[go: up one dir, main page]

WO2024115700A1 - Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof - Google Patents

Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof Download PDF

Info

Publication number
WO2024115700A1
WO2024115700A1 PCT/EP2023/083828 EP2023083828W WO2024115700A1 WO 2024115700 A1 WO2024115700 A1 WO 2024115700A1 EP 2023083828 W EP2023083828 W EP 2023083828W WO 2024115700 A1 WO2024115700 A1 WO 2024115700A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
antigen
binding fragment
identity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/EP2023/083828
Other languages
French (fr)
Inventor
Dimitri BIELI
Natascha WUILLEMIN
Niccolò PENGO
Ilca TUDOR
Micha BEETZ
Danaelle CAPRON
Tiziana SONATI
Alcide Barberis
Hans Mathias HESS HERMIDA
Douglas Phillips
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Mabylon AG
Original Assignee
Mabylon AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mabylon AG filed Critical Mabylon AG
Priority to AU2023401113A priority Critical patent/AU2023401113A1/en
Priority to KR1020257021663A priority patent/KR20250134595A/en
Priority to CN202380091643.9A priority patent/CN120897927A/en
Priority to EP23814496.8A priority patent/EP4626917A1/en
Publication of WO2024115700A1 publication Critical patent/WO2024115700A1/en
Priority to IL320880A priority patent/IL320880A/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • 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
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • 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
    • 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 antibodies binding to apoptosis-associated speck-like protein containing a CARD (ASC; also referred to as PYCARD), in particular to the PYRIN- PAAD-DAPIN domain (PYD) of ASC.
  • ASC apoptosis-associated speck-like protein containing a CARD
  • PYCARD PYRIN- PAAD-DAPIN domain
  • the present invention also relates to compositions and kits comprising such antibodies.
  • the present invention also relates to the use of such antibodies, compositions and kits, e.g. for preventing or treating a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
  • Neurodegenerative diseases are typically chronic, progressive disorders characterized by the gradual loss of neurons in discrete areas of the central nervous system (CNS), such as the brain. Such neuronal damage may involve progressive degeneration and death of neurons.
  • CNS central nervous system
  • AD Alzheimer's disease
  • Parkinson's disease As of today, neurodegenerative diseases are not curable. Although certain treatments may help to relieve some of the physical or mental symptoms associated with neurodegenerative diseases, it is currently not even possible to delay their progression.
  • Inflammasomes are cytosolic multiprotein complexes, which assemble in a stimulus-specific manner and, upon assembly, activate an inflammatory cascade.
  • the assembly of an inflammasome is driven by self-association and oligomerization of three types of proteins: sensor, adaptor, and effector.
  • Inflammasome sensors self-associate upon activation by danger signals and prompt the oligomerization of the inflammasome adaptor ASC (apoptosis-associated speck-like protein containing a CARD). Accordingly, inflammasome assembly usually requires ASC as adaptor protein.
  • ASC is composed of two protein-protein interaction domains: a N-terminal PYRIN-PAAD-DAPIN domain (PYD) and a C-terminal caspase-recruitment domain (CARD).
  • PYD and CARD domains are members of the six- helix bundle death domain-fold superfamily that mediates assembly of large signaling complexes in the inflammatory and apoptotic signaling pathways via the activation of caspase.
  • upstream sensor proteins such as NLRP3 and AIM2
  • recruit ASC via interactions between their PYDs.
  • ASC then interacts with caspase 1 via CARD/CARD interactions.
  • NLRP3 inhibitory compounds which target the NLRP3 sensor and are, thus, specific for NLRP3 inflammasomes, but not other inflammasomes (Voet S, Srinivasan S, Lamkanfi M, van Loo G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol Med. 2019 Jun;1 1(6):e10248. doi: 10.15252/emmm.201810248).
  • the object of the present invention to overcome the drawbacks of the prior art.
  • x means x ⁇ 10%, for example, x ⁇ 5%, or x ⁇ 7%, or x + 10%, or x ⁇ 12%, or x + 15%, or x + 20%.
  • disease as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
  • treatment of a subject or patient is intended to include prevention, prophylaxis, attenuation, amelioration and therapy.
  • subject or patient are used interchangeably herein to mean all mammals including humans. Examples of subjects include humans, cows, dogs, cats, horses, goats, sheep, pigs, and rabbits. Preferably, the subject or patient is a human.
  • a dose which is expressed as [g, mg, or other unit]/kg (or g, mg etc.) usually refers to [g, mg, or other unit] "per kg (or g, mg etc.) bodyweight", even if the term "bodyweight” is not explicitly mentioned.
  • binding and similar reference usually means “specifically binding”, which does not encompass non-specific sticking.
  • specific binding of an antibody means that the antibody recognizes its target antigen and binds its target with greater affinity (or at lower antibody concentrations, e.g. EC50) than it does to a structurally different antigen and/or to an antigen with a modified or mutated sequence.
  • a "greater” affinity may be at least 2fold, 3fold, 4fold, 5fold, 10fold, 15fold, 20fold, 25fold, 50fold, 75fold, 100fold 150fold, 200fold, 500fold, 750fold, 1 ,000fold, 1 ,500fold, 2,000fold, 5,000fold, 7,500fold, 10,000fold or even higher affinity as compared to the binding to a control antigen.
  • antibody-binding to the control antigen may be undetectable (below detection threshold), while antibody-binding to the specific antigen may be well detected/determined.
  • the term "antibody” encompasses various forms of antibodies including, without being limited to, whole antibodies, antibody fragments (such as antigen binding fragments), human antibodies, chimeric antibodies, humanized antibodies, recombinant antibodies and genetically engineered antibodies (e.g., variant or mutant antibodies) as long as the characteristic properties according to the invention are retained.
  • the antibody is a humanized antibody.
  • the antibody is a monoclonal antibody.
  • the antibody may be a humanized monoclonal antibody.
  • antibody generally also includes antibody fragments. Fragments of the antibodies may retain the antigen-binding activity of the antibodies. Such fragments are referred to as "antigen-binding fragments". Antigen-binding fragments include, but are not limited to, single chain antibodies, Fab, Fab', F(ab')2, Fv or scFv. Fragments of the antibodies can be obtained from the antibodies by methods that include digestion with enzymes, such as pepsin or papain, and/or by cleavage of disulfide bonds by chemical reduction.
  • enzymes such as pepsin or papain
  • fragments of the antibodies can be obtained by recombinant means, for example by cloning and expressing a part (fragment) of the sequences of the heavy and/or light chain.
  • the invention also encompasses single-chain Fv fragments (scFv) derived from the heavy and light chains of an antibody of the invention.
  • the invention includes a scFv comprising the CDRs from an antibody of the invention.
  • heavy or light chain monomers and dimers single domain heavy chain antibodies, single domain light chain antibodies, as well as single chain antibodies, e.g., single chain Fv in which the heavy and light chain variable domains are joined by a peptide linker.
  • Antibody fragments of the invention may be contained in a variety of structures known to the person skilled in the art.
  • the sequences of the invention may be a component of multispecific molecules in which the sequences of the invention target the epitopes of the invention and other regions of the molecule bind to other targets.
  • the specification, including the claims may, in some places, refer explicitly to antigen binding fragment(s), antibody fragment(s), variant(s) and/or derivative(s) of antibodies, it is understood that the term "antibody” includes all categories of antibodies, namely, antigen binding fragment(s), antibody fragment(s), variant(s) and derivative(s) of antibodies.
  • variable region (variable region of a light chain (V L ), variable region of a heavy chain (V H )) denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen.
  • Antibodies of the invention can be of any isotype e.g., IgA, IgG, IgM i.e. an a, y or heavy chain).
  • the antibody is of the IgG type or the IgA type.
  • antibodies may be IgGI , lgG2, lgG3 or lgG4 subclass, preferably lgG1 or lgG4.
  • Antibodies of the invention may have a K or a A light chain.
  • Antibodies according to the present invention may be provided in purified form. Typically, the antibody will be present in a composition that is substantially free of other polypeptides e.g., where less than 90% (by weight), usually less than 60% and more usually less than 50% of the composition is made up of other polypeptides.
  • Antibodies according to the present invention may be immunogenic in human and/or in non-human (or heterologous) hosts e.g., in mice.
  • the antibodies may have an idiotope that is immunogenic in non-human hosts, but not in a human host.
  • Antibodies of the invention for human use include those that cannot be easily isolated from hosts such as mice, goats, rabbits, rats, non-primate mammals, etc. and cannot generally be obtained by humanization or from xeno-mice.
  • the term "antigen” refers to any structural substance which serves as a target for the receptors of an adaptive immune response, in particular as a target for antibodies, T cell receptors, and/or B cell receptors.
  • An “epitope”, also known as “antigenic determinant”, is the part (or fragment) of an antigen that is recognized by the immune system, in particular by antibodies, T cell receptors, and/or B cell receptors.
  • one antigen has at least one epitope, i.e. a single antigen has one or more epitopes.
  • An antigen may be (i) a peptide, a polypeptide, or a protein, (ii) a polysaccharide, (iii) a lipid, (iv) a lipoprotein or a lipopeptide, (v) a glycolipid, (vi) a nucleic acid, or (vii) a small molecule drug or a toxin.
  • an antigen may be a peptide, a protein, a polysaccharide, a lipid, a combination thereof including lipoproteins and glycolipids, a nucleic acid (e.g.
  • the antigen is selected from (i) a peptide, a polypeptide, or a protein, (ii) a polysaccharide, (iii) a lipid, (iv) a lipoprotein or a lipopeptide and (v) a glycolipid; more preferably, the antigen is a peptide, a polypeptide, or a protein.
  • mutation relates to a change in the nucleic acid sequence and/or in the amino acid sequence in comparison to a reference sequence, e.g. a corresponding genomic sequence.
  • a mutation e.g. in comparison to a genomic sequence, may be, for example, a (naturally occurring) somatic mutation, a spontaneous mutation, an induced mutation, e.g. induced by enzymes, chemicals or radiation, or a mutation obtained by site- directed mutagenesis (molecular biology methods for making specific and intentional changes in the nucleic acid sequence and/or in the amino acid sequence).
  • mutation or “mutating” shall be understood to also include physically making a mutation, e.g.
  • a mutation includes substitution, deletion and insertion of one or more nucleotides or amino acids as well as inversion of several successive nucleotides or amino acids.
  • a mutation may be introduced into the nucleotide sequence encoding said amino acid sequence in order to express a (recombinant) mutated polypeptide.
  • a mutation may be achieved e.g., by altering, e.g., by site-directed mutagenesis, a codon of a nucleic acid molecule encoding one amino acid to result in a codon encoding a different amino acid, or by synthesizing a sequence variant, e.g., by knowing the nucleotide sequence of a nucleic acid molecule encoding a polypeptide and by designing the synthesis of a nucleic acid molecule comprising a nucleotide sequence encoding a variant of the polypeptide without the need for mutating one or more nucleotides of a nucleic acid molecule.
  • sequence variant refers to any alteration in comparison to a reference sequence.
  • sequence variant includes nucleotide sequence variants and amino acid sequence variants.
  • a reference sequence is any of the sequences listed in the "Table of Sequences and SEQ ID Numbers" (Sequence listing), i.e. SEQ ID NO: 1 to SEQ ID NO: 199.
  • a "sequence variant” shares (over the whole length of the sequence) at least 70% or at least 75%, preferably at least 80% or at least 85%, more preferably at least 90% or at least 93%, even more preferably at least 95% or at least 96%, still more preferably at least 97% or at least 98%, particularly preferably at least 99% sequence identity with its reference sequence.
  • the "sequence variant" shares at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity.
  • sequence identity In general, i.e.
  • %-identity in a sequence variant, the more it is preferred.
  • a sequence variant having at least 84% sequence identity with a reference sequence is more preferred than a sequence variant having at least 75% sequence identity with the reference sequence.
  • sequence variant maintains the (biological) function of the reference sequence.
  • sequence variants relating to antibodies of the invention preferably maintain the specific binding to ASC, in particular to the PYD domain of ASC.
  • other functions such as anti-ASC-fibrillation activity and/or reducing or blocking of ASC-Ap interaction may also be maintained.
  • Sequence identity may be calculated as described below.
  • a sequence variant may preserve the specific function of the reference sequence.
  • an amino acid sequence variant has an altered sequence in which one or more (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) of the amino acids in the reference sequence is deleted or substituted, or one or more (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) amino acids are inserted into or added to the sequence of the reference amino acid sequence.
  • the amino acid sequence variant has an amino acid sequence which is at least 70% or at least 75%, preferably at least 80% or at least 85%, more preferably at least 90% or at least 93%, even more preferably at least 95% or at least 96%, still more preferably at least 97% or at least 98%, particularly preferably at least 99% identical to the reference sequence.
  • variant sequences which are at least 90% identical have no more than 10 alterations, i.e., any combination of deletions, insertions or substitutions, per 100 amino acids of the reference sequence.
  • the "% identity" of the sequence variant is usually determined with respect to the reference sequence.
  • the substitutions are preferably conservative amino acid substitutions, wherein the substituted amino acid has similar structural or chemical properties with the corresponding amino acid in the reference sequence.
  • conservative amino acid substitutions involve substitution of one aliphatic or hydrophobic amino acids, e.g. alanine, valine, leucine and isoleucine, with another; substitution of one hydoxyl-containing amino acid, e.g. serine and threonine, with another; substitution of one acidic residue, e.g. glutamic acid or aspartic acid, with another; replacement of one amide-containing residue, e.g.
  • asparagine and glutamine with another; replacement of one aromatic residue, e.g. phenylalanine and tyrosine, with another; replacement of one basic residue, e.g. lysine, arginine and histidine, with another; and replacement of one small amino acid, e.g., alanine, serine, threonine, cysteine, and glycine, with another.
  • one aromatic residue e.g. phenylalanine and tyrosine
  • basic residue e.g. lysine, arginine and histidine
  • replacement of one small amino acid e.g., alanine, serine, threonine, cysteine, and glycine
  • the present invention provides an (isolated) antibody, or an antigen-binding fragment thereof, which (specifically) binds to ASC, in particular to the PYD domain of ASC.
  • the ASC is human ASC (SEQ ID NO: 1 ) and the PYD domain is the PYD domain (SEQ ID NO: 143) of human ASC.
  • Apoptosis-associated speck-like protein containing a CARD (ASC; also referred to as PYCARD) is an adaptor protein that is composed of two protein-protein interaction domains: PYRIN-PAAD-DAPIN domain (PYD) and a caspaserecruitment domain (CARD).
  • the PYD and CARD domains are members of the six-helix bundle death domain-fold superfamily that mediates assembly of large signaling complexes in the inflammatory and apoptotic signaling pathways via the activation of caspase.
  • the antibody, or the antigen-binding fragment thereof, of the present invention binds specifically to the PYD domain of ASC.
  • the antibody, or the antigenbinding fragment thereof, of the present invention binds in particular to a polypeptide or protein having an amino acid sequence according to SEQ ID NO: 1 (human ASC), in particular to SEQ ID NO: 143 (PYD of human ASC).
  • the antibody, or an antigen-binding fragment thereof, of the present invention does preferably not bind (specifically) to the CARD domain (SEQ ID NO: 144) of ASC.
  • the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to monomeric ASC. It is also preferred that the antibody, or the antigenbinding fragment thereof, of the present invention binds (specifically) to ASC specks.
  • ASC can assemble into a large protein complex, which is termed "speck”.
  • ASC specks are structures formed by ASC, which can reach a size of around 1 pm and which are characteristic of inflammasome activation. More preferably, the antibody, or the antigenbinding fragment thereof, of the present invention binds (specifically) to monomeric ASC and to ASC specks.
  • ASC may be of any origin, for example from the species from which it is originally derived from (e.g.
  • human ASC may be of human origin; murine ASC may be of murine origin and the like), it may be recombinantly expressed or a synthetic ASC peptide.
  • the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to physiological/endogenous ASC, in particular to human and/or murine physiological/endogenous ASC, e.g. physiological/endogenous ASC of human macrophages and/or of mouse bone-marrow derived macrophages (BMDMs).
  • BMDMs mouse bone-marrow derived macrophages
  • Binding to physiological/endogenous ASC may be tested by using cell lysates of human macrophages (such as THP-1 cells) and/or of mouse bone-marrow derived macrophages (BMDMs), e.g. as described in the appended examples.
  • human macrophages such as THP-1 cells
  • BMDMs mouse bone-marrow derived macrophages
  • ASC can occur in four different isoforms, namely (1 ) ASC (or “full-length ASC”; SEQ ID NO: 1 ); (2) ASC-b (SEQ ID NO: 145); (3) ASC-c (SEQ ID NO: 146) and (4) ASC-d (SEQ ID NO: 147).
  • the antibody, or the antigen-binding fragment thereof, of the present invention (specifically) binds to (human) ASC and (human) ASC-b.
  • the antibody, or the antigen-binding fragment thereof, of the present invention does preferably not (specifically) bind to (human) ASC-c and to (human) ASC-d.
  • the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to ASC K21 A K22A K26A (mutated ASC with K21 A, K22A and K26A mutations). Furthermore, it is preferred that the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to ASC Y146A (mutated ASC with Y146A mutation).
  • the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to a fragment or a region of human ASC, which includes amino acids 36 to 85 of human ASC (SEQ ID NO: 1 ), more preferably to a fragment or a region of human ASC, which includes amino acids 41 to 85 of human ASC (SEQ ID NO: 1 ). Accordingly, the antibody, or the antigen-binding fragment thereof, of the present invention does preferably not bind (specifically) to a region (or fragment) of ASC comprised in (or corresponding to) amino acids 1 to 35 of ASC (SEQ ID NO: 1 ).
  • the antibody, or the antigen-binding fragment thereof, of the present invention does not (specifically) bind to the ASC peptide according to SEQ ID NO: 148 (ASC aa 1 -20 peptide). Additionally (or alternatively), the antibody, or the antigen-binding fragment thereof, of the present invention may (also) not (specifically) bind to the ASC peptide according to SEQ ID NO: 149 (ASC aa 1 1 -30 peptide). In some embodiments, the antibody, or the antigenbinding fragment thereof, of the present invention does not (specifically) bind to the ASC peptide according to SEQ ID NO: 150 (ASC aa 21 -40 peptide).
  • the antibody, or the antigen-binding fragment thereof, of the present invention does preferably not bind (specifically) to a region (or fragment) of ASC comprised in (or corresponding to) amino acids 86 to 195 of ASC (SEQ ID NO: 1 ).
  • the epitope in the PYD of human ASC, to which the antibody, or the antigen-binding fragment thereof, of the present invention binds to is preferably located within amino acids 36 to 85 of ASC (SEQ ID NO: 1 ), more preferably within amino acids 41 to 85 of ASC (SEQ ID NO: 1 ).
  • the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to ASC of different species. More preferably, the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to human ASC (SEQ ID NO: 1 ) and murine ASC (SEQ ID NO: 151 ).
  • Standard methods to assess binding of the antibody according to the present invention, or the antigen-binding fragment thereof, are known to those skilled in the art and include, for example, ELISA (enzyme-linked immunosorbent assay). Thereby, the relative affinities of antibody binding may be determined by measuring the concentration of the antibody (EC 50 ) required to achieve 50% maximal binding at saturation.
  • ELISA enzyme-linked immunosorbent assay
  • the antibody, or an antigen-binding fragment thereof, according to the present invention may comprise three heavy chain complementarity determining regions (CDRs) and three light chain CDRs.
  • complementarity determining regions (CDRs) are the hypervariable regions present in the heavy chain variable domain and in the light chain variable domains.
  • the CDRs of a heavy chain and the connected light chain of an antibody together form the antigen receptor.
  • the three CDRs (CDR1 , CDR2, and CDR3) are arranged non-consecutively in the variable domain. Since antigen receptors are typically composed of two variable domains (on two different polypeptide chains, i.e.
  • heavy and light chain heavy chain variable region (VH) and light chain variable region (VL)
  • CDRs for each antigen receptor
  • the CDRs in the VH and VL are usually separated by framework regions, whereby a framework region (FR) is a region in the variable domain which is less "variable” than the CDR.
  • FR framework region
  • the sequences of the heavy chains and light chains of exemplary antibodies of the invention, comprising three different CDRs on the heavy chain and three different CDRs on the light chain were determined.
  • ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC causalAb004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 are shown in Table 1 below.
  • the Rabat CDR definition was applied (Tai Te Wu, Elvin A. Rabat; An analysis of the sequences of the variable regions of Bence Jones proteins and myeloma light chains and their implications for antibody complementarity. J Exp Med 1 August 1970; 132 (2): 211 -250; George Johnson, Tai Te Wu, Rabat Database and its applications: 30 years after the first variability plot, Nucleic Acids Research, Volume 28, Issue 1 , 1 January 2000, Pages 214-218).
  • Table 1 SEQ ID NOs for CDR and VH/VL sequences of exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_AbOO5, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_Ab009.
  • the antibody of the invention comprises a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of the specific combinations of the specific VH and VL sequences of exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 shown in Table 1 .
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 8 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 9.
  • the CDRs in the variable regions of SEQ ID NOs 8 and 9 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 8 and of a VL according to SEQ ID NO: 9, wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC, exhibits anti-ASC-fibri National activity, blocks ASC-Ap interaction and blocks the pro-inflammatory function of extracellular ASC fibrils.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 8 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 74%, at least 75%, at least 76%, at least
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 8 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 9.
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 15 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 16.
  • the CDRs in the variable regions of SEQ ID NOs 15 and 16 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 10, and SEQ ID NO: 11 , respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 15 and of a VL according to SEQ ID NO: 16, wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC and blocks ASC-Ap interaction.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 15 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 74%, at least 75%, at least 76%, at least
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 15 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 16.
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 22 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 23.
  • the CDRs in the variable regions of SEQ ID NOs 22 and 23 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 17, and SEQ ID NO: 18, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 22 and of a VL according to SEQ ID NO: 23, wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC and blocks ASC-Ap interaction.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 22 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 74%, at least 75%, at least 76%, at least
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 22 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 23.
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 30 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 31 .
  • the CDRs in the variable regions of SEQ ID NOs 30 and 31 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 24, SEQ ID NO: 25, and SEQ ID NO: 26, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 30 and of a VL according to SEQ ID NO: 31 , wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC, exhibits anti-ASC-fibri I lation activity and blocks ASC-A interaction.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 30 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 74%, at least 75%, at least 76%, at least
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 30 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 31 .
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 38 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 39.
  • the CDRs in the variable regions of SEQ ID NOs 38 and 39 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 38 and of a VL according to SEQ ID NO: 39, wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC and blocks ASC-AJ3 interaction.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 38 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 74%, at least 75%, at least 76%, at least
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 38 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 39.
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 46 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 47.
  • the CDRs in the variable regions of SEQ ID NOs 46 and 47 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 40, SEQ ID NO: 41 , and SEQ ID NO: 42, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 46 and of a VL according to SEQ ID NO: 47, wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC and blocks ASC-Ap interaction.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 46 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 74%, at least 75%, at least 76%, at least
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 46 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 47.
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 54 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 55.
  • the CDRs in the variable regions of SEQ ID NOs 54 and 55 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 48, SEQ ID NO: 49, and SEQ ID NO: 50, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 54 and of a VL according to SEQ ID NO: 55, wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC and exhibits anti-ASC-fibrillation activity.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 54 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 74%, at least 75%, at least 76%, at least
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 54 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 55.
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 61 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 62.
  • the CDRs in the variable regions of SEQ ID NOs 61 and 62 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 56, SEQ ID NO: 57, and SEQ ID NO: 58, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 61 and of a VL according to SEQ ID NO: 62, wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC and exhibits anti-ASC-fibrillation activity.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 61 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 61 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 62.
  • the antibody of the invention comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 68 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 69.
  • the CDRs in the variable regions of SEQ ID NOs 68 and 69 may be determined by different numbering schemes known in the art, such as Rabat, Chothia, EU, IMGT, Paratome and AHo numbering.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 63, SEQ ID NO: 64, and SEQ ID NO: 65, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively.
  • CDR sequences were determined as described above.
  • the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 68 and of a VL according to SEQ ID NO: 69, wherein the CDRs are determined by a distinct method.
  • such an antibody binds specifically to the PYD domain of human ASC and exhibits anti-ASC-fibri National activity.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 68 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at
  • the CDR sequences as defined above are preferably maintained.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 68 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 69.
  • the antibody, or the antigen-binding fragment thereof, of the invention may exhibit anti- ASC-fibri I lation activity.
  • Antibodies reducing or inhibiting ASC fibrillation provide a way to block either de novo or seeded ASC assembly, and thus the downstream effect of ASC speck formation such as inflammation and/or amyloid seeding.
  • Anti-fibrillation activity may be tested in a fibrillation assay or in a sedimentation assay, e.g. as described in the appended examples. Accordingly, preferred fibrillation assay is described in the examples of the present specification. Briefly, ASC assembles in large cytoplasmic macromolecular assemblies called ASC specks.
  • ASC e.g. including a marker such as green fluorescent protein (GFP)
  • GFP green fluorescent protein
  • Protein aggregates of the antibody to be tested may be compared to those obtained with controls, such as ASC only and an unrelated antibody (which are both not expected to decrease ASC fibrillation). If significantly less fibrillation is observed for the antibody to be tested as compared to an unrelated antibody, the antibody to be tested exhibits anti-fibrillation activity.
  • the antibody, or the antigen-binding fragment thereof, of the invention may reduce or block ASC-Ap interaction.
  • ASC-Ap interaction is assumed to be at the basis of increased Ap aggregation and amyloid deposition in Alzheimer disease (Venegas C, Kumar S, Franklin BS, Dierkes T, Brinkschulte R, Tejera D, Vieira-Saecker A, Schwartz S, Santarelli F, Kummer MP, Griep A, Gelpi E, Beilharz M, Riedel D, Golenbock DT, Geyer M, Walter J, Latz E, Heneka MT. Microglia-derived ASC specks cross-seed amyloid-P in Alzheimer's disease. Nature.
  • ASC fibrils may be formed by incubating ASC monomers, e.g. at about 37°C for about 1 h. Fibrillary ASC may then be incubated with Ap, e.g. including a marker such as TAMRA ((5- Carboxy)tetramethylrhodamine), in the presence of the antibody to be tested (and appropriate controls as described below), e.g. at about 37°C for about 4h, and protein aggregates may be determined in view of the marker used (e.g. by flow cytometry in case of TAMRA).
  • Ap e.g. including a marker such as TAMRA ((5- Carboxy)tetramethylrhodamine)
  • Protein aggregates obtained after incubation of ASC+Ap+anti-ASC antibodies may be compared to those obtained with controls, such as ASC only, ASC+Ap and an unrelated antibody.
  • ASC-Ap inhibition may be determined as percentage in view the binding (aggregation) observed for the ASC+Ap (without antibody) control and the ASC only control. While no inhibition (0%) is assumed for the ASC+AP control, 100% inhibition is assumed for the ASC only group (because no interaction (aggregation) is expected for ASC without Ap).
  • the %-inhibition of ASC-Ap interaction for the antibody to be tested may thus be calculated in view thereof and compared to the effects of an unrelated antibody to determine whether or not significant inhibition of ASC-Ap interaction is observed.
  • the antibody, or the antigen-binding fragment thereof, of the invention may reduce or block Ap aggregation. In some embodiments, the antibody, or the antigen-binding fragment thereof, of the invention may reduce or block protein aggregation, in particular in the context of inflammatory processes. Without being bound to any theory, it is assumed that ASC acts as a "catalyzer" on the aggregation of proteins, such as Ap. Therefore, antibodies reducing or blocking the interaction of ASC with such (aggregating) proteins - in particular by binding to ASC - are assumed to reduce or block the aggregation of proteins (which is otherwise catalyzed by ASC).
  • the antibody, or the antigen-binding fragment thereof, of the invention may reduce or block the pro-inflammatory function of extracellular ASC fibrils. It has been shown that ASC fibrils and ASC specks can propagate inflammation (Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmuller W, Latz E.
  • the adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation.
  • the NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014 Aug;15(8):738-48.
  • THP-1 efficacy assay may be used.
  • a preferred THP-1 efficacy assay is described in the examples of the present specification. Briefly, wt and asc THP-1 monocytes may be seeded, e.g. at a density of about 10'000 cells/well, in a multi-well plate. Monocytes may then be differentiated into macrophages, e.g. by incubation in 100 ng/ml phorbol 12-myristate-13- acetate (PMA)-containing medium, e.g. for about 4 days at about 37°C and about 5% CO 2 .
  • PMA phorbol 12-myristate-13- acetate
  • ASC fibrils may be prepared by incubating ASC monomers, e.g. at about 37°C for about 1 h. Before addition of the ASC fibrils to the cells, they may be pre-incubated with the antibody to be tested, e.g. for about 15 min at about 22°C. After incubation (e.g. about 16 h at about 37°C and about 5% CO 2 ), secreted IL-1 P may be measured, e.g. by Lumit Human IL-1 Immunoassay (Promega, W6010). The amount of IL-1 secretion may be compared to that obtained when an unrelated control antibody was used in the same assay in wt THP-1 cells.
  • the antibody, or the antigen-binding fragment thereof is a humanized antibody.
  • Methods for humanization of antibodies are well-known in the art.
  • a humanized antibody is engineered to comprise one or more human framework regions in the variable region together with non-human (e.g., rabbit) CDRs of the heavy and/or light chain.
  • a humanized antibody comprises sequences that are entirely human except for the CDR regions.
  • a humanized antibody may comprise residues that are found neither in the human form of the antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance.
  • a humanized antibody comprises a VH and a VL, in which all or essentially all of the CDR regions correspond to those of a parental non-human (e.g. rabbit) antibody and all or essentially all of the FR regions are those of a human antibody or a human immunoglobulin consensus sequence.
  • the FR region can be modified in any manner known in the art and/or provided herein.
  • the humanized antibody may also comprise a human Fc moiety.
  • Humanized antibodies preferably exhibit reduced immunogenicity when introduced into the human body.
  • the scatteredHumanized” antibodies may be prepared by creating a predominantlychimeric" antibody (non-human Fab grafted onto human Fc) as an initial step and selective mutation of the (non-CDR) amino acids in the Fab portion of the molecule.
  • Alternatively, handheldhumanized” antibodies can be obtain directly by grafting appropriate drapedonor" CDR coding segments derived from a non-human animal onto a human antibody traversacceptor” scaffold, and optionally mutating (non-CDR) amino acids for optimized binding.
  • the antibody, or the antigen-binding fragment thereof is a monoclonal antibody.
  • the antibody, or the antigen-binding fragment thereof may be a humanized monoclonal antibody.
  • variable regions or the CDRs of the antibody as defined herein are derived from a rabbit antibody and grafted in a scaffold of a human IgG or IgA antibody. Accordingly, the variable regions, portions thereof or the CDRs may be rabbit and grafted in an antibody framework, which is preferably of human origin. Typically, the rabbit-derived portions of the variable regions that are grafted into the antibody framework comprise the CDRs.
  • IgG IgGI and lgG4 are preferred.
  • the skilled person may use the (rabbit) VH/VL sequences of the exemplary antibodies, as described in Table 1 , and determine "extended" rabbit CDRs in said VH/VL sequences of Table 1 by combining two or more (e.g., 3, 4, 5 or 6) different antibody numbering schemes (e.g., selected from Kabat, Chothia, EU, IMGT, Paratome and AHo), such that each "extended” CDR covers the respective CDRs as defined in each of the combined numbering schemes.
  • Such "extended" CDRs determined by combining different numbering schemes may, thus, include amino acid residues of the framework region adjacent to a CDR (in particular when a single numbering scheme is used).
  • an "extended" CDR may correspond to the CDR as determined by a single numbering scheme. This applies in particular, if the CDR as determined by the single numbering scheme contains (or corresponds to) the corresponding CDR determined by one or more other numbering schemes.
  • Kabat, IMGT and Paratome may be used to obtain extended (rabbit) CDR sequences (based on the VH/VL sequences of the exemplified antibodies of Table 1 above), as described in Zhang and Ho, 2017 (Zhang YF, Ho M. Humanization of rabbit monoclonal antibodies via grafting combined Kabat/IMGT/Paratome complementarity-determining regions: Rationale and examples. MAbs. 2017 Apr;9(3):419-429. doi: 10.1080/19420862.2017.1289302). Extended rabbit CDRs may then be grafted onto human (germlined) IgG framework regions.
  • Table 2 SEQ ID NOs of exemplary extended (rabbit) CDRs of the exemplary antibodies ASC_Ab001 (two versions of extended CDRs), ASC_AbOO2, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_AbOO9 Specific examples of VH/VL sequences of humanized versions of the exemplified antibodies are shown in Table 3 below.
  • Table 3 Exemplary humanized VH/VL sequences of the exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 and human germlined V and J gene versions suitable for each of these antibodies.
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2, an extended CDRH3, an extended CDRL1 , an extended CDRL2 and an extended CDRL3 of exemplary antibodies ASC_Ab001 , ASC_AbOO2, ASC_Ab003, ASC_Ab004, ASC_AbOO5, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 shown in Table 2.
  • extended CDRs may include amino acid residues of the framework region adjacent to the CDRs shown in Table
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 159, SEQ ID NO: 3, and SEQ ID NO: 160, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively.
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 194, SEQ ID NO: 195, and SEQ ID NO: 196, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 197, SEQ ID NO: 198, and SEQ ID NO: 199, respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 66, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -5 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 159, SEQ ID NO: 3, and SEQ ID NO: 160, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 194, SEQ ID NO: 195, and SEQ ID NO: 196, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 197, SEQ ID NO: 198, and SEQ ID NO: 199, respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 57 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %,
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 1 57 and in the sequence variant of SEQ ID NO: 158, respectively.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 57 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 58.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 176 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 176 and in the sequence variant of SEQ ID NO: 177, respectively.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 176 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 177.
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 161 , SEQ ID NO: 10, and SEQ ID NO: 162, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 1 0, and SEQ ID NO: 1 1 , respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 23, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -1 6 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH 1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 61 , SEQ ID NO: 10, and SEQ ID NO: 1 62, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 12, SEQ ID NO: 1 , and SEQ ID NO: 14, respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 78 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %,
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 178 and the sequence variant of SEQ ID NO: 179.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 78 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 161 , SEQ ID NO: 17, and SEQ ID NO: 163, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 17, and SEQ ID NO: 18, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 66, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -5 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 161 , SEQ ID NO: 17, and SEQ ID NO: 163, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 180 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 74%, at least 75%, at least 76%, at least
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 180 and the sequence variant of SEQ ID NO: 181 .
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 180 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 181 .
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 164, SEQ ID NO: 25, and SEQ ID NO: 165, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 24, SEQ ID NO: 25, and SEQ ID NO: 26, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 66, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -6 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 64, SEQ ID NO: 25, and SEQ ID NO: 1 65, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 182 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 182 and the sequence variant of SEQ ID NO: 183.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 182 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 183.
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 166, SEQ ID NO: 33, and SEQ ID NO: 167, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 33, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -5 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 166, SEQ ID NO: 33, and SEQ ID NO: 167, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 184 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 184 and the sequence variant of SEQ ID NO: 185.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 184 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 185.
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 168, SEQ ID NO: 41 , and SEQ ID NO: 169, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 40, SEQ ID NO: 41 , and SEQ ID NO: 42, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 23, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -27 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 168, SEQ ID NO: 41 , and SEQ ID NO: 1 69, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 186 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 1 86 and the sequence variant of SEQ ID NO: 1 87.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 86 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 187.
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 70, SEQ ID NO: 49, and SEQ ID NO: 1 71 , respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 48, SEQ ID NO: 49, and SEQ ID NO: 50, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 1 5, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -8 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 70, SEQ ID NO: 49, and SEQ ID NO: 1 71 , respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 88 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %,
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 188 and the sequence variant of SEQ ID NO: 189.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 188 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 189.
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 172, SEQ ID NO: 57, and SEQ ID NO: 173, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 56, SEQ ID NO: 57, and SEQ ID NO: 58, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 33, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -12 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRE11 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 172, SEQ ID NO: 57, and SEQ ID NO: 173, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 190 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 190 and the sequence variant of SEQ ID NO: 1 91 .
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 90 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 91 .
  • the antibody of the invention comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 74, SEQ ID NO: 64, and SEQ ID NO: 1 75, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively.
  • the antibody of the invention comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 63, SEQ ID NO: 64, and SEQ ID NO: 65, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 23, the heavy chain J gene IGHJ6, the light chain V gene 1GKV1 -27 and the light chain J gene IGKJ3 are used to obtain the humanized framework regions.
  • such an antibody or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 74, SEQ ID NO: 64, and SEQ ID NO: 1 75, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively.
  • such an antibody of the invention may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 192 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at
  • the CDR sequences as defined above are preferably maintained.
  • the extended CDR sequences as defined above are maintained in the sequence variant of SEQ ID NO: 192 and the sequence variant of SEQ ID NO: 193.
  • the antibody, or an antigen-binding fragment thereof comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 192 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 193.
  • humanization may include back mutations, i.e. wherein amino acids of the human framework sequences are mutated back to the corresponding amino acids of the rabbit framework sequences, in particular if the result of grafting of the (extended) CDRs onto human IgG framework regions is not satisfactory.
  • the goal of humanization is always to maintain as many human sequences/amino acids as possible.
  • the amino acid residues of the vernier zones (vernier zone residues) of the human VH and VL frame work regions may be back mutated to the corresponding rabbit amino acids (of the VH/VL regions as shown in Table 1 ).
  • the first two amino acid residues in the light chain may be back mutated (from human to rabbit), if required.
  • the first 1 , 2 or 3 residues at the N terminus of the HV4/LV4 loops (light chain LV4 loop: Kabat positions 66-70; and heavy chain HV4 loop: Kabat positions 72-75 or 76) may be back mutated (from human to rabbit), if required.
  • an extra cysteine in position 80 (Kabat numbering) of the rabbit VL (as compared to the human VL) may be removed or replaced (substituted) in the humanized version.
  • T23 (Kabat numbering) of the rabbit VH is replaced, preferably by the corresponding amino acid of the human VH.
  • the amino acid residues of the vernier zones (e.g. vernier zone FR3) of the human VH frame work region may be back mutated to the corresponding rabbit amino acids (see Figure 21 , graft ID Ab001_VHhum3).
  • the back mutated amino acid residues (e.g. vernier zone FR3) are refined, e.g. (at least partially) to the human sequences (see Figure 21 , graft ID Ab001_VHhum23).
  • a mutation from IGHJ2 to IGHJ1/4/5 is introduced into the framework region FR4 (see Figure 21 , graft ID Ab001_VHhum23).
  • the above mutations are combined (see Figure 21 , graft ID Ab001_VHhum23).
  • the light chain CDRL2 may be humanized (DASSLAS) (see Figure 22, e.g. graft ID Ab001_VLhum4).
  • the light chain CDRL1 QSSESVYNNSRLS
  • RASESVYNNSRLS see Figure 22, e.g. graft ID Ab001_VLhum5
  • a (G- Q) mutation from IGKJ4 to IGKJ1 is introduced into the framework region FR4 (see Figure 22, e.g. graft ID Ab001_VLhum6).
  • a N29A mutation is introduced in LCDR1 thereby removing a glycosylation motif (see Figure 22, e.g. graft ID Ab001_VLhum10).
  • a N29Q mutation is introduced in LCDR1 (see Figure 22, graft ID Ab001_VLhum1 1 ).
  • a S31 A mutation is introduced in LCDR1 (see Figure 22, graft ID Ab001 _VLhum12).
  • one or more of the above mutations are combined (see Figure 22, graft IDs Ab001_VLhum5, VLhum6, VLhumI O, VLhum11 , VLhuml 2).
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 5 or any one of SEQ ID NOs: 230 - 233, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 5, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 230, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 231 , a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 232, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 233, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
  • the antibody, or an antigen-binding fragment thereof comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to any one of SEQ ID NO: 200, SEQ ID NO: 201 , and SEQ ID NO: 202; and a VL comprising an amino acid sequence according to any one of SEQ ID NO: 203, SEQ ID NO: 204, SEQ ID NO: 205, SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, and SEQ ID NO: 209.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to SEQ ID NO: 202; and a VL comprising an amino acid sequence according to SEQ ID NO: 208.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to SEQ ID NO: 202; and a VL comprising an amino acid sequence according to SEQ ID NO: 209.
  • the light chain CDRL2 may be humanized (GASSLAS) (see Figure 23, graft ID Ab002_VLhum6).
  • GASTLAS humanized
  • a mutation (F- Y) is introduced into frame work region FR2 (see Figure 23, graft ID Ab002_VLhum1 1 , VLhum12).
  • a further (G->A) mutation is introduced into CDRL2 (see Figure 23, graft ID Ab001_VLhum11 , VLhum12).
  • the light chain CDRL1 may be humanized (RASQNVYSNNHLS) (see Figure 23, graft ID Ab002_VLhum12).
