US20030082191A1 - Treatment for central nervous system disorders - Google Patents

Treatment for central nervous system disorders Download PDF

Info

Publication number
US20030082191A1
US20030082191A1 US09/942,253 US94225301A US2003082191A1 US 20030082191 A1 US20030082191 A1 US 20030082191A1 US 94225301 A US94225301 A US 94225301A US 2003082191 A1 US2003082191 A1 US 2003082191A1
Authority
US
United States
Prior art keywords
composition
antibody
polypeptide
patient
amyloid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/942,253
Other languages
English (en)
Inventor
Joseph Poduslo
Geoffry Curran
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Mayo Foundation for Medical Education and Research
Original Assignee
Mayo Foundation for Medical Education and Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mayo Foundation for Medical Education and Research filed Critical Mayo Foundation for Medical Education and Research
Priority to US09/942,253 priority Critical patent/US20030082191A1/en
Assigned to MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH reassignment MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CURRAN, GEOFFRY L., PODUSLO, JOSEPH F.
Priority to JP2003524526A priority patent/JP2005504779A/ja
Priority to MXPA04001940A priority patent/MXPA04001940A/es
Priority to PL02373626A priority patent/PL373626A1/xx
Priority to CA002458799A priority patent/CA2458799A1/fr
Priority to EP02761522A priority patent/EP1434801A4/fr
Priority to PCT/US2002/027291 priority patent/WO2003020212A2/fr
Publication of US20030082191A1 publication Critical patent/US20030082191A1/en
Priority to US10/796,522 priority patent/US20040142872A1/en
Priority to US12/550,944 priority patent/US7988969B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/543Lipids, e.g. triglycerides; Polyamines, e.g. spermine or spermidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/642Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a cytokine, e.g. IL2, chemokine, growth factors or interferons being the inactive part of the conjugate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6843Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1018Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against material from animals or humans
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery

