (e.g., induces phagocytosis) of beta amyloid peptide (A ⁇ ).
  • the immunoglobulin or antigen binding fragment crosses the blood-brain barrier in a subject.
  • the immunoglobulin or antigen binding fragment reduces both beta amyloid peptide (A ⁇ ) burden and neuritic dystrophy in a subject.
  • the invention features chimeric immunoglobulins that include 3D6 variable regions (e.g., the variable region sequences set forth as SEQ ID NO:2 or SEQ ID NO:4).
  • an antibody or immunoglobulin sequence comprising a VL and/or VH sequence as set forth in, for example, SEQ ID NO:2 or SEQ ID NO:4 can comprise either the full VL or VH sequence or can comprise the mature VL or VH sequence (i.e., mature peptide without the signal or leader peptide).
  • the invention features an immunoglobulin, or antigen-binding fragment thereof, including a variable heavy chain region as set forth in SEQ ID NO:8 and a variable light chain region as set forth in SEQ ID NO:5.
  • the invention features an immunoglobulin, or antigen-binding fragment thereof, including a variable heavy chain region as set forth in SEQ ID NO: 12 and a variable light chain region as set forth in SEQ ID NO: 11.
  • the invention features chimeric immunoglobulins that include 10D5 variable regions (e.g., the variable region sequences set forth as SEQ ID NO:14 or SEQ ID NO:16).
  • the immunoglobulin, or antigen-binding fragment thereof further includes constant regions from IgG1.
  • the immunoglobulins described herein are particularly suited for use in therapeutic methods aimed at preventing or treating amyloidogenic diseases.
  • the invention features a method of preventing or treating an amyloidogenic disease (e.g., Alzheimer's disease) that involves administering to the patient an effective dosage of a humanized immunoglobulin as described herein.
  • an amyloidogenic disease e.g., Alzheimer's disease
  • the invention features pharmaceutical compositions that include a humanized immunoglobulin as described herein and a pharmaceutical carrier. Also featured are isolated nucleic acid molecules, vectors and host cells for producing the immunoglobulins or immunoglobulin fragments or chains described herein, as well as methods for producing said immunoglobulins, immunoglobulin fragments or immunoglobulin chains
  • the present invention further features a method for identifying 3D6 or 10D5 residues amenable to substitution when producing a humanized 3D6 or 10D5 immunoglobulin, respectively.
  • a method for identifying variable framework region residues amenable to substitution involves modeling the three-dimensional dimensional structure of the 3D6 or 10D5 variable region on a solved homologous irnmunoglobulin structure and analyzing said model for residues capable of affecting 3D6 or 10D5 immunoglobulin variable region conformation or function, such that residues amenable to substitution are identified.
  • the invention further features use of the variable region sequence set forth as SEQ ID NO:2 or SEQ ID NO:4, or any portion thereof, in producing a three-dimensional image of a 3D6 immunoglobulin, 3D6 immunoglobulin chain, or domain thereof. Also featured is the use of the variable region sequence set forth as SEQ ID NO: 14 or SEQ ID NO: 16, or any portion thereof, in producing a three-dimensional image of a 10D5 immunoglobulin, 10D5 immunoglobulin chain, or domain thereof.
  • the present invention further features immunoglobulins having altered effector function, such as the ability to bind effector molecules, for example, complement or a receptor on an effector cell.
  • the immunoglobulin of the invention has an altered constant region, e.g., Fc region, wherein at least one amino acid residue in the Fc region has been replaced with a different residue or side chain.
  • the modified immunoglobulin is of the IgG class, comprises at least one amino acid residue replacement in the Fc region such that the immunoglobulin has an altered effector function, e.g., as compared with an unmodified immunoglobulin.
  • the immunoglobulin of the invention has an altered effector function such that it is less immunogenic (e.g., does not provoke undesired effector cell activity, lysis, or complement binding), has improved amyloid clearance properties, and/or has a desirable half-life.
  • immunoglobulin refers to an antigen-binding protein having a basic four-polypeptide chain structure consisting of two heavy and two light chains, said chains being stabilized, for example, by interchain disulfide bonds, which has the ability to specifically bind antigen. Both heavy and light chains are folded into domains.
  • domain refers to a globular region of a heavy or light chain polypeptide comprising peptide loops (e.g., comprising 3 to 4 peptide loops) stabilized, for example, by ⁇ -pleated sheet and/or intrachain disulfide bond.
  • Domains are further referred to herein as “constant” or “variable”, based on the relative lack of sequence variation within the domains of various class members in the case of a “constant” domain, or the significant variation within the domains of various class members in the case of a “variable” domain.
  • Constant domains on the light chain are referred to interchangeably as “light chain constant regions”, “light chain constant domains”, “CL” regions or “CL” domains).
  • Constant” domains on the heavy chain are referred to interchangeably as “heavy chain constant regions”, “heavy chain constant domains”, “CH” regions or “CH” domains).
  • “Variable” domains on the light chain are referred to interchangeably as “light chain variable regions”, “light chain variable domains”, “VL” regions or “VL” domains). “Variable” domains on the heavy chain are referred to interchangeably as “heavy chain constant regions”, “heavy chain constant domains”, “CH” regions or “CH” domains).
  • region refers to a part or portion of an antibody chain and includes constant or variable domains as defined herein, as well as more discrete parts or portions of said domains.
  • light chain variable domains or regions include “complementarity determining regions” or “CDRs” interspersed among “framework regions” or “FRs”, as defined herein.
  • Immunoglobulins or antibodies can exist in monomeric or polymeric form.
  • antigen-binding fragment refers to a polypeptide fragment of an immunoglobulin or antibody binds antigen or competes with intact antibody (i.e., with the intact antibody from which they were derived) for antigen binding (i.e., specific binding).
  • formation refers to the tertiary structure of a protein or polypeptide (e.g., an antibody, antibody chain, domain or region thereof).
  • the phrase “light (or heavy) chain conformation” refers to the tertiary structure of a light (or heavy) chain variable region
  • the phrase “antibody conformation” or “antibody fragment conformation” refers to the tertiary structure of an antibody or fragment thereof.
  • Specific binding” of an antibody mean that the antibody exhibits appreciable affinity for antigen or a preferred epitope and, preferably, does not exhibit significant crossreactivity.
  • Appreciable or preferred binding include binding with an affinity of at least 10 6 , 10 7 , 10 8 , 10 9 M ⁇ 1 , or 10 10 M ⁇ 1 . Affinities greater than 10 7 M ⁇ 1 , preferably greater than 10 8 M ⁇ 1 are more preferred.
  • a preferred binding affinity can be indicated as a range of affinities, for example, 10 6 to 10 10 M ⁇ 1 , preferably 10 7 to 10 10 M ⁇ 1 , more preferably 108 to 10 10 M ⁇ 1 .
  • An antibody that “does not exhibit significant crossreactivity” is one that will not appreciably bind to an undesirable entity (e.g., an undesirable proteinaceous entity).
  • an antibody that specifically binds to A ⁇ will appreciably bind A ⁇ but will not significantly react with non-A ⁇ proteins or peptides (e.g., non-A ⁇ proteins or peptides included in plaques).
  • An antibody specific for a preferred epitope will, for example, not significantly crossreact with remote epitopes on the same protein or peptide.
  • Specific binding can be determined according to any art-recognized means for determining such binding. Preferably, specific binding is determined according to Scatchard analysis and/or competitive binding assays.
  • Binding fragments are produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins. Binding fragments include Fab, Fab′, F(ab′) 2 , Fabc, Fv, single chains, and single-chain antibodies. Other than “bispecific” or “bifunctional” immunoglobulins or antibodies, an immunoglobulin or antibody is understood to have each of its binding sites identical. 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.
  • humanized immunoglobulin refers to an immunoglobulin or antibody that includes at least one humanized immunoglobulin or antibody chain (i.e., at least one humanized light or heavy chain).
  • humanized immunoglobulin chain or “humanized antibody chain” (i.e., a “humanized immunoglobulin light chain” or “humanized immunoglobulin heavy chain”) refers to an immunoglobulin or antibody chain (i.e., a light or heavy chain, respectively) having a variable region that includes a variable framework region substantially from a human immunoglobulin or antibody and complementarity determining regions (CDRs) (e.g., at least one CDR, preferably two CDRs, more preferably three CDRs) substantially from a non-human immunoglobulin or antibody, and further includes constant regions (e.g., at least one constant region or portion thereof, in the case of a light chain, and preferably three constant regions in the case of a heavy chain).
  • CDRs complementarity determining regions
  • humanized variable region refers to a variable region that includes a variable framework region substantially from a human immunoglobulin or antibody and complementarity determining regions (CDRs) substantially from a non-human immunoglobulin or antibody.
  • CDRs complementarity determining regions
  • substantially from a human immunoglobulin or antibody or “substantially human” means that, when aligned to a human immunoglobulin or antibody amino sequence for comparison purposes, the region shares at least 80-90%, preferably 90-95%, more preferably 95-99% identity (i.e., local sequence identity) with the human framework or constant region sequence, allowing, for example, for conservative substitutions, consensus sequence substitutions, germline substitutions, backmutations, and the like.
  • the introduction of conservative substitutions, consensus sequence substitutions, germline substitutions, backmutations, and the like is often referred to as “optimization” of a humanized antibody or chain.
  • substantially from a non-human immunoglobulin or antibody or “substantially non-human” means having an immunoglobulin or antibody sequence at least 80-95%, preferably 90-95%, more preferably, 96%, 97%, 98%, or 99% identical to that of a non-human organism, e.g., a non-human mammal.
  • corresponding region refers to a region or residue on a second amino acid or nucleotide sequence which occupies the same (i.e., equivalent) position as a region or residue on a first amino acid or nucleotide sequence, when the first and second sequences are optimally aligned for comparison purposes.
  • humanized immunoglobulin or “humanized antibody” are not intended to encompass chimeric immunoglobulins or antibodies, as defined infra. Although humanized immunoglobulins or antibodies are chimeric in their construction (i.e., comprise regions from more than one species of protein), they include additional features (i.e., variable regions comprising donor CDR residues and acceptor framework residues) not found in chimeric immunoglobulins or antibodies, as defined herein.
  • significant identity means that two polypeptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 50-60% sequence identity, preferably 60-70% sequence identity, more preferably 70-80% sequence identity, more preferably at least 80-90% identity, even more preferably at least 90-95% identity, and even more preferably at least 95% sequence identity or more (e.g., 99% sequence identity or more).
  • substantially identical means that two polypeptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 80-90% sequence identity, preferably 90-95% sequence identity, and more preferably at least 95% sequence identity or more (e.g., 99% sequence identity or more).
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • 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.
  • sequence identity and “sequence identity” are used interchangeably herein.
  • 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. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by visual inspection (see generally Ausubel et al., Current Protocols in Molecular Biology).
  • BLAST algorithm One example of algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described in Altschul et al., J. Mol. Biol. 215:403 (1990).
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (publicly accessible through the National Institutes of Health NCBI internet server).
  • default program parameters can be used to perform the sequence comparison, although customized parameters can also be used.
  • W wordlength
  • E expectation
  • BLOSUM62 scoring matrix see Henikoff& Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1989)).
  • residue positions which are not identical differ by conservative amino acid substitutions.
  • amino acids are grouped as follows: Group 1 (hydrophobic sidechains): leu, 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.
  • humanized immunoglobulins or antibodies bind antigen with an affinity that is within a factor of three, four, or five of that of the corresponding non-human antibody.
  • the nonhuman antibody has a binding affinity of 10 9 M ⁇
  • humanized antibodies will have a binding affinty of at least 3 ⁇ 10 9 M ⁇ 1 , 4 ⁇ 10 9 M ⁇ 1 or 10 9 M ⁇ 1 .
  • the chain can be described based on its ability to “direct antigen (e.g., A ⁇ ) binding”.
  • a chain is said to “direct antigen binding” when it confers upon an intact immunoglobulin or antibody (or antigen binding fragment thereof) a specific binding property or binding affinity.
  • a mutation e.g., a backmutation
  • a mutation is said to substantially affect the ability of a heavy or light chain to direct antigen binding if it affects (e.g., decreases) the binding affinity of an intact immunoglobulin or antibody (or antigen binding fragment thereof) comprising said chain by at least an order of magnitude compared to that of the antibody (or antigen binding fragment thereof) comprising an equivalent chain lacking said mutation.
  • a mutation “does not substantially affect (e.g., decrease) the ability of a chain to direct antigen binding” if it affects (e.g., decreases) the binding affinity of an intact immunoglobulin or antibody (or antigen binding fragment thereof) comprising said chain by only a factor of two, three, or four of that of the antibody (or antigen binding fragment thereof) comprising an equivalent chain lacking said mutation.
  • chimeric immunoglobulin refers to an immunoglobulin or antibody whose variable regions derive from a first species and whose constant regions derive from a second species.
  • Chimeric immunoglobulins or antibodies can be constructed, for example by genetic engineering, from immunogiobulin gene segments belonging to different species.
  • an “antigen” is an entity (e.g., a protenaceous entity or peptide) to which an antibody specifically binds.
  • epitopes refers to a site on an antigen to which an immunoglobulin or antibody (or antigen binding fragment thereof) specifically binds.
  • 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, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 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, G. 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, i.e., a competitive binding assay.
  • Competitive binding is determined in an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen, such as A ⁇ .
  • RIA solid phase direct or indirect radioimmunoassay
  • EIA solid phase direct or indirect enzyme immunoassay
  • sandwich competition assay see Stahli et al., Methods in Enzymology 9:242 (1983)
  • solid phase direct biotin-avidin EIA see Kirkland et al., J. Immunol . 137:3614 (1986)
  • solid phase direct labeled assay solid phase direct labeled sandwich 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 I-125 label see Morel et al., Mol. Immunol .
  • An epitope is also recognized by immunologic cells, for example, B cells and/or T cells.
  • Cellular recognition of an epitope can be determined by in vitro assays that measure antigen-dependent proliferation, as determined by 3 H-thymidine incorporation, by cytokine secretion, by antibody secretion, or by antigen-dependent killing (cytotoxic T lymphocyte assay).
  • Exemplary epitopes or antigenic determinants can be found within the human amyloid precursor protein (APP), but are preferably found within the A ⁇ peptide of APP.
  • Multiple isoforms of APP exist, for example APP 695 APP 751 and APP 770 .
  • Amino acids within APP are assigned numbers according to the sequence of the APP 770 isoform (see e.g., GenBank Accession No. P05067, also set forth as SEQ ID NO:38).
  • a ⁇ (also referred to herein as beta amyloid peptide and A-beta) peptide is a ⁇ 4-kDa internal fragment of 39-43 amino acids of APP (AP39, A ⁇ 40, A ⁇ 41, A ⁇ 42 and A ⁇ 43).
  • a ⁇ 40 for example, consists of residues 672-711 of APP and A ⁇ 42 consists of residues 673-713 of APP.
  • a ⁇ is found in both a “short form”, 40 amino acids in length, and a “long form”, ranging from 42-43 amino acids in length.
  • Preferred epitopes or antigenic determinants are located within the N-terminus of the A ⁇ peptide and include residues within amino acids 1-10 of A ⁇ , preferably from residues 1-3, 1-4, 1-5, 1-6, 1-7 or 3-7 of A ⁇ 42. Additional referred epitopes or antigenic determinants include residues 2-4, 5, 6, 7 or 8 of A ⁇ , residues 3-5, 6, 7, 8 or 9 of A ⁇ , or residues 4-7, 8, 9 or 10 of A ⁇ 42.
  • amyloidogenic disease includes any disease associated with (or caused by) the formation or deposition of insoluble amyloid fibrils.
  • exemplary amyloidogenic diseases include, but are not limited to systemic amyloidosis, Alzheimer's disease, mature onset diabetes, Parkinson's disease, Huntington's disease, fronto-temporal dementia, and the prion-related transmissible spongiform encephalopathies (kuru and Creutzfeldt-Jacob disease in humans and scrapie and BSE in sheep and cattle, respectively).
  • Different amyloidogenic diseases are defined or characterized by the nature of the polypeptide component of the fibrils deposited.
  • ⁇ -amyloid protein e.g., wild-type, variant, or truncated ⁇ -amyloid protein
  • ⁇ -amyloid protein is the characterizing polypeptide 5 component of the amyloid deposit.
  • Alzheimer's disease is an example of a “disease characterized by deposits of A ⁇ ” or a “disease associated with deposits of A ⁇ ”, e.g., in the brain of a subject or patient.
  • the terms “ ⁇ -amyloid protein”, “ ⁇ -amyloid peptide”, “ ⁇ -amyloid”, “A ⁇ ” and “A ⁇ peptide” are used interchangeably herein.
  • the term “effective dose” or “effective dosage” is defined as an amount sufficient to achieve or at least partially achieve the desired effect.
  • therapeutically effective dose is defined as an amount sufficient to cure or at least partially arrest the disease and its complications in a patient already suffering from the disease. Amounts effective for this use will depend upon the severity of the infection and the general state of the patient's own immune system.
  • the term “patient” includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment.
  • “Soluble” or “dissociated” A ⁇ refers to non-aggregating or disaggregated A ⁇ polypeptide.
  • “Insoluble” A ⁇ refers to aggregating A ⁇ polypeptide, for example, A ⁇ held together by noncovalent bonds.
  • a ⁇ e.g., A ⁇ 42
  • a ⁇ is believed to aggregate, at least in part, due to the presence of hydrophobic residues at the C-terminus of the peptide (part of the transmembrane domain of APP).
  • One method to prepare soluble A ⁇ is to dissolve lyophilized peptide in neat DMSO with sonication. The resulting solution is centrifuged to remove any insoluble particulates.
  • effector function refers to an activity that resides in the Fc region of an antibody (e.g., an IgG antibody) and includes, for example, the ability of the antibody to bind effector molecules such as complement and/or Fc receptors, which can control several immune functions of the antibody such as effector cell activity, lysis, complement-mediated activity, antibody clearance, and antibody half-life.
  • effector molecule refers to a molecule that is capable of binding to the Fc region of an antibody (e.g., an IgG antibody) including, but not limited to, a complement protein or a Fc receptor.
  • effector cell refers to a cell capable of binding to the Fc portion of an antibody (e.g., an IgG antibody) typically via an Fc receptor expressed on the surface of the effector cell including, but not limited to, lymphocytes, e.g., antigen presenting cells and T cells.
  • an antibody e.g., an IgG antibody
  • Fc receptor expressed on the surface of the effector cell including, but not limited to, lymphocytes, e.g., antigen presenting cells and T cells.
  • Fc region refers to a C-terminal region of an IgG antibody, in particular, the C-terminal region of the heavy chain(s) of said IgG antibody. Although the boundaries of the Fc region of an IgG heavy chain can vary slightly, a Fc region is typically defined as spanning from about amino acid residue Cys226 to the carboxyl-terminus of an IGg heavy chain(s).
  • Kabat numbering unless otherwise stated, is defined as the numbering of the residues in, e.g., an IgG heavy chain antibody using the EU index as in Kabat et al. (Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)), expressly incorporated herein by reference.
  • Fc receptor refers to a receptor that binds to the Fc region of an antibody.
  • Typical Fc receptors which bind to an Fc region of an antibody include, but are not limited to, receptors of the Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc receptors are reviewed in Ravetch and Kinet, Annu. Rev. Immunol 9:457-92 (1991); Capel et al., Immunomethods 4:25-34 (1994); and de Haas el al., J. Lab. Clin. Med. 126:330-41 (1995).
  • Immunological and therapeutic reagents of the invention comprise or consist of immunogens or antibodies, or functional or antigen binding fragments thereof, as defined herein.
  • 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 and are about 230 residues in length.
  • Heavy chains are classified as gamma ( ⁇ ), mu ( ⁇ ), alpha ( ⁇ ), delta ( ⁇ ), or epsilon ( ⁇ ), are about 450-600 residues in length, and define the antibody's isotype as IgG, IgM, IgA, IgD and IgE, respectively.
  • Both heavy and light chains are folded into domains.
  • domain refers to a globular region of a protein, for example, an immunoglobulin or antibody.
  • Immunoglobulin or antibody domains include, for example, 3 or four peptide loops stabilized by ⁇ -pleated sheet and an interchain disulfide bond.
  • Intact light chains have, for example, two domains (V L and C L ) and intact heavy chains have, for example, four or five domains (V H , C H 1, C H 2, and C H 3).
  • 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.
  • variable regions of each light/heavy chain pair form the antibody binding site.
  • 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.
  • FR relatively conserved framework regions
  • Naturally-occurring chains or recombinantly produced chains can be expressed with a leader sequence which is removed during cellular processing to produce a mature chain. Mature chains can also be recombinantly produced having a non-naturally occurring leader sequence, for example, to enhance secretion or alter the processing of a particular chain of interest.
  • both light and heavy chains comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • FR4 also is referred to in the art as the D/J region of the variable heavy chain and the J region of the variable light chain.
  • 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). An alternative structural definition has been proposed by Chothia et al., J. Mol. Biol .
  • Therapeutic agents of the invention include antibodies that specifically bind to A ⁇ or other component of amyloid plaques. Such antibodies can be monoclonal or polyclonal. Some such antibodies bind specifically to the aggregated form of A ⁇ without binding to the soluble form. Some bind specifically to the soluble form without binding to the aggregated form. Some bind to both aggregated and soluble forms. Some such antibodies bind to a naturally occurring short form of A ⁇ (i.e., A ⁇ 39, 40 or 41) without binding to a naturally occurring long form of A ⁇ (i.e., A ⁇ 42 and A ⁇ 43). Some antibodies bind to a long form of A ⁇ without binding to a short form. Some antibodies bind to A ⁇ without binding to full-length amyloid precursor protein.
  • Antibodies used in therapeutic methods preferably 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 A ⁇ , and the other for an Fc receptor.
  • Preferred antibodies bind to A ⁇ with a binding affinity greater than (or equal to) about 10 6 , 10 7 , 10 8 , 10 9 , or 10 10 M ⁇ 1 (including affinities intermediate of these values).
  • Polyclonal sera typically contain mixed populations of antibodies binding to several epitopes along the length of A ⁇ . However, polyclonal sera can be specific to a particular segment of A ⁇ , such as A ⁇ 1-10. Monoclonal antibodies bind to a specific epitope within A ⁇ 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 A ⁇ (with the first N terminal residue of natural A ⁇ 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 models 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 (e.g., lack the ability to reduce plaque burden and/or resolve the neuritic pathology associated with Alzheimer's disease).
  • 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 A ⁇ can also be used (e.g., administered or co-administered). For example, antibodies can be directed to the amyloid associated protein synuclein.
  • an antibody When an antibody is said to bind to an epitope within specified residues, such as A ⁇ 1-5 for example, what is meant is that the antibody specifically binds to a polypeptide containing the specified residues (i.e., A ⁇ 1-5 in this an example). Such an antibody does not necessarily contact every residue within A ⁇ 1-5. Nor does every single amino acid substitution or deletion with in A ⁇ 1-5 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 A ⁇ . The phage display library is then selected for members specifically binding to an antibody under test. A family of sequences is isolated.
  • 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 A ⁇ bind to the same or similar epitope as 3D6, i.e., within residues A ⁇ 1-5. Likewise antibodies that compete with the 10D5 antibody bind to the same or similar epitope, i.e., within residues A ⁇ 3-7. 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 A ⁇ is likely to be effective in preventing and treating Alzheimer's disease according to the methodologies of the present invention.
  • Monoclonal or polyclonal antibodies that specifically bind to a preferred segment of A ⁇ without binding to other regions of A ⁇ have a number of advantages relative to monoclonal antibodies binding to other regions or polyclonal sera to intact A ⁇ .
  • dosages of antibodies that specifically bind to preferred segments contain a higher molar dosage of antibodies effective in clearing amyloid plaques.
  • 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 side effects.
  • the present invention features non-human antibodies, for example, antibodies having specificity for the preferred A ⁇ epitopes of the invention.
  • Such antibodies can be used in formulating various therapeutic compositions of the invention or, preferably, provide complementarity determining regions for the production of humanized or chimeric antibodies (described in detail below).
  • 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 A ⁇ .
  • a longer polypeptide comprising A ⁇ or an immunogenic fragment of A ⁇ or anti-idiotypic antibodies to an antibody to A ⁇ can also be used. See Harlow & Lane, supra, incorporated by reference for all purposes).
  • an immunogen can be obtained from a natural source, by peptide synthesis or by recombinant expression.
  • the immunogen can be administered fused or otherwise complexed with a carrier protein, as described below.
  • the immunogen can be administered with an adjuvant.
  • 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.
  • adjuvants 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.
  • Exemplary preparation of polyclonal antibodies, e.g., for passive protection, can be performed as follows. 125 non-transgenic mice are immunized with 100 ⁇ g A ⁇ 1-42, plus CFA/IFA adjuvant, and euthanized at 4-5 months. Blood is collected from immunized mice. IgG is separated from other blood components. Antibody specific for the immunogen may be partially purified by affinity chromatography. An average of about 0.5-1 mg of immunogen-specific antibody is obtained per mouse, giving a total of 60-120 mg.
  • mice are typically used for making monoclonal antibodies.
  • Monoclonals can be prepared against a fragment by injecting the fragment or longer form of A ⁇ into a mouse, preparing hybridomas and screening the hybridomas for an antibody that specifically binds to A ⁇ .
  • antibodies are screened for binding to a specific region or desired fragment of A ⁇ without binding to other nonoverlapping fragments of A ⁇ . The latter screening can be accomplished by determining binding of an antibody to a collection of deletion mutants of an A ⁇ 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.
  • 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 such 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 IgG1.
  • the present invention also features chimeric and/or humanized antibodies (i.e., chimeric and/or humanized immunoglobulins) specific for beta amyloid peptide.
  • Chimeric and/or 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 antibody refers to an antibody whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin gene segments belonging to different species.
  • V variable
  • C constant
  • Human isotype IgG1 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 antibody refers to an antibody comprising at least one chain comprising variable region framework residues substantially from a human antibody chain (referred to as the acceptor immunoglobulin or antibody) and at least one complementarity determining region substantially from a mouse-antibody, (referred to as the donor immunoglobulin or antibody).
  • acceptor immunoglobulin or antibody referred to as the acceptor immunoglobulin or antibody
  • donor immunoglobulin or antibody referred to as the donor immunoglobulin or antibody.
  • the substitution of mouse CDRs into a human variable domain framework is most likely to result in retention of their correct spatial orientation if the human variable domain framework adopts the same or similar conformation to the mouse variable framework from which the CDRs originated. This is achieved by obtaining the human variable domains from human antibodies whose framework sequences exhibit a high degree of sequence identity with the murine variable framework domains from which the CDRs were derived.
  • the heavy and light chain variable framework regions 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 Kettleborough el al., Protein Engineering 4:773 (1991); Kolbinger et al., Protein Engineering 6:971 (1993) and Carter et al., WO 92/22653.
  • the next step is to determine which, if any, residues from these components should be substituted to optimize the properties of the resulting humanized antibody.
  • substitution of human amino acid residues with murine should be minimized, because introduction of nurine residues increases the risk of the antibody eliciting a human-anti-mouse-antibody (HAMA) response in humans.
  • HAMA human-anti-mouse-antibody
  • Art-recognized methods of determining immune response can be performed to monitor a HAMA response in a particular patient or during clinical trials. Patients administered humanized antibodies can be given an immunogenicity assessment at the beginning and throughout the administration of said therapy.
  • the HAMA response is measured, for example, by detecting antibodies to the humanized therapeutic reagent, in serum samples from the patient using a method known to one in the art, including surface plasmon resonance technology (BIACORE) and/or solid-phase ELISA analysis.
  • BIACORE surface plasmon resonance technology
  • 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.
  • the unnatural juxtaposition of murine CDR regions with human variable framework region can result in unnatural conformational restraints, which, unless corrected by substitution of certain amino acid residues, lead to loss of binding affinity.
  • the selection of amino acid residues for substitution is determined, in part, by computer modeling.
  • Computer hardware and software are described herein for producing three-dimensional images of immunoglobulin molecules.
  • molecular models are produced starting from solved structures for immunoglobulin chains or domains thereof.
  • the chains to be modeled are compared for amino acid sequence similarity with chains or domains of solved three-dimensional structures, and the chains or domains showing the greatest sequence similarity is/are selected as starting points for construction of the molecular model.
  • Chains or domains sharing at least 50% sequence identity are selected for modeling, and preferably those sharing at least 60%, 70%, 80%, 90% sequence identity or more are selected for modeling.
  • the solved starting structures are modified to allow for differences between the actual amino acids in the immunoglobulin chains or domains being modeled, and those in the starting structure.
  • the modified structures are then assembled into a composite immunoglobulin.
  • the model is refined by energy minimization and by verifying that all atoms are within appropriate distances from one another and that bond lengths and angles are within chemically acceptable limits.
  • amino acid residues for substitution can also be determined, in part, by 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:
  • Residues which “noncovalently bind antigen directly” include amino acids in positions in framework regions which are have a good probability of directly interacting with amino acids on the antigen according to established chemical forces, for example, by hydrogen bonding, Van der Waals forces, hydrophobic interactions, and the like.
  • CDR and framework regions are as defined by Kabat et al. or Chothia et al., supra.
  • framework residues as defined by Kabat et al., supra, constitute structural loop residues as defined by Chothia et al., supra, the amino acids present in the mouse antibody may be selected for substitution into the humanized antibody.
  • Residues which are “adjacent to a CDR region” include amino acid residues in positions immediately adjacent to one or more of the CDRs in the primary sequence of the humanized immunoglobulin chain, for example, in positions immediately adjacent to a CDR as defined by Kabat, or a CDR as defined by Chothia (See e.g., Chothia and Lesk JMB 196:901 (1987)). These amino acids are particularly likely to interact with the amino acids in the CDRs and, if chosen from the acceptor, to distort the donor CDRs and reduce affinity.
  • the adjacent amino acids may interact directly with the antigen (Amit et al., Science, 233:747 (1986), which is incorporated herein by reference) and selecting these amino acids from the donor may be desirable to keep all the antigen contacts that provide affinity in the original antibody.
  • Residues that “otherwise interact with a CDR region” include those that are determined by secondary structural analysis to be in a spatial orientation sufficient to effect a CDR region.
  • residues that “otherwise interact with a CDR region” are identified by analyzing a three-dimensional model of the donor immunoglobulin (e.g., a computer-generated model).
  • a three-dimensional model typically of the original donor antibody, shows that certain amino acids outside of the CDRs are close to the CDRs and have a good probability of interacting with amino acids in the CDRs by hydrogen bonding, Van der Waals forces, hydrophobic interactions, etc.
  • the donor immunoglobulin amino acid rather than the acceptor immunoglobulin amino acid may be selected.
  • Amino acids according to this criterion will generally have a side chain atom within about 3 angstrom units ( ⁇ ) of some atom in the CDRs and must contain an atom that could interact with the CDR atoms according to established chemical forces, such as those listed above.
  • the 3 ⁇ is measured between their nuclei, but for atoms that do not form a bond, the 3 ⁇ is measured between their Van der Waals surfaces.
  • the nuclei must be within about 6 ⁇ (3 ⁇ plus the sum of the Van der Waals radii) for the atoms to be considered capable of interacting. In many cases the nuclei will be from 4 or 5 to 6 ⁇ apart.
  • Amino acids that are capable of interacting with amino acids in the CDRs may be identified in yet another way.
  • the solvent accessible surface area of each framework amino acid is calculated in two ways: (1) in the intact antibody, and (2) in a hypothetical molecule consisting of the antibody with its CDRs removed. A significant difference between these numbers of about 10 square angstroms or more shows that access of the framework amino acid to solvent is at least partly blocked by the CDRs, and therefore that the amino acid is making contact with the CDRs.
  • Solvent accessible surface area of an amino acid may be calculated based on a three-dimensional model of an antibody, using algorithms known in the art (e.g., Connolly, J. Appl. Cryst. 16:548 (1983) and Lee and Richards, J. Mol. Biol. 55:379 (1971), both of which are incorporated herein by reference).
  • Framework amino acids may also occasionally interact with the CDRs indirectly, by affecting the conformation of another framework amino acid that in turn contacts the CDRs.
  • amino acids at several positions in the framework are known to be capable of interacting with the CDRs in many antibodies (Chothia and Lesk, supra, Chothia et al., supra and Tramontano et al., J. Mol. Biol. 215:175 (1990), all of which are incorporated herein by reference).
  • amino acids at positions 2, 48, 64 and 71 of the light chain and 26-30, 71 and 94 of the heavy chain are known to be capable of interacting with the CDRs in many antibodies.
  • the amino acids at positions 35 in the light chain and 93 and 103 in the heavy chain are also likely to interact with the CDRs.
  • Residues which “participate in the VL-VH interface” or “packing residues” include those residues at the interface between VL and VH as defined, for example, by Novotny and Haber, Proc. Natl. Acad. Sci. USA, 82:4592-66 (1985) or Chothia et al, supra. Generally, unusual packing residues should be retained in the humanized antibody if they differ from those in the human frameworks.
  • amino acids fulfilling the above criteria are substituted. In some embodiments, all or most of the amino acids fulfilling the above criteria are substituted. Occasionally, there is some ambiguity about whether a particular amino acid meets the above criteria, and alternative variant immunoglobulins are produced, one of which has that particular substitution, the other of which does not. Alternative variant inimunoglobulins so produced can be tested in any of the assays described herein for the desired activity, and the preferred immunoglobulin selected.
  • CDR regions in humanized antibodies are substantially identical, and more usually, identical to the corresponding CDR regions of the donor antibody.
  • conservative amino acid substitutions are intended combinations such as gly, ala; val, ile, leu; asp, glu; asn, gln; ser, thr; lys, arg; and phe, tyr.
  • Additional candidates for substitution are acceptor human framework amino acids that are unusual or “rare” 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. For example, substitution may be desirable when the amino acid in a human framework region of the acceptor immunoglobulin is rare for that position and the corresponding amino acid in the donor immunoglobulin is common for that position in human immunoglobulin sequences; or when the amino acid in the acceptor immunoglobulin is rare for that position and the corresponding amino acid in the donor immunoglobulin is also rare, relative to other human sequences. These criterion help ensure that an atypical amino acid in the human framework does not disrupt the antibody structure. Moreover, by replacing an unusual human acceptor amino acid with an amino acid from the donor antibody that happens to be typical for human antibodies, the humanized antibody may be made less immunogenic.
  • rare indicates an amino acid occurring at that position in less than about 20% but usually less than about 10% of sequences in a representative sample of sequences
  • common indicates an amino acid occurring in more than about 25% but usually more than about 50% of sequences in a representative sample.