  • RASQNVYSNNHLS humanized
  • graft ID Ab002_VLhum12 one or more of the above mutations are combined (see Figure 23, graft IDs Ab002_VLhum6, VLhum1 1 , VLhum12).
  • all of the above mutations are present in the VL of the antibody or antigen-binding fragment thereof (see Figure 23, graft ID Ab002_VLhum12).
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 11 , a CDRL1 according to SEQ ID NO: 12 or SEQ ID NO: 235, a CDRL2 according to SEQ ID NO: 13, SEQ ID NO: 236 or SEQ ID NO: 237, and a CDRL3 according to SEQ ID NO: 14.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 11 , a CDRL1 according to SEQ ID NO: 12, a CDRL2 according to SEQ ID NO: 236, and a CDRL3 according to SEQ ID NO: 14.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 11 , a CDRL1 according to SEQ ID NO: 12, a CDRL2 according to SEQ ID NO: 237, and a CDRL3 according to SEQ ID NO: 14.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 1 1 , a CDRL1 according to SEQ ID NO: 235, a CDRL2 according to SEQ ID NO: 237, and a CDRL3 according to SEQ ID NO: 14.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to SEQ ID NO: 210; and a VL comprising an amino acid sequence according to any one of SEQ ID NO: 211 , SEQ ID NO: 212, SEQ ID NO: 213, and SEQ ID NO: 214.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g.
  • the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to SEQ ID NO: 210; and a VL comprising an amino acid sequence according to SEQ ID NO: 214.
  • the antibody according to the present invention may comprise an Fc moiety.
  • the Fc moiety may be of any origin, preferably it is derived from human origin, e.g. from human IgA or IgG, such as IgGI , lgG2, lgG3, and/or lgG4, e.g. human IgG 1 .
  • an Fc moiety refers to a sequence derived from the portion of an immunoglobulin heavy chain beginning in the hinge region just upstream of the papain cleavage site (e.g., residue 216 in native IgG, taking the first residue of heavy chain constant region to be 1 14) and ending at the C-terminus of the immunoglobulin heavy chain.
  • an Fc moiety may be a complete Fc moiety or a portion (e.g., a domain) thereof.
  • a complete Fc moiety comprises at least a hinge domain, a CH2 domain, and a CH3 domain (e.g., EU amino acid positions 216-446).
  • An additional lysine residue (K) is sometimes present at the extreme C-terminus of the Fc moiety, but is often cleaved from a mature antibody.
  • an Fc moiety comprises at least one of: a hinge (e.g., upper, middle, and/or lower hinge region) domain, a CH2 domain, a CF13 domain, or a variant, portion, or fragment thereof.
  • An Fc moiety may comprise at least a hinge domain, a CH2 domain or a CH3 domain.
  • the Fc moiety may be a complete Fc moiety.
  • the Fc moiety may also comprise one or more amino acid insertions, deletions, or substitutions relative to a naturally-occurring Fc moiety. For example, at least one of a hinge domain, CH2 domain or CH3 domain (or portion thereof) may be deleted.
  • the antibody, or antigen binding fragment thereof, according to the present invention comprises an Fc region.
  • Fc region refers to the portion of an immunoglobulin formed by two or more Fc moieties of antibody heavy chains.
  • the Fc region may be monomeric or "single-chain" Fc region (i.e., a scFc region).
  • Single chain Fc regions are comprised of Fc moieties linked within a single polypeptide chain (e.g., encoded in a single contiguous nucleic acid sequence).
  • Exemplary scFc regions are disclosed in WO 2008/143954 A2.
  • the Fc region is dimeric.
  • a “dimeric Fc region” or “dcFc” refers to the dimer formed by the Fc moieties of two separate immunoglobulin heavy chains.
  • the dimeric Fc region may be a homodimer of two identical Fc moieties (e.g., an Fc region of a naturally occurring immunoglobulin) or a heterodimer of two non-identical Fc moieties.
  • the Fc moiety, or the Fc region comprises or consists of an amino acid sequence derived from a human immunoglobulin sequence (e.g., from an Fc region or Fc moiety from a human IgG molecule).
  • the Fc moiety, or the Fc region may comprise one or more amino acids from another mammalian species.
  • a primate Fc moiety or a primate binding site may be included in the antibody, or antigenbinding fragment.
  • one or more murine amino acids may be present in the Fc moiety or in the Fc region.
  • the Fc moieties of the Fc region may be of the same or different class and/or subclass.
  • the Fc moieties may be derived from an immunoglobulin (e.g., a human immunoglobulin) of an IgGI , lgG2, lgG3 or lgG4 subclass.
  • the Fc moiety, or Fc region may be modified such that it varies in amino acid sequence from the complete Fc moiety, or Fc region, of a naturally occurring immunoglobulin molecule.
  • the engineered Fc moiety, or Fc region retains at least one desirable function conferred by the naturally-occurring Fc moiety.
  • Fc functions which may be modified by Fc engineering, or which may be retained, include Fc receptor (FcR) binding, antibody half-life modulation, ADCC function, protein A binding, protein G binding, and complement binding.
  • FcR Fc receptor
  • the antibody according to the present invention comprises a (complete) Fc moiety/Fc region.
  • the (complete) Fc moiety/Fc region may be engineered by one or more specific amino acid substitutions.
  • the may not be engineered (i.e., as occurring in a natural immunoglobulin, e.g. IgGI or lgG4).
  • the Fc moiety/Fc region is engineered (e.g., based on a human IgG, such as IgG 1 or lgG4, Fc moiety/Fc region) to diminish Fc effector functions, in particular to reduce or inhibit FcyR and complement interactions.
  • a human IgG such as IgG 1 or lgG4, Fc moiety/Fc region
  • the antibody of the present invention may comprise an Fc modification selected from Leu235Glu, Leu234Ala/Leu235Ala, Ser228Pro/Leu235Glu, Gly237Ala, Glu318Ala, Leu234Ala/Leu235Ala/Pro329Gly, Pro331 Ser/Leu234Glu/Leu235Phe, Asp265Ala, Glu233Pro, Gly236Arg/Leu328Arg, lgG2- lgG4 cross-subclass, Ala330Leu, His268Gln/Val309Leu/Ala330Ser/Pro331 Ser, Val234Ala/Gly237Ala/Pro238Ser/His268Ala/Val309Leu/Ala330Ser/Pro331 Ser, Asp270Ala, Leu234Ala/L235Ala/Gly237Ala/P238Ser/His268Ala/Ala330Ser/Pro331 Ser, Asp
  • the "LALA” mutation refers to the two substitutions L234A and L235A (EU numbering; also known as CH2 L4A and CH2 L5A mutation).
  • the "LALA” mutation abolishes antibody binding to FcyRI, FcyRH and FcyRllla (Lund J, Winter G, Jones PT, Pound JD, Tanaka T, Walker MR et al. Human Fc gamma Rl and Fc gamma RII interact with distinct but overlapping sites on human IgG. J Immunol. 1991 ; 147:2657-2662).
  • an exemplary amino acid sequence of CH1 -CH2-CH3 comprising the "LALA” mutation is set forth in SEQ ID NO: 141. Accordingly, the antibody preferably comprises an amino acid sequence according to SEQ ID NO: 141 or a sequence variant thereof, as described herein, wherein the "LALA" mutation is maintained.
  • the "LALA” mutation may be combined with further mutations, such as P329G or G236R (EU numbering).
  • the antibody (Fc moiety/Fc region) includes the "LALA” mutation (L234A and L235A), while P329 and/or G236 are maintained (i.e., not mutated).
  • binding of the antibody to an Fc receptor may be assessed by various methods known to the skilled person, such as ELISA (Hessell AJ, Hangartner L, Hunter M, Havenith CEG, Beurskens FJ, Bakker JM, Lanigan CMS, Landucci G, Forthal DN, Parren PWHI, et al.: Fc receptor but not complement binding is important in antibody protection against HIV.
  • ELISA Hessell AJ, Hangartner L, Hunter M, Havenith CEG, Beurskens FJ, Bakker JM, Lanigan CMS, Landucci G, Forthal DN, Parren PWHI, et al.: Fc receptor but not complement binding is important in antibody protection against HIV.
  • Antibodies of the invention can be of any isotype (e.g., IgA, IgG, IgM i.e. an a, y or p heavy chain).
  • the antibody may be of the IgA or IgG type.
  • antibodies may be IgGI , lgG2, lgG3 or lgG4 subclass, preferably IgGI or lgG4.
  • Exemplified sequences for IgGI and lgG4 constant regions, which may be useful in the antibody as described herein, are provided in SEQ ID NO: 70 (IgGI ) and SEQ ID NO: 142 (lgG4).
  • the antibody of the invention may comprise an amino acid sequence according to SEQ ID NO: 70 or SEQ ID NO: 142, or a sequence variant thereof as described herein.
  • the human lgG4 constant region sequence of SEQ ID NO: 142 comprises the stable hinge mutation S228P (S. Angal, D.J. King, M.W. Bodmer, A. Turner, A.D.G. Lawson, G. Roberts, B. Pedley, J.R.
  • Adair A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (lgG4) antibody, Molecular Immunology, Volume 30, Issue 1 , 1993, Pages 105-108, ISSN 0161 -5890, https://doi.org/10.1016/0161 - 5890(93)90432-B).
  • Antibodies of the invention may have a K or a A light chain. Exemplified sequences for K and X light chain constant regions, which may be useful in the antibody as described herein, are provided in SEQ ID NO: 71 (Ckappa) and SEQ ID NO: 72 (Clambda). Accordingly, the antibody of the invention may comprise an amino acid sequence according to SEQ ID NO: 71 or 72, or a sequence variant thereof as described herein. Preferably, the antibody of the invention has a K light chain, e.g. having the constant region comprising or consisting of SEQ ID NO: 71 .
  • an antigen-binding fragment may or may not comprise an Fc moiety, in particular a portion of a complete Fc region.
  • the antibody, or antigen-binding fragment thereof is selected from Fab, Fab', F(ab')2, Fv or scFv.
  • F(ab')2 which may be obtained by pepsin cleavage or recombinant expression
  • Fab' which can be obtained from F(ab')2 or by recombinant expression
  • the antibody, or antigen-binding fragment may be a single-chain antibody (or fragment).
  • the single-chain antibody (or fragment) may encode the complete set of six CDRs, i.e. include the three heavy chain CDRs as well as the three light chain CDRs. More specifically, the single-chain antibody (or fragment) may include a heavy chain variable region (VH) as well as a light chain variable region (VL), for example including the VH and VL sequences as described above.
  • variants of the sequences recited in the application are also included within the scope of the invention.
  • variants include natural variants generated by somatic mutation in vivo during the immune response or in vitro upon culture of immortalized B cell clones.
  • variants may arise due to the degeneracy of the genetic code or may be produced due to errors in transcription or translation.
  • Antibodies of the invention, or antigen-binding fragments thereof, may be provided in purified form.
  • the antibody, or antigen-binding fragment will be present in a composition that is substantially free of other polypeptides e.g., where less than 90% (by weight), usually less than 60% and more usually less than 50% of the composition is made up of other polypeptides.
  • Antibodies of the invention may be immunogenic in non-human (or heterologous) hosts e.g., in mice.
  • the antibodies may have an idiotope that is immunogenic in non-human hosts, but not in a human host.
  • antibodies of the invention for human use include those that cannot be easily isolated from hosts such as mice, goats, rabbits, rats, non-primate mammals, etc. and cannot generally be obtained by humanization or from xeno-mice.
  • Antibodies of the invention also include hybrid antibody molecules that comprise the six CDRs from an antibody of the invention as defined above and one or more CDRs from another antibody to an antigen.
  • the antibody may be multispecific.
  • the multispecific antibody may comprise a first antigen-binding site according to the present invention (binding to ASC, in particular PYD of ASC as described herein) and a second antigen-binding site, which is different from the first antigen-binding site, i.e. which may bind to a distinct target.
  • the antibody, or the antigen-binding fragment thereof may be monospecific.
  • the invention also provides a nucleic acid molecule comprising a polynucleotide encoding the antibody according to the present invention, or an antigenbinding fragment thereof, as described above.
  • the nucleic acid molecule comprises one or more polynucleotide(s) encoding the exemplified antibodies of the invention (e.g., as described above, in particular in Table 1), or a sequence variant thereof as described herein (e.g., having at least 70%, 71 %, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity as described above).
  • polynucleotide(s) encoding the exemplified antibodies of the invention (e.g., as described above, in particular in Table 1), or a sequence variant thereof as described herein (e.g., having at least 70%, 71 %, 72%, 73%, 74%, 75%, 76%, 77%
  • Table 4 Exemplified nucleic acid sequences encoding the CDR and VH/VL sequences of exemplified antibodies as described herein are shown in Table 4 below.
  • Table 4 SEQ ID NOs for polynucleotide sequences encoding the CDR and VH/VL sequences of exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_AbOO4, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_Ab009.
  • nucleic acid molecules and/or polynucleotides include, e.g., a recombinant polynucleotide, a vector, an oligonucleotide, an RNA molecule such as an rRNA, an mRNA, an miRNA, an siRNA, or a tRNA, or a DNA molecule such as a cDNA.
  • Nucleic acids may encode the light chain and/or the heavy chain of an antibody (or a single chain antibody).
  • the light chain and the heavy chain of the antibody may be encoded by the same nucleic acid molecule (e.g., for single chain antibodies or for antibodies with separate heavy and light chains in bicistronic manner or an expression cassette containing more than one ribosome entry site such as IRES).
  • the light chain and the heavy chain of the antibody may be encoded by distinct nucleic acid molecules.
  • the present invention also comprises sequence variants of nucleic acid sequences, which encode the same amino acid sequences.
  • the polynucleotide encoding the antibody (or the complete nucleic acid molecule) may be optimized for expression of the antibody. For example, codon optimization of the nucleotide sequence may be used to improve the efficiency of translation in expression systems for the production of the antibody.
  • the nucleic acid molecule may comprise heterologous elements (i.e., elements, which in nature do not occur on the same nucleic acid molecule as the coding sequence for the (heavy or light chain of) an antibody.
  • a nucleic acid molecule may comprise a heterologous promotor, a heterologous enhancer, heterologous UTR (e.g., for optimal translation/expression), a heterologous poly-A-tail, heterologous DNA insulator elements and the like.
  • a nucleic acid molecule is a molecule comprising nucleic acid components.
  • the term nucleic acid molecule usually refers to DNA or RNA molecules. It may be used synonymous with the term "polynucleotide", i.e. the nucleic acid molecule may consist of a polynucleotide encoding the antibody. Alternatively, the nucleic acid molecule may also comprise further elements in addition to the polynucleotide encoding the antibody.
  • a nucleic acid molecule is a polymer comprising or consisting of nucleotide monomers which are covalently linked to each other by phosphodiester-bonds of a sugar/phosphate-backbone.
  • the term "nucleic acid molecule” also encompasses modified nucleic acid molecules, such as base-modified, sugar-modified or backbone-modified etc. DNA or RNA molecules.
  • the nucleic acid molecule may be manipulated to insert, delete or alter certain nucleic acid sequences. Changes from such manipulation include, but are not limited to, changes to introduce restriction sites, to amend codon usage, to add or optimize transcription and/or translation regulatory sequences, etc. It is also possible to change the nucleic acid to alter the encoded amino acids. For example, it may be useful to introduce one or more (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) amino acid substitutions, deletions and/or insertions into the antibody's amino acid sequence.
  • Such point mutations can modify effector functions, antigen-binding affinity, post-translational modifications, immunogenicity, etc., can introduce amino acids for the attachment of covalent groups (e.g., labels) or can introduce tags (e.g., for purification purposes).
  • a mutation in a nucleic acid sequence may be "silent", i.e. not reflected in the amino acid sequence due to the redundancy of the genetic code.
  • mutations can be introduced in specific sites or can be introduced at random, followed by selection (e.g., molecular evolution).
  • one or more nucleic acids encoding any of the light or heavy chains of an (exemplary) antibody can be randomly or directionally mutated to introduce different properties in the encoded amino acids.
  • Such changes can be the result of an iterative process wherein initial changes are retained and new changes at other nucleotide positions are introduced. Further, changes achieved in independent steps may be combined.
  • the polynucleotide encoding the antibody, or an antigen-binding fragment thereof, (or the (complete) nucleic acid molecule) may be codon-optimized.
  • codon optimization Online COOL
  • Grote A Hiller K
  • Scheer M Munch R
  • Nortemann B H
  • Jahn D JCat: a novel tool to adapt codon usage of a target gene to its potential expression host.
  • the nucleic acid molecule of the invention may comprise a nucleic acid sequence as set forth in any one of SEQ ID NOs 73 - 140; or a sequence variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity.
  • the nucleic acid molecule may encode any one of the exemplified antibodies ASC_Ab001, ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_AbOO5, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 (by combining the sequences as shown in Table 2), or a sequence variant thereof as described herein.
  • the nucleic acid molecule of the invention may comprise a nucleic acid sequence as set forth in any one of SEQ ID NOs 215 - 229; or a sequence variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity.
  • the nucleic acid molecule may encode any one of the exemplified humanized antibodies ASC_Ab001_hum (RP02-F8), or ASC_Ab002_hum (RP03-C12).
  • the present invention also provides a plurality of nucleic acid molecules encoding the antibody, or an antigen-binding fragment thereof, as described herein, wherein each of the nucleic acid molecules (of the plurality of nucleic acid molecules) comprises a polynucleotide encoding an immunoglobulin chain of the antibody, or an antigen-binding fragment thereof.
  • each of the nucleic acid molecules comprises a polynucleotide encoding an immunoglobulin chain of the antibody, or an antigen-binding fragment thereof.
  • the plurality of nucleic acid molecules encoding the antibody, or an antigen-binding fragment thereof, as described herein may be a combination of a first and a second nucleic acid molecule, wherein the first nucleic acid molecule comprises a polynucleotide encoding the heavy chain of the antibody, or an antigen-binding fragment thereof, of the present invention; and the second nucleic acid molecule comprises a polynucleotide encoding the corresponding light chain of the same antibody, or the same antigen-binding fragment thereof.
  • the above description regarding the (general) features of the nucleic acid molecule of the invention applies accordingly to the nucleic acid molecules of the plurality of nucleic acid molecules.
  • one or more of the polynucleotides encoding the immunoglobulin chains of the antibody, or an antigen-binding fragment thereof may be codon-optimized.
  • the plurality may comprise a nucleic acid sequence as set forth in any one of SEQ ID NOs 73 - 140; or a sequence variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity.
  • the plurality of nucleic acid molecules may encode any one of the exemplified antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_AbOO4, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_AbOO9 (by combining the sequences as shown in Table 2), or a sequence variant thereof as described herein.
  • the plurality may comprise a nucleic acid sequence as set forth in any one of SEQ ID NOs 215 - 229; or a sequence variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity.
  • the nucleic acid molecule may encode any one of the exemplified humanized antibodies ASC_Ab001_hum (RP02-F8), or ASC_AbOO2_hum (RP03-C12).
  • vectors for example, expression vectors, comprising a nucleic acid molecule according to the present invention or the plurality of nucleic acid molecules according to the present invention.
  • a vector comprises a nucleic acid molecule as described above.
  • the present invention also provides a plurality of vectors comprising the plurality of nucleic acid molecules according to invention as described above.
  • each vector of the plurality of vectors may contain one or more nucleic acid molecules of the plurality of nucleic acid molecules according to invention as described above.
  • the plurality of vectors may be a combination of a first and a second vector, wherein the first vector comprises a first nucleic acid molecule as described above (for the combination of nucleic acid molecules) and the second vector comprises a second nucleic acid molecule as described above (for the combination of nucleic acid molecules).
  • a vector is usually a recombinant nucleic acid molecule, i.e. a nucleic acid molecule which does not occur in nature.
  • the vector may comprise heterologous elements (i.e., sequence elements of different origin in nature).
  • the vector may comprise a multiple cloning site, a heterologous promotor, a heterologous enhancer, a heterologous selection marker (to identify cells comprising said vector in comparison to cells not comprising said vector), heterologous origin of replications, heterologous DNA insulator elements and the like.
  • a vector in the context of the present invention is suitable for incorporating or harboring a desired nucleic acid sequence.
  • Such vectors may be storage vectors, expression vectors, cloning vectors, transfer vectors etc.
  • a storage vector is a vector which allows the convenient storage of a nucleic acid molecule.
  • the vector may comprise a sequence corresponding, e.g., to a (heavy and/or light chain of a) desired antibody according to the present invention.
  • An expression vector may be used for production of expression products such as RNA, e.g. mRNA, or peptides, polypeptides or proteins.
  • an expression vector may comprise sequences needed for transcription of a sequence stretch of the vector, such as a (heterologous) promoter sequence.
  • a cloning vector is typically a vector that contains a cloning site, which may be used to incorporate nucleic acid sequences into the vector.
  • a cloning vector may be, e.g., a plasmid vector or a bacteriophage vector.
  • a transfer vector may be a vector which is suitable for transferring nucleic acid molecules into cells or organisms, for example, viral vectors.
  • a vector in the context of the present invention may be, e.g., an RNA vector or a DNA vector.
  • a vector in the sense of the present application comprises a cloning site, a selection marker, such as an antibiotic resistance factor, and a sequence suitable for multiplication of the vector, such as an origin of replication.
  • a vector in the context of the present application may be a plasmid vector.
  • the term "vector” may also refer to a delivery vector, e.g. for viral or non- viral delivery of a nucleic acid of the invention. Alternatively, it may be referred to viral or non-viral delivery systems. Accordingly, the present invention also provides a delivery vector/system comprising the nucleic acid molecule as described above (or comprising an expression vector as described above). The delivery vector/system may be viral or non-viral. Various examples of viral and non-viral delivery vectors/systems are known in the art and described, for example, in Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Adv Biomed Res. 2012; 1 :27.
  • Non-limiting examples of viral delivery vectors/systems include retroviral vectors; adenoviral vectors; adeno-associated viral (AAV) vectors, including helper-dependent adenoviral vectors and hybrid adenoviral vectors; herpes simplex virus vectors; lentivirus vectors; poxvirus vectors and Epstein-Barr virus vectors.
  • retroviral vectors adenoviral vectors
  • adeno-associated viral (AAV) vectors including helper-dependent adenoviral vectors and hybrid adenoviral vectors
  • herpes simplex virus vectors lentivirus vectors
  • poxvirus vectors and Epstein-Barr virus vectors Epstein-Barr virus vectors.
  • Non-limiting examples of non-viral delivery vectors/systems include chemical and non-chemical methods.
  • Non-chemical delivery includes physical methods, such as electroporation and other methods for transient penetration of the cell membrane by mechanical, electrical, ultrasonic, hydrodynamic, or laser-based energy; naked DNA or RNA delivery; gene gun; hydrodynamic delivery; ultrasound delivery and magnetofection.
  • Chemical non-viral delivery systems include cationic particles, in particular cationic lipids/liposomes, cationic polymers and lipid/polymer systems. Among non-viral vectors/systems, cationic liposomes are preferred.
  • the present invention also provides a (host) cell expressing the antibody according to the present invention, or an antigen-binding fragment thereof; and/or comprising the vector (or the plurality of vectors) according the present invention.
  • the (host) cell may be an isolated cell, which is not part of a human or animal body, e.g. a cell line or an engineered cell.
  • the cell may express the nucleic acid(s) or vector(s) of the invention in a recombinant manner, e.g. in a heterologous manner (i.e., the cel l/cel I type does not express the antibody or the antigen-binding fragment in nature).
  • the cells include, but are not limited to, eukaryotic cells, e.g., yeast cells, animal cells or plant cells. Other examples of such cells include, but are not limited to, prokaryotic cells, e.g. £. coli.
  • the cells are mammalian cells, such as a mammalian cell line. Examples include human cells, CHO cells, HEK293 cells, PER.C6 cells, NSO cells, human liver cells, myeloma cells or hybridoma cells.
  • the cell may be transfected with a vector according to the present invention, for example with an expression vector.
  • transfection refers to the introduction of nucleic acid molecules, such as DNA or RNA (e.g. mRNA) molecules, into cells, e.g. into eukaryotic or prokaryotic cells.
  • RNA e.g. mRNA
  • transfection encompasses any method known to the skilled person for introducing nucleic acid molecules into cells, such as into mammalian cells. Such methods encompass, for example, electroporation, lipofection, e.g.
  • the introduction is non-viral.
  • the cells of the present invention may be transfected stably or transiently with the vector according to the present invention, e.g. for expressing the antibody according to the present invention.
  • the cells are stably transfected with the vector according to the present invention encoding the antibody according to the present invention.
  • the cells are transiently transfected with the vector according to the present invention encoding the antibody according to the present invention.
  • the present invention also provides a recombinant host cell, which heterologously expresses the antibody of the invention or the antigen-binding fragment thereof.
  • the cell may be of another species than the antibody (e.g., CHO cells expressing human antibodies).
  • the cell type of the cell does not express (such) antibodies in nature.
  • the host cell may impart a post-translational modification (PTM; e.g., glycosylation) on the antibody that is not present in their native state.
  • PTM post-translational modification
  • the antibody of the invention, or the antigen-binding fragment thereof may have a post-translational modification, which is distinct from the naturally produced antibody (e.g., an antibody of an immune response in a human). Production of antibodies
  • Antibodies according to the invention can be made by any method known in the art.
  • the general methodology for making monoclonal antibodies using hybridoma technology is well known (Kohler, G. and Milstein, C., 1975; Kozbar et al. 1983).
  • Standard techniques of molecular biology may be used to prepare DNA sequences encoding the antibodies or antigen-binding fragments of the present invention.
  • Desired DNA sequences may be synthesized completely or in part, e.g., using oligonucleotide synthesis techniques. Site-directed mutagenesis and polymerase chain reaction (PGR) techniques may be used as appropriate.
  • PGR polymerase chain reaction
  • Any suitable host cell/vector system may be used for expression of the DNA sequences encoding the antibody molecules of the present invention.
  • Eukaryotic, e.g., mammalian, host cell expression systems may be used for production of antibody molecules, such as complete antibody molecules.
  • Suitable mammalian host cells include, but are not limited to, CHO, HEK293, PER.C6, NSO, myeloma or hybridoma cells.
  • prokaryotic, e.g. bacterial host cell expression systems may be used for the production of antibody molecules, such as complete antibody molecules.
  • Suitable bacterial host cells include, but are not limited to, coli cells.
  • the present invention provides a method for preparing the antibody, or an antigen-binding fragment or an immunoglobulin chain(s) thereof, according to the present invention, said method comprising
  • the present invention also provides a process for the production of an antibody molecule according to the present invention comprising culturing a (heterologous) host cell comprising a vector encoding a nucleic acid of the present invention, in particular under conditions suitable for expression of protein from DNA encoding the antibody molecule of the present invention, and isolating the antibody molecule.
  • a host cell such as a cell line
  • a first vector encoding a light chain polypeptide and a second vector encoding a heavy chain polypeptide e.g. as described above.
  • a single vector may be used, the vector including sequences encoding light chain and heavy chain polypeptides (e.g. for single chain antibodies or in a bicistronic manner).
  • the invention also provides a method for preparing a recombinant cell, comprising the steps of: (i) providing one or more nucleic acids that encode(s) the antibody of the invention; (ii) inserting the nucleic acid into an expression vector and (iii) transfecting the vector into a (heterologous) host cell in order to permit expression of the antibody of interest in that host cell.
  • the nucleic acid of step (i) may, but need not, be manipulated to introduce restriction sites, to change codon usage, and/or to optimize transcription and/or translation regulatory sequences.
  • the invention also provides a method of preparing a transfected host cell, comprising the step of transfecting a host cell with one or more nucleic acids that encode an antibody of interest.
  • a method of preparing a transfected host cell comprising the step of transfecting a host cell with one or more nucleic acids that encode an antibody of interest.
  • recombinant cells of the invention can then be used for expression and culture purposes. They are particularly useful for expression of antibodies for large-scale pharmaceutical production. They can also be used as the active ingredient of a pharmaceutical composition. Any suitable culture technique can be used, including but not limited to static culture, roller bottle culture, ascites fluid, hollow-fiber type bioreactor cartridge, modular minifermenter, stirred tank, microcarrier culture, ceramic core perfusion, etc.
  • the transfected host cell may be a eukaryotic cell, including yeast and animal cells, particularly mammalian cells (e.g., CHO cells, NSO cells, human cells such as PER.C6, HEK293 or HKB-11 cells, myeloma cells, or a human liver cell), as well as plant cells.
  • the transfected host cell is a mammalian cell, such as a human cell.
  • expression hosts can glycosylate the antibody of the invention, particularly with carbohydrate structures that are not themselves immunogenic in humans.
  • the transfected host cell may be able to grow in serum-free media.
  • the transfected host cell may be able to grow in culture without the presence of animal-derived products.
  • the transfected host cell may also be cultured to give a cell line.
  • the invention also provides a method of preparing the antibody of interest comprising the steps of: culturing or sub-culturing a transfected host cell population, e.g. a stably transfected host cell population, under conditions where the antibody of interest is expressed and, optionally, purifying the antibody of interest.
  • the transfected host cell population may be prepared by (i) providing nucleic acid(s) encoding a selected antibody of interest, (ii) inserting the nucleic acid(s) into an expression vector, (iii) transfecting the vector in a host cell that can express the antibody of interest, and (iv) culturing or sub-culturing the transfected host cell comprising the inserted nucleic acids to produce the antibody of interest.
  • antibodies according to the invention may be produced by (i) expressing a nucleic acid sequence according to the invention in a host cell, e.g. by use of a vector (or host cell) according to the present invention, and (ii) isolating the expressed antibody product. Additionally, the method may include (iii) purifying the isolated antibody.
  • the antibodies may be further purified, if desired, using filtration, centrifugation and various chromatographic methods such as HPLC or affinity chromatography.
  • Techniques for purification of antibodies, e.g., monoclonal antibodies, including techniques for producing pharmaceutical-grade antibodies, are well known in the art.
  • the present invention also provides a composition comprising one or more of: (i) the antibody of the present invention, or an antigen-binding fragment thereof;
  • the composition may be used for treatment or diagnostic purposes. Accordingly, the composition may be a pharmaceutical composition or a diagnostic composition.
  • the composition may comprise a (pharmaceutically acceptable) excipient, diluent or carrier.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising the antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid or the plurality of nucleic acids of the present invention, the vector or the plurality of vectors of the present invention, and/or the cell according to the present invention.
  • the pharmaceutical composition comprises the antibody, or an antigen-binding fragment thereof, according to the present invention.
  • the pharmaceutical composition may optionally also contain a pharmaceutically acceptable carrier, diluent and/or excipient.
  • a pharmaceutically acceptable carrier, diluent and/or excipient may facilitate administration, it should not itself induce the production of antibodies harmful to the individual receiving the composition. Nor should it be toxic. Suitable carriers may be large, slowly metabolized macromolecules such as proteins, polypeptides, liposomes, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers and inactive virus particles.
  • the pharmaceutically acceptable carrier, diluent and/or excipient in the pharmaceutical composition is not an active component in respect to ASC-related diseases or disorders.
  • the pharmaceutically acceptable carrier, diluent and/or excipient in the pharmaceutical composition is not an active component in respect to a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
  • Pharmaceutically acceptable salts can be used, for example mineral acid salts, such as hydrochlorides, hydrobromides, phosphates and sulphates, or salts of organic acids, such as acetates, propionates, malonates and benzoates.
  • Pharmaceutically acceptable carriers in a pharmaceutical composition may additionally contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents or pH buffering substances, may be present in such compositions. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries and suspensions, for ingestion by the subject.
  • compositions may be prepared in various forms.
  • the compositions may be prepared as injectables, either as liquid solutions or suspensions.
  • Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared (e.g., a lyophilized composition, similar to SynagisTM and Herceptin®, for reconstitution with sterile water containing a preservative).
  • the composition may be prepared for topical administration e.g., as an ointment, cream or powder.
  • the composition may be prepared for oral administration e.g., as a tablet or capsule, as a spray, or as a syrup (optionally flavored).
  • the composition may be prepared for pulmonary administration e.g., as an inhaler, using a fine powder or a spray.
  • the composition may be prepared as a suppository or pessary.
  • the composition may be prepared for nasal, aural or ocular administration e.g., as drops.
  • the composition may be in kit form, designed such that a combined composition is reconstituted just prior to administration to a subject.
  • a lyophilized antibody may be provided in kit form with sterile water or a sterile buffer.
  • the (only) active ingredient in the composition is the antibody, or the antigen-binding fragment thereof, of the present invention. As such, it may be susceptible to degradation in the gastrointestinal tract. Thus, if the composition is to be administered by a route using the gastrointestinal tract, the composition may contain agents which protect the antibody from degradation but which release the antibody once it has been absorbed from the gastrointestinal tract.
  • the present invention also provides a method of preparing a pharmaceutical composition comprising the steps of: (i) preparing an antibody of the invention; and (ii) admixing the purified antibody with one or more pharmaceutically acceptable excipients, diluents or carriers.
  • a method of preparing a pharmaceutical composition comprises the step of: admixing an antibody with one or more pharmaceutically-acceptable carriers, wherein the antibody is a monoclonal antibody.
  • compositions may generally have a pH between 5.5 and 8.5, in some embodiments this may be between 6 and 8, for example about 7.
  • the pH may be maintained by the use of a buffer.
  • the composition may be sterile and/or pyrogen free.
  • the composition may be isotonic with respect to humans.
  • pharmaceutical compositions are supplied in hermetically-sealed containers.
  • compositions present in several forms of administration include, but are not limited to, those forms suitable for parenteral administration, e.g., by injection or infusion, for example by bolus injection or continuous infusion.
  • parenteral administration e.g., by injection or infusion
  • the product may take the form of a suspension, solution or emulsion in an oily or aqueous vehicle and it may contain formulatory agents, such as suspending, preservative, stabilizing and/or dispersing agents.
  • the antibody may be in dry form, for reconstitution before use with an appropriate sterile liquid.
  • a vehicle is typically understood to be a material that is suitable for storing, transporting, and/or administering a compound, such as a pharmaceutically active compound, in particular the antibodies as described herein.
  • the vehicle may be a physiologically acceptable liquid, which is suitable for storing, transporting, and/or administering a pharmaceutically active compound, in particular the antibodies as described herein.
  • the compositions can be administered directly to the subject.
  • the compositions are adapted for administration to mammalian, e.g., human subjects.
  • Pharmaceutical compositions may include an antimicrobial, particularly if packaged in a multiple dose format. They may comprise detergent e.g., a Tween (polysorbate), such as Tween 80.
  • Detergents are generally present at low levels e.g., less than 0.01 %.
  • Compositions may also include sodium salts (e.g., sodium chloride) to give tonicity.
  • sodium salts e.g., sodium chloride
  • concentration of 10 ⁇ 2mg/ml NaCI is typical.
  • compositions may comprise a sugar alcohol (e.g., mannitol) or a disaccharide (e.g., sucrose or trehalose) e.g., at around 15-30 mg/ml (e.g., 25 mg/ml), particularly if they are to be lyophilized or if they include material which has been reconstituted from lyophilized material.
  • a sugar alcohol e.g., mannitol
  • a disaccharide e.g., sucrose or trehalose
  • the pH of a composition for lyophilization may be adjusted to between 5 and 8, or between 5.5 and 7, or around 6.1 prior to lyophilization.
  • the pharmaceutical compositions may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intraperitoneal, intrathecal, intraventricular, transdermal, transcutaneous, topical, subcutaneous, intranasal, enteral, sublingual, intravagi nal or rectal routes.
  • the pharmaceutical composition may be prepared for oral administration, e.g. as tablets, capsules and the like, for topical administration, or as injectable, e.g. as liquid solutions or suspensions.
  • the pharmaceutical composition is an injectable. Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection are also encompassed, for example the pharmaceutical composition may be in lyophilized form.
  • the active ingredient may be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection.
  • Preservatives, stabilizers, buffers, antioxidants and/or other additives may be included, as required.
  • administration is usually in an "effective amount", e.g. in a "prophylactically effective amount” or a “therapeutically effective amount” (as the case may be), this being sufficient to show benefit to the individual.
  • an effective amount e.g. in a "prophylactically effective amount” or a “therapeutically effective amount” (as the case may be)
  • the actual amount administered, and rate and time-course of administration will depend on the nature and severity of what is being treated.
  • the pharmaceutical composition may be provided for example in a pre-filled syringe.
  • the pharmaceutical composition may also be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • the active ingredient i.e. the antibody as defined above, is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • the pharmaceutical composition may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, e.g. including accessible epithelial tissue. Suitable topical formulations are readily prepared for each of these areas or organs.
  • the pharmaceutical composition may be formulated in a suitable ointment, containing the pharmaceutical composition, particularly its components as defined above, suspended or dissolved in one or more carriers.
  • Carriers for topical administration include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical composition can be formulated in a suitable lotion or cream. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
  • Dosage treatment may be a single dose schedule or a multiple dose schedule.
  • the amount of the antibody in the pharmaceutical composition may not exceed 1 g or 500 mg.
  • the amount of the antibody in the pharmaceutical composition may not exceed 200 mg, or 100 mg.
  • the amount of the antibody in the pharmaceutical composition may not exceed 50 mg.
  • the composition may include antibodies of the invention, wherein the antibodies may make up at least 50% by weight e.g., 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more) of the total protein in the composition.
  • the antibodies may be in purified form.
  • nucleic acid typically DNA
  • Suitable gene therapy and nucleic acid delivery vectors are known in the art.
  • compositions typically include an "effective" amount of one or more antibodies as described herein, i.e. an amount that is sufficient to treat, ameliorate, attenuate, decrease or prevent a desired disease or condition, or to exhibit a detectable therapeutic effect.
  • Therapeutic effects also include reduction or attenuation in pathogenic potency or physical symptoms.
  • the precise effective amount for any particular subject will depend upon their size, weight, and health, the nature and extent of the condition, and the therapeutics or combination of therapeutics selected for administration. The effective amount for a given situation is determined by routine experimentation and is within the judgment of a clinician.
  • An effective dose may generally be from about 0.005 to about 100 mg/kg, for example from about 0.0075 to about 50 mg/kg or from about 0.01 to about 10 mg/kg. In some embodiments, the effective dose will be from about 0.02 to about 5 mg/kg, of the antibody (e.g. amount of the antibody in the pharmaceutical composition) in relation to the bodyweight (e.g., in kg) of the individual to which it is administered.
  • the pharmaceutical composition may also comprise an additional active component, which may be a further antibody or a component, which is not an antibody. In other embodiments, the pharmaceutical composition may not comprise an additional active component (in addition to the antibody of the invention or respective nucleic acids, vectors or cells as described above).
  • the pharmaceutical composition may comprise one or more of the additional active components.
  • the antibody of the invention can be present either in the same pharmaceutical composition as the additional active component or, alternatively, the antibody may be comprised by a first pharmaceutical composition and the additional active component may be comprised by a second pharmaceutical composition different from the first pharmaceutical composition. Accordingly, if more than one additional active component is envisaged, each additional active component and the antibody may be comprised in a different pharmaceutical composition.
  • Such different pharmaceutical compositions may be administered either combined/simultaneously or at separate times or at separate locations (e.g. separate parts of the body), optionally by different routes of administration.
  • the antibody and the additional active component may provide an additive therapeutic effect, such as a synergistic therapeutic effect.
  • the term “synergy” is used to describe a combined effect of two or more active agents that is greater than the sum of the individual effects of each respective active agent. Thus, where the combined effect of two or more agents results in “synergistic inhibition" of an activity or process, it is intended that the inhibition of the activity or process is greater than the sum of the inhibitory effects of each respective active agent.
  • the term “synergistic therapeutic effect” refers to a therapeutic effect observed with a combination of two or more therapies wherein the therapeutic effect (as measured by any of a number of parameters) is greater than the sum of the individual therapeutic effects observed with the respective individual therapies.
  • the present invention also provides a diagnostic composition
  • a diagnostic composition comprising an antibody according to the present invention, a nucleic acid(s) according to the present invention, a vector(s) according to the present invention, and/or a cell according to the present invention.
  • the diagnostic composition may optionally comprise suitable means for detection, such as reagents conventionally used in immuno- or nucleic acid based diagnostic methods.
  • the antibodies described herein are, for example, suited for diagnostic purposes. Accordingly, they may be used in immunoassays, in which they can be utilized in liquid phase or bound to a solid phase carrier. Such immunoassays may be competitive or noncompetitive immunoassays; in either a direct or in an indirect format. Examples of such immunoassays include, but are not limited to, radioimmunoassay (RIA), enzyme-linked immunoassay (ELISA), sandwich (immunometric assay), immunohistochemistry, flow cytometry and Western blot assay. To this end, the antibody may be labelled, e.g. as described above.
