WO2007021255A1 - Antibodies to alpha-synuclein - Google Patents

Antibodies to alpha-synuclein Download PDF

Info

Publication number
WO2007021255A1
WO2007021255A1 PCT/US2005/028361 US2005028361W WO2007021255A1 WO 2007021255 A1 WO2007021255 A1 WO 2007021255A1 US 2005028361 W US2005028361 W US 2005028361W WO 2007021255 A1 WO2007021255 A1 WO 2007021255A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
monoclonal antibody
synuclein
alpha synuclein
epitope
Prior art date
Application number
PCT/US2005/028361
Other languages
French (fr)
Inventor
Tamie Chilcote
Robin Barbour
Original Assignee
Elan Pharmaceuticals, Inc.
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 Elan Pharmaceuticals, Inc. filed Critical Elan Pharmaceuticals, Inc.
Priority to PCT/US2005/028361 priority Critical patent/WO2007021255A1/en
Publication of WO2007021255A1 publication Critical patent/WO2007021255A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells

Definitions

  • Alpha-synuclein (alphaSN) brain pathology is a conspicuous feature of several neurodegenerative diseases, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), the Lewy body variant of Alzheimer's disease (LBVAD), multiple systems atrophy (MSA), and neurodegeneration with brain iron accumulation type-1 (NBIA-I).
  • Parkinson's disease PD
  • dementia with Lewy bodies LLB
  • LVAD Lewy body variant of Alzheimer's disease
  • MSA multiple systems atrophy
  • NBIA-I neurodegeneration with brain iron accumulation type-1
  • synucleinopathies proteinaceous insoluble inclusions in the neurons and the glia which are composed primarily of alphaSN.
  • Lewy bodies and Lewy neurites are intraneuronal inclusions which primarily contain of alphaSN.
  • Lewy bodies and Lewy neurites are the neuropathological hallmarks Parkinson's disease (PD).
  • PD and other synucleinopathic diseases have been collectively referred to as Lewy body disease (LBD).
  • LBD is characterized by degeneration of the dopaminergic system, motor alterations, cognitive impairment, and formation of Lewy bodies (LBs).
  • LBs Lewy body disease
  • LBDs include diffuse Lewy body disease (DLBD), Lewy body variant of Alzheimer's disease (LBVAD), combined PD and Alzheimer's disease (AD), and multiple systems atrophy.
  • DLBD diffuse Lewy body disease
  • LBD Lewy body variant of Alzheimer's disease
  • AD Alzheimer's disease
  • DLB Dementia with Lewy bodies
  • Alpha-SN is part of a large family of proteins including beta- and gamma- synuclein and synoretin. Alpha-SN is expressed in the normal state associated with synapses and is believed to play a role in neural plasticity, learning and memory. Mutations in human (h) alpha-SN that enhance the aggregation of alpha-SN have been identified (Ala30Pro and Ala53Thr) and are associated with rare forms of autosomal dominant forms of PD. The mechanism by which these mutations increase the propensity of alpha-SN to aggregate are unknown.
  • the invention provides a monoclonal antibody that competes with a monoclonal antibody shown in Table 1 for specific binding to alpha synuclein.
  • the monoclonal antibody specifically binds alpha synuclein compared to beta synuclein.
  • the monoclonal antibody specifically binds human alpha synuclein compared to non-human alpha synuclein.
  • the monoclonal specifically binds to an epitope bound by a monoclonal antibody shown in Table 1.
  • Some antibodies specifically binds to an epitope within residues 109-120, or to an epitope on the C-terminus of synuclein, or to an epitope comprising residues 139-140.
  • Preferred antibodies include 9E4 or an antibody that competes with 9E4 for specific binding to human alpha synuclein.
  • Other preferred antibodies include 11 A5 or an antibody that competes with 11 A5 for specific binding to phosphorylated human alpha synuclein.
  • Other preferred antibodies include 6H7 or an antibody that competes with 6H7 for specific binding to alpha synuclein.
  • Other preferred antibodies include 4Bl or 8A5 or an antibody that competes with 4Bl or 8A5 for specific binding to alpha synuclein.
  • Other preferred antibodies include 7G4, 6A8, 5C12, 6A12, 9G5, and 1H7, and antibodies that competes therewith for specific binding to alpha synuclein.
  • Some antibodies specifically bind to mouse alpha synuclein without specifically binding to human alpha synuclein. Some antibodies specifically bind to mouse and human alpha synuclein. Some antibodies specifically bind to alpha synuclein phosphorylated at residue 129 without specifically binding to nonphosphorylated alpha synuclein. Some antibodies are end-specific for the N-terminus or C-terminus of alpha synuclein. Some antibodies specifically bind to alpha synuclein without specifically binding to beta or gamma synuclein.
  • Some antibodies specifically bind to an epitope within a segment of amino acids selected of the group consisting of amino acids 109-120, 43-51 and 58-65, 91-96, 118-126, and 91-99.
  • Exemplary monoclonal antibodies are shown in Table 1 and include 7G4, 6A8, 5C12, 6A12, 8A5, 4Bl, 6H7, 3A12, 12Cl, 9A6, 9G5, 9E4, 23E8, 10G5, 3C12, 11A5 andlH7.
  • Some antibodies are monoclonal antibodies that specifically binds to phosphorylated alpha synuclein. Some antibodies are monoclonal antibodies that specifically bind to nitrated synuclein. Some antibodies are monoclonal antibodies capable of capturing soluble synuclein from a fluid sample, preferably alpha-synuclein and more preferably human alpha synuclein..
  • Some antibodies are monoclonal antibodies that specifically bind to an epitope within residues selected from the group consisting of the N-terminus, 118-126, 91-99 and 40-55. Some monoclonal antibodies competes with the monoclonal antibody selected from the group consisting of 6H7, 3QA12, 12C6, 12Cl, 9A6, 9G5, 9E4, 1H7 and 23E8. Exemplary antibodies include monoclonals 6H7, 3QA12, 12C6. 12Cl, 9A6, 9G5, 9E4, 1H7 and 23E8.
  • the invention further provides a pair of monoclonal antibodies, each of the monoclonal antibodies specifically binding to a different epitope within synuclein, wherein the monoclonal antibodies are capable of detecting soluble synuclein when used together in an ELISA assay.
  • one monoclonal antibody is immobilized to a solid phase.
  • the use of the monoclonal antibodies in the ELISA assay is sequential.
  • one monoclonal antibody is a capture antibody and the other monoclonal antibody is a reporter antibody.
  • the capture antibody specifically binds an epitope within residues selected from the group consisting of N-terminus, 40-55, 91-99 and 118-126, and the reporter antibody specifically binds an epitope within residues 109-120.
  • the pair is selected from the group of an antibody that competes with 6H7 and an antibody that competes with 5C12 or 12Cl, an antibody that competes with 3A12 and an antibody that competes with 5C12 or 6H7, an antibody that competes with 12Cl and an antibody that competes with 5Cl 2 or 6H7, an antibody that competes with 9A6 and an antibody that competes with 5Cl 2, 6H7 or 12Cl, an antibody that competes with 9G5 and an antibody that competes with 5C12, 6H7 or 12Cl, an antibody that competes with 9E4 and an antibody that competes with 5C12, 6H7 or 12Cl,an antibody that competes with 1H7 and an antibody that competes with 5C12, 6H7 or 12Cl, and an antibody that competes with 10G5 and an antibody that competes with 5C12, 6H7 or 12Cl.
  • the pair is selected from the group of 6H7 and 5C12 or 12Cl; 3A12 and 5C12 or 6H7; 12Cl and 5C12 or 6H7; 9A6 and 5C12, 6H7 or 12Cl; 9G5 and 5C12, 6H7 or 12Cl; 9E4 and 5C12, 6H7 or 12C1;1H7 and 5C12, 6H7 or 12Cl; and 10G5 and 5C12, 6H7 or 12Cl.
  • the invention further provides a hybridoma producing a monoclonal antibody shown in Table 1.
  • the invention further provides a humanized or chimeric version of a monoclonal antibody shown in Table 1.
  • the invention further provides a method of humanizing a monoclonal antibody shown in Table 1, comprising: determining the amino acid sequence of CDR regions of the monoclonal antibody; selecting an acceptor antibody; and producing a humanized antibody comprising the CDRs from the monoclonal antibody and variable region frameworks from the acceptor antibody.
  • the invention further provides a method of producing a chimeric form of an antibody shown in Table 1, comprising: determining the amino acid sequence of the light and heavy chain variable regions of the monoclonal antibody; selecting heavy and light chain constant region; producing a chimeric antibody comprising a light chain comprising the light chain variable region fused to the light chain constant region, and a heavy chain comprising the heavy chain variable region fused to the heavy chain constant region.
  • the invention further provides a method for detecting alpha synuclein in a fluid sample, comprising capturing the alpha synuclein using a capture antibody and detecting the alpha synuclein using a reporter antibody, wherein the capture and reporter antibodies bind to different epitopes on alpha synuclein.
  • the fluid sample contains 0.1-1.0 M guanidine.
  • the fluid sample contains 0.5 M guanidine.
  • the reporter antibody or the capture antibody is end-specific for the C-terminal end of SNl- 119 or SN1-122.
  • the capturing step is performed twice with a first reporter antibody specifically binding to an epitope within SNl-119 and a second reporter antibody specifically binding to an antibody within SN120-140 and determining a ratio of captured alpha synuclein between the two steps, a higher ratio of synuclein captured by first reporter relative to second report being indicative of pathogenicity.
  • the reporter antibody in the first capturing step specifically binds to an epitope within SN 109-120
  • the reporter antibody in the second capturing step specifically binds to an epitope within SN 120-126
  • the capture antibody in both capturing steps specifically binds to an epitope within SN 91-99.
  • the capture antibody is immobilized to a solid phase, and the reporter antibody in solution, and the detecting step comprises detecting alpha synuclein from presence of the reporter antibody linked to the solid phase via binding of the reporter antibody to alpha synuclein, which is in turn bound to the capture antibody.
  • Some methods involve a further step of comparing a signal from the reporter antibody linked to the solid phase from analyzing the sample with a signal from the reporter antibody linked to the solid phase from analyzing a control sample containing a known amount of alpha synuclein to determine the amount of alpha synuclein in the sample, hi some methods, a signal from the reporter antibody linked to the solid phase from analyzing the sample to a calibration curve of signal is compared with an amount of alpha synuclein to determine the amount of alpha synuclein in the sample. In some methods, a signal from the reporter antibody is proportional to the amount of synuclein in the sample. Some methods further comprise contacting the reporter antibody with a labeled antibody to generate a signal indicating presence of the reporter antibody.
  • the capture antibody specifically binds to an epitope within amino acids 91-99 of human alpha synuclein and the reporter antibody binds to an epitope within amino acids 109-120 of alpha synuclein.
  • the capture antibody is 1H7 and the reporter antibody is 5C12.
  • the sample is a sample from a human.
  • the capture antibody is 1H7 and the reporter antibody is 9E4.
  • the sample from a mouse is a body fluid.
  • the sample is cerebrospinal fluid (CSF) of a human, hi some methods, the sample is a brain homogenate of a human or transgenic animal.
  • CSF cerebrospinal fluid
  • the sample is a medium used to culture cells, hi some methods, the cells express recombinant alpha synuclein. hi some methods, the sample contains a fragment of full-length alpha synuclein and the detecting step detects the fragment.
  • the invention further provides a method of monitoring processing of alpha-synuclein to a fragment, comprising culturing a cell expressing alpha synuclein and processing the alpha synuclein to a fragment that is secreted to the cell media; and detecting the fragment in the cell media.
  • the cell is a PeakS or SY5Y cell transfected with alpha synuclein.
  • the cell is a cortical cell.
  • the invention further provides a method of screening an agent for activity in inhibiting processing or secretion of alpha synuclein, comprising contacting a cell expressing alpha synuclein in culture medium with an agent; detecting alpha synuclein or a fragment thereof in the medium; and comparing the amount of alpha synuclein or the fragment in the medium with an among of alpha synuclein or fragment in medium from a control cell not contacted with the agent, wherein a reduction in the amount of alpha synuclein or the fragment indicates the agent inhibits processing or secretion of the alpha synuclein.
  • the invention further provides a method for detecting soluble alpha synuclein in a fluid sample, comprising: capturing the soluble fragment from the sample using a first binding substance under conditions in which the first binding substance specifically binds to a first epitope and detecting capture of the soluble alpha synuclein using a second binding substance which binds to an epitope on a second region of the soluble alpha synuclein different from the first epitope.
  • the invention further provides a method for detecting a soluble fragment of alpha synuclein in a fluid sample in the presence of full length alpha synuclein the method comprising: capturing the soluble fragment from the sample using a first binding substance, and detecting the soluble fragment using a second binding substance, wherein both binding substances specifically bind to the fragment and at least one specifically binds to the fragment without binding to full length alpha synuclein.
  • Figs. IA and B show quantitative detection of alpha synuclein using an ELISA assay with two pairs of antibodies 1H7/5C12 and 1H7/9E4 respectively.
  • Fig. 2 shows a calibration curve for detection of alpha synuclein using 1H7/5C12 as capture and reporter antibodies.
  • Fig. 3 shows a calibration curve for detection of alpha synuclein using 1H7 and 9E4 as capture and reporter antibodies.
  • Fig. 4 shows a calibration curve for detection of phosphorylated alpha synuclein using 11A5 and 5C12 as capture and reporter antibodies.
  • an antibody “specifically binds" to a target refers to a binding reaction which is determinative of the presence of the antibody in the presence of a heterogeneous population of other biologies.
  • a specified molecule binds preferentially to a particular target and does not bind in a significant amount to other biologies present in the sample.
  • Specific binding of an antibody to a target under such conditions requires the antibody be selected for its specificity to the target.
  • a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein.
  • antibody or "immunoglobulin” is used to include intact antibodies and binding fragments thereof. Typically, fragments compete with the intact antibody from which they were derived for specific binding to an antigen fragment including separate heavy chains, light chains Fab, Fab 1 F(ab')2, Fabc, and Fv. Fragments are produced by recombinant DNA techniques, or by enzymatic or chemical separation of intact immunoglobulins.
  • antibody also includes one or more immunoglobulin chains that are chemically conjugated to, or expressed as, fusion proteins with other proteins.
  • antibody also includes bispecific antibody. A bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79:315-321 (1990); Kostelny et al., J. Immunol. 148, 1547-1553 (1992).
  • Antibodies of the invention are typically substantially pure from undesired contaminant. This means that an agent is typically at least about 50% w/w (weight/weight) purity, as well as being substantially free from interfering proteins and contaminants. Sometimes the antibodies are at least about 80% w/w and, more preferably at least 90 or about 95% w/w purity. However, using conventional protein purification techniques, homogeneous peptides of at least 99% w/w can be obtained.
  • An "antigen” is an entity to which an antibody specifically binds.
  • epitopes refers to a site on an antigen to which B and/or T cells respond.
  • B-cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance.
  • Antibodies that recognize the same epitope can be identified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen.
  • Compet means competition between antibodies is determined by an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen, such as alpha-SN.
  • a common antigen such as alpha-SN.
  • Numerous types of competitive binding assays are known, for example: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition assay (see Stahli et at, Methods in Enzymology 9:242-253 (1983)); solid phase direct biotin-avidin EIA (see Kirkland et ah, J. Immunol.
  • solid phase direct labeled assay solid phase direct labeled sandwich assay (see Harlow and Lane, Antibodies, A Laboratory Manual, Cold Spring Harbor Press (1988)); solid phase direct label RIA using 1-125 label ⁇ see Morel et al, Molec. Immunol. 25(1):7-15 (1988)); solid phase direct biotin-avidin EIA (Cheung et al, Virology 176:546-552 (1990)); and direct labeled RIA (Moldenhauer et al, Scand. J. Immunol 32:77-82 (1990)).
  • such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabelled test immunoglobulin and a labeled reference immunoglobulin.
  • Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test immunoglobulin.
  • the test immunoglobulin is present in excess.
  • Antibodies identified by competition assay include antibodies binding to the same epitope as the reference antibody and antibodies binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference antibody for steric hindrance to occur.
  • a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to a common antigen by at least 50 or 75%.
  • conditioned culture medium and “culture medium” refer to the aqueous extracellular fluid which surrounds cells grown in tissue culture (in vitro) and which contains, among other constituents, proteins and peptides secreted by the cells.
  • body fluid refers to those fluids of a mammalian host which is suspected contain measurable amounts of alpha synuclein or fragments thereof, specifically including blood, cerebrospinal fluid (CSF), • urine, and peritoneal fluid.
  • CSF cerebrospinal fluid
  • blood refers to whole blood, as well as blood plasma and serum.
  • a "pharmacological" activity means that an agent at least exhibits an activity in a screening system that indicates that the agent is or may be useful in the prophylaxis or treatment of a disease.
  • the screening system can be in vitro, cellular, animal or human. Agents can be described as having pharmacological activity notwithstanding that further testing may be required to establish actual prophylactic or therapeutic utility in treatment of a disease.
  • An end-specific antibody means an antibody whose epitope includes a terminal residue of an antigen, and the antibody specifically binds to the antigen only when that residue has a free end.
