EP3532495A1 - Anti-tau nanobodies - Google Patents

Anti-tau nanobodies

Info

Publication number
EP3532495A1
EP3532495A1 EP17798139.6A EP17798139A EP3532495A1 EP 3532495 A1 EP3532495 A1 EP 3532495A1 EP 17798139 A EP17798139 A EP 17798139A EP 3532495 A1 EP3532495 A1 EP 3532495A1
Authority
EP
European Patent Office
Prior art keywords
seq
nanobodies
tau
nanobody
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP17798139.6A
Other languages
German (de)
French (fr)
Inventor
Daniel Fagret
Marcelle MOULIN
Catherine Ghezzi
Pascale Perret
Sabine CHIERICI
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institut National de la Sante et de la Recherche Medicale INSERM
Universite Grenoble Alpes
Centre Hospitalier Universitaire Grenoble Alpes
Original Assignee
Institut National de la Sante et de la Recherche Medicale INSERM
Universite Grenoble Alpes
Centre Hospitalier Universitaire Grenoble Alpes
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 Institut National de la Sante et de la Recherche Medicale INSERM, Universite Grenoble Alpes, Centre Hospitalier Universitaire Grenoble Alpes filed Critical Institut National de la Sante et de la Recherche Medicale INSERM
Publication of EP3532495A1 publication Critical patent/EP3532495A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/22Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to anti-Tau nanobodies.
  • AD Alzheimer's disease
  • cognitive and memory tests as well as different biomarkers.
  • biomarkers Dubois et al Lancet Neurology 13, 614-629 (2014).
  • diagnostic methods to early detect and track neurodegenerative signs in the at-risk population in order to select the patients to be treated and to control the effects of treatments.
  • AD is defined by the association of progressive dementia syndrome and two characteristic brain lesions: extracellular senile plaques (PA) composed of amyloid peptide (Ab) and neurofibrillary tangles (DNFs) composed of protein aggregates Hyperphosphorylated Tau.
  • DNFs appear in enthorino-cortical structures before PAs are observed in the cortex (Braak et al., Journal of Neuropathological Experimental Neurology, 70 (11): 960-969 (201 1)).
  • the cortical distribution and the number of DNFs are correlated with memory disorders in patients with AD (Nelson et al., Journal of Neuropathological Experimental Neurology, 71 (5): 362-381 (2012).).
  • Nuclear cerebral imaging is an appropriate tool for determining the presence and progression of AD-related molecular lesions (Dubois et al., Alzheimer's & Dementia). 12 (3): 292-323 (2016)). Radioligand of PAs are already available (and Catafau Bullich // C 'n / AC / 3 and Translational Imaging (1)... 39-55 (2015)) and some radiotracers. Tracers targeting beta sheets within DNFs are currently under development.
  • 18F-AV-1451 (Ossenkoppele et al., Brain 139 (5): 1551-1567 (2016).)
  • 18F-THK51 (Jonasson et al., Journal of Nuclear Medicine, 57 ( 4): 574-581 (2016).)
  • Tau protein is normally very soluble and is in unstructured form Under certain pathological conditions, post-translational modifications of Tau, in particular hyper-phosphorylation lead the protein to structure and form aggregates: dimers, oligomers, fibers : helical pairs of filaments leading to the formation of DNFs. A prominent importance is currently attributed to Tau oligomers, both from the point of view of toxicity and the spread of AD lesions and other tauopathies.
  • ScFvs directed against the Tau protein in the form of oligomers, in particular in the form of dimers and trimers, have been described in application US 2015/0266947. These scFvs, about 30 kDa in size, are not directed against Tau as fibers.
  • nanobodies or nanobodies targeting the pathological forms, in particular the early pathological forms of Tau, more particularly Tau in the form of oligomers and optionally in the form of fibers.
  • Nbs nanobodies or nanobodies
  • the nanobodies according to the invention which are in VHH format, are small antibody fragments corresponding to the variable domains of the heavy chains of certain camelid antibodies. These are the smallest functional elements (10-15 kDa) derived from immunoglobulins possessing an antigen-binding domain and have antigen affinities of the nanomolar order. Because of their simple Fc-like domain structure and low molecular weight they are suitable tools for crossing the blood-brain barrier (Caljon et al., British Journal of Pharmacology, 165 (7): 2341-2353 (2012)). .).
  • the present invention thus relates to a nanobody which binds to the pathological forms of the Tau protein, in particular the early pathological forms of Tau, such as Tau in the form of oligomers and optionally Tau in the form of fibers.
  • the subject of the present invention is therefore a nanobodies directed against the Tau protein in the form of oligomers and directed against the Tau protein in the form of fibers.
  • the present invention also relates to a nanobodies directed against the Tau protein, said Tau protein being in the form of oligomers and said nanobodies being devoid of light chain.
  • the subject of the present invention is therefore a nanobodies directed against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers, and said nanobodies competing for binding to the Tau protein in the form of oligomers with a protein.
  • nanobodies comprising the amino acid sequences (i) GRTFSXiX 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid ⁇ ⁇ is S or R, X 2 is D or Y and X 3 is T or A ,
  • NRDPKYGNTRY SEQ ID NO: 5
  • TARRRISGTPQWHY SEQ ID NO: 8
  • the present invention also relates to a nanobody against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers and said nanobodies competing for binding to the Tau protein in the form of oligomers with a nanobody comprising:
  • the present invention also relates to nanobodies directed against the Tau protein, said nanobodies comprising the amino acid sequences:
  • NRDPKYGNTRY SEQ ID NO: 5
  • TARRRISGTPQWHY SEQ ID NO: 8
  • a functionally conservative variant of the nanobodies defined in a) or b) comprising a conservative substitution of one or two amino acids in one, two or three of the sequences SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5 respectively; , or SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8.
  • the present invention also relates to the nanobody as defined above for its use as a contrast agent in in vivo, non-invasive medical imaging for its use in diagnostic or prognostic methods, preferably a tauopathy or for its use as a medicine.
  • the subject of the invention is also the use of this nanobodies for the in vitro detection of Tau protein, in particular in pathological form, preferably an early pathological form, more particularly in the form of oligomers and optionally in the form of fibers. in a sample.
  • composition comprising this nanobody in association with a pharmaceutically acceptable vehicle.
  • tauopathy includes about twenty pathologies that share the existence of intracerebral Tau protein deposits, particularly in pathological form, and share clinical, pathological, biochemical and genetic similarities.
  • intracerebral deposits of tau protein means the presence of Tau aggregates and thus the presence of Tau in pathological form.
  • Some tauopathies such as progressive supranuclear palsy, corticobasal degeneration and Gerstmann-Straussier-Scheinker's disease, are characterized by the presence of neurofibrillary tangles (DNFs) that are indistinguishable from those of Alzheimer's disease.
  • DNFs neurofibrillary tangles
  • the term “deposits Intracerebral tau protein "or" presence of pathological forms of Tau denotes for example the presence of neurofibrillary tangles (DNFs).
  • Pick's disease for example, is characterized by spherical neural inclusions called Pick's body. These Pick bodies consist of disordered straight filaments and thus also of a pathological Tau form.
  • Neurofibrillary degeneration also referred to as neurofibrillary tangles or tangles refers to areas, for example in the cerebral cortex, within which the neuronal population has fibrillar tangles made up of paired filaments around the nucleus and in cellular processes. helically arranged (PHF) and / or straight filaments (SF).
  • PHF helically arranged
  • SF straight filaments
  • DNFs includes the aggregation of Tau, also called “fibrillar aggregation” or “fibrillogenesis”.
  • Tau protein is organized in the form of oligomers and then in the form of fibers which is organized in the form of helicoidal pairs of filaments and in the form of straight filaments.
  • the shapes of the helical pairs of filaments and the forms of straight filaments constitute the DNFs.
  • pathological form of Tau encompasses Tau protein in the form of oligomers, in the form of fibers, in the form of helicoidal pairs of filaments and in the form of straight filaments.
  • the intermediate aggregation forms i.e., Tau as oligomers, and Tau as fibers
  • Tau is toxic, with oligomers being the most toxic.
  • early pathological forms of Tau which therefore means in particular Tau in the form of oligomers and in the form of fibers, more particularly tau in oligomeric form.
  • Tau protein is an intracellular protein, however the pathological forms of Tau can, in some cases, be extracellular.
  • the pathological forms of Tau preferably Tau in the form of oligomers, are extracellular.
  • a pathological form of Tau refers to Tau in the form of oligomers, in the form of fibers, in the form of helical pairs of filaments and / or in the form of straight filaments.
  • a pathological form of Tau is an early pathological form of Tau, more particularly Tau in the form of oligomers and optionally Tau in the form of fibers, for example Tau in the form of oligomers or Tau in the form of fibers.
  • tauopathy means a disease that is associated with the existence of pathological forms of Tau, as defined above.
  • Tauopathy is selected from Alzheimer's disease, progressive supranuclear palsy (or Steele-Richardson-OIzewski's disease), corticobasal degeneration, Pick's disease, Niemann-Pick type C disease, Gerstmann-Straussler's disease - Scheinker and fronto-temporal degeneration related to a chromosome 17 mutation, preferably Alzheimer's disease, progressive supranuclear palsy, corticobasal degeneration and Gerstmann-Straussler-Scheinker's disease, especially Alzheimer's disease.
  • Tau protein refers to the Tau protein (in English: tubule-associated unit) which is a mammalian protein.
  • Tau protein is a member of the family of microtubule-associated proteins (microtubule associating protein). It is encoded by the MAPT gene located on chromosome 17 at position 17q21. Tau protein is also called MSTD; PPND; DDPAC; MAPTL; MTBT1; MTBT2; FTDP-17; PPP1 R103. In humans, Tau protein is synthesized primarily at the level of neurons.
  • Tau's primary transcript contains 16 exons. In the brain, some exons are not translated. Exons 2, 3 and 10 are alternately spliced and are specific for adult brain tissue. The alternative splicing of these 3 exons produces 6 possible combinations (2-3-10- to 2 + 3 + 10 +). At the protein level, there are therefore six isoforms of Tau proteins in the adult brain. It should be noted that the expression of Tau proteins is regulated during development. Thus, a single isoform, called fetal, is present at birth and does not include inserts coded by exons 2, 3 or 10. The other isoforms appear during the subsequent development. The length of their sequences varies from 352 to 441 amino acids.
  • the amino-terminal part of the Tau proteins also called the projection domain, has a role that is still poorly understood.
  • This projection domain may interact with the plasma membrane and some organelles such as mitochondria.
  • the carboxy-terminal domain it comprises 3 (without exon 10) or 4 (with exon 10) repetitive segments, specific binding domains (called 3R or 4R) comprising the conserved tubulin binding domain motif, which controls the stability of microtubules.
  • These R motifs are the anchor point of the tau protein on microtubules.
  • the three isoforms without the sequence encoded by exon 10 (10-) have three microtubule binding domains (3R), are named 2N3R, 1 N3R and 0N3R, and are distinguished by the amino-terminal portion.
  • the three isoforms with the sequence encoded by exon 10 have four microtubule binding domains (4R), are named 2N4R, 1 N4R and 0N4R, and are also distinguished by the amino-terminal portion.
  • 4R microtubule binding domains
  • the interaction with Tubulin dimers are stronger with this fourth domain, which further stabilizes microtubules and can modulate the length of neuritic extensions, as well as neuronal plasticity.
  • said Tau protein is selected from the group consisting of 2N4R, 1N4R, 0N4R, 2N3R, 1N3R and 0N3R isoforms.
  • the Tau protein is under the 2N4R isoform.
  • the amino acid sequence of the 2N4R isoform of the Tau protein also called Tau-F or Tau441 or htau40, with 441 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under the number d accession P10636-8 (SEQ ID NO: 21).
  • the amino acid sequence of the 1 N4R isoform of the Tau protein, also called Tau-E or Tau412, with 412 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under the number of accession P10636-7 (SEQ ID NO: 22).
  • the amino acid sequence of the 0N4R isoform of the Tau protein also called Tau-D or Tau383, with 383 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under accession number P10636-6 (SEQ ID NO: 23).
  • the amino acid sequence of the 2N3R isoform of the Tau protein, also called Tau-C or Tau410, with 410 amino acids can be found on the UniprotKb database as of November 16, 2016 under accession number P10636-5 (SEQ ID NO: 24).
  • the amino acid sequence of the 1 N3R isoform of the Tau protein, also called Tau-B or Tau 381, with 381 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under the number d accession P10636-5 (SEQ ID NO: 25).
  • the amino acid sequence of the 0N3R isoform of the Tau protein also called Fetal-Tau or Tau 352, with 352 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under the number of accession P10636-2 (SEQ ID NO: 26).
  • Tau protein is a neuronal protein that is often located in the axon, more rarely in dendrites and exceptionally in the cell body.
  • Tau is an unstructured protein and in the form of a monomer. Therefore, in the context of the present invention, the term "native Tau” means Tau protein in monomeric form and vice versa.
  • Native form is to interact with microtubules via specific binding domains (3R and 4R) and to promote assembly and stability of microtubules.
  • the interaction of Tau protein with microtubules is regulated mainly by phosphorylation.
  • Tau is a phosphoprotein that contains about 80 potential sites of phosphorylation.
  • the regulation of the phosphorylation state of the Tau protein results from the joint activities of protein kinases and protein phosphatases.
  • hyperphosphorylation of the Tau protein decreases its affinity for microtubules, which can lead to their destabilization and hence cytoskeletal disruption.
  • this hyperphosphorylation can lead to the aggregation of Tau and thus the formation of DNFs, as described above in the "tauopathy" part.
  • Abnormal phosphorylation consists of phosphorylation at sites that under physiological conditions are not affected by phosphorylation.
  • a non-physiological epitope these are, for example, the epitopes recognized by the AT100 and TG-3 antibodies.
  • hypophosphorylated means phosphorylation at physiological epitopes in greater numbers than in a normal adult brain or when for a given site a high percentage of Tau protein is phosphorylated.
  • Hyperphosphorylation of the tau protein can cause tau detachment from microtubules and an increase in intracellular tau concentration, causing the formation of oligomers and then fibers, until the helicoidal pairs of filaments and straight filaments are formed whose entanglement will constitute the fibrillar degenerations.
  • the result is an accumulation of tau protein in neurons and involves more than 20 different neurodegenerative diseases called tauopathy.
  • the accumulation of Tau protein is sometimes even the only cause of such tauopathy.
  • the inventors of the present invention have developed the nanobodies of the invention directed against non-phosphorylated Tau protein, in particular in the form of oligomers. However, as demonstrated by immunohistochemistry, the nanobodies of The invention also binds specifically to Tau protein in neurofibrillary degenerations of human brain slices in which the Tau protein is present in phosphorylated form.
  • the Tau protein is non-phosphorylated and / or phosphorylated.
  • This truncated Tau protein comprises the carboxy-terminal portion of the Tau proteins, and therefore, depending on the starting isoform, is the 3R region (R1, R2, R3, R4) or the 4R region (R1, R3 and R4).
  • This proteolysis promotes the formation of fibers and then the aggregation of pairs of helical filaments that become insoluble and are also indicative of certain Tauopathies.
  • the Tau protein is a truncated Tau protein comprising the carboxy-terminal portion of the Tau protein, particularly the R3 or R4 region of the Tau repeats, preferably the R3 region.
  • the truncated Tau protein lacks N-terminal domains.
  • the Tau protein comprises or consists of a peptide having the amino acid sequence VQIVYKPVDLSKVTSKCG (SEQ ID NO: 27) which corresponds to amino acids 306 to 323 of tau441, as defined above.
  • immunoglobulin have the same meaning and are used interchangeably.
  • the two heavy chains are linked to one another by disulfide bridges and each heavy chain is linked to a light chain by a disulfide bridge.
  • light chain There are two types of light chain: lambda ( ⁇ ) and kappa ( ⁇ ) light chains.
  • lambda
  • kappa
  • heavy chains or isotypes that determine the functional activity of an antibody: IgM, IgD, IgG, IgA and IgE.
  • Each string contains separate sequence domains.
  • the light chain has two domains: a variable domain (VL) and a constant domain (CL).
  • the heavy chain comprises four domains: a variable domain (VH) and three constant domains (CH1, CH2 and CH3, collectively called CH). Heavy (VH) and light (VL) variable regions determine binding recognition and antigen specificity.
  • the domains of the light chain (CL) and heavy (CH) constant regions confer important biological properties such as the association of antibody chains, secretion, transplacental mobility, complement binding and Fc receptor binding.
  • the Fv fragment is the N-terminal part of the Fab fragment of an immunoglobulin consisting of the variable portions of a light chain and a heavy chain.
  • the specificity of the antibody lies in the structural complementarity between the site of combination of the antibody and the antigenic determinant.
  • the antibody combining sites are made of residues that originate primarily from hypervariable regions or complementarity determining regions (CDRs). Occasionally, residues from non-hypervariable regions or "framework" (FR) regions may influence the overall structure of the domain and hence the combining site.
  • CDRs complementarity determining regions
  • FR framework
  • CDR refers to amino acid sequences, which together define the binding affinity and specificity of the natural Fv region of a native binding site of a immunoglobulin.
  • the heavy and light chains of an immunoglobulin each have three CDRs, designated H-CDR1, H-CDR2, H-CDR3 and L-CDR1, L-CDR2, L-CDR3 respectively.
  • An antigen binding site therefore includes 6 CDRs, including all the CDRs of a variable region of a heavy chain and a variable region of a light chain.
  • CDRs in the sequence of an antibody or nanobody can be determined by those skilled in the art using previously described techniques.
  • CDRs can be identified by sequencing the DNA of the antibody or nanobodies with a suitable system, such as the 3730XL DNA Analyzer and ABI PRISM BigDye Terminator cyc, and then analyzing the sequences thus obtained using bases.
  • dedicated data such as the international ImMunoGeneTics database or IMTG (Lefranc (2003) Dev Comp., Immunol 27:55) or Kabat, et al. (National Institutes of Health, Bethesda, Md., 1991), preferably IMGT.
  • framework region refers to the amino acid sequences intercalated between the CDRs.
  • nanobody In the context of the invention, the terms “nanobody”, “nanobody”, “VHH”, “VHH antibody fragment” and “unic domain antibody” are used interchangeably and denote the variable domain of the unique heavy chain. antibodies of the type found in camelids, which are naturally devoid of light chains. In the absence of light chain the nanobodies each have three CDRs, designated CDR1, CDR2, and CDR3 respectively.
  • the nanobodies according to the invention may in particular be nanobodies of camels, dromedaries, llamas or alpacas. Preferably, the nanobodies according to the invention are lamas nanobodies.
  • nanobodies directed against the Tau protein is meant here a nanobodies capable of selectively binding to the Tau protein, as defined in the “Tau” section.
  • the nanobody is specific for Tau, that is to say that it binds Tau to the exclusion of any other molecule.
  • the inventors have prepared nanobodies directed against pathological forms of the Tau protein, such as the Tau protein in the form of oligomers and optionally in the form of fibers.
  • the nanobodies can also be directed against the truncated Tau protein in fiber form.
  • the inventors have immunized llamas with a protein enriched Tau Tau protein preparation in the form of oligomers, with a protein enriched Tau protein preparation in Tau form and a Tau protein enriched truncated Tau protein preparation. truncated in the form of fibers.
  • the inventors then identified more precisely two nanobodies directed against Tau and having additional features not expected that do not have other anti-Tau nanobodies. Indeed, these two nanobodies, 2C5 and S2T2M3_E6, in particular 2C5, recognize Tau as oligomers and optionally in the form of fibers.
  • Tau protein in fiber form is also recognized when it is a truncated Tau protein as defined above.
  • these two nanobodies, 2C5 and S2T2M3_E6, in particular 2C5 do not bind to the native Tau protein in monomeric form.
  • these two nanobodies, 2C5 and S2T2M3_E6, in particular 2C5 do not bind to beta amyloid peptide fibers.
  • the nanobodies are directed against the Tau protein as oligomers.
  • oligomers in the context of the present invention relates to a Tau preparation obtained by an in vitro fibrillation method as described in the examples of this application.
  • the fibrillation of Tau (40 ⁇ l) is carried out for example at 37 ° C. in buffer, for example MOPS (3- (N-morpholino) propanesulfonic acid), typically 20 mM, typically at pH 7 in the presence typically of heparin (10 ⁇ l), NaN3 (4%) for a final volume of typically 1.5 ml.
  • Fibrillation is typically stopped by freezing the samples at -80 ° C after typically 48 hours to obtain Tau as oligomers.
  • this Tau protein preparation is enriched in Tau protein as oligomers.
  • Tau protein enriched as oligomers means a Tau protein preparation in the form of oligomers of which more than 50% of Tau proteins are in the form of oligomers. For example, more than 50%, more than 55%, more than 60%, more than 70%, more than 75%, more than 80%, more than 85%, more than 90% of Tau proteins are in the form of oligomers . It is known to those skilled in the art that an enriched preparation Tau proteins in the form of oligomers further contains Tau protein in native form and Tau protein in the form of fibers.
  • the oligomeric Tau protein is a mixture of Tau proteins in the form of monomers, in the form of oligomers and in fiber form, of which more than 50% of the Tau proteins are in the form of oligomers. Oligomers, in particular more than 55%, more than 60%, more than 70%, more than 75%, more than 80%, more than 85%, more than 90% of the Tau proteins are in the form of oligomers.
  • the oligomers preferably comprise between 2 and 45 units of Tau. Oligomers having 2 to 8 units of Tau are referred to as soluble oligomers and oligomers having more than 8 to 45 units of Tau are referred to as granular oligomers and may be soluble or insoluble. In one embodiment these oligomers can be characterized, using an atomic force microscope or electron microscope, as round points having a diameter of about 12 to 29 nm.
  • these oligomers can be characterized using size exclusion chromatography as being spherical molecules having a diameter of about 12 to 35 nm.
  • oligomer in the context of the invention refers to an aggregate or Tau polymer having from 2 to 50 Tau units, particularly 2 to 20, most preferably 2 to 12 tau units. , for example a dimer, a trimer, a tetramer, a pentamer, a hexamer, heptamer, octamer, nonamer, decamer, undecamer or dodecamer of Tau or having 20 to 45, preferably 30 to 45, units of Tau, for example at 45, preferably 38 to 42 units of Tau.
  • the tau protein is dimeric or trimeric.
  • the oligomeric Tau protein is soluble and / or non-soluble, more preferably soluble.
  • the nanobodies of the invention have an affinity for Tau protein in the form of oligomers which is ⁇ 20nM, for example ⁇ 10nM, ⁇ 8nM, ⁇ 7nM or ⁇ 5nM, for example an affinity of 0.1nM to 20 nM, in particular from 1 nM to 10 nM, or from 1 nM to 7 nM, for example 5 nM.
  • affinity is meant the binding capacity between a macromolecule and the antigen it fixes, in particular the binding capacity between a nanobody and the antigen that it fixes, for example a nanobody of the invention and the Tau protein in the pathological forms as defined above.
  • the affinity and therefore the ability of the nanobodies of the invention to bind to the Tau protein, for example in the form of oligomers or fibers, can be measured in vitro by several methods, including surface plasmon resonance (SPR). especially using a BIAcore 2000-Pharmacia Biosensor type device, Upsala, Sweden) or for example by an ELISA test, as described in the examples.
  • SPR surface plasmon resonance
  • the nanobodies of the invention further bind to Tau protein in the form of fibers.
  • fibers in the context of the invention relates to a preparation of the Tau protein obtained by a fibrillation process as described in the examples of this application.
  • the fibrillation of Tau (40 ⁇ l) is carried out for example at 37 ° C. in buffer, for example MOPS (3- (N-morpholino) propanesulfonic acid), typically 20 mM, typically at pH 7 in the presence typically of heparin (10 ⁇ l), NaN3 (4%) for a final volume of typically 1.5 ml.
  • Fibrillation is typically stopped by freezing the samples at -80 ° C after typically 72 hours to obtain Tau as fibers.
  • Such a preparation of Tau proteins in the form of fibers is therefore a preparation of Tau proteins enriched in Tau protein in the form of fibers.
  • fiber-enriched Tau means a Tau protein preparation in the form of fibers of which more than 50% of the Tau proteins are in the form of fibers. For example, more than 50%, more than 55%, more than 60%, more than 70%, more than 75%, more than 80%, more than 85%, more than 90% of the Tau proteins are in the form of fibers. It is known to a person skilled in the art that a preparation enriched with Tau proteins in the form of fibers also contains Tau protein in native form and Tau protein in the form of oligomers.
  • the Tau protein in the form of fibers is a mixture of Tau proteins in the form of monomers, in the form of oligomers and in the form of fibers, of which more than 50% of the Tau proteins are in the form of fibers, especially more than 55%, more than 60%, more than 70%, more than 75%, more than 80%, more than 85%, more than 90% of the Tau proteins are in the form of fibers.
  • Tau fibers describes a polymer or Tau aggregate obtained in an artificial manner mimicking the Tau fibrillation in vivo and thus mimicking the helical pairs of filaments and the straight filaments that make up the DNFs. Accordingly, Tau fibers as used in the context of the invention encompass pairs of helical filaments and straight filaments. By electron microscopy these fibers can be characterized as having a filamentous appearance.
  • the nanobodies of the invention have an affinity for the Tau protein in the form of fibers which is ⁇ 20nM, for example ⁇ 10nM, ⁇ 8nM, ⁇ 7nM or ⁇ 6nM, for example an affinity of 0.1nM to 20nM.
  • nM in particular from 1 nM to 10 nM, or from 1 nM to 8 nM, for example 6 nM.
  • the nanobodies of the invention further bind to the truncated Tau protein.
  • this truncated protein is in the form of fibers and may also be named R3 peptide fibers.
  • the truncated Tau protein is as defined in the "Tau" section above.
  • this truncated Tau protein comprises or consists of the amino acid sequence SEQ ID NO: 27.
  • the nanobodies of the invention have an affinity for the truncated Tau protein in the form of fibers which is ⁇ 100nM, for example ⁇ 80nM, ⁇ 70nM, ⁇ 60nM or ⁇ 50nM, for example an affinity of 1nM to 100 nM, in particular from 1 nM to 80 nM, or from 40 nM to 60 nM, for example 50 nM.
  • the nanobodies of the invention have an affinity for the truncated Tau protein in the form of fibers which is ⁇ 100 nM, for example an affinity of 1 nM to 100 nM, in particular 1 nM to 80 nM, of 1 nM at 20nM, for example an affinity of 10 to 20nM.
  • the truncated Tau protein in the form of fibers or "the peptide fibers R3" relates to an R3 peptide preparation obtained by an in vitro fibrillation method as described in the examples of this application.
  • the fibrillation of R3 (0.4 ⁇ l) is carried out for example at 37 ° C. in buffer, for example PBS (50 mM salt buffer phosphate, typically at pH 7 in the presence of heparin (0.4 ⁇ l), NaN 3 (4%) for a final volume of typically 1.5 ml Fibrillation is typically stopped by freezing the samples at -80 ° C after typically 72 hours to obtain truncated Tau (R3) as fibers.
  • buffer for example PBS (50 mM salt buffer phosphate, typically at pH 7 in the presence of heparin (0.4 ⁇ l), NaN 3 (4%) for a final volume of typically 1.5 ml
  • Fibrillation is typically stopped by freezing the samples at -80 ° C after typically 72 hours to obtain truncated Tau (R3) as fibers.
  • the affinity for the truncated Tau protein in fiber form is measured, for example, by ELISA, in, for example, a buffer phosphate buffer.
  • Saline NaCl, 137mM, KCl, 2.7mM, Na 2 HPO 4 , 10mM KH 2 PO 4 , 1.8mM
  • BSA buffer phosphate buffer
  • the inventors have also demonstrated for the nanobodies of the invention a specific labeling of the hippocampal cell bodies, the entorhinal cortex and the temporal cortex in patients with Alzheimer's disease.
  • the nanobodies of the invention additionally bind to helical pairs of filaments and / or straight filaments.
  • helical pair of filaments refers to helical paired filaments comprising at least 10, at least 12, at least 20, preferably at least 40, and in particular at least 50 or at least 60 units of Tau. Normally these filaments can be observed by electron microscopy and have a diameter of 8 to 20 nanometers, for example 10 to 20 nm and a helical pitch of about 80 nm, for example 70 to 90 nm.
  • straight filaments means filaments which are not helically matched and comprising at least 10, at least 12, at least 20, preferably at least 40 units of Tau, in particular at least 50 or at least 60 units of Tau. Tau. Normally these filaments can be observed by electron microscopy and have a diameter of about 10 nanometers, for example from 8 to 17 nm, in particular from 9 to 12 nm, for example 10 nm.
  • the helical pairs of filaments and straight filaments that make up the DNFs are not soluble.
  • the nanobodies of the invention do not interact substantially with the polymerized amyloid beta 1 -42 protein.
  • the nanobodies do not substantially interact with the Tau protein in monomeric form.
  • a nanobodies "do not substantially intercalate" with a protein for example, polymerized amyloid beta 1 -42 protein or Tau in monomeric form, when the affinities for Tau protein as an oligomer and the affinity for Tau protein under The monomer form or the polymerized amyloid beta-42 protein are very different.
  • the affinity for monomeric Tau protein can not be measured because the binding response is too weak.
  • a nanobodies do not substantially interact with the Tau protein in monomeric form or the polymerized amyloid beta 1 -42 protein, when the nanobody-binding reaction with Tau as a monomer is less than 5% of the response. binding of the same nanobodies with Tau as an oligomer in the same experimental condition and at the same concentration of the nanobody.
  • the The concentration of the nanobodies used may be the EC 50 concentration or the concentration necessary to reach the saturation plateau.
  • the affinity of the nanobodies of the invention for Tau protein in monomeric form is> 1200 nM, for example> 1400 nM,> 1600 nM,> 1800 nM, in particular> 1800 nM.
  • the affinity of the nanobodies of the invention for the polymerized amyloid beta 1 -42 protein is> 5000 nM, for example> 8000 nM,> 9000 nM,> 10000 nM, in particular> 10000 nM.
  • the 2C5 nanobodies have the amino acid sequence QVQLVQSGG
  • the nanobody S2T2M3_E6 has the amino acid sequence EVQLVESGGGLVQAGGSLRLSCAASGRTFSRYAMGWFRQAPGKEREF
  • VASISRSGGSTRYADAVKGRFTISRDNTNNTVYLLMNNLKPEDTAVYYC TARRRISGTPQWHYWGQGTQVTVSS (SEQ ID NO: 10).
  • o CDR2 ISPSGGVT (SEQ ID NO: 4)
  • CDR1, CDR2 and CDR3 are sufficient to define an antigen binding site. It is also known to those skilled in the art that two nanobodies that recognize the same antigen compete for binding to this antigen.
  • an object of the invention relates to a nanobodies directed against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers and said nanobodies competing for binding to the Tau protein in the form of oligomers with a nanobody including amino acid sequences (i) GRTFSX1X 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid ⁇ ⁇ is S or R, X 2 is D or Y and X 3 is T or A,
  • NRDPKYGNTRY SEQ ID NO: 5
  • TARRRISGTPQWHY SEQ ID NO: 8
  • a candidate nanobicide to compete for Tau protein binding for example in the form of oligomers
  • a nanobodies comprising the CDRs of a 2C5 and / or S2T2M3_E6 nanobodies as defined above, ( hereinafter a "reference" nanobodies)
  • the antigen ie Tau protein in the form of oligomers
  • two solutions containing the nanobody The candidate and the reference nanobodies, respectively, are added and the nanobodies will compete to bind to the antigen.
  • the amount of reference nanobodies bound to the antigen can then be measured and compared to the amount of reference nanobodies bound to the antigen when measured against a negative control (eg solution free of candidate nanobodies).
  • a negative control eg solution free of candidate nanobodies.
  • An amount of bound reference nanobodies in the presence of candidate nanobodies that is decreased relative to the amount of bound reference nanobodies in the presence of the negative control indicates that the candidate nanobodies compete for binding to Tau protein as oligomers.
  • the reference nanobodies can be labeled (e.g. by fluorescence) to facilitate the detection of bound reference nanobodies. Repeated measurements can be performed with successive dilutions of the candidate and / or reference nanobodies.
  • the invention in another subject of the invention, relates to a nanobodies directed against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers and said nanobodies competing for binding to the Tau protein under oligomer form with a nanobodies selected from nanobodies comprising: a) amino acid sequences (i) GRTFSSDT (SEQ ID NO: 3) such as CDR1, (ii) ISPSGGVT (SEQ ID NO: 4) such as CDR2 and ( iii) NRDPKYGNTRY (SEQ ID NO: 5) as CDR3; or
  • An object of the present invention relates to a nanobody against protein
  • Tau comprising the amino acid sequences (i) GRTFSX1X 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid ⁇ ⁇ is S or R, X 2 is D or Y and X 3 is T or A,
  • NRDPKYGNTRY SEQ ID NO: 5
  • TARRRISGTPQWHY SEQ ID NO: 8
  • the invention also relates to a nanobodies directed against the Tau protein comprising: a) the amino acid sequences (i) GRTFSSDT (SEQ ID NO: 3) as CDR1, (ii) ISPSGGVT (SEQ ID NO: 4) as CDR2 and (iii) NRDPKYGNTRY (SEQ ID NO: 5) as CDR3; or
  • a functionally conservative variant of the nanobodies defined in a) or b) comprising a conservative substitution of one or two amino acids in one, two or three of the sequences SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5 respectively; , or SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8.
  • this Tau protein is in the form of oligomers and optionally in the form of fibers.
  • the inventors have also sequenced the "framework" (FR) regions of the 2C5 and S2T2M2 E6 nanobodies.
  • the corresponding sequences are as follows:
  • Region FR1 QVQLVQSGGGLVQAGGSLRLSCAAS (SEQ ID NO: 1 1)
  • Region FR2 LAWFRQAPGKEREFVAS (SEQ ID NO: 12)