  • the present invention also provides a kit comprising one or more of
  • the kit may comprise means for administration of the antibody, or an antigen binding fragment thereof, according to the present invention, the nucleic acid according to the present invention, the vector according to the present invention, the cell according to the present invention or the pharmaceutical composition according to the present invention, such as a syringe or a vessel, a leaflet, and/or a co-agent to be administered as described herein.
  • the kit may contain a leaflet, e.g. comprising instructions for use.
  • the kit may comprise one or more reagents, e.g. for use in appropriate diagnostic assays.
  • the kit may contain a reference agent or control.
  • the composition of the invention may be provided in kit form, e.g., designed such that a combined composition is reconstituted just prior to administration to a subject.
  • a lyophilized antibody may be provided in kit form with sterile water or a sterile buffer (e.g., in a separate container).
  • the present invention provides the use of the antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention or the (pharmaceutical) composition according to the present invention as a medicament.
  • the antibody according to the present invention, or an antigenbinding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention or the (pharmaceutical) composition according to the present invention may be used in prophylaxis and/or treatment of a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
  • the present invention also provides a method of treating, ameliorating or reducing a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder, or a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder, or lowering the risk of (occurrence of) a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder, or a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder, comprising: administering to a subject (in need thereof), (a therapeutically effective amount of) the antibody, or an antigen-binding fragment thereof, according to the present invention, a nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, a vector (or the plurality of vectors) according to the present invention, a cell according to the present invention or a (pharmaceutical) composition according to the present invention.
  • the present invention also provides the use of an antibody according to the present invention, or an antigen-binding fragment thereof, a nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, a vector (or the plurality of vectors) according to the present invention, a cell according to the present invention, or a pharmaceutical composition according to the present invention in the manufacture of a medicament for prophylaxis, treatment or attenuation of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder, or of a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder.
  • the terms “treat” or “treatment” include therapeutic treatment and prophylactic or preventative measures.
  • Prophylaxis of a neurodegenerative or neuroinflammatory disease refers in particular to prophylactic settings, wherein the subject was either not diagnosed with a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder (either no diagnosis was performed or diagnosis results were negative) and/or the subject does not show symptoms of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder.
  • the subject is typically diagnosed with a neurodegenerative or neuroinflammatory disease and/or showing symptoms of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder.
  • the terms “treatment” and “therapy'7”therapeutic” include (complete) cure as well as attenuation/reduction of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder and/or related symptoms.
  • the object of the "treatment” may be to decrease, ameliorate, inhibit, prevent or slow down (lessen or delay) an undesired physiological change or disorder, such as the neurodegenerative or neuroinflammatory disease or a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder.
  • beneficial or desired clinical results of a treatment include, but are not limited to, alleviation of symptoms, diminishment of the extent of a disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival, e.g. as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the manifestation of the condition or disorder or the risk thereof is to be decreased, delayed or prevented.
  • Treatment can be a single dose schedule or a multiple dose schedule.
  • an antibody, antibody fragment, nucleic acid, vector, cell, or composition as described herein may be administered to a subject in need of such treatment.
  • a subject includes, but is not limited to, one who is particularly at risk of, or susceptible to, a neurodegenerative or neuroinflammatory disease or a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder.
  • the antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention or the (pharmaceutical) composition according to the present invention may be used for human and also for veterinary medical purposes, preferably for human medical purposes.
  • the term “humansubject”, inconveniencepatient” or thereafteri ndi vidual” as used herein thus generally includes humans and non-human animals and preferably mammals (e.g., non-human primates, including marmosets, tamarins, spider monkeys, owl monkeys, vervet monkeys, squirrel monkeys, and baboons, macaques, chimpanzees, orangutans, gorillas; cows; horses; sheep; pigs; chicken; cats; dogs; mice; rat; rabbits; guinea pigs; etc.), including chimeric and transgenic animals and disease models.
  • the term “apparject” preferably refers a non-human primate or a human.
  • the subject is a human.
  • the human may be selected from the group of a human suffering from a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder, a human at risk of developing a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder, and a human of unknown clinical history for a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
  • the "neurodegenerative or neuroinflammatory disease” is characterized by or associated with the formation of ASC aggregates ("specks") and/or amyloid-p (Ap)-related pathology, in particular the formation and spreading of amyloid-P aggregates.
  • ASC aggregates specks
  • Ap-related pathology refers to the abnormal production, deposition and aggregation of amyloid-p in the brain.
  • the neurodegenerative or neuroinflammatory disease may involve interaction of ASC and amyloid-p.
  • the neurodegenerative or neuroinflammatory disease is associated with the fibrillation activity of ASC.
  • the neurodegenerative or neuroinflammatory disease involves ASC-induced inflammation.
  • Neurodegenerative diseases are typically chronic, progressive disorders characterized by the gradual loss of neurons in discrete areas of the central nervous system (CNS), such as the brain.
  • the neurodegenerative diseases envisaged to be treated may preferably be characterized and/or accompanied by dementia.
  • “Dementia” is a general term for a decline in mental ability severe enough to interfere with daily life. Dementia may include decline or loss of memory, communication and language, ability to focus and pay attention, reasoning and judgment, visual perception, or a combination thereof. It may be caused by neurodegeneration in a variety of neurodegenerative diseases.
  • Neuroinflammation is the inflammation of nervous tissue. It may be initiated in response to a variety of cues, including infection, traumatic brain injury, toxic metabolites or autoimmunity. Neuroinflammatory diseases are characterized by inflammatory processes in the nervous system, in particular in the CNS, which may result in the gradual damage and loss of neurons. Neuroinflammation and neurodegeneration often result from the aberrant deposition of aggregated host proteins that can activate inflammasomes. ASC is a central component of the inflammasome.
  • the neurodegenerative or neuroinflammatory disease is selected from Alzheimer's Disease, Parkinsons's Disease, Huntington's disease, Multiple System Atrophy, Amyotrophic Lateral Sclerosis, Multiple Sclerosis, traumatic brain injury, Sinocerebellar ataxia, Frontotemporal Dementia, Frontotemporal Lobar Degeneration, Mild Cognitive Impairment, Parkinson-plus syndromes, Pick disease, Progressive isolated aphasia, Greymatter degeneration [Alpers], Subacute necrotizing encephalopathy, or Lewy body dementia.
  • AD Alzheimer's Disease
  • MCI Mild Cognitive Impairment
  • Alzheimer's Disease is a neurodegenerative brain disease that is a major cause of dementia among the elderly. Symptoms of AD may include progressive loss of learning and memory functions, personality changes, neuromuscular changes, seizures and occasionally psychotic behaviour. Alzheimer's disease is characterized by the deposition of amyloid-P plaques in areas of the brain that are critical for memory and other cognitive functions. It is believed that the deposition of amyloid-P plaques, in these critical areas of the brain, interferes with brain functions.
  • MCI Mild Cognitive Impairment
  • AD Alzheimer's disease
  • FDD frontotemporal dementia
  • DLB dementia with Lewy bodies
  • MCI is often referred to as an objective cognitive complaint for age, in a person with essentially normal functional activities, who does not have dementia. It affects 19% of people aged 65 and over. Around 46% of people with MCI develop dementia within 3 years compared with 3% of the population of the same age.
  • neurodegenerative or neuroinflammatory diseases envisaged for prophylaxis or treatment according to the present invention include hereditary ataxia, congenital nonprogressive ataxia, early-onset cerebellar ataxia, late-onset cerebellar ataxia, cerebellar ataxia with defective DNA repair, hereditary spastic paraplegia, infantile spinal muscular atrophy, type I [Werdnig-Hoffman], inherited spinal muscular atrophy, systemic atrophies primarily affecting the central nervous system, paraneoplastic neuromyopathy and neuropathy, postpolio syndrome, Degenerative diseases of basal ganglia, Hallervorden-Spatz disease, progressive supranuclear ophthalmoplegia [Steele- Richardson-Olszewski], Neurogenic orthostatic hypotension [Shy-Drager], dystonia, tremor, chorea, Restless legs syndrome, Stiff-man syndrome, extrapyramidal and movement disorders, Multiple sclerosis, acute disseminated demyel
  • peripheral inflammatory disorder examples include, without being limited thereto, inflammatory bowel disease, non-alcoholic steatohepatitis, Beh et disease, sarcoidosis, lupus erythematosus, vasculitis, rheumatoid arthritis.
  • the antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention, or the pharmaceutical composition according to the present invention may be administered by any route of administration including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intraperitoneal, transdermal, transcutaneous, topical, subcutaneous, intranasal, enteral, sublingual or rectal routes.
  • any gene therapy approaches may be used, e.g.
  • the antibody according to the present invention, or an antigen-binding fragment thereof may be administered as nucleic acid or vector encoding said antibody, e.g. using viral or non-viral vectors as described above.
  • the antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention or the pharmaceutical composition according to the present invention may be administered systemically, for example by intravenous or subcutaneous administration.
  • Antibodies and fragments thereof as described herein may also be used for the (jn-vitrd) diagnosis of a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
  • Methods of diagnosis may include contacting an antibody with a sample. Such samples may be isolated from a subject, for example an isolated blood sample, such as whole blood, plasma or serum.
  • the methods of diagnosis may also include the detection of an antigen/antibody complex, in particular following the contacting of an antibody with a sample.
  • it may be tested, whether the sample contains antibodies competing with the antibodies as described herein, e.g. for allergen binding. This is typically performed in vitro, i.e. without any contact to the human or animal body.
  • diagnosis may be performed in vitro, for example by using an isolated sample as described above (and an in vitro analysis step as described above).
  • the present invention also provides the use of the antibody, or an antigenbinding fragment thereof, according to the present invention, the nucleic acid molecule or the plurality of nucleic acid molecules according to the present invention, the vector or the plurality of vectors according to the present invention, the cell according to the present invention, the composition according to the present invention, or the kit according to the present invention in in-vitro) diagnosis of a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
  • Figure 1 shows for Example 3 binding curves of exemplary antibody ASC_Ab001 to full length ASC, ASG specks, CARD domain, PYD domain, murine ASC and BSA as tested by ELISA.
  • Figure 2 shows for Example 4 an alignment of the different ASC isoforms: full-length ASC (SEQ ID NO: 1 ); ASC-b (SEQ ID NO: 145); ASC-c (SEQ ID NO: 146) and ASC-d (SEQ ID NO: 147).
  • Figure 3 shows for Example 4 representative images of binding patterns of (A) ASC_Ab001 , (B) ASC_Ab008, (C) ASC_Ab004, and (D) control antibody (AL- 177, adipogen).
  • lane 1 shows binding to ASC-b 20kDa
  • lane 2 shows binding to ASC-d 1 1 .8kDa
  • lane 3 shows binding to ASC-c 15kDa
  • lane 4 shows binding to ASC Y146A 21 ,5kDa
  • lane 5 shows binding to ASC K21 A K22A K26A 21.5kDa
  • lane 6 shows binding to UT Ctrl
  • lane 7 shows binding to ASC-HIS purified 23kDa.
  • Figure 4 shows for Example 4 the binding of anti-ASC antibodies and control antibodies as indicated to ASC peptides as indicated.
  • A Binding of ASC_Ab001 , ASC_AbOO2 clonotype, ASC_Ab004, ASC_Ab005, ASC_AbOO6, ASC_AbOO7, ASC_AbOO8 clonotype, AL-177 and VHHASC to peptides covering amino acids 1 to 20 and 1 1 to 30 of ASC.
  • B Binding of ASC_Ab001 , ASC_AbOO2, AL-177, IC100 and VHHASC to peptides covering amino acids 1 to 20, 1 1 to 30 and 21 to 40 of ASC.
  • Figure s shows for Example 5 ELISA binding curves of antibodies ASC_Ab001 , ASC_AbOO2, AL-177, VHHASC and IC100 to human ASC.
  • Figure 6 shows for Example 5 ELISA binding curves of antibodies ASC_Ab001 , ASC_Ab002, AL-177, VHHASC and IC100 to murine ASC.
  • Figure 7 shows for Example 5 ELISA binding curves of antibodies ASC_Ab001 , ASC_Ab002, AL-177, VHHASC and IC100 to ASC specks.
  • Figure 8 shows for Example 5 ELISA binding curves of antibodies ASC_Ab001 , ASC_Ab002, AL-177, VHHASC and IC100 to the PYD domain of ASC.
  • Figure 9 shows for Example 6 (A) the quantification of ASC specks detected in Thp1 wt or asc ''- cells primed with LPS (1 ug/mL), stimulated with Nigericin (5uM) and stained with 1 ug/mL of anti-ASC antibodies or isotype control and (B) representative immunofluorescence pictures of THP-1 asc +/+ and THP-1 asc-/- cells stained with an ASC_Ab001 antibody.
  • Figure 10 shows for Example 7 Western blot images of antibodies ASC_Ab001 , ASC_Ab004, ASC causalAb007 and ASC_AbOO9 on cell lysates of wt and asc" human THP-1 monocytes.
  • Figure 1 1 shows for Example 7 Western blot images of antibodies ASC_Ab001 , ASC_Ab004, ASC_Ab007 and ASC_AbOO9 on cell lysates of mouse bone- marrow derived macrophages (BMDMs).
  • BMDMs mouse bone- marrow derived macrophages
  • Figure 12 shows for Example 8 that treatment with ASC_Ab001 , ASC_Ab004, ASC_Ab007, ASC_Ab008 and ASC_Ab009 significantly impaired ASC fibrillation.
  • the effect of anti-ASC antibodies on ASC fibrillation was assessed by IF of ASC fibrils formed after incubation of ASC-gfp with anti-ASC antibodies or isotype control 1 h at 37°C.
  • (A) Representative picture and (B) quantification of number of ASC fibrils per well upon treatment with anti- ASC antibodies. Pooled data from n 1 or 2 independent experiments. One way ANOVA followed by Dunett's multiple comparison test, ns: nonsignificant, ****: p-value ⁇ 0.0001
  • Figure 13 shows for Example 8 the results of a sedimentation assay to assess antifibrillation properties of anti-ASC antibodies. Soluble and insoluble fractions of ASC were separated by high-speed centrifugation upon treatment with anti-ASC antibodies or isotype control. Treatment with ASC_Ab001 lead to more soluble ASC (supernatant fraction) when compared to ASC only or isotype control after 30 min of incubation at 37°C. Or in other words, ASC_Ab001 shifts the equilibrium towards the soluble ASC in the supernatant fraction. Treatment with an anti-CARD antibody had no effect on the fibrillation.
  • Figure 14 shows for Example 9 that the anti-ASC antibodies block ASC-Ap interaction. Percentage of inhibition of ASC- A0 binding after treatment with anti-ASC antibodies (3.5uM) or isotype control (3.5uM). One way ANOVA followed by Dunett's multiple comparison test ***: p-value ⁇ 0.001
  • Figure 15 shows for Example 10 that ASC_Ab001 blocks the pro- infl mmatory function of extracellular ASC fibrils.
  • the in-vitro efficacy of ASC_Ab001 was tested in a cell-based assay using differentiated human THP-1 macrophages that were treated with extracellular ASC fibrils. Before addition of the ASC fibrils to the cells, ASC fibrils were pre-incubated with ASC_Ab001 and isotype control antibody. IL-1
  • Figure 16 shows for Example 11 quantification of IL-1 beta release by differentiated human macrophages treated with ASC fibrils and 0.0035nM or 35nM of anti- ASC antibodies (ASC_Ab001 - ASC_Ab009) and isotype controls, respectively, in an ASC seeded Inflammation assay.
  • Figure 17 shows for Example 12 that anti-ASC antibodies (ASC cetAb001 - ASC_AbOO9) do not bind inflammasome proteins AIM2 and NRLP3 having homologous sequences to ASC, determined by a binding ELISA.
  • Figure 18 shows for Example 13 that anti-ASC antibodies ASC_Ab001 and ASC_Ab002 ameliorate the severity of dextran sulfate sodium (DSS)-induced colitis in an acute inflammatory bowel disease model. Measurement of (A) body weight; (B) disease activity index; and (C) colon length. Data represent mean +SEM. *p ⁇ 0.05, **p ⁇ 0.01 , ***p ⁇ 0.001, ****p ⁇ 0.0001 in two-way ANOVA with multiple comparisons to isotype.
  • Figure 19D is a diagram illustrating infiltration of inflammatory cells into the mucosa of healthy, isotype treated and Asc_Ab001 treated mice.
  • Figure 20 shows for Example 14 that humanized ASC_Ab001 preserves binding to ASC. Binding of humanized ASC_Ab001 to ASC-His was assessed by ELISA. Similar binding of the humanized antibodies was observed compared to the rabbit IgG format.
  • Figure 21 shows the VH amino acid sequences of humanized Ab001 antibodies. All sequences for the heavy chain are based on the IGHV3-66 germline, which has the highest homology to the original rabbit Ab001 heavy chain framework and has also been reported to be a good acceptor framework for rabbit CDRs. Humanized Ab001 antibodies comprising Ab001_VHhum23 were used in Example 15.
  • Figure 22 shows the VL amino acid sequences of humanized antibodies. All sequences for the light chain are based on the IGKV1 -5 germline, which has the highest homology to the original rabbit Ab001 light chain framework and is very similar to the 1GKV1-27*O1 germline, previously used as template for a generic acceptor frame for rabbit light chain CDRs. Humanized Ab001 antibodies comprising Ab001_VLhum11 and Ab001_VLhum12, respectively, were used in Example 15.
  • Figure 23 shows the VH and VL amino acid sequences of humanized Ab002 antibodies.
  • the presented sequence for the heavy chain is based on the IGHV3-66 germline, which is has the highest homology to the original rabbit Ab002 heavy chain framework and has also been reported to be a good acceptor framework for rabbit CDRs. All sequences for the heavy chain are based on the IGKV1 -27 germline, which has the highest homology to the original rabbit Ab002 light chain framework and was previously used as template for a generic acceptor frame for rabbit light chain CDRs.
  • a humanized Ab002 antibody comprising Ab002_VHhum17 and Ab002_VLhum12 was used in Example 15.
  • Figure 24 shows for Example 15 kinetic measurements of humanized Ab001 Fabs: Ab001 VH23/VL11 (A) and Ab001 VH23/VL12 (B) versus non-humanized Ab001 VHrab/VLrab (C) by Bio-Layer Interferometry (BLI) using a RED96e OCTET system (Sartorius AG).
  • Figure 25 shows for Example 15 kinetic measurements of humanized Ab002 Fabs: Ab002 VH16/VL12 (A) versus non-humanized Ab002 VHrabA/Lrab (B) by Bio-Layer Interferometry (BLI) using a RED96e OCTET system (Sartorius AG).
  • Figure 26 shows for Example 16 binding of full IgGI humanized Ab001 antibodies Ab001 VH23/VL1 1 and Ab001 VH23/VL12, as well as Ab001 rb-huIgG (non-humanized) to human ASC (A) and murine ASC (B), measured by binding ELISA.
  • Figure 27 shows for Example 16 binding of full IgGI humanized Ab002 antibody Ab002 VH1 7/VL12 as well as Ab002 rb-huIgG (non-humanized) to human ASC (A) and murine ASC (B), measured by binding ELISA.
  • Full-length tag-free human ASC (SEQ ID NO: 1 ) and ASC-GFP (SEQ ID NO: 152) was produced as intracellular specks in HEK293 and purified according to Martin-Sanchez F, Gomez Al, Pelegrfn P. Isolation of Particles of Recombinant ASC and NLRP3. Bio Protoc. 2015 May 20;5(10):e1480. doi: 10.21769/BioProtoc.1480. In brief, suspension HEK293 cells were transiently transfected with expression plasmids encoding full length human ASC or ASC-GFP using linear PEI 40kDa.
  • Buffer A 320 mM sucrose, 20 mM HEPES-KOH (pH 7.5), 10 mM KCI, 1.5 mM MgCb, 1 mM, EDTA, 1 mM EGTA.
  • Cells were lysed by syringing (10x 20G, 20x 25G), freeze-thawing (3 x), followed by subsequent syringing (20x 25G).
  • the lysate was centrifuged at 400g for 8min, the pellet was re-suspended in 2x CHAPS buffer (40 mM HEPES-KOH (pH 7.5), 10 mM MgCb, 1 mM EGTA, 0.2 mM PMSF, 0.2 % CHAPS) and filtered using a 5 pm centrifugal filters at 2000g for 10 min.
  • the filtrate was then diluted and gently mixed with 1 volume of 2x CHAPS buffer and centrifuged at 2300g for 8 min.
  • the resulting pellet was resuspended in 1 ml of 1 x CHAPS buffer and centrifuged at 5000g for 8 min. This washing step was repeated twice.
  • the pellet was re-suspended in 1 x CHAPS buffer and loaded carefully on the top of 40% Percoll and centrifuged at 16000g for 10min.
  • the interface layer containing the ASC speck particles was collected carefully and washed once by centrifugation at 5000g for 3min and resuspension in 1 x CHAPS buffer.
  • fluorescent particles were quantified in a fluorescence microscope using a Burker chamber.
  • Monomeric human ASC-His (SEQ ID NO: 153), ASC-GFP-His (SEQ ID NO: 154) and the ASC PYD domain (ASCPYD-His-SII; SEQ ID NO: 155) as well as the ASC CARD domain (ASCCARD-His-SII; SEQ ID NO: 156) were expressed in E. coli and purified from inclusion bodies via Nickel beads. Therefore, £ co// strain BL21 (DE3) was transformed with pET- based vectors encoding the his-tagged ASC variants. Expression was done in self-inducing media (MagicMediaTM, Invitrogen) incubated at 37°C for 1 h and 67h at 20°C.
  • MagicMediaTM MagneticMediaTM, Invitrogen
  • pellets were harvested by centrifugation at 4000rpm for 30min.
  • the pellet was resuspended in 50 mM phosphate, 300 mM NaCl, pH 7.5 and sonicated for on ice 10 min at 40% power (2 sec pulse/pause).
  • the suspension was then centrifuged at 14000g at 4°C for 30min to collect the pellet containing the inclusion bodies.
  • pellets were resuspended in 50 mM phosphate, 300 mM NaCI, 6M Gua-HCl, 2 mM DTT, pH 7.5 for 30min at RT.
  • the suspension was centrifuged at 14000g at 4°C for 30min to remove residual insoluble cell debris.
  • the supernatant was then incubated with Nickel beads (Themo, #88221 ) for 3h at RT., The beads were washed once with 50 mM phosphate, 300 mM NaCI, 6M Gua-HCl, 2 mM DTT, pH 7.5 and followed by 50 mM phosphate, 300 mM NaCI, 6M Gua-HCl, 2 mM DTT, 20mM imidazole pH 7.5.
  • High binding clear flat bottom 384-well plate (Greiner bio-one, 781061 ) were coated o/n (4°C) with 10 pl/well of ASC CARD domain (1.25ug/mL; SEQ ID NO: 156), ASC PYD domain (1.25ug/mL; SEQ ID NO: 155), murine ASC (Cusabio, CSB-EP861664 MO, 1.25ug/mL; SEQ ID NO: 151 ), ASC-His (1 ug/mL; SEQ ID NO: 153), ASC-gfp specks (50 000 specks/well) or BSA (1 pg/ml) diluted in PBS.
  • both human ASC and murine ASC were immobilized at a concentration as low as 0.625 n g/mL in 10 mM sodium phosphate buffer pH 5, for 300 s, to ensure the interaction with the analyte taking place predominantly at 1 :1 stoichiometric ratio.
  • Association and dissociation kinetics were measured at 50, 16.67, 5.55, 1.85, 0.617, 0.206 and 0 nM antibody concentrations in 1 x PBS buffer (pH 7.4), supplemented with 0.1 % BSA and 0.02% Tween, for 150 s and 500 s, respectively.
  • Antibodies were released from ASC by immersing the biosensors into regeneration buffer (10 mM glycine, pH 3.0).
  • the regeneration buffer was selected following a regeneration scouting procedure, as the mildest agent which resulted in full baseline recovery and complete preservation of ligand binding capacity. This allowed for using a single immobilized biosensor for the full concentration series of each antibody.
  • a biosensor for which buffer was injected as analyte throughout the entire run was used as a reference for subtraction.
  • the association (k on ) and dissociation (k O ft) constants were calculated using a 1 :1 binding model, with a global fit.
  • human ASC and murine ASC were immobilized at concentrations ranging from 1.25 pg/mL to 20 pg/mL.
  • Antibodies were binned in sets of up to 7 antibodies per experiment, in a two-dimensional matrix format, with the antibodies added sequentially, in pairs, at saturating concentrations. The biosensors were regenerated between consecutive antibody pairs with regeneration buffer. For data analysis, the condition where the same antibody was injected both as first and second antibody was selected as reference, for subtraction. Only incoming antibodies generating an additive binding response greater than the Bmax of the first antibody alone were considered non-competing. Distinct and overlapping bins were defined manually based on relative degrees of competition between the antibodies involved.
  • ASC-b, ASC-c, ASC-d (as described by Bryan et al, 2010)
  • ASC-Y146A (as described by Prather et al 2022)
  • ASC K21 A K22A K26A (as described by Venegas-Maldonado et al, 2017) were expressed in HEK293 by transfection with PEI.
  • Cell lysates were prepared by lysing the cells with ice-cold CHAPS buffer supplemented with proteases inhibitors.
  • Protein transfer (Blotting): Gels were placed onto AmershamTM ProtranTM Premium 0.45um Nitrocellulose Blotting membrane (Cytiva) and proteins transferred at 100 V for 1 h using Mini Trans-BlotTM Cell system (Bio-Rad) and 1 X Transfer buffer (25mM Tris-Base, 192mM glycine, 10% methanol). After blotting, membranes were blocked in 5% non-fat dry milk in PBS-T (0.1 % Tween-20) for 1 h at RT.
  • PBS-T 0.1 % Tween-20
  • THP-1 wt and asc-/- cells were seeded at a density of 10 000 cells/ well in 50uL of RPMI medium supplemented with Glutamax, 10%FBS, 1 %P/S, 100ug/mL Normocin, 100ug/mL Zeocin in a 384 well plate (Greiner bio-one 781986).
  • the cells were primed with 1 ug/mL LPS (Sigma, cat: L4391 -1 MG) for 3h. After priming, the formation of ASC specks was induced by treatment with 5uM Nigericin (Invivogen cat: tlrl-nig) for 1 h.
  • the cells were fixed and permealized using BD cytofix/cytoperm (BD biosciences, cat: 554722) solution for 20min at RT. The cells were then washed twice with PBS and blocked with 2% BSA supplemented with human Fc block reagent (Miltenyi cat: 130-059-901 ). After blocking, the cells were incubated with 1 ug/mL of anti-ASC antibodies or isotype control 2h at RT, washed twice with PBS and further incubated with a detection secondary antibody coupled with Alexa fluor 647 (Jackson Immunoresearch, 1 1 1 -607-003) for 1 h at RT.
  • Alexa fluor 647 Jackson Immunoresearch, 1 1 1 1 -607-003
  • DAPI Diamidino-2'-phenylindol-dihydrochloride
  • Cell lysates were prepared by lysing the cells (wt and a sc" human THP-1 monocytes and mouse BMDMs) with ice-cold lysis buffer containing 1 % Triton X-100 for 15 min on ice. After clearing the lysates by centrifugation at 17'000 x G for 15 min at 4°C, supernatant was transferred to a new set of pre-chilled tubes. Protein concentration was determined by BCA assay (PierceTM BCA Protein Assay Kit, Thermofisher Scientific). NuPAGE LDS sample buffer (4X) (Invitrogen) and NuPAGE Sample reducing agent (10X) (Invitrogen) was added to the samples and boiled at 95°C for 5 min.
  • Fibrillation Assay 1 .75uM ASC-GFP-His was incubated with 3.5uM of anti-ASC antibody or isotype control 1 h at 37°C in protein LoBind tube (Eppendorf, cat 0030109.116)). The mix was then transferred to a clear bottom non-binding 384well-plate (Greiner bio-one, cat 781906) and protein aggregates imaged. Images were acquired using the 10X objective of a fluorescence microscope and image processing accomplished using Fiji Image],
  • Samples were prepared for Western Blot by adding NuPAGE LDS sample buffer (4X) (Invitrogen) and NuPAGE Sample reducing agent (10X) (Invitrogen) and boiling at 95°C for 5 min before loading onto NuPAGE 4-12% Bis-Tris Gels (Invitrogen). Gels were run in MES SDS Running buffer (Invitrogen) at 80 V for 10 min and then at 120 V for another 1 10 min. Gel electrophoresis was performed using the Invitrogen XCell SureLockTM Electrophoresis cell chamber system.
  • Membranes were washed 3x in PBS-T for 10 min each time and then incubated in secondary antibody in 1 % non-fat dry milk in PBS-T for 1 h at RT. Membranes were washed 3x in PBS-T and subsequently incubated in SuperSignalTM West Pico PLUS Chemiluminescent Substrate (Thermofisher Scientific).
  • ASC-His fibrils were formed by incubating ASC-His monomer at 37°C for 1 h in protein LoBind tubes (Eppendorf, 0030109.116). Fibrillary ASC (1.75uM) was then incubated with 1 uM 5'-TAMRA-A01 -42 (Bachem) with and without 3.5uM anti-ASC antibodies or isotype control for 4h at 37°C. After incubation, protein aggregates were analyzed by flow cytometry using a Novocyte Advanteon (Agilent) and Flowjo v10.7.1 . ASC_AbOO1 TH P-1 efficacy data
  • monocytes were seeded at a density of 10'000 cells/well in RPMl medium supplemented with Glutamax, 10%FBS, 1 %P/S, 100 ug/mL Normocin, 100 ug/mL Zeocin in a 384-well plate.
  • RPMl medium supplemented with Glutamax, 10%FBS, 1 %P/S, 100 ug/mL Normocin, 100 ug/mL Zeocin in a 384-well plate.
  • PMA phorbol 12 -myristate- 13 -acetate
  • ASC fibrils were prepared by incubating ASC-His monomers in sterile PBS at 37°C for 1 h. Before addition of the ASC fibrils (3.5uM) to the cells, they were pre-incubated with antibodies (3.5uM) for 15 min at RT. After 16 h incubation at 37°C and 5% CO 2 , secreted IL-1 p was measured by Lumit Human IL-1 Immunoassay (Promega, W6010) according to manufacturer's protocol.
  • Example 1 Identification and production of antibodies binding to ASC
  • Immunization of rabbits with untagged, HEK derived ASC specks was carried out by dosing rabbits on day 1 , 7, 14, 21 and 35, with spleen and bone marrow cell harvesting 7 days after the final dose. Immunization of rabbits with the His tagged PYD or CARD domain of ASC was carried out by dosing rabbits on day 1 , 7, 14, 21 and 28, with spleen and bone marrow cell harvesting 5 days after the final dose. Cells were cryopreserved.
  • Rabbit bone marrow or spleen cells were screened for plasmablasts secreting antibodies specific for ASC specks, human ASC (SEQ ID NO: 1 ), mouse ASC (SEQ ID NO: 151), or ASC PYD or CARD domains (SEQ ID NOs 143 and 144, respectively), which were then sorted in 10pl of first strand buffer (ThermoFisher, USA).
  • ThermoFisher USA
  • DNA encoding the variable domain heavy (VH) and light (VL) sequences was amplified by 2 step PCR using primers annealing to the ends of rabbit variable framework regions, sequenced (Microsynth, Switzerland), translated into amino acid sequences, annotated and aligned (Geneious Biologies) and cloned into expression vectors containing rabbit constant heavy or light regions.
  • the expression vector pDB contains an Epstein-Barr virus origin of replication (oriP), which allows episomal replication of the transfected in cells expressing Epstein-Barr virus Nuclear Antigen! (EBNA1 ). Additionally, the plasmid pDB has a cytomegalovirus-chicken actin-fusion promoter (CAG Promoter), which results in high expression of recombinant proteins.
  • oriP Epstein-Barr virus origin of replication
  • EBNA1 Epstein-Barr virus Nuclear Antigen!
  • CAG Promoter cytomegalovirus-chicken act
  • Small scale antibody expression was carried out in suspension HEK293 cells. A few minutes prior to transfection, cells were seeded into a 24-well plate 500pl cell suspension at 0.8- 1 mio cells/ml in FreeStyle medium supplemented with 0.1 % Pluronic F68, 1 % Glutamax and 1 x Geniticin (all ThermoFisher, USA). 250 ng of heavy chain plasmid DNA and 250 ng of light chain plasmid DNA was mixed in a total volume of 15 pl of FreeStyle medium without supplements.
  • TranslT-LT1 2 pl of TranslT-LT1 (Mirus, USA) was added to 50 pl of pure FreeStyle medium, mixed and incubated for 5 minutes before being added drop by drop to the FreeStyle medium -DNA mix and incubated for a further 15 minutes at room temperature. This mixture was then added drop by drop to the plated cells, which were incubated at 37°C in a humidified incubator with 5% CO2 for around 3-4 days. The supernatant was harvested after centrifugation at 8000 rpm for 5-10 minutes.
  • V H and V L sequences as well as CDR sequences of the exemplary antibodies identified as described above are shown in Table 5 below.
  • Table 5 SEQ ID NOs for CDR and VH/VL sequences of exemplary antibodies ASC_Ab001 , ASC complicatAb002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009.
  • ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_AbOO4, ASC_Ab005, ASC_AbOO6, ASC_Ab007, ASC_Ab008 and ASC_Ab009 bind to the same or distinct epitope(s).
  • a cross-competition assay was performed.
  • cloned monoclonal antibodies were tested by biolayer interferometry (BLI) on full length ASC coupled to BLI sensors. Briefly, 1.25 pg/ml monomeric human ASC was immoblized to ARG2G biosensors (Sartorius AG) via amine coupling. Firstly, the blocking antibody was applied at a concentration of 266.7 nM to saturate ASC binding sites. To monitor competition, the binding of the subsequent antibodies was measured at a concentration of
  • antibodies ASC_Ab001 , ASC_Ab002 and ASC_Ab003 compete for the same epitope on ASC, indicating that antibodies ASC_Ab001 , ASC_Ab002 and ASC_Ab003 bind to the same epitope.
  • Partial competition was observed for antibodies ASC_Ab008 with ASC_AbOO9 as well as for ASC_Ab008 with ASC_Ab001 , ASC_Ab002 and ASC_Ab003, indicating that these antibodies bind to partially overlapping epitopes. No competition was observed for the remaining antibodies, indicating that these antibodies bind to distinct, non-overlapping epitopes on ASC.
  • antibodies were grouped based on the ASC epitope targeted. Antibodies were assigned to different binning groups named "PYD-1", “PYD-2”, “PYD-4”, “PYD-6", “PYD-7a”, “PYD-7b” and “PYD-7c”, wherein each binning group represents a distinct, nonoverlapping ASC epitope, except for "PYD-7a” and “PYD-7b” and “PYD-7b” and “PYD-7c", which epitopes are partially overlapping, as shown in Table 7 below:
  • Table 8 EC50-values determined for antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC complexAbOO8 and ASC_Ab009
  • Figure 1 shows the binding curves exemplarily for antibody ASC_Ab001 , additionally including the results for binding to the PYD domain and the CARD domain.
  • the data shown in Figure 1 demonstrate that exemplary antibody ASC_Ab001 specifically binds to human monomeric ASC, ASC specks, murine ASC and to the PYD domain of ASC, whereas no binding was detected to the CARD domain of ASC and to the bovine serum albumin (BSA) control.
  • BSA bovine serum albumin
  • ASC_Ab001 , ASC_Ab004 and ASC_Ab008 to ASC binding to the different ASC isoforms (full-length ASC, SEQ ID NO: 1 ; ASC-b, SEQ ID NO: 145; ASC-c, SEQ ID NO: 146 and ASC-d, SEQ ID NO: 147) and to human ASC carrying (i) mutations K21 A, K22A and K26A ("ASC K21 A K22A K26A"); or (ii) mutation Y146A (“ASC Y146A”) was investigated by western blot using lysate of HEK293 cells transfected with expression plasmids for expression of the different ASC isoforms or ASC carrying (i) mutations K21 A, K22A and K26A; or (ii) mutation Y146A.
  • Polyclonal antibody AL-177 (AdipoGen, Prod. No. AG-25B-0006-C100, Liestal, Switzerland), which is known to target an epitope in the N-terminus of the PYD domain, which is present in all four isoforms, was used as control.
  • Figure 2 shows an alignment of the different ASC isoforms. While full-length ASC and ASC- b contain a complete PYD domain, ASC-c lacks amino acids 26 to 85 of full-length ASC, while ASC-d contains only the N-terminal 35 amino acids of the PYD domain of ASC.
  • Figure 3 shows representative images of binding patterns of ASC_Ab001 , ASC_Ab004, ASC_Ab008 and control antibody (AL-177, adipogen). While control antibody AL-177 bound to each of the four ASC isoforms and to both mutated ASC, antibodies ASC_Ab001 , ASC_Ab004 and ASC_Ab008 bound to full-length ASC and ASC-b as well as to the two mutated ASC, but not to isoforms ASC-c and ASC-d.
  • binding of antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC cognitiveAb008 and control antibody AL-177 to ASC peptides covering amino acid 1 to 20 (SEQ ID NO: 148) or 1 1 to 30 (SEQ ID NO: 149) was assessed by ELISA.
  • nanobody VH HASC as described in Schmidt et al., 2016 Schomidt Fl, Lu A, Chen J , Ruan J, Tang C, Wu H, Ploegh HL.
  • a single domain antibody fragment that recognizes the adaptor ASC defines the role of ASC domains in inflammasome assembly.
  • Figure 4(A) shows the results of the first experiment. Only control antibody AL-177 bound to the N-terminal peptides, as expected based on its binding to all four ASC isoforms. As expected, no binding to the N-terminal peptides was found for VHHASC, which binds to CARD.
  • Figure 4(B) shows the results obtained for ASC peptides covering amino acid 1 to 20 (SEQ ID NO: 148), 1 1 to 30 (SEQ ID NO: 149) or 21 to 40 (SEQ ID NO: 150).
  • AL-177 was found to bind to the ASC peptides covering amino acid 1 to 20 (SEQ ID NO: 148) and 1 1 to 30 (SEQ ID NO: 149), while none of VH HASC, ASC_Ab001 and ASC_Ab002 bound to this peptide.
  • Antibody IC-100 did not bind to the most N-terminal peptide (amino acids 1 -20; SEQ ID NO: 148), but was found to bind to the peptides covering amino acids 11 to 30 (SEQ ID NO: 149) or 21 to 40 (SEQ ID NO: 150). None of the other antibodies bound to the peptide covering 21 to 40 (SEQ ID NO: 150).
  • this ASC peptide analysis shows that AL-177 binds to an epitope within amino acids 1 - 30 of ASC, while IC100 binds to an epitope within amino acids 11 - 40 of ASC. None of the antibodies of the present invention was found to bind to such an N-terminal peptide.
  • antibodies AL-177, IC100 and anti-CARD antibody VHH ASC bind to distinct epitopes of ASC as compared to the antibodies of the present invention.
  • binding of antibodies ASC_Ab001 and ASC__Ab002 as well as prior art antibodies AL-177, VHHASC and IC100 obtained as described in Example 4 above) to (i) human ASC, (ii) murine ASC, (iii) ASC specks and (iv) the PYD domain of ASC was assessed by ELISA as described above.
  • Results are shown in Figures 5 (human ASC), 6 (murine ASC), 7 (ASC specks) and 8 (PYD domain of ASC).
  • Example 4 demonstrates binding of IC100 to peptides of SEQ ID NOs 149 and 150, no binding to full-length human ASC, murine ASC, ASC specks and the (entire) PYD domain of ASC could be detected for IC100 (human IgGI antibody containing the VH of SEQ ID NO: 19 and the VL of SEQ ID NO: 30 of US 10,961 ,306 B2, as described in US 10,961 ,306 B2 column 40, lines 60-64). For the PYD domain, also for VHHASC no binding could be detected, which was expected since this antibody was previously reported to bind to the CARD domain of ASC.
  • IC100 human IgGI antibody containing the VH of SEQ ID NO: 19 and the VL of SEQ ID NO: 30 of US 10,961 ,306 B2, as described in US 10,961 ,306 B2 column 40, lines 60-64.
  • THP-1 asc /_ cells were compared to THP-1 wt cells.
  • THP-1 is human macrophage cell line, which is commercially available.
  • THP-1 asc /_ cells lack expression of ASC, in contrast to THP-1 wt cells, such that physiological ASC speck formation is only expected in THP-1 wt cells, while THP-1 asc A cells represent negative controls.
  • ASC specks formation of ASC specks was induced in THP-1 wt and THP-1 asc A cells.
  • the cells were then incubated with the anti-ASC antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_AbOO6, ASC_Ab007, ASC_Ab008, ASC_AbOO9 or isotype control, as described above in detail. Results were obtained by fluorescent imaging. Results are shown in Figure 9.
