+

WO2014096321A1 - Anticorps spécifiques de la protéine tau phosphorylée à la sérine 422 et leurs utilisations pour traiter et diagnostiquer les tauopathies - Google Patents

Anticorps spécifiques de la protéine tau phosphorylée à la sérine 422 et leurs utilisations pour traiter et diagnostiquer les tauopathies Download PDF

Info

Publication number
WO2014096321A1
WO2014096321A1 PCT/EP2013/077608 EP2013077608W WO2014096321A1 WO 2014096321 A1 WO2014096321 A1 WO 2014096321A1 EP 2013077608 W EP2013077608 W EP 2013077608W WO 2014096321 A1 WO2014096321 A1 WO 2014096321A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
tau
antibodies
serine
chain
Prior art date
Application number
PCT/EP2013/077608
Other languages
English (en)
Inventor
Luc Buee
Laetitia TROQUIER
Philippe Lassale
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Institut Pasteur De Lille
Université De Droit Et De La Santé De Lille 2
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Institut Pasteur De Lille, Université De Droit Et De La Santé De Lille 2 filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2014096321A1 publication Critical patent/WO2014096321A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • the present invention relates to antibodies or fragments thereof that specifically bind to Tau phosphorylated at serine 422 (pS422), and to their use for treating and diagnosing Tauopathies.
  • Human Tau is a neuronal micro tubule-associated protein found predominantly in axons and functions to promote tubulin polymerization and stabilize microtubules.
  • Six isoforms (isoform A, B, C, D, E, F, G, fetal-Tau) are found in the human brain, the longest isoform comprising 441 amino acids (isoform F, Uniprot P10636-8).
  • Tau and its properties are also described by Reynolds, C. H. et al., J. Neurochem. 69 (1997) 191-198.
  • Tau, in its hyperphosphorylated form is the major component of paired helical filaments (PHF), the building block of neurofibrillary lesions in Alzheimer's disease (AD) brain.
  • PHF paired helical filaments
  • AD Alzheimer's disease
  • Tau can be phosphorylated at its serine or threonine residues by several different kinases including GSK3beta, cdk5, MARK and members of
  • Protein misfolded aggregates affect a number of different neurodegenerative diseases including Alzheimer's disease, Parkinsonism and other Tauopathies.
  • Several studies have provided the hypothesis that protein aggregates spread in predictable sequences between anatomically related brain regions (Delacourte A, David J-P, Sergeant N, Buee L, Wattez A, Vermersch P, Ghozali F, Fallet-Bianco C, Pasquier F, Lebert F, Petit H, DiMenza C. The biochemical pathway of neurofibrillary degeneration in aging and Alzheimer's disease. Neurology, 52 ⁇ 1999) 1158-1165.; Verny, M., C. Duyckaerts, Y. Agid, and J. J. Hauw.
  • Phosphorylation serine 422 on Tau protein is also found in association with developing pathology in various transgenic mouse models of Alzheimer's disease.
  • Deters, N. et al. mention in Biochem. Biophys. Res. Commun. 379 (2009) 400-405 that double-transgenic Dom5/pR5 mice showed 7-fold increased numbers of hippocampal neurons that contain Tau specifically phosphorylated the pathological S422 epitope.
  • Goetz, J. et al. (Science 293 (2001) 1491-1495) reported the appearance of Tau phosphorylated at serine 422 in the brains of Tau P301L transgenic mice injected with Abeta42 fibrils.
  • WO2010142423 describes antibodies binding to Tau that is phosphorylated at serine 422 (pS422).
  • Monoclonal antibodies against Tau pS422 are also described in EP 1 876 185.
  • Polyclonal antibodies against Tau pS422 are commercially available (e.g. ProSci Inc. and Biosource International).
  • the inventors provide an antibody that specifically binds to Tau phosphorylated at serine 422 (pS422) and demonstrate that said antibody is able to reduce the pathological Tau aggregation, to accelerate the subsequent central degradation of Tau phosphorylated at Ser422 correlated with increased of Tau in the plasma and finally to correct the cognitive impairment produced by Tauopathy pathology.
  • the present invention thus provides a publicly available source for said antibody
  • This monoclonal antibody hereinafter designated “2H9” is indeed produced by the hybridoma deposited in accordance with the terms of Budapest Treaty, at the CNCM (Collection Nationale de Cultures de Microorganismes, Institut Pasteur, 25 rue du Dondel Roux, 75724 Paris Cedex 15, France), on August 7, 2012, under the deposit number CNCM-I-4666.
  • Tau refers to all Tau isoforms derived from the MAPT gene. All numbering is given from the longest brain isoform of human Tau, comprising 441 amino acids (isoform F, Uniprot P10636-8).
  • Tau phosphorylated at serine 422 pS422
  • Tau phosphorylated at serine 422 pS422
  • tauopathy has its general meaning in the art and refers to a disease characterized by an abnormal hyperphosphorylation of Tau (Iqbal, K. et al. Biochimica et Biophysica Acta (BBA) 1739 (2005) 198-210).
  • Tauopathies include among others, Alzheimer's Disease, Down syndrome; Guam parkinsonism dementia complex; Dementia pugilistica and other chronic traumatic encephalopathies; myotonic dystrophies; Niemann- Pick disease type C; Pick disease; argyrophilic grain disease; Fronto-temporal dementia; Cortico-basal degeneration; Pallido-ponto-nigral degeneration; Progressive supranuclear palsy; and Prion disorders such as Gerstmann-Straussler-Scheinker disease with tangles.
  • "antibody” or “immunoglobulin” have the same meaning, and will be used equally in the present invention.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds an antigen.
  • antibody encompasses not only whole antibody molecules, but also antibody fragments as well as variants (including derivatives) of antibodies and antibody fragments.
  • two heavy chains are linked to each other by disulfide bonds and each heavy chain is linked to a light chain by a disulfide bond. There are two types of light chain, lambda (1) and kappa (k).
  • the heavy chain includes two domains, a variable domain (VL) and a constant domain (CL).
  • the heavy chain includes four domains, a variable domain (VH) and three constant domains (CHI, CH2 and CH3, collectively referred to as CH).
  • VL variable domain
  • VH variable domain
  • CH constant domain
  • the constant region domains of the light (CL) and heavy (CH) chains confer important biological properties such as antibody chain association, secretion, trans-placental mobility, complement binding, and binding to Fc receptors (FcR).
  • the Fv fragment is the N-terminal part of the Fab fragment of an immunoglobulin and consists of the variable portions of one light chain and one heavy chain.
  • the specificity of the antibody resides in the structural complementarity between the antibody combining site and the antigenic determinant.
  • Antibody combining sites are made up of residues that are primarily from the hypervariable or complementarity determining regions (CDRs). Occasionally, residues from nonhypervariable or framework regions (FR) influence the overall domain structure and hence the combining site.
  • Complementarity Determining Regions or CDRs refer to amino acid sequences which together define the binding affinity and specificity of the natural Fv region of a native immunoglobulin binding site.
  • the light and heavy chains of an immunoglobulin each have three CDRs, designated VL-CDR1, VL-CDR2, VL-CDR3 and VH-CDR1, VH-CDR2, VH-CDR3, respectively.
  • An antigen-binding site therefore, includes six CDRs, comprising the CDR set from each of a heavy and a light chain V region.
  • Framework Regions refer to amino acid sequences interposed between CDRs.
  • the monoclonal antibody hereinafter designated "2H9” is produced by the hybridoma deposited in accordance with the terms of Budapest Treaty, at the CNCM (Collection Nationale de Cultures de Microorganismes, Institut Pasteur, 25 rue du Dondel Roux, 75724 Paris Cedex 15, France), on August 7, 2012, under the deposit number CNCM-I-4666.