  • an antibody that binds to the N-terminus of alpha synuclein when the alpha synuclein is not linked to any other protein but which does not specifically bind when the N-terminus of alpha synuclein is fused to another protein is an end-specific antibody.
  • a synucleinopathic disease means a disease characterized by Lewy bodies, Lewy neurites or other deposits of alpha synuclein.
  • An immunogenic fragment of alpha synuclein is one capable of inducing a humoral (antibody mediated) and/or a cellular (mediated by antigen-specific T cells or their secretion products) when administered to a subject.
  • compositions or methods "comprising" one or more recited elements may include other elements not specifically recited.
  • a composition that comprises alpha-SN peptide encompasses both an isolated alpha-SN peptide and alpha-SN peptide as a component of a larger polypeptide sequence.
  • the invention provides several exemplary monoclonal antibodies to different epitopes of alpha synuclein as shown in Table 1. Hybridomas producing the respective antibodies are also provided. The first column of the table shows the name of an antibody. The same name is used to refer to an antibody and the hybridoma producing it. The second column shows the antigen used to generate the antibody. The third column indicates the epitope bound by the antibody. The fourth column indicates the isotype. The fifth column indicates applications of the antibody (immunofluorescence, Western blot, immunoprecipitation, histology, and capture and reporter antibody for a sandwich assay). The invention further provides humanized and chimeric forms of mouse monoclonals, particularly of the exemplified antibodies shown in Table 1.
  • the invention also provides other antibodies that compete with one of the exemplified antibodies for specific binding to alpha-synuclein (i.e., bind to the same epitope as an exemplified antibody or a sufficiently proximal epitope to interfere with the binding of an exemplified antibody to its epitope).
  • the invention also provides antibodies that bind to the same epitope as one of the exemplified antibodies.
  • Antibodies that compete with or bind to the same epitope as an exemplified antibody are expected to show similar functional properties.
  • Such antibodies include mouse and other nonhuman antibodies, human antibodies, chimerics and humanized antibodies.
  • Such antibodies include monoclonal antibodies and polyclonal antibody.
  • a polyclonal antibody specifically binds to an epitope if it binds to the epitope without binding to other regions of alpha synuclein. This is usually true of a monoclonal antibody as well. However, some monoclonal antibodies can be designed or selected to have specificity for two epitopes within alpha synuclein.
  • Some preferred epitope specificities include monoclonal antibodies that specifically binds to a repeated epitope comprising residues within residues 43-51 and residues within residues 58-65, monoclonal antibodies that specifically binds to an epitope within residues 118-126; monoclonal antibodies that specifically binds to an epitope comprising residues 91- 99, monoclonal antibodies that specifically binds to an epitope comprising residues 40-55; monoclonal antibodies that specifically binds to an epitope comprising residues 124-134, wherein residue 129 is phosphorylated serine, and monoclonal antibodies that specifically binds to an epitope comprising residues 123-127, wherein residue 125 is nitrated tyrosine.
  • an antibody When an antibody is said to bind to an epitope within specified residues, such as alpha-SN 109-120 , for example, what is meant is that the antibody specifically binds to a polypeptide consisting of the specified residues ⁇ i.e., alpha-SN 109-120 in this an example). Such an antibody does not necessarily contact every residue within alpha-SN 109-120. Nor does every single amino acid substitution or deletion with in alpha-SN109-120 necessarily significantly affect binding affinity.
  • Epitope specificity of an antibody can be determined, for example, by testing a collection of overlapping peptides of about 15 amino acids spanning the sequence of alpha-synuclein and differing in increments of a small number of amino acids (e.g., 3 amino acids).
  • the peptides are immobilized within the wells of a microtiter dish. Immobilization can be effected by biotinylating one terminus of the peptides.
  • different samples of the same peptide can be biotinylated at the N and C terminus and immobilized in separate wells for purposes of comparison. Such is particularly useful for identifying end-specific antibodies.
  • additional peptides can be included terminating at a particular amino acid of interest (e.g., the first and last residue of the NAC fragment).
  • a particular amino acid of interest e.g., the first and last residue of the NAC fragment.
  • An antibody is screened for specific binding to each of the various peptides.
  • the epitope is defined as occurring within a segment of amino acids that is common to all peptides to which the antibody shows specific binding.
  • the basic antibody structural unit is known to comprise a tetramer of subunits. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light” (about 25 kDa) and one "heavy" chain (about 50-70 IcDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's isotype as IgG, IgM, IgA, IgD and IgE, respectively.
  • the variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D” region of about 10 more amino acids.
  • variable regions of each light/heavy chain pair form the antibody binding site.
  • an intact antibody has two binding sites.
  • the chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs.
  • the CDRs from the two chains of each pair are aligned by the framework regions, enabling binding to a specific epitope.
  • FRl relatively conserved framework regions
  • CDRs complementarity determining regions
  • Chimeric and humanized antibodies have the same or similar binding specificity and affinity as a mouse or other nonhuman antibody that provides the starting material for construction of a chimeric or humanized antibody.
  • Chimeric antibodies are antibodies whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin gene segments belonging to different species. For example, DNA encoding the variable domains of a mouse antibody can be sequenced, and DNA construct(s) encoding the variable domains joined to human constant (C) segments, such as IgGl and IgG4 constructed. The constructs are then expressed to produce the antibody Human isotype IgGl is preferred. In some methods, the isotype of the antibody is human IgGl. IgM antibodies can also be used in some methods.
  • a typical chimeric antibody is thus a hybrid protein consisting of the V or antigen-binding domain from a mouse antibody and the C or effector domain from a human antibody.
  • Humanized antibodies have variable region framework residues substantially from a human antibody or consensus of human antibodies (termed an acceptor antibody) and some and usually all six complementarity determining regions substantially or entirely from a mouse-antibody, (referred to as the donor immunoglobulin). See, Queen et ah, Proc. Natl. Acad. ScL USA 86:10029-10033 (1989), WO 90/07861, US 5,693,762, US 5,693,761, US 5,585,089, US 5,530,101, and Winter, US 5,225,539 (each of which is incorporated by reference in its entirety for all purposes).
  • the constant region(s), if present, are also substantially or entirely from a human immunoglobulin.
  • the human variable domains are usually chosen from human antibodies whose framework sequences exhibit a high degree of sequence identity with the murine variable region domains from which the CDRs were derived.
  • the heavy and light chain variable region framework residues can be derived from the same or different human antibody sequences.
  • the human antibody sequences can be the sequences of naturally occurring human antibodies or can be consensus sequences of several human antibodies. See Carter et al, WO 92/22653.
  • Certain amino acids from the human variable region framework residues are selected for substitution based on their possible influence on CDR conformation and/or binding to antigen. Investigation of such possible influences is by modeling, examination of the characteristics of the amino acids at particular locations, or empirical observation of the effects of substitution or mutagenesis of particular amino acids.
  • the human framework amino acid should usually be substituted by the equivalent framework amino acid from the mouse antibody when it is reasonably expected that the amino acid:
  • variable region frameworks of humanized immunoglobulins usually show at least 85% sequence identity to a human variable region framework sequence or consensus of such sequences.
  • Human antibodies against alpha-SN are provided by a variety of techniques described below. Some human antibodies are selected by competitive binding experiments, or otherwise, to have the same epitope specificity as a particular mouse antibody, such as one of the mouse monoclonals shown in Table 1. Human antibodies can also be screened for a particular epitope specificity by using only a fragment of alpha-SN as the immunogen, and/or by screening antibodies against a collection of deletion mutants of alpha-SN. Human antibodies preferably have isotype specificity human IgGl. Several methods are available for producing human antibodies including the trioma method, Oestberg et al, Hybridoma 2:361- 367 (1983); Oestberg, US Patent No.
  • the heavy and light chain variable regions of chimeric, humanized, or human antibodies can be linked to at least a portion of a human constant region.
  • the choice of constant region depends, in part, whether antibody-dependent complement and/or cellular mediated toxicity is desired.
  • isotopes IgGl and IgG3 have complement activity and isotypes IgG2 and IgG4 do not.
  • Choice of isotype can also affect passage of antibody into the brain.
  • Human isotype IgGl is preferred.
  • Light chain constant regions can be lambda or kappa.
  • Antibodies can be expressed as tetramers containing two light and two heavy chains, as separate heavy chains, light chains, as Fab, Fab' F(ab')2, and Fv, or as single chain antibodies in which heavy and light chain variable domains are linked through a spacer.
  • Chimeric, humanized and human antibodies are typically produced by recombinant expression.
  • Recombinant polynucleotide constructs typically include an expression control sequence operably linked to the coding sequences of antibody chains, including naturally associated or heterologous promoter regions.
  • the expression control sequences are eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and the collection and purification of the crossreacting antibodies.
  • expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA.
  • expression vectors contain selection markers, e.g., ampicillin-resistance or hygromycin-resistance, to permit detection of those cells transformed with the desired DNA sequences.
  • E. coli is one prokaryotic host particularly useful for cloning the DNA sequences of the present invention.
  • Microbes such as yeast are also useful for expression. Saccharomyces is a preferred yeast host, with suitable vectors having expression control sequences, an origin of replication, termination sequences and the like as desired.
  • Typical promoters include 3- phosphoglycerate kinase and other glycolytic enzymes.
  • Inducible yeast promoters include, among others, promoters from alcohol dehydrogenase, isocytochrome C, and enzymes responsible for maltose and galactose utilization.
  • Mammalian cells are a preferred host for expressing nucleotide segments encoding immunoglobulins or fragments thereof. See Winnacker, From Genes to Clones, (VCH Publishers, NY, 1987).
  • a number of suitable host cell lines capable of secreting intact heterologous proteins have been developed in the art, and include CHO cell lines, various COS cell lines, HeLa cells, L cells, human embryonic kidney cell, and myeloma cell lines.
  • the cells are nonhuman.
  • Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer (Queen et al, Immunol. Rev.
  • Preferred expression control sequences are promoters derived from endogenous genes, cytomegalovirus, S V40, adenovirus, bovine papillomavirus, and the like. See Co et al, J. Immunol. 148:1149 (1992).
  • antibody coding sequences can be incorporated in transgenes for introduction into the genome of a transgenic animal and subsequent expression in the milk of the transgenic animal (see, e.g., US 5,741,957, US 5,304,489, US 5,849,992).
  • Suitable transgenes include coding sequences for light and/or heavy chains in operable linkage with a promoter and enhancer from a mammary gland specific gene, such as casein or beta lactoglobulin.
  • the vectors containing the DNA segments of interest can be transferred into the host cell by well-known methods, depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment, electroporation, lipofection, biolistics or viral-based transfection can be used for other cellular hosts. Other methods used to transform mammalian cells include the use of polybrene, protoplast fusion, liposomes, electroporation, and microinjection (see generally, Sambrook et al, supra). For production of transgenic animals, transgenes can be microinjected into fertilized oocytes, or can be incorporated into the genome of embryonic stem cells, and the nuclei of such cells transferred into enucleated oocytes.
  • antibodies can be purified according to standard procedures of the art, including HPLC purification, column chromatography, and gel electrophoresis and the like (see generally, Scopes, Protein Purification (Springer- Verlag, NY, 1982).
  • Alpha synuclein was originally identified in human brains as the precursor protein of the non-
  • Alpha-SN also termed the precursor of the non- A/3 component of AD amyloid (NACP), is a peptide of 140 amino acids.
  • Alpha-SN has the amino acid sequence:
  • NAC non-A/3 component of AD amyloid
  • NAC a highly hydrophobic domain within alpha synuclein, is a peptide consisting of at least 28 amino acids residues (residues 60-87) (SEQ ID NO: 3) and optionally 35 amino acid residues (residues 61-95) (SEQ ID NO: 2).
  • SEQ ID NO: 3 amino acids residues
  • SEQ ID NO: 2 amino acids residues
  • EQVTNVGGAVVTGVTAVAQKTVEGAGSIAAATGFV (SEQ ID NO: 2) (Jensen et al., Biochem. J. 310 (Pt 1): 91-94 (1995); GenBank accession number S56746).
  • NAC has the acid sequence:
  • Disaggregated alpha-SN or fragments thereof including NAC, means monomelic peptide units. Disaggregated alpha-SN or fragments thereof are generally soluble, and are capable of self-aggregating to form soluble oligomers. Oligomers of alpha-SN and fragments thereof are usually soluble and exist predominantly as alpha-helices. Monomelic alpha-SN may be prepared in vitro by dissolving lyophilized peptide in neat DMSO with sonication. The resulting solution is centrifuged to remove any insoluble particulates. Aggregated alpha- SN or fragments thereof, including NAC, means oligomers of alpha-SN or fragments thereof which have associate into insoluble beta-sheet assemblies.
  • Aggregated alpha-SN or fragments thereof, including NAC means also means fibrillar polymers. Fibrils are usually insoluble. Some antibodies bind either soluble alpha-SN or fragments thereof or aggregated alpha-SN or fragments thereof. Some antibodies bind both soluble and aggregated alpha-SN or fragments thereof. [0062] Unless otherwise indicated, reference to alpha-SN means the natural human amino acid sequence indicated above as well as natural allelic and species variants thereof, including full-length forms and immunogenic fragments thereof, as well as forms having undergone posttranslational modification, such as phosphorylation. Induced variants of alpha-SN or can also be used but are not preferred. Such variants typically differ from naturally occurring peptides at one, two or a few positions, often by virtue of conservative substitutions. Analogs can also unnatural amino acids, but such is not preferred.
  • Alpha-SN, its fragments, and analogs can be synthesized by solid phase peptide synthesis or recombinant expression, or can be obtained from natural sources.
  • Automatic peptide synthesizers are commercially available from numerous suppliers, such as Applied Biosystems, Foster City, California.
  • Recombinant expression can be in bacteria, such as E. coli, yeast, insect cells or mammalian cells. Procedures for recombinant expression are described by Sambrook et ah, Molecular Cloning: A Laboratory Manual (C.S.H.P. Press, NY 2d ed., 1989).
  • Some forms of alpha-SN peptide are also available commercially, for example, at BACHEM and American Peptide Company, Inc.
  • Beta synuclein shows 78% sequence similarity with alpha synuclein at the amino acid level.
  • Gamma synuclein shares 60% similarity at the amino acid level with alpha- synuclein (see Biere et al., J. Biol. Chem., Vol. 275, Issue 44, 34574-34579, November 3, 2000, incorporated by reference).
  • Antibodies of the invention can be used to detect alpha synuclein or fragments thereof in a variety of formats including immunoprecipitation, Western blotting, ELISA, radioimmunoassay, competitive and isometric assays. See Harlow & Lane, Antibodies ⁇ A Laboratory Manual (CSHP NY, 1988); U.S. Patent Nos.
  • Isometric or sandwich assays are a preferred format (see US 4,376,110, 4,486,530, 5,914,241, and 5,965,375). Such assays use one antibody immobilized to a solid phase (capture antibody), and another antibody in solution (reporter antibody). Typically, the reporter antibody is labeled, either directly or via a secondary labeling reagent, such as an anti-idiotypic antibody.
  • the capture and reporter antibodies having different binding specificities so both can bind to alpha-synuclein or a fragment thereof at the same time. Capture and reporter antibodies can be contacted with target antigen in either order or simultaneously. If the capture antibody is contacted first, the assay is referred to as being a forward assay.
  • the assay is referred to as being a reverse assay. If target is contacted with both antibodies simultaneously, the assay is referred to as a simultaneous assay. After contacting the target with antibody, a sample is incubated for a period that usually varies from about 10 min to about 24 hr and is usually about 1 hr. A wash step is then performed to remove components of the sample that do not become specifically bound to the solid phase When capture and reporter antibodies are bound in separate steps, a wash can be performed after either or both binding steps. After washing, binding is quantified, typically by detecting label linked to the solid phase through binding of labeled reporter.
