WO2006036291A2 - Antibodies directed against amyloid-beta peptide and methods using same - Google Patents

Antibodies directed against amyloid-beta peptide and methods using same Download PDF

Info

Publication number
WO2006036291A2
WO2006036291A2 PCT/US2005/027295 US2005027295W WO2006036291A2 WO 2006036291 A2 WO2006036291 A2 WO 2006036291A2 US 2005027295 W US2005027295 W US 2005027295W WO 2006036291 A2 WO2006036291 A2 WO 2006036291A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
region
peptide
amino acid
seq
Prior art date
Application number
PCT/US2005/027295
Other languages
French (fr)
Other versions
WO2006036291A3 (en
Inventor
Arnon Rosenthal
Jaume Pons
Wei-Hsien Ho
Jan Markus Grimm
Original Assignee
Rinat Neuroscience Corp.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to MX2007000998A priority Critical patent/MX2007000998A/en
Priority to AP2007003890A priority patent/AP2007003890A0/en
Priority to BRPI0513959-7A priority patent/BRPI0513959A/en
Priority to JP2007523885A priority patent/JP5042828B2/en
Priority to CA2575663A priority patent/CA2575663C/en
Priority to UAA200700982A priority patent/UA91683C2/en
Priority to EP05778628A priority patent/EP1781704A2/en
Priority to AU2005290250A priority patent/AU2005290250A1/en
Application filed by Rinat Neuroscience Corp. filed Critical Rinat Neuroscience Corp.
Priority to CN200580025690.5A priority patent/CN101124247B/en
Priority to NZ552480A priority patent/NZ552480A/en
Priority to EA200700083A priority patent/EA016357B1/en
Publication of WO2006036291A2 publication Critical patent/WO2006036291A2/en
Publication of WO2006036291A3 publication Critical patent/WO2006036291A3/en
Priority to IL180364A priority patent/IL180364A/en
Priority to NO20066058A priority patent/NO20066058L/en
Priority to TNP2007000031A priority patent/TNSN07031A1/en
Priority to HK08106860.6A priority patent/HK1116500A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • 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
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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
    • 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
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the invention concerns antibodies to amyloid-beta peptide.
  • the invention further concerns use of such antibodies in the treatment and/or prevention of diseases, such as Alzheimer's disease.
  • AD Alzheimer's disease
  • a ⁇ amyloid beta peptide
  • APP is a type I transmembrane glycoprotein that contains a large ectopic N-terminal domain, a transmembrane domain, and a small cytoplasmic C-terminal tail.
  • Alternative splicing of the transcript of the single APP gene on chromosome 21 results in several isoforms that differ in the number of amino acids.
  • a ⁇ appears to have a central role in the neuropathology of Alzheimer's disease.
  • the present invention provides methods for treating a disease characterized by aberrant deposition of a protein in the brain of a subject.
  • the methods comprise administering to the subject an effective amount of a pharmaceutical composition comprising an antibody that specifically binds to the protein or the protein deposit, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • the invention also provides methods for treating or preventing diseases associated with amyloid deposit of A ⁇ (e.g., deposit in the brain tissue and cerebral vasculature) in a subject, such as Alzheimer's disease, Down's syndrome, multi-infarct dementia, mild cognitive impairment, and cerebral amyloid angiopathy.
  • the method comprises administering to the subject an effective amount of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • the invention also provides methods of delaying development of a symptom associated with diseases associated with amyloid deposit of A ⁇ in a subject, such as Alzheimer's disease, comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • the invention also provides methods of suppressing formation of amyloid plaques and/or amyloid accumulation in a subject comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta- amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • the amyloid plaques are in the brain (brain tissue) of the subject.
  • the amyloid plaques are in the cerebral vasculature.
  • the amyloid accumulation is in the circulatory system.
  • the invention also provides methods of reducing amyloid plaques and/or amyloid accumulation in a subject comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • the amyloid plaques are in the brain (brain tissue) of the subject.
  • the amyloid plaques are in the cerebral vasculature.
  • the amyloid accumulation is in the circulatory system.
  • the invention also provides methods of removing or clearing amyloid plaques and/or amyloid accumulation in a subject comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • the amyloid plaques are in the brain (brain tissue) of the subject, hi some embodiments, the amyloid plaques are in the cerebral vasculature.
  • the amyloid accumulation is in the circulatory system.
  • the invention also provides methods for inhibiting the accumulation of A ⁇ peptide in a tissue comprising contacting the tissue with an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, wherein the antibody has impaired effector function.
  • the invention also provides methods of reducing A ⁇ peptide (such as soluble, oligomeric, and deposited form) in a subject comprising administrating to the subject an effective amount of an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • the accumulation of A ⁇ peptide is inhibited and/or reduced in the brain.
  • the toxic effects of A ⁇ peptide are inhibited and/or reduced.
  • the method of the invention can be used to treat any disease in which accumulation of A ⁇ peptide is present or suspected, such as Alzheimer's disease, Down's syndrome, Parkinson's disease, and multi-infarct dementia.
  • the invention also provides methods of improving cognition or reversing cognitive decline associated with diseases associated with amyloid deposit of A ⁇ in a subject, such as Alzheimer's disease, comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • the invention also provides methods for treating or preventing diseases associated with amyloid deposit of A ⁇ , comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, wherein the antibody comprises an Fc region with a variation from a naturally occurring Fc region, wherein the variation results in impaired effector function.
  • a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, wherein the antibody comprises an Fc region with a variation from a naturally occurring Fc region, wherein the variation results in impaired effector function.
  • the administration of the antibody causes less cerebral microhemorrhage than administration of an antibody without the variation.
  • Polypeptides that specifically bind to an A ⁇ peptide or an aggregated form of an A ⁇ peptide and comprises a heavy chain constant region having impaired effector function may also be used for any of the methods described herein.
  • the polypeptide comprises a sequence (e.g., one or more CDRs) derived from antibody 9TL or its variants shown in Table 3.
  • the polypeptide comprises a sequence (e.g., one or more CDRs) derived from antibody 6G.
  • the antibody and polypeptide used for the methods of the invention specifically bind to an A ⁇ peptide or an aggregated form of an A ⁇ peptide, but have impaired effector function.
  • the antibody or polypeptide is not a F(ab')2 fragment.
  • the antibody or polypeptide is not a Fab fragment.
  • the antibody or polypeptide is not a single chain antibody scFv.
  • the antibody or the polypeptide comprises a heavy chain constant region having impaired effector function, wherein the heavy chain constant region comprises an Fc region, hi some embodiments, the N-glycosylation in the Fc region is removed.
  • the Fc region comprises a mutation within the N-glycosylation recognition sequence, whereby the Fc region of the antibody or polypeptide is not N-glycosylated.
  • the Fc region is PEGylated.
  • the heavy chain constant region of the antibody or the polypeptide is a human heavy chain IgG2a constant region containing the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence).
  • the antibody or the polypeptide comprises a constant region of IgG4 comprising the following mutations: E233F234L235 to P233V234A235.
  • the antibody or polypeptide specifically binds to an epitope within residues 1-16 of A ⁇ peptide. In some embodiments, the antibody or polypeptide specifically binds to the N-terminus of the A ⁇ peptide. In some embodiments, the antibody or the polypeptide specifically binds to an epitope within residues 16-28 of A ⁇ peptide. In some embodiments, the antibody specifically binds to an epitope on the C-terminal side of an A ⁇ peptide, such as an epitope starting from amino acid 25 or later.
  • the antibody may specifically bind to the free C-terminus amino acid of C-terminus truncated A ⁇ peptide, for example, A ⁇ 1- 37, 1-38, 1-39, 1-40, 1-41, 1-42, 1-43.
  • the antibody or the polypeptide specifically binds to an epitope within residues 28-40 of A ⁇ 1-4 o peptide.
  • the antibody or the polypeptide specifically binds to an epitope within residues 28-42 of A ⁇ 1-42 peptide.
  • the antibody or the polypeptide specifically binds to an epitope within residues 28-43 of A ⁇ 1-43 peptide.
  • the antibody or the polypeptide specifically binds to A ⁇ peptide without binding to full-length amyloid precursor protein (APP). In some embodiments, the antibody or the polypeptide specifically binds to the aggregated form of A ⁇ without binding to the soluble form. In some embodiments, the antibody or the polypeptide specifically binds to the soluble form of A ⁇ without binding to the aggregated form. In some embodiments, the antibody or the polypeptide specifically binds to both aggregated form and soluble forms of A ⁇ .
  • APP amyloid precursor protein
  • the antibody or the polypeptide specifically binds to a C- terminal peptide 33-40 of A ⁇ 1-40 . In some embodiments, the antibody or the polypeptide specifically binds to an epitope on A ⁇ 1-40 that includes amino acid 35-40. In some embodiments, the antibody or the polypeptide specifically binds to an epitope on A ⁇ 1-40 that includes amino acid 36-40. In some embodiments, the antibody or the polypeptide specifically binds to an epitope on A ⁇ 1-40 that includes amino acid 39 and/or 40. In some embodiments, the antibody or the polypeptide specifically binds to A ⁇ 1-40 but do not specifically bind to A ⁇ 1-42 and/or Ap 1-43 .
  • the antibody comprises the variable region of antibody 9TL or an antibody derived from 9TL described herein. In some embodiments, the antibody or polypeptide competitively inhibits ⁇ binding of antibody 9TL and/or antibody or polypeptide derived from 9TL to A ⁇ 1-40 .
  • the antibody or the polypeptide binds to A ⁇ 1-4 o with higher affinity than its binding to A ⁇ 1-42 and A ⁇ 1-43 . In some embodiments, the antibody binds to an epitope on A ⁇ 1-40 that includes amino acids 25-34 and 40. In some embodiments, the antibody comprises the variable region of antibody 6G or an antibody derived from 6G described herein. In some embodiments, the antibody or polypeptide competitively inhibits binding of antibody 6G and/or antibody or polypeptide derived from 6G to A ⁇ .
  • Administration of antibody or polypeptide that specifically binds to an A ⁇ peptide and has impaired effector function may be by any means known in the art, including: intravenously, subcutaneously, via inhalation, intraarterially, intramuscularly, intracardially, intraventricularly, parenteral, intrathecally, and intraperitoneally. Administration may be systemic, e.g. intravenously, or localized. This also generally applies to polypeptides and polynucleotides of the invention.
  • the invention also provides pharmaceutical composition comprising an effective amount of any of the antibodies or polypeptides that specifically bind to an A ⁇ peptide or an aggregated form of an A ⁇ peptide and have impaired effector function, or polynucleotides encoding the antibodies or polypeptides, and a pharmaceutical acceptable excipient.
  • the invention also provides kits and compositions comprising any one or more of the compositions comprising an effective amount of any of the antibodies or polypeptides that specifically bind to an A ⁇ peptide or an aggregated form of an A ⁇ peptide and have impaired effector function, or polynucleotides encoding the antibodies or polypeptides. These kits, generally in suitable packaging and provided with appropriate instructions, are useful for any of the methods described herein.
  • the invention also provides a method of producing a therapeutic humanized antibody for treatment of a disease associated with amyloid deposits of A ⁇ peptide in the brain of a human subject, comprising selecting a first humanized antibody that specifically binds to A ⁇ peptide; and altering the Fc region of the antibody to provide a therapeutic humanized antibody having impaired effector function relative to the first humanized antibody.
  • the invention disclosed herein concerns antibodies that bind to C-terminus of A ⁇ i.
  • the invention is an antibody 9TL (interchangeably termed "9TL") that is produced by expression vectors having ATCC Accession Nos. PTA-6124 and PTA-6125.
  • the amino acid sequences of the heavy chain and light chain variable regions of 9TL are shown in Figure 1.
  • the complementarity determining region (CDR) portions of antibody 9TL are also shown in Figure 1. It is understood that reference to any part of or entire region of 9TL encompasses sequences produced by the expression vectors having ATCC Accession Nos. PTA-6124 and PTA- 6125, and/or the sequences depicted in Figure 1.
  • the invention also provides antibody variants of 9TL with amino acid sequences depicted in Table 3.
  • the invention is an antibody comprising a fragment or a region of the antibody 9TL or its variants shown in Table 3.
  • the fragment is a light chain of the antibody 9TL.
  • the fragment is a heavy chain of the antibody 9TL.
  • the fragment contains one or more variable regions from a light chain and/or a heavy chain of the antibody 9TL.
  • the fragment contains one or more variable regions from a light chain and/or a heavy chain shown in Figure 1.
  • the fragment contains one or more CDRs from a light chain and/or a heavy chain of the antibody 9TL.
  • polypeptides (which may or may not be an antibody) comprising any one or more of the following: a) one or more CDR(s) of antibody 9TL or its variants shown in Table 3; b) CDR H3 from the heavy chain of antibody 9TL or its variants shown in Table 3; c) CDR L3 from the light chain of antibody 9TL or its variants shown in Table 3; d) three CDRs from the light chain of antibody 9TL or its variants shown in Table 3; e) three CDRs from the heavy chain of antibody 9TL or its variants shown in Table 3; f) three CDRs from the light chain and three CDRs from the heavy chain of antibody 9TL or its variants shown in Table 3.
  • the invention further provides polypeptides (which may or may not be an antibody) comprising any one or more of the following: a) one or more (one, two , three, four, five, or six) CDR(s) derived from antibody 9TL or its variants shown in Table 3; b) a CDR derived from, CDR H3 from the heavy chain of antibody 9TL; and/or c) a CDR derived from CDR L3 from the light chain of antibody 9TL.
  • the CDR is a CDR shown in Figure 1.
  • the one or more CDRs derived from antibody 9TL or its variants shown in Table 3 are at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to at least one, at least two, at least three, at least four, at least five, or at least six CDRs of 9TL or its variants.
  • the CDR is a Kabat CDR. In other embodiments, the CDR is a Chothia CDR. In other embodiments, the CDR is a combination of a Kabat and a Chothia CDR (also termed “combined CDR" or "extended CDR"). In other words, for any given embodiment containing more than one CDR, the CDRs may be any of Kabat, Chothia, and/or combined.
  • the polypeptide (such as an antibody) comprises an amino acid sequence shown in SEQ ID NO:5, wherein Ll is L, V, or I; wherein Y2 is Y or W; wherein S3 is S, T, or: G; wherein L4 is L, R, A, V, S, T, Q, or E; wherein V6 is V, I, T, P, C, Q, S, N, or F; and wherein Y7 is Y, H, F, W, S, I, V, or A.
  • the amino acid sequence is a CDR3 in a heavy chain variable region.
  • amino acid refers to choices of amino acid(s) for a given position with reference to the position in the SEQ ID.
  • Ll is L, V, or I
  • amino acid L at position 1 in SEQ ID NO: 5 may be substituted with V or I.
  • the polypeptide (such as an antibody) comprises an amino acid sequence shown in SEQ ID NO: 6, wherein Y8 is Y, A, or H; and wherein Al l is A or S; and wherein Kl 2- is K or A.
  • the amino acid sequence is a CDRl in a light chain variable region.
  • the polypeptide (such as an antibody) comprises an amino acid sequence shown in SEQ ID NO:8, wherein Ll is L, M, N, C, F, V, K, S, Q, G, S; wherein G3 is G, S, or T; wherein T4 is T or S; wherein H5 is H or L; wherein Y6 is Y, P, A, W, Q, M, S, or E; wherein V8 is V, L, K, H, T, A, E, or M; and wherein L9 is L, I 5 T, S, or V.
  • the amino acid sequence is a CDR3 in a light chain variable region.
  • the polypeptide (such as an antibody) comprises a heavy chain variable region comprising (a) a CDRl region shown in SEQ ID NO:3; (b) a CDR2 region shown in SEQ ID NO:4; and (c) a CDR3 region shown in SEQ ID NO:5, wherein Ll is L, V, or I; wherein " Y2 is Y or W; wherein S3 is S, T, or G; wherein L4 is L, R, A, V, S, T, Q, or E; wherein V6 is V, I, T, P, C, Q, S 3 N, or F; and wherein Y7 is Y, H, F, W, S, I, V, or A.
  • the polypeptide (such as an antibody) comprises a light chain variable region comprising (a) a CDRl region shown in SEQ ID NO:6, wherein Y8 is Y, A, or H; and wherein Al 1 is A or S; and wherein Kl 2 is K or A; (b) a CDR2 region shown in SEQ ID NO:7; and (c) a CDR3 region shown in SEQ ID NO:8, wherein Ll is L, M, N, C, F, V, K, S, Q, G, S; wherein G3 is G, S, or T; wherein T4 is T or S; wherein H5 is H or L; wherein Y6 is Y, P, A 5 W 5 Q, M, S 5 or E; wherein V8 is V 5 L 5 K 5 H 5 T 5 A 5 E 5 or M; and wherein L9 is L 5 1 5 T 5 S 5 or V.
  • the antibody of the invention is a human antibody. In other embodiments, the antibody of the invention is a humanized antibody. In some embodiments, the antibody is monoclonal. In some embodiments, the antibody (or polypeptide) is isolated. In some embodiments, the antibody (or polypeptide) is substantially pure. [0040] I The heavy chain constant region of the antibodies may be from any types of constant region, such as IgG 5 IgM 5 IgD 5 IgA 5 and IgE; and any isotypes, such as IgGl, IgG2, IgG3, and IgG4.
  • the antibody comprises a modified constant region, such as a constant region that is immunologically inert (which includes partially immunologically inert, and is used interchangeably with the term "having impaired effector function"), e.g. 1 , does not trigger complement mediated lysis, does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC), or does not activate microglia.
  • the constant region is modified as described in Eur. J. Immunol. (1999) 29:2613-2624; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No. 9809951.8.
  • the antibody comprises a human heavy chain IgG2a constant region comprising the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence). Eur. J. Immunol. (1999) 29:2613-2624.
  • the antibody comprises a constant region of IgG4 comprising the following mutations: E233F234L235 to P233V234A235.
  • the constant region is aglycosylated for N-linked glycosylation.
  • the constant region is aglycosylated for N-linked glycosylation by mutating the oligosaccharide attachment residue (such as Asn297) and/or flanking residues that are part of the N-glycosylation recognition sequence in the constant region .
  • the constant region is aglycosylated for N-linked glycosylation.
  • the constant region may be aglycosylated for N-linked glycosylation enzymatically or by expression in a glycosylation deficient host cell.
  • the invention provides a polynucleotide (which may be isolated) comprising a polynucleotide encoding a fragment or a region of the antibody 9TL or its variants shown in Table 3.
  • the fragment is a light chain of the antibody 9TL.
  • the fragment is a heavy chain of the antibody 9TL.
  • the fragment contains one or more variable regions from a light chain and/or a heavy chain of the antibody 9TL.
  • the fragment contains one or more (i.e., one, two, three, four, five, six) complementarity determining regions (CDRs) from a light chain and/or a heavy chain of the antibody 9TL.
  • CDRs complementarity determining regions
  • the invention is a polynucleotide (which may be isolated) comprising a polynucleotide that encodes for antibody 9TL or its variants shown in Table 3.
  • the polynucleotide comprises either or both of the polynucleotides shown in SEQ ID NO:9 and SEQ ID NO: 10.
  • the invention provides polynucleotides encoding any of the antibodies (including antibody fragments) or polypeptides described herein.
  • the invention provides vectors (including expression and cloning vectors) and host cells comprising any of the polynucleotide disclosed herein.
  • the vector is ⁇ Db.9TL.hFc2a having ATCC No. PTA-6124.
  • the vector is pEb.9TL.hK having ATCC No. PTA-6125.
  • the invention is a host cell comprising a polynucleotide encoding any of the antibodies described herein.
  • the invention is a complex of A ⁇ i- 40 bound by antibody 9TL or its variants shown in Table 3.
  • the invention is a complex of Ap 1-40 bound by any of the antibodies or polypeptides described herein.
  • the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of any of the polypeptides (including antibodies, such as an antibody comprising one or more CDRs of antibody 9TL) or polynucleotides described herein, and a pharmaceutically acceptable excipient.
  • the invention is a method of generating antibody 9TL comprising culturing a host cell or progeny thereof under conditions that allow production of antibody 9TL, wherein the host cell comprises an expression vector that encodes for antibody
  • the expression vector comprises one or both of the polynucleotide sequences shown in SEQ ID NO: 9 and SEQ ID NO: 10.
  • the invention provides methods of generating any of the antibodies or polypeptides described herein by expressing one or more polynucleotides encoding the antibody (which may be separately expressed as a single light or heavy chain, or both a light and a heavy chain are expressed from one vector) or the polypeptide in a suitable cell, generally followed by recovering and/or isolating the antibody or polypeptides of interest.
  • the invention also provides a method for preventing, treating, inhibiting, or delaying the development of Alzheimer's disease and other diseases associated with altered A ⁇ or ⁇ APP expression, or accumulation of A ⁇ peptide, such as Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, and AIDS.
  • the method comprises administering an effective dosage a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to a subject.
  • the invention also provides a method of delaying development of a symptom associated with Alzheimer's disease or other diseases related to accumulation of A ⁇ peptide in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject.
  • the invention also provides a method of suppressing formation of amyloid plaques and/or amyloid accumulation in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject.
  • a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject.
  • the amyloid plaques are in the brain (brain tissue) of the subject.
  • the amyloid plaques are in the cerebral vasculature.
  • the amyloid accumulation is in the circulatory system.
  • the invention also provides a method of reducing amyloid plaques and/or amyloid accumulation in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject.
  • a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject.
  • the amyloid plaques are in the brain (brain tissue) of the subject, hi some embodiments, the amyloid plaques are in the cerebral vasculature, hi other embodiments, the amyloid accumulation is in the circulatory system.
  • the invention also provides a method of removing or clearing amyloid plaques and/or amyloid accumulation in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject.
  • a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject.
  • the amyloid plaques are in the brain (brain tissue) of the subject. Li some embodiments, the amyloid plaques are in the cerebral vasculature, hi other embodiments, the amyloid accumulation is in the circulatory system.
  • the invention provides a method for inhibiting the accumulation of A ⁇ peptide in a tissue comprising contacting the tissue with an anti ⁇ body or a polypeptide of the invention.
  • the invention also provides a method of reducing A ⁇ peptide (such as soluble, oligomeric and deposited form) in the brain of an individual comprising administering to the individual an effective amount of an antibody or a polypeptide of the invention.
  • a ⁇ peptide such as soluble, oligomeric and deposited form
  • the accumulation of A ⁇ peptide is inhibited and/or reduced in the brain.
  • the toxic effects of A ⁇ peptide are inhibited and/or reduced.
  • the method of the invention can be used to treat any disease in which accumulation of A ⁇ peptide is present or suspected, such as Alzheimer's disease, Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, and cerebral amyloid angiopathy.
  • the invention also provides methods of improving cognition or reversing cognitive decline associated with diseases associated with amyloid deposit of A ⁇ in the brain of an individual, such as Alzheimer's disease, comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the individual.
  • any antibodies, polypeptides, or polynucleotides described herein may be used for the methods of the invention.
  • the antibody is antibody 9TL.
  • Antibodies and polypeptides of the invention can further be used in the detection, diagnosis and monitoring of Alzheimer's disease and other diseases associated with altered A ⁇ or ⁇ APP expression, such as Down's syndrome, and AIDS.
  • the method comprises contacting a specimen of a patient suspected of having altered A ⁇ or ⁇ APP expression with an antibody of the invention and determining whether the level of A ⁇ or ⁇ APP differs from that of a control or comparison specimen.
  • serum level of A ⁇ is measured before and after administration of an anti-A ⁇ antibody; and any increase of serum level of A ⁇ is assessed.
  • Administration of any antibody or polypeptide of the invention may be by any means known in the art, including: intravenously, subcutaneously, via inhalation, intraarterially, intramuscularly, intracardially, intraventricularly, parenteral, intrathecally, and intraperitoneally. Administration may be systemic, e.g. intravenously, or localized. This also generally applies to polypeptides and polynucleotides of the invention.
  • kits and compositions comprising any one or more of the compositions described herein. These kits, generally in suitable packaging and provided with appropriate instructions, are useful for any of the methods described herein.
  • Figure 1 shows the amino acid sequence of the heavy chain variable region (SEQ ID NO:1) and light chain variable region (SEQ ID NO:2) of the 9TL antibody.
  • the Kabat CDRs are in bold text, and the Chothia CDRs are underlined.
  • the amino acid residu.es for the heavy chain and light chain variable region are numbered sequentially.
  • Figure 2 shows epitope mapping of antibody 9TL by peptide competition. A ⁇ M o peptide was immobilized on the SA chip.
  • Monoclonal antibody 2289 and 9TL Fab fragment (50 nM each), each of which was preincubated for 1 h with 10 ⁇ M various peptide (amino acids 28- 40, 1-40, 1-28, 28-42, 22-35, 1-16, 1-43, 33-40, 1-38, or 17-40 of A ⁇ ) or no peptide, and was then flowed onto the chip. Binding of the antibody Fab fragment to immobilized A ⁇ 1-40 peptide was measured.
  • FIG. 3 is a graph showing epitope mapping of antibody 2H6 by peptide competition.
  • a ⁇ 1-40 peptide was immobilized on the SA chip.
  • Monoclonal antibody 2289, 2286, or 2H6 (100 nM each), each of which was preincubated for 1 h with 16 ⁇ M various peptide (amino acids 1-16, 1-28, 1-38, 1-40, 1-42, 1-43, 17-40, 17-42, 22-35, 25-35, or 33-40 of A ⁇ ) or no peptide, was flowed onto the chip. Binding of the antibody to immobilized A ⁇ 1-40 peptide was measured.
  • Figure 4 is a graph showing binding of antibody 2H6, 2286, and 2289 to different A ⁇ peptide C-terminal variants.
  • GST-A ⁇ variants M35A, V36A, G37A, G38A, V39A, or V40A
  • GST-A ⁇ peptide 1-39, 1-41, 1-40, 1-42 were immobilized on ELISA plate.
  • Monoclonal antibody 2286, 2H6, or 2289 (0.3 nM each niAb) was incubated with each of the immobilized peptides, and their binding was detected by further incubating with biotinylated anti-mouse IgG (H+L) and followed by Sterptavidin-HRP.
  • Figure 5 is a graph showing spatial learning deficits in APP -transgenic mice were reversed following 16 weeks of antibody treatment with 2H6 and deglycosylated 2H6. Mice were tested in a two-day version of the radial-arm water maze. Y axis represents mean number of errors made over the 2-day trial period. Block numbers 1-5 represent tests in day 1 ; and block numbers 6-10 represent tests in day 2. "*" indicates p ⁇ 0.05 for both 2H6 (A-2H6) and deglycosylated 2H6 (A-De-2H6) treated mice when compared with anti-AMN antibody treated mice (A-AMN).
  • Figure 6A and 6B are graphs showing decreases of parenchymal Congo-red stained amyloid-beta peptide in hippocampus (Fig.6B) and frontal cortex (Fig. 6A) after 16 weeks of antibody treatment with 2H6, anti-AMN (referred to as AMN), and deglycosylated 2H6 (referred to as D-2H6) antibody.
  • Y-axes in Fig. 6A and 6B represent mean of percent area positive for Congo-red staining.
  • FIG. 6A and 6B represent type of antibody administered.
  • Figure 7A and 7B are graphs showing increases of vascular Congo-red stained amyloid-beta peptide in hippocampus (Fig. 7A) and frontal cortex (Fig. 7B) after 16 weeks of antibody treatment with 2H6, anti-AMN (referred to as AMN), and deglycosylated 2H6 (referred to as D-2H6) antibody.
  • Y-axes in Fig.7A and 7B represent mean of percent area positive for Congo-red staining.
  • X-axes in Fig. 7A and 7B represent type of antibody administered.
  • Figure 8 is a graph showing number of Prussian blue positive profiles after 16 weeks of antibody treatment with 2H6, anti-AMN (referred to as AMN), and deglycosylated 2H6 (referred to as D-2H6) antibody.
  • Y-axis represents positive profiles per section.
  • X-axis represents type of antibody administered.
  • FIG. 9 is a graph showing serum level of A ⁇ peptide after administration of anti- AMN antibody (referred to as AMN) 5 antibody 2H6 (referred to as 2H6), deglycosylated 2H6 (referred to as 2H6-D) in APP Tg2576 mice, and after administration of anti-AMN antibody and antibody 2H6 in wild type (WT) mice.
  • AMN anti- AMN antibody
  • 2H6-D deglycosylated 2H6
  • WT wild type mice.
  • Figure 10 shows immunostaining of CD45 in the hippocampus of a mouse after intracranial administration of 2H6 antibody (A) or deglycosylated 2H6 antibody (B).
  • the bottom panel shows that the ratio of the average area occupied CD45 positive staining of injected side over uninjected side in the frontal cortex and hippocampus after intracranial administration of the control antibody, 2H6 antibody, or deglycosylated 2H6 antibody.
  • "**" indicates P ⁇ 0.01 as compared to the control antibody.
  • Figure 11 shows immunostaining of Fc ⁇ receptor in the hippocampus of a mouse after intracranial administration of 2H6 antibody (A) or deglycosylated 2H6 antibody (B).
  • the bottom panel shows that the ratio of the average area occupied by Fc ⁇ receptor positive staining of injected side over uninjected side in the frontal cortex and hippocampus after intracranial administration of the control antibody, 2H6 antibody, or deglycosylated 2H6 antibody.
  • "**" indicates P ⁇ 0.01 as compared to the control antibody.
  • Figure 12 shows immunostaining of A ⁇ peptide in the hippocampus of a mouse after intracranial administration of 2H6 antibody (A) or deglycosylated 2H6 antibody (B).
  • the bottom panel shows that the ratio of the average area occupied by A ⁇ positive staining of injected side over uninjected side in the frontal cortex and hippocampus after intracranial administration of the control antibody, 2H6 antibody, or deglycosylated 2H6 antibody.
  • "**" indicates PO.01 as compared to the control antibody.
  • “*” indicates PO.05 as compared to the control antibody.
  • Figure 13 shows thioflavine-S in the hippocampus of a mouse after intracranial administration of 2H6 antibody (A) or deglycosylated 2H6 antibody (B).
  • the bottom panel shows that the ratio of the average area occupied by thioflavine-S positive staining of injected side over uninjected side in the frontal cortex and hippocampus after intracranial administration of the control antibody, 2H6 antibody, or deglycosylated 2H6 antibody.
  • "*" indicates P ⁇ 0.05 as compared to the control antibody.
  • Figure 14 shows epitope mapping of antibody 2294 and 6G by ELISA.
  • Various A ⁇ peptides were immobilized on ELISA plates. Antibodies were incubated for 1 hour with various immobilized peptides.
  • Antibody 6G bound to immobilized A ⁇ peptides were measured using goat anti-human kappa HRP conjugated secondary antibody.
  • Antibody 2294 bound to immobilized A ⁇ peptides were measured using goat anti-mouse that binds to both heavy and light chain and is HRP conjugated secondary antibody.
  • "NB" refers to no binding detected.
  • the numbers in the columns under "2294" and "6G” represent absorbance at 450 nm.
  • the invention disclosed herein provides antibodies and polypeptides that bind to
  • the invention also provides methods of making and using these antibodies. In some embodiments, the invention provides antibody 9TL, and methods of making and using this antibody. The invention also provides 9TL polypeptides (including antibodies) that bind A ⁇ 1-4 o, and polynucleotides encoding 9TL antibody and/or polypeptide.
  • the invention disclosed herein also provides methods for preventing and/or treating A ⁇ -associated diseases, such as Alzheimer's disease, Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, vascular disorder caused by deposit of A ⁇ peptide in blood vessels (such as stroke and HCHWA- D) in an individual by administration of a therapeutically effective amount of an antibody 9TL, or antibody or polypeptide derived from 9TL.
  • a ⁇ -associated diseases such as Alzheimer's disease, Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, vascular disorder caused by deposit of A ⁇ peptide in blood vessels (such as stroke and HCHWA- D) in an individual by administration of a therapeutically effective amount of an antibody 9TL, or antibody or polypeptide derived from 9TL.
  • the invention also provides methods for treating or preventing diseases associated with ⁇ -amyloid deposit in an individual, such as Alzheimer's disease, Down's syndrome, multi- infarct dementia, mild cognitive impairment, and cerebral amyloid angiopathy in an individual by administering to the individual an effective amount of a pharmaceutical composition comprising an antibody or a polypeptide that specifically binds to a ⁇ -amyloid peptide or an aggregated form of an A ⁇ peptide, or a polynucleotide encoding the antibody or the polypeptide, wherein the antibody or the polypeptide has impaired effector function.
  • diseases associated with ⁇ -amyloid deposit in an individual such as Alzheimer's disease, Down's syndrome, multi- infarct dementia, mild cognitive impairment, and cerebral amyloid angiopathy
  • an "antibody” is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule.
  • a target such as a carbohydrate, polynucleotide, lipid, polypeptide, etc.
  • the term encompasses not only intact polyclonal or monoclonal antibodies, but also fragments thereof (such as Fab, Fab', F(ab') 2 , Fv), single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site.
  • An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three- dimensional configurations of different classes of immunoglobulins are well known.
  • "monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler and Milstein, 1975, Nature, 256:495, or may be made by recombinant DNA methods such as described in U.S. Pat. No. 4,816,567.
  • the monoclonal antibodies may also be isolated from phage libraries generated using the techniques described in McCafferty et al., 1990, Nature, 348:552-554, for example.
  • humanized antibodies refer to forms of non-human (e.g. murine) antibodies that are specific chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity.
  • CDR complementary determining region
  • Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
  • Antibodies may have Fc regions modified as described in WO 99/58572.
  • Other forms of humanized antibodies have one or more CDRs (one, two, three, four, five, six) which are altered with respect to the original antibody, which are also termed one or more CDRs "derived from” one or more CDRs from the original antibody.
  • "human antibody” means an antibody having an amino acid sequence corresponding to that of an antibody produced by a human and/or has been made using any of the techniques for making human antibodies known in the art or disclosed herein.
  • This definition of a human antibody includes antibodies comprising at least one human heavy chain polypeptide or at least one human light chain polypeptide.
  • One such example is an antibody comprising murine light chain and human heavy chain polypeptides.
  • Human antibodies can be produced using various techniques known in the art.
  • the human antibody is selected from a phage library, where that phage library expresses human antibodies (Vaughan et al., 1996, Nature Biotechnology, 14:309-314; Sheets et al., 1998, PNAS, (USA) 95:6157-6162; Hoogenboom and Winter, 1991, J. MoI. Biol., 227:381; Marks et al., 1991, J. MoI. Biol., 222:581).
  • Human antibodies can also be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated.
  • the human antibody may be prepared by immortalizing human B lymphocytes that produce an antibody directed against a target antigen (such B lymphocytes may be recovered from an individual or may have been immunized in vitro). See, e.g., Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985); Boerner et al., 1991, J. Immunol., 147 (l):86-95; and U.S. Patent No. 5,750,373.
  • the terms “9TL” and “antibody 9TL” are used interchangeably to refer to an antibody produced by expression vectors having deposit numbers of ATCC PTA-6124 and ATCC PTA-6125.
  • the amino acid sequence of the heavy chain and light chain variable regions are shovm in Figure 1.
  • the CDR portions of antibody 9TL (including Chothia and Kabat CDRs) are diagrammatically depicted in Figure 1.
  • the polynucleotides encoding the heavy and light chain variable regions are shown in SEQ ID NO:9 and SEQ ID NO: 10. The characterization of 9TL is described in the Examples.
  • polypeptide oligopeptide
  • peptide protein
  • polymers of amino acids of any length may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non- amino acids.
  • Tlie terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • Polynucleotide or “nucleic acid,” as used interchangeably herein, refer to polymers of nucleotides of any length, and include DNA and RNA.
  • the nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase.
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs.
  • modification to the nucleotide structure may be imparted before or after assembly of the polymer.
  • the sequence of nucleotides may be interrupted by non-nucleotide components.
  • a polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.
  • modifications include, for example, "caps", substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, cabamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, ply-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as
  • any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonateigroups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports.
  • the 5' and 3' terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from 1 to 20 carbon atoms.
  • Other hydroxy Is may also be derivatized to standard protecting groups.
  • Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2' ⁇ O-methyl-, T- O-allyl, 2'-fluoro- or 2'-azido-ribose, carbocyclic sugar analogs, ⁇ -anomeric sugars, epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuloses, acyclic analogs and abasic nucleoside analogs such as methyl riboside.
  • One or more phosphodiester linkages may be replaced by alternative linking groups.
  • linking groups include, but are not limited to, embodiments wherein phosphate is replaced by P(O)S("thioate"), P(S)S ("dithioate"), "(O)NR 2 ("amidate"), P(O)R, P(O)OR', CO or CH 2 ("formacetal"), in which each R or R' is independently H or substituted or unsubstituted alkyl (1- 20 C) optionally containing an ether (-O-) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
  • variable region of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination.
  • the variable regions of the heavy and light chain each consist of four framework regions (FR) connected by three complementarity determining regions (CDRs) also known as hypervariable regions.
  • the CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies.
  • There are at least two techniques for determining CDRs (1) an approach based on cross-species sequence variability (i.e., Kabat et al.
  • a CDR may refer to CDRs defined by either approach or by a combination of both approaches.
  • a "constant region" of an antibody refers to the constant region of the antibody light chain or the constant region of the antibody heavy chain, either alone or in combination.
  • An epitope that "preferentially binds” or “specifically binds” (used interchangeably herein) to an antibody or a polypeptide is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art.
  • a molecule is said to exhibit "specific binding” or “preferential binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances.
  • an antibody “specifically binds” or “preferentially binds” to a target if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances.
  • an antibody that specifically or preferentially binds to an A ⁇ 1-40 epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other A ⁇ 1-40 epitopes or nbn-A ⁇ 1-4 o epitopes. It is also understood by reading this definition that, for example, an antibody (or moiety or epitope) that specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target.
  • binding does not necessarily require (although it can include) exclusive binding. Generally, but not necessarily, reference to binding means preferential binding. [0092] As used herein, “substantially pure” refers to material which is at least 50% pure
  • a "host cell” includes an individual cell or cell culture that can be or has been a recipient for vector(s) for incorporation of polynucleotide inserts.
  • Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation.
  • a host cell includes cells transfected in vivo with a polynucleotide(s) of this invention.
  • the term "Fc region” is used to define a C-terminal region of an immunoglobulin heavy chain.
  • the "Fc region” may be a native sequence Fc region or a variant Fc region.
  • the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof.
  • the numbering of the residues in the Fc region is that of the EU index as in Kabat. Kabat et al., Sequences of Proteins of Imunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991.
  • the Fc region of an immunoglobulin generally comprises two constant domains, CH2 and CH3.
  • Fc receptor and “FcR” describe a receptor that binds to the Fc region of an antibody.
  • the preferred FcR is a native sequence human FcR.
  • a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the Fc ⁇ RI, Fc ⁇ RJI, and Fc ⁇ RIII subclasses, including allelic variants and alternatively spliced forms of these receptors.
  • Fc ⁇ RII receptors include Fc ⁇ RIIA (an "activating receptor") and Fc ⁇ RIIB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • FcRs are reviewed in Ravetch and Kinet, 1991, Ann. Rev. Immunol., 9:457-92; Capel et al., 1994, Immunomethods, 4:25-34; and de Haas et al., 1995, J. Lab. Clin. Med., 126:330-41.
  • FcR also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., 1976, J. Immunol., 117:587; and Kim et al., 1994, J. Immunol., 24:249).
  • Complement dependent cytotoxicity and “CDC” refer to the lysing of a target in the presence of complement.
  • the complement activation pathway is initiated by th.e binding of the first component of the complement system (CIq) to a molecule (e.g. an antibody) complexed with a cognate antigen.
  • a CDC assay e.g. as described in Gazzano-Santoro et al., J. Immunol. Methods, 202:163 (1996), may be performed.
  • a "functional Fc region” possesses at least one effector function of a native sequence Fc region.
  • effector functions include CIq binding; complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down-regulation of cell surface receptors (e.g. B cell receptor; BCR), etc.
  • Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
  • a “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature.
  • a “variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one amino acid modification, yet retains at least one effector function of the native sequence Fc region.
  • the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide.
  • the variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% sequence identity therewith, more preferably at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% sequence identity therewith.
  • antibody-dependent cell-mediated cytotoxicity and "ADCC” refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (FcRs) (e.g. natural killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell.
  • FcRs Fc receptors
  • ADCC activity of a molecule of interest can be assessed using an in vitro ADCC assay, such as that described in U.S. Patent No. 5,500,362 or 5,821,337.
  • Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and NK cells.
  • PBMC peripheral blood mononuclear cells
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al., 1998, PNAS (USA), 95:652-656.
  • an "effective dosage” or “effective amount” drug, compound, or pharmaceutical composition is an amount sufficient to effect beneficial or desired results.
  • beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the outset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • beneficial or desired results include clinical results such as inhibiting, suppressing or reducing the formation of amyloid plaques, reducing, removing, clearing amyloid plaques, improving cognition, reversing or slowing cognitive decline, sequestering or increasing soluble A ⁇ peptide circulating in biological fluids, decreasing one or more symptoms resulting from the disease (biochemical, histological and/or behavioral), including its complications and intermediate pathological phenotypes presenting during development of the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication, delaying the progression of the disease, and/or prolonging survival of patients.
  • An effective dosage can be administered in one or more administrations.
  • an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
  • an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition.
  • an "effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • treatment or “treating” is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: inhibiting, suppressing or reducing the formation of amyloid plaques, reducing, removing, or clearing amyloid plaques, improving cognition, reversing or slowing cognitive decline, sequestering soluble A ⁇ peptide circulating in biological fluids, reducing A ⁇ peptide (including soluble, oligomeric and deposited) in a tissue (such as brain), inhibiting, slowing and/or reducing accumulation of A ⁇ peptide in the brain, inhibiting, slowing and/or reducing toxic effects of A ⁇ peptide in a tissue (such as brain), decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of patients.
  • Alzheimer's disease means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease.
  • a method that "delays" development of Alzheimer's disease is a method that reduces probability of disease development in a given time frame and/or reduces extent of the disease in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a statistically significant number of subjects.
  • Alzheimer's disease means the onset and/or progression of
  • Alzheimer's disease within an individual Alzheimer's disease development can be detectable using standard clinical techniques as described herein. However, development also refers to disease progression that may be initially undetectable. For purposes of this invention, progression refers to the biological course of the disease state, in this case, as determined by a standard neurological examination, patient interview, or may be determined by more specialized testing. A variety of these diagnostic tests include, but not limited to, neuroimaging, detecting alterations of levels of specific proteins in the serum or cerebrospinal fluid (e.g., amyloid peptides and Tau), computerized tomography (CT), and magnetic resonance imaging (MRI).
  • “Development” includes occurrence, recurrence, and onset. As used herein "onset” or "occurrence” of Alzheimer's disease includes initial onset and and/or recurrence.
  • administration "in conjunction” includes simultaneous administration and/or administration at different times. Administration in conjunction also encompasses administration as a co-formulation or administration as separate compositions. As used herein, administration in conjunction is meant to encompass any circumstance wherein an anti-A ⁇ antibody and another agent are administered to an individual, which can occur simultaneously and/or separately. As further discussed herein, it is understood that an anti-A ⁇ antibody and the other agent can be administered at different dosing frequencies or intervals. For example, an anti-A ⁇ antibody can be administered weekly, while the other agent can be administered less frequently. It is understood that the anti-A ⁇ antibody and the other agent can be administered using the same route of administration or different routes of administration.
  • a "biological sample” encompasses a variety of sample types obtained from an individual and can be used in a diagnostic or monitoring assay.
  • the definition encompasses blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom, and the progeny thereof.
  • the definition also includes samples that have been manipulated in any way after their procurement, such as by treatment with reagents, solubilization, or enrichment for certain components, such as proteins or polynucleotides, or embedding in a semi-solid or solid matrix for sectioning purposes.
  • the term "biological sample” encompasses a clinical sample, and also includes ' cells in culture, cell supernatants, cell lysates, serum, plasma, biological fluid, and tissue samples.
  • An "individual” is a mammal, more preferably a human. Mammals also include, but are not limited to, farm animals (such as cows), sport animals, pets (such as cats, dogs, horses), primates, mice and rats.
  • "vector” means a construct, which is capable of delivering, and preferably expressing, one or more gene(s) or sequence(s) of interest in a host cell.
  • vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells, such as producer cells.
  • expression control sequence means a nucleic acid sequence that directs transcription of a nucleic acid.
  • An expression control sequence can be a promoter, such as a constitutive or an inducible promoter, or an enhancer.
  • the expression control sequence is operably linked to the nucleic acid sequence to be transcribed.
  • pharmaceutically acceptable carrier includes any material which, when combined with an active ingredient, allows the ingredient to retain biological activity and is non-reactive with the subject's immune system. Examples include, but are not limited to, any of the standard pharmaceutical carriers such as a phosphate buffered saline solution, water, emulsions such as oil/water emulsion, and various types of wetting agents.
  • Preferred diluents for aerosol or parenteral administration are phosphate buffered saline or normal (0.9%) saline.
  • Compositions comprising such carriers are formulated by well known conventional methods (see, for example, Remington's Pharmaceutical Sciences, 18th edition,
  • W is intended to refer to the on rate constant for association of an antibody to an antigen.
  • k off is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.
  • K 0 is intended to refer to the equilibrium dissociation constant of an antibody-antigen interaction.
  • compositions including pharmaceutical compositions, comprising antibody 9TL and its variants shown in Table 3 or polypeptide derived from antibody 9TL and its variants shown in Table 3; and polynucleotides comprising sequences encoding 9TL antibody and its variants or the polypeptide.
  • compositions comprise one or more antibodies or polypeptides (which may or may not be an antibody) that bind to C-terminus of A ⁇ 1-40 , and/or one or more polynucleotides comprising sequences encoding one or more antibodies or polypeptides that bind to C-terminus of A ⁇ i -4 o.
  • compositions may further comprise suitable excipients, such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
  • suitable excipients such as pharmaceutically acceptable excipients including buffers, which are well known in the art.
  • the antibodies and polypeptides of the invention are characterized by any (one or more) of the following characteristics: (a) binds to C-terminal peptide 28-40 of A ⁇ 1-40 , but does not significantly bind to A ⁇ l-42 or A ⁇ l-43; (b) binds to C-terminal peptide 33-40 of A ⁇ 1-40 ; (c) suppresses formation of amyloid plaques in a subject; (d) reduces amyloid plaques in a subject; (e) treats, prevents, ameliorates one or more symptoms of Alzheimer's disease; (f) improves cognitive function.
  • compositions of the invention may not cause significant or unacceptable levels of any one or more of: bleeding in the brain vasculature (cerebral hemorrhage); meningoencephalitis (including changing magnetic resonance scan); elevated white blood count in cerebral spinal fluid; central nervous system inflammation.
  • the invention provides any of the following, or compositions
  • (including pharmaceutical compositions) comprising any of the following: (a) antibody 9TL or its variants shown in Table 3; (b) a fragment or a region of antibody 9TL or its variants shown in Table 3; (c) a light chain of antibody 9TL or its variants shown in Table 3; (d) a heavy chain of antibody 9TL or its variants shown in Table 3; (e) one or more variable region(s) from a light chain and/or a heavy chain of antibody 9TL or its variants shown in Table 3; (f) one or more CDR(s) (one, two, three, four, five or six CDRs) of antibody 9TL or its variants shown in Table 3; (g) CDR H3 from the heavy chain of antibody 9TL; (h) CDR L3 from the light chain of antibody 9TL or its variants shown in Table 3; (i) three CDRs from the light chain of antibody 9TL or its variants shown in Table 3; (j) three CDRs from the heavy chain of antibody 9TL or its variants shown in Table 3
  • the invention also provides polypeptides comprising any one or more of the above.
  • the CDR portions of antibody 9TL are diagrammatically depicted in Figure 1. Determination of CDR regions is well within the skill of the art. It is understood that in some embodiments, CDRs can be a combination of the Kabat and Chothia CDR (also termed “combined CDRs” or “extended CDRs”). In some embodiments, the CDRs are the Kabat CDRs. In other embodiments, the CDRs are the Chothia CDRs. In other words, in embodiments with more than one CDR, the CDRs may be any of Kabat, Chothia, combination CDRs, or combinations thereof.
  • the invention provides a polypeptide (which may or may not be an antibody) which comprises at least one CDR, at least two, at least three, or at least four, at least five, or all six CDRs that are substantially identical to at least one CDR, at least two, at least three, at least four, at least five or all six CDRs of 9TL or its variants shown in Table 3.
  • Other embodiments include antibodies which have at least two, three, four, five, or six CDR(s) that are substantially identical to at least two, three, four, five or six CDRs of 9TL or derived from 9TL.
  • the at least one, two, three, four, five, or six CDR(s) are at least about 85%, 86%, 87%, 88%, 89%, 90%, 95%, 96%, 97%, 98%, or 99% identical to at least one, two, three, four, five or six CDRs of 9TL or its variants shown in Table 3. It is understood that, for purposes of this invention, binding specificity and/or overall activity is generally retained, although the extent of activity may vary compared to 9TL or its variants shown in Table 3 (may be greater or lesser).
  • the invention also provides a polypeptide (which may or may not be an antibody) which comprises an amino acid sequence of 9TL or its variants shown in Table 3 that has any of the following: at least 5 contiguous amino acids, at least 8 contiguous amino acids, at least about 10 contiguous amino acids, at least about 15 contiguous amino acids, at least about 20 contiguous amino acids, at least about 25 contiguous amino acids, at least about 30 contiguous amino acids of a. sequence of 9TL or its variants shown in Table 3, wherein at least 3 of the amino acids are from a variable region of 9TL ( Figure 1) or its variants shown in Table 3. In one embodiment, the variable region is from a light chain of 9TL.
  • variable region is from a heavy chain of 9TL.
  • An exemplary polypeptide has contiguous amino acid (lengths described above) from both the heavy and light chain variable regions of 9TL.
  • the 5 (or more) contiguous amino acids are from a complementarity determining region (CDR) of 9TL shown in Figure 1.
  • the contiguous amino acids are from a variable region of 9TL.
  • the binding affinities of the antibodies and polypeptides of the invention may vary, and need not be (but can be) a particular value or range, as the exemplary embodiments described below.
  • the binding affinity of the antibodies and polypeptides of the invention to A ⁇ i_ 40 can be about 0.10 to about 0.80 nM, about 0.15 to about 0.75 nM and about 0.18 to about 0.72 nM. In some embodiments, the binding affinity is about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, about 40 pM, or greater than about 40 pM. In one embodiment, the binding affinity is between about 2 pM and 22 pM.
  • the binding affinity is less than about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, about 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM. In some embodiment, the binding affinity is about 10 nM.
  • the binding affinity is less than about 10 nM, less than about 50 nM, less than about 100 nM, less than about 150 nM, less than about 200 nM, less than about 250 nM, less than about 500 nM, or less than about 1000 nM. In other embodiments, the binding affinity is less than about 5 nM. In other embodiments, the binding affinity is less than about 1 nM. In other embodiments, the binding affinity is about 0.1 nM or about 0.07 nM. In other embodiments, the binding affinity is less than about 0.1 nM or less than about 0.07 nM.
  • the binding affinity is from any of about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, about 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM to any of about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, or about 40 pM.
  • the binding affinity is any of about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, bout 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM.
  • the binding affinity is about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, about 40 pM, or greater than about 40 pM.
  • the invention also provides methods of making any of these antibodies or polypeptides,
  • the antibodies of this invention can be made by procedures known in the art.
  • the polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis.
  • Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available.
  • an antibody could be produced by an automated polypeptide synthesizer employing the solid phase method. See also, U.S. Patent Nos. 5,807,715; 4,816,567; and 6,331,415.
  • a polynucleotide comprises a sequence encoding the heavy chain and/or the light chain variable regions of antibody 9TL shown in SEQ ID NO:9 and SEQ ID NO: 10.
  • the polynucleotide comprising the nucleotide sequence shown in SEQ ID NO: 9 and SEQ ID NO: 10 are cloned into one or more vectors for expression or propagation.
  • the sequence encoding the antibody of interest may be maintained in a vector in a host cell and the host cell can then be expanded and frozen for future use.
  • Vectors (including expression vectors) and host cells are further described herein.
  • the invention also encompasses single chain variable region fragments ("scFv") of antibodies of this invention, such as 9TL.
  • Single chain variable region fragments are made by linking light and/or heavy chain variable regions by using a short linking peptide.
  • An example of a linking peptide is (GGGGS) 3 (SEQ ID NO:40) which bridges approximately 3.5 nm between the carboxy terminus of one variable region and the amino terminus of the other variable region.
  • Linkers of other sequences have been designed and used. Bird et al. (1988). Linkers can in turn be modified for additional functions, such as attachment of drugs or attachment to solid supports.
  • the single chain variants can be produced either recombinantly or synthetically.
  • an automated synthesizer can be used for synthetic production of scFv.
  • a suitable plasmid containing polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such as E. coli.
  • Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides.
  • the resultant scFv can be isolated using standard protein, purification techniques known in the art. [0123]
  • Other forms of single chain antibodies, such as diabodies are also encompassed.
  • Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123).
  • bispecific antibodies, monoclonal antibodies that have binding specificities for at least two different antigens can be prepared using the antibodies disclosed herein.
  • bispecific antibodies are known in the art (see, e.g., Suresh et al., 1986, Methods in Enzymology 121 :210). Traditionally, the recombinant production of bispecific antibodies was based on the coexpression of two immunoglobulin heavy chain-light chain pairs, with the two heavy chains having different specificities (Millstein and Cuello, 1983, Nature 305, 537-539).
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2 and CH3 regions. It is preferred to have the first heavy chain constant region (CHl), containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are cotransfected into a suitable host organism.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm.
  • Heteroconjugate antibodies comprising two covalently joined antibodies, are also within the scope of the invention. Such antibodies have been used to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HIV infection (PCT application publication Nos. WO 91/00360 and WO 92/200373; EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents and techniques are well known in the art, and are described in U.S. Patent No. 4,676,980. [0128] Chimeric or hybrid antibodies also may be prepared in vitro using known methods of synthetic protein chemistry, including those involving cross-linking agents. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4- mercaptobutyrimidate.
  • Humanized antibody comprising one or more CDRs of antibody 9TL or one or more CDRs derived from antibody 9TL can be made using any methods known in the art. For example, four general steps may be used to humanize a monoclonal antibody. These are: (1) determining the nucleotide and predicted amino acid sequence of the starting antibody light and heavy variable domains (2) designing the humanized antibody, i.e., deciding which antibody framework region to use during the humanizing process (3) the actual humanizing methodologies/techniques and (4) the transfection and expression of the humanized antibody. See, for example, U.S. Patent Nos.
  • the Fc portion can be modified to avoid interaction with Fey receptor and the complement immune system. This type of modification was designed by Dr. Mike Clark from the Department of Pathology at Cambridge University, and techniques for preparation of such antibodies are described in WO 99/58572, published November 18, 1999.
  • the constant region may be engineered to more resemble human constant regions to avoid immune response if the antibody for use in clinical trials and treatments in humans. See, for example, U.S. Patent Nos. 5,997,867 and 5,866,692.
  • the invention encompasses modifications to antibody 9TL, including functionally equivalent antibodies which do not significantly affect their properties and variants whicli have enhanced or decreased activity and/or affinity.
  • the amino acid sequence of antibody 9TL may be mutated to obtain an antibody with the desired binding affinity to A ⁇ 1-40 peptide.
  • Modification of polypeptides is routine practice in the art and need not be described in detail herein. Modification of polypeptides is exemplified in the Examples. Examples of modified polypeptides include polypeptides with conservative substitutions of amino acid residues, one or more deletions or additions of amino acids which do not significantly deleteriously change the functional activity, or use of chemical analogs.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue or the antibody fused to an epitope tag.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C- terminus of the antibody of an enzyme or a polypeptide which increases the serum half-life of the antibody.
  • substitution variants have at least one amino acid residue in the antibody molecule removed and a different residue inserted in its place.
  • the sites of greatest interest for substitutional mutagenesis include the hypervariable regions, but FR alterations are also contemplated.
  • Conservative substitutions are shown in Table 1 under the heading of "conservative substitutions". If such substitutions result in a change in biological activity, then more substantial changes, denominated "exemplary substitutions" in Table 1, or as further described below in reference to amino acid classes, may be introduced and the products screened.
  • Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties:
  • Non-polar Norleucine, Met, Ala, VaI, Leu, He
  • Polar without charge Cys, Ser, Thr, Asn, GIn
  • Non-conservative substitutions are made by exchanging a member of one of these classes for another class.
  • cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross-linking.
  • cysteine bond(s) may be added to the antibody to improve its stability, particularly where the antibody is an antibody fragment such as an Fv fragment.
  • Amino acid modifications can range from changing or modifying one or more amino acids to complete redesign of a region, such as the variable region. Changes in the variable region can alter binding affinity and/or specificity. In some embodiments, no more than one to five conservative amino acid substitutions are made within a CDR domain. In other embodiments, no more than one to three conservative amino acid substitutions are made within a CDR domain. In still other embodiments, the CDR domain is CDR H3 and/or CDR L3. [0139] Modifications also include glycosylated and nonglycosylated polypeptides, as well as polypeptides with other post-translational modifications, such as, for example, glycosylation with different sugars, acetylation, and phosphorylation.
  • Antibodies are glycosylated at conserved positions in their constant regions (Jefferis and Lund, 1997, Chem. Immunol. 65:111-128; Wright and Morrison, 1997, TibTECH 15:26-32).
  • the oligosaccharide side chains of the immunoglobulins affect the protein's function (Boyd et al., 1996, MoI. Immunol. 32:1311-1318; Wittwe and Howard, 1990, Biochem. 29:4175-4180) and the intramolecular interaction between portions of the glycoprotein, which can affect the conformation and presented three-dimensional surface of the glycoprotein (Hefferis and Lund, supra; Wyss and Wagner, 1996, Current Opin. Biotech. 7:409-416).
  • Oligosaccharides may also serve to target a given glycoprotein to certain molecules based upon specific recognition structures. Glycosylation of antibodies has also been reported to affect antibody-dependent cellular cytotoxicity (ADCC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CHO cells with tetracycline-regulated expression of ⁇ (l,4)-N-acetylglucosaminyltransferase III (GnTIII), a glycosyltransferase catalyzing formation of bisecting GIcNAc was reported to have improved ADCC activity (Umana et al, 1999, Mature Biotech. 17:176-180).
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine, asparagine-X-threonine, and asparagine-X- cysteine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • X is any amino acid except proline
  • O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • Addition of glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
  • glycosylation pattern of antibodies may also be altered without altering the underlying nucleotide sequence. Glycosylation largely depends on the host cell used to express the antibody. Since the cell type used for expression of recombinant glycoproteins, e.g. antibodies, as potential therapeutics is rarely the native cell, variations in the glycosylation pattern of the antibodies can be expected (see, e.g. Hse et al., 1997, J. Biol. Chem. 272:9062- 9070).
  • Glycosylation or certain types of glycosylation, can be enzymatically removed from the glycoprotein, for example using endoglycosidase H (Endo H), N-glycosidase F as described in Example 3, endoglycosidase Fl 3 endoglycosidase F2, endoglycosidase F3.
  • the recombinant host cell can be genetically engineered to be defective in processing certain types of polysaccharides.
  • Modifications include using coupling techniques known in the art, including, but not limited to, enzymatic means, oxidative substitution and chelation. Modifications can be used, for example, for attachment of labels for immunoassay. Modified 9TL polypeptides are made using established procedures in the art and can be screened using standard assays known in the art, some of which are described below and in the Examples.
  • the antibody comprises a modified constant region, such as a constant region that is immunologically inert or partially inert, e.g., does not trigger complement mediated lysis, does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC), or does not activate microglia; or have reduced activities (compared to the unmodified antibody) in any one or more of the following: triggering complement mediated lysis, stimulating antibody-dependent cell mediated cytotoxicity (ADCC), or activating microglia.
  • a modified constant region such as a constant region that is immunologically inert or partially inert, e.g., does not trigger complement mediated lysis, does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC), or does not activate microglia; or have reduced activities (compared to the unmodified antibody) in any one or more of the following: triggering complement mediated lysis, stimulating antibody-dependent cell mediated cytotoxicity (ADCC), or activating microglia.
  • Different modifications of the constant region may be used to achieve optimal level and/or
  • the constant region is modified as described in Eur. J. Immunol. (1999) 29:2613-2624; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No. 9809951.8 .
  • the antibody comprises a human heavy chain IgG2a constant region comprising the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence). Eur. J. Immunol. (1999) 29:2613-2624.
  • the constant region is aglycosylated for N-linked glycosylation.
  • the constant region is aglycosylated for N-linked glycosylation by mutating the glycosylated amino acid residue or flanking residues that are part of the N-glycosylation recognition sequence in the constant region. For example ⁇ N-glycosylation site N297 may be mutated to A, Q, K, or H.
  • the constant region is aglycosylated for N-linked glycosylation.
  • the constant region may be aglycosylated for N-linked glycosylation enzymatically (such as removing carbohydrate by enzyme PNGase), or by expression in a glycosylation deficient host cell.
  • antibody modifications include antibodies that have been modified as described in PCT Publication No. WO 99/58572, published November 18, 1999. These antibodies comprise, in addition to a binding domain directed at the target molecule, an effector domain having an amino acid sequence substantially homologous to all or part of a constant domain of a human immunoglobulin heavy chain. These antibodies are capable of binding the target molecule without triggering significant complement dependent lysis, or cell-mediated destruction of the target. In some embodiments, the effector domain is capable of specifically binding FcRn and/or Fc ⁇ RIIb. These are typically based on chimeric domains derived from two or more human immunoglobulin heavy chain CH2 domains. Antibodies modified in this manner are particularly suitable for use in chronic antibody therapy, to avoid inflammatory and other adverse reactions to conventional antibody therapy.
  • affinity matured antibodies can be produced by procedures known in the art (Marks et al., 1992, Bio/Technology, 10:779-783; Barbas et al., 1994, Proc Nat. Acad. Sci, USA 91 :3809-3813; Schier et al., 1995, Gene, 169:147-155; Yelton et al., 1995, J. Immunol., 155:1994-2004; Jackson et al., 1995, J. Immunol., 154(7):3310-9; Hawkins et al, 1992, J. MoI. Biol., 226:889- 896; and WO2004/058184).
  • library scanning mutagenesis One way of characterizing a CDR of an antibody and/or altering (such as improving) the binding affinity of a polypeptide, such as an antibody, termed "library scanning mutagenesis".
  • library scanning mutagenesis works as follows. One or more amino acid positions in the CDR are replaced with two or more (such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) amino acids using art recognized methods. This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of two or more members (if two or more amino acids are substituted at every position).
  • the library also includes a clone comprising the native (uns'ubstituted) amino acid.
  • a small number of clones e.g., about 20-80 clones (depending on the complexity of the library), from each library are screened for binding affinity to the target polypeptide (or other binding target), and candidates with increased, the same, decreased or no binding are identified.
  • Methods for determining binding affinity are well-known in the art. Binding affinity may be determined using BIAcore surface plasmon resonance analysis, which detects differences in binding affinity of about 2-fold or greater. BIAcore is particularly useful when the starting antibody already binds with a relatively high affinity, for example a K D of about 10 nM or lower.
  • Binding affinity may be determined using Kinexa Biocensor, scintillation proximity assays, ELISA, ORIGEN immunoassay (IGEN), fluorescence quenching, fluorescence transfer, and/or yeast display. Binding affinity may also be screened using a suitable bioassay.
  • every amino acid position in a CDR is replaced (in some embodiments, one at a time) with all 20 natural amino acids using art recognized mutagenesis methods (some of which are described herein). This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of 20 members (if all 20 amino acids are substituted at every position).
  • the library to be screened comprises substitutions in two or more positions, which may be in the same CDR or in two or more CDRs.
  • the library may comprise substitutions in two or more positions in one CDR.
  • the library may comprise substitution in two or more positions in two or more CDRs.
  • the library may comprise substitution in 3, 4, 5, or more positions, said positions found in two, three, four, five or six CDRs.
  • the substitution may be prepared using low redundancy codons. See, e.g., Table 2 of Balint et al. , (1993) Gene 137(l):109-18).
  • the CDR may be CDRH3 and/or CDRL3.
  • the CDR may be one or more of
  • the CDR may be a Kabat CDR, a Chothia CDR, or an extended CDR.
  • Candidates with improved binding may be sequenced, thereby identifying a CDR substitution mutant which results in improved affinity (also termed an "improved" substitution).
  • Candidates that bind may also be sequenced, thereby identifying a CDR substitution which retains binding.
  • Multiple rounds of screening may be conducted. For example, candidates (each comprising an amino acid substitution at one or more position of one or more CDR) with improved binding are also useful for the design of a second library containing at least the original and substituted amino acid at each improved CDR position (i.e., amino acid position in the CDR- at which a substitution mutant showed improved binding). Preparation, and screening or selection of this library is discussed further below.
  • Library scanning mutagenesis also provides a means for characterizing a CDR, in so far as the frequency of clones with improved binding, the same binding, decreased binding or no binding also provide information relating to the importance of each amino acid position for the stability of the antibody-antigen complex. For example, if a position of the CDR retains binding when changed to all 20 amino acids, that position is identified as a position that is unlikely to be required for antigen binding. Conversely, if a position of CDR retains binding in only a small percentage of substitutions, that position is identified as a position that is important to CDR function. Thus, the library scanning mutagenesis methods generate information regarding positions in the CDRs that can be changed to many different amino acid (including all 20 amino acids), and positions in the CDRs which cannot be changed or which can only be changed to a few amino acids.
  • Candidates with improved affinity may be combined in a second library, which includes the improved amino acid, the original amino acid at that position, and may further include additional substitutions at that position, depending on the complexity of the library that is desired, or permitted using the desired screening or selection method.
  • adjacent amino acid position can be randomized to at least two or more amino acids. Randomization of adjacent amino acids may permit additional conformational flexibility in the mutant CDR, which may in turn, permit or facilitate the introduction of a larger number of improving mutations.
  • the library may also comprise substitution at positions that did not show improved affinity in the first round of screening.
  • the second library is screened or selected for library members with improved and/or altered binding affinity using any method known in the art, including screening using BIAcore surface plasmon resonance analysis, and selection using any method known in the art for selection, including phage display, yeast display, and ribosome display.
  • the invention also encompasses fusion proteins comprising one or more fragments or regions from the antibodies (such as 9TL) or polypeptides of this invention.
  • a fusion polypeptide is provided that comprises at least 10 contiguous amino acids of the variable light chain region shown in SEQ ID NO:2 ( Figure 1) and/or at least 10 amino acids of the variable heavy chain region shown in SEQ ID NO:1 ( Figure 1).
  • a fusion polypeptide comprises at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable light chain region shown in SEQ ID N0:2 ( Figure 1) and/or at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable heavy chain region shown in SEQ ID NO:1 ( Figure 1).
  • the fusion polypeptide comprises a light chain variable region arid/or a heavy chain variable region of 9TL, as shown in SEQ ID NO:2 and SEQ ID NO:1 of Figure 1.
  • the fusion polypeptide comprises one or more CDR(s) of 9TL.
  • the fusion polypeptide comprises CDR H3 and/or CDR L3 of antibody 9TL.
  • an 9TL fusion protein contains one or more 9TL antibodies and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region.
  • exemplary heterologous sequences include, but are not limited to a "tag" such as a FLAG tag or a 6His tag. Tags are well known in the art.
  • a 9TL fusion polypeptide can be created by methods known in the art, for example, synthetically or recombinantly.
  • the 9TL fusion proteins of this invention are made by preparing an expressing a polynucleotide encoding them using recombinant methods described herein, although they may also be prepared by other means known in the art, including, for example, chemical synthesis.
  • compositions comprising 9TL antibodies or polypeptides conjugated (for example, linked) to an agent that facilitate coupling to a solid support (such as biotin or avidin).
  • a solid support such as biotin or avidin.
  • Conjugation generally refers to linking these components as described herein.
  • the linking (which is generally fixing these components in proximate association at least for administration) can be achieved in any number of ways. For example, a direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other.