  • antibodies were tested by Western blot on wt and asc 7 human TH P-1 monocytes and mouse bone-marrow derived macrophages (BMDMs), as described in detail above.
  • BMDMs mouse bone-marrow derived macrophages
  • Results are shown in Figures 10 (wt and asc 7 ' human THP-1 cells) and 11 (BMDMs). All of the tested antibodies were binding to human and murine endogenous ASC in cell lysates from wt cells, while there was no binding observed in the asc 7 cel I lysates.
  • Example 8 Treatment with antibodies of the invention significantly impaired ASC fibrillation
  • ASC assembles in large cytoplasmic macromolecular assemblies called ASC specks. This is driven by ASC-ASC interaction and can be reconstituted in vitro by incubating ASC monomers at 37°C. Under these conditions ASC will rapidly fibrillate and retain their physiological and pathological functions (Friker LL, Scheibling H, Hochheiser IV, Brinkschulte R, Riedel D, Latz E, Geyer M, Heneka MT. [3-Amyloid Clustering around ASC Fibrils Boosts Its Toxicity in Microglia. Cell Rep. 2020 Mar 17;30(11 ):3743-3754.e6. doi: 10.1016/j.celrep.2020.02.025). An effect of the antibodies on fibrillation would provide a way to block either de novo or seeded ASC assembly, and thus the downstream effect of ASC speck formation such as inflammation and/or amyloid seeding.
  • ASC_Ab001 , ASC_Ab004, ASC_Ab007, ASC_Ab008 and ASC cognitively smoothing factor (ASC fibrillation) was assessed in an ASC fibrillation assay as described above. Briefly, ASC-GFP-His was incubated with the antibody to be tested and protein aggregates were imaged.
  • Results are shown in Figure 12. These data show that each of antibodies ASC_Ab001 , ASC_Ab004, ASC_Ab007, ASC_Ab008 and ASC complicatAb009 significantly impaired ASC fibrillation, as compared to an isotype control. An anti-ASC-CARD antibody used as control did not show any effects, while anti-CARD antibody VHHASC even resulted in increased ASC fibrillation.
  • Results are shown in Figure 13.
  • Treatment with ASC_Ab001 lead to more soluble ASC (supernatant fraction) when compared to ASC only or isotype control after 30 min of incubation at 37°C. Or in other words, ASC_Ab001 shifts the equilibrium towards the soluble ASC in the supernatant fraction.
  • treatment with an anti-CARD antibody had no effect on the fibrillation.
  • ASC -Ap interaction is assumed to be at the basis of increased A aggregation and amyloid deposition in Alzheimer disease (Venegas C, Kumar S, Franklin BS, Dierkes T, Brinkschulte R, Tejera D, Vieira-Saecker A, Schwartz S, Santarelli F, Kummer MP, Griep A, Gelpi E, Beilharz M, Riedel D, Golenbock DT, Geyer M, Walter J, Latz E, Heneka MT. Microglia- derived ASC specks cross-seed amyloid-p in Alzheimer's disease. Nature. 201 7 Dec 20;552(7685):355-361 .
  • ASC-A interaction assay was performed as described above. Thereafter, protein aggregates of the antibody to be tested were compared to those obtained with controls, such as ASC only, ASC+Ap, VHHASC and an isotype control antibody. ASC-Ap inhibition was determined as percentage in view the binding (aggregation) observed for the ASC+Ap (without antibody) control and the ASC only control.
  • Results are shown in Figure 14. These data demonstrate that antibodies ASC_Ab001 , ASC_AbOO2, ASC_Ab003, ASC_Ab004, ASC_Ab005 and ASC_Ab006 significantly reduce ASC-AP interaction, while anti-CARD antibody VH HASC even increased ASC-Ap interaction.
  • Example 10 ASC Ab001 blocks the pro-inflammatory function of extracellular ASC fibrils
  • ASC_Ab001 The in-vitro efficacy of ASC_Ab001 was tested in a cell-based assay using differentiated human THP-1 macrophages that were treated with extracellular ASC fibrils obtained as described above. It has been shown that ASC fibrils and ASC specks can propagate inflammation (Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels C, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmuller W, Latz E.
  • the adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation.
  • the NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014 Aug;15(8):738- 48.
  • ASC fibrils were pre-incubated with ASC_Ab001 and isotype control antibody, each with or without Leu234Ala/Leu235Ala/Pro329Gly ("PGLALA") Fc modification.
  • PGLALA Leu234Ala/Leu235Ala/Pro329Gly
  • Results are shown in Figure 15.
  • ASC_Ab001 significantly reduced the ASG fibrils-induced production of IL-1 P compared to the isotype control, demonstrating the efficacy of ASC_Ab001 .
  • This reduction was not dependent on antibody Fc function, as no difference between the wt IgG and the PGLALA (Fc-inactive) format was observed.
  • Treatment with antibody alone did not influence IL-113 production.
  • Example 11 Anti ASC antibodies decrease IL-1 beta release in a dose dependant manner in ASC seeded Inflammation assay
  • Wt THP-1 monocytes were seeded at a density of 20'000 cells/well in RPMI medium supplemented with Glutamax, 10%FBS, 1 %P/S, 100 ug/mL Normocin, 100 ug/mL Zeocin in a 384-well plate.
  • PMA phorbol 12-myristate-13-acetate
  • ASC fibrils were prepared by incubating ASC-his monomers in sterile PBS at 37°C for 1 h. Before addition of the ASC fibrils to the cells, they were pre-incubated with antibodies for 15 min at RT. After 48 h incubation at 37°C and 5% CO2, secreted IL-1 was measured by Lumit Human IL-1 Immunoassay (Promega, W6010) according to manufacturer's protocol.
  • Binding of anti-ASC antibodies (ASC_AB001 - ASC_AbOO9) to inflammasome proteins AIM2 and NLRP3 was tested by a binding ELISA.
  • High binding clear flat bottom 384-well plate (Greiner bio-one , 781061 ) were coated o/n (4°C) with 10 pl/well of AIM2 (1 ug/mL, Sino Biological, cat: 11654-H09B) or NLRP3 (1 pg/ml, cusabio, CSB-EP822275HU1 ) diluted in PBS. After o/n incubation, wells were aspirated and washed once with 100 pl/well of wash buffer (0.05% Tween-20 in PBS).
  • Wells were blocked for an hour with 50 l of 2% BSA diluted in PBS at RT. After blocking, wells were incubated 2 hours at RT with 10 pl of test antibody and serially diluted. Wells were washed four times and consecutively incubated with 10 pl/well of detection antibody for an hour at RT. Unbound detection antibody was removed by washing four times, and the wells were incubated with 20 pl TMB for 5 min. The reaction was stopped by adding 10 pl of 1 M H2SO4 per well. Absorbance was immediately read at 450 nm using EnSpire 2300 microplate reader (Perkin Elmer).
  • anti-ASC antibodies do not bind to inflammasome proteins AIM2 and NLRP3 comprising a homologous sequence to ASC, confirming the specificity of anti- ASC antibodies (ASC_AB001 - ASC_AbOO9) for ASC.
  • Example 13 Anti-ASC antibodies ameliorate the severity of dextran sulfate sodium (DSS)- induced colitis
  • DAI Disease activity index
  • Figure 19A to C show the beneficial effect of ASC_Ab001 in DSS induced colitis on the mucosal morphology in isolated bowel segments
  • Figure 19D illustrates the reduction of infiltration of inflammatory cells into the bowel mucosa in anti-ASC antibody treated animals versus isotype (normal IgG) treated animals.
  • the humanized antibody (ASC congruent humanized) contained the VH according to SEQ ID NO: 157 (which comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3 and SEQ ID NO: 4, respectively) and the VL according to SEQ ID NO: 158 (which comprises light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively).
  • SEQ ID NO: 157 which comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3 and SEQ ID NO: 4, respectively
  • the VL according to SEQ ID NO: 158 which comprises light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively.
  • Binding of the humanized ASC_Ab001 to human ASC was compared with (rabbit) ASC_Ab001 having VH and VL sequences according to SEQ ID NOs 8 and 9, respectively, as described above in an ELISA as described above.
  • Results are shown in Figure 20. These data show that similar binding of the humanized antibodies was observed compared to the rabbit IgG format.
  • Example 15 Construction and evaluation of further humanized anti-ASC antibodies and antigen binding fragments thereof
  • VL variable light chains
  • ASC_Ab001 Ab001_VLhum1 , Ab001_VLhum4, Ab001 _VLhum5, Ab001_VLhum6, Ab001_VLhum10, Ab001_VLhum1 1 , and Ab001_VLhum12
  • All sequences for the light chain are based on the IGKV1 -5 germline, which has the highest homology to the original rabbit Ab001 light chain framework and is very similar to the IGKV1 -27*01 germline, previously used as template for a generic acceptor frame for rabbit light chain CDRs.
  • humanized constructs comprising Ab001 threadVLhum11 and Ab001_VLhum12, respectively, were used.
  • VH humanized variable heavy chain
  • VL humanized variable light chains
  • the presented sequence for the heavy chain is based on the IGHV3-66 germline, which is has the highest homology to the original rabbit Ab002 heavy chain framework and has also been reported to be a good acceptor framework for rabbit CDRs.
  • association and dissociation kinetics were measured at 50, 16.67, 5.55, 1 .85, 0.61 7 and 0 nM Fabs concentrations in 1 x PBS buffer (pH 7.4), supplemented with 0.1 % BSA and 0.02% Tween, for 200 s and 500 s, respectively.
  • Antibodies were released from ASC by immersing the biosensors into regeneration buffer (10 mM glycine, pH 3.0).
  • the association (k on ) and dissociation (k O ff) rate constants were calculated using a 1 :1 binding model, with a global fit.
  • the calculated KD, k on and k o ff rate constants are presented in Table 1 0 (for ASC_Ab001 Fabs) and Table 1 1 (for ASC_Ab002 Fabs), respectively.
  • Table 1 1 KD, k O n and koff values determined for Fabs comprising Ab002 VH1 7/ VL12, and Ab002 VHrabA/Lrab, respectively
  • association and dissociation curves are graphically shown in Figure 24 (for Fabs comprising Ab001 VH23/VL1 1 , Ab001 VH23/VL12, and Ab001 VHrabA/Lrab, respectively), and Figure 25 (for Fabs comprising Ab002 VH1 7/ VL12, and Ab002 VHrab/VLrab, respectively).
  • the tested humanized Ab001 (RP02-F8) Fabs have a superior on-rate, but an inferior off-rate compared to the parental rabbit Fab.
  • the overall affinity of the humanized Ab001 Fabs to hASC is only marginally reduced compared to the parental rabbit Fab.
  • the tested humanized Ab002 (RP03-12) Fab has an on-rate and an off-rate similar to the on- and off-rates of the parental Fab.
  • Fabs comprising humanized VH and VL domains (Ab001 VH23/VL1 1 , Ab001 VH23A/L12, Ab002 VH17A/L12) have overall affinities to hASC similar to the affinity of the respective parental (non-humanized) rabbit antibodies (Ab001 VHrabA/Lrab, Ab002 VHrabA/Lrab) to hASC.
  • Example 16 Humanized IgG 1 antibodies have affinity to human and murine ASC
  • Binding of full IgGI humanized ASC_Ab001 antibodies and ASC_Ab002 antibodies to human and murine ASC was compared to (rabbit) Ab001 rb-huIgG, and Ab002 rb-huIgG, respectively (non-humanized) in an ELISA as described above.
  • Results are shown in Figure 26 (for Ab001 VH23A / L11 , Ab001 VH23A/L12 in comparison to Ab001 rb-huIgG) and Figure 27 (for Ab002 VH17A/L12 in comparison to Ab002 rb- huIgG).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Psychiatry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Hospice & Palliative Care (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention provides antibodies binding to apoptosis-associated speck-like protein containing a CARD (ASC), in particular to the PYRIN-PAAD-DAPIN domain (PYD) of ASC. The present invention also provides nucleic acids encoding such antibodies, vectors comprising such nucleic acids and host cells expressing said antibodies. Further provided herein are compositions and kits comprising such antibodies. In addition, the present invention also relates to the use of such antibodies, e.g. for prophylaxis or treatment of a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.

Description

ANTIBODIES AGAINST APOPTOSIS-ASSOCIATED SPECK-LIKE PROTEIN CONTAINING A CARD (ASC) AND USES THEREOF
The present invention relates to antibodies binding to apoptosis-associated speck-like protein containing a CARD (ASC; also referred to as PYCARD), in particular to the PYRIN- PAAD-DAPIN domain (PYD) of ASC. The present invention also relates to compositions and kits comprising such antibodies. In addition, the present invention also relates to the use of such antibodies, compositions and kits, e.g. for preventing or treating a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
Neurodegenerative diseases are typically chronic, progressive disorders characterized by the gradual loss of neurons in discrete areas of the central nervous system (CNS), such as the brain. Such neuronal damage may involve progressive degeneration and death of neurons. Within neurodegenerative diseases, it is estimated that 55 million people worldwide had dementia in 2019, and that by 2050 this figure will increase to 139 million people. In the United States, it is estimated that 6.5 million older adults suffer from Alzheimer's disease (AD) and about a million Americans suffer from Parkinson's disease. As of today, neurodegenerative diseases are not curable. Although certain treatments may help to relieve some of the physical or mental symptoms associated with neurodegenerative diseases, it is currently not even possible to delay their progression.
In the recent years, neuroinflammation has attracted increasing attention as common feature in the development of neurodegenerative diseases (Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG, Ramos-Escobar N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front Pharmacol. 2019 Sep 12; 10:1008. doi: 1 O.3389/fphar.2O19.01008; Amor S, Puentes F, Baker D, van der Valk P. Inflammation in neurodegenerative diseases. Immunology. 2010 Feb;129(2):154-69. doi: 10.1 11 1/j.l 365- 2567.2009.03225.x.; Chitnis T, Weiner HL. CNS inflammation and neurodegeneration. J Clin Invest. 2017 Oct 2;127(10):3577-3587. doi: 10.1172/JCI90609; Voet S, Srinivasan S, Lamkanfi M, van Loo G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol Med. 2019 Jun;1 1 (6):e10248. doi: 10.15252/emmm.201810248; Venegas C, Kumar S, Franklin BS, Dierkes T, Brinkschulte R, Tejera D, Vieira-Saecker A, Schwartz S, Santarelli F, Kummer MP, Griep A, Gelpi E, Beilharz M, Riedel D, Golenbock DT, Geyer M, Walter J, Latz E, Heneka MT. Microglia-derived ASC specks cross-seed amyloid-P in Alzheimer's disease. Nature. 2017 Dec 20;552(7685):355-361 ; Hulse J, Bhaskar K. Crosstalk Between the NLRP3 Inflammasome/ASC Speck and Amyloid Protein Aggregates Drives Disease Progression in Alzheimer's and Parkinson's Disease. Front Mol Neurosci. 2022 Feb 3; 15:805169. doi: 10.3389/fnmol.2022.805169). Neurodegenerative diseases often result from depositions of self-aggregated proteins, such as p-amyloid (A ), in neuronal cells. Usually, the self-aggregation products are toxic to the neurons, and eventually toxicity may spread. Aggregated proteins are known to activate inflammasomes, resulting in the production of inflammatory mediators.
Inflammasomes are cytosolic multiprotein complexes, which assemble in a stimulus-specific manner and, upon assembly, activate an inflammatory cascade. The assembly of an inflammasome is driven by self-association and oligomerization of three types of proteins: sensor, adaptor, and effector. Inflammasome sensors self-associate upon activation by danger signals and prompt the oligomerization of the inflammasome adaptor ASC (apoptosis-associated speck-like protein containing a CARD). Accordingly, inflammasome assembly usually requires ASC as adaptor protein. ASC is composed of two protein-protein interaction domains: a N-terminal PYRIN-PAAD-DAPIN domain (PYD) and a C-terminal caspase-recruitment domain (CARD). The PYD and CARD domains are members of the six- helix bundle death domain-fold superfamily that mediates assembly of large signaling complexes in the inflammatory and apoptotic signaling pathways via the activation of caspase. In inflammasome assembly, upstream sensor proteins, such as NLRP3 and AIM2, recruit ASC via interactions between their PYDs. ASC then interacts with caspase 1 via CARD/CARD interactions. ASC with its two domains, PYD and CARD, therefore represents the core structure of the inflammasome. Despite the increasing interest in targeting the inflammasome in neurodegenerative and neuroinflammatory diseases, most compounds developed in this context are NLRP3 inhibitory compounds, which target the NLRP3 sensor and are, thus, specific for NLRP3 inflammasomes, but not other inflammasomes (Voet S, Srinivasan S, Lamkanfi M, van Loo G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol Med. 2019 Jun;1 1(6):e10248. doi: 10.15252/emmm.201810248).
Recently, antibodies against ASC were developed (Schmidt Fl, Lu A, Chen JW, Ruan J, Tang C, Wu H, Ploegh HL. A single domain antibody fragment that recognizes the adaptor ASC defines the role of ASC domains in inflammasome assembly. J Exp Med. 2016 May 2;213(5):771 -90. doi: 10.1084/jem.20151790; Desu HL, Plastini M, llliano P, Bramlett HM, Dietrich WD, de Rivero Vaccari JP, Brambilla R, Keane RW. IC100: a novel anti-ASC monoclonal antibody improves functional outcomes in an animal model of multiple sclerosis. J Neuroinflammation. 2020 May 4;17(1 ):143. doi: 10.1 186/s12974-020-01826-0), which may be useful to target ASC. However, as these antibodies are still not sufficiently characterized, e.g. with regard to the epitope targeted on ASC, and early in the development, there is a need for further strategies targeting ASC, in particular in the context of neuroinflammatory or neurodegenerative diseases.
In view of the above, it is the object of the present invention to overcome the drawbacks of the prior art. In particular, it is an object of the present invention to provide antibodies, which bind specifically to the PYD domain of ASC. It is also an object of the present invention to provide antibodies, which are able to reduce or block fibrillation activity of ASC. It is also an object of the present invention to provide antibodies, which are able to reduce or block interaction of ASC and p-amyloid (Ap).
This object is achieved by means of the subject-matter set out below and in the appended claims.
Although the present invention is described in detail below, it is to be understood that this invention is not limited to the particular methodologies, protocols and reagents described herein as these may vary. It is also to be understood that the terminology used herein is not intended to limit the scope of the present invention which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art.
In the following, the elements of the present invention will be described. These elements are listed with specific embodiments, however, it should be understood that they may be combined in any manner and in any number to create additional embodiments. The variously described examples and embodiments should not be construed to limit the present invention to only the explicitly described embodiments. This description should be understood to support and encompass embodiments which combine the explicitly described embodiments with any number of the disclosed elements. Furthermore, any permutations and combinations of all described elements in this application should be considered disclosed by the description of the present application unless the context indicates otherwise.
Throughout this specification and the claims which follow, unless the context requires otherwise, the term "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated member, integer or step but not the exclusion of any other non-stated member, integer or step. The term "consist of" is a particular embodiment of the term "comprise", wherein any other non-stated member, integer or step is excluded. In the context of the present invention, the term "comprise" encompasses the term "consist of". The term "comprising" thus encompasses "including" as well as "consisting" e.g., a composition "comprising" X may consist exclusively of X or may include something additional e.g., X + Y.
The terms "a" and "an" and "the" and similar reference used in the context of describing the invention (especially in the context of the claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it were individually recited herein. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention. The word "substantially" does not exclude "completely" e.g., a composition which is "substantially free" from Y may be completely free from Y. Where necessary, the word "substantially" may be omitted from the definition of the invention.
The term "about" in relation to a numerical value x means x ± 10%, for example, x ± 5%, or x ± 7%, or x + 10%, or x ± 12%, or x + 15%, or x + 20%.
The term "disease" as used herein is intended to be generally synonymous, and is used interchangeably with, the terms "disorder" and "condition" (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
As used herein, reference to "treatment" of a subject or patient is intended to include prevention, prophylaxis, attenuation, amelioration and therapy. The terms "subject" or "patient" are used interchangeably herein to mean all mammals including humans. Examples of subjects include humans, cows, dogs, cats, horses, goats, sheep, pigs, and rabbits. Preferably, the subject or patient is a human.
Doses are often expressed in relation to the bodyweight. Thus, a dose which is expressed as [g, mg, or other unit]/kg (or g, mg etc.) usually refers to [g, mg, or other unit] "per kg (or g, mg etc.) bodyweight", even if the term "bodyweight" is not explicitly mentioned.
The term "binding" and similar reference usually means "specifically binding", which does not encompass non-specific sticking. In particular, specific binding of an antibody means that the antibody recognizes its target antigen and binds its target with greater affinity (or at lower antibody concentrations, e.g. EC50) than it does to a structurally different antigen and/or to an antigen with a modified or mutated sequence. Thereby, a "greater" affinity may be at least 2fold, 3fold, 4fold, 5fold, 10fold, 15fold, 20fold, 25fold, 50fold, 75fold, 100fold 150fold, 200fold, 500fold, 750fold, 1 ,000fold, 1 ,500fold, 2,000fold, 5,000fold, 7,500fold, 10,000fold or even higher affinity as compared to the binding to a control antigen. In some instances, antibody-binding to the control antigen may be undetectable (below detection threshold), while antibody-binding to the specific antigen may be well detected/determined. As used herein, the term "antibody" encompasses various forms of antibodies including, without being limited to, whole antibodies, antibody fragments (such as antigen binding fragments), human antibodies, chimeric antibodies, humanized antibodies, recombinant antibodies and genetically engineered antibodies (e.g., variant or mutant antibodies) as long as the characteristic properties according to the invention are retained. In some embodiments, the antibody is a humanized antibody. In some embodiments, the antibody is a monoclonal antibody. For example, the antibody may be a humanized monoclonal antibody.
As described above, the term "antibody" generally also includes antibody fragments. Fragments of the antibodies may retain the antigen-binding activity of the antibodies. Such fragments are referred to as "antigen-binding fragments". Antigen-binding fragments include, but are not limited to, single chain antibodies, Fab, Fab', F(ab')2, Fv or scFv. Fragments of the antibodies can be obtained from the antibodies by methods that include digestion with enzymes, such as pepsin or papain, and/or by cleavage of disulfide bonds by chemical reduction. Alternatively, fragments of the antibodies can be obtained by recombinant means, for example by cloning and expressing a part (fragment) of the sequences of the heavy and/or light chain. The invention also encompasses single-chain Fv fragments (scFv) derived from the heavy and light chains of an antibody of the invention. For example, the invention includes a scFv comprising the CDRs from an antibody of the invention. Also included are heavy or light chain monomers and dimers, single domain heavy chain antibodies, single domain light chain antibodies, as well as single chain antibodies, e.g., single chain Fv in which the heavy and light chain variable domains are joined by a peptide linker. Antibody fragments of the invention may be contained in a variety of structures known to the person skilled in the art. In addition, the sequences of the invention may be a component of multispecific molecules in which the sequences of the invention target the epitopes of the invention and other regions of the molecule bind to other targets. Although the specification, including the claims, may, in some places, refer explicitly to antigen binding fragment(s), antibody fragment(s), variant(s) and/or derivative(s) of antibodies, it is understood that the term "antibody" includes all categories of antibodies, namely, antigen binding fragment(s), antibody fragment(s), variant(s) and derivative(s) of antibodies. As used herein, the term "variable region" (variable region of a light chain (VL), variable region of a heavy chain (VH)) denotes each of the pair of light and heavy chains which is involved directly in binding the antibody to the antigen.
Antibodies of the invention can be of any isotype e.g., IgA, IgG, IgM i.e. an a, y or heavy chain). Preferably, the antibody is of the IgG type or the IgA type. Within the IgG isotype, antibodies may be IgGI , lgG2, lgG3 or lgG4 subclass, preferably lgG1 or lgG4. Antibodies of the invention may have a K or a A light chain.
Antibodies according to the present invention may be provided in purified form. Typically, the antibody will be present in a composition that is substantially free of other polypeptides e.g., where less than 90% (by weight), usually less than 60% and more usually less than 50% of the composition is made up of other polypeptides.
Antibodies according to the present invention may be immunogenic in human and/or in non-human (or heterologous) hosts e.g., in mice. For example, the antibodies may have an idiotope that is immunogenic in non-human hosts, but not in a human host. Antibodies of the invention for human use include those that cannot be easily isolated from hosts such as mice, goats, rabbits, rats, non-primate mammals, etc. and cannot generally be obtained by humanization or from xeno-mice.
As used herein, the term "antigen" refers to any structural substance which serves as a target for the receptors of an adaptive immune response, in particular as a target for antibodies, T cell receptors, and/or B cell receptors. An "epitope", also known as "antigenic determinant", is the part (or fragment) of an antigen that is recognized by the immune system, in particular by antibodies, T cell receptors, and/or B cell receptors. Thus, one antigen has at least one epitope, i.e. a single antigen has one or more epitopes. An antigen may be (i) a peptide, a polypeptide, or a protein, (ii) a polysaccharide, (iii) a lipid, (iv) a lipoprotein or a lipopeptide, (v) a glycolipid, (vi) a nucleic acid, or (vii) a small molecule drug or a toxin. Thus, an antigen may be a peptide, a protein, a polysaccharide, a lipid, a combination thereof including lipoproteins and glycolipids, a nucleic acid (e.g. DNA, siRNA, shRNA, antisense oligonucleotides, decoy DNA, plasmid), or a small molecule drug (e.g. cyclosporine A, paclitaxel, doxorubicin, methotrexate, 5-amino!evulinic acid), or any combination thereof. Preferably, the antigen is selected from (i) a peptide, a polypeptide, or a protein, (ii) a polysaccharide, (iii) a lipid, (iv) a lipoprotein or a lipopeptide and (v) a glycolipid; more preferably, the antigen is a peptide, a polypeptide, or a protein.
As used herein, the term "mutation" relates to a change in the nucleic acid sequence and/or in the amino acid sequence in comparison to a reference sequence, e.g. a corresponding genomic sequence. A mutation, e.g. in comparison to a genomic sequence, may be, for example, a (naturally occurring) somatic mutation, a spontaneous mutation, an induced mutation, e.g. induced by enzymes, chemicals or radiation, or a mutation obtained by site- directed mutagenesis (molecular biology methods for making specific and intentional changes in the nucleic acid sequence and/or in the amino acid sequence). Thus, the terms "mutation" or "mutating" shall be understood to also include physically making a mutation, e.g. in a nucleic acid sequence or in an amino acid sequence. A mutation includes substitution, deletion and insertion of one or more nucleotides or amino acids as well as inversion of several successive nucleotides or amino acids. To achieve a mutation in an amino acid sequence, a mutation may be introduced into the nucleotide sequence encoding said amino acid sequence in order to express a (recombinant) mutated polypeptide. A mutation may be achieved e.g., by altering, e.g., by site-directed mutagenesis, a codon of a nucleic acid molecule encoding one amino acid to result in a codon encoding a different amino acid, or by synthesizing a sequence variant, e.g., by knowing the nucleotide sequence of a nucleic acid molecule encoding a polypeptide and by designing the synthesis of a nucleic acid molecule comprising a nucleotide sequence encoding a variant of the polypeptide without the need for mutating one or more nucleotides of a nucleic acid molecule.
As used herein (i.e. throughout the present specification), the term "sequence variant" refers to any alteration in comparison to a reference sequence. The term "sequence variant" includes nucleotide sequence variants and amino acid sequence variants. Preferably, a reference sequence is any of the sequences listed in the "Table of Sequences and SEQ ID Numbers" (Sequence listing), i.e. SEQ ID NO: 1 to SEQ ID NO: 199. In particular, a "sequence variant" shares (over the whole length of the sequence) at least 70% or at least 75%, preferably at least 80% or at least 85%, more preferably at least 90% or at least 93%, even more preferably at least 95% or at least 96%, still more preferably at least 97% or at least 98%, particularly preferably at least 99% sequence identity with its reference sequence. In some embodiments, the "sequence variant" shares at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity. In general, i.e. throughout the present specification, the higher the %-identity is in a sequence variant, the more it is preferred. For example, a sequence variant having at least 84% sequence identity with a reference sequence is more preferred than a sequence variant having at least 75% sequence identity with the reference sequence.
In some embodiments, the "sequence variant" maintains the (biological) function of the reference sequence. For example, sequence variants relating to antibodies of the invention preferably maintain the specific binding to ASC, in particular to the PYD domain of ASC. In addition, other functions, such as anti-ASC-fibrillation activity and/or reducing or blocking of ASC-Ap interaction may also be maintained.
Sequence identity may be calculated as described below. Usually a sequence variant may preserve the specific function of the reference sequence. In some embodiments, an amino acid sequence variant has an altered sequence in which one or more (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) of the amino acids in the reference sequence is deleted or substituted, or one or more (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) amino acids are inserted into or added to the sequence of the reference amino acid sequence. As a result of the alterations, the amino acid sequence variant has an amino acid sequence which is at least 70% or at least 75%, preferably at least 80% or at least 85%, more preferably at least 90% or at least 93%, even more preferably at least 95% or at least 96%, still more preferably at least 97% or at least 98%, particularly preferably at least 99% identical to the reference sequence. For example, variant sequences which are at least 90% identical have no more than 10 alterations, i.e., any combination of deletions, insertions or substitutions, per 100 amino acids of the reference sequence. The same, of course, also applies similarly to nucleic acid sequences. The "% identity" of the sequence variant is usually determined with respect to the reference sequence. It is usually calculated with regard to the full length of the reference sequence (i.e. the sequence recited in the application). Percentage identity, as referred to herein, can be determined, for example, by methods known in the art, such as BLAST using the default parameters specified by the NCBI (the National Center for Biotechnology Information; http://www.ncbi.nlm.nih.gov/) [Blosum 62 matrix; gap open penalty=1 1 and gap extension penalty=1 ].
In general, while it is possible to have non-conservative amino acid substitutions, the substitutions are preferably conservative amino acid substitutions, wherein the substituted amino acid has similar structural or chemical properties with the corresponding amino acid in the reference sequence. By way of example, conservative amino acid substitutions involve substitution of one aliphatic or hydrophobic amino acids, e.g. alanine, valine, leucine and isoleucine, with another; substitution of one hydoxyl-containing amino acid, e.g. serine and threonine, with another; substitution of one acidic residue, e.g. glutamic acid or aspartic acid, with another; replacement of one amide-containing residue, e.g. asparagine and glutamine, with another; replacement of one aromatic residue, e.g. phenylalanine and tyrosine, with another; replacement of one basic residue, e.g. lysine, arginine and histidine, with another; and replacement of one small amino acid, e.g., alanine, serine, threonine, cysteine, and glycine, with another.
Several documents are cited throughout the text of this specification. Each of the documents cited herein (including all patents, patent applications, scientific publications, manufacturer's specifications, instructions, etc.), whether supra or infra, are hereby incorporated by reference in their entirety. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
It is to be understood that this invention is not limited to the particular methodology, protocols and reagents described herein as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art.
Antibodies and antigen-binding fragments thereof
In a first aspect the present invention provides an (isolated) antibody, or an antigen-binding fragment thereof, which (specifically) binds to ASC, in particular to the PYD domain of ASC. Preferably, the ASC is human ASC (SEQ ID NO: 1 ) and the PYD domain is the PYD domain (SEQ ID NO: 143) of human ASC. Apoptosis-associated speck-like protein containing a CARD (ASC; also referred to as PYCARD) is an adaptor protein that is composed of two protein-protein interaction domains: PYRIN-PAAD-DAPIN domain (PYD) and a caspaserecruitment domain (CARD). The PYD and CARD domains are members of the six-helix bundle death domain-fold superfamily that mediates assembly of large signaling complexes in the inflammatory and apoptotic signaling pathways via the activation of caspase. In particular, the antibody, or the antigen-binding fragment thereof, of the present invention binds specifically to the PYD domain of ASC. Preferably, the antibody, or the antigenbinding fragment thereof, of the present invention binds in particular to a polypeptide or protein having an amino acid sequence according to SEQ ID NO: 1 (human ASC), in particular to SEQ ID NO: 143 (PYD of human ASC). Accordingly, the antibody, or an antigen-binding fragment thereof, of the present invention does preferably not bind (specifically) to the CARD domain (SEQ ID NO: 144) of ASC.
Preferably, the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to monomeric ASC. It is also preferred that the antibody, or the antigenbinding fragment thereof, of the present invention binds (specifically) to ASC specks. ASC can assemble into a large protein complex, which is termed "speck". ASC specks are structures formed by ASC, which can reach a size of around 1 pm and which are characteristic of inflammasome activation. More preferably, the antibody, or the antigenbinding fragment thereof, of the present invention binds (specifically) to monomeric ASC and to ASC specks. ASC may be of any origin, for example from the species from which it is originally derived from (e.g. human ASC may be of human origin; murine ASC may be of murine origin and the like), it may be recombinantly expressed or a synthetic ASC peptide. Preferably, the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to physiological/endogenous ASC, in particular to human and/or murine physiological/endogenous ASC, e.g. physiological/endogenous ASC of human macrophages and/or of mouse bone-marrow derived macrophages (BMDMs). Binding to physiological/endogenous ASC may be tested by using cell lysates of human macrophages (such as THP-1 cells) and/or of mouse bone-marrow derived macrophages (BMDMs), e.g. as described in the appended examples.
ASC can occur in four different isoforms, namely (1 ) ASC (or "full-length ASC"; SEQ ID NO: 1 ); (2) ASC-b (SEQ ID NO: 145); (3) ASC-c (SEQ ID NO: 146) and (4) ASC-d (SEQ ID NO: 147). Preferably, the antibody, or the antigen-binding fragment thereof, of the present invention (specifically) binds to (human) ASC and (human) ASC-b. Moreover, the antibody, or the antigen-binding fragment thereof, of the present invention does preferably not (specifically) bind to (human) ASC-c and to (human) ASC-d. It is also preferred that the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to ASC K21 A K22A K26A (mutated ASC with K21 A, K22A and K26A mutations). Furthermore, it is preferred that the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to ASC Y146A (mutated ASC with Y146A mutation).
More preferably, the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to a fragment or a region of human ASC, which includes amino acids 36 to 85 of human ASC (SEQ ID NO: 1 ), more preferably to a fragment or a region of human ASC, which includes amino acids 41 to 85 of human ASC (SEQ ID NO: 1 ). Accordingly, the antibody, or the antigen-binding fragment thereof, of the present invention does preferably not bind (specifically) to a region (or fragment) of ASC comprised in (or corresponding to) amino acids 1 to 35 of ASC (SEQ ID NO: 1 ). In some embodiments, the antibody, or the antigen-binding fragment thereof, of the present invention does not (specifically) bind to the ASC peptide according to SEQ ID NO: 148 (ASC aa 1 -20 peptide). Additionally (or alternatively), the antibody, or the antigen-binding fragment thereof, of the present invention may (also) not (specifically) bind to the ASC peptide according to SEQ ID NO: 149 (ASC aa 1 1 -30 peptide). In some embodiments, the antibody, or the antigenbinding fragment thereof, of the present invention does not (specifically) bind to the ASC peptide according to SEQ ID NO: 150 (ASC aa 21 -40 peptide). Moreover, the antibody, or the antigen-binding fragment thereof, of the present invention does preferably not bind (specifically) to a region (or fragment) of ASC comprised in (or corresponding to) amino acids 86 to 195 of ASC (SEQ ID NO: 1 ). In other words, the epitope in the PYD of human ASC, to which the antibody, or the antigen-binding fragment thereof, of the present invention binds to, is preferably located within amino acids 36 to 85 of ASC (SEQ ID NO: 1 ), more preferably within amino acids 41 to 85 of ASC (SEQ ID NO: 1 ).
Preferably, the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to ASC of different species. More preferably, the antibody, or the antigen-binding fragment thereof, of the present invention binds (specifically) to human ASC (SEQ ID NO: 1 ) and murine ASC (SEQ ID NO: 151 ).
Standard methods to assess binding of the antibody according to the present invention, or the antigen-binding fragment thereof, are known to those skilled in the art and include, for example, ELISA (enzyme-linked immunosorbent assay). Thereby, the relative affinities of antibody binding may be determined by measuring the concentration of the antibody (EC50) required to achieve 50% maximal binding at saturation. A specific example of an ELISA, which may be used to assess binding of the antibody, is described in the appended examples.
In general, the antibody, or an antigen-binding fragment thereof, according to the present invention, may comprise three heavy chain complementarity determining regions (CDRs) and three light chain CDRs. In general, complementarity determining regions (CDRs) are the hypervariable regions present in the heavy chain variable domain and in the light chain variable domains. Typically, the CDRs of a heavy chain and the connected light chain of an antibody together form the antigen receptor. Usually, the three CDRs (CDR1 , CDR2, and CDR3) are arranged non-consecutively in the variable domain. Since antigen receptors are typically composed of two variable domains (on two different polypeptide chains, i.e. heavy and light chain: heavy chain variable region (VH) and light chain variable region (VL)), there are typically six CDRs for each antigen receptor (heavy chain: CDRH1 , CDRH2, and CDRH3; light chain: CDRL1 , CDRL2, and CDRL3). The CDRs in the VH and VL are usually separated by framework regions, whereby a framework region (FR) is a region in the variable domain which is less "variable" than the CDR. Usually, a variable region (or each variable region, respectively) is composed of four framework regions, separated by three CDR's.
For numbering of amino acids in the variable regions and identifying CDRs, different numbering schemes are known in the art including Kabat, Chothia, EU, IMGT, Paratome and AHo (Kabat, E.A.; National Institutes of Health (U.S.) Office of the Director. Sequences of Proteins of Immunological Interest, 5th ed.; DIANE Publishing: Collingdale, PA, USA, 1991 ; Tai Te Wu, Elvin A. Kabat; An analysis of the sequences of the variable regions of Bence Jones proteins and myeloma light chains and their implications for antibody complementarity. J Exp Med August 1970; 132 (2): 21 1 -250; George Johnson, Tai Te Wu, Kabat Database and its applications: 30 years after the first variability plot, Nucleic Acids Research, Volume 28, Issue 1 , 1 January 2000, Pages 214-218; Chothia C, Lesk AM. Canonical structures for the hypervariable regions of immunoglobulins. J Mol Biol. (1987) 196:901-17. doi: 10.1016/0022-2836(87)90412-8; Edelman, G.M.; Cunningham, B.A.; Gall, W.E.; Gottlieb, P.D.; Rutishauser, U.; Waxdal, MJ. The covalent structure of an entire gammaG immunoglobulin molecule. Proc. Natl. Acad. Sci. USA 1969, 63, 78-85; Lefranc, M.P.; Giudicelli, V.; Ginestoux, C.; Bodmer, J.; Muller, W.; Bontrop, R.; Lemaitre, M.; Malik, A.; Barbie, V.; Chaume, D. Imgt, the international immunogenetics database. Nucleic Acids Res. 1999, 27, 209-212; Kunik V, Ashkenazi S, Ofran Y. Paratome: an online tool for systematic identification of antigen-binding regions in antibodies based on sequence or structure. Nucleic Acids Res. 2012 Jul;40(Web Server issue):W521 -4. doi: 10.1093/nar/gks480; Honegger A, Pluckthun A. Yet another numbering scheme for immunoglobulin variable domains: an automatic modeling and analysis tool. J Mol Biol. 2001 Jun 8;309(3):657-70. doi: 10.1006/jmbi.2001 .4662). Accordingly, CDRs of the same variable region (VH or VL) may be determined according to distinct numbering schemes.
The sequences of the heavy chains and light chains of exemplary antibodies of the invention, comprising three different CDRs on the heavy chain and three different CDRs on the light chain were determined. The CDR amino acid sequences of the CDR1 of the heavy chain (CDRH1 ), the CDR2 of the heavy chain (CDRH2), the CDR3 of the heavy chain (CDRH3), the CDR1 of the light chain (CDRL1 ), the CDR2 of the light chain (CDRL2) and the CDR3 of the light chain (CDRL3) of exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC„Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 are shown in Table 1 below. To define the CDR regions, the Rabat CDR definition was applied (Tai Te Wu, Elvin A. Rabat; An analysis of the sequences of the variable regions of Bence Jones proteins and myeloma light chains and their implications for antibody complementarity. J Exp Med 1 August 1970; 132 (2): 211 -250; George Johnson, Tai Te Wu, Rabat Database and its applications: 30 years after the first variability plot, Nucleic Acids Research, Volume 28, Issue 1 , 1 January 2000, Pages 214-218).
Figure imgf000016_0001
Table 1 : SEQ ID NOs for CDR and VH/VL sequences of exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_AbOO5, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_Ab009.
Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of the specific combinations of the specific VH and VL sequences of exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 shown in Table 1 .
Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 8 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 9. As described above, the CDRs in the variable regions of SEQ ID NOs 8 and 9 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 8 and of a VL according to SEQ ID NO: 9, wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_Ab001 ) binds specifically to the PYD domain of human ASC, exhibits anti-ASC-fibri Nation activity, blocks ASC-Ap interaction and blocks the pro-inflammatory function of extracellular ASC fibrils.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 8 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 9. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively) are preferably maintained. Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 8 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 9.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 15 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 16. As described above, the CDRs in the variable regions of SEQ ID NOs 15 and 16 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 10, and SEQ ID NO: 11 , respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 15 and of a VL according to SEQ ID NO: 16, wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_Ab002) binds specifically to the PYD domain of human ASC and blocks ASC-Ap interaction.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 15 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 16. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 10, and SEQ ID NO: 1 1 , respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively) are preferably maintained.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 15 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 16.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 22 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 23. As described above, the CDRs in the variable regions of SEQ ID NOs 22 and 23 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 17, and SEQ ID NO: 18, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 22 and of a VL according to SEQ ID NO: 23, wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_Ab003) binds specifically to the PYD domain of human ASC and blocks ASC-Ap interaction.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 22 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 23. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 1 7, and SEQ ID NO: 1 8, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 1 9, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively) are preferably maintained.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 22 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 23.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 30 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 31 . As described above, the CDRs in the variable regions of SEQ ID NOs 30 and 31 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 24, SEQ ID NO: 25, and SEQ ID NO: 26, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 30 and of a VL according to SEQ ID NO: 31 , wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_Ab004) binds specifically to the PYD domain of human ASC, exhibits anti-ASC-fibri I lation activity and blocks ASC-A interaction.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 30 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 31 . Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 24, SEQ ID NO: 25, and SEQ ID NO: 26, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively) are preferably maintained.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 30 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 31 .
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 38 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 39. As described above, the CDRs in the variable regions of SEQ ID NOs 38 and 39 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 38 and of a VL according to SEQ ID NO: 39, wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_Ab005) binds specifically to the PYD domain of human ASC and blocks ASC-AJ3 interaction.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 38 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 39. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively) are preferably maintained. Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 38 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 39.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 46 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 47. As described above, the CDRs in the variable regions of SEQ ID NOs 46 and 47 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 40, SEQ ID NO: 41 , and SEQ ID NO: 42, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 46 and of a VL according to SEQ ID NO: 47, wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_Ab006) binds specifically to the PYD domain of human ASC and blocks ASC-Ap interaction.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 46 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 47. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 40, SEQ ID NO: 41 , and SEQ ID NO: 42, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively) are preferably maintained.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 46 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 47.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 54 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 55. As described above, the CDRs in the variable regions of SEQ ID NOs 54 and 55 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 48, SEQ ID NO: 49, and SEQ ID NO: 50, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 54 and of a VL according to SEQ ID NO: 55, wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_Ab007) binds specifically to the PYD domain of human ASC and exhibits anti-ASC-fibrillation activity.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 54 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 55. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 48, SEQ ID NO: 49, and SEQ ID NO: 50, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively) are preferably maintained.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 54 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 55.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 61 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 62. As described above, the CDRs in the variable regions of SEQ ID NOs 61 and 62 may be determined by different numbering schemes known in the art, such as Kabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 56, SEQ ID NO: 57, and SEQ ID NO: 58, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 61 and of a VL according to SEQ ID NO: 62, wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_Ab008) binds specifically to the PYD domain of human ASC and exhibits anti-ASC-fibrillation activity.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 61 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 62. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 56, SEQ ID NO: 57, and SEQ ID NO: 58, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively) are preferably maintained.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 61 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 62.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises a CDRH1 , a CDRH2 and a CDRH3 of a VH according to SEQ ID NO: 68 and a CDRL1 , a CDRL2 and a CDRL3 of a VL according to SEQ ID NO: 69. As described above, the CDRs in the variable regions of SEQ ID NOs 68 and 69 may be determined by different numbering schemes known in the art, such as Rabat, Chothia, EU, IMGT, Paratome and AHo numbering. Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 63, SEQ ID NO: 64, and SEQ ID NO: 65, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively. These CDR sequences were determined as described above. It is understood that the present inventions also encompasses antibodies comprising a CDRH1 , a CDRH2, a CDRH3, a CDRL1 , a CDRL2 and a CDRL3 of a VH according to SEQ ID NO: 68 and of a VL according to SEQ ID NO: 69, wherein the CDRs are determined by a distinct method.
As shown in the appended examples, such an antibody (e.g., ASC_AbOO9) binds specifically to the PYD domain of human ASC and exhibits anti-ASC-fibri Nation activity.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 68 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 69. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 63, SEQ ID NO: 64, and SEQ ID NO: 65, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively) are preferably maintained. Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 68 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 69.
The antibody, or the antigen-binding fragment thereof, of the invention may exhibit anti- ASC-fibri I lation activity. Antibodies reducing or inhibiting ASC fibrillation provide a way to block either de novo or seeded ASC assembly, and thus the downstream effect of ASC speck formation such as inflammation and/or amyloid seeding. Anti-fibrillation activity may be tested in a fibrillation assay or in a sedimentation assay, e.g. as described in the appended examples. Accordingly, preferred fibrillation assay is described in the examples of the present specification. Briefly, ASC assembles in large cytoplasmic macromolecular assemblies called ASC specks. This is driven by ASC-ASC interaction and can be reconstituted in vitro by incubating ASC monomers at 37°C. Under these conditions ASC will rapidly fibrillate and retain their physiological and pathological functions. Therefore, to test the effect of an antibody, or an antigen-binding fragment thereof, on ASC fibrillation, ASC, e.g. including a marker such as green fluorescent protein (GFP), may be incubated in the presence of the antibody to be tested (and appropriate controls as described below) at about 37°C, e.g. for about 1 h, and protein aggregates may be determined in view of the marker used (e.g. by fluorescent imaging in case of GFP). Protein aggregates of the antibody to be tested may be compared to those obtained with controls, such as ASC only and an unrelated antibody (which are both not expected to decrease ASC fibrillation). If significantly less fibrillation is observed for the antibody to be tested as compared to an unrelated antibody, the antibody to be tested exhibits anti-fibrillation activity.
The antibody, or the antigen-binding fragment thereof, of the invention may reduce or block ASC-Ap interaction. ASC-Ap interaction is assumed to be at the basis of increased Ap aggregation and amyloid deposition in Alzheimer disease (Venegas C, Kumar S, Franklin BS, Dierkes T, Brinkschulte R, Tejera D, Vieira-Saecker A, Schwartz S, Santarelli F, Kummer MP, Griep A, Gelpi E, Beilharz M, Riedel D, Golenbock DT, Geyer M, Walter J, Latz E, Heneka MT. Microglia-derived ASC specks cross-seed amyloid-P in Alzheimer's disease. Nature. 2017 Dec 20;552(7685):355-361 . doi: 10.1038/nature25158) as well as of increased neuroinflammation caused by these ASC-Ap complexes (Friker LL, Scheiblich H, Hochheiser IV, Brinkschulte R, Riedel D, Latz E, Geyer M, Heneka MT. p-Amyloid Clustering around ASC Fibrils Boosts Its Toxicity in Microglia. Cell Rep. 2020 Mar 17;30(11 ):3743-3754.e6. doi: 10.1016/j.celrep.2020.02.025). Antibodies reducing or blocking ASC-A0 interaction are thus assumed to be capable of reducing or blocking its downstream effects, in particular reducing or blocking Ap aggregation. Thereby, the antibody can reduce or block increased amyloid deposition and neuroinflammation.
The effect on ASC-A interaction may be tested in an ASC-Ap interaction assay. A preferred ASC-Ap interaction assay is described in the examples of the present specification. Briefly, ASC fibrils may be formed by incubating ASC monomers, e.g. at about 37°C for about 1 h. Fibrillary ASC may then be incubated with Ap, e.g. including a marker such as TAMRA ((5- Carboxy)tetramethylrhodamine), in the presence of the antibody to be tested (and appropriate controls as described below), e.g. at about 37°C for about 4h, and protein aggregates may be determined in view of the marker used (e.g. by flow cytometry in case of TAMRA). Protein aggregates obtained after incubation of ASC+Ap+anti-ASC antibodies may be compared to those obtained with controls, such as ASC only, ASC+Ap and an unrelated antibody. ASC-Ap inhibition may be determined as percentage in view the binding (aggregation) observed for the ASC+Ap (without antibody) control and the ASC only control. While no inhibition (0%) is assumed for the ASC+AP control, 100% inhibition is assumed for the ASC only group (because no interaction (aggregation) is expected for ASC without Ap). The %-inhibition of ASC-Ap interaction for the antibody to be tested may thus be calculated in view thereof and compared to the effects of an unrelated antibody to determine whether or not significant inhibition of ASC-Ap interaction is observed.
In some embodiments, the antibody, or the antigen-binding fragment thereof, of the invention may reduce or block Ap aggregation. In some embodiments, the antibody, or the antigen-binding fragment thereof, of the invention may reduce or block protein aggregation, in particular in the context of inflammatory processes. Without being bound to any theory, it is assumed that ASC acts as a "catalyzer" on the aggregation of proteins, such as Ap. Therefore, antibodies reducing or blocking the interaction of ASC with such (aggregating) proteins - in particular by binding to ASC - are assumed to reduce or block the aggregation of proteins (which is otherwise catalyzed by ASC). The antibody, or the antigen-binding fragment thereof, of the invention may reduce or block the pro-inflammatory function of extracellular ASC fibrils. It has been shown that ASC fibrils and ASC specks can propagate inflammation (Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmuller W, Latz E. The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation. Nat Immunol. 2014 Aug;15(8):727-37. doi: 10.1038/ni.2913; Baroja-Mazo A, Martm-Sanchez F, Gomez Al, Martfnez CM, Amores-lniesta J, Compan V, Barbera-Cremades M, Yagiie J, Ruiz-Ortiz E, Anton J, Bujan S, Couillin I, Brough D, Arostegui JI, Pelegnn P. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014 Aug;15(8):738-48. doi: 10.1038/ni.2919). To test the effect of an antibody on the pro-inflammatory function of extracellular ASC fibrils, a THP-1 efficacy assay may be used. A preferred THP-1 efficacy assay is described in the examples of the present specification. Briefly, wt and asc THP-1 monocytes may be seeded, e.g. at a density of about 10'000 cells/well, in a multi-well plate. Monocytes may then be differentiated into macrophages, e.g. by incubation in 100 ng/ml phorbol 12-myristate-13- acetate (PMA)-containing medium, e.g. for about 4 days at about 37°C and about 5% CO2. After differentiation cells may be washed and ASC fibrils may be prepared by incubating ASC monomers, e.g. at about 37°C for about 1 h. Before addition of the ASC fibrils to the cells, they may be pre-incubated with the antibody to be tested, e.g. for about 15 min at about 22°C. After incubation (e.g. about 16 h at about 37°C and about 5% CO2), secreted IL-1 P may be measured, e.g. by Lumit Human IL-1 Immunoassay (Promega, W6010). The amount of IL-1 secretion may be compared to that obtained when an unrelated control antibody was used in the same assay in wt THP-1 cells.
In some embodiments, the antibody, or the antigen-binding fragment thereof, is a humanized antibody. Methods for humanization of antibodies are well-known in the art. In general, in "humanized" antibodies, minimal portions of a non-human antibody (such as the six CDRs or (portions of) the VH and VL comprising the six CDRs) are introduced into an otherwise human antibody. Preferably, a humanized antibody is engineered to comprise one or more human framework regions in the variable region together with non-human (e.g., rabbit) CDRs of the heavy and/or light chain. More preferably, a humanized antibody comprises sequences that are entirely human except for the CDR regions. However, amino acids in the framework region (FR) of the human immunoglobulin may also be replaced by corresponding non-human residues. In addition, a humanized antibody may comprise residues that are found neither in the human form of the antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance. Preferably, a humanized antibody comprises a VH and a VL, in which all or essentially all of the CDR regions correspond to those of a parental non-human (e.g. rabbit) antibody and all or essentially all of the FR regions are those of a human antibody or a human immunoglobulin consensus sequence. The FR region can be modified in any manner known in the art and/or provided herein. The humanized antibody may also comprise a human Fc moiety. Humanized antibodies preferably exhibit reduced immunogenicity when introduced into the human body. „Humanized" antibodies may be prepared by creating a „chimeric" antibody (non-human Fab grafted onto human Fc) as an initial step and selective mutation of the (non-CDR) amino acids in the Fab portion of the molecule. Alternatively, „humanized" antibodies can be obtain directly by grafting appropriate „donor" CDR coding segments derived from a non-human animal onto a human antibody „acceptor" scaffold, and optionally mutating (non-CDR) amino acids for optimized binding. In some embodiments, the antibody, or the antigen-binding fragment thereof, is a monoclonal antibody. For example, the antibody, or the antigen-binding fragment thereof, may be a humanized monoclonal antibody.
In some embodiments, the variable regions or the CDRs of the antibody as defined herein are derived from a rabbit antibody and grafted in a scaffold of a human IgG or IgA antibody. Accordingly, the variable regions, portions thereof or the CDRs may be rabbit and grafted in an antibody framework, which is preferably of human origin. Typically, the rabbit-derived portions of the variable regions that are grafted into the antibody framework comprise the CDRs. Among IgG, IgGI and lgG4 are preferred.
Specific methods for humanization of rabbit antibodies are known in the art and described, for example, in Borras L, Gunde T, Tietz J, Bauer U, Hulmann-Cottier V, Grimshaw JP, Urech DM. Generic approach for the generation of stable humanized single-chain Fv fragments from rabbit monoclonal antibodies. J Biol Chem. 2010 Mar 19;285(12):9054-66. doi: 10.1074/jbc.M109.072876; in WO 2009/155726 A2; and in Zhang YF, Ho M. Humanization of rabbit monoclonal antibodies via grafting combined Kabat/IMGT/Paratome complementarity-determining regions: Rationale and examples. MAbs. 2017 Apr;9(3):419-429. doi: 10.1080/19420862.2017.1289302, which are incorporated herein by reference in their entirety.
In particular, for humanization of the exemplary antibodies of the invention, the skilled person may use the (rabbit) VH/VL sequences of the exemplary antibodies, as described in Table 1 , and determine "extended" rabbit CDRs in said VH/VL sequences of Table 1 by combining two or more (e.g., 3, 4, 5 or 6) different antibody numbering schemes (e.g., selected from Kabat, Chothia, EU, IMGT, Paratome and AHo), such that each "extended" CDR covers the respective CDRs as defined in each of the combined numbering schemes. Such "extended" CDRs determined by combining different numbering schemes may, thus, include amino acid residues of the framework region adjacent to a CDR (in particular when a single numbering scheme is used). In some embodiments, an "extended" CDR may correspond to the CDR as determined by a single numbering scheme. This applies in particular, if the CDR as determined by the single numbering scheme contains (or corresponds to) the corresponding CDR determined by one or more other numbering schemes. In some embodiments, Kabat, IMGT and Paratome may be used to obtain extended (rabbit) CDR sequences (based on the VH/VL sequences of the exemplified antibodies of Table 1 above), as described in Zhang and Ho, 2017 (Zhang YF, Ho M. Humanization of rabbit monoclonal antibodies via grafting combined Kabat/IMGT/Paratome complementarity-determining regions: Rationale and examples. MAbs. 2017 Apr;9(3):419-429. doi: 10.1080/19420862.2017.1289302). Extended rabbit CDRs may then be grafted onto human (germlined) IgG framework regions.
Specific examples of sets of extended CDRs of the exemplified antibodies are shown in Table 2 below.
Figure imgf000032_0001
Figure imgf000033_0001
Table 2: SEQ ID NOs of exemplary extended (rabbit) CDRs of the exemplary antibodies ASC_Ab001 (two versions of extended CDRs), ASC_AbOO2, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_AbOO9 Specific examples of VH/VL sequences of humanized versions of the exemplified antibodies are shown in Table 3 below.
Figure imgf000033_0002
Table 3: Exemplary humanized VH/VL sequences of the exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 and human germlined V and J gene versions suitable for each of these antibodies.
Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2, an extended CDRH3, an extended CDRL1 , an extended CDRL2 and an extended CDRL3 of exemplary antibodies ASC_Ab001 , ASC_AbOO2, ASC_Ab003, ASC_Ab004, ASC_AbOO5, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 shown in Table 2. As described above, "extended" CDRs may include amino acid residues of the framework region adjacent to the CDRs shown in Table
1 . Preferably, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 159, SEQ ID NO: 3, and SEQ ID NO: 160, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively. More preferably, the antibody of the invention, or the antigenbinding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 194, SEQ ID NO: 195, and SEQ ID NO: 196, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 197, SEQ ID NO: 198, and SEQ ID NO: 199, respectively.
In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 66, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -5 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 159, SEQ ID NO: 3, and SEQ ID NO: 160, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively. More preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 194, SEQ ID NO: 195, and SEQ ID NO: 196, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 197, SEQ ID NO: 198, and SEQ ID NO: 199, respectively.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 57 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 158. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 59, SEQ ID NO: 3, and SEQ ID NO: 1 60, respectively, and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively; even more preferably an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 94, SEQ ID NO: 195, and SEQ ID NO: 1 96, respectively, and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 197, SEQ ID NO: 1 98, and SEQ ID NO: 199, respectively) are maintained in the sequence variant of SEQ ID NO: 1 57 and in the sequence variant of SEQ ID NO: 158, respectively.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 57 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 58.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 176 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 177. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3, and SEQ ID NO: 4, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 159, SEQ ID NO: 3, and SEQ ID NO: 160, respectively, and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively; even more preferably an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 194, SEQ ID NO: 195, and SEQ ID NO: 196, respectively, and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 197, SEQ ID NO: 198, and SEQ ID NO: 199, respectively) are maintained in the sequence variant of SEQ ID NO: 176 and in the sequence variant of SEQ ID NO: 177, respectively.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 176 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 177.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 161 , SEQ ID NO: 10, and SEQ ID NO: 162, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively. In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 1 0, and SEQ ID NO: 1 1 , respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 23, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -1 6 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH 1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 61 , SEQ ID NO: 10, and SEQ ID NO: 1 62, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 12, SEQ ID NO: 1 , and SEQ ID NO: 14, respectively.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 78 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 79. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 10, and SEQ ID NO: 1 1 , respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 61 , SEQ ID NO: 10, and SEQ ID NO: 1 62, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 12, SEQ ID NO: 13, and SEQ ID NO: 14, respectively) are maintained in the sequence variant of SEQ ID NO: 178 and the sequence variant of SEQ ID NO: 179.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 78 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 179.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 161 , SEQ ID NO: 17, and SEQ ID NO: 163, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively.
In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 17, and SEQ ID NO: 18, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 66, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -5 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 161 , SEQ ID NO: 17, and SEQ ID NO: 163, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 180 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 181. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 17, and SEQ ID NO: 18, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 161 , SEQ ID NO: 17, and SEQ ID NO: 163, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 19, SEQ ID NO: 20, and SEQ ID NO: 21 , respectively) are maintained in the sequence variant of SEQ ID NO: 180 and the sequence variant of SEQ ID NO: 181 .
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 180 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 181 .
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 164, SEQ ID NO: 25, and SEQ ID NO: 165, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively.
In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 24, SEQ ID NO: 25, and SEQ ID NO: 26, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 66, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -6 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 64, SEQ ID NO: 25, and SEQ ID NO: 1 65, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 182 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 83. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 24, SEQ ID NO: 25, and SEQ ID NO: 26, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 64, SEQ ID NO: 25, and SEQ ID NO: 1 65, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 27, SEQ ID NO: 28, and SEQ ID NO: 29, respectively) are maintained in the sequence variant of SEQ ID NO: 182 and the sequence variant of SEQ ID NO: 183. Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 182 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 183.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 166, SEQ ID NO: 33, and SEQ ID NO: 167, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively.
In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 33, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -5 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 166, SEQ ID NO: 33, and SEQ ID NO: 167, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 184 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 185. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 32, SEQ ID NO: 33, and SEQ ID NO: 34, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 166, SEQ ID NO: 33, and SEQ ID NO: 167, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 35, SEQ ID NO: 36, and SEQ ID NO: 37, respectively) are maintained in the sequence variant of SEQ ID NO: 184 and the sequence variant of SEQ ID NO: 185.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 184 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 185.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 168, SEQ ID NO: 41 , and SEQ ID NO: 169, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively.
In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 40, SEQ ID NO: 41 , and SEQ ID NO: 42, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 23, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -27 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 168, SEQ ID NO: 41 , and SEQ ID NO: 1 69, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 186 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 187. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 40, SEQ ID NO: 41 , and SEQ ID NO: 42, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 168, SEQ ID NO: 41 , and SEQ ID NO: 1 69, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 43, SEQ ID NO: 44, and SEQ ID NO: 45, respectively) are maintained in the sequence variant of SEQ ID NO: 1 86 and the sequence variant of SEQ ID NO: 1 87.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 86 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 187. In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 70, SEQ ID NO: 49, and SEQ ID NO: 1 71 , respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively.
In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 48, SEQ ID NO: 49, and SEQ ID NO: 50, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 1 5, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -8 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 70, SEQ ID NO: 49, and SEQ ID NO: 1 71 , respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 88 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 89. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 48, SEQ ID NO: 49, and SEQ ID NO: 50, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 70, SEQ ID NO: 49, and SEQ ID NO: 171 , respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 51 , SEQ ID NO: 52, and SEQ ID NO: 53, respectively) are maintained in the sequence variant of SEQ ID NO: 188 and the sequence variant of SEQ ID NO: 189.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 188 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 189.
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 172, SEQ ID NO: 57, and SEQ ID NO: 173, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively.
In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 56, SEQ ID NO: 57, and SEQ ID NO: 58, respectively; and light chain CDR1, CDR2, and CDR3 sequences as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 33, the heavy chain J gene IGHJ2, the light chain V gene IGKV1 -12 and the light chain J gene IGKJ4 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRE11 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 172, SEQ ID NO: 57, and SEQ ID NO: 173, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively. In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 190 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 1 91 . Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 56, SEQ ID NO: 57, and SEQ ID NO: 58, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 72, SEQ ID NO: 57, and SEQ ID NO: 1 73, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 59, SEQ ID NO: 52, and SEQ ID NO: 60, respectively) are maintained in the sequence variant of SEQ ID NO: 190 and the sequence variant of SEQ ID NO: 1 91 .
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 90 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 1 91 .
In certain aspects, the antibody of the invention, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 74, SEQ ID NO: 64, and SEQ ID NO: 1 75, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively.
In some embodiments, the antibody of the invention, or the antigen-binding fragment thereof, comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 63, SEQ ID NO: 64, and SEQ ID NO: 65, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively; and humanized framework regions, wherein the heavy chain V gene IGHV3- 23, the heavy chain J gene IGHJ6, the light chain V gene 1GKV1 -27 and the light chain J gene IGKJ3 are used to obtain the humanized framework regions. Preferably, such an antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 74, SEQ ID NO: 64, and SEQ ID NO: 1 75, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively.
In some embodiments, such an antibody of the invention, or the antigen-binding fragment thereof, may comprise (i) a heavy chain variable region (VH) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 192 and a light chain variable region (VL) comprising an amino acid sequence having 70% or more (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79%, at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%) identity to SEQ ID NO: 193. Thereby, the CDR sequences as defined above (heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 63, SEQ ID NO: 64, and SEQ ID NO: 65, respectively; and light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively) are preferably maintained. More preferably, the extended CDR sequences as defined above (an extended CDRH1 , an extended CDRH2 and an extended CDRH3 as set forth in SEQ ID NO: 1 74, SEQ ID NO: 64, and SEQ ID NO: 175, respectively; and an extended CDRL1 , an extended CDRL2 and an extended CDRL3 as set forth in SEQ ID NO: 66, SEQ ID NO: 36, and SEQ ID NO: 67, respectively) are maintained in the sequence variant of SEQ ID NO: 192 and the sequence variant of SEQ ID NO: 193.
Preferably, the antibody, or an antigen-binding fragment thereof, comprises a heavy chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 192 and a light chain variable region comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 193.
In some embodiments, humanization may include back mutations, i.e. wherein amino acids of the human framework sequences are mutated back to the corresponding amino acids of the rabbit framework sequences, in particular if the result of grafting of the (extended) CDRs onto human IgG framework regions is not satisfactory. In this context, it is understood that the goal of humanization is always to maintain as many human sequences/amino acids as possible. In some embodiments, the amino acid residues of the vernier zones (vernier zone residues) of the human VH and VL frame work regions may be back mutated to the corresponding rabbit amino acids (of the VH/VL regions as shown in Table 1 ). In some embodiments, the first two amino acid residues in the light chain may be back mutated (from human to rabbit), if required. Alternatively, or additionally, the first 1 , 2 or 3 residues at the N terminus of the HV4/LV4 loops (light chain LV4 loop: Kabat positions 66-70; and heavy chain HV4 loop: Kabat positions 72-75 or 76) may be back mutated (from human to rabbit), if required. In some embodiments, an extra cysteine in position 80 (Kabat numbering) of the rabbit VL (as compared to the human VL) may be removed or replaced (substituted) in the humanized version. In some embodiments, T23 (Kabat numbering) of the rabbit VH is replaced, preferably by the corresponding amino acid of the human VH.
In some embodiments of a humanized (ASC_AB001 ) antibody, or antigen-binding fragment thereof, according to the present invention, the amino acid residues of the vernier zones (e.g. vernier zone FR3) of the human VH frame work region may be back mutated to the corresponding rabbit amino acids (see Figure 21 , graft ID Ab001_VHhum3). In some embodiments, the back mutated amino acid residues (e.g. vernier zone FR3) are refined, e.g. (at least partially) to the human sequences (see Figure 21 , graft ID Ab001_VHhum23). In some embodiments, a mutation from IGHJ2 to IGHJ1/4/5 is introduced into the framework region FR4 (see Figure 21 , graft ID Ab001_VHhum23). In particular embodiments, the above mutations are combined (see Figure 21 , graft ID Ab001_VHhum23).
In some embodiments, the light chain CDRL2 (DTSSLAS) may be humanized (DASSLAS) (see Figure 22, e.g. graft ID Ab001_VLhum4). In some embodiments, the light chain CDRL1 (QSSESVYNNSRLS) may be humanized (RASESVYNNSRLS) (see Figure 22, e.g. graft ID Ab001_VLhum5). In some embodiments, a (G- Q) mutation from IGKJ4 to IGKJ1 is introduced into the framework region FR4 (see Figure 22, e.g. graft ID Ab001_VLhum6). In some embodiments, a N29A mutation is introduced in LCDR1 thereby removing a glycosylation motif (see Figure 22, e.g. graft ID Ab001_VLhum10). In some embodiments, a N29Q mutation is introduced in LCDR1 (see Figure 22, graft ID Ab001_VLhum1 1 ). In some embodiments, a S31 A mutation is introduced in LCDR1 (see Figure 22, graft ID Ab001 _VLhum12). In particular embodiments, one or more of the above mutations are combined (see Figure 22, graft IDs Ab001_VLhum5, VLhum6, VLhumI O, VLhum11 , VLhuml 2).
Thus, in particular embodiments, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 5 or any one of SEQ ID NOs: 230 - 233, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
In a particular embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 5, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7. In a further particular embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 230, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
In a further particular embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 231 , a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
In a preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 232, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
In a further preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 233, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7.
Accordingly, in a particular embodiment, the antibody, or an antigen-binding fragment thereof according to the present invention, comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to a sequence according to any one of SEQ ID NO: 200, SEQ ID NO: 201 , and SEQ ID NO: 202; and a VL having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to a sequence according to any one of SEQ ID NO: 203, SEQ ID NO: 204, SEQ ID NO: 205, SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, and SEQ ID NO: 209, wherein the CDRH1 -3 and CDRL1 -3 sequences, as set forth above are preferably maintained.
In a preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to any one of SEQ ID NO: 200, SEQ ID NO: 201 , and SEQ ID NO: 202; and a VL comprising an amino acid sequence according to any one of SEQ ID NO: 203, SEQ ID NO: 204, SEQ ID NO: 205, SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, and SEQ ID NO: 209.
In a preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to SEQ ID NO: 202; and a VL having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to SEQ ID NO: 208, wherein the CDRH1 -3 sequences (according to SEQ ID NOs: 2, 3 and 4) and CDRL1 -3 sequences (according to SEQ ID NOs 232, 234 and 7) are preferably maintained. In a more preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to SEQ ID NO: 202; and a VL comprising an amino acid sequence according to SEQ ID NO: 208.
In a further preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to SEQ ID NO: 202; and a VL having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to SEQ ID NO: 209, wherein the CDRH1 -3 sequences (according to SEQ ID NOs: 2, 3 and 4) and CDRL1 -3 sequences (according to SEQ ID NOs 233, 234 and 7) are preferably maintained.
In a more preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to SEQ ID NO: 202; and a VL comprising an amino acid sequence according to SEQ ID NO: 209.
In some embodiments of a humanized (ASC_AB002) antibody, the light chain CDRL2 (GASTLAS) may be humanized (GASSLAS) (see Figure 23, graft ID Ab002_VLhum6). In some embodiments, a mutation (F- Y) is introduced into frame work region FR2 (see Figure 23, graft ID Ab002_VLhum1 1 , VLhum12). In some embodiments, a further (G->A) mutation is introduced into CDRL2 (see Figure 23, graft ID Ab001_VLhum11 , VLhum12). In some embodiments, the light chain CDRL1 (QSSQNVYSNNHLS) may be humanized (RASQNVYSNNHLS) (see Figure 23, graft ID Ab002_VLhum12). In particular embodiments, one or more of the above mutations are combined (see Figure 23, graft IDs Ab002_VLhum6, VLhum1 1 , VLhum12). In preferred embodiments, all of the above mutations are present in the VL of the antibody or antigen-binding fragment thereof (see Figure 23, graft ID Ab002_VLhum12).
Thus, in a particular embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 11 , a CDRL1 according to SEQ ID NO: 12 or SEQ ID NO: 235, a CDRL2 according to SEQ ID NO: 13, SEQ ID NO: 236 or SEQ ID NO: 237, and a CDRL3 according to SEQ ID NO: 14.
In a further particular embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 11 , a CDRL1 according to SEQ ID NO: 12, a CDRL2 according to SEQ ID NO: 236, and a CDRL3 according to SEQ ID NO: 14.
In a further particular embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 11 , a CDRL1 according to SEQ ID NO: 12, a CDRL2 according to SEQ ID NO: 237, and a CDRL3 according to SEQ ID NO: 14.
In a preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 1 1 , a CDRL1 according to SEQ ID NO: 235, a CDRL2 according to SEQ ID NO: 237, and a CDRL3 according to SEQ ID NO: 14. In a particular embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to a sequence according to SEQ ID NO: 210; and a VL having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to a sequence according to any one of SEQ ID NO: 21 1 , SEQ ID NO: 212, SEQ ID NO: 213, and SEQ ID NO: 214, wherein the CDRH1 -3 sequences (according to SEQ ID NOs: 2, 10 and 1 1 , respectively) and CDRL1 -3 sequences (according to SEQ ID NOs 12/235; SEQ ID NOs 13/236/237; and 14, respectively) are preferably maintained.
Thus, in a particular embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to SEQ ID NO: 210; and a VL comprising an amino acid sequence according to any one of SEQ ID NO: 211 , SEQ ID NO: 212, SEQ ID NO: 213, and SEQ ID NO: 214.
In a preferred embodiment, the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to SEQ ID NO: 210; and a VL having at least 70% identity (e.g., at least 70%, at least 71 %, at least 72%, at least 73%, at least 74%, at least 75%, at least 76%, at least 77%, at least 78%, at least 79% identity), preferably at least 80% identity (e.g. at least 80%, at least 81 %, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89% identity), more preferably at least 90% identity (e.g. at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity) to SEQ ID NO: 214, wherein the CDRH1 -3 sequences according to SEQ ID NOs: 2, 10 and 11 , respectively) and CDRL1 -3 sequences (according to SEQ ID NOs 235, 237 and 14, respectively) are preferably maintained.
Accordingly, in a more preferred embodiment the antibody, or an antigen-binding fragment thereof, according to the present invention comprises a VH comprising an amino acid sequence according to SEQ ID NO: 210; and a VL comprising an amino acid sequence according to SEQ ID NO: 214.
The antibody according to the present invention, or an antigen binding fragment thereof, may comprise an Fc moiety. The Fc moiety may be of any origin, preferably it is derived from human origin, e.g. from human IgA or IgG, such as IgGI , lgG2, lgG3, and/or lgG4, e.g. human IgG 1 .
As used herein, the term "Fc moiety" refers to a sequence derived from the portion of an immunoglobulin heavy chain beginning in the hinge region just upstream of the papain cleavage site (e.g., residue 216 in native IgG, taking the first residue of heavy chain constant region to be 1 14) and ending at the C-terminus of the immunoglobulin heavy chain. Accordingly, an Fc moiety may be a complete Fc moiety or a portion (e.g., a domain) thereof. A complete Fc moiety comprises at least a hinge domain, a CH2 domain, and a CH3 domain (e.g., EU amino acid positions 216-446). An additional lysine residue (K) is sometimes present at the extreme C-terminus of the Fc moiety, but is often cleaved from a mature antibody.
In some embodiments, in the context of the present invention an Fc moiety comprises at least one of: a hinge (e.g., upper, middle, and/or lower hinge region) domain, a CH2 domain, a CF13 domain, or a variant, portion, or fragment thereof. An Fc moiety may comprise at least a hinge domain, a CH2 domain or a CH3 domain. The Fc moiety may be a complete Fc moiety. The Fc moiety may also comprise one or more amino acid insertions, deletions, or substitutions relative to a naturally-occurring Fc moiety. For example, at least one of a hinge domain, CH2 domain or CH3 domain (or portion thereof) may be deleted.
In some embodiments, the antibody, or antigen binding fragment thereof, according to the present invention comprises an Fc region. As used herein, the term "Fc region" refers to the portion of an immunoglobulin formed by two or more Fc moieties of antibody heavy chains. For example, the Fc region may be monomeric or "single-chain" Fc region (i.e., a scFc region). Single chain Fc regions are comprised of Fc moieties linked within a single polypeptide chain (e.g., encoded in a single contiguous nucleic acid sequence). Exemplary scFc regions are disclosed in WO 2008/143954 A2. Preferably, the Fc region is dimeric. A "dimeric Fc region" or "dcFc" refers to the dimer formed by the Fc moieties of two separate immunoglobulin heavy chains. The dimeric Fc region may be a homodimer of two identical Fc moieties (e.g., an Fc region of a naturally occurring immunoglobulin) or a heterodimer of two non-identical Fc moieties.
In some embodiments, the Fc moiety, or the Fc region, comprises or consists of an amino acid sequence derived from a human immunoglobulin sequence (e.g., from an Fc region or Fc moiety from a human IgG molecule). However, the Fc moiety, or the Fc region, may comprise one or more amino acids from another mammalian species. For example, a primate Fc moiety or a primate binding site may be included in the antibody, or antigenbinding fragment. Alternatively, one or more murine amino acids may be present in the Fc moiety or in the Fc region.
The Fc moieties of the Fc region may be of the same or different class and/or subclass. For example, the Fc moieties may be derived from an immunoglobulin (e.g., a human immunoglobulin) of an IgGI , lgG2, lgG3 or lgG4 subclass.
It will be understood by one of ordinary skill in the art that the Fc moiety, or Fc region, may be modified such that it varies in amino acid sequence from the complete Fc moiety, or Fc region, of a naturally occurring immunoglobulin molecule. In some embodiments, the engineered Fc moiety, or Fc region, retains at least one desirable function conferred by the naturally-occurring Fc moiety. Fc functions, which may be modified by Fc engineering, or which may be retained, include Fc receptor (FcR) binding, antibody half-life modulation, ADCC function, protein A binding, protein G binding, and complement binding. The portions of naturally occurring Fc moieties, or Fc regions, which are responsible and/or essential for such functions are well known by those skilled in the art. Various Fc modifications are known in the art and described, for example, in Saunders KO. Conceptual Approaches to Modulating Antibody Effector Functions and Circulation Half-Life. Front Immunol. 2019 Jun 7; 10:1296. doi: 10.3389/fimmu.2019.01296, which is incorporated herein by reference in its entirety.
In some embodiments, the antibody according to the present invention comprises a (complete) Fc moiety/Fc region. The (complete) Fc moiety/Fc region may be engineered by one or more specific amino acid substitutions. In other embodiments, the may not be engineered (i.e., as occurring in a natural immunoglobulin, e.g. IgGI or lgG4).
Preferably, the Fc moiety/Fc region is engineered (e.g., based on a human IgG, such as IgG 1 or lgG4, Fc moiety/Fc region) to diminish Fc effector functions, in particular to reduce or inhibit FcyR and complement interactions. It has been demonstrated that auto-antibodies against ASC can opsonize ASC specks (via their Fc region) and, thereby, increase inflammation (Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmuller W, Latz E. The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation. Nat Immunol. 2014 Aug;15(8):727-37. doi: 10.1038/ni.2913). Various Fc modifications to reduce or inhibit FcyR and complement interactions are known in the art, which may be useful to avoid opsonization of anti-ASC antibodies. Specifically, Table 3 of Saunders KO. Conceptual Approaches to Modulating Antibody Effector Functions and Circulation Half-Life. Front Immunol. 2019 Jun 7; 10:1296. doi: 1 O.3389/fimmu.2O19.01296, which is incorporated herein by reference, provides examples of Fc modifications to silence antibody effector functions, which may be included in an antibody of the present invention. Accordingly, the antibody of the present invention may comprise an Fc modification selected from Leu235Glu, Leu234Ala/Leu235Ala, Ser228Pro/Leu235Glu, Gly237Ala, Glu318Ala, Leu234Ala/Leu235Ala/Pro329Gly, Pro331 Ser/Leu234Glu/Leu235Phe, Asp265Ala, Glu233Pro, Gly236Arg/Leu328Arg, lgG2- lgG4 cross-subclass, Ala330Leu, His268Gln/Val309Leu/Ala330Ser/Pro331 Ser, Val234Ala/Gly237Ala/Pro238Ser/His268Ala/Val309Leu/Ala330Ser/Pro331 Ser, Asp270Ala, Leu234Ala/L235Ala/Gly237Ala/P238Ser/His268Ala/Ala330Ser/Pro331 Ser, Lys322Ala, Pro329Ala, Pro331 Ala, lgG2-lgG3 cross-subclass, High mannose glycosylation, Val264Ala, Phe241 Ala, Asn297Ala_or_Gly_or_Gln, and S228P/Phe234Ala/Leu235Ala as described in Saunders KO. Conceptual Approaches to Modulating Antibody Effector Functions and Circulation Half-Life. Front Immunol. 2019 Jun 7; 10:1296. doi: 10.3389/fimmu.2019.01296, which is incorporated herein by reference.
Among such Fc modification diminishing effector functions the "LALA" mutation is preferred. The "LALA" mutation refers to the two substitutions L234A and L235A (EU numbering; also known as CH2 L4A and CH2 L5A mutation). The "LALA" mutation abolishes antibody binding to FcyRI, FcyRH and FcyRllla (Lund J, Winter G, Jones PT, Pound JD, Tanaka T, Walker MR et al. Human Fc gamma Rl and Fc gamma RII interact with distinct but overlapping sites on human IgG. J Immunol. 1991 ; 147:2657-2662). An exemplary amino acid sequence of CH1 -CH2-CH3 comprising the "LALA" mutation is set forth in SEQ ID NO: 141. Accordingly, the antibody preferably comprises an amino acid sequence according to SEQ ID NO: 141 or a sequence variant thereof, as described herein, wherein the "LALA" mutation is maintained.
In some embodiments, the "LALA" mutation may be combined with further mutations, such as P329G or G236R (EU numbering). In other embodiments, the antibody (Fc moiety/Fc region) includes the "LALA" mutation (L234A and L235A), while P329 and/or G236 are maintained (i.e., not mutated).
In general, binding of the antibody to an Fc receptor may be assessed by various methods known to the skilled person, such as ELISA (Hessell AJ, Hangartner L, Hunter M, Havenith CEG, Beurskens FJ, Bakker JM, Lanigan CMS, Landucci G, Forthal DN, Parren PWHI, et al.: Fc receptor but not complement binding is important in antibody protection against HIV. Nature 2007, 449:101-104; Grevys A, Bern M, Foss S, Bratlie DB, Moen A, Gunnarsen KS, Aase A, Michaelsen TE, Sandlie I, Andersen JT: Fc Engineering of Human IgGI for Altered Binding to the Neonatal Fc Receptor Affects Fc Effector Functions. 2015, 194:5497-5508) or flow-cytometry (Perez LG, Costa MR, Todd CA, Haynes BF, Montefiori DC: Utilization of immunoglobulin G Fc receptors by human immunodeficiency virus type 1 : a specific role for antibodies against the membrane-proximal external region of gp41. J Virol 2009, 83:7397-7410; Piccoli L, Campo I, Fregni CS, Rodriguez BMF, Minola A, Sallusto F, Luisetti M, Corti D, Lanzavecchia A: Neutralization and clearance of GM-CSF by autoantibodies in pulmonary alveolar proteinosis. Nat Commun 2015, 6:1-9).