Definitions

  • This invention relates to compositions for treating central nervous system (CNS) disorders such as Alzheimer's disease (AD), and more particularly, to compositions that contain a ⁇ amyloid (A ⁇ ) polypeptide linked to a non-A ⁇ polypeptide.
  • CNS central nervous system
  • a ⁇ ⁇ amyloid
  • the invention is based on the discovery that AP-immune complexes are transported across the BBB via a receptor-mediated process at a rate greater than that of antibody alone.
  • transport of antibodies having specific binding affinity for A ⁇ across the BBB, or other polypeptides that have low permeability at the BBB can be enhanced when linked to an A ⁇ polypeptide.
  • Polyamine modified antibodies having specific binding affinity for A ⁇ also have increased permeability at the BBB and can be used for passive immunization and treatment of AD.
  • the invention features a composition that includes an A ⁇ polypeptide and a non-A ⁇ polypeptide, wherein the A ⁇ polypeptide and the non-A ⁇ polypeptide are linked (e.g., covalently).
  • the composition further can include a pharmaceutically acceptable carrier or excipient.
  • the non-A ⁇ polypeptide can be an antibody or a fragment thereof (e.g., a Fab fragment, a single chain Fv antibody fragment, or a F(ab) 2 fragment).
  • the antibody can be labeled with a radioisotope or a contrast agent.
  • the antibody can have specific binding affinity for amyloid.
  • the non-A ⁇ polypeptide also can be an enzyme such as an antioxidant enzyme (e.g., catalase or superoxide dismutase), a cytokine such as an interferon, an interleukin, or a neurotrophic factor, or leptin.
  • an antioxidant enzyme e.g., catalase or superoxide dismutase
  • a cytokine such as an interferon, an interleukin, or a neurotrophic factor, or leptin.
  • the A ⁇ polypeptide can include residues 1-40, 1-42, or 1-43 of SEQ ID NO:1.
  • the invention also features a method of treating a patient diagnosed with AD.
  • the method includes administering to the patient an amount of a composition effective to treat AD, wherein the composition includes an A ⁇ polypeptide and an antibody having specific binding affinity for the A ⁇ polypeptide.
  • the antibody can be a Fab fragment, a single chain Fv antibody fragment, or a F(ab) 2 fragment.
  • the invention features a method of treating a patient diagnosed with AD.
  • the method includes administering to the patient an amount of an antibody effective to treat AD, wherein the antibody is polyamine modified and has specific binding affinity for an AP polypeptide.
  • the invention features a method of diagnosing AD in a patient.
  • the method includes administering a composition to the patient, wherein the composition includes an A ⁇ polypeptide and an antibody having specific binding affinity for amyloid, wherein the antibody is labeled, and detecting the presence or absence of the antibody bound to amyloid in the brain of the patient, wherein the patient is diagnosed with AD based on the presence of labeled amyloid (e.g., labeled amyloid deposits such as ⁇ -amyloid plaques).
  • labeled amyloid e.g., labeled amyloid deposits such as ⁇ -amyloid plaques
  • the detecting step can include diagnostic imaging (e.g., positron emission tomography, gamma-scintigraphy, single photon emission computerized tomography, magnetic resonance imaging, functional magnetic resonance imaging, or magnetoencephalography). Magnetic resonance imaging is particularly useful.
  • diagnostic imaging e.g., positron emission tomography, gamma-scintigraphy, single photon emission computerized tomography, magnetic resonance imaging, functional magnetic resonance imaging, or magnetoencephalography.
  • Magnetic resonance imaging is particularly useful.
  • the antibody can be labeled with a contrast agent (e.g., gadolinium, dysprosium, or iron). Gadolinium is a particularly useful contrast agent.
  • the invention features compositions containing A ⁇ polypeptides that can be used to enhance transport of non-A ⁇ polypeptides across the BBB.
  • BBB permeability of a composition containing A ⁇ bound to a monoclonal antibody was significantly greater than that of the monoclonal antibody alone. Without being bound by a particular mechanism, A ⁇ itself may be responsible for transporting the antibody across the BBB.
  • compositions of the invention can be used in the diagnosis, treatment, and/or prevention of neurodegenerative disorders such as AD, Parkinson's disease, frontotemporal dementias (e.g., Pick's disease), and amyloidotic polyneuropathies, transmissible spongiform encephalopathies (i.e., prion diseases) such as Creutzfeldt-Jakob disease (CJD), Gerstmann-St Hurssler-Scheinker syndrome, and fatal familial insomnia, demyelinating diseases such as multiple sclerosis, and amyotropic lateral sclerosis.
  • neurodegenerative disorders such as AD, Parkinson's disease, frontotemporal dementias (e.g., Pick's disease), and amyloidotic polyneuropathies, transmissible spongiform encephalopathies (i.e., prion diseases) such as Creutzfeldt-Jakob disease (CJD), Gerstmann-St syndromesler-Scheinker syndrome, and fatal familial insomnia,
  • compositions of the invention include a purified A ⁇ polypeptide linked to a purified non-A ⁇ polypeptide.
  • purified refers to a polypeptide that is separated from cellular components (e.g., other polypeptides, lipids, carbohydrates, and nucleic acids) that are naturally associated with the polypeptide.
  • a purified polypeptide is any polypeptide that is removed from its natural environment and is at least 75% pure (e.g., at least about 80, 85, 90, 95, or 99% pure).
  • a purified polypeptide will yield a single major band on a non-reducing polyacrylamide gel.
  • a ⁇ polypeptide refers to 1) the naturally occurring human A ⁇ polypeptide (DAEFRHDSGY EVHHQKLVFF AEDVGSNKGA IIGLMVGGVV IAT, SEQ ID NO: 1) 2) polypeptides having one or more substitutions or insertions in the amino acid sequence of the naturally occurring human A ⁇ polypeptide that retain the ability to cross the BBB, and 3) fragments of 1) and 2) that retain the ability to cross the BBB. Permeability of an A ⁇ polypeptide at the BBB can be assessed according to the methods of Example 1. See also Poduslo et al., Proc. Natl. Acad.
  • human A ⁇ polypeptide ranges in length from 39 to 43 amino acids (residues 1 to 39, 1 to 40, 1 to 41, 1 to 42, or 1 to 43 of SEQ ID NO:1), and is a proteolytic cleavage product of the amyloid precursor protein (APP).
  • amino acid substitutions that can be introduced into human A ⁇ include substitutions at amino acid residues 5, 10, 13, 19, and 20 of SEQ ID NO:1, or combinations thereof.