  • all human light and heavy chain variable region sequences are respectively grouped into “subgroups” of sequences that are especially homologous to each other and have the same amino acids at certain critical positions (Kabat et al., supra).
  • CDR1 in the variable heavy chain is defined as including residues 26-32.
  • Additional candidates for substitution are acceptor framework residues that correspond to a rare or unusual donor framework residue.
  • Rare or unusual donor framework residues are those that are rare or unusual (as defined herein) for murine antibodies at that position.
  • the subgroup can be determined according to Kabat and residue positions identified which differ from the consensus. These donor specific differences may point to somatic mutations in the murine sequence which enhance activity. Unusual residues that are predicted to affect binding are retained, whereas residues predicted to be unimportant for binding can be substituted.
  • Additional candidates for substitution are non-germline residues occurring in an acceptor framework region.
  • an acceptor antibody chain i.e., a human antibody chain sharing significant sequence identity with the donor antibody chain
  • a germline antibody chain likewise sharing significant sequence identity with the donor chain
  • residues not matching between acceptor chain framework and the germline chain framework can be substituted with corresponding residues from the germline sequence.
  • the framework regions of humanized immunoglobulins are usually substantially identical, and more usually, identical to the framework regions of the human antibodies from which they were derived.
  • many of the amino acids in the framework region make little or no direct contribution to the specificity or affinity of an antibody.
  • many individual conservative substitutions of framework residues can be tolerated without appreciable change of the specificity or affinity of the resulting humanized immunoglobulin.
  • the variable framework region of the humanized immunoglobulin shares at least 85% sequence identity to a human variable framework region sequence or consensus of such sequences.
  • variable framework region of the humanized immunoglobulin shares at least 90%, preferably 95%, more preferably 96%, 97%, 98% or 99% sequence identity to a human variable framework region sequence or consensus of such sequences. In general, however, such substitutions are undesirable.
  • the humanized antibodies preferably exhibit a specific binding affinity for antigen of at least 10 7 , 10 8 , 10 9 , or 10 10 M 31 1 .
  • the upper limit of binding affinity of the humanized antibodies for antigen is within a factor of three, four or five of that of the donor immunoglobulin.
  • the lower limit of binding affinity is also within a factor of three, four or five of that of donor immunoglobulin.
  • the binding affinity can be compared to that of a humanized antibody having no substitutions (e.g., an antibody having donor CDRs and acceptor FRs, but no FR substitutions).
  • the binding of the optimized antibody (with substitutions) is preferably at least two- to three-fold greater, or three- to four-fold greater, than that of the unsubstituted antibody.
  • activity of the various antibodies can be determined, for example, by BIACORE (i.e., surface plasmon resonance using unlabelled reagents) or competitive binding assays.
  • a preferred embodiment of the present invention features a humanized antibody to the N-terminus of A ⁇ , in particular, for use in the therapeutic and/or diagnostic methodologies described herein.
  • a particularly preferred starting material for production of humanized antibodies is 3D6.
  • 3D6 is specific for the N-terminus of A ⁇ and has been shown to mediate phagocytosis (e.g., induce phagocytosis) of amyloid plaque (see Examples I-V).
  • the cloning and sequencing of cDNA encoding the 3D6 antibody heavy and light chain variable regions is described in Example VI.
  • Suitable human acceptor antibody sequences are identified by computer comparisons of the amino acid sequences of the mouse variable regions with the sequences of known human antibodies. The comparison is performed separately for heavy and light chains but the principles are similar for each.
  • variable domains from human antibodies whose framework sequences exhibit a high degree of sequence identity with the murine VL and VH framework regions were identified by query of the Kabat Database using NCBI BLAST (publicly accessible through the National Institutes of Health NCBI internet server) with the respective murine framework sequences.
  • acceptor sequences sharing greater that 50% sequence identity with murine donor sequences are selected.
  • acceptor antibody sequences sharing 60%, 70%, 80%, 90% or more are selected.
  • light and heavy human framework regions are preferably derived from human antibodies of these subtypes, or from consensus sequences of such subtypes.
  • the preferred light chain human variable regions showing greatest sequence identity to the corresponding region from 3D6 are from antibodies having Kabat ID Numbers 019230, 005131, 005058, 005057, 005059, U21040 and U41645, with 019230 being more preferred.
  • the preferred heavy chain human variable regions showing greatest sequence identity to the corresponding region from 3D6 are from antibodies having Kabat ID Numbers 045919, 000459, 000553, 000386 and M23691, with 045919 being more preferred.
  • Residues are next selected for substitution, as follows.
  • the human framework amino acid should usually be substituted by the equivalent mouse amino acid if it is reasonably expected that the amino acid:
  • a CDR region e.g., is within about 3 ⁇ of a CDR region
  • VL-VH interface e.g., amino acids at positions L36, L46 and H93 of 3D6
  • the model was further refined by a series of energy minimization steps to relieve unfavorable atomic contacts and optimize electrostatic and van der Walls interactions.
  • the solved structure of 1qkz (PDB ID: 1 QKZ, Derrick et al., J. Mol. Biol . 293:81 (1999)) was chosen as a template for modeling CDR3 of the heavy chain as 3D6 and 1OPG did not exhibit significant sequence homology in this region when aligned for comparison purposes.
  • Three-dimensional structural information for the antibodies described herein is publicly available, for example, from the Research Collaboratory for Structural Bioinformatics' Protein Data Bank (PDB).
  • PDB is freely accessible via the World Wide Web internet and is described by Berman et al. (2000) Nucleic Acids Research , 28:235.
  • Computer modeling allows for the identification of CDR-interacting residues.
  • the computer model of the structure of 3D6 can in turn serve as a starting point for predicting the three-dimensional structure of an antibody containing the 3D6 complementarity determining regions substituted in human framework structures. Additional models can be constructed representing the structure as further amino acid substitutions are introduced.
  • the humanized antibodies of the present invention will usually contain a substitution of a human light chain framework residue with a corresponding 3D6 residue in at least 1, 2 or 3, and more usually 4, of the following positions: L1, L2, L36 and L46.
  • the humanized antibodies also usually contain a substitution of a human heavy chain framework residue with a corresponding 3D6 residue in at least 1, 2, and sometimes 3, of the following positions: H49, H93 and H94.
  • Humanized antibodies can also contain a substitution of a heavy chain framework residue with a corresponding germline residue in at least 1, 2, and sometimes 3, of the following positions: H74, H77 and H89.
  • the human light chain framework acceptor immunoglobulin is Kabat ID Number 019230
  • the light chain contains substitutions in at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more usually 13, of the following positions: L7, L10, L12, L15, L17, L39, L45, L63, L78, L83, L85, L100 or L104.
  • the heavy chain contains substitutions in at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or more usually 15, of the following positions: H3, H5, H13, H16, H19, H40, H41, H42, H44, H72, H77, H82A, H83, H84, or H108. These positions are substituted with the amino acid from the equivalent position of a human immunoglobulin having a more typical amino acid residue. Examples of appropriate amino acids to substitute are shown in FIGS. 1 and 2.
  • germline sequences having the greatest degree of identity to the 3D6 light chain include A1A17, A18, A2 and A19, with A19 being most preferred. Residues not matching between a selected light chain acceptor framework and one of these germline sequences could be selected for substitution with the corresponding germline residue.
  • Table 1 summarizes the sequence analysis of the 3D6 VH and VL regions. Additional mouse and human structures that can be used for computer modeling of the 3D6 antibody and additional human antibodies are set forth as well as germline sequences that can be used in selecting amino acid substitutions. Rare mouse residues are also set forth in Table 1. Rare mouse residues are identified by comparing the donor VL and/or VH sequences with the sequences of other members of the subgroup to which the donor VL and/or VH sequences belong (according to Kabat) and identifying the residue positions which differ from the consensus. These donor specific differences may point to somatic mutations which enhance activity. Unusual or rare residues close to the binding site may possibly contact the antigen, making it desirable to retain the mouse residue.
  • the unusual mouse residue is not important for binding, use of the corresponding acceptor residue is preferred as the mouse residue may create immunogenic neoepitopes in the humanized antibody.
  • the preferred residue is clearly the acceptor residue.
  • Germline gene sequences referenced herein are publicly available, for example, from the National Center for Biotechnology Information (NCBI) database of sequences in collections of Igh, Ig kappa and Ig lambda germline V genes (as a division of the National Library of Medicine (NLM) at the National Institutes of Health (NIH)). Homology searching of the NCBI “Ig Germline Genes” database is provided by IgG BLASTTM.
  • NCBI National Center for Biotechnology Information
  • a humanized antibody of the present invention contains (i) a light chain comprising a variable domain comprising murine 3D6 VL CDRs and a human acceptor framework, the framework having at least one, preferably two, three or four residues selected from the group consisting of L1, L2, L36, and L46 substituted with the corresponding 3D6 residue and (ii) a heavy chain comprising 3D6 VH CDRs and a human acceptor framework, the framework having at least one, preferably two or three residues selected from the group consisting of H49, H93 and H94 substituted with the corresponding 3D6 residue, and, optionally, at least one, preferably two or three residues selected from the group consisting of H74, H77 and H89 is substituted with a corresponding human germline residue.
  • a humanized antibody of the present invention contains (i) a light chain comprising a variable domain comprising murine 3D6 VL CDRs and a human acceptor framework, the framework having residue 1 substituted with a tyr (Y), residue 2 substituted with a val (V), residue 36 substituted with a leu (L) and/or residue 46 substituted with an arg (R), and (ii) a heavy chain comprising 3D6 VH CDRs and a human acceptor framework, the framework having residue 49 substituted with an ala (A), residue 93 substituted with a val (V) and/or residue 94 substituted with an arg (R), and, optionally, having residue 74 substituted with a ser (S), residue 77 substituted with a thr (T) and/or residue 89 substituted with a val (V).
  • a humanized antibody of the present invention has structural features, as described herein, and further has at least one (preferably two, three, four or all) of the following activities: (1) binds aggregated A ⁇ 1-42 (e.g., as determined by ELISA); (2) binds A ⁇ in plaques (e.g., staining of AD and/or PDAPP plaques); (3) binds A ⁇ with two- to three- fold higher binding affinity as compared to chimeric 3D6 (e.g., 3D6 having murine variable region sequences and human constant region sequences); (4) mediates phagocytosis of A ⁇ (e.g., in an ex vivo phagocytosis assay, as described herein); and (5) crosses the blood-brain barrier (e.g., demonstrates short-term brain localization, for example, in a PDAPP animal model, as described herein).
  • a ⁇ 1-42 e.g., as determined by ELISA
  • a ⁇ in plaques e.g., stain
  • a humanized antibody of the present invention has structural features, as described herein, binds A ⁇ in a manner or with an affinity sufficient to elicit at least one of the following in vivo effects: (1) reduce A ⁇ plaque burden; (2) prevent plaque formation; (3) reduce levels of soluble A ⁇ ; (4) reduce the neuritic pathology associated with an amyloidogenic disorder; (5) lessens or ameliorate at least one physiological symptom associated with an amyloidogenic disorder; and/or (6) improves cognitive function.
  • a humanized antibody of the present invention has structural features, as described herein, and specifically binds to an epitope comprising residues 1-5 or 3-7 of A ⁇ .
  • Activities described above can be determined utilizing any one of a variety of assays described herein or in the art (e.g., binding assays, phagocytosis assays etc.). Activities can be assayed either in vivo (e.g., using labeled assay components and/or imaging techniques) or in vitro (e.g., using samples or specimens derived from a subject). Activities can be assayed either directly or indirectly. In certain preferred embodiments, neurological endpoints (e.g., amyloid burden, neuritic burden, etc) are assayed.
  • assays described herein or in the art e.g., binding assays, phagocytosis assays etc.
  • Activities can be assayed either in vivo (e.g., using labeled assay components and/or imaging techniques) or in vitro (e.g., using samples or specimens derived from a subject). Activities can be assayed either directly or indirectly.
  • neurological endpoints
  • Such endpoints can be assayed in living subjects (e.g., in animal models of Alzheimer's disease or in human subjects, for example, undergoing immunotherapy) using non-invasive detection methodologies. Alternatively, such endpoints can be assayed in subjects post mortem. Assaying such endpoints in animal models and/or in human subjects post mortem is useful in assessing the effectiveness of various agents (e.g., humanized antibodies) to be utilized in similar immunotherapeutic applications. In other preferred embodiments, behavioral or neurological parameters can be assessed as indicators of the above neuropathological activities or endpoints.
  • agents e.g., humanized antibodies
  • Human antibodies against A ⁇ 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 herein. Human antibodies can also be screened for a particular epitope specificity by using only a fragment of A ⁇ as the immunogen, and/or by screening antibodies against a collection of deletion mutants of A ⁇ . Human antibodies preferably have human IgG1 isotype specificity.
  • 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 A ⁇ , a fragment thereof, larger polypeptide containing A ⁇ or fragment, or an anti-idiotypic antibody to an antibody to A ⁇ . 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.
  • RPMI-1640 see Engleman, supra
  • the immunized B-lymphocytes are fused to a xenogeneic hybrid cell such as SPAZ-4 by well-known methods.
  • 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 A ⁇ or a fragment thereof.
  • Triomas 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 A ⁇ or a fragment thereof.
  • 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 mamnalian, bacterial or yeast cell lines.
  • Human antibodies against A ⁇ can also be produced from non-human transgenic mammals having transgenes encoding at least a segment of the human immunoglobulin locus.
  • the endogenous immunoglobulin locus of such transgenic mammals is functionally inactivated.
  • the segment of the human immunoglobulin locus includes unrearranged sequences of heavy and light chain components.
  • 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., WO93/12227 (1993); U.S. Pat. No. 5,877,397, U.S. Pat. No. 5,874,299, U.S. Pat. No. 5,814,318, U.S. Pat. No. 5,789,650, U.S. Pat. No. 5,770,429, U.S. Pat. No. 5,661,016, U.S. Pat. No.
  • mice are particularly suitable.
  • Anti-A ⁇ antibodies are obtained by immunizing a transgenic nonhuman mammal, such as described by Lonberg or Kucherlapati, supra, with A ⁇ 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.
  • such polyclonal antibodies can be concentrated by affinity purification using A ⁇ or other amyloid peptide as an affinity reagent.
  • a further approach for obtaining human anti-A ⁇ 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).
  • B cells can be obtained from a human immunized with A ⁇ , fragments, longer polypeptides containing A ⁇ or fragments or anti-idiotypic antibodies.
  • B cells are obtained from a patient who is ultimately to receive antibody treatment.
  • Antibodies binding to A ⁇ 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.
  • libraries of phage are produced in which members display different antibodies on their outer surfaces.
  • Antibodies are usually displayed as Fv or Fab fragments.
  • Phage displaying antibodies with a desired specificity are selected by affinity enrichment to an A ⁇ peptide or fragment thereof.
  • human antibodies having the binding specificity of a selected murine antibody can be produced. See Winter, WO 92/20791.
  • 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 A ⁇ (e.g., at least 10 8 and preferably at least 10 9 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.
  • 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 A ⁇ are selected. These phage display the variable regions of completely human anti-A ⁇ antibodies. These antibodies usually have the same or similar epitope specificity as the murine starting material.
  • nucleic acid sequences will encode each immunoglobulin amino acid sequence.
  • the desired nucleic acid sequences can be produced by de novo solid-phase DNA synthesis or by PCR mutagenesis of an earlier prepared variant of the desired polynucleotide. Oligonucleotide-mediated mutagenesis is a preferred method for preparing substitution, deletion and insertion variants of target polypeptide DNA. See Adelman et al., DNA 2:183 (1983).
  • the target polypeptide DNA is altered by hybridizing an oligonucleotide encoding the desired mutation to a single-stranded DNA template. After hybridization, a DNA polymerase is used to synthesize an entire second complementary strand of the template that incorporates the oligonucleotide primer, and encodes the selected alteration in the target polypeptide DNA.
  • variable segments of antibodies produced as described supra are typically linked to at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • Human constant region DNA sequences can be isolated in accordance with well known procedures from a variety of human cells, but preferably immortalized B cells (see Kabat et al., supra, and Liu et al., W087/0267 1) (each of which is incorporated by reference in its entirety for all purposes).
  • the antibody will contain both light chain and heavy chain constant regions.
  • the heavy chain constant region usually includes CH1, hinge, CH2, CH3, and CH4 regions.
  • the antibodies described herein include antibodies having all types of constant regions, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgG1, IgG2, IgG3 and IgG4.
  • 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.
  • the constant domain is usually a complement fixing constant domain and the class is typically IgG1.
  • the constant domain may be of the IgG2 class.
  • 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.
  • the humanized antibody may comprise sequences from more than one class or isotype.
  • 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.
  • Chimeric, humanized and human antibodies are typically produced by recombinant expression.
  • Nucleic acids encoding light and heavy chain variable region's, optionally linked to constant regions, are inserted into expression vectors.
  • the light and heavy chains can be cloned in the same or different expression vectors.
  • the DNA segments encoding immunoglobulin chains are operably linked to control sequences in the expression vector(s) that ensure the expression of immunoglobulin polypeptides.
  • Expression control sequences include, but are not limited to, promoters (e.g., naturally-associated or heterologous promoters), signal sequences, enhancer elements, and transcription termination sequences.
  • the expression control sequences are eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells.
  • the vector 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.
  • expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA.
  • expression vectors contain selection markers (e.g., ampicillin-resistance, hygromycin-resistance, tetracycline resistance or neomycin resistance) to permit detection of those cells transformed with the desired DNA sequences (see, e.g., Itakura et al., U.S. Pat. No. 4,704,362).
  • E. coli is one prokaryotic host particularly useful for cloning the polynucleotides (e.g., DNA sequences) of the present invention.
  • Other microbial hosts suitable for use include bacilli, such as Bacillus subtilus , and other enterobacteriaceae such as Salmonella, Serratia, and various Pseudomonas species.
  • bacilli such as Bacillus subtilus
  • enterobacteriaceae such as Salmonella, Serratia, and various Pseudomonas species.
  • expression vectors which will typically contain expression control sequences compatible with the host cell (e.g., an origin of replication).
  • any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda.
  • the promoters will typically control expression, optionally with an operator sequence, and have ribosome binding site sequences and the like, for initiating and completing transcription and translation.
  • yeast Other microbes, such as yeast, are also useful for expression. Saccharomyces is a preferred yeast host, with suitable vectors having expression control sequences (e.g., promoters), 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.
  • 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 tissue cell culture may also be used to express and produce the polypeptides of the present invention (e.g., polynucleotides encoding immunoglobulins or fragments thereof). See Winnacker, From Genes to Clones, VCH Publishers, N.Y., N.Y. (1987).
  • Eukaryotic cells are actually preferred, because a number of suitable host cell lines capable of secreting heterologous proteins (e.g., intact immunoglobulins) have been developed in the art, and include CHO cell lines, various Cos cell lines, HeLa cells, preferably, myeloma cell lines, or transformed B-cells or hybridomas.
  • the cells are nonhuman.
  • Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, and 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 immunoglobulin genes, SV40, adenovirus, bovine papilloma virus, cytomegalovirus and the like. See Co et al., J. Immunol . 148:1149 (1992).
  • 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., Deboer et al., U.S. Pat. No. 5,741,957, Rosen, U.S. Pat. No. 5,304,489, and Meade et al., U.S. Pat. No. 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 polynucleotide sequences of interest can be transferred into the host cell by well-known methods, which vary 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-based transfection may be used for other cellular hosts. (See generally Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Press. 2nd ed., 1989) (incorporated by reference in its entirety for all purposes).
  • transgenic animals 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.
  • the vectors are co-transfected to obtain expression and assembly of intact immunoglobulins.
  • the whole antibodies, their dimers, individual light and heavy chains, or other immunoglobulin forms of the present invention can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, HPLC purification, gel electrophoresis and the like (see generally Scopes, Protein Purification (Springer-Verlag, N.Y., (1982)).
  • Substantially pure immunoglobulins of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity most preferred, for pharmaceutical uses.
  • antibody fragments are also contemplated within the scope of the instant invention.
  • fragments of non-human, chimeric and/or human antibodies are provided.
  • fragments of humanized antibodies are provided. Typically, these fragments exhibit specific binding to antigen with an affinity of at least 10 7 , and more typically 10 8 or 10 9 M ⁇ 1 .
  • Humanized antibody fragments include 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.
  • mice are injected intraperitoneally as needed over a 4 month period to maintain a circulating antibody concentration measured by ELISA titer of greater than 1/1000 defined by ELISA to A ⁇ 42 or other immunogen. Titers are monitored and mice are euthanized at the end of 6 months of injections. Histochemistry, A ⁇ levels and toxicology are performed post mortem. Ten mice are used per group.
  • the invention also 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.
  • 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 phagocytic cells can be a primary culture or a cell line, such as BV-2, C8-B4, or THP-1.
  • the components are combined on a microscope slide to facilitate microscopic monitoring.
  • multiple reactions are performed in parallel in the wells of a microtiter dish.
  • a separate miniature microscope slide can be mounted in the separate wells, or a nonmicroscopic detection format, such as ELISA detection of A ⁇ can be used.
  • 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 labeled antibody to A ⁇ 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.
  • 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.
  • tissue bearing pathological matrices between cells e.g., fibrinous pericarditis
  • tissue bearing aberrant antigens e.g., scar tissue.
  • isolated biological entities that can be used include A ⁇ , 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.
  • the antibody and biological entity are contacted with each other before adding the phagocytic cells.
  • the concentration of the biological entity and/or the associated antigen remaining in the medium, if present, 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 IV).
  • the effector function of an antibody resides in the constant or Fc region of the molecule which can mediate binding to various effector molecules, e.g., complement proteins or Fc receptors.
  • the binding of complement to the Fc region is important, for example, in the opsonization and lysis of cell pathogens and the activation of inflammatory responses.
  • the binding of antibody to Fc receptors, for example, on the surface of effector cells can trigger a number of important and diverse biological responses including, for example, engulfment and destruction of antibody-coated pathogens or particles, clearance of immune complexes, lysis of antibody-coated target cells by killer cells (i.e., antibody-dependent cell-mediated cytotoxicity, or ADCC), release of inflammatory mediators, placental transfer of antibodies, and control of immunoglobulin production.
  • killer cells i.e., antibody-dependent cell-mediated cytotoxicity, or ADCC
  • the above-mentioned immune functions may be desirable.
  • various aspects of the effector function of the molecule including enhancing or suppressing various reactions of the immune system, with beneficial effects in diagnosis and therapy, are achieved.
  • the antibodies of the invention can be produced which react only with certain types of Fc receptors, for example, the antibodies of the invention can be modified to bind to only certain Fc receptors, or if desired, lack Fc receptor binding entirely, by deletion or alteration of the Fc receptor binding site located in the Fc region of the antibody.
  • Other desirable alterations of the Fc region of an antibody of the invention are cataloged below.
  • the Kabat numbering system is used to indicate which amino acid residue(s) of the Fc region (e.g., of an IgG antibody) are altered (e.g., by amino acid substitution) in order to achieve a desired change in effector function.
  • the numbering system is also employed to compare antibodies across species such that a desired effector function observed in, for example, a mouse antibody, can then be systematically engineered into a human, humanized, or chimeric antibody of the invention.
  • antibodies e.g., IgG antibodies
  • Fc receptor e.g. an Fc receptor on human monocytes (Fc ⁇ RI)
  • Fc ⁇ RI Fc receptor on human monocytes
  • a comparison of the sequence of these proteins in the hinge-link region shows that the sequence 234 to 238, i.e., Leu-Leu-Gly-Gly-Pro in the strong binders becomes Leu-Glu-Gly-Gly-Pro in mouse gamma 2 b , i.e., weak binders. Accordingly, a corresponding change in a human antibody hinge sequence can be made if reduced Fc ⁇ I receptor binding is desired. It is understood that other alterations can be made to achieve the same or similar results.
  • the affinity of Fc ⁇ RI binding can be altered by replacing the specified residue with a residue having an inappropriate functional group on its sidechain, or by introducing a charged functional group (e.g., Glu or Asp) or for example an aromatic non-polar residue (e.g., Phe, Tyr, or Trp).
  • a charged functional group e.g., Glu or Asp
  • an aromatic non-polar residue e.g., Phe, Tyr, or Trp
  • the antibodies of the invention can also have an altered Fc region with altered binding affinity for Fc ⁇ RI as compared with the unmodified antibody.
  • Such an antibody conveniently has a modification at amino acid residue 234, 235, 236, or 237.
  • Affinity for other Fc receptors can be altered by a similar approach, for controlling the immune response in different ways.
  • the lytic properties of IgG antibodies following binding of the Cl component of complement can be altered.
  • the first component of the complement system, Cl comprises three proteins known as Clq, Clr and Cls which bind tightly together. It has been shown that Clq is responsible for binding of the three protein complex to an antibody.
  • the Clq binding activity of an antibody can be altered by providing an antibody with an altered CH 2 domain in which at least one of the amino acid residues 318, 320, and 322 of the heavy chain has been changed to a residue having a different side chain.
  • the numbering of the residues in the heavy chain is that of the EU index (see Kabat et al., supra).
  • Other suitable alterations for altering, e.g., reducing or abolishing specific Clq-binding to an antibody include changing any one of residues 318 (Glu), 320 (Lys) and 322 (Lys), to Ala.
  • Clq binding activity can be abolished by replacing any one of the three specified residues with a residue having an inappropriate functionality on its side chain. It is not necessary to replace the ionic residues only with Ala to abolish Clq binding. It is also possible to use other alkyl-substituted non-ionic residues, such as Gly, Ile, Leu, or Val, or such aromatic non-polar residues as Phe, Tyr, Trp and Pro in place of any one of the three residues in order to abolish Clq binding. In addition, it is also be possible to use such polar non-ionic residues as Ser, Thr, Cys, and Met in place of residues 320 and 322, but not 318, in order to abolish Clq binding activity.
  • the invention also provides an antibody having an altered effector function wherein the antibody has a modified hinge region.
  • the modified hinge region may comprise a complete hinge region derived from an antibody of different antibody class or subclass from that of the CH1 domain.
  • the constant domain (CH1) of a class IgG antibody can be attached to a hinge region of a class IgG4 antibody.
  • the new hinge region may comprise part of a natural hinge or a repeating unit in which each unit in the repeat is derived from a natural hinge region.
  • the natural hinge region is altered by converting one or more cysteine residues into a neutral residue, such as alanine, or by converting suitably placed residues into cysteine residues. Such alterations are carried out using art recognized protein chemistry and, preferably, genetic engineering techniques, as described herein.
  • the number of cysteine residues in the hinge region of the antibody is reduced, for example, to one cysteine residue.
  • This modification has the advantage of facilitating the assembly of the antibody, for example, bispecific antibody molecules and antibody molecules wherein the Fc portion has been replaced by an effector or reporter molecule, since it is only necessary to form a single disulfide bond.
  • This modification also provides a specific target for attaching the hinge region either to another hinge region or to an effector or reporter molecule, either directly or indirectly, for example, by chemical means.
  • the number of cysteine residues in the hinge region of the antibody is increased, for example, at least one more than the number of normally occurring cysteine residues. Increasing the number of cysteine residues can be used to stabilize the interactions between adjacent hinges. Another advantage of this modification is that it facilitates the use of cysteine thiol groups for attaching effector or reporter molecules to the altered antibody, for example, a radiolabel.
  • the invention provides for an exchange of hinge regions between antibody classes, in particular, IgG classes, and/or an increase or decrease in the number of cysteine residues in the hinge region in order to achieve an altered effector function (see for example U.S. Pat. No. 5,677,425 which is expressly incorporated herein).
  • a determination of altered antibody effector function is made using the assays described herein or other art recognized techniques.
  • the resultant antibody can be subjected to one or more assays to evaluate any change in biological activity compared to the starting antibody.
  • the ability of the antibody with an altered Fc region to bind complement or Fc receptors can be assessed using the assays disclosed herein as well as any art recognized assay.
  • Production of the antibodies of the invention is carried out by any suitable technique including techniques described herein as well as techniques known to those skilled in the art.
  • an appropriate protein sequence e.g. forming part of or all of a relevant constant domain, e.g., Fc region, i.e., CH2, and/or CH3 domain(s), of an antibody, and include appropriately altered residue(s) can be synthesized and then chemically joined into the appropriate place in an antibody molecule.
  • genetic engineering techniques are used for producing an altered antibody.
  • Preferred techniques include, for example, preparing suitable primers for use in polymerase chain reaction (PCR) such that a DNA sequence which encodes at least part of an IgG heavy chain, e.g., an Fc or constant region (e.g., CH2, and/or CH3) is altered, at one or more residues.
  • PCR polymerase chain reaction
  • the segment can then be operably linked to the remaining portion of the antibody, e.g., the variable region of the antibody and required regulatory elements for expression in a cell.
  • the present invention also includes vectors used to transform the cell line, vectors used in producing the transforming vectors, cell lines transformed with the transforming vectors, cell lines transformed with preparative vectors, and methods for their production.
  • the cell line which is transformed to produce the antibody with an altered Fc region is an immortalized mammalian cell line (e.g., CHO cell).
  • the cell line used to produce the antibody with an altered Fc region is preferably a mammalian cell line, any other suitable cell line, such as a bacterial cell line or a yeast cell line, may alternatively be used.
  • Immune responses against amyloid deposits can also be induced by administration of nucleic acids encoding antibodies and their component chains used for passive immunization.
  • 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.
  • regulatory elements such as, a promoter and enhancer
  • 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.
  • 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., J. Virol . 67:5911 (1993)); adeno-associated virus vectors (see, e.g., Zhou et al., J. Exp. Med .
  • viral vectors from the pox family including vaccinia virus and the avian pox viruses, viral vectors from the alpha virus genus such as those derived from Sindbis and Semliki Forest Viruses (see, e.g., Dubensky et al., J. Virol . 70:508 (1996)), Venezuelan equine encephalitis virus (see Johnston et al., U.S. Pat. No.
  • rhabdoviruses such as vesicular stomatitis virus (see Rose, WO 96/34625)and papillomaviruses (Ohe et al., Human Gene Therapy 6:325 (1995); Woo et al., WO 94/12629 and Xiao & Brandsma, Nucleic Acids. Res . 24, 2630-2622 (1996)).
  • DNA encoding an immunogen can be packaged into liposomes.
  • Suitable lipids and related analogs are described by Eppstein et al., U.S. Pat. No. 5,208,036, Felgner et al., U.S. Pat. No. 5,264,618, Rose, U.S. Pat. No. 5,279,833, and Epand et al., U.S. Pat. No. 5,283,185.
  • Vectors and DNA encoding an immunogen can also be adsorbed to or associated with particulate carriers, examples of which include polymethyl methacrylate polymers and polylactides and poly (lactide-co-glycolides), see, e.g., McGee et al., J. Micro Encap . (1996).
  • Gene therapy vectors or naked polypeptides 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., Anderson et al., U.S. Pat. No. 5,399,346).
  • systemic administration e.g., intravenous, intraperitoneal, nasal, gastric, intradermal, intramuscular, subdermal, or intracranial infusion
  • topical application see e.g., Anderson et al., U.S. Pat. No. 5,399,346
  • naked polynucleotide refers to a polynucleotide not complexed with colloidal materials. Naked polynucleotides are sometimes cloned in a plasmid vector.
  • Such vectors can further include facilitating agents such as bupivacine (Attardo et al., U.S. Pat. No. 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.
  • the AccelTM Gene Delivery Device manufactured by Agacetus, Inc. Middleton WI is suitable.
  • naked DNA can pass through skin into the blood stream simply by spotting the DNA onto skin with chemical or mechanical irritation (see Howell et al., WO 95/05853).
  • 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.
  • cells explanted from an individual patient e.g., lymphocytes, bone marrow aspirates, tissue biopsy
  • tissue biopsy e.g., hematopoietic stem cells
  • the present invention is directed inter alia to treatment of Alzheimer's and other amyloidogenic diseases by administration of therapeutic immunological reagents (e.g., humanized immunoglobulins) to specific epitopes within A ⁇ to a patient under conditions that generate a beneficial therapeutic response in a patient (e.g., induction of phagocytosis of A ⁇ , reduction of plaque burden, inhibition of plaque formation, reduction of neuritic dystrophy, improving cognitive function, and/or reversing, treating or preventing cognitive decline) in the patient, for example, for the prevention or treatment of an amyloidogenic disease.
  • the invention is also directed to use of the disclosed immunological reagents (e.g., humanized immunoglobulins) in the manufacture of a medicament for the treatment or prevention of an amyloidogenic disease.
  • treatment is defined as the application or administration of a therapeutic agent to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient, who has a disease, a symptom of disease or a predisposition toward a disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect the disease, the symptoms of disease or the predisposition toward disease.
  • the invention provides methods of preventing or treating a disease associated with amyloid deposits of A ⁇ in the brain of a patient.
  • 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.
  • Exemplary methods entail administering an effective dosage of an antibody that binds to A ⁇ .
  • Preferred methods entail administering an effective dosage of an antibody that specifically binds to an epitope within residues 1-10 of A ⁇ , for example, antibodies that specifically bind to an epitope within residues 1-3 of A ⁇ , antibodies that specifically bind to an epitope within residues 1-4 of A ⁇ , antibodies that specifically bind to an epitope within residues 1-5 of A ⁇ , antibodies that specifically bind to an epitope within residues 1-6 of A ⁇ , antibodies that specifically bind to an epitope within residues 1-7 of A ⁇ , or antibodies that specifically bind to an epitope within residues 3-7 of A ⁇ .
  • the invention features administering antibodies that bind to an epitope comprising a free N-terminal residue of A ⁇ .
  • the invention features administering antibodies that bind to an epitope within residues of 1-10 of A ⁇ wherein residue 1 and/or residue 7 of A ⁇ is aspartic acid.
  • the invention features administering antibodies that specifically bind to A ⁇ peptide without binding to full-length amyloid precursor protein (APP).