  • a calibration curve is prepared from samples containing known concentrations of target antigen. Concentrations of antigen in samples being tested are then read by interpolation from the calibration curve. Analyte can be measured either from the amount of labeled reporter antibody bound at equilibrium or by kinetic measurements of bound labeled solution antibody at a series of time points before equilibrium is reached. The slope of such a curve is a measure of the concentration of target in a sample. Alternatively, the amount of alpha synuclein in a sample can be determined by comparing the signal of reporter antibody bound to alpha synuclein the sample with the signal from reporter antibody bound to a known amount of alpha synuclein in a control sample.
  • Target alpha-synuclein in a sample competes with exogenously supplied labeled alpha synuclein for binding to an antibody.
  • the amount of labeled alpha synuclein bound to the antibody is inversely proportional to the amount of target alpha synuclein in the sample.
  • the antibody can be immobilized to facilitate separation of the bound complex from the sample prior to detection (heterogeneous assays) or separation may be unnecessary as practiced in homogeneous assay formats.
  • the antibody used as a detection reagent is labeled. When the antibody is labeled, its binding sites compete for binding to the target alpha synuclein in the sample and exogenously supplied form of alpha synuclein immobilized on a solid phase.
  • Suitable detectable labels for use in the above methods include any moiety that is detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical, chemical, or other means.
  • suitable labels include biotin for staining with labeled streptavidin conjugate, fluorescent dyes (e.g., fluorescein, Texas red, rhodamine, green fluorescent protein, and the like), radiolabels (e.g., .
  • Radiolabels can be detected using photographic film or scintillation counters, fluorescent markers can be detected using a photodetector to detect emitted light.
  • Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting the reaction product produced by the action of the enzyme on the substrate, and colorimetric labels are detected by simply visualizing the colored label.
  • Suitable supports for use in the above methods include, for example, nitrocellulose membranes, nylon membranes, and derivatized nylon membranes, and also particles, such as agarose, a dextran-based gel, dipsticks, particulates, microspheres, magnetic particles, test tubes, microtiter wells, SEPHADEXTM. (Amersham Pharmacia Biotech, Piscataway NJ., and the like. Immobilization can be by absorption or by covalent attachment.
  • antibodies can be joined to a linker molecule, such as biotin for attachment to a surface.
  • Solvents used to extract alpha synuclein from tissue samples can decrease the sensitivity of the assay (e.g., 5M guanidine, urea/thiourea/CHAPS, urea/thiourea, 1% SDS, 1%SDS/8M, cell lysis buffer). It is recommended that such solvents be removed or diluted such that they account for less than 1% and preferably less than 0.1% of the buffer used for the assay.
  • Preferred pairs of monoclonal antibodies for use in isometric assays are as follows.
  • the capture antibody specifically binds an epitope within residues selected from the group consisting of N-terminus, 40-55, 91-99 and 118-126
  • the reporter antibody specifically binds an epitope within residues 109-120.
  • Another suitable pairing is an antibody that specifically binds to an epitope within residues 124-134 and is specific for the phosphorylated form and another antibody that specifically binds to an epitope within residues 91-99.
  • Other suitable pairings include an antibody that competes with 6H7 and an antibody that competes with 5C12 or 12Cl; an antibody that competes with 3A12 and an antibody that competes with 5Cl 2 or 6H7; an antibody that competes with 12Cl and an antibody that competes with 5Cl 2 or 6H7; an antibody that competes with 9A6 and an antibody that competes with 5C12, 6H7 or 12Cl; an antibody that competes with 9G5 and an antibody that competes with 5C12, 6H7 or 12Cl;an antibody that competes with 9E4 and an antibody that competes with 5C12, 6H7 or 12Cl; an antibody that competes with 1H7 and an antibody that competes with 5C12, 6H7 or 12Cl; and an antibody that competes with 10G5 and an antibody that competes with 5C12, 6H7 or 12Cl, and antibody that competes with 1H7 and an antibody that competes with 11 A5.
  • Preferred pairings include 6H7 and 5C12 or 12Cl; 3A12 and 5C12 or 6H7; 12Cl and 5C12 or 6H7;9A6 and 5C12, 6H7 or 12Cl; 9G5 and 5C12, 6H7 or 12Cl; 9E4 and 5C12, 6H7 or 12Cl; 1H7 and 5C12, 6H7 or 12Cl; 10G5 and 5C12, 6H7 or 12Cl, 1H7 and 11A5.
  • Some method employ either a capture or a reporter antibody that is end-specific for a fragment of alpha-synuclein, particularly, SNl-119 (i.e., residues 1-119 of SEQ ID NO:1) or SN1-122 (residues 1-122 of SEQ ID NO:1). Applications for detection of these fragments are described below.
  • alpha synuclein in patient samples can be used for diagnosing and monitoring diseases characterized by Lewy bodies or other deposits of alpha synuclein.
  • Synucleinopathic diseases include Parkinson's disease (PD), dementia with Lewy bodies (DLB), the Lewy body variant of Alzheimer's disease (LBVAD), multiple systems atrophy (MSA), neurodegeneration with brain iron accumulation type-1 (NBIA-I), diffuse Lewy body disease (DLBD), and combined PD and Alzheimer's disease (AD).
  • Suitable patient samples include body fluids, such as blood, CSF, urine, and peritoneal fluid.
  • synucleinopathic disease is generally associated with significantly altered levels of alpha synuclein or fragments thereof in the fluid (increased or decreased) when compared to the mean values in normal individuals, i.e., individuals not suffering from a synucleinopathic disease.
  • a level is significantly altered if it departs by more than one standard deviation from the mean level in a population of normal individuals.
  • the measured concentrations of alpha synuclein and its fragments can be monitored to follow the progress of the disease, and potentially follow the effectiveness of treatment. Levels of alpha- synuclein and its fragments revert toward the mean in a population of normal individuals if the treatment regime is effective.
  • In vitro monitoring of alpha synculein and its fragment in conditioned culture medium from a suitable cell culture can be used for analyzing processing and secretion of alpha- synuclein and the effect of potential agents on the same.
  • Monitoring processing of alpha synuclein provides a means to study proteolytic cleavage, identify responsible proteases, determine what factors affect cleavage, and determine the physiological function of truncated forms of alpha synuclein.
  • Agents that inhibit processing and/or secretion of alpha synuclein have pharmacological activity potentially useful for prophylaxis of synucleinopathic disease.
  • inhibitory activity is determined by comparing levels of alpha synuclein and/or its fragments in medium from a cell treated with a test agent versus a comparable control cell not treated with the agent.
  • a test agent e.g., USSN 60/471,929 filed May 19, 2003
  • USSN 10/850,570 filed May 19, 2004
  • attorney docket 015270-011520US filed October 19, 2004, which is a CIP thereof
  • certain fragments of alpha synuclein with C- terminal truncations preferentially accumulate in Lewy bodies (e.g., SNl-119 and SN1-122).
  • media from PeakS cells transfected with alpha synuclein contain two prominent truncated species of about 12 and 7 kDa.
  • Agents inhibiting processing reactions that form such fragments therefore have pharmacological activity useful for treating diseases characterized by Lewy bodies.
  • the NAC fragment of alpha synuclein accumulates in extracellular amyloid deposits in Alzheimer's disease.
  • Agents that inhibit formation or secretion of NAC therefore have pharmacological activity useful for treating Alzheimer's disease.
  • Suitable cells include cells transfected with nucleic acids encoding alpha synuclein, preferably, human alpha synuclein and cells naturally expressing alpha synuclein, also preferably human.
  • the alpha synuclein in transfected cells can bear a mutation, such as S129A, S129D, A53T and A20P.
  • Cells include PeakS cells, SY5Y cells,human cortical cells, human neuroglioma cell lines, human HeLa cells, primary human endothelial cells (e.g. HUVEC cells), primary human fibroblasts or lymphoblasts, primary human mixed brain cells (including neurons, astrocytes, and neuroglia), Chinese hamster ovary (CHO) cells, and the like.
  • SY5Y cells are neuronal cells that can be induced to differentiate by treatment with retinoic acid/BDNF (brain derived neurotrophic factor).
  • BDNF brain derived neurotrophic factor
  • Transfected cells expressing alpha synuclein at higher levels than normal human cells are preferred. In some such cells, the alpha synuclein bears a mutation associated with synucleinopathic disease.
  • Random libraries of peptides or other compounds can also be screened for suitability. Combinatorial libraries can be produced for many types of compounds that can be synthesized in a step-by-step fashion.
  • Such compounds include polypeptides, beta-turn mimetics, polysaccharides, phospholipids, hormones, prostaglandins, steroids, aromatic compounds, heterocyclic compounds, benzodiazepines, oligomeric N-substituted glycines and oligocarbamates.
  • Large combinatorial libraries of the compounds can be constructed by the encoded synthetic libraries (ESL) method described in Affymax, WO 95/12608, Affymax, WO 93/06121, Columbia University, WO 94/08051, Pharmacopeia, WO 95/35503 and Scripps, WO 95/30642 (each of which is incorporated herein by reference for all purposes).
  • Peptide libraries can also be generated by phage display methods.
  • test compounds are typically administered to the culture medium at a concentration in the range from about 1 nM to 1 mM, usually from about 10 ⁇ M to 1 niM.
  • Test compounds which are able to inhibit formation, processing or secretion of alpha synuclein are candidates for further determinations in transgenic animals and eventually human clinical trials.
  • the antibodies of the invention and assays for detecting them can also be used to monitor alpha synuclein production, and processing in transgenic animal models of disease.
  • Transgenic animal models of Lewy body disease are described by Masliah, et al. Science 287:1265-1269 (2000); Masliah et al, PNAS USA 98:12245-12250 (2001).
  • Alpha synuclein can be analyzed either in body fluids as described above for human samples, or in tissue samples taken directly from the animal (see copending 60/423,012, filed November 1, 2002, incorporated by reference). Tissue samples can be classified as Lewy body, particulate fraction and soluble fractions.
  • Simple assays can be performed as for cell culture to screen agents for capacity to inhibit formation of alpha synuclein or its processing to fragments.
  • the inhibitory activity is determined by comparing the level of alpha synuclein or a fragment thereof in a particularly body fluid or fraction from a tissue sample from a transgenic animal treated with the agent in comparison with the level of alpha synuclein or the fragment in the same body fluid or fraction in a control transgenic animal not treated with the agent.
  • Inhibitory activity is shown by decreased levels of alpha synuclein of fragment thereof in the treated animal relative to the control.
  • Tissue samples from the brains of human patients can be subject to similar analyses.
  • analyses are usually confined to cadavers.
  • the analyses are useful, for example, in identifying fragments of alpha synuclein that preferentially accumulate in Lewy bodies, as described in copending application 60/471, 929, filed 05/19/2003 and its progeny supra. V. Kits
  • kits including one or more antibodies of the invention.
  • the antibodies are preimmobilized to a solid phase.
  • labeling reagents such as an antiidiotypic antibody are also included in the kits.
  • the labeling may also include a chart or other correspondence regime correlating levels of measured label with levels of antibodies to alpha-SN.
  • labeling refers to any written or recorded material that is attached to, or otherwise accompanies a kit at any time during its manufacture, transport, sale or use.
  • labeling encompasses advertising leaflets and brochures, packaging materials, instructions, audio or video cassettes, computer discs, as well as writing imprinted directly on kits.
  • the kits can be sold, for example, as research or diagnostic reagents.
  • IP immunoprecipitation
  • H histology
  • EC and ER ELISA capture and reporter antibody
  • WB western blot
  • the antibodies were produced by immunizing with the antigen shown. Hybridomas were produced by standard methods, and tested for appropriate binding specificity.
  • Capture antibodies were biotinylation by a modification of the Igen commercial method. Pierce sulfo NHS biotin was prepared at 4 mg/ml in H2O and added at a 40 M excess to 1 mg/ml antibody in 150 mM KPO4 pH 7.8. This was incubated with gentle mixing in the dark for two hours and then dialyzed against PBS to remove free biotin. Capture antibodies were diluted to 10 ⁇ g/ml in Well Coating Buffer 0.1 M PO4. pH 8.5. Plates were coated 100 ⁇ l per well in Dynx 4HBX plates overnight at room temperature. Plates were then blocked in 0.25% Casein/PBS.
  • Standard curves of synuclein were prepared either in Specimen Diluent- (0.6% BSA, 0.05% Triton 405, 0.5% Thimerisol in 0.1 M PO4, .15 M NaCl pH 7.4) or 0.5 M guanidine, 0.25% casein/ protease inhibitor/ PBS depending on the samples to be assayed. Samples were diluted in either Specimen Diluent or the guanidine/casein depending on sample. Standards and samples were incubated over night at 4°C. For simple samples, room temperature and a few hours are sufficient. Plates were washed 4 times.
  • ELISA assays were developed for the measurement of synuclein from various sources included human and rodent brain, cell culture and Lewy Body Preps. The follow table describes the sensitivity of various capture antibodies when 5Cl 2 biotin is used as the reporter
  • the combination of 11 A5 capture and 5C12 reporter is suitable for detecting alpha synuclein phosphorylated at residue 129.
  • the combination of 1H7 as capture antibody and 11 A5 as reporter offers about ten-fold greater sensitivity.
  • the 11 A5 antibody is specific for alpha synuclein phosphorylated at residue 129.
  • FIG. 2 A calibration curve showing OD450 vs. concentration of alpha synuclein using 1H7 and 5Cl 2 as capture and reporter antibody is shown in Fig. 2. The assay is more sensitive in the absence of guanidine.
  • a calibration curve for 1H7/9E4 in the presence of 0.5 M guanidine buffer is shown in Fig. 3. The guanidine maintains synuclein in a mildly denatured state making epitopes available for antibody binding and decreasing interactions with other proteins.
  • Fig. 4 shows a calibration curve for the combination of 11 A5/5C12 for detection of phosphorylated alpha synuclein.
  • the actual sensitivity is about twenty fold greater than the measure value of 9 ng/ml because the alpha synuclein was only about 6% phosphorylated.
  • the capacity of the ELISA assay to quantify synuclein in a sample was testing by spiking brain homogenates from nontransgenic mice with known amounts of alpha synuclein.
  • Fig. IA shows the results using 1H7/5C12 as capture and reporter antibodies. The assay detects 96% of the added alpha synuclein. Some synuclein is detected in the control because the 5C12 antibody binds to both mouse and human alpha synuclein.
  • IB shows the results using 1H7 and 9E4 as capture and reporter antibody, hi this case, no alpha synuclein is detected in the control because 9E4 binds to human alpha synuclein only. 92% of the added alpha synuclein was detected in the spiked sample, showing that detection is quantitative.
  • the ELISA assay was used to detect alpha synuclein in soluble, particulate and Lewy body preparations from DLBD patients and controls.
  • a brain homogenate was spun at 100Og.
  • the pellet was the source of Lewy bodies. These were prepared by Percoll gradient density fractionation, DNA digestion, and immunoaffinity purification using antibodies tagged with magnetic beads.
  • the alpha synuclein content of Lewy bodies can be enriched by stripping with the anionic detergent sarkosyl.
  • the soluble and particulate fractions were prepared by spinning the initial supernatant at 150,00Og. The resulting supernatant was referred to as a soluble fraction, and the pellet as the particulate fraction.
  • Synuclein can be extracted from these fractions using 0.5 and 5 M guanidine respectively. Note that the Lewy body samples were in UTC and that the soluble brain samples were prepared in Tris-buffered sucrose, and were assayed at a dilution in which these buffers did not interfere with the ELISA.
  • the soluble extract of the 8 human brains assayed contain approximately 0.03% of ⁇ - synuclein. There is a trend toward decreasing amounts of oc-synuclein in the soluble brain fraction of a diseased patient. (Table 4). There is a statistically significant increase in the amount of synuclein in the particulate fraction of the DLBD brain (Table 5). The increased level of synuclein may be due to immature Lewy bodies that are to small to pellet with mature Lewy bodies in the initial centrirugation.
  • the amount of ⁇ -synuclein in the Lewy body prep varies.
  • synuclein comprises at most 0.07% of the total Lewy body protein.
  • the 1H7/9E4 ELISA measures approximately 60% less ⁇ -synuclein in the Lewy bodies than the 1H7/5C12 ELISA .
  • the 9E4 reporter antibody recognizes an epitope more C-terminal than that of 5 C 12, and therefore the assay may simply reflect the presence of C-terminal truncations in the Lewy bodies as seen on 2-D gels.
  • 9E4 reactivity or a lowered ratio of 9E4/5C12 activity is a marker of pathogenicity.
  • the 9E4 epitope encompasses the estimated cleavage sites.
  • the soluble fraction contains less truncated synuclein and did show only a 12% difference between the ELISA assays consistent with this interpretation.
  • An alternate explanation is that 9E4's epitope is modified or masked (perhaps due to the aggregation as described above or modification of tyrosine at 125) and cannot be detected.
  • Plasma from the EDTA collection gave excellent spike and recovery of ⁇ -synuclein.
  • blood was processed to generate total blood cells, platelets and plasma.
  • the blood was spun at 350 X g to pellet cells but not platelets.
  • the plasma was removed and spun at 1000 X g to remove platelets.
  • the cell pellet was washed two times at 350 X g to remove any remaining platelets. Both plasma and cells were initially diluted/homogenized in 5 M guanidine. The cell pellet was further diluted 1:10, treated with DNAse and spun to remove insoluble material.
  • To test the synuclein specificity of the ELISA an aliquot of the platelet and the total blood cell samples were absorbed with the synuclein antibody 1H7 coupled to Sepharose.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The invention provides methods for detecting alpha-synuclein. The invention also identifies preferred epitopes of alpha synuclein for use in such detection, and provides antibodies specifically binding to such epitopes.