  • a glycine can be substituted for the arginine at residue 5
  • a phenylalanine can be substituted for the tyrosine at residue 10
  • an arginine can be substituted for the histidine at residue 13.
  • substitutions do not alter the properties of human A ⁇ polypeptide. See Fraser et al., Biochemistry 31:10716-10723 (1992); and Hilbich et al., Eur. J. Biochem. 201:61-69 (1992).
  • An isoleucine, leucine, threonine, serine, alanine, valine, or glycine can be substituted for the phenylalanine residues at positions 19 and 20.
  • Suitable fragments of A ⁇ polypeptides are about 6 to 38 amino acid residues in length (e.g., 10 to 36, 10 to 34, 10 to 30, 12 to 28, 14 to 26, 16 to 24, or 18 to 22 amino acid residues in length) and retain the ability to cross the BBB.
  • an A ⁇ polypeptide may contain residues 1 to 10, 1 to 15, 1 to 20, 5 to 15, 5 to 20, 5 to 25, 10 to 20, 10 to 25, 10 to 30, 15 to 25, 15 to 30, or 15 to 35 of SEQ ID NO:1.
  • an A ⁇ polypeptide may include residues 20 to 30, 20 to 35, 20 to 40, 25 to 35, 25 to 40, 30 to 40, 25 to 42, or 30 to 42 of SEQ ID NO:1.
  • a ⁇ polypeptides can be linked to non-A ⁇ polypeptides via covalent links.
  • Covalent cross-linking techniques are known in the art. See, for example, “Chemistry of Protein Conjugation and Cross-Linking”, Shan S. Wong, CRC Press, Ann Arbor, 1991. Suitable cross-linking reagents do not interfere with the binding of the A ⁇ polypeptide to its cognate receptor and are chosen for appropriate reactivity, specificity, spacer arm length, membrane permeability, cleavability, and solubility characteristics. Similarly, suitable cross-linking reagents do not interfere with binding of a non-A ⁇ polypeptide to its binding partner (e.g., cognate receptor or epitope on a macromolecule). Cross-linking reagents are available commercially from many sources including Pierce Chemical Co., Rockford, Ill.
  • An A ⁇ polypeptide and a non-A ⁇ polypeptide can be covalently cross-linked using, for example, glutaraldehyde, a homobifunctional cross-linker, or a heterobifunctional cross-linker.
  • Glutaraldehyde cross-links polypeptides via their amino moieties.
  • Homobifunctional cross-linkers e.g., a homobifunctional imidoester, a homobifunctional N-hydroxysuccinimidyl (NHS) ester, or a homobifunctional sulfhydryl reactive cross-linker
  • a homobifunctional imidoester e.g., a homobifunctional N-hydroxysuccinimidyl (NHS) ester, or a homobifunctional sulfhydryl reactive cross-linker
  • Homobifunctional NHS esters and imido esters cross-link amine containing polypeptides. In a mild alkaline pH, imido esters react only with primary amines to form imidoamides, and overall charge of the cross-linked polypeptides is not affected.
  • Homobifunctional sulfhydryl reactive cross-linkers include bismaleimidhexane (BMH), 1,5-difluoro-2,4-dinitrobenzene (DFDNB), and 1,4-di-(3′,2′-pyridyldithio) propionamido butane (DPDPB).
  • BMH bismaleimidhexane
  • DFDNB 1,5-difluoro-2,4-dinitrobenzene
  • DPDPB 1,4-di-(3′,2′-pyridyldithio) propionamido butane
  • Heterobifunctional cross-linkers have two or more different reactive moieties (e.g., an amine reactive moiety and a sulfhydryl-reactive moiety) and are cross-linked with one of the polypeptides via the amine or sulfhydryl reactive moiety, then reacted with the other polypeptide via the non-reacted moiety.
  • Multiple heterobifunctional haloacetyl cross-linkers are available, as are pyridyl disulfide cross-linkers.
  • Carbodiimides are a classic example of heterobifunctional cross-linking reagents for coupling carboxyls to amines, which results in an amide bond.
  • an A ⁇ polypeptide can be linked to a non-A ⁇ polypeptide such as an antibody via the specific binding affinity of the antibody for the A ⁇ polypeptide.
  • Purified A ⁇ polypeptide and antibody can be incubated together at 37° C. in an appropriate buffer (e.g., phosphate buffered saline) to form an immune complex.
  • an appropriate buffer e.g., phosphate buffered saline
  • Such an immune complex constitutes a composition of the invention.
  • a ⁇ polypeptides can be linked to any non-A ⁇ polypeptide, and in particular, to any polypeptide that is useful for diagnosing or treating a disorder of the CNS.
  • Non-A ⁇ polypeptides are at least six amino acid residues in length.
  • an A ⁇ polypeptide can be linked to an enzyme such as an antioxidant enzyme, which can protect cells against reactive oxygen species.
  • antioxidant enzymes include catalase (E.C. 1.11.1.6), superoxide dismutase (E.C. 1.15.1.1), glutathione peroxidase (E.C. 1.6.4.2), and glutathione reductase (E.C. 1.11.1.9).
  • a ⁇ polypeptides also can be linked to cytokines such as an interferon (e.g., interferon ⁇ , ⁇ , or ⁇ ), interleukin (IL) (e.g., IL-1a or b, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, or IL-12), neurotrophic factors such as neurotrophins (e.g., nerve growth factor or brain-derived neurotrophic factor), neuropoietic factors such as cholinergic differentiation factor, ciliary neurotrophic factor, oncostatin M, growth-promoting factor, and sweat gland factor, and growth factor peptides such as glial-cell line-derived neurotrophic factor, or a hormone such as leptin.
  • IL interleukin
  • neurotrophic factors such as neurotrophins (e.g., nerve growth factor or brain-derived neurotrophic factor), neuropoietic factors such as cho
  • a ⁇ polypeptides can be linked to an antibody.
  • an A ⁇ polypeptide can be linked to an antibody having specific binding affinity for amyloid deposits of A ⁇ or of a prion protein (PrP).
  • PrP prion protein
  • antibodies includes polyclonal or monoclonal antibodies, humanized or chimeric antibodies, and antibody fragments such as single chain Fv antibody fragments, Fab fragments, and F(ab) 2 fragments. Monoclonal antibodies are particularly useful.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region. Chimeric antibodies can be produced through standard techniques.
  • Antibody fragments can be generated by known techniques. For example, F(ab′) 2 fragments can be produced by pepsin digestion of the antibody molecule, and Fab fragments can be generated by reducing the disulfide bridges of F(ab′) 2 fragments. Alternatively, Fab expression libraries can be constructed. See, for example, Huse et al., Science, 246:1275 (1989). Single chain Fv antibody fragments are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge (e.g., 15 to 18 amino acids), resulting in a single chain polypeptide. See, for example, U.S. Pat. No. 4,946,778.
  • amino acid bridge e.g. 15 to 18 amino acids
  • the A ⁇ polypeptide and/or the non-A ⁇ polypeptide are labeled to facilitate diagnosis of a CNS disorder.