  • the isotype of the antibody is human IgG1.
  • the invention features administering antibodies that bind to an amyloid deposit in the patient and induce a clearing response against the amylcid deposit.
  • a clearing response can be effected by Fc receptor mediated phagocytosis.
  • 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.
  • the methods can be used on both asymptomatic patients and those currently showing symptoms of disease.
  • the antibodies used in such methods can be human, humanized, chimeric or nonhuman antibodies, or fragments thereof (e.g., antigen binding fragments) and can be monoclonal or polyclonal, as described herein.
  • the invention features administering antibodies prepared from a human immunized with A ⁇ peptide, which human can be the patient to be treated with antibody.
  • the invention features administering an antibody with a pharmaceutical carrier as a pharmaceutical composition.
  • the antibody can be administered to a patient by administering a polynucleotide encoding at least one antibody chain.
  • the polynucleotide is expressed to produce the antibody chain in the patient.
  • the polynucleotide encodes heavy and light chains of the antibody.
  • the polynucleotide is expressed to produce the heavy and light chains in the patient.
  • the patient is monitored for level of administered antibody in the blood of 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.
  • Patients amenable to treatment include individuals at risk of disease but not showing symptoms, as well as patients presently showing symptoms.
  • 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 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, supra).
  • 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.
  • a number of diagnostic tests are available for identifying individuals who have AD. These include measurement of CSF tau and AP42 levels. Elevated tau and decreased A ⁇ 42 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.
  • 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 levels 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.
  • 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.
  • 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.
  • 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.
  • agents are usually administered in several dosages until a sufficient immune response has been achieved.
  • the term “immune response” or “immunological response” includes the development of a humoral (antibody mediated) and/or a cellular (mediated by antigen-specific T cells or their secretion products) response directed against an antigen in a recipient subject. Such a response can be an active response, i.e., induced by administration of immunogen, or a passive response, i.e., induced by administration of immunoglobulin or antibody or primed T-cells.
  • 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. Typically, the immune response is monitored and repeated dosages are given if the immune response starts to wane.
  • 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.
  • the patient is a human but non-human mammals including transgenic mammals can also be treated. Treatment dosages need to be titrated to optimize safety and efficacy.
  • 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.
  • dosages can be 1 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg, preferably at least 1 mg/kg.
  • Subjects can be administered such doses daily, on alternative days, weekly or according to any other schedule determined by empirical analysis.
  • An exemplary treatment entails administration in multiple dosages over a prolonged period, for example, of at least six months. Additional exemplary treatment regimes entail administration once per every two weeks or once a month or once every 3 to 6 months.
  • Exemplary dosage schedules include 1 -10 mg/kg or 15 mg/kg on consecutive days, 30 mg/kg on alternate days or 60 mg/kg weekly.
  • 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 A ⁇ in the patient. In some methods, dosage is adjusted to achieve a plasma antibody concentration of 1-1000 ⁇ g/ml and in some methods 25-300 ⁇ g/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.
  • compositions containing the present antibodies or a cocktail thereof are administered to a patient not already in the disease state to enhance the patient's resistance. Such an amount is defined to be a “prophylactic effective dose.”
  • prophylactic effective dose the precise amounts again depend upon the patient's state of health and general immunity, but generally range from 0.1 to 25 mg per dose, especially 0.5 to 2.5 mg per dose.
  • a relatively low dosage is administered at relatively infrequent intervals over a long period of time.
  • a relatively high dosage e.g., from about 1 to 200 mg of antibody per dose, with dosages of from 5 to 25 mg being more commonly used
  • a 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 antibodies range from about 10 ng to 1 g, 100 ng to 100 mg, 1 ⁇ g to 10 mg, or 30-300 ⁇ g DNA per patient.
  • Doses for infectious viral vectors vary from 10-100, or more, virions per dose.
  • Therapeutic agents 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.
  • agents are injected directly into a particular tissue where deposits have accumulated, for example intracranial injection. Intramuscular injection or intravenous infusion are preferred for administration of antibody.
  • particular therapeutic antibodies are injected directly into the cranium.
  • 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.
  • 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.
  • 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.
  • compositions or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, noninimunogenic stabilizers and the like.
  • 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).
  • 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.
  • a pharmaceutical carrier that can be a sterile liquid such as water oils, saline, glycerol, or ethanol.
  • 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.
  • 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 HCl.
  • 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 (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.
  • 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 crosslinking or expression as a fusion protein.
  • transdermal delivery can be achieved using a skin path or using transferosomes (Paul et al., Eur. J. Immunol . 25:3521 (1995); Cevc et al., Biochem. Biophys. Acta 1368:201-15 (1998)).
  • the invention provides methods of monitoring treatment in a patient suffering from or susceptible to Alzheimer's, i.e., 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 passive immunization (e.g., measuring level of administered antibody).
  • Some methods entail determining a baseline value, for example, of an antibody level or profile in a patient, before administering a dosage of agent, and comparing this with a value for the profile or level 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
  • a positive treatment outcome i.e., that administration of the agent has achieved a desired response. If the value for immune response does not change significantly, or decreases, a negative treatment outcome is indicated.
  • a control value i.e., a mean and standard deviation
  • a control value i.e., a mean and standard deviation
  • Measured values of the level or profile 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
  • a lack of significant increase or a decrease signals a negative or insufficient treatment outcome.
  • Administration of agent is generally continued while the level is increasing relative to the control value. As before, attainment of a plateau relative to control values is an indicator that the administration of treatment can be discontinued or reduced in dosage and/or frequency.
  • a control value of the level or profile (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 levels or profiles have plateaued in response to treatment. Measured values of levels or profiles 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 may be indicated.
  • a control value of the level or profile e.g., a mean and standard deviation
  • a patient who is not presently receiving treatment but has undergone a previous course of treatment is monitored for antibody levels or profiles to determine whether a resumption of treatment is required.
  • the measured level or profile in the patient can be compared with a value 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.
  • 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.
  • 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.
  • 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 fluid or cerebrospinal fluid from the patient.
  • the sample is analyzed, for example, for levels or profiles of antibodies to A ⁇ peptide, e.g., levels or profiles of humanized antibodies.
  • ELISA methods of detecting antibodies specific to A ⁇ are described in the Examples section.
  • the level or profile of an administered antibody is determined using a clearing assay, for example, in an in vitro phagocytosis assay, as described herein.
  • a tissue sample from a patient being tested is contacted with amyloid deposits (e.g., from a PDAPP mouse) and phagocytic cells bearing Fc 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 A ⁇ in the tissue sample of the patient under test.
  • 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.
  • a baseline measurement of antibody to A ⁇ 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.
  • a predetermined percentage of the peak less baseline e.g., 50%, 25% or 10%
  • administration of a further dosage of antibody is administered.
  • 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.
  • Additional methods include monitoring, over the course of treatment, any art-recognized physiologic symptom (e.g., physical or mental symptom) routinely relied on by researchers or physicians to diagnose or monitor amyloidogenic diseases (e.g., Alzheimer's disease).
  • physiologic symptom e.g., physical or mental symptom
  • amyloidogenic diseases e.g., Alzheimer's disease
  • cognitive impairment can be monitored by determining a patient's score on the Mini-Mental State Exam in accordance with convention throughout the course of treatment.
  • kits for performing the monitoring methods described above contain an agent that specifically binds to antibodies to A ⁇ .
  • the kit can also include a label.
  • the label is typically in the form of labeled anti-idiotypic 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 A ⁇ .
  • 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.
  • the term labeling encompasses advertising leaflets and brochures, packaging materials, instructions, audio or videocassettes, computer discs, as well as writing imprinted directly on kits.
  • kits for example, research, detection and/or diagnostic kits (e.g., for performing in vivo imaging).
  • diagnostic kits typically contain an antibody for binding to an epitope of A ⁇ , preferably within residues 1-10.
  • the antibody is labeled or a secondary labeling reagent is included in the kit.
  • the kit is labeled with instructions for performing the intended application, for example, for performing an in vivo imaging assay. Exemplary antibodies are those described herein.
  • 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 Alzorer'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 A ⁇ , to the patient and then detecting the agent after it has bound.
  • a reagent such as antibody that binds to A ⁇
  • Preferred antibodies bind to A ⁇ deposits in a patient without binding to full length APP polypeptide.
  • Antibodies binding to an epitope of A ⁇ within amino acids 1-10 are particularly preferred.
  • the antibody binds to an epitope within amino acids 7-10 of A ⁇ .
  • Such antibodies typically bind without inducing a substantial clearing response.
  • the antibody binds to an epitope within amino acids 1-7 of A ⁇ .
  • Such antibodies typically bind and induce a clearing response to A ⁇ .
  • the clearing response can be avoided by using antibody fragments lacking a full-length constant region, such as Fabs.
  • the same antibody can serve as both a treatment and diagnostic reagent.
  • antibodies binding to epitopes C-terminal to residue 10 of A ⁇ do not show as strong a 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.
  • the reagent is labeled, although in some methods, the primary reagent with affinity for A ⁇ 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 labeled loci, to corresponding baseline values.
  • the base line values can represent the mean levels in a population of undiseased individuals. Baseline values can also represent previous levels determined in the same patient. For example, baseline values can be determined in a patient before beginning treatment, and measured values thereafter compared with the baseline values. A decrease in values relative to baseline signals a positive response to treatment.
  • This example tests the capacity of various monoclonal and polyclonal antibodies to A ⁇ to inhibit accumulation of A ⁇ in the brain of heterozygotic transgenic mice.
  • mice 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. Table 2 depicts the Experimental design.
  • the antibodies included four murine A ⁇ -specific monoclonal antibodies, 2H3 (directed to A ⁇ residues 1-12), 10D5 (directed to A ⁇ residues 3-7) (10D5 is produced by the cell line designated RB44-10D5.19.21, which was deposited on Apr. 8, 2003 and assigned ATCC accession number PTA-5129), 266 (directed to A ⁇ residues 13-28 and binds to soluble but not to aggregated AN1792), 21F12 (directed to A ⁇ residues 33-42).
  • a fifth group was treated with an A ⁇ -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 the the mice weighed 50 g). Antibody titers were monitored over the 28 weeks of treatment. Injections were administered intraperitoneally every seven days on average to maintain anti-A ⁇ 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.
  • antibody titers For determination 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 A 1-42-binding antibody using a sandwich ELISA with plastic multi-well plates coated with A ⁇ 1-42 as described in detail in the General Materials and Methods. Mean titers for each bleed are set forth in TABLE 3 for the polyclonal antibody and the monoclonals 10D5 and 21F 12.
  • Treatment was continued over a six-month period for a total of 196 days. Animals were euthanized one week after the final dose.
  • 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 A ⁇ and APP levels.
  • the hemisphere was dissected and homogenates of the hippocampal, cortical, and cerebellar region 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 A ⁇ peptide or APP of known concentrations in an ELISA format.
  • the levels of total A ⁇ and of A ⁇ 1-42 measured by ELISA in homogenates of the cortex, and the hippocampus and the level of total A ⁇ in the cerebellum are shown in Tables 4, 5, and 6, respectively.
  • the median concentration of total A ⁇ 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 level are similar to those previously reported for heterozygous PDAPP transgenic mice of this age (Johnson-Wood et al., supra).
  • one treatment group had a median A ⁇ level, measured as A ⁇ 1-42, which differed significantly from that of the control group (p ⁇ 0.05), those animals receiving the polyclonal anti-A ⁇ antibody as shown in Table 4.
  • the median level of A ⁇ 1-42 was reduced by 65%, compared to the control for this treatment group.
  • 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- ⁇ /FL, recognizes both APP-alpha ( ⁇ , the secreted form of APP which has been cleaved within the A ⁇ sequence), and full-length forms (FL) of APP, while the second recognizes only APP- ⁇ . In contrast to the treatment-associated diminution of A ⁇ 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 A ⁇ antibodies deplete A ⁇ without depleting APP.
  • a ⁇ levels were significantly reduced in the cortex, hippocampus and cerebellum in animals treated with the polyclonal antibody raised against AN 1792. To a lesser extent monoclonal antibodies to the amino terminal region of A ⁇ 1-42, specifically amino acids 1-16 and 13-28 also showed significant treatment effects.
  • amyloid plaques in the 10D5 group were also reduced in number and appearance, with some evidence of cell-associated A ⁇ immunoreactivity.
  • the polyclonal Ig fraction against A ⁇ 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 pAbA ⁇ 1-42 show diffuse deposits and absence of many of the larger compacted plaques in the pAbA ⁇ 1-42 treated group relative to control treated animals.
  • a ⁇ -dependent lymphoproliferation was measured using spleen cells harvested eight days following the final antibody infusion. Freshly harvested cells, 10 5 per well, were cultured for 5 days in the presence of A ⁇ 1-40 at a concentration of 5 ⁇ M 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-A ⁇ antibodies were stimulated in vitro with AN 1792 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-A ⁇ antibodies.
  • Anti-A ⁇ Antibodies mAb 2H3, mAb 10D5, mAb 266, mAb 21F12, mAb 3D6, mAb 16C11 and pAb ⁇ 1-42
  • the plaque burden was greatly reduced in these treatment groups compared to the 16C11 group.
  • 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.
  • 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).
  • the pellet was resuspended in growth medium (high glucose DMEM, 10%FBS, 25ng/ml 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 1000 rpm and resuspended in the assay medium.
  • growth medium high glucose DMEM, 10%FBS, 25ng/ml rmGM-CSF
  • microglial cells were then seeded at a density of 0.8 ⁇ 10 6 cells/ml assay medium.
  • the cultures were maintained in a humidified incubator (37° C., 5%CO 2 ) 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).
  • DAPI nuclear stain
  • 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).
  • 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 pg/ml of the pabA ⁇ 42 followed by an HRP-conjugated anti-mouse antibody, and developed with ECL (Amersham)
  • Table 7 compares A ⁇ binding versus phagocytosis for several different antibody binding specificities. It can be seen that antibodies binding to epitopes within aa 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 8 shows results obtained with several antibodies against A ⁇ , comparing their abilities to induce phagocytosis in the ex vivo assay and to reduce in vivo plaque burden in passive transfer studies.
  • 16C11 and 21F12 bound to aggregated synthetic A ⁇ peptide with high avidity, these antibodies were unable to react with ⁇ -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 A ⁇ were active by all three measures.
  • NAC synuclein
  • 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, and 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.
  • F(ab′)2 fragments of the anti-A ⁇ 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 A ⁇ occurs through Fc-receptor mediated phagocytosis.
  • This example determines the concentration of antibody delivered to the brain following intravenous injection into a peripheral tissue of either normal or PDAPP mice. Following treatment, 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. F(ab)′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 IgG1-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 indicating that the PDAPP does not have a uniquely leak blood brain barrier.
  • VH region leader peptide encompassing the translation initiation codon as the 5′ primer (DNA #3818-3829), and a g2b (DNA #3832) constant regions specific 3′ primer.
  • mice heavy variable RCT.CTT.CTG.C primer 6 MHV primer 6, MRC set; 3824 36 Yes ACT.AGT.CGA.CAT.GGR.ATG.GAG.CKG.GRT.
  • mouse heavy variable CTT.TMT.CTT primer 7 MHV primer 7, MRC set; 3825 33 Yes ACT.AGT.CGA.CAT.GAG.AGT.GCT.GAT.TCT.
  • 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 numbering convention of Kabat et al., supra.
  • variable heavy and light chain regions were re-engineered to encode splice donor sequences downstream of the respective VDJ or VJ junctions, and cloned into the mammalian expression vector pCMV-h ⁇ l for the heavy chain, and pCMV-h ⁇ l for the light chain. These vectors encode human ⁇ l and Ck constant regions as exonic fragments downstream of the inserted variable region cassette.
  • the heavy chain and light chain expression vectors were co-transfected into COS cells. Two different heavy chain clones (H2.2 & H3.2) were independently co-transfected with 3 different chimeric light chain clones (L3, L4, &L10) to confirm reproducibility of the result. A chimeric 21.6 antibody transfection was carried out as a positive control for the vectors. Conditioned media was collected 48 hrs post transfection and assayed by western blot analysis for antibody production or ELISA for A ⁇ binding.
  • the multiple transfectants all expressed heavy chain+light chain combinations which are recognized by a goat anti-human IgG (H+L) antibody on a western blot.
  • both 3D6 and PK1614 were effective at clearing A ⁇ plaques.
  • the ex vivo assay demonstrates that as the concentration of antibody increases, the amount of A ⁇ decreases in a similar manner for both murine and chimeric 3D6 antibodies. Hence, it can be concluded that the sequences encode functional 3D6 heavy chain and light chains respectively.
  • a first pass homology model of 3D6 variable region based on the antibodies noted above was constructed using the Look & SegMod Modules GeneMine (v3.5) software package. This software was purchased under a perpetual license from Molecular applications Group (Palo Alto, Calif.). This software package, authored by Drs. Michael Levitt and Chris Lee, facilitates the process of molecular modeling by automating the steps involved in structural modeling a primary sequence on a template of known structure based on sequence homology. Working on a Silicon Graphics IRIS workstation under a UNIX environment, the modeled structure is automatically refined by a series of energy minimization steps to relieve unfavorable atomic contacts and optimize electrostatic and van der Walls interactions.
  • a further refined model was built using the modeling capability of Quanta®.
  • a query of the PDB database with CDR3 of the heavy chain of 3D6 identified 1 qkz as most homologous and having the identical number of residues as 3D6.
  • CDR3 of the heavy chain of 3D6 was modeled using the crystal structure of 1 qkz as template.
  • the ⁇ -carbon backbone trace of the 3D6 model is shown in FIG. 4.
  • the VH domain is shown as a stippled line
  • VL domain is shown as a solid line
  • CDR loops are indicated in ribbon form.
  • Suitable human acceptor antibody sequences were identified by computer comparisons of the amino acid sequences of the mouse variable regions with the sequences of known human antibodies. The comparison was performed separately for the 3D6 heavy and light chains.
  • variable domains from human antibodies whose framework sequences exhibited a high degree of sequence identity with the murine VL and VH framework regions were identified by query of the Kabat Database using NCBI BLAST (publicly accessible through the National Institutes of Health NCBI internet server) with the respective murine framework sequences.
  • acceptor sequences Two candidate sequences were chosen as acceptor sequences based on the following criteria: (1) homology with the subject sequence; (2) sharing canonical CDR structures with the donor sequence; and (3) not containing any rare amino acid residues in the framework regions.
  • the selected acceptor sequence for VL is Kabat ID Number (KABID) 019230 (Genbank Accession No. S40342), and for VH is KABID 045919 (Genbank AccessionNo. AF115110).
  • First versions of humanized 3D6 antibody utilize these selected acceptor antibody sequences.
  • the humanized antibodies of the invention comprise variable framework regions substantially from a human immunoglobulin (acceptor immunoglobulin) and complementarity determining regions substantially from a mouse immunoglobulin (donor immunoglobulin) termed 3D6. Having identified the complementarity determining regions of 3D6 and appropriate human acceptor immunoglobulins, the next step was to determine which, if any, residues from these components to substitute to optimize the properties of the resulting humanized antibody. The criteria described supra were used to select residues for substitution.
  • FIGS. 1 and 2 depict alignments of the original murine 3D6 VL and VH, respectively, with the respective version 1 of the humanized sequence, the corresponding human framework acceptor sequence and, lastly, the human germline V region sequence showing highest homology to the human framework acceptor sequence.
  • the shaded residues indicate the canonical (solid fill), vernier (dotted outline), packing (bold), and rare amino acids (bold italics), and are indicated on the figure.
  • the asterisks indicate residues backmutated to murine residues in the human acceptor framework sequence, and CDR regions are shown overlined.
  • Table 12 A summary of the changes incorporated into version 1 of humanized 3D6 VH and VL is presented in Table 12.
  • Tables 13 and 14 set forth Kabat numbering keys for the various light and heavy chains, respectively. TABLE 13 Key to Kabat Numbering for Light Chain A19- mouse HUM KABID Germ- KAB # # TYPE 3D6VL 3D6VL 019230 line Comment 1 1 FR1 Y Y D D Rare mouse, may contact CDR 2 2 V V I I Canonical/CDR contact 3 3 V V V V 4 4 M M M 5 5 T T T T 6 6 Q Q Q Q 7 7 T S S S S 8 8 P P P P 9 9 L L L L 10 10 T S S S 11 11 L L L L 12 12 S P P P 13 13 V V V V 14 14 T T T 15 15 I P P P 16 16 G G G G G 17 17 Q E E E 18 18 P P P P P 19 19 A A A A 20 20 S S S S 21 21 I I I I I 22 22 S S S S S 23 23 C C C C 24 24 CDR1 K K R R 25 25 S S S S 26 26 S S S S 27 27 Q Q Q Q
  • the humanized antibodies preferably exhibit a specific binding affinity for A ⁇ of at least 10 7 , 10 8 , 10 9 or 10 10 M ⁇ 1 .
  • the upper limit of binding affinity of the humanized antibodies for A ⁇ is within a factor of three, four or five of that of 3D6 (i.e., ⁇ 10 9 M ⁇ 1 ).
  • the lower limit of binding affinity is also within a factor of three, four or five of that of 3D6.
  • the humanized light chain was assembled using PCR. DNA sequence analysis of greater than two dozen clones revealed scattered point mutations and deletions throughout the VL region with respect to the expected sequence. Analysis of the sequences indicated that clone 2.3 was amenable to repair of 2 closely spaced single nucleotide deletions in the amino-terminal region. Hence site directed mutagenesis was performed on clone pCRShum3D6vl2.3 using oligonucleotides to introduce the 2 deleted nucleotides, and repair of the point mutations was confirmed by DNA sequence analysis, and the VL insert was cloned into the light chain expression vector pCMV-cK.
  • Kabat numbering for the version 2 light and heavy chains is the same as that depicted in Tables 13 and 14, respectively, except that residue 1 of the version 2 light chain is asp (D), residue 74 of the heavy chain is ser (S), residue 77 of the heavy chain is thr (T) and residue 89 of the heavy chain is val (V).
  • the nucleotide sequence of humanized 3D6 version 1 light and heavy chains are set forth as SEQ ID NOs: 34 and 36, respectively.
  • the nucleotide sequence of humanized 3D6 version 2 light and heavy chain are set forth as SEQ ID NOs: 35 and 37, respectively.
  • Binding of humanized 3D6v1 to aggregated A ⁇ Functional testing of humanized 3D6v1 was conducted using conditioned media from transiently transfected COS cells. The cells were transfected with fully chimeric antibody, a mixture of either chimeric heavy chain+humanized light chain, or chimeric light chain+humanized heavy chain, and lastly, fully humanized antibody. The conditioned media was tested for binding to aggregated A ⁇ 1-42 by ELISA assay. The humanized antibody showed good activity within experimental error, and displayed binding properties indistinguishable from the chimeric 3D6 reference sample. The results are shown in Table 16.
  • rNET Replacement NET analysis of h3D6v2.
  • the rNET epitope map assay provides information about the contribution of individual residues within the epitope to the overall binding activity of the antibody.
  • rNET analysis uses synthesized systematic single substituted peptide analogs. Binding of an antibody being tested is determined against native peptide (native antigen) and against 19 alternative “single substituted” peptides, each peptide being substituted at a first position with one of 19 non-native amino acids for that position. A profile is generated reflecting the effect of substitution at that position with the various non-native residues. Profiles are likewise generated at successive positions along the antigenic peptide.
  • the combined profile, or epitope map (reflecting substitution at each position with all 19 non-native residues) can then be compared to a map similarly generated for a second antibody.
  • Substantially similar or identical maps indicate that antibodies being compared have the same or similar epitope specificity.
  • the profiles are virtually identical for 3D6 and h3D6v2 when one looks at the substitutions at each position (i.e., the values fluctuate in an identical manner when comparing the data in column 1 (3D6) versus column 2 (h3D6v2).
  • both h3D6v1 and h3D6v2 had about a 2 to 3 fold less binding affinity, measured at 2.06 nM and 2.24 nM for h3D6v1 and h3D6v2, respectively.
  • the ELISA competitive binding assay revealed an approximate 6-fold less binding affinity for h3D6v1 and h3D6v2.
  • humanized antibodies lose about 3-4 fold in binding affinity in comparison to their murine counterparts. Therefore, a loss of about 3 fold (average of ELISA and BlAcore results) for h3D6v1 and h3D6v2 is within the accepted range.
  • This Example describes the efficacy of murine mAb 3D6 on various neuropathological endpoints. A comparison is made between passive immunization with 3D6 (at varying doses) and active immunization with an A ⁇ peptide.
  • PDAPP mice were passively immunized with mAb 3D6 at three different doses, 10 mg/kg, 1 mg/kg and 10 mg/kg once a month (1 ⁇ 4).
  • An unrelated IgG ⁇ 2a antibody (TY 11/15) and PBS injections served as controls.
  • Active immunization with A ⁇ peptide served as a comparison. Between 20 and 35 animals were analyzed in each group.
  • the neuropathological endpoints assayed include amyloid burden and neuritic burden.
  • Neuritic burden following passive immunization with 3D6 was determined in PDAPP mice by immunostaining of brain sections with anti-APP antibody 8E5 followed by quantitative image analysis. Neuritic dystrophy is indicated by the appearance of dystrophic neurites (e.g., neurites with a globular appearance) located in the immediate vicinity of amyloid plaques. The results of this analysis are shown in Table 7. 3D6 (IgG ⁇ 2a isotype, recognizing A ⁇ 1-5) did not significantly reduce neuritic burden as compared to active immunization with A ⁇ peptide. Previously, it had been observed that 10D5 (IgG ⁇ 1 isotype recognizing A ⁇ 3-7) was unable to significantly reduce neuritic burden.
  • 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.
  • 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.
  • 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.
  • the anti-A ⁇ 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 pg of AN1792 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.
  • the anti-A ⁇ 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 CaCl 2 was then added with stirring to reach a final concentration of 64 mM. This solution was centrifuged at 10,000 ⁇ 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 ⁇ g and the supernatant was discarded.
  • the pellet was resuspended and dialyzed against 20 mM Tris-HCl, 0.4 M NaCl, 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 NaCl in 20 mM Tris-HCl, 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.
  • mice were bled by making a small nick in the tail vein and collecting about 200 ⁇ l 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 ⁇ 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.
  • RT room temperature
  • Antibody titers were measured by ELISA. 96-well microtiter plates (Costar EIA plates) were coated with 100 ⁇ pl of a solution containing either 10 ⁇ g/ml either A ⁇ 42 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 NaCl, 0.6% bovine serum albumin, 0.05% thimerosal).
  • Well Coating Buffer 0.1 M sodium phosphate, pH 8.5, 0.1% sodium azide
  • 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.
  • Tissues destined for ELISAs were homogenized in 10 volumes of ice-cold guanidine buffer (5.0 M guanidine-HCl, 50 mM Tris-HCl, pH 8.0). The homogenates were mixed by gentle agitation using an Adams Nutator (Fisher) for three to four hr at RT, then stored at ⁇ 20° C. prior to quantitation of A ⁇ and APP. Previous experiments had shown that the analytes were stable under this storage condition, and that synthetic A ⁇ protein (Bachem) could be quantitatively recovered when spiked into homogenates of control brain tissue from mouse littermates (Johnson-Wood et al., supra).
  • the brain homogenates were diluted 1:10 with ice cold Casein Diluent (0.25% casein, PBS, 0.05% sodium azide, 20 ⁇ g/ml aprotinin, 5 mM EDTA pH 8.0, 10 ⁇ g/ml leupeptin) and then centrifuged at 16,000 ⁇ g for 20 min at 4° C.
  • Casein Diluent 0.25% casein, PBS, 0.05% sodium azide, 20 ⁇ g/ml aprotinin, 5 mM EDTA pH 8.0, 10 ⁇ g/ml leupeptin
  • the synthetic A ⁇ 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.
  • BSA bovine serum albumin
  • the “total” A ⁇ sandwich ELISA utilizes monoclonal antibody monoclonal antibody 266, specific for amino acids 13-28 of A ⁇ (Seubert et al., supra), as the capture antibody, and biotinylated monoclonal antibody 3D6, specific for amino acids 1-5 of AD (Johnson-Wood et al., supra), as the reporter antibody.
  • the 3D6 monoclonal antibody does not recognize secreted APP or full-length APP, but detects only A ⁇ species with an amino-terminal aspartic acid.
  • the cell line producing the antibody 3D6 has the ATCC accession number PTA-5130, having been deposited on Apr. 8, 2003.
  • This assay has a lower limit of sensitivity of ⁇ 50 ng/ml (11 nM) and shows no cross-reactivity to the endogenous murine A ⁇ protein at concentrations up to 1 ng/ml (Johnson-Wood et al., supra).
  • the A ⁇ 1-42 specific sandwich ELISA employs mA ⁇ 21F12, specific for amino acids 33-42 of A ⁇ (Johnson-Wood, et al. supra), as the capture antibody.
  • Biotinylated mA ⁇ 3D6 is also the reporter antibody in this assay which has a lower limit of sensitivity of about 125 ⁇ g/ml (28 ⁇ M, Johnson-Wood et al., supra).
  • mA ⁇ 266 at 10 ⁇ g/ml
  • mA ⁇ 2 1F12 at 5 ⁇ g/ml
  • 100 ⁇ l of either mA ⁇ 266 (at 10 ⁇ g/ml) or mA ⁇ 2 1F12 at (5 ⁇ g/ml) was coated into the wells of 96-well immunoassay plates (Costar) by overnight incubation at RT.
  • the solution was removed by aspiration and the wells were blocked by the addition of 200 ⁇ l 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 NaCl, 0.01 M Tris-HCl, pH 7.5), plus 0.05% Tween 20] prior to use.
  • the samples and standards were added in triplicate aliquots of 100 ⁇ l 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 ⁇ g/ml 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, Calif.), 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 ⁇ l 2 N H2SO4.
  • the reaction product was quantified using a Molecular Devices Vmax measuring the difference in absorbance at 450 nm and 650 nm.
  • APP- ⁇ /FL recognizes both APP-alpha ( ⁇ ) and full-length (FL) forms of APP.
  • the second assay is specific for APP- ⁇ .
  • the APP- ⁇ /FL assay recognizes secreted APP including the first 12 amino acids of A ⁇ . Since the reporter antibody (2H3) is not specific to the ⁇ -clip-site, occurring between amino acids 612-613 of APP 695 (Esch et al., Science 248:1122-1124 (1990)); this assay also recognizes full length APP (APP-FL).
  • the reporter mAb for the APP- ⁇ /FL assay is mAb 2H3, specific for amino acids 597-608 of APP 695 (Johnson-Wood et al., supra) and the reporter antibody for the APP- ⁇ assay is a biotinylated derivative of mAb 16H9, raised to amino acids 605 to611 of APP.
  • the lower limit of sensitivity of the APP- ⁇ FL assay is about 11 ng/ml (150 ⁇ M) (Johnson-Wood et al.) and that of the APP- ⁇ specific assay is 22 ng/ml (0.3 nM).
  • mAb 8E5 was coated onto the wells of 96-well EIA plates as described above for mAb 266.
  • Purified, recombinant secreted APP- ⁇ was used as the reference standard for the APP- ⁇ assay and the APP- ⁇ /FL assay (Esch et al., supra).
  • the brain homogenate samples in 5 M guanidine were diluted 1:10 in ELISA Specimen Diluent (0.014 M phosphate buffer, pH 7.4, 0.6% bovine serum albumin, 0.05% thimerosal, 0.5 M NaCl, 0.1% NP40). They were then diluted 1:4 in Specimen Diluent containing 0.5 M guanidine.
  • the fluorescent substrate 4-methyl-umbellipheryl-phosphate was added for a 30-min RT incubation and the plates were read on a Cytofluor tm 2350 fluorimeter (Millipore) at 365 nm excitation and 450 nm emission.
  • 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% paraformaldehyde, PBS until sectioned. Forty-micron-thick coronal sections were cut on a vibratome at RT and stored in cryoprotectant (30% glycerol, 30% ethylene glycol in phosphate buffer) at ⁇ 20° C. prior to immunohistochemical processing.
  • a biotinylated anti-A ⁇ (mAb, 3D6, specific for human A ⁇ ) diluted to a concentration of 2 ⁇ g/ml in PBS and 1% horse serum; or (2) a biotinylated mAb specific for human APP, 8E5, diluted to a concentration of 3 ⁇ g/ml 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 CD11b, MAC-1 antigen, (Chemicon International) diluted 1:100 with 0.25% Triton X-l00 and 1% rabbit serum in
  • 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” 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, Calif.).
  • Sections were developed in 0.01% hydrogen peroxide, 0.05% 3,3′ ⁇ diaminobenzidine (DAB) at RT. Sections destined for incubation with the GFAP-,MAC-1- AND MHC II-specific antibodies were pretreated with 0.6% hydrogen peroxide at RT to block endogenous peroxidase then incubated overnight with the primary antibody at 4° C. Sections reacted with the GFAP antibody were incubated for 1 hr at RT with biotinylated anti-mouse IgG made in horse (Vector Laboratories; Vectastain Elite ABC Kit) diluted 1:200 with TBS.
  • DAB 3,3′ ⁇ diaminobenzidine
  • 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 slides), air dried overnight, dipped in Propar (Anatech) and overlaid with coverslips using Permount (Fisher) as the mounting medium.
  • a Videometric 150 Image Analysis System (Oncor, Inc., Gaithersburg, Md.) linked to a Nikon Microphot-FX microscope through a CCD video camera and a Sony Trinitron monitor was used for quantification of the immunoreactive slides.
  • the image of the section was stored in a video buffer and a color-and saturation-based threshold was determined to select and calculate the total pixel area occupied by the immunolabeled structures.
  • the hippocampus was manually outlined and the total pixel area occupied by the hippocampus was calculated.
  • the percent amyloid burden was measured as: (the fraction of the hippocampal area containing A ⁇ deposits immunoreactive with mAb 3D6) ⁇ 100.
  • the percent neuritic burden was measured as: (the fraction of the hippocampal area containing dystrophic neurites reactive with monoclonal antibody 8E5) ⁇ 100.
  • the C-Imaging System (Compix, Inc., Cranberry Township, Pa.) operating the Simple 32 Software application program was linked to a Nikon Microphot-FX microscope through an Optronics camera and used to quantitate the percentage of the retrospenial cortex occupied by GFAP-positive astrocytes and MAC-1-and MHC II-positive microglia.
  • the image of the immunoreacted section was stored in a video buffer and a monochrome-based threshold was determined to select and calculate the total pixel area occupied by immunolabeled cells.
  • the retrosplenial cortex 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.
  • percent microgliosis was defined as: (the fraction of the RSC occupied by MAC-1- or MHC II-reactive microglia) X 100.
  • the invention provides for a number of uses.
  • the invention provides for the use of any of the antibodies to A ⁇ 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.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Neurosurgery (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Toxicology (AREA)
  • Virology (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Mycology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US10/703,713 1997-12-02 2003-11-07 Humanized antibodies that recognize beta amyloid peptide Abandoned US20040171815A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US10/703,713 US20040171815A1 (en) 1997-12-02 2003-11-07 Humanized antibodies that recognize beta amyloid peptide
US11/516,724 US20080050367A1 (en) 1998-04-07 2006-09-05 Humanized antibodies that recognize beta amyloid peptide
US11/842,023 US7893214B2 (en) 1997-12-02 2007-08-20 Humanized antibodies that recognize beta amyloid peptide
US11/842,042 US9051363B2 (en) 1997-12-02 2007-08-20 Humanized antibodies that recognize beta amyloid peptide
US11/842,056 US20080281082A1 (en) 1997-12-02 2007-08-20 Humanized antibodies that recognize beta amyloid peptide