Antibodies of the invention can be of any isotype (e.g., IgA, IgG, IgM i.e. an a, y or p heavy chain). Preferably, the antibody may be of the IgA or IgG type. Within the IgG isotype, antibodies may be IgGI , lgG2, lgG3 or lgG4 subclass, preferably IgGI or lgG4. Exemplified sequences for IgGI and lgG4 constant regions, which may be useful in the antibody as described herein, are provided in SEQ ID NO: 70 (IgGI ) and SEQ ID NO: 142 (lgG4). Accordingly, the antibody of the invention may comprise an amino acid sequence according to SEQ ID NO: 70 or SEQ ID NO: 142, or a sequence variant thereof as described herein. The human lgG4 constant region sequence of SEQ ID NO: 142 comprises the stable hinge mutation S228P (S. Angal, D.J. King, M.W. Bodmer, A. Turner, A.D.G. Lawson, G. Roberts, B. Pedley, J.R. Adair, A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (lgG4) antibody, Molecular Immunology, Volume 30, Issue 1 , 1993, Pages 105-108, ISSN 0161 -5890, https://doi.org/10.1016/0161 - 5890(93)90432-B).
Antibodies of the invention may have a K or a A light chain. Exemplified sequences for K and X light chain constant regions, which may be useful in the antibody as described herein, are provided in SEQ ID NO: 71 (Ckappa) and SEQ ID NO: 72 (Clambda). Accordingly, the antibody of the invention may comprise an amino acid sequence according to SEQ ID NO: 71 or 72, or a sequence variant thereof as described herein. Preferably, the antibody of the invention has a K light chain, e.g. having the constant region comprising or consisting of SEQ ID NO: 71 .
As outlined above, the present invention encompasses antigen-binding fragments. An antigen-binding fragment may or may not comprise an Fc moiety, in particular a portion of a complete Fc region. In some embodiments, the antibody, or antigen-binding fragment thereof, is selected from Fab, Fab', F(ab')2, Fv or scFv. For example, F(ab')2 (which may be obtained by pepsin cleavage or recombinant expression) as well as Fab' (which can be obtained from F(ab')2 or by recombinant expression) usually includes the hinge region.
In some embodiments, the antibody, or antigen-binding fragment, may be a single-chain antibody (or fragment). The single-chain antibody (or fragment) may encode the complete set of six CDRs, i.e. include the three heavy chain CDRs as well as the three light chain CDRs. More specifically, the single-chain antibody (or fragment) may include a heavy chain variable region (VH) as well as a light chain variable region (VL), for example including the VH and VL sequences as described above.
Variant antibodies are also included within the scope of the invention. Thus, variants of the sequences recited in the application are also included within the scope of the invention. Such variants include natural variants generated by somatic mutation in vivo during the immune response or in vitro upon culture of immortalized B cell clones. Alternatively, variants may arise due to the degeneracy of the genetic code or may be produced due to errors in transcription or translation.
Antibodies of the invention, or antigen-binding fragments thereof, may be provided in purified form. Typically, the antibody, or antigen-binding fragment, will be present in a composition that is substantially free of other polypeptides e.g., where less than 90% (by weight), usually less than 60% and more usually less than 50% of the composition is made up of other polypeptides.
Antibodies of the invention may be immunogenic in non-human (or heterologous) hosts e.g., in mice. In particular, the antibodies may have an idiotope that is immunogenic in non-human hosts, but not in a human host. In particular, antibodies of the invention for human use include those that cannot be easily isolated from hosts such as mice, goats, rabbits, rats, non-primate mammals, etc. and cannot generally be obtained by humanization or from xeno-mice.
Antibodies of the invention also include hybrid antibody molecules that comprise the six CDRs from an antibody of the invention as defined above and one or more CDRs from another antibody to an antigen. For example, the antibody may be multispecific. The multispecific antibody may comprise a first antigen-binding site according to the present invention (binding to ASC, in particular PYD of ASC as described herein) and a second antigen-binding site, which is different from the first antigen-binding site, i.e. which may bind to a distinct target. In other embodiments, the antibody, or the antigen-binding fragment thereof, may be monospecific.
Nucleic Acids
In another aspect, the invention also provides a nucleic acid molecule comprising a polynucleotide encoding the antibody according to the present invention, or an antigenbinding fragment thereof, as described above.
In some embodiments, the nucleic acid molecule comprises one or more polynucleotide(s) encoding the exemplified antibodies of the invention (e.g., as described above, in particular in Table 1), or a sequence variant thereof as described herein (e.g., having at least 70%, 71 %, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity as described above).
Exemplified nucleic acid sequences encoding the CDR and VH/VL sequences of exemplified antibodies as described herein are shown in Table 4 below.
Figure imgf000061_0001
Table 4: SEQ ID NOs for polynucleotide sequences encoding the CDR and VH/VL sequences of exemplary antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_AbOO4, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_Ab009.
Examples of nucleic acid molecules and/or polynucleotides include, e.g., a recombinant polynucleotide, a vector, an oligonucleotide, an RNA molecule such as an rRNA, an mRNA, an miRNA, an siRNA, or a tRNA, or a DNA molecule such as a cDNA. Nucleic acids may encode the light chain and/or the heavy chain of an antibody (or a single chain antibody). In other words, the light chain and the heavy chain of the antibody may be encoded by the same nucleic acid molecule (e.g., for single chain antibodies or for antibodies with separate heavy and light chains in bicistronic manner or an expression cassette containing more than one ribosome entry site such as IRES). Alternatively, the light chain and the heavy chain of the antibody may be encoded by distinct nucleic acid molecules.
Due to the redundancy of the genetic code, the present invention also comprises sequence variants of nucleic acid sequences, which encode the same amino acid sequences. The polynucleotide encoding the antibody (or the complete nucleic acid molecule) may be optimized for expression of the antibody. For example, codon optimization of the nucleotide sequence may be used to improve the efficiency of translation in expression systems for the production of the antibody. Moreover, the nucleic acid molecule may comprise heterologous elements (i.e., elements, which in nature do not occur on the same nucleic acid molecule as the coding sequence for the (heavy or light chain of) an antibody. For example, a nucleic acid molecule may comprise a heterologous promotor, a heterologous enhancer, heterologous UTR (e.g., for optimal translation/expression), a heterologous poly-A-tail, heterologous DNA insulator elements and the like.
A nucleic acid molecule is a molecule comprising nucleic acid components. The term nucleic acid molecule usually refers to DNA or RNA molecules. It may be used synonymous with the term "polynucleotide", i.e. the nucleic acid molecule may consist of a polynucleotide encoding the antibody. Alternatively, the nucleic acid molecule may also comprise further elements in addition to the polynucleotide encoding the antibody. Typically, a nucleic acid molecule is a polymer comprising or consisting of nucleotide monomers which are covalently linked to each other by phosphodiester-bonds of a sugar/phosphate-backbone. The term "nucleic acid molecule" also encompasses modified nucleic acid molecules, such as base-modified, sugar-modified or backbone-modified etc. DNA or RNA molecules.
In general, the nucleic acid molecule may be manipulated to insert, delete or alter certain nucleic acid sequences. Changes from such manipulation include, but are not limited to, changes to introduce restriction sites, to amend codon usage, to add or optimize transcription and/or translation regulatory sequences, etc. It is also possible to change the nucleic acid to alter the encoded amino acids. For example, it may be useful to introduce one or more (e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, etc.) amino acid substitutions, deletions and/or insertions into the antibody's amino acid sequence. Such point mutations can modify effector functions, antigen-binding affinity, post-translational modifications, immunogenicity, etc., can introduce amino acids for the attachment of covalent groups (e.g., labels) or can introduce tags (e.g., for purification purposes). Alternatively, a mutation in a nucleic acid sequence may be "silent", i.e. not reflected in the amino acid sequence due to the redundancy of the genetic code. In general, mutations can be introduced in specific sites or can be introduced at random, followed by selection (e.g., molecular evolution). For instance, one or more nucleic acids encoding any of the light or heavy chains of an (exemplary) antibody can be randomly or directionally mutated to introduce different properties in the encoded amino acids. Such changes can be the result of an iterative process wherein initial changes are retained and new changes at other nucleotide positions are introduced. Further, changes achieved in independent steps may be combined.
In some embodiments, the polynucleotide encoding the antibody, or an antigen-binding fragment thereof, (or the (complete) nucleic acid molecule) may be codon-optimized. The skilled artisan is aware of various tools for codon optimization, such as those described in: Ju Xin Chin, Bevan Kai-Sheng Chung, Dong-Yup Lee, Codon Optimization Online (COOL): a web-based multi -objective optimization platform for synthetic gene design, Bioinformatics, Volume 30, Issue 15, 1 August 2014, Pages 2210-2212; or in: Grote A, Hiller K, Scheer M, Munch R, Nortemann B, Hempel DC, Jahn D, JCat: a novel tool to adapt codon usage of a target gene to its potential expression host. Nucleic Acids Res. 2005 Jul 1 ; 33(Web Server issue):W526-31 ; or, for example, Genscript's OptimumGene™ algorithm (as described in US 201 1/0081708 A1 ).
For example, the nucleic acid molecule of the invention may comprise a nucleic acid sequence as set forth in any one of SEQ ID NOs 73 - 140; or a sequence variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity. Thereby, the nucleic acid molecule may encode any one of the exemplified antibodies ASC_Ab001, ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_AbOO5, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009 (by combining the sequences as shown in Table 2), or a sequence variant thereof as described herein.
In particular embodiments, the nucleic acid molecule of the invention may comprise a nucleic acid sequence as set forth in any one of SEQ ID NOs 215 - 229; or a sequence variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity. Thereby, the nucleic acid molecule may encode any one of the exemplified humanized antibodies ASC_Ab001_hum (RP02-F8), or ASC_Ab002_hum (RP03-C12).
The present invention also provides a plurality of nucleic acid molecules encoding the antibody, or an antigen-binding fragment thereof, as described herein, wherein each of the nucleic acid molecules (of the plurality of nucleic acid molecules) comprises a polynucleotide encoding an immunoglobulin chain of the antibody, or an antigen-binding fragment thereof. Thereby, the plurality of nucleic acid molecules, taken together, encodes (all of the immunoglobulin chains of) the antibody, or an antigen-binding fragment thereof, as described herein. In some embodiments, the plurality of nucleic acid molecules encoding the antibody, or an antigen-binding fragment thereof, as described herein, may be a combination of a first and a second nucleic acid molecule, wherein the first nucleic acid molecule comprises a polynucleotide encoding the heavy chain of the antibody, or an antigen-binding fragment thereof, of the present invention; and the second nucleic acid molecule comprises a polynucleotide encoding the corresponding light chain of the same antibody, or the same antigen-binding fragment thereof. In general, the above description regarding the (general) features of the nucleic acid molecule of the invention applies accordingly to the nucleic acid molecules of the plurality of nucleic acid molecules. Accordingly, one or more of the polynucleotides encoding the immunoglobulin chains of the antibody, or an antigen-binding fragment thereof, may be codon-optimized. For example, the plurality may comprise a nucleic acid sequence as set forth in any one of SEQ ID NOs 73 - 140; or a sequence variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity. Thereby, the plurality of nucleic acid molecules may encode any one of the exemplified antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_AbOO4, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_AbOO9 (by combining the sequences as shown in Table 2), or a sequence variant thereof as described herein.
In particular embodiments, the plurality may comprise a nucleic acid sequence as set forth in any one of SEQ ID NOs 215 - 229; or a sequence variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity. Thereby, the nucleic acid molecule may encode any one of the exemplified humanized antibodies ASC_Ab001_hum (RP02-F8), or ASC_AbOO2_hum (RP03-C12).
Vectors
Further included within the scope of the invention are vectors, for example, expression vectors, comprising a nucleic acid molecule according to the present invention or the plurality of nucleic acid molecules according to the present invention. Usually, a vector comprises a nucleic acid molecule as described above.
The present invention also provides a plurality of vectors comprising the plurality of nucleic acid molecules according to invention as described above. Thereby, each vector of the plurality of vectors may contain one or more nucleic acid molecules of the plurality of nucleic acid molecules according to invention as described above. In some embodiments, the plurality of vectors may be a combination of a first and a second vector, wherein the first vector comprises a first nucleic acid molecule as described above (for the combination of nucleic acid molecules) and the second vector comprises a second nucleic acid molecule as described above (for the combination of nucleic acid molecules).
A vector is usually a recombinant nucleic acid molecule, i.e. a nucleic acid molecule which does not occur in nature. Accordingly, the vector may comprise heterologous elements (i.e., sequence elements of different origin in nature). For example, the vector may comprise a multiple cloning site, a heterologous promotor, a heterologous enhancer, a heterologous selection marker (to identify cells comprising said vector in comparison to cells not comprising said vector), heterologous origin of replications, heterologous DNA insulator elements and the like. A vector in the context of the present invention is suitable for incorporating or harboring a desired nucleic acid sequence. Such vectors may be storage vectors, expression vectors, cloning vectors, transfer vectors etc. A storage vector is a vector which allows the convenient storage of a nucleic acid molecule. Thus, the vector may comprise a sequence corresponding, e.g., to a (heavy and/or light chain of a) desired antibody according to the present invention. An expression vector may be used for production of expression products such as RNA, e.g. mRNA, or peptides, polypeptides or proteins. For example, an expression vector may comprise sequences needed for transcription of a sequence stretch of the vector, such as a (heterologous) promoter sequence. A cloning vector is typically a vector that contains a cloning site, which may be used to incorporate nucleic acid sequences into the vector. A cloning vector may be, e.g., a plasmid vector or a bacteriophage vector. A transfer vector may be a vector which is suitable for transferring nucleic acid molecules into cells or organisms, for example, viral vectors. A vector in the context of the present invention may be, e.g., an RNA vector or a DNA vector. For example, a vector in the sense of the present application comprises a cloning site, a selection marker, such as an antibiotic resistance factor, and a sequence suitable for multiplication of the vector, such as an origin of replication. A vector in the context of the present application may be a plasmid vector.
As used herein, the term "vector" may also refer to a delivery vector, e.g. for viral or non- viral delivery of a nucleic acid of the invention. Alternatively, it may be referred to viral or non-viral delivery systems. Accordingly, the present invention also provides a delivery vector/system comprising the nucleic acid molecule as described above (or comprising an expression vector as described above). The delivery vector/system may be viral or non-viral. Various examples of viral and non-viral delivery vectors/systems are known in the art and described, for example, in Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Adv Biomed Res. 2012; 1 :27. doi: 10.4103/2277-9175.98152, which is incorporated herein by reference. Non-limiting examples of viral delivery vectors/systems include retroviral vectors; adenoviral vectors; adeno-associated viral (AAV) vectors, including helper-dependent adenoviral vectors and hybrid adenoviral vectors; herpes simplex virus vectors; lentivirus vectors; poxvirus vectors and Epstein-Barr virus vectors. Among the viral vectors, adenoviral vectors and adeno-associated viral (AAV) vectors are preferred. Non-limiting examples of non-viral delivery vectors/systems include chemical and non-chemical methods. Non-chemical delivery includes physical methods, such as electroporation and other methods for transient penetration of the cell membrane by mechanical, electrical, ultrasonic, hydrodynamic, or laser-based energy; naked DNA or RNA delivery; gene gun; hydrodynamic delivery; ultrasound delivery and magnetofection. Chemical non-viral delivery systems include cationic particles, in particular cationic lipids/liposomes, cationic polymers and lipid/polymer systems. Among non-viral vectors/systems, cationic liposomes are preferred.
Cells
In a further aspect, the present invention also provides a (host) cell expressing the antibody according to the present invention, or an antigen-binding fragment thereof; and/or comprising the vector (or the plurality of vectors) according the present invention. The (host) cell may be an isolated cell, which is not part of a human or animal body, e.g. a cell line or an engineered cell. The cell may express the nucleic acid(s) or vector(s) of the invention in a recombinant manner, e.g. in a heterologous manner (i.e., the cel l/cel I type does not express the antibody or the antigen-binding fragment in nature).
Examples of such cells include, but are not limited to, eukaryotic cells, e.g., yeast cells, animal cells or plant cells. Other examples of such cells include, but are not limited to, prokaryotic cells, e.g. £. coli. In some embodiments, the cells are mammalian cells, such as a mammalian cell line. Examples include human cells, CHO cells, HEK293 cells, PER.C6 cells, NSO cells, human liver cells, myeloma cells or hybridoma cells.
The cell may be transfected with a vector according to the present invention, for example with an expression vector. The term "transfection" refers to the introduction of nucleic acid molecules, such as DNA or RNA (e.g. mRNA) molecules, into cells, e.g. into eukaryotic or prokaryotic cells. In the context of the present invention, the term "transfection" encompasses any method known to the skilled person for introducing nucleic acid molecules into cells, such as into mammalian cells. Such methods encompass, for example, electroporation, lipofection, e.g. based on cationic lipids and/or liposomes, calcium phosphate precipitation, nanoparticle based transfection, virus based transfection, or transfection based on cationic polymers, such as DEAE-dextran or polyethylenimine etc. In some embodiments, the introduction is non-viral.
Moreover, the cells of the present invention may be transfected stably or transiently with the vector according to the present invention, e.g. for expressing the antibody according to the present invention. In some embodiments, the cells are stably transfected with the vector according to the present invention encoding the antibody according to the present invention. In other embodiments, the cells are transiently transfected with the vector according to the present invention encoding the antibody according to the present invention.
Accordingly, the present invention also provides a recombinant host cell, which heterologously expresses the antibody of the invention or the antigen-binding fragment thereof. For example, the cell may be of another species than the antibody (e.g., CHO cells expressing human antibodies). In some embodiments, the cell type of the cell does not express (such) antibodies in nature. Moreover, the host cell may impart a post-translational modification (PTM; e.g., glycosylation) on the antibody that is not present in their native state. Such a PTM may result in a functional difference (e.g., decreased immunogenicity). Accordingly, the antibody of the invention, or the antigen-binding fragment thereof, may have a post-translational modification, which is distinct from the naturally produced antibody (e.g., an antibody of an immune response in a human). Production of antibodies
Antibodies according to the invention can be made by any method known in the art. For example, the general methodology for making monoclonal antibodies using hybridoma technology is well known (Kohler, G. and Milstein, C., 1975; Kozbar et al. 1983).
Standard techniques of molecular biology may be used to prepare DNA sequences encoding the antibodies or antigen-binding fragments of the present invention. Desired DNA sequences may be synthesized completely or in part, e.g., using oligonucleotide synthesis techniques. Site-directed mutagenesis and polymerase chain reaction (PGR) techniques may be used as appropriate.
Any suitable host cell/vector system may be used for expression of the DNA sequences encoding the antibody molecules of the present invention. Eukaryotic, e.g., mammalian, host cell expression systems may be used for production of antibody molecules, such as complete antibody molecules. Suitable mammalian host cells include, but are not limited to, CHO, HEK293, PER.C6, NSO, myeloma or hybridoma cells. Also, prokaryotic, e.g. bacterial host cell expression systems may be used for the production of antibody molecules, such as complete antibody molecules. Suitable bacterial host cells include, but are not limited to, coli cells.
Accordingly, the present invention provides a method for preparing the antibody, or an antigen-binding fragment or an immunoglobulin chain(s) thereof, according to the present invention, said method comprising
(i) culturing the host cell as described above; and
(ii) isolating the antibody or immunoglobulin chain(s) thereof from the culture.
In other words, the present invention also provides a process for the production of an antibody molecule according to the present invention comprising culturing a (heterologous) host cell comprising a vector encoding a nucleic acid of the present invention, in particular under conditions suitable for expression of protein from DNA encoding the antibody molecule of the present invention, and isolating the antibody molecule. For production of the antibody comprising both heavy and light chains, a host cell, such as a cell line, may be transfected with two vectors, a first vector encoding a light chain polypeptide and a second vector encoding a heavy chain polypeptide, e.g. as described above. Alternatively, a single vector may be used, the vector including sequences encoding light chain and heavy chain polypeptides (e.g. for single chain antibodies or in a bicistronic manner).
Thus, the invention also provides a method for preparing a recombinant cell, comprising the steps of: (i) providing one or more nucleic acids that encode(s) the antibody of the invention; (ii) inserting the nucleic acid into an expression vector and (iii) transfecting the vector into a (heterologous) host cell in order to permit expression of the antibody of interest in that host cell. The nucleic acid of step (i) may, but need not, be manipulated to introduce restriction sites, to change codon usage, and/or to optimize transcription and/or translation regulatory sequences.
Furthermore, the invention also provides a method of preparing a transfected host cell, comprising the step of transfecting a host cell with one or more nucleic acids that encode an antibody of interest. Thus the procedures for first preparing the nucleic acid(s) and then using it to transfect a host cell can be performed at different times by different people in different places (e.g., in different countries).
These recombinant cells of the invention can then be used for expression and culture purposes. They are particularly useful for expression of antibodies for large-scale pharmaceutical production. They can also be used as the active ingredient of a pharmaceutical composition. Any suitable culture technique can be used, including but not limited to static culture, roller bottle culture, ascites fluid, hollow-fiber type bioreactor cartridge, modular minifermenter, stirred tank, microcarrier culture, ceramic core perfusion, etc.
The transfected host cell may be a eukaryotic cell, including yeast and animal cells, particularly mammalian cells (e.g., CHO cells, NSO cells, human cells such as PER.C6, HEK293 or HKB-11 cells, myeloma cells, or a human liver cell), as well as plant cells. In some embodiments, the transfected host cell is a mammalian cell, such as a human cell. In some embodiments, expression hosts can glycosylate the antibody of the invention, particularly with carbohydrate structures that are not themselves immunogenic in humans. In some embodiments the transfected host cell may be able to grow in serum-free media. In further embodiments the transfected host cell may be able to grow in culture without the presence of animal-derived products. The transfected host cell may also be cultured to give a cell line.
The invention also provides a method of preparing the antibody of interest comprising the steps of: culturing or sub-culturing a transfected host cell population, e.g. a stably transfected host cell population, under conditions where the antibody of interest is expressed and, optionally, purifying the antibody of interest. The transfected host cell population may be prepared by (i) providing nucleic acid(s) encoding a selected antibody of interest, (ii) inserting the nucleic acid(s) into an expression vector, (iii) transfecting the vector in a host cell that can express the antibody of interest, and (iv) culturing or sub-culturing the transfected host cell comprising the inserted nucleic acids to produce the antibody of interest.
In some embodiments, antibodies according to the invention may be produced by (i) expressing a nucleic acid sequence according to the invention in a host cell, e.g. by use of a vector (or host cell) according to the present invention, and (ii) isolating the expressed antibody product. Additionally, the method may include (iii) purifying the isolated antibody.
Accordingly, after production, the antibodies may be further purified, if desired, using filtration, centrifugation and various chromatographic methods such as HPLC or affinity chromatography. Techniques for purification of antibodies, e.g., monoclonal antibodies, including techniques for producing pharmaceutical-grade antibodies, are well known in the art.
Compositions and kits
The present invention also provides a composition comprising one or more of: (i) the antibody of the present invention, or an antigen-binding fragment thereof;
(ii) the nucleic acid or the plurality of nucleic acids of the present invention;
(iii) the vector or the plurality of vectors of the present invention; or
(iv) the cell expressing the antibody according to the present invention or comprising the vector according to the present invention.
The composition may be used for treatment or diagnostic purposes. Accordingly, the composition may be a pharmaceutical composition or a diagnostic composition. The composition may comprise a (pharmaceutically acceptable) excipient, diluent or carrier.
Accordingly, the present invention also provides a pharmaceutical composition comprising the antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid or the plurality of nucleic acids of the present invention, the vector or the plurality of vectors of the present invention, and/or the cell according to the present invention. Preferably, the pharmaceutical composition comprises the antibody, or an antigen-binding fragment thereof, according to the present invention.
The pharmaceutical composition may optionally also contain a pharmaceutically acceptable carrier, diluent and/or excipient. Although the carrier or excipient may facilitate administration, it should not itself induce the production of antibodies harmful to the individual receiving the composition. Nor should it be toxic. Suitable carriers may be large, slowly metabolized macromolecules such as proteins, polypeptides, liposomes, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers and inactive virus particles. In some embodiments, the pharmaceutically acceptable carrier, diluent and/or excipient in the pharmaceutical composition is not an active component in respect to ASC-related diseases or disorders. In some embodiments, the pharmaceutically acceptable carrier, diluent and/or excipient in the pharmaceutical composition is not an active component in respect to a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
Pharmaceutically acceptable salts can be used, for example mineral acid salts, such as hydrochlorides, hydrobromides, phosphates and sulphates, or salts of organic acids, such as acetates, propionates, malonates and benzoates. Pharmaceutically acceptable carriers in a pharmaceutical composition may additionally contain liquids such as water, saline, glycerol and ethanol. Additionally, auxiliary substances, such as wetting or emulsifying agents or pH buffering substances, may be present in such compositions. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries and suspensions, for ingestion by the subject.
Pharmaceutical compositions may be prepared in various forms. For example, the compositions may be prepared as injectables, either as liquid solutions or suspensions. Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared (e.g., a lyophilized composition, similar to Synagis™ and Herceptin®, for reconstitution with sterile water containing a preservative). The composition may be prepared for topical administration e.g., as an ointment, cream or powder. The composition may be prepared for oral administration e.g., as a tablet or capsule, as a spray, or as a syrup (optionally flavored). The composition may be prepared for pulmonary administration e.g., as an inhaler, using a fine powder or a spray. The composition may be prepared as a suppository or pessary. The composition may be prepared for nasal, aural or ocular administration e.g., as drops. The composition may be in kit form, designed such that a combined composition is reconstituted just prior to administration to a subject. For example, a lyophilized antibody may be provided in kit form with sterile water or a sterile buffer.
In some embodiments, the (only) active ingredient in the composition is the antibody, or the antigen-binding fragment thereof, of the present invention. As such, it may be susceptible to degradation in the gastrointestinal tract. Thus, if the composition is to be administered by a route using the gastrointestinal tract, the composition may contain agents which protect the antibody from degradation but which release the antibody once it has been absorbed from the gastrointestinal tract.
A thorough discussion of pharmaceutically acceptable carriers is available in Gennaro (2000) Remington: The Science and Practice of Pharmacy, 20th edition, ISBN: 0683306472. The present invention also provides a method of preparing a pharmaceutical composition comprising the steps of: (i) preparing an antibody of the invention; and (ii) admixing the purified antibody with one or more pharmaceutically acceptable excipients, diluents or carriers.
In other embodiments, a method of preparing a pharmaceutical composition comprises the step of: admixing an antibody with one or more pharmaceutically-acceptable carriers, wherein the antibody is a monoclonal antibody.
Pharmaceutical compositions may generally have a pH between 5.5 and 8.5, in some embodiments this may be between 6 and 8, for example about 7. The pH may be maintained by the use of a buffer. The composition may be sterile and/or pyrogen free. The composition may be isotonic with respect to humans. In some embodiments pharmaceutical compositions are supplied in hermetically-sealed containers.
Within the scope of the invention are compositions present in several forms of administration; the forms include, but are not limited to, those forms suitable for parenteral administration, e.g., by injection or infusion, for example by bolus injection or continuous infusion. Where the product is for injection or infusion, it may take the form of a suspension, solution or emulsion in an oily or aqueous vehicle and it may contain formulatory agents, such as suspending, preservative, stabilizing and/or dispersing agents. Alternatively, the antibody may be in dry form, for reconstitution before use with an appropriate sterile liquid.
A vehicle is typically understood to be a material that is suitable for storing, transporting, and/or administering a compound, such as a pharmaceutically active compound, in particular the antibodies as described herein. For example, the vehicle may be a physiologically acceptable liquid, which is suitable for storing, transporting, and/or administering a pharmaceutically active compound, in particular the antibodies as described herein. Once formulated, the compositions can be administered directly to the subject. In some embodiments the compositions are adapted for administration to mammalian, e.g., human subjects. Pharmaceutical compositions may include an antimicrobial, particularly if packaged in a multiple dose format. They may comprise detergent e.g., a Tween (polysorbate), such as Tween 80. Detergents are generally present at low levels e.g., less than 0.01 %. Compositions may also include sodium salts (e.g., sodium chloride) to give tonicity. For example, a concentration of 10±2mg/ml NaCI is typical.
Further, pharmaceutical compositions may comprise a sugar alcohol (e.g., mannitol) or a disaccharide (e.g., sucrose or trehalose) e.g., at around 15-30 mg/ml (e.g., 25 mg/ml), particularly if they are to be lyophilized or if they include material which has been reconstituted from lyophilized material. The pH of a composition for lyophilization may be adjusted to between 5 and 8, or between 5.5 and 7, or around 6.1 prior to lyophilization.
The pharmaceutical compositions may be administered by any number of routes including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intramedullary, intraperitoneal, intrathecal, intraventricular, transdermal, transcutaneous, topical, subcutaneous, intranasal, enteral, sublingual, intravagi nal or rectal routes. Optionally, the pharmaceutical composition may be prepared for oral administration, e.g. as tablets, capsules and the like, for topical administration, or as injectable, e.g. as liquid solutions or suspensions. In some embodiments, the pharmaceutical composition is an injectable. Solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection are also encompassed, for example the pharmaceutical composition may be in lyophilized form.
For injection, e.g. intravenous, cutaneous or subcutaneous injection, or injection at the site of affliction, the active ingredient may be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability. Those of relevant skill in the art are well able to prepare suitable solutions using, for example, isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection. Preservatives, stabilizers, buffers, antioxidants and/or other additives may be included, as required. Whether it is an antibody, a peptide, a nucleic acid molecule, or another pharmaceutically useful compound that is to be given to an individual, administration is usually in an "effective amount", e.g. in a "prophylactically effective amount" or a "therapeutically effective amount" (as the case may be), this being sufficient to show benefit to the individual. The actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. For injection, the pharmaceutical composition may be provided for example in a pre-filled syringe.
The pharmaceutical composition may also be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient, i.e. the antibody as defined above, is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
The pharmaceutical composition may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, e.g. including accessible epithelial tissue. Suitable topical formulations are readily prepared for each of these areas or organs. For topical applications, the pharmaceutical composition may be formulated in a suitable ointment, containing the pharmaceutical composition, particularly its components as defined above, suspended or dissolved in one or more carriers. Carriers for topical administration include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutical composition can be formulated in a suitable lotion or cream. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2- octyldodecanol, benzyl alcohol and water.
Dosage treatment may be a single dose schedule or a multiple dose schedule. For a single dose, e.g. a daily, weekly or monthly dose, the amount of the antibody in the pharmaceutical composition, may not exceed 1 g or 500 mg. In some embodiments, for a single dose, the amount of the antibody in the pharmaceutical composition, may not exceed 200 mg, or 100 mg. For example, for a single dose, the amount of the antibody in the pharmaceutical composition, may not exceed 50 mg. In some embodiments, the composition may include antibodies of the invention, wherein the antibodies may make up at least 50% by weight e.g., 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more) of the total protein in the composition. In the composition, the antibodies may be in purified form.
As an alternative to delivering antibodies for therapeutic purposes, it is possible to deliver nucleic acid (typically DNA) that encodes the monoclonal antibody of interest to a subject, such that the nucleic acid can be expressed in the subject in situ to provide a desired therapeutic effect. Suitable gene therapy and nucleic acid delivery vectors are known in the art.
Pharmaceutical compositions typically include an "effective" amount of one or more antibodies as described herein, i.e. an amount that is sufficient to treat, ameliorate, attenuate, decrease or prevent a desired disease or condition, or to exhibit a detectable therapeutic effect. Therapeutic effects also include reduction or attenuation in pathogenic potency or physical symptoms. The precise effective amount for any particular subject will depend upon their size, weight, and health, the nature and extent of the condition, and the therapeutics or combination of therapeutics selected for administration. The effective amount for a given situation is determined by routine experimentation and is within the judgment of a clinician. An effective dose may generally be from about 0.005 to about 100 mg/kg, for example from about 0.0075 to about 50 mg/kg or from about 0.01 to about 10 mg/kg. In some embodiments, the effective dose will be from about 0.02 to about 5 mg/kg, of the antibody (e.g. amount of the antibody in the pharmaceutical composition) in relation to the bodyweight (e.g., in kg) of the individual to which it is administered.
In some embodiments, the pharmaceutical composition may also comprise an additional active component, which may be a further antibody or a component, which is not an antibody. In other embodiments, the pharmaceutical composition may not comprise an additional active component (in addition to the antibody of the invention or respective nucleic acids, vectors or cells as described above).
Accordingly, the pharmaceutical composition may comprise one or more of the additional active components. The antibody of the invention can be present either in the same pharmaceutical composition as the additional active component or, alternatively, the antibody may be comprised by a first pharmaceutical composition and the additional active component may be comprised by a second pharmaceutical composition different from the first pharmaceutical composition. Accordingly, if more than one additional active component is envisaged, each additional active component and the antibody may be comprised in a different pharmaceutical composition. Such different pharmaceutical compositions may be administered either combined/simultaneously or at separate times or at separate locations (e.g. separate parts of the body), optionally by different routes of administration.
The antibody and the additional active component may provide an additive therapeutic effect, such as a synergistic therapeutic effect. The term "synergy" is used to describe a combined effect of two or more active agents that is greater than the sum of the individual effects of each respective active agent. Thus, where the combined effect of two or more agents results in "synergistic inhibition" of an activity or process, it is intended that the inhibition of the activity or process is greater than the sum of the inhibitory effects of each respective active agent. The term "synergistic therapeutic effect" refers to a therapeutic effect observed with a combination of two or more therapies wherein the therapeutic effect (as measured by any of a number of parameters) is greater than the sum of the individual therapeutic effects observed with the respective individual therapies.
The present invention also provides a diagnostic composition comprising an antibody according to the present invention, a nucleic acid(s) according to the present invention, a vector(s) according to the present invention, and/or a cell according to the present invention. The diagnostic composition may optionally comprise suitable means for detection, such as reagents conventionally used in immuno- or nucleic acid based diagnostic methods.
The antibodies described herein are, for example, suited for diagnostic purposes. Accordingly, they may be used in immunoassays, in which they can be utilized in liquid phase or bound to a solid phase carrier. Such immunoassays may be competitive or noncompetitive immunoassays; in either a direct or in an indirect format. Examples of such immunoassays include, but are not limited to, radioimmunoassay (RIA), enzyme-linked immunoassay (ELISA), sandwich (immunometric assay), immunohistochemistry, flow cytometry and Western blot assay. To this end, the antibody may be labelled, e.g. as described above.
In a further aspect the present invention also provides a kit comprising one or more of
(i) the antibody according to the present invention, or an antigen-binding fragment thereof, as described above,
(ii) the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention as described above,
(iii) the vector (or the plurality of vectors) according to the present invention as described above,
(iv) the cell according to the present invention as described above, and/or
(v) the composition according to the present invention as described above.
In addition, the kit may comprise means for administration of the antibody, or an antigen binding fragment thereof, according to the present invention, the nucleic acid according to the present invention, the vector according to the present invention, the cell according to the present invention or the pharmaceutical composition according to the present invention, such as a syringe or a vessel, a leaflet, and/or a co-agent to be administered as described herein. For example, the kit may contain a leaflet, e.g. comprising instructions for use. In addition or alternatively, the kit may comprise one or more reagents, e.g. for use in appropriate diagnostic assays. In some instances, the kit may contain a reference agent or control. In some embodiments, the composition of the invention may be provided in kit form, e.g., designed such that a combined composition is reconstituted just prior to administration to a subject. For example, a lyophilized antibody may be provided in kit form with sterile water or a sterile buffer (e.g., in a separate container).
Medical treatments and other uses
In a further aspect, the present invention provides the use of the antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention or the (pharmaceutical) composition according to the present invention as a medicament. In particular, the antibody according to the present invention, or an antigenbinding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention or the (pharmaceutical) composition according to the present invention may be used in prophylaxis and/or treatment of a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
Accordingly, the present invention also provides a method of treating, ameliorating or reducing a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder, or a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder, or lowering the risk of (occurrence of) a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder, or a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder, comprising: administering to a subject (in need thereof), (a therapeutically effective amount of) the antibody, or an antigen-binding fragment thereof, according to the present invention, a nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, a vector (or the plurality of vectors) according to the present invention, a cell according to the present invention or a (pharmaceutical) composition according to the present invention. Moreover, the present invention also provides the use of an antibody according to the present invention, or an antigen-binding fragment thereof, a nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, a vector (or the plurality of vectors) according to the present invention, a cell according to the present invention, or a pharmaceutical composition according to the present invention in the manufacture of a medicament for prophylaxis, treatment or attenuation of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder, or of a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder.
As used herein, the terms "treat" or "treatment" include therapeutic treatment and prophylactic or preventative measures. Prophylaxis of a neurodegenerative or neuroinflammatory disease refers in particular to prophylactic settings, wherein the subject was either not diagnosed with a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder (either no diagnosis was performed or diagnosis results were negative) and/or the subject does not show symptoms of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder. In therapeutic settings, in contrast, the subject is typically diagnosed with a neurodegenerative or neuroinflammatory disease and/or showing symptoms of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder. Of note, the terms "treatment" and "therapy'7"therapeutic" include (complete) cure as well as attenuation/reduction of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder and/or related symptoms.
In general, the object of the "treatment" may be to decrease, ameliorate, inhibit, prevent or slow down (lessen or delay) an undesired physiological change or disorder, such as the neurodegenerative or neuroinflammatory disease or a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder. Beneficial or desired clinical results of a treatment include, but are not limited to, alleviation of symptoms, diminishment of the extent of a disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival, e.g. as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder as well as those prone to have the condition or disorder or those in which the manifestation of the condition or disorder or the risk thereof is to be decreased, delayed or prevented.
One way of checking efficacy of therapeutic treatment involves monitoring disease symptoms after administration of the antibody or of the composition. Treatment can be a single dose schedule or a multiple dose schedule. In some embodiments, an antibody, antibody fragment, nucleic acid, vector, cell, or composition as described herein may be administered to a subject in need of such treatment. Such a subject includes, but is not limited to, one who is particularly at risk of, or susceptible to, a neurodegenerative or neuroinflammatory disease or a symptom of a neurodegenerative or neuroinflammatory disease or of a peripheral inflammatory disorder. The antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention or the (pharmaceutical) composition according to the present invention may be used for human and also for veterinary medical purposes, preferably for human medical purposes. The term „subject", „patient" or „i ndi vidual" as used herein thus generally includes humans and non-human animals and preferably mammals (e.g., non-human primates, including marmosets, tamarins, spider monkeys, owl monkeys, vervet monkeys, squirrel monkeys, and baboons, macaques, chimpanzees, orangutans, gorillas; cows; horses; sheep; pigs; chicken; cats; dogs; mice; rat; rabbits; guinea pigs; etc.), including chimeric and transgenic animals and disease models. In the context of the present invention, the term „subject" preferably refers a non-human primate or a human. Most preferably, the subject is a human. The human may be selected from the group of a human suffering from a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder, a human at risk of developing a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder, and a human of unknown clinical history for a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
Preferably, the "neurodegenerative or neuroinflammatory disease" is characterized by or associated with the formation of ASC aggregates ("specks") and/or amyloid-p (Ap)-related pathology, in particular the formation and spreading of amyloid-P aggregates. The term "Ap- related pathology" refers to the abnormal production, deposition and aggregation of amyloid-p in the brain. In particular, the neurodegenerative or neuroinflammatory disease may involve interaction of ASC and amyloid-p. In some embodiments, the neurodegenerative or neuroinflammatory disease is associated with the fibrillation activity of ASC. In some embodiments, the neurodegenerative or neuroinflammatory disease involves ASC-induced inflammation.
Neurodegenerative diseases are typically chronic, progressive disorders characterized by the gradual loss of neurons in discrete areas of the central nervous system (CNS), such as the brain. The neurodegenerative diseases envisaged to be treated may preferably be characterized and/or accompanied by dementia. "Dementia" is a general term for a decline in mental ability severe enough to interfere with daily life. Dementia may include decline or loss of memory, communication and language, ability to focus and pay attention, reasoning and judgment, visual perception, or a combination thereof. It may be caused by neurodegeneration in a variety of neurodegenerative diseases.
Neuroinflammation is the inflammation of nervous tissue. It may be initiated in response to a variety of cues, including infection, traumatic brain injury, toxic metabolites or autoimmunity. Neuroinflammatory diseases are characterized by inflammatory processes in the nervous system, in particular in the CNS, which may result in the gradual damage and loss of neurons. Neuroinflammation and neurodegeneration often result from the aberrant deposition of aggregated host proteins that can activate inflammasomes. ASC is a central component of the inflammasome.