  • Typical labels that are useful include radioisotopes and contrast agents used for imaging procedures in humans.
  • Non-limiting examples of labels include radioisotope such as 123 I(iodine), 18 F (fluorine), 99m Tc (technetium), 111 In (indium), and 67 Ga (gallium), and contrast agents such as gadolinium (Gd), dysprosium, and iron.
  • Radioactive Gd isotopes ( 153 Gd) also are available and suitable for imaging procedures in non-human mammals.
  • Polypeptides can be labeled through standard techniques.
  • polypeptides can be iodinated using chloramine T or 1,3,4,6-tetrachloro-3 ⁇ ,6 ⁇ -diphenylglycouril.
  • fluorination polypeptides are synthesized and fluorine is added during the synthesis by a fluoride ion displacement reaction. See, Muller-Gartner, H., TIB Tech., 16:122-130 (1998) and Saji, H., Crit. Rev. Ther. Drug Carrier Syst., 16(2):209-244 (1999) for a review of synthesis of proteins with such radioisotopes.
  • Polypeptides also can be labeled with a contrast agent through standard techniques.
  • polypeptides can be labeled with Gd by conjugating low molecular Gd chelates such as Gd diethylene triamine pentaacetic acid (GdDTPA) or Gd tetraazacyclododecanetetraacetic (GdDOTA) to the polypeptide.
  • GdDTPA Gd diethylene triamine pentaacetic acid
  • GdDOTA Gd tetraazacyclododecanetetraacetic
  • Antibodies can be labeled with Gd by, for example, conjugating polylysine-Gd chelates to the antibody.
  • antibodies can be labeled with Gd by incubating paramagnetic polymerized liposomes that include Gd chelator lipid with avidin and biotinylated antibody. See, for example, Sipkins et al. Nature Med., 4 623-626 (1998).
  • Isolated nucleic acid molecules encoding A ⁇ and non-A ⁇ polypeptides of the invention can be produced by standard techniques.
  • isolated refers to a sequence corresponding to part or all of a gene encoding an A ⁇ or non-A ⁇ polypeptide, but free of sequences that normally flank one or both sides of the wild-type gene in a mammalian genome.
  • An isolated nucleic acid can be, for example, a recombinant DNA molecule, provided one or both of the nucleic acid sequences normally found immediately flanking that DNA molecule in a naturally-occurring genome is removed or absent.
  • isolated nucleic acids include, without limitation, a DNA that exists as a separate molecule (e.g., a cDNA or genomic DNA fragment produced by PCR or restriction endonuclease treatment) independent of other sequences as well as recombinant DNA that is incorporated into a vector, an autonomously replicating plasmid, a virus (e.g., a retrovirus, adenovirus, or herpes virus), or into the genomic DNA of a prokaryote or eukaryote.
  • an isolated nucleic acid can include a recombinant DNA molecule that is part of a hybrid or fusion nucleic acid.
  • a nucleic acid existing among hundreds to millions of other nucleic acids within, for example, cDNA or genomic libraries, or gel slices containing a genomic DNA restriction digest, is not to be considered an isolated nucleic acid.
  • Isolated nucleic acid molecules are at least about 18 nucleotides in length.
  • the nucleic acid molecule can be about 18 to 20, 20-50, 50-100, or greater than 150 nucleotides in length.
  • Nucleic acid molecules can be DNA or RNA, linear or circular, and in sense or antisense orientation.
  • Specific point changes can be introduced into the nucleic acid sequence encoding the naturally-occurring human A ⁇ polypeptide by, for example, oligonucleotide-directed mutagenesis.
  • a desired change is incorporated into an oligonucleotide, which then is hybridized to the wild-type nucleic acid.
  • the oligonucleotide is extended with a DNA polymerase, creating a heteroduplex that contains a mismatch at the introduced point change, and a single-stranded nick at the 5′ end, which is sealed by a DNA ligase. The mismatch is repaired upon transformation of E.
  • kits for introducing site-directed mutations can be purchased commercially.
  • Muta-Gene® in-vitro mutagenesis kits can be purchased from Bio-Rad Laboratories, Inc. (Hercules, Calif.).
  • PCR Polymerase chain reaction
  • PCR can be used to amplify specific sequences from DNA as well as RNA, including sequences from total genomic DNA or total cellular RNA.
  • Primers are typically 14 to 40 nucleotides in length, but can range from 10 nucleotides to hundreds of nucleotides in length.
  • General PCR techniques are described, for example in PCR Primer: A Laboratory Manual, Ed. by Dieffenbach, C. and Dveksler, G., Cold Spring Harbor Laboratory Press, 1995.
  • Nucleic acids encoding A ⁇ and non-A ⁇ polypeptides also can be produced by chemical synthesis, either as a single nucleic acid molecule or as a series of oligonucleotides. For example, one or more pairs of long oligonucleotides (e.g., >100 nucleotides) can be synthesized that contain the desired sequence, with each pair containing a short segment of complementarity (e.g., about 15 nucleotides) such that a duplex is formed when the oligonucleotide pair is annealed. DNA polymerase is used to extend the oligonucleotides, resulting in a double-stranded nucleic acid molecule per oligonucleotide pair, which then can be ligated into a vector.
  • oligonucleotides e.g., >100 nucleotides
  • DNA polymerase is used to extend the oligonucleotides, resulting in a double-
  • Purified A ⁇ and non-A ⁇ polypeptides of the invention can be obtained from commercial sources, or alternatively, can be obtained by extraction from a natural source (e.g., liver tissue), chemical synthesis, or by recombinant production in a host cell.
  • recombinant polypeptides are produced by introducing an expression vector that contains a nucleic acid encoding the polypeptide of interest operably linked to regulatory elements necessary for expression of the polypeptide into a bacterial or eukaryotic host cell (e.g., insect, yeast, or mammalian cells). Regulatory elements do not typically encode a gene product, but instead affect the expression of the nucleic acid sequence.
  • a strain of Escherichia coli such as BL-21 can be used.
  • Suitable E. coli vectors include the pGEX series of vectors that produce fusion proteins with glutathione S-transferase (GST). Transformed E. coli are typically grown exponentially then stimulated with isopropylthiogalactopyranoside (IPTG) prior to harvesting. Such fusion proteins typically are soluble and can be purified easily from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • a nucleic acid encoding a polypeptide of the invention can be cloned into, for example, a baculoviral vector such as pBlueBac (Invitrogen, San Diego, Calif.) and then used to co-transfect insect cells such as Spodoptera frugiperda (Sf9) cells with wild type DNA from Autographa californica multinuclear polyhedrosis virus (AcMNPV). Recombinant viruses producing polypeptides of the invention can be identified by standard methodology.