  • a nucleophilic group such as an amino or sulfhydryl group
  • a carbonyl-containing group such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide) on the other.
  • An antibody or polypeptide of this invention may be linked to a labeling agent
  • Label such as a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art which generally provide (either directly or indirectly) a signal.
  • compositions including pharmaceutical compositions
  • kits comprising antibody 9TL, and, a.s this disclosure makes clear, any or all of the antibodies and/or polypeptides described herein.
  • Anti-A ⁇ peptide antibodies and polypeptides having impaired effector function use antibodies or polypeptides (including pharmaceutical compositions comprising the antibodies or polypeptides) that specifically bind to a beta-amyloid peptide and have impaired effector function.
  • the antibodies and polypeptides are further characterized by any (one or more) of the following characteristics: (a) suppresses formation of amyloid plaques in a subject; (b) reduces amyloid plaques in a subject; (c) treats, prevents, ameliorates one or more symptoms of Alzheimer's disease; (d) improves cognitive function.
  • the antibodies and polypeptides described herein may exhibit a desirable safety profile, for example, the compositions of the invention do not cause significant or unacceptable levels or have a reduced level of any one or more of: bleeding in the brain vasculature (cerebral hemorrhage); meningoencephalitis (including changing magnetic resonance scan); elevated white blood count in cerebral spinal fluid; central nervous system inflammation.
  • an anti-A ⁇ antibody having N-linked glycosylation removed in the Fc region was effective in removing amyloid plaques in the brain and improving cognitive function with significantly less microhemorrhage than the intact antibody in an animal model for Alzheimer's disease.
  • an antibody or a polypeptide having an "impaired effector function” refers to antibodies or polypeptides that do not have any effector function or have reduced activity or activities of effector function (compared to antibody or polypeptide having an unmodified or a naturally occurring constant region), e.g., having no activity or reduced activity in any one or more of the following: a) triggering complement mediated lysis; b) stimulating antibody-dependent cell mediated cytotoxicity (ADCC); and c) activating microglia.
  • the effector function activity may be reduced by about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, and 100%.
  • the antibody binds to a beta-amyloid peptide without triggering significant complement dependent lysis, or cell mediated destruction of the target.
  • the Fc receptor binding site on the constant region may be modified or mutated to remove or reduce binding affinity to certain Fc receptors, such as Fc ⁇ RI, Fc ⁇ RII, and/or Fc ⁇ RIII.
  • Fc ⁇ RI Fc ⁇ RII
  • Fc ⁇ RIII Fc ⁇ RIII
  • the antibody that specifically binds to the an A ⁇ peptide comprises a heavy chain constant region having impaired effector function.
  • the heavy chain constant region may have naturally occurring sequence or is a variant.
  • the amino acid sequence of a naturally occurring heavy chain constant region is mutated, e.g., by amino acid substitution, insertion and/or deletion, whereby the effector function of the constant region is impaired.
  • the N-glycosylation of the Fc region of a heavy chain constant region may also be changed, e.g., may be removed completely or partially, whereby the effector function of the constant region is impaired.
  • the effector function is impaired by removing N- glycosylation of the Fc region (e.g., in the CH 2 domain of IgG) of the anti-A ⁇ peptide.
  • N-glycosylation of the Fc region is removed by mutating the glycosylated amino acid residue or flanking residues that are part of the glycosylation recognition sequence in the constant region.
  • the tripeptide sequences asparagine-X-serine (N-X-S), asparagine-X-thxeonine (N-X-T) and asparagine-X-cysteine (N-X-C), where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain for N-glycosylation. Mutating any of the amino acid in the tripeptide sequences in the constant region yields an aglycosylated IgG.
  • N-glycosylation site N297 of human IgGl and IgG3 may be mutated to A, D, Q, K, or H. See, Tao et al., J.
  • the amino acid N in the tripeptide sequences is mutated to any one of amino acid A, C, D, E, F, G, H, I, K, L 5 M, P, Q, R, S, T, V 5 W, Y.
  • the amino acid N in the tripeptide sequences is mutated to a conservative substitution.
  • the amino acid X in the tripeptide sequences is mutated to proline.
  • the amino acid S in the tripeptide sequences is mutated to Ai D, E, F, G, H, I, K, L, M, N, P, Q, R, V, W, Y.
  • the amino acid T in the tripeptide sequences is mutated to A 5 D, E, F 5 G, H, I 5 K, L, M 5 N, P, Q 5 R 5 V, W 5 Y.
  • the amino acid C in the tripeptide sequences is mutated to A 5 D, E, F, G 5 H 5 1 5 K 5 L, M 5 N, P, Q, R 5 V 5 W, Y. In some embodiments, the amino acid following the tripeptide is mutated to P.
  • the N-glycosylation in the constant region is removed enzymatically (such as N-glycosidase F as described in Example 3, endoglycosidase Fl , endoglycosidase F2, endoglycosidase F3, and englycosidase H). Removing N-glycosylation may also be achieved by producing the antibody in a cell line having deficiency for N-glycosylation. Wright et al. J J Immunol. 160(7):3393-402 (1998).
  • amino acid residue interacting with oligosaccharide attached to the N-glycosylation site of the constant region is mutated to reduce binding affinity to Fc ⁇ RI.
  • F241, V264, D265 of human IgG3 may be mutated. See, Lund et al., J. Immunology 157:4963-4969 (1996).
  • the effector function is impaired by modifying regions such as 233-236, 297, and/or 327-331 of human IgG as described in PCT WO 99/58572 and Armoxir et al., Molecular Immunology 40: 585-593 (2003); Reddy et al., J. Immunology 164:1925-1933 (2000).
  • Antibodies described in PCT WO 99/58572 and Armour et al. comprise, in addition to a binding domain directed at the target molecule, an effector domain having an amino acid sequence substantially homologous to all or part of a constant region of a human immunoglobulin heavy chain.
  • the effector domain has a reduced affinity for Fc ⁇ RI, Fc ⁇ RIIa, and Fc ⁇ RIII.
  • the effector domain is capable of specifically binding FcRn and/or Fc ⁇ RIIb. These are typically based on chimeric domains derived from two or more human immunoglobulin heavy chain CR2 domains. Antibodies modified in this manner are particularly suitable for use in chronic antibody therapy, to avoid inflammatory and other adverse reactions to conventional antibody therapy.
  • the heavy chain constant region of the antibody is a human heavy chain IgGl with any of the following mutations: 1) A327A330P331 to G327S330S331; 2) E233L234L235G236 to P233V234A235 with G236 deleted; 3) E233L234L235 to P233V234A235; 4) E233L234L235G236A327A330P331 to P233V234A235G327S330S331 with G236 deleted; 5) E233L234L235A327A330P331 to P233V234A235G327S330S331; and 6) N297 to A297 or any other amino acid except N.
  • the heavy chain constant region of the antibody is a human heavy chain IgG2 with the following mutations: A330P331 to S330S331. In some embodiments, the heavy chain constant region of the antibody is a human heavy chain IgG4 with any of the following mutations: E233F234L235G236 to P233V234A235 with G236 deleted; E233F234L235 to P233V234A235; and S228L235 to P228E235.
  • the constant region of the antibodies may also be modified to impair complement activation.
  • complement activation of IgG antibodies following binding of the Cl component of complement may be reduced by mutating amino acid residues in the constant region in a Cl binding motif (e.g., CIq binding motif).
  • CIq binding motif constitutes residues E318, K320, and K322.
  • CIq binding motif E318, K320, and K322 identified for murine IgG2b is believed to be common for other antibody isotypes.
  • CIq binding activity for IgG2b 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 CIq binding.
  • the invention also provides antibodies having impaired effector function wherein the antibody has a modified hinge region. Binding affinity of human IgG for its Fc receptors can be modulated by modifying the hinge region. Canfield et al., J.
  • the modified hinge region may comprise a complete hinge region derived from an antibody of different antibody class or subclass from that of the CHl domain.
  • the constant domain (CHl) 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.
  • a neutral residue such as alanine
  • Such alterations are carried out using art recognized protein chemistry and, preferably, genetic engineering techniques and as described herein.
  • polypeptides that specifically bind to an A ⁇ peptide and fused to a heavy chain constant region having impaired effector function may also be used for the methods described herein.
  • the polypeptide comprises a sequence derived from antibody 9TL or its variants shown in Table 3.
  • the polypeptide is derived from a single domain antibody that binds to an A ⁇ peptide. Single domain antibodies can be generated using methods known in the art. Omidfar et al., Tumour Biol 25:296-305 (2004); Herring et al., Trends in Biotechnology 21:484-489 (2003).
  • the antibody or polypeptide is not a F(ab') 2 fragment. In some embodiments, the antibody or polypeptide is not a Fab fragment. In some embodiments, the antibody or polypeptide is not a single chain antibody scFv. In some embodiments, the antibody or polypeptide is a PEGylated F(ab') 2 fragment. In some embodiments, the antibody or polypeptide is a PEGylated Fab fragment. In some embodiments, the antibody or polypeptide is a PEGylated single chain antibody scFv.
  • Antibodies and polypeptides with modified constant regions can be tested in one or more assays to evaluate level of effector function reduction in biological activity compared to the starting antibody. For example, the ability of the antibody or polypeptide with an altered Fc region to bind complement or Fc receptors (for example, Fc receptors on microglia), or altered hinge region can be assessed using the assays disclosed herein as well as any art recognized assay.
  • the antibody that specifically binds to beta-amyloid peptide is a polyclonal antibody.
  • the antibody is a monoclonal antibody.
  • the antibody is a human antibody.
  • the antibody is a chimeric antibody.
  • the antibody is a humanized antibody.
  • the antibody is a primatized antibody. See, e.g., Yocum et al., J. Rheumatol. 25:1257-62 (1998); Bugelski et al., Human & Experimental Toxicoloy 19:230-243 (2000).
  • the antibody is deimmunized by mutation so that the antibody does not activate human immune system.
  • a ⁇ peptide includes any fragments of the enzymatic cleavage products of amyloid precursor protein.
  • a ⁇ peptide includes any fragments of A ⁇ i. 40 , A ⁇ i 42 , or A ⁇ M a; and peptides which are truncated with various number of amino acids at the N-terminus or the C-terminus of A ⁇ o, A ⁇ 1-4 2, or A ⁇ 1-43 .
  • Amino acid numbering used herein is based on the numbering for A ⁇ l-43 (SEQ ID NO: 17).
  • the antibody or polypeptide specifically binds to an epitope within residues 1-16 of A ⁇ peptide. In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 16-28 of A ⁇ peptide. In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 28-40 of A ⁇ i -4 o peptide. In some embodiments, the antibody or polypeptide specifically binds to an epitope within ⁇ residues 28-42 of A ⁇ 1-42 peptide. In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 28-43 of Ap 1-43 peptide.
  • the antibody or polypeptide specifically binds to an A ⁇ peptide without binding to full-length amyloid precursor protein (APP). In some embodiments, the antibody or the polypeptide specifically binds to the aggregated form of A ⁇ without binding to the soluble form. In some embodiments, the antibody or the polypeptide specifically binds to the soluble form of A ⁇ without binding to the aggregated form. In some embodiments, the antibody or the polypeptide specifically binds to both aggregated form and soluble forms of A ⁇ .
  • Antibodies that bind to various aggregated form of A ⁇ are known in the art, for example, antibodies that bind to amyloid beta-derived diffusible ligands (ADDLs); antibodies that bind to amyloid fibrils and/or deposit. WO 03/104437; U.S. Pub. No. 2003/0147887; U.S. Pub. No. 2004/0219146.
  • the antibody or polypeptide comprises one, two, or three
  • the antibody or polypeptide comprises a variable heavy chain region as set forth in SEQ ID NO:8 in U.S. Pub. No. 2003/0165496 and a variable light chain region as set forth in SEQ ID NO:5 in U.S. Pub. No. 2003/0165496.
  • the antibody or polypeptide comprises a variable heavy chain region as set forth in SEQ ID NO: 12 in U.S. Pub. No. 2003/0165496 and a variable light chain region as set forth in SEQ ID NO:11 in U.S. Pub. No. 2003/0165496.
  • the antibody or polypeptide comprises one, two, or three CDRs from the 10D5 immunoglobulin light chain (SEQ ID NO:14 in U.S. Pub. Nos. 2003/0165496, or 2004/0087777), and/or one, two, or three CDRs from the 10D5 immunoglobulin heavy chain (SEQ ID NO:16 in U.S. Pub. Nos. 2003/0165496, or 2004/0087777).
  • the antibody or polypeptide specifically binds to an epitope within residues 33-40 of A ⁇ mo. In some embodiments, the antibody or polypeptide specifically binds to an epitope on A ⁇ 1-40 that includes amino acid 35-40. In some embodiments, the antibody or polypeptide specifically binds to an epitope on AP M0 that includes amino acid 36-40. In some embodiments, the antibody or polypeptide specifically binds to an epitope on A ⁇ i- 4 o that includes amino acid 39 and/or 40. In some embodiments, the antibody or polypeptide specifically binds to A ⁇ i -4 o but does not specifically bind to A ⁇ i- 42 and/or A ⁇ 1-43 .
  • the antibody or polypeptide is antibody 9TL or an antibody or a polypeptide derived from 9TL described herein. In some embodiments, the antibody or polypeptide competitively inhibits binding of antibody 9TL and/or antibody or polypeptide derived from 9TL to A ⁇ 1-4 o. In some embodiments, the antibody is not antibody 2286 described in PCT WO 2004/032868. [0181]
  • the binding affinities of the antibodies and polypeptides of the invention may vary, and need not be (but can be) a particular value or range, as the exemplary embodiments described below.
  • the binding affinity is less than about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, about 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM; about 30 pM, about 10 pM. In some embodiments, the binding affinity is about 10 nM.
  • the binding affinity is less than about 10 nM, less than about 50 nM, less than about 100 nM, less than about 150 nM, less than about 200 nM, less than about 250 nM, less than about 500 nM, or less than about 1000 nM. In other embodiments, the binding affinity is less than about 5 nM. In other embodiments, the binding affinity is less than about 1 nM. In other embodiments, the binding affinity is about 0.1 nM or about 0.07 nM. In other embodiments, the binding affinity is less than about 0.1 nM or less than about 0.07 nM.
  • the binding affinity is from any of about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, about 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM to any of about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, or about 40 pM.
  • the binding affinity is any of about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, bout 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM.
  • the binding affinity is about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, about 40 pM, or greater than about 4O pM.
  • Competition assays can be used to determine whether two antibodies bind the same epitope by recognizing identical or sterically overlapping epitopes or one antibody competitively inhibits binding of another antibody to the antigen. These assays are known in the art. Typically, antigen is immobilized on a multi-well plate and the ability of unlabeled antibodies to block the binding of labeled antibodies is measured. Common labels for such competition assays are radioactive labels or enzyme labels.
  • Antibodies and polypeptides that specifically bind to A ⁇ can be screened for efficacy in removing amyloid deposit and other beneficial effects, such as improving cognition.
  • antibodies or polypeptides may be administered to an animal having Alzheimer's pathology.
  • Various animal models for Alzheimer's disease are known in the art.
  • level of compact and diffuse amyloid plaques, behavior analysis for cognition, and microglia activation and microhemorrhage may tested using methods known in the art and described in detail in Example 2.
  • the invention also provides isolated polynucleotides encoding the antibodies and polypeptides of the invention (including an antibody comprising the polypeptide sequences of the light chain and heavy chain variable regions shown in Figure 1), and vectors and host cells comprising the polynucleotide.
  • the invention provides polynucleotides (or compositions, including pharmaceutical compositions), comprising polynucleotides encoding any of the following: (a) antibody 9TL or its variants shown in Table 3; (b) a fragment or a region of antibody 9TL or its variants shown in Table 3; (c) a light chain of antibody 9TL or its variants shown in Table 3; (d) a heavy chain of antibody 9TL or its variants shown in Table 3; (e) one or more variable region(s) from a light chain and/or a heavy chain of antibody 9TL or its variants shown in Table 3; (f) one or more CDR(s) (one, two, three, four, five or six CDRs) of antibody 9TL or its variants shown in Table 3; (g) CDR H3 from the heavy chain of antibody 9TL; (h) CDR L3 from the light chain of antibody 9TL or its variants shown in Table 3; (i) three CDRs from the light chain of antibody 9TL or its
  • the invention provides polynucleotides encoding any of the antibodies (including antibody fragments) and polypeptides described herein, such as antibodies and polypeptides having impaired effector function.
  • Polynucleotides can be made by procedures known in the art.
  • the invention provides compositions (such as a pharmaceutical compositions) comprising any of the polynucleotides of the invention.
  • the composition comprises an expression vector comprising a polynucleotide encoding the 9TL antibody as described herein.
  • the composition comprises an expression vector comprising a polynucleotide encoding any of the antibodies or polypeptides described herein.
  • the composition comprises either or both of the polynucleotides shown in SEQ ID NO:9 and SEQ ID NO: 10. Expression vectors, and administration of polynucleotide compositions are further described herein.
  • the invention provides a method of making any of the polynucleotides described herein.
  • Polynucleotides complementary to any such sequences are also encompassed by the present invention.
  • Polynucleotides may be single-stranded (coding or antisense) or double- stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules.
  • RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one- to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non- coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence.
  • Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule.
  • the effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein.
  • Variants preferably exhibit at least about 70% identity, more preferably at least about 80% identity and most preferably at least about 90% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof.
  • Two polynucleotide or polypeptide sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • the "percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e. gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e. the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • Variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof.
  • Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementary sequence).
  • Suitable “moderately stringent conditions” include prewashing in a solution of 5 X
  • SSC 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50°C-65°C, 5 X SSC, overnight; followed by washing twice at 65°C for 20 minutes with each of 2X, 0.5X and 0.2X SSC containing 0. 1 % SDS.
  • highly stringent conditions or “high stringency conditions” are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 5O 0 C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/5 OmM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42 0 C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 ⁇ g/ml), 0.1% SDS, and 10% dextran
  • the resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison).
  • the polynucleotides of this invention can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well kno ⁇ vn in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence.
  • a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein.
  • Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome.
  • the polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al. (1989).
  • RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., (1989), for example.
  • Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector.
  • Suitable examples include plasmids and bacterial viruses, e.g., pUCl 8, ⁇ UC19, Bluescript (e.g., pBS SK+) and its derivatives, mpl8, mpl9, pBR322, pMB9, CoIEl, pCRl, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28.
  • plasmids and bacterial viruses e.g., pUCl 8, ⁇ UC19, Bluescript (e.g., pBS SK+) and its derivatives, mpl8, mpl9, pBR322, pMB9, CoIEl, pCRl, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28.
  • Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462.
  • Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons.
  • the vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus).
  • electroporation employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances
  • microprojectile bombardment e.g., where the vector is an infectious agent such as vaccinia virus.
  • infection e.g., where the vector is an infectious agent such as vaccinia virus.
  • the invention also provides host cells comprising any of the polynucleotides described herein.
  • Any host cells capable of over-expressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest.
  • mammalian host cells include but not limited to COS, HeLa, and CHO cells. See also PCT Publication No. WO 87/04462.
  • Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis) and yeast (such as S. cerevisae, S. pombe; or K. lactis).
  • the host cells express the cDNAs at a level of about 5 fold higher, more preferably 10 fold higher, even more preferably 20 fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells.
  • Screening the host cells for a specific binding to A ⁇ 1-40 is effected by an immunoassay or FACS.
  • a cell overexpressing the antibody or protein of interest can be identified.
  • Antibody 9TL which binds to C-terminus of Ap 1-40 may be used to identify or detect the presence or absence of Ap 1-40 .
  • Detection generally involves contacting a biological sample with an antibody described herein that binds to A ⁇ i_ 40 and the formation of a complex between A ⁇ 1-40 and an antibody ⁇ e.g., 9TL) which binds specifically to Ap 1-40 .
  • the formation of such a complex can be in vitro or in vivo.
  • the term "detection" as used herein includes qualitative and/or quantitative detection (measuring levels) with or without reference to a control.
  • any of a variety of known methods can be used for detection, including, but not limited to, immunoassay, using antibody that binds the polypeptide, e.g. by enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA) and the like; and functional assay for the encoded polypeptide, e.g. binding activity or enzymatic assay.
  • the antibody is detectably labeled.
  • Other embodiments are known in the art and described herein.
  • Antibodies and polypeptides of the invention can be used in the detection, diagnosis and monitoring of a disease, condition, or disorder associated with altered or aberrant A ⁇ or ⁇ APP expression, such as Alzheimer's disease and Down's syndrome.
  • the invention provides methods comprises contacting a specimen (sample) of an individual suspected of having altered or aberrant A ⁇ expression with an antibody or polypeptide of the invention and determining whether the level of A ⁇ 1-4 o differs from that of a control or comparison specimen.
  • the invention provides methods comprises contacting a specimen (sample) of an individual and determining level of A ⁇ 1-40 expression.
  • the antibody may be labeled with a detectable moiety including but not limited to radioisotopes, fluorescent labels, and various enzyme-substrate labels. Methods of conjugating labels to an antibody are known in the art.
  • antibodies of the invention need not be labeled, and the presence thereof can be detected using a labeled antibody which binds to the antibodies of the invention.
  • the antibodies of the present invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques, pp.147- 158 (CRC Press, Inc. 1987).
  • the antibodies may also be used for in vivo diagnostic assays, such as in vivo imaging.
  • the antibody is labeled with a radionuclide (such as 111 In, 99 Tc, 14 C, 131 1, 125 I, or 3 H) so that the cells or tissue of interest can be localized using immunoscintiography.
  • a radionuclide such as 111 In, 99 Tc, 14 C, 131 1, 125 I, or 3 H
  • the antibody may also be used as staining reagent in pathology, following techniques well known in the art.
  • Anti-A ⁇ antibodies having impaired effector function may be used for measuring brain amyloid burden for diagnosis of subject at risk of or diagnosed with AD, and assessing progress of any treatment and disease stage. It has been reported that peripheral administration of a monoclonal anti-A ⁇ antibody results in a rapid increase in plasma A ⁇ and the magnitude of this increase is highly correlated with amyloid burden in the hippocampus and cortex. DeMattos et al., Science 295:2264-2267 (2002).
  • an anti-A ⁇ antibody having impaired effector function is administered to a subject, and level of A ⁇ in the plasma is measured, whereby an increase in plasma A ⁇ indicates presence and/or level of brain amyloid burden in the subject. These methods may be used to monitor effectiveness of the treatment and disease stage and to determine future dosing and frequency. Antibodies having impaired effector function may have a better safety profile and provide advantage for these diagnostic uses.
  • the antibodies can be used in methods for treating, preventing and inhibiting the development of a disease characterized by aberrant deposition of a protein in the brain of a subject.
  • the methods comprise administering to the subject an effective amount of an antibody that specifically binds to the protein or the protein deposit or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
  • an antibody which specifically binds to prion protein or aggregated form of prion protein and has impaired effector function may be administered to a subject for prophylactic and/or therapeutic treatment of Prion diseases; an antibody which specifically binds to synuclein (e.g., alpha-synuclein) or aggregated form of synuclein and has impaired effector function may be administered to a subject for prophylactic and/or therapeutic treatment of Parkinson's disease.
  • synuclein e.g., alpha-synuclein
  • Parkinson's disease e.g., Parkinson's disease
  • the antibodies (including polypeptides), polynucleotides, and pharmaceutical compositions described herein can be used in methods for treating, preventing and inhibiting the development of Alzheimer's disease and other diseases associated with altered A ⁇ or ⁇ APP expression, or accumulation or deposit of A ⁇ peptide (collectively termed "A ⁇ -associated diseases"), such as Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, vascular disorder caused by deposit of A ⁇ peptide in blood vessels (such as stroke and HCHWA-D).
  • Such methods comprise administering the antibodies, polypeptides, or polynucleotides, or a pharmaceutical composition to the subject.
  • compositions or medicaments are administered to a patient susceptible to, or otherwise at risk of, Alzheimer's disease (or other A ⁇ -associated disease) in an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the outset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • compositions or medicaments are administered to a patient suspected of, or already suffering from such a disease in amount sufficient to cure, or at least partially arrest, the symptoms of the disease (biochemical, histological and/or behavioral), including its complications and intermediate pathological phenotypes in development of the disease.
  • the invention also provides a method of delaying development of a symptom associated with Alzheimer's disease (or other A ⁇ -associated disease) in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
  • a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
  • Alzheimer disease includes, but not limited to, abnormalities of memory, problem solving, language, calculation, visuospatial perception, judgment, and behavior.
  • This invention also provides methods of inhibiting or suppressing the formation of amyloid plaques and/or A ⁇ accumulation in a subject comprising administering an effective dose of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein described herein to the subject.
  • a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein described herein to the subject.
  • the amyloid plaques are in the brain of the subject.
  • the amyloid plaques are in the cerebral vasculature of the subject, hi other embodiments, the A ⁇ accumulation is in the circulatory system of the subject.
  • This invention also provides methods of reducing amyloid plaques and/or reducing or slowing A ⁇ accumulation in a subject comprising administering an effective dose of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
  • a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
  • the amyloid plaques are in the brain of the subject, hi some embodiments, the amyloid plaques are in the cerebral vasculature of the subject.
  • the A ⁇ accumulation is in the circulatory system of the subject.
  • This invention also provides methods of removing or clearing amyloid plaques and/or A ⁇ accumulation in a subject comprising administering an effective dose of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
  • a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
  • the amyloid plaques are in the brain of the subject, hi some embodiments, the amyloid plaques are in the cerebral vasculature of the subject, hi other embodiments, the A ⁇ accumulation is in the circulatory system of the subject.
  • This invention also provides methods of reducing A ⁇ peptide in a tissue (such as brain), inhibiting and/or reducing accumulation of A ⁇ peptide in a tissue (such as brain), and inhibiting and/or reducing toxic effects of A ⁇ peptide in a tissue (such as brain) in a subject comprising administering an effective dose of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
  • a ⁇ polypeptide may be in soluble, oligomeric, or deposited form. Oligomeric form of A ⁇ may be composed of 2-50
  • a ⁇ polypeptides which can be a mixture of full length 1-40 and 1-42 peptides and/or any truncated version of the these peptides.
  • the invention also provides methods of improving cognition or reversing cognitive decline associated with diseases associated with amyloid deposit of A ⁇ in a subject, such as Alzheimer's disease, comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
  • the invention also provides methods for treating or preventing diseases associated with amyloid deposit of A ⁇ , comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, wherein the antibody comprises an Fc region with a variation from a naturally occurring Fc region, wherein the variation results in impaired effector function, whereby the administration of the antibody causes less cerebral microhemorrhage than administration of an antibody without the variation.
  • Amyloidoses are disorders that are characterized by extracellular deposition of protein fibrils, which form amyloid deposits. While the majority of these conditions are associated with amyloid deposition in the periphery, there are a number of amyloidoses in which central nervous system fibril deposition predominates. WO 00/72876 describes a number of central and peripheral amyloidoses.
  • Alzheimer's disease is the most well-known and probably the most common amyloid disease of the central nervous system. This condition is characterized by A-beta- containing plaques and neurofibrillatory tangles, as summarized elsewhere. Various forms of senile dementia are also associated with similar, but less progressive, A-beta plaque formation, as is Down's syndrome.
  • a variant of the amyloidogenic protein cystatin C L68Q cystatin C, is associated with massive cerebral amyloidosis leading to brain hemorrhage and death in early adult life in a hereditary form of amyloid angiopathy (hereditary cystatin C amyloid angiopathy).
  • cystatin C wt 1 cystatin C
  • a normal variant of cystatin C can be found associated with A-beta peptide as a component of amyloid plaques in Alzheimer's disease.
  • Nilsson and co-workers incubated antibodies directed to wt 1 cystatin C with monomeric forms of the two variant proteins in solution and observed decreased dimerization of the proteins. Nilsson, M. et al. (2004) J. Biol. Chem. 279(3): 24236-45.
  • Down syndrome is characterized by A-beta peptide plaque deposition, apoptotic cell death and aberrant dendritic arborization, in part due to constitutively increased expression of genes that include amyloid precursor protein (APP) and other proteins (superoxide dismutase I, and SlOO-beta) - all located within the Down locus.
  • genes that are not linked to the Down locus include amyloid precursor protein (APP) and other proteins (superoxide dismutase I, and SlOO-beta) - all located within the Down locus.
  • genes that are not linked to the Down locus genes w/in a segment of chromosome 21) - GAP-43, nitric oxide synthase 3, neuronal thread protein, pro-apoptosis genes such as p53, Bax and LI-I beta- converting enzyme. Expression of these non-Down locus genes correlates with proliferation of dystrophic neuritis and apoptotic cell death, de Ia Monte
  • Brain deposition of amyloid plaques formed from A-beta peptide is a also a common pathologic feature in HIV-AIDS patients. Green, D. A., et al. (2005) AIDS 19(4): 407- 11. Similarly, deposition of A-beta peptide-containing plaques have also been observed shortly following traumatic brain injury in humans, where A-beta co-localizes with APP and neurofilament proteins in swollen axons. Smith, D.H., et al. (2003): 98(5): 1072-7. Brains of patients with end-stage acquired immunodeficiency syndrome (AIDS) were also shown to have increased levels of ubiquitin-stained dotlike deposits (Ub-dots). Gelman, B.B,, and Schuenke, K.
  • AIDS end-stage acquired immunodeficiency syndrome
  • Amyloidoma is a relatively rare form of CNS amyloidosis, presenting in the form of an amyloid tumor, usually in the choroid plexus, with secondary extensions into white matter.
  • Primary amyloidomas of the brain parenchyma comprise lesions composed of amyloid AL lambda light chain. Tabatbai, G., et al. (2005) Arch Neurol. 62(3): 477-80.
  • Familial leptomeningeal amyloidosis is associated with a genetic abnormality of the transthyretin (TTR) variant As ⁇ l8Gly (D18G).
  • TTR transthyretin
  • Alpha-synuclein inclusions in oligodendroglia characterize multiple system atrophy (MSA). Kahle, P.M., et al. 2002. EMBO rep. 3(6): 583-8.
  • PrP is aberrant form of cellular prion protein (PrP ), a copper-binding glycoprotein attached to the cell membrane of neurons and other cells. PrP amyloid accumulation is commonly associated with PrP cerebral amyloid angiopathy (PrP-CAA), where the accumulation is in neurofibrillary tangles and vascular amyloid, and in Gerstmann-Straussler-
  • PrP-CAA PrP cerebral amyloid angiopathy
  • PrP Sc deposition is also found in human spongiform encephalopathy (variant Creutzfeld- Jacob Disease). Paracrine inhibition of prion propagation by anti-PrP single-chain F ⁇ v miniantibodies has been reported. Heppner et al., J. Viol. 79:8330-8; 2005.
  • the following table provides a summary of examples of diseases associated with aberrant brain protein deposition and protein components for the deposit. Antibody against these components may be generated using methods known in the art and methods described herein, or antibodies known in the art.
  • the methods described herein can be accomplished by a single direct injection at a single time point or multiple time points to a single or multiple sites. Administration can also be nearly simultaneous to multiple sites. Frequency of administration may be determined and adjusted over the course of therapy, and is base on accomplishing desired results.
  • sustained continuous release formulations of antibodies (including polypeptides), polynucleotides, and pharmaceutical compositions of the invention may be appropriate.
  • Various formulations and devices for achieving sustained release are known in the art.
  • Patients, subjects, or individuals include mammals, such as human, bovine, equine, canine, feline, porcine, and ovine animals.
  • the subject is preferably a human, and may or may not be afflicted with disease or presently show symptoms.
  • 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 useful for individuals who do have a known genetic risk of Alzheimer's disease. Such individuals include those having relatives who have experienced this disease, and those whose risk is determined by analysis of genetic or biochemical markers.
  • Genetic markers of risk toward Alzheimer's disease include mutations in the APP gene, particularly mutations at position 717 and positions 670 and 671 referred to as the Hardy and Swedish mutations respectively (see Hardy (1997) Trends Neurosci. 20: 154-9).
  • Other markers of risk are mutations in the presenilin genes, PSl 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 A ⁇ 42 levels. Elevated tau and decreased A ⁇ 42 levels signify the presence of AD.
  • Alzheimer's disease can also be diagnosed by ADRDA (Alzheimer's Disease and Related Disorders Association) criteria.
  • ADRDA Alzheimer's Disease and Related Disorders Association
  • 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 various ways known in the art over time. In the case of potential Down's syndrome patients, treatment can begin antenatally by administering therapeutic agent to the mother or shortly after birth.
  • the pharmaceutical composition that can be used in the above methods include, any of the antibodies, polypeptides, and/or polynucleotides described herein.
  • antibody is antibody 9TL or its variants sho ⁇ vn in Table 3.
  • the antibody is an antibody that specifically binds to an A ⁇ peptide and comprises a constant region having impaired effector function.
  • the antibody is preferably administered to the mammal in a carrier; preferably a pharmaceutically-acceptable carrier.
  • a carrier preferably a pharmaceutically-acceptable carrier.
  • Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, PA, 1990; and Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing, 2000.
  • an appropriate amount of a priarmaceutically-acceptable salt is used in the formulation to render the formulation isotonic.
  • the carrier include saline, Ringer's solution and dextrose solution.
  • the pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5.
  • Further carriers include sustained release preparations such as semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, liposomes or microparticles. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentration of antibody being administered.
  • the antibody can be administered to the mammal by injection (e.g., systemic, intravenous, intraperitoneal, subcutaneous, intramuscular, intraportal, intracerebral, intracerebralventricular, and intranasal), or by other methods, such as infusion, which ensure its delivery to the bloodstream in an effective form.
  • the antibody may also be administered by isolated perfusion techniques, such as isolated tissue perfusion, to exiert local therapeutic effects. Intravenous injection is preferred.
  • Effective dosages and schedules for administering the antibody may be determined empirically, and making such determinations is within the skill in the art. Those skilled in the art will understand that the dosage of antibody that must be administered will vary depending on, for example, the mammal that will receive the antibody, the route of administration, the particular type of antibody used and other drugs being administered to the mammal. Guidance in selecting appropriate doses for antibody is found in the literature on therapeutic uses of antibodies, e.g., Handbook of Monoclonal Antibodies, Ferrone et al., eds., Noges Publications, Park Ridge, N. J., 1985, ch. 22 ⁇ nd pp. 303-357; Smith et al.
  • a typical daily dosage of the antibody used alone might range £rom about 1 ⁇ g/kg to up to 100 mg/kg of body weight or more per day, depending on the factors mentioned above.
  • any of the following doses may be used: a dose of at least about 50 mg/kg body weight; at least about 10 mg/kg body weight; at least about 3 mg/kg body weight; at least about 1 mg/kg body weight; at least about 750 ⁇ g/kg body weight; at least about 500 ⁇ g/kg body weight; at least about 250 ug/kg body weight; at least about 100 ⁇ g /kg body weight; at least about 50 ⁇ g /kg body weight; at least about 10 ug /kg body weight; at least about 1 ⁇ g/kg body weight, or more, is administered.
  • Antibodies may be administered at lower doses or less frequent at the beginning of the treatment to avoid potential side effect, such as temporary cerebral amyloid angiopathy (CAA).
  • CAA temporary cerebral amyloid angiopathy
  • compositions may contain at least one, at least two, at least three, at least four, at least five different antibodies (including polypeptides) of the invention.
  • the antibody may also be administered to the mammal in combination with effective amounts of one or more other therapeutic agents.
  • the antibody may be administered sequentially or concurrently with the one or more other therapeutic agents.
  • the amounts of antibody and therapeutic agent depend, for example, on what type of drugs are used, the pathological condition being treated, and the scheduling and routes of administration but would generally be less than if each were used individually.
  • a polynucleotide encoding an antibody or a polypeptide described herein may also be used for delivery and expression of the antibody or the polypeptide in a desired cell. It is apparent that an expression vector can be used to direct expression of the antibody.
  • the expression vector can be administered systemically, intraperitoneally, intravenously, intramuscularly, subcutaneously, intrathecally, intraventricularly, orally, enterally, parenterally, intranasally, dermally, or by inhalation.
  • administration of expression vectors includes local or systemic administration, including injection, oral administration, particle gun or catheterized administration, and topical administration.
  • One skilled in the art is familiar with administration of expression vectors to obtain expression of an exogenous protein in vivo. See, e.g., U.S. Patent Nos. 6,436,908; 6,413,942; and 6,376,471.
  • Targeted delivery of therapeutic compositions comprising a polynucleotide encoding an antibody of the invention can also be used.
  • Receptor-mediated DNA delivery techniques are described in, for example, Findeis et al., Trends Biotechnol. (1993) 11 :202; Chio ⁇ et al., Gene Therapeutics: Methods And Applications Of Direct Gene Transfer (J.A. Wolff, ed.) (1994); Wu et al., J. Biol. Chem. (1988) 263:621; Wu et al., J. Biol Chem. (1994) 269:542; Zenke et al, Proc. Natl. Acad. Sd.
  • compositions containing a polynucleotide are administered in a range of about 100 ng to about 200 mg of DNA for local administration in a gene therapy protocol. Concentration ranges of about 500 ng to about 50 mg, about 1 ⁇ g to about 2 mg, about 5 ⁇ g to about 500 ⁇ g, and about 20 ⁇ g to about 100 ⁇ g of DNA can also be used during a gene therapy protocol.
  • the therapeutic polynucleotides and polypeptides of the present invention can be delivered using gene delivery vehicles.
  • the gene delivery vehicle can be of viral or non- viral origin (see generally, Jolly, Cancer Gene Therapy (1994) 1 :51; Kirnura, Human Gene Therapy (1994) 5:845; Connelly, Human Gene Therapy (1995) 1:185; and Kaplitt, Nature Genetics (1994) 6:148). Expression of such coding sequences can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence can be either constitutive or regulated.
  • Viral-based vectors for delivery of a desired polynucleotide and expression in a desired cell are well known in the art.
  • Exemplary viral-based vehicles include, but are not limited to, recombinant retroviruses (see, e.g., PCT Publication Nos. WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; WO 93/11230; WO 93/10218; WO 91/02805; U.S. Patent Nos. 5, 219,740; 4,777,127; GB Patent No.
  • alphavirus-based vectors e.g., Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR- 1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249; ATCC VR-532)
  • AAV adeno-associated virus
  • Non-viral delivery vehicles and methods can also be employed, including, but not limited to, polycationic condensed DNA linked or unlinked to killed adenovirus alone (see, e.g., Curiel, Hum. Gene Ther. (1992) 3:147); ligand-linked DNAfcee, e.g., Wu, J Biol. Chem. (1 989) 264: 16985); eukaryotic cell delivery vehicles cells (see, e.g., U.S. Patent No. 5,814,482; PCT Publication Nos.
  • WO 95/07994 WO 96/17072; WO 95/30763; and WO 97/423378
  • nucleic charge neutralization or fusion with cell membranes can also be employed.
  • Exemplary naked DNA introduction methods are described in PCT Publication No. WO 90/11092 and U.S. Patent No. 5,580,859.
  • Liposomes that can act as gene delivery vehicles are described in U.S. Patent No. 5,422,120; PCT Publication Nos. WO 95/13796; WO 94/23697; WO 91/14445; and EP 0 524 968. Additional approaches are described in Philip, MoI Cell Biol (1994) 14:2411, and in Woffendin, Proc. Natl. Acad. Sci. (1994) 91 :1581.
  • the invention also provides articles of manufacture and kits containing materials useful for treating pathological conditions described herein, such as Alzheimer's disease or other A ⁇ -associated diseases (such as Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, vascular disorder caused by deposit of A ⁇ peptide in blood vessels (such as stroke and HCHWA-D)), or detecting or purifying A ⁇ or ⁇ APP.
  • the article of manufacture comprises a container with a label. Suitable containers include, for example, bottles, vials, and test tubes. The containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition having an active agent which is effective for treating pathological conditions or for detecting or purifying A ⁇ or ⁇ APP.
  • the active agent in the composition is an antibody and preferably, comprises monoclonal antibodies specific for A ⁇ or ⁇ APP.
  • the active agent comprises antibody 9TL or any antibodies or polypeptides derived from antibody 9TL.
  • the active agent comprises an anti-A ⁇ antibody or polypeptide having impaired effector function.
  • the anti-A ⁇ antibody or polypeptide comprises a heavy chain constant region, wherein the constant region has impaired effector function.
  • the label on the container indicates that the composition is used for treating pathological conditions such as Alzheimer's disease or detecting or purifying A ⁇ or ⁇ APP, and may also indicate directions for either in vivo or in vitro use, such as those described above.
  • kits comprising any of the antibodies (such as 9TL), polypeptides, polynucleotides described herein.
  • the kit of the invention comprises the container described above.
  • the kit of the invention comprises the container described above and a second container comprising a buffer. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for performing any methods described herein (such as methods for treating Alzheimer's disease, and methods for inhibiting or reducing accumulation of A ⁇ peptide in the brain).
  • kits to be used for detecting or purifying A ⁇ or ⁇ APP the antibody is typically labeled with a detectable marker, such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme.
  • a detectable marker such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme.
  • the invention provides compositions (described herein) for use in any of the methods described herein, whether in the context of use as a medicament and/or use for manufacture of a medicament.
  • HBS-P 10 mM HEPES buffer pH 7.4, 150 mM NaCl, 0.005% P20. Lysis of HBS-P resuspended cells was attained by one cycle of freezing (-80 0 C) then thawing at 37°C. Cell lysates were centrifuged at 5000 rpm for 30 min to separate cell debris from supernatants containing Fabs. The supernatants were then injected into the BIAcore plasmon resonance apparatus to obtain affinity information for each Fab. Clones expressing Fabs were rescued from the master plate to sequence the DNA and for large scale Fab production and detailed characterization as described below.
  • Fabs were expressed and purified from large cultures. Erlenmeyer flasks containing 200 mL of LB+Ampicillin (50 ⁇ g/ml) + 2% Glucose were inoculated with 5 mL of over night culture from a selected Fab-expressing E. coli clone. Clones were incubated at 30°C until an OD 550nm of 1.0 was attained and then induced by replacing the media for 200 ml, of LB+Ampicillin (50 ⁇ g/ml) + 1 mM IPTG.
  • Vector pDb.9TL.hFc2a is an expression vector comprising the heavy chain of the
  • Vector pDb.9TL.hFc2a has nucleotide sequences corresponding to the following regions: the murine cytomegalovirus promoter region (nucleotides 1-612); a synthetic intron (nucleotides 619-1507); the DHFR coding region (nucleotides 707-1267); human growth hormone signal peptide (nucleotides 1525-1602); heavy chain variable region of 9TL (nucleotides 1603-1951); human heavy chain IgG2a constant region containing the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence; see Eur.
  • Vector pDb.9TL.hFc2a was deposited at the ATCC on July 20, 2004, and was assigned ATCC Accession No. PTA-6124.
  • Vector pEb.9TL.hK is an expression vector comprising the light chain of the 9TL antibody, and is suitable for transient expression of the light chain.
  • Vector pEb.9TL.hK has nucleotide sequences corresponding to the following regions: the murine cytomegalovirus promoter region (nucleotides 1-612); human EF-I intron (nucleotides 619-1142); human growth hormone signal peptide (nucleotides 1173-1150); antibody 9TL light chain variable region (nucleotides 1251-1593); human kappa chain constant region (nucleotides 1594-1914); SV40 late polyadenylation signal (nucleotides 1932-2175); SV40 enhancer region (nucleotides 2176- 2421); phage fl region (nucleotides 2509-2964) and beta lactamase (AmpR) coding region (nucleotides 3401-4258).
  • Vector ⁇ Eb.9TL.hK was deposited at the ATCC on July 20, 2004, and was assigned ATCC Accession No. PTA-6125.
  • Affinities of 9TL monoclonal antibody were determined using the BlAcore3000TM surface plasmon resonance (SPR) system (BIAcore, INC, Piscaway NJ).
  • SPR surface plasmon resonance
  • One way of determining the affinity was immobilizing of 9TL on CM5 chip and measuring binding kinetics of A ⁇ 1-4 o peptide to the antibody.
  • CM5 chips were activated with N-ethyl-N'-(3-dimethylaminopropyl)- carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions.
  • EDC N-ethyl-N'-(3-dimethylaminopropyl)- carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Antibody 9TL or its variants was diluted into 10 mM sodium acetate pH 4.0 or 5.0 and injected over the activated chip at a concentration of 0.005 mg/mL. Using variable flow time across the individual chip channels, a range of antibody density was achieved: 1000-2000 or 2000-3000 response units (RU). The chip was blocked with ethanolamine. Regeneration studies showed that a solution containing 2 volumes of PIERCE elution buffer and 1 volumes of 4 M NaCl effectively removed the bound A ⁇ 1-40 peptide while keeping the activity of 9TL on the chip for over 200 injections.
  • HBS-EP buffer (0.0 IM HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20) was used as running buffer for all the BIAcore assays.
  • Serial dilutions (0.1-1Ox estimated K D ) of purified Ap 1-40 synthetic peptide samples were injected for 1 min at 100 ⁇ L/min and dissociation times of 10 min were allowed.
  • Kinetic association rates (Ic 0n ) and dissociation rates (k off ) were obtained simultaneously by fitting the data to a 1 : 1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program.
  • Equilibrium dissociation constant (KD) values were calculated as kWk o n.
  • affinity was determined by immobilizing A ⁇ i -40 peptide on SA chip and measuring binding kinetics of 9TL Fab and Fab of 9TL variants to the immobilized A PM 0 peptide. Affinities of 9TL Fab fragment and its variants Fab fragments were determined by Surface Plasmon Resonance (SPR) system (BIAcore 3000TM, BIAcore, Inc., Piscaway, NJ). SA chips (streptavidin) were used according to the supplier's instructions.
  • SPR Surface Plasmon Resonance
  • Biotinylated A ⁇ peptide 1-40 was diluted into HBS-EP (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% P20) and injected over the chip at a concentration of 0.005 mg/mL.
  • HBS-EP 10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% P20
  • two ranges of antigen density were achieved: 10-200 response units (RU) for detailed kinetic studies and 500-600 RU for concentration studies and screening.
  • Regeneration studies showed that 100 mM phosphoric acid (may also be followed by a solution containing 2 volumes of 50 mM NaOH and 1 volume of 70% ethanol) effectively removed the bound Fab while keeping the activity of A ⁇ peptide on the chip for over 200 injections.
  • HBS-EP buffer was used as running buffer for all the BIAcore assays.
  • Serial dilutions (0.1-1Ox estimated KD) of purified Fab samples were injected for 2 min at 100 ⁇ L/min and dissociation times of 10 min were allowed.
  • the concentrations of the Fab proteins were determined by ELISA and/or SDS-PAGE electrophoresis using a standard Fab of known concentration (determined by amino acid analysis).
  • Kinetic association rates (Ic 0n ) and dissociation rates (k off ) were obtained simultaneously by fitting the data to a 1 : 1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program.
  • Equilibrium dissociation constant (KD) values were calculated as IWk 0n .
  • I AIl CDRs are extended CDRs including both Kabat and Chotbia CDRs. Amino acid residues are numbered sequentially.
  • Example 2 Characterization of epitope on A ⁇ l-40 peptide that antibody 9TL binds [0268] To determine the epitope on A ⁇ polypeptide that is recognized by antibody 9TL,
  • a ⁇ 1-40 polypeptide coupled to biotin was immobilized on a streptavidin-coated chip (SA chip).
  • SA chip streptavidin-coated chip
  • a ⁇ antibodies Fab fragments at 50 nM
  • a ⁇ o ⁇ i the absence or presence of different soluble fragments of the A ⁇ peptide (at 10 ⁇ M, from American Peptide Company Inc., CA).
  • Amino acid sequences of A ⁇ 1-40 , A ⁇ i- 42 , and A ⁇ i. 43 are shown in below in Table 4.
  • the A ⁇ peptides which displaced binding of antibody 9TL Fab fragment to A ⁇ 1-4 o were A ⁇ 28 - 40 , A ⁇ M o, A ⁇ 33-4 o, and A ⁇ 17-40 , respectively ( Figure 2).
  • antibody 9TL binds to a C-terminal peptide (33-40) of A ⁇ 1-40 .
  • the A ⁇ i -28 , A ⁇ 28- 42> A ⁇ 22 - 35 , A ⁇ i- 16 , A ⁇ 1-43 , and A ⁇ i-38 peptide did not inhibit the binding of antibody 9TL Fab fragment, suggesting that antibody 9TL binds to the C-terminus of A ⁇ o peptide.
  • a ⁇ 28 - 4 2 and A ⁇ 1-43 peptide did not inhibit binding of antibody 9TL to
  • mice were iminunized with 25-100 ⁇ g of a peptide (amino acid 28-40 of Ap 1-40 ) conjugated to KLH in adjuvant (50 ⁇ l per footpad, 100 ⁇ l total per mouse) at about 16 consecutive week intervals as described in Geerligs HJ et al., 1989, J. Immunol. Methods 124:95- 102; Kenney JS et al., 1989, J. Immunol. Methods 121 :157-166; and Wicher K et al., 1989, Int. Arch. Allergy Appl. Immunol. 89:128-135.
  • a peptide amino acid 28-40 of Ap 1-40 conjugated to KLH in adjuvant
  • mice were first immunized with 50 ⁇ g of the peptide in CFA (complete Freud's adjuvant). After 21 days, mice were secondly immunized with 25 ⁇ g of the peptide in IFA (incomplete Freud's adjuvant). Twenty three days later after the second immunization, third immunization was performed with 25 ⁇ g of the peptide in IFA. Ten days later, antibody titers were tested using ELISA. Forth immunization was performed with 25 ⁇ g of the peptide in IFA 34 days after the third immunization. Final booster was performed with 100 ⁇ g soluble peptide 32 days after the forth immunization.
  • ⁇ Splenocytes were obtained from the immunized mouse and fused with NSO myeloma cells at a ratio of 10: 1 , with polyethylene glycol 1500, The hybrids were plated out into 96-well plates in DMEM containing 20% horse serum and 2-oxaloacetate/pyruvate/insulin (Sigma), and;hypoxanthine/a-minopterin/thymidine selection was begun. On day 8, 100 ⁇ l of DMEM containing 20% horse serum was added to all the wells. Supernatants of the hybrids were screened by using antibody capture immunoassay. Determination of antibody class was done with class-specific second antibodies. [0272] A panel of monoclonal antibody-producing cell lines was selected for characterization. One cell line selected produces as antibody designated 2H6. This antibody was determined to have IgG2b heavy chain.
  • the affinity of antibody 2H6 to A ⁇ i- 4 o was determined.
  • Monoclonal antibody 2H6 was purified from supernatants of hybridoma cultures using protein A affinity chromatography. The supernatants was equilibrated to pH 8. The supernatants " were then loaded to the protein A column MabSelect (Amersham Biosciences # 17-5199-02) equilibrated with PBS to pH 8. The column was washed with 5 column volumes of PBS, pH 8. Tlie antibody was eluted with 50 mM citrate-phosphate buffer, pH 3. The eluted antibody was neutralized with IM Phosphate Buffer, pH 8. The purified antibody was dialyzed with PBS. The antibody concentration was determined by SDS-PAGE, using a murine mAb standard curve.
  • Affinities of 2H6 monoclonal antibody were determined using the BlAcore3000TM surface plasmon resonance (SPR) system (BIAcore, INC, Piscaway NJ).
  • SPR surface plasmon resonance
  • One way of determining the affinity was immobilizing 2H6 antibody on CM5 chip and measuring binding kinetics of A ⁇ ⁇ - 4 o peptide to the antibody.
  • CM5 chips were activated with N-ethyl-N'-(3-dimethylaminopropyl)- carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions.
  • EDC N-ethyl-N'-(3-dimethylaminopropyl)- carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • 2H6 monoclonal antibody was diluted into 10 mM sodium acetate pH 4.0 or 5.0 and injected over the activated chip at a concentration of 0.005 mg/mL. Using variable flow time across the individual chip channels, a range of antibody density was achieved: 1000-2000 or 2000-3000 response units (RU). The chip was blocked with ethanolamine. Regeneration studies showed that a mixture of Pierce elution buffer (Product No. 21004, Pierce Biotechnology, Rockford, IL) and 4 M NaCl (2:1) effectively removed the bound A ⁇ i- 4 0 peptide while keeping the activity of 2H6 antibody on the chip for over 200 injections.
  • Pierce elution buffer Product No. 21004, Pierce Biotechnology, Rockford, IL
  • 4 M NaCl (2:1) effectively removed the bound A ⁇ i- 4 0 peptide while keeping the activity of 2H6 antibody on the chip for over 200 injections.
  • HBS-EP buffer (0.01M HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20) was used as running buffer for all the BIAcore assays.
  • Serial dilutions (0.1-1Ox estimated KD) of purified APi -40 synthetic peptide samples were injected for 1 min at 100 ⁇ L/min and dissociation times of 10 min were allowed.
  • Kinetic association rates (Ij 0n ) and dissociation rates (k off ) were obtained simultaneously by fitting the data to a 1:1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program.
  • Equilibrium dissociation constant (K D ) values were calculated as k O fi/kon-
  • affinity was determined by immobilizing A ⁇ 1-4 o peptide on SA chip and measuring binding kinetics of 2H6 Fab to the immobilized A ⁇ i- 4 o peptide. Affinities of 2H6 Fab fragment was determined by Surface Plasmon Resonance (SPR) system (BIAcore 3000TM, BIAcore, In ⁇ , Piscaway, NJ). SA chips (streptavidin) were used according to the supplier's instructions.
  • SPR Surface Plasmon Resonance
  • Biotinylated A ⁇ peptide 1-40 (SEQ ID NO: 15) was diluted into HBS-EP (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% P20) and injected over the chip at a concentration of 0.005 mg/mL. Using variable flow time across the individual chip channels, two ranges of antigen density were achieved: 10-200 response units (RU) for detailed kinetic studies and 500-600 RU for concentration studies. Regeneration studies showed that a mixture of Pierce elution buffer and 4 M NaCl (2:1) effectively removed the bound Fab while keeping the activity of A ⁇ peptide on the chip for over 200 injections.
  • HBS-EP buffer was used as running buffer for all the BIAcore assays.
  • Serial dilutions (0.1-1Ox estimated KD) of purified Fab samples were injected for 2 min at 100 ⁇ L/min and dissociation times of 10 min were allowed.
  • the concentrations of the Fab proteins were determined by ELISA and/or SDS-PAGE electrophoresis using a standard Fab of known concentration (determined by amino acid analysis).
  • Kinetic association rates (k on ) and dissociation rates (koff) were obtained simultaneously by fitting the data to a 1:1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program.
  • Equilibrium dissociation constant (KD) values were calculated as koff/kcm- The affinity of 2H6 antibody determined using both methods described above is shown in Table 5 below.
  • a ⁇ 1-4 o polypeptide (SEQ ID NO: 15) coupled to biotin (Global Peptide Services, CO) was immobilized on a streptavidin- coated chip (SA chip).
  • SA chip streptavidin- coated chip
  • the A ⁇ peptides which displaced binding of antibody 2H6 to A ⁇ i-w were A ⁇ 17 - 40 , A ⁇ 33-40 , and A ⁇ 1-40 , respectively ( Figure 3).
  • antibody 2H6 binds to a C-terminal peptide (33-40) of A ⁇ 1-40 .
  • this C-terminal peptide (33-40) of A ⁇ 1-40 did not displace binding of antibody 2286 to A ⁇ 1-40 at the concentration tested.
  • the A ⁇ 1-38 peptide did not inhibit the binding of antibody 2H6 or antibody 2286 to A ⁇ M o, suggesting that, similar to antibody 2286, the epitope that antibody 2H6 binds includes amino acids 39 and/or 40 of the A ⁇ 1-4 o peptide ( Figure 3).
  • a ⁇ 1-4 o variants in which each of the last 6 amino acids (A ⁇ 1-4 o amino acid residues 35-40) was individually replaced by an alanine (alanine scanning mutagenesis), were generated by site directed mutagenesis. These A ⁇ 1-4 o variants (sequences shown in Tab' Ie 6) were expressed in E.
  • Glutathione-S-Transferase (GST) fusion proteins (Amersham Pharmacia Biotech, Piscataway, NJ USA) followed by affinity purification on a Glutathione-Agarose beads (Sigma-Aldrich Corp., St. Louis, MO, USA).
  • GST Glutathione-S-Transferase
  • Wild-type (WT) A ⁇ M o as well as A ⁇ i-_u, Ap 1-42, , and Ap 1-39 were also expressed as GST fusion proteins. A ⁇ i.
  • assay plates were incubated with 100 ⁇ l of 0.03 ⁇ g/ml per well of Biotin-conjugated Goat-anti-Mouse (H+L) antibody (Vector Laboratories, vector #BA-9200, Burllingame CA, USA) followed by 100 ⁇ l of 0.025 ⁇ g/ml per well of HRP- conjugated Streptavidin (Aniersham Biosciences Corp., #RPN4401V, NJ, USA). The absorbance of the plate was read at 450 nm.
  • H+L Biotin-conjugated Goat-anti-Mouse
  • HRP- conjugated Streptavidin Aniersham Biosciences Corp., #RPN4401V, NJ, USA
  • a ⁇ recognized all variants with the same intensity and served as internal positive control of protein concentration and protein integrity on the plate.
  • Antibody 2H6 did not recognize A ⁇ im, Ap 1-39 , or A ⁇ M2 as shown in Fig. 4.
  • a ⁇ 1-40 variants V40A, V39A, G38A, G37A, V36A, and M35A showed reduced binding to antibody 2H6, demonstrating that antibody 2H6 epitope extended for at least 6 amino acids at the C terminal end of Ap 1-4O .
  • Antibody density was 1625 response units (RU) for 2H6; 4000 RU for 9TL; and 2200 RU for 2289. Each channel was blocked with ethanolamine. A ⁇ 1-40 peptide (150 uM) was flowed onto the chip for 2 min. Then antibody 2H6 (to be tested for competition of binding) at 0.6 uM was flowed onto the chip for 1 min. HBS-EP buffer (0.01M HEPES, pH 7.4, 0.15 MNaCl, 3 mM EDTA, 0.005% Surfactant P20) was used as running buffer for all the BIAcore assays.
  • Antibody 2H6 does not bind to APP
  • 2H6 to cells transfected with wildtype APP was determined.
  • 293 cells were transfected with a cDNA encoding wild type human amyloid precursor protein. Forty eight hours after the transfection, cells were incubated oh ice for 45 minutes with monoclonal antibodies anti-A ⁇ i.i 6 , anti-A ⁇ 16-28 , or 2H6 (5 ug/ml in DMEM with 10% FCS). The cells were then washed three times in PBS for 5 minutes, fixed with 4% PFA. The cells were washed three times again in PBS, and antibody binding was detected with secondary Cy3 -conjugated goat anti-mouse antibody (dilution of 1:500) from Jackson Immunoresearch under fluorescence microscope.
  • Anti-A ⁇ i -16 and anti-A ⁇ 16 - 28 antibodies which recognize N-terminal or central epitopes in A ⁇ , both showed significant binding to APP precursor proteins expressed on cells. In contrast, 2H6 did not bind to APP expressing cells.
  • Example 4 Reversal of cognitive deficits and histological symptoms with less microhemorrhage in an animal model of Alzheimer's disease by administration of deglycosylated,2H6 A.
  • mice over-expressing the "Swedish" mutant amyloid precursor protein (APP Tg2576 with K670N/M671; Hsiao et al., Science 274:99- 102 (1996)) were used for the experiments.
  • the Alzheimer's-like phenotype present in these mice has been well-characterized. Holcomb et al., Nat. Med. 4:97-100 (1998); Holcomb et al., Behav. Gen. 29:177-185 (1999); and McGowan E, Neurobiol. Dis. 6:231-244 (1999).
  • APP-transgenic mice, aged 20 months were assigned to one of the four groups.
  • mice from the study were subjected to a two-day radial-arm water-maze paradigm as described previously.
  • Wilcock et al. J Neuroinflammation 1 :24 (2004).
  • the apparatus was a 6-arm maze as described previously.
  • Gordon et al. Neurobiol. Aging 22:377-385 (2001).
  • 15 trials were run in three blocks of 5.
  • a cohort of 4 mice were run sequentially for each block (i.e., each of 4 mice get trial one, then the same mice get trial two, etc.).
  • a second cohort of mice was run permitting an extended rest period before mice were exposed to the second block of 5 trials.
  • the goal arm was different for each mouse in a cohort to minimize odor cues.
  • the start arm was varied for each trial, with the goal arm remaining constant for a given individual for both days.
  • the platform was alternately visible then hidden (hidden for the last 4 trials).
  • the mice were run in exactly the same manner as day one except that the platform was hidden for all trials.
  • the number of errors was measured in a one-minute time frame. Mice failing to make an arm choice in 20 seconds were assigned one error, but no mice in this study had to be assigned an error in this manner. Due to the numbers of mice in the study, the tester was unaware of treatment group identity of each mouse.
  • mice were weighed, overdosed with 100 mg/kg Nembutal (Abbott laboratories, North Chicago, IL), and then intracardially perfused with 25 mL of 0.9% sodium chloride.
  • a series of 8 equally spaced tissue sections 600 ⁇ apart were randomly selected spanning the entire brain and stained using free- floating immunohistochemistry for total A ⁇ (rabbit polyclonal anti-pan A ⁇ ; Biosource, Camarillo, CA, 1:10,000) as previously described. Gordon et al., Exp. Neurol. 173:183-195 (2002); Wilcock et al., J. Neurosci. 24:6144-6151 (2004).
  • a second series of tissue sections 600 ⁇ m apart were stained using 0.2% Congo red in NaCl-saturated 80% ethanol.
  • Detection was done using a streptavidin-horseradish peroxidase conjugate (Amersham Biosciences), followed by TMB substrate (KPL, Gaithersburg, MD).
  • a ⁇ i_ 40 American Peptide scaling from 6-400 pM were used for standard curves.
  • Nontransgenic normal mice including 2 mice treated with 2H6 antibody and 4 mice treated with anti-AMN antibody; these two groups were combined since no behavior difference was observed) were also tested in the two-day version of the radial-arm water maze.
  • APP- transgenic mice treated with the control antibody (anti-AMN) failed to learn platform location over two days of testing and were significantly impaired compared to the nontransgenic mice as previously described. Wilcock et al., J Neuroinflammation 1:24 (2004).
  • APP- transgenic mice administered the anti-A ⁇ antibody 2H6 and deglycosylated 2H6 demonstrated a significant reversal of the impairment observed in the control-treated APP-transgenic mice, ending day trwo with a mean performance near 1 error per trial (Fig. 5).
  • the control-treated APP- transgenic mice has a mean performance near 3 errors per trial at the ending of day two (Fig. 5).
  • the data sho ⁇ vn in Fig. 5 indicate that the deglycosylated antibody works as well as the intact antibody for reversal of cognitive deficits in the APP-transgenic mice.
  • Table 8 Total A ⁇ load for frontal cortex after 16 weeks of antibody treatment. Mean percent area occupied by positive immunohistochemical stain for A ⁇ , and standard deviation and standard error of the mean for the frontal cortex are shown.
  • parenchymal Congo-red staining in the hippocampus was also significantly reduced after 16 weeks immunotherapy with antibody 2H6 (about 96% reduction, p ⁇ 0.0001) and deglycosylated 2H6 (about 63% reduction, pO.OOOl) as. compared to the control antibody-treated group (anti-AMN).
  • the deglycosylated 2H6 was less effective than the intact 2H6 antibody in reducing Congo-red load in the frontal cortex and hippocampus.
  • Serum level of A ⁇ after administration of deglycosylated 2H6 antibody As shown in Fig. 9, administration of both antibody 2H6 and deglycosylated 2H6 to the APP Tg2576 mice significantly increased serum level of beta-amyloid peptide. However, no significant increase of serum level of beta-amyloid peptide was observed in the APP Tg2576 mice after administration of anti-AMN antibody or in the wild type mice after administration of anti-AMN antibody or antibody 2H6. This indicates that the increase of serum level of beta-amyloid peptide may be used to assist disgnosis of AD and monitor the response to AD therapy, such as immunotherapy.
  • Binding affinity of the antibody Fc regions to Fc ⁇ receptors or complement were measured using BIAcore as described above. Briefly, purified human or murine Fc ⁇ receptors (from R&D Systems) and human CIq (from Quidel) were immobilized on BIAcore CM5 chip by amine chemistry. Serial dilutions of monoclonal antibodies (ranging from 2 nM to the maximum concentration as indicated in Tables 16 and 17) were injected. HBS-EP (0.01M HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20) as running and sample buffer.
  • Binding data were analyzed using 1 : 1 langmuir interaction model for high affinity interactions, or steady state affinity model for low affinity interactions.
  • Table 16 shows the binding affinity of the anti- ⁇ -amyloid antibodies as measured by KD (nM) to murine Fc ⁇ PJ, Fc ⁇ PJIb, Fc ⁇ PJII, and human CIq (hClcL).
  • Deglycosylated antibodies have a constant region with removed N-glycosylation.
  • 9TL(MgGl) and 9TL(hIgG2 ⁇ a) have the same variable region (shown in SEQ ID NO: 1 and SEQ ID NO:2), but different constant region.
  • 9TL(MgGl) has a human IgGl constant region; and 9TL(MgG2 ⁇ a) has a human IgG2a with mutations of A330P331 to S330S33 1 (Kabat amino acid numbering with reference to the wildtype IgG2a sequence).
  • deglycosylated 2H6, 2294, and 2286 had reduced affinity to all murine Fc ⁇ receptors tested as compared to each corresponding antibody without removed N-glycosylation.
  • Deglycosylated 2H6 also had reduced affinity to human complement as compared to 2H6.
  • 9TL(MgGl) had no significant binding to mFc ⁇ RIIb or mFc ⁇ RIII; and 9TL(hIgG2 ⁇ a) had no significant binding to any of murine Fc ⁇ RI, Fc ⁇ RIIb, Fc ⁇ RIII, or hClq.
  • Table 17 below shows the binding affinity of the anti- ⁇ -amyloid antibodies as measured by K 0 (nM) to human Fc ⁇ RI, Fc ⁇ RIIb/c, Fc ⁇ RIIIb, and hClq.
  • Example 6 Effect of deglycosylated 2H6 antibody on microglial activation, Fc ⁇ receptor binding, and amyloid clearance after intracranial administration
  • Surgical procedure and intracranial administration of antibodies Tg2576 transgenic mice aged 19.5 months were assigned to one of the three groups, all groups received intracranial injections into the frontal cortex and hippocampus.
  • the first group received anti-A ⁇ antibody 2H6 at a concentration of 2 ⁇ g/2 ⁇ l in each region.
  • the second group received deglycosylated 2H6 antibody at 2 ⁇ g/2 ⁇ l in each region.
  • the third group received IgG directed against drosophila amnesiac protein as a control for nonspecific aspects of intact IgG injection.
  • mice Tg2576 transgenic mice
  • mice were weighed, anesthetized with isoflurane and placed in a stereotaxic apparatus (51603 dual manipulator lab standard, Stoelting, Wood Dale, IL).
  • a midsagittal incision was made to expose the cranium and two burr holes were drilled using a dental drill over the right frontal cortex and hippocampus to the following coordinates: Cortex: AP +1.5 mm, L -2.0 mm, hippocampus: AP -2.7 mm, L -2.5 mm, all taken from bregma.
  • a 26 gauge needle attached to a 10 ⁇ l Hamilton (Reno, NV) syringe was lowered 3 mm ventral to bregma and a 2 ⁇ l injection was made over a 2 minute period.
  • the incision was cleaned with saline and closed with surgical staples.
  • Biotinylated goat anti-rat at 1 : 3, 000 dilution was used as the secondary antibody for anti-CD45 and Fc ⁇ receptor antibody staining.
  • immunostaining some sections were omitted from the primary antibody to assess non-specific immunohistochemical reactions. Adjacent sections were mounted on slides and stained using 4% thiofiavine-S (Sigma- Aldrich, St Louis MO) for 10 minutes. It should be noted that there were a limited number of sections that include the injection volume.
  • Data analysis The immunohistochemical reaction product on all stained sections was measured using a videometric Vl 50 image analysis system (Oncor, San Diego, CA) in the injected area of cortex and hippocampus and corresponding regions on the contralateral side of the brain.
  • CD45 staining in the frontal cortex and the hippocampus was about the same after intracranial administration of deglycosylated 2H6 antibody as the control antibody.
  • CD45 staining in the frontal cortex was significantly higher (p ⁇ 0.01) and was generally higher in the hippocampus after intracranial administration of 2H6 antibody than the control antibody. This indicates that, unlike antibody 2H6, administration of deglycosylated 2H6 did not activate microglia in the frontal cortex and the hippocampus 72 hours after the administration.
  • Fc ⁇ ll and Fc ⁇ lll receptor staining in the frontal cortex and the hippocampus was about the same after intracranial administration of deglycosylated 2H6 antibody as the control antibody.
  • Fc ⁇ receptor staining in the frontal cortex and the hippocampus was significantly higher (p ⁇ 0.01) after intracranial administration of 2H6 antibody than the control antibody. This indicates that, unlike antibody 2H6, administration of deglycosylated 2H6 did not activate microglia in the frontal cortex and the hippocampus 72 hours after the administration.
  • a ⁇ staining was lower in frontal cortex and hippocampus 72 hours after intracranial administration of 2H6 antibody or deglycosylated 2H6 antibody as compared to the control antibody.
  • thioflavin-S stained compact plaque was also lower in frontal cortex and hippocampus 72 hours after intracranial administration of 2H6 antibody or deglycosylated 2H6 antibody as compared to the control antibody.
  • Antibody 2294 is a murine antibody raised by immunizing a mouse with A ⁇ i.
  • Binding affinity for antibody 2294 to A ⁇ 1-40 , A ⁇ 1-42 , or A ⁇ 22 - 37 was measured using Biacore as described above. Table 18 below shows the affinity of antibody 2294 Fab fragment to various A ⁇ peptides.
  • antibody 2294 binds to A ⁇ peptides with amino acids 20-34, 21-35, 22-36, 23-37, 24-38, 25-39, 26-40, and 25-34 with a glycine at the C-terminus; but does not bind to A ⁇ peptides with amino acids 19-33, 27-41, 24-33, and 27-35 having a glycine at the C-terminus of these peptides.
  • the epitope of antibody 2294 binds includes amino acids from 26 to 34.
  • Table 19 below shows binding comparison of 2294 to A ⁇ 1-4 o to other A ⁇ peptide as measured by Biacore assay.
  • Antibody 2294 full antibody
  • a ⁇ 4 o has the strongest binding to A ⁇ 4 o as compared to other peptides, with significantly lower binding to truncated A ⁇ 1-40 (such as 1-36, 1-37, 1-38, and 1-39), A ⁇ 1-42 and A ⁇ 1-43 .
  • Table 19 shows binding comparison of 2294 to A ⁇ 1-4 o to other A ⁇ peptide as measured by Biacore assay.
  • Antibody 2294 full antibody
  • a ⁇ 4 o has significantly lower binding to truncated A ⁇ 1-40 (such as 1-36, 1-37, 1-38, and 1-39), A ⁇ 1-42 and A ⁇ 1-43 .
  • the epitope that antibody 2294 binds seems to include amino acids 26-34 and 40.
  • Antibody 2294 binds to an epitope very similar to antibody 6G described in U.S. provisional application serial no. 60/676,093 (the amino acid and nucleic acid sequences of this antibody is shown in SEQ ID NOS:36-39; vectors encoding 6G are deposited at American Type Culture Collection on June 15, 2005, with accession numbers PTA-6786 and PTA-6787).
  • the epitope that antibody 6G binds includes amino acids from 25 to 34, and 40.
  • the epitope comparison of antibody 2294 and 6G are shown in Figure 14.
  • Antibody 6G Fab fragment binds to A ⁇ 1-42 with Ic 0n (1/Ms) of 1.8 x 10 4 , k off (1/s) of 1.6 x 10 "3 , and K 0 of 80 nM.
  • Antibody 6G Fab fragment binds to A ⁇ 22 - 37 with Ic 0n (I/Ms) of 3.6 x 10 5 , k off (1/s) of 3.9 x 10 " 3 , and K D of 11 nM.
  • Antibody 6G has significant higher affinity to A ⁇ i -42 , and A ⁇ 22-37 than antibody 2294. Data indicate that binding of antibody 6G is less dependent on amino acid 40 than antibody 2294.
  • Biacore assay were performed as described in Example 3. Competition experiments were performed using Biacore assay.
  • Antibody 2294, 6G, 2H6, and 2289 were immobilized on different channels of a CM5 chip. CM5 chip channels were activated with N-ethyl-N'-(3- dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions.
  • EDC N-ethyl-N'-(3- dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • a ⁇ 1-40 peptide (150 uM) was flowed onto the chip for 2 min. Then antibody 2294 (to be tested for competition of binding) at 0.6 uM was flowed onto the chip for 1 min.
  • HBS-EP buffer (0.0 IM HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20) was used as running buffer for all the BIAcore assays. After measuring binding of A ⁇ 1-40 , all channels of the chip were regenerated by washing twice with a mixture of Pierce elution buffer (Product No. 21004, Pierce Biotechnology, Rockford, IL) and 4 M NaCl (2:1) for 6 sec.
  • Example 8 Effect of antibody 2294 and deglycosylated antibody 2294 in reducing A ⁇ deposit and cognition in animal model of Alzheimer's disease
  • Deglyeosylated antibody 2294 was prepared as by incubating purified antibody
  • Tg2576 for effect on reversal of cognitive deficits, histological symptoms, and microhemorrhage as described in Example 4.
  • transgenic mice (aged 20 months) were assigned to were assigned to one of the four groups.
  • the first group received weekly intraperitoneal anti-A ⁇ antibody 2294 injections for a period of 16 weeks (n - 4).
  • Vector pEb.9TL.hK is a polynucleotide encoding the 9TL light chain variable region and the light chain kappa constant region
  • vector pDb.9TL.hFc2a is a polynucleotide encoding the 9TL heavy chain variable region and the heavy chain IgG2a constant region containing the following mutations: A330P331 to S33OS331 (amino acid numbering with reference to the wildtype IgG2a sequence; see Eur. J. Immunol. (1999) 29:2613-2624).
  • Vector pEb.6G.hK is a polynucleotide encoding the 6G light chain variable region and the light chain kappa constant region
  • vector pDb.6G.hFc2a is a polynucleotide encoding the 6G heavy chain variable region and the heavy chain IgG2a constant region containing the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence; see Eur. J. Immunol. (1999) 29:2613-2624).
  • 9TL light chain full antibody amino acid sequence (SEO ID NO: 12) DVVMTQSPLSLPVTLGQP ASISCKSSQSLLYSDAKTYLNWFQQRPGQSPRULIYQISR LDPGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCLQGTHYPVLFGQGTRXEIKRTV AAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQD SKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
  • 6G heavy chain full antibody amino acid sequence (Including modified IgG2a as described herein) fSEO ID NQ-.36)