  • the inventors have cloned and sequenced the variable domain (VL) of the light chain, and the variable domain (VH) of the heavy chain of the monoclonal antibody 2H9.
  • the location of the sequences encoding the complementarity determining regions (CDRs) of said antibody have been determined with reference to other antibody sequences (Kabat EA et al., 1991).
  • chimeric antibody refers to an antibody which comprises a VH domain and a VL domain of an antibody, and a CH domain and a CL domain of a human antibody.
  • humanized antibody refers to an antibody having variable region framework and constant regions from a human antibody but retains the CDRs of a previous non human antibody.
  • antibody fragment refers to a fragment of an antibody which contain the variable domains comprising the CDRs of said antibody.
  • the basic antibody fragments include Fab, Fab', F(ab')2 Fv, scFv, dsFv.
  • Fab fragment of an antibody which contain the variable domains comprising the CDRs of said antibody.
  • Fab denotes an antibody fragment having a molecular weight of about 50,000 and antigen binding activity, in which about a half of the N-terminal side of H chain and the entire L chain, among fragments obtained by treating IgG with a protease, papaine, are bound together through a disulfide bond.
  • F(ab')2 refers to an antibody fragment having a molecular weight of about
  • Fab' refers to an antibody fragment having a molecular weight of about 50,000 and antigen binding activity, which is obtained by cutting a disulfide bond of the hinge region of the F(ab')2.
  • a single chain Fv (“scFv”) polypeptide is a covalently linked VH::VL heterodimer which is usually expressed from a gene fusion including VH and VL encoding genes linked by a peptide-encoding linker.
  • dsFv is a VH::VL heterodimer stabilised by a disulfide bond.
  • Divalent and multivalent antibody fragments can form either spontaneously by association of monovalent scFvs, or can be generated by coupling monovalent scFvs by a peptide linker, such as divalent sc(Fv)2.
  • diabodies refers to small antibody fragments with multivalent antigen-binding sites (2, 3 or four), which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) in the same polypeptide chain (VH-VL).
  • VH heavy-chain variable domain
  • VL light-chain variable domain
  • purified preferably means at least 75% by weight, more preferably at least 85% by weight, more preferably still at least 95% by weight, and most preferably at least 98% by weight, of biological macromolecules of the same type are present.
  • An "isolated" nucleic acid molecule which encodes a particular polypeptide refers to a nucleic acid molecule which is substantially free of other nucleic acid molecules that do not encode the polypeptide; however, the molecule may include some additional bases or moieties which do not deleteriously affect the basic characteristics of the composition.
  • the present invention relates to an antibody characterized in that it is capable of binding human Tau phosphorylated at serine 422 and having a variable light chain (VL) comprising the VL-CDRl, VL-CDR2 and VL-CDR3 of the VL chain and a variable heavy chain (VH) comprising the VH-CDR1, VH-CDR2 and VH-CDR3 of the VH chain of the antibody 2H9 obtainable from hybridoma deposited as CNCM-I-4666.
  • VL variable light chain
  • VH variable heavy chain
  • the antibody of the invention which is characterized in that it is capable of binding human Tau phosphorylated at serine 422, has the following 6 CDRs:
  • said antibody is selected from :
  • said antibody is selected from the group consisting of, chimeric antibodies, and humanized antibodies.
  • the antibody of the invention comprises the VL chain and the VH chain of the antibody 2H9 obtainable from hybridoma deposited as CNCM-I-4666.
  • the antibody of the invention is a chimeric antibody, which comprises the variable chains (VL and VH) of the antibody 2H9 obtainable from hybridoma deposited as CNCM-I-4666.
  • the antibody of the invention is a humanized antibody comprising the CDRs of the antibody 2H9 obtainable from hybridoma deposited as CNCM-I- 4666.
  • a further aspect of the invention thus relates to a murine monoclonal antibody (2H9) obtainable from the hybridoma available under CNCM deposit number 1-4666.
  • the invention further provides fragments of said antibodies which include but are not limited to Fv, Fab, F(ab')2, Fab', dsFv, scFv, sc(Fv)2 diabodies, tribodies and tetrabodies.
  • the antibody of the invention is able to reduce the brain level of the Tau phosphorylated at Ser422 and its aggregation.
  • the antibodies of the invention may be produced by any technique known in the art, such as, without limitation, any chemical, biological, genetic or enzymatic technique, either alone or in combination.
  • antibodies of the desired sequence can readily produce said antibodies, by standard techniques for production of polypeptides. For instance, they can be synthesized using well-known solid phase method, preferably using a commercially available peptide synthesis apparatus (such as that made by Applied Biosystems, Foster City, California) and following the manufacturer's instructions.
  • antibodies of the invention can be synthesized by recombinant DNA techniques well-known in the art.
  • antibodies can be obtained as DNA expression products after incorporation of DNA sequences encoding the antibodies into expression vectors and introduction of such vectors into suitable eukaryotic or prokaryotic hosts that will express the desired antibodies, from which they can be later isolated using well-known techniques.
  • a further object of the invention relates to a nucleic acid sequence encoding an antibody according to the invention.
  • the invention relates to a nucleic acid sequence encoding the VH domain of the antibody of the invention (e.g. the antibody obtainable from hybridoma deposited as CNCM-I-4666 (2H9)) and/ or the VL domain of the antibody of the invention (e.g. the antibody obtainable from hybridoma deposited as CNCM-I-4666 (2H9)).
  • said nucleic acid is a DNA or RNA molecule, which may be included in any suitable vector, such as a plasmid, cosmid, episome, artificial chromosome, phage or a viral vector.
  • vector means the vehicle by which a DNA or RNA sequence (e.g. a foreign gene) can be introduced into a host cell, so as to transform the host and promote expression (e.g. transcription and translation) of the introduced sequence.
  • a further object of the invention relates to a vector comprising a nucleic acid of the invention.
  • Such vectors may comprise regulatory elements, such as a promoter, enhancer, terminator and the like, to cause or direct expression of said antibody upon administration to a subject.
  • promoters and enhancers used in the expression vector for animal cell include early promoter and enhancer of SV40, LTR promoter and enhancer of Moloney mouse leukemia virus, promoter and enhancer of immunoglobulin H chain and the like.
  • plasmids include replicating plasmids comprising an origin of replication, or integrative plasmids, such as for instance pUC, pcDNA, pBR, and the like.
  • viral vector include adenoviral, retroviral, herpes virus and AAV vectors. Such recombinant viruses may be produced by techniques known in the art, such as by transfecting packaging cells or by transient transfection with helper plasmids or viruses..
  • a further object of the present invention relates to a host cell which has been transfected, infected or transformed by a nucleic acid and/or a vector according to the invention and expressing an antibody according to the invention.
  • transformation means the introduction of a "foreign” (i.e. extrinsic or extracellular) gene, DNA or RNA sequence to a host cell, so that the host cell will express the introduced gene or sequence to produce a desired substance, typically a protein or enzyme coded by the introduced gene or sequence.