  • a nucleic acid encoding a polypeptide of the invention can be introduced into a SV40, retroviral, or vaccinia based viral vector and used to infect suitable host cells.
  • Mammalian cell lines that stably express a polypeptide of interest can be produced using an expression vector that contains a selectable marker and standard techniques.
  • the eukaryotic expression vector pCR3.1 (Invitrogen, San Diego, Calif.) can be used to express polypeptides of interest in, for example, Chinese hamster ovary (CHO) cells, COS-1 cells, human embryonic kidney 293 cells, NIH3T3 cells, BHK21 cells, MDCK cells, and human vascular endothelial cells (HUVEC).
  • a nucleic acid encoding the polypeptide of interest can be ligated into a mammalian expression vector such as pcDNA3 (Invitrogen, San Diego, Calif.) then transcribed and translated in vitro using wheat germ extract or rabbit reticulocyte lysate.
  • pcDNA3 Invitrogen, San Diego, Calif.
  • Polypeptides of interest can be purified by known chromatographic methods including DEAE ion exchange, gel filtration, and hydroxylapatite chromatography
  • Polypeptides can be “engineered” to contain an amino acid sequence that allows the polypeptide to be captured onto an affinity matrix.
  • a tag such as c-myc, hemagglutinin, polyhistidine, or FlagTM tag (Kodak) can be used to aid polypeptide purification.
  • tags can be inserted anywhere within the polypeptide including at either the carboxyl or amino termini.
  • Other fusions that could be useful include enzymes that aid in the detection of the polypeptide, such as alkaline phosphatase.
  • Immunoaffinity chromatography also can be used to purify polypeptides of interest.
  • polyamine modification of an antibody having specific binding affinity for A ⁇ enhances permeability of the modified antibody at the BBB.
  • polyamine-modified monoclonal antibody against A ⁇ has a PS product that is 36 fold higher in the cortex compared to unmodified antibody and may provide a better approach to passive immunization for AD.
  • Antibodies having specific binding affinity for A ⁇ can be modified with polyamines that are either naturally occurring or synthetic. See, for example, U.S. Pat. No. 5,670,477.
  • Useful naturally occurring polyamines include putrescine, spermidine, spermine, 1,3-diaminopropane, norspermidine, syn-homospermidine, thermine, thermospermine, caldopentamine, homocaldopentamine, and canavalmine. Putrescine, spermidine, and spermine are particularly useful.
  • Synthetic polyamines are composed of the empirical formula C x H y N z , and can be cyclic or acyclic, branched or unbranched, hydrocarbyl chains of 3-12 carbon atoms that further include 1-6 NR or N(R) 2 moieties, wherein R is H, (C 1 -C 4 ) alkyl, phenyl, or benzyl. Polyamines can be linked to an antibody using the cross-linking techniques described above.
  • compositions of the invention can be formulated with a pharmaceutically acceptable carrier and administered to a mammal.
  • a composition of the invention can be administered to a non-human animal (e.g., a transgenic mouse model of Alzheimer's disease) or to a human to aid in the diagnosis of a CNS disorder such as Alzheimer's disease or for treating a human patient that has been diagnosed with a CNS disorder.
  • treatment refers to administering a composition of the invention to a patient, regardless of whether the patient responds to the treatment, with the proviso that when the same composition is administered to a population of patients, a statistically significant number of patients within the population exhibit a clinically recognized improvement or stabilization of one or more clinical features of the disorder.
  • compositions of the invention are administered intravenously (i.v.), although other parenteral routes of administration, including subcutaneous, intramuscular, intra-arterial, intranasal, intracarotid, and intrathecal also can be used.
  • parenteral routes of administration including subcutaneous, intramuscular, intra-arterial, intranasal, intracarotid, and intrathecal also can be used.
  • Formulations for parenteral administration may contain pharmaceutically acceptable carriers such as sterile water or saline, polyalkylene glycols such as polyethylene glycol, vegetable oils, hydrogenated naphthalenes, and the like.
  • the dosage of the composition to be administered can be determined by the attending physician taking into account various factors known to modify the action of drugs. These include health status, body weight, sex, diet, time and route of administration, other medications, and any other relevant clinical factors. Typically, the dosage is about 1-3000 ⁇ g/kg body weight (e.g., from about 10-1000 ⁇ g/kg body weight or 50-500 ⁇ g/kg body weight). Therapeutically effective dosages may be determined by either in vitro or in vivo methods.
  • Treatment of a CNS disorder can be assessed by determining if one or more clinical features of the disorder (e.g., cognitive function, memory, behavior, language skills, motor skills, or rigidity of the patient) improve or are stabilized in the patient.
  • one or more clinical features of the disorder e.g., cognitive function, memory, behavior, language skills, motor skills, or rigidity of the patient
  • the composition that is administered to the patient contains at least one polypeptide that is labeled as described above. Presence or absence of the labeled polypeptide (e.g., labeled antibody or labeled A ⁇ polypeptide) is detected in the CNS in vivo (e.g., in the brain of the patient) using, for example, imaging techniques such as positron emission tomography (PET), gamma-scintigraphy, magnetic resonance imaging (MRI), functional magnetic resonance imaging (FMRI), magnetoencephalography (MEG), and single photon emission computerized tomography (SPECT).
  • PET positron emission tomography
  • MRI magnetic resonance imaging
  • FMRI functional magnetic resonance imaging
  • MEG magnetoencephalography
  • SPECT single photon emission computerized tomography
  • MRI is particularly useful as the spatial resolution and signal-to-noise ratio provided by MRI (30 microns) is suitable for detecting amyloid deposits, which can reach up to 200 microns in size.
  • the CNS disorder can be diagnosed based on the presence, for example, of labeled amyloid (e.g., labeled amyloid deposits).