Description

ANTIBODIES TO ALPHA-SYNUCLEIN
BACKGROUND OF THE INVENTION
[0001] Alpha-synuclein (alphaSN) brain pathology is a conspicuous feature of several neurodegenerative diseases, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), the Lewy body variant of Alzheimer's disease (LBVAD), multiple systems atrophy (MSA), and neurodegeneration with brain iron accumulation type-1 (NBIA-I). Common to all of these diseases, termed synucleinopathies, are proteinaceous insoluble inclusions in the neurons and the glia which are composed primarily of alphaSN.
[0002] Lewy bodies and Lewy neurites are intraneuronal inclusions which primarily contain of alphaSN. Lewy bodies and Lewy neurites are the neuropathological hallmarks Parkinson's disease (PD). PD and other synucleinopathic diseases have been collectively referred to as Lewy body disease (LBD). LBD is characterized by degeneration of the dopaminergic system, motor alterations, cognitive impairment, and formation of Lewy bodies (LBs). (McKeith et ah, Clinical and pathological diagnosis of dementia with Lewy bodies (DLB): Report of the CDLB International Workshop, Neurology (1996) 47:1113-24). Other LBDs include diffuse Lewy body disease (DLBD), Lewy body variant of Alzheimer's disease (LBVAD), combined PD and Alzheimer's disease (AD), and multiple systems atrophy. Dementia with Lewy bodies (DLB) is a term coined to reconcile differences in the terminology of LBDs.
[0003] Disorders with LBs continue to be a common cause for movement disorders and cognitive deterioration in the aging population (Galasko et al., Arch. Neurol. (1994) 51:888- 95). Although their incidence continues to increase creating a serious public health problem, to date these disorders are neither curable nor preventable and understanding the causes and pathogenesis of PD is critical towards developing new treatments (Tanner et ah, Curr. Opin. Neurol. (2000) 13:427-30). The cause for PD is controversial and multiple factors have been proposed to play a role, including various neurotoxins and genetic susceptibility factors.
[0004] In recent years, new hope for understanding the pathogenesis of PD has emerged. Specifically, several studies have shown that the synaptic protein alpha-SN plays a central role in PD pathogenesis since: (1) this protein accumulates in LBs (Spillantini et al, Nature (1997) 388:839-40; Takeda et al., AM. J. Pathol. (1998) 152:367-72; Wakabayashi et al, Neurosci. Lett. (1997) 239:45-8), (2) mutations in the alpha-SN gene co-segregate with rare familial forms of parkinsonism (Rruger et al, Nature Gen. (1998) 18:106-8; Polymeropoulos MH, et al, Science (1997) 276:2045-7) and, (3) its overexpression in transgenic mice (Masliah et al, Science (2000) 287:1265-9) and Drosophila (Feany et al, Nature (2000) 404:394-8) mimics several pathological aspects of PD. Thus, the fact that accumulation of alpha-SN in the brain is associated with similar morphological and neurological alterations in species as diverse as humans, mice, and flies suggests that this molecule contributes to the development of PD.
[0005] Alpha-SN is part of a large family of proteins including beta- and gamma- synuclein and synoretin. Alpha-SN is expressed in the normal state associated with synapses and is believed to play a role in neural plasticity, learning and memory. Mutations in human (h) alpha-SN that enhance the aggregation of alpha-SN have been identified (Ala30Pro and Ala53Thr) and are associated with rare forms of autosomal dominant forms of PD. The mechanism by which these mutations increase the propensity of alpha-SN to aggregate are unknown.
SUMMARY OF THE CLAIMED INVENTION
[0006] The invention provides a monoclonal antibody that competes with a monoclonal antibody shown in Table 1 for specific binding to alpha synuclein. Optionally, the monoclonal antibody specifically binds alpha synuclein compared to beta synuclein. Optionally, the monoclonal antibody specifically binds human alpha synuclein compared to non-human alpha synuclein. Optionally, the monoclonal specifically binds to an epitope bound by a monoclonal antibody shown in Table 1. Some antibodies specifically binds to an epitope within residues 109-120, or to an epitope on the C-terminus of synuclein, or to an epitope comprising residues 139-140. Some antibodies specifically bind to an epitope on the N-terminus of synuclein. Some antibodies specifically bind to an epitope within residues 43- 51 and within residues 58-65. Some antibodies specifically bind to a repeated epitope comprising residues within residues 43-51 and residues within residues 58-65. Some antibodies specifically bind to an epitope within residues 118-126. Some antibodies specifically bind to an epitope comprising residues 91-99. Some antibodies specifically bind to an epitope comprising residues 40-55. Some antibodies specifically bind to an epitope comprising residues 124-134, wherein residue 129 is phosphorylated serine. Some antibodies specifically bind to an epitope comprising residues 123-127, wherein residue 125 is nitrated tyrosine. Some antibodies specifically binds to human alpha synuclein without specifically binding to mouse alpha synuclein.
[0007] Preferred antibodies include 9E4 or an antibody that competes with 9E4 for specific binding to human alpha synuclein. Other preferred antibodies include 11 A5 or an antibody that competes with 11 A5 for specific binding to phosphorylated human alpha synuclein. Other preferred antibodies include 6H7 or an antibody that competes with 6H7 for specific binding to alpha synuclein. Other preferred antibodies include 4Bl or 8A5 or an antibody that competes with 4Bl or 8A5 for specific binding to alpha synuclein. Other preferred antibodies include 7G4, 6A8, 5C12, 6A12, 9G5, and 1H7, and antibodies that competes therewith for specific binding to alpha synuclein.
[0008] Some antibodies specifically bind to mouse alpha synuclein without specifically binding to human alpha synuclein. Some antibodies specifically bind to mouse and human alpha synuclein. Some antibodies specifically bind to alpha synuclein phosphorylated at residue 129 without specifically binding to nonphosphorylated alpha synuclein. Some antibodies are end-specific for the N-terminus or C-terminus of alpha synuclein. Some antibodies specifically bind to alpha synuclein without specifically binding to beta or gamma synuclein.
[0009] Some antibodies specifically bind to an epitope within a segment of amino acids selected of the group consisting of amino acids 109-120, 43-51 and 58-65, 91-96, 118-126, and 91-99. Exemplary monoclonal antibodies are shown in Table 1 and include 7G4, 6A8, 5C12, 6A12, 8A5, 4Bl, 6H7, 3A12, 12Cl, 9A6, 9G5, 9E4, 23E8, 10G5, 3C12, 11A5 andlH7.
[0010] Some antibodies are monoclonal antibodies that specifically binds to phosphorylated alpha synuclein. Some antibodies are monoclonal antibodies that specifically bind to nitrated synuclein. Some antibodies are monoclonal antibodies capable of capturing soluble synuclein from a fluid sample, preferably alpha-synuclein and more preferably human alpha synuclein..
[0011] Some antibodies are monoclonal antibodies that specifically bind to an epitope within residues selected from the group consisting of the N-terminus, 118-126, 91-99 and 40-55. Some monoclonal antibodies competes with the monoclonal antibody selected from the group consisting of 6H7, 3QA12, 12C6, 12Cl, 9A6, 9G5, 9E4, 1H7 and 23E8. Exemplary antibodies include monoclonals 6H7, 3QA12, 12C6. 12Cl, 9A6, 9G5, 9E4, 1H7 and 23E8. [0012] The invention further provides a pair of monoclonal antibodies, each of the monoclonal antibodies specifically binding to a different epitope within synuclein, wherein the monoclonal antibodies are capable of detecting soluble synuclein when used together in an ELISA assay. Optionally, one monoclonal antibody is immobilized to a solid phase. In some methods, the use of the monoclonal antibodies in the ELISA assay is sequential. In some methods, one monoclonal antibody is a capture antibody and the other monoclonal antibody is a reporter antibody. In some methods, the capture antibody specifically binds an epitope within residues selected from the group consisting of N-terminus, 40-55, 91-99 and 118-126, and the reporter antibody specifically binds an epitope within residues 109-120. In some methods, the pair is selected from the group of an antibody that competes with 6H7 and an antibody that competes with 5C12 or 12Cl, an antibody that competes with 3A12 and an antibody that competes with 5C12 or 6H7, an antibody that competes with 12Cl and an antibody that competes with 5Cl 2 or 6H7, an antibody that competes with 9A6 and an antibody that competes with 5Cl 2, 6H7 or 12Cl, an antibody that competes with 9G5 and an antibody that competes with 5C12, 6H7 or 12Cl, an antibody that competes with 9E4 and an antibody that competes with 5C12, 6H7 or 12Cl,an antibody that competes with 1H7 and an antibody that competes with 5C12, 6H7 or 12Cl, and an antibody that competes with 10G5 and an antibody that competes with 5C12, 6H7 or 12Cl. Optionally, the pair is selected from the group of 6H7 and 5C12 or 12Cl; 3A12 and 5C12 or 6H7; 12Cl and 5C12 or 6H7; 9A6 and 5C12, 6H7 or 12Cl; 9G5 and 5C12, 6H7 or 12Cl; 9E4 and 5C12, 6H7 or 12C1;1H7 and 5C12, 6H7 or 12Cl; and 10G5 and 5C12, 6H7 or 12Cl.
[0013] The invention further provides a hybridoma producing a monoclonal antibody shown in Table 1.
[0014] The invention further provides a humanized or chimeric version of a monoclonal antibody shown in Table 1.
[0015] The invention further provides a method of humanizing a monoclonal antibody shown in Table 1, comprising: determining the amino acid sequence of CDR regions of the monoclonal antibody; selecting an acceptor antibody; and producing a humanized antibody comprising the CDRs from the monoclonal antibody and variable region frameworks from the acceptor antibody.
[0016] The invention further provides a method of producing a chimeric form of an antibody shown in Table 1, comprising: determining the amino acid sequence of the light and heavy chain variable regions of the monoclonal antibody; selecting heavy and light chain constant region; producing a chimeric antibody comprising a light chain comprising the light chain variable region fused to the light chain constant region, and a heavy chain comprising the heavy chain variable region fused to the heavy chain constant region.
[0017] The invention further provides a method for detecting alpha synuclein in a fluid sample, comprising capturing the alpha synuclein using a capture antibody and detecting the alpha synuclein using a reporter antibody, wherein the capture and reporter antibodies bind to different epitopes on alpha synuclein. In some methods, the fluid sample contains 0.1-1.0 M guanidine. hi some methods, the fluid sample contains 0.5 M guanidine. In some methods, the reporter antibody or the capture antibody is end-specific for the C-terminal end of SNl- 119 or SN1-122. In some methods, the capturing step is performed twice with a first reporter antibody specifically binding to an epitope within SNl-119 and a second reporter antibody specifically binding to an antibody within SN120-140 and determining a ratio of captured alpha synuclein between the two steps, a higher ratio of synuclein captured by first reporter relative to second report being indicative of pathogenicity. In some methods, the reporter antibody in the first capturing step specifically binds to an epitope within SN 109-120, and the reporter antibody in the second capturing step specifically binds to an epitope within SN 120-126, and the capture antibody in both capturing steps specifically binds to an epitope within SN 91-99. hi some methods, the capture antibody is immobilized to a solid phase, and the reporter antibody in solution, and the detecting step comprises detecting alpha synuclein from presence of the reporter antibody linked to the solid phase via binding of the reporter antibody to alpha synuclein, which is in turn bound to the capture antibody. Some methods involve a further step of comparing a signal from the reporter antibody linked to the solid phase from analyzing the sample with a signal from the reporter antibody linked to the solid phase from analyzing a control sample containing a known amount of alpha synuclein to determine the amount of alpha synuclein in the sample, hi some methods, a signal from the reporter antibody linked to the solid phase from analyzing the sample to a calibration curve of signal is compared with an amount of alpha synuclein to determine the amount of alpha synuclein in the sample. In some methods, a signal from the reporter antibody is proportional to the amount of synuclein in the sample. Some methods further comprise contacting the reporter antibody with a labeled antibody to generate a signal indicating presence of the reporter antibody. In some methods, the capture antibody specifically binds to an epitope within amino acids 91-99 of human alpha synuclein and the reporter antibody binds to an epitope within amino acids 109-120 of alpha synuclein. In some methods, the capture antibody is 1H7 and the reporter antibody is 5C12. Li some methods, the sample is a sample from a human. In some methods, the capture antibody is 1H7 and the reporter antibody is 9E4. In some methods, the sample from a mouse. In some methods, the sample is a body fluid. In some methods, the sample is cerebrospinal fluid (CSF) of a human, hi some methods, the sample is a brain homogenate of a human or transgenic animal. In some methods, the sample is a medium used to culture cells, hi some methods, the cells express recombinant alpha synuclein. hi some methods, the sample contains a fragment of full-length alpha synuclein and the detecting step detects the fragment.
[0018] The invention further provides a method of monitoring processing of alpha-synuclein to a fragment, comprising culturing a cell expressing alpha synuclein and processing the alpha synuclein to a fragment that is secreted to the cell media; and detecting the fragment in the cell media. Li some methods, the cell is a PeakS or SY5Y cell transfected with alpha synuclein. Li some methods, the cell is a cortical cell.
[0019] The invention further provides a method of screening an agent for activity in inhibiting processing or secretion of alpha synuclein, comprising contacting a cell expressing alpha synuclein in culture medium with an agent; detecting alpha synuclein or a fragment thereof in the medium; and comparing the amount of alpha synuclein or the fragment in the medium with an among of alpha synuclein or fragment in medium from a control cell not contacted with the agent, wherein a reduction in the amount of alpha synuclein or the fragment indicates the agent inhibits processing or secretion of the alpha synuclein.
[0020] The invention further provides a method for detecting soluble alpha synuclein in a fluid sample, comprising: capturing the soluble fragment from the sample using a first binding substance under conditions in which the first binding substance specifically binds to a first epitope and detecting capture of the soluble alpha synuclein using a second binding substance which binds to an epitope on a second region of the soluble alpha synuclein different from the first epitope.
[0021] The invention further provides a method for detecting a soluble fragment of alpha synuclein in a fluid sample in the presence of full length alpha synuclein the method comprising: capturing the soluble fragment from the sample using a first binding substance, and detecting the soluble fragment using a second binding substance, wherein both binding substances specifically bind to the fragment and at least one specifically binds to the fragment without binding to full length alpha synuclein.
BRIEF DESCRIPTION OF THE DRAWINGS
[0022] Figs. IA and B show quantitative detection of alpha synuclein using an ELISA assay with two pairs of antibodies 1H7/5C12 and 1H7/9E4 respectively.
[0023] Fig. 2 shows a calibration curve for detection of alpha synuclein using 1H7/5C12 as capture and reporter antibodies.
[0024] Fig. 3 shows a calibration curve for detection of alpha synuclein using 1H7 and 9E4 as capture and reporter antibodies.
[0025] Fig. 4 shows a calibration curve for detection of phosphorylated alpha synuclein using 11A5 and 5C12 as capture and reporter antibodies.
DEFINITIONS
[0026] The phrase that an antibody "specifically binds" to a target refers to a binding reaction which is determinative of the presence of the antibody in the presence of a heterogeneous population of other biologies. Thus, under designated immunoassay conditions, a specified molecule binds preferentially to a particular target and does not bind in a significant amount to other biologies present in the sample. Specific binding of an antibody to a target under such conditions requires the antibody be selected for its specificity to the target. A variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See, e.g., Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity. Specific binding between two entities means an affinity of at least 106, 107, 108, 109 M"1, or 1010 M"1. Affinities greater than 108 M"1 are preferred. Lack of specific binding means an affinity of less than 106 M"1.
[0027] The term "antibody" or "immunoglobulin" is used to include intact antibodies and binding fragments thereof. Typically, fragments compete with the intact antibody from which they were derived for specific binding to an antigen fragment including separate heavy chains, light chains Fab, Fab1 F(ab')2, Fabc, and Fv. Fragments are produced by recombinant DNA techniques, or by enzymatic or chemical separation of intact immunoglobulins. The term "antibody" also includes one or more immunoglobulin chains that are chemically conjugated to, or expressed as, fusion proteins with other proteins. The term "antibody" also includes bispecific antibody. A bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann, Clin. Exp. Immunol. 79:315-321 (1990); Kostelny et al., J. Immunol. 148, 1547-1553 (1992).
[0028] Antibodies of the invention are typically substantially pure from undesired contaminant. This means that an agent is typically at least about 50% w/w (weight/weight) purity, as well as being substantially free from interfering proteins and contaminants. Sometimes the antibodies are at least about 80% w/w and, more preferably at least 90 or about 95% w/w purity. However, using conventional protein purification techniques, homogeneous peptides of at least 99% w/w can be obtained.
[0029] An "antigen" is an entity to which an antibody specifically binds.
[0030] The term "epitope" or "antigenic determinant" refers to a site on an antigen to which B and/or T cells respond. B-cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Methods of determining spatial conformation of epitopes include, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, Glenn E. Morris, Ed. (1996). Antibodies that recognize the same epitope can be identified in a simple immunoassay showing the ability of one antibody to block the binding of another antibody to a target antigen.
[0031] The term "compete" means competition between antibodies is determined by an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody to a common antigen, such as alpha-SN. Numerous types of competitive binding assays are known, for example: solid phase direct or indirect radioimmunoassay (RIA), solid phase direct or indirect enzyme immunoassay (EIA), sandwich competition assay (see Stahli et at, Methods in Enzymology 9:242-253 (1983)); solid phase direct biotin-avidin EIA (see Kirkland et ah, J. Immunol. 137:3614-3619 (1986)); solid phase direct labeled assay, solid phase direct labeled sandwich assay (see Harlow and Lane, Antibodies, A Laboratory Manual, Cold Spring Harbor Press (1988)); solid phase direct label RIA using 1-125 label {see Morel et al, Molec. Immunol. 25(1):7-15 (1988)); solid phase direct biotin-avidin EIA (Cheung et al, Virology 176:546-552 (1990)); and direct labeled RIA (Moldenhauer et al, Scand. J. Immunol 32:77-82 (1990)). Typically, such an assay involves the use of purified antigen bound to a solid surface or cells bearing either of these, an unlabelled test immunoglobulin and a labeled reference immunoglobulin. Competitive inhibition is measured by determining the amount of label bound to the solid surface or cells in the presence of the test immunoglobulin. Usually the test immunoglobulin is present in excess. Antibodies identified by competition assay (competing antibodies) include antibodies binding to the same epitope as the reference antibody and antibodies binding to an adjacent epitope sufficiently proximal to the epitope bound by the reference antibody for steric hindrance to occur. Usually, when a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to a common antigen by at least 50 or 75%.
[0032] The terms "conditioned culture medium" and "culture medium" refer to the aqueous extracellular fluid which surrounds cells grown in tissue culture (in vitro) and which contains, among other constituents, proteins and peptides secreted by the cells.
[0033] The term "body fluid" refers to those fluids of a mammalian host which is suspected contain measurable amounts of alpha synuclein or fragments thereof, specifically including blood, cerebrospinal fluid (CSF), urine, and peritoneal fluid. The term "blood" refers to whole blood, as well as blood plasma and serum.
[0034] A "pharmacological" activity means that an agent at least exhibits an activity in a screening system that indicates that the agent is or may be useful in the prophylaxis or treatment of a disease. The screening system can be in vitro, cellular, animal or human. Agents can be described as having pharmacological activity notwithstanding that further testing may be required to establish actual prophylactic or therapeutic utility in treatment of a disease.
[0035] An end-specific antibody means an antibody whose epitope includes a terminal residue of an antigen, and the antibody specifically binds to the antigen only when that residue has a free end. For example, an antibody that binds to the N-terminus of alpha synuclein when the alpha synuclein is not linked to any other protein but which does not specifically bind when the N-terminus of alpha synuclein is fused to another protein is an end-specific antibody.
[0036] A synucleinopathic disease means a disease characterized by Lewy bodies, Lewy neurites or other deposits of alpha synuclein.
An immunogenic fragment of alpha synuclein is one capable of inducing a humoral (antibody mediated) and/or a cellular (mediated by antigen-specific T cells or their secretion products) when administered to a subject.
[0037] Compositions or methods "comprising" one or more recited elements may include other elements not specifically recited. For example, a composition that comprises alpha-SN peptide encompasses both an isolated alpha-SN peptide and alpha-SN peptide as a component of a larger polypeptide sequence.
DETAILED DESCRIPTION
I. Antibodies of the Invention
[0038] The invention provides several exemplary monoclonal antibodies to different epitopes of alpha synuclein as shown in Table 1. Hybridomas producing the respective antibodies are also provided. The first column of the table shows the name of an antibody. The same name is used to refer to an antibody and the hybridoma producing it. The second column shows the antigen used to generate the antibody. The third column indicates the epitope bound by the antibody. The fourth column indicates the isotype. The fifth column indicates applications of the antibody (immunofluorescence, Western blot, immunoprecipitation, histology, and capture and reporter antibody for a sandwich assay). The invention further provides humanized and chimeric forms of mouse monoclonals, particularly of the exemplified antibodies shown in Table 1.