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US6774097P 1997-12-02 1997-12-02
US8097098P 1998-04-07 1998-04-07
US09/201,430 US6787523B1 (en) 1997-12-02 1998-11-30 Prevention and treatment of amyloidogenic disease
US09/322,289 US7964192B1 (en) 1997-12-02 1999-05-28 Prevention and treatment of amyloidgenic disease
US58001500A 2000-05-26 2000-05-26
US25189200P 2000-12-06 2000-12-06
US10/010,942 US7189819B2 (en) 2000-12-06 2001-12-06 Humanized antibodies that recognize beta amyloid peptide
US10/388,389 US7179892B2 (en) 2000-12-06 2003-03-12 Humanized antibodies that recognize beta amyloid peptide
US10/703,713 US20040171815A1 (en) 1997-12-02 2003-11-07 Humanized antibodies that recognize beta amyloid peptide

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US10/388,389 Continuation US7179892B2 (en) 1997-12-02 2003-03-12 Humanized antibodies that recognize beta amyloid peptide
US11/520,438 Continuation US7790856B2 (en) 1997-12-02 2006-09-12 Humanized antibodies that recognize beta amyloid peptide

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US09/201,430 Continuation US6787523B1 (en) 1997-12-02 1998-11-30 Prevention and treatment of amyloidogenic disease
US11/516,724 Continuation US20080050367A1 (en) 1997-12-02 2006-09-05 Humanized antibodies that recognize beta amyloid peptide
US11/516,724 Continuation-In-Part US20080050367A1 (en) 1997-12-02 2006-09-05 Humanized antibodies that recognize beta amyloid peptide

Publications (1)

Publication Number Publication Date
US20040171815A1 true US20040171815A1 (en) 2004-09-02

Family

ID=26748211

Family Applications (26)

Application Number Title Priority Date Filing Date
US10/703,713 Abandoned US20040171815A1 (en) 1997-12-02 2003-11-07 Humanized antibodies that recognize beta amyloid peptide
US10/704,070 Abandoned US20040171816A1 (en) 1997-12-02 2003-11-07 Humanized antibodies that recognize beta amyloid peptide
US10/789,273 Abandoned US20050249725A1 (en) 1997-12-02 2004-02-27 Humanized antibodies that recognize beta amyloid peptide
US10/816,380 Expired - Lifetime US7014855B2 (en) 1997-12-02 2004-03-31 Prevention and treatment of amyloidogenic disease
US10/815,353 Expired - Lifetime US6808712B2 (en) 1997-12-02 2004-03-31 Prevention and treatment of amyloidogenic disease
US10/815,404 Expired - Lifetime US6982084B2 (en) 1997-12-02 2004-03-31 Prevention and treatment of amyloidogenic disease
US10/923,474 Abandoned US20050048049A1 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/923,267 Abandoned US20050191292A1 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/923,605 Abandoned US20050249727A1 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/923,469 Expired - Fee Related US8034339B2 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/923,471 Expired - Fee Related US8535673B2 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/928,926 Abandoned US20050196399A1 (en) 1997-12-02 2004-08-27 Prevention and treatment of amyloidogenic disease
US10/934,609 Expired - Lifetime US6946135B2 (en) 1997-12-02 2004-09-02 Prevention and treatment of amyloidogenic disease
US10/934,819 Abandoned US20050163788A1 (en) 1997-12-02 2004-09-02 Prevention and treatment of amyloidogenic disease
US10/933,559 Expired - Lifetime US6972127B2 (en) 1997-12-02 2004-09-02 Prevention and treatment of amyloidogenic disease
US11/058,757 Abandoned US20050142132A1 (en) 1997-12-02 2005-02-14 Prevention and treatment of amyloidogenic disease
US11/108,102 Abandoned US20050191314A1 (en) 1997-12-02 2005-04-15 Prevention and treatment of amyloidogenic disease
US11/245,916 Abandoned US20060029611A1 (en) 1997-12-02 2005-10-07 Prevention and treatment of amyloidogenic disease
US11/245,524 Abandoned US20060034858A1 (en) 1997-12-02 2005-10-07 Prevention and treatment of amyloidogenic disease
US11/842,113 Expired - Fee Related US8034348B2 (en) 1997-12-02 2007-08-20 Prevention and treatment of amyloidogenic disease
US11/842,085 Abandoned US20080096818A1 (en) 1997-12-02 2007-08-20 Prevention and treatment of amyloidogenic disease
US11/842,116 Expired - Lifetime US8642044B2 (en) 1997-12-02 2007-08-20 Prevention and treatment of amyloidogenic disease
US11/842,120 Abandoned US20080227719A1 (en) 1997-12-02 2007-08-20 Prevention and treatment of amyloidogenic disease
US13/270,015 Abandoned US20130058869A1 (en) 1997-12-02 2011-10-10 Prevention And Treatment Of Amyloidogenic Disease
US13/271,081 Abandoned US20120276116A1 (en) 1997-12-02 2011-10-11 Prevention and Treatment of Amyloidogenic Disease
US14/017,177 Abandoned US20140227274A1 (en) 1997-12-02 2013-09-03 Prevention and Treatment of Amyloidogenic Disease

Family Applications After (25)

Application Number Title Priority Date Filing Date
US10/704,070 Abandoned US20040171816A1 (en) 1997-12-02 2003-11-07 Humanized antibodies that recognize beta amyloid peptide
US10/789,273 Abandoned US20050249725A1 (en) 1997-12-02 2004-02-27 Humanized antibodies that recognize beta amyloid peptide
US10/816,380 Expired - Lifetime US7014855B2 (en) 1997-12-02 2004-03-31 Prevention and treatment of amyloidogenic disease
US10/815,353 Expired - Lifetime US6808712B2 (en) 1997-12-02 2004-03-31 Prevention and treatment of amyloidogenic disease
US10/815,404 Expired - Lifetime US6982084B2 (en) 1997-12-02 2004-03-31 Prevention and treatment of amyloidogenic disease
US10/923,474 Abandoned US20050048049A1 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/923,267 Abandoned US20050191292A1 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/923,605 Abandoned US20050249727A1 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/923,469 Expired - Fee Related US8034339B2 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/923,471 Expired - Fee Related US8535673B2 (en) 1997-12-02 2004-08-20 Prevention and treatment of amyloidogenic disease
US10/928,926 Abandoned US20050196399A1 (en) 1997-12-02 2004-08-27 Prevention and treatment of amyloidogenic disease
US10/934,609 Expired - Lifetime US6946135B2 (en) 1997-12-02 2004-09-02 Prevention and treatment of amyloidogenic disease
US10/934,819 Abandoned US20050163788A1 (en) 1997-12-02 2004-09-02 Prevention and treatment of amyloidogenic disease
US10/933,559 Expired - Lifetime US6972127B2 (en) 1997-12-02 2004-09-02 Prevention and treatment of amyloidogenic disease
US11/058,757 Abandoned US20050142132A1 (en) 1997-12-02 2005-02-14 Prevention and treatment of amyloidogenic disease
US11/108,102 Abandoned US20050191314A1 (en) 1997-12-02 2005-04-15 Prevention and treatment of amyloidogenic disease
US11/245,916 Abandoned US20060029611A1 (en) 1997-12-02 2005-10-07 Prevention and treatment of amyloidogenic disease
US11/245,524 Abandoned US20060034858A1 (en) 1997-12-02 2005-10-07 Prevention and treatment of amyloidogenic disease
US11/842,113 Expired - Fee Related US8034348B2 (en) 1997-12-02 2007-08-20 Prevention and treatment of amyloidogenic disease
US11/842,085 Abandoned US20080096818A1 (en) 1997-12-02 2007-08-20 Prevention and treatment of amyloidogenic disease
US11/842,116 Expired - Lifetime US8642044B2 (en) 1997-12-02 2007-08-20 Prevention and treatment of amyloidogenic disease
US11/842,120 Abandoned US20080227719A1 (en) 1997-12-02 2007-08-20 Prevention and treatment of amyloidogenic disease
US13/270,015 Abandoned US20130058869A1 (en) 1997-12-02 2011-10-10 Prevention And Treatment Of Amyloidogenic Disease
US13/271,081 Abandoned US20120276116A1 (en) 1997-12-02 2011-10-11 Prevention and Treatment of Amyloidogenic Disease
US14/017,177 Abandoned US20140227274A1 (en) 1997-12-02 2013-09-03 Prevention and Treatment of Amyloidogenic Disease

Country Status (38)

Country Link
US (26) US20040171815A1 (fr)
EP (5) EP1679080B1 (fr)
JP (4) JP4677094B2 (fr)
KR (3) KR101248711B1 (fr)
CN (1) CN100374151C (fr)
AR (1) AR020050A1 (fr)
AT (4) ATE433760T1 (fr)
AU (1) AU1706199A (fr)
BG (1) BG104562A (fr)
BR (1) BR9815357A (fr)
CA (1) CA2312920C (fr)
CO (1) CO4980846A1 (fr)
CY (3) CY1108331T1 (fr)
CZ (1) CZ303137B6 (fr)
DE (7) DE06075704T1 (fr)
DK (4) DK1679080T3 (fr)
EA (3) EA200000608A1 (fr)
EE (1) EE05377B1 (fr)
ES (4) ES2428740T3 (fr)
GE (2) GEP20053715B (fr)
HK (2) HK1095265A1 (fr)
HR (2) HRP20000443B1 (fr)
HU (2) HU228493B1 (fr)
ID (1) ID25504A (fr)
IL (5) IL136252A0 (fr)
IS (1) IS5500A (fr)
MY (1) MY134906A (fr)
NO (3) NO328865B1 (fr)
NZ (1) NZ504569A (fr)
PE (1) PE20001383A1 (fr)
PL (2) PL202706B1 (fr)
PT (4) PT1994937E (fr)
SA (1) SA99191269B1 (fr)
SG (1) SG111083A1 (fr)
SI (4) SI2305282T1 (fr)
TR (1) TR200001608T2 (fr)
TW (1) TWI239847B (fr)
WO (1) WO1999027944A1 (fr)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040213800A1 (en) * 2003-02-01 2004-10-28 Seubert Peter A. Active immunization to generate antibodies to soluble A-beta
US20050118651A1 (en) * 2003-05-30 2005-06-02 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US20050158304A1 (en) * 2000-05-26 2005-07-21 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20060193850A1 (en) * 2005-01-28 2006-08-31 Warne Nicholas W Anti a beta antibody formulation
US20080181902A1 (en) * 2000-07-07 2008-07-31 Lars Lannfelt Prevention and treatment of alzheimer's disease
US20080292639A1 (en) * 2005-01-24 2008-11-27 Amgen Inc. Humanized Anti-Amyloid Antibody
US20090155246A1 (en) * 2004-06-21 2009-06-18 Bioartic Neuroscience Ab Antibodies Specific For Soluble Amyloid Beta Peptide Protofibrils and Uses Thereof
US20090155256A1 (en) * 2007-10-17 2009-06-18 Wyeth Immunotherapy Regimes Dependent On APOE Status
WO2009085200A2 (fr) 2007-12-21 2009-07-09 Amgen Inc. Anticorps anti-amyloïde et utilisations de ceux-ci
US20090258009A1 (en) * 2006-03-23 2009-10-15 Bioarctic Neuroscience Ab Protofibril selective antibodies and the use thereof
US20090285806A1 (en) * 2004-10-05 2009-11-19 Martin Sinacore Methods and compositions for improving recombinant protein production
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US20100167313A1 (en) * 2008-12-22 2010-07-01 Ulrich Essig Anti-idiotype antibody against an antibody against the amyloid beta peptide
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US20100322932A1 (en) * 1998-05-21 2010-12-23 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US7893214B2 (en) 1997-12-02 2011-02-22 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
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
US8034339B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
US8168188B1 (en) 2004-08-11 2012-05-01 Kyoto University Antibody and utilization of the same
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US9573994B2 (en) 2014-07-10 2017-02-21 Bioarctic Neuroscience Ab Aβ protofibril binding antibodies