  • a ⁇ Proteins Human A ⁇ 1-42 was synthesized by using f-moc chemistry in a Perkin-Elmer peptide synthesizer in the Mayo Protein Core Facility. The amino acid sequence of human A ⁇ is provided in SEQ ID NO:1. Purity of the peptide was evaluated by peptide sequencing and laser desorption mass spectrometry.
  • B-cell hybridomas were generated following the procedure of St. Groths and Scheidegger ( J. Immunol. Methods 35:1 (1980)) in the Mayo Monoclonal Core Facility. Human A ⁇ 1-42 that was aggregated and fibrilized by incubating at 37° C. for 24 hours was used as antigen. Positive subclones were isotyped and cryopreserved and further characterized by ELISA and immunohistochemistry labeling of AD transgenic mouse brain sections. A non-specific, monoclonal antibody was obtained from ATCC HB96 L227 (anti-human Ia).
  • PS/V p Measurements at the BBB for Radioiodinated Monoclonal IgG (MoIgG): Aliquots of the proteins (MoIgG or A ⁇ ) were labeled with 125 I or 131 I using the chloramine T procedure described by Poduslo et al., Proc. Natl. Acad. Sci. USA 9:5705-5709 (1994). PS/V p measurements were performed as described by Poduslo et al., Neurobiol. Disease, 8:555-567 (2001) and Poduslo et al., Proc. Natl. Acad. Sci. USA 89:2218-2222 (1992).
  • the procedure for quantifying BBB permeability of proteins was adapted from the rat to the mouse and included catheterizing the femoral artery and vein of the mouse instead of the brachial artery and vein as for the rat. Because of the smaller blood volume in the mouse, serial sampling of 20 ⁇ l of blood from the femoral artery was performed and directly TCA precipitated to generate a whole blood washout curve for the intact protein. Briefly, an I.V. bolus injection of phosphate-buffered saline (PBS) containing 125 I-MoIgG (100 ⁇ C) was rapidly injected into the femoral vein in pentobarbital-anesthetized mice.
  • PBS phosphate-buffered saline
  • I-MoIgG 100 ⁇ C
  • V p and PS measurements were calculated as described by Poduslo et al., Neurobiol. Disease, 8:555-567 (2001) and Poduslo et al., Proc. Natl. Acad. Sci. USA 89:2218-2222 (1992). All procedures were performed using humane and ethical protocols approved by the Mayo Clinic Institutional Animal Care and Use Committee, in accordance with the National Institute of Health's Guide for the Care and Use of Laboratory Animals. All efforts were made to minimize both the suffering and the number of animals used.
  • the BBB permeability of a non-specific monoclonal antibody (anti-human Ia; L227; IgG 1 ⁇ ), monoclonal antibody against human A ⁇ 1-42 (PC2; IgG 1 ⁇ ), and the immune complex [(human A ⁇ 42)-L227 or (human A ⁇ 42)-PC2)] at various mole ratios was determined in the normal adult mouse (B6SJL) as described in Example 1 by quantifying the permeability coefficient x surface area (PS) product for each protein after correction for the residual plasma volume (V p ) occupied by the protein in blood vessels in different brain regions following an I.V. bolus injection.
  • PS permeability coefficient x surface area
  • the V p value was determined with a second aliquot of the same protein radioiodinated with a different isotope of iodine ( 125 I vs. 131 I) given 30-60 seconds before the end of the experiment.
  • a different isotope of iodine 125 I vs. 131 I
  • a dual isotope approach allows for the determination of the vascular space in each individual animal. The PS product at the BBB for different radioiodinated proteins is corrected, therefore, for the V p with a second tracer of the same protein.
  • the PS product for the non-specific monoclonal antibody ranged from 0.5-1.1 ⁇ 10 ⁇ 6 ml/g/sec in six different brain regions (Table 1).
  • the PS values for the monoclonal antibody to human A ⁇ 1-42 ranged from 0.6-1.4 ⁇ 10 ⁇ 6 ml/g/sec in the same brain regions and were not significantly different.
  • V p values ranged from 12.8-28.4 ⁇ l/g for L227 and from 11.8-28.0 ⁇ l/g for PC2 and were not significantly different (Table 1).
  • the PS values for both monoclonal antibodies are low and less than that observed for albumin. Both IgG and albumin are considered to be transported at the BBB by passive diffusion or fluid phase endocytosis.
  • insulin has very high PS values in mice (27.7-43.0 ⁇ 10 ⁇ 6 ml/g/sec) and is transported at the BBB by a receptor-mediated transport.
  • Insulin has a PS product at the BBB that is approximately 28.3-49.9 fold greater than that of the monoclonal antibody to human A ⁇ 42 (PC2).
  • the V p values for insulin and the monoclonal antibody to human A ⁇ 42 (PC2) are similar.
  • PS values ranging from 21.5-33.0 ⁇ 10 ⁇ 6 ml/g/sec in six different brain regions were observed for a polyamine modified monoclonal antibody to human A ⁇ (PC2).
  • These PS values for PUT-PC2 were highly significant (P ⁇ 0.0001) and ranged from 22.8-37.9 fold higher than the antibody (PC2) alone.
  • Polyamine modification of the monoclonal antibody may allow for better delivery across the BBB. This approach is not dependant upon circulating A ⁇ levels and may allow for a more dramatic reduction in amyloid burden in the Alzheimer brain following passive immunization.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Organic Chemistry (AREA)
  • Nanotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Neurology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medical Informatics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Zoology (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Biochemistry (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Magnetic Resonance Imaging Apparatus (AREA)
US09/942,253 2001-08-29 2001-08-29 Treatment for central nervous system disorders Abandoned US20030082191A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US09/942,253 US20030082191A1 (en) 2001-08-29 2001-08-29 Treatment for central nervous system disorders
PCT/US2002/027291 WO2003020212A2 (fr) 2001-08-29 2002-08-27 Traitement pour des troubles du systeme nerveux central
CA002458799A CA2458799A1 (fr) 2001-08-29 2002-08-27 Traitement pour des troubles du systeme nerveux central
MXPA04001940A MXPA04001940A (es) 2001-08-29 2002-08-27 Tratamiento para desordenes del sistema nervioso central.
PL02373626A PL373626A1 (en) 2001-08-29 2002-08-27 Treatment for central nervous system disorders
JP2003524526A JP2005504779A (ja) 2001-08-29 2002-08-27 中枢神経系障害の治療法
EP02761522A EP1434801A4 (fr) 2001-08-29 2002-08-27 Traitement pour des troubles du systeme nerveux central
US10/796,522 US20040142872A1 (en) 2001-08-29 2004-03-09 Treatment for central nervous system disorders
US12/550,944 US7988969B2 (en) 2001-08-29 2009-08-31 Treatment for central nervous system disorders