Abstract

Monoclonal antibody 9TL and antibodies derived from 9TL directed against amyloid-beta peptide and methods of using same for diagnosing and treatment of Alzheimer's disease and Aβ peptide associated diseases are described. Methods of using antibodies directed against amyloid-beta peptide having impaired effector function for treatment of Alzheimer's disease and Aβ peptide associated diseases are also described.

Description

ANTIBODIES DIRECTED AGAINST AMYLOID-BETA PEPTIDE AND METHODS
USING SAME
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the priority benefit of U.S. provisional application serial nos. 60/592,494, filed July 30, 2004; 60/653,197, filed February 14, 2005; and 60/676,093, filed April 29, 2005; all of which are incorporated herein by reference in their entirety.
FIELD OF THE INVENTION
[0002] The invention concerns antibodies to amyloid-beta peptide. The invention further concerns use of such antibodies in the treatment and/or prevention of diseases, such as Alzheimer's disease.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT [0003] Not applicable.
BACKGROUND OF THE INVENTION
[0004] Alzheimer's disease (AD) is a degenerative brain disorder characterized clinically by progressive memory deficits, confusion, gradual physical deterioration and, ultimately, death. Approximately 15 million people worldwide are affected by Alzheimer's disease, and the number is expected to increase dramatically as lifespan increases. Histologically, the disease is characterized by neuritic plaques, found primarily in the association cortex, limbic system and basal ganglia. The major constituent of these plaques is amyloid beta peptide (Aβ), which is the cleavage product of beta amyloid precursor protein (βAPP or APP). APP is a type I transmembrane glycoprotein that contains a large ectopic N-terminal domain, a transmembrane domain, and a small cytoplasmic C-terminal tail. Alternative splicing of the transcript of the single APP gene on chromosome 21 results in several isoforms that differ in the number of amino acids.
[0005] Aβ appears to have a central role in the neuropathology of Alzheimer's disease.
Familial forms of the disease have been linked to mutations in APP and the presenilin genes (Tanzi et al.. 1996, Neurobiol. Dis. 3:159-168; Hardy, 1996, Ann. Med. 28:255-258). Diseased- linked mutations in these genes result in increased production of the 42-amino acid form of Aβ, the predominant form found in amyloid plaques. Moreover, immunization of transgenic mice that overexpress a disease-linked mutant form of APP with human Aβ reduces plaque burden and associated pathologies (Schenk et al., 1999, Nature 400:173-177; WO 99/27944), and peripheral administration of antibodies directed against Aβ also reduces plaque burden in the brain (Bard et al., 2000, Nature Medicine 6(8):916-919; WO 2004/032868; WO 00/72880). [0006] It has been reported that Fc-mediated phagocytosis by microglial cells and/or macrophages is important to the process of plaque clearance in vivo. Bard et al., Proc. Natl. Acad. ScL USA 100, 2023-2028 (2003). However, it has also been reported that non-Fc-mediated mechanisms are involved in clearance of amyloid-β in vivo by immunotherapy. Bacskai et al., J. Neurosci. 22:7873-7878 (2002); Das et al., J Neurosci. 23:8532-8538 (2003). [0007] Antibody therapy therefore provides a promising approach to the treatment and prevention of Alzheimer's disease. However, human clinical trials with a vaccine including Aβl- 42 were suspended due to meningoencephalititis in a subset of patients. Orgogozo et al., Neruology 61 :7-8 (2003); Ferrer et al., Brain Pathol. 14:1 1-20 (2004). It has been reported that passive immunization with an N-terminal specific anti-A(3 antibody results in a significant reduction of mainly diffuse amyloid, but induces an increase of cerebral microhemorrhage frequency in transgenic mice that exhibit the age-related development of amyloid plaques and neurodegenef ation as well as cerebral amyloid angiopathy (CAA) similar to that observed in the human AD brain. Pfeifer et al., Science 298:1379 (2002). It has been suggested that exacerbation of cerebral amyloid angiopathy (CAA)-associated microhemorrhage in APP transgenic mice by passive immunization with antibody directed to beta-amyloid is dependent on antibody recognition of deposited forms of amyloid beta peptide. Racke et al., J Neurosci. 25:629-636 (2005). Passive immunization with antibodies against a peptide component of an amyloid deposit, which antibodies are devoid of Fc regions, has been suggested in order to decrease the risk of inflammation. WO 03/086310. There remains a need for antibodies and other immunotherapeutic agents directed against Aβ having improved efficacy and safety profile, and which are suitable for use with human patients. [0008] Throughout this application various publications (including patents and patent applications) are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference.
BRIEF SUMMARY OF THE INVENTION Section I
[0009] The present invention provides methods for treating a disease characterized by aberrant deposition of a protein in the brain of a subject. The methods comprise administering to the subject an effective amount of a pharmaceutical composition comprising an antibody that specifically binds to the protein or the protein deposit, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
[0010] The invention also provides methods for treating or preventing diseases associated with amyloid deposit of Aβ (e.g., deposit in the brain tissue and cerebral vasculature) in a subject, such as Alzheimer's disease, Down's syndrome, multi-infarct dementia, mild cognitive impairment, and cerebral amyloid angiopathy. The method comprises administering to the subject an effective amount of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function. [0011] The invention also provides methods of delaying development of a symptom associated with diseases associated with amyloid deposit of Aβ in a subject, such as Alzheimer's disease, comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
[0012] The invention also provides methods of suppressing formation of amyloid plaques and/or amyloid accumulation in a subject comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta- amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function. In some embodiments, the amyloid plaques are in the brain (brain tissue) of the subject. In some embodimeats, the amyloid plaques are in the cerebral vasculature. In some embodiments, the amyloid accumulation is in the circulatory system.
[0013] The invention also provides methods of reducing amyloid plaques and/or amyloid accumulation in a subject comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function. In some embodiments, the amyloid plaques are in the brain (brain tissue) of the subject. In some embodiments, the amyloid plaques are in the cerebral vasculature. In some embodiments, the amyloid accumulation is in the circulatory system.
[0014] The invention also provides methods of removing or clearing amyloid plaques and/or amyloid accumulation in a subject comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function. In some embodiments, the amyloid plaques are in the brain (brain tissue) of the subject, hi some embodiments, the amyloid plaques are in the cerebral vasculature. In some embodiments, the amyloid accumulation is in the circulatory system.
[0015] The invention also provides methods for inhibiting the accumulation of Aβ peptide in a tissue comprising contacting the tissue with an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, wherein the antibody has impaired effector function.
[0016] The invention also provides methods of reducing Aβ peptide (such as soluble, oligomeric, and deposited form) in a subject comprising administrating to the subject an effective amount of an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function. In some embodiments, the accumulation of Aβ peptide is inhibited and/or reduced in the brain. In some embodiments, the toxic effects of Aβ peptide are inhibited and/or reduced. Thus, the method of the invention can be used to treat any disease in which accumulation of Aβ peptide is present or suspected, such as Alzheimer's disease, Down's syndrome, Parkinson's disease, and multi-infarct dementia.
[0017] The invention also provides methods of improving cognition or reversing cognitive decline associated with diseases associated with amyloid deposit of Aβ in a subject, such as Alzheimer's disease, comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function.
[0018] The invention also provides methods for treating or preventing diseases associated with amyloid deposit of Aβ, comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, wherein the antibody comprises an Fc region with a variation from a naturally occurring Fc region, wherein the variation results in impaired effector function. In some embodiments, the administration of the antibody causes less cerebral microhemorrhage than administration of an antibody without the variation. [0019] Polypeptides that specifically bind to an Aβ peptide or an aggregated form of an Aβ peptide and comprises a heavy chain constant region having impaired effector function may also be used for any of the methods described herein. In some embodiments, the polypeptide comprises a sequence (e.g., one or more CDRs) derived from antibody 9TL or its variants shown in Table 3. In some embodiments, the polypeptide comprises a sequence (e.g., one or more CDRs) derived from antibody 6G.
[0020] The antibody and polypeptide used for the methods of the invention specifically bind to an Aβ peptide or an aggregated form of an Aβ peptide, but have impaired effector function. In some embodiments, the antibody or polypeptide is not a F(ab')2 fragment. In some embodiments, the antibody or polypeptide is not a Fab fragment. In some embodiments, the antibody or polypeptide is not a single chain antibody scFv.
[0021] In some embodiments, the antibody or the polypeptide comprises a heavy chain constant region having impaired effector function, wherein the heavy chain constant region comprises an Fc region, hi some embodiments, the N-glycosylation in the Fc region is removed. In some embodiments, the Fc region comprises a mutation within the N-glycosylation recognition sequence, whereby the Fc region of the antibody or polypeptide is not N-glycosylated. In some embodiments, the Fc region is PEGylated. In some embodiments, the heavy chain constant region of the antibody or the polypeptide is a human heavy chain IgG2a constant region containing the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence). In some embodiments, the antibody or the polypeptide comprises a constant region of IgG4 comprising the following mutations: E233F234L235 to P233V234A235.
[0022] In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 1-16 of Aβ peptide. In some embodiments, the antibody or polypeptide specifically binds to the N-terminus of the Aβ peptide. In some embodiments, the antibody or the polypeptide specifically binds to an epitope within residues 16-28 of Aβ peptide. In some embodiments, the antibody specifically binds to an epitope on the C-terminal side of an Aβ peptide, such as an epitope starting from amino acid 25 or later. The antibody may specifically bind to the free C-terminus amino acid of C-terminus truncated Aβ peptide, for example, Aβ 1- 37, 1-38, 1-39, 1-40, 1-41, 1-42, 1-43. In some embodiments, the antibody or the polypeptide specifically binds to an epitope within residues 28-40 of Aβ1-4o peptide. In some embodiments, the antibody or the polypeptide specifically binds to an epitope within residues 28-42 of Aβ1-42 peptide. In some embodiments, the antibody or the polypeptide specifically binds to an epitope within residues 28-43 of Aβ1-43 peptide. In some embodiments, the antibody or the polypeptide specifically binds to Aβ peptide without binding to full-length amyloid precursor protein (APP). In some embodiments, the antibody or the polypeptide specifically binds to the aggregated form of Aβ without binding to the soluble form. In some embodiments, the antibody or the polypeptide specifically binds to the soluble form of Aβ without binding to the aggregated form. In some embodiments, the antibody or the polypeptide specifically binds to both aggregated form and soluble forms of Aβ.
[0023] In some embodiments, the antibody or the polypeptide specifically binds to a C- terminal peptide 33-40 of Aβ1-40. In some embodiments, the antibody or the polypeptide specifically binds to an epitope on Aβ1-40 that includes amino acid 35-40. In some embodiments, the antibody or the polypeptide specifically binds to an epitope on Aβ1-40 that includes amino acid 36-40. In some embodiments, the antibody or the polypeptide specifically binds to an epitope on Aβ1-40 that includes amino acid 39 and/or 40. In some embodiments, the antibody or the polypeptide specifically binds to Aβ1-40 but do not specifically bind to Aβ1-42 and/or Ap1-43. In some embodiments, the antibody comprises the variable region of antibody 9TL or an antibody derived from 9TL described herein. In some embodiments, the antibody or polypeptide competitively inhibits^binding of antibody 9TL and/or antibody or polypeptide derived from 9TL to Aβ1-40.
[0024] In some embodiments, the antibody or the polypeptide binds to Aβ1-4o with higher affinity than its binding to Aβ1-42 and Aβ1-43. In some embodiments, the antibody binds to an epitope on Aβ1-40 that includes amino acids 25-34 and 40. In some embodiments, the antibody comprises the variable region of antibody 6G or an antibody derived from 6G described herein. In some embodiments, the antibody or polypeptide competitively inhibits binding of antibody 6G and/or antibody or polypeptide derived from 6G to Aβ.
[0025] Administration of antibody or polypeptide that specifically binds to an Aβ peptide and has impaired effector function may be by any means known in the art, including: intravenously, subcutaneously, via inhalation, intraarterially, intramuscularly, intracardially, intraventricularly, parenteral, intrathecally, and intraperitoneally. Administration may be systemic, e.g. intravenously, or localized. This also generally applies to polypeptides and polynucleotides of the invention.
[0026] The invention also provides pharmaceutical composition comprising an effective amount of any of the antibodies or polypeptides that specifically bind to an Aβ peptide or an aggregated form of an Aβ peptide and have impaired effector function, or polynucleotides encoding the antibodies or polypeptides, and a pharmaceutical acceptable excipient. [0027] The invention also provides kits and compositions comprising any one or more of the compositions comprising an effective amount of any of the antibodies or polypeptides that specifically bind to an A β peptide or an aggregated form of an A β peptide and have impaired effector function, or polynucleotides encoding the antibodies or polypeptides. These kits, generally in suitable packaging and provided with appropriate instructions, are useful for any of the methods described herein.
[0028] The invention also provides a method of producing a therapeutic humanized antibody for treatment of a disease associated with amyloid deposits of Aβ peptide in the brain of a human subject, comprising selecting a first humanized antibody that specifically binds to Aβ peptide; and altering the Fc region of the antibody to provide a therapeutic humanized antibody having impaired effector function relative to the first humanized antibody.
Section II
[0029] The invention disclosed herein concerns antibodies that bind to C-terminus of Aβi.
4o peptide (SEQ ID NO: 15 shown in Table 4). Accordingly, in one aspect, the invention is an antibody 9TL (interchangeably termed "9TL") that is produced by expression vectors having ATCC Accession Nos. PTA-6124 and PTA-6125. The amino acid sequences of the heavy chain and light chain variable regions of 9TL are shown in Figure 1. The complementarity determining region (CDR) portions of antibody 9TL (including Chothia and Kabat CDRs) are also shown in Figure 1. It is understood that reference to any part of or entire region of 9TL encompasses sequences produced by the expression vectors having ATCC Accession Nos. PTA-6124 and PTA- 6125, and/or the sequences depicted in Figure 1.
[0030] hi another aspect, the invention also provides antibody variants of 9TL with amino acid sequences depicted in Table 3.
[0031] In another aspect, the invention is an antibody comprising a fragment or a region of the antibody 9TL or its variants shown in Table 3. In one embodiment, the fragment is a light chain of the antibody 9TL. In another embodiment, the fragment is a heavy chain of the antibody 9TL. In yet another embodiment, the fragment contains one or more variable regions from a light chain and/or a heavy chain of the antibody 9TL. In yet another embodiment, the fragment contains one or more variable regions from a light chain and/or a heavy chain shown in Figure 1. In yet another embodiment, the fragment contains one or more CDRs from a light chain and/or a heavy chain of the antibody 9TL.
[0032] In another aspect, the invention provides polypeptides (which may or may not be an antibody) comprising any one or more of the following: a) one or more CDR(s) of antibody 9TL or its variants shown in Table 3; b) CDR H3 from the heavy chain of antibody 9TL or its variants shown in Table 3; c) CDR L3 from the light chain of antibody 9TL or its variants shown in Table 3; d) three CDRs from the light chain of antibody 9TL or its variants shown in Table 3; e) three CDRs from the heavy chain of antibody 9TL or its variants shown in Table 3; f) three CDRs from the light chain and three CDRs from the heavy chain of antibody 9TL or its variants shown in Table 3. The invention further provides polypeptides (which may or may not be an antibody) comprising any one or more of the following: a) one or more (one, two , three, four, five, or six) CDR(s) derived from antibody 9TL or its variants shown in Table 3; b) a CDR derived from, CDR H3 from the heavy chain of antibody 9TL; and/or c) a CDR derived from CDR L3 from the light chain of antibody 9TL. In some embodiments, the CDR is a CDR shown in Figure 1. In some embodiments, the one or more CDRs derived from antibody 9TL or its variants shown in Table 3 are at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% identical to at least one, at least two, at least three, at least four, at least five, or at least six CDRs of 9TL or its variants.
[0033] In some embodiments, the CDR is a Kabat CDR. In other embodiments, the CDR is a Chothia CDR. In other embodiments, the CDR is a combination of a Kabat and a Chothia CDR (also termed "combined CDR" or "extended CDR"). In other words, for any given embodiment containing more than one CDR, the CDRs may be any of Kabat, Chothia, and/or combined.
[0034] In some embodiments, the polypeptide (such as an antibody) comprises an amino acid sequence shown in SEQ ID NO:5, wherein Ll is L, V, or I; wherein Y2 is Y or W; wherein S3 is S, T, or: G; wherein L4 is L, R, A, V, S, T, Q, or E; wherein V6 is V, I, T, P, C, Q, S, N, or F; and wherein Y7 is Y, H, F, W, S, I, V, or A. In some embodiments, the amino acid sequence is a CDR3 in a heavy chain variable region. For convenience herein, "is" in this context or reference to an amino acid refers to choices of amino acid(s) for a given position with reference to the position in the SEQ ID. For example, "Ll is L, V, or I" refers to amino acid L at position 1 in SEQ ID NO: 5 may be substituted with V or I.
[0035] In some embodiments, the polypeptide (such as an antibody) comprises an amino acid sequence shown in SEQ ID NO: 6, wherein Y8 is Y, A, or H; and wherein Al l is A or S; and wherein Kl 2- is K or A. In some embodiments, the amino acid sequence is a CDRl in a light chain variable region. [0036] In some embodiments, the polypeptide (such as an antibody) comprises an amino acid sequence shown in SEQ ID NO:8, wherein Ll is L, M, N, C, F, V, K, S, Q, G, S; wherein G3 is G, S, or T; wherein T4 is T or S; wherein H5 is H or L; wherein Y6 is Y, P, A, W, Q, M, S, or E; wherein V8 is V, L, K, H, T, A, E, or M; and wherein L9 is L, I5 T, S, or V. In some embodiments, the amino acid sequence is a CDR3 in a light chain variable region. [0037] In some embodiments, the polypeptide (such as an antibody) comprises a heavy chain variable region comprising (a) a CDRl region shown in SEQ ID NO:3; (b) a CDR2 region shown in SEQ ID NO:4; and (c) a CDR3 region shown in SEQ ID NO:5, wherein Ll is L, V, or I; wherein "Y2 is Y or W; wherein S3 is S, T, or G; wherein L4 is L, R, A, V, S, T, Q, or E; wherein V6 is V, I, T, P, C, Q, S3 N, or F; and wherein Y7 is Y, H, F, W, S, I, V, or A. [0038] In some embodiments, the polypeptide (such as an antibody) comprises a light chain variable region comprising (a) a CDRl region shown in SEQ ID NO:6, wherein Y8 is Y, A, or H; and wherein Al 1 is A or S; and wherein Kl 2 is K or A; (b) a CDR2 region shown in SEQ ID NO:7; and (c) a CDR3 region shown in SEQ ID NO:8, wherein Ll is L, M, N, C, F, V, K, S, Q, G, S; wherein G3 is G, S, or T; wherein T4 is T or S; wherein H5 is H or L; wherein Y6 is Y, P, A5 W5 Q, M, S5 or E; wherein V8 is V5 L5 K5 H5 T5 A5 E5 or M; and wherein L9 is L5 15 T5 S5 or V.
[0039] In some embodiments, the antibody of the invention is a human antibody. In other embodiments, the antibody of the invention is a humanized antibody. In some embodiments, the antibody is monoclonal. In some embodiments, the antibody (or polypeptide) is isolated. In some embodiments, the antibody (or polypeptide) is substantially pure. [0040] I The heavy chain constant region of the antibodies may be from any types of constant region, such as IgG5 IgM5 IgD5 IgA5 and IgE; and any isotypes, such as IgGl, IgG2, IgG3, and IgG4.
[0041] . In some embodiments, the antibody comprises a modified constant region, such as a constant region that is immunologically inert (which includes partially immunologically inert, and is used interchangeably with the term "having impaired effector function"), e.g.1, does not trigger complement mediated lysis, does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC), or does not activate microglia. In some embodiments, the constant region is modified as described in Eur. J. Immunol. (1999) 29:2613-2624; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No. 9809951.8. In other embodiments, the antibody comprises a human heavy chain IgG2a constant region comprising the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence). Eur. J. Immunol. (1999) 29:2613-2624. In some embodiments, the antibody comprises a constant region of IgG4 comprising the following mutations: E233F234L235 to P233V234A235. In still other embodiments, the constant region is aglycosylated for N-linked glycosylation. In some embodiments, the constant region is aglycosylated for N-linked glycosylation by mutating the oligosaccharide attachment residue (such as Asn297) and/or flanking residues that are part of the N-glycosylation recognition sequence in the constant region . In some embodiments, the constant region is aglycosylated for N-linked glycosylation. The constant region may be aglycosylated for N-linked glycosylation enzymatically or by expression in a glycosylation deficient host cell.
[0042] In another aspect, the invention provides a polynucleotide (which may be isolated) comprising a polynucleotide encoding a fragment or a region of the antibody 9TL or its variants shown in Table 3. In one embodiment, the fragment is a light chain of the antibody 9TL. In another embodiment, the fragment is a heavy chain of the antibody 9TL. In yet another embodiment, the fragment contains one or more variable regions from a light chain and/or a heavy chain of the antibody 9TL. In yet another embodiment, the fragment contains one or more (i.e., one, two, three, four, five, six) complementarity determining regions (CDRs) from a light chain and/or a heavy chain of the antibody 9TL.
[0043] In another aspect, the invention is a polynucleotide (which may be isolated) comprising a polynucleotide that encodes for antibody 9TL or its variants shown in Table 3. In some embodiments, the polynucleotide comprises either or both of the polynucleotides shown in SEQ ID NO:9 and SEQ ID NO: 10.
[0044] In another aspect, the invention provides polynucleotides encoding any of the antibodies (including antibody fragments) or polypeptides described herein. [0045] In another aspect, the invention provides vectors (including expression and cloning vectors) and host cells comprising any of the polynucleotide disclosed herein. In some embodiments, the vector is ρDb.9TL.hFc2a having ATCC No. PTA-6124. In other embodiments, the vector is pEb.9TL.hK having ATCC No. PTA-6125. [0046] In another aspect, the invention is a host cell comprising a polynucleotide encoding any of the antibodies described herein.
[0047] In another aspect, the invention is a complex of Aβi-40 bound by antibody 9TL or its variants shown in Table 3.
[0048] In another aspect, the invention is a complex of Ap1-40 bound by any of the antibodies or polypeptides described herein.
[0049] In another aspect, the invention is a pharmaceutical composition comprising an effective amount of any of the polypeptides (including antibodies, such as an antibody comprising one or more CDRs of antibody 9TL) or polynucleotides described herein, and a pharmaceutically acceptable excipient.
[0050] In another aspect, the invention is a method of generating antibody 9TL comprising culturing a host cell or progeny thereof under conditions that allow production of antibody 9TL, wherein the host cell comprises an expression vector that encodes for antibody
9TL; and, in some embodiments, purifying the antibody 9TL. In some embodiments, the expression vector comprises one or both of the polynucleotide sequences shown in SEQ ID NO: 9 and SEQ ID NO: 10.
[0051] In another aspect, the invention provides methods of generating any of the antibodies or polypeptides described herein by expressing one or more polynucleotides encoding the antibody (which may be separately expressed as a single light or heavy chain, or both a light and a heavy chain are expressed from one vector) or the polypeptide in a suitable cell, generally followed by recovering and/or isolating the antibody or polypeptides of interest.
[0052] The invention also provides a method for preventing, treating, inhibiting, or delaying the development of Alzheimer's disease and other diseases associated with altered Aβ or βAPP expression, or accumulation of Aβ peptide, such as Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, and AIDS. The method comprises administering an effective dosage a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to a subject.
[0053] The invention also provides a method of delaying development of a symptom associated with Alzheimer's disease or other diseases related to accumulation of Aβ peptide in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject.
[0054] The invention also provides a method of suppressing formation of amyloid plaques and/or amyloid accumulation in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject. In some embodiments, the amyloid plaques are in the brain (brain tissue) of the subject. In some embodiments, the amyloid plaques are in the cerebral vasculature. In other embodiments, the amyloid accumulation is in the circulatory system.
[0055] The invention also provides a method of reducing amyloid plaques and/or amyloid accumulation in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject. In some embodiments, the amyloid plaques are in the brain (brain tissue) of the subject, hi some embodiments, the amyloid plaques are in the cerebral vasculature, hi other embodiments, the amyloid accumulation is in the circulatory system.
[0056] The invention also provides a method of removing or clearing amyloid plaques and/or amyloid accumulation in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the subject. In some embodiments, the amyloid plaques are in the brain (brain tissue) of the subject. Li some embodiments, the amyloid plaques are in the cerebral vasculature, hi other embodiments, the amyloid accumulation is in the circulatory system.
[0057] Additionally, the invention provides a method for inhibiting the accumulation of Aβ peptide in a tissue comprising contacting the tissue with an anti~body or a polypeptide of the invention.
[0058] The invention also provides a method of reducing Aβ peptide (such as soluble, oligomeric and deposited form) in the brain of an individual comprising administering to the individual an effective amount of an antibody or a polypeptide of the invention. In some embodiments, the accumulation of Aβ peptide is inhibited and/or reduced in the brain. In some embodiments, the toxic effects of Aβ peptide are inhibited and/or reduced. Thus, the method of the invention can be used to treat any disease in which accumulation of Aβ peptide is present or suspected, such as Alzheimer's disease, Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, and cerebral amyloid angiopathy. [0059] The invention also provides methods of improving cognition or reversing cognitive decline associated with diseases associated with amyloid deposit of Aβ in the brain of an individual, such as Alzheimer's disease, comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide of the invention to the individual.
[0060] Any antibodies, polypeptides, or polynucleotides described herein may be used for the methods of the invention. In some embodiments, the antibody is antibody 9TL. [0061] Antibodies and polypeptides of the invention can further be used in the detection, diagnosis and monitoring of Alzheimer's disease and other diseases associated with altered Aβ or βAPP expression, such as Down's syndrome, and AIDS. The method comprises contacting a specimen of a patient suspected of having altered Aβ or βAPP expression with an antibody of the invention and determining whether the level of Aβ or βAPP differs from that of a control or comparison specimen. In some embodiments, serum level of Aβ is measured before and after administration of an anti-Aβ antibody; and any increase of serum level of Aβ is assessed. [0062] Administration of any antibody or polypeptide of the invention may be by any means known in the art, including: intravenously, subcutaneously, via inhalation, intraarterially, intramuscularly, intracardially, intraventricularly, parenteral, intrathecally, and intraperitoneally. Administration may be systemic, e.g. intravenously, or localized. This also generally applies to polypeptides and polynucleotides of the invention.
[0063] ' In another aspect, the invention provides kits and compositions comprising any one or more of the compositions described herein. These kits, generally in suitable packaging and provided with appropriate instructions, are useful for any of the methods described herein.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING(S) [0064] ; Figure 1 shows the amino acid sequence of the heavy chain variable region (SEQ ID NO:1) and light chain variable region (SEQ ID NO:2) of the 9TL antibody. The Kabat CDRs are in bold text, and the Chothia CDRs are underlined. The amino acid residu.es for the heavy chain and light chain variable region are numbered sequentially. [0065] Figure 2 shows epitope mapping of antibody 9TL by peptide competition. AβMo peptide was immobilized on the SA chip. Monoclonal antibody 2289 and 9TL Fab fragment (50 nM each), each of which was preincubated for 1 h with 10 μM various peptide (amino acids 28- 40, 1-40, 1-28, 28-42, 22-35, 1-16, 1-43, 33-40, 1-38, or 17-40 of Aβ) or no peptide, and was then flowed onto the chip. Binding of the antibody Fab fragment to immobilized Aβ1-40 peptide was measured.
[0066] Figure 3 is a graph showing epitope mapping of antibody 2H6 by peptide competition. Aβ1-40 peptide was immobilized on the SA chip. Monoclonal antibody 2289, 2286, or 2H6 (100 nM each), each of which was preincubated for 1 h with 16 μM various peptide (amino acids 1-16, 1-28, 1-38, 1-40, 1-42, 1-43, 17-40, 17-42, 22-35, 25-35, or 33-40 of Aβ) or no peptide, was flowed onto the chip. Binding of the antibody to immobilized Aβ1-40 peptide was measured.
[0067] Figure 4 is a graph showing binding of antibody 2H6, 2286, and 2289 to different Aβ peptide C-terminal variants. GST-Aβ variants (M35A, V36A, G37A, G38A, V39A, or V40A), or GST-Aβ peptide 1-39, 1-41, 1-40, 1-42 were immobilized on ELISA plate. Monoclonal antibody 2286, 2H6, or 2289 (0.3 nM each niAb) was incubated with each of the immobilized peptides, and their binding was detected by further incubating with biotinylated anti-mouse IgG (H+L) and followed by Sterptavidin-HRP.
[0068] Figure 5 is a graph showing spatial learning deficits in APP -transgenic mice were reversed following 16 weeks of antibody treatment with 2H6 and deglycosylated 2H6. Mice were tested in a two-day version of the radial-arm water maze. Y axis represents mean number of errors made over the 2-day trial period. Block numbers 1-5 represent tests in day 1 ; and block numbers 6-10 represent tests in day 2. "*" indicates p<0.05 for both 2H6 (A-2H6) and deglycosylated 2H6 (A-De-2H6) treated mice when compared with anti-AMN antibody treated mice (A-AMN). "**" indicates pO.Ol for both 2H6 (A-2H6) and deglycosylated 2H6 (A-De- 2H6) treated mice when compared with anti-AMN antibody treated mice (A-AMN). [0069] Figure 6A and 6B are graphs showing decreases of parenchymal Congo-red stained amyloid-beta peptide in hippocampus (Fig.6B) and frontal cortex (Fig. 6A) after 16 weeks of antibody treatment with 2H6, anti-AMN (referred to as AMN), and deglycosylated 2H6 (referred to as D-2H6) antibody. Y-axes in Fig. 6A and 6B represent mean of percent area positive for Congo-red staining. X-axes in Fig.6A and 6B represent type of antibody administered. [0070] Figure 7A and 7B are graphs showing increases of vascular Congo-red stained amyloid-beta peptide in hippocampus (Fig. 7A) and frontal cortex (Fig. 7B) after 16 weeks of antibody treatment with 2H6, anti-AMN (referred to as AMN), and deglycosylated 2H6 (referred to as D-2H6) antibody. Y-axes in Fig.7A and 7B represent mean of percent area positive for Congo-red staining. X-axes in Fig. 7A and 7B represent type of antibody administered. [0071] Figure 8 is a graph showing number of Prussian blue positive profiles after 16 weeks of antibody treatment with 2H6, anti-AMN (referred to as AMN), and deglycosylated 2H6 (referred to as D-2H6) antibody. Y-axis represents positive profiles per section. X-axis represents type of antibody administered.
[0072] Figure 9 is a graph showing serum level of Aβ peptide after administration of anti- AMN antibody (referred to as AMN)5 antibody 2H6 (referred to as 2H6), deglycosylated 2H6 (referred to as 2H6-D) in APP Tg2576 mice, and after administration of anti-AMN antibody and antibody 2H6 in wild type (WT) mice.
[0073] Figure 10 shows immunostaining of CD45 in the hippocampus of a mouse after intracranial administration of 2H6 antibody (A) or deglycosylated 2H6 antibody (B). The bottom panel shows that the ratio of the average area occupied CD45 positive staining of injected side over uninjected side in the frontal cortex and hippocampus after intracranial administration of the control antibody, 2H6 antibody, or deglycosylated 2H6 antibody. "**" indicates P<0.01 as compared to the control antibody.
[0074] Figure 11 shows immunostaining of Fcγ receptor in the hippocampus of a mouse after intracranial administration of 2H6 antibody (A) or deglycosylated 2H6 antibody (B). The bottom panel shows that the ratio of the average area occupied by Fcγ receptor positive staining of injected side over uninjected side in the frontal cortex and hippocampus after intracranial administration of the control antibody, 2H6 antibody, or deglycosylated 2H6 antibody. "**" indicates P<0.01 as compared to the control antibody.
[0075] Figure 12 shows immunostaining of Aβ peptide in the hippocampus of a mouse after intracranial administration of 2H6 antibody (A) or deglycosylated 2H6 antibody (B). The bottom panel shows that the ratio of the average area occupied by Aβ positive staining of injected side over uninjected side in the frontal cortex and hippocampus after intracranial administration of the control antibody, 2H6 antibody, or deglycosylated 2H6 antibody. "**" indicates PO.01 as compared to the control antibody. "*" indicates PO.05 as compared to the control antibody. [0076] Figure 13 shows thioflavine-S in the hippocampus of a mouse after intracranial administration of 2H6 antibody (A) or deglycosylated 2H6 antibody (B). The bottom panel shows that the ratio of the average area occupied by thioflavine-S positive staining of injected side over uninjected side in the frontal cortex and hippocampus after intracranial administration of the control antibody, 2H6 antibody, or deglycosylated 2H6 antibody. "*" indicates P<0.05 as compared to the control antibody.
[0077] Figure 14 shows epitope mapping of antibody 2294 and 6G by ELISA. Various Aβ peptides were immobilized on ELISA plates. Antibodies were incubated for 1 hour with various immobilized peptides. Antibody 6G bound to immobilized Aβ peptides were measured using goat anti-human kappa HRP conjugated secondary antibody. Antibody 2294 bound to immobilized Aβ peptides were measured using goat anti-mouse that binds to both heavy and light chain and is HRP conjugated secondary antibody. "NB" refers to no binding detected. The numbers in the columns under "2294" and "6G" represent absorbance at 450 nm.
DETAILED DESCRIPTION OF THE INVENTION
[0078] The invention disclosed herein provides antibodies and polypeptides that bind to
C-terminus of Aβ1-40. These antibodies and polypeptides are derived from 9TL or its variants shown in Table 3. The invention also provides methods of making and using these antibodies. In some embodiments, the invention provides antibody 9TL, and methods of making and using this antibody. The invention also provides 9TL polypeptides (including antibodies) that bind Aβ1-4o, and polynucleotides encoding 9TL antibody and/or polypeptide.
[0079] The invention disclosed herein also provides methods for preventing and/or treating Aβ-associated diseases, such as Alzheimer's disease, Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, vascular disorder caused by deposit of Aβ peptide in blood vessels (such as stroke and HCHWA- D) in an individual by administration of a therapeutically effective amount of an antibody 9TL, or antibody or polypeptide derived from 9TL. [0080] The invention also provides methods for treating or preventing diseases associated with β-amyloid deposit in an individual, such as Alzheimer's disease, Down's syndrome, multi- infarct dementia, mild cognitive impairment, and cerebral amyloid angiopathy in an individual by administering to the individual an effective amount of a pharmaceutical composition comprising an antibody or a polypeptide that specifically binds to a β-amyloid peptide or an aggregated form of an Aβ peptide, or a polynucleotide encoding the antibody or the polypeptide, wherein the antibody or the polypeptide has impaired effector function.
General Techniques
[0081] The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as, Molecular Cloning: A Laboratory Manual, second edition (Sambrook et al., 1989) Cold Spring Harbor Press; Oligonucleotide Synthesis (MJ. Gait, ed., 1984); Methods in Molecular Biology, Humana Press; Cell Biology: A Laboratory Notebook (J.E. Cellis, ed., 1998) Academic Press; Animal Cell Culture (R. I. Freshney, ed., 1987); Introduction to Cell and Tissue Culture (J.P. Mather and P.E. Roberts, 1998) Plenum Press; Cell and Tissue Culture: Laboratory Procedures (A. Doyle, J.B. Griffiths, and D. G. Newell, eds., 1993-1998) J. Wiley and Sons; Methods in Enzymology (Academic Press, Inc.); Handbook of Experimental Immunology (D .M. Weir and CC. Blackwell, eds.); Gene Transfer Vectors for Mammalian Cells (J.M. Miller and M.P. Calos, eds., 1987); Current Protocols in Molecular Biology (F.M. Ausubel et al., eds., 1987); PCR: The Polymerase Chain Reaction, (Mullis et al., eds., 1994); Current Protocols in Immunology (J.E. Coligan et al., eds., 1991); Short Protocols in Molecular Biology (Wiley and Sons, 1999); Immunobiology (CA. Janeway and P. Travers, 1997); Antibodies (P. Finch, 1997); Antibodies: a practical approach (D. Catty., ed., IRL Press, 1988-1989); Monoclonal antibodies: a practical approach (P. Shepherd and C. Dean, eds., Oxford University Press, 2000); Using antibodies: a laboratory manual (E. Harlow and D. Lane (Cold Spring Harbor Laboratory Press, 1999); The Antibodies (M. Zanetti and J.D. Capra, eds., Harwood Academic Publishers, 1995). Definitions
[0082] An "antibody" is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule. As used herein, the term encompasses not only intact polyclonal or monoclonal antibodies, but also fragments thereof (such as Fab, Fab', F(ab')2, Fv), single chain (ScFv), mutants thereof, fusion proteins comprising an antibody portion, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site. An antibody includes an antibody of any class, such as IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant domain of its heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three- dimensional configurations of different classes of immunoglobulins are well known. [0083] As used herein, "monoclonal antibody" refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler and Milstein, 1975, Nature, 256:495, or may be made by recombinant DNA methods such as described in U.S. Pat. No. 4,816,567. The monoclonal antibodies may also be isolated from phage libraries generated using the techniques described in McCafferty et al., 1990, Nature, 348:552-554, for example.
[0084] As used herein, "humanized" antibodies refer to forms of non-human (e.g. murine) antibodies that are specific chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Antibodies may have Fc regions modified as described in WO 99/58572. Other forms of humanized antibodies have one or more CDRs (one, two, three, four, five, six) which are altered with respect to the original antibody, which are also termed one or more CDRs "derived from" one or more CDRs from the original antibody. [0085] As used herein, "human antibody" means an antibody having an amino acid sequence corresponding to that of an antibody produced by a human and/or has been made using any of the techniques for making human antibodies known in the art or disclosed herein. This definition of a human antibody includes antibodies comprising at least one human heavy chain polypeptide or at least one human light chain polypeptide. One such example is an antibody comprising murine light chain and human heavy chain polypeptides. Human antibodies can be produced using various techniques known in the art. In one embodiment, the human antibody is selected from a phage library, where that phage library expresses human antibodies (Vaughan et al., 1996, Nature Biotechnology, 14:309-314; Sheets et al., 1998, PNAS, (USA) 95:6157-6162; Hoogenboom and Winter, 1991, J. MoI. Biol., 227:381; Marks et al., 1991, J. MoI. Biol., 222:581). Human antibodies can also be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. This approach is described in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661,016. Alternatively, the human antibody may be prepared by immortalizing human B lymphocytes that produce an antibody directed against a target antigen (such B lymphocytes may be recovered from an individual or may have been immunized in vitro). See, e.g., Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985); Boerner et al., 1991, J. Immunol., 147 (l):86-95; and U.S. Patent No. 5,750,373.
[0086] As used herein, the terms "9TL" and "antibody 9TL" are used interchangeably to refer to an antibody produced by expression vectors having deposit numbers of ATCC PTA-6124 and ATCC PTA-6125. The amino acid sequence of the heavy chain and light chain variable regions are shovm in Figure 1. The CDR portions of antibody 9TL (including Chothia and Kabat CDRs) are diagrammatically depicted in Figure 1. The polynucleotides encoding the heavy and light chain variable regions are shown in SEQ ID NO:9 and SEQ ID NO: 10. The characterization of 9TL is described in the Examples.
[0087] The terms "polypeptide", "oligopeptide", "peptide" and "protein" are used interchangeably herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non- amino acids. Tlie terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. It is understood that, because the polypeptides of this invention are based upon an antibody, the polypeptides can occur as single chains or associated chains. [0088] "Polynucleotide," or "nucleic acid," as used interchangeably herein, refer to polymers of nucleotides of any length, and include DNA and RNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and their analogs. If present, modification to the nucleotide structure may be imparted before or after assembly of the polymer. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. Other types of modifications include, for example, "caps", substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, cabamates, etc.) and with charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), those containing pendant moieties, such as, for example, proteins (e.g., nucleases, toxins, antibodies, signal peptides, ply-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as well as unmodified forms of the polynucleotide(s). Further, any of the hydroxyl groups ordinarily present in the sugars may be replaced, for example, by phosphonateigroups, phosphate groups, protected by standard protecting groups, or activated to prepare additional linkages to additional nucleotides, or may be conjugated to solid supports. The 5' and 3' terminal OH can be phosphorylated or substituted with amines or organic capping group moieties of from 1 to 20 carbon atoms. Other hydroxy Is may also be derivatized to standard protecting groups. Polynucleotides can also contain analogous forms of ribose or deoxyribose sugars that are generally known in the art, including, for example, 2'~O-methyl-, T- O-allyl, 2'-fluoro- or 2'-azido-ribose, carbocyclic sugar analogs, α-anomeric sugars, epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, sedoheptuloses, acyclic analogs and abasic nucleoside analogs such as methyl riboside. One or more phosphodiester linkages may be replaced by alternative linking groups. These alternative linking groups include, but are not limited to, embodiments wherein phosphate is replaced by P(O)S("thioate"), P(S)S ("dithioate"), "(O)NR2 ("amidate"), P(O)R, P(O)OR', CO or CH2 ("formacetal"), in which each R or R' is independently H or substituted or unsubstituted alkyl (1- 20 C) optionally containing an ether (-O-) linkage, aryl, alkenyl, cycloalkyl, cycloalkenyl or araldyl. Not all linkages in a polynucleotide need be identical. The preceding description applies to all polynucleotides referred to herein, including RNA and DNA.
[0089] A "variable region" of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination. The variable regions of the heavy and light chain each consist of four framework regions (FR) connected by three complementarity determining regions (CDRs) also known as hypervariable regions. The CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies. There are at least two techniques for determining CDRs: (1) an approach based on cross-species sequence variability (i.e., Kabat et al. Sequences of Proteins of Immunological Interest, (5th ed., 1991, National Institutes of Health, Bethesda MD)); and (2) an approach based on crystallographic studies of antigen-antibody complexes (Al-lazikani et al (1997) J. Molec. Biol. 273:927-948)). As used herein, a CDR may refer to CDRs defined by either approach or by a combination of both approaches.
[0090] A "constant region" of an antibody refers to the constant region of the antibody light chain or the constant region of the antibody heavy chain, either alone or in combination. [0091] An epitope that "preferentially binds" or "specifically binds" (used interchangeably herein) to an antibody or a polypeptide is a term well understood in the art, and methods to determine such specific or preferential binding are also well known in the art. A molecule is said to exhibit "specific binding" or "preferential binding" if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances. An antibody "specifically binds" or "preferentially binds" to a target if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances. For example, an antibody that specifically or preferentially binds to an Aβ1-40 epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other Aβ1-40 epitopes or nbn-Aβ1-4o epitopes. It is also understood by reading this definition that, for example, an antibody (or moiety or epitope) that specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target. As such, "specific binding" or "preferential binding" does not necessarily require (although it can include) exclusive binding. Generally, but not necessarily, reference to binding means preferential binding. [0092] As used herein, "substantially pure" refers to material which is at least 50% pure
(i.e., free from contaminants), more preferably at least 90 % pure, more preferably at least 95% pure, more preferably at least 98% pure, more preferably at least 99% pure. [0093] A "host cell" includes an individual cell or cell culture that can be or has been a recipient for vector(s) for incorporation of polynucleotide inserts. Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation. A host cell includes cells transfected in vivo with a polynucleotide(s) of this invention. [0094] The term "Fc region" is used to define a C-terminal region of an immunoglobulin heavy chain. The "Fc region" may be a native sequence Fc region or a variant Fc region. Although the boundaries of the Fc region of an immunoglobulin heavy chain might vary, the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof. The numbering of the residues in the Fc region is that of the EU index as in Kabat. Kabat et al., Sequences of Proteins of Imunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991. The Fc region of an immunoglobulin generally comprises two constant domains, CH2 and CH3.
[0095] As used herein, "Fc receptor" and "FcR" describe a receptor that binds to the Fc region of an antibody. The preferred FcR is a native sequence human FcR. Moreover, a preferred FcR is one which binds an IgG antibody (a gamma receptor) and includes receptors of the FcγRI, FcγRJI, and FcγRIII subclasses, including allelic variants and alternatively spliced forms of these receptors. FcγRII receptors include FcγRIIA (an "activating receptor") and FcγRIIB (an "inhibiting receptor"), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof. FcRs are reviewed in Ravetch and Kinet, 1991, Ann. Rev. Immunol., 9:457-92; Capel et al., 1994, Immunomethods, 4:25-34; and de Haas et al., 1995, J. Lab. Clin. Med., 126:330-41. "FcR" also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., 1976, J. Immunol., 117:587; and Kim et al., 1994, J. Immunol., 24:249).
[0096] "Complement dependent cytotoxicity" and "CDC" refer to the lysing of a target in the presence of complement. The complement activation pathway is initiated by th.e binding of the first component of the complement system (CIq) to a molecule (e.g. an antibody) complexed with a cognate antigen. To assess complement activation, a CDC assay, e.g. as described in Gazzano-Santoro et al., J. Immunol. Methods, 202:163 (1996), may be performed. [0097] A "functional Fc region" possesses at least one effector function of a native sequence Fc region. Exemplary "effector functions" include CIq binding; complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down-regulation of cell surface receptors (e.g. B cell receptor; BCR), etc. Such effector functions generally require the Fc region to be combined with a binding domain (e.g. an antibody variable domain) and can be assessed using various assays known in the art for evaluating such antibody effector functions.
[0098] A "native sequence Fc region" comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature. A "variant Fc region" comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one amino acid modification, yet retains at least one effector function of the native sequence Fc region. Preferably, the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g. from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide. The variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% sequence identity therewith, more preferably at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99% sequence identity therewith. [0099] As used herein "antibody-dependent cell-mediated cytotoxicity" and "ADCC" refer to a cell-mediated reaction in which nonspecific cytotoxic cells that express Fc receptors (FcRs) (e.g. natural killer (NK) cells, neutrophils, and macrophages) recognize bound antibody on a target cell and subsequently cause lysis of the target cell. ADCC activity of a molecule of interest can be assessed using an in vitro ADCC assay, such as that described in U.S. Patent No. 5,500,362 or 5,821,337. Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and NK cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al., 1998, PNAS (USA), 95:652-656.
[0100] As used herein, an "effective dosage" or "effective amount" drug, compound, or pharmaceutical composition is an amount sufficient to effect beneficial or desired results. For prophylactic use, beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity, or delaying the outset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, beneficial or desired results include clinical results such as inhibiting, suppressing or reducing the formation of amyloid plaques, reducing, removing, clearing amyloid plaques, improving cognition, reversing or slowing cognitive decline, sequestering or increasing soluble Aβ peptide circulating in biological fluids, decreasing one or more symptoms resulting from the disease (biochemical, histological and/or behavioral), including its complications and intermediate pathological phenotypes presenting during development of the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication, delaying the progression of the disease, and/or prolonging survival of patients. An effective dosage can be administered in one or more administrations. For purposes of this invention, an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. As is understood in the clinical context, an effective dosage of a drug, compound, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an "effective dosage" may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved. [0101] As used herein, "treatment" or "treating" is an approach for obtaining beneficial or desired results including clinical results. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: inhibiting, suppressing or reducing the formation of amyloid plaques, reducing, removing, or clearing amyloid plaques, improving cognition, reversing or slowing cognitive decline, sequestering soluble Aβ peptide circulating in biological fluids, reducing Aβ peptide (including soluble, oligomeric and deposited) in a tissue (such as brain), inhibiting, slowing and/or reducing accumulation of Aβ peptide in the brain, inhibiting, slowing and/or reducing toxic effects of Aβ peptide in a tissue (such as brain), decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of patients.
[0102] As used herein, "delaying" development of Alzheimer's disease means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. A method that "delays" development of Alzheimer's disease is a method that reduces probability of disease development in a given time frame and/or reduces extent of the disease in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a statistically significant number of subjects.
[0103] "Development" of Alzheimer's disease means the onset and/or progression of
Alzheimer's disease within an individual. Alzheimer's disease development can be detectable using standard clinical techniques as described herein. However, development also refers to disease progression that may be initially undetectable. For purposes of this invention, progression refers to the biological course of the disease state, in this case, as determined by a standard neurological examination, patient interview, or may be determined by more specialized testing. A variety of these diagnostic tests include, but not limited to, neuroimaging, detecting alterations of levels of specific proteins in the serum or cerebrospinal fluid (e.g., amyloid peptides and Tau), computerized tomography (CT), and magnetic resonance imaging (MRI). "Development" includes occurrence, recurrence, and onset. As used herein "onset" or "occurrence" of Alzheimer's disease includes initial onset and and/or recurrence.