  • a host cell that receives and expresses introduced DNA or RNA bas been "transformed”.
  • the nucleic acids of the invention may be used to produce an antibody of the invention in a suitable expression system.
  • expression system means a host cell and compatible vector under suitable conditions, e.g. for the expression of a protein coded for by foreign DNA carried by the vector and introduced to the host cell.
  • Common expression systems include E. coli host cells and plasmid vectors, insect host cells and Baculovirus vectors, and mammalian host cells and vectors.
  • Other examples of host cells include, without limitation, prokaryotic cells (such as bacteria) and eukaryotic cells (such as yeast cells, mammalian cells, insect cells, plant cells, etc.).
  • E.coli Escherreocoli
  • Kluyveromyces or Saccharomyces yeasts mammalian cell lines (e.g., Vero cells, CHO cells, 3T3 cells, COS cells, etc.) as well as primary or established mammalian cell cultures (e.g., produced from lymphoblasts, fibroblasts, embryonic cells, epithelial cells, nervous cells, adipocytes, etc.).
  • mammalian cell lines e.g., Vero cells, CHO cells, 3T3 cells, COS cells, etc.
  • primary or established mammalian cell cultures e.g., produced from lymphoblasts, fibroblasts, embryonic cells, epithelial cells, nervous cells, adipocytes, etc.
  • Examples also include mouse SP2/0-Agl4 cell (ATCC CRL1581), mouse P3X63-Ag8.653 cell (ATCC CRL1580), CHO cell in which a dihydrofolate reductase gene (hereinafter referred to as "DHFR gene") is defective (Urlaub G et al; 1980), rat YB2/3HL.P2.G11.16Ag.20 cell (ATCC CRL1662, hereinafter referred to as "YB2/0 cell”), and the like.
  • DHFR gene dihydrofolate reductase gene
  • the present invention also relates to a method of producing an antibody according to the invention, said method comprising the steps of: (i) introducing in vitro or ex vivo a recombinant nucleic acid or a vector as described above into a competent host cell, (ii) culturing in vitro or ex vivo the recombinant host cell obtained (iii), recovering the expressed antibody.
  • the method comprises the steps of:
  • Antibodies of the invention are suitably separated from the culture medium by conventional immunoglobulin purification procedures such as, for example, protein A- Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • the human chimeric antibody of the present invention can be produced by obtaining nucleic sequences encoding VL and VH domains as previously described, constructing a human chimeric antibody expression vector by inserting them into an expression vector for animal cell having genes encoding human antibody CH and human antibody CL, and expressing the coding sequence by introducing the expression vector into an animal cell.
  • the CH domain of a human chimeric antibody it may be any region which belongs to human immunoglobulin, but those of IgG class are suitable and any one of subclasses belonging to IgG class, such as IgGl, IgG2, IgG3 and IgG4, can also be used.
  • the CL of a human chimeric antibody may be any region which belongs to Ig, and those of kappa class or lambda class can be used. Methods for producing chimeric antibodies involve conventional recombinant DNA and gene transfection techniques are well known in the art.
  • the humanized antibody of the present invention may be produced by obtaining nucleic acid sequences encoding CDR domains, as previously described, constructing a humanized antibody expression vector by inserting them into an expression vector for animal cell having genes encoding (i) a heavy chain constant region identical to that of a human antibody and (ii) a light chain constant region identical to that of a human antibody, and expressing the genes by introducing the expression vector into an animal cell.
  • the humanized antibody expression vector may be either of a type in which a gene encoding an antibody heavy chain and a gene encoding an antibody light chain exists on separate vectors or of a type in which both genes exist on the same vector (tandem type). In respect of easiness of construction of a humanized antibody expression vector, easiness of introduction into animal cells, and balance between the expression levels of antibody H and L chains in animal cells, humanized antibody expression vector of the tandem type is preferred. Methods for producing humanized antibodies based on conventional recombinant DNA and gene transfection techniques are well known in the art.
  • Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT publication WO91/09967; U.S. Pat. Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596), and chain shuffling (U.S. Pat. No.5, 565, 332).
  • CDR-grafting EP 239,400
  • PCT publication WO91/09967 U.S. Pat. Nos. 5,225,539; 5,530,101; and 5,585,089
  • veneering or resurfacing EP 592,106; EP 519,596
  • chain shuffling U.S. Pat. No.5, 565, 332
  • the general recombinant DNA technology for preparation of such antibodies is also known (see European Patent Application EP 125023 and International Patent Application WO 96/02576).
  • the Fab of the present invention can be obtained by treating an antibody which specifically reacts with human HER3 with a protease, papain. Also, the Fab can be produced by inserting DNA encoding Fab of the antibody into a vector for prokaryotic expression system, or for eukaryotic expression system, and introducing the vector into a procaryote or eucaryote (as appropriate) to express the Fab.
  • the F(ab')2 of the present invention can be obtained treating an antibody which specifically reacts with human HER3 with a protease, pepsin. Also, the F(ab')2 can be produced by binding Fab' described below via a thioether bond or a disulfide bond.
  • the Fab' of the present invention can be obtained treating F(ab')2 which specifically reacts with human HER3 with a reducing agent, dithiothreitol.
  • the Fab' can be produced by inserting DNA encoding Fab' fragment of the antibody into an expression vector for prokaryote, or an expression vector for eukaryote, and introducing the vector into a prokaryote or eukaryote (as appropriate) to perform its expression.
  • the scFv of the present invention can be produced by obtaining cDNA encoding the
  • CDR grafting involves selecting the complementary determining regions (CDRs) from a donor scFv fragment, and grafting them onto a human scFv fragment framework of known three dimensional structure (see, e.
  • Amino acid sequence modification(s) of the antibodies described herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody. It is known that when a humanized antibody is produced by simply grafting only CDRs in VH and VL of an antibody derived from a non-human animal in FRs of the VH and VL of a human antibody, the antigen binding activity is reduced in comparison with that of the original antibody derived from a non-human animal.
  • the reduced antigen binding activity could be increased by replacing the identified amino acids with amino acid residues of the original antibody derived from a non- human animal. Modifications and changes may be made in the structure of the antibodies of the present invention, and in the DNA sequences encoding them, and still obtain a functional molecule that encodes an antibody with desirable characteristics.
  • the hydropathic index of amino acids may be considered.
  • the importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art. It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
  • Each amino acid has been assigned a hydropathic index on the basis of their hydrophobicity and charge characteristics these are: isoleucine (+4.5); valine (+4.2); leucine (+3.8) ; phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (- 0.4); threonine (-0.7); serine (-0.8); tryptophane (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamate (-3.5); glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