  • o Region FR3 YYEDSVKGRFTISRDNSKNTVLLQMNSLTPEDTAVYYC (SEQ ID NO: 13)
  • Region FR1 EVQLVESGGGLVQAGGSLRLSCAAS (SEQ ID NO: 15)
  • Region FR2 MGWFRQAPGKEREFVAS (SEQ ID NO: 16)
  • Region FR3 RYADAVKGRFTISRDNTNNTVYLLMNNLKPEDTAVYYC (SEQ ID NO: 17)
  • Region FR4 WGQGTQVTVSS (SEQ ID NO: 18)
  • the invention relates to a nanobody against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers and said nanobodies competing for binding to the Tau protein as oligomers with a nanobodies selected from a nanobodies comprising an amino acid sequence selected from the group consisting of SEQ amino acid sequences ID NO: 9 and SEQ ID NO: 10.
  • the invention relates to a nanobody comprising or consisting of the sequence of FR1 -CDR1 -FR2-CDR2-FR3-CDR3-FR4 sequences as defined above of one of the nanobodies identified by the inventors.
  • the nanobody according to the invention is therefore a nanobodies comprising or consisting of an amino acid sequence selected from the group consisting of the amino acid sequences SEQ ID NO: 9 and SEQ ID NO: 10 or a functionally variant conservative thereof comprising a conservative substitution of one or two amino acids in one, two or three of the CDRs included respectively in the amino acid sequence SEQ ID NO: 9 or SEQ ID NO: 10.
  • the functionally conservative variant as as defined above may further comprise one or more substitutions, in particular one or more conservative substitutions in the regions respectively of the amino acid sequences SEQ ID NO: 9 or SEQ ID NO: 10 which are not CDRs, such as that the regions "frame", especially the "framework” regions defined above.
  • the nanobody according to the invention is a nanobodies comprising or consisting of an amino acid sequence selected from the group consisting of amino acid sequences SEQ ID NO: 9 and SEQ ID NO: 10.
  • the nanobody according to the invention is a nanobody comprising or consisting of the amino acid sequence SEQ ID NO: 9.
  • the term "functionally conservative variant” refers to variants in which a given amino acid in a nanobody according to the invention is substituted without altering the overall conformation and function of the nanobody, including replacement. from one amino acid to another having similar properties (eg polarity, hydrogen bonding potential, acidity, basicity, hydrophobicity, presence of an aromatic group, etc.).
  • Amino acids with similar properties are well known to those skilled in the art. For example, arginine, histidine and lysine of hydrophilic-basic amino acids and may be interchangeable.
  • isoleucine, a hydrophobic amino acid may be replaced by leucine, methionine or valine.
  • a natural amino acid can be replaced by an unnatural amino acid, such as an amino acid in D-configuration, beta-amino acid or gamma. Examples of conservative substitutions are shown in Table 1 below.
  • conservative amino acids can be grouped as described in Lehninger (1975, Biochemistry, 2nd Edition, Worth Publishers, Inc. New York: NY, pp. 71-77), as shown in Table 2 below.
  • these functionally conservative variants retain the ability to bind the Tau protein, in particular the Tau protein in oligomeric form and optionally in the form of fibers.
  • these functionally conservative variants have a binding affinity with Tau, in particular with Tau in the form of an oligomer and optionally in the form of fibers, which is equal to or greater than that of the corresponding nanobody.
  • nanobodies of interest are capable of producing the nanobodies according to the invention, in particular the 2C5 and S2T2M2 E6 nanobodies defined above, by conventional techniques for the production of polypeptides.
  • they can be synthesized using the well known method of solid phase synthesis (Merrifield (1996) Proc Soc Ex Boil 21: 412 Merrifield (1963) J. Am Chem Soc 85: 2149 Tarn et al (1983) J Am Chem Soc 105: 6442), preferably using a commercially available peptide synthesizer (such as that made by Applied Biosystems, Foster City, Calif.) And following the manufacturer's instructions.
  • the nanobodies according to the invention can be synthesized by recombinant DNA techniques well known to those skilled in the art (Maniatis et al., (1982) Molecular Cloning: a laboratory manual, Cold Spring Harbor Laboratories, NY, 51- 54 and 412-430). For example, they can be obtained as DNA expression products after incorporation of DNA sequences encoding the polypeptide of interest into expression vectors and introduction of these vectors into appropriate prokaryotic or eukaryotic hosts which will express the polypeptide. of interest, from which they can then be isolated using techniques well known to those skilled in the art.
  • the nanobodies according to the invention comprise a myc and / or hexahistidine label.
  • an object of the invention is also constituted by a nanobody comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 9, and SEQ ID NO: 10 further comprising at their C-terminus or N-terminus, preferably at their C-terminus, a myc tag and / or six histidine residues, more preferably a myc tag and six histidine residues.
  • a protein when associated with a label facilitating its purification, such a protein comprises between the native sequence and this label a sequence allowing an enzymatic cleavage between the protein and this label.
  • a nanobody comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 19 and SEQ ID NO: 20 is also part of the invention.
  • Another subject of the invention relates to a nucleic acid comprising a nucleic sequence coding for the nanobody according to the present invention.
  • the nucleic acid according to the invention comprises or consists of a nucleic sequence encoding a nanobody defined by one of the amino acid sequences SEQ ID NO: 9 or SEQ ID NO: 10.
  • the nucleic acid according to the invention comprises or consists of a nucleic acid sequence encoding the nanobodies defined by the amino acid sequence SEQ ID NO: 9.
  • said nucleic acid is a DNA or RNA molecule, which can be included in any suitable vector, such as a plasmid, a cosmid, an episome, an artificial chromosome, a phage or a viral vector .
  • vector means the vehicle by which the DNA or RNA sequence can be introduced into the host cell, so as to transform the host and promote the expression (eg transcription and translation) of the introduced sequence.
  • Another object of the invention relates to a vector comprising a nucleic acid according to the invention.
  • Such vectors may include regulatory elements, such as a promoter, activator, terminator, etc., to cause or direct the expression of the polypeptide.
  • regulatory elements such as a promoter, activator, terminator, etc.
  • promoters and enhancers used in animal cell expression vectors include the SV40 early promoter and activator (Mizukami et al (1987) J. Biochem 101: 1307-1310), the LTR promoter. and Moloney mouse leukemia virus activator, the promoter (Mason et al (1985) Ce / 41: 479-487) and the enhancer (Gillies et al (1983) Cell 33: 717- 728) of the immunoglobulin chain, etc.
  • Any expression vector for animal cells can be used.
  • suitable vectors include pAGE107 (Miyaji et al (1990) Cytotechnology 3: 133-140), pAGE103 (Mizukami et al (1987) J. Biochem 101: 1307-1310), pHSG274. (Brady et al (1984) Gene 27: 223-232), pKCR (O'Hare et al (1981) Proc Natl Acad Sci USA 78: 1527-1531), pSG1 beta d2-4 (Miyaji et al (1990) Cytotechnology 3: 133-140), etc.
  • Plasmids include replicative plasmids comprising an origin of replication, or integrative plasmids, such as for example pUC, pcDNA, pBR, etc.
  • viral vectors include adenoviral, retroviral, herpesvirus and AAV vectors.
  • recombinant viruses can be produced by techniques well known to those skilled in the art, such as by transfection of packaging cells or by transient transfection with plasmids or helper viruses.
  • Typical examples of virus packaging cells include PA317 cells, PsiCRIP cells, GPenv + cells, 293 cells, and the like.
  • Detailed protocols for producing such replication-deficient recombinant viruses can be found, for example, in WO 95/14785, WO 96/22378, US 5,882,887, US 6,013,516, US 4,861,719, US 5,278,056, and WO 94/19478.
  • Another object of the present invention relates to a cell which has been transfected, transduced or transformed with a nucleic acid and / or a vector according to the invention.
  • the terminating "transformation” means the introduction of a “foreign” (extrinsic or extracellular) DNA or RNA gene or sequence into a host cell, such that the host cell will express the gene. or the sequence introduced to produce the substance of interest, typically a protein encoded by the gene or sequence introduced.
  • a host cell that receives and expresses the introduced DNA or RNA has been "transformed".
  • the nucleic acids according to the invention can be used to produce a nanobody according to the invention in an appropriate expression system.
  • expression system means a host cell and a compatible vector under appropriate conditions, e.g. for the expression of a protein encoded by the foreign DNA carried by the vector and introduced into the host cell.
  • Conventional expression systems include the host cells Escherichia co // 'and plasmid vectors, insect host cells and Baculovirus vectors, and mammalian host cells and vectors.
  • host cells include prokaryotic cells (such as bacteria), and eukaryotic cells (such as yeast cells, mammalian cells, insect cells, plant cells, etc.).
  • eukaryotic cells such as yeast cells, mammalian cells, insect cells, plant cells, etc.
  • Specific examples include Escherichia coli, Kluyveromyces or Saccharomyces yeasts, mammalian cell lines (eg Vero cells, CHO cells, 3T3 cells, COS cells, etc.) as well as primary or established mammalian cell cultures. (eg produced from lymphoblasts, fibroblasts, epithelial cells, nerve cells, adipocytes, etc.).
  • Examples also include mouse SP2 / 0-Ag14 cells (ATCC CRU 581), mouse P3X63-Ag8.653 cells (ATCC CRU 580), CHO cells in which a dihydrofolate reductase gene is defective, YB2 cells / Rat 3HL.P2.G1 1 .16Ag.20 (ATCC CRU 662), etc.
  • the present invention also relates to a method for producing a recombinant host cell expressing a nanobody according to the invention, said method comprising the steps of:
  • nanobodies according to the invention may be produced by any technique known to those skilled in the art, such as for example any chemical, biological, genetic or enzymatic technique, alone or in combination.
  • the invention furthermore relates to a method for producing a nanobodies according to the invention, said method comprising the steps of:
  • the nanobodies according to the invention can be conveniently separated from the culture medium by conventional immunoglobulin purification procedures, such as, for example, protein A-Sepharose, hydroxilapatite chromatography, gel electrophoresis, dialysis or affinity chromatography.
  • immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxilapatite chromatography, gel electrophoresis, dialysis or affinity chromatography.
  • the nanobodies according to the invention are particularly useful for medical imaging.
  • the present inventors have shown that the 2C5 and S2T2M3_E6 nanobodies, in particular 2C5, retained their properties when they were associated with a detectable marker.
  • the present invention therefore also relates to a nanobody as defined above linked to a detectable marker.
  • nanobodies bound to a detectable marker is meant herein that the detectable label is directly or indirectly linked to the nanobody, for example via a cleavable or non-cleavable linker peptide, or is incorporated into the nanobody.
  • the detectable marker may in particular be bound to the nanobodies by substitution (for example, by substituting an H for an I at the tyrosine residues), by complexing or by chelation.
  • detectable marker is meant here a compound that produces a detectable signal. When it is associated with a tracer, it makes it possible to follow the fate of the tracer in the body.
  • the detectable marker may be an MRI contrast agent, a scintigraphic contrast agent, an X-ray contrast agent, an ultrasound contrast agent, an optical imaging contrast agent.
  • detectable markers include radioelements, fluorophores such as fluorescein, Alexa, cyanine; chemiluminescent compounds such as luminol; bioluminescent compounds such as luciferase or alkaline phosphatase; and contrast agents such as nanoparticles or gadolinium.
  • the detectable label is preferably an iron oxide nanoparticle or gadolinium; when the detection system is fluorescence imaging, the detectable marker is preferably Fluorescein, Alexa or cyanine; when the detection system is chemiluminescence imaging, the detectable label is preferably luminol; when the detection system is bioluminescence imaging, the detectable label is preferably luciferase or alkaline phosphatase; when the detection system is nuclear imaging, the detectable marker is preferably a radioelement such as gallium ( 68 Ga) for PET imaging, or technetium 99m ( 99m Tc) for SPECT imaging.
  • the detectable marker is preferably a radioelement such as gallium ( 68 Ga) for PET imaging, or technetium 99m ( 99m Tc) for SPECT imaging.
  • the detectable marker is a radioelement.
  • radioelements which are more particularly used in nuclear imaging techniques, include Technetium 99m ( 99m Tc), Iodine 123 ( 123 I), 125 I ( 125 I), and fluorine 18 ( 18 F), gallium 68 ( 68 Ga), and any other radioelements usable in humans. Therefore, preferably, the radioelement is selected from the group consisting of 99m Tc, 123 l, 125 l, 18 F, and 68 Ga. Most preferably, the radioelement is 99m Tc or 68 Ga, still favorite 99m Tc.
  • the 2C5 nanobody allows specific labeling of the hippocampus, entorhinal cortex and temporal cortex cell bodies in patients suffering from Alzheimer's disease and in pure tauopathy.
  • the nanobodies of the invention in particular 2C5 constitute tracers specific for the pathological forms of the Tau protein, in particular early pathological forms of Tau, for example Tau in the form of oligomer and optionally in the form of fibers, and allow its detection by imaging.
  • the invention therefore proposes a nanobody as defined above for its use as a contrast agent in medical imaging, in particular in vivo, non-invasive medical imaging.
  • nanobody as defined above for the manufacture of a contrast agent useful for medical imaging, in particular in vivo, non-invasive medical imaging.
  • contrast agent is meant here a substance or a composition which, administered in the body, makes it possible to detectably mark organs or structures (tissue, cell, receptor) which, without a contrast agent, are or not visible in medical imaging.
  • contrast agent is used to designate a tracer associated with a marker as defined above.
  • imaging methods refer to methods that allow the visualization of the interior of an organism or organs of an organism.
  • imaging methods include invasive techniques such as endocoronary ultrasound, and non-invasive techniques such as magnetic resonance imaging, x-ray imaging, ultrasound, optical imaging, or nuclear medicine such as scintigraphy, especially single photon emission computed tomography (SPECT) and positron emission tomography (PET).
  • SPECT single photon emission computed tomography
  • PET positron emission tomography
  • the imaging method according to the invention is scintigraphy, in particular SPECT or PET scintigraphy.
  • the scintigraphy is based on the administration (usually intravenously) of a contrast agent, also called radiopharmaceutical, consisting of a tracer labeled with a radioelement.
  • a contrast agent also called radiopharmaceutical
  • the specific localization of this contrast agent in the body is then determined by detection of gamma or beta emitted radiation.
  • Single-photon emission computed tomography (PET) and positron emission tomography (PET) are scintigraphic tomography-based nuclear tomographic imaging techniques that provide three-dimensional images and reconstructions of organs and their metabolism by means of a set of cameras that revolve around the patient.
  • the present invention also relates to a medical imaging method, in particular non-invasive in vivo medical imaging, in which a nanobody as defined above is administered to a patient.
  • the medical imaging method according to the invention may further comprise the steps of detecting the link or the absence binding nanobodies in patient's body areas and viewing patient's body areas in which nanobody binding can be detected.
  • a "patient” designates a human or non-human mammal, such as a rodent (rat, mouse, rabbit), a primate (chimpanzee), a feline (cat), a canine (dog ).
  • the individual is human.
  • the term "patient” refers to a human who exhibits symptoms associated with tauopathy. Depending on the tauopathy, these symptoms may be for example Parkinson's syndrome, axial dystonia, capricious hand phenomenon, or cognitive disorders of the patient.
  • the nanobody can be administered, for example, orally, by inhalation or parenterally (in particular by intravenous injection).
  • parenteral route the nanobody can be in the form of injectable solutions and suspensions, packaged in ampoules or flasks.
  • the forms of parenteral administration are conventionally obtained by mixing the nanobodies according to the invention with buffers, stabilizers, preservatives, solubilizing agents, isotonic agents and suspending agents. According to known techniques, these mixtures can be sterilized and packaged in the form of intravenous injections.
  • buffers based on phosphate salts may for example use buffers based on phosphate salts as buffers.
  • suspending agents include methylcellulose, acacia, and sodium carbocymethylcellulose.
  • stabilizers include sodium sulfite and sodium metasulfite, and examples of preservatives include sodium p-hydroxybenzoate, sorbic acid, cresol and chlorocresol.
  • the amount of nanobodies administered depends naturally on the route of administration, the size and / or weight of the patient, and the detection technique used.
  • body area refers to a specific region of the body. It may be, for example, an organ, a part of an organ or a tissue, such as the brain, in particular the hippocampus, the entorhinal cortex or the temporal cortex.
  • the nanobody according to the invention is used as a contrast agent in medical imaging to visualize the Tau protein in pathological form, in particular to visualize Tau in the form chosen from oligomers, fibers, helicoidal pairs of filaments and straight filaments, of oligomers and fibers, more preferably oligomers, in a patient.
  • the nanobody according to the invention is used as a contrast agent in medical imaging to visualize neurofibrillary tangles (DNFs) in a patient.
  • DNFs neurofibrillary tangles
  • cortical distribution and the number of, for example, DNFs are correlated with memory disorders in patients with, for example, AD.
  • pathological forms of Tau protein is a marker of tauopathy.
  • detection of pathological forms of Tau in particular early pathological forms, such as Tau protein in the form of oligomers and possibly fibers, makes it possible to identify the presence of the toxic forms involved in the propagation or development of Tau. one of the tauopathies.
  • the possibility of imaging the evolution, that is to say the progression or regression, of a pathological form of Tau previously identified represents a modality for evaluating the efficacy of a therapeutic treatment. in a patient diagnosed with tauopathy.
  • the invention therefore also relates to a nanobody as defined above for its use in diagnostic or prognostic methods.
  • diagnosis method or “diagnosis” is meant here a method for determining whether an individual suffers from a pathology.
  • prognostic method or “prognosis” is meant here a method for determining whether an individual is at risk of developing a pathology.
  • the nanobody as defined above is used for the diagnosis or prognosis of a tauopathy.
  • the nanobody according to the invention is used for the diagnosis or prognosis of Alzheimer's disease.
  • the presence, for example, of Tau in oligomeric form exposes the subject to a risk of developing tauopathy, especially Alzheimer's disease.
  • the nanobody according to the invention can therefore be used to detect a risk of occurrence of tauopathy, in particular Alzheimer's disease, in a patient.
  • risk of occurrence is meant here the probability that an individual develops a pathology.
  • the present invention also relates to a method for diagnosing a tauopathy and / or detecting a risk of occurrence of tauopathy in a patient, said method comprising the steps of administering a nanobody as defined above to said patient and detecting said nanobodies in the body of said patient, detecting a preferential location of said nanobody in the brain indicative of tauopathy and / or risk of occurrence of tauopathy.
  • said method further comprises the steps of administering an amyloid plaque marker to said patient and detecting said amyloid plaque marker in said patient's body, detecting a preferential location of said nanobody and said marker amyloid plaques in the brain indicative of tauopathy and / or risk of occurrence of tauopathy, especially Alzheimer's disease.
  • the present invention also relates to a method for diagnosing a tauopathy, in particular Alzheimer's disease and / or to detecting a risk of occurrence of tauopathy, in particular Alzheimer's disease, in a patient. patient, said method comprising the steps of:
  • these methods of diagnosing tauopathy are in vivo methods.
  • amyloid plaque marker is meant tracers bound to a detectable marker which binds specifically to amyloid plaques and is useful as a contrast agent in non-invasive, in vivo medical imaging. Markers of amyloid plaques are, for example tracer 18F-18F as florbetapir, florbetaben 18F, 18F and 18F-flutemetanol-AZD4694 bookmark.
  • tauopathies which brain region is affected by the presence of pathological forms of Tau.
  • the localization in the brain of degeneration neurofibrillary or other pathological form of Tau and possibly amyloid plaques is described for several tauopathies, for example in Catafau, AM and Bullich, S (Clin Transi Imaging, 2015; 3 (1): 39-55. Epub 2015 Jan 21 .) or in Tranchant, C. (Medicine / Science A1997; 13: 989-97).
  • preferential localization it is meant that the amount of nanobodies detected in the brain, in particular for example at the levels of the hippocampus, frontal neocortex, entorhinal cortex and temporal cortex cell bodies, is greater than the background noise which corresponds to a non-specific localization of the nanobody in the body.
  • progressive supranuclear paralyzation is characterized by the presence of DNF in the brain stem and frontal neocortex.
  • corticobasal degeneration is characterized by the presence of DNF in the parietal cortex.
  • Alzheimer's disease is characterized by the presence of DNF at enthorino-cortical structures.
  • the invention also relates to the nanobody according to the invention for its use for the therapeutic monitoring of a tauopathy in a subject in whom a tauopathy has been diagnosed.
  • nanobodies according to the invention for the manufacture of a contrast agent useful for the therapeutic monitoring of a tauopathy in a subject in whom a tauopathy has been diagnosed.
  • therapeutic monitoring is meant here the observation of the subject's response to the treatment administered to him.
  • the therapeutic effect of a treatment is generally associated with slowing or inhibiting the progression of a disease, a reversion of the disease, or one or more symptoms associated with this disease.
  • an absence of a therapeutic effect can result in a stability or an acceleration of the progression of the disease or one or more of its symptoms.
  • the use according to the invention may comprise the steps of:
  • the "pathological form of Tau” is as defined in the "tauopathy” section above.
  • the treatment is a treatment of a taupopathy and comprises for example the use of inhibitors of Tau aggregation.
  • the treatment of a taupopathy includes the use of the nanobodies of the invention.
  • tauopathies For example extracellular toxic forms, have been shown to represent the toxic forms of Tau (Usenovic et al., 2015) involved in the propagation (Goedert et al., 2014) of such tauopathies. than AD in the brain, by contamination of the neurons. Consequently, targeting the toxic forms of Tau to inhibit their aggregation and thus the formation of helical paired filament pairs (PHFs) and straight filaments (SFs) is a promising treatment for tauopathies, particularly Alzheimer's disease.
  • PHFs helical paired filament pairs
  • SFs straight filaments
  • the invention further relates to the use of a nanobody as defined above for the manufacture of a medicament, in particular a medicament for the treatment of tauopathy.
  • a method of treatment comprising administering a therapeutically effective amount of the nanobody as defined above to a patient in need thereof is also part of the present invention.
  • treatment of a tauopathy is meant the “therapeutic treatment” (or curative) of a tauopathy, which includes slowing or inhibiting the evolution of a tauopathy.
  • the “prophylactic treatment” of tauopathy is also understood to include the prevention of DNF formation.
  • Prevention refers to preventing or delaying the onset or decreasing the intensity of the clinical or biochemical manifestations associated with tauopathy.
  • a biological parameter of interest may be the presence and location of Tau in pathological form, in particular Tau in early pathological form, such as Tau protein in the form of oligomers and optionally Tau in the form of fibers.
  • the invention more particularly relates to the nanobody as defined above for its use for the treatment of tauopathy and / or the prevention of tauopathy, preferably for the treatment and / or prevention of the disease of tauopathy. Alzheimer.
  • the invention also relates to the use of a nanobody as defined above for the manufacture of a medicament for the treatment of tauopathy and / or the prevention of tauopathy in a patient likely to present a tauopathy .
  • the invention also relates to a method of treating tauopathy and / or preventing tauopathy in a patient in need thereof, comprising administering a therapeutically effective amount of a nanobody as defined above to a patient in need.
  • the nanobodies according to the invention are used to treat the pathological forms of Tau, in particular the early pathological forms, such as the Tau oligomer forms and optionally the Tau fiber forms.
  • the nanobody according to the invention can be administered, for example, orally, by inhalation, parenterally (in particular by intravenous injection), in a suitable form.
  • parenterally in particular by intravenous injection
  • the nanobody can be in the form of injectable solutes and suspensions packaged in ampoules or flasks.
  • Forms for parenteral administration are conventionally obtained by mixing the nanobody with buffers, stabilizing agents, preservatives, solubilizing agents, isotonic agents and suspending agents. In accordance with known techniques, these mixtures are then sterilized and then packaged in the form of intravenous injections.
  • buffers those skilled in the art may use buffers based on organic phosphate salts.
  • suspending agents examples include methylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, acacia and sodium carboxymethylcellulose.
  • useful stabilizers according to the invention are sodium sulfite and sodium metasulfite, while sodium p-hydroxybenzoate, sorbic acid, cresol and chlorocresol can be mentioned as preservatives.
  • the amount of nanobodies administered depends naturally on the mode of administration, the size and / or weight of the patient, and the nature of the cytotoxic agent that may be associated with it.
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a nanobody as defined above in association with a pharmaceutically acceptable vehicle.
  • pharmaceutically or “pharmaceutically acceptable” refers to molecular entities and compositions which do not produce side, allergic or otherwise objectionable reactions when administered to a mammal, particularly a human.
  • the term "pharmaceutically acceptable carrier” includes any solvent, dispersion medium, coating, antibacterial or antifungal agent, isotonic or absorption delaying agent, and the like.
  • the use of such media and agents for pharmaceutically active substances is well known to those skilled in the art. With the exception of the case where a medium or conventional agent is incompatible with the active ingredient, its use in pharmaceutical compositions is envisaged. Additional active ingredients may also be incorporated into the compositions.
  • the present invention also relates to the use of a nanobody as defined above for the in vitro detection of Tau protein as oligomers in a sample.
  • sample is meant here a part of a larger element.
  • the sample is a substance of biological origin.
  • biological samples include, but are not limited to, parts of organs or tissues such as the brain, particularly the hippocampus, the entorhinal cortex or temporal cortex, blood, particularly blood brain, the cerebrospinal fluid.
  • a sample in the context of the invention relates to a brain sample.
  • the present invention will be further illustrated by the figures, sequences and the example below.
  • SEQ ID NO: 1 shows the consensus amino acid sequence of CDR1 of the nanobodies of the invention wherein the amino acid Xi is S or R, X 2 is D or Y and X 3 is T or A.
  • SEQ ID NO: 2 shows the consensus amino acid sequence of CDR2 of the nanobodies of the invention wherein the amino acid Xi is P or R and X 2 is S or V.
  • SEQ ID NO: 3 shows the amino acid sequence of CDR1 of 2C5 nanobody.
  • SEQ ID NO: 4 shows the amino acid sequence of CDR2 of the 2C5 nanobody.
  • SEQ ID NO: 5 shows the amino acid sequence of CDR3 of the 2C5 nanobody.
  • SEQ ID NO: 6 shows the amino acid sequence of CDR1 of the S2T2M3_E6 nanobody.
  • SEQ ID NO: 7 shows the amino acid sequence of CDR2 of the S2T2M3_E6 nanobody.
  • SEQ ID NO: 8 shows the amino acid sequence of CDR3 of the S2T2M3_E6 nanobody.
  • SEQ ID NO: 9 shows the amino acid sequence of the 2C5 nanobody variable region.
  • SEQ ID NO: 10 shows the amino acid sequence of the variable region of the S2T2M3_E6 nanobody.
  • SEQ ID NO: 11 shows the amino acid sequence of the FR1 region of the 2C5 nanobody.
  • SEQ ID NO: 12 shows the amino acid sequence of the FR2 region of the 2C5 nanobody.
  • SEQ ID NO: 13 shows the amino acid sequence of the FR3 region of the 2C5 nanobody.
  • SEQ ID NO: 14 shows the amino acid sequence of the FR4 region of the 2C5 nanobody.
  • SEQ ID NO: 15 shows the amino acid sequence of the FR1 region of the S2T2M2_E6 nanobody.
  • SEQ ID NO: 16 shows the amino acid sequence of the FR2 region of the S2T2M2_E6 nanobody.
  • SEQ ID NO: 17 shows the amino acid sequence of the FR3 region of the S2T2M2_E6 nanobody.
  • SEQ ID NO: 18 shows the amino acid sequence of the FR4 region of the S2T2M2_E6 nanobody
  • SEQ ID NO: 19 shows the amino acid sequence of the variable region of 2C5 nanobodies with a myc tag and six histidine residues.
  • SEQ ID NO: 20 shows the amino acid sequence of the S2T2M3_E6 nanowind variable region with a myc tag and six histidine residues.
  • SEQ ID NO: 21 shows the amino acid sequence of the 2N4R isoform of Tau protein, also called Tau-F, with 441 amino acids.
  • SEQ ID NO: 22 shows the amino acid sequence of the N4R 1 isoform of the Tau protein, also called Tau-E, with 412 amino acids.
  • SEQ ID NO: 23 shows the amino acid sequence of the 0N4R isoform of Tau protein, also called Tau-D, with 383 amino acids.
  • SEQ ID NO: 24 shows the amino acid sequence of the 2N3R isoform of Tau protein, also called Tau-C, with 410 amino acids.
  • SEQ ID NO: 25 shows the amino acid sequence of the 1 N3R isoform of the Tau protein, also called Tau-B, with 381 amino acids.
  • SEQ ID NO: 26 shows the amino acid sequence of the 0N3R isoform of Tau protein, also called Fetal-Tau, with 352 amino acids.
  • SEQ ID NO: 27 shows the amino acid sequence of the truncated Tau protein consisting of the 3R region of the Tau protein.
  • FIG. 1 Graph which shows the results of the ELISA tests and therefore of the affinity curves of the 2C5 nanobodies for Tau-0 (Tau oligomers), Tau-F (tau fibers), Mimes (the R3 region of Tau polymerized) and Tau-N (native tau).
  • Figure 2 Graph showing the lack of affinity for the polymerized Abeta1 -42 amyloid peptide
  • Figure 3 Comparative immunohistochemistry on sections of temporal cortex of a case of AD; A) AT8 is a monoclonal antibody directed against phosphorylated PHFs in Ser2O2 and Thr205; B) 2C5 is the nanobodies of the invention directed against Tau as oligomers C) T22 is a monoclonal antibody directed against tau oligomers, D) Ab4G8 is a monoclonal antibody directed against aggregated forms of amyloid beta peptide).
  • Figure 4 Alignment of the variable sequences of the nanobodies of the invention.
  • Figure 5 Graph which shows the results of the ELISA tests and thus affinity curves of the 2C5 nanobodies (without radiolabeling) for polymers of the R3 sequence of the tau protein.
  • Figure 6 Graph which shows the results of the ELISA tests and thus the affinity curves of the 2C5 nanobodies (radiolabeled with iodine 125) for polymers of the R3 sequence of the tau protein.
  • the cDNA (htau40) encoding the longest Tau isoform (441 aa-45.8 kDa) was cloned into plasmid pRK172, downstream of the T7 RNA polymerase promoter.
  • the recombinant plasmids were transformed into Escherichia coli BL21 bacteria (obtained from Goedert, M et al., Neuron, 3 (4): 519-526 (1989)) (bacteria provided). by the team of Prof. Baulieu, INSERM, Paris).
  • Fibrillation of Tau (40 ⁇ l) is carried out at 37 ° C. in MOPS buffer (20 mM 3- (N-morpholino) propane sulphonic acid), pH 7 in the presence of heparin (10 ⁇ l) of NaN 3 (4%). for a final volume of 1.5 mL [21] [14]. Fibrillation is analyzed as described above. Fibrillation is stopped by freezing the samples at -80 ° C after 48 hours and 72 hours. The duration of the polymerization process is suitable for the production of (a) oligomers and (b) fibers. In parallel, a peptide covering the R3 region of the Tau repeats was synthesized and polymerized in a similar manner.
  • the fibrillation of R3 (0.4 ⁇ l) is carried out at 37 ° C. in PBS buffer (50 mM saline phosphate buffer, at pH 7 in the presence of heparin (0.4 ⁇ l), NaN 3 (4%) for a volume of The fibrillation is stopped by freezing the samples at -80 ° C after typically 72 hours to obtain truncated Tau (R3) in the form of fibers.
  • a llama was immunized under the same conditions with a mixture of oligomers and Tau fibers as well as polymers of R3 ((a), (b) (c)). Three successive injections are performed in days 0, 9, 18. The blood samples are taken in days 28 and 42. The blood bags are centrifuged in a Ficoll gradient in order to separate and isolate the lymphocytes and the total RNAs are extracted.
  • Nanobodies have been characterized for their affinity and specificity through two stages.
  • tissue sections are deparaffinized and then treated in floating sections after permeabilization in 0.2% PBS-Triton buffer and unmasking of the antigen using a commercial unmixing solution (Vector H-3300).
  • the primary antibody (Nb) is applied at different concentrations ( ⁇ 20 nM) overnight at 4 ° C. After rinsing, a rabbit anti-histdine antibody is applied for 1 h at room temperature and then a third goat anti-rabbit antibody for 1 h is finally applied after rinsing.
  • the revelation is carried out through two incubation steps in the presence of avidine biotin complex (Vector ABC kit) and then di-amino-benzidine (Vector ABC kit). Control sections are systematically performed in the absence of the first antibody (Nb).
  • ELISA II Tests The nanobody 2C5 was radiolabeled with iodine 125.
  • the ELISAs are performed before (FIG. 5) and after radiolabelling (FIG. 6). It is found that after radiolabelling, the ligand retains a good affinity for its target (FIG. 6). 3. Results:
  • Nbs single variable domain camelide antibodies
  • Nb 2C5 was sequenced and characterized as VHH and characterized in vitro. It has respective affinities (Kd) of 5nM, 6nM and 50nM for oligomers, fibers and truncated polymerized forms of Tau. This Nb binds neither native tau protein (Kd> 1800 nM) nor beta amyloid peptide fibers (Kd> 10,000 nM). (Figs 1 and 2). This Nb has been tested in immunohistochemistry on sections of human brains from anatomopathology (Alzheimer's of different grades and tauopathy). It demonstrates specific staining of hippocampal, entorhinal and temporal cortex cell bodies in patients with Alzheimer's disease and in dementia with pure tauopathy (Table 4, Figure 3).
  • Nb S2T2M3 E6 was selected for its affinity for low molecular weight tau polymers.