[0104] As used herein, administration "in conjunction" includes simultaneous administration and/or administration at different times. Administration in conjunction also encompasses administration as a co-formulation or administration as separate compositions. As used herein, administration in conjunction is meant to encompass any circumstance wherein an anti-Aβ antibody and another agent are administered to an individual, which can occur simultaneously and/or separately. As further discussed herein, it is understood that an anti-Aβ antibody and the other agent can be administered at different dosing frequencies or intervals. For example, an anti-Aβ antibody can be administered weekly, while the other agent can be administered less frequently. It is understood that the anti-Aβ antibody and the other agent can be administered using the same route of administration or different routes of administration.
[0105] A "biological sample" encompasses a variety of sample types obtained from an individual and can be used in a diagnostic or monitoring assay. The definition encompasses blood and other liquid samples of biological origin, solid tissue samples such as a biopsy specimen or tissue cultures or cells derived therefrom, and the progeny thereof. The definition also includes samples that have been manipulated in any way after their procurement, such as by treatment with reagents, solubilization, or enrichment for certain components, such as proteins or polynucleotides, or embedding in a semi-solid or solid matrix for sectioning purposes. The term "biological sample" encompasses a clinical sample, and also includes' cells in culture, cell supernatants, cell lysates, serum, plasma, biological fluid, and tissue samples.
[0106] An "individual" (alternatively referred to as a "subject") is a mammal, more preferably a human. Mammals also include, but are not limited to, farm animals (such as cows), sport animals, pets (such as cats, dogs, horses), primates, mice and rats. [0107] As used herein, "vector" means a construct, which is capable of delivering, and preferably expressing, one or more gene(s) or sequence(s) of interest in a host cell. Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, DNA or RNA expression vectors encapsulated in liposomes, and certain eukaryotic cells, such as producer cells.
[0108] As used herein, "expression control sequence" means a nucleic acid sequence that directs transcription of a nucleic acid. An expression control sequence can be a promoter, such as a constitutive or an inducible promoter, or an enhancer. The expression control sequence is operably linked to the nucleic acid sequence to be transcribed.
[0109] As used herein, "pharmaceutically acceptable carrier" includes any material which, when combined with an active ingredient, allows the ingredient to retain biological activity and is non-reactive with the subject's immune system. Examples include, but are not limited to, any of the standard pharmaceutical carriers such as a phosphate buffered saline solution, water, emulsions such as oil/water emulsion, and various types of wetting agents.
Preferred diluents for aerosol or parenteral administration are phosphate buffered saline or normal (0.9%) saline. Compositions comprising such carriers are formulated by well known conventional methods (see, for example, Remington's Pharmaceutical Sciences, 18th edition,
A. Gennaro, ed., Mack Publishing Co., Easton, PA, 1990; and Remington, The Science and
Practice of Pharmacy 20th Ed. Mack Publishing, 2000).
[0110] The term "W, as used herein, is intended to refer to the on rate constant for association of an antibody to an antigen.
[0111] The term "koff ", as used herein, is intended to refer to the off rate constant for dissociation of an antibody from the antibody/antigen complex.
[0112] The term "K0", as used herein, is intended to refer to the equilibrium dissociation constant of an antibody-antigen interaction.
Compositions and Methods of Making the Compositions
Antibody 9TL and 9TL derived antibodies and polypeptides
[0113] This invention encompasses compositions, including pharmaceutical compositions, comprising antibody 9TL and its variants shown in Table 3 or polypeptide derived from antibody 9TL and its variants shown in Table 3; and polynucleotides comprising sequences encoding 9TL antibody and its variants or the polypeptide. As used herein, compositions comprise one or more antibodies or polypeptides (which may or may not be an antibody) that bind to C-terminus of Aβ1-40, and/or one or more polynucleotides comprising sequences encoding one or more antibodies or polypeptides that bind to C-terminus of Aβi-4o. These compositions may further comprise suitable excipients, such as pharmaceutically acceptable excipients including buffers, which are well known in the art. [0114] The antibodies and polypeptides of the invention are characterized by any (one or more) of the following characteristics: (a) binds to C-terminal peptide 28-40 of Aβ1-40, but does not significantly bind to Aβl-42 or Aβl-43; (b) binds to C-terminal peptide 33-40 of Aβ1-40; (c) suppresses formation of amyloid plaques in a subject; (d) reduces amyloid plaques in a subject; (e) treats, prevents, ameliorates one or more symptoms of Alzheimer's disease; (f) improves cognitive function. The antibodies and polypeptides of the invention may also exhibit a desirable safety profile in contrast to other reported anti-Aβ antibodies. For example, the compositions of the invention may not cause significant or unacceptable levels of any one or more of: bleeding in the brain vasculature (cerebral hemorrhage); meningoencephalitis (including changing magnetic resonance scan); elevated white blood count in cerebral spinal fluid; central nervous system inflammation. [0115] Accordingly, the invention provides any of the following, or compositions
(including pharmaceutical compositions) comprising any of the following: (a) antibody 9TL or its variants shown in Table 3; (b) a fragment or a region of antibody 9TL or its variants shown in Table 3; (c) a light chain of antibody 9TL or its variants shown in Table 3; (d) a heavy chain of antibody 9TL or its variants shown in Table 3; (e) one or more variable region(s) from a light chain and/or a heavy chain of antibody 9TL or its variants shown in Table 3; (f) one or more CDR(s) (one, two, three, four, five or six CDRs) of antibody 9TL or its variants shown in Table 3; (g) CDR H3 from the heavy chain of antibody 9TL; (h) CDR L3 from the light chain of antibody 9TL or its variants shown in Table 3; (i) three CDRs from the light chain of antibody 9TL or its variants shown in Table 3; (j) three CDRs from the heavy chain of antibody 9TL or its variants shown in Table 3; (k) three CDRs from the light chain and three CDRs from the heavy chain, of antibody 9TL or its variants shown in Table 3; and (1) an antibody comprising any one of (b) through (k). The invention also provides polypeptides comprising any one or more of the above. [0116] The CDR portions of antibody 9TL (including Chothia and Kabat CDRs) are diagrammatically depicted in Figure 1. Determination of CDR regions is well within the skill of the art. It is understood that in some embodiments, CDRs can be a combination of the Kabat and Chothia CDR (also termed "combined CDRs" or "extended CDRs"). In some embodiments, the CDRs are the Kabat CDRs. In other embodiments, the CDRs are the Chothia CDRs. In other words, in embodiments with more than one CDR, the CDRs may be any of Kabat, Chothia, combination CDRs, or combinations thereof.
[0117] In some embodiments, the invention provides a polypeptide (which may or may not be an antibody) which comprises at least one CDR, at least two, at least three, or at least four, at least five, or all six CDRs that are substantially identical to at least one CDR, at least two, at least three, at least four, at least five or all six CDRs of 9TL or its variants shown in Table 3. Other embodiments include antibodies which have at least two, three, four, five, or six CDR(s) that are substantially identical to at least two, three, four, five or six CDRs of 9TL or derived from 9TL. In some embodiments, the at least one, two, three, four, five, or six CDR(s) are at least about 85%, 86%, 87%, 88%, 89%, 90%, 95%, 96%, 97%, 98%, or 99% identical to at least one, two, three, four, five or six CDRs of 9TL or its variants shown in Table 3. It is understood that, for purposes of this invention, binding specificity and/or overall activity is generally retained, although the extent of activity may vary compared to 9TL or its variants shown in Table 3 (may be greater or lesser).
[0118] The invention also provides a polypeptide (which may or may not be an antibody) which comprises an amino acid sequence of 9TL or its variants shown in Table 3 that has any of the following: at least 5 contiguous amino acids, at least 8 contiguous amino acids, at least about 10 contiguous amino acids, at least about 15 contiguous amino acids, at least about 20 contiguous amino acids, at least about 25 contiguous amino acids, at least about 30 contiguous amino acids of a. sequence of 9TL or its variants shown in Table 3, wherein at least 3 of the amino acids are from a variable region of 9TL (Figure 1) or its variants shown in Table 3. In one embodiment, the variable region is from a light chain of 9TL. In another embodiment, the variable region is from a heavy chain of 9TL. An exemplary polypeptide has contiguous amino acid (lengths described above) from both the heavy and light chain variable regions of 9TL. In another embodiment, the 5 (or more) contiguous amino acids are from a complementarity determining region (CDR) of 9TL shown in Figure 1. In some embodiments, the contiguous amino acids are from a variable region of 9TL.
[0119] The binding affinities of the antibodies and polypeptides of the invention may vary, and need not be (but can be) a particular value or range, as the exemplary embodiments described below. The binding affinity of the antibodies and polypeptides of the invention to Aβi_ 40 can be about 0.10 to about 0.80 nM, about 0.15 to about 0.75 nM and about 0.18 to about 0.72 nM. In some embodiments, the binding affinity is about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, about 40 pM, or greater than about 40 pM. In one embodiment, the binding affinity is between about 2 pM and 22 pM. In other embodiments, the binding affinity is less than about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, about 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM. In some embodiment, the binding affinity is about 10 nM. In other embodiments, the binding affinity is less than about 10 nM, less than about 50 nM, less than about 100 nM, less than about 150 nM, less than about 200 nM, less than about 250 nM, less than about 500 nM, or less than about 1000 nM. In other embodiments, the binding affinity is less than about 5 nM. In other embodiments, the binding affinity is less than about 1 nM. In other embodiments, the binding affinity is about 0.1 nM or about 0.07 nM. In other embodiments, the binding affinity is less than about 0.1 nM or less than about 0.07 nM. In other embodiments, the binding affinity is from any of about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, about 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM to any of about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, or about 40 pM. In some embodiments, the binding affinity is any of about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, bout 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM. In still other embodiments, the binding affinity is about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, about 40 pM, or greater than about 40 pM. [0120] The invention also provides methods of making any of these antibodies or polypeptides, The antibodies of this invention can be made by procedures known in the art. The polypeptides can be produced by proteolytic or other degradation of the antibodies, by recombinant methods (i.e., single or fusion polypeptides) as described above or by chemical synthesis. Polypeptides of the antibodies, especially shorter polypeptides up to about 50 amino acids, are conveniently made by chemical synthesis. Methods of chemical synthesis are known in the art and are commercially available. For example, an antibody could be produced by an automated polypeptide synthesizer employing the solid phase method. See also, U.S. Patent Nos. 5,807,715; 4,816,567; and 6,331,415.
[0121] In another alternative, the antibodies can be made recombinantly using procedures that are well known in the art. In one embodiment, a polynucleotide comprises a sequence encoding the heavy chain and/or the light chain variable regions of antibody 9TL shown in SEQ ID NO:9 and SEQ ID NO: 10. In another embodiment, the polynucleotide comprising the nucleotide sequence shown in SEQ ID NO: 9 and SEQ ID NO: 10 are cloned into one or more vectors for expression or propagation. The sequence encoding the antibody of interest may be maintained in a vector in a host cell and the host cell can then be expanded and frozen for future use. Vectors (including expression vectors) and host cells are further described herein. [0122] The invention also encompasses single chain variable region fragments ("scFv") of antibodies of this invention, such as 9TL. Single chain variable region fragments are made by linking light and/or heavy chain variable regions by using a short linking peptide. Bird et al. (1988) Science 242:423-426. An example of a linking peptide is (GGGGS)3 (SEQ ID NO:40) which bridges approximately 3.5 nm between the carboxy terminus of one variable region and the amino terminus of the other variable region. Linkers of other sequences have been designed and used. Bird et al. (1988). Linkers can in turn be modified for additional functions, such as attachment of drugs or attachment to solid supports. The single chain variants can be produced either recombinantly or synthetically. For synthetic production of scFv, an automated synthesizer can be used. For recombinant production of scFv, a suitable plasmid containing polynucleotide that encodes the scFv can be introduced into a suitable host cell, either eukaryotic, such as yeast, plant, insect or mammalian cells, or prokaryotic, such as E. coli. Polynucleotides encoding the scFv of interest can be made by routine manipulations such as ligation of polynucleotides. The resultant scFv can be isolated using standard protein, purification techniques known in the art. [0123] Other forms of single chain antibodies, such as diabodies are also encompassed.
Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al. (1993) Proc. Natl. Acad Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). [0124] For example, bispecific antibodies, monoclonal antibodies that have binding specificities for at least two different antigens, can be prepared using the antibodies disclosed herein. Methods for making bispecific antibodies are known in the art (see, e.g., Suresh et al., 1986, Methods in Enzymology 121 :210). Traditionally, the recombinant production of bispecific antibodies was based on the coexpression of two immunoglobulin heavy chain-light chain pairs, with the two heavy chains having different specificities (Millstein and Cuello, 1983, Nature 305, 537-539).
[0125] According to one approach to making bispecific antibodies, antibody variable domains with the desired binding specificities (antibody-antigen combining sites) are fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2 and CH3 regions. It is preferred to have the first heavy chain constant region (CHl), containing the site necessary for light chain binding, present in at least one of the fusions. DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are cotransfected into a suitable host organism. This provides for great flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yields. It is, however, possible to insert the coding sequences for two or all three polypeptide chains in one expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios are of mo particular significance. [0126] In one approach, the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. This asymmetric structure, with an immunoglobulin light chain in only one half of the bispecific molecule, facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations. This approach is described in PCT Publication No. WO 94/04690, published March 3, 1994.
[0127] Heteroconjugate antibodies, comprising two covalently joined antibodies, are also within the scope of the invention. Such antibodies have been used to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HIV infection (PCT application publication Nos. WO 91/00360 and WO 92/200373; EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents and techniques are well known in the art, and are described in U.S. Patent No. 4,676,980. [0128] Chimeric or hybrid antibodies also may be prepared in vitro using known methods of synthetic protein chemistry, including those involving cross-linking agents. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4- mercaptobutyrimidate.
[0129] Humanized antibody comprising one or more CDRs of antibody 9TL or one or more CDRs derived from antibody 9TL can be made using any methods known in the art. For example, four general steps may be used to humanize a monoclonal antibody. These are: (1) determining the nucleotide and predicted amino acid sequence of the starting antibody light and heavy variable domains (2) designing the humanized antibody, i.e., deciding which antibody framework region to use during the humanizing process (3) the actual humanizing methodologies/techniques and (4) the transfection and expression of the humanized antibody. See, for example, U.S. Patent Nos. 4,816,567; 5,807,715; 5,866,692; 6,331,415; 5,530,101; 5,693,761; 5,693,762; 5,585,089; 6,180,370; 5,225,539; 6,548,640.
[0130] In the recombinant humanized antibodies, the Fc portion can be modified to avoid interaction with Fey receptor and the complement immune system. This type of modification was designed by Dr. Mike Clark from the Department of Pathology at Cambridge University, and techniques for preparation of such antibodies are described in WO 99/58572, published November 18, 1999. [0131] For example, the constant region may be engineered to more resemble human constant regions to avoid immune response if the antibody for use in clinical trials and treatments in humans. See, for example, U.S. Patent Nos. 5,997,867 and 5,866,692.
[0132] The invention encompasses modifications to antibody 9TL, including functionally equivalent antibodies which do not significantly affect their properties and variants whicli have enhanced or decreased activity and/or affinity. For example, the amino acid sequence of antibody 9TL may be mutated to obtain an antibody with the desired binding affinity to Aβ1-40 peptide. Modification of polypeptides is routine practice in the art and need not be described in detail herein. Modification of polypeptides is exemplified in the Examples. Examples of modified polypeptides include polypeptides with conservative substitutions of amino acid residues, one or more deletions or additions of amino acids which do not significantly deleteriously change the functional activity, or use of chemical analogs.
[0133] Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an antibody with an N-terminal methionyl residue or the antibody fused to an epitope tag. Other insertional variants of the antibody molecule include the fusion to the N- or C- terminus of the antibody of an enzyme or a polypeptide which increases the serum half-life of the antibody.
[0134] Substitution variants have at least one amino acid residue in the antibody molecule removed and a different residue inserted in its place. The sites of greatest interest for substitutional mutagenesis include the hypervariable regions, but FR alterations are also contemplated. Conservative substitutions are shown in Table 1 under the heading of "conservative substitutions". If such substitutions result in a change in biological activity, then more substantial changes, denominated "exemplary substitutions" in Table 1, or as further described below in reference to amino acid classes, may be introduced and the products screened.
Table 1 : Amino Acid Substitutions
Figure imgf000037_0001
Figure imgf000038_0001
[0135] Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Naturally occurring residues are divided into groups based on common side-chain properties:
(1) Non-polar: Norleucine, Met, Ala, VaI, Leu, He; (2) Polar without charge: Cys, Ser, Thr, Asn, GIn;
(3) Acidic (negatively charged): Asp, GIu;
(4) Basic (positively charged): Lys, Arg;
(5) Residues that influence chain orientation: GIy, Pro; and
(6) Aromatic: Trp, Tyr, Phe, His.
[0136] Non-conservative substitutions are made by exchanging a member of one of these classes for another class.
[0137] Any cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross-linking. Conversely, cysteine bond(s) may be added to the antibody to improve its stability, particularly where the antibody is an antibody fragment such as an Fv fragment.
[0138] Amino acid modifications can range from changing or modifying one or more amino acids to complete redesign of a region, such as the variable region. Changes in the variable region can alter binding affinity and/or specificity. In some embodiments, no more than one to five conservative amino acid substitutions are made within a CDR domain. In other embodiments, no more than one to three conservative amino acid substitutions are made within a CDR domain. In still other embodiments, the CDR domain is CDR H3 and/or CDR L3. [0139] Modifications also include glycosylated and nonglycosylated polypeptides, as well as polypeptides with other post-translational modifications, such as, for example, glycosylation with different sugars, acetylation, and phosphorylation. Antibodies are glycosylated at conserved positions in their constant regions (Jefferis and Lund, 1997, Chem. Immunol. 65:111-128; Wright and Morrison, 1997, TibTECH 15:26-32). The oligosaccharide side chains of the immunoglobulins affect the protein's function (Boyd et al., 1996, MoI. Immunol. 32:1311-1318; Wittwe and Howard, 1990, Biochem. 29:4175-4180) and the intramolecular interaction between portions of the glycoprotein, which can affect the conformation and presented three-dimensional surface of the glycoprotein (Hefferis and Lund, supra; Wyss and Wagner, 1996, Current Opin. Biotech. 7:409-416). Oligosaccharides may also serve to target a given glycoprotein to certain molecules based upon specific recognition structures. Glycosylation of antibodies has also been reported to affect antibody-dependent cellular cytotoxicity (ADCC). In particular, CHO cells with tetracycline-regulated expression of β(l,4)-N-acetylglucosaminyltransferase III (GnTIII), a glycosyltransferase catalyzing formation of bisecting GIcNAc, was reported to have improved ADCC activity (Umana et al, 1999, Mature Biotech. 17:176-180).
[0140] Glycosylation of antibodies is typically either N-linked or O-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine, asparagine-X-threonine, and asparagine-X- cysteine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain. Thus, the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
[0141] Addition of glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites). The alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
[0142] The glycosylation pattern of antibodies may also be altered without altering the underlying nucleotide sequence. Glycosylation largely depends on the host cell used to express the antibody. Since the cell type used for expression of recombinant glycoproteins, e.g. antibodies, as potential therapeutics is rarely the native cell, variations in the glycosylation pattern of the antibodies can be expected (see, e.g. Hse et al., 1997, J. Biol. Chem. 272:9062- 9070).
[0143] In addition to the choice of host cells, factors that affect glycosylation during recombinant production of antibodies include growth mode, media formulation, culture density, oxygenation, pH, purification schemes and the like. Various methods have been proposed to alter the glycosylation pattern achieved in a particular host organism including introducing or overexpressing certain enzymes involved in oligosaccharide production (U. S. Patent Nos. 5,047,335; 5,510,261 and 5.278,299). Glycosylation, or certain types of glycosylation, can be enzymatically removed from the glycoprotein, for example using endoglycosidase H (Endo H), N-glycosidase F as described in Example 3, endoglycosidase Fl3 endoglycosidase F2, endoglycosidase F3. In addition, the recombinant host cell can be genetically engineered to be defective in processing certain types of polysaccharides. These and similar techniques are well known in the art.
[0144] Other methods of modification include using coupling techniques known in the art, including, but not limited to, enzymatic means, oxidative substitution and chelation. Modifications can be used, for example, for attachment of labels for immunoassay. Modified 9TL polypeptides are made using established procedures in the art and can be screened using standard assays known in the art, some of which are described below and in the Examples. [0145] In some embodiments of the invention, the antibody comprises a modified constant region, such as a constant region that is immunologically inert or partially inert, e.g., does not trigger complement mediated lysis, does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC), or does not activate microglia; or have reduced activities (compared to the unmodified antibody) in any one or more of the following: triggering complement mediated lysis, stimulating antibody-dependent cell mediated cytotoxicity (ADCC), or activating microglia. Different modifications of the constant region may be used to achieve optimal level and/or combination of effector functions. See, for example, Morgan et al., Immunology 86:319-324 (1995); Lund et al., J Immunology 157:4963-9 157:4963-4969 (1996); Idusogie et al., J Immunology 164:4178-4184 (2000); Tao et al., J Immunology 143: 2595-2601 (1989); and Jefferis et al.. Immunological Reviews 163:59-76 (1998). In some embodiments, the constant region is modified as described in Eur. J. Immunol. (1999) 29:2613-2624; PCT Application No. PCT/GB99/01441; and/or UK Patent Application No. 9809951.8 . In other embodiments, the antibody comprises a human heavy chain IgG2a constant region comprising the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence). Eur. J. Immunol. (1999) 29:2613-2624. In still other embodiments, the constant region is aglycosylated for N-linked glycosylation. In some embodiments, the constant region is aglycosylated for N-linked glycosylation by mutating the glycosylated amino acid residue or flanking residues that are part of the N-glycosylation recognition sequence in the constant region. For example^ N-glycosylation site N297 may be mutated to A, Q, K, or H. See, Tao et al., J. Immunology 143: 2595-2601 (1989); and Jefferis et al., Immunological Reviews 163:59-76 (1998). In some embodiments, the constant region is aglycosylated for N-linked glycosylation. The constant region may be aglycosylated for N-linked glycosylation enzymatically (such as removing carbohydrate by enzyme PNGase), or by expression in a glycosylation deficient host cell.
[0146] Other antibody modifications include antibodies that have been modified as described in PCT Publication No. WO 99/58572, published November 18, 1999. These antibodies comprise, in addition to a binding domain directed at the target molecule, an effector domain having an amino acid sequence substantially homologous to all or part of a constant domain of a human immunoglobulin heavy chain. These antibodies are capable of binding the target molecule without triggering significant complement dependent lysis, or cell-mediated destruction of the target. In some embodiments, the effector domain is capable of specifically binding FcRn and/or FcγRIIb. These are typically based on chimeric domains derived from two or more human immunoglobulin heavy chain CH2 domains. Antibodies modified in this manner are particularly suitable for use in chronic antibody therapy, to avoid inflammatory and other adverse reactions to conventional antibody therapy.
[0147] The invention includes affinity matured embodiments. For example, affinity matured antibodies can be produced by procedures known in the art (Marks et al., 1992, Bio/Technology, 10:779-783; Barbas et al., 1994, Proc Nat. Acad. Sci, USA 91 :3809-3813; Schier et al., 1995, Gene, 169:147-155; Yelton et al., 1995, J. Immunol., 155:1994-2004; Jackson et al., 1995, J. Immunol., 154(7):3310-9; Hawkins et al, 1992, J. MoI. Biol., 226:889- 896; and WO2004/058184).
[0148] The following methods may be used for adjusting the affinity of an antibody and for characterizing a CDR. One way of characterizing a CDR of an antibody and/or altering (such as improving) the binding affinity of a polypeptide, such as an antibody, termed "library scanning mutagenesis". Generally, library scanning mutagenesis works as follows. One or more amino acid positions in the CDR are replaced with two or more (such as 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) amino acids using art recognized methods. This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of two or more members (if two or more amino acids are substituted at every position). Generally, the library also includes a clone comprising the native (uns'ubstituted) amino acid. A small number of clones, e.g., about 20-80 clones (depending on the complexity of the library), from each library are screened for binding affinity to the target polypeptide (or other binding target), and candidates with increased, the same, decreased or no binding are identified. Methods for determining binding affinity are well-known in the art. Binding affinity may be determined using BIAcore surface plasmon resonance analysis, which detects differences in binding affinity of about 2-fold or greater. BIAcore is particularly useful when the starting antibody already binds with a relatively high affinity, for example a KD of about 10 nM or lower. Screening using BIAcore surface plasmon resonance is described in the Examples, herein. [0149J Binding affinity may be determined using Kinexa Biocensor, scintillation proximity assays, ELISA, ORIGEN immunoassay (IGEN), fluorescence quenching, fluorescence transfer, and/or yeast display. Binding affinity may also be screened using a suitable bioassay. [0150] In some embodiments, every amino acid position in a CDR is replaced (in some embodiments, one at a time) with all 20 natural amino acids using art recognized mutagenesis methods (some of which are described herein). This generates small libraries of clones (in some embodiments, one for every amino acid position that is analyzed), each with a complexity of 20 members (if all 20 amino acids are substituted at every position).
[0151] In some embodiments, the library to be screened comprises substitutions in two or more positions, which may be in the same CDR or in two or more CDRs. Thus, the library may comprise substitutions in two or more positions in one CDR. The library may comprise substitution in two or more positions in two or more CDRs. The library may comprise substitution in 3, 4, 5, or more positions, said positions found in two, three, four, five or six CDRs. The substitution may be prepared using low redundancy codons. See, e.g., Table 2 of Balint et al. , (1993) Gene 137(l):109-18).
[0152] The CDR may be CDRH3 and/or CDRL3. The CDR may be one or more of
CDRLl, CDRL2, CDRL3, CDRHl, CDRH2, and/or CDRH3. The CDR may be a Kabat CDR, a Chothia CDR, or an extended CDR.
[0153] Candidates with improved binding may be sequenced, thereby identifying a CDR substitution mutant which results in improved affinity (also termed an "improved" substitution). Candidates that bind may also be sequenced, thereby identifying a CDR substitution which retains binding. [0154] Multiple rounds of screening may be conducted. For example, candidates (each comprising an amino acid substitution at one or more position of one or more CDR) with improved binding are also useful for the design of a second library containing at least the original and substituted amino acid at each improved CDR position (i.e., amino acid position in the CDR- at which a substitution mutant showed improved binding). Preparation, and screening or selection of this library is discussed further below.
[0155] Library scanning mutagenesis also provides a means for characterizing a CDR, in so far as the frequency of clones with improved binding, the same binding, decreased binding or no binding also provide information relating to the importance of each amino acid position for the stability of the antibody-antigen complex. For example, if a position of the CDR retains binding when changed to all 20 amino acids, that position is identified as a position that is unlikely to be required for antigen binding. Conversely, if a position of CDR retains binding in only a small percentage of substitutions, that position is identified as a position that is important to CDR function. Thus, the library scanning mutagenesis methods generate information regarding positions in the CDRs that can be changed to many different amino acid (including all 20 amino acids), and positions in the CDRs which cannot be changed or which can only be changed to a few amino acids.
[0156] Candidates with improved affinity may be combined in a second library, which includes the improved amino acid, the original amino acid at that position, and may further include additional substitutions at that position, depending on the complexity of the library that is desired, or permitted using the desired screening or selection method. In addition, if desired, adjacent amino acid position can be randomized to at least two or more amino acids. Randomization of adjacent amino acids may permit additional conformational flexibility in the mutant CDR, which may in turn, permit or facilitate the introduction of a larger number of improving mutations. The library may also comprise substitution at positions that did not show improved affinity in the first round of screening.
[0157] The second library is screened or selected for library members with improved and/or altered binding affinity using any method known in the art, including screening using BIAcore surface plasmon resonance analysis, and selection using any method known in the art for selection, including phage display, yeast display, and ribosome display. [0158] The invention also encompasses fusion proteins comprising one or more fragments or regions from the antibodies (such as 9TL) or polypeptides of this invention. In one embodiment, a fusion polypeptide is provided that comprises at least 10 contiguous amino acids of the variable light chain region shown in SEQ ID NO:2 (Figure 1) and/or at least 10 amino acids of the variable heavy chain region shown in SEQ ID NO:1 (Figure 1). In other embodiments, a fusion polypeptide is provided that comprises at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable light chain region shown in SEQ ID N0:2 (Figure 1) and/or at least about 10, at least about 15, at least about 20, at least about 25, or at least about 30 contiguous amino acids of the variable heavy chain region shown in SEQ ID NO:1 (Figure 1). In another embodiment, the fusion polypeptide comprises a light chain variable region arid/or a heavy chain variable region of 9TL, as shown in SEQ ID NO:2 and SEQ ID NO:1 of Figure 1. In another embodiment, the fusion polypeptide comprises one or more CDR(s) of 9TL. In still other embodiments, the fusion polypeptide comprises CDR H3 and/or CDR L3 of antibody 9TL. For purposes of this invention, an 9TL fusion protein contains one or more 9TL antibodies and another amino acid sequence to which it is not attached in the native molecule, for example, a heterologous sequence or a homologous sequence from another region. Exemplary heterologous sequences include, but are not limited to a "tag" such as a FLAG tag or a 6His tag. Tags are well known in the art. [0159] A 9TL fusion polypeptide can be created by methods known in the art, for example, synthetically or recombinantly. Typically, the 9TL fusion proteins of this invention are made by preparing an expressing a polynucleotide encoding them using recombinant methods described herein, although they may also be prepared by other means known in the art, including, for example, chemical synthesis.
[0160] This invention also provides compositions comprising 9TL antibodies or polypeptides conjugated (for example, linked) to an agent that facilitate coupling to a solid support (such as biotin or avidin). For simplicity, reference will be made generally to 9TL or antibodies with the understanding that these methods apply to any of the Ap1-40 binding embodiments described herein. Conjugation generally refers to linking these components as described herein. The linking (which is generally fixing these components in proximate association at least for administration) can be achieved in any number of ways. For example, a direct reaction between an agent and an antibody is possible when each possesses a substituent capable of reacting with the other. For example, a nucleophilic group, such as an amino or sulfhydryl group, on one may be capable of reacting with a carbonyl-containing group, such as an anhydride or an acid halide, or with an alkyl group containing a good leaving group (e.g., a halide) on the other.
[0161] An antibody or polypeptide of this invention may be linked to a labeling agent
(alternatively termed "label") such as a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art which generally provide (either directly or indirectly) a signal.
[0162] The invention also provides compositions (including pharmaceutical compositions) and kits comprising antibody 9TL, and, a.s this disclosure makes clear, any or all of the antibodies and/or polypeptides described herein.
Anti-Aβ peptide antibodies and polypeptides having impaired effector function [0163] The methods of the invention use antibodies or polypeptides (including pharmaceutical compositions comprising the antibodies or polypeptides) that specifically bind to a beta-amyloid peptide and have impaired effector function. The antibodies and polypeptides are further characterized by any (one or more) of the following characteristics: (a) suppresses formation of amyloid plaques in a subject; (b) reduces amyloid plaques in a subject; (c) treats, prevents, ameliorates one or more symptoms of Alzheimer's disease; (d) improves cognitive function. The antibodies and polypeptides described herein may exhibit a desirable safety profile, for example, the compositions of the invention do not cause significant or unacceptable levels or have a reduced level of any one or more of: bleeding in the brain vasculature (cerebral hemorrhage); meningoencephalitis (including changing magnetic resonance scan); elevated white blood count in cerebral spinal fluid; central nervous system inflammation. As shown in Example 4, an anti-Aβ antibody having N-linked glycosylation removed in the Fc region was effective in removing amyloid plaques in the brain and improving cognitive function with significantly less microhemorrhage than the intact antibody in an animal model for Alzheimer's disease. [0164] As used herein, an antibody or a polypeptide having an "impaired effector function" (used interchangeably with "immunologically inert" or "partially immunologically inert") refers to antibodies or polypeptides that do not have any effector function or have reduced activity or activities of effector function (compared to antibody or polypeptide having an unmodified or a naturally occurring constant region), e.g., having no activity or reduced activity in any one or more of the following: a) triggering complement mediated lysis; b) stimulating antibody-dependent cell mediated cytotoxicity (ADCC); and c) activating microglia. The effector function activity may be reduced by about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, and 100%. In some embodiments, the antibody binds to a beta-amyloid peptide without triggering significant complement dependent lysis, or cell mediated destruction of the target. For example, the Fc receptor binding site on the constant region may be modified or mutated to remove or reduce binding affinity to certain Fc receptors, such as FcγRI, FcγRII, and/or FcγRIII. For simplicity, reference will be made to antibodies with the understanding that embodiments also apply to polypeptides. EU numbering system (Kabat et al., Sequences of Proteins of Immunological Interest; 5th ed. Public Health Service, National Institutes of Healthy, Bethesda, Md., 1991) is used to indicate which amino acid residue(s) of the constant region (e.g., of an IgG antibody) are altered or mutated. The numbering may be used for a specific type of antibody (e.g., IgGl) or a species (e.g., human) with the understanding that similar changes can be made across types of antibodies and species.
O
[0165] In some embodiments, the antibody that specifically binds to the an Aβ peptide comprises a heavy chain constant region having impaired effector function. The heavy chain constant region may have naturally occurring sequence or is a variant. In some embodiments, the amino acid sequence of a naturally occurring heavy chain constant region is mutated, e.g., by amino acid substitution, insertion and/or deletion, whereby the effector function of the constant region is impaired. In some embodiments, the N-glycosylation of the Fc region of a heavy chain constant region may also be changed, e.g., may be removed completely or partially, whereby the effector function of the constant region is impaired.
[0166] In some embodiments, the effector function is impaired by removing N- glycosylation of the Fc region (e.g., in the CH 2 domain of IgG) of the anti-Aβ peptide. In some embodiments, N-glycosylation of the Fc region is removed by mutating the glycosylated amino acid residue or flanking residues that are part of the glycosylation recognition sequence in the constant region. The tripeptide sequences asparagine-X-serine (N-X-S), asparagine-X-thxeonine (N-X-T) and asparagine-X-cysteine (N-X-C), where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain for N-glycosylation. Mutating any of the amino acid in the tripeptide sequences in the constant region yields an aglycosylated IgG. For example, N-glycosylation site N297 of human IgGl and IgG3 may be mutated to A, D, Q, K, or H. See, Tao et al., J. Immunology 143: 2595- 2601 (1989); and Jefferis et al., Immunological Reviews 163:59-76 (1998). It has been reported that human IgGl and IgG3 with substitution of Asn-297 with GIn, His, or Lys do not bind to the human FcγRI and do not activate complement with CIq binding ability completely lost for IgGl and dramatically decreased for IgG3. In some embodiments, the amino acid N in the tripeptide sequences is mutated to any one of amino acid A, C, D, E, F, G, H, I, K, L5 M, P, Q, R, S, T, V5 W, Y. In some embodiments, the amino acid N in the tripeptide sequences is mutated to a conservative substitution. In some embodiments, the amino acid X in the tripeptide sequences is mutated to proline. In some embodiments, the amino acid S in the tripeptide sequences is mutated to Ai D, E, F, G, H, I, K, L, M, N, P, Q, R, V, W, Y. In some embodiments, the amino acid T in the tripeptide sequences is mutated to A5 D, E, F5 G, H, I5 K, L, M5 N, P, Q5 R5 V, W5 Y. In some embodiments, the amino acid C in the tripeptide sequences is mutated to A5 D, E, F, G5 H5 15 K5 L, M5 N, P, Q, R5 V5 W, Y. In some embodiments, the amino acid following the tripeptide is mutated to P. In some embodiments, the N-glycosylation in the constant region is removed enzymatically (such as N-glycosidase F as described in Example 3, endoglycosidase Fl , endoglycosidase F2, endoglycosidase F3, and englycosidase H). Removing N-glycosylation may also be achieved by producing the antibody in a cell line having deficiency for N-glycosylation. Wright et al. J J Immunol. 160(7):3393-402 (1998).
[0167] : In some embodiments, amino acid residue interacting with oligosaccharide attached to the N-glycosylation site of the constant region is mutated to reduce binding affinity to FcγRI. For example, F241, V264, D265 of human IgG3 may be mutated. See, Lund et al., J. Immunology 157:4963-4969 (1996).
[0168] = In some embodiments, the effector function is impaired by modifying regions such as 233-236, 297, and/or 327-331 of human IgG as described in PCT WO 99/58572 and Armoxir et al., Molecular Immunology 40: 585-593 (2003); Reddy et al., J. Immunology 164:1925-1933 (2000). Antibodies described in PCT WO 99/58572 and Armour et al. comprise, in addition to a binding domain directed at the target molecule, an effector domain having an amino acid sequence substantially homologous to all or part of a constant region of a human immunoglobulin heavy chain. These antibodies are capable of binding the target molecule without triggering significant complement dependent lysis, or cell-mediated destruction of the target. In some embodiments, the effector domain has a reduced affinity for FcγRI, FcγRIIa, and FcγRIII. In some embodiments, the effector domain is capable of specifically binding FcRn and/or FcγRIIb. These are typically based on chimeric domains derived from two or more human immunoglobulin heavy chain CR2 domains. Antibodies modified in this manner are particularly suitable for use in chronic antibody therapy, to avoid inflammatory and other adverse reactions to conventional antibody therapy. In some embodiments, the heavy chain constant region of the antibody is a human heavy chain IgGl with any of the following mutations: 1) A327A330P331 to G327S330S331; 2) E233L234L235G236 to P233V234A235 with G236 deleted; 3) E233L234L235 to P233V234A235; 4) E233L234L235G236A327A330P331 to P233V234A235G327S330S331 with G236 deleted; 5) E233L234L235A327A330P331 to P233V234A235G327S330S331; and 6) N297 to A297 or any other amino acid except N. In some embodiments, the heavy chain constant region of the antibody is a human heavy chain IgG2 with the following mutations: A330P331 to S330S331. In some embodiments, the heavy chain constant region of the antibody is a human heavy chain IgG4 with any of the following mutations: E233F234L235G236 to P233V234A235 with G236 deleted; E233F234L235 to P233V234A235; and S228L235 to P228E235.
[0169] The constant region of the antibodies may also be modified to impair complement activation. For example, complement activation of IgG antibodies following binding of the Cl component of complement may be reduced by mutating amino acid residues in the constant region in a Cl binding motif (e.g., CIq binding motif). It has been reported that Ala mutation for each of D270, K322, P329, P331 of human IgGl significantly reduced the ability of the antibody to bind to CIq and activating complement. For murine IgG2b, CIq binding motif constitutes residues E318, K320, and K322. Idusogie et al, J Immunology 164:4178-4184 (2000); Duncan et al., Nature 322: 738-740 (1988).
[0170] CIq binding motif E318, K320, and K322 identified for murine IgG2b is believed to be common for other antibody isotypes. Duncan et al., Nature 322: 738-740 (1988). CIq binding activity for IgG2b 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 CIq binding. It is also possible to use other alkyl-substituted non-ionic residues, such as GIy, He, Leu, or VaI, or such aromatic non-polar residues as Phe, Tyr, Tip and Pro in place of any one of the three residues in order to abolish CIq 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 CIq binding activity. [0171] The invention also provides antibodies having impaired effector function wherein the antibody has a modified hinge region. Binding affinity of human IgG for its Fc receptors can be modulated by modifying the hinge region. Canfield et al., J. Exp. Med. 173:1483-1491 (1991); Hezareh et al., J Virol. 75:12161-12168 (2001); Redpath et al., Human Immunology 59:720-727 (1998). Specific amino acid residues may be mutated or deleted. The modified hinge region may comprise a complete hinge region derived from an antibody of different antibody class or subclass from that of the CHl domain. For example, the constant domain (CHl) of a class IgG antibody can be attached to a hinge region of a class IgG4 antibody. Alternatively, 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. In some embodiments, 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. U.S. Pat. No.5,677,425. Such alterations are carried out using art recognized protein chemistry and, preferably, genetic engineering techniques and as described herein.
[0172] Polypeptides that specifically bind to an Aβ peptide and fused to a heavy chain constant region having impaired effector function may also be used for the methods described herein. In some embodiments, the polypeptide comprises a sequence derived from antibody 9TL or its variants shown in Table 3. In some embodiments, the polypeptide is derived from a single domain antibody that binds to an Aβ peptide. Single domain antibodies can be generated using methods known in the art. Omidfar et al., Tumour Biol 25:296-305 (2004); Herring et al., Trends in Biotechnology 21:484-489 (2003).
[0173] In some embodiments, the antibody or polypeptide is not a F(ab')2 fragment. In some embodiments, the antibody or polypeptide is not a Fab fragment. In some embodiments, the antibody or polypeptide is not a single chain antibody scFv. In some embodiments, the antibody or polypeptide is a PEGylated F(ab')2 fragment. In some embodiments, the antibody or polypeptide is a PEGylated Fab fragment. In some embodiments, the antibody or polypeptide is a PEGylated single chain antibody scFv.
[0174] Other methods to make antibodies having impaired effector function known in the art may also be used.
[0175] Antibodies and polypeptides with modified constant regions can be tested in one or more assays to evaluate level of effector function reduction in biological activity compared to the starting antibody. For example, the ability of the antibody or polypeptide with an altered Fc region to bind complement or Fc receptors (for example, Fc receptors on microglia), or altered hinge region can be assessed using the assays disclosed herein as well as any art recognized assay. PCT WO 99/58572; Armour et al., Molecular Immunology 40: 585-593 (2003); Reddy et al., J. Immunology 164:1925-1933 (2000); Song et al., Infection and Immunity 70:5177-5184 (2002).
[0176] In some embodiments, the antibody that specifically binds to beta-amyloid peptide is a polyclonal antibody. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a human antibody. In some embodiments, the antibody is a chimeric antibody. In some embodiments, the antibody is a humanized antibody. In some embodiments, the antibody is a primatized antibody. See, e.g., Yocum et al., J. Rheumatol. 25:1257-62 (1998); Bugelski et al., Human & Experimental Toxicoloy 19:230-243 (2000). In some embodiments, the antibody is deimmunized by mutation so that the antibody does not activate human immune system. See, e.g., Nanus, et al., J. Urology 170:S84-S89 (2003). [0177] As used herein, Aβ peptide includes any fragments of the enzymatic cleavage products of amyloid precursor protein. For example, Aβ peptide includes any fragments of Aβi. 40, Aβi42, or AβMa; and peptides which are truncated with various number of amino acids at the N-terminus or the C-terminus of Aβ^o, Aβ1-42, or Aβ1-43. Amino acid numbering used herein is based on the numbering for Aβl-43 (SEQ ID NO: 17).
[0178] In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 1-16 of A β peptide. In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 16-28 of Aβ peptide. In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 28-40 of Aβi-4o peptide. In some embodiments, the antibody or polypeptide specifically binds to an epitope within ~ residues 28-42 of Aβ1-42 peptide. In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 28-43 of Ap1-43 peptide. In some embodiments, the antibody or polypeptide specifically binds to an Aβ peptide without binding to full-length amyloid precursor protein (APP). In some embodiments, the antibody or the polypeptide specifically binds to the aggregated form of Aβ without binding to the soluble form. In some embodiments, the antibody or the polypeptide specifically binds to the soluble form of Aβ without binding to the aggregated form. In some embodiments, the antibody or the polypeptide specifically binds to both aggregated form and soluble forms of Aβ. Antibodies that bind to various aggregated form of Aβ are known in the art, for example, antibodies that bind to amyloid beta-derived diffusible ligands (ADDLs); antibodies that bind to amyloid fibrils and/or deposit. WO 03/104437; U.S. Pub. No. 2003/0147887; U.S. Pub. No. 2004/0219146.
[0179] In some embodiments, the antibody or polypeptide comprises one, two, or three
CDRs from the 3D6 immunoglobulin light chain (SEQ ID NO:2 in U.S. Pub. Nos. 2003/0165496, or 2004/0087777), and/or one, two, or three CDRs from the 3D6 immunoglobulin heavy chain (SEQ ID NO:4 in U.S. Pub. Nos. 2003/0165496, or 2004/0087777). In some embodiments, the antibody or polypeptide comprises a variable heavy chain region as set forth in SEQ ID NO:8 in U.S. Pub. No. 2003/0165496 and a variable light chain region as set forth in SEQ ID NO:5 in U.S. Pub. No. 2003/0165496. In some embodiments, the antibody or polypeptide comprises a variable heavy chain region as set forth in SEQ ID NO: 12 in U.S. Pub. No. 2003/0165496 and a variable light chain region as set forth in SEQ ID NO:11 in U.S. Pub. No. 2003/0165496. In some embodiments, the antibody or polypeptide comprises one, two, or three CDRs from the 10D5 immunoglobulin light chain (SEQ ID NO:14 in U.S. Pub. Nos. 2003/0165496, or 2004/0087777), and/or one, two, or three CDRs from the 10D5 immunoglobulin heavy chain (SEQ ID NO:16 in U.S. Pub. Nos. 2003/0165496, or 2004/0087777).
[0180] In some embodiments, the antibody or polypeptide specifically binds to an epitope within residues 33-40 of Aβmo. In some embodiments, the antibody or polypeptide specifically binds to an epitope on Aβ1-40 that includes amino acid 35-40. In some embodiments, the antibody or polypeptide specifically binds to an epitope on APM0 that includes amino acid 36-40. In some embodiments, the antibody or polypeptide specifically binds to an epitope on Aβi-4o that includes amino acid 39 and/or 40. In some embodiments, the antibody or polypeptide specifically binds to Aβi-4o but does not specifically bind to Aβi-42 and/or Aβ1-43. In some embodiments, the antibody or polypeptide is antibody 9TL or an antibody or a polypeptide derived from 9TL described herein. In some embodiments, the antibody or polypeptide competitively inhibits binding of antibody 9TL and/or antibody or polypeptide derived from 9TL to Aβ1-4o. In some embodiments, the antibody is not antibody 2286 described in PCT WO 2004/032868. [0181] The binding affinities of the antibodies and polypeptides of the invention may vary, and need not be (but can be) a particular value or range, as the exemplary embodiments described below. The binding affinity of the antibodies and polypeptides of the invention to Aβ^ 40, Aβi-40, or Aβ1-4ocan be about 0.10 to about 0.80 nM, about 0.15 to about 0.75 nM and about 0.18 to about 0.72 nM. In some embodiments, the binding affinity is about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, about 40 pM, or greater than about 40 pM. In one embodiment, the binding affinity is between about 2 pM and 22 pM. In other embodiments, the binding affinity is less than about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, about 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM; about 30 pM, about 10 pM. In some embodiments, the binding affinity is about 10 nM. In other embodiments, the binding affinity is less than about 10 nM, less than about 50 nM, less than about 100 nM, less than about 150 nM, less than about 200 nM, less than about 250 nM, less than about 500 nM, or less than about 1000 nM. In other embodiments, the binding affinity is less than about 5 nM. In other embodiments, the binding affinity is less than about 1 nM. In other embodiments, the binding affinity is about 0.1 nM or about 0.07 nM. In other embodiments, the binding affinity is less than about 0.1 nM or less than about 0.07 nM. In other embodiments, the binding affinity is from any of about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, about 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM to any of about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, or about 40 pM. In some embodiments, the binding affinity is any of about 10 nM, about 5 nM, about 1 nM, about 900 pM, about 800 pM, bout 700 pM, about 600 pM, about 500 pM, about 400 pM, about 300 pM, about 200 pM, about 150 pM, about 100 pM, about 90 pM, about 80 pM, about 70 pM, about 60 pM, about 50 pM, about 40 pM, about 30 pM, about 10 pM. In still other embodiments, the binding affinity is about 2 pM, about 5 pM, about 10 pM, about 15 pM, about 20 pM, about 40 pM, or greater than about 4O pM.