  • a further object of the present invention also encompasses function-conservative variants of the antibodies of the present invention.
  • “Function-conservative variants” are those in which a given amino acid residue in a protein or enzyme has been changed without altering the overall conformation and function of the polypeptide, including, but not limited to, replacement of an amino acid with one having similar properties (such as, for example, polarity, hydrogen bonding potential, acidic, basic, hydrophobic, aromatic, and the like).
  • Amino acids other than those indicated as conserved may differ in a protein so that the percent protein or amino acid sequence similarity between any two proteins of similar function may vary and may be, for example, from 70 % to 99 % as determined according to an alignment scheme such as by the Cluster Method, wherein similarity is based on the MEGALIGN algorithm.
  • a "function-conservative variant" also includes a polypeptide which has at least 60 % amino acid identity as determined by BLAST or FASTA algorithms, preferably at least 75 %, more preferably at least 85%, still preferably at least 90 %, and even more preferably at least 95%, and which has the same or substantially similar properties or functions as the native or parent protein to which it is compared.
  • Two amino acid sequences are "substantially homologous” or “substantially similar” when greater than 80 %, preferably greater than 85 %, preferably greater than 90 % of the amino acids are identical, or greater than about 90 %, preferably grater than 95 %, are similar (functionally identical) over the whole length of the shorter sequence.
  • the similar or homologous sequences are identified by alignment using, for example, the GCG (Genetics Computer Group, Program Manual for the GCG Package, Version 7, Madison, Wisconsin) pileup program, or any of sequence comparison algorithms such as BLAST, FASTA, etc.
  • amino acids may be substituted by other amino acids in a protein structure without appreciable loss of activity. Since the interactive capacity and nature of a protein define the protein's biological functional activity, certain amino acid substitutions can be made in a protein sequence, and, of course, in its DNA encoding sequence, while nevertheless obtaining a protein with like properties. It is thus contemplated that various changes may be made in the antibodies sequences of the invention, or corresponding DNA sequences which encode said antibodies, without appreciable loss of their biological activity. It is known in the art that certain amino acids may be substituted by other amino acids having a similar hydropathic index or score and still result in a protein with similar biological activity, i.e. still obtain a biological functionally equivalent protein.
  • amino acid substitutions are generally therefore based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
  • Exemplary substitutions which take various of the foregoing characteristics into consideration are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
  • variable domain comprises:
  • VH-CDR1 having at least 90% or 95% identity with the VH-CDR1 of of the VH chain of the antibody obtainable from hybridoma deposited as CNCM-I-4666,
  • VH-CDR2 having at least 90% or 95% identity with the VH-CDR2 of of the VH chain of the antibody obtainable from hybridoma deposited as CNCM-I-4666,
  • VH-CDR3 having at least 90% or 95% identity with the VH-CDR3 of of the VH chain of the antibody obtainable from hybridoma deposited as CNCM-I-4666
  • a VL-CDR1 having at least 90% or 95% identity with the VL-CDR1 of of the VL chain of the antibody obtainable from hybridoma deposited as CNCM-I-4666
  • VL-CDR2 having at least 90% or 95% identity with the VL-CDR2 of of the VL chain of the antibody obtainable from hybridoma deposited as CNCM-I-4666,
  • VL-CDR3 having at least 90% or 95% identity with the VL-CDR3 of of the VL chain of the antibody obtainable from hybridoma deposited as CNCM-I-4666,
  • VL variable light chain
  • VH variable heavy chain
  • the biological activities of the antibody of the invention are, for example, to reduce the phosphorylated Tau level and its pathological aggregation as described above.
  • the evaluation of the phosphorylated Tau level and its pathological aggregation allows to determine the therapeutic properties of the antibody such as the correction of cognitive impairment produced by Tauopathy.
  • Said antibodies may be assayed for specific binding by any method known in the art.
  • Many different competitive binding assay format(s) can be used for epitope binding.
  • the immunoassays which can be used include, but are not limited to, competitive assay systems using techniques such western blots, radioimmunoassays, ELISA, "sandwich” immunoassays, immunoprecipitation assays, precipitin assays, gel diffusion precipitin assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and complement-fixation assays.
  • Such assays are routine and well known in the art (see, e.g., Ausubel et al., eds, 1994 Current Protocols in Molecular Biology, Vol. 1, John Wiley & sons, Inc., New York).
  • the BIACORE® GE Healthcare, Piscaataway, NJ
  • routine cross-blocking assays such as those described in Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory, Ed Harlow and David Lane, 1988, can be performed.
  • Another type of amino acid modification of the antibody of the invention may be useful for altering the original glycosylation pattern of the antibody.
  • altering is meant deleting one or more carbohydrate moieties found in the antibody, and/or adding one or more glycosylation sites that are not present in the antibody.
  • Glycosylation of antibodies is typically N-linked.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X- serine and asparagines-X- threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • glycosylation sites are conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the sugar(s) may be attached to (a) arginine and histidine, (b) free carboxyl groups, (c) free sulfhydryl groups such as thoseof cysteine, (d) free hydroxyl groups such as those of serine, threonine, orhydroxyproline, (e) aromatic residues such as those of phenylalanine, tyrosine, or tryptophan, or (f) the amide group of glutamine.
  • Removal of any carbohydrate moieties present on the antibody may be accomplished chemically or enzymatically.
  • Chemical deglycosylation requires exposure of the antibody to the compound trifluoromethanesulfonic acid, or an equivalent compound. This treatment results in the cleavage of most or all sugars except the linking sugar (N-acetylglucosamine or N-acetylgalactosamine), while leaving the antibody intact.
  • Chemical deglycosylation is described by Sojahr H. et al. (1987) and by Edge, AS. et al. (1981).
  • Enzymatic cleavage of carbohydrate moieties on antibodies can be achieved by the use of a variety of endo-and exo- glycosidases as described by Thotakura, NR. et al. (1987).
  • Another type of covalent modification of the antibody comprises linking the antibody to one of a variety of non proteinaceous polymers, eg. , polyethylene glycol, polypropylene glycol, or polyoxyalkylenes, in the manner set forth in US Patent Nos. 4,640, 835; 4,496, 689; 4,301, 144; 4,670, 417; 4,791, 192 or 4,179,337.
  • non proteinaceous polymers eg. , polyethylene glycol, polypropylene glycol, or polyoxyalkylenes
  • Immunoconjugates An antibody of the invention can be conjugated with a detectable label to form an immunoconjugate.
  • Suitable detectable labels include, for example, a radioisotope, a fluorescent label, a chemiluminescent label, an enzyme label, a bioluminescent label or colloidal gold. Methods of making and detecting such detectably-labeled immunoconjugates are well-known to those of ordinary skill in the art, and are described in more detail below.
  • the detectable label can be a radioisotope that is detected by autoradiography.
