WO2017009327A1 - Antibodies that bind to sortilin and inhibit the binding of progranulin - Google Patents

Antibodies that bind to sortilin and inhibit the binding of progranulin Download PDF

Info

Publication number
WO2017009327A1
WO2017009327A1 PCT/EP2016/066516 EP2016066516W WO2017009327A1 WO 2017009327 A1 WO2017009327 A1 WO 2017009327A1 EP 2016066516 W EP2016066516 W EP 2016066516W WO 2017009327 A1 WO2017009327 A1 WO 2017009327A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
antigen
variable domain
chain variable
binding fragment
Prior art date
Application number
PCT/EP2016/066516
Other languages
English (en)
French (fr)
Inventor
Lars Christian BIILMANN RØNN
Ibrahim John MALIK
Jeffrey B STAVENHAGEN
Søren CHRISTENSEN
Jan Egebjerg
Arnout Gerritsen
Edward Van Den Brink
Paul Parren
Rob DE JONG
Original Assignee
H. Lundbeck A/S
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EA201890038A priority Critical patent/EA201890038A1/ru
Priority to TNP/2017/000534A priority patent/TN2017000534A1/en
Priority to CR20180002A priority patent/CR20180002A/es
Priority to CN201680040476.5A priority patent/CN107849135A/zh
Priority to EP16739087.1A priority patent/EP3322726A1/en
Priority to CA2989739A priority patent/CA2989739A1/en
Priority to JP2018501182A priority patent/JP6979397B2/ja
Priority to UAA201800574A priority patent/UA125136C2/uk
Priority to CN202210124752.3A priority patent/CN114478775A/zh
Priority to MX2018000506A priority patent/MX2018000506A/es
Priority to US15/743,549 priority patent/US10479835B2/en
Priority to BR112018000771-4A priority patent/BR112018000771A2/pt
Application filed by H. Lundbeck A/S filed Critical H. Lundbeck A/S
Priority to AU2016292980A priority patent/AU2016292980B2/en
Priority to KR1020187001192A priority patent/KR20180030045A/ko
Priority to RU2018100824A priority patent/RU2735639C2/ru
Priority to CN202210124751.9A priority patent/CN114478774A/zh
Publication of WO2017009327A1 publication Critical patent/WO2017009327A1/en
Priority to ZA2017/08613A priority patent/ZA201708613B/en
Priority to CONC2017/0012988A priority patent/CO2017012988A2/es
Priority to IL256503A priority patent/IL256503B/en
Priority to PH12018500100A priority patent/PH12018500100A1/en
Priority to HK18113432.9A priority patent/HK1254356A1/zh
Priority to US16/521,279 priority patent/US10889650B2/en
Priority to US17/097,457 priority patent/US11548950B2/en
Priority to US18/057,104 priority patent/US20230159643A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/286Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against neuromediator receptors, e.g. serotonin receptor, dopamine receptor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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 monoclonal anti-Sortilin antibodies useful in correcting a deficient level of progranulin (PGRN).
  • PGRN progranulin
  • these antibodies can be used in the treatment of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS).
  • FTD frontotemporal dementia
  • ALS amyotrophic lateral sclerosis
  • the monoclonal antibodies may also be useful to treat neurodegenerative disorders such as Alzheimer's Disease (AD).
  • Sortilin is a receptor that has been reported to mediate pro-apoptotic effects of pro-neurotrophins and to mediate trafficking and sorting of neurotrophin receptors (Nykjaer et al, 201 2, Trends Neurosci. 201 2;35(4):261 - 70; Glerup et al, Handb Exp Pharmacol, 2014;220:165-89, Carlo et al, J Mol Med (Berl). 2014 Sep;92(9):905-1 1 ).
  • sortilin ligands have been identified including neurotensin for which a high affinity binding site was localized by x-ray crystallography to inside a beta propeller tunnel in the sortilin molecule (Quistgaard et al, Nat Struct Mol Biol. 2009 Jan; 16(1 ):96-8; Quistgaard et al, Protein Sci. 2014, Sep;23(9):1291 -300). More recently, sortilin was shown to function as a high affinity receptor for the growth factor progranulin (PGRN, Hu et al. Neuron. 2010 Nov 18;68(4):654-67.
  • PGRN Hu et al. Neuron. 2010 Nov 18;68(4):654-67.
  • PGRN (proepithelin, granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin)) is a secreted glycosylated protein with antiinflammatory and neurotrophic-like actions (For a recent review, see Nguyen, Trends Endocrinol Metab. 2013 Dec;24(12):597-606). PGRN is proteolytically cleaved to granulins, but much remains to be learned regarding the physiological role of PGRN and granulins and the identity of their receptors. PGRN has been implicated in several cellular functions including cell cycle regulation and cell motility (He, Z. & Bateman, A., J. Mol. Med.
  • VEGF vascular endothelial growth factor
  • sortilin is a key regulator of extracellular PGRN.
  • PGRN has been linked to frontotemporal dementia (FTD), a progressive dementia characterized by behavioral and semantic changes, as well as frontotemporal lobar degeneration (FTLD) and neuronal inclusions containing TAR DNA Binding Protein-43 (TDP-43) or tau inclusions (Baker et al, 2006, Nature. 2006 Aug 24;442(7105):916-9; Cruts et al, Nature 442: 920-924 (2006); Am J Hum Genet. 2010 Dec 10;87(6):890-7,M et al, Trends in Genetics 24: 186-194 (2008)).
  • FDD frontotemporal dementia
  • TDP-43 frontotemporal lobar degeneration
  • TDP-43 pathology 50%) similar to ALS and FTD-TDP43 and ALS are by some considered to constitute a disease spectrum (Ito D Neurology. 201 1 Oct 25;77(17):1636-43; Boxer AL et al, Alzheimers Dement. 201 3 Mar;9(2):176-88; Rademakers et al, Nat Rev Neurol. 2012 August; 8(8): 423-434) due to common pathologies and genetic factors and some overlap in symptomatology. No disease-modifying treatment options are available for FTD.
  • a subset of frontotemporal dementia patients with TDP-43 pathology have loss of function mutations in the granulin gene (GRN) resulting in PGRN haplo-insufficiency.
  • GNN granulin gene
  • PGRN haplo-insufficiency 69 different mutations in the granulin gene, all resulting in reduced PGRN levels and/or function, have been associated with FTD and it is believed that raising extracellular PGRN in plasma and brain would counteract the disease process.
  • PGRN mutations have also been linked with Alzheimer's disease (AD) (Sheng et al., 2014, Gene. 2014 Jun 1 ;542(2):141 -5; Brouwers et al., 2008, Neurology.
  • AD Alzheimer's disease
  • the present application describes the generation and identification of anti-human Sortilin antibodies which can regulate PGRN in cellular models and in mice. Those antibodies surprisingly bind to a region on Sortilin which is distant to the previously reported progranulin binding site, the so-called neurotensin-site, and yet are capable of inhibiting Sortilin-PGRN interaction and of thereby increasing extracellular PGRN.
  • Sortilin binding regions As PGRN has neuroprotective and anti-inflammatory effects, the inventors' findings indicate that such antibodies targeting Sortilin are likely to have a beneficial effect in the treatment of a range of neurodegenerative disorders including FTD/FTLD. A subgroup of these patients carry a mutation in the gene encoding PGRN leading to haploinsufficiency. Sortilin antibodies are therefore likely to have similar therapeutic benefits for patients suffering from other TDP- 43 proteinopathies and diseases in which PGRN levels may influence TDP43- function and pathology, including ALS and AD.
  • the inventors of the present invention have generated monoclonal antibodies which are able to inhibit the binding of PGRN to Sortilin, and which surprisingly bind to a novel Sortilin region denominated the "D-region" as defined in SEQ ID NO:170.
  • Several antibodies identified by the inventors have properties similar to the D-region antibodies and experimental evidence indicates that they also bind within the D-region, and those antibodies are herein referred to as D+ antibodies. Accordingly, in one aspect, the invention relates to such antibodies, to compositions and/or kits comprising such antibodies, and to methods and uses thereof.
  • the invention also relates to a method of preventing or treating a disease associated with decreased PGRN levels in the brain of a patient, comprising administering an effective dosage of an antibody or an antigen- binding fragment thereof that binds to the D-region of Sortilin.
  • diseases include i.a. FTD, ALS and TDP43 proteinopathies such as AD.
  • Figure 1 provides an overview of the generation of region assignment of human antibodies based on Sortilin region binding, effect on PGRN-binding and PG RN-levels and on cross-blocking between the antibodies.
  • Sortilin-binding antibodies were selected and assigned to regions A- E based on binding to shuffle constructs in which the sequence corresponded to the tetraodon Sortilin sequence within selected regions of the protein (Example 1 , Figure 2).
  • Figure 2 shows the region assignment of antibodies based on binding to Sortilin shuffle constructs.
  • Panel A shows a linear illustration of the shuffle constructs used for region assignment of antibodies as described in Example 1. Sortilin shuffle constructs were generated based on the human Sortilin sequence (SEQ ID NO:169) (sections depicted in grey) in which amino acid residues were exchanged to the corresponding amino acid from the tetraodon Sortilin sequence (depicted in black) (SEQ ID NO:173) (Examples 1 -3).
  • Panel B shows predicted structure of the shuffle constructs illustrated linearly in A. Dark residues indicate residues changed to the corresponding tetraodon sequence in the shuffle constructs.
  • Panel C illustrates the binding pattern of antibodies assigned to the D-region and the E region classes respectively.
  • a "+” indicates binding to a given shuffle construct and a "-" indicates lack of binding.
  • antibodies were assigned to regions.
  • the resultant antibody region classes are indicated by A-E.
  • D and E region antibodies both bound the human sequences (all grey) as indicated by "+” and neither bound the tetraodon sequence (all black) as indicated by whereas the E region antibody bound the hB45678 shuffle construct while the D Region antibody did not bind resulting in the localisation of binding as illustrated in Panel A.
  • D Region antibodies binding to the following shuffle regions was observed: hsort, hB06-10, B12390. The antibodies did not bind to hB01 -05, B45678, tet.
  • D+ antibodies binding to the following shuffle region was observed: hsort, B1 2390. The antibodies did not bind to hB01 -05, hB06-10, B45678, tet.
  • the F binding pattern was similar to the D binding pattern except that no binding to the hB06-10 was observed for D+ antibodies.
  • the antibodies did not bind to the fully tetraodon Sortilin protein, except two. The two antibodies capable of binding the tetraodon sequence were denoted "tet". "Other" refers to an antibody which could not be assigned to one region.
  • Figure 4 shows the binding affinities of mouse anti-human antibodies to Sortilin shuffle constructs as obtained by bioLayer interferometry using Octet 384RED as described in Example 8 (EC50, ng/ml). No shading indicates binding and grey shading indicates no binding (NB). Region assignment based on binding patterns is illustrated in Figure 2.
  • FIG. 5 shows the effect of Sortilin antibody on Sortilin PGRN binding.
  • the D Region Sortilin human monoclonal (humAb) antibody 45 (filled circles) prevented PGRN binding to Sortilin, in contrast to a control Sortilin E region antibody (filled triangles) and an IgG control, lgG1 -b1 2 (open triangles) that did not interfere with the binding.
  • the binding of antibodies was determined by measuring the displacement of PGRN binding to Sortilin using Homogenous Time Resolved Fluorescent (HTRF) (Example 10).
  • HTRF Homogenous Time Resolved Fluorescent
  • Dose-response evaluation of antibodies was performed with ten concentrations covering 50pM to 1 ⁇ in a 3-fold dilution curve.
  • the half-maximal inhibitory concentration (IC50) values were calculated by non-linear regression using sigmoidal concentration response (variable slope) in XLfit 4 (IDBS, UK).
  • FIG. 6 Summary of effect of antibodies on Sortilin-PGRN binding determined by homogenous time resolved fluorescent (HTRF) analysis as shown in Figure 5.
  • HTRF time resolved fluorescent
  • FIG. 7 shows cross-blocking between antibodies. Human antibodies and the mouse antibodies were all tested in a single experiment where each antibody was bound to human wild type (WT) Sortilin ( Figure 7). Subsequently all other antibodies were tested for binding to the preformed sortilin:antibody complex (Example 9). The selected 15 D-region and 3 D+ human antibodies (based on their effect in the HTRF PGRN-sortilin assay, ( Figure 5 and Figure 6) and two mouse D Region antibodies all inhibited binding of each other to human WT Sortilin.
  • the antibodies did not cross-block with antibodies designated to other region classes (as illustrated for A-region, E-region and tetraodon recognizing antibodies numbered AbA1 -x, AbE1 -x and Abtet in the table respectively) except for one cross blocking A region antibody, one antibody with unknown region assignment ("other") and a partial block for a D+ antibody 548.
  • region classes as illustrated for A-region, E-region and tetraodon recognizing antibodies numbered AbA1 -x, AbE1 -x and Abtet in the table respectively.
  • Cross blocking between Sortilin antibodies from the same or different regions was determined by analyzing interference with antibody-Sortilin binding. Binding of antibodies to Sortilin-ECD-His was measured by BioLayer Interferometry using Octet 384RED (Example 9). The left column indicates primary (immobilized) antibodies and the top row indicates the secondary antibodies (antibodies being tested against the immobilized antibodies). Binding of both the primary and secondary antibodies to Sortilin-ECD-His would results in a response value higher than 0.1 and indicate that both antibodies were binding to different regions of the protein. Response value less than 0.1 shows lack of binding of the secondary antibody and an effective cross blocking by the immobilized (primary) antibody, which suggests that both antibodies bind to the same region of Sortilin.
  • Figure 8 shows the effect of D-region and D+ Sortilin antibodies on the binding of the selective small molecule ligand AF38469 to Sortilin.
  • the binding site for AF38469 has been shown to be similar to the binding site of neurotensin and characterized by X-ray crystallography (Schroder et al. Bioorg Med Chem Lett. 2014 Jan 1 ;24(1 ):177-80).
  • PGRN has been reported to bind to the same site (Lee ef al. Hum Mol Genet. 2013) antibodies 45 and 68, binding to D-region, and D+ respectively, did not inhibit the binding of AF38469 to sortilin.
  • Antibodies (100nM) to be tested were added to S18 cells 30 min before addition of recombinant PGRN for 4 hr. The cells were then fixed, stained for PGRN and analyzed by Cellomics. PGRN fluorescence was measured as mean fluorescence per cell. Data is presented as mean ⁇ SD. Data analyzed by one-way Anova followed by Dunnett's analysis, all groups were compared to PGRN. * p ⁇ 0.05; ** p ⁇ 0.01
  • FIG. 10 Extracellular PGRN levels estimated by ELISA in media from cultures of sortilin over-expressing HEK cells (S18). Sortilin D-region (45, 81 1 ) and D+ (68) antibodies increased PGRN levels and a similar effect of the sortilin ligand neurotensin was observed whereas the control antibody B12 had no effect. These observations indicate that D-region and D+ Sortilin antibodies were capable of inhibiting sortilin-mediated clearance of PGRN thereby increasing extracellular PGRN. All antibodies were tested at 100nM. Neurotensin was tested at 10uM. PGRN levels have been normalized to control. Data is presented as mean ⁇ SD. Data was analyzed by one-way Anova followed by Dunnett's analysis, all groups were compared to CTRL * p ⁇ 0.05; ** p ⁇ 0.01 . (Example 13).
  • Figure 12 (Upper and Lower Panels) shows the effect of Sortilin antibody on extracellular PGRN in neuronally differentiated iPSC cells (Example 14). The sortilin D-region antibody 45 and the D+ antibody 68 increased PGRN levels whereas the control antibodies B12 and anti-HEL had no effect.
  • Neuronally differentiated iPSC cells were plated into 96 wells plate. One week later, antibodies were added to the cells. Media from the cells were collected at 48 hrs or 96 hrs and analysed by human PGRN ELISA (Enzo Life sciences) and samples analysed as per the manufacturer's instructions. Sortilin human antibodies 45 and 68 increased PGRN levels in the media at both time points. Control isotype antibodies B12 and Anti-Hel (negative control) did not change extracellular PGRN. Data is presented as mean ⁇ SD.
  • FIG. 13 shows plasma PGRN levels in human Sortilin expressing knock in (Kl) mice treated with Sortilin human antibody (Example 15). Sortilin antibody 45 increased plasma PGRN levels whereas the control antibody had no effect.
  • C Dose response study Different doses (4 doses: 0.1 , 0.4, 2 and 10 mg/kg) of the Sortilin (45) and control antibodies (Anti-Hel) were injected and mice were sacrificed on day 2. Plasma PGRN was elevated in mice treated with 45 (10 and 2mg/kg). lower doses (0.4 and 0.1 mg/kg) did not have an effect on plasma PGRN. Data is presented as mean ⁇ SD. Data was analyzed by two- way Anova followed by Bonferroni's analysis * p ⁇ 0.05; ** p ⁇ 0.01 ; *** p ⁇ 0.001 (Example 15).
  • Figure 14 provides an illustration of sortilin regions, based on sortilin shuffle constructs, imposed onto the predicted sortilin structure.
  • the regions constitute the sections of the sortilin protein in which the change of selected amino acid residues from the human sequence to the tetraodon sequence inhibited binding for antibodies of that region class.
  • the arrow indicates the reported high affinity binding site of neurotensin and PGRN (Quistgaard Nat Struct Mol Biol. 2009 Jan;16(1 ):96-8, Lee et al, Hum Mol Genet. 2013).
  • Figure 15a Panels 1 -6) and 15b (Panels 1-3) Shows representative peptides covering the conformational epitope of antibody 45, 68 and 81 1 .
  • Peptide 1 15-125 is an example of a peptide which is unaffected by the presence of antibody 45, 68 or 81 1 , and thereby is not a part of the conformational binding epitope (Example 16)
  • Figure 16a (Panels 1-6) and 16b (Panels 1-3) shows representative peptides covering the conformational epitope of antibody 30. All of the shown peptides show a protection from exchange larger than 0.5D, except peptide 563-572, peptide 646-656 and peptide 704-714. These three peptides are examples of peptides which are unaffected by the presence of antibody 30, and thereby is not a part of the conformational binding (Example 16)
  • Figure 17 shows an illustration of the microdialysis for procedure.
  • Figure 18a shows a time course: effect of systemic administration of antibody 45 or PBS (50 mg/kg, 10 ml/kg, s.c.) 24h prior to the microdialysis experiments on the levels of PRGN in the hippocampus of freely moving hSORTI mice over time (24h).
  • PBS 50 mg/kg, 10 ml/kg, s.c.
  • Figure 18b shows the results of a pooled 24h dialysis: basal PRGN in the hippocampus of freely moving hSORTI mice in mab#45- and PBS- treated mice, 3.3 ⁇ 0.3 ng/ml and 1 .1 ⁇ 0.1 ng/ml, respectively, as assessed by push-pull microdialysis (Example 17).
  • Sortilin is synonymous with the Sortilin protein (identified in for example UniProt as Q99523, 1 and 2).
  • the amino acid numbering of Sortilin is given with respect to SEQ ID NO:169 as shown below, Met being amino acid 1 :
  • D Region is intended to refer to the region on Sortilin (corresponding to residues 523-610 of SEQ ID NO:169) consisting of the amino acids in SEQ ID NO:170 as shown below:
  • D Region antibodies binding to the following shuffle regions was observed: hsort, hB06-10, B12390. The antibodies did not bind to hB01 -05, B45678, tet.
  • D+ antibodies binding to the following shuffle regions was observed: hsort, B12390. The antibodies did not bind to hB01 -05, hB06-10, B45678, tet.
  • hsort, B12390 The antibodies did not bind to hB01 -05, hB06-10, B45678, tet.
  • D+ a similar binding pattern was observed as for "D" region antibodies except that no binding to the hB06-1 0 was observed for D+ antibodies.