Abstract

The invention relates to anti-Tau nanobodies.

Description

Nanocorps anti-Tau  Anti-Tau nanobody
La présente invention concerne des nanocorps anti-Tau. II existe actuellement un consensus considérant la forme sporadique de la maladie d'Alzheimer (MA) comme un continuum, depuis une forme asymptomatique préclinique jusqu'à une maladie caractérisée cliniquement au moyen de tests cognitifs et mnésiques, ainsi qu'à l'aide de différents biomarqueurs (Dubois et al. Lancet Neurology. 13, 614-629 (2014)). Cependant, le développement de nouvelles stratégies thérapeutiques exige des méthodes diagnostiques permettant de détecter de façon précoce et de suivre les signes neurodégénératifs dans la population à risque afin de choisir les patients qui doivent être traités et pour contrôler les effets des traitements. The present invention relates to anti-Tau nanobodies. There is currently a consensus that the sporadic form of Alzheimer's disease (AD) is a continuum, from a preclinical, asymptomatic form to a clinically-characterized disease using cognitive and memory tests, as well as different biomarkers (Dubois et al Lancet Neurology 13, 614-629 (2014)). However, the development of new therapeutic strategies requires diagnostic methods to early detect and track neurodegenerative signs in the at-risk population in order to select the patients to be treated and to control the effects of treatments.
La MA est définie par l'association d'un syndrome de démence progressive et de deux lésions cérébrales caractéristiques: les plaques séniles extracellulaires (PA), composées de peptide amyloïde (Ab) et les dégénérescences neurofibrillaires (DNFs) composées d'agrégats de protéine Tau hyper-phosphorylée. Les DNFs apparaissent dans les structures enthorino-corticales avant que les PAs soient observées dans le cortex (Braak et al. Journal of Neuropathological Expérimental Neurology, 70 (1 1 ) : 960-969. (201 1 )). De plus la distribution corticale et le nombre des DNFs sont corrélés avec les troubles mnésiques chez le patients atteints de MA (Nelson et al. Journal of Neuropathological Expérimental Neurology, 71 (5): 362-381 (2012).). Récemment, il a été montré que les formes oligomèriques de Tau représentaient les formes toxiques (Usenovic et al. Journal of Neuroscience. 35 (42) : 14234-14250. (2015)) impliquées dans la propagation (Goedert et al. Current Neurology and Neuroscience 14 : 495 (2014) des tauopathies, telles que la MA, par contamination de proche en proche des neurones.  AD is defined by the association of progressive dementia syndrome and two characteristic brain lesions: extracellular senile plaques (PA) composed of amyloid peptide (Ab) and neurofibrillary tangles (DNFs) composed of protein aggregates Hyperphosphorylated Tau. DNFs appear in enthorino-cortical structures before PAs are observed in the cortex (Braak et al., Journal of Neuropathological Experimental Neurology, 70 (11): 960-969 (201 1)). In addition, the cortical distribution and the number of DNFs are correlated with memory disorders in patients with AD (Nelson et al., Journal of Neuropathological Experimental Neurology, 71 (5): 362-381 (2012).). Recently, the oligomeric forms of Tau have been shown to be toxic forms (Usenovic et al., Journal of Neuroscience, 35 (42): 14234-14250 (2015)) involved in propagation (Goedert et al., Current Neurology and Neuroscience 14: 495 (2014) tauopathies, such as AD, by gradually contaminating neurons.
L'imagerie cérébrale nucléaire (tomographie à émission monophotonique (TEMP) et tomographie à émission de positon (TEP)) est un outil approprié pour déterminer la présence et l'évolution des lésions moléculaires liées à la MA (Dubois et al. Alzheimer's & Dementia. 12 (3): 292-323 (2016)). Des radioligands des PAs sont déjà disponibles (Catafau and Bullich. C//'n/'ca/ and Translational Imaging. 3 (1 ) : 39-55 (2015).) ainsi que quelques radiotraceurs. Des traceurs ciblant les feuillets beta au sein des DNFs sont actuellement en cours de développement. Deux d'entre eux, le 18F-AV-1451 (Ossenkoppele et al. Brain. 139 (5) : 1551 -1567 (2016).) et le 18F-THK51 17 (Jonasson et al. Journal of Nuclear Medicine. 57 (4) : 574-581 (2016).) pourraient faciliter la stratification des patients atteints de lésions de type DNFs. En revanche aucun ne permet la détection et le suivi des formes oligomèriques solubles de Tau. Dans les conditions physiologiques, la protéine Tau, très soluble, interagit avec les microtubules pour les stabiliser, sa phosphorylation partielle permettant son détachement alternatif des microtubules. La protéine Tau est normalement très soluble et se trouve sous forme non structurée Dans certaines conditions pathologiques, des modifications post-traductionnelles de Tau, en particulier une hyper-phosphorylation conduisent la protéine à se structurer et à former des agrégats : dimères, oligomères, fibres: paires hélicoïdales de filaments aboutissant à la formation des DNFs. Une importance prépondérante est actuellement attribuée aux oligomères de Tau, tant du point de vue de la toxicité que de la propagation des lésions de la MA et d'autres tauopathies. Nuclear cerebral imaging (single-photon emission computed tomography (SPECT) and positron emission tomography (PET)) is an appropriate tool for determining the presence and progression of AD-related molecular lesions (Dubois et al., Alzheimer's & Dementia). 12 (3): 292-323 (2016)). Radioligand of PAs are already available (and Catafau Bullich // C 'n / AC / 3 and Translational Imaging (1)... 39-55 (2015)) and some radiotracers. Tracers targeting beta sheets within DNFs are currently under development. Two of them, 18F-AV-1451 (Ossenkoppele et al., Brain 139 (5): 1551-1567 (2016).) And 18F-THK51 (Jonasson et al., Journal of Nuclear Medicine, 57 ( 4): 574-581 (2016).) Could facilitate the stratification of patients with DNF lesions. On the other hand, none allows the detection and monitoring of soluble oligomeric forms of Tau. Under physiological conditions, the highly soluble Tau protein interacts with microtubules to stabilize them, its partial phosphorylation allowing its alternative detachment of microtubules. Tau protein is normally very soluble and is in unstructured form Under certain pathological conditions, post-translational modifications of Tau, in particular hyper-phosphorylation lead the protein to structure and form aggregates: dimers, oligomers, fibers : helical pairs of filaments leading to the formation of DNFs. A prominent importance is currently attributed to Tau oligomers, both from the point of view of toxicity and the spread of AD lesions and other tauopathies.
Des scFv dirigés contre la protéine Tau sous forme d'oligomères, en particulier sous forme de dimères et trimères, ont été décrits dans la demande US 2015/0266947. Ces scFv, d'une taille d'environ 30 kDa, ne sont pas dirigés contre Tau sous forme de fibres.  ScFvs directed against the Tau protein in the form of oligomers, in particular in the form of dimers and trimers, have been described in application US 2015/0266947. These scFvs, about 30 kDa in size, are not directed against Tau as fibers.
Dans le but de développer de nouveaux marqueurs des formes pathologiques de Tau pour l'imagerie nucléaire ciblant les différentes formes agrégées de cette molécule, les présents inventeurs ont mis au point des nanobodies ou nanocorps (Nbs), en particulier des VHH, en ciblant les formes pathologiques, en particulier les formes pathologiques précoces de Tau, plus particulièrement Tau sous forme d'oligomères et éventuellement sous forme de fibres. Ces nanocorps sont ainsi susceptibles d'améliorer le diagnostic et le suivi de la MA, ainsi que l'évaluation des effets de traitements potentiels.  In order to develop new markers of pathological forms of Tau for nuclear imaging targeting the various aggregated forms of this molecule, the present inventors have developed nanobodies or nanobodies (Nbs), in particular VHHs, targeting the pathological forms, in particular the early pathological forms of Tau, more particularly Tau in the form of oligomers and optionally in the form of fibers. These nanobodies are thus likely to improve the diagnosis and monitoring of AD, as well as the evaluation of the effects of potential treatments.
Ces nanobodies permettent également de cibler et donc identifier la présence des formes pathologiques de Tau dans d'autres tauopathies que la MA. Les nanobodies selon l'invention, qui sont en format VHH, sont de petits fragments d'anticorps correspondant aux domaines variables des chaînes lourdes de certains anticorps de camélidés. Ce sont les plus petits éléments fonctionnels (10-15 kDa) dérivés des immunoglobulines possédant un domaine de liaison à l'antigène et ils présentent des affinités envers l'antigène de l'ordre du nanomolaire. Du fait de leur structure simple sans domaine de type Fc et de leur poids moléculaire faible ce sont des outils appropriés pour traverser la barrière hémato-encéphalique (Caljon et al. British Journal of Pharmacology. 165 (7) : 2341 -2353 (2012).).  These nanobodies also make it possible to target and thus identify the presence of pathological forms of Tau in other tauopathies than AD. The nanobodies according to the invention, which are in VHH format, are small antibody fragments corresponding to the variable domains of the heavy chains of certain camelid antibodies. These are the smallest functional elements (10-15 kDa) derived from immunoglobulins possessing an antigen-binding domain and have antigen affinities of the nanomolar order. Because of their simple Fc-like domain structure and low molecular weight they are suitable tools for crossing the blood-brain barrier (Caljon et al., British Journal of Pharmacology, 165 (7): 2341-2353 (2012)). .).
Résumé de l'invention Summary of the invention
La présente invention se rapporte ainsi à un nanocorps qui se lie aux formes pathologiques de la protéine Tau, en particulier les formes pathologiques précoces de Tau, telles que Tau sous forme d'oligomères et éventuellement Tau sous forme de fibres.  The present invention thus relates to a nanobody which binds to the pathological forms of the Tau protein, in particular the early pathological forms of Tau, such as Tau in the form of oligomers and optionally Tau in the form of fibers.
La présente invention a donc pour objet un nanocorps dirigé contre la protéine Tau sous forme d'oligomères et dirigé contre la protéine Tau sous forme de fibres. La présente invention concerne également un nanocorps dirigé contre la protéine Tau, ladite protéine Tau étant sous forme d'oligomères et ledit nanocorps étant dépourvu de chaîne légère. The subject of the present invention is therefore a nanobodies directed against the Tau protein in the form of oligomers and directed against the Tau protein in the form of fibers. The present invention also relates to a nanobodies directed against the Tau protein, said Tau protein being in the form of oligomers and said nanobodies being devoid of light chain.
La présente invention a donc pour objet un nanocorps dirigé contre la protéine Tau, ladite protéine Tau étant sous forme pathologique, en particulier sous forme d'oligomères et ledit nanocorps entrant en compétition pour la liaison à la protéine Tau sous forme d'oligomères avec un nanocorps comprenant les séquences d'acides aminés (i) GRTFSXiX2X3 (SEQ ID NO: 1 ) comme CDR1 dans laquelle l'acide aminé ΧΊ est S ou R, X2 est D ou Y et X3 est T ou A, The subject of the present invention is therefore a nanobodies directed against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers, and said nanobodies competing for binding to the Tau protein in the form of oligomers with a protein. nanobodies comprising the amino acid sequences (i) GRTFSXiX 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid Χ Ί is S or R, X 2 is D or Y and X 3 is T or A ,
(ii) ISX1SGGX2T (SEQ ID NO: 2) comme CDR2 dans laquelle l'acide aminé ΧΊ est P ou R et X2 est S ou V, et (ii) ISX1SGGX 2 T (SEQ ID NO: 2) as CDR2 in which the amino acid Χ Ί is P or R and X 2 is S or V, and
(iii) NRDPKYGNTRY (SEQ ID NO: 5) ou TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3.  (iii) NRDPKYGNTRY (SEQ ID NO: 5) or TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3.
La présente invention se rapporte encore à un nanocorps dirigé contre la protéine Tau, ladite protéine Tau étant sous forme pathologique, en particulier sous forme d'oligomères et ledit nanocorps entrant en compétition pour la liaison à la protéine Tau sous forme d'oligomères avec un nanocorps comprenant : The present invention also relates to a nanobody against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers and said nanobodies competing for binding to the Tau protein in the form of oligomers with a nanobody comprising:
a) les séquences d'acides aminés (i) GRTFSSDT (SEQ ID NO: 3) comme CDR1 , (ii) ISPSGGVT (SEQ ID NO: 4) comme CDR2 et (iii) NRDPKYGNTRY (SEQ ID NO: 5) comme CDR3 ; ou  a) the amino acid sequences (i) GRTFSSDT (SEQ ID NO: 3) as CDR1, (ii) ISPSGGVT (SEQ ID NO: 4) as CDR2 and (iii) NRDPKYGNTRY (SEQ ID NO: 5) as CDR3; or
b) les séquences d'acides aminés (i) GRTFSRYA (SEQ ID NO: 6) comme CDR1 , (ii) ISRSGGST (SEQ ID NO: 7) comme CDR2 et (iii) TARRRISGTPQWHY (SEQ ID NO: b) the amino acid sequences (i) GRTFSRYA (SEQ ID NO: 6) as CDR1, (ii) ISRSGGST (SEQ ID NO: 7) as CDR2 and (iii) TARRRISGTPQWHY (SEQ ID NO:
8) comme CDR3. 8) as CDR3.
La présente invention se rapporte également à des nanocorps dirigés contre la protéine Tau, lesdits nanocorps comprenant les séquences d'acides aminés : The present invention also relates to nanobodies directed against the Tau protein, said nanobodies comprising the amino acid sequences:
(i) GRTFSX1X2X3 (SEQ ID NO: 1 ) comme CDR1 dans laquelle l'acide aminé ΧΊ est S ou R, X2 est D ou Y et X3 est T ou A, (i) GRTFSX1X 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid Χ Ί is S or R, X 2 is D or Y and X 3 is T or A,
(ii) ISX1SGGX2T (SEQ ID NO: 2) comme CDR2 dans laquelle l'acide aminé ΧΊ est P ou R et X2 est S ou V, et (ii) ISX1SGGX 2 T (SEQ ID NO: 2) as CDR2 in which the amino acid Χ Ί is P or R and X 2 is S or V, and
(iii) NRDPKYGNTRY (SEQ ID NO: 5) ou TARRRISGTPQWHY (SEQ ID NO: 8) comme L'invention concerne encore des nanocorps, dirigés contre la protéine Tau, comprenant : (iii) NRDPKYGNTRY (SEQ ID NO: 5) or TARRRISGTPQWHY (SEQ ID NO: 8) as The invention also relates to nanobodies, directed against the Tau protein, comprising:
a) les séquences d'acides aminés (i) GRTFSSDT (SEQ ID NO: 3) comme CDR1 , (ii) ISPSGGVT (SEQ ID NO: 4) comme CDR2 et (iii) NRDPKYGNTRY (SEQ ID NO: 5) comme CDR3 ; ou  a) the amino acid sequences (i) GRTFSSDT (SEQ ID NO: 3) as CDR1, (ii) ISPSGGVT (SEQ ID NO: 4) as CDR2 and (iii) NRDPKYGNTRY (SEQ ID NO: 5) as CDR3; or
b) les séquences d'acides aminés (i) GRTFSRYA (SEQ ID NO: 6) comme CDR1 , (ii ISRSGGST (SEQ ID NO: 7) comme CDR2 et (iii) TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3 ; ou  b) the amino acid sequences (i) GRTFSRYA (SEQ ID NO: 6) such as CDR1, (ii ISRSGGST (SEQ ID NO: 7) as CDR2 and (iii) TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3, or
un variant fonctionnellement conservatif du nanocorps défini en a) ou b) comprenant une substitution conservative d'un ou deux acides aminés dans respectivement une, deux ou trois des séquences SEQ ID NO: 3, SEQ ID NO: 4 et SEQ ID NO: 5, ou SEQ ID NO: 6, SEQ ID NO: 7 et SEQ ID NO: 8. a functionally conservative variant of the nanobodies defined in a) or b) comprising a conservative substitution of one or two amino acids in one, two or three of the sequences SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5 respectively; , or SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8.
La présente invention concerne également le nanocorps tel que défini ci-dessus pour son utilisation comme agent de contraste dans l'imagerie médicale in vivo, non invasive, pour son utilisation dans des méthodes de diagnostic ou de pronostic, de préférence d'une tauopathie ou pour son utilisation comme médicament. The present invention also relates to the nanobody as defined above for its use as a contrast agent in in vivo, non-invasive medical imaging for its use in diagnostic or prognostic methods, preferably a tauopathy or for its use as a medicine.
L'invention a également pour objet l'utilisation de ce nanocorps pour la détection in vitro de la protéine Tau, en particulier sous forme pathologique, de préférence une forme pathologique précoce, plus particulièrement sous forme d'oligomères et éventuellement sous formes de fibres, dans un échantillon.  The subject of the invention is also the use of this nanobodies for the in vitro detection of Tau protein, in particular in pathological form, preferably an early pathological form, more particularly in the form of oligomers and optionally in the form of fibers. in a sample.
Elle concerne enfin une composition pharmaceutique comprenant ce nanocorps en association avec un véhicule pharmaceutiquement acceptable.  Finally, it relates to a pharmaceutical composition comprising this nanobody in association with a pharmaceutically acceptable vehicle.
Description détaillée de l'invention Detailed description of the invention
Tauopathie  tauopathy
Le terme «tauopathie » regroupe une vingtaine de pathologies qui ont en commun l'existence de dépôts intracérébraux de protéine Tau, en particulier sous forme pathologique, et qui partagent des similitudes cliniques, pathologiques, biochimiques et génétiques. Le terme «dépôts intracérébraux de protéine tau» signifie la présence d'agrégats de Tau et donc la présence de Tau sous forme pathologique.  The term "tauopathy" includes about twenty pathologies that share the existence of intracerebral Tau protein deposits, particularly in pathological form, and share clinical, pathological, biochemical and genetic similarities. The term "intracerebral deposits of tau protein" means the presence of Tau aggregates and thus the presence of Tau in pathological form.
Certaines tauopathies, telles que la paralysie supranucléaire progressive, la dégénérescence corticobasale et la maladie de Gerstmann-Straussier-Scheinker, sont caractérisées par la présence des dégénérescences neurofibrillaires (DNFs) qui ne se distinguent pas de celles de la maladie d'Alzheimer. Dans ce cas le terme "dépôts intracérébraux de protéine tau" ou "présence des formes pathologiques de Tau" désigne par exemple la présence des dégénérescences neurofibrillaires (DNFs). Some tauopathies, such as progressive supranuclear palsy, corticobasal degeneration and Gerstmann-Straussier-Scheinker's disease, are characterized by the presence of neurofibrillary tangles (DNFs) that are indistinguishable from those of Alzheimer's disease. In this case the term "deposits Intracerebral tau protein "or" presence of pathological forms of Tau "denotes for example the presence of neurofibrillary tangles (DNFs).
La maladie de Pick par exemple est caractérisée par des inclusions neuronales sphériques appelées corps de Pick. Ces corps de Pick sont constitués de filaments droits désorganisés et donc également d'une forme de Tau pathologique.  Pick's disease, for example, is characterized by spherical neural inclusions called Pick's body. These Pick bodies consist of disordered straight filaments and thus also of a pathological Tau form.
La « dégénérescence neurofibrillaire » (également appelée écheveaux ou enchevêtrements neurofibrillaires) désigne des zones, par exemple dans le cortex cérébral, à l'intérieur desquelles la population neuronale présente, autour du noyau et dans les prolongements cellulaires, des enchevêtrements fibrillaires constitués de filaments appariés organisés en hélice (PHF) et/ou de filaments droits (SF, en anglais « straight filaments).  "Neurofibrillary degeneration" (also referred to as neurofibrillary tangles or tangles) refers to areas, for example in the cerebral cortex, within which the neuronal population has fibrillar tangles made up of paired filaments around the nucleus and in cellular processes. helically arranged (PHF) and / or straight filaments (SF).
Il faut noter que la formation des DNFs comprend l'agrégation de Tau, également nommée «agrégation fibrillaire» ou «fibrillogenèse». Pendant ce processus, la protéine Tau s'organise sous forme d'oligomères puis sous forme de fibres qui s'organise sous forme de paires hélicoïdales de filaments et sous formes de filaments droits. Les formes des paires hélicoïdales de filaments et les formes de filaments droits constituent les DNFs.  It should be noted that the formation of DNFs includes the aggregation of Tau, also called "fibrillar aggregation" or "fibrillogenesis". During this process, the Tau protein is organized in the form of oligomers and then in the form of fibers which is organized in the form of helicoidal pairs of filaments and in the form of straight filaments. The shapes of the helical pairs of filaments and the forms of straight filaments constitute the DNFs.
Le terme «forme pathologigue de Tau» englobe la protéine Tau sous forme d'oligomères, sous forme de fibres, sous forme de paires hélicoïdales de filaments et sous forme de filaments droits. Cependant, il a été découvert que les formes d'agrégation intermédiaires, c'est-à-dire Tau sous formes d'oligomères, et Tau sous forme de fibres, sont toxiques, les oligomères étant les plus toxiques. Celles-ci sont visées par le terme «formes pathologigues précoces de Tau» qui signifie donc en particulier Tau sous forme d'oligomères et sous formes de fibres, plus particulièrement tau sous forme oligomères.  The term "pathological form of Tau" encompasses Tau protein in the form of oligomers, in the form of fibers, in the form of helicoidal pairs of filaments and in the form of straight filaments. However, it has been discovered that the intermediate aggregation forms, i.e., Tau as oligomers, and Tau as fibers, are toxic, with oligomers being the most toxic. These are referred to by the term "early pathological forms of Tau" which therefore means in particular Tau in the form of oligomers and in the form of fibers, more particularly tau in oligomeric form.
La protéine Tau est une protéine intracellulaire, cependant les formes pathologiques de Tau peuvent, dont certaines cas, être extracellulaire.  Tau protein is an intracellular protein, however the pathological forms of Tau can, in some cases, be extracellular.
Dans un mode de réalisation particulier, les formes pathologiques de Tau, de préférence Tau sous forme d'oligomères, sont extracellulaires.  In a particular embodiment, the pathological forms of Tau, preferably Tau in the form of oligomers, are extracellular.
En conséquence, dans un mode de réalisation « une forme pathologigue de Tau » se réfère à Tau sous forme d'oligomères, sous formes de fibres, sous forme de paires hélicoïdales de filaments et/ou sous formes de filaments droits. De préférence, une forme pathologique de Tau est une forme pathologique précoce de Tau, plus particulièrement Tau sous forme d'oligomères et éventuellement Tau sous forme de fibres, par exemple Tau sous forme d'oligomères ou Tau sous forme de fibres.  Accordingly, in one embodiment "a pathological form of Tau" refers to Tau in the form of oligomers, in the form of fibers, in the form of helical pairs of filaments and / or in the form of straight filaments. Preferably, a pathological form of Tau is an early pathological form of Tau, more particularly Tau in the form of oligomers and optionally Tau in the form of fibers, for example Tau in the form of oligomers or Tau in the form of fibers.
Dans un mode de réalisation, la tauopathie signifie une maladie qui est associée à l'existence des formes pathologiques de Tau, telles que définies ci-dessus. La tauopathie est choisie parmi la maladie d'Alzheimer, la paralysie supranucléaire progressive (ou la maladie de Steele-Richardson-OIzewski), la dégénérescence corticobasale, la maladie de Pick, la maladie de Niemann-Pick de type C, la maladie de Gerstmann-Straussler- Scheinker et la dégérescence fronto-temporale liée à une mutation du chromosome 17, de préférence la maladie d'Alzheimer, la paralysie supranucléaire progressive, la dégénérescence corticobasale et la maladie de Gerstmann-Straussler-Scheinker, en particulier la maladie d'Alzheimer. In one embodiment, tauopathy means a disease that is associated with the existence of pathological forms of Tau, as defined above. Tauopathy is selected from Alzheimer's disease, progressive supranuclear palsy (or Steele-Richardson-OIzewski's disease), corticobasal degeneration, Pick's disease, Niemann-Pick type C disease, Gerstmann-Straussler's disease - Scheinker and fronto-temporal degeneration related to a chromosome 17 mutation, preferably Alzheimer's disease, progressive supranuclear palsy, corticobasal degeneration and Gerstmann-Straussler-Scheinker's disease, especially Alzheimer's disease.
Tau Tau
Le terme «protéine Tau» ou «Tau» désigne la protéine Tau (en anglais : tubule- associated unit) qui est une protéine de mammifère. La protéine Tau est membre de la famille des protéines associées aux microtubules (protéines MAP (microtubule associating protein)). Elle est encodée par le gène MAPT localisé sur le chromosome 17, à la position 17q21 . La protéine Tau est également appelée MSTD; PPND; DDPAC; MAPTL; MTBT1 ; MTBT2; FTDP-17; PPP1 R103. Chez les humains, la protéine Tau est synthétisée essentiellement au niveau des neurones.  The term "Tau protein" or "Tau" refers to the Tau protein (in English: tubule-associated unit) which is a mammalian protein. Tau protein is a member of the family of microtubule-associated proteins (microtubule associating protein). It is encoded by the MAPT gene located on chromosome 17 at position 17q21. Tau protein is also called MSTD; PPND; DDPAC; MAPTL; MTBT1; MTBT2; FTDP-17; PPP1 R103. In humans, Tau protein is synthesized primarily at the level of neurons.
Le transcrit primaire de Tau contient 16 exons. Dans le cerveau, certains exons ne sont pas traduits. Les exons 2, 3 et 10 sont épissés de manière alternative et sont spécifiques du tissu cérébral adulte. L'épissage alternatif de ces 3 exons produit 6 combinaisons possibles (2-3-10- à 2+3+10+) Au niveau protéique, il y a donc six isoformes de protéines Tau dans le cerveau adulte. Il faut noter que l'expression des protéines Tau est régulée au cours du développement. Ainsi, une seule isoforme, dite fœtale, est présente à la naissance et ne comporte pas d'inserts codés par les exons 2, 3 ou 10. Les autres isoformes apparaissent au cours du développement ultérieur. La longueur de leurs séquences varie de 352 à 441 acides aminés.  Tau's primary transcript contains 16 exons. In the brain, some exons are not translated. Exons 2, 3 and 10 are alternately spliced and are specific for adult brain tissue. The alternative splicing of these 3 exons produces 6 possible combinations (2-3-10- to 2 + 3 + 10 +). At the protein level, there are therefore six isoforms of Tau proteins in the adult brain. It should be noted that the expression of Tau proteins is regulated during development. Thus, a single isoform, called fetal, is present at birth and does not include inserts coded by exons 2, 3 or 10. The other isoforms appear during the subsequent development. The length of their sequences varies from 352 to 441 amino acids.
La partie amino-terminale des protéines Tau, encore appelée domaine de projection, a un rôle encore mal connu. Ce domaine de projection pourrait interagir avec la membrane plasmique et certains organites comme les mitochondries. Quant au domaine carboxy-terminal, il comporte 3 (sans exon 10) ou 4 (avec exon 10) segments répétitifs, des domaines spécifiques de liaison (appelés 3R ou 4R) comportant le motif conservé de domaine de liaison à la tubuline, qui contrôle la stabilité des microtubules. Ces motifs R constituent le point d'ancrage de la protéine tau sur les microtubules. Les trois isoformes sans la séquence codée par l'exon 10 (10-) possèdent trois domaines de liaison aux microtubules (3R), sont nommées 2N3R, 1 N3R et 0N3R, et se distinguent par la partie amino-terminale. Les trois isoformes avec la séquence codée par l'exon 10 possèdent quatre domaines de liaison aux microtubules (4R), sont nommés 2N4R, 1 N4R et 0N4R, et se distinguent également par la partie amino-terminale. L'interaction avec les dimères de tubuline est plus forte avec ce quatrième domaine, ce qui stabilise davantage les microtubules et peut moduler la longueur des extensions neuritiques, ainsi que la plasticité neuronale. The amino-terminal part of the Tau proteins, also called the projection domain, has a role that is still poorly understood. This projection domain may interact with the plasma membrane and some organelles such as mitochondria. As for the carboxy-terminal domain, it comprises 3 (without exon 10) or 4 (with exon 10) repetitive segments, specific binding domains (called 3R or 4R) comprising the conserved tubulin binding domain motif, which controls the stability of microtubules. These R motifs are the anchor point of the tau protein on microtubules. The three isoforms without the sequence encoded by exon 10 (10-) have three microtubule binding domains (3R), are named 2N3R, 1 N3R and 0N3R, and are distinguished by the amino-terminal portion. The three isoforms with the sequence encoded by exon 10 have four microtubule binding domains (4R), are named 2N4R, 1 N4R and 0N4R, and are also distinguished by the amino-terminal portion. The interaction with Tubulin dimers are stronger with this fourth domain, which further stabilizes microtubules and can modulate the length of neuritic extensions, as well as neuronal plasticity.
Dans un mode de réalisation de l'invention, ladite protéine Tau est sélectionnée dans le groupe consistant en les isoformes 2N4R, 1 N4R, 0N4R, 2N3R, 1 N3R et 0N3R. De préférence la protéine Tau est sous l'isoforme 2N4R.  In one embodiment of the invention, said Tau protein is selected from the group consisting of 2N4R, 1N4R, 0N4R, 2N3R, 1N3R and 0N3R isoforms. Preferably the Tau protein is under the 2N4R isoform.
La séquence d'acides aminés de l'isoforme 2N4R de la protéine Tau, également appelée Tau-F ou Tau441 ou htau40, avec 441 acides aminés peut être trouvée sur la base de données UniprotKb dans sa version du 16 novembre 2016 sous le numéro d'accession P10636-8 (SEQ ID NO: 21 ).  The amino acid sequence of the 2N4R isoform of the Tau protein, also called Tau-F or Tau441 or htau40, with 441 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under the number d accession P10636-8 (SEQ ID NO: 21).
La séquence d'acides aminés de l'isoforme 1 N4R de la protéine Tau, également appelée Tau-E ou Tau412, avec 412 acides aminés peut être trouvée sur la base de données UniprotKb dans sa version du 16 novembre 2016 sous le numéro d'accession P10636-7 (SEQ ID NO: 22).  The amino acid sequence of the 1 N4R isoform of the Tau protein, also called Tau-E or Tau412, with 412 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under the number of accession P10636-7 (SEQ ID NO: 22).
La séquence d'acides aminés de l'isoforme 0N4R de la protéine Tau, également appelée Tau-D ou Tau383, avec 383 acides aminés peut être trouvée sur la base de données UniprotKb dans sa version du 16 novembre 2016 sous le numéro d'accession P10636-6 (SEQ ID NO: 23).  The amino acid sequence of the 0N4R isoform of the Tau protein, also called Tau-D or Tau383, with 383 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under accession number P10636-6 (SEQ ID NO: 23).
La séquence d'acides aminés de l'isoforme 2N3R de la protéine Tau, également appelée Tau-C ou Tau410, avec 410 acides aminés peut être trouvée sur la base de données UniprotKb dans sa version du 16 novembre 2016 sous le numéro d'accession P10636-5 (SEQ ID NO: 24).  The amino acid sequence of the 2N3R isoform of the Tau protein, also called Tau-C or Tau410, with 410 amino acids can be found on the UniprotKb database as of November 16, 2016 under accession number P10636-5 (SEQ ID NO: 24).
La séquence d'acides aminés de l'isoforme 1 N3R de la protéine Tau, également appelée Tau-B ou Tau 381 , avec 381 acides aminés peut être trouvée sur la base de données UniprotKb dans sa version du 16 novembre 2016 sous le numéro d'accession P10636-5 (SEQ ID NO: 25).  The amino acid sequence of the 1 N3R isoform of the Tau protein, also called Tau-B or Tau 381, with 381 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under the number d accession P10636-5 (SEQ ID NO: 25).
La séquence d'acides aminés de l'isoforme 0N3R de la protéine Tau, également appelée Fetal-Tau ou Tau 352, avec 352 acides aminés peut être trouvée sur la base de données UniprotKb dans sa version du 16 novembre 2016 sous le numéro d'accession P10636-2 (SEQ ID NO: 26).  The amino acid sequence of the 0N3R isoform of the Tau protein, also called Fetal-Tau or Tau 352, with 352 amino acids can be found on the UniprotKb database in its version of November 16, 2016 under the number of accession P10636-2 (SEQ ID NO: 26).
La protéine Tau est une protéine neuronale qui est localisée souvent dans l'axone, plus rarement dans les dendrites et exceptionnellement dans le corps cellulaire. Sous forme native, Tau est une protéine non structurée et sous forme d'un monomère. Par conséquent, dans le contexte de la présente invention, le terme « Tau native » signifie la protéine Tau sous forme monomérique et vice versa. La fonction des protéines tau sous forme native est d'interagir avec les microtubules via des domaines spécifiques de liaison (3R et 4R) et de favoriser l'assemblage et la stabilité des microtubules. L'interaction de la protéine Tau avec les microtubules est régulée principalement par la phosphorylation. Tau est une phosphoprotéine qui contient environ 80 sites potentiels de phosphorylation. La régulation de l'état de phosphorylation de la protéine Tau résulte des activités conjointes de protéines kinases et de protéines phosphatases. Tau protein is a neuronal protein that is often located in the axon, more rarely in dendrites and exceptionally in the cell body. In native form, Tau is an unstructured protein and in the form of a monomer. Therefore, in the context of the present invention, the term "native Tau" means Tau protein in monomeric form and vice versa. The function of tau proteins Native form is to interact with microtubules via specific binding domains (3R and 4R) and to promote assembly and stability of microtubules. The interaction of Tau protein with microtubules is regulated mainly by phosphorylation. Tau is a phosphoprotein that contains about 80 potential sites of phosphorylation. The regulation of the phosphorylation state of the Tau protein results from the joint activities of protein kinases and protein phosphatases.
En général, une hyperphosphorylation de la protéine Tau, en particulier dans les domaines spécifiques de liaison (3R et 4R), diminue son affinité pour les microtubules, ce qui peut entraîner leur déstabilisation et par conséquent une désorganisation du cytosquelette. En outre, cette hyperphosphorylation peut entraîner l'agrégation de Tau et donc la formation des DNFs, comme décrit ci-dessus dans la partie «tauopathie».  In general, hyperphosphorylation of the Tau protein, particularly in the specific binding domains (3R and 4R), decreases its affinity for microtubules, which can lead to their destabilization and hence cytoskeletal disruption. In addition, this hyperphosphorylation can lead to the aggregation of Tau and thus the formation of DNFs, as described above in the "tauopathy" part.
L'homme du métier distingue la phosphorylation anormale et l'hyperphosphorylation de la protéine Tau.  One skilled in the art distinguishes the abnormal phosphorylation and hyperphosphorylation of Tau protein.