[0039] The invention also provides other antibodies that compete with one of the exemplified antibodies for specific binding to alpha-synuclein (i.e., bind to the same epitope as an exemplified antibody or a sufficiently proximal epitope to interfere with the binding of an exemplified antibody to its epitope). The invention also provides antibodies that bind to the same epitope as one of the exemplified antibodies. Antibodies that compete with or bind to the same epitope as an exemplified antibody are expected to show similar functional properties. Such antibodies include mouse and other nonhuman antibodies, human antibodies, chimerics and humanized antibodies. Such antibodies include monoclonal antibodies and polyclonal antibody. A polyclonal antibody specifically binds to an epitope if it binds to the epitope without binding to other regions of alpha synuclein. This is usually true of a monoclonal antibody as well. However, some monoclonal antibodies can be designed or selected to have specificity for two epitopes within alpha synuclein. [0040] Some preferred epitope specificities include monoclonal antibodies that specifically binds to a repeated epitope comprising residues within residues 43-51 and residues within residues 58-65, monoclonal antibodies that specifically binds to an epitope within residues 118-126; monoclonal antibodies that specifically binds to an epitope comprising residues 91- 99, monoclonal antibodies that specifically binds to an epitope comprising residues 40-55; monoclonal antibodies that specifically binds to an epitope comprising residues 124-134, wherein residue 129 is phosphorylated serine, and monoclonal antibodies that specifically binds to an epitope comprising residues 123-127, wherein residue 125 is nitrated tyrosine.
[0041] When an antibody is said to bind to an epitope within specified residues, such as alpha-SN 109-120 , for example, what is meant is that the antibody specifically binds to a polypeptide consisting of the specified residues {i.e., alpha-SN 109-120 in this an example). Such an antibody does not necessarily contact every residue within alpha-SN 109-120. Nor does every single amino acid substitution or deletion with in alpha-SN109-120 necessarily significantly affect binding affinity. Epitope specificity of an antibody can be determined, for example, by testing a collection of overlapping peptides of about 15 amino acids spanning the sequence of alpha-synuclein and differing in increments of a small number of amino acids (e.g., 3 amino acids). The peptides are immobilized within the wells of a microtiter dish. Immobilization can be effected by biotinylating one terminus of the peptides. Optionally, different samples of the same peptide can be biotinylated at the N and C terminus and immobilized in separate wells for purposes of comparison. Such is particularly useful for identifying end-specific antibodies. Optionally, additional peptides can be included terminating at a particular amino acid of interest (e.g., the first and last residue of the NAC fragment). Such is particularly useful for identifying end-specific antibodies to internal fragments of alpha synuclein. An antibody is screened for specific binding to each of the various peptides. The epitope is defined as occurring within a segment of amino acids that is common to all peptides to which the antibody shows specific binding.
i. General Characteristics of Immunoglobulins [0042] The basic antibody structural unit is known to comprise a tetramer of subunits. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa) and one "heavy" chain (about 50-70 IcDa). The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
[0043] Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's isotype as IgG, IgM, IgA, IgD and IgE, respectively. Within light and heavy chains, the variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D" region of about 10 more amino acids. (See generally, Fundamental Immunology, Paul, W., ed., 2nd ed. Raven Press, N. Y., 1989, Ch. 7 (incorporated by reference in its entirety for all purposes).
[0044] The variable regions of each light/heavy chain pair form the antibody binding site. Thus, an intact antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same. The chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. The CDRs from the two chains of each pair are aligned by the framework regions, enabling binding to a specific epitope. From N-terminal to C-terminal, both light and heavy chains comprise the domains FRl, CDRl, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is in accordance with the definitions of Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD, 1987 and 1991); Chothia & Lesk, J. MoI. Biol. 196:901-917 (1987); or Chothia et ah, Nature 342:878-883 (1989).
ii. Production of Nonhuman Antibodies
[0045] Chimeric and humanized antibodies have the same or similar binding specificity and affinity as a mouse or other nonhuman antibody that provides the starting material for construction of a chimeric or humanized antibody. Chimeric antibodies are antibodies whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin gene segments belonging to different species. For example, DNA encoding the variable domains of a mouse antibody can be sequenced, and DNA construct(s) encoding the variable domains joined to human constant (C) segments, such as IgGl and IgG4 constructed. The constructs are then expressed to produce the antibody Human isotype IgGl is preferred. In some methods, the isotype of the antibody is human IgGl. IgM antibodies can also be used in some methods. A typical chimeric antibody is thus a hybrid protein consisting of the V or antigen-binding domain from a mouse antibody and the C or effector domain from a human antibody.
[0046] Humanized antibodies have variable region framework residues substantially from a human antibody or consensus of human antibodies (termed an acceptor antibody) and some and usually all six complementarity determining regions substantially or entirely from a mouse-antibody, (referred to as the donor immunoglobulin). See, Queen et ah, Proc. Natl. Acad. ScL USA 86:10029-10033 (1989), WO 90/07861, US 5,693,762, US 5,693,761, US 5,585,089, US 5,530,101, and Winter, US 5,225,539 (each of which is incorporated by reference in its entirety for all purposes). The constant region(s), if present, are also substantially or entirely from a human immunoglobulin. The human variable domains are usually chosen from human antibodies whose framework sequences exhibit a high degree of sequence identity with the murine variable region domains from which the CDRs were derived. The heavy and light chain variable region framework residues can be derived from the same or different human antibody sequences. The human antibody sequences can be the sequences of naturally occurring human antibodies or can be consensus sequences of several human antibodies. See Carter et al, WO 92/22653. Certain amino acids from the human variable region framework residues are selected for substitution based on their possible influence on CDR conformation and/or binding to antigen. Investigation of such possible influences is by modeling, examination of the characteristics of the amino acids at particular locations, or empirical observation of the effects of substitution or mutagenesis of particular amino acids.
[0047] For example, when an amino acid differs between a murine variable region framework residue and a selected human variable region framework residue, the human framework amino acid should usually be substituted by the equivalent framework amino acid from the mouse antibody when it is reasonably expected that the amino acid:
( 1 ) nonco valently binds antigen directly,
(2) is adjacent to a CDR region, (3) otherwise interacts with a CDR region (e.g. is within about 6 A of a CDR region), or
(4) participates in the VL-VH interface.
[0048] Other candidates for substitution are acceptor human framework amino acids that are unusual for a human immunoglobulin at that position. These amino acids can be substituted with amino acids from the equivalent position of the mouse donor antibody or from the equivalent positions of more typical human immunoglobulins. Other candidates for substitution are acceptor human framework amino acids that are unusual for a human immunoglobulin at that position. The variable region frameworks of humanized immunoglobulins usually show at least 85% sequence identity to a human variable region framework sequence or consensus of such sequences.
iii. Human Antibodies
[0049] Human antibodies against alpha-SN are provided by a variety of techniques described below. Some human antibodies are selected by competitive binding experiments, or otherwise, to have the same epitope specificity as a particular mouse antibody, such as one of the mouse monoclonals shown in Table 1. Human antibodies can also be screened for a particular epitope specificity by using only a fragment of alpha-SN as the immunogen, and/or by screening antibodies against a collection of deletion mutants of alpha-SN. Human antibodies preferably have isotype specificity human IgGl. Several methods are available for producing human antibodies including the trioma method, Oestberg et al, Hybridoma 2:361- 367 (1983); Oestberg, US Patent No. 4,634,664; and Engleman et al, US Patent 4,634,666 (each of which is incorporated by reference in its entirety for all purposes); transgenic non- human mammals described in detail by, e.g., Lonberg et al, WO93/1222, US 5,877,397, US 5,874,299, US 5,814,318, US 5,789,650, US 5,770,429, US 5,661,016, US 5,633,425, US 5,625,126, US 5,569,825, US 5,545,806, Nature 148, 1547-1553 (1994), Nature Biotechnology 14, 826 (1996), Kucherlapati, WO 91/10741(each of which is incorporated by reference in its entirety for all purposes); and phage display methods See, e.g., Dower et al, WO 91/17271 and McCafferty et al, WO 92/01047, US 5,877,218, US 5,871,907, US 5,858,657, US 5,837,242, US 5,733,743 and US 5,565,332 (each of which is incorporated by reference in its entirety for all purposes).
iv. Selection of Constant Region [0050] The heavy and light chain variable regions of chimeric, humanized, or human antibodies can be linked to at least a portion of a human constant region. The choice of constant region depends, in part, whether antibody-dependent complement and/or cellular mediated toxicity is desired. For example, isotopes IgGl and IgG3 have complement activity and isotypes IgG2 and IgG4 do not. Choice of isotype can also affect passage of antibody into the brain. Human isotype IgGl is preferred. Light chain constant regions can be lambda or kappa. Antibodies can be expressed as tetramers containing two light and two heavy chains, as separate heavy chains, light chains, as Fab, Fab' F(ab')2, and Fv, or as single chain antibodies in which heavy and light chain variable domains are linked through a spacer.
v. Expression of Recombinant Antibodies
[0051] Chimeric, humanized and human antibodies are typically produced by recombinant expression. Recombinant polynucleotide constructs typically include an expression control sequence operably linked to the coding sequences of antibody chains, including naturally associated or heterologous promoter regions. Preferably, the expression control sequences are eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and the collection and purification of the crossreacting antibodies.
[0052] These expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA. Commonly, expression vectors contain selection markers, e.g., ampicillin-resistance or hygromycin-resistance, to permit detection of those cells transformed with the desired DNA sequences.
[0053] E. coli is one prokaryotic host particularly useful for cloning the DNA sequences of the present invention. Microbes, such as yeast are also useful for expression. Saccharomyces is a preferred yeast host, with suitable vectors having expression control sequences, an origin of replication, termination sequences and the like as desired. Typical promoters include 3- phosphoglycerate kinase and other glycolytic enzymes. Inducible yeast promoters include, among others, promoters from alcohol dehydrogenase, isocytochrome C, and enzymes responsible for maltose and galactose utilization.
[0054] Mammalian cells are a preferred host for expressing nucleotide segments encoding immunoglobulins or fragments thereof. See Winnacker, From Genes to Clones, (VCH Publishers, NY, 1987). A number of suitable host cell lines capable of secreting intact heterologous proteins have been developed in the art, and include CHO cell lines, various COS cell lines, HeLa cells, L cells, human embryonic kidney cell, and myeloma cell lines. Preferably, the cells are nonhuman. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer (Queen et al, Immunol. Rev. 89:49 (1986)), and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Preferred expression control sequences are promoters derived from endogenous genes, cytomegalovirus, S V40, adenovirus, bovine papillomavirus, and the like. See Co et al, J. Immunol. 148:1149 (1992).
[0055] Alternatively, antibody coding sequences can be incorporated in transgenes for introduction into the genome of a transgenic animal and subsequent expression in the milk of the transgenic animal (see, e.g., US 5,741,957, US 5,304,489, US 5,849,992). Suitable transgenes include coding sequences for light and/or heavy chains in operable linkage with a promoter and enhancer from a mammary gland specific gene, such as casein or beta lactoglobulin.
[0056] The vectors containing the DNA segments of interest can be transferred into the host cell by well-known methods, depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment, electroporation, lipofection, biolistics or viral-based transfection can be used for other cellular hosts. Other methods used to transform mammalian cells include the use of polybrene, protoplast fusion, liposomes, electroporation, and microinjection (see generally, Sambrook et al, supra). For production of transgenic animals, transgenes can be microinjected into fertilized oocytes, or can be incorporated into the genome of embryonic stem cells, and the nuclei of such cells transferred into enucleated oocytes.
[0057] Once expressed, antibodies can be purified according to standard procedures of the art, including HPLC purification, column chromatography, and gel electrophoresis and the like (see generally, Scopes, Protein Purification (Springer- Verlag, NY, 1982).
II. Alpha Svnuclein
[0058] Alpha synuclein was originally identified in human brains as the precursor protein of the non-|8-amyloid component of (NAC) of AD plaques. (Ueda et al., Proc. Natl. Acad. Sd. U.S.A. 90 (23): 11282-11286 (1993). Alpha-SN, also termed the precursor of the non- A/3 component of AD amyloid (NACP), is a peptide of 140 amino acids. Alpha-SN has the amino acid sequence:
MDVFMKGLSKAKEGVVAAAEKTKQGVAEAAGKTKEGVLYVGSKTK EGVVHGVATVAEKTKEQVTNVGGAWTGVTAVAQKTVEGAGSIAAATGFVKKDQ LGKNEEGAPQEGILEDMPVDPDNEAYEMPSEEGYQDYEPEA (SEQ ID NO: 1)
(Ueda et al., Ibid.; GenBank accession number: P37840).
[0059] The non-A/3 component of AD amyloid (NAC) is derived from alpha-SN. NAC, a highly hydrophobic domain within alpha synuclein, is a peptide consisting of at least 28 amino acids residues (residues 60-87) (SEQ ID NO: 3) and optionally 35 amino acid residues (residues 61-95) (SEQ ID NO: 2). See Fig. 1. NAC displays a tendency to form a beta-sheet structure (Iwai, et al, Biochemistry, 34:10139-10145). Jensen et al. have reported NAC has the amino acid sequence:
EQVTNVGGAVVTGVTAVAQKTVEGAGSIAAATGFV (SEQ ID NO: 2) (Jensen et al., Biochem. J. 310 (Pt 1): 91-94 (1995); GenBank accession number S56746).
[0060] Ueda et al. have reported NAC has the acid sequence:
KEQVTNVGGAWTGVTAVAQKTVEGAGS (SEQ ID NO: 3)
(Ueda et al., PNAS USA 90:11282-11286 (1993).
[0061] Disaggregated alpha-SN or fragments thereof, including NAC, means monomelic peptide units. Disaggregated alpha-SN or fragments thereof are generally soluble, and are capable of self-aggregating to form soluble oligomers. Oligomers of alpha-SN and fragments thereof are usually soluble and exist predominantly as alpha-helices. Monomelic alpha-SN may be prepared in vitro by dissolving lyophilized peptide in neat DMSO with sonication. The resulting solution is centrifuged to remove any insoluble particulates. Aggregated alpha- SN or fragments thereof, including NAC, means oligomers of alpha-SN or fragments thereof which have associate into insoluble beta-sheet assemblies. Aggregated alpha-SN or fragments thereof, including NAC, means also means fibrillar polymers. Fibrils are usually insoluble. Some antibodies bind either soluble alpha-SN or fragments thereof or aggregated alpha-SN or fragments thereof. Some antibodies bind both soluble and aggregated alpha-SN or fragments thereof. [0062] Unless otherwise indicated, reference to alpha-SN means the natural human amino acid sequence indicated above as well as natural allelic and species variants thereof, including full-length forms and immunogenic fragments thereof, as well as forms having undergone posttranslational modification, such as phosphorylation. Induced variants of alpha-SN or can also be used but are not preferred. Such variants typically differ from naturally occurring peptides at one, two or a few positions, often by virtue of conservative substitutions. Analogs can also unnatural amino acids, but such is not preferred.
[0063] Alpha-SN, its fragments, and analogs can be synthesized by solid phase peptide synthesis or recombinant expression, or can be obtained from natural sources. Automatic peptide synthesizers are commercially available from numerous suppliers, such as Applied Biosystems, Foster City, California. Recombinant expression can be in bacteria, such as E. coli, yeast, insect cells or mammalian cells. Procedures for recombinant expression are described by Sambrook et ah, Molecular Cloning: A Laboratory Manual (C.S.H.P. Press, NY 2d ed., 1989). Some forms of alpha-SN peptide are also available commercially, for example, at BACHEM and American Peptide Company, Inc.
[0064] Beta synuclein shows 78% sequence similarity with alpha synuclein at the amino acid level. Gamma synuclein shares 60% similarity at the amino acid level with alpha- synuclein (see Biere et al., J. Biol. Chem., Vol. 275, Issue 44, 34574-34579, November 3, 2000, incorporated by reference).
III. Assays for Detecting Alpha Synuclein
[0065] Antibodies of the invention can be used to detect alpha synuclein or fragments thereof in a variety of formats including immunoprecipitation, Western blotting, ELISA, radioimmunoassay, competitive and isometric assays. See Harlow & Lane, Antibodies^ A Laboratory Manual (CSHP NY, 1988); U.S. Patent Nos. 3,791,932; 3,839,153; 3,850,752; 3,879,262;4,034,074, 3,791,932; 3,817,837; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; and 4,098,876.
[0066] Isometric or sandwich assays are a preferred format (see US 4,376,110, 4,486,530, 5,914,241, and 5,965,375). Such assays use one antibody immobilized to a solid phase (capture antibody), and another antibody in solution (reporter antibody). Typically, the reporter antibody is labeled, either directly or via a secondary labeling reagent, such as an anti-idiotypic antibody. The capture and reporter antibodies having different binding specificities so both can bind to alpha-synuclein or a fragment thereof at the same time. Capture and reporter antibodies can be contacted with target antigen in either order or simultaneously. If the capture antibody is contacted first, the assay is referred to as being a forward assay. Conversely, if the reporter antibody is contacted first, the assay is referred to as being a reverse assay. If target is contacted with both antibodies simultaneously, the assay is referred to as a simultaneous assay. After contacting the target with antibody, a sample is incubated for a period that usually varies from about 10 min to about 24 hr and is usually about 1 hr. A wash step is then performed to remove components of the sample that do not become specifically bound to the solid phase When capture and reporter antibodies are bound in separate steps, a wash can be performed after either or both binding steps. After washing, binding is quantified, typically by detecting label linked to the solid phase through binding of labeled reporter. Usually for a given pair of antibodies, a calibration curve is prepared from samples containing known concentrations of target antigen. Concentrations of antigen in samples being tested are then read by interpolation from the calibration curve. Analyte can be measured either from the amount of labeled reporter antibody bound at equilibrium or by kinetic measurements of bound labeled solution antibody at a series of time points before equilibrium is reached. The slope of such a curve is a measure of the concentration of target in a sample. Alternatively, the amount of alpha synuclein in a sample can be determined by comparing the signal of reporter antibody bound to alpha synuclein the sample with the signal from reporter antibody bound to a known amount of alpha synuclein in a control sample.
[0067] Competitive assays can also be used, hi some methods, target alpha-synuclein in a sample competes with exogenously supplied labeled alpha synuclein for binding to an antibody. The amount of labeled alpha synuclein bound to the antibody is inversely proportional to the amount of target alpha synuclein in the sample. The antibody can be immobilized to facilitate separation of the bound complex from the sample prior to detection (heterogeneous assays) or separation may be unnecessary as practiced in homogeneous assay formats. In other methods, the antibody used as a detection reagent is labeled. When the antibody is labeled, its binding sites compete for binding to the target alpha synuclein in the sample and exogenously supplied form of alpha synuclein immobilized on a solid phase.
[0068] Suitable detectable labels for use in the above methods include any moiety that is detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical, chemical, or other means. For example, suitable labels include biotin for staining with labeled streptavidin conjugate, fluorescent dyes (e.g., fluorescein, Texas red, rhodamine, green fluorescent protein, and the like), radiolabels (e.g., . 3H, 1251, 35S, 14C, or 32P), enzymes (e.g., horseradish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex beads). Patents that described the use of such labels include U.S. Pat. Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241. See also Handbook of Fluorescent Probes and Research Chemicals (6th Ed., Molecular Probes, Inc., Eugene Oreg.). Radiolabels can be detected using photographic film or scintillation counters, fluorescent markers can be detected using a photodetector to detect emitted light. Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting the reaction product produced by the action of the enzyme on the substrate, and colorimetric labels are detected by simply visualizing the colored label.
[0069] Suitable supports for use in the above methods include, for example, nitrocellulose membranes, nylon membranes, and derivatized nylon membranes, and also particles, such as agarose, a dextran-based gel, dipsticks, particulates, microspheres, magnetic particles, test tubes, microtiter wells, SEPHADEX™. (Amersham Pharmacia Biotech, Piscataway NJ., and the like. Immobilization can be by absorption or by covalent attachment. Optionally, antibodies can be joined to a linker molecule, such as biotin for attachment to a surface.