  • D+ antibodies shared functional characteristics (cell assays etc.) to D-region antibodies.
  • Certain D and D + binding antibodies binds particular the region as defined in SEQ ID NO:185, 186 or 187.
  • the invention relates in certain embodiments to antibodies or antigen binding fragments thereof that binds to the D region sequences of SEQ ID 170, and within that region SEQ ID NO:185, 186 or 187.
  • these antibodies or antigen-binding fragments influence the PGRN levels and thus may be used to treat disease associated with PGRN, such as FTD, ALS and AD.
  • the invention relates to antibodies or antigen binding fragments thereof that binds the D region as defined above and in SEQ ID Nos 170, 185, 186 or 187, and further have affinity for the A region identified in SEQ ID NO 180. Within the A region certain antibodies or antigen binding fragments thereof have affinity for the A region amino acids identified in SEQ ID Nos 181 , 182, 183 or 184.
  • PGRN proepithelin, granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin
  • PGRN proteolytically cleaved from the precursor PGRN (He, Z. & Bateman, A., J. Mol. Med. 81 :600-6X2 (2003)).
  • PGRN has been associated with a variety of events, such as cell cycle regulation and cell motility (He, Z. & Bateman, A., J. Mol. Med.
  • VEGF vascular endothelial growth factor
  • PGRN mutations result in haploinsufficiency (Baker, M., et ah, Nature 442:916-919 (2006); Cruts, M., et ah, Nature 442:920-924 (2006)) and are known to be present in nearly 50% of familial FTD cases, making PGRN mutation a major genetic contributor to FTD (Cruts, M. & Van Broeckhoven, C, Trends Genet. 24:186-194 (2008); Le Ber, I., et ah, Brain 129:3051 - 3065 (2006)).
  • antibody in the context of the present invention refers to an immunoglobulin molecule or according to some embodiments of the invention, a fragment of an immunoglobulin molecule which has the ability to bind to an epitope of a molecule ("antigen").
  • Naturally occurring antibodies typically comprise a tetramer which is usually composed of at least two heavy (H) chains and at least two light (L) chains.
  • Each heavy chain is comprised of a heavy chain variable domain (abbreviated herein as VH) and a heavy chain constant domain, usually comprised of three domains (CH 1 , CH2 and CH3).
  • Heavy chains can be of any isotype, including IgG (lgG 1 , lgG2, lgG3 and lgG4 subtypes), IgA (lgA1 and lgA2 subtypes), IgM and IgE.
  • Each light chain is comprised of a light chain variable domain (abbreviated herein as VL) and a light chain constant domain (CL).
  • Light chains include kappa chains and lambda chains.
  • the heavy and light chain variable domain is typically responsible for antigen recognition, while the heavy and light chain constant domain may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1 q) of the classical complement system.
  • the VH and VL domains can be further subdivided into regions of hypervariability, termed “complementarity determining regions,” that are interspersed with domains of more conserved sequence, termed “framework regions" (FR).
  • Each VH and VL is composed of three CDR Domains and four FR Domains arranged from amino-terminus to carboxy-terminus in the following order: FR1 -CDR1 -FR2-CDR2-FR3-CDR3- FR4.
  • the variable domains of the heavy and light chains contain a binding domain that interacts with an antigen.
  • antibodies and their antigen-binding fragments that have been "isolated” so as to exist in a physical milieu distinct from that in which it may occur in nature or that have been modified so as to differ from a naturally occurring antibody in amino acid sequence.
  • epitope means an antigenic determinant capable of specific binding to an antibody.
  • Epitopes usually consist of surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics. Conformational and linear epitopes are distinguished in that the binding to the former, but not the latter, is always lost in the presence of denaturing solvents.
  • the epitope may comprise amino acid residues directly involved in the binding and other amino acid residues, which are not directly involved in the binding, such as amino acid residues which are effectively blocked by the specifically antigen-binding peptide (in other words, the amino acid residue is within the footprint of the specifically antigen-binding peptide).
  • the term "antigen-binding fragment of an antibody” means a fragment, portion, region or domain of an antibody (regardless of how it is produced (e.g., via cleavage, recombinantly, synthetically, etc.)) that is capable of binding to an epitope, and thus the term “antigen-binding” is intended to mean the same as “epitope-binding” so that, for example, an "antigen-binding fragment of an antibody” is intended to be the same as an “epitope-binding fragment of an antibody”.
  • An antigen-binding fragment may contain 1 , 2, 3, 4, 5 or all 6 of the CDR Domains of such antibody and, although capable of binding to such epitope, may exhibit a specificity, affinity or selectivity toward such epitope that differs from that of such antibody. Preferably, however, an antigen- binding fragment will contain all 6 of the CDR Domains of such antibody.
  • An antigen-binding fragment of an antibody may be part of, or comprise, a single polypeptide chain (e.g., an scFv), or may be part of, or comprise, two or more polypeptide chains, each having an amino-terminus and a carboxyl terminus (e.g., a diabody, a Fab fragment, a Fab 2 fragment, etc.).
  • Fragments of antibodies that exhibit antigen-binding ability can be obtained, for example, by protease cleavage of intact antibodies. More preferably, although the two domains of the Fv fragment, VL and VH, are naturally encoded by separate genes, or polynucleotides that encode such gene sequences (e.g., their encoding cDNA) can be joined, using recombinant methods, by a flexible linker that enables them to be made as a single protein chain in which the VL and VH regions associate to form monovalent antigen-binding molecules (known as single-chain Fv (scFv); see e.g., Bird ef al. , (1988) Science 242:423-426; and
  • scFv single-chain Fv
  • a flexible linker that is too short (e.g., less than about 9 residues) to enable the VL and VH domains of a single polypeptide chain to associate together, one can form a bispecific antibody, diabody, or similar molecule (in which two such polypeptide chains associate together to form a bivalent antigen-binding molecule) (see for instance PNAS USA 90(14), 6444-8 (1993) for a description of diabodies).
  • antigen-binding fragments encompassed within the present invention include (i) a Fab' or Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains, or a monovalent antibody as described in WO2007059782; (ii) F(ab')2 fragments, bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge domain; (iii) an Fd fragment consisting essentially of the VH and CH1 domains; (iv) a Fv fragment consisting essentially of a VL and VH domains, (v) a dAb fragment (Ward et al., Nature 341 , 544-546 (1989)), which consists essentially of a VH domain and also called domain antibodies (Holt et al; Trends Biotechnol.
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they may be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH domains pair to form monovalent molecules (known as single chain antibodies or single chain Fv (scFv), see for instance Bird et al., Science 242, 423-426 (1988) and Huston et al., PNAS USA 85, 5879-5883 (1988)).
  • scFv single chain antibodies or single chain Fv
  • antibody also includes antibody-like polypeptides, such as chimeric antibodies and humanized antibodies, and antibody fragments retaining the ability to bind to the antigen (antigen-binding fragments) provided by any known technique, such as enzymatic cleavage, peptide synthesis, and recombinant techniques.
  • An antibody as generated can possess any isotype.
  • isotype refers to the immunoglobulin class (for instance lgG1 , lgG2, lgG3 or lgG4) that is encoded by heavy chain constant domain genes.
  • Such antibody fragments are obtained using conventional techniques known to those of skill in the art; suitable fragments capable of binding to a desired epitope may be readily screened for utility in the same manner as an intact antibody.
  • bispecific antibody refers to an antibody containing two independent antigen-binding fragments that each target independent targets. These targets can be epitopes present on different proteins or different epitopes present on the same target. Bispecific antibody molecules can be made using compensatory amino acid changes in the constant domains of the HCs of the parent monospecific bivalent antibody molecules. The resulting heterodimeric antibody contains one Fabs contributed from two different parent monospecific antibodies. Amino acid changes in the Fc domain leads to increased stability of the heterodimeric antibody with bispecificity that is stable over time.
  • Bispecific antibodies can also include molecules that are generated using ScFv fusions.
  • bispecific molecules Two monospecific scfv are then independently joined to Fc domains able to form stable heterodimers to generate a single bispecific molecule (Mabry et al., PEDS 23:3 1 15-127 (2010). Bispecific molecules have dual binding capabilities.
  • an "anti-Sortilin antibody” or “Sortilin antibody” (used interchangeably herein, depending on the context wherein its written) is an antibody an antigen- binding fragment thereof which binds specifically to Sortilin, and especially to the Sortilin D Region, SEQ ID NO:170.
  • An anti-Sortilin antibody that binds to the Sortilin D Region will usually bind to a conformational epitope or a linear epitope of 3, 4, 5, 6 or 7 consecutive amino acids within the D-Region (for example SEQ ID NOs:185, 186 or 187) with an affinity (IC50) at or below 22 nM, such as between 22 nM and 1 nM, between 10 nM and 1 nM or between 5 nM and 1 nM.
  • the anti-Sortilin antibodies may also bind to the A region (SEQ ID NOs:180, 181 , 182, 183 or 184) although it's emphasized that their main biological function is believed to be achieved by binding to the D Region.
  • the binding site identified is rather unique as shown with for example the binding of the selective small molecule ligand AF38469 to Sortilin.
  • the binding site for AF38469 has been shown to be similar to the binding site of neurotensin and characterized by X-ray crystallography (Schroder et al. Bioorg Med Chem Lett. 2014 Jan 1 ;24(1 ):177-80).
  • PGRN has been reported to bind to the same site (Lee ef al. Hum Mol Genet. 2013).
  • the invention relates to an antibody, or an antigen-binding fragment thereof, capable of specifically binding to Sortilin and inhibiting the binding of PGRN to Sortilin, but which binding does not inhibit or substantially inhibit the binding of neurotensin or AF38469 to Sortilin.
  • This can be shown using for example displacement of binding to Sortilin using a scintillation proximity assay (SPA) (Example 1 1 ).
  • SPA scintillation proximity assay
  • human antibody (which may be abbreviated to "humAb” or “HuMab”), as used herein, is intended to include antibodies having variable and constant domains derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or during gene rearrangement or by somatic mutation in vivo).
  • the terms "monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition.
  • a conventional monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
  • a monoclonal antibody can be composed of more than one Fab domain thereby increasing the specificity to more than one target.
  • the terms “monoclonal antibody” or “monoclonal antibody composition” are not intended to be limited by any particular method of production (e.g., recombinant, transgenic, hybridoma, etc.).
  • the antibodies of the present invention, and their sortilin antigen- binding fragments will preferably be human or, for example for the mouse antibodies (denoted 1 F2, 5E1 ), "humanized,” particularly if employed for therapeutic purposes.
  • the term “humanized” refer to a molecule, generally prepared using recombinant techniques, having an antigen-binding site derived from an immunoglobulin from a non-human species and a remaining immunoglobulin structure based upon the structure and /or sequence of a human immunoglobulin.
  • the antigen-binding site may comprise either complete non-human antibody variable domains fused to human constant domains, or only the complementarity determining regions (CDRs) of such variable domains grafted to appropriate human framework regions of human variable domains.
  • the framework residues of such humanized molecules may be wild type (e.g., fully human) or they may be modified to contain one or more amino acid substitutions not found in the human antibody whose sequence has served as the basis for humanization. Humanization lessens or eliminates the likelihood that a constant domain of the molecule will act as an immunogen in human individuals, but the possibility of an immune response to the foreign variable domain remains (LoBuglio, A.F. et al. (1989) "Mouse/Human Chimeric Monoclonal Antibody In Man: Kinetics And Immune Response," Proc. Natl. Acad. Sci. (U.S.A.) 86:4220-4224).
  • variable domains of both heavy and light chains contain three complementarity- determining regions (CDRs) which vary in response to the antigens in question and determine binding capability, flanked by four framework regions (FRs) which are relatively conserved in a given species and which putatively provide a scaffolding for the CDRs.
  • CDRs complementarity- determining regions
  • FRs framework regions
  • humanized antibodies preserve all CDR sequences (for example, a humanized mouse antibody which contains all six CDRs from the mouse antibodies).
  • humanized antibodies have one or more CDRs (one, two, three, four, five, six) which are altered with respect to the original antibody, which are also termed one or more CDRs "derived from" one or more CDRs from the original antibody.
  • the ability to humanize an antigen is well known (see, e.g., US Patents No. 5,225,539; 5,530,101 ; 5,585,089; 5,859,205; 6,407,213; 6,881 ,557).
  • antibody “XX” is intended to denote an antibody or antigen-binding fragment thereof (for example antibody “5E1 "), comprising or consisting of the Light Chain, the Light Chain Variable domain, or the Light Chain Variable domain CDR1 -3, as defined by its respective SEQ ID NO, and the Heavy Chain, Heavy Chain Variable Domain, or Heavy Chain Variable Domain CDR1 -3 as defined by its respective SEQ ID NO.
  • the antibody or antigen-binding fragment thereof are defined by their entire Heavy Chain Variable Domain comprising as defined by their SEQ ID NO and their Light Chain Variable Domain as defined by their SEQ ID NO.
  • IMGT® the international ImMunoGeneTics information system® or, Kabat, E. A., Wu, T. T., Perry, H. M., Gottesmann, K. S. & Foeller, C. (1 991 ). Sequences of Proteins of Immunological Interest, 5th edit., NIH Publication no. 91 -3242 U.S. Department of Health and Human Services; Chothia, C. & Lesk, A. M. (1 987). Canonical structures For The Hypervariable domains Of Immunoglobu- lins. J. Mol. Biol. 196, 901 -917.
  • an antibody or an antigen-binding fragment thereof is said to "specifically" bind a region of another molecule (i.e., an epitope) if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity or avidity with that epitope relative to alternative epitopes.
  • the antibody, or antigen-binding fragment thereof, of the invention binds at least 10-fold more strongly to its target (Sortilin) than to another molecule; preferably at least 50-fold more strongly and more preferably at least 1 00-fold more strongly.
  • the antibody, or antigen-binding fragment thereof binds under physiological conditions, for example, in vivo.
  • binding in the context of the binding of an antibody to a predetermined antigen typically refers to binding with an affinity corresponding to a KD of about 10 ⁇ 7 M or less, such as about 10 ⁇ 8 M or less, such as about 10 ⁇ 9 M or less when determined by for instance surface plasmon resonance (SPR) technology in either a BIAcore® 3000 or T200instrument using the antigen as the ligand and the antibody as the analyte, and binds to the predetermined antigen with an affinity corresponding to a KD that is at least ten-fold lower, such as at least 100 fold lower, for instance at least 1 ,000 fold lower, such as at least 10,000 fold lower, for instance at least 100,000 fold lower than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely- related antigen.
  • a non-specific antigen e.g., BSA, casein
  • the amount with which the affinity is lower is dependent on the KD of the antibody, so that when the KD of the antibody is very low (that is, the antibody is highly specific), then the amount with which the affinity for the antigen is lower than the affinity for a non-specific antigen may be at least 10,000 fold.
  • the invention pertains to anti-Sortilin antibodies that exhibit a binding affinity corresponding to at or below 22 nM, such as between 22 nM and 1 nM, between 10 nM and 1 nM or between 5 nM and 1 nM, when determined by, for instance, bioLayer interferometry using an Octet 384RED (Example 8).
  • the invention relates to an antibody or antigen-binding fragment thereof able to compete with humAb antibody 45 or humAb antibody 68 for binding to Sortilin.
  • the invention relates to an antibody or antigen-binding fragment thereof that is able to compete with antibody 45 for binding to the D Region of Sortilin as defined in SEQ ID NO:170.
  • Such competitive binding inhibition can be determined using assays and methods well known in the art, for example using BIAcore® chips with immobilised human Sortilin and incubating with a reference antibody (such as antibody "45” or "68") with and without an antibody polypeptide to be tested.
  • a pair-wise mapping approach can be used, in which a reference antibody (such as antibody “45” or “68") is immobilised to the surface of the BIAcore® chip, human Sortilin antigen is bound to the immobilised antibody, and then a second antibody is tested for simultaneous binding ability to human Sortilin (see 'BIAcore® Assay Handbook', GE Healthcare Life Sciences, 29-0194-00 AA 05/2012; the disclosures of which are incorporated herein by reference).
  • the term "kd” (sec -1 or 1 /s), as used herein, refers to the dissociation rate constant of a particular antibody-antigen interaction. Said value is also referred to as the koff value.
  • ka (M-1 x sec-1 or 1 /Msec), as used herein, refers to the association rate constant of a particular antibody-antigen interaction.
  • KD (M) refers to the dissociation equilibrium constant of a particular antibody-antigen interaction and is obtained by dividing the kd by the ka.
  • KA (M-1 or 1 /M), as used herein, refers to the association equilibrium constant of a particular antibody-antigen interaction and is obtained by dividing the ka by the kd.
  • the invention relates to an antibody, or antigen- binding fragment thereof, which exhibits one or more of the following properties:
  • a binding affinity (K D ) for Sortilin of between 0.5-10 nM, such as 1 -5 nM or 1 -2 nM;
  • the term "capability to reduce and/or inhibit PGRN binding to Sortilin” includes an antibody that has the ability to inhibit binding to PGRN at an IC50 less than 50nM but preferably between 10nM and 0.2 nM using a time resolved fluorescence assay (HTFR) disclosed in Example 10.
  • HTFR time resolved fluorescence assay
  • Sortilin-expressing cells includes the ability to increase the concentration of
  • PGRN in the medium by at least 25%, such as between 25% and 500%, between 25% and 400% or between 25% and 200% as measured by an ELISA assay as disclosed in Example 13.
  • the "capability to reduce and/or inhibit the endocytosis of PGRN by Sortilin-expressing cells” includes the ability reduce the intracellular concentration of PGRN by at least 1 0% but preferably between 20 and 100 % as measured by a cellomics based assay as disclosed in Example 12.