La « phosphorylation anormale » consiste en la phosphorylation en des sites qui dans des conditions physiologiques ne sont pas concernés par la phosphorylation. On parle d'épitope non-physiologique ; ce sont par exemple les épitopes reconnus par les anticorps AT100 et TG-3.  "Abnormal phosphorylation" consists of phosphorylation at sites that under physiological conditions are not affected by phosphorylation. We speak of a non-physiological epitope; these are, for example, the epitopes recognized by the AT100 and TG-3 antibodies.
En revanche, le terme « hvperphosphorylée » signifie une phosphorylation au niveau d'épitopes physiologiques en plus grand nombre que dans un cerveau adulte normal ou lorsque pour un site donné un pourcentage élevé de protéine Tau est phosphorylé.  In contrast, the term "hyperphosphorylated" means phosphorylation at physiological epitopes in greater numbers than in a normal adult brain or when for a given site a high percentage of Tau protein is phosphorylated.
L'hyperphosphorylation de la protéine tau peut entraîner le détachement de tau des microtubules et une augmentation de la concentration intracellulaires de tau provoquant la formation d'oligomères puis de fibres, jusqu'à aboutir à la formation de paires hélicoïdales de filaments et de filaments droit dont l'enchevêtrement vont constituer les dégénérescences fibrillaires. Le résultat est une accumulation de protéine tau dans les neurones et concerne plus de 20 maladies neurodégénératives différentes dénommées tauopathie. L'accumulation de la protéine Tau est même parfois la seule cause d'une telle tauopathie.  Hyperphosphorylation of the tau protein can cause tau detachment from microtubules and an increase in intracellular tau concentration, causing the formation of oligomers and then fibers, until the helicoidal pairs of filaments and straight filaments are formed whose entanglement will constitute the fibrillar degenerations. The result is an accumulation of tau protein in neurons and involves more than 20 different neurodegenerative diseases called tauopathy. The accumulation of Tau protein is sometimes even the only cause of such tauopathy.
II est connu de l'homme du métier que l'agrégation de Tau peut être étudiée en utilisant la protéine Tau non-phosphorylée (Xu, S et al. Alzheimers Dément. 2010 Mar;6(2):1 10-7, Kumar S. et al. J Biol Chem. 2014 Jul 18;289(29):20318-32 ; Flach, K et al. J Biol Chem. 2012 Dec 21 ;287(52):43223-33).  It is known to those skilled in the art that Tau aggregation can be studied using non-phosphorylated Tau protein (Xu, S et al., Alzheimers Dément, 2010 Mar; 6 (2): 1-10-7, Kumar S. et al., J Biol Chem., 2014 Jul 18; 289 (29): 20318-32; Flach, K et al., J Biol Chem., 2012 Dec 21; 287 (52): 43223-33).
Les inventeurs de la présente invention ont mis au point les nanocorps de l'invention dirigés contre la protéine Tau non-phosphorylée, en particulier sous forme d'oligomères. Cependant, comme démontré par immunohistochimie, les nanobodies de l'invention se lient également de façon spécifique à la protéine Tau dans des dégénérescences neurofibrillaires de coupes de cerveaux humains dans lesquels la protéine Tau est présente sous forme phosphorylée. The inventors of the present invention have developed the nanobodies of the invention directed against non-phosphorylated Tau protein, in particular in the form of oligomers. However, as demonstrated by immunohistochemistry, the nanobodies of The invention also binds specifically to Tau protein in neurofibrillary degenerations of human brain slices in which the Tau protein is present in phosphorylated form.
En conséquence, dans un mode de réalisation de l'invention la protéine Tau est non-phosphorylée et/ou phosphorylée.  Accordingly, in one embodiment of the invention the Tau protein is non-phosphorylated and / or phosphorylated.
Dans certaines dépôts intracérébraux de protéine Tau, celle-ci peut également être dégradée et perdre la partie N-terminale en conservant les domaines de liaison aux microtobules. Cette protéine Tau tronquée comprend la partie carboxy-terminale des protéines Tau, et donc, selon l'isoforme de départ soit la région 3R (R1 , R2, R3, R4) soit la région 4R (R1 , R3 et R4). Cette protéolyse favorise la formation des fibres et puis l'agrégation des paires de filaments en hélice qui deviennent insolubles et sont également indicateurs de certaines Tauopathies.  In some intracerebral deposits of Tau protein, it may also be degraded and lose the N-terminal portion while retaining the microtobule binding domains. This truncated Tau protein comprises the carboxy-terminal portion of the Tau proteins, and therefore, depending on the starting isoform, is the 3R region (R1, R2, R3, R4) or the 4R region (R1, R3 and R4). This proteolysis promotes the formation of fibers and then the aggregation of pairs of helical filaments that become insoluble and are also indicative of certain Tauopathies.
En conséquence, dans une mode de réalisation la protéine Tau est une protéine Tau tronquée comprenant la partie carboxy-terminale de la protéine Tau, en particulier la région R3 ou R4 des motifs répétés de Tau, de manière préférée la région R3. De préférence, la protéine Tau tronquée est dépourvue des domaines N-terminaux. Dans une mode de réalisation particulier la protéine Tau comprend ou consiste en un peptide ayant la séquence d'acides aminés VQIVYKPVDLSKVTSKCG (SEQ ID NO : 27) qui correspond aux acides aminé 306 à 323 de tau441 , telle que défini ci-dessus. Nanocorps anti-Tau  Accordingly, in one embodiment the Tau protein is a truncated Tau protein comprising the carboxy-terminal portion of the Tau protein, particularly the R3 or R4 region of the Tau repeats, preferably the R3 region. Preferably, the truncated Tau protein lacks N-terminal domains. In a particular embodiment, the Tau protein comprises or consists of a peptide having the amino acid sequence VQIVYKPVDLSKVTSKCG (SEQ ID NO: 27) which corresponds to amino acids 306 to 323 of tau441, as defined above. Anti-Tau nanobody
Dans le contexte de la présente invention, les termes "anticorps" et In the context of the present invention, the terms "antibodies" and
"immunoqlobuline" ont la même signification et sont utilisés indifféremment. Dans les anticorps conventionnels, les deux chaînes lourdes sont liées l'une à l'autre par des ponts disulfures et chaque chaîne lourde est liée à une chaîne légère par un pont disulfure. Il y a deux types de chaîne légère : les chaînes légères lambda (λ) et kappa (κ). Il existe cinq principales classes de chaînes lourdes (ou isotypes) qui déterminent l'activité fonctionnelle d'un anticorps : IgM, IgD, IgG, IgA et IgE. Chaque chaîne contient des domaines de séquence distincts. La chaîne légère comprend deux domaines : un domaine variable (VL) et un domaine constant (CL). La chaîne lourde comprend quatre domaines : un domaine variable (VH) et trois domaines constants (CH1 , CH2 et CH3, collectivement nommés CH). Les régions variables des chaînes lourde (VH) et légère (VL) déterminent la reconnaissance de liaison et la spécificité à l'antigène. Les domaines des régions constantes des chaînes légère (CL) et lourde (CH) confèrent des propriétés biologiques importantes telles que l'association des chaînes des anticorps, la sécrétion, la mobilité transplacentaire, la liaison au complément et la liaison aux récepteurs Fc. Le fragment Fv est la partie N-terminale du fragment Fab d'une immunoglobuline consistant en les portions variables d'une chaîne légère et d'une chaîne lourde. La spécificité de l'anticorps réside dans la complémentarité structurale entre le site de combinaison de l'anticorps et le déterminant antigénique. Les sites de combinaison de l'anticorps sont faits de résidus qui proviennent principalement des régions hypervariables ou régions de détermination de la complémentarité (CDRs). Occasionnellement, des résidus provenant des régions non-hypervariables ou régions "charpente" ("framework", FR) peuvent influencer la structure globale du domaine et par conséquent le site de combinaison. "immunoglobulin" have the same meaning and are used interchangeably. In conventional antibodies, the two heavy chains are linked to one another by disulfide bridges and each heavy chain is linked to a light chain by a disulfide bridge. There are two types of light chain: lambda (λ) and kappa (κ) light chains. There are five major classes of heavy chains (or isotypes) that determine the functional activity of an antibody: IgM, IgD, IgG, IgA and IgE. Each string contains separate sequence domains. The light chain has two domains: a variable domain (VL) and a constant domain (CL). The heavy chain comprises four domains: a variable domain (VH) and three constant domains (CH1, CH2 and CH3, collectively called CH). Heavy (VH) and light (VL) variable regions determine binding recognition and antigen specificity. The domains of the light chain (CL) and heavy (CH) constant regions confer important biological properties such as the association of antibody chains, secretion, transplacental mobility, complement binding and Fc receptor binding. The Fv fragment is the N-terminal part of the Fab fragment of an immunoglobulin consisting of the variable portions of a light chain and a heavy chain. The specificity of the antibody lies in the structural complementarity between the site of combination of the antibody and the antigenic determinant. The antibody combining sites are made of residues that originate primarily from hypervariable regions or complementarity determining regions (CDRs). Occasionally, residues from non-hypervariable regions or "framework" (FR) regions may influence the overall structure of the domain and hence the combining site.
Dans le contexte de l'invention, le terme "CDR" fait référence aux séquences d'acides aminés, qui, ensemble, définissent l'affinité de liaison et la spécificité de la région Fv naturelle d'un site de liaison natif d'une immunoglobuline. Les chaînes lourde et légère d'une immunoglobuline ont chacune trois CDRs, désignés H-CDR1 , H-CDR2, H-CDR3 et L-CDR1 , L-CDR2, L-CDR3 respectivement. Un site de liaison à l'antigène inclut donc 6 CDRs, comprenant l'ensemble des CDRs d'une région variable d'une chaîne lourde et d'une région variable d'une chaîne légère.  In the context of the invention, the term "CDR" refers to amino acid sequences, which together define the binding affinity and specificity of the natural Fv region of a native binding site of a immunoglobulin. The heavy and light chains of an immunoglobulin each have three CDRs, designated H-CDR1, H-CDR2, H-CDR3 and L-CDR1, L-CDR2, L-CDR3 respectively. An antigen binding site therefore includes 6 CDRs, including all the CDRs of a variable region of a heavy chain and a variable region of a light chain.
La localisation des CDRs dans la séquence d'un anticorps ou d'un nanocorps peut être déterminée par l'homme du métier en utilisant des techniques précédemment décrites. Typiquement, les CDRs peuvent être identifiés en séquençant l'ADN de l'anticorps ou du nanocorps avec un système approprié, tel que le 3730XL DNA Analyser et ABI PRISM BigDye Terminator cyc, puis en analysant les séquences ainsi obtenues à l'aide de bases de données dédiées telle que le international ImMunoGeneTics database ou IMTG (Lefranc (2003) Dev. Comp. Immunol. 27:55) ou Kabat, et al. (Séquences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md., 1991 ), de préférence IMGT.  The location of the CDRs in the sequence of an antibody or nanobody can be determined by those skilled in the art using previously described techniques. Typically, CDRs can be identified by sequencing the DNA of the antibody or nanobodies with a suitable system, such as the 3730XL DNA Analyzer and ABI PRISM BigDye Terminator cyc, and then analyzing the sequences thus obtained using bases. dedicated data such as the international ImMunoGeneTics database or IMTG (Lefranc (2003) Dev Comp., Immunol 27:55) or Kabat, et al. (National Institutes of Health, Bethesda, Md., 1991), preferably IMGT.
Dans le contexte de l'invention, le terme "région charpente", "framework" ou "FR" font référence aux séquences d'acides aminés intercalées entre les CDRs.  In the context of the invention, the term "framework region", "framework" or "FR" refers to the amino acid sequences intercalated between the CDRs.
Dans le contexte de l'invention, les termes "nanocorps", "nanobody", "VHH", "fragment d'anticorps VHH" et "anticorps à domaine unigue" sont utilisés indifféremment et désignent le domaine variable de la chaîne lourde unique d'anticorps du type de ceux trouvés chez les camélidés, qui sont naturellement dépourvus de chaînes légères. En absence de chaîne légère les nanocorps ont chacun trois CDRs, désignés CDR1 , CDR2, et CDR3 respectivement. Les nanocorps selon l'invention peuvent en particulier être des nanocorps de chameaux, de dromadaires, de lamas ou d'alpagas. De préférence, les nanocorps selon l'invention sont des nanocorps de lamas.  In the context of the invention, the terms "nanobody", "nanobody", "VHH", "VHH antibody fragment" and "unic domain antibody" are used interchangeably and denote the variable domain of the unique heavy chain. antibodies of the type found in camelids, which are naturally devoid of light chains. In the absence of light chain the nanobodies each have three CDRs, designated CDR1, CDR2, and CDR3 respectively. The nanobodies according to the invention may in particular be nanobodies of camels, dromedaries, llamas or alpacas. Preferably, the nanobodies according to the invention are lamas nanobodies.
Par "nanocorps dirigé contre la protéine Tau", on entend ici un nanocorps capable de se lier de manière sélective à la protéine Tau, telle que définie dans la section « Tau ». Préférentiellement, le nanocorps est spécifique de Tau, c'est-à-dire qu'il se lie à Tau à l'exclusion de toute autre molécule. By "nanobodies directed against the Tau protein" is meant here a nanobodies capable of selectively binding to the Tau protein, as defined in the "Tau" section. Preferably, the nanobody is specific for Tau, that is to say that it binds Tau to the exclusion of any other molecule.
Les inventeurs ont préparé des nanocorps dirigés contre des formes pathologiques de la protéine Tau, telles que la protéine Tau sous forme d'oligomères et éventuellement sous forme de fibres. Les nanocorps peuvent également être dirigés contre la protéine Tau tronquée sous formes de fibres.  The inventors have prepared nanobodies directed against pathological forms of the Tau protein, such as the Tau protein in the form of oligomers and optionally in the form of fibers. The nanobodies can also be directed against the truncated Tau protein in fiber form.
En particulier, les inventeurs ont immunisé des lamas avec une préparation de protéines Tau enrichie en protéine Tau sous forme d'oligomères, avec une préparation de protéines Tau enrichie en protéine Tau sous forme de fibres et une préparation de protéines Tau tronquées enrichie en protéine Tau tronquée sous forme de fibres. Les inventeurs ont ensuite identifié plus précisément deux nanocorps dirigés contre Tau et présentant des caractéristiques additionnelles non attendues que ne possèdent pas d'autres nanocorps anti-Tau. En effet, ces deux nanocorps, 2C5 et S2T2M3_E6, en particulier 2C5, reconnaissent Tau sous forme d'oligomères et éventuellement sous formes de fibres. La protéine Tau sous forme de fibres est également reconnue quand il s'agit d'une protéine Tau tronquée telle que définie ci-dessus. En même temps ces deux nanocorps, 2C5 et S2T2M3_E6, en particulier 2C5, ne se lient pas à la protéine Tau native, sous forme de monomère. En outre, ces deux nanocorps, 2C5 et S2T2M3_E6, en particulier 2C5, ne se lient pas aux fibres de peptide beta amyloïde.  In particular, the inventors have immunized llamas with a protein enriched Tau Tau protein preparation in the form of oligomers, with a protein enriched Tau protein preparation in Tau form and a Tau protein enriched truncated Tau protein preparation. truncated in the form of fibers. The inventors then identified more precisely two nanobodies directed against Tau and having additional features not expected that do not have other anti-Tau nanobodies. Indeed, these two nanobodies, 2C5 and S2T2M3_E6, in particular 2C5, recognize Tau as oligomers and optionally in the form of fibers. Tau protein in fiber form is also recognized when it is a truncated Tau protein as defined above. At the same time, these two nanobodies, 2C5 and S2T2M3_E6, in particular 2C5, do not bind to the native Tau protein in monomeric form. In addition, these two nanobodies, 2C5 and S2T2M3_E6, in particular 2C5, do not bind to beta amyloid peptide fibers.
En conséquence, dans un mode de réalisation préféré, les nanocorps sont dirigés contre la protéine Tau sous forme d'oligomères.  Accordingly, in a preferred embodiment, the nanobodies are directed against the Tau protein as oligomers.
Le terme « oliqomères » dans le contexte de la présente invention concerne une préparation de Tau obtenue par un procédé de fibrillation in vitro tel que décrit dans les exemples de cette demande. Typiquement, la fibrillation de Tau (40 μΜ) est réalisée par exemple à 37°C dans du tampon, par exemple, MOPS (acide 3-(N-morpholino) propane sulfonique) typiquement 20 mM, typiquement à pH 7 en présence typiquement d'héparine (10 μΜ), de NaN3 (4%) pour un volume final de typiquement 1 ,5 ml. La fibrillation est stoppée typiquement par congélation des échantillons à -80°C au bout de typiquement 48 heures pour obtenir Tau sous forme d'oligomères. Selon le temps exact de fibrillation cette préparation de protéines Tau est enrichie en protéine Tau sous forme d'oligomères.  The term "oligomers" in the context of the present invention relates to a Tau preparation obtained by an in vitro fibrillation method as described in the examples of this application. Typically, the fibrillation of Tau (40 μl) is carried out for example at 37 ° C. in buffer, for example MOPS (3- (N-morpholino) propanesulfonic acid), typically 20 mM, typically at pH 7 in the presence typically of heparin (10 μl), NaN3 (4%) for a final volume of typically 1.5 ml. Fibrillation is typically stopped by freezing the samples at -80 ° C after typically 48 hours to obtain Tau as oligomers. Depending on the exact time of fibrillation this Tau protein preparation is enriched in Tau protein as oligomers.
Le terme « enrichie en protéine Tau sous forme d'oligomères» signifie une préparation de protéines Tau sous forme d'oligomères dont plus de 50% de protéines Tau sont sous forme d'oligomères. Par exemple plus de 50%, plus de 55%, plus de 60%, plus de 70%, plus de 75%, plus de 80%, plus de 85%, plus de 90% des protéines Tau sont sous forme d'oligomères. Il est connu de l'homme du métier qu'une préparation enrichie en protéines Tau sous forme d'oligomères contient en outre de la protéine Tau sous forme native et de la protéine Tau sous forme de fibres. The term "Tau protein enriched as oligomers" means a Tau protein preparation in the form of oligomers of which more than 50% of Tau proteins are in the form of oligomers. For example, more than 50%, more than 55%, more than 60%, more than 70%, more than 75%, more than 80%, more than 85%, more than 90% of Tau proteins are in the form of oligomers . It is known to those skilled in the art that an enriched preparation Tau proteins in the form of oligomers further contains Tau protein in native form and Tau protein in the form of fibers.
En conséquence, dans un mode de réalisation, la protéine Tau sous forme d'oligomères est un mélange de protéines Tau sous forme de monomères, sous forme d'oligomères et sous forme de fibres, dont plus de 50% des protéines Tau sont sous forme d'oligomères, en particulier plus de 55%, plus de 60%, plus de 70%, plus de 75%, plus de 80%, plus de 85%, plus de 90% des protéines Tau sont sous forme d'oligomères.  Accordingly, in one embodiment, the oligomeric Tau protein is a mixture of Tau proteins in the form of monomers, in the form of oligomers and in fiber form, of which more than 50% of the Tau proteins are in the form of oligomers. Oligomers, in particular more than 55%, more than 60%, more than 70%, more than 75%, more than 80%, more than 85%, more than 90% of the Tau proteins are in the form of oligomers.
Il est également connu de l'homme du métier qu'il est possible de purifier la protéine Tau sous formes d'oligomères à partir d'une telle préparation, en utilisant plusieurs méthodes connues de l'homme du métier, dont par exemple la chromatographie d'exclusion stérique.  It is also known to those skilled in the art that it is possible to purify the Tau protein in the form of oligomers from such a preparation, using several methods known to those skilled in the art, including for example chromatography. steric exclusion.
Les oligomères comprennent de préférence entre 2 et 45 unités de Tau. Des oligomères ayant 2 à 8 unités de Tau sont appelés oligomères solubles et les oligomères ayant plus de 8 jusqu'à 45 unités de Tau sont appelés oligomères granulaires et peuvent être solubles ou non solubles. Dans un mode de réalisation ces oligomères peuvent être caractérisées, en utilisant un microscope à force atomique ou microscope électronique, comme étant des points ronds ayant un diamètre d'environ 12 à 29 nm.  The oligomers preferably comprise between 2 and 45 units of Tau. Oligomers having 2 to 8 units of Tau are referred to as soluble oligomers and oligomers having more than 8 to 45 units of Tau are referred to as granular oligomers and may be soluble or insoluble. In one embodiment these oligomers can be characterized, using an atomic force microscope or electron microscope, as round points having a diameter of about 12 to 29 nm.
Dans un autre mode de réalisation ces oligomères peuvent être caractérisés en utilisant une chromatographie d'exclusion stérique comme étant des molécules sphériques ayant un diamètre d'environ 12 à 35 nm.  In another embodiment these oligomers can be characterized using size exclusion chromatography as being spherical molecules having a diameter of about 12 to 35 nm.
En conséquence, le terme «oliqomère» dans le contexte de l'invention fait référence à un agrégat ou polymère de Tau ayant de 2 jusqu'à 50 unités de Tau, en particulier 2 à 20, de manière préférée 2 à 12 unités de tau, par exemple un dimère, un trimère, un tétramère, un pentamère, un hexamère, heptamère, octamère, nonamère, décamère, undécamère ou dodécamère de Tau ou ayant 20 à 45, de préférence, 30 à 45 unités de Tau, par exemple 35 à 45, de préférence 38 à 42 unités de Tau. Dans certains modes de réalisation, la protéine tau est dimérique ou trimérique. Dans certains modes de réalisation, la protéine Tau sous forme d'oligomères est soluble et/ou non soluble, de manière plus préférée soluble.  Accordingly, the term "oligomer" in the context of the invention refers to an aggregate or Tau polymer having from 2 to 50 Tau units, particularly 2 to 20, most preferably 2 to 12 tau units. , for example a dimer, a trimer, a tetramer, a pentamer, a hexamer, heptamer, octamer, nonamer, decamer, undecamer or dodecamer of Tau or having 20 to 45, preferably 30 to 45, units of Tau, for example at 45, preferably 38 to 42 units of Tau. In some embodiments, the tau protein is dimeric or trimeric. In some embodiments, the oligomeric Tau protein is soluble and / or non-soluble, more preferably soluble.
Dans une mode de réalisation le nanocorps de l'invention a une affinité pour la protéine Tau sous forme d'oligomères qui est < 20nM, par exemple < 10nM, < 8nM, < 7nM ou < 5nM, par exemple une affinité de 0.1 nM à 20 nM, en particulier de 1 nM à 10 nM, ou de 1 nM à 7 nM, par exemple 5 nM.  In one embodiment, the nanobodies of the invention have an affinity for Tau protein in the form of oligomers which is <20nM, for example <10nM, <8nM, <7nM or <5nM, for example an affinity of 0.1nM to 20 nM, in particular from 1 nM to 10 nM, or from 1 nM to 7 nM, for example 5 nM.
Par « affinité » on entend la capacité de fixation entre une macromolécule et l'antigène qu'elle fixe, en particulier la capacité de fixation entre un nanocorps et l'antigène qu'il fixe, par exemple un nanocorps de l'invention et la protéine Tau sous les formes pathologiques telles que définies ci-dessus. By "affinity" is meant the binding capacity between a macromolecule and the antigen it fixes, in particular the binding capacity between a nanobody and the antigen that it fixes, for example a nanobody of the invention and the Tau protein in the pathological forms as defined above.
L'affinité et donc la capacité du nanocorps de l'invention à se lier à la protéine Tau, par exemple sous forme d'oligomère ou de fibres, peut être mesurée in vitro par plusieurs méthodes, dont la résonance plasmonique de surface (RPS, notamment à l'aide d'un appareil type BIAcore 2000- Pharmacia Biosensor, Upsala, Suède) ou par exemple par un test ELISA, comme décrit dans les exemples.  The affinity and therefore the ability of the nanobodies of the invention to bind to the Tau protein, for example in the form of oligomers or fibers, can be measured in vitro by several methods, including surface plasmon resonance (SPR). especially using a BIAcore 2000-Pharmacia Biosensor type device, Upsala, Sweden) or for example by an ELISA test, as described in the examples.
Dans une mode de réalisation les nanocorps de l'invention se lient en outre à la protéine Tau sous forme de fibres.  In one embodiment, the nanobodies of the invention further bind to Tau protein in the form of fibers.
Le terme « fibres » dans le contexte de l'invention concerne une préparation de la protéine Tau obtenue par un procédé de fibrillation tel que décrit dans les exemples de cette demande. Typiquement, la fibrillation de Tau (40 μΜ) est réalisée par exemple à 37°C dans du tampon, par exemple, MOPS (acide 3-(N-morpholino) propane sulfonique) typiquement 20 mM, typiquement à pH 7 en présence typiquement d'héparine (10 μΜ), de NaN3 (4%) pour un volume final de typiquement 1 ,5 ml. La fibrillation est stoppée typiquement par congélation des échantillons à -80°C au bout de typiquement 72 heures pour obtenir Tau sous forme de fibres. Une telle préparation de protéines Tau sous forme de fibres est donc une préparation de protéines Tau enrichie en protéine Tau sous forme de fibres.  The term "fibers" in the context of the invention relates to a preparation of the Tau protein obtained by a fibrillation process as described in the examples of this application. Typically, the fibrillation of Tau (40 μl) is carried out for example at 37 ° C. in buffer, for example MOPS (3- (N-morpholino) propanesulfonic acid), typically 20 mM, typically at pH 7 in the presence typically of heparin (10 μl), NaN3 (4%) for a final volume of typically 1.5 ml. Fibrillation is typically stopped by freezing the samples at -80 ° C after typically 72 hours to obtain Tau as fibers. Such a preparation of Tau proteins in the form of fibers is therefore a preparation of Tau proteins enriched in Tau protein in the form of fibers.
Le terme « enrichie par Tau sous forme de fibres» signifie une préparation de protéines Tau sous forme de fibres dont plus de 50% des protéines Tau sont sous forme de fibres. Par exemple plus de 50%, plus de 55%, plus de 60%, plus de 70%, plus de 75%, plus de 80%, plus de 85%, plus de 90% des protéines Tau sont sous forme de fibres. Il est connu de l'homme du métier qu'une préparation enrichie en protéines Tau sous forme de fibres contient en outre de la protéine Tau sous forme native et de la protéine Tau sous forme d'oligomères.  The term "fiber-enriched Tau" means a Tau protein preparation in the form of fibers of which more than 50% of the Tau proteins are in the form of fibers. For example, more than 50%, more than 55%, more than 60%, more than 70%, more than 75%, more than 80%, more than 85%, more than 90% of the Tau proteins are in the form of fibers. It is known to a person skilled in the art that a preparation enriched with Tau proteins in the form of fibers also contains Tau protein in native form and Tau protein in the form of oligomers.
En conséquence, dans un mode de réalisation, la protéine Tau sous forme de fibres est un mélange de protéines Tau sous forme de monomères, sous forme d'oligomères et sous forme de fibres, dont plus de 50% des protéines Tau sont sous forme de fibres, en particulier plus de 55%, plus de 60%, plus de 70%, plus de 75%, plus de 80%, plus de 85%, plus de 90% des protéines Tau sont sous forme de fibres.  Accordingly, in one embodiment, the Tau protein in the form of fibers is a mixture of Tau proteins in the form of monomers, in the form of oligomers and in the form of fibers, of which more than 50% of the Tau proteins are in the form of fibers, especially more than 55%, more than 60%, more than 70%, more than 75%, more than 80%, more than 85%, more than 90% of the Tau proteins are in the form of fibers.
Il est également connu de l'homme du métier qu'il est possible de purifier la protéine Tau sous formes de fibres à partir d'une telle préparation, en utilisant plusieurs méthodes connu de l'homme du métier, dont par exemple la chromatographie d'exclusion stérique. Dans le contexte de l'invention, le terme fibre décrit un polymère ou agrégat de Tau obtenu d'une façon artificielle mimant la fibrillation de Tau in vivo et donc mimant les paires hélicoïdales de filaments et les filaments droits qui composent les DNFs. En conséquence, les fibres de Tau telles qu'utilisées dans le contexte de l'invention englobe les paires de filaments en hélice et les filaments droits. Par microscopie électronique ces fibres peuvent être caractérisées comme ayant une apparence filamenteuse. Dans un mode de réalisation le nanocorps de l'invention a une affinité pour la protéine Tau sous forme de fibres qui est < 20nM, par exemple < 10nM, < 8nM, < 7nM ou < 6nM, par exemple une affinité de 0.1 nM à 20 nM, en particulier de 1 nM à 10 nM, ou de 1 nM à 8 nM, par exemple 6 nM. It is also known to those skilled in the art that it is possible to purify Tau protein in fiber form from such a preparation, using several methods known to those skilled in the art, including for example chromatography. steric exclusion. In the context of the invention, the term fiber describes a polymer or Tau aggregate obtained in an artificial manner mimicking the Tau fibrillation in vivo and thus mimicking the helical pairs of filaments and the straight filaments that make up the DNFs. Accordingly, Tau fibers as used in the context of the invention encompass pairs of helical filaments and straight filaments. By electron microscopy these fibers can be characterized as having a filamentous appearance. In one embodiment, the nanobodies of the invention have an affinity for the Tau protein in the form of fibers which is <20nM, for example <10nM, <8nM, <7nM or <6nM, for example an affinity of 0.1nM to 20nM. nM, in particular from 1 nM to 10 nM, or from 1 nM to 8 nM, for example 6 nM.
Dans un autre mode de réalisation les nanocorps de l'invention se lient en outre à la protéine Tau tronquée. De préférence, cette protéine tronquée est sous forme de fibres et peut également être nommé fibres de peptide R3. La protéine Tau tronquée est telle que définie dans la section «Tau» ci-dessus. En particulier, cette protéine Tau tronquée comprend ou consiste en la séquence d'acides aminés SEQ ID NO : 27.  In another embodiment, the nanobodies of the invention further bind to the truncated Tau protein. Preferably, this truncated protein is in the form of fibers and may also be named R3 peptide fibers. The truncated Tau protein is as defined in the "Tau" section above. In particular, this truncated Tau protein comprises or consists of the amino acid sequence SEQ ID NO: 27.
Dans un mode de réalisation le nanocorps de l'invention a une affinité pour la protéine Tau tronquée sous forme de fibres qui est < 100nM, par exemple < 80nM, < 70nM, < 60nM ou < 50nM, par exemple une affinité de 1 nM à 100 nM, en particulier de 1 nM à 80 nM, ou de 40 nM à 60 nM, par exemple 50 nM.  In one embodiment, the nanobodies of the invention have an affinity for the truncated Tau protein in the form of fibers which is <100nM, for example <80nM, <70nM, <60nM or <50nM, for example an affinity of 1nM to 100 nM, in particular from 1 nM to 80 nM, or from 40 nM to 60 nM, for example 50 nM.
Dans un mode de réalisation le nanocorps de l'invention a une affinité pour la protéine Tau tronquée sous forme de fibres qui est < 100nM, par exemple une affinité de 1 nM à 100 nM, en particulier de 1 nM à 80 nM, de 1 nM à 20nM, par exemple une affinité de 10 à 20nM.  In one embodiment, the nanobodies of the invention have an affinity for the truncated Tau protein in the form of fibers which is <100 nM, for example an affinity of 1 nM to 100 nM, in particular 1 nM to 80 nM, of 1 nM at 20nM, for example an affinity of 10 to 20nM.
Dans le contexte de l'invention, « la protéine Tau tronquée sous forme de fibres » ou « les fibres de peptide R3 » concerne une préparation de peptide R3 obtenue par un procédé de fibrillation in vitro tel que décrit dans les exemples de cette demande. In the context of the invention, "the truncated Tau protein in the form of fibers" or "the peptide fibers R3" relates to an R3 peptide preparation obtained by an in vitro fibrillation method as described in the examples of this application.
Typiquement, la fibrillation de R3 (0.4 μΜ) est réalisée par exemple à 37°C dans du tampon, par exemple, PBS (phosphate buffer saline 50 mM, typiquement à pH 7 en présence typiquement d'héparine (0.4 μΜ), de NaN3 (4%) pour un volume final de typiquement 1 ,5 ml. La fibrillation est stoppée typiquement par congélation des échantillons à -80°C au bout de typiquement 72 heures pour obtenir Tau tronquée (R3) sous forme de fibres. Typically, the fibrillation of R3 (0.4 μl) is carried out for example at 37 ° C. in buffer, for example PBS (50 mM salt buffer phosphate, typically at pH 7 in the presence of heparin (0.4 μl), NaN 3 (4%) for a final volume of typically 1.5 ml Fibrillation is typically stopped by freezing the samples at -80 ° C after typically 72 hours to obtain truncated Tau (R3) as fibers.
Dans un exemple l'affinité pour la protéine Tau tronquée sous forme de fibres est mesurée, par exemple, par ELISA, dans, par exemple, un tampon Phosphate Buffer Saline (NaCI, 137mM ; KCI, 2.7mM ; Na2HP04, 10mM KH2P04, 1 .8mM) comprenant, par exemple, BSA 1 % et- ayant, par exemple un pH de 7.5. In one example, the affinity for the truncated Tau protein in fiber form is measured, for example, by ELISA, in, for example, a buffer phosphate buffer. Saline (NaCl, 137mM, KCl, 2.7mM, Na 2 HPO 4 , 10mM KH 2 PO 4 , 1.8mM) comprising, for example, 1% BSA and having, for example, a pH of 7.5.
Les inventeurs ont également démontré pour les nanocorps de l'invention un marquage spécifique des corps cellulaires de l'hippocampe, du cortex entorhinal et du cortex temporal chez des patients atteints de la maladie d'Alzheimer.  The inventors have also demonstrated for the nanobodies of the invention a specific labeling of the hippocampal cell bodies, the entorhinal cortex and the temporal cortex in patients with Alzheimer's disease.
En conséquence, dans un mode de réalisation les nanocorps de l'invention se lient en outre aux paires hélicoïdales de filaments et/ou les filaments droits.  Accordingly, in one embodiment, the nanobodies of the invention additionally bind to helical pairs of filaments and / or straight filaments.
Le terme «paires hélicoïdales de filaments» désigne des filaments appariés en hélice comprenant au moins 10, au moins 12, au moins 20, de préférence au moins 40 unités de Tau, en particulier au moins 50 ou au moins 60 unités de Tau. Normalement ces filaments peuvent être observés en microscopie électronique et ont un diamètre de 8 à 20 nanomètres, par exemple 10 à 20 nm et un pas d'hélice d'environ 80 nm, par exemple 70 jusqu'à 90nm.  The term "helical pair of filaments" refers to helical paired filaments comprising at least 10, at least 12, at least 20, preferably at least 40, and in particular at least 50 or at least 60 units of Tau. Normally these filaments can be observed by electron microscopy and have a diameter of 8 to 20 nanometers, for example 10 to 20 nm and a helical pitch of about 80 nm, for example 70 to 90 nm.