[0070] Solvents used to extract alpha synuclein from tissue samples can decrease the sensitivity of the assay (e.g., 5M guanidine, urea/thiourea/CHAPS, urea/thiourea, 1% SDS, 1%SDS/8M, cell lysis buffer). It is recommended that such solvents be removed or diluted such that they account for less than 1% and preferably less than 0.1% of the buffer used for the assay.
[0071] Preferred pairs of monoclonal antibodies for use in isometric assays are as follows. In one combination, the capture antibody specifically binds an epitope within residues selected from the group consisting of N-terminus, 40-55, 91-99 and 118-126, and the reporter antibody specifically binds an epitope within residues 109-120. Another suitable pairing is an antibody that specifically binds to an epitope within residues 124-134 and is specific for the phosphorylated form and another antibody that specifically binds to an epitope within residues 91-99. Other suitable pairings include an antibody that competes with 6H7 and an antibody that competes with 5C12 or 12Cl; an antibody that competes with 3A12 and an antibody that competes with 5Cl 2 or 6H7; an antibody that competes with 12Cl and an antibody that competes with 5Cl 2 or 6H7; an antibody that competes with 9A6 and an antibody that competes with 5C12, 6H7 or 12Cl; an antibody that competes with 9G5 and an antibody that competes with 5C12, 6H7 or 12Cl;an antibody that competes with 9E4 and an antibody that competes with 5C12, 6H7 or 12Cl; an antibody that competes with 1H7 and an antibody that competes with 5C12, 6H7 or 12Cl; and an antibody that competes with 10G5 and an antibody that competes with 5C12, 6H7 or 12Cl, and antibody that competes with 1H7 and an antibody that competes with 11 A5. Preferred pairings include 6H7 and 5C12 or 12Cl; 3A12 and 5C12 or 6H7; 12Cl and 5C12 or 6H7;9A6 and 5C12, 6H7 or 12Cl; 9G5 and 5C12, 6H7 or 12Cl; 9E4 and 5C12, 6H7 or 12Cl; 1H7 and 5C12, 6H7 or 12Cl; 10G5 and 5C12, 6H7 or 12Cl, 1H7 and 11A5.
[0072] Some method employ either a capture or a reporter antibody that is end-specific for a fragment of alpha-synuclein, particularly, SNl-119 (i.e., residues 1-119 of SEQ ID NO:1) or SN1-122 (residues 1-122 of SEQ ID NO:1). Applications for detection of these fragments are described below.
IV. Applications
A. Body fluids
[0073] In vivo detection of alpha synuclein in patient samples can be used for diagnosing and monitoring diseases characterized by Lewy bodies or other deposits of alpha synuclein. Synucleinopathic diseases include Parkinson's disease (PD), dementia with Lewy bodies (DLB), the Lewy body variant of Alzheimer's disease (LBVAD), multiple systems atrophy (MSA), neurodegeneration with brain iron accumulation type-1 (NBIA-I), diffuse Lewy body disease (DLBD), and combined PD and Alzheimer's disease (AD). Suitable patient samples include body fluids, such as blood, CSF, urine, and peritoneal fluid. The presence of a synucleinopathic disease is generally associated with significantly altered levels of alpha synuclein or fragments thereof in the fluid (increased or decreased) when compared to the mean values in normal individuals, i.e., individuals not suffering from a synucleinopathic disease. A level is significantly altered if it departs by more than one standard deviation from the mean level in a population of normal individuals.
[0074] In addition to initial diagnosis of synucleinopathic disease, condition, the measured concentrations of alpha synuclein and its fragments can be monitored to follow the progress of the disease, and potentially follow the effectiveness of treatment. Levels of alpha- synuclein and its fragments revert toward the mean in a population of normal individuals if the treatment regime is effective.
B. Cell culture
[0075] In vitro monitoring of alpha synculein and its fragment in conditioned culture medium from a suitable cell culture can be used for analyzing processing and secretion of alpha- synuclein and the effect of potential agents on the same. Monitoring processing of alpha synuclein provides a means to study proteolytic cleavage, identify responsible proteases, determine what factors affect cleavage, and determine the physiological function of truncated forms of alpha synuclein. Agents that inhibit processing and/or secretion of alpha synuclein have pharmacological activity potentially useful for prophylaxis of synucleinopathic disease. Typically, inhibitory activity is determined by comparing levels of alpha synuclein and/or its fragments in medium from a cell treated with a test agent versus a comparable control cell not treated with the agent. As discussed in copending application USSN 60/471,929 filed May 19, 2003, USSN 10/850,570 filed May 19, 2004 and attorney docket 015270-011520US filed October 19, 2004, which is a CIP thereof, certain fragments of alpha synuclein with C- terminal truncations preferentially accumulate in Lewy bodies (e.g., SNl-119 and SN1-122). For example, media from PeakS cells transfected with alpha synuclein contain two prominent truncated species of about 12 and 7 kDa. Agents inhibiting processing reactions that form such fragments therefore have pharmacological activity useful for treating diseases characterized by Lewy bodies. The NAC fragment of alpha synuclein accumulates in extracellular amyloid deposits in Alzheimer's disease. Agents that inhibit formation or secretion of NAC therefore have pharmacological activity useful for treating Alzheimer's disease.
[0076] Suitable cells include cells transfected with nucleic acids encoding alpha synuclein, preferably, human alpha synuclein and cells naturally expressing alpha synuclein, also preferably human. The alpha synuclein in transfected cells can bear a mutation, such as S129A, S129D, A53T and A20P. Cells include PeakS cells, SY5Y cells,human cortical cells, human neuroglioma cell lines, human HeLa cells, primary human endothelial cells (e.g. HUVEC cells), primary human fibroblasts or lymphoblasts, primary human mixed brain cells (including neurons, astrocytes, and neuroglia), Chinese hamster ovary (CHO) cells, and the like. SY5Y cells are neuronal cells that can be induced to differentiate by treatment with retinoic acid/BDNF (brain derived neurotrophic factor). Transfected cells expressing alpha synuclein at higher levels than normal human cells are preferred. In some such cells, the alpha synuclein bears a mutation associated with synucleinopathic disease. [0077] Random libraries of peptides or other compounds can also be screened for suitability. Combinatorial libraries can be produced for many types of compounds that can be synthesized in a step-by-step fashion. Such compounds include polypeptides, beta-turn mimetics, polysaccharides, phospholipids, hormones, prostaglandins, steroids, aromatic compounds, heterocyclic compounds, benzodiazepines, oligomeric N-substituted glycines and oligocarbamates. Large combinatorial libraries of the compounds can be constructed by the encoded synthetic libraries (ESL) method described in Affymax, WO 95/12608, Affymax, WO 93/06121, Columbia University, WO 94/08051, Pharmacopeia, WO 95/35503 and Scripps, WO 95/30642 (each of which is incorporated herein by reference for all purposes). Peptide libraries can also be generated by phage display methods. See, e.g., Devlin, WO 91/18980. The test compounds are typically administered to the culture medium at a concentration in the range from about 1 nM to 1 mM, usually from about 10 μM to 1 niM. Test compounds which are able to inhibit formation, processing or secretion of alpha synuclein are candidates for further determinations in transgenic animals and eventually human clinical trials.
C. Transgenic Animals
[0078] The antibodies of the invention and assays for detecting them can also be used to monitor alpha synuclein production, and processing in transgenic animal models of disease. Transgenic animal models of Lewy body disease are described by Masliah, et al. Science 287:1265-1269 (2000); Masliah et al, PNAS USA 98:12245-12250 (2001). Alpha synuclein can be analyzed either in body fluids as described above for human samples, or in tissue samples taken directly from the animal (see copending 60/423,012, filed November 1, 2002, incorporated by reference). Tissue samples can be classified as Lewy body, particulate fraction and soluble fractions. Simple assays can be performed as for cell culture to screen agents for capacity to inhibit formation of alpha synuclein or its processing to fragments. Typically, the inhibitory activity is determined by comparing the level of alpha synuclein or a fragment thereof in a particularly body fluid or fraction from a tissue sample from a transgenic animal treated with the agent in comparison with the level of alpha synuclein or the fragment in the same body fluid or fraction in a control transgenic animal not treated with the agent. Inhibitory activity is shown by decreased levels of alpha synuclein of fragment thereof in the treated animal relative to the control.
[0079] Tissue samples from the brains of human patients can be subject to similar analyses. However, as obtaining samples from the brains of patient is an undesirably invasive procedure, such, analyses are usually confined to cadavers. The analyses are useful, for example, in identifying fragments of alpha synuclein that preferentially accumulate in Lewy bodies, as described in copending application 60/471, 929, filed 05/19/2003 and its progeny supra. V. Kits
[0080] The invention further provides kits including one or more antibodies of the invention. In some kits the antibodies are preimmobilized to a solid phase. For example, several different antibodies can be immobilized to the wells of a microtiter dish. Optionally, labeling reagents, such as an antiidiotypic antibody are also included in the kits. The labeling may also include a chart or other correspondence regime correlating levels of measured label with levels of antibodies to alpha-SN. The term labeling refers to any written or recorded material that is attached to, or otherwise accompanies a kit at any time during its manufacture, transport, sale or use. For example, the term labeling encompasses advertising leaflets and brochures, packaging materials, instructions, audio or video cassettes, computer discs, as well as writing imprinted directly on kits. The kits can be sold, for example, as research or diagnostic reagents.
[0081] Although the invention has been described in detail for purposes of clarity of understanding, it will be obvious that certain modifications may be practiced within the scope of the appended claims. AU publications and patent documents cited in this application are hereby incorporated by reference in their entirety for all purposes to the same extent as if each were so individually denoted.
EXAMPLES
I. Antibodies of the Invention
TABLE l
Figure imgf000026_0001
Abbreviations: IP (immunoprecipitation); H (histology); EC and ER (ELISA capture and reporter antibody); WB (western blot)., IF immunofluorescence.
[0082] The antibodies were produced by immunizing with the antigen shown. Hybridomas were produced by standard methods, and tested for appropriate binding specificity.
II. ELISA Assays
[0083] Capture antibodies were biotinylation by a modification of the Igen commercial method. Pierce sulfo NHS biotin was prepared at 4 mg/ml in H2O and added at a 40 M excess to 1 mg/ml antibody in 150 mM KPO4 pH 7.8. This was incubated with gentle mixing in the dark for two hours and then dialyzed against PBS to remove free biotin. Capture antibodies were diluted to 10 μg/ml in Well Coating Buffer 0.1 M PO4. pH 8.5. Plates were coated 100 μl per well in Dynx 4HBX plates overnight at room temperature. Plates were then blocked in 0.25% Casein/PBS.
[0084] Standard curves of synuclein were prepared either in Specimen Diluent- (0.6% BSA, 0.05% Triton 405, 0.5% Thimerisol in 0.1 M PO4, .15 M NaCl pH 7.4) or 0.5 M guanidine, 0.25% casein/ protease inhibitor/ PBS depending on the samples to be assayed. Samples were diluted in either Specimen Diluent or the guanidine/casein depending on sample. Standards and samples were incubated over night at 4°C. For simple samples, room temperature and a few hours are sufficient. Plates were washed 4 times.
[0085] Plates were then incubated with the appropriate biotinylated antibody at 2 μg/ml in Specimen Diluent for 1 hour. Plates were washed 4 times. Plates were then incubated with Vector Strepavidin HRP 1/5000 or Amersham Strepavidin HRP at 1/10000 in Specimen Diluent at 100 μl/well for 1 hour. Plates were washed 4 times. BioFx TMB was added 100 μl/well and incubated 5 minutes at room temperature. The reaction was stopped with 100 μl BioFx stop and read on a Spectromax plate reader at 450 nm. Samples were assayed from the standard curve using the machine software and a 4 parameter curve fit.
[0086] ELISA assays were developed for the measurement of synuclein from various sources included human and rodent brain, cell culture and Lewy Body Preps. The follow table describes the sensitivity of various capture antibodies when 5Cl 2 biotin is used as the reporter
TABLE 2
Figure imgf000027_0001
[0087] 1H17/5C12 was shown to have the best sensitivity in the above assays. 1H7 was then used as a plate coat to investigate if any other antibodies used as a reporter offered any increases in sensitivity. Table 3 describes the results.
TABLE 3
Figure imgf000028_0001
[0088] 9A6 and 9G5 appeared to fail as a reporter due to cross blocking of the capture and reporter. The failure of 3Al 2 is due to poor biotinylation of the antibody and can be revisited if needed. [0089] The cell line designated JHl 7.6H7.1.54.28 producing the antibody 6H7 has the
ATCC accession number having been deposited under the provisions of the
Budapest Treaty with the American Type Culture Collection (ATCC, Manassas, VA 20108) on August 4, 2005. The cell line designated JH4.8A5.25.7.36 producing the antibody 8A5 has the ATCC accession number having been deposited on August 4, 2005.
[0090] From these results, the combination of 1H7 capture and 5C12 reporter for total synuclein in human samples is recommended. Although the 5C12 antibody binds to both mouse and human alpha synuclein, this is not a problem in human samples because no mouse synuclein is present. The combination of 1H7 capture 9E4 reporter for detecting human alpha synuclein in transgenic mouse brains is recommended. 9E4 is specific to human alpha synuclein. Both formats are specific for alpha-synuclein. The combination of 1H7 capture and 8A5 reporter is recommended for measurement of alpha synuclein having an intact C- terminus. The combination of 11 A5 capture and 5C12 reporter is suitable for detecting alpha synuclein phosphorylated at residue 129. The combination of 1H7 as capture antibody and 11 A5 as reporter offers about ten-fold greater sensitivity. The 11 A5 antibody is specific for alpha synuclein phosphorylated at residue 129.
[0091] A calibration curve showing OD450 vs. concentration of alpha synuclein using 1H7 and 5Cl 2 as capture and reporter antibody is shown in Fig. 2. The assay is more sensitive in the absence of guanidine. A calibration curve for 1H7/9E4 in the presence of 0.5 M guanidine buffer is shown in Fig. 3. The guanidine maintains synuclein in a mildly denatured state making epitopes available for antibody binding and decreasing interactions with other proteins. Fig. 4 shows a calibration curve for the combination of 11 A5/5C12 for detection of phosphorylated alpha synuclein. The actual sensitivity is about twenty fold greater than the measure value of 9 ng/ml because the alpha synuclein was only about 6% phosphorylated. [0092] The capacity of the ELISA assay to quantify synuclein in a sample was testing by spiking brain homogenates from nontransgenic mice with known amounts of alpha synuclein. Fig. IA shows the results using 1H7/5C12 as capture and reporter antibodies. The assay detects 96% of the added alpha synuclein. Some synuclein is detected in the control because the 5C12 antibody binds to both mouse and human alpha synuclein. Fig. IB shows the results using 1H7 and 9E4 as capture and reporter antibody, hi this case, no alpha synuclein is detected in the control because 9E4 binds to human alpha synuclein only. 92% of the added alpha synuclein was detected in the spiked sample, showing that detection is quantitative.
III. α-Synuclein Detection in Samples
[0093] The ELISA assay was used to detect alpha synuclein in soluble, particulate and Lewy body preparations from DLBD patients and controls. A brain homogenate was spun at 100Og. The pellet was the source of Lewy bodies. These were prepared by Percoll gradient density fractionation, DNA digestion, and immunoaffinity purification using antibodies tagged with magnetic beads. Optionally, the alpha synuclein content of Lewy bodies can be enriched by stripping with the anionic detergent sarkosyl. The soluble and particulate fractions were prepared by spinning the initial supernatant at 150,00Og. The resulting supernatant was referred to as a soluble fraction, and the pellet as the particulate fraction. Synuclein can be extracted from these fractions using 0.5 and 5 M guanidine respectively. Note that the Lewy body samples were in UTC and that the soluble brain samples were prepared in Tris-buffered sucrose, and were assayed at a dilution in which these buffers did not interfere with the ELISA.
[0094] The soluble extract of the 8 human brains assayed contain approximately 0.03% of α- synuclein. There is a trend toward decreasing amounts of oc-synuclein in the soluble brain fraction of a diseased patient. (Table 4). There is a statistically significant increase in the amount of synuclein in the particulate fraction of the DLBD brain (Table 5). The increased level of synuclein may be due to immature Lewy bodies that are to small to pellet with mature Lewy bodies in the initial centrirugation.
[0095] The amount of α-synuclein in the Lewy body prep varies. According to the 1H7/5C12 ELISA, synuclein comprises at most 0.07% of the total Lewy body protein. The 1H7/9E4 ELISA measures approximately 60% less α-synuclein in the Lewy bodies than the 1H7/5C12 ELISA . The 9E4 reporter antibody recognizes an epitope more C-terminal than that of 5 C 12, and therefore the assay may simply reflect the presence of C-terminal truncations in the Lewy bodies as seen on 2-D gels. In this case loss of 9E4 reactivity or a lowered ratio of 9E4/5C12 activity (or other antibodies with the same or similar epitopes) is a marker of pathogenicity. Indeed, the 9E4 epitope encompasses the estimated cleavage sites. The soluble fraction contains less truncated synuclein and did show only a 12% difference between the ELISA assays consistent with this interpretation. An alternate explanation is that 9E4's epitope is modified or masked (perhaps due to the aggregation as described above or modification of tyrosine at 125) and cannot be detected.
TABLE 4
Soluble fraction
Contr
DLB
Figure imgf000031_0001
TABLE 5
(α-Synuclein ELISA Results From Lewy Bodies
contr
DLB
Figure imgf000032_0001
TABLE 6: Alpha synuclein in Lewy bodies
Figure imgf000032_0002
Detection of α-Synuclein in Blood
[0096] Blood samples from four healthy volunteers were collected into both EDTA- and heparin-containing tubes and plasma samples were prepared. The plasma was initially diluted 2-fold with IM guanidine for a final concentration of 0.5 M guanidine. Samples were assayed in 1H7/5C12 ELISA assay using the standard buffer of 0.25% casein, 0.5 M guanidine, and protease inhibitors plus 300 μg/ml mouse IgG (added to prevent interference from a human anti-mouse response that can be quite common in patient samples).
[0097] Plasma from the EDTA collection gave excellent spike and recovery of α-synuclein.
The amount of synuclein varied almost 2-fold between patients. The amount of synuclein in the heparin collected plasma was up to 10-fold lower than that of the EDTA collected plasma.
Not only was the total α-synuclein concentration lower but so was the spike and recovery indicating that heparin interferes with the assay possibly by binding to synuclein. In all further experiments, EDTA collection tubes were used.
[0098] The presence of synuclein in the plasma indicates that it is produced in cells from the blood. Secretion is unlikely because synuclein is not a secreted protein. Release during cell breakage is more likely. Which cells in the blood, i.e. white blood cells (WBC), red blood cells (RBC), and platelets produce synuclein was investigated. To prepare WBC, whole blood was spun down at 1000 X g and the plasma removed. The packed cell pellet was washed two times at 300 X g to remove any platelet contamination. The RBC were lysed in de-ionized water for 1 minute and the sample was readjusted to IX PBS. There was a small amount of RBC contamination. The resulting pellet was homogenized in 5 M guanidine, diluted 1/10 in assay buffer, treated with DNAse, and then spun in the microfuge to remove insoluble material.
[0099] hi an alternate fractionation, blood was processed to generate total blood cells, platelets and plasma. The blood was spun at 350 X g to pellet cells but not platelets. The plasma was removed and spun at 1000 X g to remove platelets. The cell pellet was washed two times at 350 X g to remove any remaining platelets. Both plasma and cells were initially diluted/homogenized in 5 M guanidine. The cell pellet was further diluted 1:10, treated with DNAse and spun to remove insoluble material. To test the synuclein specificity of the ELISA, an aliquot of the platelet and the total blood cell samples were absorbed with the synuclein antibody 1H7 coupled to Sepharose.
[0100] The amount of synuclein measured in whole blood is too large to be accounted for by platelets or by WBC as previously shown, thus strongly suggesting that RBC, the most abundant cells in the blood, are the major source of synuclein in blood. [0101] All samples (except plasma) were assayed for spike and recovery and well as % synuclein absorbable with 1H7 resin. In addition, all samples were run in the phospho- synuclein assay.
TABLE 7
Figure imgf000033_0001
[0102] The results confirm that low levels of synuclein are present in plasma and that low levels are also present in the cellular components of blood, WBC and platelets, leaving only the RBC unaccounted for. Thus by subtractive analysis, greater than 99% of the synuclein in blood resides in the RBC, with most of the remaining synuclein residing in the platelets. Less than 1% of the synuclein was phosphorylated in the 1H7/11 A5 assay. The spike and recoveries were very good in all but the WBC prep. The absorption of synuclein by 1H7- conjugated Sepharose was not as robust as in the previous experiment but nonetheless indicated that the majority of the ELISA signal could be removed with a synuclein-specific antibody and therefore was likely due to synuclein.
[0103] The large amount of synuclein in RBC makes it difficult to accurately assess the amount of synuclein in plasma and WBC. A minor amount of contamination or lysis of RBC will greatly elevate the concentration found in these compartments. Platelets are difficult to work with in a clinical setting since all manipulations must be done within several hours of blood collection. In addition, platelets are notorious for activating even under the best of assay circumstances. Therefore the most practical measure of synuclein in blood is from total blood cells.