Families Citing this family (268)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0866805A1 (fr) 1995-12-12 1998-09-30 Karolinska Innovations AB PEPTIDE FIXANT LA SEQUENCE KLVFF DE L'AMYLOIDE $g(b)
US8173127B2 (en) * 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US6703015B1 (en) 1999-09-03 2004-03-09 Ramot At Tel-Aviv University Ltd. Filamentous bacteriophage displaying an β-amyloid epitope
US6905686B1 (en) 1997-12-02 2005-06-14 Neuralab Limited Active immunization for treatment of alzheimer's disease
US7179892B2 (en) * 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US7588766B1 (en) * 2000-05-26 2009-09-15 Elan Pharma International Limited Treatment of amyloidogenic disease
US20050059802A1 (en) * 1998-04-07 2005-03-17 Neuralab Ltd Prevention and treatment of amyloidogenic disease
JP2002515235A (ja) * 1998-05-21 2002-05-28 ザ ユニヴァーシティ オブ テネシー リサーチ コーポレイション 坑アミロイド抗体を用いるアミロイドの除去方法
US7060670B1 (en) 1999-05-05 2006-06-13 Neurochem (International) Limited Stereoselective antifibrillogenic peptides and peptidomimetics thereof
TR200103266T2 (tr) * 1999-05-13 2002-01-21 American Cyanamid Company Yardımcı kombinasyon formülasyonları
AU2008203784B2 (en) * 1999-05-28 2012-01-19 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US6787637B1 (en) 1999-05-28 2004-09-07 Neuralab Limited N-Terminal amyloid-β antibodies
AU2005202644B2 (en) * 1999-06-01 2009-03-05 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
PE20010212A1 (es) * 1999-06-01 2001-02-22 Neuralab Ltd Composiciones del peptido a-beta y procesos para producir las mismas
AU780474B2 (en) 1999-06-16 2005-03-24 Boston Biomedical Research Institute Incorporated Immunological control of beta-amyloid levels in vivo
WO2001018169A2 (fr) * 1999-09-03 2001-03-15 Ramot University Authority For Applied Research & Industrial Development Ltd. Agents et compositions et procedes utilisant lesdits agents et lesdites compositions pour le diagnostic et/ou le traitement ou la prevention des maladies formant des plaques.
US7384640B1 (en) 1999-09-30 2008-06-10 Wyeth Holdings Corporation Mutant cholera holotoxin as an adjuvant
US20070135337A2 (en) * 1999-11-29 2007-06-14 Neurochem (International) Limited 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
JP4804690B2 (ja) 1999-12-08 2011-11-02 インテレクト・ニューロサイエンシズ・インコーポレーテッド 免疫原としてのキメラペプチド、それに対する抗体、およびキメラペプチドまたは抗体を用いた免疫法
EP1752472A3 (fr) * 1999-12-08 2007-04-25 Intellect Neurosciences, Inc. Peptides amyloides beta chimériques
PL204878B1 (pl) * 2000-02-21 2010-02-26 Lundbeck & Co As H Zastosowanie analogu autologicznego zwierzęcego Aβ albo polipeptydu APP lub sekwencji kodującej taki analog lub mikroorganizmu lub wirusa, który niesie nukleotydową sekwencję kodującą taki analog, analog polipeptydu amyloidogennego, immunogenna kompozycja, fragment kwasu nukleinowego kodujący analog, wektor, stransformowana komórka, kompozycja do indukowania produkcji przeciwciał i stabilna linia komórkowa
AU783144B2 (en) * 2000-02-21 2005-09-29 H. Lundbeck A/S Novel method for down-regulation of amyloid
BRPI0108676B8 (pt) 2000-02-24 2021-05-25 Lilly Co Eli anticorpos humanizados que sequestram peptídeo amilóide beta e seus usos no tratamento de condições caracterizadas por formação de placas amiloides, bem como composição farmacêutica que compreende os referidos anticorpos
WO2001077167A2 (fr) * 2000-04-05 2001-10-18 University Of Tennessee Research Corporation Techniques d'etude, de diagnostic et de traitement d'amylose
JP2003534351A (ja) 2000-05-22 2003-11-18 ニュー・ヨーク・ユニヴァーシティー アミロイドβ及びアミロイド沈着物に対する免疫応答誘導のための、アミロイドβと相同的な、合成の、免疫原性だが非アミロイド原性ペプチド
JP2003516929A (ja) * 2000-06-01 2003-05-20 ニユーララブ・リミテツド アミロイド形成性疾患の予防および治療
US7371920B2 (en) 2000-06-20 2008-05-13 The Governing Council Of The University Of Toronto Transgenic mouse model of neurodegenerative disorders
SI1292187T1 (sl) 2000-06-20 2006-10-31 Univ Toronto Transgenski zivalski model nevrodegenerativnih motenj
WO2002000885A2 (fr) 2000-06-23 2002-01-03 American Cyanamid Company Assemblage sauvage-chimerique de particules viraloïdes de la grippe (vlp)
ES2389811T3 (es) * 2000-06-28 2012-10-31 Prana Biotechnology Limited Oligómeros de amiloide beta para uso en el tratamiento, el alivio o la prevención de la enfermedad de Alzheimer
AU2007200047B2 (en) * 2000-07-07 2009-11-26 Bioarctic Neuroscience Ab Prevention and treatment of Alzheimer's disease
CA2421374C (fr) * 2000-09-06 2017-03-07 Aventis Pharma S.A. Procedes et compositions relatifs a des maladies associees a l'amyloide
AU1225702A (en) * 2000-09-19 2002-04-02 Evotec Neurosciences Gmbh Methods and compounds for treating brain amyloidosis
IL139308A0 (en) * 2000-10-26 2001-11-25 Marikovsky Moshe Peptides from amyloid precursor protein which inhibit tumor growth and metastasis
EE200300172A (et) 2000-11-10 2003-06-16 Wyeth Holdings Corporation Antigeenne kompositsioon ja adjuvantsegu
DK1346036T3 (da) 2000-11-29 2010-07-19 Univ Rochester Virusfri virusamplicon hjælpepartikler samt anvendelser deraf
TWI255272B (en) * 2000-12-06 2006-05-21 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
ES2359625T3 (es) 2000-12-28 2011-05-25 Wyeth Llc Proteina protectora recombinante de.
US7320793B2 (en) 2001-01-19 2008-01-22 Cytos Biotechnology Ag Molecular antigen array
US6815175B2 (en) 2001-03-16 2004-11-09 Cornell Research Foundation, Inc. Anti-amyloid peptide antibody based diagnosis and treatment of a neurological disease or disorder
GB0109297D0 (en) 2001-04-12 2001-05-30 Glaxosmithkline Biolog Sa Vaccine
ES2437875T3 (es) 2001-04-30 2014-01-14 Eli Lilly And Company Anticuerpos humanizados que reconocen el péptido beta-amiloide
US7320790B2 (en) 2001-04-30 2008-01-22 Eli Lilly And Company Humanized antibodies
US8092791B2 (en) 2001-05-23 2012-01-10 University Of Rochester Method of producing herpes simplex virus amplicons, resulting amplicons, and their use
US6906169B2 (en) 2001-05-25 2005-06-14 United Biomedical, Inc. Immunogenic peptide composition comprising measles virus Fprotein Thelper cell epitope (MUFThl-16) and N-terminus of β-amyloid peptide
IL159209A0 (en) 2001-06-07 2004-06-01 Wyeth Corp Mutant forms of cholera holotoxin as an adjuvant
CN1977971A (zh) 2001-06-07 2007-06-13 惠氏控股有限公司 作为佐剂的霍乱全毒素的突变体形式
WO2003006893A2 (fr) 2001-07-09 2003-01-23 Elan Pharmaceuticals, Inc. Procedes inhibant la toxicite amyloide
US7829087B2 (en) 2001-07-09 2010-11-09 Elan Pharmaceuticals, Inc. Methods of treating cognitive impairment
AU2002329775C1 (en) 2001-08-17 2011-04-07 Eli Lilly And Company Assay method for alzheimer's disease
EP1432444A4 (fr) * 2001-08-17 2005-11-02 Lilly Co Eli Anticorps anti-a beta
CA2452104A1 (fr) * 2001-08-17 2003-02-27 Eli Lilly And Company Utilisation d'anticorps ayant une forte affinite pour l'ass soluble dans le traitement de pathologies et de maladies liees a l'ass
AU2002324468A1 (en) * 2001-08-17 2003-03-03 Eli Lilly And Company Rapid improvement of cognition in conditions related to abeta
ATE381346T1 (de) * 2001-08-17 2008-01-15 Univ Washington Assayverfahren für alzheimer-krankheit
MY144532A (en) 2001-08-20 2011-09-30 Lundbeck & Co As H Novel method for down-regulation of amyloid
US20030082191A1 (en) * 2001-08-29 2003-05-01 Poduslo Joseph F. Treatment for central nervous system disorders
MX339524B (es) 2001-10-11 2016-05-30 Wyeth Corp Composiciones inmunogenicas novedosas para la prevencion y tratamiento de enfermedad meningococica.
US7479482B2 (en) 2001-11-21 2009-01-20 New York University Synthetic immunogenic but non-deposit-forming polypeptides and peptides homologous to amyloid β, prion protein, amylin, α-synuclein, or polyglutamine repeats for induction of an immune response thereto
US20040052928A1 (en) 2002-09-06 2004-03-18 Ehud Gazit Peptides and methods using same for diagnosing and treating amyloid-associated diseases
AR038568A1 (es) 2002-02-20 2005-01-19 Hoffmann La Roche Anticuerpos anti-a beta y su uso
US20040001848A1 (en) * 2002-03-01 2004-01-01 Szu-Yi Chou Method of producing disease-specific antigens
WO2003076455A2 (fr) * 2002-03-05 2003-09-18 Ramot At Tel-Aviv University Ltd. Composition immunisante et procede permettant d'induire une reponse immunitaire contre le site de clivage de $g(b)-secretase
EP1575529A4 (fr) * 2002-07-17 2007-08-08 Intellect Neurosciences Inc Peptides et procedes de criblage de vaccins a base de peptides immunogenes contre la maladie d'alzheimer
DK1524994T3 (da) 2002-07-19 2011-08-15 Cytos Biotechnology Ag Vaccinesammensætninger indeholdende amyloid beta 1-6-antigen-arrays
WO2004014296A2 (fr) * 2002-08-13 2004-02-19 U.S. Department Of Veterans Affairs Methode de detection et de prevention de la maladie d'alzheimer, en particulier a des stades prodromiques et precoces de la maladie
US7785608B2 (en) 2002-08-30 2010-08-31 Wyeth Holdings Corporation Immunogenic compositions for the prevention and treatment of meningococcal disease
CA2498407A1 (fr) * 2002-09-12 2004-03-25 The Regents Of The University Of California Immunogenes et anticorps correspondant specifiques pour des intermediaires d'agregation de poids moleculaire eleve communs a des amyloides constitues par des proteines de sequences differentes
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
EP1633786A4 (fr) * 2002-10-09 2007-07-25 Rinat Neuroscience Corp Methodes de traitement de la maladie d'alzheimer au moyen d'anticorps diriges contre le peptide g(b)-amyloide et compositions les comprenant
US9034337B2 (en) 2003-10-31 2015-05-19 Prothena Biosciences Limited Treatment and delay of outset of synucleinopathic and amyloidogenic disease
TW200509968A (en) 2002-11-01 2005-03-16 Elan Pharm Inc Prevention and treatment of synucleinopathic disease
US8697082B2 (en) 2002-11-01 2014-04-15 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US20080014194A1 (en) * 2003-10-31 2008-01-17 Elan Pharmaceuticals, Inc. Prevention and Treatment of Synucleinopathic and Amyloidogenic Disease
US8506959B2 (en) * 2002-11-01 2013-08-13 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
CN100450551C (zh) * 2002-11-29 2009-01-14 中国医学科学院基础医学研究所 用于治疗和预防阿尔茨海默病的重组腺相关病毒基因疫苗及其用途
WO2004056318A2 (fr) 2002-12-19 2004-07-08 New York University Methode permettant de traiter une maladie amyloide
DE10303974A1 (de) * 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid-β(1-42)-Oligomere, Verfahren zu deren Herstellung und deren Verwendung
US7575747B2 (en) 2003-02-10 2009-08-18 Applied Molecular Evolution Aβ binding molecules
CA2515603C (fr) * 2003-02-10 2016-06-07 To-Bbb Holding B.V. Acides nucleiques exprimes de facon differentielle dans la barriere hemato-encephalique dans des conditions inflammatoires
US8663650B2 (en) 2003-02-21 2014-03-04 Ac Immune Sa Methods and compositions comprising supramolecular constructs
US20060251675A1 (en) * 2003-03-17 2006-11-09 Michael Hagen Mutant cholera holotoxin as an adjuvant and an antigen carrier protein
KR100546066B1 (ko) * 2003-03-21 2006-01-26 한국생명공학연구원 베타아밀로이드 유전자의 다중 연결체를 발현하는 형질전환 식물 세포 및 이로부터 배양된 식물
WO2004087733A2 (fr) * 2003-03-28 2004-10-14 New York University Prevention et traitement du depot d'amyloide dans la maladie d'alzheimer
US7732162B2 (en) 2003-05-05 2010-06-08 Probiodrug Ag Inhibitors of glutaminyl cyclase for treating neurodegenerative diseases
ES2246178B1 (es) 2003-05-08 2007-03-01 Universidad De Zaragoza. Uso de anticuerpos para el tratamiento de enfermedades amiloideas.
US7358331B2 (en) * 2003-05-19 2008-04-15 Elan Pharmaceuticals, Inc. Truncated fragments of alpha-synuclein in Lewy body disease
JP4888876B2 (ja) * 2003-06-13 2012-02-29 田平 武 アルツハイマー病の治療のための組換えアデノ随伴ウィルスベクター
EP1638517A4 (fr) 2003-06-30 2010-01-06 Univ Tel Aviv Future Tech Dev Peptides, anticorps diriges contre les maladies associees a l'amyloide et procedes d'utilisation pour le diagnostic et le traitement de ces maladies
WO2005014041A2 (fr) * 2003-07-24 2005-02-17 Novartis Ag Substances et procedes pour le traitement des amyloses
US7807171B2 (en) 2003-07-25 2010-10-05 Ac Immune Sa Therapeutic vaccine targeted against P-glycoprotein 170 for inhibiting multidrug resistance in the treatment of cancers
GB0321615D0 (en) 2003-09-15 2003-10-15 Glaxo Group Ltd Improvements in vaccination
US20070264280A1 (en) * 2003-11-07 2007-11-15 Federoff Howard J Compositions and Methods for Treating Neurological Diseases
US7674599B2 (en) 2003-11-08 2010-03-09 Elan Pharmaceuticals, Inc. Methods of using antibodies to detect alpha-synuclein in fluid samples
SG149039A1 (en) 2003-12-17 2009-01-29 Elan Pharm Inc Ass IMMUNOGENIC PEPTIDE CARRIER CONJUGATES AND METHODS OF PRODUCING SAME
US20050225165A1 (en) * 2004-04-13 2005-10-13 Naik Sanjeev M Brake by-wire control system
WO2005102385A1 (fr) 2004-04-26 2005-11-03 Innate Pharma Composition adjuvante et methodes d'utilisation correspondantes
GB0410220D0 (en) 2004-05-07 2004-06-09 Kirkham Lea Ann Mutant pneumolysin proteins
CA2571035A1 (fr) * 2004-06-25 2006-01-12 Id Biomedical Corporation Of Quebec Compositions et procedes de traitement de troubles neurologiques
US7955812B2 (en) 2004-07-19 2011-06-07 The General Hospital Corporation Methods of diagnosing alzheimer's disease by detecting antibodies to cross-linked β-amyloid oligomers
EP2298807A3 (fr) * 2004-07-30 2011-05-18 Rinat Neuroscience Corp. Anticorps anti peptide amyloide beta, et leurs procedes d' utilisation
WO2006044666A2 (fr) * 2004-10-15 2006-04-27 Northeastern University Detection de maladie associee a la proteolyse
GB0424563D0 (en) 2004-11-05 2004-12-08 Novartis Ag Organic compounds
TW200635608A (en) * 2004-12-15 2006-10-16 Neuralab Ltd Aβ antibodies for use in improving cognition
AU2006211625A1 (en) * 2005-01-14 2006-08-10 The Regents Of The University Of California Compositions and methods for inhibiting drusen formation and for diagnosing or treating drusen-related disorders
CA2595380A1 (fr) * 2005-01-28 2006-08-03 Wyeth Formulations polypeptidiques liquides stabilisees
KR20080031167A (ko) * 2005-04-20 2008-04-08 디나벡크 가부시키가이샤 알츠하이머병 치료용의 고도로 안전한 비강투여용 유전자백신
CN101228272A (zh) * 2005-04-20 2008-07-23 生物载体株式会社 用于治疗阿尔茨海默病的具有较高安全性的经鼻腔内给药基因疫苗
UY29504A1 (es) 2005-04-29 2006-10-31 Rinat Neuroscience Corp Anticuerpos dirigidos contra el péptido amiloide beta y métodos que utilizan los mismos.
WO2006119352A2 (fr) * 2005-05-03 2006-11-09 University Of South Florida Procede pour traiter le declin cognitif et la perte synaptique lies a la maladie d'alzheimer
MX2007013825A (es) * 2005-05-05 2008-01-18 Merck & Co Inc Composiciones de un conjugado peptidico y metodos para la prevencion y tratamiento de la enfermedad de alzheimer.
JPWO2006126682A1 (ja) * 2005-05-27 2008-12-25 財団法人化学及血清療法研究所 アルツハイマー病の予防・治療用ワクチン
CN104926938B (zh) 2005-06-17 2019-06-04 惠氏有限责任公司 纯化含Fc区蛋白的方法
BRPI0613105A2 (pt) 2005-07-10 2010-12-28 Adaptive Spectrum & Signal método para configurar um primeiro sistema dsl, produto de programa de computador e controlador
WO2007056401A1 (fr) * 2005-11-09 2007-05-18 Merck & Co., Inc. Methode d'identification de modulateurs d'osbp convenant pour le traitement de la maladie d'alzheimer
US20070122413A1 (en) 2005-11-28 2007-05-31 Sivakumar Pallavur V Il-21 antagonists
WO2007114861A2 (fr) * 2005-11-28 2007-10-11 Zymogenetics, Inc. Antagonistes du recepteur de il-21
DK1976877T4 (en) 2005-11-30 2017-01-16 Abbvie Inc Monoclonal antibodies to amyloid beta protein and uses thereof
KR20080090408A (ko) 2005-11-30 2008-10-08 아보트 러보러터리즈 항-Aβ 글로불로머 항체, 이의 항원-결합 잔기, 상응하는하이브리도마, 핵산, 벡터, 숙주 세포, 당해 항체의 제조방법, 당해 항체를 포함하는 조성물, 당해 항체의 용도 및당해 항체의 사용 방법
WO2007067512A2 (fr) * 2005-12-08 2007-06-14 Merck & Co., Inc. Procede pour identifier des modulateurs d'adprh convenant pour traiter la maladie d'alzheimer
RU2551782C2 (ru) * 2005-12-12 2015-05-27 Ац Иммуне Са Специфические в отношении амилоида бета (а бета) 1-42 моноклональные антитела, обладающие терапевтическими свойствами
WO2007068429A1 (fr) 2005-12-12 2007-06-21 F. Hoffmann-La Roche Ag Anticorps anti-amyloïde bêta 4 avec asparagine glycosylée dans la région variable
EP1992639A4 (fr) * 2006-02-22 2009-08-26 Hayashibara Biochem Lab Vaccin peptidique destiné à induire la production d'anticorps anti-peptide amyloide
US7479550B2 (en) 2006-06-02 2009-01-20 The Board Of Regents Of The University Of Texas System Amyloid β gene vaccines
PL2046833T3 (pl) * 2006-07-14 2014-01-31 Ac Immune Sa Humanizowane przeciwciało przeciw amyloidowi beta
US20090123488A1 (en) * 2006-08-14 2009-05-14 Thymon, Llc Compositions and methods for the treatment and prophylaxis of Alzheimer's disease
JP2010506839A (ja) * 2006-10-12 2010-03-04 ワイス エルエルシー 乳光の低減を伴う方法および組成物
US8188046B2 (en) * 2006-10-16 2012-05-29 University Of South Florida Amyloid beta peptides and methods of use
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
AR064642A1 (es) 2006-12-22 2009-04-15 Wyeth Corp Polinucleotido vector que lo comprende celula recombinante que comprende el vector polipeptido , anticuerpo , composicion que comprende el polinucleotido , vector , celula recombinante polipeptido o anticuerpo , uso de la composicion y metodo para preparar la composicion misma y preparar una composi
US20080214987A1 (en) 2006-12-22 2008-09-04 Nanomed Devices, Inc. Microdevice And Method For Transdermal Delivery And Sampling Of Active Substances
SI2436696T1 (sl) 2007-01-05 2017-10-30 University Of Zurich Anti-beta-amiloidno protitelo in načini njegove uporabe
IL199534A (en) * 2007-01-05 2013-01-31 Univ Zuerich An isolated human antibody capable of detecting a neoepitope in a disease-related protein, a polynucleotide encoding an antibody, a vector containing the polynucleotide, a host cell containing the polynucleotide or vector, a preparation containing the antibody and related methods and uses.
US7939075B2 (en) 2007-01-11 2011-05-10 Philipps-Universitaet Marburg Human monoclonal anti-amyloid-beta antibodies
LT2842967T (lt) 2007-01-18 2017-02-27 Eli Lilly And Company Beta amiloido pegilintas fab
US8147833B2 (en) 2007-02-23 2012-04-03 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
EP2118300B1 (fr) 2007-02-23 2015-05-27 Prothena Biosciences Limited Prévention et traitement de maladies synucléinopathiques et amyloïdogéniques
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
WO2008106657A2 (fr) * 2007-03-01 2008-09-04 Intezyne Technologies, Inc. Peptides bêta-amyloïde encapsulés
CN101668525A (zh) 2007-03-01 2010-03-10 前体生物药物股份公司 谷氨酰胺酰环化酶抑制剂的新用途
RU2523894C2 (ru) * 2007-04-18 2014-07-27 Янссен Альцгеймер Иммунотерапи Предупреждение и лечение церебральной амилоидной ангиопатии
EP2142514B1 (fr) 2007-04-18 2014-12-24 Probiodrug AG Dérivés de thio-urée utilisés comme inhibiteurs de la glutaminyl cyclase
EP2012122A1 (fr) * 2007-07-06 2009-01-07 Medigene AG Protéines structurelles de parvovirus muté
JP2010528583A (ja) * 2007-06-11 2010-08-26 エーシー イミューン ソシエテ アノニム アミロイドβに対するヒト化抗体
KR20150055103A (ko) * 2007-06-12 2015-05-20 에이씨 이뮨 에스.에이. 아밀로이드 베타에 대해 인간화된 항체
US8613923B2 (en) 2007-06-12 2013-12-24 Ac Immune S.A. Monoclonal antibody
US8048420B2 (en) * 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
ES2466916T3 (es) 2007-06-27 2014-06-11 Admune Therapeutics Llc Complejos de IL-15 e IL-15R alfa y usos de los mismos
JP5731198B2 (ja) 2007-09-27 2015-06-10 イムノバクシーン・テクノロジーズ・インコーポレイテッドImmunovaccine Technologies Inc. インビボでのポリヌクレオチドの送達のための連続疎水相を含む担体におけるリポソームの使用
US9403902B2 (en) 2007-10-05 2016-08-02 Ac Immune S.A. Methods of treating ocular disease associated with amyloid-beta-related pathology using an anti-amyloid-beta antibody
NZ601858A (en) * 2007-10-05 2014-03-28 Genentech Inc Methods and compositions for diagnosis and treatment of amyloidosis
CA2701790A1 (fr) * 2007-10-05 2009-04-16 Ac Immune S.A. Utilisation d'anticorps anti-amyloides beta humanises dans les maladies oculaires
CN101878301B (zh) * 2007-10-29 2014-08-20 道健康生活医药株式会社 抗体及其应用
RU2504552C2 (ru) 2007-12-07 2014-01-20 Займодженетикс, Инк. Моноклональные антитела против il-21 человека
US9345753B2 (en) * 2008-01-16 2016-05-24 Yeda Research And Development Co. Ltd. At The Weizmann Institute Of Science Vaccine for alzheimer's disease
JP5715051B2 (ja) * 2008-06-05 2015-05-07 イムノバクシーン・テクノロジーズ・インコーポレイテッドImmunovaccine Technologies Inc. リポソーム、抗原、ポリヌクレオチド及び疎水性物質の連続相を含む担体を含む組成物
WO2010028246A2 (fr) * 2008-09-05 2010-03-11 Id Biomedical Corporation Of Quebec Nouvelles compositions et nouveaux adjuvants
AU2009295357A1 (en) * 2008-09-26 2010-04-01 The University Of Melbourne Alzheimer's disease biomarkers
WO2010050585A1 (fr) * 2008-10-31 2010-05-06 ディナベック株式会社 Vecteur pour le traitement de la maladie d’alzheimer
AU2009313615B2 (en) 2008-11-05 2012-11-29 Regents Of The University Of Minnesota Multicomponent immunogenic composition for the prevention of beta-hemolytic streptococcal (BHS) disease
JP5810413B2 (ja) 2008-12-19 2015-11-11 バイオジェン インターナショナル ニューロサイエンス ゲーエムベーハー ヒト抗アルファシヌクレイン自己抗体
US9925282B2 (en) 2009-01-29 2018-03-27 The General Hospital Corporation Cromolyn derivatives and related methods of imaging and treatment
EP3108949A1 (fr) 2009-03-02 2016-12-28 Dignity Health Dispositifs diagnostiques et procédés d'utilisation
FR2945538B1 (fr) 2009-05-12 2014-12-26 Sanofi Aventis Anticorps humanises specifiques de la forme protofibrillaire du peptide beta-amyloide.
EP3381937A3 (fr) 2009-08-13 2018-10-31 The Johns Hopkins University Procédés de modulation de la fonction immune
US9125846B2 (en) 2010-10-15 2015-09-08 The Board Of Regents Of The University Of Texas System Antibodies that bind amyloid oligomers
SG178953A1 (en) 2009-09-11 2012-04-27 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
WO2011102901A1 (fr) * 2010-02-20 2011-08-25 Ebiotec (Euroespes Biotechnology) Prévention et traitement de la maladie d'alzheimer par le peptide bêta amyloïde et le sphingosine-1-phosphate
TW201144437A (en) 2010-03-03 2011-12-16 Boehringer Ingelheim Int A-beta binding polypeptides
ES2586231T3 (es) 2010-03-03 2016-10-13 Probiodrug Ag Inhibidores de glutaminil ciclasa
EP3056510B1 (fr) * 2010-03-03 2018-10-03 The University Of British Columbia Épitope de bêta amyloïde spécifique à des oligomères et anticorps
DK2545047T3 (da) 2010-03-10 2014-07-28 Probiodrug Ag Heterocycliske inhibitorer af glutaminylcyclase (QC, EC 2.3.2.5)
JP2013523182A (ja) 2010-04-15 2013-06-17 アボット・ラボラトリーズ アミロイドベータ結合タンパク質
WO2011131748A2 (fr) 2010-04-21 2011-10-27 Probiodrug Ag Nouveaux inhibiteurs
WO2011151076A2 (fr) 2010-06-04 2011-12-08 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts, Universitätsmedizin Anticorps monoclonaux ciblant des oligomères aβ
JP6081356B2 (ja) 2010-07-30 2017-02-15 エーシー イミューン エス.エー. 安全で機能的なヒト化抗βアミロイド抗体
US9289488B2 (en) 2010-08-12 2016-03-22 Ac Immune Sa Vaccine engineering
CN105348387B (zh) 2010-08-14 2020-08-25 Abbvie 公司 β淀粉样蛋白结合蛋白
ES2850973T3 (es) 2010-08-23 2021-09-01 Wyeth Llc Formulaciones estables de antígenos rLP2086 de Neisseria meningitidis
WO2012032489A1 (fr) 2010-09-10 2012-03-15 Wyeth Llc Variants non-lipidiques des antigènes orf 2086 de neisseria meningitidis
WO2012055933A1 (fr) 2010-10-26 2012-05-03 Ac Immune S.A. Préparation d'un produit de recombinaison antigénique
EP2640737B1 (fr) 2010-11-15 2018-08-29 Ramot at Tel-Aviv University Ltd. Analogues dipeptidiques pour le traitement d'états pathologiques associé à la formation de fibrilles amyloïdes
ES2570167T3 (es) 2011-03-16 2016-05-17 Probiodrug Ag Derivados de benzimidazol como inhibidores de glutaminil ciclasa
RU2473133C2 (ru) * 2011-03-30 2013-01-20 Государственное образовательное учреждение высшего профессионального образования "Северо-Осетинская государственная медицинская академия" Министерства здравоохранения и социального развития РФ Способ профилактики системного амилоидоза и его нефропатической формы у экспериментальных животных
WO2012131539A1 (fr) 2011-03-31 2012-10-04 Pfizer Inc. Nouvelles pyridones bicycliques
WO2012163289A1 (fr) * 2011-06-01 2012-12-06 厦门大学 Protéine de fusion comprenant le mutant non toxique de la toxine diphtérique crm197 ou un fragment de celui-ci
WO2012172449A1 (fr) 2011-06-13 2012-12-20 Pfizer Inc. Lactames convenant comme inhibiteurs des bêta-sécrétases
WO2012177972A1 (fr) 2011-06-23 2012-12-27 Biogen Idec International Neuroscience Gmbh Molécules de liaison anti-alpha-synucléine
US9926353B2 (en) 2011-07-19 2018-03-27 New York University Immunotherapeutic modulation of amyloidogenic disease using non-fibrillogenic, non-amyloidogenic polymerized proteins and peptides
US8906382B2 (en) 2011-07-19 2014-12-09 New York University Method for treating amyloid disease
CN102895659B (zh) * 2011-07-29 2014-10-29 复旦大学 一种防治老年痴呆症复合疫苗及其制备方法
US20150065449A1 (en) 2011-08-12 2015-03-05 Florida State University Research Foundation, Inc. Treating Amyloidoses With A Vitamin B12 Composition Including Melatonin, Resveratrol, and EGCG
US20150065448A1 (en) * 2011-08-12 2015-03-05 The Florida State University Research Foundation Inc. Methods of treating amyloidoses with vitamin b12 and diagnostic test for detecting the presence of amyloid-beta peptides
JP6043355B2 (ja) 2011-08-31 2016-12-14 ファイザー・インク ヘキサヒドロピラノ[3,4−d][1,3]チアジン−2−アミン化合物
PL2579042T3 (pl) 2011-10-04 2014-12-31 Affiris Ag Sposób wykrywania przeciwciał swoistych wobec Aß w próbce biologicznej
CA2853100A1 (fr) * 2011-10-24 2013-05-02 Intellect Neurosciences, Inc. Compositions et methodes de traitement de proteinopathies
JP2012102131A (ja) * 2012-01-05 2012-05-31 Elan Pharma Internatl Ltd アミロイド形成性疾患の予防および治療
NZ628449A (en) 2012-03-09 2016-04-29 Pfizer Neisseria meningitidis compositions and methods thereof
SA115360586B1 (ar) 2012-03-09 2017-04-12 فايزر انك تركيبات لعلاج الالتهاب السحائي البكتيري وطرق لتحضيرها
EP2852597B1 (fr) 2012-05-04 2016-06-08 Pfizer Inc Composés d'hexahydropyrano[3,4-d][1,3]thiazin-2-amine substitués par des hétérocycliques à titre d'inhibiteurs d'app, bace1 et bace2
CN105732639A (zh) 2012-06-29 2016-07-06 辉瑞大药厂 作为LRRK2抑制剂的4-(取代的氨基)-7H-吡咯并〔2,3-d〕嘧啶类
JP2015529239A (ja) 2012-09-20 2015-10-05 ファイザー・インク アルキル置換ヘキサヒドロピラノ[3,4−d][1,3]チアジン−2−アミン化合物
UA110688C2 (uk) 2012-09-21 2016-01-25 Пфайзер Інк. Біциклічні піридинони
JP6499077B2 (ja) 2012-10-25 2019-04-10 ザ ジェネラル ホスピタル コーポレイション アルツハイマー病と関連疾患の治療のための併用療法
US10058530B2 (en) 2012-10-25 2018-08-28 The General Hospital Corporation Combination therapies for the treatment of Alzheimer's disease and related disorders
US9414752B2 (en) 2012-11-09 2016-08-16 Elwha Llc Embolism deflector
WO2014091352A1 (fr) 2012-12-11 2014-06-19 Pfizer Inc. Composés d'hexahydropyrano [3,4-d][1,3]thiazin-2-amine en tant qu'inhibiteurs de bace1
US9403846B2 (en) 2012-12-19 2016-08-02 Pfizer Inc. Carbocyclic- and heterocyclic-substituted hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
AU2014203873A1 (en) 2013-01-07 2015-07-30 Biomedical Research Models, Inc. Therapeutic vaccines for treating herpes simplex virus type 2 infections
CA2897678A1 (fr) 2013-02-13 2014-08-21 Pfizer Inc. Composes hexahydropyrano [3,4-d][1,3] thiazin-2-amine substitues par un heteroaryle
US9233981B1 (en) 2013-02-15 2016-01-12 Pfizer Inc. Substituted phenyl hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
MX2015010714A (es) 2013-02-19 2016-06-14 Pfizer Compuestos de azabencimidazol.
JP6446377B2 (ja) 2013-03-08 2018-12-26 ファイザー・インク 免疫原性融合ポリペプチド
WO2014159614A1 (fr) * 2013-03-14 2014-10-02 Flow Pharma, Inc. Formulation de particules à base d'adjuvants et d'antigènes
US9102752B2 (en) 2013-03-15 2015-08-11 United Biomedical, Inc. Peptide vaccine for prevention and immunotherapy of dementia of the Alzheimer's type
EP2787347A1 (fr) 2013-04-03 2014-10-08 Affiris AG Procédé pour la détection d'anticorps Aß-spécifiques dans un échantillon biologique
US10525005B2 (en) 2013-05-23 2020-01-07 The General Hospital Corporation Cromolyn compositions and methods thereof
WO2015017280A1 (fr) 2013-07-28 2015-02-05 Qantu Therapeutics, Inc. Formulation de vaccins induisant une réponse immunitaire de type th2
CA2923129C (fr) 2013-09-08 2020-06-09 Pfizer Inc. Compositions utilisables contre neisseria meningitidis et procedes associes
CA2925743C (fr) 2013-10-04 2018-03-06 Pfizer Inc. Nouvelles pyridinones bicycliques utilisees comme modulateurs de gamma-secretase
CN110305095A (zh) 2013-10-22 2019-10-08 综合医院公司 色甘酸衍生物以及成像和治疗的相关方法
EP3083618B1 (fr) 2013-12-17 2018-02-21 Pfizer Inc Nouvelles 1h-pyrrolo[2,3- b]pyridines 3,4-disubstituées et 7h-pyrrolo[2,3-c]pyridazines 4,5-disubstituées en tant qu'inhibiteurs de la lrrk2
MD20160102A2 (ro) 2014-04-01 2017-04-30 Pfizer Inc. Cromen şi 1,1a,2,7b-tetrahidrociclopropa[c]cromen piridopirazindione ca modulatori ai gama-secretazei
AP2016009493A0 (en) 2014-04-10 2016-10-31 Pfizer 2-AMINO-6-METHYL-4,4a,5,6-TETRAHYDROPYRANO[3,4-d][1,3]THIAZIN-8a(8H)-YL-1,3-THIAZOL-4-YL AMIDES
CA2946931A1 (fr) * 2014-04-29 2015-11-05 Affiris Ag Traitement et prevention de la maladie d'alzheimer
CA2946929A1 (fr) * 2014-04-29 2015-11-05 Affiris Ag Traitement et prevention de la maladie d'alzheimer
WO2015165961A1 (fr) 2014-04-29 2015-11-05 Affiris Ag Traitement et prévention de la maladie d'alzheimer
WO2015165971A1 (fr) * 2014-04-29 2015-11-05 Affiris Ag Traitement et prévention de la maladie d'alzheimer
ES2824763T3 (es) * 2014-04-29 2021-05-13 Frank Mattner Tratamiento y a la prevención de la enfermedad de Alzheimer (AD)
CA2943588C (fr) * 2014-06-17 2019-04-30 Academia Sinica Anticorps humanises anti-ige qui reticulent cd23 sur des lymphocytes b mais qui ne sensibilisent pas les mastocytes
EP3172210B1 (fr) 2014-07-24 2020-01-15 Pfizer Inc Composés de pyrazolopyrimidine
KR102061952B1 (ko) 2014-08-06 2020-01-02 화이자 인코포레이티드 이미다조피리다진 화합물
MA41115A (fr) 2014-12-02 2017-10-10 Biogen Int Neuroscience Gmbh Procédé de traitement de la maladie d'alzheimer
PE20171318A1 (es) 2015-02-03 2017-09-07 Pfizer Ciclopropabenzofuranil-piridopirazindionas novedosas
BR112017017460A2 (pt) 2015-02-19 2018-04-10 Pfizer Inc. composições de neisseria meningitidis e métodos das mesmas
WO2016203347A1 (fr) 2015-06-17 2016-12-22 Pfizer Inc. Composés tricycliques et leur utilisation en tant qu'inhibiteurs de la phosphodiesterase
LT3313879T (lt) 2015-06-24 2022-03-25 F. Hoffmann-La Roche Ag Antikūnai prieš transferino receptorių su pritaikytu giminingumu
JP6873980B2 (ja) 2015-09-14 2021-05-19 ファイザー・インク LRRK2阻害薬としての新規のイミダゾ[4,5−c]キノリンおよびイミダゾ[4,5−c][1,5]ナフチリジン誘導体
JP2018531924A (ja) 2015-09-24 2018-11-01 ファイザー・インク テトラヒドロピラノ[3,4−d][1,3]オキサジン誘導体、およびbace阻害剤としてのその使用
WO2017051276A1 (fr) 2015-09-24 2017-03-30 Pfizer Inc. N-[2-(2-amino-6,6-disubstitués-4,4a,5,6-tétrahydropyrano[3,4-d][1,3]thiazin-8a(8h)-yl)-1,3-thiazol-4-yl]amides
WO2017051294A1 (fr) 2015-09-24 2017-03-30 Pfizer Inc. N-[2-(3-amino-2,5-diméthyl-1,1-dioxido-5,6-dihydro-2h-1,2,4-thiadiazin-5-yl)-1,3-thiazol-4-yl] amides utiles comme inhibiteurs de bace
AR106189A1 (es) 2015-10-02 2017-12-20 Hoffmann La Roche ANTICUERPOS BIESPECÍFICOS CONTRA EL A-b HUMANO Y EL RECEPTOR DE TRANSFERRINA HUMANO Y MÉTODOS DE USO
TWI819458B (zh) 2015-10-02 2023-10-21 瑞士商赫孚孟拉羅股份公司 雙特異性抗‐人類cd20/人類轉鐵蛋白受體抗體及使用方法
WO2017079835A1 (fr) * 2015-11-09 2017-05-18 The University Of British Columbia Épitopes de la bêta-amyloïde et anticorps associés
KR20180085736A (ko) 2015-11-09 2018-07-27 더 유니버시티 오브 브리티쉬 콜롬비아 아밀로이드 베타 중간-영역 내 에피토프 및 이에 대해 구조적으로 선택성인 항체
CN108350051A (zh) 2015-11-09 2018-07-31 英属哥伦比亚大学 淀粉样蛋白β中的N-末端表位及其构象选择性抗体
AU2017223132B2 (en) 2016-02-23 2019-12-05 Pfizer Inc. 6,7-Dihydro-5H-pyrazolo[5,1-b][1,3]oxazine-2-carboxamide compounds
CN109641898B (zh) 2016-07-01 2022-04-08 辉瑞公司 用于治疗神经和神经退行性疾病的5,7-二氢-吡咯并-吡啶衍生物
JP2019524865A (ja) 2016-08-31 2019-09-05 ザ ジェネラル ホスピタル コーポレイション 神経変性疾患と関連する神経炎症におけるマクロファージ/ミクログリア
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
CN110234658B (zh) 2017-01-31 2024-03-12 辉瑞大药厂 脑膜炎奈瑟菌组合物及其使用方法
CA3056030A1 (fr) 2017-03-10 2018-09-13 Pfizer Inc. Nouveaux derives imidazo[4,5-c]quinoleine utilises en tant qu'inhibiteurs de lrrk2
IL269215B (en) 2017-03-10 2022-09-01 Pfizer Disubstituted imidazole[4,5-c]quinoline derivatives
RU2678987C2 (ru) 2017-04-28 2019-02-05 Общество с ограниченной ответственностью "Научно-производственная фирма ВЕРТА" Пептид для лечения болезни альцгеймера
WO2018226992A1 (fr) 2017-06-07 2018-12-13 Adrx, Inc. Inhibiteur d'agrégation de tau
AU2018287787A1 (en) 2017-06-22 2020-01-16 Pfizer Inc. Dihydro-pyrrolo-pyridine derivatives
US11209638B2 (en) * 2017-07-05 2021-12-28 West Virginia University Systems and methods for supporting and positioning body tissue samples in a microscope
AU2017423862A1 (en) 2017-07-20 2020-02-06 Aztherapies, Inc. Powdered formulations of cromolyn sodium and ibuprofen
EP3668886A2 (fr) 2017-08-18 2020-06-24 Adrx, Inc. Inhibiteurs peptidiques d'agrégation de tau
SG11202001281WA (en) 2017-08-22 2020-03-30 Biogen Ma Inc Pharmaceutical compositions containing anti-beta amyloid antibodies
PL3461819T3 (pl) 2017-09-29 2020-11-30 Probiodrug Ag Inhibitory cyklazy glutaminylowej
HUE061533T2 (hu) 2018-03-23 2023-07-28 Pfizer Piperazin azaspiro származékok
US12018069B2 (en) 2018-06-28 2024-06-25 The Trustees Of Columbia University In The City Of New York Methods and compositions for imaging amyloid deposits
JP2021529771A (ja) 2018-07-02 2021-11-04 ザ ジェネラル ホスピタル コーポレイション クロモリンナトリウムおよびα−ラクトースの粉末製剤
JP2023526500A (ja) 2020-05-19 2023-06-21 オター プロシーナ リミテッド アルツハイマー病の処置のためのマルチエピトープワクチン