  • Isotopes that are particularly useful for the purpose of the present invention are 3 125 131 35 S and 14 C.
  • Immunoconjugates can also be labeled with a fluorescent compound.
  • the presence of a fluorescently-labeled antibody is determined by exposing the immunoconjugate to light of the proper wavelength and detecting the resultant fluorescence.
  • Fluorescent labeling compounds include fluorescein isothiocyanate, rhodamine, phycoerytherin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine.
  • immunoconjugates can be detectably labeled by coupling an antibody to a chemiluminescent compound.
  • the presence of the chemiluminescent-tagged immunoconjugate is determined by detecting the presence of luminescence that arises during the course of a chemical reaction.
  • chemiluminescent labeling compounds include luminol, isoluminol, an aromatic acridinium ester, an imidazole, an acridinium salt and an oxalate ester.
  • Bioluminescent compound can be used to label immunoconjugates of the present invention.
  • Bioluminescence is a type of chemiluminescence found in biological systems in which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence.
  • Bioluminescent compounds that are useful for labeling include luciferin, luciferase and aequorin.
  • immunoconjugates can be detectably labeled by linking a monoclonal antibody to an enzyme.
  • the enzyme conjugate When the enzyme conjugate is incubated in the presence of the appropriate substrate, the enzyme moiety reacts with the substrate to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorometric or visual means.
  • enzymes that can be used to detectably label polyspecific immunoconjugates include ⁇ -galactosidase, glucose oxidase, peroxidase and alkaline phosphatase.
  • An antibody of the invention may be labelled with a metallic chemical element such as lanthanides.
  • Lanthanides offer several advantages over other labels in that they are stable isotopes, there are a large number of them available, up to 100 or more distinct labels, they are relatively stable, and they are highly detectable and easily resolved between detection channels when detected using mass spectrometry.
  • Lanthanide labels also offer a wide dynamic range of detection. Lanthanides exhibit high sensitivity, are insensitive to light and time, and are therefore very flexible and robust and can be utilized in numerous different settings. Lanthanides are a series of fifteen metallic chemical elements with atomic numbers 57-71. They are also referred to as rare earth elements. Lanthanides may be detected using CyTOF technology. CyTOF is inductively coupled plasma time-of-flight mass spectrometry (ICP-MS). CyTOF instruments are capable of analyzing up to 1000 cells per second for as many parameters as there are available stable isotope tags.
  • ICP-MS inductively coupled plasma time-of-flight mass spectrometry
  • Antibodies of the present invention and immunoconjugates can be used for detecting human Tau phosphorylated at serine 422 (e.g. Fibrillary aggregates comprising thereof), and/or evaluating its amount in a biological sample, in particular a culture medium sample, a whole blood sample, a serum sample, a plasma sample, a cerebrospinal fluid sample, or a brain tissue sample. Therefore they can be used for diagnosing all diseases associated with abnormal Tau phosphorylated at serine 422 levels, whether they are associated with Tau phosphorylated at serine 422 excess.
  • a biological sample in particular a culture medium sample, a whole blood sample, a serum sample, a plasma sample, a cerebrospinal fluid sample, or a brain tissue sample. Therefore they can be used for diagnosing all diseases associated with abnormal Tau phosphorylated at serine 422 levels, whether they are associated with Tau phosphorylated at serine 422 excess.
  • the method of detection of the invention is consequently useful for the in vitro diagnosis of tauopathy.
  • An object of the invention is a method for detecting human Tau phosphorylated at serine 422, and/or evaluating its amount in a biological sample, wherein said method comprises contacting said sample with an antibody or immunoconjugate of the invention under conditions allowing the formation of an immune complex between human Tau phosphorylated at serine 422 and said antibody/immunoconjugate, and detecting or measuring the immune complex formed.
  • the immune complex formed can be detected or measured by a variety of methods using standard techniques, including, by way of non-limitative examples, enzyme-linked immunosorbent assay (ELISA) or other solid phase immunoassays, radioimmunoassay, electrophoresis, immunofluorescence, or Western blot.
  • ELISA enzyme-linked immunosorbent assay
  • radioimmunoassay radioimmunoassay
  • electrophoresis electrophoresis
  • immunofluorescence or Western blot.
  • a further object of the invention is a method for diagnosing a tauopathy, wherein said method comprising evaluating the amount of Tau phosphorylated at serine 422, as indicated above, in a biological sample from a subject to be tested, and comparing the determined amount with a control value of Tau in a normal subject.
  • kits comprising at least one antibody of the invention or a fragment thereof.
  • Kits of the invention can contain an antibody coupled to a solid support, e.g., a tissue culture plate or beads (e.g., sepharose beads).
  • Kits can be provided which contain antibodies for detection and quantification of Tau phosphorylated at serine 422in vitro, e.g. in an ELISA or a Western blot.
  • Such antibody useful for detection may be provided with a label such as a fluorescent or radiolabel.
  • a further object of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an antibody of the invention or a fragment thereof for use in the treatment of tauopathy.
  • a further object of the invention relates to a method for treating a tauopathy comprising administering a subject in need thereof with a therapeutically effective amount of an antibody of the invention or a fragment thereof.
  • a “therapeutically effective amount” of the antibody of the invention is meant a sufficient amount of the antibody to treat said tauopathy, at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the antibodies and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific antibody employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific antibody employed; the duration of the treatment; drugs used in combination or coincidental with the specific antibody employed; and like factors well known in the medical arts. For example, it is well known within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.
  • the antibody of the invention or the fragment thereof is formulated as a pharmaceutical composition.
  • a pharmaceutical composition comprising an antibody of the invention or a fragment thereof can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the therapeutic molecule is combined in a mixture with a pharmaceutically acceptable carrier.
  • a composition is said to be a "pharmaceutically acceptable carrier” if its administration can be tolerated by a recipient patient.
  • Sterile phosphate-buffered saline is one example of a pharmaceutically acceptable carrier.
  • Formulations may further include one or more excipients, preservatives, solubilizers, buffering agents, albumin to prevent protein loss on vial surfaces, etc.
  • the form of the pharmaceutical compositions, the route of administration, the dosage and the regimen naturally depend upon the condition to be treated, the severity of the illness, the age, weight, and sex of the patient, etc.
  • the pharmaceutical compositions of the invention can be formulated for a topical, oral, parenteral, intranasal, intravenous, intramuscular, subcutaneous or intraocular.
  • an effective amount of the antibody may be dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium.