Claims

WHAT IS CLAIMED IS:
1. A monoclonal antibody that competes with a monoclonal antibody shown in Table 1 for specific binding to alpha synuclein.
2. The monoclonal antibody of claim 1, wherein the monoclonal antibody specifically binds alpha synuclein compared to beta synuclein.
3. The monoclonal antibody of claim IA, wherein the monoclonal antibody specifically binds human alpha synuclein compared to non-human alpha synuclein.
4. A monoclonal antibody of claim 1 that specifically binds to an epitope bound by a monoclonal antibody shown in Table 1.
5. The monoclonal antibody of claim 4, wherein the monoclonal antibody specifically binds to an epitope within residues 109-120.
6. The monoclonal antibody of claim 4, wherein the monoclonal antibody specifically binds to an epitope on the C-terminus of synuclein.
7. The monoclonal antibody of claim 6, wherein the epitope comprises residues 139-140.
8. The monoclonal antibody of claim 4, wherein the monoclonal antibody specifically binds to an epitope on the N-terminus of synuclein.
9. The monoclonal antibody of claim 4, wherein the monoclonal antibody specifically binds to an epitope within residues 43-51 and within residues 58-65.
10. The monoclonal antibody of Claim 4, wherein the monoclonal antibody specifically binds to a discontinuous epitope comprising residues within residues 43-51 and residues within residues 58-65.
11. The monoclonal antibody of Claim 4, wherein the monoclonal antibody specifically binds to an epitope within residues 118-126.
12. The monoclonal antibody of Claim 4, wherein the monoclonal antibody specifically binds to an epitope comprising residues 91-99.
13. The monoclonal antibody of Claim 4, wherein the monoclonal antibody specifically binds to an epitope comprising residues 40-55.
14. The monoclonal antibody of Claim 4, wherein the monoclonal antibody specifically binds to an epitope comprising residues 124-134, wherein residue 129 is phosphorylated serine.
15. The monoclonal antibody of Claim 4, wherein the monoclonal antibody specifically binds to an epitope comprising residues 123-127, wherein residue 125 is nitrated tyrosine.
16. A monoclonal antibody of claims 1 that specifically binds to human alpha synuclein without specifically binding to mouse alpha synuclein.
17. The monoclonal antibody of claim 16 that is 9E4 or an antibody that competes with 9E4 for specific binding to human alpha synuclein.
18. A monoclonal antibody of claim 1 that specifically binds to mouse alpha synuclein without specifically binding to human alpha synuclein.
19. A monoclonal antibody of claim 1 that specifically binds to mouse and human alpha synuclein.
20. A monoclonal antibody of claim 1 that specifically binds to alpha synuclein phosphorylated at residue 129 without specifically binding to nonphosphorylated alpha synuclein.
21. The monoclonal antibody of claim 20 that is 11 A5 or an antibody that competes with 11 A5 for specific binding to phosphorylated human alpha synuclein.
22. A monoclonal antibody of claim 1 that is end-specific for the N- terminus of alpha synuclein.
23. The monoclonal antibody of claim 22 that is 6H7 or an antibody that competes with 6H7 for specific binding to alpha synuclein.
24. A monoclonal antibody of claim 1 that is end-specific for the C- terminus of alpha synuclein.
25. The monoclonal antibody of claim 24 that is 4Bl or 8A5 or an antibody that competes with 4Bl or 8A5 for specific binding to alpha synuclein.
26. A monoclonal antibody of claim 1 that specifically binds to alpha synuclein without specifically binding to beta or gamma synuclein.
27. The monoclonal antibody of claim 26, wherein the monoclonal antibody is selected from the group consisting of 7G4, 6A8, 5C12, 6A12, 9G5, and 1H7, and antibodies that competes therewith for specific binding to alpha synuclein.
28. A monoclonal antibody of claim 1 that binds to an epitope within a segment of amino acids selected of the group consisting of amino acids 109-12Oj 43-51 and 58-65, 91-96, 118-126, and 91-99.
29. A monoclonal antibody shown in Table 1, selected from the group consisting of 7G4, 6A8, 5C12, 6A12, 8A5, 4Bl, 6H7, 3A12, 12Cl5 9A6, 9G5, 9E4, 23E8, 10G5, 3C12, llA5 andlH7.
30. A monoclonal antibody that specifically binds to phosphorylated alpha synuclein.
31. A monoclonal antibody that specifically binds to nitrated synuclein.
32. A monoclonal antibody capable of capturing soluble synuclein from a fluid sample.
33. The monoclonal antibody of claim 32, wherein the synuclein is alpha- synuclein.
34. The monoclonal antibody of claim 32, wherein the synuclein is human alpha-synuclein.
35. The monoclonal antibody of claim 32, wherein the monoclonal antibody specifically binds to an epitope within residues selected from the group consisting ofthe N-terminus, 118-126, 91-99 and 40-55.
36. The monoclonal antibody of claim 32, wherein the monoclonal antibody competes with the monoclonal antibody selected from the group consisting of 6H7, 3A12, 12Cl, 9A6, 9G5, 9E4, 1H7 and 23E8.
37. The monoclonal antibody of claim 32, wherein the monoclonal antibody is selected from the group consisting of 6H7, 3QA12, 12Cl, 9A6, 9G5, 9E4, 1H7 and 23E8.
38. A pair of monoclonal antibodies, each of the monoclonal antibodies specifically binding to a different epitope within synuclein, wherein the monoclonal antibodies are capable of detecting soluble synuclein when used together in an ELISA assay.
39. The pair of claim 38, wherein one monoclonal antibody is immobilized to a solid phase.
40. The pair of claim 38, wherein the use of the monoclonal antibodies in the ELISA assay is sequential.
41. The pair of claim 38, wherein one monoclonal antibody is a capture antibody and the other monoclonal antibody is a reporter antibody.
42. The pair of claim 38, wherein
(i) the capture antibody specifically binds an epitope within residues selected from the group consisting of N-terminus, 40- 55, 91-99 and 118-126, and
(ii) the reporter antibody specifically binds an epitope within residues 109-120.
43. The pair of claim 38, wherein the pair is selected from the group of
(i) an antibody that competes with 6H7 and an antibody that competes with 5C12 or 12Cl,
(ii) an antibody that competes with 3Al 2 and an antibody that competes with 5C12 or 6H7,
(iii) an antibody that competes with 12Cl and an antibody that competes with 5C12 or 6H7,
(iv) an antibody that competes with 9A6 and an antibody that competes with 5C12, 6H7 or 12Cl,
(v) an antibody that competes with 9G5 and an antibody that competes with 5C12, 6H7 or 12Cl,
(vi) an antibody that competes with 9E4 and an antibody that competes with 5C12, 6H7 or 12Cl,
(vii) an antibody that competes with 1H7 and an antibody that competes with 5C12, 6H7 or 12Cl, and
(viii) an antibody that competes with 10G5 and an antibody that competes with 5C12, 6H7 or 12Cl.
44. The pair of claim 43, wherein the pair is selected from the group of (ix) 6H7 and 5C12 or 12Cl,
(x) 3A12 and 5C12 or 6H7, (xi) 12Cl and 5C12 or 6H7, (xii) 9A6 and 5C12, 6H7 or 12Cl, (xiii) 9G5 and 5C12, 6H7 or 12Cl, (xiv) 9E4 and 5C12, 6H7 or 12Cl, (xv) 1H7 and 5C12, 6H7 or 12Cl, and (xvi) 10G5 and 5C12, 6H7 or 12Cl.
45. A hybridoma producing a monoclonal antibody shown in Table 1.
46. A humanized or chimeric version of a monoclonal antibody shown in
Table 1.
47. A method of humanizing a monoclonal antibody shown in Table 1, comprising: determining the amino acid sequence of CDR regions of the monoclonal antibody; selecting an acceptor antibody; and producing a humanized antibody comprising the CDRs from the monoclonal antibody and variable region frameworks from the acceptor antibody.
48. A method of producing a chimeric form of an antibody shown in Table 1, comprising: determining the amino acid sequence of the light and heavy chain variable regions of the monoclonal antibody; selecting heavy and light chain constant region; producing a chimeric antibody comprising a light chain comprising the light chain variable region fused to the light chain constant region, and a heavy chain comprising the heavy chain variable region fused to the heavy chain constant region.
49. A method for detecting alpha synuclein in a fluid sample, comprising capturing the alpha synuclein using a capture antibody and detecting the alpha synuclein using a reporter antibody, wherein the capture and reporter antibodies bind to different epitopes on alpha synuclein.
50. The method of claim 49, wherein the fluid sample contains 0.1-1.0 M guanidine.
51. The method of claim 49, wherein the fluid sample contains 0.5 M guanidine.
52. The method of claim 49, wherein the reporter antibody or the capture antibody is end-specific for the C-terminal end of SNl-119 or SN1-122.
53. The method of claim 49, wherein the capturing step is performed twice with a first reporter antibody specifically binding to an epitope within SNl-119 and a second reporter antibody specifically binding to an antibody within SN120-140 and determining a ratio of captured alpha synuclein between the two steps, a higher ratio of synuclein captured by first reporter relative to second report being indicative of pathogenicity.
54. The method of claim 49, wherein the reporter antibody in the first capturing step specifically binds to an epitope within SN 109-120, and the reporter antibody in the second capturing step specifically binds to an epitope within SN 120-126, and the capture antibody in both capturing steps specifically binds to an epitope within SN 91-99.
55. The method of claim 49, wherein the capture antibody is immobilized to a solid phase, and the reporter antibody in solution, and the detecting step comprises detecting alpha synuclein from presence of the reporter antibody linked to the solid phase via binding of the reporter antibody to alpha synuclein, which is in turn bound to the capture antibody.
56. The method of claim 55, further comprising: comparing a signal from the reporter antibody linked to the solid phase from analyzing the sample with a signal from the reporter antibody linked to the solid phase from analyzing a control sample containing a known amount of alpha synuclein to determine the amount of alpha synuclein in the sample.
57. The method of claim 56, further comprising comparing a signal from the reporter antibody linked to the solid phase from analyzing the sample to a calibration curve of signal versus amount of alpha synuclein to determine the amount of alpha synuclein in the sample.
58. The method of claim 56, wherein a signal from the reporter antibody is proportional to the amount of synuclein in the sample.
59. The method of claim 56, further comprising contacting the reporter antibody with a labeled antibody to generate a signal indicating presence of the reporter antibody.
60. The method of claim 56, wherein the capture antibody specifically binds to an epitope within amino acids 91-99 of human alpha synuclein and the reporter antibody binds to an epitope within amino acids 109-120 of alpha synuclein.
61. The method of claim 60, wherein the capture antibody is 1H7 and the reporter antibody is 5C12.
62. The method of claim 61, wherein the sample is a sample from a human.
63. The method of claim 56, wherein the capture antibody is 1H7 and the reporter antibody is 9E4.
64. The method of claim 63, wherein the sample from a mouse.
65. The method of claim 49, wherein the sample is a body fluid.
66. The method of claim 65, wherein the sample is cerebrospinal fluid (CSF) of a human.
67. The method of claim 49, wherein the sample is a brain homogenate of a human or transgenic animal.
68. The method of claim 49, wherein the sample is a medium used to culture cells.
69. The method of claim 68, wherein the cells express recombinant alpha synuclein.
70. The method of claim 49, wherein the sample contains a fragment of full-length alpha synuclein and the detecting step detects the fragment.
71. A method of monitoring processing of alpha-synuclein to a fragment, comprising culturing a cell expressing alpha synuclein and processing the alpha synuclein to a fragment that is secreted to the cell media. detecting the fragment in the cell media.
72. The method of claim 71, wherein the cell is a PeakS or SY5Y cell transfected with alpha synuclein.
73. The method of claim 71 , wherein the cell is a cortical cell.
74. A method of screening an agent for activity in inhibiting processing or secretion of alpha synuclein, comprising contacting a cell expressing alpha synuclein in culture medium with an agent; detecting alpha synuclein or a fragment thereof in the medium; comparing the amount of alpha synuclein or the fragment in the medium with an among of alpha synuclein or fragment in medium from a control cell not contacted with the agent, wherein a reduction in the amount of alpha synuclein or the fragment indicates the agent inhibits processing or secretion of the alpha synuclein.
75. A method for detecting soluble alpha synuclein in a fluid sample said method comprising: capturing the soluble fragment from the sample using a first binding substance under conditions in which the first binding substance specifically binds to a first epitope and detecting capture of the soluble alpha synuclein using a second binding substance which binds to an epitope on a second region of the soluble alpha synuclein different from the first epitope.
76. A method for detecting a soluble fragment of alpha synuclein in a fluid sample in the presence of full length alpha synuclein the method comprising: capturing the soluble fragment from the sample using a first binding substance, and detecting the soluble fragment using a second binding substance, wherein both binding substances specifically bind to the fragment and at least one specifically binds to the fragment without binding to full length alpha-synuclein.
PCT/US2005/028361 2005-08-09 2005-08-09 Antibodies to alpha-synuclein WO2007021255A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2005/028361 WO2007021255A1 (en) 2005-08-09 2005-08-09 Antibodies to alpha-synuclein