  • the "capability to increase the amount and/or concentration of PGRN in the plasma in human-Sortilin-expressing knock-in mice” includes the ability to increase the concentration of PGRN in the plasma by at least 25% but preferably between 50 and 500 percent as measured by an ELISA assay as disclosed in Example 15.
  • the capability to increase PGRN in the brain may also be assayed by for example microdialysis.
  • capability to increase the amount and/or concentration of PGRN in the brain includes the ability to increase the concentration of PGRN in the brain by at least 25% but preferably between 50 and 500 percent as measured by microdialysis.
  • CDR residues not contacting the relevant epitope and not in the SDRs can be identified based on previous studies (for example residues H60-H65 in CDR H2 are often not required), from regions of Kabat CDRs lying outside Chothia hypervariable loops (see, Kabat ei al. (1 992) SEQUENCES OF PROTEINS OF IMMUNOLOG ICAL INTEREST, National Institutes of Health Publication No. 91 -3242; Chothia, C. et al.
  • substitutions of acceptor for donor amino acids in the CDRs to include reflects a balance of competing considerations. Such substitutions are potentially advantageous in decreasing the number of mouse amino acids in a humanized antibody and consequently decreasing potential immunogenicity. However, substitutions can also cause changes of affinity, and significant reductions in affinity are preferably avoided. Positions for substitution within CDRs and amino acids to substitute can also be selected empirically. [0082] The fact that a single amino acid alteration of a CDR residue can result in loss of functional binding (Rudikoff, S. etc. (1982) "Single Amino Acid Substitution Altering Antigen-binding Specificity," Proc. Natl. Acad. Sci.
  • a polynucleotide encoding the CDR is mutagenized (for example via random mutagenesis or by a site-directed method (e.g., polymerase chain- mediated amplification with primers that encode the mutated locus)) to produce a CDR having a substituted amino acid residue.
  • a site-directed method e.g., polymerase chain- mediated amplification with primers that encode the mutated locus
  • the BLOSUM system provides a matrix of amino acid substitutions created by analyzing a database of sequences for trusted alignments (Eddy, S.R. (2004) "Where Did The BLOSUM62 Alignment Score Matrix Come From?,” Nature Biotech. 22(8) : 1035- 1036; Henikoff, J.G. (1992) "Amino acid substitution matrices from protein blocks,” Proc. Natl. Acad. Sci. (USA) 89:10915-1091 9; Karlin, S. ef al. (1 990) "Methods For Assessing The Statistical Significance Of Molecular Sequence Features By Using General Scoring Schemes," Proc. Natl. Acad. Sci.
  • BLOSUM62.iij (BLOSUM62.iij). Table 1 presents the BLOSUM62.iij substitution scores (the higher the score the more conservative the substitution and thus the more likely the substitution will not affect function). If an antigen-binding fragment comprising the resultant CDR fails to bind to Sortilin, for example, then the BLOSUM62.iij substitution score is deemed to be insufficiently conservative, and a new candidate substitution is selected and produced having a higher substitution score. Thus, for example, if the original residue was glutamate (E), and the non-functional substitute residue was histidine (H), then the BLOSUM62.iij substitution score will be 0, and more conservative changes (such as to aspartate, asparagine, glutamine, or lysine) are preferred.
  • E glutamate
  • H histidine
  • the invention thus contemplates the use of random mutagenesis to identify improved CDRs.
  • conservative substitutions may be defined by substitutions within the classes of amino acids reflected in one or more of the following three tables:
  • More conservative substitutions groupings include: valine-leucine- isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, and asparagine-glutamine.
  • Phage display technology can alternatively be used to increase (or decrease) CDR affinity.
  • This technology referred to as affinity maturation, employs mutagenesis or "CDR walking" and re-selection uses the target antigen or an antigenic antigen-binding fragment thereof to identify antibodies having CDRs that bind with higher (or lower) affinity to the antigen when compared with the initial or parental antibody (See, e.g. Glaser et al. (1992) J. Immunology 149:3903).
  • Libraries can be constructed consisting of a pool of variant clones each of which differs by a single amino acid alteration in a single CDR and which contain variants representing each possible amino acid substitution for each CDR residue.
  • Mutants with increased (or decreased) binding affinity for the antigen can be screened by contacting the immobilized mutants with labeled antigen. Any screening method known in the art can be used to identify mutant antibodies with increased or decreased affinity to the antigen (e.g., ELISA) (See Wu ef al. 1998, Proc. Natl. Acad. Sci.
  • Rat Antibody For Anti-RAGE Therapy Comprehensive Mutagenesis Reveals A High Level Of Mutational Plasticity Both Inside And Outside The Complementarity-Determining Regions," J. Mol. Biol. 388(3):541 -558; Bostrom, J. et al. (2009) “Improving Antibody Binding Affinity And Specificity For Therapeutic Development,” Methods Mol. Biol. 525:353-376; Steidl, S. ef al. (2008) “In Vitro Affinity Maturation Of Human GM-CSF Antibodies By Targeted CDR-Diversification," Mol. Immunol. 46(1 ):135-144; and Barderas, R. et al. (2008) “Affinity Maturation Of Antibodies Assisted By In Silico Modeling," Proc. Natl. Acad. Sci. (USA) 105(26) :9029-9034.
  • sequence of CDR variants of encompassed antibodies or their antigen-binding fragments may differ from the sequence of the CDR of the parent antibody through substitutions; for instance substituted 4 amino acid residue, 3 amino acid residue, 2 amino acid residue or 1 of the amino acid residues.
  • amino acids in the CDR regions may be substituted with conservative substitutions, as defined in the below 3 tables.
  • transgenic non-human animal refers to a non-human animal having a genome comprising one or more human heavy and/or light chain transgenes or trans-chromosomes (either integrated or non-integrated into the animal's natural genomic DNA) and which is capable of expressing fully human antibodies.
  • a transgenic mouse can have a human light chain transgene and either a human heavy chain transgene or human heavy chain trans-chromosome, such that the mouse produces human anti-Sortilin antibody when immunized with Sortilin antigen and/or cells expressing Sortilin.
  • the human heavy chain transgene may be integrated into the chromosomal DNA of the mouse, as is the case for transgenic mice, for instance HuMAb mice, such as HCo7 or HCo12 mice, or the human heavy chain transgene may be maintained extra-chromosomally, as is the case for trans-chromosomal KM mice as described in WO02/43478.
  • transgenic and trans-chromosomal mice are capable of producing multiple isotypes of human monoclonal antibodies to a given antigen (such as IgG, IgA, IgM, IgD and/or IgE) by undergoing V-D-J recombination and isotype switching.
  • Transgenic, nonhuman animal can also be used for production of antibodies against a specific antigen by introducing genes encoding such specific antibody, for example by operatively linking the genes to a gene which is expressed in the milk of the animal.
  • treatment means ameliorating, slowing, attenuating or reversing the progress or severity of a disease or disorder, or ameliorating, slowing, attenuating or reversing one or more symptoms or side effects of such disease or disorder.
  • treatment or “treating” further means an approach for obtaining beneficial or desired clinical results, where "beneficial or desired clinical results” include, without limitation, alleviation of a symptom, diminishment of the extent of a disorder or disease, stabilized (i.e., not worsening) disease or disorder state, delay or slowing of the progression a disease or disorder state, amelioration or palliation of a disease or disorder state, and remission of a disease or disorder, whether partial or total detectable or undetectable.
  • an “effective amount,” when applied to an antibody or antigen-binding fragment thereof of the invention, refers to an amount sufficient, at dosages and for periods of time necessary, to achieve an intended biological effect or a desired therapeutic result including, without limitation, clinical results.
  • the phrase "therapeutically effective amount,” when applied to an antibody or antigen-binding fragment thereof of the invention, is intended to denote an amount of the antibody, or antigen-binding fragment thereof, that is sufficient to ameliorate, palliate, stabilize, reverse, slow, attenuate or delay the progression of a disorder or disease state, or of a symptom of the disorder or disease.
  • the method of the present invention provides for administration of the antibody, or antigen-binding fragment thereof, in combinations with other compounds.
  • the "effective amount” is the amount of the combination sufficient to cause the intended biological effect.
  • a therapeutically effective amount of an anti-Sortilin antibody or antigen-binding fragment thereof of the invention may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the anti-Sortilin antibody or antigen-binding fragment thereof to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody or antibody portion are outweighed by the therapeutically beneficial effects.
  • the antibodies are preferably a human or humanized antibody.
  • IMGT® the international ImMunoGeneTics information system® or, Kabat, E. A., Wu, T. T., Perry, H. M., Gottesmann, K. S. & Foeller, C. (1 991 ). Sequences of Proteins of Immunological Interest, 5th edit., NIH Publication no. 91 -3242 U.S. Department of Health and Human Services; Chothia, C. & Lesk, A. M. (1 987). Canonical structures For The Hypervariable domains Of Immunoglobulins. J. Mol. Biol. 196, 901 -917.
  • the invention relates to an antibody, or an antigen- binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:8 and the light chain variable domain of SEQ ID NO:7.
  • Antibody 1 F2 [0098] According to another embodiment the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:16 and the light chain variable domain of SEQ ID NO:15.
  • Antibody 068 [00100] According to another embodiment the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:24 and the light chain variable domain of SEQ ID NO:23.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:32 and the light chain variable domain of SEQ ID NO:31.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:40 and the light chain variable domain of SEQ ID NO:39.
  • Antibody 93-01 is a group consisting of SEQ ID NO:40 and the light chain variable domain of SEQ ID NO:39.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:48 and the light chain variable domain of SEQ ID NO:47.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:56 and the light chain variable domain of SEQ ID NO:55.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of: (a) a Light Chain CDR1 having the amino acid sequence of SEQ ID NO:57; (b) a Light Chain CDR2 having the amino acid sequence of SEQ ID NO:58;
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:64 and the light chain variable domain of SEQ ID NO:63.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of: (a) a Light Chain CDR1 having the amino acid sequence of SEQ ID NO:65;
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:72 and the light chain variable domain of SEQ ID NO:71.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:80 and the light chain variable domain of SEQ ID NO:79.
  • Antibody 548-01 :
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of: (a) a Light Chain CDR1 having the amino acid sequence of SEQ ID NO:81 ; (b) a Light Chain CDR2 having the amino acid sequence of SEQ ID NO:82;
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:88 and the light chain variable domain of SEQ ID NO:87.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of: (a) a Light Chain CDR1 having the amino acid sequence of SEQ ID NO:89; (b) a Light Chain CDR2 having the amino acid sequence of SEQ ID NO:90;
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:96 and the light chain variable domain of SEQ ID NO:95.
  • Antibody 1289-02 [00120] According to another embodiment the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:104 and the light chain variable domain of SEQ ID NO:103.
  • Antibody 811-02 [00122] According to another embodiment the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:112 and the light chain variable domain of SEQ ID NO:111 .
  • Antibody 566-01
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of: (a) a Light Chain CDR1 having the amino acid sequence of SEQ ID NO:113; (b) a Light Chain CDR2 having the amino acid sequence of SEQ ID NO:114;
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:120 and the light chain variable domain of SEQ ID NO:119.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of: (a) a Light Chain CDR1 having the amino acid sequence of SEQ ID NO:121 ; (b) a Light Chain CDR2 having the amino acid sequence of SEQ ID NO:122;
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:128 and the light chain variable domain of SEQ ID NO:127.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:136 and the light chain variable domain of SEQ ID NO:135.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:144 and the light chain variable domain of SEQ ID NO:143.
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of:
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:152 and the light chain variable domain of SEQ ID NO:151 .
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of: (a) a Light Chain CDR1 having the amino acid sequence of SEQ ID NO:153; (b) a Light Chain CDR2 having the amino acid sequence of SEQ ID NO:154;
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:160 and the light chain variable domain of SEQ ID NO:159.
  • Antibody 002 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • the invention relates to an antibody, or an antigen-binding fragment thereof, comprising or consisting of: (a) a Light Chain CDR1 having the amino acid sequence of SEQ ID NO:161 ;
  • the monoclonal antibody may comprise or consist of the heavy chain variable domain of SEQ ID NO:168 and the light chain variable domain of SEQ ID NO:167.
  • the antibodies mentioned above may, according to one embodiment, further comprise a variant with no more than 4 amino acid differences, or no more than 3 amino acid differences, or no more than 2 amino acid differences, or no more than 1 amino acid difference from said CDR1 , CDR2, and/or CDR3 (VH and/or VL) sequences.
  • the antibodies may be in a composition together with a pharmaceutically acceptable carrier.
  • the antibodies of the invention may be used in therapy.
  • the antibodies of the invention may be used in treating FTD or ALS or TDP43 proteinopathies such as Alzheimer's Disease (AD).
  • AD Alzheimer's Disease
  • the treatment envisioned by the present invention may be chronic and the patient may be treated at least 2 weeks, such as at least for 1 month, 6, months, 1 year or more.
  • the antibodies of the present invention may, for example, be monoclonal antibodies produced by the hybridoma method first described by Kohler et al., Nature 256, 495 (1 975), or may be monoclonal antibodies produced by recombinant DNA or other methods. Monoclonal antibodies may also be isolated from phage antibody libraries using the techniques described in, for example, Clackson et al., Nature 352, 624-628 (1991 ) and Marks et al., J. Mol. Biol. 222, 581 -597 (1 991 ). Monoclonal antibodies may be obtained from any suitable source.
  • monoclonal antibodies may be obtained from hybridomas prepared from murine splenic B lymphocyte cells obtained from mice immunized with an antigen of interest, for instance, in the form of cells expressing the antigen on the surface, or a nucleic acid encoding an antigen of interest.
  • Monoclonal antibodies may also be obtained from hybridomas derived from antibody-expressing cells of immunized humans or from non-human mammals such as rats, rabbits, dogs, sheep, goats, primates, etc.
  • the antibody of the invention is a human antibody.
  • Human monoclonal antibodies directed against Sortilin may be generated using transgenic or transchromosomal mice carrying parts of the human immune system rather than the mouse system.
  • transgenic and trans-chromosomic mice include mice referred to herein as HuMAb mice and KM mice, respectively.
  • the HuMAb mouse contains a human immunoglobulin gene minilocus that encodes unrearranged human heavy variable and constant ( ⁇ and Y) and light variable and constant ( ⁇ ) chain immunoglobulin sequences, together with targeted mutations that inactivate the endogenous ⁇ and K chain loci (Lonberg, N. et al., Nature 368, 856-859 (1994)). Accordingly, the mice exhibit reduced expression of mouse IgM or K and in response to immunization, the introduced human heavy and light chain transgenes, undergo class switching and somatic mutation to generate high affinity human IgG, ⁇ ; monoclonal antibodies (Lonberg, N. et al.
  • HuMAb mice The preparation of HuMAb mice is described in detail in Taylor, L. et al., Nucleic Acids Research 20, 6287-6295 (1992), Chen, J. et al., International Immunology 5, 647-656 (1 993), Tuaillon et al., J. Immunol. 1 52, 2912-2920 (1994), Taylor, L.
  • mice have a JKD disruption in their endogenous light chain (kappa) genes (as described in Chen et al., EMBO J. 12, 81 1 -820 (1993)), a CMD disruption in their endogenous heavy chain genes (as described in Example 1 of WO 01 /14424), and a KCo5 human kappa light chain transgene (as described in Fishwild et al., Nature Biotechnology 14, 845-851 (1996)).
  • kappa endogenous light chain
  • CMD disruption in their endogenous heavy chain genes as described in Example 1 of WO 01 /14424
  • KCo5 human kappa light chain transgene as described in Fishwild et al., Nature Biotechnology 14, 845-851 (1996).
  • the HCo7 mice have a HCo7 human heavy chain transgene (as described in US 5,770,429), the HCo12 mice have a HCo1 2 human heavy chain transgene (as described in Example 2 of WO 01 /14424), the HCo17 mice have a HCo17 human heavy chain transgene (as described in Example 2 of WO 01/09187) and the HCo20 mice have a HCo20 human heavy chain transgene.
  • the resulting mice express human immunoglobulin heavy and kappa light chain transgenes in a background homozygous for disruption of the endogenous mouse heavy and kappa light chain loci.
  • the endogenous mouse kappa light chain gene has been homozygously disrupted as described in Chen et al., EMBO J. 12, 81 1 -820 (1 993) and the endogenous mouse heavy chain gene has been homozygously disrupted as described in Example 1 of WO 01/09187.
  • This mouse strain carries a human kappa light chain transgene, KCo5, as described in Fishwild et al., Nature Biotechnology 14, 845-851 (1996).
  • This mouse strain also carries a human heavy chain transchromosome composed of chromosome 14 fragment hCF (SC20) as described in WO 02/43478.
  • HCo12-Balb/c, HCo17- Balb/c and HCo20-Balb/c mice can be generated by crossing HCo1 2, HCo17 and HCo20 to KCo5[J/K](Balb) as described in WO 09/097006.
  • the endogenous mouse kappa light chain gene has been homozygously disrupted as described in Chen et al., EMBO J. 12, 81 1 -820 (1 993) and the endogenous mouse heavy chain gene has been homozygously disrupted as described in Example 1 of WO 01/09187.
  • This mouse strain carries a human kappa light chain transgene, KCo5, as described in Fishwild et al., Nature Biotechnology 14, 845-851 (1996).
  • This mouse strain also carries a human heavy chain trans-chromosome composed of chromosome 14 antigen-binding fragment hCF (SC20) as described in WO 02/43478.
  • Splenocytes from these transgenic mice may be used to generate hybridomas that secrete human monoclonal antibodies according to well-known techniques.
  • Human monoclonal or polyclonal antibodies of the present invention, or antibodies of the present invention originating from other species may also be generated transgenically through the generation of another non- human mammal or plant that is transgenic for the immunoglobulin heavy and light chain sequences of interest and production of the antibody in a recoverable form therefrom.