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US09/942,253 US20030082191A1 (en) 2001-08-29 2001-08-29 Treatment for central nervous system disorders

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/796,522 Continuation US20040142872A1 (en) 2001-08-29 2004-03-09 Treatment for central nervous system disorders

Publications (1)

Publication Number Publication Date
US20030082191A1 true US20030082191A1 (en) 2003-05-01

Family

ID=25477805

Family Applications (3)

Application Number Title Priority Date Filing Date
US09/942,253 Abandoned US20030082191A1 (en) 2001-08-29 2001-08-29 Treatment for central nervous system disorders
US10/796,522 Abandoned US20040142872A1 (en) 2001-08-29 2004-03-09 Treatment for central nervous system disorders
US12/550,944 Expired - Fee Related US7988969B2 (en) 2001-08-29 2009-08-31 Treatment for central nervous system disorders

Family Applications After (2)

Application Number Title Priority Date Filing Date
US10/796,522 Abandoned US20040142872A1 (en) 2001-08-29 2004-03-09 Treatment for central nervous system disorders
US12/550,944 Expired - Fee Related US7988969B2 (en) 2001-08-29 2009-08-31 Treatment for central nervous system disorders

Country Status (7)

Country Link
US (3) US20030082191A1 (fr)
EP (1) EP1434801A4 (fr)
JP (1) JP2005504779A (fr)
CA (1) CA2458799A1 (fr)
MX (1) MXPA04001940A (fr)
PL (1) PL373626A1 (fr)
WO (1) WO2003020212A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070032501A1 (en) * 2005-08-02 2007-02-08 Augeri David J Aryl pyridines and methods of their use
WO2008084402A2 (fr) 2007-01-11 2008-07-17 Philipps-Universitaet Marburg Diagnostic et traitement de la maladie d'alzheimer, et d'autres maladies neurodémentielles
US20080292639A1 (en) * 2005-01-24 2008-11-27 Amgen Inc. Humanized Anti-Amyloid Antibody
WO2009085200A2 (fr) 2007-12-21 2009-07-09 Amgen Inc. Anticorps anti-amyloïde et utilisations de ceux-ci
US20090324492A1 (en) * 2001-08-29 2009-12-31 Mayo Foundation For Medical Education And Research Treatment for Central Nervous System Disorders

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US6913745B1 (en) * 1997-12-02 2005-07-05 Neuralab Limited Passive immunization of Alzheimer's disease
US6787523B1 (en) 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20080050367A1 (en) 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
US7344702B2 (en) 2004-02-13 2008-03-18 Bristol-Myers Squibb Pharma Company Contrast agents for myocardial perfusion imaging
MY139983A (en) 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
DE10303974A1 (de) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid-β(1-42)-Oligomere, Verfahren zu deren Herstellung und deren Verwendung
PE20050627A1 (es) 2003-05-30 2005-08-10 Wyeth Corp Anticuerpos humanizados que reconocen el peptido beta amiloideo
GB0404187D0 (en) * 2004-02-25 2004-03-31 Biotransformations Ltd Binding agents
US8916165B2 (en) 2004-12-15 2014-12-23 Janssen Alzheimer Immunotherapy Humanized Aβ antibodies for use in improving cognition
PT2289909E (pt) 2005-11-30 2015-02-10 Abbvie Inc Método de rastreio, processo de purificação de globulómeros a-beta não difundíveis, anticorpos selectivos contra os referidos globulómeros a-beta não difundíveis e processo para o fabrico dos referidos anticorpos
KR20140087058A (ko) 2005-11-30 2014-07-08 애브비 인코포레이티드 아밀로이드 베타 단백질에 대한 모노클로날 항체 및 이의 용도
BRPI0619748B8 (pt) * 2005-12-12 2021-05-25 Ac Immune Sa anticorpo monoclonal, polinucleotídeo, composição, mistura, uso de um anticorpo monoclonal ou uma parte funcional do mesmo, método para preparar uma composição farmacêutica, linhagem celular de hibridoma isolado, método de diagnóstico de uma doença ou condição associada a amilóide em um paciente, método para diagnosticar uma predisposição para uma doença ou condição, método para monitorar doença residual mínima, método para prever a responsividade de um paciente e kits de teste
WO2009017467A1 (fr) 2007-07-27 2009-02-05 Elan Pharma International Limited Traitement de maladies amyloïdogéniques
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
CA2657681C (fr) * 2006-07-14 2019-03-19 Ac Immune S.A. Anticorps humanises contre la proteine beta-amyloide
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
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
US8048420B2 (en) * 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
US8613923B2 (en) * 2007-06-12 2013-12-24 Ac Immune S.A. Monoclonal antibody
CA2697032C (fr) 2007-08-22 2021-09-14 The Regents Of The University Of California Polypeptides de liaison activables et procedes d'identification et utilisation de ceux-ci
SG178809A1 (en) * 2007-10-05 2012-03-29 Genentech Inc Use of anti-amyloid beta antibody in ocular diseases
PT2238166E (pt) 2007-10-05 2014-02-11 Genentech Inc Utilização do anticorpo anti-amilóide beta em doenças oculares
JO3076B1 (ar) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap نظم العلاج المناعي المعتمد على حالة apoe
CA2967254C (fr) * 2008-02-29 2019-03-26 Lantheus Medical Imaging, Inc. Agents de contraste destine aux applications comprenant l'imagerie du cancer
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
EP2385955B1 (fr) 2009-01-12 2020-08-12 CytomX Therapeutics, Inc. Compositions d anticorps modifiées et leurs procédés de production et d utilisation
WO2010096838A2 (fr) 2009-02-23 2010-08-26 Cytomx Therapeutics, Llc Proprotéines et leurs procédés d'utilisation
SG10201500929XA (en) 2010-02-08 2015-04-29 Lantheus Medical Imaging Inc Methods and apparatus for synthesizing imaging agents, and intermediates thereof
ES2684475T3 (es) 2010-04-15 2018-10-03 Abbvie Inc. Proteínas que se unen a beta amiloide
JP6081356B2 (ja) 2010-07-30 2017-02-15 エーシー イミューン エス.エー. 安全で機能的なヒト化抗βアミロイド抗体
MX358739B (es) 2010-08-14 2018-09-03 Abbvie Inc Star Proteinas de union a amiloide beta.
AU2013203000B9 (en) 2012-08-10 2017-02-02 Lantheus Medical Imaging, Inc. Compositions, methods, and systems for the synthesis and use of imaging agents
WO2019060298A1 (fr) 2017-09-19 2019-03-28 Neuroenhancement Lab, LLC Procédé et appareil de neuro-activation
US11717686B2 (en) 2017-12-04 2023-08-08 Neuroenhancement Lab, LLC Method and apparatus for neuroenhancement to facilitate learning and performance
US11318277B2 (en) 2017-12-31 2022-05-03 Neuroenhancement Lab, LLC Method and apparatus for neuroenhancement to enhance emotional response
US11364361B2 (en) 2018-04-20 2022-06-21 Neuroenhancement Lab, LLC System and method for inducing sleep by transplanting mental states
CA3112564A1 (fr) 2018-09-14 2020-03-19 Neuroenhancement Lab, LLC Systeme et procede d'amelioration du sommeil
US11786694B2 (en) 2019-05-24 2023-10-17 NeuroLight, Inc. Device, method, and app for facilitating sleep
WO2022169211A1 (fr) * 2021-02-03 2022-08-11 한양대학교 산학협력단 Composition contenant le peptide bêta-amyloïde pour la prévention ou le traitement d'une douleur chronique