Le terme « filaments droits » signifie des filaments qui ne sont pas appariés en hélice et comprenant au moins 10, au moins 12, au moins 20, de préférence au moins 40 unités de Tau, en particulier au moins 50 ou au moins 60 unités de Tau. Normalement ces filaments peuvent être observés en microscopie électronique et ont un diamètre d'environ 10 nanomètres, par exemple de 8 à 17nm, en particulier de 9 à 12nm, par exemple 10 nm.  The term "straight filaments" means filaments which are not helically matched and comprising at least 10, at least 12, at least 20, preferably at least 40 units of Tau, in particular at least 50 or at least 60 units of Tau. Tau. Normally these filaments can be observed by electron microscopy and have a diameter of about 10 nanometers, for example from 8 to 17 nm, in particular from 9 to 12 nm, for example 10 nm.
Les paires hélicoïdales de filaments et les filaments droits qui composent les DNFs ne sont pas solubles.  The helical pairs of filaments and straight filaments that make up the DNFs are not soluble.
Dans un autre mode de réalisation les nanocorps de l'invention n'interagissent pas substantiellement avec la protéine amyloïde beta 1 -42 polymérisée.  In another embodiment, the nanobodies of the invention do not interact substantially with the polymerized amyloid beta 1 -42 protein.
Dans un mode de réalisation les nanocorps n'interagissent pas substantiellement avec la protéine Tau sous forme monomérique.  In one embodiment, the nanobodies do not substantially interact with the Tau protein in monomeric form.
Un nanocorps « n'interaqit pas substantiellement » avec une protéine, par exemple la protéine amyloïde beta 1 -42 polymérisé ou Tau sous forme monomérique, lorsque les affinités pour la protéine Tau sous forme d'oligomère et l'affinité pour la protéine Tau sous forme de monomère ou la protéine amyloïde beta 1 -42 polymérisé sont très différentes. Dans un exemple, l'affinité pour la protéine Tau sous forme monomérique ne peut pas être mesurée car la réponse de liaison est trop faible. Dans un autre exemple, un nanocorps n'interagit pas substantiellement avec la protéine Tau sous forme monomérique ou la protéine amyloïde beta 1 -42 polymérisé, lorsque la réaction de liaison du nanocorps avec Tau sous forme de monomère est inférieure à 5% de la réponse de liaison du même nanocorps avec Tau sous forme d'oligomère dans la même condition expérimentale et à la même concentration du nanocorps. Dans la pratique, la concentration du nanocorps utilisée peut être la concentration CE50 ou la concentration nécessaire pour atteindre le plateau de saturation. A nanobodies "do not substantially intercalate" with a protein, for example, polymerized amyloid beta 1 -42 protein or Tau in monomeric form, when the affinities for Tau protein as an oligomer and the affinity for Tau protein under The monomer form or the polymerized amyloid beta-42 protein are very different. In one example, the affinity for monomeric Tau protein can not be measured because the binding response is too weak. In another example, a nanobodies do not substantially interact with the Tau protein in monomeric form or the polymerized amyloid beta 1 -42 protein, when the nanobody-binding reaction with Tau as a monomer is less than 5% of the response. binding of the same nanobodies with Tau as an oligomer in the same experimental condition and at the same concentration of the nanobody. In practice, the The concentration of the nanobodies used may be the EC 50 concentration or the concentration necessary to reach the saturation plateau.
Par exemple, l'affinité des nanocorps de l'invention pour la protéine Tau sous forme monomérique est > 1200 nM, par exemple > 1400 nM, > 1600 nM, > 1800 nM, en particulier > 1800 nM.  For example, the affinity of the nanobodies of the invention for Tau protein in monomeric form is> 1200 nM, for example> 1400 nM,> 1600 nM,> 1800 nM, in particular> 1800 nM.
Par exemple, l'affinité des nanocorps de l'invention pour la protéine amyloïde beta 1 -42 polymérisée est > 5000 nM, par exemple> 8000 nM, > 9000 nM, > 10000 nM, en particulier > 10000 nM.  For example, the affinity of the nanobodies of the invention for the polymerized amyloid beta 1 -42 protein is> 5000 nM, for example> 8000 nM,> 9000 nM,> 10000 nM, in particular> 10000 nM.
Les nanocorps de l'invention ont été séquencés :  The nanobodies of the invention have been sequenced:
- le nanocorps 2C5 présente la séquence d'acides aminés QVQLVQSGG the 2C5 nanobodies have the amino acid sequence QVQLVQSGG
GLVQAGGSLRLSCAASGRTFSSDTLAWFRQAPGKEREFVASISPSGGV TYYEDSVK.GRFTISRDNSKNTVLLQMNSLTPEDTAVYYCNRDPKYGNT RYWGQGTQVTVSS (SEQ ID NO: 9), GLVQAGGSLRLSCAASGRTFSSDTLAWFRQAPGKEREFVASISPSGGV TYYEDSVK.GRFTISRDNSKNTVLLQMNSLTPEDTAVYYCNRDPKYGNT RYWGQGTQVTVSS (SEQ ID NO: 9),
le nanocorps S2T2M3_E6 présente la séquence d'acides aminés EVQLVESGGGLVQAGGSLRLSCAASGRTFSRYAMGWFRQAPGKEREF the nanobody S2T2M3_E6 has the amino acid sequence EVQLVESGGGLVQAGGSLRLSCAASGRTFSRYAMGWFRQAPGKEREF
VASISRSGGSTRYADAVKGRFTISRDNTNNTVYLLMNNLKPEDTAVYYC TARRRISGTPQWHYWGQGTQVTVSS (SEQ ID NO: 10). VASISRSGGSTRYADAVKGRFTISRDNTNNTVYLLMNNLKPEDTAVYYC TARRRISGTPQWHYWGQGTQVTVSS (SEQ ID NO: 10).
Les CDRs de ces deux nanocorps ont été plus spécifiquement séquencés et sont les suivants :  The CDRs of these two nanobodies have been more specifically sequenced and are as follows:
· 2C5  · 2C5
o CDR1 : GRTFSSDT (SEQ ID NO: 3)  o CDR1: GRTFSSDT (SEQ ID NO: 3)
o CDR2 : ISPSGGVT (SEQ ID NO: 4)  o CDR2: ISPSGGVT (SEQ ID NO: 4)
o CDR3 : NRDPKYGNTRY (SEQ ID NO: 5)  o CDR3: NRDPKYGNTRY (SEQ ID NO: 5)
• S2T2M2_E6  • S2T2M2_E6
o CDR1 : GRTFSRYA (SEQ ID NO: 6)  o CDR1: GRTFSRYA (SEQ ID NO: 6)
o CDR2 : ISRSGGST (SEQ ID NO: 7)  o CDR2: ISRSGGST (SEQ ID NO: 7)
o CDR3: TARRRISGTPQWHY (SEQ ID NO: 8)  o CDR3: TARRRISGTPQWHY (SEQ ID NO: 8)
Comme il est bien connu de l'homme du métier, la combinaison des CDR1 , CDR2 et CDR3 suffit pour définir un site de liaison à l'antigène. Il est également connu par l'homme de métier que deux nanocorps qui reconnaissent le même antigène entrent en compétition pour la liaison à cet antigène.  As is well known to those skilled in the art, the combination of CDR1, CDR2 and CDR3 is sufficient to define an antigen binding site. It is also known to those skilled in the art that two nanobodies that recognize the same antigen compete for binding to this antigen.
En conséquence, un objet de l'invention concerne un nanocorps dirigé contre la protéine Tau, ladite protéine Tau étant sous forme pathologique, en particulier sous forme d'oligomères et ledit nanocorps entrant en compétition pour la liaison à la protéine Tau sous forme d'oligomères avec un nanocorps comprenant les séquences d'acides aminés (i) GRTFSX1X2X3 (SEQ ID NO: 1 ) comme CDR1 dans laquelle l'acide aminé ΧΊ est S ou R, X2 est D ou Y et X3 est T ou A, Accordingly, an object of the invention relates to a nanobodies directed against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers and said nanobodies competing for binding to the Tau protein in the form of oligomers with a nanobody including amino acid sequences (i) GRTFSX1X 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid Χ Ί is S or R, X 2 is D or Y and X 3 is T or A,
(ii) ISX1SGGX2T (SEQ ID NO: 2) comme CDR2 dans laquelle l'acide aminé ΧΊ est P ou R et X2 est S ou V, et (ii) ISX1SGGX 2 T (SEQ ID NO: 2) as CDR2 in which the amino acid Χ Ί is P or R and X 2 is S or V, and
(iii) NRDPKYGNTRY (SEQ ID NO: 5) ou TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3. (iii) NRDPKYGNTRY (SEQ ID NO: 5) or TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3.
La capacité d'un nanocorps candidat à entrer en compétition pour la liaison à la protéine Tau, par exemple sous forme d'oligomères, avec un nanocorps comprenant les CDR d'un nanocorps 2C5 et/ou S2T2M3_E6 tels que définis ci-dessus, (ci-après un nanocorps «de référence») peut être facilement vérifiée, par exemple, par ELISA compétitive dans laquelle l'antigène (à savoir la protéine Tau sous forme d'oligomères) est lié à un support solide et deux solutions contenant le nanocorps candidat et le nanocorps de référence, respectivement, sont ajoutés et les nanocorps vont entrer en compétition pour se lier à l'antigène. La quantité de nanocorps de référence lié à l'antigène peut ensuite être mesurée et comparée à la quantité de nanocorps de référence lié à l'antigène lorsqu'il est mesuré contre un contrôle négatif (solution par exemple dépourvue de nanocorps de candidat). Une quantité de nanocorps de référence lié en présence de nanocorps candidat qui est diminuée par rapport à la quantité de nanocorps de référence lié en présence du témoin négatif indique que le nanocorps candidat entre en compétition pour la liaison à la protéine Tau sous forme d'oligomères. Idéalement, le nanocorps de référence peut être marqué (par exemple par fluorescence) pour faciliter la détection des nanocorps de référence liés. Des mesures répétées peuvent être réalisées avec des dilutions successives du candidat et/ou nanocorps de référence.  The ability of a candidate nanobicide to compete for Tau protein binding, for example in the form of oligomers, with a nanobodies comprising the CDRs of a 2C5 and / or S2T2M3_E6 nanobodies as defined above, ( hereinafter a "reference" nanobodies) can easily be verified, for example, by competitive ELISA in which the antigen (ie Tau protein in the form of oligomers) is bound to a solid support and two solutions containing the nanobody The candidate and the reference nanobodies, respectively, are added and the nanobodies will compete to bind to the antigen. The amount of reference nanobodies bound to the antigen can then be measured and compared to the amount of reference nanobodies bound to the antigen when measured against a negative control (eg solution free of candidate nanobodies). An amount of bound reference nanobodies in the presence of candidate nanobodies that is decreased relative to the amount of bound reference nanobodies in the presence of the negative control indicates that the candidate nanobodies compete for binding to Tau protein as oligomers. . Ideally, the reference nanobodies can be labeled (e.g. by fluorescence) to facilitate the detection of bound reference nanobodies. Repeated measurements can be performed with successive dilutions of the candidate and / or reference nanobodies.
Dans un autre objet de l'invention, l'invention concerne un nanocorps dirigé contre la protéine Tau, ladite protéine Tau étant sous forme pathologique, en particulier sous forme d'oligomères et ledit nanocorps entrant en compétition pour la liaison à la protéine Tau sous forme d'oligomères avec un nanocorps choisi parmi un nanocorps comprenant : a) les séquences d'acides aminés (i) GRTFSSDT (SEQ ID NO: 3) comme CDR1 , (ii) ISPSGGVT (SEQ ID NO: 4) comme CDR2 et (iii) NRDPKYGNTRY (SEQ ID NO: 5) comme CDR3 ; ou  In another subject of the invention, the invention relates to a nanobodies directed against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers and said nanobodies competing for binding to the Tau protein under oligomer form with a nanobodies selected from nanobodies comprising: a) amino acid sequences (i) GRTFSSDT (SEQ ID NO: 3) such as CDR1, (ii) ISPSGGVT (SEQ ID NO: 4) such as CDR2 and ( iii) NRDPKYGNTRY (SEQ ID NO: 5) as CDR3; or
b) les séquences d'acides aminés (i) GRTFSRYA (SEQ ID NO: 6) comme CDR1 , (ii) ISRSGGST (SEQ ID NO: 7) comme CDR2 et (iii) TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3. Un objet de la présente invention concerne un nanocorps dirigé contre la protéineb) the amino acid sequences (i) GRTFSRYA (SEQ ID NO: 6) as CDR1, (ii) ISRSGGST (SEQ ID NO: 7) as CDR2 and (iii) TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3. An object of the present invention relates to a nanobody against protein
Tau comprenant les séquences d'acides aminés (i) GRTFSX1X2X3 (SEQ ID NO: 1 ) comme CDR1 dans laquelle l'acide aminé ΧΊ est S ou R, X2 est D ou Y et X3 est T ou A, Tau comprising the amino acid sequences (i) GRTFSX1X 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid Χ Ί is S or R, X 2 is D or Y and X 3 is T or A,
(ii) ISX1SGGX2T (SEQ ID NO: 2) comme CDR2 dans laquelle l'acide aminé ΧΊ est P ou R et X2 est S ou V, et (ii) ISX1SGGX 2 T (SEQ ID NO: 2) as CDR2 in which the amino acid Χ Ί is P or R and X 2 is S or V, and
(iii) NRDPKYGNTRY (SEQ ID NO: 5) ou TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3. (iii) NRDPKYGNTRY (SEQ ID NO: 5) or TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3.
L'invention concerne également un nanocorps dirigé contre la protéine Tau comprenant : a) les séquences d'acides aminés (i) GRTFSSDT (SEQ ID NO: 3) comme CDR1 , (ii) ISPSGGVT (SEQ ID NO: 4) comme CDR2 et (iii) NRDPKYGNTRY (SEQ ID NO: 5) comme CDR3 ; ou The invention also relates to a nanobodies directed against the Tau protein comprising: a) the amino acid sequences (i) GRTFSSDT (SEQ ID NO: 3) as CDR1, (ii) ISPSGGVT (SEQ ID NO: 4) as CDR2 and (iii) NRDPKYGNTRY (SEQ ID NO: 5) as CDR3; or
b) les séquences d'acides aminés (i) GRTFSRYA (SEQ ID NO: 6) comme CDR1 , (ii ISRSGGST (SEQ ID NO: 7) comme CDR2 et (iii) TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3 ; ou  b) the amino acid sequences (i) GRTFSRYA (SEQ ID NO: 6) such as CDR1, (ii ISRSGGST (SEQ ID NO: 7) as CDR2 and (iii) TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3, or
un variant fonctionnellement conservatif du nanocorps défini en a) ou b) comprenant une substitution conservative d'un ou deux acides aminés dans respectivement une, deux ou trois des séquences SEQ ID NO: 3, SEQ ID NO: 4 et SEQ ID NO: 5, ou SEQ ID NO: 6, SEQ ID NO: 7 et SEQ ID NO: 8. a functionally conservative variant of the nanobodies defined in a) or b) comprising a conservative substitution of one or two amino acids in one, two or three of the sequences SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5 respectively; , or SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8.
Dans un mode de réalisation préféré cette protéine Tau est sous forme d'oligomères et éventuellement sous forme de fibres.  In a preferred embodiment, this Tau protein is in the form of oligomers and optionally in the form of fibers.
Les inventeurs ont également séquencé les régions "charpente" (FR) des nanocorps 2C5 et S2T2M2 E6. Les séquences correspondantes sont les suivantes :  The inventors have also sequenced the "framework" (FR) regions of the 2C5 and S2T2M2 E6 nanobodies. The corresponding sequences are as follows:
• 2C5  • 2C5
o Région FR1 : QVQLVQSGGGLVQAGGSLRLSCAAS (SEQ ID NO: 1 1 ) o Région FR2 : LAWFRQAPGKEREFVAS (SEQ ID NO: 12)  o Region FR1: QVQLVQSGGGLVQAGGSLRLSCAAS (SEQ ID NO: 1 1) o Region FR2: LAWFRQAPGKEREFVAS (SEQ ID NO: 12)
o Région FR3 : YYEDSVKGRFTISRDNSKNTVLLQMNSLTPEDTAVYYC (SEQ ID NO: 13)  o Region FR3: YYEDSVKGRFTISRDNSKNTVLLQMNSLTPEDTAVYYC (SEQ ID NO: 13)
o Région FR4 : WGQGTQVTVSS (SEQ ID NO: 14)  o Region FR4: WGQGTQVTVSS (SEQ ID NO: 14)
• S2T2M2_E6  • S2T2M2_E6
o Région FR1 : EVQLVESGGGLVQAGGSLRLSCAAS (SEQ ID NO: 15) o Région FR2 : MGWFRQAPGKEREFVAS (SEQ ID NO: 16)  Region FR1: EVQLVESGGGLVQAGGSLRLSCAAS (SEQ ID NO: 15) Region FR2: MGWFRQAPGKEREFVAS (SEQ ID NO: 16)
o Région FR3 : RYADAVKGRFTISRDNTNNTVYLLMNNLKPEDTAVYYC (SEQ ID NO: 17)  Region FR3: RYADAVKGRFTISRDNTNNTVYLLMNNLKPEDTAVYYC (SEQ ID NO: 17)
o Région FR4 : WGQGTQVTVSS (SEQ ID NO: 18)  Region FR4: WGQGTQVTVSS (SEQ ID NO: 18)
Dans un mode de réalisation particulier, l'invention concerne un nanocorps dirigé contre la protéine Tau, ladite protéine Tau étant sous forme pathologique, en particulier sous forme d'oligomères et ledit nanocorps entrant en compétition pour la liaison à la protéine Tau sous forme d'oligomères avec un nanocorps choisi parmi un nanocorps comprenant une séquence d'acides aminés sélectionnée dans le groupe consistant en les séquences d'acides aminés SEQ ID NO: 9 et SEQ ID NO: 10. In a particular embodiment, the invention relates to a nanobody against the Tau protein, said Tau protein being in pathological form, in particular in the form of oligomers and said nanobodies competing for binding to the Tau protein as oligomers with a nanobodies selected from a nanobodies comprising an amino acid sequence selected from the group consisting of SEQ amino acid sequences ID NO: 9 and SEQ ID NO: 10.
Dans un mode de réalisation particulier, l'invention concerne un nanocorps comprenant ou consistant en l'enchaînement de séquences FR1 -CDR1 -FR2-CDR2-FR3- CDR3-FR4 telles que définies ci-dessus de l'un des nanocorps identifiés par les inventeurs.  In a particular embodiment, the invention relates to a nanobody comprising or consisting of the sequence of FR1 -CDR1 -FR2-CDR2-FR3-CDR3-FR4 sequences as defined above of one of the nanobodies identified by the inventors.
De préférence, le nanocorps selon l'invention est donc un nanocorps comprenant ou consistant en une séquence d'acides aminés sélectionnée dans le groupe consistant en les séquences d'acides aminés SEQ ID NO: 9 et SEQ ID NO: 10 ou un variant fonctionnellement conservatif de celui-ci comprenant une substitution conservative d'un ou deux acides aminés dans un, deux ou trois des CDRs compris respectivement dans la séquence d'acides aminé SEQ ID NO: 9 ou SEQ ID NO: 10. Le variant fonctionnellement conservatif tel que défini ci-dessus peut en outre comprendre une ou plusieurs substitutions, en particulier une ou plusieurs substitutions conservatives dans les régions respectivement des séquences d'acides aminés SEQ ID NO: 9 ou SEQ ID NO: 10 qui ne sont pas des CDRs, telles que les régions "charpente", en particulier les régions "charpente" définies ci-dessus. De manière davantage préférée, le nanocorps selon l'invention est un nanocorps comprenant ou consistant en une séquence d'acides aminés sélectionnée dans le groupe consistant en les séquences d'acides aminés SEQ ID NO: 9 et SEQ ID NO: 10. De manière préférée entre toutes, le nanocorps selon l'invention est un nanocorps comprenant ou consistant en la séquence d'acides aminés SEQ ID NO: 9.  Preferably, the nanobody according to the invention is therefore a nanobodies comprising or consisting of an amino acid sequence selected from the group consisting of the amino acid sequences SEQ ID NO: 9 and SEQ ID NO: 10 or a functionally variant conservative thereof comprising a conservative substitution of one or two amino acids in one, two or three of the CDRs included respectively in the amino acid sequence SEQ ID NO: 9 or SEQ ID NO: 10. The functionally conservative variant as as defined above may further comprise one or more substitutions, in particular one or more conservative substitutions in the regions respectively of the amino acid sequences SEQ ID NO: 9 or SEQ ID NO: 10 which are not CDRs, such as that the regions "frame", especially the "framework" regions defined above. More preferably, the nanobody according to the invention is a nanobodies comprising or consisting of an amino acid sequence selected from the group consisting of amino acid sequences SEQ ID NO: 9 and SEQ ID NO: 10. most preferred, the nanobody according to the invention is a nanobody comprising or consisting of the amino acid sequence SEQ ID NO: 9.
Dans le contexte de l'invention, l'expression "variant fonctionnellement conservatif" fait référence à des variants dans lesquels un acide aminé donné dans un nanocorps selon l'invention est substitué sans altérer la conformation globale et la fonction du nanocorps, incluant un remplacement d'un acide aminé par un autre ayant des propriétés similaires (par exemple polarité, potentiel de liaison hydrogène, acidité, basicité, hydrophobicité, présence d'un groupe aromatique, etc). Les acides aminés ayant des propriétés similaires sont bien connus de l'homme du métier. Par exemple, l'arginine, l'histidine et la lysine des acides aminés hydrophiles-basiques et peuvent être interchangeables. De manière similaire, l'isoleucine, un acide aminé hydrophobe, peut être remplacé par la leucine, la méthionine ou la valine. De tels changements devraient avoir peu ou pas d'effet sur le poids moléculaire apparent ou le point isoélectrique du nanocorps. Un acide aminé naturel peut être remplacé par un acide aminé non naturel, tel qu'un acide aminé en configuration D, acide aminé bêta ou gamma. Des exemples de substitutions conservatives sont présentés dans le tableau 1 ci- dessous. In the context of the invention, the term "functionally conservative variant" refers to variants in which a given amino acid in a nanobody according to the invention is substituted without altering the overall conformation and function of the nanobody, including replacement. from one amino acid to another having similar properties (eg polarity, hydrogen bonding potential, acidity, basicity, hydrophobicity, presence of an aromatic group, etc.). Amino acids with similar properties are well known to those skilled in the art. For example, arginine, histidine and lysine of hydrophilic-basic amino acids and may be interchangeable. Similarly, isoleucine, a hydrophobic amino acid, may be replaced by leucine, methionine or valine. Such changes should have little or no effect on the apparent molecular weight or isoelectric point of the nanobodies. A natural amino acid can be replaced by an unnatural amino acid, such as an amino acid in D-configuration, beta-amino acid or gamma. Examples of conservative substitutions are shown in Table 1 below.
Tableau 1 : Substitutions conservatives I  Table 1: Conservative Substitutions I
Alternativement, les acides aminés conservatifs peuvent être groupés comme décrit dans Lehninger (1975, Biochemistry, 2nde Edition, Worth Publishers, Inc. New- York: NY., p. 71 -77), comme montré dans le tableau 2 ci-dessous. Alternatively, the conservative amino acids can be grouped as described in Lehninger (1975, Biochemistry, 2nd Edition, Worth Publishers, Inc. New York: NY, pp. 71-77), as shown in Table 2 below.
Tableau 2: Substitutions conservatives II Table 2: Conservative Substitutions II
Selon une autre alternative, des exemples de substitutions conservatives sont présentés dans le tableau 3 ci-dessous. According to another alternative, examples of conservative substitutions are shown in Table 3 below.
Tableau 3: Substitutions conservatives III  Table 3: Conservative Substitutions III
Résidu d'origine Exemple de substitution  Residue of origin Example of substitution
A V L I A V L I
R K Q NR K Q N
N Q H K RN Q H K R
D ED E
C SC S
G NG N
E DE D
H N Q K RH N Q K R
I L V M A FI L V M A F
L I V M A FL I V M A F
K R Q NK R Q N
M L F IM L F I
F L V I AF L V I A
P GP G
S T τ s ST τ s
w Y  w Y
Y W F T S Y W F T S
V I L M F A V I L M F A
Ces variants fonctionnellement conservatifs conservent la capacité de lier la protéine Tau, en particulier la protéine Tau sous forme d'oligomère et éventuellement sous forme de fibres. Préférentiellement ces variants fonctionnellement conservatifs présentent une affinité de liaison avec Tau, en particulier avec Tau sous forme d'oligomère et éventuellement sous forme de fibres, égale ou accrue par rapport au nanocorps correspondant. These functionally conservative variants retain the ability to bind the Tau protein, in particular the Tau protein in oligomeric form and optionally in the form of fibers. Preferably, these functionally conservative variants have a binding affinity with Tau, in particular with Tau in the form of an oligomer and optionally in the form of fibers, which is equal to or greater than that of the corresponding nanobody.
Connaissant la séquence d'acides aminés du nanocorps d'intérêt, l'homme du métier est capable de produire les nanocorps selon l'invention, en particulier les nanocorps 2C5 et S2T2M2 E6 définis ci-dessus, par des techniques conventionnelles de production de polypeptides. Par exemple, ils peuvent être synthétisés en utilisant la méthode bien connue de synthèse en phase solide (Merrifield (19962) Proc. Soc. Ex. Boil. 21 :412; Merrifield (1963) J. Am. Chem. Soc. 85:2149; Tarn et al. (1983) J. Am. Chem. Soc. 105:6442), de préférence en utilisant un appareil de synthèse peptidique disponible commercialement (tel que celui fait par Applied Biosystems, Foster City, Californie) et en suivant les instructions du fabricant.  Knowing the amino acid sequence of the nanobodies of interest, those skilled in the art are capable of producing the nanobodies according to the invention, in particular the 2C5 and S2T2M2 E6 nanobodies defined above, by conventional techniques for the production of polypeptides. . For example, they can be synthesized using the well known method of solid phase synthesis (Merrifield (1996) Proc Soc Ex Boil 21: 412 Merrifield (1963) J. Am Chem Soc 85: 2149 Tarn et al (1983) J Am Chem Soc 105: 6442), preferably using a commercially available peptide synthesizer (such as that made by Applied Biosystems, Foster City, Calif.) And following the manufacturer's instructions.
Alternativement, les nanocorps selon l'invention peuvent être synthétisés par des techniques d'ADN recombinant bien connues de l'homme du métier (Maniatis et al. (1982) Molecular Cloning: a laboratory manual, Cold Spring Harbor Laboratories, NY, 51 -54 et 412-430). Par exemple, ils peuvent être obtenus comme produits d'expression d'ADN après incorporation des séquences d'ADN codant le polypeptide d'intérêt dans des vecteurs d'expression et introduction de ces vecteurs dans les hôtes procaryotes ou eucaryotes appropriés qui exprimeront le polypeptide d'intérêt, à partir desquels ils pourront ensuite être isolés en utilisant des techniques bien connues de l'homme du métier. Comme il est bien connu de l'homme du métier, lorsqu'une protéine est synthétisée par des techniques d'ADN recombinant, elle est généralement synthétisée associée à une étiquette facilitant sa purification. De telles étiquettes sont bien connues de l'homme du métier et incluent par exemple l'hexahistidine (6His), la glutathion-S- transférase (GST), étiquette myc ou l'hémagglutinine du virus de la grippe (HA). De préférence, les nanocorps selon l'invention comprennent une étiquette myc et/ou hexahistidine. Par conséquent, un objet de l'invention est également constitué par un nanocorps comprenant ou étant constitué de la séquence d'acides aminés choisie dans le groupe constitué des séquences SEQ ID NO: 9, et SEQ ID NO: 10 comprenant en outre à leur extrémité C-terminale ou N-terminale, de préférence à leur extrémité C-terminale, une étiquette myc et/ou six résidus histidine, de manière plus préférée une étiquette myc et six résidus histidine. Comme il est bien connu de l'homme du métier, lorsqu'une protéine est associée à une étiquette facilitant sa purification, une telle protéine comprend entre la séquence native et cette étiquette une séquence permettant une coupure enzymatique entre la protéine et cette étiquette. Un nanocorps comprenant ou étant constitué de la séquence d'acides aminés choisie dans le groupe constitué des séquences SEQ ID NO: 19 et SEQ ID NO: 20 fait donc également partie de l'invention. Alternatively, the nanobodies according to the invention can be synthesized by recombinant DNA techniques well known to those skilled in the art (Maniatis et al., (1982) Molecular Cloning: a laboratory manual, Cold Spring Harbor Laboratories, NY, 51- 54 and 412-430). For example, they can be obtained as DNA expression products after incorporation of DNA sequences encoding the polypeptide of interest into expression vectors and introduction of these vectors into appropriate prokaryotic or eukaryotic hosts which will express the polypeptide. of interest, from which they can then be isolated using techniques well known to those skilled in the art. As is well known to those skilled in the art, when a protein is synthesized by recombinant DNA techniques, it is generally synthesized associated with a label facilitating its purification. Such labels are well known to those skilled in the art and include, for example, hexahistidine (6His), glutathione-S-transferase (GST), myc tag or hemagglutinin of influenza virus (HA). Preferably, the nanobodies according to the invention comprise a myc and / or hexahistidine label. Accordingly, an object of the invention is also constituted by a nanobody comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 9, and SEQ ID NO: 10 further comprising at their C-terminus or N-terminus, preferably at their C-terminus, a myc tag and / or six histidine residues, more preferably a myc tag and six histidine residues. As is well known to those skilled in the art, when a protein is associated with a label facilitating its purification, such a protein comprises between the native sequence and this label a sequence allowing an enzymatic cleavage between the protein and this label. A nanobody comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NO: 19 and SEQ ID NO: 20 is also part of the invention.
Un autre objet de l'invention concerne un acide nucléique comprenant une séquence nucléique codant le nanocorps selon la présente invention.  Another subject of the invention relates to a nucleic acid comprising a nucleic sequence coding for the nanobody according to the present invention.
Dans un mode de réalisation particulier, l'acide nucléique selon l'invention comprend ou consiste en une séquence nucléique codant un nanocorps défini par l'une des séquences d'acides aminés SEQ ID NO: 9 ou SEQ ID NO: 10. De manière préférée, l'acide nucléique selon l'invention comprend ou consiste en une séquence nucléique codant le nanocorps défini par la séquence d'acides aminés SEQ ID NO: 9.  In a particular embodiment, the nucleic acid according to the invention comprises or consists of a nucleic sequence encoding a nanobody defined by one of the amino acid sequences SEQ ID NO: 9 or SEQ ID NO: 10. preferred, the nucleic acid according to the invention comprises or consists of a nucleic acid sequence encoding the nanobodies defined by the amino acid sequence SEQ ID NO: 9.
Typiquement, ledit acide nucléique est une molécule d'ADN ou d'ARN, qui peut être incluse dans n'importe quel vecteur approprié, tel qu'un plasmide, un cosmide, un épisome, un chromosome artificiel, un phage ou un vecteur viral.  Typically, said nucleic acid is a DNA or RNA molecule, which can be included in any suitable vector, such as a plasmid, a cosmid, an episome, an artificial chromosome, a phage or a viral vector .
Les termes "vecteur", "vecteur de clonage" et "vecteur d'expression" signifient le véhicule par lequel la séquence d'ADN ou d'ARN peut être introduite dans la cellule hôte, de manière à transformer l'hôte et promouvoir l'expression (e.g. transcription et traduction) de la séquence introduite.  The terms "vector", "cloning vector" and "expression vector" mean the vehicle by which the DNA or RNA sequence can be introduced into the host cell, so as to transform the host and promote the expression (eg transcription and translation) of the introduced sequence.
Par conséquent, un autre objet de l'invention concerne un vecteur comprenant un acide nucléique selon l'invention.  Therefore, another object of the invention relates to a vector comprising a nucleic acid according to the invention.
De tels vecteurs peuvent comprendre des éléments régulateurs, tels qu'un promoteur, un activateur, un terminateur, etc., pour causer ou diriger l'expression du polypeptide. Des exemples de promoteurs et d'activateurs utilisés dans les vecteurs d'expression pour cellule animale incluent le promoteur précoce et l'activateur de SV40 (Mizukami et al. (1987) J. Biochem. 101 :1307-1310), le promoteur LTR et l'activateur de virus de la leucémie de la souris Moloney, le promoteur (Mason et a/ (1985) Ce// 41 :479- 487) et l'activateur (Gillies et al. (1983) Cell 33:717-728) de la chaîne de l'immunoglobuline, etc.  Such vectors may include regulatory elements, such as a promoter, activator, terminator, etc., to cause or direct the expression of the polypeptide. Examples of promoters and enhancers used in animal cell expression vectors include the SV40 early promoter and activator (Mizukami et al (1987) J. Biochem 101: 1307-1310), the LTR promoter. and Moloney mouse leukemia virus activator, the promoter (Mason et al (1985) Ce / 41: 479-487) and the enhancer (Gillies et al (1983) Cell 33: 717- 728) of the immunoglobulin chain, etc.
N'importe quel vecteur d'expression pour cellules animales peut être utilisé. Des exemples de vecteurs appropriés incluent pAGE107 (Miyaji et al. (1990) Cytotechnology 3:133-140), pAGE103 (Mizukami et al. (1987) J. Biochem. 101 :1307-1310), pHSG274 (Brady et al. (1984) Gene 27:223-232), pKCR (O'Hare et al. (1981 ) Proc. Natl. Acad. Sci. USA 78:1527-1531 ), pSG1 beta d2-4 (Miyaji et al. (1990) Cytotechnology 3: 133- 140), etc. Any expression vector for animal cells can be used. Examples of suitable vectors include pAGE107 (Miyaji et al (1990) Cytotechnology 3: 133-140), pAGE103 (Mizukami et al (1987) J. Biochem 101: 1307-1310), pHSG274. (Brady et al (1984) Gene 27: 223-232), pKCR (O'Hare et al (1981) Proc Natl Acad Sci USA 78: 1527-1531), pSG1 beta d2-4 (Miyaji et al (1990) Cytotechnology 3: 133-140), etc.
D'autres exemples de plasmides incluent des plasmides réplicatifs comprenant une origine de réplication, ou des plasmides intégratifs, tels que par exemple pUC, pcDNA, pBR, etc.  Other examples of plasmids include replicative plasmids comprising an origin of replication, or integrative plasmids, such as for example pUC, pcDNA, pBR, etc.
D'autres exemples de vecteurs viraux incluent les vecteurs adénoviraux, rétroviraux, du virus de l'herpès et AAV. De tels virus recombinants peuvent être produits par des techniques bien connues de l'homme du métier, telles que par transfection des cellules d'empaquetage ou par transfection transitoire avec des plasmides ou virus auxiliaires. Des exemples typiques de cellules d'empaquetage de virus incluent les cellules PA317, les cellules PsiCRIP, les cellules GPenv+, les cellules 293, etc. Des protocoles détaillés de production de tels virus recombinants déficients pour leur réplication peuvent être trouvés par exemple dans les demandes WO 95/14785, WO 96/22378, US 5,882,887, US 6,013,516, US 4,861 ,719, US 5,278,056 et WO 94/19478.  Other examples of viral vectors include adenoviral, retroviral, herpesvirus and AAV vectors. Such recombinant viruses can be produced by techniques well known to those skilled in the art, such as by transfection of packaging cells or by transient transfection with plasmids or helper viruses. Typical examples of virus packaging cells include PA317 cells, PsiCRIP cells, GPenv + cells, 293 cells, and the like. Detailed protocols for producing such replication-deficient recombinant viruses can be found, for example, in WO 95/14785, WO 96/22378, US 5,882,887, US 6,013,516, US 4,861,719, US 5,278,056, and WO 94/19478.
Un autre objet de la présente invention concerne une cellule qui a été transfectée, transduite ou transformée avec un acide nucléique et/ou un vecteur selon l'invention.  Another object of the present invention relates to a cell which has been transfected, transduced or transformed with a nucleic acid and / or a vector according to the invention.
Le termine "transformation" signifie l'introduction d'un gène ou d'une séquence d'ADN ou d'ARN "étranger" ( e. extrinsèque ou extracellulaire) dans une cellule hôte, de telle sorte que la cellule hôte exprimera le gène ou la séquence introduit pour produire la substance d'intérêt, typiquement une protéine codée par le gène ou la séquence introduit. Une cellule hôte qui reçoit et exprime l'ADN ou l'ARN introduit a été "transformée".  The terminating "transformation" means the introduction of a "foreign" (extrinsic or extracellular) DNA or RNA gene or sequence into a host cell, such that the host cell will express the gene. or the sequence introduced to produce the substance of interest, typically a protein encoded by the gene or sequence introduced. A host cell that receives and expresses the introduced DNA or RNA has been "transformed".