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2005/028361 WO2007021255A1 (en) 2005-08-09 2005-08-09 Antibodies to alpha-synuclein

Publications (1)

Publication Number Publication Date
WO2007021255A1 true WO2007021255A1 (en) 2007-02-22

Family

ID=37757838

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/028361 WO2007021255A1 (en) 2005-08-09 2005-08-09 Antibodies to alpha-synuclein

Country Status (1)

Country Link
WO (1) WO2007021255A1 (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2118300A2 (en) * 2007-02-23 2009-11-18 Elan Pharmaceuticals Inc. Prevention and treatment of synucleinopathic and amyloidogenic disease
US7674599B2 (en) 2003-11-08 2010-03-09 Elan Pharmaceuticals, Inc. Methods of using antibodies to detect alpha-synuclein in fluid samples
US7727957B2 (en) 2002-11-01 2010-06-01 Janssen Alzheimer Immunotherapy Treatment and delay of outset of Parkinson's disease
WO2010069603A1 (en) 2008-12-19 2010-06-24 Neurimmune Therapeutics Ag Human anti-alpha-synuclein autoantibodies
US7919088B2 (en) 2003-10-31 2011-04-05 Elan Pharmaceuticals, Inc. Treatment and delay of onset of synucleinopathic and amyloidogenic disease
US8147833B2 (en) 2007-02-23 2012-04-03 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
WO2012177972A1 (en) 2011-06-23 2012-12-27 Biogen Idec International Neuroscience Gmbh Anti-alpha synuclein binding molecules
US8506959B2 (en) 2002-11-01 2013-08-13 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
CN103320391A (en) * 2013-07-12 2013-09-25 首都医科大学 Hybridoma cell and application thereof
US8546532B2 (en) 2008-04-17 2013-10-01 Declion Pharmaceuticals, Inc. Synthesis of directed sequence polymer compositions and antibodies thereof for the treatment of protein conformational disorders
US8697082B2 (en) 2002-11-01 2014-04-15 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
CN103983778A (en) * 2014-05-08 2014-08-13 首都医科大学宣武医院 Method for detecting in-vitro disease-related protein polymerization promotion capability of body fluid of subject
JP2014159439A (en) * 2007-02-23 2014-09-04 Janssen Alzheimer Immunotherapy Prevention and treatment of synucleinopathy and amyloidogenic disease
US9034337B2 (en) 2003-10-31 2015-05-19 Prothena Biosciences Limited Treatment and delay of outset of synucleinopathic and amyloidogenic disease
WO2015075011A1 (en) * 2013-11-21 2015-05-28 F. Hoffmann-La Roche Ag ANTI-alpha-SYNUCLEIN ANTIBODIES AND METHODS OF USE
JP2016500673A (en) * 2012-10-08 2016-01-14 プロセナ バイオサイエンシーズ リミテッド Alpha-synuclein recognition antibody
JP2016208981A (en) * 2011-10-28 2016-12-15 プロセナ バイオサイエンシーズ リミテッド Humanized antibodies that recognize alpha-synuclein
US9670272B2 (en) 2007-01-05 2017-06-06 University Of Zurich Method of providing disease-specific binding molecules and targets
US9879071B2 (en) 2003-05-19 2018-01-30 Prothena Biosciences Limited Antibodies to alpha synuclein
WO2018109058A1 (en) 2016-12-16 2018-06-21 H. Lundbeck A/S Agents, uses and methods
DE102011008153B4 (en) 2011-01-08 2018-08-09 Aj Roboscreen Gmbh Process for the preparation of a monoclonal antibody against oligomeric alpha-synuclein
WO2019115671A1 (en) * 2017-12-15 2019-06-20 Ucb Biopharma Sprl Anti-alpha synuclein antibodies
WO2019175883A1 (en) * 2018-03-14 2019-09-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd Detection of phospho-serine 129 alpha-synuclein in blood cells as a biomarker for synucleinopathies
EP3551228A4 (en) * 2016-12-12 2020-08-12 The Michael J. Fox Foundation For Parkinson's Research Antibodies to human alpha-synuclein
US10842871B2 (en) 2014-12-02 2020-11-24 Biogen International Neuroscience Gmbh Methods for treating Alzheimer's disease
US11142570B2 (en) 2017-02-17 2021-10-12 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
US11261242B2 (en) 2017-12-15 2022-03-01 UCB Biopharma SRL Anti-alpha-synuclein antibodies
US11655289B2 (en) 2017-08-22 2023-05-23 Biogen Ma Inc. Pharmaceutical compositions containing anti-beta amyloid antibodies
TWI860014B (en) 2017-12-15 2024-10-21 比利時商Ucb生物製藥公司 Antibodies

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHOI J.-Y. ET AL.: "Fine epitope mapping of monoclonal antibodies specific to human alpha-synuclein", NEUROSCIENCE LETTERS, vol. 397, 10 April 2006 (2006-04-10), pages 53 - 58, XP005323072 *

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8506959B2 (en) 2002-11-01 2013-08-13 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US7727957B2 (en) 2002-11-01 2010-06-01 Janssen Alzheimer Immunotherapy Treatment and delay of outset of Parkinson's disease
US8697082B2 (en) 2002-11-01 2014-04-15 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US9879071B2 (en) 2003-05-19 2018-01-30 Prothena Biosciences Limited Antibodies to alpha synuclein
US7919088B2 (en) 2003-10-31 2011-04-05 Elan Pharmaceuticals, Inc. Treatment and delay of onset of synucleinopathic and amyloidogenic disease
US9034337B2 (en) 2003-10-31 2015-05-19 Prothena Biosciences Limited Treatment and delay of outset of synucleinopathic and amyloidogenic disease
US7910333B2 (en) 2003-11-08 2011-03-22 Elan Pharmaceuticals, Inc. Antibodies to alpha-synuclein
US7674599B2 (en) 2003-11-08 2010-03-09 Elan Pharmaceuticals, Inc. Methods of using antibodies to detect alpha-synuclein in fluid samples
US8673593B2 (en) 2003-11-08 2014-03-18 Elan Pharmaceuticals, Llc Antibodies to alpha-synuclein
US9670272B2 (en) 2007-01-05 2017-06-06 University Of Zurich Method of providing disease-specific binding molecules and targets
US10131708B2 (en) 2007-01-05 2018-11-20 University Of Zürich Methods of treating Alzheimer's disease
US10202445B2 (en) 2007-01-05 2019-02-12 University Of Zurich Method of providing disease-specific binding molecules and targets
US9828420B2 (en) 2007-01-05 2017-11-28 University Of Zürich Method of providing disease-specific binding molecules and targets
EP2118300A4 (en) * 2007-02-23 2010-10-20 Elan Pharm Inc Prevention and treatment of synucleinopathic and amyloidogenic disease
EP2118300A2 (en) * 2007-02-23 2009-11-18 Elan Pharmaceuticals Inc. Prevention and treatment of synucleinopathic and amyloidogenic disease
EP2583978A3 (en) * 2007-02-23 2013-09-11 The Regents of the University of California Prevention and treatment of synucleinopathic and amyloidogenic disease
JP2016145211A (en) * 2007-02-23 2016-08-12 ヤンセン・サイエンシズ・アイルランド・ユーシー Prevention and treatment of synucleinopathic and amyloidogenic disease
EP3067066B1 (en) 2007-02-23 2019-03-27 Prothena Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US8147833B2 (en) 2007-02-23 2012-04-03 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US8092801B2 (en) 2007-02-23 2012-01-10 Neotope Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
JP2014159439A (en) * 2007-02-23 2014-09-04 Janssen Alzheimer Immunotherapy Prevention and treatment of synucleinopathy and amyloidogenic disease
JP2010526028A (en) * 2007-02-23 2010-07-29 エラン・フアーマシユーチカルズ・インコーポレーテツド Prevention and treatment of synucleinopathies and amyloidogenic diseases
EP3067066A1 (en) * 2007-02-23 2016-09-14 Prothena Biosciences Limited Prevention and treatment of synucleinopathic and amyloidogenic disease
US8546532B2 (en) 2008-04-17 2013-10-01 Declion Pharmaceuticals, Inc. Synthesis of directed sequence polymer compositions and antibodies thereof for the treatment of protein conformational disorders
US9896504B2 (en) 2008-12-19 2018-02-20 Biogen International Neuroscience Gmbh Human anti-alpha-synuclein antibodies
US8940276B2 (en) 2008-12-19 2015-01-27 Biogen Idec International Neuroscience Gmbh Human anti-alpha-synuclein antibodies
EP2949666A1 (en) 2008-12-19 2015-12-02 Biogen Idec International Neuroscience GmbH Human anti-alpha-synuclein antibodies
EP3521309A1 (en) 2008-12-19 2019-08-07 Biogen International Neuroscience GmbH Human anti-alpha-synuclein antibodies
US10703808B2 (en) 2008-12-19 2020-07-07 Biogen International Neuroscience Gmbh Human anti-alpha-synuclein antibodies
WO2010069603A1 (en) 2008-12-19 2010-06-24 Neurimmune Therapeutics Ag Human anti-alpha-synuclein autoantibodies
DE102011008153B4 (en) 2011-01-08 2018-08-09 Aj Roboscreen Gmbh Process for the preparation of a monoclonal antibody against oligomeric alpha-synuclein
JP2017000160A (en) * 2011-06-23 2017-01-05 バイオジェン インターナショナル ニューロサイエンス ゲーエムベーハー Anti-alpha synuclein binding molecules
KR20140053974A (en) * 2011-06-23 2014-05-08 바이오겐 아이덱 인터내셔널 뉴로사이언스 게엠베하 Anti-alpha synuclein binding molecules
WO2012177972A1 (en) 2011-06-23 2012-12-27 Biogen Idec International Neuroscience Gmbh Anti-alpha synuclein binding molecules
US9580493B2 (en) 2011-06-23 2017-02-28 Biogen International Neuroscience Gmbh Anti-α synuclein binding molecules
US10301381B2 (en) 2011-06-23 2019-05-28 Biogen International Neuroscience Gmbh Anti-alpha synuclein binding molecules
KR101976887B1 (en) 2011-06-23 2019-05-09 바이오겐 인터내셔널 뉴로사이언스 게엠베하 Anti-alpha synuclein binding molecules
EP2723379A4 (en) * 2011-06-23 2015-03-04 Biogen Idec Internat Neuroscience Gmbh Anti-alpha synuclein binding molecules
US9975947B2 (en) 2011-06-23 2018-05-22 Biogen International Neuroscience Gmbh Anti-alpha synuclein binding molecules
EP2723379A1 (en) * 2011-06-23 2014-04-30 Biogen IDEC International Neuroscience GmbH Anti-alpha synuclein binding molecules
JP2014528695A (en) * 2011-06-23 2014-10-30 バイオジェン アイデック インターナショナル ニューロサイエンス ゲーエムベーハー Anti-alpha synuclein binding molecule
JP2016208981A (en) * 2011-10-28 2016-12-15 プロセナ バイオサイエンシーズ リミテッド Humanized antibodies that recognize alpha-synuclein
JP2016500673A (en) * 2012-10-08 2016-01-14 プロセナ バイオサイエンシーズ リミテッド Alpha-synuclein recognition antibody
CN103320391A (en) * 2013-07-12 2013-09-25 首都医科大学 Hybridoma cell and application thereof
CN111499742B (en) * 2013-11-21 2024-05-07 豪夫迈·罗氏有限公司 Anti-alpha-synuclein antibodies and methods of use
US9670274B2 (en) 2013-11-21 2017-06-06 Hoffmann-La Roche Inc. Anti-alpha-synuclein antibodies and methods of use
CN111499742A (en) * 2013-11-21 2020-08-07 豪夫迈·罗氏有限公司 Anti- α -synuclein antibodies and methods of use
US9890209B2 (en) 2013-11-21 2018-02-13 Hoffmann-La Roche Inc. Anti-alpha-synuclein antibodies and methods of use
WO2015075011A1 (en) * 2013-11-21 2015-05-28 F. Hoffmann-La Roche Ag ANTI-alpha-SYNUCLEIN ANTIBODIES AND METHODS OF USE
US10316082B2 (en) 2013-11-21 2019-06-11 Hoffmann-La Roche Inc. Anti-alpha-synuclein antibodies and methods of use
EP3783020A1 (en) * 2013-11-21 2021-02-24 F. Hoffmann-La Roche AG Anti-alpha-synuclein antibodies and methods of use
US9493553B2 (en) 2013-11-21 2016-11-15 Hoffmann-La Roche Inc. Anti-alpha-synuclein antibodies and methods of use
AU2014351996B2 (en) * 2013-11-21 2020-01-02 F. Hoffmann-La Roche Ag Anti-alpha-synuclein antibodies and methods of use
CN103983778B (en) * 2014-05-08 2016-04-13 首都医科大学宣武医院 Method for detecting in-vitro disease-related protein polymerization promotion capability of body fluid of subject
CN103983778A (en) * 2014-05-08 2014-08-13 首都医科大学宣武医院 Method for detecting in-vitro disease-related protein polymerization promotion capability of body fluid of subject
US10842871B2 (en) 2014-12-02 2020-11-24 Biogen International Neuroscience Gmbh Methods for treating Alzheimer's disease
EP3551228A4 (en) * 2016-12-12 2020-08-12 The Michael J. Fox Foundation For Parkinson's Research Antibodies to human alpha-synuclein
WO2018109058A1 (en) 2016-12-16 2018-06-21 H. Lundbeck A/S Agents, uses and methods
US11142570B2 (en) 2017-02-17 2021-10-12 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
US11827695B2 (en) 2017-02-17 2023-11-28 Bristol-Myers Squibb Company Antibodies to alpha-synuclein and uses thereof
US11655289B2 (en) 2017-08-22 2023-05-23 Biogen Ma Inc. Pharmaceutical compositions containing anti-beta amyloid antibodies
US11261242B2 (en) 2017-12-15 2022-03-01 UCB Biopharma SRL Anti-alpha-synuclein antibodies
US11292831B2 (en) 2017-12-15 2022-04-05 UCB Biopharma SRL Anti-alpha synuclein antibodies
IL275223B1 (en) * 2017-12-15 2023-07-01 UCB Biopharma SRL Anti-alpha synuclein antibodies
IL275223B2 (en) * 2017-12-15 2023-11-01 UCB Biopharma SRL Anti-alpha synuclein antibodies
WO2019115671A1 (en) * 2017-12-15 2019-06-20 Ucb Biopharma Sprl Anti-alpha synuclein antibodies
TWI860014B (en) 2017-12-15 2024-10-21 比利時商Ucb生物製藥公司 Antibodies
WO2019175883A1 (en) * 2018-03-14 2019-09-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd Detection of phospho-serine 129 alpha-synuclein in blood cells as a biomarker for synucleinopathies

Similar Documents

Publication Publication Date Title
US8673593B2 (en) Antibodies to alpha-synuclein
WO2007021255A1 (en) Antibodies to alpha-synuclein
US20230228772A1 (en) Methods For Detecting Phosphorylated Alpha-Synuclein
EP2189526B1 (en) Antibody binding specifically to tdp-43 aggregate
US20070280935A1 (en) Antibody that recognizes phosphorylated peptides
US7682795B2 (en) Method of diagnosing Alzheimer's Disease
KR20190066027A (en) As a protein antigen for use in diagnostic assays, the REP protein
AU653536B2 (en) Measuring non-dystrophin proteins and diagnosing muscular dystrophy
JP3259768B2 (en) Testing methods for kidney disease
EP1876185A1 (en) An antibody which recognizes phosphorylated polypeptides
EP1242450B1 (en) Organ transplant rejection and associated conditions
EP3482210B1 (en) Assay for detecting total and s129 phosphorylated alpha-synuclein
CN113508300A (en) Novel biomarkers for human alzheimer's disease
JP2003130880A (en) Reagent and kit for measuring immunity of abnormal prion, and method of measuring immunity of abnormal prion by using the same
US20030166568A1 (en) Organic compounds

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 05785617

Country of ref document: EP

Kind code of ref document: A1