Preferably, the neurodegenerative or neuroinflammatory disease is selected from Alzheimer's Disease, Parkinsons's Disease, Huntington's disease, Multiple System Atrophy, Amyotrophic Lateral Sclerosis, Multiple Sclerosis, traumatic brain injury, Sinocerebellar ataxia, Frontotemporal Dementia, Frontotemporal Lobar Degeneration, Mild Cognitive Impairment, Parkinson-plus syndromes, Pick disease, Progressive isolated aphasia, Greymatter degeneration [Alpers], Subacute necrotizing encephalopathy, or Lewy body dementia. Alzheimer's Disease (AD) and Mild Cognitive Impairment (MCI) are particularly preferred.
"Alzheimer's Disease" is a neurodegenerative brain disease that is a major cause of dementia among the elderly. Symptoms of AD may include progressive loss of learning and memory functions, personality changes, neuromuscular changes, seizures and occasionally psychotic behaviour. Alzheimer's disease is characterized by the deposition of amyloid-P plaques in areas of the brain that are critical for memory and other cognitive functions. It is believed that the deposition of amyloid-P plaques, in these critical areas of the brain, interferes with brain functions.
"Mild Cognitive Impairment" is a syndrome defined as a subjective and objective decline in cognition and function greater than expected for an individual's age and education level that may not meet the criteria for a diagnosis of dementia. However, elderly patients diagnosed with MCI constitute a high-risk population for developing dementia, in particular Alzheimer's disease (AD) but also other kinds of dementia, such as vascular dementia, frontotemporal dementia (FTD) and dementia with Lewy bodies (DLB). MCI is often referred to as an objective cognitive complaint for age, in a person with essentially normal functional activities, who does not have dementia. It affects 19% of people aged 65 and over. Around 46% of people with MCI develop dementia within 3 years compared with 3% of the population of the same age.
Further non-limiting examples of neurodegenerative or neuroinflammatory diseases envisaged for prophylaxis or treatment according to the present invention include hereditary ataxia, congenital nonprogressive ataxia, early-onset cerebellar ataxia, late-onset cerebellar ataxia, cerebellar ataxia with defective DNA repair, hereditary spastic paraplegia, infantile spinal muscular atrophy, type I [Werdnig-Hoffman], inherited spinal muscular atrophy, systemic atrophies primarily affecting the central nervous system, paraneoplastic neuromyopathy and neuropathy, postpolio syndrome, Degenerative diseases of basal ganglia, Hallervorden-Spatz disease, progressive supranuclear ophthalmoplegia [Steele- Richardson-Olszewski], Neurogenic orthostatic hypotension [Shy-Drager], dystonia, tremor, chorea, Restless legs syndrome, Stiff-man syndrome, extrapyramidal and movement disorders, Multiple sclerosis, acute disseminated demyelination, Neuromyelitis optica [Devic], Acute and subacute haemorrhagic leukoencephalitis [Hurst], Periaxial encephalitis, Schilder disease, Central demyelination of corpus callosum, Central pontine myelinolysis, Acute transverse myelitis in demyelinating disease of central nervous system, Subacute necrotizing myelitis, Concentric sclerosis, Epilepsy, Localization-related (focal)(partial) idiopathic epilepsy and epileptic syndromes with seizures of localized onset, Localization- related (focal)(partial) symptomatic epilepsy and epileptic syndromes with simple partial seizures, Localization-related (focal)(partial) symptomatic epilepsy and epileptic syndromes with complex partial seizures, myoclonic epilepsy in infancy, neonatal convulsions (familial), Childhood absence epilepsy [pyknolepsy], absence epilepsy, myoclonic epilepsy [impulsive petit mal], epilepsy with myoclonic absences, myoclonic-astatic seizures, Infantile spasms, Lennox-Gastaut syndrome, Salaam attacks, Symptomatic early myoclonic encephalopathy, West syndrome, Epilepsia partialis continua [Kozhevnikof], Grand mal seizures, Petit mal, Status epilepticus, Grand mal status epilepticus, Petit mal status epilepticus, Complex partial status epilepticus, Migraine, Cluster headache syndrome, Vascular headache, Tension-type headache, Chronic post-traumatic headache, Narcolepsy and cataplexy, Kleine-Levin syndrome.
Examples of a peripheral inflammatory disorder are, without being limited thereto, inflammatory bowel disease, non-alcoholic steatohepatitis, Beh et disease, sarcoidosis, lupus erythematosus, vasculitis, rheumatoid arthritis.
The antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention, or the pharmaceutical composition according to the present invention may be administered by any route of administration including, but not limited to, oral, intravenous, intramuscular, intra-arterial, intraperitoneal, transdermal, transcutaneous, topical, subcutaneous, intranasal, enteral, sublingual or rectal routes. In addition, any gene therapy approaches may be used, e.g. the antibody according to the present invention, or an antigen-binding fragment thereof, may be administered as nucleic acid or vector encoding said antibody, e.g. using viral or non-viral vectors as described above. In some embodiments, the antibody according to the present invention, or an antigen-binding fragment thereof, the nucleic acid molecule (or the plurality of nucleic acid molecules) according to the present invention, the vector (or the plurality of vectors) according to the present invention, the cell according to the present invention or the pharmaceutical composition according to the present invention may be administered systemically, for example by intravenous or subcutaneous administration.
Further uses
Antibodies and fragments thereof as described herein may also be used for the (jn-vitrd) diagnosis of a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder. Methods of diagnosis may include contacting an antibody with a sample. Such samples may be isolated from a subject, for example an isolated blood sample, such as whole blood, plasma or serum. The methods of diagnosis may also include the detection of an antigen/antibody complex, in particular following the contacting of an antibody with a sample. Furthermore, it may be tested, whether the sample contains antibodies competing with the antibodies as described herein, e.g. for allergen binding. This is typically performed in vitro, i.e. without any contact to the human or animal body. Examples of analytical methods are well-known to the person skilled in the art and include immunoassays such as flow cytometry, dot or slot blots, Western blots, ELISA (enzyme- linked immunosorbent assay), e.g. for cross-competition, immunohistochemistry and immunoprecipitation followed by SDS-PAGE immunocytochemistry. Accordingly, the diagnosis may be performed in vitro, for example by using an isolated sample as described above (and an in vitro analysis step as described above).
Accordingly, the present invention also provides the use of the antibody, or an antigenbinding fragment thereof, according to the present invention, the nucleic acid molecule or the plurality of nucleic acid molecules according to the present invention, the vector or the plurality of vectors according to the present invention, the cell according to the present invention, the composition according to the present invention, or the kit according to the present invention in in-vitro) diagnosis of a neurodegenerative or neuroinflammatory disease or a peripheral inflammatory disorder.
BRIEF DESCRIPTION OF THE FIGURES
In the following a brief description of the appended figures will be given. The figures are intended to illustrate the present invention in more detail. However, they are not intended to limit the subject matter of the invention in any way.
Figure 1 shows for Example 3 binding curves of exemplary antibody ASC_Ab001 to full length ASC, ASG specks, CARD domain, PYD domain, murine ASC and BSA as tested by ELISA.
Figure 2 shows for Example 4 an alignment of the different ASC isoforms: full-length ASC (SEQ ID NO: 1 ); ASC-b (SEQ ID NO: 145); ASC-c (SEQ ID NO: 146) and ASC-d (SEQ ID NO: 147). Figure 3 shows for Example 4 representative images of binding patterns of (A) ASC_Ab001 , (B) ASC_Ab008, (C) ASC_Ab004, and (D) control antibody (AL- 177, adipogen). For all blots, lane 1 shows binding to ASC-b 20kDa, lane 2 shows binding to ASC-d 1 1 .8kDa , lane 3 shows binding to ASC-c 15kDa, lane 4 shows binding to ASC Y146A 21 ,5kDa, lane 5 shows binding to ASC K21 A K22A K26A 21.5kDa, lane 6 shows binding to UT Ctrl, and lane 7 shows binding to ASC-HIS purified 23kDa.
Figure 4 shows for Example 4 the binding of anti-ASC antibodies and control antibodies as indicated to ASC peptides as indicated. (A) Binding of ASC_Ab001 , ASC_AbOO2 clonotype, ASC_Ab004, ASC_Ab005, ASC_AbOO6, ASC_AbOO7, ASC_AbOO8 clonotype, AL-177 and VHHASC to peptides covering amino acids 1 to 20 and 1 1 to 30 of ASC. (B) Binding of ASC_Ab001 , ASC_AbOO2, AL-177, IC100 and VHHASC to peptides covering amino acids 1 to 20, 1 1 to 30 and 21 to 40 of ASC.
Figure s shows for Example 5 ELISA binding curves of antibodies ASC_Ab001 , ASC_AbOO2, AL-177, VHHASC and IC100 to human ASC.
Figure 6 shows for Example 5 ELISA binding curves of antibodies ASC_Ab001 , ASC_Ab002, AL-177, VHHASC and IC100 to murine ASC.
Figure 7 shows for Example 5 ELISA binding curves of antibodies ASC_Ab001 , ASC_Ab002, AL-177, VHHASC and IC100 to ASC specks.
Figure 8 shows for Example 5 ELISA binding curves of antibodies ASC_Ab001 , ASC_Ab002, AL-177, VHHASC and IC100 to the PYD domain of ASC.
Figure 9 shows for Example 6 (A) the quantification of ASC specks detected in Thp1 wt or asc ''- cells primed with LPS (1 ug/mL), stimulated with Nigericin (5uM) and stained with 1 ug/mL of anti-ASC antibodies or isotype control and (B) representative immunofluorescence pictures of THP-1 asc +/+ and THP-1 asc-/- cells stained with an ASC_Ab001 antibody.
Figure 10 shows for Example 7 Western blot images of antibodies ASC_Ab001 , ASC_Ab004, ASC„Ab007 and ASC_AbOO9 on cell lysates of wt and asc" human THP-1 monocytes.
Figure 1 1 shows for Example 7 Western blot images of antibodies ASC_Ab001 , ASC_Ab004, ASC_Ab007 and ASC_AbOO9 on cell lysates of mouse bone- marrow derived macrophages (BMDMs).
Figure 12 shows for Example 8 that treatment with ASC_Ab001 , ASC_Ab004, ASC_Ab007, ASC_Ab008 and ASC_Ab009 significantly impaired ASC fibrillation. The effect of anti-ASC antibodies on ASC fibrillation was assessed by IF of ASC fibrils formed after incubation of ASC-gfp with anti-ASC antibodies or isotype control 1 h at 37°C. (A) Representative picture and (B) quantification of number of ASC fibrils per well upon treatment with anti- ASC antibodies. Pooled data from n=1 or 2 independent experiments. One way ANOVA followed by Dunett's multiple comparison test, ns: nonsignificant, ****: p-value<0.0001
Figure 13 shows for Example 8 the results of a sedimentation assay to assess antifibrillation properties of anti-ASC antibodies. Soluble and insoluble fractions of ASC were separated by high-speed centrifugation upon treatment with anti-ASC antibodies or isotype control. Treatment with ASC_Ab001 lead to more soluble ASC (supernatant fraction) when compared to ASC only or isotype control after 30 min of incubation at 37°C. Or in other words, ASC_Ab001 shifts the equilibrium towards the soluble ASC in the supernatant fraction. Treatment with an anti-CARD antibody had no effect on the fibrillation.
Figure 14 shows for Example 9 that the anti-ASC antibodies block ASC-Ap interaction. Percentage of inhibition of ASC- A0 binding after treatment with anti-ASC antibodies (3.5uM) or isotype control (3.5uM). One way ANOVA followed by Dunett's multiple comparison test ***: p-value<0.001
Figure 15 shows for Example 10 that ASC_Ab001 blocks the pro- infl mmatory function of extracellular ASC fibrils. The in-vitro efficacy of ASC_Ab001 was tested in a cell-based assay using differentiated human THP-1 macrophages that were treated with extracellular ASC fibrils. Before addition of the ASC fibrils to the cells, ASC fibrils were pre-incubated with ASC_Ab001 and isotype control antibody. IL-1 |3 measurement was used as a readout for inflammasome activation. ASC_Ab001 significantly reduced the ASC fibrils-induced production of IL-10 compared to the isotype control.
Figure 16 shows for Example 11 quantification of IL-1 beta release by differentiated human macrophages treated with ASC fibrils and 0.0035nM or 35nM of anti- ASC antibodies (ASC_Ab001 - ASC_Ab009) and isotype controls, respectively, in an ASC seeded Inflammation assay.
Figure 17 shows for Example 12 that anti-ASC antibodies (ASC„Ab001 - ASC_AbOO9) do not bind inflammasome proteins AIM2 and NRLP3 having homologous sequences to ASC, determined by a binding ELISA.
Figure 18 shows for Example 13 that anti-ASC antibodies ASC_Ab001 and ASC_Ab002 ameliorate the severity of dextran sulfate sodium (DSS)-induced colitis in an acute inflammatory bowel disease model. Measurement of (A) body weight; (B) disease activity index; and (C) colon length. Data represent mean +SEM. *p<0.05, **p<0.01 , ***p<0.001, ****p<0.0001 in two-way ANOVA with multiple comparisons to isotype.
Figure 19 shows for Example 13 representative photos of haematoxylin and eosin staining of colon, N=8 mice per group (A: healthy; B: isotype control; C: ASC_Ab001 ). Figure 19D is a diagram illustrating infiltration of inflammatory cells into the mucosa of healthy, isotype treated and Asc_Ab001 treated mice. Figure 20 shows for Example 14 that humanized ASC_Ab001 preserves binding to ASC. Binding of humanized ASC_Ab001 to ASC-His was assessed by ELISA. Similar binding of the humanized antibodies was observed compared to the rabbit IgG format.
Figure 21 shows the VH amino acid sequences of humanized Ab001 antibodies. All sequences for the heavy chain are based on the IGHV3-66 germline, which has the highest homology to the original rabbit Ab001 heavy chain framework and has also been reported to be a good acceptor framework for rabbit CDRs. Humanized Ab001 antibodies comprising Ab001_VHhum23 were used in Example 15.
Figure 22 shows the VL amino acid sequences of humanized antibodies. All sequences for the light chain are based on the IGKV1 -5 germline, which has the highest homology to the original rabbit Ab001 light chain framework and is very similar to the 1GKV1-27*O1 germline, previously used as template for a generic acceptor frame for rabbit light chain CDRs. Humanized Ab001 antibodies comprising Ab001_VLhum11 and Ab001_VLhum12, respectively, were used in Example 15.
Figure 23 shows the VH and VL amino acid sequences of humanized Ab002 antibodies. The presented sequence for the heavy chain is based on the IGHV3-66 germline, which is has the highest homology to the original rabbit Ab002 heavy chain framework and has also been reported to be a good acceptor framework for rabbit CDRs. All sequences for the heavy chain are based on the IGKV1 -27 germline, which has the highest homology to the original rabbit Ab002 light chain framework and was previously used as template for a generic acceptor frame for rabbit light chain CDRs. A humanized Ab002 antibody comprising Ab002_VHhum17 and Ab002_VLhum12 was used in Example 15. Figure 24 shows for Example 15 kinetic measurements of humanized Ab001 Fabs: Ab001 VH23/VL11 (A) and Ab001 VH23/VL12 (B) versus non-humanized Ab001 VHrab/VLrab (C) by Bio-Layer Interferometry (BLI) using a RED96e OCTET system (Sartorius AG).
Figure 25 shows for Example 15 kinetic measurements of humanized Ab002 Fabs: Ab002 VH16/VL12 (A) versus non-humanized Ab002 VHrabA/Lrab (B) by Bio-Layer Interferometry (BLI) using a RED96e OCTET system (Sartorius AG).
Figure 26 shows for Example 16 binding of full IgGI humanized Ab001 antibodies Ab001 VH23/VL1 1 and Ab001 VH23/VL12, as well as Ab001 rb-huIgG (non-humanized) to human ASC (A) and murine ASC (B), measured by binding ELISA.
Figure 27 shows for Example 16 binding of full IgGI humanized Ab002 antibody Ab002 VH1 7/VL12 as well as Ab002 rb-huIgG (non-humanized) to human ASC (A) and murine ASC (B), measured by binding ELISA.
EXAMPLES
In the following, particular examples illustrating various embodiments and aspects of the invention are presented. However, the present invention shall not to be limited in scope by the specific embodiments described herein. The following preparations and examples are given to enable those skilled in the art to more clearly understand and to practice the present invention. The present invention, however, is not limited in scope by the exemplified embodiments, which are intended as illustrations of single aspects of the invention only, and methods which are functionally equivalent are within the scope of the invention. Indeed, various modifications of the invention in addition to those described herein will become readily apparent to those skilled in the art from the foregoing description, accompanying figures and the examples below. All such modifications fall within the scope of the appended claims. Methods
Production of recombinant ASC specks in HEK293
Full-length tag-free human ASC (SEQ ID NO: 1 ) and ASC-GFP (SEQ ID NO: 152) was produced as intracellular specks in HEK293 and purified according to Martin-Sanchez F, Gomez Al, Pelegrfn P. Isolation of Particles of Recombinant ASC and NLRP3. Bio Protoc. 2015 May 20;5(10):e1480. doi: 10.21769/BioProtoc.1480. In brief, suspension HEK293 cells were transiently transfected with expression plasmids encoding full length human ASC or ASC-GFP using linear PEI 40kDa. After 7 days of expression at 37°C and 5% CO2, cells were harvested and resuspended in Buffer A (320 mM sucrose, 20 mM HEPES-KOH (pH 7.5), 10 mM KCI, 1.5 mM MgCb, 1 mM, EDTA, 1 mM EGTA. Cells were lysed by syringing (10x 20G, 20x 25G), freeze-thawing (3 x), followed by subsequent syringing (20x 25G). Afterwards the lysate was centrifuged at 400g for 8min, the pellet was re-suspended in 2x CHAPS buffer (40 mM HEPES-KOH (pH 7.5), 10 mM MgCb, 1 mM EGTA, 0.2 mM PMSF, 0.2 % CHAPS) and filtered using a 5 pm centrifugal filters at 2000g for 10 min. The filtrate was then diluted and gently mixed with 1 volume of 2x CHAPS buffer and centrifuged at 2300g for 8 min. The resulting pellet was resuspended in 1 ml of 1 x CHAPS buffer and centrifuged at 5000g for 8 min. This washing step was repeated twice. Afterwards the pellet was re-suspended in 1 x CHAPS buffer and loaded carefully on the top of 40% Percoll and centrifuged at 16000g for 10min. The interface layer containing the ASC speck particles was collected carefully and washed once by centrifugation at 5000g for 3min and resuspension in 1 x CHAPS buffer. Lastly, fluorescent particles were quantified in a fluorescence microscope using a Burker chamber.
Production of monomeric ASC in £ coli
Monomeric human ASC-His (SEQ ID NO: 153), ASC-GFP-His (SEQ ID NO: 154) and the ASC PYD domain (ASCPYD-His-SII; SEQ ID NO: 155) as well as the ASC CARD domain (ASCCARD-His-SII; SEQ ID NO: 156) were expressed in E. coli and purified from inclusion bodies via Nickel beads. Therefore, £ co// strain BL21 (DE3) was transformed with pET- based vectors encoding the his-tagged ASC variants. Expression was done in self-inducing media (MagicMedia™, Invitrogen) incubated at 37°C for 1 h and 67h at 20°C. Subsequently, cells were harvested by centrifugation at 4000rpm for 30min. For cell lysis, the pellet was resuspended in 50 mM phosphate, 300 mM NaCl, pH 7.5 and sonicated for on ice 10 min at 40% power (2 sec pulse/pause). The suspension was then centrifuged at 14000g at 4°C for 30min to collect the pellet containing the inclusion bodies. To solubilize the inclusion bodies, pellets were resuspended in 50 mM phosphate, 300 mM NaCI, 6M Gua-HCl, 2 mM DTT, pH 7.5 for 30min at RT. Afterwards the suspension was centrifuged at 14000g at 4°C for 30min to remove residual insoluble cell debris. The supernatant was then incubated with Nickel beads (Themo, #88221 ) for 3h at RT., The beads were washed once with 50 mM phosphate, 300 mM NaCI, 6M Gua-HCl, 2 mM DTT, pH 7.5 and followed by 50 mM phosphate, 300 mM NaCI, 6M Gua-HCl, 2 mM DTT, 20mM imidazole pH 7.5. To elute the His-tagged proteins, beads were incubated with 50 mM phosphate, 300 mM NaCI, 6M Gua-HCl, 2 mM DTT, 500mM imidazole pH 7.5. The pH of the eluate was adjusted to pH 3.8 with diluted HCI. Next, the eluate was dialysed against 50 mM Glycine, 150 mM NaCI, pH 3.8 in 3500 Da cassette O/N at 4°C. To remove higher order aggregates, the dialysed samples were purified using a preparative SEC column (HiLoad 16/600 Superdex 75 pg). The monomeric proteins were finally concentrated using a VivaSpin 3000 Da column.
ELISA
High binding clear flat bottom 384-well plate (Greiner bio-one, 781061 ) were coated o/n (4°C) with 10 pl/well of ASC CARD domain (1.25ug/mL; SEQ ID NO: 156), ASC PYD domain (1.25ug/mL; SEQ ID NO: 155), murine ASC (Cusabio, CSB-EP861664 MO, 1.25ug/mL; SEQ ID NO: 151 ), ASC-His (1 ug/mL; SEQ ID NO: 153), ASC-gfp specks (50 000 specks/well) or BSA (1 pg/ml) diluted in PBS. For testing of the binding to ASC peptides (SEQ ID NOs 148 - 150), the same plates were pre-coated with 1 ug/mL streptavidin (Sigma, cat S4762) then incubated with 1 ug/mL of peptide o/n (4°C). After o/n incubation, wells were aspirated and washed once with 100 pl/well of wash buffer (0.05% Tween-20 in PBS). Wells were blocked for an hour with 50 pl of 2% BSA diluted in PBS at RT. After blocking, wells were incubated 2 hours at RT with 10 pl of test antibody and serially diluted. Wells were washed four times and consecutively incubated with 10 pl/well of detection antibody for an hour at RT. Unbound detection antibody was removed by washing four times, and the wells were incubated with 20 pl TMB for 5 min. The reaction was stopped by adding 10 pl of 1 M H2SO4 per well. Absorbance was immediately read at 450 nm with using EnSpire 2300 microplate reader (Perkin Elmer).
BLI assays Kinetic measurements and epitope binning experiments were performed by Bio-Layer Interferometry (BLI) using a RED96e OCTET system (Sartorius AG). In both set-ups, recombinant human ASC, expressed in house, or murine ASC (Cusabio, CSB-EP861664 MO) were covalently immobilized onto AR2G biosensors (Sartorius AG) via EDC/NHS- mediated amine coupling.
For kinetic measurements, both human ASC and murine ASC were immobilized at a concentration as low as 0.625 n g/mL in 10 mM sodium phosphate buffer pH 5, for 300 s, to ensure the interaction with the analyte taking place predominantly at 1 :1 stoichiometric ratio. Association and dissociation kinetics were measured at 50, 16.67, 5.55, 1.85, 0.617, 0.206 and 0 nM antibody concentrations in 1 x PBS buffer (pH 7.4), supplemented with 0.1 % BSA and 0.02% Tween, for 150 s and 500 s, respectively. Antibodies were released from ASC by immersing the biosensors into regeneration buffer (10 mM glycine, pH 3.0). The regeneration buffer was selected following a regeneration scouting procedure, as the mildest agent which resulted in full baseline recovery and complete preservation of ligand binding capacity. This allowed for using a single immobilized biosensor for the full concentration series of each antibody. In the analysis part, a biosensor for which buffer was injected as analyte throughout the entire run was used as a reference for subtraction. The association (kon) and dissociation (kOft) constants were calculated using a 1 :1 binding model, with a global fit.
In the set-up used for epitope binning experiments, human ASC and murine ASC were immobilized at concentrations ranging from 1.25 pg/mL to 20 pg/mL. Antibodies were binned in sets of up to 7 antibodies per experiment, in a two-dimensional matrix format, with the antibodies added sequentially, in pairs, at saturating concentrations. The biosensors were regenerated between consecutive antibody pairs with regeneration buffer. For data analysis, the condition where the same antibody was injected both as first and second antibody was selected as reference, for subtraction. Only incoming antibodies generating an additive binding response greater than the Bmax of the first antibody alone were considered non-competing. Distinct and overlapping bins were defined manually based on relative degrees of competition between the antibodies involved.
Binding to ASC isoforms Protein expression and cell lysate: ASC-b, ASC-c, ASC-d (as described by Bryan et al, 2010) ASC-Y146A (as described by Prather et al 2022) and ASC K21 A K22A K26A (as described by Venegas-Maldonado et al, 2017) were expressed in HEK293 by transfection with PEI. Cell lysates were prepared by lysing the cells with ice-cold CHAPS buffer supplemented with proteases inhibitors. After clearing the lysates by centrifugation at 17'000 x G for 15 min at 4°C, supernatant was transferred to a new set of pre-chilled tubes. Protein concentration was determined by BCA assay (Pierce™ BCA Protein Assay Kit, Thermpfisher Scientific). NuPAGE LDS sample buffer (4X) (Invitrogen) and NuPAGE Sample reducing agent (10X) (Invitrogen) was added to the samples and they were boiled at 95°C for 5 min. SDS PAGE: 15ug of lysate or 150ng of ASC-His monomer were loaded onto NuPAGE 4- 12% Bis-Tris Gels (Invitrogen). Gels were run in MES SDS Running buffer (Invitrogen) at 120 V for 40min. Gel electrophoresis was performed using the Invitrogen XCell SureLock™ Electrophoresis cell chamber system.
Protein transfer (Blotting): Gels were placed onto Amersham™ Protran™ Premium 0.45um Nitrocellulose Blotting membrane (Cytiva) and proteins transferred at 100 V for 1 h using Mini Trans-Blot™ Cell system (Bio-Rad) and 1 X Transfer buffer (25mM Tris-Base, 192mM glycine, 10% methanol). After blotting, membranes were blocked in 5% non-fat dry milk in PBS-T (0.1 % Tween-20) for 1 h at RT.
Staining: After blocking, primary antibodies were added at 1 ug/ml in 1 % non-fat dry milk in PBS-T to the membranes for 2h at RT. Membranes were washed 3x in PBS-T for 10 min each time and then incubated in secondary antibody in 1% non-fat dry milk in PBS-T for 1 h at RT. Membranes were washed 3x in PBS-T and subsequently incubated in SuperSignal™ West Pico PLUS Chemiluminescent Substrate (Thermofisher Scientific).
Immunofluorescence of ASC specks
THP-1 wt and asc-/- cells were seeded at a density of 10 000 cells/ well in 50uL of RPMI medium supplemented with Glutamax, 10%FBS, 1 %P/S, 100ug/mL Normocin, 100ug/mL Zeocin in a 384 well plate (Greiner bio-one 781986). The cells were primed with 1 ug/mL LPS (Sigma, cat: L4391 -1 MG) for 3h. After priming, the formation of ASC specks was induced by treatment with 5uM Nigericin (Invivogen cat: tlrl-nig) for 1 h. Thereafter, the cells were fixed and permealized using BD cytofix/cytoperm (BD biosciences, cat: 554722) solution for 20min at RT. The cells were then washed twice with PBS and blocked with 2% BSA supplemented with human Fc block reagent (Miltenyi cat: 130-059-901 ). After blocking, the cells were incubated with 1 ug/mL of anti-ASC antibodies or isotype control 2h at RT, washed twice with PBS and further incubated with a detection secondary antibody coupled with Alexa fluor 647 (Jackson Immunoresearch, 1 1 1 -607-003) for 1 h at RT. 4 ',6- Diamidino-2'-phenylindol-dihydrochloride (DAPI) (Sigma, cat: MBD0015-1 ML) was used as a counterstain at 1 ug/mL for 10 min in PBS. Images were taken using a 10X objective. All images were acquired using a fluorescence microscope. Image processing and ASC speck count was accomplished using Fiji Image] and Cellprofiler.
Western Blot of selected antibodies on THP-1 cells and BMDMs
-Cell lysis: Cell lysates were prepared by lysing the cells (wt and a sc" human THP-1 monocytes and mouse BMDMs) with ice-cold lysis buffer containing 1 % Triton X-100 for 15 min on ice. After clearing the lysates by centrifugation at 17'000 x G for 15 min at 4°C, supernatant was transferred to a new set of pre-chilled tubes. Protein concentration was determined by BCA assay (Pierce™ BCA Protein Assay Kit, Thermofisher Scientific). NuPAGE LDS sample buffer (4X) (Invitrogen) and NuPAGE Sample reducing agent (10X) (Invitrogen) was added to the samples and boiled at 95°C for 5 min.
-SDS PAGE: 10 ug of protein were loaded onto NuPAGE 4-12% Bis-Tris Gels (Invitrogen). Gels were run in MES SDS Running buffer (Invitrogen) at 80 V for 10 min and then at 120 V for another 1 10 min. Gel electrophoresis was performed using the Invitrogen XCell SureLock™ Electrophoresis cell chamber system.
-Protein transfer (Blotting): Gels were placed onto Amersham™ Protran™ Premium 0.45um Nitrocellulose Blotting membrane (Cytiva) and proteins transferred at 100 V for 1 h using Mini Trans-Blot™ Cell system (Bio-Rad) and 1 X Transfer buffer (25mM Tris-Base, 192mM glycine, 10% methanol). After blotting, membranes were blocked in 5% non-fat dry milk in PBS-T (0.1 % Tween-20) for 1 h at RT.
-Staining: After blocking, primary antibodies were added at 1 ug/ml in 1 % non-fat dry milk in PBS-T to the membranes for 2h at RT. Membranes were washed 3x in PBS-T for 10 min each time and then incubated in secondary antibody in 1 % non-fat dry milk in PBS-T for 1 h at RT. Membranes were washed 3x in PBS-T and subsequently incubated in SuperSignal™ West Pico PLUS Chemiluminescent Substrate (Thermofisher Scientific).
Fibrillation Assay: 1 .75uM ASC-GFP-His was incubated with 3.5uM of anti-ASC antibody or isotype control 1 h at 37°C in protein LoBind tube (Eppendorf, cat 0030109.116)). The mix was then transferred to a clear bottom non-binding 384well-plate (Greiner bio-one, cat 781906) and protein aggregates imaged. Images were acquired using the 10X objective of a fluorescence microscope and image processing accomplished using Fiji Image],
Sedimentation assay
1.75 uM of ASC-GFP-His monomers were incubated with 3.5 uM of MY_anti-ASC antibodies or isotype control in Protein LoBind tubes (Eppendorf, cat 0030109.116) for 30 min at 37°C. After incubation, tubes were centrifuged for 15 min at 17'000 x G and 4°C. Supernatants were transferred to a new set of tubes. Pellets were washed once with PBS, centrifuged for 15 min at 17'000 x G and 4°C and supernatant discarded. Samples were prepared for Western Blot by adding NuPAGE LDS sample buffer (4X) (Invitrogen) and NuPAGE Sample reducing agent (10X) (Invitrogen) and boiling at 95°C for 5 min before loading onto NuPAGE 4-12% Bis-Tris Gels (Invitrogen). Gels were run in MES SDS Running buffer (Invitrogen) at 80 V for 10 min and then at 120 V for another 1 10 min. Gel electrophoresis was performed using the Invitrogen XCell SureLock™ Electrophoresis cell chamber system. Gels were placed onto Amersham™ Protran™ Premium 0.45um Nitrocellulose Blotting membrane (Cytiva) and proteins transferred at 100 V for 1 h in 1 X Transfer buffer (25 mM Tris-Base, 192 mM glycine, 10% methanol) using Mini Trans-Blot™ Cell system (Bio-Rad). After blotting, membranes were blocked in 5% non-fat dry milk in PBS-T (0.1 % Tween-20) for 1 h at RT. Primary antibodies were added at 1 ug/ml in 1 % nonfat dry milk in PBS-T to the membranes for 2h at RT. Membranes were washed 3x in PBS-T for 10 min each time and then incubated in secondary antibody in 1 % non-fat dry milk in PBS-T for 1 h at RT. Membranes were washed 3x in PBS-T and subsequently incubated in SuperSignal™ West Pico PLUS Chemiluminescent Substrate (Thermofisher Scientific).
Blocking of ASC-A0 interaction
ASC-His fibrils were formed by incubating ASC-His monomer at 37°C for 1 h in protein LoBind tubes (Eppendorf, 0030109.116). Fibrillary ASC (1.75uM) was then incubated with 1 uM 5'-TAMRA-A01 -42 (Bachem) with and without 3.5uM anti-ASC antibodies or isotype control for 4h at 37°C. After incubation, protein aggregates were analyzed by flow cytometry using a Novocyte Advanteon (Agilent) and Flowjo v10.7.1 . ASC_AbOO1 TH P-1 efficacy data
- Wt and asc' THP-1 monocytes were seeded at a density of 10'000 cells/well in RPMl medium supplemented with Glutamax, 10%FBS, 1 %P/S, 100 ug/mL Normocin, 100 ug/mL Zeocin in a 384-well plate. To differentiate the monocytes into macrophages, cells were incubated in 100 ng/ml phorbol 12 -myristate- 13 -acetate (PMA)-containing medium for 4 days at 37°C, 5% CO2. After differentiation cells were washed twice (10 min each) with fresh cell culture medium. ASC fibrils were prepared by incubating ASC-His monomers in sterile PBS at 37°C for 1 h. Before addition of the ASC fibrils (3.5uM) to the cells, they were pre-incubated with antibodies (3.5uM) for 15 min at RT. After 16 h incubation at 37°C and 5% CO2, secreted IL-1 p was measured by Lumit Human IL-1 Immunoassay (Promega, W6010) according to manufacturer's protocol.
Example 1 : Identification and production of antibodies binding to ASC
Immunization of Rabbits
Immunization of rabbits with untagged, HEK derived ASC specks, was carried out by dosing rabbits on day 1 , 7, 14, 21 and 35, with spleen and bone marrow cell harvesting 7 days after the final dose. Immunization of rabbits with the His tagged PYD or CARD domain of ASC was carried out by dosing rabbits on day 1 , 7, 14, 21 and 28, with spleen and bone marrow cell harvesting 5 days after the final dose. Cells were cryopreserved.
Identification and Cloning of Rabbit ASC Specific Antibodies
Rabbit bone marrow or spleen cells were screened for plasmablasts secreting antibodies specific for ASC specks, human ASC (SEQ ID NO: 1 ), mouse ASC (SEQ ID NO: 151), or ASC PYD or CARD domains (SEQ ID NOs 143 and 144, respectively), which were then sorted in 10pl of first strand buffer (ThermoFisher, USA). Cell lysis and reverse transcription of RNA was carried out using maxima H minus reverse transcriptase (ThermoFisher, USA), and DNA encoding the variable domain heavy (VH) and light (VL) sequences was amplified by 2 step PCR using primers annealing to the ends of rabbit variable framework regions, sequenced (Microsynth, Switzerland), translated into amino acid sequences, annotated and aligned (Geneious Biologies) and cloned into expression vectors containing rabbit constant heavy or light regions. The expression vector pDB contains an Epstein-Barr virus origin of replication (oriP), which allows episomal replication of the transfected in cells expressing Epstein-Barr virus Nuclear Antigen! (EBNA1 ). Additionally, the plasmid pDB has a cytomegalovirus-chicken actin-fusion promoter (CAG Promoter), which results in high expression of recombinant proteins.
Small scale antibody expression was carried out in suspension HEK293 cells. A few minutes prior to transfection, cells were seeded into a 24-well plate 500pl cell suspension at 0.8- 1 mio cells/ml in FreeStyle medium supplemented with 0.1 % Pluronic F68, 1 % Glutamax and 1 x Geniticin (all ThermoFisher, USA). 250 ng of heavy chain plasmid DNA and 250 ng of light chain plasmid DNA was mixed in a total volume of 15 pl of FreeStyle medium without supplements. 2 pl of TranslT-LT1 (Mirus, USA) was added to 50 pl of pure FreeStyle medium, mixed and incubated for 5 minutes before being added drop by drop to the FreeStyle medium -DNA mix and incubated for a further 15 minutes at room temperature. This mixture was then added drop by drop to the plated cells, which were incubated at 37°C in a humidified incubator with 5% CO2 for around 3-4 days. The supernatant was harvested after centrifugation at 8000 rpm for 5-10 minutes.
Initially, non-purified cell supernatants were tested by ELISA to confirm binding specificity. High binding clear flat bottom 384-well plates (Greiner bio-one , 781061 ) were coated overnight at 4°C with 10 pl/well of ASG CARD domain (1.25ug/mL), ASC PYD domain (1.25ug/mL), murine ASC (Cusabio, CSB-EP861664 MO, 1.25ug/mL), ASC-His (1 ug/mL), ASC-GFP specks (50 000 specks/well) or BSA (1 pg/ml) diluted in PBS. After overnight incubation, wells were aspirated and washed once with 100 pl/well of wash buffer (0.05% Tween-20 in PBS). Wells were blocked for an hour with 50 pl of 2% BSA diluted in PBS at RT. After blocking, wells were incubated 2 hours at RT with 10 pl of test antibody and serially diluted. Wells were washed four times and consecutively incubated with 10 pl/well of detection antibody for an hour at RT. Unbound detection antibody was removed by washing four times, and the wells were incubated with 20 pl TMB for 5 min. The reaction was stopped by adding 10 pl of 1 M H2SO4 per well. Absorbance was immediately read at 450 nm using EnSpire 2300 microplate reader (Perkin Elmer). Antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_Ab009, which were found to bind to the PYD-domain of ASC, were identified and selected for further characterization. VH and VL sequences as well as CDR sequences of the exemplary antibodies identified as described above are shown in Table 5 below.
Figure imgf000100_0001
Table 5: SEQ ID NOs for CDR and VH/VL sequences of exemplary antibodies ASC_Ab001 , ASC„Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009.
Antibody Expression and Purification
Larger scale antibody expression was carried out in suspension HEK293 cells, using a method scalable from 20 to 1000 mL expression volumes. Transfection was achieved by adding a mixture of polyethylenimine (PEI Max, Polysciences, USA) and DNA encoding rabbit light and heavy chain IgG to cells. To reach a high level of IgGs, the transfected cells are supplemented with Tryptone N1 (OrganoTechni, France) after 24 hours. After expression for 5-7 days the supernatant, which contains the expressed antibodies, was harvested. Purification was carried out using a HiTrap Protein A HP column (Cytiva, US) or Protein A Sepharose 4 Fast Flow beads (Cytiva, US). These beads contain Staphylococcus Protein A immobilized on an agarose matrix. Protein A has a strong affinity to the Fc region of IgG. Binding of protein A to IgG occurs at a neutral pH, and antibody is eluted using acidic pH. Purified antibodies were dialyzed into PBS, concentrated and stored at -80°C. Example 2: Cross-competition of antibodies binding to PYD of ASC
To identify whether antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_AbOO4, ASC_Ab005, ASC_AbOO6, ASC_Ab007, ASC_Ab008 and ASC_Ab009 bind to the same or distinct epitope(s), a cross-competition assay was performed. To this end, cloned monoclonal antibodies were tested by biolayer interferometry (BLI) on full length ASC coupled to BLI sensors. Briefly, 1.25 pg/ml monomeric human ASC was immoblized to ARG2G biosensors (Sartorius AG) via amine coupling. Firstly, the blocking antibody was applied at a concentration of 266.7 nM to saturate ASC binding sites. To monitor competition, the binding of the subsequent antibodies was measured at a concentration of
66.7 nM. Only incoming antibodies generating an additive binding response greater than the max response of the blocking antibody alone were considered non-competing. Partial competition is seen in combinations were the incoming antibody results in a response increase, however somewhat reduced to what could be anticipated based on the expected increase of response levels.
Results are summarized in Table 6 below, wherein "X" means (full) competition, means no competition; and "pc" means partial competition:
Figure imgf000101_0001
Figure imgf000102_0001
ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_AbOO8 and ASC_Ab009
As can be retrieved from Table 4, antibodies ASC_Ab001 , ASC_Ab002 and ASC_Ab003 compete for the same epitope on ASC, indicating that antibodies ASC_Ab001 , ASC_Ab002 and ASC_Ab003 bind to the same epitope. Partial competition was observed for antibodies ASC_Ab008 with ASC_AbOO9 as well as for ASC_Ab008 with ASC_Ab001 , ASC_Ab002 and ASC_Ab003, indicating that these antibodies bind to partially overlapping epitopes. No competition was observed for the remaining antibodies, indicating that these antibodies bind to distinct, non-overlapping epitopes on ASC.