Les acides nucléiques selon l'invention peuvent être utilisés pour produire un nanocorps selon l'invention dans un système d'expression approprié. Le terme "système d'expression" signifie une cellule hôte et un vecteur compatible dans des conditions appropriées, e.g. pour l'expression d'une protéine codée par l'ADN étranger porté par le vecteur et introduit dans la cellule hôte.  The nucleic acids according to the invention can be used to produce a nanobody according to the invention in an appropriate expression system. The term "expression system" means a host cell and a compatible vector under appropriate conditions, e.g. for the expression of a protein encoded by the foreign DNA carried by the vector and introduced into the host cell.
Des systèmes d'expression conventionnels incluent les cellules hôtes Escherichia co//' et les vecteurs plasmidiques, les cellules hôtes d'insecte et les vecteurs Baculovirus, et les cellules hôtes de mammifère et leurs vecteurs. D'autres exemples de cellules hôtes incluent les cellules procaryotes (telles que les bactéries), et les cellules eucaryotes (telles que les cellules de levure, les cellules de mammifère, les cellules d'insecte, les cellules de plantes, etc.). Des exemples spécifiques incluent Escherichia coli, les levures Kluyveromyces ou Saccharomyces, les lignées cellulaires de mammifère (e.g. les cellules Vero, les cellules CHO, les cellules 3T3, les cellules COS, etc.) ainsi que les cultures de cellules de mammifères primaires ou établies (e.g. produites à partir de lymphoblastes, fibroblastes, cellules épithéliales, cellules nerveuses, adipocytes, etc.). Des exemples incluent également les cellules SP2/0-Ag14 de souris (ATCC CRU 581 ), les cellules P3X63-Ag8.653 de souris (ATCC CRU 580), les cellules CHO dans lesquelles un gène de la réductase dihydrofolate est défectueux, les cellules YB2/3HL.P2.G1 1 .16Ag.20 de rat (ATCC CRU 662), etc. Conventional expression systems include the host cells Escherichia co // 'and plasmid vectors, insect host cells and Baculovirus vectors, and mammalian host cells and vectors. Other examples of host cells include prokaryotic cells (such as bacteria), and eukaryotic cells (such as yeast cells, mammalian cells, insect cells, plant cells, etc.). Specific examples include Escherichia coli, Kluyveromyces or Saccharomyces yeasts, mammalian cell lines (eg Vero cells, CHO cells, 3T3 cells, COS cells, etc.) as well as primary or established mammalian cell cultures. (eg produced from lymphoblasts, fibroblasts, epithelial cells, nerve cells, adipocytes, etc.). Examples also include mouse SP2 / 0-Ag14 cells (ATCC CRU 581), mouse P3X63-Ag8.653 cells (ATCC CRU 580), CHO cells in which a dihydrofolate reductase gene is defective, YB2 cells / Rat 3HL.P2.G1 1 .16Ag.20 (ATCC CRU 662), etc.
La présente invention concerne également une méthode de production d'une cellule hôte recombinante exprimant un nanocorps selon l'invention, ladite méthode comprenant les étapes consistant à :  The present invention also relates to a method for producing a recombinant host cell expressing a nanobody according to the invention, said method comprising the steps of:
(i) introduire in vitro ou ex vivo un acide nucléique tel que décrit ci-dessus dans une cellule hôte compétente,  (i) introducing in vitro or ex vivo a nucleic acid as described above into a competent host cell,
(ii) cultiver in vitro ou ex vivo la cellule hôte recombinante obtenue et  (ii) culturing in vitro or ex vivo the recombinant host cell obtained and
(iii) éventuellement sélectionner les cellules qui expriment et/ou sécrètent ledit nanocorps. De telles cellules hôtes recombinantes peuvent être utilisées pour la production de nanocorps selon l'invention.  (iii) optionally selecting the cells that express and / or secrete said nanobody. Such recombinant host cells can be used for the production of nanobodies according to the invention.
Les nanocorps selon l'invention peuvent être produits par n'importe quelle technique connue de l'homme du métier, telles que par exemple n'importe quelle technique chimique, biologique, génétique ou enzymatique, seule ou en combinaison.  The nanobodies according to the invention may be produced by any technique known to those skilled in the art, such as for example any chemical, biological, genetic or enzymatic technique, alone or in combination.
En particulier, l'invention concerne en outre une méthode de production d'un nanocorps selon l'invention, ladite méthode comprenant les étapes consistant à:  In particular, the invention furthermore relates to a method for producing a nanobodies according to the invention, said method comprising the steps of:
(i) cultiver une cellule transduite ou transfectée ou transformée selon l'invention dans des conditions appropriées pour permettre l'expression dudit nanocorps, et  (i) culturing a transduced or transfected or transformed cell according to the invention under conditions suitable to allow the expression of said nanobody, and
(ii) récupérer le nanocorps exprimé.  (ii) recover the expressed nanobodies.
Les nanocorps selon l'invention peuvent être convenablement séparés du milieu de culture par des procédures de purification d'immunoglobulines conventionnelles, telles que par exemple protéine A-Sépharose, chromatographie hydroxilapatite, électrophorèse sur gel, dialyse ou chromatographie d'affinité.  The nanobodies according to the invention can be conveniently separated from the culture medium by conventional immunoglobulin purification procedures, such as, for example, protein A-Sepharose, hydroxilapatite chromatography, gel electrophoresis, dialysis or affinity chromatography.
Nanocorps marqués Nanobodies marked
Les nanocorps selon l'invention sont particulièrement utiles pour l'imagerie médicale. Dans ce contexte, il est particulièrement intéressant de disposer de nanocorps associés à un marqueur détectable. Les présents inventeurs ont montré que les nanocorps 2C5 et S2T2M3_E6, en particulier 2C5, conservaient leurs propriétés lorsqu'ils étaient associés à un marqueur détectable.  The nanobodies according to the invention are particularly useful for medical imaging. In this context, it is particularly advantageous to have nanobodies associated with a detectable marker. The present inventors have shown that the 2C5 and S2T2M3_E6 nanobodies, in particular 2C5, retained their properties when they were associated with a detectable marker.
La présente invention concerne donc également un nanocorps tel que défini ci- dessus lié à un marqueur détectable.  The present invention therefore also relates to a nanobody as defined above linked to a detectable marker.
Par "nanocorps lié à un marqueur détectable", on entend ici que le marqueur détectable est lié, de manière directe ou indirecte, au nanocorps, par exemple via un peptide de liaison clivable ou non-clivable, ou est incorporé dans le nanocorps. Le marqueur détectable peut en particulier être lié au nanocorps par substitution (par exemple, en substituant un H par un I au niveau des résidus tyrosine), par complexion ou par chélation. By "nanobodies bound to a detectable marker" is meant herein that the detectable label is directly or indirectly linked to the nanobody, for example via a cleavable or non-cleavable linker peptide, or is incorporated into the nanobody. The detectable marker may in particular be bound to the nanobodies by substitution (for example, by substituting an H for an I at the tyrosine residues), by complexing or by chelation.
Par "marqueur détectable", on entend ici un composé qui produit un signal détectable. Lorsqu'il est associé à un traceur, il permet de suivre le devenir du traceur dans l'organisme. Le marqueur détectable peut être un agent de contraste IRM, un agent de contraste en scintigraphie, un agent de contraste en imagerie aux rayons X, un agent de contraste ultrason, un agent de contraste en imagerie optique. Des exemples de marqueurs détectables incluent des radioéléments, des fluorophores tels que la fluorescéine, l'Alexa, la cyanine; les composés chimioluminescents tels que le luminol; les composés bioluminescents tels que la luciférase ou la phosphatase alcaline; et les agents de contraste tels que les nanoparticules ou le Gadolinium. Le choix du marqueur détectable approprié, qui dépend du système de détection utilisé, est à la portée de l'homme du métier. A titre d'exemple lorsque le système de détection est l'IRM, le marqueur détectable est de préférence une nanoparticule d'oxyde de fer ou du Gadolinium; lorsque le système de détection est l'imagerie par fluorescence, le marqueur détectable est de préférence la Fluorescéine, l'Alexa ou la cyanine; lorsque le système de détection est l'imagerie par chimioluminescence, le marqueur détectable est de préférence le luminol; lorsque le système de détection est l'imagerie par bioluminescence, le marqueur détectable est de préférence la luciférase ou la phosphatase alcaline; lorsque le système de détection est l'imagerie nucléaire, le marqueur détectable est de préférence un radioélément tel que le Gallium (68Ga) pour l'imagerie TEP, ou le technetium 99m (99mTc) pour l'imagerie SPECT. By "detectable marker" is meant here a compound that produces a detectable signal. When it is associated with a tracer, it makes it possible to follow the fate of the tracer in the body. The detectable marker may be an MRI contrast agent, a scintigraphic contrast agent, an X-ray contrast agent, an ultrasound contrast agent, an optical imaging contrast agent. Examples of detectable markers include radioelements, fluorophores such as fluorescein, Alexa, cyanine; chemiluminescent compounds such as luminol; bioluminescent compounds such as luciferase or alkaline phosphatase; and contrast agents such as nanoparticles or gadolinium. The choice of the appropriate detectable marker, which depends on the detection system used, is within the abilities of those skilled in the art. For example, when the detection system is MRI, the detectable label is preferably an iron oxide nanoparticle or gadolinium; when the detection system is fluorescence imaging, the detectable marker is preferably Fluorescein, Alexa or cyanine; when the detection system is chemiluminescence imaging, the detectable label is preferably luminol; when the detection system is bioluminescence imaging, the detectable label is preferably luciferase or alkaline phosphatase; when the detection system is nuclear imaging, the detectable marker is preferably a radioelement such as gallium ( 68 Ga) for PET imaging, or technetium 99m ( 99m Tc) for SPECT imaging.
De préférence, le marqueur détectable est un radioélément. Des exemples de radioéléments, qui sont plus particulièrement utilisés dans les techniques d'imagerie nucléaires, incluent le Technetium 99m (99mTc), l'iode 123 (123l), l'iode 125 (125l), , le fluor 18 (18F), le gallium 68 (68Ga), et n'importe quel autre radioélément utilisable chez les êtres humains. Par conséquent, de préférence, le radioélément est choisi dans le groupe constitué du 99mTc, 123l, 125l, 18F, et 68Ga. De manière préférée entre toutes, le radioélément est le 99mTc ou 68Ga, de manière encore préférée 99mTc. Preferably, the detectable marker is a radioelement. Examples of radioelements, which are more particularly used in nuclear imaging techniques, include Technetium 99m ( 99m Tc), Iodine 123 ( 123 I), 125 I ( 125 I), and fluorine 18 ( 18 F), gallium 68 ( 68 Ga), and any other radioelements usable in humans. Therefore, preferably, the radioelement is selected from the group consisting of 99m Tc, 123 l, 125 l, 18 F, and 68 Ga. Most preferably, the radioelement is 99m Tc or 68 Ga, still favorite 99m Tc.
Utilisation comme agent de contraste Use as a contrast agent
Les inventeurs ont démontré que le nanocorps 2C5 permet un marquage spécifique des corps cellulaires de l'hippocampe, du cortex entorhinal et du cortex temporal chez des patients atteints de la maladie d'Alzheimer et dans une tauopathie pure. Les inventeurs ont donc démontré que les nanocorps de l'invention, en particulier 2C5, constituaient des traceurs spécifiques des formes pathologiques de la protéine Tau, en particulier des formes pathologiques précoces de Tau, par exemple Tau sous forme d'oligomère et éventuellement sous forme de fibres, et permettent sa détection par imagerie. The inventors have demonstrated that the 2C5 nanobody allows specific labeling of the hippocampus, entorhinal cortex and temporal cortex cell bodies in patients suffering from Alzheimer's disease and in pure tauopathy. The inventors have therefore demonstrated that the nanobodies of the invention, in particular 2C5, constitute tracers specific for the pathological forms of the Tau protein, in particular early pathological forms of Tau, for example Tau in the form of oligomer and optionally in the form of fibers, and allow its detection by imaging.
L'invention propose donc un nanocorps tel que défini ci-dessus pour son utilisation comme agent de contraste dans l'imagerie médicale, en particulier l'imagerie médicale in vivo, non invasive.  The invention therefore proposes a nanobody as defined above for its use as a contrast agent in medical imaging, in particular in vivo, non-invasive medical imaging.
Elle concerne également l'utilisation d'un nanocorps tel que défini ci-dessus pour la fabrication d'un agent de contraste utile pour l'imagerie médicale, en particulier l'imagerie médicale in vivo, non invasive.  It also relates to the use of a nanobody as defined above for the manufacture of a contrast agent useful for medical imaging, in particular in vivo, non-invasive medical imaging.
Par "agent de contraste", on entend ici une substance ou une composition qui, administrée dans l'organisme, permet de marquer de manière détectable des organes ou des structures (tissu, cellule, récepteur) qui, sans agent de contraste, sont peu ou non visibles en imagerie médicale. Par extension, l'expression "agent de contraste" est utilisée pour désigner un traceur associé à un marqueur tel que défini ci-dessus.  By "contrast agent" is meant here a substance or a composition which, administered in the body, makes it possible to detectably mark organs or structures (tissue, cell, receptor) which, without a contrast agent, are or not visible in medical imaging. By extension, the term "contrast agent" is used to designate a tracer associated with a marker as defined above.
Dans le contexte de l'invention, les "méthodes d'imagerie" se réfèrent à des méthodes qui permettent de visualiser l'intérieur d'un organisme ou des organes d'un organisme. Des exemples de méthodes d'imagerie incluent des techniques invasives telles que l'échographie endocoronaire, et des techniques non-invasives telles que l'imagerie par résonnance magnétique, l'imagerie aux rayons X, l'échographie, l'imagerie optique, ou la médecine nucléaire telle que la scintigraphie, en particulier la tomographie d'émission monophotonique (SPECT) et la tomographie d'émission de positons (PET). De préférence, la méthode d'imagerie selon l'invention est la scintigraphie, en particulier la scintigraphie SPECT ou PET.  In the context of the invention, "imaging methods" refer to methods that allow the visualization of the interior of an organism or organs of an organism. Examples of imaging methods include invasive techniques such as endocoronary ultrasound, and non-invasive techniques such as magnetic resonance imaging, x-ray imaging, ultrasound, optical imaging, or nuclear medicine such as scintigraphy, especially single photon emission computed tomography (SPECT) and positron emission tomography (PET). Preferably, the imaging method according to the invention is scintigraphy, in particular SPECT or PET scintigraphy.
La scintigraphie repose sur l'administration (généralement par voie intraveineuse) d'un agent de contraste, aussi appelé radio-pharmaceutique, constitué d'un traceur marqué par un radioélément. La localisation spécifique de cet agent de contraste dans l'organisme est ensuite déterminée par détection des rayons gamma ou bêta émis.  The scintigraphy is based on the administration (usually intravenously) of a contrast agent, also called radiopharmaceutical, consisting of a tracer labeled with a radioelement. The specific localization of this contrast agent in the body is then determined by detection of gamma or beta emitted radiation.
La tomographie d'émission monophotonique et la tomographie d'émission de positons sont des techniques d'imagerie médicale nucléaire tomographique basées sur la scintigraphie et qui permettent de réaliser des images et des reconstructions en trois dimensions des organes et de leur métabolisme au moyen d'un ensemble de caméras qui tournent autour du patient.  Single-photon emission computed tomography (PET) and positron emission tomography (PET) are scintigraphic tomography-based nuclear tomographic imaging techniques that provide three-dimensional images and reconstructions of organs and their metabolism by means of a set of cameras that revolve around the patient.
La présente invention concerne également une méthode d'imagerie médicale, en particulier d'imagerie médicale in vivo, non invasive, dans laquelle un nanocorps tel que défini ci-dessus est administré à un patient. La méthode d'imagerie médicale selon l'invention peut en outre comprendre les étapes de détection de la liaison ou de l'absence de liaison du nanocorps dans des zones corporelles du patient et de visualisation des zones corporelles du patient dans lesquelles la liaison du nanocorps peut être détectée. The present invention also relates to a medical imaging method, in particular non-invasive in vivo medical imaging, in which a nanobody as defined above is administered to a patient. The medical imaging method according to the invention may further comprise the steps of detecting the link or the absence binding nanobodies in patient's body areas and viewing patient's body areas in which nanobody binding can be detected.
Dans le contexte de l'invention, un "patient" désigne un mammifère humain ou non-humain, tel qu'un rongeur (rat, souris, lapin), un primate (chimpanzé), un félin (chat), un canin (chien). De préférence, l'individu est humain.  In the context of the invention, a "patient" designates a human or non-human mammal, such as a rodent (rat, mouse, rabbit), a primate (chimpanzee), a feline (cat), a canine (dog ). Preferably, the individual is human.
Dans un mode de réalisation particulier, le terme « patient » désigne un humain qui présente des symptômes associés à une tauopathie. Selon la tauopathie, ces symptômes peuvent être par exemple le syndrome de parkinson, la dystonie axiale, le phénomène de main capricieuse, ou des troubles cognitifs du patient.  In a particular embodiment, the term "patient" refers to a human who exhibits symptoms associated with tauopathy. Depending on the tauopathy, these symptoms may be for example Parkinson's syndrome, axial dystonia, capricious hand phenomenon, or cognitive disorders of the patient.
N'importe quelle méthode d'administration, connue de l'homme du métier, peut être utilisée pour administrer le nanocorps selon l'invention au patient. En particulier, le nanocorps peut être administré par exemple par voie orale, par inhalation or par voie parentérale (en particulier par injection intraveineuse). Quand la voie parentérale est choisie, le nanocorps peut être sous forme de solutions et de suspensions injectables, conditionné en ampoules ou flacons. Les formes d'administration parentérale sont conventionnellement obtenues par mélange du nanocorps selon l'invention avec des tampons, stabilisants, conservateurs, agents solubilisants, agents isotoniques et agents de mise en suspension. Selon des techniques connues, ces mélanges peuvent être stérilisés et conditionnés sous la forme d'injections intraveineuses. L'homme du métier peut par exemple utiliser des tampons à base de sels de phosphate en tant que tampons. Des exemples d'agents de mise en suspension incluent la méthylcellulose, l'acacia, et la carbocyméthylcellulose de sodium. Des exemples de stabilisants incluent le sulfite de sodium et le métasulfite de sodium, et des exemples de conservateurs incluent le p- hydroxybenzoate de sodium, l'acide sorbique, le crésol et le chlorocrésol.  Any method of administration known to those skilled in the art can be used to administer the nanobody according to the invention to the patient. In particular, the nanobody can be administered, for example, orally, by inhalation or parenterally (in particular by intravenous injection). When the parenteral route is chosen, the nanobody can be in the form of injectable solutions and suspensions, packaged in ampoules or flasks. The forms of parenteral administration are conventionally obtained by mixing the nanobodies according to the invention with buffers, stabilizers, preservatives, solubilizing agents, isotonic agents and suspending agents. According to known techniques, these mixtures can be sterilized and packaged in the form of intravenous injections. Those skilled in the art may for example use buffers based on phosphate salts as buffers. Examples of suspending agents include methylcellulose, acacia, and sodium carbocymethylcellulose. Examples of stabilizers include sodium sulfite and sodium metasulfite, and examples of preservatives include sodium p-hydroxybenzoate, sorbic acid, cresol and chlorocresol.
La quantité de nanocorps administrée dépend naturellement de la voie d'administration, de la taille et/ou du poids du patient, et de la technique de détection utilisée.  The amount of nanobodies administered depends naturally on the route of administration, the size and / or weight of the patient, and the detection technique used.
Dans le contexte de l'invention, le terme "zone corporelle" fait référence à une région déterminée de l'organisme. Il peut s'agir par exemple d'un organe, d'une partie d'un organe ou d'un tissu, tel que le cerveau, en particulier l'hippocampe, le cortex entorhinal ou le cortex temporal.  In the context of the invention, the term "body area" refers to a specific region of the body. It may be, for example, an organ, a part of an organ or a tissue, such as the brain, in particular the hippocampus, the entorhinal cortex or the temporal cortex.
Dans un mode de réalisation particulier, le nanocorps selon l'invention est utilisé comme agent de contraste dans l'imagerie médicale pour visualiser la protéine Tau sous forme pathologique, en particulier, pour visualiser Tau sous forme choisie parmi les oligomères, les fibres, les paires hélicoïdales de filaments et les filaments droit, de préférence les oligomères et les fibres, de manière encore préférée les oligomères, chez un patient. In a particular embodiment, the nanobody according to the invention is used as a contrast agent in medical imaging to visualize the Tau protein in pathological form, in particular to visualize Tau in the form chosen from oligomers, fibers, helicoidal pairs of filaments and straight filaments, of oligomers and fibers, more preferably oligomers, in a patient.
Dans un mode de réalisation, le nanocorps selon l'invention est utilisé comme agent de contraste dans l'imagerie médicale pour visualiser les dégénérescences neurofibrillaires (DNFs) chez un patient.  In one embodiment, the nanobody according to the invention is used as a contrast agent in medical imaging to visualize neurofibrillary tangles (DNFs) in a patient.
Dans un exemple, la distribution corticale et le nombre des, par exemple, DNFs sont corrélés avec les troubles mnésiques chez le patient atteint par exemple de MA. In one example, cortical distribution and the number of, for example, DNFs are correlated with memory disorders in patients with, for example, AD.
Utilisation diagnostique Diagnostic use
L'apparition des formes pathologiques de la protéine Tau, telles que définies ci- dessus, est un marqueur d'une tauopathie. En outre, la détection des formes pathologiques de Tau, en particulier des formes pathologiques précoces, telles que la protéine Tau sous formes d'oligomères et éventuellement de fibres, permet d'identifier la présence des formes toxiques impliquées dans la propagation ou le développement d'une des tauopathies. L'identification des formes pathologiques, en particulier des formes pathologiques précoces, telles que Tau sous forme d'oligomères et éventuellement de fibres, constitue donc la caractérisation d'une tauopathie et/ou l'identification du début d'une tauopathie. D'autre part, la possibilité de suivre par imagerie l'évolution, c'est-à-dire la progression ou la régression, d'une forme pathologique de Tau précédemment identifiée représente une modalité pour évaluer l'efficacité d'un traitement thérapeutique chez un patient chez qui on a diagnostiqué une tauopathie.  The appearance of pathological forms of Tau protein, as defined above, is a marker of tauopathy. In addition, the detection of pathological forms of Tau, in particular early pathological forms, such as Tau protein in the form of oligomers and possibly fibers, makes it possible to identify the presence of the toxic forms involved in the propagation or development of Tau. one of the tauopathies. The identification of pathological forms, in particular early pathological forms, such as Tau in the form of oligomers and possibly fibers, therefore constitutes the characterization of a tauopathy and / or the identification of the beginning of a tauopathy. On the other hand, the possibility of imaging the evolution, that is to say the progression or regression, of a pathological form of Tau previously identified represents a modality for evaluating the efficacy of a therapeutic treatment. in a patient diagnosed with tauopathy.
L'invention concerne donc également un nanocorps tel que défini ci-dessus pour son utilisation dans des méthodes de diagnostic ou de pronostic.  The invention therefore also relates to a nanobody as defined above for its use in diagnostic or prognostic methods.
Par "méthode diagnostique" ou "diagnostic", on entend ici une méthode permettant de déterminer si un individu souffre d'une pathologie.  By "diagnostic method" or "diagnosis" is meant here a method for determining whether an individual suffers from a pathology.
Par "méthode pronostigue" ou "pronostic", on entend ici une méthode permettant de déterminer si un individu risque de développer une pathologie.  By "prognostic method" or "prognosis" is meant here a method for determining whether an individual is at risk of developing a pathology.
De préférence, le nanocorps tel que défini ci-dessus est utilisé pour le diagnostic ou le pronostic d'une tauopathie.  Preferably, the nanobody as defined above is used for the diagnosis or prognosis of a tauopathy.
Les tauopathies sont telles que définies ci-dessus. De préférence, le nanocorps selon l'invention est utilisé pour le diagnostic ou le pronostic de la maladie d'Alzheimer.  The tauopathies are as defined above. Preferably, the nanobody according to the invention is used for the diagnosis or prognosis of Alzheimer's disease.
La présence, par exemple, de Tau sous forme d'oligomère expose le sujet à un risque de développer une tauopathie, notamment la maladie d'Alzheimer. Le nanocorps selon l'invention peut donc être utilisé pour détecter un risque de survenue d'une tauopathie, en particulier une maladie d'Alzheimer, chez un patient. Par "risque de survenue", on entend ici la probabilité qu'un individu développe une pathologie. The presence, for example, of Tau in oligomeric form exposes the subject to a risk of developing tauopathy, especially Alzheimer's disease. The nanobody according to the invention can therefore be used to detect a risk of occurrence of tauopathy, in particular Alzheimer's disease, in a patient. By "risk of occurrence" is meant here the probability that an individual develops a pathology.
La présente invention concerne également une méthode de diagnostic d'une tauopathie et/ou de détection d'un risque de survenue d'une tauopathie chez un patient, ladite méthode comprenant les étapes consistant à administrer un nanocorps tel que défini ci-dessus audit patient et à détecter ledit nanocorps dans l'organisme dudit patient, la détection d'une localisation préférentielle dudit nanocorps au niveau du cerveau étant indicatrice d'une tauopathie et/ou d'un risque de survenue d'une tauopathie.  The present invention also relates to a method for diagnosing a tauopathy and / or detecting a risk of occurrence of tauopathy in a patient, said method comprising the steps of administering a nanobody as defined above to said patient and detecting said nanobodies in the body of said patient, detecting a preferential location of said nanobody in the brain indicative of tauopathy and / or risk of occurrence of tauopathy.
Dans un mode de réalisation, ladite méthode comprend en outre les étapes consistant à administrer un marqueur de plaques amyloïdes audit patient et à détecter ledit marqueur de plaques amyloïdes dans l'organisme dudit patient, la détection d'une localisation préférentielle dudit nanocorps et dudit marqueur de plaques amyloïdes au niveau du cerveau étant indicatrice d'une tauopathie et/ou d'un risque de survenue d'une tauopathie, en particulier de la maladie d'Alzheimer.  In one embodiment, said method further comprises the steps of administering an amyloid plaque marker to said patient and detecting said amyloid plaque marker in said patient's body, detecting a preferential location of said nanobody and said marker amyloid plaques in the brain indicative of tauopathy and / or risk of occurrence of tauopathy, especially Alzheimer's disease.
En conséquence, la présente invention concerne également une méthodede diagnostic d'une tauopathie, en particulier la maladie d'Alzheimer et/ou de détection d'un risque de survenue d'une tauopathie, en particulier de la maladie d'Alzheimer, chez un patient, ladite méthode comprenant les étapes consistant à :  Consequently, the present invention also relates to a method for diagnosing a tauopathy, in particular Alzheimer's disease and / or to detecting a risk of occurrence of tauopathy, in particular Alzheimer's disease, in a patient. patient, said method comprising the steps of:
i) administrer un nanocorps selon l'invention audit patient et à détecter ledit nanocorps dans l'organisme dudit patient, et i) administering a nanobody according to the invention to said patient and detecting said nanobodies in the body of said patient, and
ii) administrer un marqueur de plaques amyloïdes audit patient et à détecter ledit marqueur de plaques amyloïdes dans l'organisme dudit patient, ii) administering an amyloid plaque marker to said patient and detecting said amyloid plaque marker in the body of said patient,
la détection d'une localisation préférentielle dudit nanocorps et dudit marqueur de plaques amyloïdes au niveau du cerveau étant indicatrice d'une tauopathie, en particulier de la maladie d'Alzheimer et/ou d'un risque de survenue d'une tauopathie, en particulier de la maladie d'Alzheimer. detecting a preferential location of said nanobody and said amyloid plaque marker in the brain indicative of tauopathy, particularly Alzheimer's disease and / or risk of occurrence of tauopathy, particularly of Alzheimer's disease.
Dans une mode de réalisation ces méthodes de diagnostic d'une tauopathie sont des méthodes in vivo.  In one embodiment, these methods of diagnosing tauopathy are in vivo methods.
Par "marqueur de plaques amyloïdes" on entend des traceurs liés à un marqueur détectable qui se lie d'une façon spécifique aux plaques amyloïdes et qui sont utilisables comme agent de contraste dans l'imagerie médicale in vivo, non invasive. Des marqueurs de plaques amyloïdes sont par exemple des traceurs 18F telle que 18F-florbetapir, 18F- florbetaben, 18F-flutemetanol et 18F-AZD4694-marque. By "amyloid plaque marker" is meant tracers bound to a detectable marker which binds specifically to amyloid plaques and is useful as a contrast agent in non-invasive, in vivo medical imaging. Markers of amyloid plaques are, for example tracer 18F-18F as florbetapir, florbetaben 18F, 18F and 18F-flutemetanol-AZD4694 bookmark.
Il est décrit dans l'art antérieur et il est donc connu de l'homme du métier, selon les tauopathies particulières, quelle région cérébrale est affecté par la présence des formes pathologiques de Tau. La localisation dans le cerveau des dégénérescences neurofibrillaires ou d'autres forme pathologiques de Tau et éventuellement des plaques amyloides est décrite pour plusieurs tauopathies, par exemple dans Catafau, AM et Bullich, S (Clin Transi Imaging. 2015;3(1 ):39-55. Epub 2015 Jan 21 .) ou dans Tranchant, C. (médicine/sciences A1997 ; 13 : 989-97). It is described in the prior art and it is therefore known to those skilled in the art, according to particular tauopathies, which brain region is affected by the presence of pathological forms of Tau. The localization in the brain of degeneration neurofibrillary or other pathological form of Tau and possibly amyloid plaques is described for several tauopathies, for example in Catafau, AM and Bullich, S (Clin Transi Imaging, 2015; 3 (1): 39-55. Epub 2015 Jan 21 .) or in Tranchant, C. (Medicine / Science A1997; 13: 989-97).
Par "localisation préférentielle", on entend que la quantité de nanocorps détectée au niveau du cerveau, en particulier par exemple aux niveaux des corps cellulaires de l'hippocampe, néocortex frontal, du cortex entorhinal et du cortex temporal est supérieure au bruit de fond qui correspond à une localisation non spécifique du nanocorps dans l'organisme.  By "preferential localization" it is meant that the amount of nanobodies detected in the brain, in particular for example at the levels of the hippocampus, frontal neocortex, entorhinal cortex and temporal cortex cell bodies, is greater than the background noise which corresponds to a non-specific localization of the nanobody in the body.
Par exemple, la paralyse supranucléaire progressive est caractérisée par la présence de DNF au niveau du tronc cérébral et du néocortex frontal.  For example, progressive supranuclear paralyzation is characterized by the presence of DNF in the brain stem and frontal neocortex.
Dans un autre exemple, la dégénérescence corticobasale est caractérisée par la présence de DNF au niveau du cortex pariétal.  In another example, corticobasal degeneration is characterized by the presence of DNF in the parietal cortex.
Dans un autre exemple, la maladie d'Alzheimer est caractérisée par la présence de DNF au niveau des structures enthorino-corticales.  In another example, Alzheimer's disease is characterized by the presence of DNF at enthorino-cortical structures.
L'invention concerne également le nanocorps selon l'invention pour son utilisation pour le suivi thérapeutique d'une tauopathie chez un sujet chez qui une tauopathie a été diagnostiquée.  The invention also relates to the nanobody according to the invention for its use for the therapeutic monitoring of a tauopathy in a subject in whom a tauopathy has been diagnosed.
Elle concerne aussi l'utilisation du nanocorps selon l'invention pour la fabrication d'un agent de contraste utile pour le suivi thérapeutique d'une tauopathie chez un sujet chez qui une tauopathie a été diagnostiquée.  It also relates to the use of the nanobodies according to the invention for the manufacture of a contrast agent useful for the therapeutic monitoring of a tauopathy in a subject in whom a tauopathy has been diagnosed.