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5004697A (en) * 1987-08-17 1991-04-02 Univ. Of Ca Cationized antibodies for delivery through the blood-brain barrier
US5231000A (en) * 1987-10-08 1993-07-27 The Mclean Hospital Antibodies to A4 amyloid peptide
US5262332A (en) * 1989-04-05 1993-11-16 Brigham And Women's Hospital Diagnostic method for Alzheimer's disease: examination of non-neural tissue
US5260308A (en) * 1991-11-06 1993-11-09 Mayo Foundation For Medical Education And Research Method to increase permeability of the blood-nerve/brain barriers to proteins
AU683703B2 (en) * 1993-10-20 1997-11-20 Duke University Method of binding material to the beta-amyloid peptide
US5604198A (en) * 1994-05-12 1997-02-18 Poduslo; Joseph F. Method to enhance permeability of the blood/brain blood/nerve barriers to therapeutic agents
EP0815134B1 (fr) * 1995-03-14 2002-06-05 Praecis Pharmaceuticals Incorporated Modulateurs de l'agregation de substances amyloides
US5670477A (en) * 1995-04-20 1997-09-23 Joseph F. Poduslo Method to enhance permeability of the blood/brain blood/nerve bariers to therapeutic agents
WO1996040248A1 (fr) * 1995-06-07 1996-12-19 Eukarion, Inc. Liberation transvasculaire et intracellulaire de proteines lipidisees
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
JP3422740B2 (ja) * 2000-01-26 2003-06-30 旭ファイバーグラス株式会社 サンドイッチ構造体用ハニカム芯材の製造方法
US7371365B2 (en) * 2000-04-04 2008-05-13 Mayo Foundation For Medical Education And Research Methods for detecting parenchymal plaques in vivo
WO2001074374A1 (fr) * 2000-04-04 2001-10-11 Mayo Foundation For Medical Education And Research Methodes de detection de plaques in vivo
DK1346041T3 (da) * 2000-11-27 2007-07-02 Praecis Pharm Inc Terapeutiske midler og fremgangsmåder til anvendelse deraf til behandling af en amyloidogen sygdom
US20030082191A1 (en) * 2001-08-29 2003-05-01 Poduslo Joseph F. Treatment for central nervous system disorders

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090324492A1 (en) * 2001-08-29 2009-12-31 Mayo Foundation For Medical Education And Research Treatment for Central Nervous System Disorders
US7988969B2 (en) 2001-08-29 2011-08-02 Mayo Foundation For Medical Education And Research Treatment for central nervous system disorders
US20080292639A1 (en) * 2005-01-24 2008-11-27 Amgen Inc. Humanized Anti-Amyloid Antibody
US7906625B2 (en) 2005-01-24 2011-03-15 Amgen Inc. Humanized anti-amyloid antibody
US20070032501A1 (en) * 2005-08-02 2007-02-08 Augeri David J Aryl pyridines and methods of their use
US8153804B2 (en) 2005-08-02 2012-04-10 Lexicon Pharmaceuticals, Inc. Aryl pyridines and methods of their use
WO2008084402A2 (fr) 2007-01-11 2008-07-17 Philipps-Universitaet Marburg Diagnostic et traitement de la maladie d'alzheimer, et d'autres maladies neurodémentielles
WO2009085200A2 (fr) 2007-12-21 2009-07-09 Amgen Inc. Anticorps anti-amyloïde et utilisations de ceux-ci
EP2261254A2 (fr) 2007-12-21 2010-12-15 Amgen, Inc Anticorps anti-amyloïdes et leurs utilisations
US8414893B2 (en) 2007-12-21 2013-04-09 Amgen Inc. Anti-amyloid antibodies and uses thereof

Also Published As

Publication number Publication date
WO2003020212A2 (fr) 2003-03-13
EP1434801A4 (fr) 2007-12-19
US7988969B2 (en) 2011-08-02
PL373626A1 (en) 2005-09-05
WO2003020212A3 (fr) 2004-02-19
US20090324492A1 (en) 2009-12-31
EP1434801A2 (fr) 2004-07-07
JP2005504779A (ja) 2005-02-17
US20040142872A1 (en) 2004-07-22
MXPA04001940A (es) 2004-07-23
CA2458799A1 (fr) 2003-03-13

Similar Documents

Publication Publication Date Title
US7988969B2 (en) Treatment for central nervous system disorders
US20220064333A1 (en) Humanised Anti Kallikrein-2 Antibody
US9447180B2 (en) Methods of treating a tauopathy
EP2450056B1 (fr) Prévention et traitement de la maladie synucléinopathique et amyloidogénique
ES2699801T3 (es) Moléculas de unión anti-alfa sinucleína
CN111471106A (zh) 人源抗转甲状腺素蛋白抗体、多核苷酸、载体及其应用
JP2017528449A (ja) アミロイドーシスのための標的化免疫療法
CN111569063A (zh) 治疗tau病变的方法
WO2008122441A9 (fr) Anticorps anti-amyloïdes et leur utilisation pour le diagnostic et le traitement de maladies amyloïdes
WO2008030251A1 (fr) Anticorps deglycosylés anti-bêta amyloïde
US20100048488A1 (en) Immunomodulatory peptides
JP2022519586A (ja) 糖ポリシアル酸化治療用タンパク質を使用する方法
US20200385461A1 (en) Fusion constructs and uses thereof
CN108136013B (zh) 抗体多肽及其用途
AU2002326780A1 (en) Treatment for central nervous system disorders
JP2018531045A6 (ja) 抗体ポリペプチド及びそれらの使用
JP2019089763A (ja) タウペプチド、抗タウ抗体、およびそれらの使用方法
EP4153634A1 (fr) Protéines de fusion d'antigène de cellules souches anti-prostatiques génétiquement modifiées et leurs utilisations
JP2022516574A (ja) 新規ペプチドとその使用
IL302111A (en) A radioactive complex of an antibody against HER2, and a radioactive drug

Legal Events

Date Code Title Description
AS Assignment

Owner name: MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PODUSLO, JOSEPH F.;CURRAN, GEOFFRY L.;REEL/FRAME:012790/0537

Effective date: 20011107

STCB Information on status: application discontinuation

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