  • antibodies may be produced in, and recovered from, the milk of goats, cows, or other mammals. See for instance US 5,827,690, US 5,756,687, US 5,750,172 and US 5,741 ,957.
  • the antibody of the invention may be of any isotype.
  • the choice of isotype typically will be guided by the desired effector functions, such as ADCC induction.
  • Exemplary isotypes are lgG1 , lgG2, lgG3, and lgG4. Either of the human light chain constant domains, kappa or lambda, may be used.
  • the class of an anti-Sortilin antibody of the present invention may be switched by known methods. For example, an antibody of the present invention that was originally IgM may be class switched to an IgG antibody of the present invention. Further, class switching techniques may be used to convert one IgG subclass to another, for instance from IgGI to lgG2.
  • an antibody of the present invention is an lgG1 antibody, for instance an lgG1 , ⁇ ;- An antibody is said to be of a particular isotype if its amino acid sequence is most homologous to that isotype, relative to other isotypes.
  • the antibody of the invention is a full-length antibody, preferably an IgG antibody, in particular an lgG1 , ⁇ ; antibody.
  • the antibody of the invention is an antibody antigen- binding fragment or a single-chain antibody.
  • Antibodies and antigen-binding fragments thereof may e.g. be obtained by antigen-binding fragmentation using conventional techniques, and antigen-binding fragments screened for utility in the same manner as described herein for whole antibodies. For example, F(ab')2 antigen-binding fragments may be generated by treating antibody with pepsin.
  • the resulting F(ab')2 antigen-binding fragment may be treated to reduce disulfide bridges to produce Fab' antigen-binding fragments.
  • Fab antigen-binding fragments may be obtained by treating an IgG antibody with papain; Fab' antigen-binding fragments may be obtained with pepsin digestion of IgG antibody.
  • An F(ab') antigen-binding fragment may also be produced by binding Fab'-described below via a thioether bond or a disulfide bond.
  • a Fab' antigen-binding fragment is an antibody antigen-binding fragment obtained by cutting a disulfide bond of the hinge domain of the F(ab') 2 .
  • a Fab'- antigen-binding fragment may be obtained by treating an F(ab') 2 antigen-binding fragment with a reducing agent, such as dithiothreitol.
  • Antibody antigen-binding fragment may also be generated by expression of nucleic acids encoding such antigen-binding fragments in recombinant cells (see for instance Evans et al., J. Immunol. Meth. 184, 1 23-38 (1 995)).
  • a chimeric gene encoding a portion of an F(ab')2 antigen- binding fragment could include DNA sequences encoding the CH1 domain and hinge domain of the H chain, followed by a translational stop codon to yield such a truncated antibody antigen-binding fragment molecule.
  • the anti-Sortilin antibody is a monovalent antibody, preferably a monovalent antibody as described in WO2007059782 (which is incorporated herein by reference in its entirety) having a deletion of the hinge region.
  • the antibody is a monovalent antibody, wherein said anti-Sortilin antibody is constructed by a method comprising : i) providing a nucleic acid construct encoding the light chain of said monovalent antibody, said construct comprising a nucleotide sequence encoding the VL region of a selected antigen specific anti-Sortilin antibody and a nucleotide sequence encoding the constant CL region of an Ig, wherein said nucleotide sequence encoding the VL region of a selected antigen specific antibody and said nucleotide sequence encoding the CL region of an Ig are operably linked together, and wherein, in case of an lgG1 subtype, the nucleotide sequence encoding the CL region has
  • the anti-Sortilin antibody is a monovalent antibody, which comprises:
  • variable domain of an antibody of the invention as described herein or an antigen-binding part of the said domain (i) a variable domain of an antibody of the invention as described herein or an antigen-binding part of the said domain
  • a CH domain of an immunoglobulin or a domain thereof comprising the CH2 and CH3 domains, wherein the CH domain or domain thereof has been modified such that the domain corresponding to the hinge domain and, if the immunoglobulin is not an lgG4 subtype, other domains of the CH domain, such as the CH3 domain, do not comprise any amino acid residues, which are capable of forming disulfide bonds with an identical CH domain or other covalent or stable non-covalent inter-heavy chain bonds with an identical CH domain in the presence of polyclonal human IgG.
  • the heavy chain of the monovalent antibody has been modified such that the entire hinge region has been deleted.
  • the sequence of the monovalent antibody has been modified so that it does not comprise any acceptor sites for N-linked glycosylation.
  • the invention also includes "Bispecific Antibodies," wherein an anti- Sortilin binding region ⁇ e.g., a Sortilin-binding region of an anti-Sortilin monoclonal antibody) is part of a bivalent or polyvalent bispecific scaffold that targets more than one epitope, (for example a second epitope could comprise an epitope of an active transport receptor, such that the Bispecific Antibody would exhibit improved transcytosis across a biological barrier, such as the Blood Brain Barrier).
  • the monovalent Fab of an anti-Sortilin antibody may be joined to an additional Fab or scfv that targets a different protein to generate a bispecific antibody.
  • a bispecific antibody can have a dual function, for example a therapeutic function imparted by an anti-sortilin binding domain and a transport function that can bind to a receptor molecule to enhance transfer cross a biological barrier, such as the blood brain barrier.
  • Antibodies and antigen-binding fragments thereof of the invention also include single chain antibodies.
  • Single chain antibodies are peptides in which the heavy and light chain Fv domains are connected.
  • the present invention provides a single-chain Fv (scFv) wherein the heavy and light chains in the Fv of an anti-Sortilin antibody of the present invention are joined with a flexible peptide linker (typically of about 10, 12, 15 or more amino acid residues) in a single peptide chain.
  • scFv single-chain Fv
  • Methods of producing such antibodies are described in for instance US 4,946,778, Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 1 1 3, Rosenburg and Moore eds. Springer-Verlag, New York, pp.
  • the single chain antibody may be monovalent, if only a single VH and VL are used, bivalent, if two VH and VL are used, or polyvalent, if more than two VH and VL are used.
  • the antibodies and antigen-binding fragments thereof described herein may be modified by inclusion of any suitable number of modified amino acids and/or associations with such conjugated substituents.
  • Suitability in this context is generally determined by the ability to at least substantially retain the Sortilin selectivity and/or Sortilin specificity associated with the non-derivatized parent anti-Sortilin antibody.
  • the inclusion of one or more modified amino acids may be advantageous in, for example, increasing polypeptide serum half-life, reducing polypeptide antigenicity, or increasing polypeptide storage stability.
  • Amino acid(s) are modified, for example, co-translationally or post- translationally during recombinant production (e.g., N-linked glycosylation at N- X-S/T motifs during expression in mammalian cells) or modified by synthetic means.
  • Non-limiting examples of a modified amino acid include a glycosylated amino acid, a sulfated amino acid, a prenylated (e. g., farnesylated, geranyl- geranylated) amino acid, an acetylated amino acid, an acylated amino acid, a PEGylated amino acid, a biotinylated amino acid, a carboxylated amino acid, a phosphorylated amino acid, and the like.
  • the modified amino acid may, for instance, be selected from a glycosylated amino acid, a PEGylated amino acid, a farnesylated amino acid, an acetylated amino acid, a biotinylated amino acid, an amino acid conjugated to a lipid moiety, or an amino acid conjugated to an organic derivatizing agent.
  • the antibodies and antigen-binding fragments thereof of the invention may also be chemically modified by covalent conjugation to a polymer to for instance increase their circulating half-life.
  • Exemplary polymers, and methods to attach them to peptides are illustrated in for instance US 4,766, 106; US 4,179,337; US 4,495,285 and US 4,609,546.
  • Additional illustrative polymers include polyoxyethylated polyols and polyethylene glycol (PEG) (e.g., a PEG with a molecular weight of between about 1 ,000 and about 40,000, such as between about 2,000 and about 20,000, e.g., about 3,000- 12,000 g/mol).
  • PEG polyethylene glycol
  • the antibodies and antigen-binding fragments thereof of the present invention may further be used in a diagnostic method or as a diagnostic imaging ligand.
  • antibodies and antigen-binding fragments thereof of the invention comprising one or more radiolabeled amino acids are provided.
  • a radiolabeled anti-Sortilin antibody may be used for both diagnostic and therapeutic purposes (conjugation to radiolabeled molecules is another possible feature).
  • Non-limiting examples of such labels include, but are not limited to bismuth ( 213 Bi), carbon ( 11 C, 13 C, 1 C), chromium ( 51 Cr), cobalt ( 57 Co, 60 Co), copper ( 6 Cu), dysprosium ( 165 Dy), erbium ( 169 Er), fluorine ( 18 F), gadolinium ( 153 Gd, 159 Gd), gallium ( 68 Ga, 67 Ga), germanium ( 68 Ge), gold ( 198 Au), holmium ( 166 Ho), hydrogen ( 3 H), indium ( 11 1 ln, 112 ln, 113 ln, 115 ln), iodine ( 121 l, 123 l, 125 l, 131 1), iridium ( 192 lr), iron ( 59 Fe), krypton ( 81 m Kr), lanthanium ( 1 0 La), lutelium ( 177 Lu), manganese ( 5 Mn), molybdenum ( 99 Mo), nitrogen (
  • radiolabeled amino acids and related peptide derivatives are known in the art (see for instance Junghans et al., in Cancer Chemotherapy and Biotherapy 655- 686 (2nd edition, Chafner and Longo, eds., Lippincott Raven (1996)) and US 4,681 ,581 , US 4,735,210, US 5,1 01 ,827, US 5, 102,990 (US RE35,500), US 5,648,471 and US 5,697,902.
  • a radioisotope may be conjugated by a chloramine T method (Lindegren, S. et al.
  • the invention also provides anti-Sortilin antibodies and antigen- binding fragments thereof that are detectably labeled using a fluorescent label (such as a rare earth chelate (e.g., a europium chelate)), a fluorescein-type label (e.g., fluorescein, fluorescein isothiocyanate, 5-carboxyfluorescein, 6- carboxy fluorescein, dichlorotriazinylamine fluorescein), a rhodamine-type label (e.g., ALEXA FLUOR® 568 (Invitrogen), TAMRA® or dansyl chloride), VIVOTAG 680 XL FLUOROCHROMETM (Perkin Elmer), phycoerythrin; umbelliferone, Lissamine; a cyanine; a phycoerythrin, Texas Red, BODIPY FL- SE® (Invitrogen) or an analogue thereof, all of which are suitable
  • Chemiluminescent labels may be employed (e.g., luminol, luciferase, luciferin, and aequorin). Such diagnosis and detection can also be accomplished by coupling the diagnostic molecule of the present invention to detectable substances including, but not limited to, various enzymes, enzymes including, but not limited to, horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase, or to prosthetic group complexes such as, but not limited to, streptavidin/biotin and avidin/biotin.
  • Chemiluminescent labels may be employed (e.g., luminol, luciferase, luciferin, and aequorin). Such diagnosis and detection can also be accomplished by coupling the diagnostic molecule of the present invention to detectable substances including, but not limited to, various enzymes, enzymes including, but not limited to, horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase, or to prosthetic group complexes such as, but not limited to, streptavidin/biotin and avidin/biotin.
  • Paramagnetic labels can also be employed, and are preferably detected using Positron Emission Tomography (PET) or Single-Photon Emission Computed Tomography (SPECT).
  • Such paramagnetic labels include, but are not limited to compounds containing paramagnetic ions of Aluminum (Al), Barium (Ba), Calcium (Ca), Cerium (Ce), Dysprosium (Dy), Erbium (Er), Europium (Eu), Gandolinium (Gd), Holmium (Ho), Iridium (Ir), Lithium (Li), Magnesium (Mg), Manganese (Mn), Molybdenum (M), Neodymium (Nd), Osmium (Os), Oxygen (O), Palladium (Pd), Platinum (Pt), Rhodium (Rh), Ruthenium (Ru), Samarium (Sm), Sodium (Na), Strontium (Sr), Terbium (Tb), Thulium (Tm), Tin (Sn), Titanium (Ti
  • the anti-Sortilin antibody or Sortilin-binding fragment thereof of the invention may be labelled with a fluorescent label, a chemiluminescent label, a paramagnetic label, a radioisotopic label or an enzyme label.
  • the labelled antibody of fragment may be used in detecting or measuring the presence or amount of said Sortilin in the brain of a subject.
  • This method may comprise the detection or measurement of in vivo imaging of anti- Sortilin antibody or Sortilin-binding fragment bound to said Sortilin and may comprises ex vivo imaging of said anti-Sortilin antibody or Sortilin-binding fragment bound to such Sortilin.
  • the invention relates to an expression vector encoding one or more polypeptide chains of an antibody of the invention or an antigen-binding-domain thereof.
  • Such expression vectors may be used for recombinant production of the antibodies and antigen-binding fragments of the invention.
  • An expression vector in the context of the present invention may be any suitable DNA or RNA vector, including chromosomal, non-chromosomal, and synthetic nucleic acid vectors (a nucleic acid sequence comprising a suitable set of expression control elements).
  • suitable DNA or RNA vector including chromosomal, non-chromosomal, and synthetic nucleic acid vectors (a nucleic acid sequence comprising a suitable set of expression control elements).
  • examples of such vectors include derivatives of SV40, bacterial plasmids, phage DNA, baculovirus, yeast plasmids, vectors derived from combinations of plasmids and phage DNA, and viral nucleic acid (RNA or DNA) vectors.
  • an anti-Sortilin antibody-encoding nucleic acid is comprised in a naked DNA or RNA vector, including, for example, a linear expression element (as described in, for instance, Sykes and Johnston, Nat Biotech 12, 355-59 (1997)), a compacted nucleic acid vector (as described in for instance US 6,077,835 and/or WO 00/70087), a plasmid vector such as pBR322, pUC 1 9/18, or pUC 1 18/1 1 9, a "midge" minimally-sized nucleic acid vector (as described in, for instance, Schakowski et al., Mol Ther 3, 793-800 (2001 )), or as a precipitated nucleic acid vector construct, such as a CaPO 4 -precipitated construct (as described in, for instance, WO 00/46147, Benvenisty and Reshef, PNAS USA 83, 9551 -55 (1 986), Wigler et al.,
  • the vector is suitable for expression of anti- Sortilin antibodies or antigen-binding fragments thereof in a bacterial cell.
  • expression vectors such as BlueScript (Stratagene), pIN vectors (Van Heeke & Schuster, J Biol Chem 264, 5503-5509 (1 989), pET vectors (Novagen, Madison, Wl) and the like).
  • An expression vector may also or alternatively be a vector suitable for expression in a yeast system. Any vector suitable for expression in a yeast system may be employed. Suitable vectors include, for example, vectors comprising constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH (reviewed in: F. Ausubel et al., ed. Current Protocols in Molecular Biology, Greene Publishing and Wiley InterScience New York (1987), Grant et al., Methods in Enzymol 153, 516-544 (1987), Mattanovich, D. et al. Methods Mol. Biol. 824, 329-358 (2012), Celik, E. et al. Biotechnol. Adv.
  • constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH
  • anti-Sortilin antibody- encoding nucleic acids may comprise or be associated with any suitable promoter, enhancer, and other expression-facilitating elements.
  • suitable promoter, enhancer, and other expression-facilitating elements include strong expression promoters (e. g., human CMV IE promoter/enhancer as well as RSV, SV40, SL3-3, MMTV, and HIV LTR promoters), effective poly (A) termination sequences, an origin of replication for plasmid product in E. coli, an antibiotic resistance gene as selectable marker, and/or a convenient cloning site (e.g., a polylinker).
  • Nucleic acids may also comprise an inducible promoter as opposed to a constitutive promoter such as CMV IE (the skilled artisan will recognize that such terms are actually descriptors of a degree of gene expression under certain conditions).
  • the invention relates to a recombinant eukaryotic or prokaryotic host cell, such as a transfectoma, which produces an antibody or antigen-binding fragment thereof of the invention as defined herein or a bispecific molecule of the invention as defined herein.
  • host cells include yeast, bacteria, and mammalian cells, such as CHO or HEK cells.
  • the present invention provides a cell comprising a nucleic acid stably integrated into the cellular genome that comprises a sequence coding for expression of an anti-Sortilin antibody of the present invention or an antigen-binding fragment thereof.
  • the present invention provides a cell comprising a non-integrated nucleic acid, such as a plasmid, cosmid, phagemid, or linear expression element, which comprises a sequence coding for expression of an anti-Sortilin antibody or antigen-binding fragment thereof of the invention.
  • a non-integrated nucleic acid such as a plasmid, cosmid, phagemid, or linear expression element, which comprises a sequence coding for expression of an anti-Sortilin antibody or antigen-binding fragment thereof of the invention.
  • the invention relates to a method for producing an anti-Sortilin antibody of the invention, said method comprising the steps of a) culturing a hybridoma or a host cell of the invention as described herein above, and b) purifying the antibody of the invention from the culture media.
  • the invention relates to a preparation that, as such term is used herein, comprises an anti-Sortilin antibody as defined herein, and that is substantially free of naturally-arising antibodies that are either not capable of binding to sortilin or that do not materially alter the anti-Sortilin functionality of the preparation.
  • a preparation does not encompass naturally-arising serum, or a purified derivative of such serum, that comprises a mixture of an anti-Sortilin antibody and another antibody that does not alter the functionality of the anti-Sortilin antibody of the preparation, wherein such functionality is:
  • FTD frontotemporal dementia
  • ALS amyotrophic lateral sclerosis
  • the invention particularly relates to preparations of such an anti- Sortilin antibody having a structural change in its amino acid sequence (in any of its CDRs, variable domains, framework residues and/or constant domains) relative to the structure of a naturally-occurring anti-Sortilin antibody, wherein said structural change causes the anti-Sortilin antibody monoclonal antibody to exhibit a markedly altered functionality (i.e., more than a 20% difference, more than a 40% difference, more than a 60% difference, more than an 80% difference, more than a 100% difference, more than a 150% difference, more than a 2-fold difference, more than a 4-fold difference, more than a 5-fold difference, or more than a 10-fold difference in functionality) relative to the functionality exhibited by said naturally-occurring anti-Sortilin antibody; wherein such functionality is:
  • FTD frontotemporal dementia
  • ALS amyotrophic lateral sclerosis
  • AD Alzheimer's Disease
  • substantially free of naturally-arising antibodies refers to the complete absence of such naturally-arising antibodies in such preparations, or of the inclusion of a concentration of such naturally-arising antibodies in such preparations that does not materially affect the Sortilin-binding properties of the preparations.