Par "suivi thérapeutique" on entend ici l'observation de la réponse du sujet au traitement qui lui est administré. L'effet thérapeutique d'un traitement est généralement associé à un ralentissement ou une inhibition de la progression d'une maladie, à une réversion de la maladie, ou d'un ou plusieurs symptômes associés à cette maladie. Inversement, une absence d'effet thérapeutique peut se traduire par une stabilité, voire une accélération, de la progression de la maladie ou d'un ou plusieurs de ses symptômes. Par exemple si une tauopathie a été diagnostiquée à cause de la présence des formes pathologiques de Tau, le suivi thérapeutique peut être réalisé en observant la disparition, la régression, le maintien, ou la croissance des formes pathologiques de Tau. Ainsi l'utilisation selon l'invention peut comprendre les étapes consistant à :  By "therapeutic monitoring" is meant here the observation of the subject's response to the treatment administered to him. The therapeutic effect of a treatment is generally associated with slowing or inhibiting the progression of a disease, a reversion of the disease, or one or more symptoms associated with this disease. Conversely, an absence of a therapeutic effect can result in a stability or an acceleration of the progression of the disease or one or more of its symptoms. For example, if a tauopathy was diagnosed because of the presence of pathological forms of Tau, the therapeutic follow-up can be realized by observing the disappearance, the regression, the maintenance, or the growth of the pathological forms of Tau. Thus the use according to the invention may comprise the steps of:
a) administrer un nanocorps tel que défini ci-dessus à un sujet chez qui on a détecté une forme pathologique de Tau; a) administering a nanobody as defined above to a subject in whom a pathological form of Tau has been detected;
b) détecter la liaison du nanocorps au niveau de ladite forme pathologique de Tau; c) répéter les étapes a) et b) avant et après administration d'un traitement audit sujet ; une absence ou une diminution de la liaison du nanocorps au niveau de ladite forme pathologique de Tau étant indicatrice d'un traitement ayant un effet thérapeutique. b) detecting the binding of the nanobody to said pathological form of Tau; c) repeating steps a) and b) before and after administration of a treatment to said subject; an absence or a decrease in the binding of the nanobody to said Tau pathological form indicative of a treatment having a therapeutic effect.
La « forme pathologique de Tau » est telle que défini dans la section « tauopathie » ci-dessus.  The "pathological form of Tau" is as defined in the "tauopathy" section above.
De préférence le traitement est un traitement d'une taupopathie et comprend par exemple l'utilisation d'inhibiteurs de l'agrégation de Tau. Dans un autre exemple, le traitement d'une taupopathie comprend l'utilisation des nanobodies de l'invention.  Preferably the treatment is a treatment of a taupopathy and comprises for example the use of inhibitors of Tau aggregation. In another example, the treatment of a taupopathy includes the use of the nanobodies of the invention.
Utilisation comme médicament Use as medicine
Récemment, il a été montré que les formes pathologiques de Tau, par exemple, des formes toxiques extracellulaires, représentaient les formes toxiques de Tau (Usenovic et al., 2015) impliquées dans la propagation (Goedert et al., 2014) des tauopathies telles que la MA au sein du cerveau, par contamination de proche en proche des neurones. En conséquence, cibler les formes toxiques de Tau pour inhiber leur agrégation et donc la formation des paires de filaments appariés en hélice (PHFs) et des filaments droits (SFs) est un traitement prometteur des tauopathies, en particulier de la maladie d'Alzheimer.  Recently, pathological forms of Tau, for example extracellular toxic forms, have been shown to represent the toxic forms of Tau (Usenovic et al., 2015) involved in the propagation (Goedert et al., 2014) of such tauopathies. than AD in the brain, by contamination of the neurons. Consequently, targeting the toxic forms of Tau to inhibit their aggregation and thus the formation of helical paired filament pairs (PHFs) and straight filaments (SFs) is a promising treatment for tauopathies, particularly Alzheimer's disease.
En conséquence, l'invention concerne en outre l'utilisation d'un nanocorps tel que défini ci-dessus pour la fabrication d'un médicament, en particulier un médicament destiné au traitement d'une tauopathie. Une méthode de traitement comprenant l'administration d'une quantité thérapeutiquement efficace du nanocorps tel que défini ci-dessus à un patient en ayant besoin, fait également partie de la présente invention.  Accordingly, the invention further relates to the use of a nanobody as defined above for the manufacture of a medicament, in particular a medicament for the treatment of tauopathy. A method of treatment comprising administering a therapeutically effective amount of the nanobody as defined above to a patient in need thereof is also part of the present invention.
L'utilisation d'un nanocorps anti- Tau permet donc d'inhiber l'avancement de l'agrégation de Tau.  The use of an anti-Tau nanobodies therefore makes it possible to inhibit the progress of Tau aggregation.
Par "traitement" d'une tauopathie on entend le "traitement thérapeutique" (ou curatif) d'une tauopathie, qui inclut le ralentissement ou l'inhibition de l'évolution d'une tauopathie. On entend également le "traitement prophylactique" d'une tauopathie qui inclut notamment la prévention des formations des DNF.  By "treatment" of a tauopathy is meant the "therapeutic treatment" (or curative) of a tauopathy, which includes slowing or inhibiting the evolution of a tauopathy. The "prophylactic treatment" of tauopathy is also understood to include the prevention of DNF formation.
Par « prévention », on entend le fait de d'empêcher ou de retarder la survenue ou de diminuer l'intensité des manifestations cliniques ou biochimiques associées à la tauopathie.  "Prevention" refers to preventing or delaying the onset or decreasing the intensity of the clinical or biochemical manifestations associated with tauopathy.
L'homme du métier sait, de par ses connaissances générales, déterminer quelles sont les manifestations cliniques ou biochimiques associées à une tauopathie donnée et qui sont susceptibles d'être améliorées (traitement) ou bien empêchées, retardées ou diminuées en intensité (prévention). Dans le cadre des tauopathies, un paramètre biologique d'intérêt peut-être la présence et la localisation de Tau sous forme pathologique, en particulier Tau sous forme pathologique précoce, telle que la protéine Tau sous forme d'oligomères et éventuellement Tau sous forme de fibres. L'invention concerne plus particulièrement le nanocorps tel que défini ci-dessus pour son utilisation pour le traitement d'une tauopathie et/ou à la prévention d'une tauopathie, de préférence pour le traitement et/ou la prévention de la maladie d'Alzheimer. The person skilled in the art knows, from his general knowledge, to determine what are the clinical or biochemical manifestations associated with a given tauopathy and which are likely to be improved (treatment) or prevented, delayed or diminished in intensity (prevention). In the context of tauopathies, a biological parameter of interest may be the presence and location of Tau in pathological form, in particular Tau in early pathological form, such as Tau protein in the form of oligomers and optionally Tau in the form of fibers. The invention more particularly relates to the nanobody as defined above for its use for the treatment of tauopathy and / or the prevention of tauopathy, preferably for the treatment and / or prevention of the disease of tauopathy. Alzheimer.
L'invention concerne également l'utilisation d'un nanocorps tel que défini ci-dessus pour la fabrication d'un médicament destiné au traitement d'une tauopathie et/ou à la prévention d'une tauopathie chez un patient susceptible de présenter une tauopathie.  The invention also relates to the use of a nanobody as defined above for the manufacture of a medicament for the treatment of tauopathy and / or the prevention of tauopathy in a patient likely to present a tauopathy .
L'invention concerne également une méthode de traitement d'une tauopathie et/ou de prévention d'une tauopathie chez un patient en ayant besoin, comprenant l'administration d'une quantité thérapeutiquement efficace d'un nanocorps tel que défini ci- dessus à un patient en ayant besoin.  The invention also relates to a method of treating tauopathy and / or preventing tauopathy in a patient in need thereof, comprising administering a therapeutically effective amount of a nanobody as defined above to a patient in need.
Préférentiellement, le nanocorps selon l'invention est utilisé pour traiter les formes pathologiques de Tau, en particulier les formes pathologiques précoces, tel que les formes d'oligomères de Tau et éventuellement les formes de fibres de Tau.  Preferably, the nanobodies according to the invention are used to treat the pathological forms of Tau, in particular the early pathological forms, such as the Tau oligomer forms and optionally the Tau fiber forms.
Le nanocorps selon l'invention peut être administré par exemple par voie orale, par inhalation, par voie parentérale (en particulier par injection intraveineuse), sous une forme appropriée. Lorsque la voie parentérale est envisagée, le nanocorps peut être sous la forme de solutés et suspensions injectables conditionnées en ampoules ou flacons. Les formes pour l'administration parentérale sont obtenues de façon conventionnelle par mélange du nanocorps avec des tampons, des agents stabilisants, des conservateurs, des agents solubilisants, des agents isotoniques et des agents de mise en suspension. Conformément aux techniques connues, ces mélanges sont ensuite stérilisés puis conditionnés sous la forme d'injections intraveineuses. A titre de tampon, l'homme du métier pourra utiliser des tampons à base de sels de phosphate organique. Des exemples d'agents de mise en suspension englobent la méthylcellulose, l'hydroxyéthylcellulose, l'hydroxy-propylcellulose, l'acacia et la carboxyméthylcellulose sodique. En outre, des stabilisants utiles selon l'invention sont le sulfite de sodium et le métasulfite de sodium, tandis que l'on peut citer le p-hydroxybenzoate de sodium, l'acide sorbique, le crésol et le chlorocrésol en tant que conservateurs.  The nanobody according to the invention can be administered, for example, orally, by inhalation, parenterally (in particular by intravenous injection), in a suitable form. When the parenteral route is envisaged, the nanobody can be in the form of injectable solutes and suspensions packaged in ampoules or flasks. Forms for parenteral administration are conventionally obtained by mixing the nanobody with buffers, stabilizing agents, preservatives, solubilizing agents, isotonic agents and suspending agents. In accordance with known techniques, these mixtures are then sterilized and then packaged in the form of intravenous injections. As a buffer, those skilled in the art may use buffers based on organic phosphate salts. Examples of suspending agents include methylcellulose, hydroxyethylcellulose, hydroxypropylcellulose, acacia and sodium carboxymethylcellulose. In addition, useful stabilizers according to the invention are sodium sulfite and sodium metasulfite, while sodium p-hydroxybenzoate, sorbic acid, cresol and chlorocresol can be mentioned as preservatives.
La quantité de nanocorps administrée dépend naturellement du mode d'administration, de la taille et/ou du poids du patient, et de la nature de l'agent cytotoxique qui peut lui être associé.  The amount of nanobodies administered depends naturally on the mode of administration, the size and / or weight of the patient, and the nature of the cytotoxic agent that may be associated with it.
La présente invention concerne également une composition pharmaceutique comprenant un nanocorps tel que défini ci-dessus en association avec un véhicule pharmaceutiquement acceptable. Le terme "pharmaceutiquement" ou "pharmaceutiquement acceptable" se réfère à des entités moléculaires et compositions qui ne produisent pas de réactions secondaires, allergiques ou autrement fâcheuses lorsqu'elles sont administrées à un mammifère, en particulier un humain. The present invention also relates to a pharmaceutical composition comprising a nanobody as defined above in association with a pharmaceutically acceptable vehicle. The term "pharmaceutically" or "pharmaceutically acceptable" refers to molecular entities and compositions which do not produce side, allergic or otherwise objectionable reactions when administered to a mammal, particularly a human.
Dans le contexte de l'invention, l'expression "véhicule pharmaceutiquement acceptable" inclut n'importe quel solvant, milieu de dispersion, revêtement, agent antibactérien ou antifongique, agent isotonique ou retardant l'absorption, et analogues. L'utilisation de tels milieux et agents pour les substances pharmaceutiquement actives est bien connue de l'homme du métier. A l'exception du cas où un milieu ou agent conventionnel est incompatible avec l'ingrédient actif, son utilisation dans les compositions pharmaceutiques est envisagée. Des ingrédients actifs supplémentaires peuvent également être incorporés dans les compositions.  In the context of the invention, the term "pharmaceutically acceptable carrier" includes any solvent, dispersion medium, coating, antibacterial or antifungal agent, isotonic or absorption delaying agent, and the like. The use of such media and agents for pharmaceutically active substances is well known to those skilled in the art. With the exception of the case where a medium or conventional agent is incompatible with the active ingredient, its use in pharmaceutical compositions is envisaged. Additional active ingredients may also be incorporated into the compositions.
Utilisation pour détecter in vitro Tau Use to detect in vitro Tau
La présente invention concerne également l'utilisation d'un nanocorps tel que défini ci- dessus pour la détection in vitro de la protéine Tau sous forme d'oligomères dans un échantillon.  The present invention also relates to the use of a nanobody as defined above for the in vitro detection of Tau protein as oligomers in a sample.
Par "échantillon", on entend ici une partie d'un élément plus grand. De préférence, l'échantillon est une substance d'origine biologique. Des exemples d'échantillons biologiques incluent, mais ne sont pas limités à, des parties d'organes ou de tissus tels que tel que le cerveau, en particulier l'hippocampe, le cortex entorhinal ou cortex temporal, le sang, en particulier le sang cérébral, le liquide céphalo-rachidien. De préférence un échantillon dans le contexte de l'invention concerne un échantillon de cerveau. La présente invention sera illustrée plus en détail par les figures, les séquences et l'exemple ci-dessous.  By "sample" is meant here a part of a larger element. Preferably, the sample is a substance of biological origin. Examples of biological samples include, but are not limited to, parts of organs or tissues such as the brain, particularly the hippocampus, the entorhinal cortex or temporal cortex, blood, particularly blood brain, the cerebrospinal fluid. Preferably a sample in the context of the invention relates to a brain sample. The present invention will be further illustrated by the figures, sequences and the example below.
BRÈVE DESCRIPTION DES SÉQUENCES BRIEF DESCRIPTION OF SEQUENCES
SEQ ID NO: 1 montre la séquence d'acides aminés consensus de CDR1 des nanocorps de l'invention dans laquelle l'acide aminé Xi est S ou R, X2 est D ou Y et X3 est T ou A .SEQ ID NO: 1 shows the consensus amino acid sequence of CDR1 of the nanobodies of the invention wherein the amino acid Xi is S or R, X 2 is D or Y and X 3 is T or A.
SEQ ID NO: 2 montre la séquence d'acides aminés consensus de CDR2 des nanocorps de l'invention dans laquelle l'acide aminé Xi est P ou R et X2 est S ou V. SEQ ID NO: 2 shows the consensus amino acid sequence of CDR2 of the nanobodies of the invention wherein the amino acid Xi is P or R and X 2 is S or V.
SEQ ID NO: 3 montre la séquence d'acides aminés de CDR1 du nanocorps 2C5.  SEQ ID NO: 3 shows the amino acid sequence of CDR1 of 2C5 nanobody.
SEQ ID NO: 4 montre la séquence d'acides aminés de CDR2 du nanocorps 2C5.  SEQ ID NO: 4 shows the amino acid sequence of CDR2 of the 2C5 nanobody.
SEQ ID NO: 5 montre la séquence d'acides aminés de CDR3 du nanocorps 2C5. SEQ ID NO: 6 montre la séquence d'acides aminés de CDR1 du nanocorps S2T2M3_E6. SEQ ID NO: 7 montre la séquence d'acides aminés de CDR2 du nanocorps S2T2M3_E6. SEQ ID NO: 8 montre la séquence d'acides aminés de CDR3 du nanocorps S2T2M3_E6. SEQ ID NO: 9 montre la séquence d'acides aminés de la région variable du nanocorps 2C5. SEQ ID NO: 5 shows the amino acid sequence of CDR3 of the 2C5 nanobody. SEQ ID NO: 6 shows the amino acid sequence of CDR1 of the S2T2M3_E6 nanobody. SEQ ID NO: 7 shows the amino acid sequence of CDR2 of the S2T2M3_E6 nanobody. SEQ ID NO: 8 shows the amino acid sequence of CDR3 of the S2T2M3_E6 nanobody. SEQ ID NO: 9 shows the amino acid sequence of the 2C5 nanobody variable region.
SEQ ID NO: 10 montre la séquence d'acides aminés de la région variable du nanocorps S2T2M3_E6.  SEQ ID NO: 10 shows the amino acid sequence of the variable region of the S2T2M3_E6 nanobody.
SEQ ID NO: 11 montre la séquence d'acides aminés de la région FR1 du nanocorps 2C5. SEQ ID NO: 12 montre la séquence d'acides aminés de la région FR2 du nanocorps 2C5. SEQ ID NO: 13 montre la séquence d'acides aminés de la région FR3 du nanocorps 2C5. SEQ ID NO: 14 montre la séquence d'acides aminés de la région FR4 du nanocorps 2C5. SEQ ID NO: 15 montre la séquence d'acides aminés de la région FR1 du nanocorps S2T2M2_E6.  SEQ ID NO: 11 shows the amino acid sequence of the FR1 region of the 2C5 nanobody. SEQ ID NO: 12 shows the amino acid sequence of the FR2 region of the 2C5 nanobody. SEQ ID NO: 13 shows the amino acid sequence of the FR3 region of the 2C5 nanobody. SEQ ID NO: 14 shows the amino acid sequence of the FR4 region of the 2C5 nanobody. SEQ ID NO: 15 shows the amino acid sequence of the FR1 region of the S2T2M2_E6 nanobody.
SEQ ID NO: 16 montre la séquence d'acides aminés de la région FR2 du nanocorps S2T2M2_E6.  SEQ ID NO: 16 shows the amino acid sequence of the FR2 region of the S2T2M2_E6 nanobody.
SEQ ID NO: 17 montre la séquence d'acides aminés de la région FR3 du nanocorps S2T2M2_E6.  SEQ ID NO: 17 shows the amino acid sequence of the FR3 region of the S2T2M2_E6 nanobody.
SEQ ID NO: 18 montre la séquence d'acides aminés de la région FR4 du nanocorps S2T2M2_E6  SEQ ID NO: 18 shows the amino acid sequence of the FR4 region of the S2T2M2_E6 nanobody
SEQ ID NO: 19 montre la séquence d'acides aminés de la région variable du nanocorps 2C5 avec une étiquette myc et six résidus histidine. SEQ ID NO: 19 shows the amino acid sequence of the variable region of 2C5 nanobodies with a myc tag and six histidine residues.
SEQ ID NO: 20 montre la séquence d'acides aminés de la région variable du nanocorps S2T2M3_E6 avec une une étiquette myc et six résidus histidine.  SEQ ID NO: 20 shows the amino acid sequence of the S2T2M3_E6 nanowind variable region with a myc tag and six histidine residues.
SEQ ID NO: 21 montre la séquence d'acides aminés de l'isoforme 2N4R de la protéine Tau, également appelée Tau-F, avec 441 acides aminés.  SEQ ID NO: 21 shows the amino acid sequence of the 2N4R isoform of Tau protein, also called Tau-F, with 441 amino acids.
SEQ ID NO: 22 montre la séquence d'acides aminés de l'isoforme 1 N4R de la protéine Tau, également appelée Tau-E, avec 412 acides aminés.  SEQ ID NO: 22 shows the amino acid sequence of the N4R 1 isoform of the Tau protein, also called Tau-E, with 412 amino acids.
SEQ ID NO: 23 montre la séquence d'acides aminés de l'isoforme 0N4R de la protéine Tau, également appelée Tau-D, avec 383 acides aminés.  SEQ ID NO: 23 shows the amino acid sequence of the 0N4R isoform of Tau protein, also called Tau-D, with 383 amino acids.
SEQ ID NO: 24 montre la séquence d'acides aminés de la isoforme 2N3R de la protéine Tau, également appelée Tau-C, avec 410 acides aminés. SEQ ID NO: 24 shows the amino acid sequence of the 2N3R isoform of Tau protein, also called Tau-C, with 410 amino acids.
SEQ ID NO: 25 montre la séquence d'acides aminés de l'isoforme 1 N3R de la protéine Tau, également appelée Tau-B, avec 381 acides aminés.  SEQ ID NO: 25 shows the amino acid sequence of the 1 N3R isoform of the Tau protein, also called Tau-B, with 381 amino acids.
SEQ ID NO: 26 montre la séquence d'acides aminés de l'isoforme 0N3R de la protéine Tau, également appelée Fetal-Tau, avec 352 acides aminés. SEQ ID NO: 27 montre la séquence d'acides aminés de la protéine Tau tronquée consistant en la région3R de la protéine Tau. SEQ ID NO: 26 shows the amino acid sequence of the 0N3R isoform of Tau protein, also called Fetal-Tau, with 352 amino acids. SEQ ID NO: 27 shows the amino acid sequence of the truncated Tau protein consisting of the 3R region of the Tau protein.
DESCRIPTION DES FIGURES Figure 1 : Graphique qui montre les résultats des tests ELISA et donc des courbes d'affinité du nanocorps 2C5 pour Tau-0 (Tau oligomères), Tau-F (tau fibres), Mimes (la région R3 de Tau polymérisé) et Tau-N (tau natives). DESCRIPTION OF THE FIGURES FIG. 1: Graph which shows the results of the ELISA tests and therefore of the affinity curves of the 2C5 nanobodies for Tau-0 (Tau oligomers), Tau-F (tau fibers), Mimes (the R3 region of Tau polymerized) and Tau-N (native tau).
Figure 2 : Graphique qui montre l'absence d'affinité pour le peptide amyloïde Abeta1 -42 polymérisé Figure 2: Graph showing the lack of affinity for the polymerized Abeta1 -42 amyloid peptide
Figure 3 : Immunohistochimie comparative sur des coupes de cortex temporal d'un cas de MA ; A) AT8 est un: anticorps monoclonal dirigé contre les PHF phosphorylé en Ser202 et Thr 205 ; B) 2C5 est le nanocorps de l'invention dirigé contre Tau sous forme d'oligomères C) T22 est un anticorps monoclonal dirigé contre les oligomères de tau, D) Ab4G8 est un anticorps monoclonal dirigé contre les formes agrégées de peptide beta amyloïde). Figure 3: Comparative immunohistochemistry on sections of temporal cortex of a case of AD; A) AT8 is a monoclonal antibody directed against phosphorylated PHFs in Ser2O2 and Thr205; B) 2C5 is the nanobodies of the invention directed against Tau as oligomers C) T22 is a monoclonal antibody directed against tau oligomers, D) Ab4G8 is a monoclonal antibody directed against aggregated forms of amyloid beta peptide).
Figure 4 : Alignement des séquences variables des nanocorps de l'invention. Figure 5 : Graphique qui montre les résultats des tests ELISA et donc des courbes d'affinité du nanocorps 2C5 (sans radiomarquage) pour des polymères de la séquence R3 de la protéine tau. Figure 4: Alignment of the variable sequences of the nanobodies of the invention. Figure 5: Graph which shows the results of the ELISA tests and thus affinity curves of the 2C5 nanobodies (without radiolabeling) for polymers of the R3 sequence of the tau protein.
Figure 6 : Graphique qui montre les résultats des tests ELISA et donc des courbes d'affinité du nanocorps 2C5 (radiomarqué à iode 125) pour des polymères de la séquence R3 de la protéine tau. Figure 6: Graph which shows the results of the ELISA tests and thus the affinity curves of the 2C5 nanobodies (radiolabeled with iodine 125) for polymers of the R3 sequence of the tau protein.
EXAMPLE EXAMPLE
1) Génération des nanobodies : 1) Generation of nanobodies:
(1 ) Préparation des antigènes nécessaires à l'immunisation des camélidés : (1) Preparation of antigens necessary for the immunization of camelids:
L'ADNc (htau40) codant pour l'isoforme la plus longue de Tau (441 aa - 45,8 kDa) a été cloné dans un plasmide pRK172, en aval du promoteur de l'ARN polymérase T7. Les plasmides recombinants ont été transformés dans des bactéries Escherichia coli BL21 (procurées auprès de Goedert, M et al., Neuron, 3(4): 519-526 (1989))(bactéries fournies par l'équipe du Pr Baulieu, INSERM, Paris). La purification de la protéine est réalisée par deux étapes de FPLC (Fast Protein Liquid Chromatography) : (1 ) par colonne à échange de cations chargée en S04- (HiTrap SP-XL, Thermoscientfic) puis (2) par chromatographie d'exclusion (Colonne Hi Trap Sepharose HP - Amersham Biosciences). Pour obtenir des préparations enrichies avec différentes formes polymérisées de tau (oligomère et fibres) mimant la fibrillation de Tau in vivo, nous avons utilisé la méthode de Haase et al. (Journal of Neurochemistry, 88(6): 1509-1520, 2004) modifiée par Flach et al. (Journal of Biological Chemistry, 2012, (287) 52: 43223-43233). La fibrillation de Tau (40 μΜ) est réalisée à 37°C dans du tampon MOPS (acide 3-(N-morpholino) propane sulfonique) 20 mM, pH 7 en présence d'héparine (10 μΜ) de NaN3 (4%) pour un volume final de 1 ,5 mL [21 ] [14]. La fibrillation est analysée comme décrit ci-dessus. La fibrillation est stoppée par congélation des échantillons à -80°C au bout de 48 heures et de 72h. La durée du processus de polymérisation est adaptée à la production (a) d'oligomères et (b) de fibres. En parallèle, un peptide recouvrant la région R3 des motifs répétés de Tau a été synthétisé et polymérisé de manière similaire. En particulier, la fibrillation de R3 (0.4 μΜ) est réalisée à 37°C dans du tampon PBS (phosphate buffer saline 50 mM, à pH 7 en présence d'héparine (0.4 μΜ), de NaN3 (4%) pour un volume final de 1 ,5 ml. La fibrillation est stoppée par congélation des échantillons à -80°C au bout de typiquement 72 heures pour obtenir Tau tronquée (R3) sous forme de fibres. The cDNA (htau40) encoding the longest Tau isoform (441 aa-45.8 kDa) was cloned into plasmid pRK172, downstream of the T7 RNA polymerase promoter. The recombinant plasmids were transformed into Escherichia coli BL21 bacteria (obtained from Goedert, M et al., Neuron, 3 (4): 519-526 (1989)) (bacteria provided). by the team of Prof. Baulieu, INSERM, Paris). Purification of the protein is carried out by two FPLC (Fast Protein Liquid Chromatography) steps: (1) by cation exchange column loaded with SO 4 - (HiTrap SP-XL, Thermoscientfic) followed by (2) by exclusion chromatography ( HP Sepharose Hi Trap column - Amersham Biosciences). To obtain enriched preparations with different polymerized forms of tau (oligomer and fiber) mimicking Tau fibrillation in vivo, we used the method of Haase et al. (Journal of Neurochemistry, 88 (6): 1509-1520, 2004) modified by Flach et al. (Journal of Biological Chemistry, 2012, (287) 52: 43223-43233). Fibrillation of Tau (40 μl) is carried out at 37 ° C. in MOPS buffer (20 mM 3- (N-morpholino) propane sulphonic acid), pH 7 in the presence of heparin (10 μl) of NaN 3 (4%). for a final volume of 1.5 mL [21] [14]. Fibrillation is analyzed as described above. Fibrillation is stopped by freezing the samples at -80 ° C after 48 hours and 72 hours. The duration of the polymerization process is suitable for the production of (a) oligomers and (b) fibers. In parallel, a peptide covering the R3 region of the Tau repeats was synthesized and polymerized in a similar manner. In particular, the fibrillation of R3 (0.4 μl) is carried out at 37 ° C. in PBS buffer (50 mM saline phosphate buffer, at pH 7 in the presence of heparin (0.4 μl), NaN 3 (4%) for a volume of The fibrillation is stopped by freezing the samples at -80 ° C after typically 72 hours to obtain truncated Tau (R3) in the form of fibers.
(2) Immunisation des lamas : (2) Immunization of llamas:
Un lama a été immunisé dans les mêmes conditions avec un mélange constitué d'oligomères et de fibres de Tau ainsi que de polymères de R3 ((a), (b) (c)). Trois injections successive sont réalisés en jours 0, 9, 18. Les prélèvements de sang sont réalisés en jour 28 et 42. Les poches de sang sont centrifugées en gradient de Ficoll afin de séparer et isoler les lymphocytes et les ARN totaux sont extraits.  A llama was immunized under the same conditions with a mixture of oligomers and Tau fibers as well as polymers of R3 ((a), (b) (c)). Three successive injections are performed in days 0, 9, 18. The blood samples are taken in days 28 and 42. The blood bags are centrifuged in a Ficoll gradient in order to separate and isolate the lymphocytes and the total RNAs are extracted.
(3) Production et criblage des nanobodies : Ils comportent plusieurs étapes :  (3) Production and screening of nanobodies: They comprise several stages:
- Amplification et production des mRNA des lymphocytes du lama  - Amplification and production of mRNA of lymphocytes of the llama
- Génération d'une librairie de phages exprimant les nanobodies du dromadaire précédemment immunisé.  - Generation of a library of phages expressing the nanobodies of the previously immunized dromedary.
- Sélection, par des tests ELISA et par FACS, de phages exprimant des nanobodies anti-tau-oligomères.  - Selection, by ELISA and FACS, of phage expressing anti-tau-oligomeric nanobodies.
- Séquençage des clones sélectionnés et identification de clones distincts  - Sequencing of selected clones and identification of distinct clones
- Détermination des séquences du Nbs, - construction et production du fragment d'ARNm codant pour un Nb comportant un tag Myc et histidine en Cterm - Determination of Nbs sequences, construction and production of the mRNA fragment encoding a Nb comprising a Myc tag and histidine in Cterm
- Insertion en bactéries et production en quantité du Nb.  - Insertion in bacteria and quantity production of Nb.
2. Caractérisation in vitro des nanobodies : 2. In vitro characterization of nanobodies:
Les nanobodies ont été caractérisés pour leur affinité et leur spécificité au travers de deux étapes. Nanobodies have been characterized for their affinity and specificity through two stages.
a) Tests ELISA I : avec les antigènes de départ soit préparation enrichie en tau oligomères, préparation enrichie en tau fibres, R3 polymérisé, ainsi que la protéine tau native et des fibres de peptide beta amyloïde (réalisé à partir de peptide du commerce). Les constantes d'affinités des nanobodies ont été déterminées pour chacun de ces immunogènes. b) Immunohistochimie : sur des coupes de tissus en paraffine provenant de l'anatomo- pathologie humaine (collaboration avec le Département de Santé Mentale et de Psychiatrie à Genève). Ces coupes recouvrent différents cas de tauopathies plus ou moins sévères, a) ELISA I tests: with the starting antigens either tau-oligomer enriched preparation, tau fiber-enriched preparation, polymerized R3, as well as the native tau protein and beta amyloid peptide fibers (made from commercial peptide). The affinity constants of the nanobodies have been determined for each of these immunogens. (b) Immunohistochemistry: on sections of paraffin tissue from human pathology (collaboration with the Department of Mental Health and Psychiatry in Geneva). These cuts cover different cases of more or less severe tauopathies,
c) vs)
Les coupes de tissu sont déparaffiné puis traitées en coupes flottantes après perméabilisation en tampon PBS-Triton 0.2 % et démasquage de l'antigène au moyen d'une solution de démasquage du commerce (Vector H-3300). L'anticorps primaire (Nb) est appliqué à différentes concentrations (≥ 20 nM) toute la nuit à 4°C. Après rinçage un anticorps anti-histdine de lapin est appliqué pendant 1 h à température ambiante puis un troisième anticorps anti lapin de chèvre pendant 1 h est enfin appliqué près rinçage. La révélation est réalisée au travers de deux étapes incubation en présence de complexe avidine biotine (kit ABC Vector) puis di-amino-benzidine(kit ABC Vector). Des coupes contrôles sont systématiquement réalisées en absence du premier anticorps (Nb). Les coupes sont montées sur lame et contre colorées au moyen de l'hématoxiline. d) Tests ELISA II : Le nanobody 2C5 a été radiomarqué à iode 125. Les test d'affinité pour des polymères de la séquence R3 de la protéine tau, sont réalisés en ELISA , dans un tampon Phosphate Buffer Saline (NaCI, 137mM ; KCI, 2.7mM ; Na2HP04, 10mM KH2P04, 1 .8mM) BSA 1 % ; pH = 7.5 . Les ELISAs sont réalisés avant (Figure 5) et après radiomarquage (Figure 6). On constate qu'après radiomarquage, le ligand conserve une bonne affinité pour sa cible (Figure 6). 3. Résultats : The tissue sections are deparaffinized and then treated in floating sections after permeabilization in 0.2% PBS-Triton buffer and unmasking of the antigen using a commercial unmixing solution (Vector H-3300). The primary antibody (Nb) is applied at different concentrations (≥ 20 nM) overnight at 4 ° C. After rinsing, a rabbit anti-histdine antibody is applied for 1 h at room temperature and then a third goat anti-rabbit antibody for 1 h is finally applied after rinsing. The revelation is carried out through two incubation steps in the presence of avidine biotin complex (Vector ABC kit) and then di-amino-benzidine (Vector ABC kit). Control sections are systematically performed in the absence of the first antibody (Nb). The sections are mounted on a slide and counterstained with hematoxylin. d) ELISA II Tests: The nanobody 2C5 was radiolabeled with iodine 125. The affinity tests for polymers of the R3 sequence of the tau protein are carried out in ELISA, in a phosphate buffered saline buffer (NaCl, 137 mM; KCl 2.7mM Na2HPO4, 10mM KH2PO4, 1.8mM BSA 1%; pH = 7.5. The ELISAs are performed before (FIG. 5) and after radiolabelling (FIG. 6). It is found that after radiolabelling, the ligand retains a good affinity for its target (FIG. 6). 3. Results:
Nous avons produit plusieurs anticorps de camélidé à domaine variable unique (Nbs), dirigés spécifiquement contre des formes pathologiques précoces de la protéine Tau (oligomères et fibres) de faible poids moléculaires (15 kDa) : 2Ç5 (VHH), S2T2M3 E6 (VHH). We have produced several single variable domain camelide antibodies (Nbs), specifically directed against early pathological forms of Tau protein (oligomers and fibers) of low molecular weight (15 kDa): 2C5 (VHH), S2T2M3 E6 (VHH) .
Le Nb 2C5 a été séquencé et caractérisé comme VHH et caractérisé in vitro. Il présente des affinités respectives (Kd) de 5nM, 6 nM et 50 nM pour les oligomères, les fibres et des formes tronquées polymérisées de Tau. Ce Nb ne se lie ni à la protéine tau native (Kd>1800 nM) ni aux fibres de peptide beta amyloïde (Kd>10 000 nM). (Figue 1 et 2). Ce Nb a été testé en immunohistochimie sur des coupes de cerveaux humains provenant de l'anatomopathologie (Alzheimer de différents grade et tauopathie). Il démontre un marquage spécifique des corps cellulaires de l'hippocampe, du cortex entorhinal et du cortex temporal chez des patients atteint de la maladie d'Alzheimer et dans un cas de démence avec tauopathie pure (Tableau 4, Figure 3).  Nb 2C5 was sequenced and characterized as VHH and characterized in vitro. It has respective affinities (Kd) of 5nM, 6nM and 50nM for oligomers, fibers and truncated polymerized forms of Tau. This Nb binds neither native tau protein (Kd> 1800 nM) nor beta amyloid peptide fibers (Kd> 10,000 nM). (Figs 1 and 2). This Nb has been tested in immunohistochemistry on sections of human brains from anatomopathology (Alzheimer's of different grades and tauopathy). It demonstrates specific staining of hippocampal, entorhinal and temporal cortex cell bodies in patients with Alzheimer's disease and in dementia with pure tauopathy (Table 4, Figure 3).
Le radiomarquage au technétium 99m de ce SdAbs a été réalisé avec succès. The technetium-99m radiolabelling of this SdAbs has been successfully performed.
Le Nb S2T2M3 E6 a été sélectionné pour son affinité aux polymères de tau de bas poids moléculaires.  Nb S2T2M3 E6 was selected for its affinity for low molecular weight tau polymers.
Tableau 4: Cas d'anatomo-pathologie humaine testés pour 2C5 Table 4: Cases of human pathology tested for 2C5
Type Abbréviation Stade de Type Abbreviation Stage de
Age Sexe CDR MMS pathologie type Break  Age Sex CDR MMS pathology type Break
Vieillissement  Aging
C ? ? 1 ? ? normal  VS ? ? 1? ? normal
AD AD ? ? 5 ? ? AD AD? ? 5? ?
AD AD 74 F 5 na 15/30AD AD 74 F 5 na 15/30
AD AD 81 F 5 na 17/30AD AD 81 F 5 na 17/30
Tangle only Tangle only
T 73 M 4 na na dementia  T 73 M 4 na na dementia
PSP/CBD PSP/CBD ? ? Non ? ?  PSP / CBD PSP / CBD? ? No ? ?