  • the pharmaceutical forms include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • An antibody of the invention can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • the antibodies of the invention may be formulated within a therapeutic mixture to comprise about 0.0001 to 1.0 milligrams, or about 0.001 to 0.1 milligrams, or about 0.1 to 1.0 or even about 10 milligrams per dose or so. Multiple doses can also be administered.
  • FIGURES
  • THY-Tau22 control mice did not.
  • THY-Tau22 mice injected with 2H9 at lOmg/kg spent significantly more time in the target quadrant supporting a beneficial effect of active immunotherapy.
  • PBS, 5 or lOmg/Kg 2H9 antibody were intraperitoneally injected in THY-Tau22 mice (5.5 months).
  • Tau plasma levels were analyzed and found significantly increased with a dose-dependent effect.
  • 2H9 Hybridoma supernatants were analyzed by serial dilution of medium using ELISA as follows: 96-well microtiter plates (Maxisorp F8; Nunc, Inc.) were coated overnight at 4°C with lOOng/well of S422-Tau peptide, pS422-Tau peptide and an irrelevant peptide (ATP synthase alpha chain; NeoMPS, France) in 50 mM NaHC03 , pH 9.6. After washes, plates were blocked and several dilutions of medium tested by use of goat anti-mouse IgG horseradish peroxidase-conjugated antibody. Tetramethyl was the substrate. Plates were measured with a spectrophotometer (Multiskan Ascent, Thermo Labsystem) at 450 nm. 2H9 antibody only recognizes the phospho-S422 Tau peptide.
  • mice were immunized with phospho-peptide (NeoMPS, France) containing the epitope Ser422 phosphorylated (in bold) and three amino-acids Tyr-Gly-Gly conjugated to KLH emulsified in Freund' s adjuvant: Y14T [Tyr-Gly-Gly- He- Asp-Met- Val-Asp- Ser(P03H2)-Pro-Gln-Leu-Ala-Thr]. Briefly, the lymphocytes from the spleen of the immunized mice were fused with myeloma cells, according to the method described in (Pandey, 2010).
  • hybridoma supernatants were screened in ELISA as described in (Troquier et al., 2012). Then, the selected hybridomas were tested by immunohistochemistry and Western blot in both brain homogenates from AD patients and THY-Tau22 mice. The selected monoclonal antibody was specific for the phosphor-Y14T peptide. It was then produced, purified by protein G affinity chromatography (HiTrap Protein G HP) and controlled for endotoxin levels (2H9 ⁇ 0,15U/mg).
  • SEQ ID N°l YGGIDM VDS (P)PQLAT (phospho-peptide used for immunization)
  • SEQ ID N°2 IDMVDS(P)PQLAT epipitope Ser422 phosphorylated
  • PBS phosphate buffered saline
  • THY-Tau22 mice aged of 5 months were anesthetized with ketamine (10 mg/mL) and xylazine (1.5 mg/mL), and then positioned on a stereotaxic frame. Two injections were made with ⁇ of monoclonal antibodies (AT8, AT100 (Inno genetics, Gent, Belgium) and 2H9) at ⁇ g ⁇ L and/or the same volume of PBS buffer or isotype control into the hippocampus in the left and right hemispheres at a rate of 0.2 ⁇ / ⁇ .
  • monoclonal antibodies AT8, AT100 (Inno genetics, Gent, Belgium) and 2H9
  • mice were sacrificed and transcardially perfused sequentially with 0.9% NaCl and 4% paraformaldehyde in 0.1 mol/L phosphate-buffered saline (pH 7.4). Brains were post fixed for 2 days in 4% paraformaldehyde and then incubated in 20% sucrose for 24 hours and finally kept frozen at - 80°C until use. Behavioral testing
  • mice were tested for hippocampus-dependent spatial memory using a two-trial Y- maze task.
  • the arms of the maze were 22cm long, 6.4cm wide and 15cm deep.
  • the floor of the maze was covered with sawdust that was mixed after each trial in order to eliminate olfactory cues.
  • Various extra-maze cues were placed on the surrounding walls. Experiments were conducted with an ambient light level of 6 lux.
  • mice were assigned to two arms (the "start arm” and the "other arm”) that they were allowed to freely explore during 5 min, without access to the third arm of the maze (the "novel arm”) blocked by an opaque door. The assignment of arms was counterbalanced within each experimental group.
  • mice were then removed from the maze and returned to their home cage for 2 min. During the test phase, mice were placed at the end of the same "start arm” and allowed to freely explore all three arms during 1 min. The amount of time spent in each of the arms was recorded using EthovisionXT (Noldus, Netherlands).
  • the start position (1, 2, 3, or 4) was pseudo- randomized across trials. Mice that failed to find the submerged platform within 2 min were guided to the platform, where they remained for 15 s before being returned to their cages. Time required to locate the hidden escape platform (escape latency) and distance traveled (path length) were recorded using a video tracking system.
  • Tau concentrations in plasma were determined at different times (6 hours; 24 hours and 1 week) following a single i.p injection using the INNOTEST® hTau Ag (Inno genetics, Belgium) that is a sandwich ELISA microplate assay for the quantitative determination of human Tau antigen in fluids.
  • Capture antibody is the AT 120 antibody and biotinylated antibodies HT7 and BT2 are detecting antibodies (Schraen-Maschke and al., 2008).
  • Photomicrographs were taken using a Leica digital camera, imported in ImageJ software (Scion) and converted to black and white images. Threshold intensity was set and kept constant and the number of pixel, expressing staining density, was determined for both THY-Tau22 and immunized THY-Tau22- mice. Quantifications were performed blindly by at least two observers and averaged from five to nine animals per group.
  • Data are presented as mean + standard error of the mean (SEM). Data were analysed using analysis of variance (ANOVA), and where appropriate, followed by post-hoc Tukey's test. Differences of p ⁇ 0.05 were considered significant. Data were analyzed by Prism Graphpad (San Diego, CA, USA).
  • THY-Tau22 mice have deficits in learning and memory in several behavioral testing including the Morris water maze and the Y-Maze at 9-12 months of age (Van der Jeugd et al., 2011; Burnouf et al., 2012). In the present work, THY-Tau22 mice were assessed for behavioral testing at 9 months of age.
  • Tau concentrations in plasma were measured one week after a single i.p injection of 2H9 at 5mg/kg - lOmg/kg or PBS in THY-Tau22.
  • Plasma from THY-Tau22 mice exhibited higher Tau concentrations than those of non-vaccinated mice (Fig 4).
  • This pathological phospho-epitope is found in multiple Tauopathies (Buissiere et al., 1999; Augustinack et al., 2002; Guillozet-Bongaarts et al., 2007) and animals models (for review Gotz et al., 2010).
  • Phosphorylation of Ser422 increases aggregation propensity (Haase et al., 2004) and correlates with tangle formation in P301L model (Deters et al., 2008) as in THY Tau22 model (Schindowski et al., 2006).
  • Tau pSer422 is early events that precede caspase cleavage (DeCalumble et al., 2010).
  • Caspases cuts Tau at the aspartate residue at position 421 (Asp421), yielding a truncated Tau protein that has been suggested to be a nucleation agent. Then immunotherapy targeting pSer422 may prevent this cleavage.
  • immunotherapy targeting pSer422 may prevent this cleavage.
  • i.p injections of an antibody raised against pS422 delayed cognitive deficits in the THY-Tau22 model as observed in the Y-Maze and the Morris Water Maze tests.
  • Immunohistochemical analysis also revealed a reduction of abnormally phosphorylated Tau proteins in the hippocampus. We are convinced that immunotherapy targeting Tau pS422 has a great potential as treatment for Tauopathies.
  • anti-Tau antibodies may easily recognize extracellular epitopes found in ghost tangles and trigger microglia-related clearance. However, they may also enter the cell through the endosome-lysosome pathway and activate mechanisms of Tau clearance as propose by several groups (Asuni et al., 2007; Krishnamurthy et al., 2011; Boimel et al., 2010).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Neurology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