  • An antibody is said to be "isolated” if it has no naturally-arising counterpart or has been separated or purified from components which naturally accompany it.
  • naturally-arising antibodies refers to antibodies (including naturally-arising autoantibodies) elicited within living humans or other animals, as a natural consequence to the functioning of their immune systems.
  • the preparations of the present invention do not exclude, and indeed explicitly encompass, such preparations that contain an anti-Sortilin antibody and a deliberately added additional antibody capable of binding to an epitope that is not possessed by Sortilin.
  • Such preparations particularly include embodiments thereof wherein the preparation exhibits enhanced efficacy in treating frontotemporal dementia (FTD) and/or amyotrophic lateral sclerosis (ALS).
  • FTD frontotemporal dementia
  • ALS amyotrophic lateral sclerosis
  • the antibodies of antigen-binding fragments thereof of the present invention may be produced in different cell lines, such as a human cell line, a mammal non-human cell line, and insect cell line, for example a CHO cell line, HEK cell line, BHK-21 cell line, murine cell line (such as a myeloma cell line), fibrosarcoma cell line, PER.C6 cell line, HKB-1 1 cell line, CAP cell line and HuH-7 human cell line (Dumont et al, 2015, Crit Rev Biotechnol. Sep 18:1 -13., the contents which is included herein by reference).
  • insect cell line for example a CHO cell line, HEK cell line, BHK-21 cell line, murine cell line (such as a myeloma cell line), fibrosarcoma cell line, PER.C6 cell line, HKB-1 1 cell line, CAP cell line and HuH-7 human cell line (Dumont et al, 2015, Crit Rev Biotechnol.
  • an anti-Sortilin antibody or antigen-binding fragment thereof both as defined herein, or a preparation, as such term is defined herein, that comprises such an anti-Sortilin antibody or antigen-binding fragment thereof;
  • compositions may be formulated with pharmaceutically acceptable carriers or diluents as well as any other known adjuvants and excipients in accordance with conventional techniques such as those disclosed in Remington: The Science and Practice of Pharmacy, 22nd Edition, Gennaro, Ed., Mack Publishing Co., Easton, PA, 2013.
  • the pharmaceutically acceptable carriers or diluents as well as any other known adjuvants and excipients should be suitable for the chosen compound of the present invention and the chosen mode of administration. Suitability for carriers and other components of pharmaceutical compositions is determined based on the lack of significant negative impact on the desired biological properties of the chosen compound or pharmaceutical composition of the present invention (e.g., less than a substantial impact (10% or less relative inhibition, 5% or less relative inhibition, etc.)) on epitope binding.
  • a pharmaceutical composition of the present invention may also include diluents, fillers, salts, buffers, detergents (e.g., a non-ionic detergent, such as Tween-20 or Tween- 80), stabilizers (e.g., sugars or protein-free amino acids), preservatives, tissue fixatives, solubilizers, and/or other materials suitable for inclusion in a pharmaceutical composition.
  • the diluent is selected to not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution.
  • compositions or formulation may also include other carriers, or non-toxic, nontherapeutic, non-immunogenic stabilizers and the like.
  • the compositions may also include large, slowly metabolized macromolecules, such as proteins, polysaccharides like chitosan, polylactic acids, polyglycolic acids and copolymers (e.g., latex functionalized sepharose, agarose, cellulose, and the like), polymeric amino acids, amino acid copolymers, and lipid aggregates (e.g., oil droplets or liposomes).
  • the actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration.
  • the selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • the pharmaceutical composition may be administered by any suitable route and mode, including: parenteral, topical, oral or intranasal means for prophylactic and/or therapeutic treatment.
  • a pharmaceutical composition of the present invention is administered parenterally.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and include epidermal, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, intratendinous, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, intracranial, intrathoracic, epidural and intrasternal injection and infusion. Additional suitable routes of administering a compound of the present invention in vivo and in vitro are well known in the art and may be selected by those of ordinary skill in the art. In one embodiment that pharmaceutical composition is administered by intra
  • Pharmaceutically acceptable carriers include any and all suitable solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonicity agents, antioxidants and absorption delaying agents, and the like that are physiologically compatible with a compound of the present invention.
  • aqueous and non-aqueous carriers examples include water, saline, phosphate buffered saline, ethanol, dextrose, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, corn oil, peanut oil, cottonseed oil, and sesame oil, carboxymethyl cellulose colloidal solutions, tragacanth gum and injectable organic esters, such as ethyl oleate, and/or various buffers.
  • Other carriers are well known in the pharmaceutical arts.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the present invention is contemplated.
  • Proper fluidity may be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions of the present invention may also comprise pharmaceutically acceptable antioxidants for instance (1 ) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha- tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butyl
  • compositions of the present invention may also comprise isotonicity agents, such as sugars, polyalcohols, such as mannitol, sorbitol, glycerol or sodium chloride in the compositions.
  • isotonicity agents such as sugars, polyalcohols, such as mannitol, sorbitol, glycerol or sodium chloride in the compositions.
  • compositions of the present invention may also contain one or more adjuvants appropriate for the chosen route of administration such as preservatives, wetting agents, emulsifying agents, dispersing agents, preservatives or buffers, which may enhance the shelf life or effectiveness of the pharmaceutical composition.
  • adjuvants appropriate for the chosen route of administration such as preservatives, wetting agents, emulsifying agents, dispersing agents, preservatives or buffers, which may enhance the shelf life or effectiveness of the pharmaceutical composition.
  • the compounds of the present invention may be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • Such carriers may include gelatin, glyceryl monostearate, glyceryl distearate, biodegradable, biocompatible polymers such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid alone or with a wax, or other materials well known in the art. Methods for the preparation of such formulations are generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1 978.
  • the compounds of the present invention may be formulated to ensure proper distribution in vivo.
  • Pharmaceutically acceptable carriers for parenteral administration include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the present invention is contemplated. Supplementary active compounds may also be incorporated into the compositions.
  • compositions for injection must typically be sterile and stable under the conditions of manufacture and storage.
  • the composition may be formulated as a solution, micro-emulsion, liposome, or other ordered structure suitable to high drug concentration.
  • the carrier may be an aqueous or non-aqueous solvent or dispersion medium containing for instance water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • the proper fluidity may be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • a coating such as lecithin
  • surfactants it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as glycerol, mannitol, sorbitol, or sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions may be brought about by including in the composition an agent that delays antibody absorption, for example, monostearate salts and gelatin.
  • Sterile injectable solutions may be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients e.g.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients e.g. from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions examples of methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Sterile injectable solutions may be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • examples of methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Dosage regimens in the above methods of treatment and uses described herein are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • Parenteral compositions may be formulated in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the present invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals.
  • the effective dosages and the dosage regimens for the anti-Sortilin antibodies depend on the disease or condition to be treated and may be determined by the persons skilled in the art.
  • An exemplary, non-limiting range for a therapeutically effective amount of an antibody of the present invention is about 0.1 -10 mg/kg/body weight, such as about 0.1 -5 mg/kg/body weight, for example about 0.1 -2 mg/kg/body weight, such as about 0.1 -1 mg/kg/body weight, for instance about 0.15, about 0.2, about 0.5, about 1 , about 1 .5 or about 2 mg/kg/body weight.
  • a physician or veterinarian having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the anti-Sortilin antibody employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a composition of the present invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect.
  • Such an effective dose will generally depend upon the factors described above.
  • Administration may e.g. be intravenous, intramuscular, intraperitoneal, or subcutaneous, and for instance administered proximal to the site of the target.
  • the effective daily dose of a pharmaceutical composition may be administered as two, three, four, five, six or more sub- doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical composition as described above.
  • the labelled antibodies or antigen-binding fragments thereof of the invention can be used for diagnostic purposes to detect, diagnose, or monitor diseases or disorders.
  • the invention provides for the detection or diagnosis of a neurodegenerative or cognitive disease or disorder, including but not limited to FTD, ALS or TDP43 proteinopathies such as Alzheimer's Disease (AD), comprising: (a) assaying the existence of pyroglutamated ⁇ fragments in cells or tissue samples of a subject using one or more antibodies that specifically bind to Sortilin; and (b) comparing the level of the antigen with a control level, e.g. levels in normal tissue samples, whereby an increase in the assayed level of antigen compared to the control level of antigen is indicative of the disease or disorder, or indicative of the severity of the disease or disorder.
  • a control level e.g. levels in normal tissue samples
  • the antibodies or antigen-binding fragments thereof of the invention can be used to assay Sortilin or antigen-binding fragments of Sortilin in a biological sample using immuno-histochemical methods well-known in the art.
  • Other antibody-based methods useful for detecting protein include immunoassays such as the enzyme linked immunoassay (ELISA) and the radioimmunoassay assay (RIA) and mesoscale discovery platform based assays (MSD).
  • Suitable antibody labels may be used in such kits and methods, and labels known in the art include enzyme labels, such as alkaline phosphatase and glucose oxidase; radioisotope labels, such as iodine ( 125 l, 131 1), carbon ( 1 C), sulfur ( 35 S), tritium ( 3 H), indium ( 121 ln), and technetium ( 99m Tc); and luminescent labels, such as luminol and luciferase; and fluorescent labels, such as fluorescein and rhodamine.
  • enzyme labels such as alkaline phosphatase and glucose oxidase
  • radioisotope labels such as iodine ( 125 l, 131 1), carbon ( 1 C), sulfur ( 35 S), tritium ( 3 H), indium ( 121 ln), and technetium ( 99m Tc)
  • luminescent labels such as luminol and luciferase
  • fluorescent labels such as fluorescein and rh
  • diagnosis comprises: a) administering to a subject an effective amount of such labeled molecule; b) waiting for a time interval following administration to allow the labeled molecule to concentrate at sites (if any) of ⁇ deposition and to allow for unbound labeled molecule to be cleared to background level; c) determining a background level; and d) detecting the labeled molecule in the subject, such that detection of labeled molecule above the background level is indicative that the subject has the disease or disorder, or is indicative of the severity of the disease or disorder.
  • the molecule is labeled with an imaging moiety suitable for detection using a particular imaging system known to those skilled in the art.
  • Background levels may be determined by various methods known in the art, including comparing the amount of labeled antibody detected to a standard value previously determined for a particular imaging system.
  • Methods and systems that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as positron emission tomography (PET), magnetic resonance imaging (MRI), and sonography.
  • CT computed tomography
  • PET positron emission tomography
  • MRI magnetic resonance imaging
  • sonography sonography
  • the invention relates to an antibody, or antigen- binding fragment thereof, of the invention, for use in medicine.
  • the invention relates to an antibody, or antigen- binding fragment thereof, of the invention, for use in treating a disease associated with decreased PGRN levels in the brain of a patient,
  • the invention relates to the use of the antibody, or antigen-binding fragment thereof, of the invention, in the manufacture of a medicament for treating a disease associated with decreased PGRN levels in the brain of a patient, [00202] In a further aspect, the invention relates to a method of preventing or treating a disease associated with decreased PGRN levels in the brain of a patient, comprising administering an effective dosage of an antibody of the invention, or an antigen-binding fragment thereof.
  • the disease is: FTD; ALS; or TDP43 proteinopathies, such as AD.
  • the treatment is chronic, and is preferably for at least 2 weeks, such as at least for 1 month, 6, months, 1 year or more.
  • the invention provides a kit comprising the antibody, or antigen-binding fragment thereof, of the invention.
  • an antigen-binding fragment selected from the group consisting of: an Fv fragment (e.g. single chain Fv or a disulphide- bonded Fv); a Fab-like fragment (e.g. Fab fragment or F(ab') 2 fragment); and a domain antibody (e.g. a single V H variable domain or V L variable domain).
  • an Fv fragment e.g. single chain Fv or a disulphide- bonded Fv
  • a Fab-like fragment e.g. Fab fragment or F(ab') 2 fragment
  • a domain antibody e.g. a single V H variable domain or V L variable domain
  • the antibody, or antigen-binding fragment thereof, according to Embodiment 7, wherein the capability of the antibody or fragment thereof to reduce PGRN binding to Sortilin comprises reducing PGRN binding to Sortilin by 1 0% or more; for example, by 20% or more; or by 30% or more.
  • the antibody, or antigen-binding fragment thereof, according to Embodiment 7 or 8, wherein the capability of said antibody or fragment thereof the antibody or fragment thereof to reduce and/or inhibit PGRN binding to Sortilin comprises reducing and/or inhibiting PGRN binding to Sortilin with an IC50 at or below 22 nM, such as between 22 nM and 1 nM, or between 10 nM and 1 nM, or between 5 nM and 1 nM.
  • the antibody, or antigen-binding fragment thereof according to any previous Embodiment, wherein the antibody or antigen-binding fragment thereof is human or is humanized.
  • the antibody, or antigen-binding fragment thereof, according to any preceding Embodiment comprising a light chain variable domain comprising one or more of the CDR 1 -3 Light Chain as listed for each of the antibodies defined in Table 5, or an amino acid sequence having no more than 4 amino acid differences, or no more than 3 amino acid differences, or no more than 2 amino acid differences, or no more than 1 amino acid difference.
  • the antibody, or antigen-binding fragment thereof, according to Embodiment 1 1 comprising a light chain variable domain comprising the
  • CDR 1 -3 Light Chain as listed for each of the antibodies defined in Table 5.
  • the antibody, or antigen-binding fragment thereof comprising a heavy chain variable domain comprising one or more CDR 1 -3 Heavy Chain as listed for each of the antibodies defined in Table 5, or an amino acid sequence having no more than 4 amino acid differences, or no more than 3 amino acid differences, or no more than 2 amino acid differences, or no more than 1 amino acid difference.
  • an antibody, or antigen-binding fragment thereof, according to Embodiment 15 or 16 comprising a heavy chain variable domain comprising or consisting of the amino acid sequence of VH as listed for each of the antibodies defined in Table 5.
  • the antibody, or antigen-binding fragment thereof, according to any of Embodiments 15 to 17, comprising a heavy chain comprising or consisting of the amino acid sequence VL as listed for each of the antibodies defined in Table 5.
  • the antibody, or antigen-binding fragment thereof, according to any preceding embodiment comprising a light chain variable domain comprising or consisting of the amino acid sequence of VL as listed for each of the antibodies defined in Table 5, and a heavy chain variable domain comprising or consisting of the amino acid sequence of VH as listed for each of the antibodies defined in Table 5.
  • the antibody, or antigen-binding fragment thereof, according to any preceding embodiment, comprising a light chain comprising or consisting of the amino acid sequence of VL as listed for each of the antibodies defined in Table 5, and a heavy chain comprising or consisting of the amino acid sequence of VH as listed for each of the antibodies defined in Table 5.
  • the antibody, or antigen-binding fragment thereof, according to any preceding Embodiment, wherein said antibody or antigen-binding fragment thereof competes with the antibody or antigen-binding fragment thereof defined in Embodiment 20 for binding to Sortilin.
  • the antibody, or antigen-binding fragment thereof, according to any preceding Embodiment, wherein the antibody or antigen-binding fragment comprises an Fc region.
  • the moiety for increasing the in vivo half-life is selected from the group consisting of polyethylene glycol (PEG), human serum albumin, glycosylation groups, fatty acids and dextran.
  • the antibody or antigen-binding fragment further comprises a detectable moiety.
  • the radioisotope is selected from the group consisting of 99m Tc, 11 1 In, 67 Ga, 68 Ga, 72 As, 89 Zr, 123 l and 201 TI.
  • the antibody, or antigen-binding fragment thereof, according to Embodiment 29 wherein the paramagnetic isotope is selected from the group consisting of 157 Gd, 55 Mn, 162 Dy, 52 Cr and 56 Fe.
  • DOTA deferoxamine
  • DTPA diethylenetriaminepentaacetic avid
  • NOTA 1,4,8,1 1 -tetraazacyclodocedan-1 ,4,8, 1 1 - tetraacetic acid
  • a vector comprising a nucleic acid molecule as defined in Embodiment 34 or 35.
  • a recombinant host cell comprising a nucleic acid molecule as defined in any of Embodiments 34-36.
  • a method for producing an antibody or antigen-binding fragment as defined in any of Embodiments 1 -33 comprising culturing a host cell as defined in Embodiment 37 under conditions which permit expression of the encoded antibody or antigen-binding fragment thereof.
  • a preparation comprising the antibody or antigen-binding fragment thereof according to any one of the previous Embodiments, wherein said preparation is substantially free of naturally-arising antibodies that are either not capable of binding to Sortilin or that do not materially alter an an- ti-Sortilin functionality of the preparation, said functionality being selected from the group consisting of:
  • FTD frontotemporal dementia
  • ALS amyotrophic lateral sclerosis
  • a preparation comprising the monoclonal antibody or antigen-binding fragment thereof according to any one of the previous Embodiments, wherein said monoclonal antibody possesses a structural change in its amino acid sequence, relative to the structure of a naturally-occurring an- ti-Sortilin antibody, wherein said structural change causes said monoclonal antibody to exhibit an altered functionality relative to the functionality exhibited by said naturally-occurring anti-Sortilin antibody, wherein said functionality is:
  • a pharmaceutical composition comprising an antibody, or antigen- binding fragment thereof, as defined in any of Embodiments 1 -33, or the preparation of any one of embodiments 39-40, and a pharmaceutically- acceptable carrier.
  • Embodiment 45 The antibody or antigen-binding fragment thereof for use according to Embodiment 43, or the use according to Embodiment 44, wherein the disease is selected from the group consisting of: FTD; ALS; TDP43 proteinopathies, such as AD.
  • a method of preventing or treating a disease associated with decreased PGRN levels in the brain of a patient comprising administering an effective dosage of an antibody or a fragment thereof as defined in any of Embodiments 1 -33, the preparation of any one of Embodiments 39-40, or the pharmaceutical composition of Embodiment 41 .
  • Embodiment 43 or the use according to Embodiment 44, or the method according to Embodiment 46, wherein the disease is selected from the group consisting of: FTD; ALS; or TDP43 proteinopathies, such as AD. 48.
  • Embodiments 50 The antibody, or antigen-binding fragment thereof, as defined in any of Embodiments 1 -33, the preparation of any one of Embodiments 39-40, or the pharmaceutical composition of Embodiment 41 , which is capable of specifically binding to Sortilin and inhibiting the binding of PGRN to Sortilin, but which binding does not inhibit or substantially inhibit the binding of neurotensin or AF38469 to Sortilin.
  • a kit comprising the antibody, or antigen-binding fragment thereof, as defined in any of Embodiments 1 -33, the preparation, as defined in any one of Embodiments 39-40, or the pharmaceutical composition as defined in Embodiment 41 .
  • Example 1 discloses the shuffle constructs.
  • Example 2 discloses the expression of sortilin constructs.
  • Example 3 discloses the purification of sortilin constructs.
  • Example 4 discloses the immunization and the hybridomas.
  • Example 5 discloses the sequence analysis.
  • Example 6 discloses the purification of antibodies.
  • Example 7 discloses the generation of mouse antibodies.
  • Example 8 discloses the binding to sortilin.
  • Example 9 discloses the cross blocking ability of Sortilin antibodies.
  • Example 10 discloses HTRF PGRN- sortilin binding.
  • Example 11 discloses NTS binding.
  • Example 12 discloses cellular PGRN binding and endocytosis.
  • Example 13 discloses extracellular PGRN levels.
  • Example 14 discloses iPSC PGRN levels.
  • Example 15 discloses a plasma PGRN levels.
  • Example 16 disclose epitope mapping by HDX.
  • Example 17 disclose microdialysis of PGRN in the brain.
  • Example 1 discloses the binding to sortilin.
  • Example 9 discloses the cross blocking ability of Sortilin antibodies.
  • Example 10 discloses HTRF PGRN- sortilin binding.
  • Example 11 discloses NTS binding.
  • Example 12 discloses cellular PGRN binding and endocytosis.
  • Example 13 discloses extracellular PGRN levels.
  • Example 14 discloses iPSC PGRN levels.
  • Soluble extracellular region (ECD, aa 1 - 755) constructs were tagged with either a BAP tag (biotin acceptor peptide), enabling the "in vitro" biotinylation of the proteins by co-expression of biotin ligase or a His tag, enabling easy purification.
  • Expression vectors encoding the following proteins were prepared: SORT-ECDBAP, SORT-ECDBAP-hB01 -05, SORT-ECDBAP-hB06-10, SORT-ECDBAP-hB12390, SORT-ECDBAP- hB45678, SORT-ECDBAP-tetra, SORT, SORT-tetra.
  • Sortilin sequences can be found in SEQ ID Nos:169-180 and
  • Figure 2 shows schematic presentation of the region assignment of antibodies based on binding to Sortilin shuffle constructs.
  • SORTECDHis was expressed in HEK-293F cells.
  • the His-tag in the proteins enables purification with immobilized metal affinity chromatography.
  • NiNTA Superflow Cartridge Qiagen
  • NiNTA Superflow Cartridge Qiagen
  • Column is loaded with His tagged protein with a residence time of 1 minute.
  • Column is washed with 50mM NAH 2 P0 , 300 mM NaCI and 20mM Imidazole pH 8.0.
  • Protein is eluted with 50mM NAH 2 P0 , 300 mM NaCI and 250mM Imidazole pH 8.0.
  • the protein is dialyzed to PBS using a Slide-A-Lyzer with a cut off of 10.000 mwco (Thermo Scientific). After dialyzing the protein is sterile filtered using a 0.2 micron SFCA filter(Thermo Scientific).
  • the S18-HEK cell line was generated by transfecting HEK293 cells with a human wild type (WT) sortilin expression vector. Stable transfected cells were derived after passage in the presence of a selection agent. Individual clones were selected by dilution cloning. Clones were characterized for sortilin mRNA expression using QPCR. Highest expressing clones were than analyzed by FACS (Guava, Millipore) using an anti-sortilin polyclonal antibody (Polyclonal Goat Sortilin Biotinylated Ab, Cat.No: BAF2934JR&D Systems)) to determine the surface expressed levels of Sortilin.
  • WT human wild type
  • Antibodies HuMab Sortilin were derived from the immunizations of HuMAb mouse strains HCo12, HCo17, HCo20, HCo12-BALB/c, HCo17-BALB/c and HCo20-BALB/c (human monoclonal antibody; Medarex Inc., San Jose, CA, USA), These mice are double knock out for the mouse immunoglobulin (Ig) heavy and mouse kappa light chain, which substantially inactivate the expression of antibodies that are completely murine.
  • Ig immunoglobulin
  • mice The various mouse strains were made transgenic by the insertion of human Ig heavy and human Ig kappa light chain loci and differ in the number of human VH (variable domain of heavy chain) and VL (variable domain of light chain) genes.
  • HCo12-BALB/c mice were derived by crossbreeding with KCo5-BALB/c (kappa light chain transgenic) mice.
  • mice were immunized alternating intraperitoneal ⁇ (IP) with 20 ⁇ g SORTECDHis (SEQ ID NO: 179) and subcutaneously (SC, at the tail base) with the same protein, with an interval of 14 days. A maximum of eight immunizations were performed, 4 IP and 4 SC.
  • IP intraperitoneal ⁇
  • SC subcutaneously
  • mice were additionally boosted twice intravenously (IV) with 10 ⁇ g SORTECDHis protein in 100 ⁇ _ PBS, four and three days before fusion.
  • CFA complete Freund's adjuvant
  • IFA incomplete Freund's adjuvant
  • HuMAb mice with sufficient antigen-specific titer development as defined above were sacrificed and the spleen and lymph nodes flanking the abdominal aorta and caval vein were collected. Fusion of splenocytes and lymph node cells with a mouse myeloma cell line was done by electrofusion using a CEEF 50 Electrofusion System (Cyto Pulse Sciences, Glen Burnie, MD, USA), essentially according to the manufacturer's instructions.
  • Fused cells were seeded in fusion medium containing 10% Fetal Clone I Bovine serum (Perbio), 1 mM sodium pyruvate (Cambrex), 0.5 U/mL penicillin, 0.5 U/mL streptomycin (Cambrex), 50 ⁇ 2-mercaptoethanol (Invitrogen), 600 ng/mL interleukin 6 (IL-6) (Strathmann), 1 x HAT (Sigma) and 0.5 mg/mL kanamycin (Invitrogen) in HyQ mADCF-Mab (Perbio).
  • Hybridoma cells from the best primary wells were seeded in semisolid medium made from 40% CloneMedia (Genetix, Hampshire, UK) and 60% HyQ 2x complete medium (Hyclone, Waltham, USA).
  • a well of a Genetix black 6-well plate was seeded.
  • 25 sub clones were picked, using the ClonePix system (Genetix).
  • the sub clones were picked in harvest medium.
  • the supernatants of the sub clones were screened again for Sortilin-specific human IgG binding and the human IgG concentration was measured using Octet 384red (Fortebio, Menlo Park, USA).
  • anti-sortilin HuMab antibodies of the invention were identified and subjected to sequence analysis.
  • Example 5 Sequence analysis of the Sortilin-specific HuMab variable domains and cloning in expression vectors
  • RNA was prepared from 0.2 to 5x106 hybridoma cells and 5'- RACE-Complementary DNA (cDNA) was prepared from 100 ng total RNA, using the SMART RACE cDNA Amplification kit (Clontech), according to the manufacturer's instructions.
  • VH and VL coding regions were amplified by PCR and cloned directly, in frame, in the p33G1 f and p33Kappa expression vectors (containing the human lgG1 ./ kappa constant domain encoding sequences), by ligation independent cloning (Aslanidis, C. and P.J. de Jong, Nucleic Acids Res 1990;18(20): 6069-74).
  • a synthetic gene coding for the chimeric immunogen hSortilin-FC, (human Sortilin AA (78-756) from SEQ ID NO:169) and human lgG1 -FC AA (1 04-330) from SEQ ID NO:169 was cloned into pcDNA3.1 and used for expression using the freestyle system from Invitrogen.
  • the antigen was purified from cell culture supernatants by protein-A affinity chromatography using standard procedures for antibody purification as described above for human antibodies.
  • hSortilin-FC was used as immunogen and 5 BALB/c mice were immunized. A mouse with satisfactory immune response was selected for cell fusion and hybridoma generation. Hybridoma supernatants were screened by ELISA using hSortilin-ECD as coating antigen. A total of eighteen hybridoma cell lines derived from nine parental clones were generated. Expression
  • Hybridomas were initially grown in complete growth medium, DMEM with 10%FBS+antibiotics, and subsequently adapted to CDhybridoma media (Invitrogen) for expression experiments.
  • Binding kinetics of anti-Sortilin HuMab antibodies to Sortilin were determined using Octet 384RED (Fortebio, Menlo Park, USA). HuMab solutions of 2 ⁇ g/ml were made by dilution in sample diluent (ForteBio, art. No. 18-5028). Prot A sensors (ForteBio, art.no. 18-0004) were prewetted with kinetics buffer (1 :10 sample diluent in PBS) for at least 600 seconds. Subsequently sensors were immobilized with HuMab solution for 600 seconds. A baseline response was obtained by dipping in kinetics buffer for 120 seconds.
  • Antibody cross-block studies were performed using Octet 384RED (Fortebio, Menlo Park, USA).
  • HuMab antibody solutions of 2 ⁇ g/ml were made by dilution in sample diluent (ForteBio, art. No. 18-5028).
  • Amine reactive sensors (ForteBio, art.no. 18-0008) were used for immobilization of HuMabs. Prior to coupling to amine reactive sensors, HuMabs were diluted in MES pH 6.0 buffer (18-5027). Coupling was performed at 30°C and 1000 rpm as follows: Amine reactive sensors were prewet in PBS and subsequently activated with EDC/NHS (ForteBio. Art.no.
  • Antibodies were grouped based on their binding profiles on the different Sortilin shuffle constructs ( Figure 2, Figure 3 and Figure 4). To confirm that all the antibodies from Region D (and region F) bind to the same region on human wild type Sortilin ECD, their ability to block each other's binding to the wild type human Sortilin ECD was characterised in a cross blocking study using the Octet384 red. For example, when antibodies from the same region were tested, the primary antibody would block binding of the secondary antibody and vice versa. Whereas, when antibodies from different regions were tested, there would be no cross blocking as only one region is blocked by the primary antibody and the remaining regions are available for the secondary antibody to bind.
  • Figure 7 shows that all D-region and D+ antibodies cross block each other which confirms classification of the antibodies to Region D and D+ based on shuffle constructs. Further, these data also confirm that the chimeric Sortilin constructs retain similarity to the native human wild type Sortilin ECD.
  • Example 10 Characterization of sortilin - PGRN ligand binding in the presence of anti-sortilin antibodies.
  • IC50 values for antibodies were determined by measuring the displacement of PGRN binding to Sortilin using a homogenous time resolved fluorescent (HTRF, CisBio) assay, see Figure 5 and Figure 6.
  • the assay plate was incubated for 60min at room temperature and overnight at 4 Q C before the plate was read in EnVision reader (Perkin Elmer).
  • IC50 half-maximal inhibitory concentration
  • Example 11 Characterization of sortilin -neurotensin binding in the presence of anti-sortilin antibodies
  • the IC50 for the Sortilin specific compound AF38469 was determined by measuring the displacement of 3 H-Neurotensin binding to Sortilin using a scintillation proximity assay (SPA).
  • Example 12 Characterization of sortilin - PGRN ligand binding on the surface of cells in the presence of anti-sortilin antibodies.
  • Sortilin transiently transfected cells and the stable cell line S18- HEK cells (human Sortilin over-expressing HEK cells) were used in this assay.
  • Cells were trypsinized and plated at density of 42,000 cells per well in a 96 well plate. In the case of transiently transfected cells, the cells were plated 24 hrs after transfection in 96 well plates. Next day, media was changed completely and test compounds diluted in media were added to cells for 30 min. followed by addition of PGRN for 4hr. At the end of the study (after 4.5 hrs), the cells were fixed and stained for PGRN.
  • PGRN used in the assay was harvested from media following transient transfection of PGRN expression plasmids in HEK 293 cells. PGRN levels were measured using PGRN ELISA kit (R&D).
  • PGRN added to cells was readily bound and endocytosed which led to an increased fluorescence signal in Sortilin transfected wells. Addition of neurotensin, prevented Sortilin binding to PGRN and a PGRN fluorescence intensity is similar to control levels indicating that PGRN was not bound and endocytosed in the presence of neurotensin.
  • Antibodies and control agents were added to S18-HEK cells to evaluate the effect on PGRN.
  • Addition of Neurotensin, a known peptide sortilin ligand, or human antibodies, 45, 68 and 81 1 to S18-HEK cells led to an increase of PGRN in the cell culture medium.
  • Two of the Sortilin human antibodies (45 and 68) had an effect similar to neurotensin elevating PGRN levels to 202% and 201 % respectively.
  • the antibody 81 1 increased PGRN to 146% in the medium as compared to the controls B12, an isotype control antibody was used as negative control in all our studies and did not show any effect on PGRN levels.
  • Induced pluripotent stem cells were generated by non- integrative reprogramming of human fibroblasts (Normal human dermal fibroblasts 1 8y male; Lonza) as described elsewhere (Rasmussen et al., Stem Cell Reports. 2014 Sep 9;3(3):404-13.).
  • the NHDF K1 _shp53 line was used for these studies.
  • the iPSCs were initially generated in mTESR media and subsequently cultured in monolayer in Pluripro (Cell Guidance System).
  • Neuronal differentiation was initiated day 0 by replating the cells on poly-L- ornithine/laminin coated dishes and culturing them in N3 media (50% DMEM/F12 + 50% Neurobasal media supplemented with 0.5% N2, 1 % B27 with RA, 0,5 mM GlutaMAX, 0,5& NEA, 50 ⁇ 2-mercaptoethanol and 2.5 mg/mL insulin) with 500 ng/mL noggin and 10 ⁇ SB431542. The media was refreshed every day. After 1 1 days of noggin/SB431542 induction, the cells were split with dispase and re-plated on poly-L-ornithine/laminin in N3 media. From that point forward, N3 media was refreshed every 2-3 days and the cells were split approximately every 1 0-14 day using accutase.
  • N3 media was refreshed every 2-3 days and the cells were split approximately every 1 0-14 day using accutase.
  • Neuronal differentiated iPSC cells were plated into 96 wells plate. One week later, antibodies were added to the cells. Media from the cells were collected at 48hrs or 96 hrs and analysed by human PGRN ELISA (Enzo Life sciences) and samples analysed as per the manufacturer's instructions.
  • mice were treated with antibodies (a Sortilin humab or a control ab) and sacrificed at different time points.
  • Mice treated with control antibodies did not show change in plasma PGRN whereas in mice treated with Sortilin humab 45, there was an increase in PGRN levels which seemed to peak between 24 and 48hrs and then gradually decreased from around day 4.
  • PGRN levels were still elevated at day 7.
  • mice were anaesthetized with 0,4 ml Avertin IP and heart blood was collected and transferred to a 500 ul kEDTA vial. Samples were kept on ice until centrifuged at 3600G for 15 min at 4C. The plasma was pipetted in to a micronic vial and frozen at -20C. PGRN in the samples was measured using PGRN ELISA kit (Adipogen) as per the manufacturer's instructions. Results can be seen in Figure 13.
  • Example 16 Epitope mapping of antibodies targeting the progranu- lin-sortilin interaction by hydrogen/deuterium ex- change followed by mass spectrometry
  • the protein is treated with an acid stable protease (e.g. pepsin), which generates local regions of typically ten to fifteen amino acids. Regions that shows a perturbation in the presence of a ligand is either directly involved in the binding interface or allosterically affected by the binding event.
  • an acid stable protease e.g. pepsin
  • the peptides were ionized by electrospray ionization into a mass spectrometer (Synapt G2 mass spectrometer, Waters Inc, UK), where the peptides were further separated by ion mobility before final mass determination.
  • MSe data independent
  • Determination of deuterium incorporation The acquired mass spectra were lock mass corrected against GFP and the software DynamX 3.0 (Waters Inc., USA) was used to determine the deuterium incorporation for all peptides of sortilin either in absence or presence of antibodies. [00261] A peptide was considered to be a part of the binding epitope if a protection from exchange larger than 0.5D was observed in presence of an antibody.
  • Example 17 Microdialysis to assess progranulin levels in the brain of awake freely moving animals
  • Push-pull microdialysis method was used to assess brain ISF progranulin (PRGN) from awake and freely moving mice. Mice were single- housed in controlled temperature (22 ⁇ 1 .5°C) and humidity conditions (55-65%) and kept in a 12:12 hour light/dark cycle (lights on at 06:00h). Food and water were available ad libitum. The current study was performed in the hippocampus of human sortilin knock-in (hSORTI ) mice (22 weeks old).
  • hSORTI human sortilin knock-in
  • mice were anaesthetized with isoflurane and an intracerebral guide cannula (CMA) was stereotaxically implanted into the brain, positioning the microdialysis probe in the hippocampus (co-ordinates of probe tip: 3.1 mm posterior and 2.8 mm lateral from bregma, and 1 .3 mm relative dura mater) according to the atlas of Paxinos and Franklin 2001 .
  • Acrylic cement was used for the fixation of the guide cannulas. After implantation of the cannula mice were allowed to recover from the surgery for 7 days before dialysis.
  • PRGN levels are significantly increased in animals-treated with mab #45 increased when compared to the ones from animals treated with PBS (Figure 18a).
  • PRGN levels are stable over time from 4h until 1 6h after probe insertion in hippocampus.
  • PRGN levels are elevated likely due to the probe insertion into hippocampus. It is speculated that PRGN levels are decreasing ⁇ 18h/20h after probe insertion, likely due to the clogging of the probe membrane, as it occurred in both groups (and has been previously observed in other push-pull studies).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Neurosurgery (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Endocrinology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
PCT/EP2016/066516 2015-07-13 2016-07-12 Antibodies that bind to sortilin and inhibit the binding of progranulin WO2017009327A1 (en)