Claims

Revendications claims
1. Nanocorps dirigé contre la protéine Tau, ladite protéine Tau étant sous forme d'oligomères et ledit nanocorps étant dépourvu de chaîne légère. 1. Nanobodies directed against the Tau protein, said Tau protein being in the form of oligomers and said nanobodies being devoid of light chain.
2. Nanocorps selon la revendication 1 , ledit nanocorps entre en compétition pour la liaison à la protéine Tau sous forme d'oligomères avec un nanocorps comprenant les séquences d'acides aminés The nanobody according to claim 1, said nanobodies competing for binding to the Tau protein as oligomers with a nanobodies comprising the amino acid sequences.
(i) GRTFSXiX2X3 (SEQ ID NO: 1 ) comme CDR1 dans laquelle l'acide aminé ΧΊ est S ou R, X2 est D ou Y et X3 est T ou A, (i) GRTFSXiX 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid Χ Ί is S or R, X 2 is D or Y and X 3 is T or A,
(ii) ISX1SGGX2T (SEQ ID NO: 2) comme CDR2 dans laquelle l'acide aminé ΧΊ est P ou R et X2 est S ou V, et (ii) ISX1SGGX 2 T (SEQ ID NO: 2) as CDR2 in which the amino acid Χ Ί is P or R and X 2 is S or V, and
(iii) NRDPKYGNTRY (SEQ ID NO: 5) ou TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3.  (iii) NRDPKYGNTRY (SEQ ID NO: 5) or TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3.
3. Nanocorps selon la revendication 1 ou 2, ledit nanocorps entre en compétition pour la liaison à la protéine Tau sous forme d'oligomères avec un nanocorps choisi parmi un nanocorps comprenant : The nanobody according to claim 1 or 2, said nanobodies compete for binding to the Tau protein as oligomers with a nanobodies selected from nanobodies comprising:
a) les séquences d'acides aminés (i) GRTFSSDT (SEQ ID NO: 3) comme CDR1 , (ii) ISPSGGVT (SEQ ID NO: 4) comme CDR2 et (iii) NRDPKYGNTRY (SEQ ID NO: 5) comme CDR3 ; ou a) the amino acid sequences (i) GRTFSSDT (SEQ ID NO: 3) as CDR1, (ii) ISPSGGVT (SEQ ID NO: 4) as CDR2 and (iii) NRDPKYGNTRY (SEQ ID NO: 5) as CDR3; or
b) les séquences d'acides aminés (i) GRTFSRYA (SEQ ID NO: 6) comme CDR1 , (ii) ISRSGGST (SEQ ID NO: 7) comme CDR2 et (iii) TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3. b) the amino acid sequences (i) GRTFSRYA (SEQ ID NO: 6) as CDR1, (ii) ISRSGGST (SEQ ID NO: 7) as CDR2 and (iii) TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3.
4. Nanocorps selon l'une quelconque des revendications 1 à 3, ledit nanocorps comprenant les séquences d'acides aminés 4. Nanobodies according to any one of claims 1 to 3, said nanobodies comprising the amino acid sequences
(i) GRTFSX1X2X3 (SEQ ID NO: 1 ) comme CDR1 dans laquelle l'acide aminé ΧΊ est S ou R, X2 est D ou Y et X3 est T ou A, (i) GRTFSX1X 2 X 3 (SEQ ID NO: 1) as CDR1 in which the amino acid Χ Ί is S or R, X 2 is D or Y and X 3 is T or A,
(ii) ISX1SGGX2T (SEQ ID NO: 2) comme CDR2 dans laquelle l'acide aminé ΧΊ est P ou R et X2 est S ou V, et (ii) ISX1SGGX 2 T (SEQ ID NO: 2) as CDR2 in which the amino acid Χ Ί is P or R and X 2 is S or V, and
(iii) NRDPKYGNTRY (SEQ ID NO: 5) ou TARRRISGTPQWHY (SEQ ID NO: 8) comme CDR3.  (iii) NRDPKYGNTRY (SEQ ID NO: 5) or TARRRISGTPQWHY (SEQ ID NO: 8) as CDR3.
5. Nanocorps selon l'une quelconque des revendications 1 à 4, ledit nanocorps comprenant : a) les séquences d'acides aminés (i) GRTFSSDT (SEQ ID NO: 3) comme CDR1 , (ii) ISPSGGVT (SEQ ID NO: 4) comme CDR2 et (iii) NRDPKYGNTRY (SEQ ID NO: 5) comme CDR3 ; ou The nanobody according to any one of claims 1 to 4, said nanobody comprising: a) the amino acid sequences (i) GRTFSSDT (SEQ ID NO: 3) as CDR1, (ii) ISPSGGVT (SEQ ID NO: 4) as CDR2 and (iii) NRDPKYGNTRY (SEQ ID NO: 5) as CDR3; or
b) les séquences d'acides aminés (i) GRTFSRYA (SEQ ID NO: 6) comme CDR1 , (ii) ISRSGGST (SEQ ID NO: 7) comme CDR2 et (iii) TARRRISGTPQWHY (SEQ ID NO: b) the amino acid sequences (i) GRTFSRYA (SEQ ID NO: 6) as CDR1, (ii) ISRSGGST (SEQ ID NO: 7) as CDR2 and (iii) TARRRISGTPQWHY (SEQ ID NO:
8) comme CDR3 ; ou 8) as CDR3; or
un variant fonctionnellement conservatif du nanocorps défini en a) ou b) comprenant une substitution conservative d'un ou deux acides aminés dans respectivement une, deux ou trois des séquences SEQ ID NO: 3, SEQ ID NO: 4 et SEQ ID NO: 5, ou SEQ ID NO: 6, SEQ ID NO: 7 et SEQ ID NO: 8. a functionally conservative variant of the nanobodies defined in a) or b) comprising a conservative substitution of one or two amino acids in one, two or three of the sequences SEQ ID NO: 3, SEQ ID NO: 4 and SEQ ID NO: 5 respectively; , or SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8.
6. Nanocorps selon l'une quelconque des revendications 1 à 5, ledit nanocorps comprenant une séquence d'acides aminés sélectionnée dans le groupe consistant en les séquences d'acides aminés SEQ ID NO: 9 ou SEQ ID NO: 10, de préférence SEQ ID NO: 9. The nanobody according to any one of claims 1 to 5, said nanobodies comprising an amino acid sequence selected from the group consisting of amino acid sequences SEQ ID NO: 9 or SEQ ID NO: 10, preferably SEQ ID NO: 9.
7. Nanocorps selon l'une quelconque des revendications 1 à 6, ledit nanocorps étant liée à un marqueur détectable. The nanobody according to any one of claims 1 to 6, said nanobodies being bound to a detectable marker.
8. Nanocorps selon la revendication 7, ledit marqueur détectable étant un radioélément. 8. Nanobodies according to claim 7, said detectable marker being a radioelement.
9. Nanocorps selon l'une quelconque des revendications 1 à 8 pour son utilisation comme agent de contraste dans l'imagerie médicale in vivo, non invasive. 9. Nanobody according to any one of claims 1 to 8 for its use as a contrast agent in non-invasive, in vivo medical imaging.
10. Nanocorps selon l'une quelconque des revendications 1 à 9, pour son utilisation dans des méthodes de diagnostic ou de pronostic, de préférence d'une Tauopathies. The nanobody according to any one of claims 1 to 9 for use in diagnostic or prognostic methods, preferably Tauopathies.
11. Nanocorps selon l'une quelconque des revendications 1 à 6 pour son utilisation comme médicament. 11. Nanobody according to any one of claims 1 to 6 for its use as a medicament.
12. Utilisation d'un nanocorps tel que défini dans l'une quelconque des revendications 1 à 6 pour la détection in vitro de la protéine Tau sous forme d'oligomères dans un échantillon. 12. Use of a nanobody as defined in any one of claims 1 to 6 for the in vitro detection of Tau protein as oligomers in a sample.
13. Composition pharmaceutique comprenant un nanocorps tel que défini dans l'une quelconque des revendications 1 à 6 en association avec un véhicule pharmaceutiquement acceptable. 13. A pharmaceutical composition comprising a nanobody as defined in any one of claims 1 to 6 in association with a pharmaceutically acceptable carrier.
14. Acide nucléique comprenant une séquence nucléique codant le nanocorps tel que défini dans l'une quelconque des revendications 1 à 6. 14. Nucleic acid comprising a nucleic acid sequence encoding the nanobody as defined in any one of claims 1 to 6.
15. Vecteur comprenant un acide nucléique tel que défini dans la revendication 14. A vector comprising a nucleic acid as defined in claim 14.
16. Cellule transfectée, transduite ou transformée avec un acide nucléique selon la revendication 14 ou un vecteur selon la revendication 15. 16. A cell transfected, transduced or transformed with a nucleic acid according to claim 14 or a vector according to claim 15.
17. Méthode de production d'une cellule hôte recombinante exprimant un nanocorps tel que défini selon l'une quelconque des revendications 1 à 6, ladite méthode comprenant les étapes consistant à : A method of producing a recombinant host cell expressing a nanobody as defined in any one of claims 1 to 6, said method comprising the steps of:
(i) introduire in vitro ou ex vivo un acide nucléique selon la revendication 14 ou un vecteur selon la revendication 15 dans une cellule hôte compétente,  (i) introducing in vitro or ex vivo a nucleic acid according to claim 14 or a vector according to claim 15 into a competent host cell,
(ii) cultiver in vitro ou ex vivo la cellule hôte recombinante obtenue et  (ii) culturing in vitro or ex vivo the recombinant host cell obtained and
(iii) éventuellement sélectionner les cellules qui expriment et/ou sécrètent ledit nanocorps.  (iii) optionally selecting the cells that express and / or secrete said nanobody.
18. Méthode de production d'un nanocorps tel que défini selon l'une quelconque des revendications 1 à 6, ladite méthode comprenant les étapes consistant à : 18. A method of producing a nanobody as defined in any one of claims 1 to 6, said method comprising the steps of:
(i) cultiver une cellule transduite ou transfectée ou transformée selon la revendication 16, dans des conditions appropriées pour permettre l'expression dudit nanocorps, et  (i) culturing a transduced or transfected or transformed cell according to claim 16 under conditions suitable to allow the expression of said nanobody, and
(ii) récupérer le nanocorps exprimé.  (ii) recover the expressed nanobodies.
EP17798139.6A 2016-10-27 2017-10-27 Anti-tau nanobodies Pending EP3532495A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
FR1660450A FR3058143B1 (en) 2016-10-27 2016-10-27 ANTI-TAU NANOCBODIES
PCT/EP2017/077691 WO2018078140A1 (en) 2016-10-27 2017-10-27 Anti-tau nanobodies

Publications (1)

Publication Number Publication Date
EP3532495A1 true EP3532495A1 (en) 2019-09-04

Family

ID=58314353

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17798139.6A Pending EP3532495A1 (en) 2016-10-27 2017-10-27 Anti-tau nanobodies

Country Status (6)

Country Link
US (1) US10590191B2 (en)
EP (1) EP3532495A1 (en)
JP (1) JP7197494B2 (en)
CA (1) CA3041066A1 (en)
FR (1) FR3058143B1 (en)
WO (1) WO2018078140A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3583123A1 (en) 2017-02-17 2019-12-25 Denali Therapeutics Inc. Anti-tau antibodies and methods of use thereof
CN112649615A (en) * 2020-12-21 2021-04-13 大连理工大学 Method for detecting soluble Abeta oligomers with different sizes and application
FR3134390A1 (en) 2022-04-11 2023-10-13 Universite Grenoble Alpes PEPTIDE USEFUL FOR THE TRANSPORT OF MOLECULES THROUGH CELL BARRIERS

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4861719A (en) 1986-04-25 1989-08-29 Fred Hutchinson Cancer Research Center DNA constructs for retrovirus packaging cell lines
US5278056A (en) 1988-02-05 1994-01-11 The Trustees Of Columbia University In The City Of New York Retroviral packaging cell lines and process of using same
CA2156725A1 (en) 1993-02-22 1994-09-01 Warren S. Pear Production of high titer helper-free retroviruses by transient transfection
FR2712812B1 (en) 1993-11-23 1996-02-09 Centre Nat Rech Scient Composition for the production of therapeutic products in vivo.
EP0781334A1 (en) 1994-09-12 1997-07-02 Schering Aktiengesellschaft Process for manufacture of a modified collagen-induced platelet aggregation inhibitor pallidipin
IL116816A (en) 1995-01-20 2003-05-29 Rhone Poulenc Rorer Sa Cell for the production of a defective recombinant adenovirus or an adeno-associated virus and the various uses thereof
US6013516A (en) 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
JP6124591B2 (en) * 2009-08-28 2017-05-10 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム Antibodies that bind to tau oligomers
GB201112056D0 (en) * 2011-07-14 2011-08-31 Univ Leuven Kath Antibodies
SG10201912964PA (en) * 2011-09-19 2020-02-27 Axon Neuroscience Se Protein-based therapy and diagnosis of tau-mediated pathology in alzheimer's disease
EP2906596B1 (en) * 2012-10-12 2023-12-27 Arizona Board of Regents on behalf of Arizona State University Antibody based reagents that specifically recognize toxic oligomeric forms of tau
EP2899208A1 (en) * 2014-01-28 2015-07-29 F.Hoffmann-La Roche Ag Camelid single-domain antibody directed against phosphorylated tau proteins and methods for producing conjugates thereof

Also Published As

Publication number Publication date
JP7197494B2 (en) 2022-12-27
WO2018078140A1 (en) 2018-05-03
US10590191B2 (en) 2020-03-17
CN110225926A (en) 2019-09-10
JP2020505056A (en) 2020-02-20
FR3058143B1 (en) 2021-03-12
CA3041066A1 (en) 2018-05-03
FR3058143A1 (en) 2018-05-04
US20190263898A1 (en) 2019-08-29

Similar Documents

Publication Publication Date Title
EP3160999B1 (en) Antibodies and antigen-binding fragments that specifically bind to microtubule-associated protein tau
AU2013286680B2 (en) Antibodies to tau
JP5765857B2 (en) Antibody specifically binding to Aβ oligomer and use thereof
EP2731969B1 (en) Antibodies to phosphorylated tau aggregates
US11028157B2 (en) Binding molecules that specifically bind to tau
GB2507207A (en) Treatment of Tauopathies
HUE027649T2 (en) Antibodies recognising phospho-tau
BR112020018868A2 (en) antibody-based methods to detect and treat alzheimer&#39;s disease
US20200369754A1 (en) Binding molecules that specifically bind to tau
US20130184441A1 (en) Detection of Neurodegenerative Disease
JP2018510617A (en) Anti-transthyretin antibody
JP2019530428A (en) Anti-transthyretin antibody
EP3532495A1 (en) Anti-tau nanobodies
CA2846251C (en) Anti-vcam-1 nanobodies
JP2023521763A (en) Anti-PHF-tau antibody and uses thereof
BR112020014940A2 (en) ADRENOMEDULIN (ADM) FOR DIAGNOSIS AND / OR PREDICTION OF DEMENTIA AND ANTIADRENOMEDULIN BINDER FOR USE IN THERAPY AND PREVENTION OF DEMENTIA
Jiang et al. Single-domain antibody–based noninvasive in vivo imaging of α-synuclein or tau pathology
JP2018535921A (en) Antibody-based molecules selective for tau&#39;s {p} Ser 404 epitope and their use in the diagnosis and treatment of tauopathy
CN110225926B (en) Anti-Tau nanobodies

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20190506

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20220217

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: INSTITUT NATIONAL DE LA SANTE ET DE LA RECHERCHE MEDICALE

Owner name: CENTRE HOSPITALIER UNIVERSITAIRE GRENOBLE ALPES

Owner name: UNIVERSITE GRENOBLE ALPES