[0182] Methods of making antibodies and polypeptides are known in the art and described herein.
[0183] Competition assays can be used to determine whether two antibodies bind the same epitope by recognizing identical or sterically overlapping epitopes or one antibody competitively inhibits binding of another antibody to the antigen. These assays are known in the art. Typically, antigen is immobilized on a multi-well plate and the ability of unlabeled antibodies to block the binding of labeled antibodies is measured. Common labels for such competition assays are radioactive labels or enzyme labels.
[0184] Antibodies and polypeptides that specifically bind to Aβ can be screened for efficacy in removing amyloid deposit and other beneficial effects, such as improving cognition. For example, antibodies or polypeptides may be administered to an animal having Alzheimer's pathology. Various animal models for Alzheimer's disease are known in the art. Following administration, level of compact and diffuse amyloid plaques, behavior analysis for cognition, and microglia activation and microhemorrhage may tested using methods known in the art and described in detail in Example 2. PCT WO 2004/032868; Wilcock et al., J Neurosci. 23:3745- 3751 (20O3); Wilcock et al., J. Neuroinflammation 1:24 (2004).
Polynucleotides, vectors and host cells
[0185] The invention also provides isolated polynucleotides encoding the antibodies and polypeptides of the invention (including an antibody comprising the polypeptide sequences of the light chain and heavy chain variable regions shown in Figure 1), and vectors and host cells comprising the polynucleotide.
[0186] Accordingly, the invention provides polynucleotides (or compositions, including pharmaceutical compositions), comprising polynucleotides encoding any of the following: (a) antibody 9TL or its variants shown in Table 3; (b) a fragment or a region of antibody 9TL or its variants shown in Table 3; (c) a light chain of antibody 9TL or its variants shown in Table 3; (d) a heavy chain of antibody 9TL or its variants shown in Table 3; (e) one or more variable region(s) from a light chain and/or a heavy chain of antibody 9TL or its variants shown in Table 3; (f) one or more CDR(s) (one, two, three, four, five or six CDRs) of antibody 9TL or its variants shown in Table 3; (g) CDR H3 from the heavy chain of antibody 9TL; (h) CDR L3 from the light chain of antibody 9TL or its variants shown in Table 3; (i) three CDRs from the light chain of antibody 9TL or its variants shown in Table 3; Q) three CDRs from the heavy chain of antibody 9TL or its variants shown in Table 3; (k) three CDRs from the light chain and three CDRs from the heavy chain, of antibody 9TL or its variants shown in Table 3; and (1) an antibody comprising any one of (b) through (k). In some embodiments, the polynucleotide comprises either or both of the polynucleotide(s) shown in SEQ ID NO:9 and SEQ ID NO: 10.
[0187] In another aspect, the invention provides polynucleotides encoding any of the antibodies (including antibody fragments) and polypeptides described herein, such as antibodies and polypeptides having impaired effector function. Polynucleotides can be made by procedures known in the art.
[0188] In another aspect, the invention provides compositions (such as a pharmaceutical compositions) comprising any of the polynucleotides of the invention. In some embodiments, the composition comprises an expression vector comprising a polynucleotide encoding the 9TL antibody as described herein. In other embodiment, the composition comprises an expression vector comprising a polynucleotide encoding any of the antibodies or polypeptides described herein. In still other embodiments, the composition comprises either or both of the polynucleotides shown in SEQ ID NO:9 and SEQ ID NO: 10. Expression vectors, and administration of polynucleotide compositions are further described herein. [0189] In another aspect, the invention provides a method of making any of the polynucleotides described herein.
[0190] Polynucleotides complementary to any such sequences are also encompassed by the present invention. Polynucleotides may be single-stranded (coding or antisense) or double- stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules. RNA molecules include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one- to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non- coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials. [0191] Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes an antibody or a portion thereof) or may comprise a variant of such a sequence. Polynucleotide variants contain one or more substitutions, additions, deletions and/or insertions such that the immunoreactivity of the encoded polypeptide is not diminished, relative to a native immunoreactive molecule. The effect on the immunoreactivity of the encoded polypeptide may generally be assessed as described herein. Variants preferably exhibit at least about 70% identity, more preferably at least about 80% identity and most preferably at least about 90% identity to a polynucleotide sequence that encodes a native antibody or a portion thereof. [0192] Two polynucleotide or polypeptide sequences are said to be "identical" if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A "comparison window" as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
[0193] Optimal alignment of sequences for comparison may be conducted using the
Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, WI), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M.O. (1978) A model of evolutionary change in proteins - Matrices for detecting distant relationships. In Dayhoff, M.O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington DC Vol. 5, Suppl. 3, pp. 345-358; Hein J., 1990, Unified Approach, to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, CA; Higgins, D.G. and Sharp, P.M., 1989, CABIOS 5:151-153; Myers, KW. and Muller W., 1988, CABIOS 4:11-17; Robinson, E.D., 1971, Comb.Theor. 11:105; Santou, N., Nes, M., 1987, MoI. Biol. Evol. 4:406-425; Sneath, P.H.A. and Sbkal, R.R., 1973, Numerical Taxonomy the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, CA; Wilbur, WJ. and Lipman, D.J., 1983, Proc. Natl. Acad. Sci. USA 80:726-730.
[0194] Preferably, the "percentage of sequence identity" is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e. gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e. the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
[0195] Variants may also, or alternatively, be substantially homologous to a native gene, or a portion or complement thereof. Such polynucleotide variants are capable of hybridizing under moderately stringent conditions to a naturally occurring DNA sequence encoding a native antibody (or a complementary sequence).
[0196] Suitable "moderately stringent conditions" include prewashing in a solution of 5 X
SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50°C-65°C, 5 X SSC, overnight; followed by washing twice at 65°C for 20 minutes with each of 2X, 0.5X and 0.2X SSC containing 0. 1 % SDS.
[0197] As used herein, "highly stringent conditions" or "high stringency conditions" are those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 5O0C; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/5 OmM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 420C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 μg/ml), 0.1% SDS, and 10% dextran sulfate at 42°C, with washes at 420C in 0.2 x SSC (sodium chloride/sodium citrate) and 50% formamide at 550C, followed by a high- stringency wash consisting of 0.1 x SSC containing EDTA at 550C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.
[0198] It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison). [0199] The polynucleotides of this invention can be obtained using chemical synthesis, recombinant methods, or PCR. Methods of chemical polynucleotide synthesis are well knoΛvn in the art and need not be described in detail herein. One of skill in the art can use the sequences provided herein and a commercial DNA synthesizer to produce a desired DNA sequence. [0200] For preparing polynucleotides using recombinant methods, a polynucleotide comprising a desired sequence can be inserted into a suitable vector, and the vector in turn can be introduced into a suitable host cell for replication and amplification, as further discussed herein. Polynucleotides may be inserted into host cells by any means known in the art. Cells are transformed by introducing an exogenous polynucleotide by direct uptake, endocytosis, transfection, F-mating or electroporation. Once introduced, the exogenous polynucleotide can be maintained within the cell as a non-integrated vector (such as a plasmid) or integrated into the host cell genome. The polynucleotide so amplified can be isolated from the host cell by methods well known within the art. See, e.g., Sambrook et al. (1989).
[0201] Alternatively, PCR allows reproduction of DNA sequences. PCR technology is well known in the art and is described in U.S. Patent Nos. 4,683,195, 4,800,159, 4,754,065 and 4,683,202, as well as PCR: The Polymerase Chain Reaction, Mullis et al. eds., Birkauswer Press, Boston (1994). [0202] RNA can be obtained by using the isolated DNA in an appropriate vector and inserting it into a suitable host cell. When the cell replicates and the DNA is transcribed into RNA, the RNA can then be isolated using methods well known to those of skill in the art, as set forth in Sambrook et al., (1989), for example.
[0203] Suitable cloning vectors may be constructed according to standard techniques, or may be selected from a large number of cloning vectors available in the art. While the cloning vector selected may vary according to the host cell intended to be used, useful cloning vectors will generally have the ability to self-replicate, may possess a single target for a particular restriction endonuclease, and/or may carry genes for a marker that can be used in selecting clones containing the vector. Suitable examples include plasmids and bacterial viruses, e.g., pUCl 8, ρUC19, Bluescript (e.g., pBS SK+) and its derivatives, mpl8, mpl9, pBR322, pMB9, CoIEl, pCRl, RP4, phage DNAs, and shuttle vectors such as pSA3 and pAT28. These and many other cloning vectors are available from commercial vendors such as BioRad, Strategene, and Invitrogen.
[0204] Expression vectors generally are replicable polynucleotide constructs that contain a polynucleotide according to the invention. It is implied that an expression vector must be replicable in the host cells either as episomes or as an integral part of the chromosomal DNA. Suitable expression vectors include but are not limited to plasmids, viral vectors, including adenoviruses, adeno-associated viruses, retroviruses, cosmids, and expression vector(s) disclosed in PCT Publication No. WO 87/04462. Vector components may generally include, but are not limited to, one or more of the following: a signal sequence; an origin of replication; one or more marker genes; suitable transcriptional controlling elements (such as promoters, enhancers and terminator). For expression (i.e., translation), one or more translational controlling elements are also usually required, such as ribosome binding sites, translation initiation sites, and stop codons. [0205] The vectors containing the polynucleotides of interest can be introduced into the host cell by any of a number of appropriate means, including electroporation, transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (e.g., where the vector is an infectious agent such as vaccinia virus). The choice of introducing vectors or polynucleotides will often depend on features of the host cell. [0206] The invention also provides host cells comprising any of the polynucleotides described herein. Any host cells capable of over-expressing heterologous DNAs can be used for the purpose of isolating the genes encoding the antibody, polypeptide or protein of interest. Non- limiting examples of mammalian host cells include but not limited to COS, HeLa, and CHO cells. See also PCT Publication No. WO 87/04462. Suitable non-mammalian host cells include prokaryotes (such as E. coli or B. subtillis) and yeast (such as S. cerevisae, S. pombe; or K. lactis). Preferably, the host cells express the cDNAs at a level of about 5 fold higher, more preferably 10 fold higher, even more preferably 20 fold higher than that of the corresponding endogenous antibody or protein of interest, if present, in the host cells. Screening the host cells for a specific binding to Aβ1-40 is effected by an immunoassay or FACS. A cell overexpressing the antibody or protein of interest can be identified.
Diagnostic uses of 9TL derived antibodies and anti-Aβ antibodies having impaired effector function
[0207] Antibody 9TL which binds to C-terminus of Ap1-40 may be used to identify or detect the presence or absence of Ap1-40. For simplicity, reference will be made generally to 9TL or antibodies with the understanding that these methods apply to any of Aβ1-40 binding embodiments (such as polypeptides) described herein. Detection generally involves contacting a biological sample with an antibody described herein that binds to Aβi_40 and the formation of a complex between Aβ1-40 and an antibody {e.g., 9TL) which binds specifically to Ap1-40. The formation of such a complex can be in vitro or in vivo. The term "detection" as used herein includes qualitative and/or quantitative detection (measuring levels) with or without reference to a control.
[0208] Any of a variety of known methods can be used for detection, including, but not limited to, immunoassay, using antibody that binds the polypeptide, e.g. by enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA) and the like; and functional assay for the encoded polypeptide, e.g. binding activity or enzymatic assay. In some embodiments, the antibody is detectably labeled. Other embodiments are known in the art and described herein. [0209] Antibodies and polypeptides of the invention can be used in the detection, diagnosis and monitoring of a disease, condition, or disorder associated with altered or aberrant Aβ or βAPP expression, such as Alzheimer's disease and Down's syndrome. Thus, in some embodiments, the invention provides methods comprises contacting a specimen (sample) of an individual suspected of having altered or aberrant Aβ expression with an antibody or polypeptide of the invention and determining whether the level of Aβ1-4o differs from that of a control or comparison specimen. In other embodiments, the invention provides methods comprises contacting a specimen (sample) of an individual and determining level of Aβ1-40 expression. [0210] For diagnostic applications, the antibody may be labeled with a detectable moiety including but not limited to radioisotopes, fluorescent labels, and various enzyme-substrate labels. Methods of conjugating labels to an antibody are known in the art. In other embodiment of the invention, antibodies of the invention need not be labeled, and the presence thereof can be detected using a labeled antibody which binds to the antibodies of the invention. [0211] The antibodies of the present invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques, pp.147- 158 (CRC Press, Inc. 1987).
[0212] The antibodies may also be used for in vivo diagnostic assays, such as in vivo imaging. Generally, the antibody is labeled with a radionuclide (such as 111In, 99Tc, 14C, 1311, 125I, or 3H) so that the cells or tissue of interest can be localized using immunoscintiography. [0213] The antibody may also be used as staining reagent in pathology, following techniques well known in the art.
[0214] Anti-Aβ antibodies having impaired effector function may be used for measuring brain amyloid burden for diagnosis of subject at risk of or diagnosed with AD, and assessing progress of any treatment and disease stage. It has been reported that peripheral administration of a monoclonal anti-Aβ antibody results in a rapid increase in plasma Aβ and the magnitude of this increase is highly correlated with amyloid burden in the hippocampus and cortex. DeMattos et al., Science 295:2264-2267 (2002). In some embodiments, an anti-Aβ antibody having impaired effector function is administered to a subject, and level of Aβ in the plasma is measured, whereby an increase in plasma Aβ indicates presence and/or level of brain amyloid burden in the subject. These methods may be used to monitor effectiveness of the treatment and disease stage and to determine future dosing and frequency. Antibodies having impaired effector function may have a better safety profile and provide advantage for these diagnostic uses.
Methods of using anti-Aβ antibody for therapeutic purposes
[0215] The antibodies (including polypeptides), polynucleotides, and pharmaceutical compositions described herein can be used in methods for treating, preventing and inhibiting the development of a disease characterized by aberrant deposition of a protein in the brain of a subject. The methods comprise administering to the subject an effective amount of an antibody that specifically binds to the protein or the protein deposit or a polynucleotide encoding the antibody, wherein the antibody has impaired effector function. For example, an antibody which specifically binds to prion protein or aggregated form of prion protein and has impaired effector function may be administered to a subject for prophylactic and/or therapeutic treatment of Prion diseases; an antibody which specifically binds to synuclein (e.g., alpha-synuclein) or aggregated form of synuclein and has impaired effector function may be administered to a subject for prophylactic and/or therapeutic treatment of Parkinson's disease.
[0216] The antibodies (including polypeptides), polynucleotides, and pharmaceutical compositions described herein can be used in methods for treating, preventing and inhibiting the development of Alzheimer's disease and other diseases associated with altered Aβ or βAPP expression, or accumulation or deposit of Aβ peptide (collectively termed "Aβ-associated diseases"), such as Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, vascular disorder caused by deposit of Aβ peptide in blood vessels (such as stroke and HCHWA-D). Such methods comprise administering the antibodies, polypeptides, or polynucleotides, or a pharmaceutical composition to the subject. In prophylactic applications, pharmaceutical compositions or medicaments are administered to a patient susceptible to, or otherwise at risk of, Alzheimer's disease (or other Aβ-associated disease) in an amount sufficient to eliminate or reduce the risk, lessen the severity, or delay the outset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. In therapeutic applications, compositions or medicaments are administered to a patient suspected of, or already suffering from such a disease in amount sufficient to cure, or at least partially arrest, the symptoms of the disease (biochemical, histological and/or behavioral), including its complications and intermediate pathological phenotypes in development of the disease.
[0217] The invention also provides a method of delaying development of a symptom associated with Alzheimer's disease (or other Aβ-associated disease) in a subject comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject. Symptoms associated with
Alzheimer disease includes, but not limited to, abnormalities of memory, problem solving, language, calculation, visuospatial perception, judgment, and behavior.
[0218] This invention also provides methods of inhibiting or suppressing the formation of amyloid plaques and/or Aβ accumulation in a subject comprising administering an effective dose of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein described herein to the subject. In some embodiments, the amyloid plaques are in the brain of the subject. In some embodiments, the amyloid plaques are in the cerebral vasculature of the subject, hi other embodiments, the Aβ accumulation is in the circulatory system of the subject.
[0219] This invention also provides methods of reducing amyloid plaques and/or reducing or slowing Aβ accumulation in a subject comprising administering an effective dose of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject. In some embodiments, the amyloid plaques are in the brain of the subject, hi some embodiments, the amyloid plaques are in the cerebral vasculature of the subject. In other embodiments, the Aβ accumulation is in the circulatory system of the subject.
[0220] This invention also provides methods of removing or clearing amyloid plaques and/or Aβ accumulation in a subject comprising administering an effective dose of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject. In some embodiments, the amyloid plaques are in the brain of the subject, hi some embodiments, the amyloid plaques are in the cerebral vasculature of the subject, hi other embodiments, the Aβ accumulation is in the circulatory system of the subject.
[0221] This invention also provides methods of reducing Aβ peptide in a tissue (such as brain), inhibiting and/or reducing accumulation of Aβ peptide in a tissue (such as brain), and inhibiting and/or reducing toxic effects of Aβ peptide in a tissue (such as brain) in a subject comprising administering an effective dose of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject. Aβ polypeptide may be in soluble, oligomeric, or deposited form. Oligomeric form of Aβ may be composed of 2-50
Aβ polypeptides, which can be a mixture of full length 1-40 and 1-42 peptides and/or any truncated version of the these peptides.
[0222] The invention also provides methods of improving cognition or reversing cognitive decline associated with diseases associated with amyloid deposit of Aβ in a subject, such as Alzheimer's disease, comprising administering an effective dosage of a pharmaceutical composition comprising an antibody, a polypeptide, or a polynucleotide described herein to the subject.
[0223] The invention also provides methods for treating or preventing diseases associated with amyloid deposit of Aβ, comprising administering to the subject an effective dosage of a pharmaceutical composition comprising an antibody that specifically binds to a beta-amyloid peptide or an aggregated form of a beta-amyloid peptide, wherein the antibody comprises an Fc region with a variation from a naturally occurring Fc region, wherein the variation results in impaired effector function, whereby the administration of the antibody causes less cerebral microhemorrhage than administration of an antibody without the variation.
[0224] Aberrant deposition of proteins in the brain is associated with a number of disorders, some of which can also be characterized as amyloid diseases or amyloidoses, due to concurrent deposition of amyloid-forming proteins.
[0225] Amyloidoses are disorders that are characterized by extracellular deposition of protein fibrils, which form amyloid deposits. While the majority of these conditions are associated with amyloid deposition in the periphery, there are a number of amyloidoses in which central nervous system fibril deposition predominates. WO 00/72876 describes a number of central and peripheral amyloidoses.
[0226] Alzheimer's disease is the most well-known and probably the most common amyloid disease of the central nervous system. This condition is characterized by A-beta- containing plaques and neurofibrillatory tangles, as summarized elsewhere. Various forms of senile dementia are also associated with similar, but less progressive, A-beta plaque formation, as is Down's syndrome.
[0227] Aberrant deposition of endostatin (a 20 kDa C-terminal fragment of collagen
XVIII) has been observed in brains of Alzheimer's patients, where it co-localizes with amyloid- beta(l-40). Deininger, M.H., et al., (2003) J. Neurosci 22(24): 10621-10626. [0228] A variant of the amyloidogenic protein cystatin C, L68Q cystatin C, is associated with massive cerebral amyloidosis leading to brain hemorrhage and death in early adult life in a hereditary form of amyloid angiopathy (hereditary cystatin C amyloid angiopathy). A normal variant of cystatin C (wt1 cystatin C) can be found associated with A-beta peptide as a component of amyloid plaques in Alzheimer's disease.
[0229] In studies to determine whether exogenous agents could suppress formation of dimers of cystatin C (either L68Q or wt cystatin C), Nilsson and co-workers incubated antibodies directed to wt1 cystatin C with monomeric forms of the two variant proteins in solution and observed decreased dimerization of the proteins. Nilsson, M. et al. (2004) J. Biol. Chem. 279(3): 24236-45.
[0230] Down syndrome is characterized by A-beta peptide plaque deposition, apoptotic cell death and aberrant dendritic arborization, in part due to constitutively increased expression of genes that include amyloid precursor protein (APP) and other proteins (superoxide dismutase I, and SlOO-beta) - all located within the Down locus. There is also aberrant expression of genes that are not linked to the Down locus (genes w/in a segment of chromosome 21) - GAP-43, nitric oxide synthase 3, neuronal thread protein, pro-apoptosis genes such as p53, Bax and LI-I beta- converting enzyme. Expression of these non-Down locus genes correlates with proliferation of dystrophic neuritis and apoptotic cell death, de Ia Monte, S.M. 1999. , J. Neural Transm. Suppl. 57: 1-19.
[0231] Brain deposition of amyloid plaques formed from A-beta peptide is a also a common pathologic feature in HIV-AIDS patients. Green, D. A., et al. (2005) AIDS 19(4): 407- 11. Similarly, deposition of A-beta peptide-containing plaques have also been observed shortly following traumatic brain injury in humans, where A-beta co-localizes with APP and neurofilament proteins in swollen axons. Smith, D.H., et al. (2003): 98(5): 1072-7. Brains of patients with end-stage acquired immunodeficiency syndrome (AIDS) were also shown to have increased levels of ubiquitin-stained dotlike deposits (Ub-dots). Gelman, B.B,, and Schuenke, K.
{2004) J. Neurovirol 10(2): 98-108.
[0232] Amyloidoma is a relatively rare form of CNS amyloidosis, presenting in the form of an amyloid tumor, usually in the choroid plexus, with secondary extensions into white matter.
Primary amyloidomas of the brain parenchyma comprise lesions composed of amyloid AL lambda light chain. Tabatbai, G., et al. (2005) Arch Neurol. 62(3): 477-80.
[0233] Multifocal white matter lesions have been observed in brain MRIs in patients carrying the gene(s) for the transthyretin Tyr77 (Tyr77 FAP) variant of familial amyloid polyneuropathy (FAP). Lossos, A., et al. Eur. Neurol. 2005. 53(2): 55-9.
[0234] Familial leptomeningeal amyloidosis is associated with a genetic abnormality of the transthyretin (TTR) variant Asρl8Gly (D18G). Jin, K., et al J. Neurol. Neurosurg.
Psychiatry (2004) 75(10): 1463-66. The D18G variant form of TTR has been also shown to lead to CNS amyloidosis in Hungarian patients. According to one report, small molecule stabilizers of a tetrameric form of the protein may prevent amyloidogenesis. Hammarstrom, P. et al. (2003)
Biochemistry 42(22): 6656-63.
[0235] Mutations in genes encoding alpha-synuclein have been found to be responsible for at least some familial forms of Parkinson's disease, where alpha-synuclein has been shown to be abnormally derivatized and to form neuronal and glial inclusions. Alpha synuclein also forms fibrils in vitro, leading to the categorization of Parkinson's disease as a brain amyloidosis.
Trojanowski, J.Q. and Lee, V.M. (2003) Ann NY Acad. Sci. 991 : 107-110.
[0236] Alpha-synuclein inclusions in oligodendroglia characterize multiple system atrophy (MSA). Kahle, P.M., et al. 2002. EMBO rep. 3(6): 583-8.
[0237] PrP is aberrant form of cellular prion protein (PrP ), a copper-binding glycoprotein attached to the cell membrane of neurons and other cells. PrP amyloid accumulation is commonly associated with PrP cerebral amyloid angiopathy (PrP-CAA), where the accumulation is in neurofibrillary tangles and vascular amyloid, and in Gerstmann-Straussler-
Scheinker disease, where parenchymal amyloidosis may be present in association with spongiform degeneration of neurofibrillary tangles. Ghetti, B., et al. Clin. Lab. Med. (2003)
23(1): 65-85. PrPSc deposition is also found in human spongiform encephalopathy (variant Creutzfeld- Jacob Disease). Paracrine inhibition of prion propagation by anti-PrP single-chain F~v miniantibodies has been reported. Heppner et al., J. Viol. 79:8330-8; 2005. [0238] The following table provides a summary of examples of diseases associated with aberrant brain protein deposition and protein components for the deposit. Antibody against these components may be generated using methods known in the art and methods described herein, or antibodies known in the art.
Figure imgf000067_0001
[0239] The methods described herein (including prophylaxis or therapy) can be accomplished by a single direct injection at a single time point or multiple time points to a single or multiple sites. Administration can also be nearly simultaneous to multiple sites. Frequency of administration may be determined and adjusted over the course of therapy, and is base on accomplishing desired results. In some cases, sustained continuous release formulations of antibodies (including polypeptides), polynucleotides, and pharmaceutical compositions of the invention may be appropriate. Various formulations and devices for achieving sustained release are known in the art.
[0240] Patients, subjects, or individuals include mammals, such as human, bovine, equine, canine, feline, porcine, and ovine animals. The subject is preferably a human, and may or may not be afflicted with disease or presently show symptoms. In the case of Alzheimer's disease, virtually anyone is at risk of suffering from Alzheimer's disease if he or she lives long enough. Therefore, the present methods can be administered prophylactically to the general population without the need for any assessment of the risk of the subject patient. The present methods are useful for individuals who do have a known genetic risk of Alzheimer's disease. Such individuals include those having relatives who have experienced this disease, and those whose risk is determined by analysis of genetic or biochemical markers. Genetic markers of risk toward Alzheimer's disease include mutations in the APP gene, particularly mutations at position 717 and positions 670 and 671 referred to as the Hardy and Swedish mutations respectively (see Hardy (1997) Trends Neurosci. 20: 154-9). Other markers of risk are mutations in the presenilin genes, PSl and PS2, and ApoE4, family history of AD, hypercholesterolemia or atherosclerosis. Individuals presently suffering from Alzheimer's disease can be recognized from characteristic dementia, as well as the presence of risk factors described above. In addition, a number of diagnostic tests are available for identifying individuals who have AD. These include measurement of CSF tau and Aβ42 levels. Elevated tau and decreased Aβ42 levels signify the presence of AD. Individuals suffering from Alzheimer's disease can also be diagnosed by ADRDA (Alzheimer's Disease and Related Disorders Association) criteria. In asymptomatic patients, treatment can begin at any age (e.g., 10, 20, 30). Usually, however, it is not necessary to begin treatment until a patient reaches 40, 50, 60 or 70. Treatment typically entails multiple dosages over a period of time. Treatment can be monitored by various ways known in the art over time. In the case of potential Down's syndrome patients, treatment can begin antenatally by administering therapeutic agent to the mother or shortly after birth.
[0241] The pharmaceutical composition that can be used in the above methods include, any of the antibodies, polypeptides, and/or polynucleotides described herein. In some embodiments, antibody is antibody 9TL or its variants shoΛvn in Table 3. In some embodiments, the antibody is an antibody that specifically binds to an Aβ peptide and comprises a constant region having impaired effector function.
Administration and Dosage
[0242] The antibody is preferably administered to the mammal in a carrier; preferably a pharmaceutically-acceptable carrier. Suitable carriers and their formulations are described in Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, PA, 1990; and Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing, 2000. Typically, an appropriate amount of a priarmaceutically-acceptable salt is used in the formulation to render the formulation isotonic. Examples of the carrier include saline, Ringer's solution and dextrose solution. The pH of the solution is preferably from about 5 to about 8, and more preferably from about 7 to about 7.5. Further carriers include sustained release preparations such as semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, liposomes or microparticles. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentration of antibody being administered. [0243] The antibody can be administered to the mammal by injection (e.g., systemic, intravenous, intraperitoneal, subcutaneous, intramuscular, intraportal, intracerebral, intracerebralventricular, and intranasal), or by other methods, such as infusion, which ensure its delivery to the bloodstream in an effective form. The antibody may also be administered by isolated perfusion techniques, such as isolated tissue perfusion, to exiert local therapeutic effects. Intravenous injection is preferred.
[0244] : Effective dosages and schedules for administering the antibody may be determined empirically, and making such determinations is within the skill in the art. Those skilled in the art will understand that the dosage of antibody that must be administered will vary depending on, for example, the mammal that will receive the antibody, the route of administration, the particular type of antibody used and other drugs being administered to the mammal. Guidance in selecting appropriate doses for antibody is found in the literature on therapeutic uses of antibodies, e.g., Handbook of Monoclonal Antibodies, Ferrone et al., eds., Noges Publications, Park Ridge, N. J., 1985, ch. 22 έnd pp. 303-357; Smith et al.5 Antibodies in Human Diagnosis and Therapy, Haber et al., eds., Raven Press, New York, 1977, pp. 365-389. A typical daily dosage of the antibody used alone might range £rom about 1 μg/kg to up to 100 mg/kg of body weight or more per day, depending on the factors mentioned above. Generally, any of the following doses may be used: a dose of at least about 50 mg/kg body weight; at least about 10 mg/kg body weight; at least about 3 mg/kg body weight; at least about 1 mg/kg body weight; at least about 750 μg/kg body weight; at least about 500 μg/kg body weight; at least about 250 ug/kg body weight; at least about 100 μg /kg body weight; at least about 50 μg /kg body weight; at least about 10 ug /kg body weight; at least about 1 μg/kg body weight, or more, is administered. Antibodies may be administered at lower doses or less frequent at the beginning of the treatment to avoid potential side effect, such as temporary cerebral amyloid angiopathy (CAA).
[0245] In some embodiments, more than one antibody may be present. Such compositions may contain at least one, at least two, at least three, at least four, at least five different antibodies (including polypeptides) of the invention.
[0246] The antibody may also be administered to the mammal in combination with effective amounts of one or more other therapeutic agents. The antibody may be administered sequentially or concurrently with the one or more other therapeutic agents. The amounts of antibody and therapeutic agent depend, for example, on what type of drugs are used, the pathological condition being treated, and the scheduling and routes of administration but would generally be less than if each were used individually.
[0247] Following administration of antibody to the mammal, the mammal's physiological condition can be monitored in various ways well known to the skilled practitioner. [0248] The above principles of administration and dosage can be adapted for polypeptides described herein.
[0249] A polynucleotide encoding an antibody or a polypeptide described herein may also be used for delivery and expression of the antibody or the polypeptide in a desired cell. It is apparent that an expression vector can be used to direct expression of the antibody. The expression vector can be administered systemically, intraperitoneally, intravenously, intramuscularly, subcutaneously, intrathecally, intraventricularly, orally, enterally, parenterally, intranasally, dermally, or by inhalation. For example, administration of expression vectors includes local or systemic administration, including injection, oral administration, particle gun or catheterized administration, and topical administration. One skilled in the art is familiar with administration of expression vectors to obtain expression of an exogenous protein in vivo. See, e.g., U.S. Patent Nos. 6,436,908; 6,413,942; and 6,376,471.
[0250] Targeted delivery of therapeutic compositions comprising a polynucleotide encoding an antibody of the invention can also be used. Receptor-mediated DNA delivery techniques are described in, for example, Findeis et al., Trends Biotechnol. (1993) 11 :202; Chioυ et al., Gene Therapeutics: Methods And Applications Of Direct Gene Transfer (J.A. Wolff, ed.) (1994); Wu et al., J. Biol. Chem. (1988) 263:621; Wu et al., J. Biol Chem. (1994) 269:542; Zenke et al, Proc. Natl. Acad. Sd. (USA) (1990) 87:3655; Wu et al., J. Biol. Chem. (1991) 266:338. Therapeutic compositions containing a polynucleotide are administered in a range of about 100 ng to about 200 mg of DNA for local administration in a gene therapy protocol. Concentration ranges of about 500 ng to about 50 mg, about 1 μg to about 2 mg, about 5 μg to about 500 μg, and about 20 μg to about 100 μg of DNA can also be used during a gene therapy protocol. The therapeutic polynucleotides and polypeptides of the present invention can be delivered using gene delivery vehicles. The gene delivery vehicle can be of viral or non- viral origin (see generally, Jolly, Cancer Gene Therapy (1994) 1 :51; Kirnura, Human Gene Therapy (1994) 5:845; Connelly, Human Gene Therapy (1995) 1:185; and Kaplitt, Nature Genetics (1994) 6:148). Expression of such coding sequences can be induced using endogenous mammalian or heterologous promoters. Expression of the coding sequence can be either constitutive or regulated.
[0251] Viral-based vectors for delivery of a desired polynucleotide and expression in a desired cell are well known in the art. Exemplary viral-based vehicles include, but are not limited to, recombinant retroviruses (see, e.g., PCT Publication Nos. WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; WO 93/11230; WO 93/10218; WO 91/02805; U.S. Patent Nos. 5, 219,740; 4,777,127; GB Patent No. 2,200,651; and EP 0 345 242), alphavirus-based vectors (e.g., Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR- 1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249; ATCC VR-532)), and adeno-associated virus (AAV) vectors (see, e.g., PCT Publication Nos. WO 94/12649, WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655). Administration of DNA linked to killed adenovirus as described in Curiel, Hum. Gene Ther. (1992) 3:147 can also be employed. [0252] Non-viral delivery vehicles and methods can also be employed, including, but not limited to, polycationic condensed DNA linked or unlinked to killed adenovirus alone (see, e.g., Curiel, Hum. Gene Ther. (1992) 3:147); ligand-linked DNAfcee, e.g., Wu, J Biol. Chem. (1 989) 264: 16985); eukaryotic cell delivery vehicles cells (see, e.g., U.S. Patent No. 5,814,482; PCT Publication Nos. WO 95/07994; WO 96/17072; WO 95/30763; and WO 97/42338) and nucleic charge neutralization or fusion with cell membranes. Naked DNA can also be employed. Exemplary naked DNA introduction methods are described in PCT Publication No. WO 90/11092 and U.S. Patent No. 5,580,859. Liposomes that can act as gene delivery vehicles are described in U.S. Patent No. 5,422,120; PCT Publication Nos. WO 95/13796; WO 94/23697; WO 91/14445; and EP 0 524 968. Additional approaches are described in Philip, MoI Cell Biol (1994) 14:2411, and in Woffendin, Proc. Natl. Acad. Sci. (1994) 91 :1581.
Kits
[0253] The invention also provides articles of manufacture and kits containing materials useful for treating pathological conditions described herein, such as Alzheimer's disease or other Aβ-associated diseases (such as Down's syndrome, Parkinson's disease, multi-infarct dementia, mild cognitive impairment, cerebral amyloid angiopathy, vascular disorder caused by deposit of Aβ peptide in blood vessels (such as stroke and HCHWA-D)), or detecting or purifying Aβ or βAPP. The article of manufacture comprises a container with a label. Suitable containers include, for example, bottles, vials, and test tubes. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition having an active agent which is effective for treating pathological conditions or for detecting or purifying Aβ or βAPP. The active agent in the composition is an antibody and preferably, comprises monoclonal antibodies specific for Aβ or βAPP. In some embodiments, the active agent comprises antibody 9TL or any antibodies or polypeptides derived from antibody 9TL. In some embodiments, the active agent comprises an anti-Aβ antibody or polypeptide having impaired effector function. In some embodiments, the anti-Aβ antibody or polypeptide comprises a heavy chain constant region, wherein the constant region has impaired effector function. The label on the container indicates that the composition is used for treating pathological conditions such as Alzheimer's disease or detecting or purifying Aβ or βAPP, and may also indicate directions for either in vivo or in vitro use, such as those described above.
[0254] The invention also provides kits comprising any of the antibodies (such as 9TL), polypeptides, polynucleotides described herein. In some embodiments, the kit of the invention comprises the container described above. In other embodiments, the kit of the invention comprises the container described above and a second container comprising a buffer. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for performing any methods described herein (such as methods for treating Alzheimer's disease, and methods for inhibiting or reducing accumulation of Aβ peptide in the brain). In kits to be used for detecting or purifying Aβ or βAPP, the antibody is typically labeled with a detectable marker, such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme.
[0255] In some embodiments, the invention provides compositions (described herein) for use in any of the methods described herein, whether in the context of use as a medicament and/or use for manufacture of a medicament.
[0256] The following examples are provided to illustrate, but not to limit, the invention.
EXAMPLES
Example 1. Binding affinity determination of antibody 9TL and its variants A. General methods [0257] The following general methods were used in this example.
Expression vector used in clone characterization
[0258] Expression of the Fab fragment of the antibodies was under control of an IPTG inducible lacZ promotor similar to that described in Barbas (2001) Phage display: a laboratory manual, Cold Spring Harbor, NY, Cold Spring Harbor Laboratory Press pg 2.10. Vector pComb3X), however, modifications included addition and expression of the following additional domains: the human Kappa light chain constant domain and the CHI constant domain of IgG2a human immunoglobulin, Ig gamma-2 chain C region, protein accession number POl 859; Immunoglobulin kappa light chain (homosapiens), protein accession number CAA09181.
Small scale Fab preparation
[0259] Small scale expression of Fabs in 96 wells plates was carried out as follows.
Starting from E. coli transformed with a Fab library, colonies were picked to inoculate both a master plate (agar LB + Ampicillin (50 μg/ml) + 2% Glucose) and a working plate (2 ml/well, 96 well/plate containing 1.5 mL of LB + Ampicillin (50 μg/ml) + 2% Glucose). Both plates were grown at 30°C for 8-12 hours. The master plate was stored at 4°C and the cells from the working plate were pelleted at 5000 rpm and resuspended with 1 mL of LB+Ampicillin (50 μg/ml)+ 1 mM IPTG to induce expression of Fabs. Cells were harvested by centrifugation after 5 h expression time at 30°C, then resuspended in 500 μL of buffer HBS-P (10 mM HEPES buffer pH 7.4, 150 mM NaCl, 0.005% P20). Lysis of HBS-P resuspended cells was attained by one cycle of freezing (-800C) then thawing at 37°C. Cell lysates were centrifuged at 5000 rpm for 30 min to separate cell debris from supernatants containing Fabs. The supernatants were then injected into the BIAcore plasmon resonance apparatus to obtain affinity information for each Fab. Clones expressing Fabs were rescued from the master plate to sequence the DNA and for large scale Fab production and detailed characterization as described below.
Large Scale Fab preparation
[0260] To obtain detailed kinetic parameters, Fabs were expressed and purified from large cultures. Erlenmeyer flasks containing 200 mL of LB+Ampicillin (50 μg/ml) + 2% Glucose were inoculated with 5 mL of over night culture from a selected Fab-expressing E. coli clone. Clones were incubated at 30°C until an OD550nm of 1.0 was attained and then induced by replacing the media for 200 ml, of LB+Ampicillin (50 μg/ml) + 1 mM IPTG. After 5h expression time at 300C, cells were pelleted by centrifugation, then resuspended in 10 mL PBS (pH 8). Lysis of the cells was obtained by two cycles of freeze/thaw (at -80°C and 37°C, respectively). Supernatant of the cell lysates were loaded onto Ni-NTA superflow sepharose (Qiagen, Valencia. CA) columns equilibrated with PBS, pH 8, then washed with 5 column volumes of PBS, pH 8. Individual Fabs eluted in different fractions with PBS (pH 8) + 300 mM Imidazol. Fractions containing Fabs were pooled and dialized in PBS5 then quantified by ELISA prior to affinity characterization.
Full antibody preparation [0261] For expression of full antibodies, heavy and light chain variable regions were cloned in mammalian expression vectors and transfected using lipofectamine into HEK 293 cells for transient expression. Antibodies were purified using protein A using standard methods. [0262] Vector pDb.9TL.hFc2a is an expression vector comprising the heavy chain of the
9TL antibody, and is suitable for transient or stable expression of the heavy chain. Vector pDb.9TL.hFc2a has nucleotide sequences corresponding to the following regions: the murine cytomegalovirus promoter region (nucleotides 1-612); a synthetic intron (nucleotides 619-1507); the DHFR coding region (nucleotides 707-1267); human growth hormone signal peptide (nucleotides 1525-1602); heavy chain variable region of 9TL (nucleotides 1603-1951); human heavy chain IgG2a constant region containing the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence; see Eur. J. Immunol. (1999) 29:2613-2624); SV40 late polyadenylation signal (nucleotides 2960-3203); SV40 enhancer region (nucleotides 3204-3449); phage fl region (nucleotides 3537-4992) and beta lactamase (AmpR) coding region (nucleotides 4429-5286). Vector pDb.9TL.hFc2a was deposited at the ATCC on July 20, 2004, and was assigned ATCC Accession No. PTA-6124. [0263] Vector pEb.9TL.hK is an expression vector comprising the light chain of the 9TL antibody, and is suitable for transient expression of the light chain. Vector pEb.9TL.hK has nucleotide sequences corresponding to the following regions: the murine cytomegalovirus promoter region (nucleotides 1-612); human EF-I intron (nucleotides 619-1142); human growth hormone signal peptide (nucleotides 1173-1150); antibody 9TL light chain variable region (nucleotides 1251-1593); human kappa chain constant region (nucleotides 1594-1914); SV40 late polyadenylation signal (nucleotides 1932-2175); SV40 enhancer region (nucleotides 2176- 2421); phage fl region (nucleotides 2509-2964) and beta lactamase (AmpR) coding region (nucleotides 3401-4258). Vector ρEb.9TL.hK was deposited at the ATCC on July 20, 2004, and was assigned ATCC Accession No. PTA-6125.
Biacore Assay
[0264] Affinities of 9TL monoclonal antibody were determined using the BlAcore3000™ surface plasmon resonance (SPR) system (BIAcore, INC, Piscaway NJ). One way of determining the affinity was immobilizing of 9TL on CM5 chip and measuring binding kinetics of Aβ1-4o peptide to the antibody. CM5 chips were activated with N-ethyl-N'-(3-dimethylaminopropyl)- carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Antibody 9TL or its variants was diluted into 10 mM sodium acetate pH 4.0 or 5.0 and injected over the activated chip at a concentration of 0.005 mg/mL. Using variable flow time across the individual chip channels, a range of antibody density was achieved: 1000-2000 or 2000-3000 response units (RU). The chip was blocked with ethanolamine. Regeneration studies showed that a solution containing 2 volumes of PIERCE elution buffer and 1 volumes of 4 M NaCl effectively removed the bound Aβ1-40 peptide while keeping the activity of 9TL on the chip for over 200 injections. HBS-EP buffer (0.0 IM HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20) was used as running buffer for all the BIAcore assays. Serial dilutions (0.1-1Ox estimated KD) of purified Ap1-40 synthetic peptide samples were injected for 1 min at 100 μL/min and dissociation times of 10 min were allowed. Kinetic association rates (Ic0n) and dissociation rates (koff) were obtained simultaneously by fitting the data to a 1 : 1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program. Equilibrium dissociation constant (KD) values were calculated as kWkon.
[0265] Alternatively, affinity was determined by immobilizing Aβi-40 peptide on SA chip and measuring binding kinetics of 9TL Fab and Fab of 9TL variants to the immobilized A PM0 peptide. Affinities of 9TL Fab fragment and its variants Fab fragments were determined by Surface Plasmon Resonance (SPR) system (BIAcore 3000™, BIAcore, Inc., Piscaway, NJ). SA chips (streptavidin) were used according to the supplier's instructions. Biotinylated Aβ peptide 1-40 was diluted into HBS-EP (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% P20) and injected over the chip at a concentration of 0.005 mg/mL. Using variable flow time across the individual chip channels, two ranges of antigen density were achieved: 10-200 response units (RU) for detailed kinetic studies and 500-600 RU for concentration studies and screening. Regeneration studies showed that 100 mM phosphoric acid (may also be followed by a solution containing 2 volumes of 50 mM NaOH and 1 volume of 70% ethanol) effectively removed the bound Fab while keeping the activity of Aβ peptide on the chip for over 200 injections. HBS-EP buffer was used as running buffer for all the BIAcore assays. Serial dilutions (0.1-1Ox estimated KD) of purified Fab samples were injected for 2 min at 100 μL/min and dissociation times of 10 min were allowed. The concentrations of the Fab proteins were determined by ELISA and/or SDS-PAGE electrophoresis using a standard Fab of known concentration (determined by amino acid analysis). Kinetic association rates (Ic0n) and dissociation rates (koff) were obtained simultaneously by fitting the data to a 1 : 1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program. Equilibrium dissociation constant (KD) values were calculated as IWk0n.
B. Binding affinity of antibody 9TL and its variants to Aβi.40
[0266] The amino acid sequences of the heavy chain and light chain variable regions of antibody 9TL is shown in Figure 1. The binding affinity of 9TL antibody to Aβ1-40 determined using both methods of Biacore described above is shown in Table 2 below.
Table 2. Binding affinity of antibody 9TL and Fab fragment
Figure imgf000077_0001
[0267] The amino acid sequence of the variants of 9TL is shown in Table 3 below. All amino acid substitutions of the variants shown in Table 3 are described relative to the sequence of 9TL. The binding affinity of Fab fragment of 9TL variants are also shown in Table 3. KD and other kinetic parameters were determined by BIAcore analysis described above with Aβ1-4o immobilized on SA chip.
Figure imgf000077_0002
Figure imgf000078_0001
Figure imgf000079_0002
I=AIl CDRs are extended CDRs including both Kabat and Chotbia CDRs. Amino acid residues are numbered sequentially.
2=underlined Ic0n were experimentally determined. Others were estimated to be the same as 9TL.
Figure imgf000079_0001
Example 2: Characterization of epitope on Aβl-40 peptide that antibody 9TL binds [0268] To determine the epitope on Aβ polypeptide that is recognized by antibody 9TL,
Surface Plasmon Resonance (SPR, Biacore 3000) binding analysis was used. Aβ1-40 polypeptide coupled to biotin (Global Peptide Services, CO) was immobilized on a streptavidin-coated chip (SA chip). The binding of Aβ antibodies Fab fragments (at 50 nM) to the immobilized Aβ^o ύi the absence or presence of different soluble fragments of the Aβ peptide (at 10 μM, from American Peptide Company Inc., CA). Amino acid sequences of Aβ1-40, Aβi-42, and Aβi.43 are shown in below in Table 4. The Aβ peptides which displaced binding of antibody 9TL Fab fragment to Aβ1-4o were Aβ28-40, AβMo, Aβ33-4o, and Aβ17-40, respectively (Figure 2). Thus, antibody 9TL binds to a C-terminal peptide (33-40) of Aβ1-40. As shown in Figure 2, the Aβi-28, Aβ28-42>22-35, Aβi-16, Aβ1-43, and Aβi-38 peptide did not inhibit the binding of antibody 9TL Fab fragment, suggesting that antibody 9TL binds to the C-terminus of Aβ^o peptide. [0269] In addition, Aβ28-42 and Aβ1-43 peptide did not inhibit binding of antibody 9TL to
Mo although they could readily inhibit Aβ^o binding to control antibody (antibody 2289, this antibody is described in U.S. Appl. Pub. No. 2004/0146512 and WO04/032868) which bind to 16-28 of ApI-40. These results show that antibody 9TL preferentially binds to Aβi-40, but not to
Figure imgf000080_0001
Table 4. Amino acid se uences of beta am loid e tides
Figure imgf000080_0002
Example 3. Generation of monoclonal antibody 2H6 and deglycosylated 2H6 A. Generation and characterization of monoclonal antibody 2H6
[0270] Mice were iminunized with 25-100 μg of a peptide (amino acid 28-40 of Ap1-40) conjugated to KLH in adjuvant (50 μl per footpad, 100 μl total per mouse) at about 16 consecutive week intervals as described in Geerligs HJ et al., 1989, J. Immunol. Methods 124:95- 102; Kenney JS et al., 1989, J. Immunol. Methods 121 :157-166; and Wicher K et al., 1989, Int. Arch. Allergy Appl. Immunol. 89:128-135. Mice were first immunized with 50 μg of the peptide in CFA (complete Freud's adjuvant). After 21 days, mice were secondly immunized with 25 μg of the peptide in IFA (incomplete Freud's adjuvant). Twenty three days later after the second immunization, third immunization was performed with 25 μg of the peptide in IFA. Ten days later, antibody titers were tested using ELISA. Forth immunization was performed with 25 μg of the peptide in IFA 34 days after the third immunization. Final booster was performed with 100 μg soluble peptide 32 days after the forth immunization.
[0271] Splenocytes were obtained from the immunized mouse and fused with NSO myeloma cells at a ratio of 10: 1 , with polyethylene glycol 1500, The hybrids were plated out into 96-well plates in DMEM containing 20% horse serum and 2-oxaloacetate/pyruvate/insulin (Sigma), and;hypoxanthine/a-minopterin/thymidine selection was begun. On day 8, 100 μl of DMEM containing 20% horse serum was added to all the wells. Supernatants of the hybrids were screened by using antibody capture immunoassay. Determination of antibody class was done with class-specific second antibodies. [0272] A panel of monoclonal antibody-producing cell lines was selected for characterization. One cell line selected produces as antibody designated 2H6. This antibody was determined to have IgG2b heavy chain.