Priority Applications (24)

Application Number Priority Date Filing Date Title
AU2016292980A AU2016292980B2 (en) 2015-07-13 2016-07-12 Antibodies that bind to Sortilin and inhibit the binding of progranulin
TNP/2017/000534A TN2017000534A1 (en) 2015-07-13 2016-07-12 Antibodies that bind to sortilin and inhibit the binding of progranulin
KR1020187001192A KR20180030045A (ko) 2015-07-13 2016-07-12 소르틸린에 결합하고 프로그라눌린의 결합을 억제하는 항체
EP16739087.1A EP3322726A1 (en) 2015-07-13 2016-07-12 Antibodies that bind to sortilin and inhibit the binding of progranulin
CA2989739A CA2989739A1 (en) 2015-07-13 2016-07-12 Antibodies that bind to sortilin and inhibit the binding of progranulin
JP2018501182A JP6979397B2 (ja) 2015-07-13 2016-07-12 ソルチリンに結合し、プログラニュリンの結合を阻害する抗体
UAA201800574A UA125136C2 (uk) 2015-07-13 2016-07-12 Антитіла, які зв'язуються з сортиліном і пригнічують зв'язування програнуліну
CN202210124752.3A CN114478775A (zh) 2015-07-13 2016-07-12 与分拣蛋白结合并抑制颗粒蛋白前体的结合的抗体
MX2018000506A MX2018000506A (es) 2015-07-13 2016-07-12 Agentes, usos y metodos para el tratamiento.
US15/743,549 US10479835B2 (en) 2015-07-13 2016-07-12 Agent, uses and methods for treatment
BR112018000771-4A BR112018000771A2 (pt) 2015-07-13 2016-07-12 anticorpos que se ligam à sortilina e inibem a ligação de progranulina, composições farmacêuticas, preparações dos mesmos, uso de anticorpos, kit e método de prevenção ou tratamento de uma doença associada a níveis diminuídos de pgrn
EA201890038A EA201890038A1 (ru) 2015-07-13 2016-07-12 Антитела, которые связываются с сортилином и подавляют связывание програнулина
CR20180002A CR20180002A (es) 2015-07-13 2016-07-12 Agentes, usos y métodos para el tratamiento
CN201680040476.5A CN107849135A (zh) 2015-07-13 2016-07-12 与分拣蛋白结合并抑制颗粒蛋白前体的结合的抗体
RU2018100824A RU2735639C2 (ru) 2015-07-13 2016-07-12 Антитела, которые связываются с сортилином и подавляют связывание програнулина
CN202210124751.9A CN114478774A (zh) 2015-07-13 2016-07-12 与分拣蛋白结合并抑制颗粒蛋白前体的结合的抗体
ZA2017/08613A ZA201708613B (en) 2015-07-13 2017-12-18 Antibodies that bind to sortilin and inhibit the binding of progranulin
CONC2017/0012988A CO2017012988A2 (es) 2015-07-13 2017-12-18 Agentes, usos y métodos para el tratamiento
IL256503A IL256503B (en) 2015-07-13 2017-12-22 Antibodies that bind sortilin and inhibit binding to progranulin
PH12018500100A PH12018500100A1 (en) 2015-07-13 2018-01-11 Antibodies that bind to sortilin and inhibit the binding of progranulin
HK18113432.9A HK1254356A1 (zh) 2015-07-13 2018-10-19 與分揀蛋白結合並抑制顆粒蛋白前體的結合的抗體
US16/521,279 US10889650B2 (en) 2015-07-13 2019-07-24 Agent, uses and methods for treatment
US17/097,457 US11548950B2 (en) 2015-07-13 2020-11-13 Agent, uses and methods for treatment
US18/057,104 US20230159643A1 (en) 2015-07-13 2022-11-18 Agent, uses and methods for treatment