Citations (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4666829A (en) * 1985-05-15 1987-05-19 University Of California Polypeptide marker for Alzheimer's disease and its use for diagnosis
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4816397A (en) * 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
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
US5004697A (en) * 1987-08-17 1991-04-02 Univ. Of Ca Cationized antibodies for delivery through the blood-brain barrier
US5096706A (en) * 1986-03-25 1992-03-17 National Research Development Corporation Antigen-based treatment for adiposity
US5192753A (en) * 1991-04-23 1993-03-09 Mcgeer Patrick L Anti-rheumatoid arthritic drugs in the treatment of dementia
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
US5220013A (en) * 1986-11-17 1993-06-15 Scios Nova Inc. DNA sequence useful for the detection of Alzheimer's disease
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5231000A (en) * 1987-10-08 1993-07-27 The Mclean Hospital Antibodies to A4 amyloid peptide
US5231170A (en) * 1986-08-27 1993-07-27 Paul Averback Antibodies to dense microspheres
US5278049A (en) * 1986-06-03 1994-01-11 Incyte Pharmaceuticals, Inc. Recombinant molecule encoding human protease nexin
US5387742A (en) * 1990-06-15 1995-02-07 Scios Nova Inc. Transgenic mice displaying the amyloid-forming pathology of alzheimer's disease
US5434170A (en) * 1993-12-23 1995-07-18 Andrulis Pharmaceuticals Corp. Method for treating neurocognitive disorders
US5441870A (en) * 1992-04-15 1995-08-15 Athena Neurosciences, Inc. Methods for monitoring cellular processing of β-amyloid precursor protein
US5514548A (en) * 1993-02-17 1996-05-07 Morphosys Gesellschaft Fur Proteinoptimerung Mbh Method for in vivo selection of ligand-binding proteins
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5593846A (en) * 1992-07-10 1997-01-14 Athena Neurosciences Methods for the detection of soluble β-amyloid peptide
US5605811A (en) * 1992-10-26 1997-02-25 Athena Neurosciences, Inc. Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
US5612486A (en) * 1993-10-27 1997-03-18 Athena Neurosciences, Inc. Transgenic animals harboring APP allele having swedish mutation
US5618920A (en) * 1985-11-01 1997-04-08 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
US5622701A (en) * 1994-06-14 1997-04-22 Protein Design Labs, Inc. Cross-reacting monoclonal antibodies specific for E- and P-selectin
US5624821A (en) * 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5641474A (en) * 1987-06-24 1997-06-24 Autoimmune, Inc. Prevention of autoimmune diseases by aerosol administration of autoantigens
US5641473A (en) * 1987-06-24 1997-06-24 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
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
US5733547A (en) * 1987-06-24 1998-03-31 Autoimmune, Inc. Treatment of autoimmune arthritis by oral administration of type I or type III collagen
US5736142A (en) * 1993-09-14 1998-04-07 Cytel Corporation Alteration of immune response using pan DR-binding peptides
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)
US5750361A (en) * 1995-11-02 1998-05-12 The Regents Of The University Of California Formation and use of prion protein (PRP) complexes
US5750349A (en) * 1993-01-25 1998-05-12 Takeda Chemical Industries Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5753624A (en) * 1990-04-27 1998-05-19 Milkhaus Laboratory, Inc. Materials and methods for treatment of plaquing disease
US5776468A (en) * 1993-03-23 1998-07-07 Smithkline Beecham Biologicals (S.A.) Vaccine compositions containing 3-0 deacylated monophosphoryl lipid A
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
US5786180A (en) * 1995-02-14 1998-07-28 Bayer Corporation Monoclonal antibody 369.2B specific for β A4 peptide
US5859205A (en) * 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US5869046A (en) * 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5869054A (en) * 1987-06-24 1999-02-09 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of autoantigens
US5877399A (en) * 1994-01-27 1999-03-02 Johns Hopkins University Transgenic mice expressing APP-Swedish mutation develop progressive neurologic disease
US5891991A (en) * 1992-04-20 1999-04-06 The General Hospital Corporation Amyloid precursor-like protein and uses thereof
US5935927A (en) * 1994-02-03 1999-08-10 The Picower Institute For Medical Research Compositions and methods for stimulating amyloid removal in amyloidogenic diseases using advanced glycosylation endproducts
US5955079A (en) * 1992-02-11 1999-09-21 Henry Jackson Foundation For The Advancement Of Military Medicine Dual carrier immunogenic construct
US5955317A (en) * 1993-01-25 1999-09-21 Takeda Chemical Industries, Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5958883A (en) * 1992-09-23 1999-09-28 Board Of Regents Of The University Of Washington Office Of Technology Animal models of human amyloidoses
US6022859A (en) * 1996-11-15 2000-02-08 Wisconsin Alumni Research Foundation Inhibitors of β-amyloid toxicity
US6054297A (en) * 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
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
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)
US6194551B1 (en) * 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6218506B1 (en) * 1997-02-05 2001-04-17 Northwestern University Amyloid β protein (globular assembly and uses thereof)
US6261569B1 (en) * 1992-08-27 2001-07-17 Deakin Research Limited Retro-, inverso- and retro-inverso synthetic peptide analogues
US6262335B1 (en) * 1994-01-27 2001-07-17 Johns Hopkins University Transgenic mice expressing APP mutant at amino acids 717, 721 and 722
US6277375B1 (en) * 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US20010018053A1 (en) * 1999-09-14 2001-08-30 Mcmichael John Methods for treating disease states comprising administration of low levels of antibodies
US20020009445A1 (en) * 2000-07-12 2002-01-24 Yansheng Du Human beta-amyloid antibody and use thereof for treatment of alzheimer's disease
US20020058267A1 (en) * 1997-04-16 2002-05-16 American Home Products Corporation Beta-amyloid peptide-binding proteins and polynucleotides encoding the same
US6399314B1 (en) * 1999-12-29 2002-06-04 American Cyanamid Company Methods of detection of amyloidogenic proteins
US6407213B1 (en) * 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
US20020077288A1 (en) * 2000-05-22 2002-06-20 New York University Synthetic immunogenic but non-amyloidogenic peptides homologous to amyloid beta for induction of an immune response to amyloid beta and amyloid deposits
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
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
US20020094335A1 (en) * 1999-11-29 2002-07-18 Robert Chalifour Vaccine for the prevention and treatment of alzheimer's and amyloid related diseases
US20020102261A1 (en) * 1999-06-16 2002-08-01 Boston Biomedical Research Institute Immunological control of beta-amyloid levels in vivo
US6528624B1 (en) * 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US20030068325A1 (en) * 2001-05-25 2003-04-10 Wang Chang Yi Immunogenic peptide composition for the prevention and treatment of Altzheimers Disease
US20030068316A1 (en) * 1997-02-05 2003-04-10 Klein William L. Anti-ADDL antibodies and uses thereof
US6548640B1 (en) * 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US20030073655A1 (en) * 1997-04-09 2003-04-17 Chain Daniel G. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US6562341B2 (en) * 1995-09-14 2003-05-13 The Regents Of The University Of California Antibodies specific for native PrPSc
US20030147882A1 (en) * 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US20040043418A1 (en) * 2000-02-24 2004-03-04 Holtzman David M. Humanized antibodies that sequester Abeta peptide
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
US6743427B1 (en) * 1997-12-02 2004-06-01 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6750324B1 (en) * 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US6761888B1 (en) * 2000-05-26 2004-07-13 Neuralab Limited Passive immunization treatment of Alzheimer's disease
US20050009150A1 (en) * 1998-11-30 2005-01-13 Elan Pharmaceuticals, Inc. Humanized antibodies that recognize beta amyloid peptide
US6866849B2 (en) * 1997-12-02 2005-03-15 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050059591A1 (en) * 1998-04-07 2005-03-17 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050059802A1 (en) * 1998-04-07 2005-03-17 Neuralab Ltd Prevention and treatment of amyloidogenic disease
US6875434B1 (en) * 1997-12-02 2005-04-05 Neuralab Limited Methods of treatment of Alzheimer's disease
US20050123534A1 (en) * 1989-12-21 2005-06-09 Celltech R&D Limited Humanised antibodies
US20050163788A1 (en) * 1997-12-02 2005-07-28 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6933368B2 (en) * 1992-03-09 2005-08-23 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation of immunoglobulin variable region
US6936698B2 (en) * 1998-04-28 2005-08-30 Smithkline Beecham Monoclonal antibodies with reduced immunogenicity