[0273] The affinity of antibody 2H6 to Aβi-4o was determined. Monoclonal antibody 2H6 was purified from supernatants of hybridoma cultures using protein A affinity chromatography. The supernatants was equilibrated to pH 8. The supernatants "were then loaded to the protein A column MabSelect (Amersham Biosciences # 17-5199-02) equilibrated with PBS to pH 8. The column was washed with 5 column volumes of PBS, pH 8. Tlie antibody was eluted with 50 mM citrate-phosphate buffer, pH 3. The eluted antibody was neutralized with IM Phosphate Buffer, pH 8. The purified antibody was dialyzed with PBS. The antibody concentration was determined by SDS-PAGE, using a murine mAb standard curve.
[0274] 2H6 Fabs were prepared by papain proteolysis of the 2H6 full antibody using
Immunopure Fab kit (pierce # 44885) and purified by flow through protein A chromatography following manufacturer instructions. Concentration was determined by SDS-PAGE and A280 using 1OD=O.6 mg/ml.
[0275] Affinities of 2H6 monoclonal antibody were determined using the BlAcore3000™ surface plasmon resonance (SPR) system (BIAcore, INC, Piscaway NJ). One way of determining the affinity was immobilizing 2H6 antibody on CM5 chip and measuring binding kinetics of Aβϊ- 4o peptide to the antibody. CM5 chips were activated with N-ethyl-N'-(3-dimethylaminopropyl)- carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. 2H6 monoclonal antibody was diluted into 10 mM sodium acetate pH 4.0 or 5.0 and injected over the activated chip at a concentration of 0.005 mg/mL. Using variable flow time across the individual chip channels, a range of antibody density was achieved: 1000-2000 or 2000-3000 response units (RU). The chip was blocked with ethanolamine. Regeneration studies showed that a mixture of Pierce elution buffer (Product No. 21004, Pierce Biotechnology, Rockford, IL) and 4 M NaCl (2:1) effectively removed the bound Aβi-40 peptide while keeping the activity of 2H6 antibody on the chip for over 200 injections. HBS-EP buffer (0.01M HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20) was used as running buffer for all the BIAcore assays. Serial dilutions (0.1-1Ox estimated KD) of purified APi-40 synthetic peptide samples were injected for 1 min at 100 μL/min and dissociation times of 10 min were allowed. Kinetic association rates (Ij0n) and dissociation rates (koff) were obtained simultaneously by fitting the data to a 1:1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program. Equilibrium dissociation constant (KD) values were calculated as kOfi/kon-
[0276] Alternatively, affinity was determined by immobilizing Aβ1-4o peptide on SA chip and measuring binding kinetics of 2H6 Fab to the immobilized Aβi-4o peptide. Affinities of 2H6 Fab fragment was determined by Surface Plasmon Resonance (SPR) system (BIAcore 3000™, BIAcore, Inα, Piscaway, NJ). SA chips (streptavidin) were used according to the supplier's instructions. Biotinylated Aβ peptide 1-40 (SEQ ID NO: 15) was diluted into HBS-EP (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.005% P20) and injected over the chip at a concentration of 0.005 mg/mL. Using variable flow time across the individual chip channels, two ranges of antigen density were achieved: 10-200 response units (RU) for detailed kinetic studies and 500-600 RU for concentration studies. Regeneration studies showed that a mixture of Pierce elution buffer and 4 M NaCl (2:1) effectively removed the bound Fab while keeping the activity of Aβ peptide on the chip for over 200 injections. HBS-EP buffer was used as running buffer for all the BIAcore assays. Serial dilutions (0.1-1Ox estimated KD) of purified Fab samples were injected for 2 min at 100 μL/min and dissociation times of 10 min were allowed. The concentrations of the Fab proteins were determined by ELISA and/or SDS-PAGE electrophoresis using a standard Fab of known concentration (determined by amino acid analysis). Kinetic association rates (kon) and dissociation rates (koff) were obtained simultaneously by fitting the data to a 1:1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110) using the BIAevaluation program. Equilibrium dissociation constant (KD) values were calculated as koff/kcm- The affinity of 2H6 antibody determined using both methods described above is shown in Table 5 below.
[0277] Affinity for murine antibody 2286, which binds to a peptide of amino acid 28-40 of Aβi.40, was tested as described above. Antibody 2286 is described Ln U.S. Appl. Ser. No. 10/683,815 and PCT/US03/32080.
Table 5. Binding affinity of antibody 2H6 and 2286
Figure imgf000082_0001
Figure imgf000083_0001
[0278] To determine the epitope on Aβ polypeptide recognized by antibody 2H6, Surface
Plasmon Resonance (SPR5 Biacore 3000) binding analysis was used. Aβ1-4o polypeptide (SEQ ID NO: 15) coupled to biotin (Global Peptide Services, CO) was immobilized on a streptavidin- coated chip (SA chip). The binding of Aβ antibodies (at 100 nM) to the immobilized ApM0 in the absence or presence of different soluble fragments of the Aβ peptide (at 16 μ.M, from American Peptide Company Inc., CA). The Aβ peptides which displaced binding of antibody 2H6 to Aβi-w were Aβ17-40, Aβ33-40, and Aβ1-40, respectively (Figure 3). Thus, antibody 2H6 binds to a C-terminal peptide (33-40) of Aβ1-40. However, this C-terminal peptide (33-40) of Aβ1-40 did not displace binding of antibody 2286 to Aβ1-40 at the concentration tested. As shown in Figure 3, the Aβ1-38 peptide did not inhibit the binding of antibody 2H6 or antibody 2286 to AβMo, suggesting that, similar to antibody 2286, the epitope that antibody 2H6 binds includes amino acids 39 and/or 40 of the Aβ1-4o peptide (Figure 3).
[0279] In addition, Ap1-42 and Aβ1-43 peptide did not inhibit binding of antibody 2H6 to
1-4o although they could readily inhibit Ap1-40 binding to control antibody (antibody 2289, this antibody is described in U.S. Appl. Ser. No. 10/683,815 and PCT/USO3/3208O) -which bind to 16- 28 of Aβi.40 (Figure 3). These results show that antibody 2H6 preferentially binds to Ap1-40, but not to APi_42;and Ap1-43.
[0280] To further assess the involvement of discrete amino acid residues of the β-amyloid peptide that antibody 2H6 binds, different Aβ1-4o variants, in which each of the last 6 amino acids (Aβ1-4o amino acid residues 35-40) was individually replaced by an alanine (alanine scanning mutagenesis), were generated by site directed mutagenesis. These Aβ1-4o variants (sequences shown in Tab' Ie 6) were expressed in E. coli as Glutathione-S-Transferase (GST) fusion proteins (Amersham Pharmacia Biotech, Piscataway, NJ USA) followed by affinity purification on a Glutathione-Agarose beads (Sigma-Aldrich Corp., St. Louis, MO, USA). As control, Wild-type (WT) AβMo as well as Aβi-_u, Ap1-42,, and Ap1-39 were also expressed as GST fusion proteins. Aβi.40, Aβ1-41, Aβ1-42;, Aβi-39, as well as the six different variants (M35A(l-40), V36A(l-40), G37A(l-40), G38A(l-40), V39A(l-40), V40A(l-40) shown in Table 6) were then immobilized (100 μl of 0.025 μg/μl of GST-peptide per well) onto ELISA assay plates and incubated with either of mAb 2286, 2289, and 2H6 in serial dilution from 0.3 nM down (data using 0.3 nM mAb are shown in Figure 4). After 10 consecutive washes, assay plates were incubated with 100 μl of 0.03 μg/ml per well of Biotin-conjugated Goat-anti-Mouse (H+L) antibody (Vector Laboratories, vector #BA-9200, Burllingame CA, USA) followed by 100 μl of 0.025 μg/ml per well of HRP- conjugated Streptavidin (Aniersham Biosciences Corp., #RPN4401V, NJ, USA). The absorbance of the plate was read at 450 nm.
Table 6. Amino acid sequences of beta amyloid peptides and variants
Figure imgf000084_0001
[0281] As shown in Figure 4, Mab 2289 which was directed to amino acid 16 to 28 of
Aβ, recognized all variants with the same intensity and served as internal positive control of protein concentration and protein integrity on the plate. Antibody 2H6 did not recognize Aβim, Ap1-39, or AβM2 as shown in Fig. 4. Aβ1-40 variants V40A, V39A, G38A, G37A, V36A, and M35A showed reduced binding to antibody 2H6, demonstrating that antibody 2H6 epitope extended for at least 6 amino acids at the C terminal end of Ap1-4O. Mutations of V and G to A are very conservative and are not likely to produce important conformational changes in proteins, therefore, the large effect of these mutations to antibody 2H6 binding might be due to the ability of the antibody to differentiate between the mentioned amino acids in the context of Aβ and these data demonstrated a very high degree of specificity for this antibody.
[0282] To determine whether 2H6 and 9TL compete for binding to Ap1-40, competition experiments were performed using Biacore assay. Antibody 2H6, 9TL and 2289 were immobilized on different channels of a CM5 chip. CM5 chip channels were activated with N- ethyl-N'-(3-dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N- hydroxysuccinimide (NHS) according to the supplier's instructions. Antibody 2H6, 9TL, and 2289 were each diluted into 10 mM sodium acetate pH 4.0 and injected over an activated chip at a concentration of 0.005 mg/mL. Antibody density was 1625 response units (RU) for 2H6; 4000 RU for 9TL; and 2200 RU for 2289. Each channel was blocked with ethanolamine. Aβ1-40 peptide (150 uM) was flowed onto the chip for 2 min. Then antibody 2H6 (to be tested for competition of binding) at 0.6 uM was flowed onto the chip for 1 min. HBS-EP buffer (0.01M HEPES, pH 7.4, 0.15 MNaCl, 3 mM EDTA, 0.005% Surfactant P20) was used as running buffer for all the BIAcore assays. After measuring binding OfAp1-40, all channels of the chip were regenerated by washing twice with a mixture of Pierce elution buffer (Product No. 21004, Pierce Biotechnology, Rockford, IL) and 4 M NaCl (2:1) for 6 sec. Competition binding was then performed for antibody 9TL, and then antibody 2289. Competition between 9TL and 2H6 for binding to Aβ^o was observed, but no competition was observed between 9TL and 2289 or between 2H6 and 2289. Observations of competition between the antibody immobilized and the same antibody flowed onto the chip served as the positive control.
B. Antibody 2H6 does not bind to APP
[0283] To determine whether 2H6 binds to amyloid precursor proteins (APP)5 binding of
2H6 to cells transfected with wildtype APP was determined. 293 cells were transfected with a cDNA encoding wild type human amyloid precursor protein. Forty eight hours after the transfection, cells were incubated oh ice for 45 minutes with monoclonal antibodies anti-Aβi.i6, anti-Aβ16-28, or 2H6 (5 ug/ml in DMEM with 10% FCS). The cells were then washed three times in PBS for 5 minutes, fixed with 4% PFA. The cells were washed three times again in PBS, and antibody binding was detected with secondary Cy3 -conjugated goat anti-mouse antibody (dilution of 1:500) from Jackson Immunoresearch under fluorescence microscope. [0284] Anti-Aβi-16 and anti-Aβ16-28 antibodies, which recognize N-terminal or central epitopes in Aβ, both showed significant binding to APP precursor proteins expressed on cells. In contrast, 2H6 did not bind to APP expressing cells.
C. Generation of deglycosylated antibody 2H6
[0285] To generate deglycosylated antibody 2H6, purified antibody 2H6 was incubated at
37°C for 7 days with peptide-N-glycosidase F (Prozyme, 0.05 U per mg of antibody) in 20 mM Tris-HCl pH:8.0. Completeness of deglycosylation was verified by MALDI-TOF-MS and protein gel electrophoresis. Deglycosylated antibodies were purified by Protein A chromatography and endotoxin was removed by Q-Sepharose. The binding affinity to Aβ1-4o of the deglycosylated 2H6 was tested using Biacore assay described above, and the binding affinity of the deglycosylated 2H6 to Aβ1-4o was found to be identical to the intact antibody 2H6.
Example 4. Reversal of cognitive deficits and histological symptoms with less microhemorrhage in an animal model of Alzheimer's disease by administration of deglycosylated,2H6 A. Experimental Protocol
[0286] Administration of antibodies. Transgenic mice over-expressing the "Swedish" mutant amyloid precursor protein (APP Tg2576 with K670N/M671; Hsiao et al., Science 274:99- 102 (1996)) were used for the experiments. The Alzheimer's-like phenotype present in these mice has been well-characterized. Holcomb et al., Nat. Med. 4:97-100 (1998); Holcomb et al., Behav. Gen. 29:177-185 (1999); and McGowan E, Neurobiol. Dis. 6:231-244 (1999). For the sixteen weeks treatment study, APP-transgenic mice, aged 20 months, were assigned to one of the four groups. The first group received weekly intraperitoneal anti-Aβ antibody 2H6 (mouse monoclonal anti-human Aβ28-40 IgG2b described in Example 3) injections for a period of 16 weeks (n = 4). The second group received weekly intraperitoneal deglycosylated anti-Aβ antibody 2H6 (produced as described in Example 3) injections for a period of 16 weeks (n = 5). The third group received weekly intraperitoneal anti-AMN antibody (2906; mouse-monoclonal anti-Drosophila amnesiac protein IgGl) injections for a period of 16 weeks (n = 6). Non- transgenic littermates were treated for 16 weeks with either anti-AMN antibody (n = 4) or 2H6 (n = 2).
[0287] Behavioral analysis. Following 16 weeks of antibody treatment, the mice from the study were subjected to a two-day radial-arm water-maze paradigm as described previously. Wilcock et al., J Neuroinflammation 1 :24 (2004). The apparatus was a 6-arm maze as described previously. Gordon et al., Neurobiol. Aging 22:377-385 (2001). On day one, 15 trials were run in three blocks of 5. A cohort of 4 mice were run sequentially for each block (i.e., each of 4 mice get trial one, then the same mice get trial two, etc.). After each 5-trial block, a second cohort of mice was run permitting an extended rest period before mice were exposed to the second block of 5 trials. The goal arm was different for each mouse in a cohort to minimize odor cues. The start arm was varied for each trial, with the goal arm remaining constant for a given individual for both days. For the first 11 trials, the platform was alternately visible then hidden (hidden for the last 4 trials). On day two, the mice were run in exactly the same manner as day one except that the platform was hidden for all trials. The number of errors (incorrect arm entries) was measured in a one-minute time frame. Mice failing to make an arm choice in 20 seconds were assigned one error, but no mice in this study had to be assigned an error in this manner. Due to the numbers of mice in the study, the tester was unaware of treatment group identity of each mouse. Since the dependent measures in the radial-arm water-maze task were quantitative, not evaluative, the potential for tester bias was reduced. In order to minimize the influence of individual trial variability, each mouse's errors for 3 consecutive trials were averaged producing 5 data points for each day, which were analyzed statistically by ANOVA using StatView (SAS Institute Inc., NC). [0288] Histological analysis. On the day of sacrifice, mice were weighed, overdosed with 100 mg/kg Nembutal (Abbott laboratories, North Chicago, IL), and then intracardially perfused with 25 mL of 0.9% sodium chloride. Brains were rapidly removed, and the left half of the brain was immersion fixed for 24 h in freshly prepared 4% paraformaldehyde in 100 mM KPO4 (pH 7.2) for histopathology. The hemi-brains were then incubated for 24 h in 10%, 20% and 30% sucrose sequentially for cyroprotection, Horizontal sections of 25 μ thickness were collected using a sliding microtome and stored at 40C in Dulbecco's phosphate-buffered saline with sodium azide (pH 7.2) to prevent microbial growth. A series of 8 equally spaced tissue sections 600 μ apart were randomly selected spanning the entire brain and stained using free- floating immunohistochemistry for total Aβ (rabbit polyclonal anti-pan Aβ; Biosource, Camarillo, CA, 1:10,000) as previously described. Gordon et al., Exp. Neurol. 173:183-195 (2002); Wilcock et al., J. Neurosci. 24:6144-6151 (2004). A second series of tissue sections 600 μm apart were stained using 0.2% Congo red in NaCl-saturated 80% ethanol. Another set of sections were also mounted and stained for hemosiderin using 2% potassium ferrocyanide in 2% hydrochloric acid for 15 min, followed by a counterstain in a 1% neutral red solution for 10 min. Quantification of Congo red staining and Aβ immunohistochemistry was performed using the Image-Pro Plus (Media Cybernetics, Silver Spring, MD) to analyze the percent area occupied by positive stain. One region of the frontal cortex and three regions of the hippocampus were analyzed (to ensure that there was no regional bias in the hippocampal values). The initial analysis of Congo red was performed to give a total value. A second analysis was performed after manually editing out all of the parenchymal amyloid deposits to yield a percent area restricted to vascular Congo red staining. To estimate the parenchymal area of Congo red, the vascular amyloid values were subtracted from the total percentage. For the hemosiderin stain the numbers of Prussian blue-positive sites were counted on all sections and the average number of sites per section calculated. Qualitative differences between animals were observed at the sections at a low magnification. Eight equally spaced sections were examined and the number of positive profiles was determined and averaged to a per-section value. To assess possible treatment-related differences, the values for each treatment group were analyzed by one-way ANOVA followed by Fisher's LSD means comparisons.
[0289] Measurement of serum level of Aβ peptide using ELISA. Serum collected one day after the last dosing of antibodies was diluted and incubated in 96-well microtiter plates (MaxiSorp; Nunc, Rosklide, Denmark), which were precoated with antibody 6E10 (anti-beta amyloid antibody that binds to Aβi.π; Signet, Dedham, MA) at 5 ug/ml in PBS buffer, pH 7.4. The secondary antibody was biotinylated 4G8 (anti-beta amyloid antibody that binds to Aβi7.24; Signet) at a 1 :5000 dilution. Detection was done using a streptavidin-horseradish peroxidase conjugate (Amersham Biosciences), followed by TMB substrate (KPL, Gaithersburg, MD). Aβi_ 40 (American Peptide) scaling from 6-400 pM were used for standard curves.
B. Results
[0290] Reversal of cognitive deficits by administration ofdeglycosylated antibody. The radial-arm water-maze task detects spatial learning and memory deficits in transgenic mouse models. Gordon et al., Neurobiol. Aging. 22:377-385 (2001); Morgan et al., Nature 408:982-985 (2000). Animal treated with antibody 2H6, deglycosylated 2H6, or anti-AMN for 16 weeks were tested for spatial navigation learning in a two-day version of the radial-arm water maze. Nontransgenic normal mice (including 2 mice treated with 2H6 antibody and 4 mice treated with anti-AMN antibody; these two groups were combined since no behavior difference was observed) were also tested in the two-day version of the radial-arm water maze. As shown in Fig. 5, APP- transgenic mice treated with the control antibody (anti-AMN) failed to learn platform location over two days of testing and were significantly impaired compared to the nontransgenic mice as previously described. Wilcock et al., J Neuroinflammation 1:24 (2004). However, APP- transgenic mice administered the anti-Aβ antibody 2H6 and deglycosylated 2H6 demonstrated a significant reversal of the impairment observed in the control-treated APP-transgenic mice, ending day trwo with a mean performance near 1 error per trial (Fig. 5). The control-treated APP- transgenic mice has a mean performance near 3 errors per trial at the ending of day two (Fig. 5). The data shoΛvn in Fig. 5 indicate that the deglycosylated antibody works as well as the intact antibody for reversal of cognitive deficits in the APP-transgenic mice.
[0291] Reduction of A β deposits without increasing microhemorrhage. As shown Table 7 below, total Aβ immunostaining in the hippocampus was significantly reduced after 16 weeks immunotherapy with antibody 2H6 (about 56% reduction, p=0.0001) and deglycosylated 2H6 (about 58% reduction, p<0.0001) as compared to the control antibody-treated group (anti-AMN). As shown in Table 8 below, total Aβ immunostaining in the frontal cortex was significantly reduced after 16 weeks immunotherapy with antibody 2H6 (about 50% reduction, pO.OOOl) and deglycosylated 2H6 (about 51% reduction, pO.OOOl) as compared to the control antibody-treated group (anti-AMN). Table 7. Total Aβ load for hippocampus after 16 weeks of antibody treatment. Mean percent area occupied by positive immunohistochemical stain for Aβ, and standard deviation and standard error of the mean for the hippocampus are shown.
Figure imgf000090_0001
Table 8. Total Aβ load for frontal cortex after 16 weeks of antibody treatment. Mean percent area occupied by positive immunohistochemical stain for Aβ, and standard deviation and standard error of the mean for the frontal cortex are shown.
Figure imgf000090_0002
[0292] As shown Table 9, total Congo-red staining in the hippocampus was significantly reduced after 16 weeks immunotherapy with antibody 2H6 (about 77% redaction, p<0.0001) and deglycosylated 2H6 (about 53 % reduction, p<0.0001) as compared to the control antibody-treated group (anti-AMN). As showrx in Table 10, total Congo-red staining in the frontal cortex was also significantly reduced after 16 "weeks immunotherapy with antibody 2H6 (about 79% reduction, p<0.0001) and deglycosylated 2H6 (about 68% reduction, pO.OOOl) as compared to the control antibody-treated group (anti- A-MN).
Table 9. Total Congo-red staining for hippocampus after 16 weeks of antibody treatment. Mean percent area occupied by positive Congo-red stain for Aβ, and standard deviation and standard error of the mean for the hippocampus are shown.
Figure imgf000090_0003
Table 10. Total Congo-red staining for frontal cortex after 16 weeks of antibody treatment. Mean percent area occupied by positive Congo-red stain for Aβ, and standard deviation and standard error of the mean for the frontal cortex are shown.
Figure imgf000091_0001
[0293] Parenchymal (Tables 11 and 12; and Figure 6) and vascular (Tables 13 and 14-;
Figure 7) Congo-red staining were analyzed separately for both frontal cortex and hippocampus. As shown Table 11 and Fig.6A, parenchymal Congo-red staining in the frontal cortex was significantly reduced after 16 weeks immunotherapy with antibody 2H6 (about 98% reduction, pO.OOOl) and deglycosylated 2H6 (about 77% reduction, pO.OOOl) as compared to the control antibody-treated group (anti-AMN). As shown in Table 12 and Fig. 6B, parenchymal Congo-red staining in the hippocampus was also significantly reduced after 16 weeks immunotherapy with antibody 2H6 (about 96% reduction, p<0.0001) and deglycosylated 2H6 (about 63% reduction, pO.OOOl) as. compared to the control antibody-treated group (anti-AMN). The deglycosylated 2H6 was less effective than the intact 2H6 antibody in reducing Congo-red load in the frontal cortex and hippocampus.
Table 11. Parenchymal Congo-red staining for frontal cortex after 16 weeks of antibody treatment. Mean percent area occupied by positive Congo-red stain for Aβ, and standard deviation and standard error of the mean for the frontal cortex are shown.
Figure imgf000091_0002
Table 12. Parenchymal Congo-red staining for hippocampus after 16 weeks of antibody treatment. Mean percent area occupied by positive Congo-red stain for Aβ, and standard deviation and standard error of the mean for the hippocampus are shown.
Figure imgf000092_0001
[0294] As shown Table 13 and Fig. 7 A, vascular Congo-red staining in the hippocampus was significantly increased after 16 weeks immunotherapy with antibody 2H6 (about 2.7 fold, pO.OOOl) and deglycosylated 2H6 (about 1.7 fold, p=0.0185) as compared to the control antibody-treated group (anti-AMN). As shown in Table 14 and Fig. 7B, vascular Congo-red staining in the frontal cortex was also significantly increased after 16 weeks immunotherapy -with antibody 2H6 (about 3.5 fold, pO.OOOl) and deglycosylated 2H6 (about 1.8 fold, p=0.0048) as compared to the control antibody-treated group (anti-AtMN). The increase of vascular Congo-red staining in the deglycosylated 2H6 treated group was significantly less than in the intact 2H6 antibody treated group for both hippocampus (p=0.0025) and frontal cortex (pO.OOOl).
Table 13. Vascular Congo-red staining for hippocampαs after 16 weeks of antibody treatment. Mean percent area occupied by positive Congo-red stain for Aβ, and standard deviation and standard error of the mean for the hippocampus are shown.
Figure imgf000092_0002
Table 14. Vascular Congo-red staining for frontal cortex after 16 weeks of antibody treatment. Mean percent area occupied by positive Congo-red stain for Aβ, and standard deviation and standard error of the mean for the frontal cortex are shown.
Figure imgf000092_0003
[0295] Prussian blue histological stain was used to label hemosiderin, a feric oxide material produced in the breakdown of hemoglobin. Extravenous blood in the brain leads to microglial phagocytosis of the erythrocytes and breakdown of the hemoglobin within them. Thus, ferric oxide-containing microglia are thus markers of past hemorrhage. The number Prussian blue-positive profiles in antibody treated animals were counted. As shoΛvn in Table 15 and Fig. 8, treatment with antibody 2H6 significantly (p<0.0001) increased Prussian blue staining for about 5.5 fold as compared to the control antibody treated group (anti-AMN). Treatment with deglycosylated 2H6 antibody only increased Prussian blue staining for about 1.8 fold (p=O.0364) as compared to the control antibody treated group.
Table 15. Prussian blue staining for the entire section after 16 weeks of antibody treatment. Mean percent area occupied by positive Prussian blue stain, and standard deviation and standard error of the mean for the entire section are shown.
Figure imgf000093_0001
[0296] Serum level of Aβ after administration of deglycosylated 2H6 antibody. As shown in Fig. 9, administration of both antibody 2H6 and deglycosylated 2H6 to the APP Tg2576 mice significantly increased serum level of beta-amyloid peptide. However, no significant increase of serum level of beta-amyloid peptide was observed in the APP Tg2576 mice after administration of anti-AMN antibody or in the wild type mice after administration of anti-AMN antibody or antibody 2H6. This indicates that the increase of serum level of beta-amyloid peptide may be used to assist disgnosis of AD and monitor the response to AD therapy, such as immunotherapy.
C. Conclusion
[0297] The above data demonstrate that 1) the deglycosylated antibody 2H6 is as effective as the intact antibody 2H6 in reversing learning and memory deficits in APP Tg2576 mice; 2) the deglycosylated antibody 2H6 is a little less effective as the intact antibody 2H6 in depleting Aβ deposits in hippocampus and frontal cortex as measured by Congo-red staining, but is as effective in depleting Aβ load in hippocampus and frontal cortex as measured by Aβ immunostaining; 3) increase of Aβ deposit (as measured by Congo-red staining) in hippocampous and frontal cortex vasculature by the deglycosylated antibody 2H6 is much less than the intact 2H6 antibody; and 4) microhemorrhage as measured by Prussian blue staining in APP Tg2576 mice treated with the deglycosylated 2H6 antibody was much lower than mice treated with the intact 2H6 antibody. These data suggest that deglycosylated antibody is as effective for improving indications of Alzheimer's disease with lower risk of microhemorrhage in APP Tg2576 mice.
Example 5. Binding affinity of various antibody Fc regions to murine and human Fcγ receptors and complement
[0298] Binding affinity of the antibody Fc regions to Fcγ receptors or complement were measured using BIAcore as described above. Briefly, purified human or murine Fcγ receptors (from R&D Systems) and human CIq (from Quidel) were immobilized on BIAcore CM5 chip by amine chemistry. Serial dilutions of monoclonal antibodies (ranging from 2 nM to the maximum concentration as indicated in Tables 16 and 17) were injected. HBS-EP (0.01M HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20) as running and sample buffer. Binding data were analyzed using 1 : 1 langmuir interaction model for high affinity interactions, or steady state affinity model for low affinity interactions. [0299] Table 16 below shows the binding affinity of the anti-β-amyloid antibodies as measured by KD (nM) to murine FcγPJ, FcγPJIb, FcγPJII, and human CIq (hClcL). Deglycosylated antibodies have a constant region with removed N-glycosylation. 9TL(MgGl) and 9TL(hIgG2Δa) have the same variable region (shown in SEQ ID NO: 1 and SEQ ID NO:2), but different constant region. 9TL(MgGl) has a human IgGl constant region; and 9TL(MgG2Δa) has a human IgG2a with mutations of A330P331 to S330S33 1 (Kabat amino acid numbering with reference to the wildtype IgG2a sequence). As shown in Table 16, deglycosylated 2H6, 2294, and 2286 had reduced affinity to all murine Fcγ receptors tested as compared to each corresponding antibody without removed N-glycosylation. Deglycosylated 2H6 also had reduced affinity to human complement as compared to 2H6. 9TL(MgGl) had no significant binding to mFcγRIIb or mFcγRIII; and 9TL(hIgG2Δa) had no significant binding to any of murine FcγRI, FcγRIIb, FcγRIII, or hClq. [0300] Table 17 below shows the binding affinity of the anti-β -amyloid antibodies as measured by K0 (nM) to human FcγRI, FcγRIIb/c, FcγRIIIb, and hClq. As shown in Table 17, 9TL(hIgG2Δa) had no significant binding to human FcγRI or hClq; and significant lower affinity to human FcγRIIb/c and FcγRIIIb as compared to the affinity of the antibody with human IgGl constant region to these molecules.
Table 16. Binding affinity of antibodies to murine Fcγ receptors and human complement as measured by KD (nM)
Figure imgf000095_0001
NB: no significant binding when antibody was used at the maximum concentration tested. Maximum antibody concentration tested for binding to hClq was 30,000 nM. Table 17. Binding affinity of antibodies to human Fcγ receptors and human complement as measured by Kp (nM)
Figure imgf000096_0001
NB: no significant binding when antibody is used at 30 μM concentration. Maximum antibody concentration tested for binding was 30,000 nM.
Example 6. Effect of deglycosylated 2H6 antibody on microglial activation, Fcγ receptor binding, and amyloid clearance after intracranial administration
[0301] Surgical procedure and intracranial administration of antibodies Tg2576 transgenic mice aged 19.5 months were assigned to one of the three groups, all groups received intracranial injections into the frontal cortex and hippocampus. The first group received anti-Aβ antibody 2H6 at a concentration of 2μg/2μl in each region. The second group received deglycosylated 2H6 antibody at 2 μg/2μl in each region. The third group received IgG directed against drosophila amnesiac protein as a control for nonspecific aspects of intact IgG injection. AU mice survived for 72 hours after surgery. [0302] On the day of surgery the mice (Tg2576 transgenic mice) were weighed, anesthetized with isoflurane and placed in a stereotaxic apparatus (51603 dual manipulator lab standard, Stoelting, Wood Dale, IL). A midsagittal incision was made to expose the cranium and two burr holes were drilled using a dental drill over the right frontal cortex and hippocampus to the following coordinates: Cortex: AP +1.5 mm, L -2.0 mm, hippocampus: AP -2.7 mm, L -2.5 mm, all taken from bregma. A 26 gauge needle attached to a 10 μl Hamilton (Reno, NV) syringe was lowered 3 mm ventral to bregma and a 2 μl injection was made over a 2 minute period. The incision was cleaned with saline and closed with surgical staples.
[0303] Tissue preparations. On the day of sacrifice mice were weighed, overdosed with 100 mg/kg pentobarbital (Nembutal sodium solution, Abbott laboratories, North Chicago IL) and intracardially perfused with 25 ml 0.9% sodium chloride followed by 50 ml freshly prepared 4% paraformaldehyde (pH=7.4). Brains were rapidly removed and immersion fixed for 24 hours in freshly prepared 4% paraformaldehyde. The brains were then incubated for 24 hours in 10, 20 and 30% sucrose sequentially to cyroprotect them. Horizontal sections of 25 μm thickness were then collected using a sliding microtome and stored at 40C in DPBS buffer with sodium azide to prevent microbial growth.
[0304] Six to eight sections approximately 100 μm apart were selected spanning the injection site and stained using free-floating immunohistochemistry methods for total Aβ (rabbit antiserum reacting with Aβi_4o and Aβ1-42; dilution used 1 : 10,000), anti-CD45 antibody (Cat. No. MCA1031G, Serotec, Raleigh NC; dilution used 1:5000), and anti-Fcγ receptor (CD16/CD32) antibody (Cat. No. 553141 from BD Biosciences; used as 1 : 1,000 dilution). Biotinylated goat anti-rabbit at 1 :3,000 dilution was used as the secondary antibody for anti-Aβ antibody staining. Biotinylated goat anti-rat at 1 : 3, 000 dilution was used as the secondary antibody for anti-CD45 and Fcγ receptor antibody staining. For immunostaining, some sections were omitted from the primary antibody to assess non-specific immunohistochemical reactions. Adjacent sections were mounted on slides and stained using 4% thiofiavine-S (Sigma- Aldrich, St Louis MO) for 10 minutes. It should be noted that there were a limited number of sections that include the injection volume. [0305] Data analysis. The immunohistochemical reaction product on all stained sections was measured using a videometric Vl 50 image analysis system (Oncor, San Diego, CA) in the injected area of cortex and hippocampus and corresponding regions on the contralateral side of the brain. Data were presented as the ratio of injected side to non-injected side for Aβ staining, thioflavine-S staining, CD45 staining, and Fcγ receptor staining. Normalizing each injection site to the corresponding contralateral site diminishes the influence of mteranimal variability and permits reliable measurements of drug effects with a smaller number of mice. To assess possible treatment-related differences, the ratio values for each treatment group were analyzed by ANOVA using StatView software version 5.0.1 (SAS Institute Inc., NC) followed by Fischer's LSD means comparisons. Results
[0306] As shown in Figure 10, CD45 staining in the frontal cortex and the hippocampus was about the same after intracranial administration of deglycosylated 2H6 antibody as the control antibody. In contrast, CD45 staining in the frontal cortex was significantly higher (p<0.01) and was generally higher in the hippocampus after intracranial administration of 2H6 antibody than the control antibody. This indicates that, unlike antibody 2H6, administration of deglycosylated 2H6 did not activate microglia in the frontal cortex and the hippocampus 72 hours after the administration.
[0307] As shown in Figure 11, Fcγll and Fcγlll receptor staining in the frontal cortex and the hippocampus was about the same after intracranial administration of deglycosylated 2H6 antibody as the control antibody. In contrast, Fcγ receptor staining in the frontal cortex and the hippocampus was significantly higher (p<0.01) after intracranial administration of 2H6 antibody than the control antibody. This indicates that, unlike antibody 2H6, administration of deglycosylated 2H6 did not activate microglia in the frontal cortex and the hippocampus 72 hours after the administration.
[0308] As shown in Figure 12, Aβ staining was lower in frontal cortex and hippocampus 72 hours after intracranial administration of 2H6 antibody or deglycosylated 2H6 antibody as compared to the control antibody. As shown in Figure 13, thioflavin-S stained compact plaque was also lower in frontal cortex and hippocampus 72 hours after intracranial administration of 2H6 antibody or deglycosylated 2H6 antibody as compared to the control antibody.
[0309] These data indicate that deglycosylated 2H6 antibody was able to reduce Aβ and compact plaques in frontal cortex and hippocampus without inducing microglia activation and inflammatory response.
Example 7. Characterization of epitope on Aβ peptide that antibody 2294 binds
[0310] Antibody 2294 is a murine antibody raised by immunizing a mouse with Aβi.
40. This antibody is described in US 2004/0146512 and WO 04/032868.
[0311] Binding affinity for antibody 2294 to Aβ1-40, Aβ1-42, or Aβ22-37 was measured using Biacore as described above. Table 18 below shows the affinity of antibody 2294 Fab fragment to various Aβ peptides.
Table 18. Binding affinity of antibody 2294 Fab fragment
Figure imgf000098_0001
Figure imgf000099_0001
[0312] Epitope mapping of antibody 2294 was performed by ELISA assay. Biotinylated
15-mer or 10-mer of various Aβ peptides (these peptides have glycine added to the C-terminal end) were immobilized on streptavidin coated plates. NUNC maxisorp plates were coated with 6 ug/ml of streptavidin (Pierce, 21122) in PBS pH 7.4 for more than 1 h at 40C. Plates were blocked with 1% BSA in PBS buffer pH 7.4. After washing, biotinylated Aβ peptides in PBS pH 7.4 were incubated 1 hour at room temperature. Antibody 2294 (from 2.5 ug/ml to 10 ug/ml) was incubated with the immobilized Aβ peptides for 1 h at room temperature. After washing, plates were incubated with secondary antibody, a HRP conjugated goat anti-human kappa chain antibody (MP Biomedicals, 55233) at 1:5000 dilution. After washing, bound secondary antibody was measured by adding TMB substrate (KPL, 50-76-02, 50-65-02). HRP reaction was stopped by adding IM phosphoric acid and absorbance at 450 nm was measured. As shown in Figure 14, antibody 2294 binds to Aβ peptides with amino acids 20-34, 21-35, 22-36, 23-37, 24-38, 25-39, 26-40, and 25-34 with a glycine at the C-terminus; but does not bind to Aβ peptides with amino acids 19-33, 27-41, 24-33, and 27-35 having a glycine at the C-terminus of these peptides. This suggests that the epitope of antibody 2294 binds includes amino acids from 26 to 34. [0313] To further determine the epitope on Aβ peptide that is recognized by antibody
2294, ELISA binding analysis was used. Various Aβ peptides (Global Peptide Services, CO) was immobilized on a ELISA plate. The binding of 2294 full antibody (at 20 nM) to the immobilized Aβ was determined by ELISA as described above. Antibody 2294 binds to Aβ peptides 17-40, 17-42, 28-40, 1-38, 1-40, 1-42, and 1-43. Antibody 2294 did not bind to Aβ peptide 1-16, 1-28, 28-42, 22-35, and 33-40. Thus, antibody 2294 binds to the C-terminus of various truncated Aβ peptide, for example, 1-38, 1-40, 1-42, and 1-43. [0314] Table 19 below shows binding comparison of 2294 to Aβ1-4o to other Aβ peptide as measured by Biacore assay. Antibody 2294 (full antibody) has the strongest binding to Aβμ4o as compared to other peptides, with significantly lower binding to truncated Aβ1-40 (such as 1-36, 1-37, 1-38, and 1-39), Aβ1-42 and Aβ1-43. This indicates that the side chain or backbone of amino acid 40 (Valine) of Aβ is involved in binding of 2294 to Aβ1-40; and binding is significantly reduced in absence of this amino acid. Table 19.
Figure imgf000100_0001
"-" indicates no binding; "+" indicates very low binding; "++" indicates medium binding; -" indicates strong binding; and "++++" indicates very strong binding.
[0315] Based on data shown above, the epitope that antibody 2294 binds seems to include amino acids 26-34 and 40. Antibody 2294 binds to an epitope very similar to antibody 6G described in U.S. provisional application serial no. 60/676,093 (the amino acid and nucleic acid sequences of this antibody is shown in SEQ ID NOS:36-39; vectors encoding 6G are deposited at American Type Culture Collection on June 15, 2005, with accession numbers PTA-6786 and PTA-6787). The epitope that antibody 6G binds includes amino acids from 25 to 34, and 40. The epitope comparison of antibody 2294 and 6G are shown in Figure 14.
[0316] The binding affinity of antibody 6G Fab fragment to Aβ1-40, Aβ1-42, or Aβ22-37 was measured using Biacore. Biotinylated Aβmo, Aβ1-42, or Aβ22-37 was immobilized on a streptavidin chip, and 6G Fab was flowed onto it. Antibody 6G Fab fragment binds to Aβ1-40 with kon (I/Ms) of 3.0 x 105, koff (1/s) of 7.0 x 10"4, and KD of 2 nM. Antibody 6G Fab fragment binds to Aβ1-42 with Ic0n (1/Ms) of 1.8 x 104, koff (1/s) of 1.6 x 10"3, and K0 of 80 nM. Antibody 6G Fab fragment binds to Aβ22-37 with Ic0n (I/Ms) of 3.6 x 105, koff (1/s) of 3.9 x 10" 3, and KD of 11 nM. Antibody 6G has significant higher affinity to Aβi-42, and Aβ22-37 than antibody 2294. Data indicate that binding of antibody 6G is less dependent on amino acid 40 than antibody 2294.
[0317] Antibody competition experiments between 2294, 6G, 2H6, and 2289 using
Biacore assay were performed as described in Example 3. Competition experiments were performed using Biacore assay. Antibody 2294, 6G, 2H6, and 2289 were immobilized on different channels of a CM5 chip. CM5 chip channels were activated with N-ethyl-N'-(3- dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Antibody 2294, 6G, 2H6, and 2289 were each diluted into 10 mM sodium acetate pH 4.0 and injected over an activated chip at a concentration of 0.005 mg/niL. Each channel was blocked with ethanolamine. Aβ1-40 peptide (150 uM) was flowed onto the chip for 2 min. Then antibody 2294 (to be tested for competition of binding) at 0.6 uM was flowed onto the chip for 1 min. HBS-EP buffer (0.0 IM HEPES, pH 7.4, 0.15 M NaCl, 3 mM EDTA, 0.005% Surfactant P20) was used as running buffer for all the BIAcore assays. After measuring binding of Aβ1-40, all channels of the chip were regenerated by washing twice with a mixture of Pierce elution buffer (Product No. 21004, Pierce Biotechnology, Rockford, IL) and 4 M NaCl (2:1) for 6 sec. Competition binding was then performed for antibody 6G5 2H6, and then antibody 2289. Competition between 2294 and 6G and between 2294 and 2H6 for binding to Aβ1-40 was observed, but no competition was observed between 2294 and 2289 or between 6G and 2289. Observations of competition between the antibody immobilized and the same antibody flowed onto the chip served as the positive control. Data indicate that antibody 2294 competes with 2H6 and 6G for binding to Aβ1-40.
Example 8. Effect of antibody 2294 and deglycosylated antibody 2294 in reducing Aβ deposit and cognition in animal model of Alzheimer's disease
[0318] Deglyeosylated antibody 2294 was prepared as by incubating purified antibody
2294 at 370C for 7 days with peptide-N-glycosidase F (Prozyme, 0.05 U per mg of antibody) in 20 niM Tris-HCl pH 8.0. Completeness of deglycosylation was verified by MALDI-TOF- MS and protein gel electrophoresis. Deglycosylated antibodies were purified by Protein A chromatography and endotoxin was removed by Q-Sepharose. The binding affinity to APi-40 of the deglycosylated 2294 was tested using Biacore assay described above, and the binding affinity of the deglycosylated 2294 to Ap1-40 was found to be identical to the intact antibody 2294.
[0319] Antibody 2294 and deglycosylated 2294 were tested in transgenic mice APP
Tg2576 for effect on reversal of cognitive deficits, histological symptoms, and microhemorrhage as described in Example 4. For the sixteen weeks treatment study, transgenic mice (aged 20 months) were assigned to were assigned to one of the four groups. The first group received weekly intraperitoneal anti-Aβ antibody 2294 injections for a period of 16 weeks (n - 4). The second group received weekly intraperitoneal deglycosylated anti- Aβ antibody 2294 injections for a period of 16 weeks (n = 5). The third group received weekly intraperitoneal anti-AMN antibody (2906; mouse-monoclonal mti-Drosophila amnesiac protein IgGl) injections for a period of 16 weeks (n = 6). Non-transgenic littermates were treated for 16 weeks with either anti-AMN antibody (n = 4) or 2294 (n = 2). [0320] Histological and behavioral analysis are performed as described in Example 4.
[0321] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application. All publications, patents and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, patent or patent application were specifically and individually indicated to be so incorporated by reference. !
Deposit of Biological Material
[0322] ■ The following materials have been deposited with the American Type Culture
Collection, 10801 University Boulevard, Manassas, Virginia 20110-2209, USA (ATCC): Material Antibody No. ATCC Accession No. Date of Deposit pDb.9TL.hFc2a 9TL heavy chain PTA-6124 July 20, 2004 pEb.9TL.hK 9TL light chain PTA-6125 July 20, 2004 pDb.6G.hPc2a 6G heavy chain PTA-6786 June 15, 2005 pEb.6G.hK 6G light chain PTA-6787 June 15, 2005
[0323] Vector pEb.9TL.hK is a polynucleotide encoding the 9TL light chain variable region and the light chain kappa constant region; and vector pDb.9TL.hFc2a is a polynucleotide encoding the 9TL heavy chain variable region and the heavy chain IgG2a constant region containing the following mutations: A330P331 to S33OS331 (amino acid numbering with reference to the wildtype IgG2a sequence; see Eur. J. Immunol. (1999) 29:2613-2624). [0324] Vector pEb.6G.hK is a polynucleotide encoding the 6G light chain variable region and the light chain kappa constant region; and vector pDb.6G.hFc2a is a polynucleotide encoding the 6G heavy chain variable region and the heavy chain IgG2a constant region containing the following mutations: A330P331 to S330S331 (amino acid numbering with reference to the wildtype IgG2a sequence; see Eur. J. Immunol. (1999) 29:2613-2624). [0325] These deposits were made under the provisions of the Budapest Treaty on the
International Recognition of the Deposit of Microorganisms for the Purpose of Patent Procedure and the Regulations thereunder (Budapest Treaty). This assures maintenance of a viable culture of the deposit for 30 years from the date of deposit. The deposit will be made available by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Rinat Neuroscience Corp. and ATCC, which assures permanent and unrestricted availability of the progeny of the culture of the deposit to the public upon issuance of the pertinent U.S. patent or upon laying open to the public of any U.S. or foreign patent application, whichever comes first, and assures availability of the progeny to one determined by the U.S. Commissioner of Patents and Trademarks to be entitled thereto according to 35 USC Section 122 and the Commissioner's rules pursuant thereto (including 37 CFR Section 1.14 with particular reference to 886 OG 638). [0326] The assignee of the present application has agreed that if a culture of the materials on deposit should die or be lost or destroyed when cultivated under suitable conditions, the materials will be promptly replaced on notification with another of the same. Availability of the deposited material is not to be construed as a license to practice the invention in contravention of the rights granted under the authority of any government in accordance with its patent laws.
Antibody sequences
9TL heavy chain variable region amino acid sequence (SEQ ID NQ: 1)
QVQLVQSGAEVKKPGASVKVSCKASGYYTEAYYIHWVRQAPGQGLEWMGRIDPAT GNTKYAPRLQDRVTMTRDTSTSTVYMELSSLRSEDTAVYYCASLYSLPVYWGQGTT VTVSS
9TL light chain variable region amino acid sequence (SEQ ID NO:2)
DVVMTQSPLSLPVTLGQPASISCKSSQSLLYSDAKTYLNWFQQRPGQSPRRLIYQISRL DPGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCLQGTHYPVLFGQGTRLEIKRT
9TL CDR Hl (extended CDR) (SEO ID NO: 3) GYYTEAYYIH
9TL CDR H2 (extended CDR) (SEQ ID NO: 4) RIDPATGNTKYAPRLQD
9TL CDR H3 (extended CDR) (SEO ID NO: 5) LYSLPVY
9TL CDR Ll (extended CDR) (SEO ID NO: 6) KSSQSLLYSDAKTYLN
9TL CDR L2 (extended CDR) (SEQ ID NO: 7) QISRLDP
9TL CDR L3 (extended CDR) (SEQ ID NO: 8) LQGTHYPVL
9TL heavy chain variable region nucleotide sequence (SEO ID N0:9) CAGGTGCAGCTGGTGCAGTCTGGTGCTGAGGTGAAGAAGCCTGGCG-CTTCCGTGA
AGGTTTCCTGCAAAGCATCTGGTTACTATACGGAGGCTTACTATATCCACTGGGTG
CGCCAAGCCCCTGGTCAAGGCCTGGAGTGGATGGGCAGGATTGATCCTGCGACTG
GTAATACTAAATATGCCCCGAGGTTACAGGACCGGGTGACCATGACTCGCGATAC
CTCCACCAGCACTGTCTACATGGAACTGAGCTCTCTGCGCTCTGAGGACACTGCTG
TGTATTACTGTGCCTCCCTTTATAGTCTCCCTGTCTACTGGGGCCAGOGTACCACT
GTTACCGTGTCCTCT
9TL light chain variable region nucleotide sequence (SEQ ID NO: 10)
GATGTTGTGATGACCCAGTCCCCACTGTCTTTGCCAGTTACCCTGGaACAACCAG
CCTCCATATCTTGCAAGTCAAGTCAGAGCCTCTTATATAGTGATGCCAAGACATA
TTTGAATTGGTTCCAACAGAGGCCTGGCCAGTCTCCACGCCGCCTAATCTATCAG
ATTTCCCGGCTGGACCCTGGCGTGCCTGACAGGTTCAGTGGCAGTGGATCAGGCA
CAGATTTTACACTTAAAATCAGCAGAGTGGAGGCTGAAGATGTGGGAGTTTATTA
CTGCTTACAAGGTACACATTATCCGGTGCTCTTCGGTCAAGGGACCCGCCTGGAG
ATCAAACGCACT
9TL heavy chain full antibody amino acid sequence (including modified IgG2a as described herein) (SEQ ID NO: ID
QVQLVQSGAEVKKPGASVKVSCKASGYYTEAYYIHWVRQAPGQGLEΛVMGRIDPAT
GNTKYAPRLQDRVTMTRDTSTSTVYMELSSLRSEDTAVYYCASLYSLPVYWGQGTT
VTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKD YFPEPVTVSWNSaALTSGVHTFP
AVLQSSGLYSLSSVVTVPSSNFGTQTYTCNVDHKPSNTKVDKTVERKCCVECPPCPAP
PVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDαVEVHNAKTK
PREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPSSIEKTISKTKGQPREPQV
YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPMLDSDGSFFL
YSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
9TL light chain full antibody amino acid sequence (SEO ID NO: 12) DVVMTQSPLSLPVTLGQP ASISCKSSQSLLYSDAKTYLNWFQQRPGQSPRULIYQISR LDPGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCLQGTHYPVLFGQGTRXEIKRTV AAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQD SKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
9TL heavy chain fall antibody nucleotide sequence (including modified IgG2a as described herein) (SEO ID NO: 13)
CAGGTGCAGCTGGTGCAGTCTGGTGCTGAGGTGAAGAAGCCTGGCGCTTCCGTGA
AGGTTTCCTGCAAAGCATCTGGTTACTATACGGAGGCTTACTATATCCACTGGGT
GCGCCAAGCCCCTGGTCAAGGCCTGGAGTGGATGGGCAGGATTGATCCTGCGACT
GGTAATACTAAATATGCCCCGAGGTTACAGGACCGGGTGACCATGACTCGCGATA
CCTCCACCAGCACTGTCTACATGGAACTGAGCTCTCTGCGCTCTGAGaACACTGC
TGTGTATTACTGTGCCTCCCTTTATAGTCTCCCTGTCTACTGGGGCCA(JGGTACCA
CTGTTACCGTGTCCTCTGCCTCCACCAAGGGCCCATCTGTCTTCCCACTGGCCCCA
TGCTCCCGCAGCACCTCCGAGAGCACAGCCGCCCTGGGCTGCCTGGTCAAGGACT
ACTTCCCAGAACCTGTGACCGTGTCCTGGAACTCTGGCGCTCTGACCA--GCGGCGT
GCACACCTTCCCAGCTGTCCTGCAGTCCTCAGGTCTCTACTCCCTCAGCAGCGTGG
TGACCGTGCCATCCAGCAACTTCGGCACCCAGACCTACACCTGCAACGTAGATCA
CAAGCCAAGCAACACCAAGGTCGACAAGACCGTGGAGAGAAAGTGTTGTGTGGA
GTGTCCACCTTGTCCAGCCCCTCCAGTGGCCGGACCATCCGTGTTCCTGTTCCCTC
CAAAGCCAAAGGACACCCTGATGATCTCCAGAACCCCAGAGGTGACCTGTGTGGT
GGTGGACGTGTCCCACGAGGACCCAGAGGTGCAGTTCAACTGGTATGTGGACGG
AGTGGAGGTGCACAACGCCAAGACCAAGCCAAGAGAGGAGCAGTTCAACTCCAC
CTTCAGAGTGGTGAGCGTGCTGACCGTGGTGCACCAGGACTGGCTGAACGGAAA
GGAGTATAAGTGTAAGGTGTCCAACAAGGGACTGCCATCCAGCATCG-AGAAGAC
CATCTCCAAGACCAAGGGACAGCCAAGAGAGCCACAGGTGTATACCCTGCCCCC
ATCCAGAGAGGAGATGACCAAGAACCAGGTGTCCCTGACCTGTCTGGTGAAGGG
ATTCTATCCATCCGACATCGCCGTGGAGTGGGAGTCCAACGGACAGCCAGAGAAC
AACTATAAGACCACCCCTCCAATGCTGGACTCCGACGGATCCTTCTTCCTGTATTC
CAAGCTGACCGTGGACAAGTCCAGATGGCAGCAGGGAAACGTGTTCrCTTGTTCC
GTGATGCACGAGGCCCTGCACAACCACTATACCCAGAAGAGCCTGTCCCTGTCTC
CAGGAAAGTAATTCTAGA
9TL light chain fall antibody nucleotide sequence (SEQ ID NO: 14)
GATGTTGTGATGACCCAGTCCCCACTGTCTTTGCCAGTTACCCTGGGACAACCAG
CCTCCATATCTTGCAAGTCAAGTCAGAGCCTCTTATATAGTGATGCCAAGACATA
TTTGAATTGGTTCCAACAGAGGCCTGGCCAGTCTCCACGCCGCCTAArCTATCAG
ATTTCCCGGCTGGACCCTGGCGTGCCTGACAGGTTCAGTGGCAGTGGATCAGGCA
CAGATTTTACACTTAAAATCAGCAGAGTGGAGGCTGAAGATGTGGGA-GTTTATTA
CTGCTTAQAAGGTACACATTATCCGGTGCTCTTCGGTCAAGGGACCCGCCTGGAG
ATCAAACGCACTGTGGCTGCACCATCTGTCTTCATCTTCCCTCCATCTGATGAGCA GTTGAAATCCGGAACTGCCTCTGTTGTGTGCCTGCTGAATAACTTCTATCCACGCG
AGGCCAAAGTACAGTGGAAGGTGGATAACGCCCTCCAATCCGGTAACTCCCAGG
AGAGTGTCACAGAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGCAGCACCC
TGACCCTGAGCAAAGCAGACTACGAGAAACACAAAGTCTACGCCTGCGAAGTCA
CCCATCAGGGCCTGAGTTCTCCAGTCACAAAGAGCTTCAACCGCGGTGAGTGCTA
ATTCTAG
6G heavy chain variable region amino acid sequence (SEO ID NO:26)
QVQLVQSGAEVKKPGASVKVSCKASGYTFTTYAIHWVRQ APGQGLEWMGFTSPYSGVSNYNQKFKGRVTMTRDTSTST VYMELSSLRSEDTAVYYCARFDNYDRGYVRDYWGQGTLV
TVS
6G light chain variable region amino acid sequence (SEQ ID NO:27)
DIVMTQSPDSLAVSLGERATINCRASESVDNDRISFLNW
YQQKPGQPPKLLIYAATKQGTGVPDRFSGSGSGTDFTLT
ISSLQAEDVAVYYCQQSKEFPWSFGGGTKVEIKRTV
6G CDR Hl (extended CDR) (SEQ ID NO:28) GYTFTTYAIH
6G CDR H2 (extended CDR) (SEO ID NO:29) FTSPYSGVSNYNQKFKG
6G CDR H3 (extended CDR) (SEO ID NO:30) FDNYDRGYVRDY
6G CDR Ll (extended CDR) (SEO ID N0:31) RASESVDNDRISFLN
6G CDR L2 (extended CDR) (SEO ID NO:32) AATKQGT
6G CDR L3 (extended CDR) (SEO ID NO:33) QQSKEFPWS 6G heavy chain variable region nucleotide sequence (SEQ ID NO:34)
CAGGTGCAACTGGTGCAATCCGGTGCCGAGGTGAAAAAGCCAGGCGCCTCCGTGA
AAGTGTCCTGCAAAGCCTCCGGTTACACCTTTACCACCTATGCCATCCATTGGGTG
CGCCAGGCCCCAGGCCAGGGTCTGGAGTGGATGGGCTTTACTTCCCCCTACTCCG
GGGTGTCGAATTACAATCAGAAGTTCAAAGGCCGCGTCACCATGACCCGCGACAC
CTCCACCTCCACAGTGTATATGGAGCTGTCCTCTCTGCGCTCCGAAGACACCGCCG
TGTATTACTGTGCCCGCTTCGACAATTACGATCGCGGCTATGTGCGTGACTATTGG
GGCCAGGGCACCCTGGTCACCGTCTCC
6G light chain variable region nucleotide sequence (SEQ ID NO:35)
GACATCGTGATGACCCAGTCCCCAGACTCCCTGGCCGTGTCCCTGGGCGAGCGCG
CCACCATCAACTGCCGCGCCAGCGAATCCGTGGATAACGATCGTATTTCCTTTCT
GAACTGGTACCAGCAGAAACCAGGCCAGCCTCCTAAGCTGCTCATTTACGCCGC
CACCAAACAGGGTACCGGCGTGCCTGACCGCTTCTCCGGCAGCGGTTCCGGCAC
CGATTTCACTCTGACCATCTCCTCCCTGCAGGCCGAAGATGTGGCAGTGTATTAC
TGTCAGCAGTCCAAAGAGTTTCCCTGGTCCTTTGGCGGTGGCACCAAGGTGGAGA
TCAAACGCACTGTG
6G heavy chain full antibody amino acid sequence (Including modified IgG2a as described herein) fSEO ID NQ-.36)
QVQLVQSGAEVKKPGASVKVSCKASGYTFTTYAIHWVRQAPGQGLEWMGFTSPYSG VSNYNQKFKGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARFDNYDRGYVRDYW GQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKD YFPEPVTVSWNSGALTS GVHTFPAVLQSSGL YSLSSWTVPSSNFGTQTYTCNVDHKPSNTKVDKWERKCCVE CPPCP APPVAGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVQFNWYVDGVEV HNAKTKPREEQFNSTFRVVSVLTVVHQDWLNGKEYKCKVSNKGLPSSIEKTISKTKG QPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPML DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 6G light chain full antibody amino acid sequence (SEQ ID NO:37)
DIVMTQSPDSLAVSLGERATINCRASESVDNDRISFLNWYQQKPGQPPKLLIYAATK
QGTGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQSKEFPWSFGGGTKVEIKRTV
AAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQD
SKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
6G heavy chain full antibody nucleotide sequence (includinfi modified IgG2a as described herein) (SEO ID NO:38)
CAGGTGCAACTGGTGCAATCCGGTGCCGAGGTGAAAAAGCCAGGCGCCTCCGTG
AAAGTGTCCTGCAAAGCCTCCGGTTACACCTTTACCACCTATGCCATCCATTGGGT
GCGCCAGGCCCCAGGCCAGGGTCTGGAGTGGATGGGCTTTACTTCCCCCTACTCC
GGGGTGTCGAATTACAATCAGAAGTTCAAAGGCCGCGTCACCATGACCCGCGAC
ACCTCCACCTCCACAGTGTATATGGAGCTGTCCTCTCTGCGCTCCGAAGACACCG
CCGTGTATTACTGTGCCCGCTTCGACAATTACGATCGCGGCTATGTGCGTGACTAT
TGGGGCCAGGGCACCCTGGTCACCGTCTCCTCAGCCTCCACCAAGGGCCCATCTG
TCTTCCCACTGGCCCCATGCTCCCGCAGCACCTCCGAGAGCACAGCCGCCCTGGG
CTGCCTGGTCAAGGACTACTTCCCAGAACCTGTGACCGTGTCCTGGAACTCTGGC
GCTCTGACCAGCGGCGTGCACACCTTCCCAGCTGTCCTGCAGTCCTCAGGTCTCTA
CTCCCTCAGCAGCGTGGTGACCGTGCCATCCAGCAACTTCGGCACCCAGACCTAC
ACCTGCAACGTAGATCACAAGCCAAGCAACACCAAGGTCGACAAGACCGTGGAG
AGAAAGTGTTGTGTGGAGTGTCCACCTTGTCCAGCCCCTCCAGTGGCCGGACCAT
CCGTGTTCCTGTTCCCTCCAAAGCCAAAGGACACCCTGATGATCTCCAGAACCCC
AGAGGTGACCTGTGTGGTGGTGGACGTGTCCCACGAGGACCCAGAGGTGCAGTTC
AACTGGTATGTGGACGGAGTGGAGGTGCACAACGCCAAGACCAAGCCAAGAGAG
GAGCAGTTCAACTCCACCTTCAGAGTGGTGAGCGTGCTGACCGTGGTGCACCAGG
ACTGGCTGAACGGAAAGGAGTATAAGTGTAAGGTGTCCAACAAGGGACTGCCAT
CCAGCATCGAGAAGACCATCTCCAAGACCAAGGGACAGCCAAGAGAGCCACAGG
TGTATACCCTGCCCCCATCCAGAGAGGAGATGACCAAGAACCAGGTGTCCCTGAC
CTGTCTGGTGAAGGGATTCTATCCATCCGACATCGCCGTGGAGTGGGAGTCCAAC
GGACAGCCAGAGAACAACTATAAGACCACCCCTCCAATGCTGGACTCCGACGGA
TCCTTCTTCCTGTATTCCAAGCTGACCGTGGACAAGTCCAGATGGCAGCAGGGAA
ACGTGTTCTCTTGTTCCGTGATGCACGAGGCCCTGCACAACCACTATACCCAGAA
GAGCCTGTCCCTGTCTCCAGGAAAG
6G light chain full antibody nucleotide sequence (SEQ ID NO: 39)
GACATCGfGATGACCCAGTCCCCAGACTCCCTGGCCGTGTCCCTGGGCGAGCGCG
CCACCATCAACTGCCGCGCCAGCGAATCCGTGGATAACGATCGTATTTCCTTTCTG
AACTGGTACCAGCAGAAACCAGGCCAGCCTCCTAAGCTGCTCATTTACGCCGCCA
CCAAACAGGGTACCGGCGTGCCTGACCGCTTCTCCGGCAGCGGTTCCGGCACCGA
TTTCACTCTGACCATCTCCTCCCTGCAGGCCGAAGATGTGGCAGTGTATTACTGTC AGCAGTCCAAAGAGTTTCCCTGGTCCTTTGGCGGTGGCACCAAGGTGGAGATCAA
ACGCACTGTGGCTGCACCATCTGTCTTCATCTTCCCTCCATCTGATGAGCAGTTGA
AATCCGGAACTGCCTCTGTTGTGTGCCTGCTGAATAACTTCTATCCACGCGAGGCC
AAAGTACAGTGGAAGGTGGATAACGCCCTCCAATCCGGTAACTCCCAGGAGAGT
GTCACAGAGCAGGACAGCAAGGACAGCACCTACAGCCTCAGCAGCACCCTGACC
CTGAGCAAAGCAGACTACGAGAAACACAAAGTCTACGCCTGCGAAGTCACCCAT
CAGGGCCTGAGTTCTCCAGTCACAAAGAGCTTCAACCGCGGTGAGTGC