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1512215.3A GB201512215D0 (en) 2015-07-13 2015-07-13 Agents,uses and methods
GB1512215.3 2015-07-13

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/743,549 A-371-Of-International US10479835B2 (en) 2015-07-13 2016-07-12 Agent, uses and methods for treatment
US16/521,279 Continuation US10889650B2 (en) 2015-07-13 2019-07-24 Agent, uses and methods for treatment

Publications (1)

Publication Number Publication Date
WO2017009327A1 true WO2017009327A1 (en) 2017-01-19

Family

ID=54013862

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/066516 WO2017009327A1 (en) 2015-07-13 2016-07-12 Antibodies that bind to sortilin and inhibit the binding of progranulin

Country Status (31)

Country Link
US (5) US10428147B2 (US20070167479A1-20070719-C00034.png)
EP (1) EP3322726A1 (US20070167479A1-20070719-C00034.png)
JP (2) JP6979397B2 (US20070167479A1-20070719-C00034.png)
KR (1) KR20180030045A (US20070167479A1-20070719-C00034.png)
CN (3) CN114478775A (US20070167479A1-20070719-C00034.png)
AR (1) AR105335A1 (US20070167479A1-20070719-C00034.png)
AU (1) AU2016292980B2 (US20070167479A1-20070719-C00034.png)
BR (1) BR112018000771A2 (US20070167479A1-20070719-C00034.png)
CA (1) CA2989739A1 (US20070167479A1-20070719-C00034.png)
CL (2) CL2018000092A1 (US20070167479A1-20070719-C00034.png)
CO (1) CO2017012988A2 (US20070167479A1-20070719-C00034.png)
CR (1) CR20180002A (US20070167479A1-20070719-C00034.png)
DO (1) DOP2018000014A (US20070167479A1-20070719-C00034.png)
EA (1) EA201890038A1 (US20070167479A1-20070719-C00034.png)
EC (1) ECSP18002725A (US20070167479A1-20070719-C00034.png)
GB (1) GB201512215D0 (US20070167479A1-20070719-C00034.png)
HK (1) HK1254356A1 (US20070167479A1-20070719-C00034.png)
IL (1) IL256503B (US20070167479A1-20070719-C00034.png)
JO (1) JO3710B1 (US20070167479A1-20070719-C00034.png)
MA (1) MA42440A (US20070167479A1-20070719-C00034.png)
MX (1) MX2018000506A (US20070167479A1-20070719-C00034.png)
NI (1) NI201800007A (US20070167479A1-20070719-C00034.png)
PE (1) PE20181014A1 (US20070167479A1-20070719-C00034.png)
PH (1) PH12018500100A1 (US20070167479A1-20070719-C00034.png)
RU (1) RU2735639C2 (US20070167479A1-20070719-C00034.png)
SV (1) SV2018005613A (US20070167479A1-20070719-C00034.png)
TN (1) TN2017000534A1 (US20070167479A1-20070719-C00034.png)
TW (1) TWI760305B (US20070167479A1-20070719-C00034.png)
UA (1) UA125136C2 (US20070167479A1-20070719-C00034.png)
WO (1) WO2017009327A1 (US20070167479A1-20070719-C00034.png)
ZA (1) ZA201708613B (US20070167479A1-20070719-C00034.png)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3280441A4 (en) * 2015-04-07 2018-09-12 Alector LLC Anti-sortilin antibodies and methods of use thereof
WO2019016247A3 (en) * 2017-07-20 2019-06-20 H. Lundbeck A/S Agents, uses and methods for treatment
US10428147B2 (en) 2015-07-13 2019-10-01 H. Lundbeck A/S Anti-sortilin antibodies, uses and methods for treatment
WO2020014617A1 (en) * 2018-07-13 2020-01-16 Alector Llc Anti-sortilin antibodies and methods of use thereof
US10849992B1 (en) 2015-04-07 2020-12-01 Alector Llc Methods of screening for sortilin binding antagonists
WO2023247754A1 (en) 2022-06-23 2023-12-28 Draupnir Bio Aps Bifunctional molecules that selectively induce degradation of extracellular targets in lysosomes

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010022175A1 (en) * 2008-08-19 2010-02-25 Yale University Identification of sortilin as a neuronal receptor for the frontotemporal dementia protein, progranulin

Family Cites Families (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
JPS5896026A (ja) 1981-10-30 1983-06-07 Nippon Chemiphar Co Ltd 新規ウロキナ−ゼ誘導体およびその製造法ならびにそれを含有する血栓溶解剤
US4609546A (en) 1982-06-24 1986-09-02 Japan Chemical Research Co., Ltd. Long-acting composition
US4681581A (en) 1983-12-05 1987-07-21 Coates Fredrica V Adjustable size diaper and folding method therefor
US4766106A (en) 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US5101827A (en) 1985-07-05 1992-04-07 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US4735210A (en) 1985-07-05 1988-04-05 Immunomedics, Inc. Lymphographic and organ imaging method and kit
US5776093A (en) 1985-07-05 1998-07-07 Immunomedics, Inc. Method for imaging and treating organs and tissues
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5750172A (en) 1987-06-23 1998-05-12 Pharming B.V. Transgenic non human mammal milk
US5648471A (en) 1987-12-03 1997-07-15 Centocor, Inc. One vial method for labeling antibodies with Technetium-99m
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
JP3070763B2 (ja) 1989-08-09 2000-07-31 ロメッド インコーポレイティド テクネチウムまたはレニウムでの抗体または他のタンパク質の直接放射能標識
US5633076A (en) 1989-12-01 1997-05-27 Pharming Bv Method of producing a transgenic bovine or transgenic bovine embryo
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US5101990A (en) 1990-03-23 1992-04-07 Continental Pet Technologies, Inc. Stretch blow molded oblong or oval container
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5874299A (en) 1990-08-29 1999-02-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
WO1992022645A1 (en) 1991-06-14 1992-12-23 Genpharm International, Inc. Transgenic immunodeficient non-human animals
EP1400536A1 (en) 1991-06-14 2004-03-24 Genentech Inc. Method for making humanized antibodies
EP0593592B1 (en) 1991-07-08 1998-03-25 The University Of Massachusetts At Amherst Thermotropic liquid crystal segmented block copolymer
DK0804070T3 (da) 1993-03-09 2000-08-07 Genzyme Corp Fremgangsmåde til isolering af proteiner fra mælk
EP0754225A4 (en) 1993-04-26 2001-01-31 Genpharm Int HETEROLOGIC ANTIBODY-PRODUCING TRANSGENIC NON-HUMAN ANIMALS
US5827690A (en) 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
US6077835A (en) 1994-03-23 2000-06-20 Case Western Reserve University Delivery of compacted nucleic acid to cells
KR970029803A (ko) 1995-11-03 1997-06-26 김광호 반도체 메모리장치의 프리차지 회로
CA2361421A1 (en) 1999-02-03 2000-08-10 Biosante Pharmaceuticals, Inc. Therapeutic calcium phosphate particles and methods of manufacture and use
US6281005B1 (en) 1999-05-14 2001-08-28 Copernicus Therapeutics, Inc. Automated nucleic acid compaction device
JP2003516718A (ja) 1999-07-29 2003-05-20 メダレックス インク HER2/neuに対するヒトモノクローナル抗体
MXPA02001911A (es) 1999-08-24 2003-07-21 Medarex Inc Anticuerpos ctla-4 humanos y sus usos.
ES2405944T3 (es) 2000-11-30 2013-06-04 Medarex, Inc. Ácidos nucleicos que codifican las secuencias de inmunoglobulina humana reorganizadas a partir de ratones transcromoscómicos transgénicos zadas
EP1539233B1 (en) 2001-07-12 2011-04-27 FOOTE, Jefferson Super humanized antibodies
US8066997B2 (en) 2002-12-20 2011-11-29 Anders Nykjaer Modulation of activity of neurotrophins
EP1973576B1 (en) 2005-11-28 2019-05-15 Genmab A/S Recombinant monovalent antibodies and methods for production thereof
WO2008076262A2 (en) 2006-12-15 2008-06-26 Merck & Co., Inc. Receptor for amyloid beta and uses thereof
PL3225251T3 (pl) 2006-12-21 2020-06-29 H. Lundbeck A/S Modulacja aktywności proneurotrofin
EP2185692A4 (en) 2007-08-10 2012-05-02 Medarex Inc HCO32 AND HCO27 AND RELATED EXAMPLES
EP2274327B1 (en) 2008-04-27 2019-03-27 H. Lundbeck A/S Crystal structure of human sortilin and uses thereof for identifying ligands to sortilin
WO2009154995A2 (en) 2008-05-27 2009-12-23 Kyowa Hakko Kirin Co., Ltd. Interleukin 10 receptor (il-10r) antibodies and methods of use
US8703125B2 (en) 2008-12-19 2014-04-22 H. Lundbeck A/S Modulation of the Vps10p-domain receptor family for the treatment of mental and behavioural disorders
RU2536232C2 (ru) * 2011-07-01 2014-12-20 Олег Ильич Эпштейн Лекарственное средство для лечения болезни альцгеймера и способ лечения болезни альцгеймера
US9822366B2 (en) 2012-11-02 2017-11-21 The Brigham And Women's Hospital, Inc. Sortilin 1 is a novel inducer of vascular calcification
PT3280441T (pt) 2015-04-07 2021-11-30 Alector Llc Anticorpos anti-sortilina e métodos para a sua utilização
GB201512215D0 (en) 2015-07-13 2015-08-19 Lundbeck & Co As H Agents,uses and methods
US10894833B2 (en) 2017-07-20 2021-01-19 H. Lundbeck A/S Agents, uses and methods for treatment
SI3618928T1 (sl) 2018-07-13 2023-04-28 Alector Llc Protitelesa proti sortilinu in postopki za uporabo le-teh

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010022175A1 (en) * 2008-08-19 2010-02-25 Yale University Identification of sortilin as a neuronal receptor for the frontotemporal dementia protein, progranulin

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BD TRANSDUCTION LABORATORIES: "Purified Mouse Anti-Neurotensin Receptor 3 Product Information - Technical Data Sheet", 1 January 2008 (2008-01-01), pages 1 - 2, XP055304695, Retrieved from the Internet <URL:http://www.bdbiosciences.com/ds/pm/tds/612100.pdf> [retrieved on 20160922] *
DE MUYNCK LOUIS ET AL: "The neurotrophic properties of progranulin depend on the granulin E domain but do not require sortilin binding", NEUROBIOLOGY OF AGING, vol. 34, no. 11, 24 May 2013 (2013-05-24), pages 2541 - 2547, XP028691739, ISSN: 0197-4580, DOI: 10.1016/J.NEUROBIOLAGING.2013.04.022 *
FATEMEH GHAEMIMANESH ET AL: "Production and Characterization of a Novel Monoclonal Antibody Against Human Sortilin", MONOCLONAL ANTIBODIES IN IMMUNODIAGNOSIS AND IMMUNOTHERAPY, vol. 34, no. 6, 1 December 2015 (2015-12-01), pages 390 - 395, XP055305165, ISSN: 2167-9436, DOI: 10.1089/mab.2015.0042 *
W. C. LEE ET AL: "Targeted manipulation of the sortilin-progranulin axis rescues progranulin haploinsufficiency", HUMAN MOLECULAR GENETICS, vol. 23, no. 6, 26 October 2013 (2013-10-26), pages 1467 - 1478, XP055193340, ISSN: 0964-6906, DOI: 10.1093/hmg/ddt534 *
YANQIU ZHENG ET AL: "C-Terminus of Progranulin Interacts with the Beta-Propeller Region of Sortilin to Regulate Progranulin Trafficking", PLOS ONE, vol. 6, no. 6, 15 June 2011 (2011-06-15), pages e21023, XP055305162, DOI: 10.1371/journal.pone.0021023 *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10849992B1 (en) 2015-04-07 2020-12-01 Alector Llc Methods of screening for sortilin binding antagonists
US10087255B2 (en) 2015-04-07 2018-10-02 Alector Llc Anti-sortilin antibodies and methods of use thereof
US10308718B2 (en) 2015-04-07 2019-06-04 Alector Llc Anti-sortilin antibodies and methods of use thereof
EP3991748A3 (en) * 2015-04-07 2022-08-24 Alector LLC Anti-sortilin antibodies and methods of use thereof
US10428150B2 (en) 2015-04-07 2019-10-01 Alector Llc Anti-sortilin antibodies and methods of use thereof
US11339223B2 (en) 2015-04-07 2022-05-24 Alector Llc Methods of use of anti-Sortilin antibodies for treating a disease, disorder, or injury
EP3280441A4 (en) * 2015-04-07 2018-09-12 Alector LLC Anti-sortilin antibodies and methods of use thereof
US11208488B2 (en) 2015-04-07 2021-12-28 Alector Llc Methods of increasing progranulin levels using anti-Sortilin antibodies
US11186645B2 (en) 2015-04-07 2021-11-30 Alector Llc Isolated nucleic acids encoding anti-sortilin antibodies
US10479835B2 (en) 2015-07-13 2019-11-19 H. Lundbeck A/S Agent, uses and methods for treatment
US11548950B2 (en) 2015-07-13 2023-01-10 H. Lundbeck A/S Agent, uses and methods for treatment
US10889650B2 (en) 2015-07-13 2021-01-12 H. Lundbeck A/S Agent, uses and methods for treatment
US10428147B2 (en) 2015-07-13 2019-10-01 H. Lundbeck A/S Anti-sortilin antibodies, uses and methods for treatment
CN110945029A (zh) * 2017-07-20 2020-03-31 H.隆德贝克有限公司 用于治疗的药剂、用途和方法
US10894833B2 (en) 2017-07-20 2021-01-19 H. Lundbeck A/S Agents, uses and methods for treatment
WO2019016247A3 (en) * 2017-07-20 2019-06-20 H. Lundbeck A/S Agents, uses and methods for treatment
JP2020527152A (ja) * 2017-07-20 2020-09-03 ハー・ルンドベック・アクチエゼルスカベット 治療のための薬剤、使用及び方法
KR20210030185A (ko) * 2018-07-13 2021-03-17 알렉터 엘엘씨 항-소틸린 항체 및 이들의 사용 방법
WO2020014617A1 (en) * 2018-07-13 2020-01-16 Alector Llc Anti-sortilin antibodies and methods of use thereof
US11396546B2 (en) 2018-07-13 2022-07-26 Alector Llc Anti-Sortilin antibodies and methods of use thereof
KR102530517B1 (ko) 2018-07-13 2023-05-09 알렉터 엘엘씨 항-소틸린 항체 및 이들의 사용 방법
EP4212210A1 (en) * 2018-07-13 2023-07-19 Alector LLC Anti-sortilin antibodies and methods of use thereof
TWI809147B (zh) * 2018-07-13 2023-07-21 美商阿列克特有限責任公司 抗分揀蛋白抗體及其使用方法
IL270214B1 (en) * 2018-07-13 2024-03-01 Alector Llc Anti-sortilin antibodies and methods of using them
WO2023247754A1 (en) 2022-06-23 2023-12-28 Draupnir Bio Aps Bifunctional molecules that selectively induce degradation of extracellular targets in lysosomes

Also Published As

Publication number Publication date
RU2018100824A3 (US20070167479A1-20070719-C00034.png) 2020-01-17
CO2017012988A2 (es) 2018-05-21
US11548950B2 (en) 2023-01-10
US20170267761A1 (en) 2017-09-21
AU2016292980B2 (en) 2022-10-06
JP2018529635A (ja) 2018-10-11
US20230159643A1 (en) 2023-05-25
UA125136C2 (uk) 2022-01-19
US10479835B2 (en) 2019-11-19
JP6979397B2 (ja) 2021-12-15
TWI760305B (zh) 2022-04-11
HK1254356A1 (zh) 2019-07-19
JP2022003058A (ja) 2022-01-11
NI201800007A (es) 2018-10-18
CL2018000092A1 (es) 2018-07-06
CN114478774A (zh) 2022-05-13
EA201890038A1 (ru) 2018-06-29
CL2020002468A1 (es) 2021-01-29
TN2017000534A1 (en) 2019-04-12
US20180305455A1 (en) 2018-10-25
US10889650B2 (en) 2021-01-12
GB201512215D0 (en) 2015-08-19
JO3710B1 (ar) 2021-01-31
IL256503B (en) 2022-02-01
DOP2018000014A (es) 2018-04-15
AR105335A1 (es) 2017-09-27
CN107849135A (zh) 2018-03-27
RU2018100824A (ru) 2019-08-13
KR20180030045A (ko) 2018-03-21
MA42440A (fr) 2018-05-23
EP3322726A1 (en) 2018-05-23
CA2989739A1 (en) 2017-01-19
US20210269524A1 (en) 2021-09-02
AU2016292980A1 (en) 2018-01-18
CR20180002A (es) 2018-04-09
IL256503A (en) 2018-02-28
US20200190188A1 (en) 2020-06-18
CN114478775A (zh) 2022-05-13
BR112018000771A2 (pt) 2018-09-25
ECSP18002725A (es) 2018-03-31
MX2018000506A (es) 2018-05-11
ZA201708613B (en) 2021-03-31
RU2735639C2 (ru) 2020-11-05
TW201702273A (zh) 2017-01-16
PE20181014A1 (es) 2018-06-26
SV2018005613A (es) 2018-02-23
PH12018500100A1 (en) 2018-07-30
US10428147B2 (en) 2019-10-01

Similar Documents

Publication Publication Date Title
US11542323B2 (en) Agents, uses and methods for the treatment of synucleinopathy
US11548950B2 (en) Agent, uses and methods for treatment
US20210292414A1 (en) Agents, uses and methods for treatment

Legal Events

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

Ref document number: 16739087

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2989739

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: NC2017/0012988

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 11201800115Y

Country of ref document: SG

WWE Wipo information: entry into national phase

Ref document number: CR2018-000002

Country of ref document: CR

ENP Entry into the national phase

Ref document number: 14671

Country of ref document: GE

WWE Wipo information: entry into national phase

Ref document number: 15743549

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2018-000007 I

Country of ref document: NI

Ref document number: 12018500100

Country of ref document: PH

Ref document number: 201890038

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20187001192

Country of ref document: KR

Kind code of ref document: A

Ref document number: 2018501182

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 000064-2018

Country of ref document: PE

Ref document number: MX/A/2018/000506

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2016292980

Country of ref document: AU

Date of ref document: 20160712

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201800574

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: 2018100824

Country of ref document: RU

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018000771

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112018000771

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20180112