Cette invention concerne des anticorps ou des fragments de ceux-ci qui se lient spécifiquement à la Tau phosphorylée à la sérine 422 (p S422) et leur utilisation pour traiter et diagnostiquer la maladie d'Alzheimer, les tauopathies et autres troubles liés à la protéine tau.
PCT/EP2013/077608 2012-12-21 2013-12-20 Anticorps spécifiques de la protéine tau phosphorylée à la sérine 422 et leurs utilisations pour traiter et diagnostiquer les tauopathies WO2014096321A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12306649.0 2012-12-21
EP12306649 2012-12-21

Publications (1)

Publication Number Publication Date
WO2014096321A1 true WO2014096321A1 (fr) 2014-06-26

Family

ID=47552858

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/077608 WO2014096321A1 (fr) 2012-12-21 2013-12-20 Anticorps spécifiques de la protéine tau phosphorylée à la sérine 422 et leurs utilisations pour traiter et diagnostiquer les tauopathies

Country Status (1)

Country Link
WO (1) WO2014096321A1 (fr)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9527909B2 (en) 2015-02-26 2016-12-27 Eli Lilly And Company Antibodies to tau and uses thereof
US9834596B2 (en) 2012-07-03 2017-12-05 Washington University Antibodies to tau
US9957317B2 (en) 2014-06-27 2018-05-01 C2N Diagnostics, Llc Humanized anti-tau antibodies
JP2018520133A (ja) * 2015-06-24 2018-07-26 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft ヒト化抗タウ(pS422)抗体及び使用法
US10358485B2 (en) 2017-10-16 2019-07-23 Eisai R&D Management Co., Ltd. Anti-Tau antibodies and uses thereof
US10465000B2 (en) 2013-12-20 2019-11-05 Hoffmann-La Roche Inc. Humanized anti-Tau(pS422) antibodies and methods of use
US10988529B2 (en) 2016-08-09 2021-04-27 Eli Lilly And Company Combination therapy
WO2023041805A1 (fr) 2021-09-20 2023-03-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés pour l'amélioration de l'efficacité d'une thérapie par inhibiteur de hdac et la prédiction de la réponse à un traitement comprenant un inhibiteur de hdac
WO2023092004A1 (fr) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions et méthodes pour le traitement de troubles liés à tau
US11839654B2 (en) 2018-07-31 2023-12-12 Eli Lilly And Company Combination therapy
WO2024133723A1 (fr) 2022-12-22 2024-06-27 Institut National de la Santé et de la Recherche Médicale Méthodes permettant de diminuer la résistance thérapeutique acquise à la chimiothérapie et/ou à la radiothérapie