Family Cites Families (348)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US589566A (en) * 1897-09-07 meevten
US6096318A (en) 1973-05-07 2000-08-01 The Ohio State University Antigenically modified HCG polypeptides
JPS57212347A (en) * 1981-06-25 1982-12-27 Nissan Motor Co Ltd Air-fuel ratio control system
US4902506A (en) 1983-07-05 1990-02-20 The University Of Rochester Immunogenic conjugates
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
IT1190383B (it) * 1985-08-01 1988-02-16 Eniricerche Spa Sostanze peptidominetiche ad azione ipotensiva
DE3689123T2 (de) 1985-11-01 1994-03-03 Xoma Corp Modulare einheit von antikörpergenen, daraus hergestellte antikörper und verwendung.
US4713366A (en) * 1985-12-04 1987-12-15 The Ohio State University Research Foundation Antigenic modification of polypeptides
US5223482A (en) 1986-11-17 1993-06-29 Scios Nova Inc. Recombinant Alzheimer's protease inhibitory amyloid protein and method of use
US5187153A (en) 1986-11-17 1993-02-16 Scios Nova Inc. Methods of treatment using Alzheimer's amyloid polypeptide derivatives
US4879213A (en) 1986-12-05 1989-11-07 Scripps Clinic And Research Foundation Synthetic polypeptides and antibodies related to Epstein-Barr virus early antigen-diffuse
US4818397A (en) * 1986-12-22 1989-04-04 Sundstrand Corporation Shut-off valve seal
DE3702789A1 (de) 1987-01-30 1988-08-18 Bayer Ag Vorlaeuferprotein des apc-polypeptids, dafuer codierende dna und diagnostische verwendung der dna und des proteins
US4883666A (en) 1987-04-29 1989-11-28 Massachusetts Institute Of Technology Controlled drug delivery system for treatment of neural disorders
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
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
JP2512796B2 (ja) 1987-06-24 1996-07-03 オートイミュン・インコーポレイテッド 自己抗原の経口投与による自己免疫疾患の治療法
US5571500A (en) * 1987-06-24 1996-11-05 Autoimmune, Inc. Treatment of autoimmune diseases through administration by inhalation of autoantigens
US5571499A (en) * 1987-06-24 1996-11-05 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
US4912208A (en) * 1987-06-29 1990-03-27 Abbott Laboratories Fluorophores for encapsulation into liposomes
US4966753A (en) 1987-08-18 1990-10-30 Molecular Rx, Inc. Immunotherapeutic methods and compositions employing antigens characteristic of malignant neoplasms
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
CA1339014C (fr) 1987-10-08 1997-03-25 Ronald E. Majocha Anticorps pour peptide a4-amyloide
AU2728588A (en) 1987-10-23 1989-05-23 Genetics Institute Inc. Composition and method for treating cancers characterized by over-expression of the c-fms proto-oncogene
US5089603A (en) 1989-06-21 1992-02-18 Tanox Biosystems, Inc. Antigenic epitopes present on membrane-bound but not secreted iga
WO1989006689A1 (fr) 1988-01-13 1989-07-27 The Mclean Hospital Corporation Constructions genetiques renfermant le gene amyloide cerebral d'alzheimer
US4912094B1 (en) 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
US5576184A (en) * 1988-09-06 1996-11-19 Xoma Corporation Production of chimeric mouse-human antibodies with specificity to human tumor antigens
US5098706A (en) * 1988-11-01 1992-03-24 The Regents Of The University Of California Juvenile hormone esterase for insect control
EP0442926A1 (fr) 1988-11-10 1991-08-28 Imperial Cancer Research Technology Limited Polypeptides
IL162181A (en) * 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5227159A (en) * 1989-01-31 1993-07-13 Miller Richard A Anti-idiotype antibodies reactive with shared idiotopes expressed by B cell lymphomas and autoantibodies
US5262332A (en) 1989-04-05 1993-11-16 Brigham And Women's Hospital Diagnostic method for Alzheimer's disease: examination of non-neural tissue
AU5525090A (en) 1989-04-14 1990-11-16 Research Foundation For Mental Hygiene, Inc. Monoclonal antibody to amyloid peptide
AU5439790A (en) 1989-04-14 1990-11-16 Research Foundation For Mental Hygiene, Inc. Cerebrovascular amyloid protein-specific monoclonal antibody sv17-6e10
CA2017507C (fr) * 1989-05-25 1996-11-12 Gary Van Nest Adjuvant constitue d'une emulsion de gouttelettes submicron d'huile
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
BR9007950A (pt) 1989-12-20 1992-10-27 Brigham & Womens Hospital Metodo para tratamento ou prevencao da manifestacao de uma doenca auto-imune em um mamifero,formulacao farmaceutica,metodo para tratamento de uma doenca auto-imune,e metodo para tratamento de um mamifero acometido de doenca auto-imune
AU633698B2 (en) 1990-01-12 1993-02-04 Amgen Fremont Inc. Generation of xenogeneic antibodies
ATE157257T1 (de) 1990-03-02 1997-09-15 Autoimmune Inc Verstärkung der unterdrückungsregulation von autoimmunkrankheiten durch orale oder enterale verabreichung von autoantigenen
GB9005705D0 (en) 1990-03-14 1990-05-09 Health Lab Service Board Particle manipulation
US5279833A (en) 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
EP0451700A1 (fr) 1990-04-10 1991-10-16 Miles Inc. Minigènes APP recombinants pour l'expression dans des souris transgéniques comme modèles de la maladie d'Alzheimer
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
EP0527823A1 (fr) 1990-04-24 1993-02-24 The Regents Of The University Of California Purification, detection et procedes d'utilisation de protease nexine-2
EP0526511B1 (fr) * 1990-04-27 1997-05-28 MCMICHAEL, John Procede et composition servant a traiter des etats pathologiques du systeme nerveux central associes a la proteine beta amyloide anormale
GB9009548D0 (en) 1990-04-27 1990-06-20 Celltech Ltd Chimeric antibody and method
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
AU8081491A (en) 1990-06-01 1991-12-31 Cetus Corporation Compositions and methods for identifying biologically active molecules
US5914109A (en) * 1990-06-15 1999-06-22 New York University Heterohybridomas producing human monoclonal antibodies to HIV-1
EP0537247A4 (en) 1990-06-19 1993-09-08 Immuvax Nonpathogenic variant virus
GB9014932D0 (en) 1990-07-05 1990-08-22 Celltech Ltd Recombinant dna product and method
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
KR970005049B1 (ko) * 1990-07-16 1997-04-11 더 리젠츠 오브 더 유니벌시티 오브 캘리포니아 바쿨로비루스 발현 시스템, 이 벡터 시스템을 포함하는 이종성 무척추동물 숙주세포, 상기 시스템에 의해 생산되는 가용성의 정제된 야생형 망막아세포종 폴리펩티드 및 이 폴리펩티드의 생산 방법
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)
JPH06502071A (ja) 1990-09-28 1994-03-10 ジ・アップジョン・カンパニー アルツハイマーのアミロイド前駆遺伝子を有するトランスジェニック動物
ES2129029T5 (es) * 1990-10-05 2005-10-16 Celldex Therapeutics, Inc. Inmunoestimulacion dirigida con reactivos biespecificos.
JP2635444B2 (ja) 1990-10-15 1997-07-30 オートイミューン インク 自己抗体の経口投与による自己免疫性疾患の治療
GB9023352D0 (en) 1990-10-26 1990-12-05 Lynxvale Ltd Vaccinia vectors,vaccinia genes and expression products thereof
JP3510244B2 (ja) 1991-01-21 2004-03-22 エラン ファーマシューティカルス,インコーポレイテッド アルツハイマー病に関するテストおよびモデル
DE69231232T2 (de) * 1991-03-01 2001-01-04 Merial Lyon Verfahren zur Immunoneutralisierung von nichtkastrierten männlichen Haustieren gegen LMRH und Peptide dafür
DE4107857A1 (de) * 1991-03-12 1992-09-17 Thomae Gmbh Dr K Cyclische harnstoffderivate, diese verbindungen enthaltende arzneimittel und verfahren zu ihrer herstellung
DE69229703T2 (de) 1991-05-08 2000-04-27 Schweiz Serum & Impfinst Wiederhergestellte Influenza-Virosomen mit immunostimulierenden und immunoverstärkenden Eigenschaften und diese enthaltende Impfstoffe
US5672805A (en) 1991-07-18 1997-09-30 The Regents Of The University Of California Transgenic mice expressing the neurotoxic C-terminus of β-amyloid precursor protein
US5434050A (en) 1991-08-13 1995-07-18 Regents Of The University Of Minnesota Labelled β-amyloid peptide and methods of screening for Alzheimer's disease
US5283185A (en) 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
CA2118806A1 (fr) 1991-09-18 1993-04-01 William J. Dower Methode pour la synthese de diverses series d'oligomeres
US5837268A (en) 1991-10-16 1998-11-17 University Of Saskatchewan GnRH-leukotoxin chimeras
EP0746609A4 (fr) 1991-12-17 1997-12-17 Genpharm Int Animaux transgeniques non humains capables de produire des anticorps heterologues
ATE243746T1 (de) 1992-01-07 2003-07-15 Elan Pharm Inc Transgene tiermodelle fur alzheimer-krankheit
US5679348A (en) * 1992-02-03 1997-10-21 Cedars-Sinai Medical Center Immunotherapy for recurrent HSV infections
US5314813A (en) 1992-02-19 1994-05-24 Scripps Research Institute Drosophila cell lines expressing genes encoding MHC class I antigens and B2-microglobulin and capable of assembling empty complexes and methods of making said cell lines
KR950700082A (ko) 1992-02-28 1995-01-16 로버트 씨. 비숍 바이스탠더(bystander)에 의한 자가면역질환을 처치하는 방법
EP0561087B1 (fr) 1992-03-20 1999-08-04 N.V. Innogenetics S.A. Forme mutée du gène de la protéine du précurseur beta-amyloide
AU4116793A (en) 1992-04-24 1993-11-29 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
GB9209118D0 (en) 1992-04-28 1992-06-10 Sb 120 Amsterdam Bv Vaccine compositions
JP3428646B2 (ja) * 1992-06-18 2003-07-22 プレジデント アンド フェローズ オブ ハーバードカレッジ ジフテリア毒素ワクチン
ATE188613T1 (de) * 1992-06-25 2000-01-15 Smithkline Beecham Biolog Adjuvantien enthaltende impfstoffzusammensetzung
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
US5837672A (en) 1992-07-10 1998-11-17 Athena Neurosciences, Inc. Methods and compositions for the detection of soluble β-amyloid peptide
WO1994001772A1 (fr) 1992-07-13 1994-01-20 The Children's Medical Center Corporation CRIBLAGE DE TRAITEMENTS DE LA MALADIE D'ALZHEIMER SUR LA BASE DE LA PRODUCTION DE β-AMYLOIDE
IL102687A (en) 1992-07-30 1997-06-10 Yeda Res & Dev Conjugates of poorly immunogenic antigens and synthetic pepide carriers and vaccines comprising them
ES2127829T3 (es) 1992-07-31 1999-05-01 Medeva Holdings Bv Expresion de proteinas recombinantes fusionadas en bacterias atenuadas.
FI96267C (sv) 1992-09-16 1996-06-10 Formit Foodprocessing Ab Oy Vals och maskin för skalning eller formning av potatis och liknande produkter
DE69311048T2 (de) * 1992-09-29 1997-12-11 Nippon Telegraph & Telephone Multi/Demultiplexer mit Gitter aus gruppierten Wellenleitern und zurückgefürten optischen Wegen
IL107166A (en) 1992-10-01 2000-10-31 Univ Columbia Complex combinatorial chemical libraries encoded with tags
WO1994009364A1 (fr) 1992-10-13 1994-04-28 Duke University Procede permettant d'inhiber la liaison de la proteine precurseur d'amyloide a la proteine beta-amyloide
EP1298436B1 (fr) 1992-10-26 2010-07-14 Elan Pharmaceuticals, Inc. Procédés de l' identification des composés inhibiteurs de la libération du peptide beta-amyloide (BAP)
US5972336A (en) * 1992-11-03 1999-10-26 Oravax Merieux Co. Urease-based vaccine against helicobacter infection
US5733647A (en) * 1992-11-05 1998-03-31 Polymer Innovations, Inc. Insole
US6210671B1 (en) * 1992-12-01 2001-04-03 Protein Design Labs, Inc. Humanized antibodies reactive with L-selectin
US5674703A (en) 1992-12-02 1997-10-07 Woo; Savio L. C. Episomal vector systems and related methods
AU685776B2 (en) * 1993-01-22 1998-01-29 Sloan-Kettering Institute For Cancer Research Ganglioside-KLH conjugate vaccines with QS-21
US5750106A (en) * 1993-01-28 1998-05-12 Novartis Ag Human monoclonal antibodies to cytomegalovirus
US5358708A (en) 1993-01-29 1994-10-25 Schering Corporation Stabilization of protein formulations
US5989565A (en) * 1993-01-29 1999-11-23 University Of Pittsburgh Elution and identification of T cell epitopes from viable cells
US5472693A (en) * 1993-02-16 1995-12-05 The Dow Chemical Company Family of anti-carcinoembryonic antigen chimeric antibodies
CA2115900A1 (fr) 1993-02-22 1994-08-23 Gerald W. Becker Cribles pharmaceutiques et anticorps
JPH08508252A (ja) 1993-03-17 1996-09-03 アメリカ合衆国 小胞体シグナル配列ペプチドと少なくとも1個の他のペプチドをエンコードする核酸配列を含有する免疫原性キメラ、及びこのキメラのワクチン及び疾患の治療における使用
CA2158475C (fr) * 1993-03-18 2009-06-02 Lawrence Tamarkin Composition et methode pour la reduction de facteurs biologiquement actifs
CA2119090A1 (fr) * 1993-03-26 1994-09-27 Wayne R. Gombotz Compositions pour la liberation controlee de tgf-.beta. biologiquement actif
IT1270939B (it) 1993-05-11 1997-05-26 Angeletti P Ist Richerche Bio Procedimento per la preparazione di immunogeni e reagenti diagnostici,e immunogeni e reagenti diagnostici cosi' ottenibili.
US5723130A (en) 1993-05-25 1998-03-03 Hancock; Gerald E. Adjuvants for vaccines against respiratory syncytial virus
AU7043894A (en) 1993-05-28 1994-12-20 Miriam Hospital, The Composition and method for (in vivo) imaging of amyloid deposits
EP0700445B1 (fr) 1993-06-04 2002-01-23 Whitehead Institute For Biomedical Research Proteines du stress et leurs utilisations
US5464823A (en) * 1993-07-20 1995-11-07 The Regents Of The University Of California Mammalian antibiotic peptides
JPH09500540A (ja) 1993-07-30 1997-01-21 メデヴァ ホールディングス ビー.ヴイ. ワクチン組成物
WO1995005393A2 (fr) 1993-08-18 1995-02-23 Morphosys Gesellschaft Für Proteinoptimierung Mbh Peptides liant et neturalisant les lipopolysaccharides
AU707083B2 (en) 1993-08-26 1999-07-01 Bavarian Nordic Inc. Inducing antibody response against self-proteins with the aid of foreign T-cell epitopes
DK96493D0 (da) 1993-08-26 1993-08-26 Mouritsen Og Elsner Aps Fremgangsmaade til at inducere antistofresponser mod selvproteiner og autovaccine fremstillet ved fremgangsmaaden
AU705889B2 (en) 1993-08-26 1999-06-03 Regents Of The University Of California, The Method, compositions and devices for administration of naked polynucleotides which encode antigens and immunostimulatory peptides
FR2709247B1 (fr) * 1993-08-27 1995-09-29 Martin Jean Raymond Dispositif d'ancrage d'une instrumentation rachidienne sur une vertèbre.
WO1995006407A1 (fr) 1993-08-30 1995-03-09 The Regents Of The University Of California Nouveau composant amyloide lie a la maladie d'alzheimer et procedes d'utilisation
ATE286510T1 (de) 1993-09-07 2005-01-15 Smithkline Beecham Corp In der behandlung von il4 auslösenden krankheiten nützliche rekombinante il4 antikörper
US5385887A (en) * 1993-09-10 1995-01-31 Genetics Institute, Inc. Formulations for delivery of osteogenic proteins
US5415584A (en) 1993-09-21 1995-05-16 Tomco2 Equipment Company Particle blast cleaning apparatus
US5470951A (en) * 1993-09-29 1995-11-28 City Of Hope Peptides for antagonizing the effects of amyloid βprotein
US5858981A (en) * 1993-09-30 1999-01-12 University Of Pennsylvania Method of inhibiting phagocytosis
DE69429925T2 (de) 1993-10-20 2002-10-02 Univ Durham METHODE ZUR BINDUNG VON MATERIAL AN DAS Beta-AMYLOID-PEPTID
ES2182850T3 (es) 1993-10-22 2003-03-16 Genentech Inc Procedimientos y composiciones para la microencapsulacion de adyuvantes.
BR9407947A (pt) 1993-11-02 1996-11-26 Affymax Tech Nv Processo para sintetizar moléculas diversas em uma pluralidade de substratos e uma coleçao molecular etiquetade para realizaçao reaçoes de copulaçao em contas em paralelo para triar uma coleçao molecular etiquetada para detectar a presença de um ou mais diferentes etiquetas e para determinar a sequência de síntese aparelho para reaçoes de copulaçao paralelas em suportes sólidos bloco de alinhamento ótico para uso com um detector ótico sistemas de distribuiçao para distribuir reagentes dispositiv
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
AUPM411994A0 (en) 1994-02-25 1994-03-24 Deakin Research Limited Epitopes
ES2129809T3 (es) 1994-02-28 1999-06-16 Sumitomo Chemical Co Composicion para resina poliolefinica y pelicula de resina.
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 (fr) 1994-05-06 1995-11-16 John J. Baldwin Bibliotheque combinatoire de dihydrobenzopyranes
CA2191101C (fr) * 1994-05-25 2001-08-07 Ellis L. Kline Compositions et procedes de traitement de maladies a formation de plaque
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
EP0769963A4 (fr) 1994-07-27 1999-07-28 Commw Scient Ind Res Org Vaccins a base de polyepitopes
CA2200097A1 (fr) 1994-09-16 1996-03-21 Cancer Research Fund Of Contra Costa Peptides recombinants derives de l'anticorps mc3 dirige contre l'antigene ba46, leurs procedes d'utilisation et procedes d'humanisation d'anticorps peptidiques
DE69508521T2 (de) * 1994-10-14 1999-10-07 Oriental Sangyo Co Ltd Elektrolytische Vorrichtung zur Herstellung von Kohlenstoff-Dioxyd
NL9401735A (nl) * 1994-10-19 1996-06-03 Crown Gear Bv Tandwieloverbrenging van een cilindrisch rondsel met een kroonwiel, het kroonwiel dat toegepast wordt in deze tandwieloverbrenging, een werkwijze volgens welke het kroonwiel gemaakt kan worden alsmede een gereedschap waarmee het kroonwiel gemaakt kan worden.
US5872005A (en) * 1994-11-03 1999-02-16 Cell Genesys Inc. Packaging cell lines for adeno-associated viral vectors
JPH08137927A (ja) * 1994-11-07 1996-05-31 Hitachi Ltd 部品配置配線表示方法
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
DE69620877T2 (de) 1995-02-06 2002-12-12 Genetics Inst Arzneimittelformulierungen für il-12
US5624937A (en) * 1995-03-02 1997-04-29 Eli Lilly And Company Chemical compounds as inhibitors of amyloid beta protein production
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
US5854215A (en) 1995-03-14 1998-12-29 Praecis Pharmaceuticals Incorporated Modulators of β-amyloid peptide aggregation
EP0815134B1 (fr) 1995-03-14 2002-06-05 Praecis Pharmaceuticals Incorporated Modulateurs de l'agregation de substances amyloides
WO1996029421A1 (fr) * 1995-03-23 1996-09-26 Cantab Pharmaceuticals Research Limited Vecteurs d'apport de genes
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
UA56132C2 (uk) 1995-04-25 2003-05-15 Смітклайн Бічем Байолоджікалс С.А. Композиція вакцини (варіанти), спосіб стабілізації qs21 відносно гідролізу (варіанти), спосіб приготування композиції вакцини
CA2222055A1 (fr) 1995-05-23 1996-11-28 Morphosys Gesellschaft Fur Proteinoptimierung Mbh Proteines multimeriques
AU6113896A (en) 1995-06-05 1996-12-24 Brigham And Women's Hospital Use of oral tolerance to suppress both th1 and th2 immune re sponses and to suppress antibody production
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
EP0832205A1 (fr) 1995-06-07 1998-04-01 Athena Neurosciences, Inc. Procede d'identification de therapies de la maladie d'alzheimer a l'aide de modeles animaux transgeniques
IT1276680B1 (it) * 1995-06-08 1997-11-03 Oris Spa Processo di sintesi dell'1-piridiniopropan-3-solfonato
US5910427A (en) 1995-06-22 1999-06-08 La Jolla Institute For Allergy And Immunology Antigen non-specific glycosylation inhibiting factor derivatives
US5780361A (en) * 1995-06-23 1998-07-14 Nec Corporation Salicide process for selectively forming a monocobalt disilicide film on a silicon region
PT750907E (pt) * 1995-06-30 2002-08-30 American Cyanamid Co Formulacoes de vacinas estaveis para administracao parenterica metodo para a sua utilizacao e processo para a sua preparacao
WO1997003192A2 (fr) * 1995-07-07 1997-01-30 Darwin Molecular Corporation Gene du chromosome 1 et produits genetiques lies a la maladie d'alzheimer
US6685940B2 (en) 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
AUPN443995A0 (en) 1995-07-27 1995-08-17 Csl Limited Papillomavirus polyprotein
US6267958B1 (en) * 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
CA2229043C (fr) 1995-08-18 2016-06-07 Morphosys Gesellschaft Fur Proteinoptimierung Mbh Banques de proteines/(poly)peptides
WO1997006809A1 (fr) * 1995-08-21 1997-02-27 Cytrx Corporation Compositions et procedes pour la stimulation de la croissance
US5664823A (en) * 1995-09-18 1997-09-09 Ford Global Technologies, Inc. Instrument panel brace
US5731284A (en) 1995-09-28 1998-03-24 Amgen Inc. Method for treating Alzheimer's disease using glial line-derived neurotrophic factor (GDNF) protein product
AU3804995A (en) 1995-10-10 1997-04-30 Novartis Ag Melanoma-associated protein
US5985242A (en) * 1995-10-27 1999-11-16 Praecis Pharmaceuticals, Inc. Modulators of β-amyloid peptide aggregation comprising D-amino acids
EP0876615A1 (fr) 1995-11-10 1998-11-11 Elan Corporation Plc Peptides accroissant le transport dans les tissus et procedes d'identification et d'utilisation correspondants
WO1997018855A1 (fr) 1995-11-21 1997-05-29 Eduard Naumovich Lerner Dispositif pour ameliorer l'administration a l'organisme de substances et de composes biologiquement actifs
EP0866805A1 (fr) 1995-12-12 1998-09-30 Karolinska Innovations AB PEPTIDE FIXANT LA SEQUENCE KLVFF DE L'AMYLOIDE $g(b)
JPH09178743A (ja) 1995-12-27 1997-07-11 Oriental Yeast Co Ltd 可溶性appの定量法
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
ZA97452B (en) * 1996-01-25 1997-08-15 Trinity College Dublin Streptococcus equi vaccine.
AU1832797A (en) 1996-01-26 1997-08-20 Trustees Of Columbia University In The City Of New York, The A polypeptide from lung extract which binds amyloid-beta peptide
GB9602294D0 (en) * 1996-02-05 1996-04-03 Zeneca Ltd Heterocyclic compounds
US6096313A (en) 1996-02-09 2000-08-01 Ludwig Institute For Cancer Research Compositions containing immunogenic molecules and granulocyte-macrophage colony stimulating factor, as an adjuvant
WO1997032017A1 (fr) 1996-02-26 1997-09-04 Morphosys Gesellschaft Für Proteinoptimierung Mbh Nouvelle methode pour l'identification de sequences d'acides nucleiques codant pour deux (poly)peptides interactifs, ou plus
US6150091A (en) * 1996-03-06 2000-11-21 Baylor College Of Medicine Direct molecular diagnosis of Friedreich ataxia
US5870587A (en) * 1996-03-20 1999-02-09 International Business Machines Corporation Information-handling system, method, and article of manufacture including a mechanism for providing an improved application binary interface
DE69731357T2 (de) 1996-03-23 2006-02-02 The Research Foundation For Microbial Diseases Of Osaka University, Suita Funktionelles antigenfragment von tetanustoxin und tetanusvakzine
HUP9902438A3 (en) * 1996-03-29 2000-03-28 Bayer Ag Parapoxvirus vectors
WO1997036601A1 (fr) * 1996-04-03 1997-10-09 Anergen, Inc. Vaccins a peptides cycliques destines au traitement et a la prevention du diabete
EP0904365A1 (fr) * 1996-04-19 1999-03-31 The Government of the United States Of America, as represented by The Secretary, Department of Health and Human Services Regions reactives aux antigenes de la polyproteine du virus de l'hepatite-a
US6284533B1 (en) * 1996-05-01 2001-09-04 Avant Immunotherapeutics, Inc. Plasmid-based vaccine for treating atherosclerosis
DE69726003T2 (de) 1996-07-16 2004-08-26 Andreas Plückthun Immunglobulin-superfamilie domainen und fragmente mit erhöhter löslichkeit
WO1998004720A1 (fr) 1996-07-26 1998-02-05 Sloan-Kettering Institute For Cancer Research Procedes et reactifs destines a l'immunisation genetique
US7147851B1 (en) 1996-08-15 2006-12-12 Millennium Pharmaceuticals, Inc. Humanized immunoglobulin reactive with α4β7 integrin
CA2183901A1 (fr) 1996-08-22 1998-02-23 Johanna E. Bergmann Cibles pour le traitement et le diagnostic de la maladie d'alzheimer et du syndrome de down chez les humains
GB9617616D0 (en) 1996-08-22 1996-10-02 Osteometer Biotech As Assaying protein fragments in body fluids
ATE298765T1 (de) 1996-08-27 2005-07-15 Praecis Pharm Inc Beta-amyloid peptidaggregation regulierende peptide mit d-aminosäuren
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
WO1998022120A1 (fr) 1996-11-19 1998-05-28 The Wistar Institute Of Anatomy & Biology Reactifs pour traiter et diagnostiquer la maladie d'alzheimer
US6962984B2 (en) 1996-12-05 2005-11-08 Nihon University IgA nephropathy-related DNA
DK0966447T3 (da) 1997-03-03 2003-06-23 Boehringer Ingelheim Pharma Anvendelige små molekyler til behandling af betændelsessygdomme
US5798102A (en) 1997-03-04 1998-08-25 Milkhaus Laboratory, Inc. Treatment of cardiomyopathy
US6611610B1 (en) * 1997-04-02 2003-08-26 Gentex Corporation Vehicle lamp control
US6057098A (en) 1997-04-04 2000-05-02 Biosite Diagnostics, Inc. Polyvalent display libraries
JP3816111B2 (ja) 1997-04-09 2006-08-30 インテレクト・ニューロサイエンシズ・インコーポレーテッド β−アミロイド末端に特異的な組換え抗体、それをコードするDNA及びその使用法
PL194221B1 (pl) 1997-04-15 2007-05-31 Pharmexa As Zmodyfikowana cząsteczka ludzkiego TNFalfa, dimery, oligomery lub multimery zmodyfikowanej cząsteczki ludzkiego TNFalfa, wyizolowana cząsteczka DNA, wektor, wektor ekspresyjny, komórka gospodarza, sposób wytwarzania zmodyfikowanej cząsteczki ludzkiego TNFalfa, szczepionki przeciwko TNFalfa oraz zastosowanie przynajmniej jednej zmodyfikowanej cząsteczki ludzkiego TNFalfa
US5858205A (en) * 1997-05-13 1999-01-12 Uop Llc Multizone catalytic reforming process
EP1003531B1 (fr) 1997-05-20 2007-08-22 Ottawa Health Research Institute Procedes de preparation de constructions d'acides nucleiques
DE69810481T2 (de) 1997-06-13 2003-09-25 Genentech Inc Stabilisierte antikörperformulierung
WO1999000150A2 (fr) 1997-06-27 1999-01-07 Regents Of The University Of California Ciblage d'un medicament radiopharmaceutique peptidique utilisant in vivo la barriere hemato-encephalique d'un primate et un anticorps monoclonal dirige contre le recepteur humain de l'insuline
IT1293511B1 (it) 1997-07-30 1999-03-01 Gentili Ist Spa Anticorpi monoclonali catalitici ad attivita' proteasica per la lisi selettiva della componente proteica di placche e aggregati correlati
WO1999006587A2 (fr) 1997-08-01 1999-02-11 Morphosys Ag Nouvelle methode et nouveau phage d'identification d'une sequence d'acide nucleique
ATE491721T1 (de) 1997-08-01 2011-01-15 Max Planck Gesellschaft Zusammensetzung und verfahren zum nachweis von krankheiten in zusammenhang mit der bildung von amyloid-ähnlichen fibrillen oder protein- aggregaten
US6645495B1 (en) 1997-08-29 2003-11-11 Antigenics, Inc. Compositions of saponin adjuvants and excipients
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
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US7588766B1 (en) * 2000-05-26 2009-09-15 Elan Pharma International Limited 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
US20080050367A1 (en) * 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
US6923964B1 (en) 1997-12-02 2005-08-02 Neuralab Limited Active immunization of AScr for prion disorders
NZ504711A (en) 1997-12-03 2002-02-01 Fujisawa Pharmaceutical Co Soft-pellet drug and process for the preparation thereof
ES2253839T3 (es) 1997-12-03 2006-06-01 Neuralab, Ltd. Supresion de cambios relacionados con amiloide beta en la enfermedad de alzheimer.
FR2777015B3 (fr) 1998-02-23 2000-09-15 Financ De Biotechnologie Procede et moyens pour l'obtention de modeles cellulaires et animaux de maladies neurodegeneratives
NO314086B1 (no) 1998-05-08 2003-01-27 Gemvax As Peptider og farmasöytiske sammensetninger inneholdende disse, nukleinsyresekvenser som koder for slike peptider, plasmider og virusvektoreromfattende slike DNA-sekvenser samt anvendelse av disse for fremstilling avfarmasöytiske preparater til
WO1999060021A2 (fr) 1998-05-19 1999-11-25 Yeda Research And Development Co. Ltd. Lymphocytes t actives, antigenes specifiques du systeme nerveux et leur utilisation
JP2002515235A (ja) 1998-05-21 2002-05-28 ザ ユニヴァーシティ オブ テネシー リサーチ コーポレイション 坑アミロイド抗体を用いるアミロイドの除去方法
US6432710B1 (en) 1998-05-22 2002-08-13 Isolagen Technologies, Inc. Compositions for regenerating tissue that has deteriorated, and methods for using such compositions
US6710228B1 (en) * 1998-05-29 2004-03-23 Mycogen Corporation Cotton cells, plants, and seeds genetically engineered to express insecticidal and fungicidal chitin binding proteins (lectins)
US6727349B1 (en) 1998-07-23 2004-04-27 Millennium Pharmaceuticals, Inc. Recombinant anti-CCR2 antibodies and methods of use therefor
EA003634B1 (ru) 1998-10-05 2003-08-28 Фармэкса А/С Новые способы терапевтической вакцинации
WO2000023082A1 (fr) * 1998-10-19 2000-04-27 Yeda Research And Development Co. Ltd. Traitement du lupus erythemateux systemique par regulation negative de la reponse auto-immune a des autoantigenes
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
ES2216509T3 (es) 1999-01-19 2004-10-16 PHARMACIA & UPJOHN COMPANY Procedimiento de envasado para sustancias medicinales sensibles a la oxidacion.
EP1146898A1 (fr) 1999-01-22 2001-10-24 Matthew John During Traitement de troubles psychologiques a base de vaccin
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
UA81216C2 (en) 1999-06-01 2007-12-25 Prevention and treatment of amyloid disease
PE20010212A1 (es) 1999-06-01 2001-02-22 Neuralab Ltd Composiciones del peptido a-beta y procesos para producir las mismas
EP1192462A1 (fr) 1999-07-01 2002-04-03 Scios Inc. Prevention et traitement des troubles associes a l'amyloide
MXPA02000525A (es) 1999-07-15 2005-07-01 Inst Genetics Llc Formulaciones de il-11.
CA2381323C (fr) 1999-08-04 2009-10-06 University Of Southern California Proteine d'amyloide .beta. (ensemble globulaire et ses utilisations)
WO2001018169A2 (fr) 1999-09-03 2001-03-15 Ramot University Authority For Applied Research & Industrial Development Ltd. Agents et compositions et procedes utilisant lesdits agents et lesdites compositions pour le diagnostic et/ou le traitement ou la prevention des maladies formant des plaques.
US6824780B1 (en) 1999-10-29 2004-11-30 Genentech, Inc. Anti-tumor antibody compositions and methods of use
CN1433321A (zh) 1999-11-29 2003-07-30 尼奥切姆公司 预防和治疗Alzheimer症和其他与淀粉样蛋白有关的疾病的疫苗
JP4804690B2 (ja) 1999-12-08 2011-11-02 インテレクト・ニューロサイエンシズ・インコーポレーテッド 免疫原としてのキメラペプチド、それに対する抗体、およびキメラペプチドまたは抗体を用いた免疫法
AU783144B2 (en) 2000-02-21 2005-09-29 H. Lundbeck A/S Novel method for down-regulation of amyloid
PL204878B1 (pl) 2000-02-21 2010-02-26 Lundbeck & Co As H Zastosowanie analogu autologicznego zwierzęcego Aβ albo polipeptydu APP lub sekwencji kodującej taki analog lub mikroorganizmu lub wirusa, który niesie nukleotydową sekwencję kodującą taki analog, analog polipeptydu amyloidogennego, immunogenna kompozycja, fragment kwasu nukleinowego kodujący analog, wektor, stransformowana komórka, kompozycja do indukowania produkcji przeciwciał i stabilna linia komórkowa
US6294221B1 (en) * 2000-03-08 2001-09-25 E. I. Du Pont De Nemours And Company Process for spray-coating with frequent changes between aqueous and non-aqueous coating agents inside a spray-coating chamber
US6548840B1 (en) * 2000-04-03 2003-04-15 Hrl Laboratories, Llc Monolithic temperature compensation scheme for field effect transistor integrated circuits
WO2001077167A2 (fr) 2000-04-05 2001-10-18 University Of Tennessee Research Corporation Techniques d'etude, de diagnostic et de traitement d'amylose
EP1385541B1 (fr) 2000-04-13 2008-06-18 Corixa Corporation Compositions immunostimulantes comprenant un phosphate d'aminoalkyl glucosaminide et du qs-21
AU2001268005A1 (en) 2000-07-07 2002-01-21 Lars Lannfelt Prevention and treatment of alzheimer's disease
EP1172378A1 (fr) 2000-07-12 2002-01-16 Richard Dr. Dodel Anticorps humains anti-beta-amyloid et leur utilisation pour le traitement de la maladie d'Alzheimer
US7199102B2 (en) * 2000-08-24 2007-04-03 The Regents Of The University Of California Orally administered peptides synergize statin activity
US20030092145A1 (en) 2000-08-24 2003-05-15 Vic Jira Viral vaccine composition, process, and methods of use
CA2421374C (fr) 2000-09-06 2017-03-07 Aventis Pharma S.A. Procedes et compositions relatifs a des maladies associees a l'amyloide
IT1319277B1 (it) 2000-10-24 2003-09-26 Chiesi Farma Spa Proteine di fusione utili per il trattamento di immunizzazione dellamalattia di alzheimer.
IL139308A0 (en) 2000-10-26 2001-11-25 Marikovsky Moshe Peptides from amyloid precursor protein which inhibit tumor growth and metastasis
ATE360392T1 (de) 2000-11-02 2007-05-15 Cornell Res Foundation Inc In vivo multiphoton diagnostische detektion und bilddarstellung einer neurodegenerativen erkrankung
PT1346041E (pt) * 2000-11-27 2007-06-05 Praecis Pharm Inc Agentes terapêuticos e métodos para a sua utilização no tratamento de uma doença amiloidogénica.
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-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
US20020160394A1 (en) * 2001-01-24 2002-10-31 Bayer Corporation Regulation of transthyretin to treat obesity
EP1251138B1 (fr) * 2001-04-19 2006-07-26 Hermann Dr. Schätzl Dimères de prions comme vaccin
US7320790B2 (en) 2001-04-30 2008-01-22 Eli Lilly And Company Humanized antibodies
ES2437875T3 (es) 2001-04-30 2014-01-14 Eli Lilly And Company Anticuerpos humanizados que reconocen el péptido beta-amiloide
US6524624B1 (en) * 2001-05-16 2003-02-25 Alcide Corporation Two-part disinfecting systems and compositions and methods related thereto
GB0113179D0 (en) 2001-05-31 2001-07-25 Novartis Ag Organic compounds
US6888849B2 (en) * 2001-06-15 2005-05-03 Lucent Technologies Inc. Method for evaluating capacity utilization of a terminus in a communication system
US20020197258A1 (en) 2001-06-22 2002-12-26 Ghanbari Hossein A. Compositions and methods for preventing protein aggregation in neurodegenerative diseases
US20030113316A1 (en) 2001-07-25 2003-06-19 Kaisheva Elizabet A. Stable lyophilized pharmaceutical formulation of IgG antibodies
US20030135035A1 (en) 2001-08-09 2003-07-17 Mark Shannon Human ZZAP1 protein
AU2002324468A1 (en) 2001-08-17 2003-03-03 Eli Lilly And Company Rapid improvement of cognition in conditions related to abeta
EP1432444A4 (fr) 2001-08-17 2005-11-02 Lilly Co Eli Anticorps anti-a beta
CA2452104A1 (fr) 2001-08-17 2003-02-27 Eli Lilly And Company Utilisation d'anticorps ayant une forte affinite pour l'ass soluble dans le traitement de pathologies et de maladies liees a l'ass
US20030082191A1 (en) 2001-08-29 2003-05-01 Poduslo Joseph F. Treatment for central nervous system disorders
DE60237459D1 (de) 2001-09-10 2010-10-07 Anticancer Inc Verbesserte beobachtungsgenauigkeit bei der absiedelung von tumoren
US6736142B2 (en) * 2001-09-13 2004-05-18 Gines Sanchez Gomez Protective tube and harness
US6907297B2 (en) 2001-09-28 2005-06-14 Ethicon, Inc. Expandable intracardiac return electrode and method of use
US6597198B2 (en) * 2001-10-05 2003-07-22 Intel Corporation Current mode bidirectional port with data channel used for synchronization
GB0124446D0 (en) * 2001-10-11 2001-12-05 Short Brothers Ltd Aircraft propulsive power unit
US7781413B2 (en) 2001-10-31 2010-08-24 Board Of Regents, The University Of Texas System SEMA3B inhibits tumor growth and induces apoptosis in cancer cells
PT1441589E (pt) 2001-11-08 2012-08-13 Abbott Biotherapeutics Corp Formulação farmacêutica líquida estável de anticorpos igg
US7479482B2 (en) * 2001-11-21 2009-01-20 New York University Synthetic immunogenic but non-deposit-forming polypeptides and peptides homologous to amyloid β, prion protein, amylin, α-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 (es) 2002-02-20 2005-01-19 Hoffmann La Roche Anticuerpos anti-a beta y su uso
ATE482719T1 (de) 2002-02-21 2010-10-15 Univ Duke Behandlungsverfahren unter verwendung von anti- cd22-antikörpern
US6688651B2 (en) * 2002-02-21 2004-02-10 Dmt Co., Ltd. Device for locking cap nut for coupling
MY139983A (en) 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
CA2390700C (fr) * 2002-06-11 2004-09-07 Henry Tsang Pain de savon lumineux sonore
GB0213437D0 (en) 2002-06-12 2002-07-24 Univ Cranfield Temporary tattoos: A novel vehicle for skin testing
US6892535B2 (en) * 2002-06-14 2005-05-17 Volvo Construction Equipment Holding Sweden Ab Hydraulic circuit for boom cylinder combination having float function
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
DK1524994T3 (da) 2002-07-19 2011-08-15 Cytos Biotechnology Ag Vaccinesammensætninger indeholdende amyloid beta 1-6-antigen-arrays
CA2487528A1 (fr) 2002-07-24 2004-02-12 Innogenetics N.V. Prevention, traitement et diagnostic de maladies associees a la formation et/ou a l'agregation de la beta-amyloide
JP2006508072A (ja) 2002-10-01 2006-03-09 ノースウエスタン ユニバーシティ アミロイドベータ由来拡散性リガンド(ADDLs)、ADDL代替物、ADDL結合性分子、およびそれらの使用
US20040080762A1 (en) * 2002-10-25 2004-04-29 Xerox Corporation Half-tone based enhancement of black
US20060019850A1 (en) * 2002-10-31 2006-01-26 Korzenski Michael B Removal of particle contamination on a patterned silicon/silicon dioxide using dense fluid/chemical formulations
FR2846667B1 (fr) 2002-11-06 2004-12-31 Pasteur Institut Fragments variables d'anticorps de camelides a chaine unique diriges contre le peptide beta-amyloide 1-42 et leurs applications pour le diagnostic et le traitement des maladies neuroagregatives
AU2003293543A1 (en) 2002-12-13 2004-07-09 Abgenix, Inc. System and method for stabilizing antibodies with histidine
US6787129B1 (en) 2003-01-13 2004-09-07 Zenitech Llc Castor polyester as gloss agents in anionic systems
CA2513722A1 (fr) 2003-02-01 2004-08-19 Neuralab Limited Immunisation active permettant de produire des anticorps contre a-beta soluble
US7575747B2 (en) 2003-02-10 2009-08-18 Applied Molecular Evolution Aβ binding molecules
JP4869064B2 (ja) 2003-04-04 2012-02-01 ジェネンテック, インコーポレイテッド 高濃度抗体及びタンパク質製剤
EP1480041A1 (fr) 2003-05-22 2004-11-24 Innogenetics N.V. Méthode de prédiction, diagnostic et diagnostic différential de la maladie d'Alzheimer
PE20050627A1 (es) 2003-05-30 2005-08-10 Wyeth Corp Anticuerpos humanizados que reconocen el peptido beta amiloideo
US20060182321A1 (en) 2003-07-07 2006-08-17 Agency For Science, Technology And Research Method and apparatus for extracting third ventricle information
WO2005014041A2 (fr) 2003-07-24 2005-02-17 Novartis Ag Substances et procedes pour le traitement des amyloses
EP1670827A2 (fr) 2003-09-05 2006-06-21 Eli Lilly And Company Anticorps anti-ghreline
CA2445743A1 (fr) 2003-10-08 2005-04-08 The University Of British Columbia Methodes pour moduler les reponses neuronales
AU2003271174A1 (en) 2003-10-10 2005-04-27 Chugai Seiyaku Kabushiki Kaisha Double specific antibodies substituting for functional protein
SG149039A1 (en) 2003-12-17 2009-01-29 Elan Pharm Inc Ass IMMUNOGENIC PEPTIDE CARRIER CONJUGATES AND METHODS OF PRODUCING SAME
TWI355390B (en) 2003-12-17 2012-01-01 Wyeth Llc Immunogenic peptide carrier conjugates and methods
US20050214222A1 (en) 2004-02-13 2005-09-29 Mckinnon Stuart J In vivo imaging of amyloid plaques in glaucoma using intravenous injectable dyes
ME01601B (me) 2004-03-15 2014-09-20 Janssen Pharmaceutica Nv Modulatori opioidnog receptora
ES2427963T3 (es) 2004-07-02 2013-11-05 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Uso de derivados de tioflavina radiomarcados en formación de imágenes de amiloide para evaluación de las terapias antiamiloide
EP2298807A3 (fr) 2004-07-30 2011-05-18 Rinat Neuroscience Corp. Anticorps anti peptide amyloide beta, et leurs procedes d' utilisation
US7328838B2 (en) * 2004-09-09 2008-02-12 Igt Counterfeit cashless instrument detection methods and systems
WO2006031653A2 (fr) 2004-09-10 2006-03-23 Wyeth Anticorps anti-5t4 humanises et conjugues anticorps anti-5t4/calicheamicine
BRPI0516572A (pt) 2004-10-05 2008-09-16 Wyeth Corp métodos e composições para melhorar a produção de proteìnas recombinantes
WO2006047670A2 (fr) 2004-10-26 2006-05-04 Wyeth Procedes permettant d'evaluer les anticorps diriges contre des antigenes associes aux maladies neurodegeneratives
US20060026911A1 (en) * 2004-11-18 2006-02-09 Sutton Adam F Footer track with moisture vent
TW200635608A (en) 2004-12-15 2006-10-16 Neuralab Ltd Aβ antibodies for use in improving cognition
US20060240486A1 (en) 2004-12-15 2006-10-26 Johnson-Wood Kelly L Immunoprecipitation-based assay for predicting in vivo efficacy of beta-amyloid antibodies
WO2006066118A2 (fr) 2004-12-15 2006-06-22 Neuralab Limited Conditionnement contextuel des peurs pour predire l'efficacite immunotherapeutique
EP1838854B1 (fr) 2004-12-15 2012-10-31 Janssen Alzheimer Immunotherapy Anticorps reconnaissant le peptide beta amyloide
WO2006066089A1 (fr) 2004-12-15 2006-06-22 Neuralab Limited Anticorps du peptide beta-amyloide humanises utilises dans l'amelioration de la cognition
GT200600031A (es) 2005-01-28 2006-08-29 Formulacion anticuerpo anti a beta
CA2595380A1 (fr) 2005-01-28 2006-08-03 Wyeth Formulations polypeptidiques liquides stabilisees
MX2007013825A (es) 2005-05-05 2008-01-18 Merck & Co Inc Composiciones de un conjugado peptidico y metodos para la prevencion y tratamiento de la enfermedad de alzheimer.
CN104926938B (zh) 2005-06-17 2019-06-04 惠氏有限责任公司 纯化含Fc区蛋白的方法
ATE550338T1 (de) * 2005-07-18 2012-04-15 Merck Sharp & Dohme Spiropiperidininhibitoren von beta-secretase zur behandlung von alzheimer-krankheit
AU2006315686A1 (en) * 2005-11-10 2007-05-24 Roskamp Research, Llc Modulation of angiogenesis by A-beta peptide fragments
UA94734C2 (ru) 2006-03-30 2011-06-10 Глаксо Груп Лимитед ТЕРАПЕВТИЧЕСКОЕ АНТИТЕЛО, КОТОРОЕ СВЯЗЫВАЕТСЯ С b-АМИЛОИДНЫМ ПЕПТИДОМ
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2010044803A1 (fr) 2008-10-17 2010-04-22 Elan Pharma International Limited Traitement de maladies amyloïdogènes
WO2009017467A1 (fr) 2007-07-27 2009-02-05 Elan Pharma International Limited Traitement de maladies amyloïdogéniques
PL2046833T3 (pl) 2006-07-14 2014-01-31 Ac Immune Sa Humanizowane przeciwciało przeciw amyloidowi beta
US20100055036A1 (en) 2007-03-12 2010-03-04 National Institute of Radiolotgical Sciences Pet visualization of amyloid-associated neuroinflammation in the brain
RU2523894C2 (ru) 2007-04-18 2014-07-27 Янссен Альцгеймер Иммунотерапи Предупреждение и лечение церебральной амилоидной ангиопатии
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
JO3076B1 (ar) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap نظم العلاج المناعي المعتمد على حالة apoe
KR101603076B1 (ko) * 2007-12-28 2016-03-14 프로테나 바이오사이언시즈 리미티드 아밀로이드증의 치료 및 예방
JP5771525B2 (ja) 2008-09-18 2015-09-02 セダーズ−シナイ メディカル センター アルツハイマー病を検出するための光学的方法
JP2013521233A (ja) 2010-02-25 2013-06-10 ヤンセン アルツハイマー イミュノセラピー Aβを標的とする免疫療法のPETモニタリング
EP2560681A4 (fr) 2010-04-22 2013-09-25 Janssen Alzheimer Immunotherap Utilisation de tau pour un suivi d'immunothérapie