Claims

CLAIMS The claimed invention is:
1. A method for treating a disease characterized by aberrant deposition of β- amyloid in a subject, comprising administering to the subject an effective amount of an antibody that specifically binds to a β-amyloid peptide or an aggregated form of a β-amyloid peptide, wherein the antibody comprises an Fc region having impaired effector function.
2. The method of claim 1, wherein the subject is a human.
3. The method of claim 1 , wherein the disease is Alzheimer's disease.
4. The method of claim 1, wherein the disease is Down's syndrome.
5. The method of claim 1, wherein the disease is cerebral amyloid angiopathy.
6. The method of claim 1, wherein the antibody is a monoclonal antibody.
7. The method of claim 1 , wherein the antibody is a humanized antibody.
8. The method of claim 1 , wherein the antibody is a human antibody.
9. The method of claim 1 , wherein the antibody binds to the Aβ peptide with a KD of about 100 nM or less.
10. The method of claim 1 , wherein the antibody binds to the Aβ peptide with a KD of about 20 nM or less.
11. The method of claim 1 , wherein the antibody binds to the Aβ peptide with a KD of about 2 nM or less.
12. The method of claim 1, wherein the antibody binds to the C-terminus of the Aβ peptide.
13. The method of claim 1, wherein the antibody specifically binds to an epitope within residues 28-40 of Aβ1-40, 28-42 of Aβ1-42, or 28-43 of Aβ1-43.
14. The method of claim 1, wherein the antibody specifically binds to the C- terminus of the Aβ peptide selected from the group consisting of Aβ1-36, Aβ1-37, Aβ1-38, Aβi. 39, Aβ1-40, Aβ1-42, and Aβ1-43.
15. The method of claim 1, wherein the antibody specifically binds to an epitope on Aβ1-40 that includes amino acid 39 and/or 40.
16. The method of claim 15, wherein the antibody comprises a heavy chain variable region comprising amino acid sequence shown in SEQ ID NO: I5 and a light chain variable region comprising amino acid sequence shown in SEQ ID NO:2.
17. The method of claim 1, wherein the antibody binds to Aβ1-40 with higher affinity than to Aβi_42 or Aβ1-43.
18. The method of claim I5 wherein the antibody binds to an epitope on Aβ1-40 that includes amino acids 25-34 and 40.
19. The method of claim 18, wherein the antibody comprises a heavy chain variable region comprising amino acid sequence shown in SEQ ID NO: 26, and a light chain variable region comprising amino acid sequence shown in SEQ ID NO:27.
20. The method of claim I5 wherein the antibody specifically binds to an epitope within residues 1-1 6 of the Aβ peptide.
I l l
21. The method of claim 1 , Λvherein the antibody specifically binds to the N- terminus of the Aβ peptide.
22. The method of claim 1 , -wherein the antibody specifically binds to an epitope within residues 16-28 of the Aβ peptide.
23. The method of claim 1 , "wherein the Fc region of the antibody is not N- glycosylated or has an N-glycosylation pattern that is altered with respect to a native Fc region.
24. The method of claim I5 "wherein the Fc region of the antibody comprises a mutation within the N-glycosylation recognition sequence, whereby the Fc region is not N- glycosylated.
25. The method of claim 1 , wherein the Fc region of the antibody is the Fc region of a human heavy chain IgG2a comprising the amino acid mutation from alanine to serine at position 330 and from proline to serine at position 331, wherein the amino acid position is based on Kabat numbering with reference to human wildtype IgG2a sequence.
26. A method for treating a disease associated with aberrant deposition of β-amyloid in a subject, comprising administering to the subject an effective amount of an antibody that specifically binds to a β-amyloid peptide or an aggregated form of a β-amyloid peptide, wherein the antibody comprises an Fc region with a variation from a naturally occurring Fc region, wherein the variation results in impaired effector function.
27. The method of claim 26, wherein the administration of the antibody with the variation in the Fc region causes less cerebral microhemorrhage than administration of an antibody without the variation.
28. The method of claim 26, wherein the subject is a human.
29. The method of claim 26, wherein the disease is Alzheimer's disease.
30. The method of claim 26, wherein the antibody is a monoclonal antibody.
31. The method of claim 26, wherein the antibody is a humanized antibody.
32. The method of claim 26, wherein the antibody is a human antibody.
33. The method of claim 26, wherein the antibody binds to the Aβ peptide with a KD of about 100 nM or less.
34. The method of claim 26, wherein the antibody binds to the Aβ peptide with a KD of about 20 nM or less.
35. The method of claim 26, wherein the antibody binds to the Aβ peptide with a KD of about 2 nM or less.
36. The method of claim 26, wherein the antibody specifically binds to an epitope within residues 28-40 of Aβ1-40, 28-42 of Aβ1-42, or 28-43 of Aβ1-43.
37. The method of claim 26, wherein the antibody specifically binds to the C- terminus of the Aβ peptide selected from the group consisting of Aβ1-36, Aβt-37, Aβi-38, Aβ!. 39, Aβ1-40, Aβ1-42, and Aβ1-43.
38. The method of claim 26, wherein the antibody specifically binds to an epitope on Aβ1-4o that includes amino acid 39 and/or 40.
39. The method of claim 38, wherein the antibody comprises a heavy chain variable region comprising amino acid sequence shown in SEQ ID NO: 1, and a light chain variable region comprising amino acid sequence shown in SEQ ID NO:2.
40. The method of claim 26, wherein the antibody binds to Ap1-40 with higher affinity than to Aβ1-42 or Aβ1-43.
41. : The method of claim 26, wherein the antibody binds to an epitope on Aβ 1-40 that includes amino acids 25-34 and 40.
42. The method of claim 41, wherein the antibody comprises a heavy chain variable region comprising amino acid sequence shown in SEQ ID NO:26, and a light chain variable region comprising amino acid sequence shown in SEQ ID NO: 27.
43. The method of claim 26, wherein the antibody specifically binds to an epitope within residues 1-16 of the Aβ peptide.
44. The method of claim 26, wherein the antibody specifically binds to the N- terminus of the Aβ peptide.
45. : The method of claim 26, wherein the antibody specifically binds to an epitope within residues 16-28 of the Aβ peptide.
46. : The method of claim 26, wherein the Fc region of the antibody is not N- glycosylated or has an N-glycosylation pattern that is altered with respect to a native Fc region.
47. The method of claim 26, wherein the Fc region of the antibody comprises a mutation within the N-glycosylation recognition sequence, whereby the Fc region is not N- glycosylated.
48. The method of claim 26, wherein the Fc region of the antibody is the Fc region of a human heavy chain IgG2a comprising the amino acid mutation from alanine to serine at position 330 and from proline to serine at position 331, wherein the amino acid position is based on Kabat numbering with reference to human wildtype IgG2a sequence.
49. An antibody comprising a heavy chain variable region comprising:
(a) a CDRl region shown in SEQ ID NO:3;
(b) a CDR2 region shown in SEQ ID NO:4; and
(c) a CDR3 region shown in SEQ ID NO:5, wherein Ll is L, V, or I; wherein Y2 is Y" or W; wherein S3 is S, T, or G; wherein L4 is L, R, A, V, S, T, Q, or E; wherein V6 is V, I, T, P", C, Q, S, N, or F; and wherein Y7 is Y, H, F, W, S, I, V, or A. wherein the antibody specifically binds to an Aβ peptide.
50. The antibody of claim 49, wherein the heavy chain variable region comprises: (a) a CDRl region shown in SEQ ID NO:3;
Qa) a CDR2 region shown in SEQ ID NO:4; and (c) a CDR3 region shown in SEQ ID NO:5.
51. The antibody of claim 49, wherein the antibody further comprises a light chain variable region.
52. The antibody of claim 49, wherein the antibody is a humanized antibody.
53. An antibody comprising a light chain variable region comprising:
(a) a CDRl region shown in SEQ ID NO:6, wherein Y8 is Y, A, or H; wherein Al 1 is A or S; and wherein Kl 2 is K or A;
(b) a CDR2 region shown in SEQ ID NO:7; and (c) a CDR3 region shown in SEQ ID NO:8, wherein Ll is L, M, N, C, F, V, K, S, Q, G, S; wherein G3 is G, S, or T; wherein T4 is T or S; wherein H5 is H or L; wherein Y6 is Y, P, A, W, Q, M, S, or E; wherein V8 is V, L, K, H, T, A, E, or M; and wherein L9 is L, I, T, S, or V; wherein the antibody specifically binds to an Aβ peptide.
54. The antibody of claim 53, wherein the antibody is a humanized antibody.
55. The antibody of claim 53, wherein the light chain variable region comprises:
(a) a CDRl region shown in SEQ ID NO:6;
(b) a CDR2 region shown in SEQ ID NO:7; and
(c) a CDR3 region shown in SEQ ID NO:8.
56. The antibody of claim 53, wherein the antibody further comprises a heavy chain variable region.
57. The antibody of claim 55, wherein the antibody further comprises a heavy chain variable region comprising:
(a) a CDRl region shown in SEQ ID NO:3;
(b) a CDR2 region shown in SEQ ID NO:4; and
(c) a CDR3 region shown in SEQ ID NO:5.
58. The antibody of claim 57, wherein the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:1.
59. The antibody of claim 57, wherein the light chain variable region comprises the amino acid sequence shown in SEQ ID NO:2.
60. The antibody of claim 59, wherein the heavy chain variable region comprises the amino acid sequence shown in SEQ ID NO:1.
61. The antibody of claim 60, wherein the heavy chain comprises the amino acid sequence shown in SEQ ID NO:11; and the light chain comprises the amino acid sequence shown in SEQ ID NO: 12.
62. The antibody of claim 49, wherein the antibody comprises an Fc region having impaired effector function.
63. The antibody of claim 62, wherein the Fc region of the antibody is not N- glycosylated or has an N-glycosylation pattern that is altered with respect to a native Fc region.
64. The antibody of claim 62, wherein the Fc region of the antibody comprises a mutation within the N-glycosylation recognition sequence, whereby the Fc region is not N- glycosylated.
65. The antibody of claim 62, wherein the Fc region of the antibody is the Fc region of a human heavy chain IgG2a comprising the amino acid mutation from alanine to serine at position 330 and from proline to serine at position 331, wherein the amino acid position is based on Kabat numbering with reference to human wildtype IgG2a sequence.
66. The antibody of claim 53, wherein the antibody comprises an Fc region having impaired effector function.
67. The antibody of claim 66, wherein the Fc region of the antibody is not N- glycosylated or has an N-glycosylation pattern that is altered with respect to a native Fc region.
68. The antibody of claim 66, wherein the Fc region of the antibody comprises a mutation within the N-glycosylation recognition sequence, whereby the Fc region is not N- glycosylated.
69. The antibody of claim 66, wherein the Fc region of the antibody is the Fc region of a human heavy chain IgG2a comprising the amino acid mutation from alanine to serine at position 330 and from proline to serine at position 331, wherein the amino acid position is based on Kabat numbering with reference to human wildtype IgG2a sequence.
70. A polynucleotide comprising a sequence encoding the antibody of any of claims 49-69.
71. A vector comprising the polynucleotide of claim 70.
72. A host cell comprising the polynucleotide of claim 70.
73. A pharmaceutical composition comprising (a) the antibody of any of claims 49-69, and (b) a pharmaceutically acceptable excipient.
74. A kit comprising the antibody of any of claims 49-69.
75. A method of making an antibody that specifically binds to an Aβ peptide, said method comprising culturing a host cell comprising the polynucleotide of claim 70 under a condition that the antibody is produced.
76. The method of claim 75, said method further comprising isolating the produced antibody.
PCT/US2005/027295 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using same WO2006036291A2 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
CN200580025690.5A CN101124247B (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and uses thereof
AP2007003890A AP2007003890A0 (en) 2004-07-30 2005-08-01 Antibodies directed against amy-loid-beta peptide and methods using same
NZ552480A NZ552480A (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods of using same
CA2575663A CA2575663C (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using same
UAA200700982A UA91683C2 (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using same
EP05778628A EP1781704A2 (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using same
AU2005290250A AU2005290250A1 (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using same
MX2007000998A MX2007000998A (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using same.
BRPI0513959-7A BRPI0513959A (en) 2004-07-30 2005-08-01 antibodies directed against beta-amyloid peptide, their pharmaceutical compositions, kit and methods of manufacture thereof
JP2007523885A JP5042828B2 (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using the antibodies
EA200700083A EA016357B1 (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using same
IL180364A IL180364A (en) 2004-07-30 2006-12-26 Use of antibodies that specifically bind to amyloid-beta peptide for the manufacture of medicaments for treating diseases characterized by aberrant deposition of amyloid-beta, pharmaceutical compositions comprising them and methods for making them
NO20066058A NO20066058L (en) 2004-07-30 2006-12-29 Antibody directed to amyloid beta-peptide, and methods for the use of the same
TNP2007000031A TNSN07031A1 (en) 2004-07-30 2007-01-29 ANTIBODIES AGAINST THE AMYLOID-BETA PEPTIDE AND METHODS FOR THEIR USE
HK08106860.6A HK1116500A1 (en) 2004-07-30 2008-06-20 Antibodies directed against amyloid-beta peptide and methods using same

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US59249404P 2004-07-30 2004-07-30
US60/592,494 2004-07-30
US65319705P 2005-02-14 2005-02-14
US60/653,197 2005-02-14
US67609305P 2005-04-29 2005-04-29
US60/676,093 2005-04-29

Publications (2)

Publication Number Publication Date
WO2006036291A2 true WO2006036291A2 (en) 2006-04-06
WO2006036291A3 WO2006036291A3 (en) 2006-07-20

Family

ID=36010949

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/027295 WO2006036291A2 (en) 2004-07-30 2005-08-01 Antibodies directed against amyloid-beta peptide and methods using same

Country Status (21)

Country Link
US (4) US7807165B2 (en)
EP (2) EP2298807A3 (en)
JP (2) JP5042828B2 (en)
KR (2) KR101068289B1 (en)
AP (1) AP2007003890A0 (en)
AU (2) AU2005290250A1 (en)
BR (1) BRPI0513959A (en)
CA (1) CA2575663C (en)
EA (1) EA016357B1 (en)
GE (1) GEP20115195B (en)
HK (1) HK1116500A1 (en)
IL (1) IL180364A (en)
MA (1) MA28757B1 (en)
MX (1) MX2007000998A (en)
NI (1) NI200700021A (en)
NO (1) NO20066058L (en)
NZ (1) NZ552480A (en)
SG (1) SG190665A1 (en)
TN (1) TNSN07031A1 (en)
TW (1) TWI355389B (en)
WO (1) WO2006036291A2 (en)

Cited By (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008030251A1 (en) * 2006-09-08 2008-03-13 Georgetown University Deglycosylated anti-amyloid beta antibodies
WO2008104580A1 (en) 2007-03-01 2008-09-04 Probiodrug Ag New use of glutaminyl cyclase inhibitors
WO2008110885A2 (en) * 2007-03-09 2008-09-18 Rinat Neuroscience Corp. Methods of treating ophthalmic diseases
WO2008156621A1 (en) * 2007-06-12 2008-12-24 Ac Immune S.A. Monoclonal anti beta amyloid antibody
WO2009033309A1 (en) * 2007-09-13 2009-03-19 Esbatech Ag HUMANIZED ANTIBODIES AGAINST THE ß-AMYLOYD PEPTIDE
WO2009052439A2 (en) 2007-10-17 2009-04-23 Elan Pharma International Limited Immunotherapy regimes dependent on apoe status
WO2009100309A2 (en) 2008-02-08 2009-08-13 Medimmune, Llc Anti-ifnar1 antibodies with reduced fc ligand affinity
US7763250B2 (en) 2005-04-29 2010-07-27 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and nucleic acids encoding same
EP2211886A2 (en) * 2007-10-15 2010-08-04 Centocor Ortho Biotech Inc. Human anti-amyloid antibodies, compositions, methods and uses
US7807165B2 (en) 2004-07-30 2010-10-05 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
WO2011029920A1 (en) 2009-09-11 2011-03-17 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
WO2011110613A1 (en) 2010-03-10 2011-09-15 Probiodrug Ag Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
WO2011131748A2 (en) 2010-04-21 2011-10-27 Probiodrug Ag Novel inhibitors
WO2012021469A1 (en) * 2010-08-12 2012-02-16 Eli Lilly And Company ANTI-N3pGlu AMYLOID BETA PEPTIDE ANTIBODIES AND USES THEREOF
WO2012123563A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Benz imidazole derivatives as inhibitors of glutaminyl cyclase
WO2012131539A1 (en) 2011-03-31 2012-10-04 Pfizer Inc. Novel bicyclic pyridinones
WO2012172449A1 (en) 2011-06-13 2012-12-20 Pfizer Inc. Lactams as beta secretase inhibitors
WO2013030713A1 (en) 2011-08-31 2013-03-07 Pfizer Inc. Hexahydropyrano [3,4-d][1,3] thiazin-2-amine compounds
WO2013106572A1 (en) * 2012-01-11 2013-07-18 Arizona Board Of Regents, A Body Corporate Of The State Of Arizona Acting For And On Behalf Of Arizona State University Bispecific antibody fragments for neurological disease proteins and methods of use
WO2013164730A1 (en) 2012-05-04 2013-11-07 Pfizer Inc. Heterocyclic substituted hexahydropyrano [3,4-d] [1,3] thiazin- 2 -amine compounds as inhibitors of app, bace1 and bace 2.
WO2014001973A1 (en) 2012-06-29 2014-01-03 Pfizer Inc. NOVEL 4-(SUBSTITUTED-AMINO)-7H-PYRROLO[2,3-d]PYRIMIDINES AS LRRK2 INHIBITORS
WO2014045156A1 (en) 2012-09-21 2014-03-27 Pfizer Inc. Novel bicyclic pyridinones
WO2014045162A1 (en) 2012-09-20 2014-03-27 Pfizer Inc. ALKYL-SUBSTITUTED HEXAHYDROPYRANO[3,4-d] [1,3]THIAZIN-2-ANIME COMPOUNDS
WO2014091352A1 (en) 2012-12-11 2014-06-19 Pfizer Inc. Hexahydropyrano [3,4-d][1,3]thiazin-2-amine compounds as inhibitors of bace1
WO2014100740A1 (en) 2012-12-21 2014-06-26 Seattle Genetics, Inc. Anti-ntb-a antibodies and related compositions and methods
WO2014097038A1 (en) 2012-12-19 2014-06-26 Pfizer Inc. CARBOCYCLIC- AND HETEROCYCLIC-SUBSTITUTED HEXAHYDROPYRANO[3,4-d][1,3]THIAZIN-2-AMINE COMPOUNDS
US8796439B2 (en) 2006-07-14 2014-08-05 Ac Immune S.A. Nucleic acid molecules encoding a humanized antibody
WO2014125394A1 (en) 2013-02-13 2014-08-21 Pfizer Inc. HETEROARYL-SUBSTITUTED HEXAHYDROPYRANO [3,4-d][1,3] THIAZIN-2-AMINE COMPOUNDS
WO2014125397A1 (en) 2013-02-15 2014-08-21 Pfizer Inc. SUBSTITUTED PHENYL HEXAHYDROPYRANO[3,4-d][1,3]THIAZIN-2-AMINE COMPOUNDS
WO2014128585A1 (en) 2013-02-19 2014-08-28 Pfizer Inc. Azabenzimidazole compounds as inhibitors of pde4 isozymes for the treatment of cns and other disorders
WO2015049616A1 (en) 2013-10-04 2015-04-09 Pfizer Inc. Novel bicyclic pyridinones as gamma-secretase modulators
EP2865670A1 (en) 2007-04-18 2015-04-29 Probiodrug AG Thiourea derivatives as glutaminyl cyclase inhibitors
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
WO2015092592A1 (en) 2013-12-17 2015-06-25 Pfizer Inc. Novel 3,4-disubstituted-1h-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7h-pyrrolo[2,3-c]pyridazines as lrrk2 inhibitors
WO2015150957A1 (en) 2014-04-01 2015-10-08 Pfizer Inc. Chromene and 1,1 a,2,7b-tetrahydrocyclopropa[c]chromene pyridopyrazinediones as gamma-secretase modulators
WO2015155626A1 (en) 2014-04-10 2015-10-15 Pfizer Inc. 2-AMINO-6-METHYL-4,4a,5,6-TETRAHYDROPYRANO[3,4-d][1,3]THIAZIN-8a(8H)-YL-1,3-THIAZOL-4-YL AMIDES
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
US9221900B2 (en) 2010-07-30 2015-12-29 Ac Immune S.A. Methods for identifying safe and functional humanized antibodies
EP2977385A1 (en) * 2009-05-12 2016-01-27 Sanofi Humanised antibodies specific to the protofibrillar form of the amyloid-beta peptide
WO2016012896A1 (en) 2014-07-24 2016-01-28 Pfizer Inc. Pyrazolopyrimidine compounds
WO2016020786A1 (en) 2014-08-06 2016-02-11 Pfizer Inc. Imidazopyridazine compounds
WO2016081640A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor / anti-bace1 multispecific antibodies and methods of use
WO2016081643A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
WO2016094566A2 (en) 2014-12-10 2016-06-16 Genentech, Inc. Blood brain barrier receptor antibodies and methods of use
WO2016125048A1 (en) 2015-02-03 2016-08-11 Pfizer Inc. Novel cyclopropabenzofuranyl pyridopyrazinediones
WO2016203347A1 (en) 2015-06-17 2016-12-22 Pfizer Inc. Tricyclic compounds and their use as phosphodiesterase inhibitors
WO2017004330A1 (en) 2015-06-30 2017-01-05 Seattle Genetics, Inc. Anti-ntb-a antibodies and related compositions and methods
WO2017046675A1 (en) 2015-09-14 2017-03-23 Pfizer Inc. Novel imidazo [4,5-c] quinoline and imidazo [4,5-c][1,5] naphthyridine derivatives as lrrk2 inhibitors
WO2017051303A1 (en) 2015-09-24 2017-03-30 Pfizer Inc. Tetrahydropyrano[3,4-d][1,3]oxazin derivatives and their use as bace inhibitors
WO2017051294A1 (en) 2015-09-24 2017-03-30 Pfizer Inc. N-[2-(3-amino-2,5-dimethyl-1,1-dioxido-5,6-dihydro-2h-1,2,4-thiadiazin-5-yl)-1,3-thiazol-4-yl] amides useful as bace inhibitors
WO2017051276A1 (en) 2015-09-24 2017-03-30 Pfizer Inc. N-[2-(2-amino-6,6-disubstituted-4, 4a, 5, 6-tetrahydropyrano [3,4-d][1,3] thiazin-8a (8h)-yl) -1, 3-thiazol-4-yl] amides
EP3156420A1 (en) 2010-12-06 2017-04-19 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
WO2017145013A1 (en) 2016-02-23 2017-08-31 Pfizer Inc. 6,7-dihydro-5h-pyrazolo[5,1-b][1,3]oxazine-2-carboxamide compounds
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
WO2018002760A1 (en) 2016-07-01 2018-01-04 Pfizer Inc. 5,7-dihydro-pyrrolo-pyridine derivatives for treating neurological and neurodegenerative diseases
RU2641256C2 (en) * 2011-06-30 2018-01-16 Чугаи Сейяку Кабусики Кайся Heterodimerizated polypeptide
US9944696B2 (en) 2016-01-15 2018-04-17 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
US10006920B2 (en) 2013-04-26 2018-06-26 Korea Institute Of Science And Technology Diagnostic kit for diagnosis of abnormal protein aggregation- or misfolding-related diseases using dissociation of protein aggregates
WO2018163030A1 (en) 2017-03-10 2018-09-13 Pfizer Inc. Cyclic substituted imidazo[4,5-c]quinoline derivatives
WO2018163066A1 (en) 2017-03-10 2018-09-13 Pfizer Inc. Novel imidazo[4,5-c]quinoline derivatives as lrrk2 inhibitors
WO2018226992A1 (en) 2017-06-07 2018-12-13 Adrx, Inc. Tau aggregation inhibitors
WO2018234953A1 (en) 2017-06-22 2018-12-27 Pfizer Inc. Dihydro-pyrrolo-pyridine derivatives
EP3421048A1 (en) 2012-05-18 2019-01-02 Seattle Genetics, Inc. Cd33 antibodies and use of same to treat cancer
EP3431496A1 (en) * 2017-07-19 2019-01-23 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Anti- isoasp7 amyloid beta antibodies and uses thereof
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
WO2019036725A2 (en) 2017-08-18 2019-02-21 Adrx, Inc. Tau aggregation peptide inhibitors
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase
EP3508499A1 (en) 2018-01-08 2019-07-10 iOmx Therapeutics AG Antibodies targeting, and other modulators of, an immunoglobulin gene associated with resistance against anti-tumour immune responses, and uses thereof
WO2019183636A1 (en) 2018-03-23 2019-09-26 Pfizer Inc. Piperazine azaspiro derivaves
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
EP3594240A1 (en) 2013-05-20 2020-01-15 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
WO2020041541A2 (en) 2018-08-23 2020-02-27 Seattle Genetics, Inc. Anti-tigit antibodies
WO2020072357A1 (en) 2018-10-04 2020-04-09 University Of Rochester Improvement of glymphatic delivery by manipulating plasma osmolarity
US10647759B2 (en) 2017-04-20 2020-05-12 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
WO2020132230A2 (en) 2018-12-20 2020-06-25 Genentech, Inc. Modified antibody fcs and methods of use
WO2020163225A1 (en) 2019-02-05 2020-08-13 Seattle Genetics, Inc. Anti-cd228 antibodies and antibody-drug conjugates
US10766960B2 (en) 2012-12-27 2020-09-08 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
WO2021005009A1 (en) 2019-07-05 2021-01-14 Iomx Therapeutics Ag Antibodies binding igc2 of igsf11 (vsig3) and uses thereof
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
WO2021067776A2 (en) 2019-10-04 2021-04-08 Seagen Inc. Anti-pd-l1 antibodies and antibody-drug conjugates
EP3822288A1 (en) 2019-11-18 2021-05-19 Deutsches Krebsforschungszentrum, Stiftung des öffentlichen Rechts Antibodies targeting, and other modulators of, the cd276 antigen, and uses thereof
US11078291B2 (en) 2016-02-17 2021-08-03 Seagen Inc. BCMA antibodies and use of same to treat cancer and immunological disorders
WO2021195362A1 (en) 2020-03-26 2021-09-30 Seagen Inc. Methods of treating multiple myeloma
US11142563B2 (en) 2012-06-14 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified Fc region
WO2022008027A1 (en) 2020-07-06 2022-01-13 Iomx Therapeutics Ag Antibodies binding igv of igsf11 (vsig3) and uses thereof
WO2022031652A1 (en) 2020-08-04 2022-02-10 Seagen Inc. Anti-cd228 antibodies and antibody-drug conjugates
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11312763B2 (en) 2016-07-01 2022-04-26 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
WO2022098972A1 (en) 2020-11-08 2022-05-12 Seagen Inc. Combination-therapy antibody drug conjugate with immune cell inhibitor
WO2022096536A1 (en) 2020-11-03 2022-05-12 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Target-cell restricted, costimulatory, bispecific and bivalent anti-cd28 antibodies
WO2022162203A1 (en) 2021-01-28 2022-08-04 Vaccinvent Gmbh Method and means for modulating b-cell mediated immune responses
US11434284B2 (en) 2020-07-23 2022-09-06 Othair Prothena Limited Anti-Abeta antibodies
WO2022217026A1 (en) 2021-04-09 2022-10-13 Seagen Inc. Methods of treating cancer with anti-tigit antibodies
WO2023016826A2 (en) 2021-08-05 2023-02-16 Vaccinvent Gmbh Method and means for enhancing therapeutic antibodies
WO2023049694A1 (en) 2021-09-23 2023-03-30 Seagen Inc. Methods of treating multiple myeloma
WO2024068777A1 (en) 2022-09-28 2024-04-04 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Modified ace2 proteins with improved activity against sars-cov-2