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020086009A1 (en) * 1996-03-13 2002-07-04 Koichi Ishiguro Anti-phosphorylated tau protein antibodies and methods for detecting alzheimer`s disease with the use of the same
WO2010142423A2 (fr) * 2009-06-10 2010-12-16 F. Hoffmann-La Roche Ag Utilisation d'un anticorps anti-tau ps422 pour le traitement de maladies cérébrales

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020086009A1 (en) * 1996-03-13 2002-07-04 Koichi Ishiguro Anti-phosphorylated tau protein antibodies and methods for detecting alzheimer`s disease with the use of the same
WO2010142423A2 (fr) * 2009-06-10 2010-12-16 F. Hoffmann-La Roche Ag Utilisation d'un anticorps anti-tau ps422 pour le traitement de maladies cérébrales

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
TROQUIER L ET AL: "Targeting phospho-Ser422 by active Tau immunotherapy in the THY-Tau22 mouse model: A suitable therapeutic approach", CURRENT ALZHEIMER RESEARCH, SAIF ZONE, SHARJAH [U.A.] : BENTHAM, AE, vol. 9, no. 4, 1 May 2012 (2012-05-01), pages 397 - 405, XP008160263, ISSN: 1567-2050, DOI: 10.2174/156720512800492503 *
YANAMANDRA KIRAN ET AL: "Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo.", NEURON 16 OCT 2013, vol. 80, no. 2, 16 October 2013 (2013-10-16), pages 402 - 414, XP002720174, ISSN: 1097-4199 *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9834596B2 (en) 2012-07-03 2017-12-05 Washington University Antibodies to tau
US10465000B2 (en) 2013-12-20 2019-11-05 Hoffmann-La Roche Inc. Humanized anti-Tau(pS422) antibodies and methods of use
US9957317B2 (en) 2014-06-27 2018-05-01 C2N Diagnostics, Llc Humanized anti-tau antibodies
US10011653B2 (en) 2015-02-26 2018-07-03 Eli Lilly And Company Antibodies to tau and methods of treatment therewith
US9527909B2 (en) 2015-02-26 2016-12-27 Eli Lilly And Company Antibodies to tau and uses thereof
US10808027B2 (en) 2015-02-26 2020-10-20 Eli Lilly And Company DNA molecules encoding antibodies to tau and methods of making thereof
JP2018520133A (ja) * 2015-06-24 2018-07-26 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft ヒト化抗タウ(pS422)抗体及び使用法
JP2020037580A (ja) * 2015-06-24 2020-03-12 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft ヒト化抗タウ(pS422)抗体及び使用法
US10822402B2 (en) 2015-06-24 2020-11-03 Hoffmann-La Roche Inc. Humanized anti-tau(pS422) antibodies and methods of use
JP2022023149A (ja) * 2015-06-24 2022-02-07 エフ.ホフマン-ラ ロシュ アーゲー ヒト化抗タウ(pS422)抗体及び使用法
JP7257482B2 (ja) 2015-06-24 2023-04-13 エフ. ホフマン-ラ ロシュ アーゲー ヒト化抗タウ(pS422)抗体及び使用法
US10988529B2 (en) 2016-08-09 2021-04-27 Eli Lilly And Company Combination therapy
US10358485B2 (en) 2017-10-16 2019-07-23 Eisai R&D Management Co., Ltd. Anti-Tau antibodies and uses thereof
US10829547B2 (en) 2017-10-16 2020-11-10 Eisai R&D Management Co., Ltd. Anti-tau antibodies and uses thereof
US11578120B2 (en) 2017-10-16 2023-02-14 Eisai R&D Management Co., Ltd. Anti-Tau antibodies and uses thereof
US11839654B2 (en) 2018-07-31 2023-12-12 Eli Lilly And Company Combination therapy
WO2023041805A1 (fr) 2021-09-20 2023-03-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés pour l'amélioration de l'efficacité d'une thérapie par inhibiteur de hdac et la prédiction de la réponse à un traitement comprenant un inhibiteur de hdac
WO2023092004A1 (fr) 2021-11-17 2023-05-25 Voyager Therapeutics, Inc. Compositions et méthodes pour le traitement de troubles liés à tau
WO2024133723A1 (fr) 2022-12-22 2024-06-27 Institut National de la Santé et de la Recherche Médicale Méthodes permettant de diminuer la résistance thérapeutique acquise à la chimiothérapie et/ou à la radiothérapie

Similar Documents

Publication Publication Date Title
WO2014096321A1 (fr) Anticorps spécifiques de la protéine tau phosphorylée à la sérine 422 et leurs utilisations pour traiter et diagnostiquer les tauopathies
US10208111B2 (en) Alpha-synuclein antibodies and uses thereof
JP7219928B2 (ja) 抗トランスサイレチン抗体
JP6748086B2 (ja) 抗トランスサイレチン抗体
ES2925716T3 (es) Anticuerpos anti-transtiretina
ES2800827T3 (es) Procedimientos de tratamiento de una tauopatía
EP2885010A2 (fr) Méthodes de traitement d'une tauopathie
KR20210143858A (ko) 피로글루타메이트 아밀로이드-β에 대한 항체 및 이의 용도
JP2019530428A (ja) 抗トランスサイレチン抗体
JP6979147B2 (ja) 抗EphA4抗体
CN113631573A (zh) 通过靶向新Tau物种治疗Tau蛋白病的方法
US20220056118A1 (en) Antibodies to misfolded tdp-43 and methods of use
WO2020100779A1 (fr) Anticorps monoclonal se liant de manière spécifique à pser46-marcks
JP2018508193A (ja) メディンを認識する抗体
KR20250004705A (ko) 항-tdp-43 결합 분자
EA045911B1 (ru) АНТИТЕЛА К ПИРОГЛУТАМАТ-β-АМИЛОИДУ И ИХ ПРИМЕНЕНИЕ

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13819031

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13819031

Country of ref document: EP

Kind code of ref document: A1

点击 这是indexloc提供的php浏览器服务,不要输入任何密码和下载