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816397A (en) * 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
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
US5096706A (en) * 1986-03-25 1992-03-17 National Research Development Corporation Antigen-based treatment for adiposity
US6548640B1 (en) * 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making 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
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
US5624821A (en) * 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5648260A (en) * 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5733547A (en) * 1987-06-24 1998-03-31 Autoimmune, Inc. Treatment of autoimmune arthritis by oral administration of type I or type III collagen
US5641474A (en) * 1987-06-24 1997-06-24 Autoimmune, Inc. Prevention of autoimmune diseases by aerosol administration of autoantigens
US5869054A (en) * 1987-06-24 1999-02-09 Autoimmune Inc. Treatment of multiple sclerosis by oral administration of autoantigens
US5869093A (en) * 1987-06-24 1999-02-09 Autoimmune Inc. Treatment of immune diseases by oral administration of autoantigens
US5645820A (en) * 1987-06-24 1997-07-08 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
US5641473A (en) * 1987-06-24 1997-06-24 Autoimmune, Inc. Treatment of autoimmune diseases by aerosol administration of autoantigens
US5004697A (en) * 1987-08-17 1991-04-02 Univ. Of Ca Cationized antibodies for delivery through the blood-brain barrier
US5231000A (en) * 1987-10-08 1993-07-27 The Mclean Hospital Antibodies to A4 amyloid peptide
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US6180370B1 (en) * 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
US20050123534A1 (en) * 1989-12-21 2005-06-09 Celltech R&D Limited Humanised antibodies
US20030039645A1 (en) * 1989-12-21 2003-02-27 Adair John Robert Humanised antibodies
US5859205A (en) * 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US20050136054A1 (en) * 1989-12-21 2005-06-23 Celltech R&D Limited Humanised antibodies
US5753624A (en) * 1990-04-27 1998-05-19 Milkhaus Laboratory, Inc. Materials and methods for treatment of plaquing disease
US5387742A (en) * 1990-06-15 1995-02-07 Scios Nova Inc. Transgenic mice displaying the amyloid-forming pathology of alzheimer's disease
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
US5192753A (en) * 1991-04-23 1993-03-09 Mcgeer Patrick L Anti-rheumatoid arthritic drugs in the treatment of dementia
US6054297A (en) * 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
US6407213B1 (en) * 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
US5955079A (en) * 1992-02-11 1999-09-21 Henry Jackson Foundation For The Advancement Of Military Medicine Dual carrier immunogenic construct
US6933368B2 (en) * 1992-03-09 2005-08-23 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation of immunoglobulin variable region
US5721130A (en) * 1992-04-15 1998-02-24 Athena Neurosciences, Inc. Antibodies and fragments thereof which bind the carboxyl-terminus of an amino-terminal fragment of βAPP
US5441870A (en) * 1992-04-15 1995-08-15 Athena Neurosciences, Inc. Methods for monitoring cellular processing of β-amyloid precursor protein
US5891991A (en) * 1992-04-20 1999-04-06 The General Hospital Corporation Amyloid precursor-like protein and uses thereof
US5766846A (en) * 1992-07-10 1998-06-16 Athena Neurosciences Methods of screening for compounds which inhibit soluble β-amyloid peptide production
US5593846A (en) * 1992-07-10 1997-01-14 Athena Neurosciences Methods for the detection of soluble β-amyloid peptide
US6261569B1 (en) * 1992-08-27 2001-07-17 Deakin Research Limited 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
US5605811A (en) * 1992-10-26 1997-02-25 Athena Neurosciences, Inc. Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
US5955317A (en) * 1993-01-25 1999-09-21 Takeda Chemical Industries, Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5750349A (en) * 1993-01-25 1998-05-12 Takeda Chemical Industries Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5514548A (en) * 1993-02-17 1996-05-07 Morphosys Gesellschaft Fur Proteinoptimerung Mbh Method for in vivo selection of ligand-binding proteins
US5776468A (en) * 1993-03-23 1998-07-07 Smithkline Beecham Biologicals (S.A.) Vaccine compositions containing 3-0 deacylated monophosphoryl lipid A
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
US5736142A (en) * 1993-09-14 1998-04-07 Cytel Corporation Alteration of immune response using pan DR-binding peptides
US5612486A (en) * 1993-10-27 1997-03-18 Athena Neurosciences, Inc. Transgenic animals harboring APP allele having swedish mutation
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)
US5434170A (en) * 1993-12-23 1995-07-18 Andrulis Pharmaceuticals Corp. Method for treating neurocognitive disorders
US6262335B1 (en) * 1994-01-27 2001-07-17 Johns Hopkins University Transgenic mice expressing APP mutant at amino acids 717, 721 and 722
US5877399A (en) * 1994-01-27 1999-03-02 Johns Hopkins University Transgenic mice expressing APP-Swedish mutation develop progressive neurologic disease
US5935927A (en) * 1994-02-03 1999-08-10 The Picower Institute For Medical Research Compositions and methods for stimulating amyloid removal in amyloidogenic diseases using advanced glycosylation endproducts
US5622701A (en) * 1994-06-14 1997-04-22 Protein Design Labs, Inc. Cross-reacting monoclonal antibodies specific for E- and P-selectin
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
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)
US5786180A (en) * 1995-02-14 1998-07-28 Bayer Corporation Monoclonal antibody 369.2B specific for β A4 peptide
US5869046A (en) * 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6562341B2 (en) * 1995-09-14 2003-05-13 The Regents Of The University Of California Antibodies specific for native PrPSc
US5750361A (en) * 1995-11-02 1998-05-12 The Regents Of The University Of California Formation and use of prion protein (PRP) complexes
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
US6022859A (en) * 1996-11-15 2000-02-08 Wisconsin Alumni Research Foundation Inhibitors of β-amyloid toxicity
US6218506B1 (en) * 1997-02-05 2001-04-17 Northwestern University Amyloid β protein (globular assembly and uses thereof)
US20030068316A1 (en) * 1997-02-05 2003-04-10 Klein William L. Anti-ADDL antibodies and uses thereof
US6277375B1 (en) * 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
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
US20030073655A1 (en) * 1997-04-09 2003-04-17 Chain Daniel G. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US20020058267A1 (en) * 1997-04-16 2002-05-16 American Home Products Corporation Beta-amyloid peptide-binding proteins and polynucleotides encoding the same
US20050163788A1 (en) * 1997-12-02 2005-07-28 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6875434B1 (en) * 1997-12-02 2005-04-05 Neuralab Limited Methods of treatment of Alzheimer's disease
US6866850B2 (en) * 1997-12-02 2005-03-15 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6890535B1 (en) * 1997-12-02 2005-05-10 Neuralab Limited Pharmaceutical compositions and methods for treatment of amyloid diseases
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
US6905686B1 (en) * 1997-12-02 2005-06-14 Neuralab Limited Active immunization for treatment of alzheimer's disease
US6866849B2 (en) * 1997-12-02 2005-03-15 Neuralab Limited Prevention and treatment of amyloidogenic disease
US6750324B1 (en) * 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
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
US6528624B1 (en) * 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6194551B1 (en) * 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6538124B1 (en) * 1998-04-02 2003-03-25 Genentech, Inc. Polypeptide variants
US20050059591A1 (en) * 1998-04-07 2005-03-17 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050059802A1 (en) * 1998-04-07 2005-03-17 Neuralab Ltd Prevention and treatment of amyloidogenic disease
US6936698B2 (en) * 1998-04-28 2005-08-30 Smithkline Beecham Monoclonal antibodies with reduced immunogenicity
US20030147882A1 (en) * 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US20050009150A1 (en) * 1998-11-30 2005-01-13 Elan Pharmaceuticals, Inc. Humanized antibodies that recognize beta amyloid peptide
US20020102261A1 (en) * 1999-06-16 2002-08-01 Boston Biomedical Research Institute Immunological control of beta-amyloid levels in vivo
US20010018053A1 (en) * 1999-09-14 2001-08-30 Mcmichael John Methods for treating disease states comprising administration of low levels of antibodies
US20020094335A1 (en) * 1999-11-29 2002-07-18 Robert Chalifour Vaccine for the prevention and treatment of alzheimer's and amyloid related diseases
US6399314B1 (en) * 1999-12-29 2002-06-04 American Cyanamid Company Methods of detection of amyloidogenic proteins
US20040043418A1 (en) * 2000-02-24 2004-03-04 Holtzman David M. Humanized antibodies that sequester Abeta peptide
US6713450B2 (en) * 2000-05-22 2004-03-30 New York University Synthetic immunogenic but non-amyloidogenic peptides homologous to amyloid β for induction of an immune response to amyloid β and amyloid deposits
US20020077288A1 (en) * 2000-05-22 2002-06-20 New York University Synthetic immunogenic but non-amyloidogenic peptides homologous to amyloid beta for induction of an immune response to amyloid beta and amyloid deposits
US6761888B1 (en) * 2000-05-26 2004-07-13 Neuralab Limited Passive immunization 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
US20030068325A1 (en) * 2001-05-25 2003-04-10 Wang Chang Yi Immunogenic peptide composition for the prevention and treatment of Altzheimers Disease

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9051363B2 (en) 1997-12-02 2015-06-09 Janssen Sciences Ireland Uc Humanized antibodies that recognize beta amyloid peptide
US8642044B2 (en) 1997-12-02 2014-02-04 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8535673B2 (en) 1997-12-02 2013-09-17 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8034348B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US8034339B2 (en) 1997-12-02 2011-10-11 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidogenic disease
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US7893214B2 (en) 1997-12-02 2011-02-22 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US8105594B2 (en) 1998-05-21 2012-01-31 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US20100322932A1 (en) * 1998-05-21 2010-12-23 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
US20050158304A1 (en) * 2000-05-26 2005-07-21 Neuralab Limited Prevention and treatment of amyloidogenic disease
US8409575B2 (en) 2000-07-07 2013-04-02 Bioarctic Neuroscience Ab Antibodies specific for amyloid beta protofibril
US20080181902A1 (en) * 2000-07-07 2008-07-31 Lars Lannfelt Prevention and treatment of alzheimer's disease
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US8128928B2 (en) 2002-03-12 2012-03-06 Wyeth Llc Humanized antibodies that recognize beta amyloid peptide
US20040213800A1 (en) * 2003-02-01 2004-10-28 Seubert Peter A. Active immunization to generate antibodies to soluble A-beta
US7871615B2 (en) 2003-05-30 2011-01-18 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US20050118651A1 (en) * 2003-05-30 2005-06-02 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US8404459B2 (en) 2004-06-21 2013-03-26 Bioarctic Neuroscience Ab Antibodies specific for soluble amyloid beta peptide protofibrils and uses thereof
US8999936B2 (en) 2004-06-21 2015-04-07 Bioarctic Neuroscience Ab Antibodies specific for soluble amyloid beta peptide protofibrils and uses thereof
US20090155246A1 (en) * 2004-06-21 2009-06-18 Bioartic Neuroscience Ab Antibodies Specific For Soluble Amyloid Beta Peptide Protofibrils and Uses Thereof
US8106164B2 (en) 2004-06-21 2012-01-31 Bioarctic Neuroscience Ab Antibodies specific for soluble amyloid beta peptide protofibrils and uses thereof
US8168188B1 (en) 2004-08-11 2012-05-01 Kyoto University Antibody and utilization of the same
US20090285806A1 (en) * 2004-10-05 2009-11-19 Martin Sinacore Methods and compositions for improving recombinant protein production
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
US7906625B2 (en) 2005-01-24 2011-03-15 Amgen Inc. Humanized anti-amyloid antibody
US20080292639A1 (en) * 2005-01-24 2008-11-27 Amgen Inc. Humanized Anti-Amyloid Antibody
US20060193850A1 (en) * 2005-01-28 2006-08-31 Warne Nicholas W Anti a beta antibody formulation
US20100166752A1 (en) * 2005-01-28 2010-07-01 Janssen Alzheimer Immunotherapy Anti A Beta Antibody Formulation
US8318164B2 (en) 2005-01-28 2012-11-27 Janssen Alzheimer Immunotherapy Anti A beta antibody formulation
US7635473B2 (en) 2005-01-28 2009-12-22 Janssen Alzheimer Immunotherapy Anti Aβ antibody formulation
US20090258009A1 (en) * 2006-03-23 2009-10-15 Bioarctic Neuroscience Ab Protofibril selective antibodies and the use thereof
US8025878B2 (en) 2006-03-23 2011-09-27 Bioarctic Neuroscience Ab Protofibril selective antibodies and the use thereof
US9034334B2 (en) 2006-03-23 2015-05-19 Bioarctic Neuroscience Ab Protofibril selective antibodies and the use thereof
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
US8613920B2 (en) 2007-07-27 2013-12-24 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US9644025B2 (en) 2007-10-17 2017-05-09 Wyeth Llc Immunotherapy regimes dependent on ApoE status
US20090155256A1 (en) * 2007-10-17 2009-06-18 Wyeth Immunotherapy Regimes Dependent On APOE Status
WO2009085200A2 (fr) 2007-12-21 2009-07-09 Amgen Inc. Anticorps anti-amyloïde et utilisations de ceux-ci
EP2261254A2 (fr) 2007-12-21 2010-12-15 Amgen, Inc Anticorps anti-amyloïdes et leurs utilisations
US8414893B2 (en) 2007-12-21 2013-04-09 Amgen Inc. Anti-amyloid antibodies and uses thereof
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
WO2010072384A1 (fr) 2008-12-22 2010-07-01 F. Hoffmann-La Roche Ag Anticorps anti-idiotype dirigé contre un anticorps dirigé contre le peptide amyloïde β
US8614297B2 (en) 2008-12-22 2013-12-24 Hoffmann-La Roche Inc. Anti-idiotype antibody against an antibody against the amyloid β peptide
US20100167313A1 (en) * 2008-12-22 2010-07-01 Ulrich Essig Anti-idiotype antibody against an antibody against the amyloid beta peptide
US9573994B2 (en) 2014-07-10 2017-02-21 Bioarctic Neuroscience Ab Aβ protofibril binding antibodies

Also Published As

Publication number Publication date
CO4980846A1 (es) 2000-11-27
US6972127B2 (en) 2005-12-06
US8642044B2 (en) 2014-02-04
US20050037026A1 (en) 2005-02-17
JP2010215651A (ja) 2010-09-30
IL191904A (en) 2014-05-28
EP1690547A1 (fr) 2006-08-16
US20130058869A1 (en) 2013-03-07
CN1281366A (zh) 2001-01-24
EP1994937B1 (fr) 2010-12-29
HRP20090568A2 (hr) 2010-06-30
DE69839647D1 (de) 2008-08-07
TWI239847B (en) 2005-09-21
US8034348B2 (en) 2011-10-11
EA200401473A1 (ru) 2005-04-28
HUP0100627A1 (hu) 2001-06-28
US20050191292A1 (en) 2005-09-01
EA200000608A1 (ru) 2000-12-25
US20140227274A1 (en) 2014-08-14
EA009283B1 (ru) 2007-12-28
US20050249727A1 (en) 2005-11-10
DE1033996T1 (de) 2001-06-07
PT1679080E (pt) 2009-09-10
EE200000379A (et) 2001-04-16
ES2262460T1 (es) 2006-12-01
CA2312920A1 (fr) 1999-06-10
EP1033996A4 (fr) 2004-05-26
US8034339B2 (en) 2011-10-11
ID25504A (id) 2000-10-05
BG104562A (en) 2001-01-31
BR9815357A (pt) 2000-10-24
CY1109368T1 (el) 2014-07-02
SI1033996T1 (sl) 2009-02-28
DE69840969D1 (de) 2009-08-20
EP1033996B1 (fr) 2008-06-25
US20050142132A1 (en) 2005-06-30
EA007218B1 (ru) 2006-08-25
KR20120135915A (ko) 2012-12-17
NO20100061L (no) 2000-07-31
US20050249725A1 (en) 2005-11-10
KR20010032635A (ko) 2001-04-25
JP2002502802A (ja) 2002-01-29
ATE399016T1 (de) 2008-07-15
IL191904A0 (en) 2008-12-29
EP1033996A1 (fr) 2000-09-13
SI1994937T1 (sl) 2011-04-29
CY1111370T1 (el) 2015-08-05
KR20090078366A (ko) 2009-07-17
HU228061B1 (en) 2012-09-28
NO332813B1 (no) 2013-01-21
CY1108331T1 (el) 2014-02-12
SA99191269B1 (ar) 2006-03-15
DE06075704T1 (de) 2006-12-28
US6946135B2 (en) 2005-09-20
EP1679080A2 (fr) 2006-07-12
US20050163788A1 (en) 2005-07-28
CZ20001706A3 (cs) 2000-11-15
US20050019343A1 (en) 2005-01-27
US20050196399A1 (en) 2005-09-08
US7014855B2 (en) 2006-03-21
PT1994937E (pt) 2011-02-22
HU228493B1 (en) 2013-03-28
HUP0100627A3 (en) 2006-03-28
SI2305282T1 (sl) 2013-10-30
IL193245A0 (en) 2009-09-22
US20080227719A1 (en) 2008-09-18
EP1690547B1 (fr) 2009-07-08
WO1999027944A1 (fr) 1999-06-10
HK1094535A1 (en) 2007-04-04
NO328865B1 (no) 2010-06-07
DK2305282T3 (da) 2013-10-07
NO20002784L (no) 2000-07-31
SG111083A1 (en) 2005-05-30
PT2305282E (pt) 2013-09-27
HRP20000443A2 (en) 2000-10-31
US20050019330A1 (en) 2005-01-27
NO20002784D0 (no) 2000-05-31
US8535673B2 (en) 2013-09-17
EA200601132A1 (ru) 2007-02-27
ES2263408T1 (es) 2006-12-16
DK1033996T3 (da) 2008-09-22
EP1679080B1 (fr) 2009-06-17
US20050031629A1 (en) 2005-02-10
PL202698B1 (pl) 2009-07-31
AR020050A1 (es) 2002-04-10
US20050191314A1 (en) 2005-09-01
NO20092874L (no) 2000-07-31
AU1706199A (en) 1999-06-16
NZ504569A (en) 2005-01-28
KR100936419B1 (ko) 2010-01-12
CA2312920C (fr) 2016-07-12
EP2305282A2 (fr) 2011-04-06
PE20001383A1 (es) 2000-11-25
GEP20043319B (en) 2004-05-10
KR101248711B1 (ko) 2013-03-28
DK1994937T3 (da) 2011-02-07
US20040228865A1 (en) 2004-11-18
DE69842082D1 (de) 2011-02-10
SI1679080T1 (sl) 2009-12-31
ATE433760T1 (de) 2009-07-15
HRP20000443B1 (en) 2009-11-30
CN100374151C (zh) 2008-03-12
EP2305282B1 (fr) 2013-07-10
IL136252A0 (en) 2001-05-20
US6808712B2 (en) 2004-10-26
EP2305282A3 (fr) 2011-05-18
US20080227718A1 (en) 2008-09-18
US20050048049A1 (en) 2005-03-03
MY134906A (en) 2007-12-31
DE69840922D1 (de) 2009-07-30
PT1033996E (pt) 2008-10-02
JP4677334B2 (ja) 2011-04-27
ES2263408T3 (es) 2009-11-16
US6982084B2 (en) 2006-01-03
JP4677333B2 (ja) 2011-04-27
CZ303137B6 (cs) 2012-04-25
IL136252A (en) 2010-11-30
HK1095265A1 (en) 2007-05-04
US20080096818A1 (en) 2008-04-24
JP4677094B2 (ja) 2011-04-27
NO331425B1 (no) 2011-12-27
GEP20053715B (en) 2005-12-26
US20120276116A1 (en) 2012-11-01
EE05377B1 (et) 2011-02-15
US20060034858A1 (en) 2006-02-16
TR200001608T2 (tr) 2001-07-23
US20050013815A1 (en) 2005-01-20
WO1999027944A9 (fr) 1999-09-23
EP1994937A3 (fr) 2008-12-24
ES2310017T3 (es) 2008-12-16
ATE493149T1 (de) 2011-01-15
DK1679080T3 (da) 2009-09-28
ATE435659T1 (de) 2009-07-15
EP1994937A2 (fr) 2008-11-26
DE06075479T1 (de) 2006-11-16
IS5500A (is) 2000-05-17
JP2006131639A (ja) 2006-05-25
JP2006077030A (ja) 2006-03-23
ES2428740T3 (es) 2013-11-11
PL342649A1 (en) 2001-06-18
US20040170641A1 (en) 2004-09-02
PL202706B1 (pl) 2009-07-31
US20090191231A1 (en) 2009-07-30
US20060029611A1 (en) 2006-02-09
US20040171816A1 (en) 2004-09-02
IL205298A0 (en) 2011-07-31
EP1679080A3 (fr) 2006-07-19

Similar Documents

Publication Publication Date Title
CA2518275C (fr) Anticorps humanises reconnaissant le peptide beta-amyloide
US7893214B2 (en) Humanized antibodies that recognize beta amyloid peptide
US7700751B2 (en) Humanized antibodies that recognize β-amyloid peptide
US7790856B2 (en) Humanized antibodies that recognize beta amyloid peptide
US7582733B2 (en) Humanized antibodies that recognize beta amyloid peptide
US20040171815A1 (en) Humanized antibodies that recognize beta amyloid peptide
AU2002225921A1 (en) Humanized antibodies that recognize beta amyloid peptide
AU2007231637B2 (en) Humanized antibodies that recognize beta amyloid peptide

Legal Events

Date Code Title Description
AS Assignment

Owner name: ELAN PHARMACEUTICALS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SCHENK, DALE B.;YEDNOCK, TED;BASI, GURIQ;REEL/FRAME:014668/0397;SIGNING DATES FROM 20040419 TO 20040422

AS Assignment

Owner name: WYETH, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ELAN PHARMACEUTICALS, INC.;REEL/FRAME:014722/0711

Effective date: 20040512

Owner name: NEURALAB LIMITED, BERMUDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ELAN PHARMACEUTICALS, INC.;REEL/FRAME:014722/0711

Effective date: 20040512

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: ELAN PHARMA INTERNATIONAL LIMITED, IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NEURALAB LIMITED;REEL/FRAME:023385/0706

Effective date: 20060828

Owner name: WYETH, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CRIMAGUA LIMITED;REEL/FRAME:023385/0731

Effective date: 20090914

Owner name: WYETH, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NEURALAB LIMITED;REEL/FRAME:023385/0706

Effective date: 20060828

Owner name: CRIMAGUA LIMITED, IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ELAN PHARMA INTERNATIONAL LIMITED;REEL/FRAME:023385/0713

Effective date: 20090914

Owner name: WYETH, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ELAN PHARMA INTERNATIONAL LIMITED;REEL/FRAME:023385/0713

Effective date: 20090914

Owner name: JANSSEN ALZHEIMER IMMUNOTHERAPY, IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CRIMAGUA LIMITED;REEL/FRAME:023385/0731

Effective date: 20090914

AS Assignment

Owner name: WYETH LLC,NEW JERSEY

Free format text: CHANGE OF NAME;ASSIGNOR:WYETH;REEL/FRAME:024160/0365

Effective date: 20091109

Owner name: WYETH LLC, NEW JERSEY

Free format text: CHANGE OF NAME;ASSIGNOR:WYETH;REEL/FRAME:024160/0365

Effective date: 20091109