Families Citing this family (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080050367A1 (en) * 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
TWI239847B (en) * 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US7790856B2 (en) * 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
MY139983A (en) * 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
EP1633786A4 (en) * 2002-10-09 2007-07-25 Rinat Neuroscience Corp Methods of treating alzheimer s disease using antibodies directed against amyloid beta peptide and compositions thereof
CA2513722A1 (en) * 2003-02-01 2004-08-19 Neuralab Limited Active immunization to generate antibodies to soluble a-beta
CA2590337C (en) * 2004-12-15 2017-07-11 Neuralab Limited Humanized amyloid beta antibodies for use in improving cognition
US8420093B2 (en) 2005-02-14 2013-04-16 Merck Sharp & Dohme Corp. Anti-ADDL monoclonal antibody and use thereof
US7731962B2 (en) 2005-02-14 2010-06-08 Merck & Co., Inc. Anti-ADDL monoclonal antibody and use thereof
JP5419131B2 (en) * 2005-12-12 2014-02-19 エーシー イミューン ソシエテ アノニム Β1-42 specific monoclonal antibodies with therapeutic properties
WO2009017467A1 (en) 2007-07-27 2009-02-05 Elan Pharma International Limited Treatment of amyloidogenic diseases
US8784810B2 (en) * 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
US8003097B2 (en) * 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
BRPI0810118A8 (en) * 2007-04-18 2015-09-29 Janssen Alzheimer Immunotherap METHOD TO TREAT DISEASE, METHOD TO EFFECT CAA PROPHYLAXIS, USE OF AN AGENT, METHOD TO REDUCE VASCULAR AMYLOID IN A PATIENT, AND, KIT FOR TREATMENT
US8323654B2 (en) * 2007-05-14 2012-12-04 Medtronic, Inc. Anti-amyloid beta antibodies conjugated to sialic acid-containing molecules
TW201518320A (en) * 2007-06-12 2015-05-16 Ac Immune Sa Antibody specifically binding to beta amyloid and relevant nucleic acid molecule, expression vector, cell, composition, kit, method, and use
US8048420B2 (en) 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
US20100239570A1 (en) * 2007-09-13 2010-09-23 Roger Nitsch Moncolonal amyloid beta (abeta) - specific antibody and uses thereof
WO2009048538A2 (en) * 2007-10-05 2009-04-16 Genentech, Inc. Humanized antibody
NZ601858A (en) * 2007-10-05 2014-03-28 Genentech Inc Methods and compositions for diagnosis and treatment of amyloidosis
RS53174B (en) 2007-10-05 2014-06-30 Genentech Inc. Use of anti-amyloid beta antibody in ocular diseases
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US8614297B2 (en) * 2008-12-22 2013-12-24 Hoffmann-La Roche Inc. Anti-idiotype antibody against an antibody against the amyloid β peptide
MX2011007916A (en) 2009-01-30 2011-11-18 Adlyfe Inc Conformationally dynamic peptides.
SI2448968T1 (en) * 2009-06-29 2021-07-30 Bioarctic Ab ANTIBODIES SELECTIVE FOR N-TERMINALTRUNCATED AMYLOID-p PROTOFIBRILS/OLIGOMERS
US9320793B2 (en) 2010-07-14 2016-04-26 Acumen Pharmaceuticals, Inc. Method for treating a disease associated with soluble, oligomeric species of amyloid beta 1-42
US9176151B2 (en) 2010-07-14 2015-11-03 Acumen Pharmaceuticals, Inc. Antibodies, kit and method for detecting amyloid beta oligomers
WO2012009442A2 (en) * 2010-07-14 2012-01-19 Merck Sharp & Dohme Corp. Anti-addl monoclonal antibody and uses thereof
KR102129220B1 (en) * 2011-09-23 2020-07-02 에이씨 이뮨 에스.에이. Vaccine therapy
US9534048B2 (en) * 2012-08-24 2017-01-03 University Health Network Antibodies to TTR and methods of use
WO2016081746A2 (en) 2014-11-21 2016-05-26 Bristol-Myers Squibb Company Antibodies comprising modified heavy constant regions
ES2807182T3 (en) 2014-11-21 2021-02-22 Bristol Myers Squibb Co CD73 Antibodies and Their Uses
JO3555B1 (en) 2015-10-29 2020-07-05 Merck Sharp & Dohme Antibody neutralizing human respiratory syncytial virus
WO2017079369A2 (en) * 2015-11-03 2017-05-11 Glaxosmithkline Llc Novel antibodies
US10851164B2 (en) 2017-04-13 2020-12-01 Aduro Biotech Holdings, Europe B.V. Anti-SIRPα antibodies
MX2019013072A (en) 2017-05-02 2019-12-16 Merck Sharp & Dohme Formulations of anti-lag3 antibodies and co-formulations of anti-lag3 antibodies and anti-pd-1 antibodies.
JOP20190260A1 (en) 2017-05-02 2019-10-31 Merck Sharp & Dohme Stable formulations of programmed death receptor 1 (pd-1) antibodies and methods of use thereof
CA3067597C (en) 2017-06-29 2023-03-21 The Trustees Of Columbia University In The City Of New York Chimeric antibodies for treatment of amyloid deposition diseases
US11382974B2 (en) 2017-08-01 2022-07-12 The Trustees Of Columbia University In The City Of New York Methods and compositions for treatment of amyloid deposition diseases
WO2019148412A1 (en) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Anti-pd-1/lag3 bispecific antibodies
CN111334480B (en) * 2020-05-18 2020-09-01 苏州仁端生物医药科技有限公司 Hybridoma cell strain secreting anti-Abeta 1-42 monoclonal antibody and application thereof
EP4252243A2 (en) 2020-11-30 2023-10-04 Enigma Biointelligence, Inc. Non-invasive assessment of alzheimer's disease
TW202300517A (en) 2021-03-12 2023-01-01 美商美國禮來大藥廠 Anti-amyloid beta antibodies and uses thereof
WO2022251048A1 (en) 2021-05-24 2022-12-01 Eli Lilly And Company Anti-amyloid beta antibodies and uses thereof

Family Cites Families (181)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3143171A (en) * 1961-04-11 1964-08-04 Shell Oil Co Underwater well guide system
US3787637A (en) * 1971-07-26 1974-01-22 Audichron Co Announcing system
FR2413974A1 (en) 1978-01-06 1979-08-03 David Bernard DRYER FOR SCREEN-PRINTED SHEETS
US4403112A (en) 1981-05-18 1983-09-06 Modafferi Acoustical Systems, Ltd. Phase shift low frequency loudspeaker system
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4769330A (en) 1981-12-24 1988-09-06 Health Research, Incorporated Modified vaccinia virus and methods for making and using the same
US4603112A (en) 1981-12-24 1986-07-29 Health Research, Incorporated Modified vaccinia virus
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4891268A (en) 1984-03-26 1990-01-02 Metal Coatings International Inc. Coated metal substrates with anticorrosion coating composition
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US4754065A (en) 1984-12-18 1988-06-28 Cetus Corporation Precursor to nucleic acid probe
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4751180A (en) 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
GB8508845D0 (en) 1985-04-04 1985-05-09 Hoffmann La Roche Vaccinia dna
US4666829A (en) 1985-05-15 1987-05-19 University Of California Polypeptide marker for Alzheimer's disease and its use for diagnosis
GB8516081D0 (en) 1985-06-25 1985-07-31 Ciba Geigy Ag Assay & purification of amyloid components
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4777127A (en) 1985-09-30 1988-10-11 Labsystems Oy Human retrovirus-related products and methods of diagnosing and treating conditions associated with said retrovirus
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
US4800159A (en) 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US5231170A (en) 1986-08-27 1993-07-27 Paul Averback Antibodies to dense microspheres
US5525339A (en) 1986-08-27 1996-06-11 Dms Pharmaceutical Inc. Isolated components of dense microspheres derived from mammalian brain tissue and antibodies thereto
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
GB8702816D0 (en) 1987-02-07 1987-03-11 Al Sumidaie A M K Obtaining retrovirus-containing fraction
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
US5231000A (en) 1987-10-08 1993-07-27 The Mclean Hospital Antibodies to A4 amyloid peptide
CA1339014C (en) 1987-10-08 1997-03-25 Ronald E. Majocha Antibodies to a4 amyloid peptide
US5422120A (en) 1988-05-30 1995-06-06 Depotech Corporation Heterovesicular liposomes
AP129A (en) 1988-06-03 1991-04-17 Smithkline Biologicals S A Expression of retrovirus gag protein eukaryotic cells
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5047335A (en) 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO1990007936A1 (en) 1989-01-23 1990-07-26 Chiron Corporation Recombinant therapies for infection and hyperproliferative disorders
AU5344190A (en) 1989-03-21 1990-10-22 Vical, Inc. Expression of exogenous polynucleotide sequences in a vertebrate
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US6673776B1 (en) 1989-03-21 2004-01-06 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate, mammal, fish, bird or human
US5262332A (en) 1989-04-05 1993-11-16 Brigham And Women's Hospital Diagnostic method for Alzheimer's disease: examination of non-neural tissue
US5234814A (en) 1989-06-01 1993-08-10 Du Pont Merck Pharmaceutical Company Diagnostic assay for alzheimer's disease
EP0479909B1 (en) 1989-06-29 1996-10-30 Medarex, Inc. Bispecific reagents for aids therapy
EP0845537A1 (en) 1989-08-18 1998-06-03 Chiron Corporation Recombinant retroviruses delivering vector constructs to target cells
US5585362A (en) 1989-08-22 1996-12-17 The Regents Of The University Of Michigan Adenovirus vectors for gene therapy
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
ZA911974B (en) 1990-03-21 1994-08-22 Res Dev Foundation Heterovesicular liposomes
CA2079880A1 (en) 1990-04-24 1991-10-25 William E. Van Nostrand Purification, detection and methods of use of protease nexin-2
US5753624A (en) 1990-04-27 1998-05-19 Milkhaus Laboratory, Inc. Materials and methods for treatment of plaquing disease
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
US5385915A (en) 1990-05-16 1995-01-31 The Rockefeller University Treatment of amyloidosis associated with Alzheimer disease using modulators of protein phosphorylation
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
JP2938569B2 (en) 1990-08-29 1999-08-23 ジェンファーム インターナショナル,インコーポレイティド Method for producing xenogeneic immunoglobulin and transgenic mouse
WO1992009690A2 (en) 1990-12-03 1992-06-11 Genentech, Inc. Enrichment method for variant proteins with altered binding properties
US5164295A (en) 1991-03-06 1992-11-17 The Upjohn Company Method for identifying amyloid protein-extracellular matrix protein affinity altering compounds
US5278299A (en) 1991-03-18 1994-01-11 Scripps Clinic And Research Foundation Method and composition for synthesizing sialylated glycosyl compounds
WO1992020373A1 (en) 1991-05-14 1992-11-26 Repligen Corporation Heteroconjugate antibodies for treatment of hiv infection
EP0519596B1 (en) 1991-05-17 2005-02-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
DE69233254T2 (en) 1991-06-14 2004-09-16 Genentech, Inc., South San Francisco Humanized Heregulin antibody
WO1994004679A1 (en) 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
GB9114324D0 (en) 1991-07-03 1991-08-21 Kontor Moulding Systems Ltd Injection moulding of thermoplastic polymers
GB9115364D0 (en) 1991-07-16 1991-08-28 Wellcome Found Antibody
ATE237694T1 (en) 1991-08-20 2003-05-15 Us Gov Health & Human Serv ADENOVIRUS-MEDIATED GENE TRANSFER INTO THE GASTROINTESTINAL TRACT
CA2078539C (en) 1991-09-18 2005-08-02 Kenya Shitara Process for producing humanized chimera antibody
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1993010218A1 (en) 1991-11-14 1993-05-27 The United States Government As Represented By The Secretary Of The Department Of Health And Human Services Vectors including foreign genes and negative selective markers
WO1993010260A1 (en) 1991-11-21 1993-05-27 The Board Of Trustees Of The Leland Stanford Junior University Controlling degradation of glycoprotein oligosaccharides by extracellular glycosisases
GB9125623D0 (en) 1991-12-02 1992-01-29 Dynal As Cell modification
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
FR2688514A1 (en) 1992-03-16 1993-09-17 Centre Nat Rech Scient Defective recombinant adenoviruses expressing cytokines and antitumour drugs containing them
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5604102A (en) 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
US5441870A (en) 1992-04-15 1995-08-15 Athena Neurosciences, Inc. Methods for monitoring cellular processing of β-amyloid precursor protein
US5851787A (en) 1992-04-20 1998-12-22 The General Hospital Corporation Nucleic acid encoding amyloid precursor-like protein and uses thereof
TW327194B (en) 1992-05-01 1998-02-21 American Cyanamid Co Novel amyloid precursor proteins and methods of using same
EP0650370A4 (en) 1992-06-08 1995-11-22 Univ California Methods and compositions for targeting specific tissue.
WO1993025698A1 (en) 1992-06-10 1993-12-23 The United States Government As Represented By The Vector particles resistant to inactivation by human serum
US5766846A (en) 1992-07-10 1998-06-16 Athena Neurosciences Methods of screening for compounds which inhibit soluble β-amyloid peptide production
GB2269175A (en) 1992-07-31 1994-02-02 Imperial College Retroviral vectors
JPH08500017A (en) 1992-08-17 1996-01-09 ジェネンテク,インコーポレイテッド Bispecific immune adhesin
WO1994009370A1 (en) 1992-10-09 1994-04-28 Massachusetts Institute Of Technology Release of alzheimer amyloid precursor stimulated by activation of muscarinic acetylcholine receptors
US5962463A (en) 1992-10-09 1999-10-05 Massachusetts Institute Of Technology Methods of stimulating non-amyloidogenic processing of the amyloid precursor protein
WO1994009371A1 (en) 1992-10-09 1994-04-28 Massachusetts Institute Of Technology Antemortem diagnostic test for alzheimer's disease
US6210671B1 (en) 1992-12-01 2001-04-03 Protein Design Labs, Inc. Humanized antibodies reactive with L-selectin
EP1024198A3 (en) 1992-12-03 2002-05-29 Genzyme Corporation Pseudo-adenoviral vectors for the gene therapy of haemophiliae
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
CA2115900A1 (en) 1993-02-22 1994-08-23 Gerald W. Becker Pharmaceutical screens and antibodies
JP3545403B2 (en) 1993-04-22 2004-07-21 スカイファルマ インコーポレイテッド Cyclodextrin liposomes encapsulating pharmaceutical compounds and methods of use
US6107050A (en) 1993-05-03 2000-08-22 The United States Of America As Represented By The Department Of Health And Human Services Diagnostic test for alzheimers disease
US6180377B1 (en) 1993-06-16 2001-01-30 Celltech Therapeutics Limited Humanized antibodies
WO1995000655A1 (en) 1993-06-24 1995-01-05 Mc Master University Adenovirus vectors for gene therapy
ATE440957T1 (en) 1993-09-15 2009-09-15 Novartis Vaccines & Diagnostic RECOMBINANT ALPHAVIRUS VECTORS
US6015686A (en) 1993-09-15 2000-01-18 Chiron Viagene, Inc. Eukaryotic layered vector initiation systems
ATE213507T1 (en) 1993-10-20 2002-03-15 Univ Duke METHOD FOR BINDING MATERIAL TO BETA-AMYLOID PEPTIDE
DE69435223D1 (en) 1993-10-25 2009-09-03 Canji Inc Recombinant adenovirus vector and method of use
RU2160093C2 (en) 1993-11-16 2000-12-10 Скайефарма Инк. Vesicles with controlled active ingredient release
US6436908B1 (en) 1995-05-30 2002-08-20 Duke University Use of exogenous β-adrenergic receptor and β-adrenergic receptor kinase gene constructs to enhance myocardial function
EP1881075B1 (en) 1994-05-09 2013-07-24 Oxford BioMedica (UK) Limited Retroviral vectors having a reduced recombination rate
US5525126A (en) 1994-10-31 1996-06-11 Agricultural Utilization Research Institute Process for production of esters for use as a diesel fuel substitute using a non-alkaline catalyst
US6114133A (en) 1994-11-14 2000-09-05 Elan Pharmaceuticals, Inc. Methods for aiding in the diagnosis of Alzheimer's disease by measuring amyloid-β peptide (x-≧41)
US5589154A (en) 1994-11-22 1996-12-31 Rutgers, The State University Of New Jersey Methods for the prevention or treatment of vascular hemorrhaging and Alzheimer's disease
AU4594996A (en) 1994-11-30 1996-06-19 Chiron Viagene, Inc. Recombinant alphavirus vectors
US5688651A (en) 1994-12-16 1997-11-18 Ramot University Authority For Applied Research And Development Ltd. Prevention of protein aggregation
US5804560A (en) 1995-01-06 1998-09-08 Sibia Neurosciences, Inc. Peptide and peptide analog protease inhibitors
US5786180A (en) 1995-02-14 1998-07-28 Bayer Corporation Monoclonal antibody 369.2B specific for β A4 peptide
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
US6265150B1 (en) 1995-06-07 2001-07-24 Becton Dickinson & Company Phage antibodies
AU6383396A (en) 1995-06-07 1996-12-30 Athena Neurosciences, Inc. Beta-secretase, antibodies to beta-secretase, and assays for detecting beta-secretase inhibition
US6221645B1 (en) 1995-06-07 2001-04-24 Elan Pharmaceuticals, Inc. β-secretase antibody
WO1997012992A2 (en) 1995-10-02 1997-04-10 Royal Netherlands Academy Of Arts And Sciences Diagnosis method for a disease caused by a frameshift mutation in a gene and reagents therefore
EP0953052B1 (en) 1996-05-06 2009-03-04 Oxford BioMedica (UK) Limited Crossless retroviral vectors
US6107029A (en) 1996-07-31 2000-08-22 Message Pharmaceticals, Inc. Universal method for detecting interactions between RNA molecules and RNA binding proteins
CA2279651A1 (en) 1996-09-05 1998-03-12 Massachusetts Institute Of Technology Compositions and methods for treatment of neurological disorders and neurodegenerative diseases
US5976817A (en) 1996-10-31 1999-11-02 Trustees Of Boston University Diagnostic method for detecting Alzheimer's disease in living patients
US20030068316A1 (en) 1997-02-05 2003-04-10 Klein William L. Anti-ADDL antibodies and uses thereof
US6221670B1 (en) 1997-03-21 2001-04-24 Scios Inc. Methods to identify β-amyloid reducing agents
US20020086847A1 (en) 1997-04-09 2002-07-04 Mindset Biopharmaceuticals (Usa) Recombinant antibodies specific for beta-amyloid ends, DNA encoding and methods of use thereof
US8173127B2 (en) 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US6376471B1 (en) 1997-10-10 2002-04-23 Johns Hopkins University Gene delivery compositions and methods
US6761888B1 (en) 2000-05-26 2004-07-13 Neuralab Limited Passive immunization treatment of Alzheimer's disease
US6710226B1 (en) 1997-12-02 2004-03-23 Neuralab Limited Transgenic mouse assay to determine the effect of Aβ antibodies and Aβ Fragments on alzheimer's disease characteristics
US6750324B1 (en) 1997-12-02 2004-06-15 Neuralab Limited Humanized and chimeric N-terminal amyloid beta-antibodies
US6743427B1 (en) 1997-12-02 2004-06-01 Neuralab Limited Prevention and treatment of amyloidogenic disease
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US7179892B2 (en) 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
GB9809951D0 (en) * 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
US20030147882A1 (en) 1998-05-21 2003-08-07 Alan Solomon Methods for amyloid removal using anti-amyloid antibodies
EP1002862A1 (en) 1998-11-12 2000-05-24 Nitsch, Roger M., Prof. Dr. Methods of diagnosing or treating neurological diseases
US6143171A (en) 1999-04-07 2000-11-07 Van Aarsen; Freda Martha Magnetic device for treatment of fluids
AU4753900A (en) 1999-05-03 2000-11-17 Evotec Biosystems Ag Methods of diagnosing or treating alzheimer's disease
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
DE60040024D1 (en) 1999-06-16 2008-10-02 Boston Biomedical Res Inst IMMUNOLOGICAL CONTROL OF THE BETA-AMYLOID CONTENT IN VIVO
US6255054B1 (en) 1999-09-09 2001-07-03 Jacques Hugon Polymorphism of the human GluR-5 gene and risk factor for alzheimer disease
US6294171B2 (en) 1999-09-14 2001-09-25 Milkhaus Laboratory, Inc. 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
AU2001253158A1 (en) 2000-04-05 2001-10-23 University Of Tennessee Research Corporation Methods of investigating, diagnosing, and treating amyloidosis
US20020009445A1 (en) 2000-07-12 2002-01-24 Yansheng Du Human beta-amyloid antibody and use thereof for treatment of alzheimer's disease
EP1172378A1 (en) 2000-07-12 2002-01-16 Richard Dr. Dodel Human beta-amyloid antibody and use thereof for treatment of alzheimer's disease
AU1225702A (en) 2000-09-19 2002-04-02 Evotec Neurosciences Gmbh Methods and compounds for treating brain amyloidosis
PE20020574A1 (en) 2000-12-06 2002-07-02 Wyeth Corp HUMANIZED ANTIBODIES THAT RECOGNIZE THE AMYLOID PEPTIDE BETA
AU2002306436A1 (en) 2001-02-12 2002-10-15 Asm America, Inc. Improved process for deposition of semiconductor films
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
US6573276B2 (en) 2001-05-09 2003-06-03 Boehringer Ingelheim Pharma Kg Muscarinic M1 agonist as an inhibitor of beta-amyloid (Aβ40 and Aβ42)-synthesis
US20020197258A1 (en) 2001-06-22 2002-12-26 Ghanbari Hossein A. Compositions and methods for preventing protein aggregation in neurodegenerative diseases
CA2452104A1 (en) 2001-08-17 2003-02-27 Eli Lilly And Company Use of antibodies having high affinity for soluble ass to treat conditions and diseases related to ass
WO2003039467A2 (en) 2001-11-02 2003-05-15 Diagenics International Corporation Monoclonal antibodies specific for beta-amyloid.
JP2005508171A (en) 2001-11-02 2005-03-31 タノックス インコーポレーテッド B cell lymphoma specific antigen for use in diagnosis and prevention of B cell malignancy
MY139983A (en) * 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
AU2003226356A1 (en) * 2002-04-12 2003-10-27 Ramot At Tel Aviv University Ltd. Prevention of brain inflammation as a result of induced autoimmune response
ES2318123T3 (en) 2002-04-25 2009-05-01 Eli Lilly And Company PROCEDURE TO TREAT ANXIETY IN SUBJECTS OF ADVANCED AGE.
EP1535075A2 (en) 2002-06-20 2005-06-01 Axonyx, Inc. Assay method
AUPS306402A0 (en) 2002-06-20 2002-07-11 Aguilar, Marie-Isabel Assay method
ES2201929B1 (en) 2002-09-12 2005-05-16 Araclon Biotech, S.L. POLYCLONAL ANTIBODIES, METHOD OF PREPARATION AND USE OF THE SAME.
DE60336848D1 (en) 2002-09-12 2011-06-01 Univ California IMMUNOGENOUS AND CORRESPONDING ANTIBODIES SPECIFIC FOR COMMON HIGH-MOLECULAR AGGREGATION INTERMEDIATE PRODUCTS OF AMYLOIDS FROM PROTEINS OF DIFFERENT SEQUENCE
WO2004029629A1 (en) 2002-09-27 2004-04-08 Janssen Pharmaceutica N.V. N-11 truncated amyloid-beta nomoclonal antibodies, compositions, methods and uses
EP1633786A4 (en) 2002-10-09 2007-07-25 Rinat Neuroscience Corp Methods of treating alzheimer s disease using antibodies directed against amyloid beta peptide and compositions thereof
WO2004056318A2 (en) 2002-12-19 2004-07-08 New York University Method for treating amyloid disease
ES2382918T3 (en) 2002-12-24 2012-06-14 Rinat Neuroscience Corp. Anti-NGF antibodies and procedures using them
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
EP1597280B2 (en) 2003-02-26 2016-08-24 Institute for Research in Biomedicine Monoclonal antibody production by ebv transformation of b cells
US20050129695A1 (en) 2003-03-28 2005-06-16 Marc Mercken Anti-amyloid antibodies, compositions, methods and uses
WO2004095031A1 (en) 2003-04-24 2004-11-04 Universität Zürich Method of monitoring immunotherapy
ES2246177B1 (en) 2003-05-08 2007-03-01 Universidad De Zaragoza. USE OF ANTIBODIES FOR THE TREATMENT OF AMYLOID DISEASES.
US20050124016A1 (en) 2003-08-01 2005-06-09 Enh Research Institute Antibodies specific for toxic amyloid beta protein oligomers
WO2005018424A2 (en) 2003-08-18 2005-03-03 Research Foundation For Mental Hygiene, Inc. Antibodies specific for fibrillar amyloid and a procedure to detect fibrillar amyloid deposits
EP1666061A1 (en) 2003-09-09 2006-06-07 Takeda Pharmaceutical Company Limited Use of antibody
US20070110750A1 (en) 2003-09-12 2007-05-17 The Regents Of The University Of California Monoclonal antibodies specific for high molecular weight aggregation intermediates common to amyloids formed from proteins of differing sequence
US20110142858A1 (en) * 2004-06-07 2011-06-16 Ramot At Tel Aviv University Ltd. Method of Passsive Immunization Against Disease or Disorder Charcterized by Amyloid Aggregation with Diminished Risk of Neuroinflammation
SG190665A1 (en) 2004-07-30 2013-06-28 Rinat Neuroscience Corp Antibodies directed against amyloid-beta peptide and methods using same
MY148086A (en) 2005-04-29 2013-02-28 Rinat Neuroscience Corp Antibodies directed against amyloid-beta peptide and methods using same
TWI453217B (en) 2007-06-12 2014-09-21 Ac Immune Sa Monoclonal anti-beta-amyloid antibody

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (157)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10464976B2 (en) 2003-01-31 2019-11-05 AbbVie Deutschland GmbH & Co. KG Amyloid β(1-42) oligomers, derivatives thereof and antibodies thereto, methods of preparation thereof and use thereof
US7927594B2 (en) 2004-07-30 2011-04-19 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide
US8268593B2 (en) 2004-07-30 2012-09-18 Rinat Neuroscience Corp. Polynucleotides encoding antibodies directed against amyloid-beta peptide
US7807165B2 (en) 2004-07-30 2010-10-05 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
AU2006242546B2 (en) * 2005-04-29 2011-04-21 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US8398978B2 (en) 2005-04-29 2013-03-19 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and methods using same
US7763250B2 (en) 2005-04-29 2010-07-27 Rinat Neuroscience Corp. Antibodies directed against amyloid-beta peptide and nucleic acids encoding same
US10538581B2 (en) 2005-11-30 2020-01-21 Abbvie Inc. Anti-Aβ globulomer 4D10 antibodies
US10208109B2 (en) 2005-11-30 2019-02-19 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US10323084B2 (en) 2005-11-30 2019-06-18 Abbvie Inc. Monoclonal antibodies against amyloid beta protein and uses thereof
US8796439B2 (en) 2006-07-14 2014-08-05 Ac Immune S.A. Nucleic acid molecules encoding a humanized antibody
WO2008030251A1 (en) * 2006-09-08 2008-03-13 Georgetown University Deglycosylated anti-amyloid beta antibodies
US9951125B2 (en) 2006-11-30 2018-04-24 Abbvie Inc. Aβ conformer selective anti-Aβ globulomer monoclonal antibodies
EP2481408A2 (en) 2007-03-01 2012-08-01 Probiodrug AG New use of glutaminyl cyclase inhibitors
WO2008104580A1 (en) 2007-03-01 2008-09-04 Probiodrug Ag New use of glutaminyl cyclase inhibitors
JP2009062358A (en) * 2007-03-09 2009-03-26 Rinat Neuroscience Corp Method of treating ophthalmic disease
WO2008110885A3 (en) * 2007-03-09 2009-01-15 Rinat Neuroscience Corp Methods of treating ophthalmic diseases
WO2008110885A2 (en) * 2007-03-09 2008-09-18 Rinat Neuroscience Corp. Methods of treating ophthalmic diseases
AU2008224600B2 (en) * 2007-03-09 2011-09-29 Rinat Neuroscience Corp. Methods of treating ophthalmic diseases
EP2865670A1 (en) 2007-04-18 2015-04-29 Probiodrug AG Thiourea derivatives as glutaminyl cyclase inhibitors
US9585956B2 (en) 2007-06-12 2017-03-07 Ac Immune S.A. Polynucleotides encoding anti-amyloid beta monoclonal antibodies
US9175094B2 (en) 2007-06-12 2015-11-03 Ac Immune S.A. Monoclonal antibody
EP2527366A1 (en) * 2007-06-12 2012-11-28 AC Immune S.A. Monoclonal anti beta amyloid antibody
WO2008156621A1 (en) * 2007-06-12 2008-12-24 Ac Immune S.A. Monoclonal anti beta amyloid antibody
RU2475500C2 (en) * 2007-09-13 2013-02-20 Деленекс Терапьютикс Аг HUMANISED ANTIBODIES TO β-AMYLOID PEPTIDE
WO2009033309A1 (en) * 2007-09-13 2009-03-19 Esbatech Ag HUMANIZED ANTIBODIES AGAINST THE ß-AMYLOYD PEPTIDE
US8323647B2 (en) 2007-09-13 2012-12-04 Delenex Therapeutics Ag Humanized antibodies against the β-amyloid peptide
US9273126B2 (en) 2007-09-13 2016-03-01 Delenex Therapeutics Ag Humanized antibodies against the beta-amyloid peptide
CN101842388B (en) * 2007-09-13 2013-09-04 德勒尼克斯治疗股份公司 Humanized antibodies against the beta-amyloyd peptide
EP2211886A4 (en) * 2007-10-15 2011-07-27 Centocor Ortho Biotech Inc Human anti-amyloid antibodies, compositions, methods and uses
EP2211886A2 (en) * 2007-10-15 2010-08-04 Centocor Ortho Biotech Inc. Human anti-amyloid antibodies, compositions, methods and uses
EP2952524A1 (en) 2007-10-17 2015-12-09 Janssen Sciences Ireland UC Immunotherapy regimes dependent on apoe status
WO2009052439A2 (en) 2007-10-17 2009-04-23 Elan Pharma International Limited Immunotherapy regimes dependent on apoe status
EP2250279A4 (en) * 2008-02-08 2011-03-30 Medimmune Llc Anti-ifnar1 antibodies with reduced fc ligand affinity
US9988459B2 (en) 2008-02-08 2018-06-05 Astrazeneca Ab Anti-IFNAR1 antibodies with reduced Fc ligand affinity
EP2250279A2 (en) * 2008-02-08 2010-11-17 MedImmune, LLC Anti-ifnar1 antibodies with reduced fc ligand affinity
US10301390B2 (en) 2008-02-08 2019-05-28 Astrazeneca Ab Anti-IFNAR1 antibodies with reduced Fc ligand affinity
WO2009100309A2 (en) 2008-02-08 2009-08-13 Medimmune, Llc Anti-ifnar1 antibodies with reduced fc ligand affinity
EP3050898A1 (en) * 2009-05-12 2016-08-03 Sanofi Humanized antibodies specific to the protofibrillar form of the beta-amyloid peptide
EP2977385A1 (en) * 2009-05-12 2016-01-27 Sanofi Humanised antibodies specific to the protofibrillar form of the amyloid-beta peptide
WO2011029920A1 (en) 2009-09-11 2011-03-17 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
WO2011110613A1 (en) 2010-03-10 2011-09-15 Probiodrug Ag Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
US9822171B2 (en) 2010-04-15 2017-11-21 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
WO2011131748A2 (en) 2010-04-21 2011-10-27 Probiodrug Ag Novel inhibitors
US9221900B2 (en) 2010-07-30 2015-12-29 Ac Immune S.A. Methods for identifying safe and functional humanized antibodies
EP3339323A1 (en) * 2010-08-12 2018-06-27 Eli Lilly And Company Anti-n3pglu amyloid beta peptide antibodies and uses thereof
CN105111308A (en) * 2010-08-12 2015-12-02 伊莱利利公司 Anti-N3pGlu amyloid BETA peptide antibodies and uses thereof
WO2012021469A1 (en) * 2010-08-12 2012-02-16 Eli Lilly And Company ANTI-N3pGlu AMYLOID BETA PEPTIDE ANTIBODIES AND USES THEREOF
US8961972B2 (en) 2010-08-12 2015-02-24 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
US8679498B2 (en) 2010-08-12 2014-03-25 Eli Lilly And Company Anti-N3PGLU amyloid beta peptide antibodies and uses thereof
CN105111308B (en) * 2010-08-12 2019-03-12 伊莱利利公司 Anti- N3pGlu A BETA antibody and application thereof
EP3042917A1 (en) * 2010-08-12 2016-07-13 Eli Lilly and Company Anti-n3pglu amyloid beta peptide antibodies and uses thereof
EA023021B1 (en) * 2010-08-12 2016-04-29 Эли Лилли Энд Компани ANTI-N3pGlu AMYLOID BETA PEPTIDE ANTIBODIES AND USES THEREOF
CN103068848A (en) * 2010-08-12 2013-04-24 伊莱利利公司 Anti-n3pglu amyloid beta peptide antibodies and uses thereof
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
US10047121B2 (en) 2010-08-14 2018-08-14 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
EP3786185A1 (en) 2010-12-06 2021-03-03 Seagen Inc. Humanized antibodies to liv-1 and use of same to treat cancer
EP3461847A1 (en) 2010-12-06 2019-04-03 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
EP3156420A1 (en) 2010-12-06 2017-04-19 Seattle Genetics, Inc. Humanized antibodies to liv-1 and use of same to treat cancer
WO2012123563A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Benz imidazole derivatives as inhibitors of glutaminyl cyclase
WO2012131539A1 (en) 2011-03-31 2012-10-04 Pfizer Inc. Novel bicyclic pyridinones
WO2012172449A1 (en) 2011-06-13 2012-12-20 Pfizer Inc. Lactams as beta secretase inhibitors
US9890218B2 (en) 2011-06-30 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
RU2641256C2 (en) * 2011-06-30 2018-01-16 Чугаи Сейяку Кабусики Кайся Heterodimerizated polypeptide
WO2013030713A1 (en) 2011-08-31 2013-03-07 Pfizer Inc. Hexahydropyrano [3,4-d][1,3] thiazin-2-amine compounds
US9550795B2 (en) 2011-08-31 2017-01-24 Pfizer Inc. Hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
US8933221B2 (en) 2011-08-31 2015-01-13 Pfizer Inc. Hexahydropyrano[3,4-d][1,3]thiazin-2-amine compounds
WO2013106572A1 (en) * 2012-01-11 2013-07-18 Arizona Board Of Regents, A Body Corporate Of The State Of Arizona Acting For And On Behalf Of Arizona State University Bispecific antibody fragments for neurological disease proteins and methods of use
US9512212B2 (en) 2012-01-11 2016-12-06 Arizona Board Of Regents, A Body Corporate Of The State Of Arizona, Acting For And On Behalf Of Arizona State University Bispecific antibody fragments for neurological disease proteins and methods of use
US10407495B2 (en) 2012-01-11 2019-09-10 Arizona Board Of Regents, A Body Corporate Of The State Of Arizona, Acting For And On Behalf Of Arizona State University Bispecific antibody fragments for neurological disease proteins and methods of use
WO2013164730A1 (en) 2012-05-04 2013-11-07 Pfizer Inc. Heterocyclic substituted hexahydropyrano [3,4-d] [1,3] thiazin- 2 -amine compounds as inhibitors of app, bace1 and bace 2.
EP3421048A1 (en) 2012-05-18 2019-01-02 Seattle Genetics, Inc. Cd33 antibodies and use of same to treat cancer
US11142563B2 (en) 2012-06-14 2021-10-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule containing modified Fc region
US9642855B2 (en) 2012-06-29 2017-05-09 Pfizer Inc. Substituted pyrrolo[2,3-d]pyrimidines as LRRK2 inhibitors
WO2014001973A1 (en) 2012-06-29 2014-01-03 Pfizer Inc. NOVEL 4-(SUBSTITUTED-AMINO)-7H-PYRROLO[2,3-d]PYRIMIDINES AS LRRK2 INHIBITORS
EP3255049A1 (en) 2012-06-29 2017-12-13 Pfizer Inc Novel 4-(substituted-amino)-7h-pyrrolo[2,3-d]pyrimidines as lrrk2 inhibitors
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
WO2014045162A1 (en) 2012-09-20 2014-03-27 Pfizer Inc. ALKYL-SUBSTITUTED HEXAHYDROPYRANO[3,4-d] [1,3]THIAZIN-2-ANIME COMPOUNDS
WO2014045156A1 (en) 2012-09-21 2014-03-27 Pfizer Inc. Novel bicyclic pyridinones
WO2014091352A1 (en) 2012-12-11 2014-06-19 Pfizer Inc. Hexahydropyrano [3,4-d][1,3]thiazin-2-amine compounds as inhibitors of bace1
WO2014097038A1 (en) 2012-12-19 2014-06-26 Pfizer Inc. CARBOCYCLIC- AND HETEROCYCLIC-SUBSTITUTED HEXAHYDROPYRANO[3,4-d][1,3]THIAZIN-2-AMINE COMPOUNDS
WO2014100740A1 (en) 2012-12-21 2014-06-26 Seattle Genetics, Inc. Anti-ntb-a antibodies and related compositions and methods
US10766960B2 (en) 2012-12-27 2020-09-08 Chugai Seiyaku Kabushiki Kaisha Heterodimerized polypeptide
WO2014125394A1 (en) 2013-02-13 2014-08-21 Pfizer Inc. HETEROARYL-SUBSTITUTED HEXAHYDROPYRANO [3,4-d][1,3] THIAZIN-2-AMINE COMPOUNDS
WO2014125397A1 (en) 2013-02-15 2014-08-21 Pfizer Inc. SUBSTITUTED PHENYL HEXAHYDROPYRANO[3,4-d][1,3]THIAZIN-2-AMINE COMPOUNDS
WO2014128585A1 (en) 2013-02-19 2014-08-28 Pfizer Inc. Azabenzimidazole compounds as inhibitors of pde4 isozymes for the treatment of cns and other disorders
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US10006920B2 (en) 2013-04-26 2018-06-26 Korea Institute Of Science And Technology Diagnostic kit for diagnosis of abnormal protein aggregation- or misfolding-related diseases using dissociation of protein aggregates
EP4324480A2 (en) 2013-05-20 2024-02-21 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
EP3594240A1 (en) 2013-05-20 2020-01-15 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies and methods of use
WO2015049616A1 (en) 2013-10-04 2015-04-09 Pfizer Inc. Novel bicyclic pyridinones as gamma-secretase modulators
WO2015092592A1 (en) 2013-12-17 2015-06-25 Pfizer Inc. Novel 3,4-disubstituted-1h-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7h-pyrrolo[2,3-c]pyridazines as lrrk2 inhibitors
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
WO2015150957A1 (en) 2014-04-01 2015-10-08 Pfizer Inc. Chromene and 1,1 a,2,7b-tetrahydrocyclopropa[c]chromene pyridopyrazinediones as gamma-secretase modulators
WO2015155626A1 (en) 2014-04-10 2015-10-15 Pfizer Inc. 2-AMINO-6-METHYL-4,4a,5,6-TETRAHYDROPYRANO[3,4-d][1,3]THIAZIN-8a(8H)-YL-1,3-THIAZOL-4-YL AMIDES
WO2016012896A1 (en) 2014-07-24 2016-01-28 Pfizer Inc. Pyrazolopyrimidine compounds
WO2016020786A1 (en) 2014-08-06 2016-02-11 Pfizer Inc. Imidazopyridazine compounds
WO2016081643A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor antibodies and methods of use
WO2016081640A1 (en) 2014-11-19 2016-05-26 Genentech, Inc. Anti-transferrin receptor / anti-bace1 multispecific antibodies and methods of use
WO2016094566A2 (en) 2014-12-10 2016-06-16 Genentech, Inc. Blood brain barrier receptor antibodies and methods of use
WO2016125048A1 (en) 2015-02-03 2016-08-11 Pfizer Inc. Novel cyclopropabenzofuranyl pyridopyrazinediones
EP3766885A1 (en) 2015-06-17 2021-01-20 Pfizer Inc Tricyclic compounds and their use as phosphodiesterase inhibitors
WO2016203347A1 (en) 2015-06-17 2016-12-22 Pfizer Inc. Tricyclic compounds and their use as phosphodiesterase inhibitors
WO2017004330A1 (en) 2015-06-30 2017-01-05 Seattle Genetics, Inc. Anti-ntb-a antibodies and related compositions and methods
EP4302784A2 (en) 2015-06-30 2024-01-10 Seagen Inc. Anti-ntb-a antibodies and related compositions and methods
US10039753B2 (en) 2015-09-14 2018-08-07 Pfizer Inc. Imidazo[4,5-c]quinoline and imidazo[4,5-c][1,5]naphthyridine derivatives as LRRK2 inhibitors
WO2017046675A1 (en) 2015-09-14 2017-03-23 Pfizer Inc. Novel imidazo [4,5-c] quinoline and imidazo [4,5-c][1,5] naphthyridine derivatives as lrrk2 inhibitors
WO2017051294A1 (en) 2015-09-24 2017-03-30 Pfizer Inc. N-[2-(3-amino-2,5-dimethyl-1,1-dioxido-5,6-dihydro-2h-1,2,4-thiadiazin-5-yl)-1,3-thiazol-4-yl] amides useful as bace inhibitors
WO2017051303A1 (en) 2015-09-24 2017-03-30 Pfizer Inc. Tetrahydropyrano[3,4-d][1,3]oxazin derivatives and their use as bace inhibitors
WO2017051276A1 (en) 2015-09-24 2017-03-30 Pfizer Inc. N-[2-(2-amino-6,6-disubstituted-4, 4a, 5, 6-tetrahydropyrano [3,4-d][1,3] thiazin-8a (8h)-yl) -1, 3-thiazol-4-yl] amides
US9944696B2 (en) 2016-01-15 2018-04-17 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
US11767365B2 (en) 2016-02-17 2023-09-26 Seagen Inc. BCMA antibodies and use of same to treat cancer and immunological disorders
US11078291B2 (en) 2016-02-17 2021-08-03 Seagen Inc. BCMA antibodies and use of same to treat cancer and immunological disorders
WO2017145013A1 (en) 2016-02-23 2017-08-31 Pfizer Inc. 6,7-dihydro-5h-pyrazolo[5,1-b][1,3]oxazine-2-carboxamide compounds
US11312763B2 (en) 2016-07-01 2022-04-26 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
WO2018002760A1 (en) 2016-07-01 2018-01-04 Pfizer Inc. 5,7-dihydro-pyrrolo-pyridine derivatives for treating neurological and neurodegenerative diseases
EP3872078A1 (en) 2016-07-01 2021-09-01 Pfizer Inc. 5,7-dihydro-pyrrolo-pyridine derivatives for use in the treament of depression, anxiety or panic disorders
WO2018163066A1 (en) 2017-03-10 2018-09-13 Pfizer Inc. Novel imidazo[4,5-c]quinoline derivatives as lrrk2 inhibitors
WO2018163030A1 (en) 2017-03-10 2018-09-13 Pfizer Inc. Cyclic substituted imidazo[4,5-c]quinoline derivatives
US10647759B2 (en) 2017-04-20 2020-05-12 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
US11078261B2 (en) 2017-04-20 2021-08-03 Eli Lilly And Company Anti-N3pGlu amyloid beta peptide antibodies and uses thereof
WO2018226992A1 (en) 2017-06-07 2018-12-13 Adrx, Inc. Tau aggregation inhibitors
WO2018234953A1 (en) 2017-06-22 2018-12-27 Pfizer Inc. Dihydro-pyrrolo-pyridine derivatives
EP3431496A1 (en) * 2017-07-19 2019-01-23 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Anti- isoasp7 amyloid beta antibodies and uses thereof
WO2019016213A1 (en) * 2017-07-19 2019-01-24 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. ANTI- ISOASP7 AMYLOID β (Aβ) ANTIBODIES AND USES THEREOF
US11703511B2 (en) 2017-07-19 2023-07-18 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Anti-isoAsp7 amyloid β (Aβ) antibodies and uses thereof
WO2019036725A2 (en) 2017-08-18 2019-02-21 Adrx, Inc. Tau aggregation peptide inhibitors
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase
EP3508499A1 (en) 2018-01-08 2019-07-10 iOmx Therapeutics AG Antibodies targeting, and other modulators of, an immunoglobulin gene associated with resistance against anti-tumour immune responses, and uses thereof
WO2019134991A1 (en) 2018-01-08 2019-07-11 Iomx Therapeutics Ag Antibodies targeting, and other modulators of, an immunoglobulin gene associated with resistance against anti-tumour immune responses, and uses thereof
EP4219464A1 (en) 2018-03-23 2023-08-02 Pfizer Inc. Piperazine azaspiro derivaves
WO2019183636A1 (en) 2018-03-23 2019-09-26 Pfizer Inc. Piperazine azaspiro derivaves
WO2020041541A2 (en) 2018-08-23 2020-02-27 Seattle Genetics, Inc. Anti-tigit antibodies
WO2020072357A1 (en) 2018-10-04 2020-04-09 University Of Rochester Improvement of glymphatic delivery by manipulating plasma osmolarity
WO2020132230A2 (en) 2018-12-20 2020-06-25 Genentech, Inc. Modified antibody fcs and methods of use
US11617798B2 (en) 2019-02-05 2023-04-04 Seagen Inc. Anti-CD228 antibodies and antibody-drug conjugates
WO2020163225A1 (en) 2019-02-05 2020-08-13 Seattle Genetics, Inc. Anti-cd228 antibodies and antibody-drug conjugates
WO2021005009A1 (en) 2019-07-05 2021-01-14 Iomx Therapeutics Ag Antibodies binding igc2 of igsf11 (vsig3) and uses thereof
WO2022008514A1 (en) 2019-07-05 2022-01-13 Iomx Therapeutics Ag Antibodies binding igc2 of igsf11 (vsig3) and uses thereof
WO2021067776A2 (en) 2019-10-04 2021-04-08 Seagen Inc. Anti-pd-l1 antibodies and antibody-drug conjugates
WO2021099347A1 (en) 2019-11-18 2021-05-27 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Antibodies targeting, and other modulators of, the cd276 antigen, and uses thereof
EP3822288A1 (en) 2019-11-18 2021-05-19 Deutsches Krebsforschungszentrum, Stiftung des öffentlichen Rechts Antibodies targeting, and other modulators of, the cd276 antigen, and uses thereof
WO2021195362A1 (en) 2020-03-26 2021-09-30 Seagen Inc. Methods of treating multiple myeloma
WO2022008027A1 (en) 2020-07-06 2022-01-13 Iomx Therapeutics Ag Antibodies binding igv of igsf11 (vsig3) and uses thereof
US11434285B2 (en) 2020-07-23 2022-09-06 Othair Prothena Limited Anti-Abeta antibodies
US11440953B2 (en) 2020-07-23 2022-09-13 Othair Prothena Limited Anti-abeta antibodies
US11434283B2 (en) 2020-07-23 2022-09-06 Othair Prothena Limited Anti-abeta antibodies
US11434284B2 (en) 2020-07-23 2022-09-06 Othair Prothena Limited Anti-Abeta antibodies
WO2022031652A1 (en) 2020-08-04 2022-02-10 Seagen Inc. Anti-cd228 antibodies and antibody-drug conjugates
WO2022096536A1 (en) 2020-11-03 2022-05-12 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Target-cell restricted, costimulatory, bispecific and bivalent anti-cd28 antibodies
WO2022098972A1 (en) 2020-11-08 2022-05-12 Seagen Inc. Combination-therapy antibody drug conjugate with immune cell inhibitor
WO2022162203A1 (en) 2021-01-28 2022-08-04 Vaccinvent Gmbh Method and means for modulating b-cell mediated immune responses
WO2022217026A1 (en) 2021-04-09 2022-10-13 Seagen Inc. Methods of treating cancer with anti-tigit antibodies
WO2023016826A2 (en) 2021-08-05 2023-02-16 Vaccinvent Gmbh Method and means for enhancing therapeutic antibodies
WO2023049694A1 (en) 2021-09-23 2023-03-30 Seagen Inc. Methods of treating multiple myeloma
WO2024068777A1 (en) 2022-09-28 2024-04-04 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Modified ace2 proteins with improved activity against sars-cov-2

Also Published As

Publication number Publication date
NO20066058L (en) 2007-01-22
KR20090046974A (en) 2009-05-11
US8425905B2 (en) 2013-04-23
HK1116500A1 (en) 2008-12-24
WO2006036291A3 (en) 2006-07-20
SG190665A1 (en) 2013-06-28
JP2012197292A (en) 2012-10-18
JP5042828B2 (en) 2012-10-03
US20060057702A1 (en) 2006-03-16
EP1781704A2 (en) 2007-05-09
US20060057701A1 (en) 2006-03-16
US20120269805A1 (en) 2012-10-25
NI200700021A (en) 2008-01-22
NZ552480A (en) 2010-01-29
TWI355389B (en) 2012-01-01
MA28757B1 (en) 2007-07-02
US7927594B2 (en) 2011-04-19
MX2007000998A (en) 2007-07-11
IL180364A0 (en) 2007-06-03
BRPI0513959A (en) 2008-05-20
AU2005290250A1 (en) 2006-04-06
EP2298807A3 (en) 2011-05-18
GEP20115195B (en) 2011-04-11
TNSN07031A1 (en) 2008-06-02
IL180364A (en) 2015-10-29
US20110008834A1 (en) 2011-01-13
US7807165B2 (en) 2010-10-05
AP2007003890A0 (en) 2007-02-28
EA016357B1 (en) 2012-04-30
JP2008511291A (en) 2008-04-17
KR101068289B1 (en) 2011-09-28
AU2010200115B2 (en) 2012-05-17
CA2575663C (en) 2013-04-23
US8268593B2 (en) 2012-09-18
TW200617023A (en) 2006-06-01
EP2298807A2 (en) 2011-03-23
EA200700083A1 (en) 2008-02-28
KR20070040824A (en) 2007-04-17
AU2010200115A1 (en) 2010-01-28
CA2575663A1 (en) 2006-04-06

Similar Documents

Publication Publication Date Title
US8425905B2 (en) Antibodies directed against amyloid-beta peptide and methods using same
US8398978B2 (en) Antibodies directed against amyloid-beta peptide and methods using same
AU2008224600B2 (en) Methods of treating ophthalmic diseases
ZA200700128B (en) Antibodies directed against amyloid-beta peptide and methods using same

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 180364

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2007/00128

Country of ref document: ZA

Ref document number: 200700128

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 552480

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2005290250

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: MX/a/2007/000998

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 200580025690.5

Country of ref document: CN

Ref document number: CR2007-008883

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 9836

Country of ref document: GE

Ref document number: 2575663

Country of ref document: CA

Ref document number: 12007500265

Country of ref document: PH

Ref document number: 2007523885

Country of ref document: JP

Ref document number: 07008780

Country of ref document: CO

Ref document number: DZP2007000081

Country of ref document: DZ

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 786/DELNP/2007

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2005290250

Country of ref document: AU

Date of ref document: 20050801

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005290250

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2005778628

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 1020077004986

Country of ref document: KR

Ref document number: 200700083

Country of ref document: EA

WWP Wipo information: published in national office

Ref document number: 2005778628

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0513959

Country of ref document: BR