In view thereof, antibodies were grouped based on the ASC epitope targeted. Antibodies were assigned to different binning groups named "PYD-1", "PYD-2", "PYD-4", "PYD-6", "PYD-7a", "PYD-7b" and "PYD-7c", wherein each binning group represents a distinct, nonoverlapping ASC epitope, except for "PYD-7a" and "PYD-7b" and "PYD-7b" and "PYD-7c", which epitopes are partially overlapping, as shown in Table 7 below:
Figure imgf000102_0002
Table 7: Binning groups based on cross-competition results
Example 3: Characterization of antibody binding
For further characterization of the binding of antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC„Ab004, ASC_Ab005, ASC„Ab006, ASC_Ab007, ASC_Ab008 and ASC_Ab009, an ELISA was performed as described above, wherein binding of the antibodies to monomeric human ASC (SEQ ID NO: 1 ), HEK-derived ASC-GFP-specks and murine ASC (SEQ ID NO: 151 ) was determined. EC50 values were calculated using Graphpad Prism. Results are shown in Table 8 below:
Figure imgf000103_0001
Table 8: EC50-values determined for antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC„AbOO8 and ASC_Ab009
These data demonstrate that antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC„Ab005, ASC_Ab006, ASC„AbOO7, ASC_Ab008 and ASC_Ab009 bind specifically to monomeric ASC as well as to ASC specks.
Figure 1 shows the binding curves exemplarily for antibody ASC_Ab001 , additionally including the results for binding to the PYD domain and the CARD domain. The data shown in Figure 1 demonstrate that exemplary antibody ASC_Ab001 specifically binds to human monomeric ASC, ASC specks, murine ASC and to the PYD domain of ASC, whereas no binding was detected to the CARD domain of ASC and to the bovine serum albumin (BSA) control.
Next, Bio-Layer Interferometry (BLI), as described above, was used to test antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_AbOO6, ASC_Ab007, ASC_Ab008 and ASC_Ab009 for their binding affinity to human full-length ASC captured on BLI sensors. The KD, off-rates and on-rates were calculated using a 1 :1 binding model with a global fit. Results are shown in Table 9: Binning
Antibody Domain KD on rate off rate group
ASC Ab004 FYD-1 PYD 6.873E-11 1.378ED6 9.469E-05
ASC_Ab005 PYD- 2 PYD <1.0E-12 4.744E05 <1.0E-07
ASC_Ab006 PYD-4 PYD <1.0E-12 2.614E05 1.615E-07
ASC_Ab007 PYD-6 PYD 1.176E-09 4.408B05 5.185E-04
ASC_Ab001 PYD-7a PYD 1.869E-11 2.401E06 4.487E-05
ASC_Ab002 FYD-7a FYD 2.796E-12 3.053E06 8.536E-06
ASC_Ab003 FYD-7a PYD 2.442E-09 4.677E05 1.142E-03
ASC_Ab008 FYD-7b PYD 4.058E-10 1.361E06 5.524E-04
ASC_Ab009 FYD-7C PYD 1.575E-12 1.576E05 2.483E-07
Table 9: Binding affinities determined for antibodies ASC_Ab001 , ASC_AbOO2, ASC_Ab003, ASC_AbOO4, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC_Ab008 and ASC_AbOO9 by BLI
Example 4: ASC isoform and peptide mapping
For further characterization of the binding of antibodies ASC_Ab001 , ASC_Ab004 and ASC_Ab008 to ASC, binding to the different ASC isoforms (full-length ASC, SEQ ID NO: 1 ; ASC-b, SEQ ID NO: 145; ASC-c, SEQ ID NO: 146 and ASC-d, SEQ ID NO: 147) and to human ASC carrying (i) mutations K21 A, K22A and K26A ("ASC K21 A K22A K26A"); or (ii) mutation Y146A ("ASC Y146A") was investigated by western blot using lysate of HEK293 cells transfected with expression plasmids for expression of the different ASC isoforms or ASC carrying (i) mutations K21 A, K22A and K26A; or (ii) mutation Y146A. Polyclonal antibody AL-177 (AdipoGen, Prod. No. AG-25B-0006-C100, Liestal, Switzerland), which is known to target an epitope in the N-terminus of the PYD domain, which is present in all four isoforms, was used as control.
Figure 2 shows an alignment of the different ASC isoforms. While full-length ASC and ASC- b contain a complete PYD domain, ASC-c lacks amino acids 26 to 85 of full-length ASC, while ASC-d contains only the N-terminal 35 amino acids of the PYD domain of ASC.
Figure 3 shows representative images of binding patterns of ASC_Ab001 , ASC_Ab004, ASC_Ab008 and control antibody (AL-177, adipogen). While control antibody AL-177 bound to each of the four ASC isoforms and to both mutated ASC, antibodies ASC_Ab001 , ASC_Ab004 and ASC_Ab008 bound to full-length ASC and ASC-b as well as to the two mutated ASC, but not to isoforms ASC-c and ASC-d.
This demonstrates that the epitope, to which antibodies ASC_Ab001 , ASC„Ab004 and ASC_Ab008 bind to, is located within a region of ASC spanning amino acids 36 to 85 (which is not present in ASC-c and ASC-d).
To confirm and broaden these findings to further antibodies of the present invention, binding of antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab004, ASC_Ab005, ASC_Ab006, ASC_Ab007, ASC„Ab008 and control antibody AL-177 to ASC peptides covering amino acid 1 to 20 (SEQ ID NO: 148) or 1 1 to 30 (SEQ ID NO: 149) was assessed by ELISA. As an additional control, nanobody VH HASC as described in Schmidt et al., 2016 (Schmidt Fl, Lu A, Chen J , Ruan J, Tang C, Wu H, Ploegh HL. A single domain antibody fragment that recognizes the adaptor ASC defines the role of ASC domains in inflammasome assembly. J Exp Med. 2016 May 2;213 (5): 771 -90. doi: 10.1084/jem.20151790) was used. As described in Schmidt et al., 2016, VHHASC binds to the CARD domain of ASC, such that no binding to the N-terminal peptides is expected.
In a further experiment, binding of antibodies ASC_Ab001 and ASC_AbOO2 as well as prior art antibodies AL-177 and VHHASC to ASC peptides covering amino acid 1 to 20 (SEQ ID NO: 148), 1 1 to 30 (SEQ ID NO: 149) or 21 to 40 (SEQ ID NO: 150) was assessed by ELISA. In addition, a human IgG 1 antibody containing the VH of SEQ ID NO: 19 and the VL of SEQ ID NO: 30 of US 10,961 ,306 B2, which can be referred to as "IC-100", as described in US 10,961 ,306 B2 column 40, lines 60-64, was produced. According to US 10,961 ,306 B2, said antibody is named "IC100" and was also included in this experiment.
Results of the peptide binding experiments are shown in Figure 4. Figure 4(A) shows the results of the first experiment. Only control antibody AL-177 bound to the N-terminal peptides, as expected based on its binding to all four ASC isoforms. As expected, no binding to the N-terminal peptides was found for VHHASC, which binds to CARD. None of antibodies ASC AbOOl, ASC_Ab002, ASC_AbOO4, ASC_Ab005, ASC_Ab006, ASC_Ab007 and ASC_Ab008 bound to the N-terminal peptides, confirming the results obtained with the distinct ASC isoforms for ASC_Ab001 , ASC_Ab004 and ASC_Ab008 and demonstrating that also ASC_AbOO2, ASC_Ab005, ASC_Ab006 and ASC_Ab007 do not bind to the N-terminal part of ASC. Figure 4(B) shows the results obtained for ASC peptides covering amino acid 1 to 20 (SEQ ID NO: 148), 1 1 to 30 (SEQ ID NO: 149) or 21 to 40 (SEQ ID NO: 150). In line with the previous experiment, AL-177 was found to bind to the ASC peptides covering amino acid 1 to 20 (SEQ ID NO: 148) and 1 1 to 30 (SEQ ID NO: 149), while none of VH HASC, ASC_Ab001 and ASC_Ab002 bound to this peptide. Antibody IC-100 did not bind to the most N-terminal peptide (amino acids 1 -20; SEQ ID NO: 148), but was found to bind to the peptides covering amino acids 11 to 30 (SEQ ID NO: 149) or 21 to 40 (SEQ ID NO: 150). None of the other antibodies bound to the peptide covering 21 to 40 (SEQ ID NO: 150).
In summary, this ASC peptide analysis shows that AL-177 binds to an epitope within amino acids 1 - 30 of ASC, while IC100 binds to an epitope within amino acids 11 - 40 of ASC. None of the antibodies of the present invention was found to bind to such an N-terminal peptide.
Example 5: Comparison with antibodies of the prior art
As described above (see also Schmidt et al., 2016) and demonstrated in Example 4, antibodies AL-177, IC100 and anti-CARD antibody VHH ASC bind to distinct epitopes of ASC as compared to the antibodies of the present invention. For further comparison of the antibodies of the present invention with antibodies of the prior art, binding of antibodies ASC_Ab001 and ASC__Ab002 as well as prior art antibodies AL-177, VHHASC and IC100 (obtained as described in Example 4 above) to (i) human ASC, (ii) murine ASC, (iii) ASC specks and (iv) the PYD domain of ASC was assessed by ELISA as described above.
Results are shown in Figures 5 (human ASC), 6 (murine ASC), 7 (ASC specks) and 8 (PYD domain of ASC).
All tested antibodies were found to bind to human ASC and ASC specks, except for 1C100. Surprisingly, despite the data previously obtained by others for an antibody "IC100" or "IC- 100" (see US 10,961 ,306 B2 and Desu HL, Plastini M, llliano P, Bramlett HM, Dietrich WD, de Rivero Vaccari JP, Brambilla R, Keane RW. IC100: a novel anti-ASC monoclonal antibody improves functional outcomes in an animal model of multiple sclerosis. J Neuroinflammation. 2020 May 4;17(1 ):143. doi: 10.1 186/sl 2974-020-01826-0) and despite the data obtained in Example 4 demonstrating binding of IC100 to peptides of SEQ ID NOs 149 and 150, no binding to full-length human ASC, murine ASC, ASC specks and the (entire) PYD domain of ASC could be detected for IC100 (human IgGI antibody containing the VH of SEQ ID NO: 19 and the VL of SEQ ID NO: 30 of US 10,961 ,306 B2, as described in US 10,961 ,306 B2 column 40, lines 60-64). For the PYD domain, also for VHHASC no binding could be detected, which was expected since this antibody was previously reported to bind to the CARD domain of ASC.
For murine ASC, only antibodies ASC_Ab001 and ASC_AbOO2 showed binding, while no specific binding was detected for AL-177, IC100 and VHHASC to murine ASC. Accordingly, only antibodies ASC_Ab001 and ASC_AbOO2 showed binding to murine ASC (in addition to human ASC) and, thus, advantageous cross-reactivity, which is absent in AL-177, IC100 and VH HASC
Example 6: Binding of antibodies to physiological ASC speck
To investigate binding of the antibodies of the present invention to physiological ASC speck, THP-1 asc/_ cells were compared to THP-1 wt cells. THP-1 is human macrophage cell line, which is commercially available. THP-1 asc/_ cells lack expression of ASC, in contrast to THP-1 wt cells, such that physiological ASC speck formation is only expected in THP-1 wt cells, while THP-1 asc A cells represent negative controls.
As described in detail above, formation of ASC specks was induced in THP-1 wt and THP-1 asc A cells. The cells were then incubated with the anti-ASC antibodies ASC_Ab001 , ASC_Ab002, ASC_Ab003, ASC_Ab004, ASC_Ab005, ASC_AbOO6, ASC_Ab007, ASC_Ab008, ASC_AbOO9 or isotype control, as described above in detail. Results were obtained by fluorescent imaging. Results are shown in Figure 9. These data show that each of the antibodies ASC_Ab001 , ASC_AbOO2, ASC_Ab003, ASC_AbOO4, ASC_Ab005, ASC_Ab006, ASC_AbOO7, ASC_Ab008 and ASC_AbOO9 binds to physiological ASC speck.
Example 7: Binding specificity of antibodies
To confirm binding of antibodies ASC_Ab001 , ASC_Ab004, ASC_Ab007 and ASC_Ab009 to endogenous ASC and species cross-reactivity, antibodies were tested by Western blot on wt and asc7 human TH P-1 monocytes and mouse bone-marrow derived macrophages (BMDMs), as described in detail above.
Results are shown in Figures 10 (wt and asc7' human THP-1 cells) and 11 (BMDMs). All of the tested antibodies were binding to human and murine endogenous ASC in cell lysates from wt cells, while there was no binding observed in the asc7 cel I lysates.
Example 8: Treatment with antibodies of the invention significantly impaired ASC fibrillation
ASC assembles in large cytoplasmic macromolecular assemblies called ASC specks. This is driven by ASC-ASC interaction and can be reconstituted in vitro by incubating ASC monomers at 37°C. Under these conditions ASC will rapidly fibrillate and retain their physiological and pathological functions (Friker LL, Scheiblich H, Hochheiser IV, Brinkschulte R, Riedel D, Latz E, Geyer M, Heneka MT. [3-Amyloid Clustering around ASC Fibrils Boosts Its Toxicity in Microglia. Cell Rep. 2020 Mar 17;30(11 ):3743-3754.e6. doi: 10.1016/j.celrep.2020.02.025). An effect of the antibodies on fibrillation would provide a way to block either de novo or seeded ASC assembly, and thus the downstream effect of ASC speck formation such as inflammation and/or amyloid seeding.
To investigate the effect of anti-ASC antibodies ASC_Ab001 , ASC_Ab004, ASC_Ab007, ASC_Ab008 and ASC„AbOO9 on ASC fibrillation was assessed in an ASC fibrillation assay as described above. Briefly, ASC-GFP-His was incubated with the antibody to be tested and protein aggregates were imaged.
Results are shown in Figure 12. These data show that each of antibodies ASC_Ab001 , ASC_Ab004, ASC_Ab007, ASC_Ab008 and ASC„Ab009 significantly impaired ASC fibrillation, as compared to an isotype control. An anti-ASC-CARD antibody used as control did not show any effects, while anti-CARD antibody VHHASC even resulted in increased ASC fibrillation.
In addition, a sedimentation assay to assess the antibody's anti-fibrillation properties was performed as described above. Briefly, soluble and insoluble fractions of ASC were separated by high-speed centrifugation upon treatment with anti-ASC antibodies or isotype control.
Results are shown in Figure 13. Treatment with ASC_Ab001 lead to more soluble ASC (supernatant fraction) when compared to ASC only or isotype control after 30 min of incubation at 37°C. Or in other words, ASC_Ab001 shifts the equilibrium towards the soluble ASC in the supernatant fraction. Similarly as in the ASC fibrillation assay described above, treatment with an anti-CARD antibody had no effect on the fibrillation.
Example 9: Anti-ASC antibodies block ASC -AB interaction
ASC -Ap interaction is assumed to be at the basis of increased A aggregation and amyloid deposition in Alzheimer disease (Venegas C, Kumar S, Franklin BS, Dierkes T, Brinkschulte R, Tejera D, Vieira-Saecker A, Schwartz S, Santarelli F, Kummer MP, Griep A, Gelpi E, Beilharz M, Riedel D, Golenbock DT, Geyer M, Walter J, Latz E, Heneka MT. Microglia- derived ASC specks cross-seed amyloid-p in Alzheimer's disease. Nature. 201 7 Dec 20;552(7685):355-361 . doi: 10.1038/nature25158) as well as of increased neuroinflammation caused by these ASC-A(3 complexes (Friker LL, Scheiblich H, Hochheiser IV, Brinkschulte R, Riedel D, Latz E, Geyer M, Heneka MT. [3-Amyloid Clustering around ASC Fibrils Boosts Its Toxicity in Microglia. Cell Rep. 2020 Mar 17;30(11 ):3743-3754.e6. doi: 10.1016/j.celrep.2020.02.025). Thus, by measuring the effects of antibody on ASC-Ap interaction we can test their potential to block these downstream effects of increases amyloid deposition and neuroinflammation.
In view thereof, it was investigated whether the antibodies of the present invention are able to reduce or inhibit ASC-Ap binding. To this end, an ASC-A interaction assay was performed as described above. Thereafter, protein aggregates of the antibody to be tested were compared to those obtained with controls, such as ASC only, ASC+Ap, VHHASC and an isotype control antibody. ASC-Ap inhibition was determined as percentage in view the binding (aggregation) observed for the ASC+Ap (without antibody) control and the ASC only control.
Results are shown in Figure 14. These data demonstrate that antibodies ASC_Ab001 , ASC_AbOO2, ASC_Ab003, ASC_Ab004, ASC_Ab005 and ASC_Ab006 significantly reduce ASC-AP interaction, while anti-CARD antibody VH HASC even increased ASC-Ap interaction.
Example 10: ASC Ab001 blocks the pro-inflammatory function of extracellular ASC fibrils
The in-vitro efficacy of ASC_Ab001 was tested in a cell-based assay using differentiated human THP-1 macrophages that were treated with extracellular ASC fibrils obtained as described above. It has been shown that ASC fibrils and ASC specks can propagate inflammation (Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels C, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmuller W, Latz E. The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation. Nat Immunol. 2014 Aug;15(8):727-37. doi: 10.1038/ni.2913; Baroja-Mazo A, Martm-Sanchez F, Gomez Al, Mart ez CM, Amores- Iniesta J, Compan V, Barbera-Cremades M, Yagiie J, Ruiz-Ortiz E, Anton J, Bujan S, Couillin I, Brough D, Arostegui JI, Pelegrm P. The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response. Nat Immunol. 2014 Aug;15(8):738- 48. doi: 10.1038/ni.2919). As described in detail above, before addition of the ASC fibrils to the cells, ASC fibrils were pre-incubated with ASC_Ab001 and isotype control antibody, each with or without Leu234Ala/Leu235Ala/Pro329Gly ("PGLALA") Fc modification. IL-1 p measurement was used as a readout for inflammasome activation.
Results are shown in Figure 15. ASC_Ab001 significantly reduced the ASG fibrils-induced production of IL-1 P compared to the isotype control, demonstrating the efficacy of ASC_Ab001 . This reduction was not dependent on antibody Fc function, as no difference between the wt IgG and the PGLALA (Fc-inactive) format was observed. Treatment with antibody alone did not influence IL-113 production.
Example 11 : Anti ASC antibodies decrease IL-1 beta release in a dose dependant manner in ASC seeded Inflammation assay
Wt THP-1 monocytes were seeded at a density of 20'000 cells/well in RPMI medium supplemented with Glutamax, 10%FBS, 1 %P/S, 100 ug/mL Normocin, 100 ug/mL Zeocin in a 384-well plate. To differentiate the monocytes into macrophages, cells were incubated in 100 ng/ml phorbol 12-myristate-13-acetate (PMA)-containing medium for 4 days at 37°C, 5% CO2. After differentiation cells were washed twice (10 min each) with fresh cell culture medium and primed with LPS (100ng/mL) for 3h. ASC fibrils were prepared by incubating ASC-his monomers in sterile PBS at 37°C for 1 h. Before addition of the ASC fibrils to the cells, they were pre-incubated with antibodies for 15 min at RT. After 48 h incubation at 37°C and 5% CO2, secreted IL-1 was measured by Lumit Human IL-1 Immunoassay (Promega, W6010) according to manufacturer's protocol.
As shown in Figure 16, all anti-ASC antibodies tested (ASC_AB001 - ASC_AbOO9) decrease IL-1 beta release in a dose dependant manner.
Example 12: Anti-ASC antibodies do no bind protein with homologous sequence to ASC
Binding of anti-ASC antibodies (ASC_AB001 - ASC_AbOO9) to inflammasome proteins AIM2 and NLRP3 was tested by a binding ELISA. High binding clear flat bottom 384-well plate (Greiner bio-one , 781061 ) were coated o/n (4°C) with 10 pl/well of AIM2 (1 ug/mL, Sino Biological, cat: 11654-H09B) or NLRP3 (1 pg/ml, cusabio, CSB-EP822275HU1 ) diluted in PBS. After o/n incubation, wells were aspirated and washed once with 100 pl/well of wash buffer (0.05% Tween-20 in PBS). Wells were blocked for an hour with 50 l of 2% BSA diluted in PBS at RT. After blocking, wells were incubated 2 hours at RT with 10 pl of test antibody and serially diluted. Wells were washed four times and consecutively incubated with 10 pl/well of detection antibody for an hour at RT. Unbound detection antibody was removed by washing four times, and the wells were incubated with 20 pl TMB for 5 min. The reaction was stopped by adding 10 pl of 1 M H2SO4 per well. Absorbance was immediately read at 450 nm using EnSpire 2300 microplate reader (Perkin Elmer).
As shown in Figure 17, anti-ASC antibodies do not bind to inflammasome proteins AIM2 and NLRP3 comprising a homologous sequence to ASC, confirming the specificity of anti- ASC antibodies (ASC_AB001 - ASC_AbOO9) for ASC.
Example 13: Anti-ASC antibodies ameliorate the severity of dextran sulfate sodium (DSS)- induced colitis
Acute colitis was induced in eight- week-old female C57BL/6 mice by giving mice 2.5% dextran sodium sulfate (DSS) in drinking water for 8 days. Healthy animal (n=8) were given normal drinking water. Anti ASC antibody (n = 8 per group) or normal IgG as control (n = 8) were injected intraperitoneally 2h before induction of colitis. Mice were sacrificed on Day 8. Disease activity index (DAI) was measured based on parameters of bodyweight change, stool consistency and stool blood presence during DSS administration. For H&E staining, sections were cut from paraffin blocks of colon tissues prefixed in 10% neutral buffered formalin (FUJIFILM Wako Pure Chemical Corporation, Japan) and stained with Lillie- Mayer's Hematoxylin (Muto Pure Chemicals Co., Ltd., Japan) and eosin solution (FUJIFILM Wako Pure Chemical Corporation). Representative photos of the changes observed were captured from each group. As shown in Figure 18 (A: Measurement of body weight; B: measurement of disease activity index (DAI) score; C: measurement of colon length), anti-ASC antibodies ASC_Ab001 and ASC_AbOO2 ameliorate the severity of DSS-induced colitis compared to the isotype control.
This finding is supported in Figure 19, wherein Figure 19A to C (haematoxylin and eosin staining of colon) show the beneficial effect of ASC_Ab001 in DSS induced colitis on the mucosal morphology in isolated bowel segments, and Figure 19D illustrates the reduction of infiltration of inflammatory cells into the bowel mucosa in anti-ASC antibody treated animals versus isotype (normal IgG) treated animals.
Example 14: Humanization of antibodies preserves binding to ASC
To test the effect of humanization, a humanized version of ASC_Ab001 was produced. The humanized antibody (ASC„AbOO1 humanized) contained the VH according to SEQ ID NO: 157 (which comprises heavy chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 2, SEQ ID NO: 3 and SEQ ID NO: 4, respectively) and the VL according to SEQ ID NO: 158 (which comprises light chain CDR1 , CDR2, and CDR3 sequences as set forth in SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7, respectively).
Binding of the humanized ASC_Ab001 to human ASC was compared with (rabbit) ASC_Ab001 having VH and VL sequences according to SEQ ID NOs 8 and 9, respectively, as described above in an ELISA as described above.
Results are shown in Figure 20. These data show that similar binding of the humanized antibodies was observed compared to the rabbit IgG format.
Example 15: Construction and evaluation of further humanized anti-ASC antibodies and antigen binding fragments thereof
Further humanized antibodies were designed based on "parental" antibodies ASC_Ab001 and ASC_Ab002, respectively. The amino acid sequences of humanized variable heavy chains (VH) based on ASC_Ab001 (Ab001_VHhum1 , AbOO1_VHhum3 and Ab001_VHhum23) are shown in Figure 21. All sequences for the heavy chain are based on the IGHV3-66 germline, which has the highest homology to the original rabbit Ab001 heavy chain framework and has also been reported to be a good acceptor framework for rabbit CDRs. In the kinetic measurement and binding studies, respectively, humanized constructs comprising Ab001_VHhum23 were used.
The amino acid sequences of the humanized variable light chains (VL) based on ASC_Ab001 (Ab001_VLhum1 , Ab001_VLhum4, Ab001 _VLhum5, Ab001_VLhum6, Ab001_VLhum10, Ab001_VLhum1 1 , and Ab001_VLhum12) are shown in Figure 22. All sequences for the light chain are based on the IGKV1 -5 germline, which has the highest homology to the original rabbit Ab001 light chain framework and is very similar to the IGKV1 -27*01 germline, previously used as template for a generic acceptor frame for rabbit light chain CDRs. In the kinetic measurement and binding studies, respectively, humanized constructs comprising Ab001„VLhum11 and Ab001_VLhum12, respectively, were used.
An amino acid sequence of a humanized variable heavy chain (VH) based on ASC_AbOO2 (Ab002_VHhum17), as well as the amino acid sequences of humanized variable light chains (VL) based on ASC_Ab002 (Ab002_VLhum1 , Ab002_VL hum6, AbOO2_VLhum1 1 , Ab002_VL hum12) are shown in Figure 23. The presented sequence for the heavy chain is based on the IGHV3-66 germline, which is has the highest homology to the original rabbit Ab002 heavy chain framework and has also been reported to be a good acceptor framework for rabbit CDRs. All sequences for the heavy chain are based on the IGKV1 -27 germline, which has the highest homology to the original rabbit Ab002 light chain framework and was previously used as template for a generic acceptor frame for rabbit light chain CDRs. A humanized Ab002 construct comprising Ab002_VHhum17 and Ab002_VLhum12 was used in the kinetic measurement and binding studies, respectively,
Kinetic measurements of Fabs
Kinetic measurements of Fabs were performed by Bio-Layer Interferometry (BLI) using a RED96e OCTET system (Sartorius AG). Recombinant hASC, expressed in house was covalently immobilized onto AR2G biosensors (Sartorius AG) via EDC/NHS-mediated amine coupling a concentration of 1 .25pg/mL in 10 mM sodium phosphate buffer pH 5, for 300 s. Association and dissociation kinetics were measured at 50, 16.67, 5.55, 1 .85, 0.61 7 and 0 nM Fabs concentrations in 1 x PBS buffer (pH 7.4), supplemented with 0.1 % BSA and 0.02% Tween, for 200 s and 500 s, respectively. Antibodies were released from ASC by immersing the biosensors into regeneration buffer (10 mM glycine, pH 3.0). The association (kon) and dissociation (kOff) rate constants were calculated using a 1 :1 binding model, with a global fit. The calculated KD, kon and koff rate constants are presented in Table 1 0 (for ASC_Ab001 Fabs) and Table 1 1 (for ASC_Ab002 Fabs), respectively.
Figure imgf000115_0001
AbOO1 VHrab/VLrab, AbOO1 VH23/VL1 1 , and AbOO1 VH23/VL12, respectively
Figure imgf000115_0002
Table 1 1 : KD, kOn and koff values determined for Fabs comprising Ab002 VH1 7/ VL12, and Ab002 VHrabA/Lrab, respectively
The association and dissociation curves are graphically shown in Figure 24 (for Fabs comprising Ab001 VH23/VL1 1 , Ab001 VH23/VL12, and Ab001 VHrabA/Lrab, respectively), and Figure 25 (for Fabs comprising Ab002 VH1 7/ VL12, and Ab002 VHrab/VLrab, respectively).
As shown in Table 10 and Figure 24, the tested humanized Ab001 (RP02-F8) Fabs have a superior on-rate, but an inferior off-rate compared to the parental rabbit Fab. Thus, the overall affinity of the humanized Ab001 Fabs to hASC is only marginally reduced compared to the parental rabbit Fab.
As shown in Table 1 1 and Figure 25, the tested humanized Ab002 (RP03-12) Fab has an on-rate and an off-rate similar to the on- and off-rates of the parental Fab. These data demonstrate that Fabs comprising humanized VH and VL domains (Ab001 VH23/VL1 1 , Ab001 VH23A/L12, Ab002 VH17A/L12) have overall affinities to hASC similar to the affinity of the respective parental (non-humanized) rabbit antibodies (Ab001 VHrabA/Lrab, Ab002 VHrabA/Lrab) to hASC.
Example 16: Humanized IgG 1 antibodies have affinity to human and murine ASC
Binding of full IgGI humanized ASC_Ab001 antibodies and ASC_Ab002 antibodies to human and murine ASC was compared to (rabbit) Ab001 rb-huIgG, and Ab002 rb-huIgG, respectively (non-humanized) in an ELISA as described above.
Results are shown in Figure 26 (for Ab001 VH23A/L11 , Ab001 VH23A/L12 in comparison to Ab001 rb-huIgG) and Figure 27 (for Ab002 VH17A/L12 in comparison to Ab002 rb- huIgG).
These data show a superior affinity of the humanized (bivalent) Ab001 IgGI antibodies to human and murine ASC compared to the rabbit Ab001 IgG format (Figure 26), and a similar apparent affinity of the humanized (bivalent) Ab002 IgGI antibody compared to the rabbit Ab002 IgG format (Figure 27).
TABLE OF SEQUENCES AND SEQ ID NUMBERS (SEQUENCE LISTING):
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000121_0002
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000125_0002
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000131_0002
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001

Claims

CLAIMS An antibody, or an antigen-binding fragment thereof, which binds to the PYRIN- PAAD-DAPIN domain (PYD) of apoptosis-associated speck-like protein containing a CARD (ASC). The antibody, or an antigen-binding fragment thereof, according to claim 1, wherein the antibody or the antigen-binding fragment thereof, binds to the PYD domain of human ASC (SEQ ID NO: 1 ). The antibody, or an antigen-binding fragment thereof, according to claim 1 or 2, wherein the antibody or the antigen-binding fragment thereof, binds specifically to ASC-b (SEQ ID NO: 145). The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, does not specifically bind to ASC-c (SEQ ID NO: 146). The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, does not specifically bind to ASC-d (SEQ ID NO: 147). The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, binds to human ASC K21 A K22A K26A and/or to human ASC Y146A. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, does not bind to an ASC peptide according to SEQ ID NO: 148 (ASC aa 1 -20 peptide) or SEQ ID NO: 149 (ASC aa 1 1 -30 peptide). The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, does not bind to an ASC peptide according to SEQ ID NO: 150 (ASC aa 21 -40 peptide). The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, binds to an epitope located in a region of the PYD domain spanning amino acids 36 to 85 of human ASC according to SEQ ID NO: 1 , preferably to an epitope located in a region of the PYD domain spanning amino acids 41 to 85 of human ASC according to SEQ ID NO: 1. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, binds to murine ASC. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, exhibits anti-ASC-fibri Nation activity. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, reduces or blocks ASC-Ap interaction. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, reduces or blocks the pro-inflammatory function of extracellular ASC fibrils. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 5, a CDRL2 according to SEQ ID NO: 6, and a CDRL3 according to SEQ ID NO: 7. The antibody, or an antigen-binding fragment thereof, according to claim 14, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 159, an extended CDRH2 according to SEQ ID NO: 3, an extended CDRH3 according to SEQ ID NO: 160, an extended CDRL1 according to SEQ ID NO: 5, an extended CDRL2 according to SEQ ID NO: 6, and an extended CDRL3 according to SEQ ID NO: 7. The antibody, or an antigen-binding fragment thereof, according to claim 14 or 15, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 (including CDRH1 and adjacent framework residues) according to SEQ ID NO: 194, an extended CDRH2 (including CDRH2 and adjacent framework residues) according to SEQ ID NO: 195, an extended CDRH3 (including CDRH3 and adjacent framework residues) according to SEQ ID NO: 196, an extended CDRL1 (including CDRL1 and adjacent framework residues) according to SEQ ID NO: 197, an extended CDRL2 (including CDRL2 and adjacent framework residues) according to SEQ ID NO: 198, and an extended CDRL3 (including CDRL3 and adjacent framework residues) according to SEQ ID NO: 199. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 8; and a VL having at least 70% identity to SEQ ID NO: 9. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 8; and a VL having at least 80% identity to SEQ ID NO: 9. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 8; and a VL having at least 90% identity to SEQ ID NO: 9. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 8 and a VL comprising an amino acid sequence according to SEQ ID NO: 9. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 157; and a VL having at least 70% identity to SEQ ID NO: 158. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 157; and a VL having at least 80% identity to SEQ ID NO: 158. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 157; and a VL having at least 90% identity to SEQ ID NO: 158. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 157 and a VL comprising an amino acid sequence according to SEQ ID NO: 158. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 176; and a VL having at least 70% identity to SEQ ID NO: 177. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 176; and a VL having at least 80% identity to SEQ ID NO: 177. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 176; and a VL having at least 90% identity to SEQ ID NO: 177. The antibody, or an antigen-binding fragment thereof, according to any one of claims 14 to 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 176 and a VL comprising an amino acid sequence according to SEQ ID NO: 177. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 1 1 , a CDRL1 according to SEQ ID NO: 12, a CDRL2 according to SEQ ID NO: 13, and a CDRL3 according to SEQ ID NO: 14. The antibody, or an antigen-binding fragment thereof, according to claim 29, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 161 , an extended CDRH2 according to SEQ ID NO: 10, an extended CDRH3 according to SEQ ID NO: 162, an extended CDRL1 according to SEQ ID NO: 12, an extended CDRL2 according to SEQ ID NO: 13, and an extended CDRL3 according to SEQ ID NO: 14. The antibody, or an antigen-binding fragment thereof, according to claim 29 or 30, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 15; and a VL having at least 70% identity to SEQ ID NO: 16. The antibody, or an antigen-binding fragment thereof, according to claim 29 or 30, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 15; and a VL having at least 80% identity to SEQ ID NO: 16. The antibody, or an antigen-binding fragment thereof, according to claim 29 or 30, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 15; and a VL having at least 90% identity to SEQ ID NO: 16. The antibody, or an antigen-binding fragment thereof, according to claim 29 or 30, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 15 and a VL comprising an amino acid sequence according to SEQ ID NO: 16. The antibody, or an antigen-binding fragment thereof, according to claim 29 or 30, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 178; and a VL having at least 70% identity to SEQ ID NO: 179. The antibody, or an antigen-binding fragment thereof, according to claim 29 or 30, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 178; and a VL having at least 80% identity to SEQ ID NO: 179. The antibody, or an antigen-binding fragment thereof, according to claim 29 or 30, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 178; and a VL having at least 90% identity to SEQ ID NO: 179. The antibody, or an antigen-binding fragment thereof, according to claim 29 or 30, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 178 and a VL comprising an amino acid sequence according to SEQ ID NO: 179. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 17, a CDRH3 according to SEQ ID NO: 18, a CDRL1 according to SEQ ID NO: 19, a CDRL2 according to SEQ ID NO: 20, and a CDRL3 according to SEQ ID NO: 21 . The antibody, or an antigen-binding fragment thereof, according to claim 39, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 161 , an extended CDRH2 according to SEQ ID NO: 17, an extended CDRH3 according to SEQ ID NO: 163, an extended CDRL1 according to SEQ ID NO: 19, an extended CDRL2 according to SEQ ID NO: 20, and an extended CDRL3 according to SEQ ID NO: 21 . The antibody, or an antigen-binding fragment thereof, according to claim 39 or 40, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 22; and a VL having at least 70% identity to SEQ ID NO: 23. The antibody, or an antigen-binding fragment thereof, according to claim 39 or 40, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 22; and a VL having at least 80% identity to SEQ ID NO: 23. The antibody, or an antigen-binding fragment thereof, according to claim 39 or 40, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 22; and a VL having at least 90% identity to SEQ ID NO: 23. The antibody, or an antigen-binding fragment thereof, according to claim 39 or 40, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 22 and a VL comprising an amino acid sequence according to SEQ ID NO: 23. The antibody, or an antigen-binding fragment thereof, according to claim 39 or 40, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 180; and a VL having at least 70% identity to SEQ ID NO: 181 . The antibody, or an antigen-binding fragment thereof, according to claim 39 or 40, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 180; and a VL having at least 80% identity to SEQ ID NO: 181 . The antibody, or an antigen-binding fragment thereof, according to claim 39 or 40, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 180; and a VL having at least 90% identity to SEQ ID NO: 181 . The antibody, or an antigen-binding fragment thereof, according to claim 39 or 40, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 180 and a VL comprising an amino acid sequence according to SEQ ID NO: 181 . The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 24, a CDRH2 according to SEQ ID NO: 25, a CDRH3 according to SEQ ID NO: 26, a CDRL1 according to SEQ ID NO: 27, a CDRL2 according to SEQ ID NO: 28, and a CDRL3 according to SEQ ID NO: 29. The antibody, or an antigen-binding fragment thereof, according to claim 49, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 164, an extended CDRH2 according to SEQ ID NO: 25, an extended CDRH3 according to SEQ ID NO: 165, an extended CDRL1 according to SEQ ID NO: 27, an extended CDRL2 according to SEQ ID NO: 28, and an extended CDRL3 according to SEQ ID NO: 29. The antibody, or an antigen-binding fragment thereof, according to claim 49 or 50, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 30; and a VL having at least 70% identity to SEQ ID NO: 31. The antibody, or an antigen-binding fragment thereof, according to claim 49 or 50, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 30; and a VL having at least 80% identity to SEQ ID NO: 31. The antibody, or an antigen-binding fragment thereof, according to claim 49 or 50, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 30; and a VL having at least 90% identity to SEQ ID NO: 31. The antibody, or an antigen-binding fragment thereof, according to claim 49 or 50, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 30 and a VL comprising an amino acid sequence according to SEQ ID NO: 31 . The antibody, or an antigen-binding fragment thereof, according to claim 49 or 50, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 182; and a VL having at least 70% identity to SEQ ID NO: 183. The antibody, or an antigen-binding fragment thereof, according to claim 49 or 50, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 182; and a VL having at least 80% identity to SEQ ID NO: 183. The antibody, or an antigen-binding fragment thereof, according to claim 49 or 50, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 182; and a VL having at least 90% identity to SEQ ID NO: 183. The antibody, or an antigen-binding fragment thereof, according to claim 49 or 50, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 182 and a VL comprising an amino acid sequence according to SEQ ID NO: 183. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 32, a CDRH2 according to SEQ ID NO: 33, a CDRH3 according to SEQ ID NO: 34, a CDRL1 according to SEQ ID NO: 35, a CDRL2 according to SEQ ID NO: 36, and a CDRL3 according to SEQ ID NO: 37. The antibody, or an antigen-binding fragment thereof, according to claim 59, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 166, an extended CDRH2 according to SEQ ID NO: 33, an extended CDRH3 according to SEQ ID NO: 167, an extended CDRL1 according to SEQ ID NO: 35, an extended CDRL2 according to SEQ ID NO: 36, and an extended CDRL3 according to SEQ ID NO: 37. The antibody, or an antigen-binding fragment thereof, according to claim 59 or 60, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 38; and a VL having at least 70% identity to SEQ ID NO: 39. The antibody, or an antigen-binding fragment thereof, according to claim 59 or 60, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 38; and a VL having at least 80% identity to SEQ ID NO: 39. The antibody, or an antigen-binding fragment thereof, according to claim 59 or 60, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 38; and a VL having at least 90% identity to SEQ ID NO: 39. The antibody, or an antigen-binding fragment thereof, according to claim 59 or 60, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 38 and a VL comprising an amino acid sequence according to SEQ ID NO: 39. The antibody, or an antigen-binding fragment thereof, according to claim 59 or 60, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 184; and a VL having at least 70% identity to SEQ ID NO: 185. The antibody, or an antigen-binding fragment thereof, according to claim 59 or 60, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 184; and a VL having at least 80% identity to SEQ ID NO: 185. The antibody, or an antigen-binding fragment thereof, according to claim 59 or 60, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 184; and a VL having at least 90% identity to SEQ ID NO: 185. The antibody, or an antigen-binding fragment thereof, according to claim 59 or 60, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 184 and a VL comprising an amino acid sequence according to SEQ ID NO: 185. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 40, a CDRH2 according to SEQ ID NO: 41 , a CDRH3 according to SEQ ID NO: 42, a CDRL1 according to SEQ ID NO: 43, a CDRL2 according to SEQ ID NO: 44, and a CDRL3 according to SEQ ID NO: 45. The antibody, or an antigen-binding fragment thereof, according to claim 69, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 168, an extended CDRH2 according to SEQ ID NO: 41 , an extended CDRH3 according to SEQ ID NO: 169, an extended CDRL1 according to SEQ ID NO: 43, an extended CDRL2 according to SEQ ID NO: 44, and an extended CDRL3 according to SEQ ID NO: 45. The antibody, or an antigen-binding fragment thereof, according to claim 69 or 70, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 46; and a VL having at least 70% identity to SEQ ID NO: 47. The antibody, or an antigen-binding fragment thereof, according to claim 69 or 70, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 46; and a VL having at least 80% identity to SEQ ID NO: 47. The antibody, or an antigen-binding fragment thereof, according to claim 69 or 70, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 46; and a VL having at least 90% identity to SEQ ID NO: 47. The antibody, or an antigen-binding fragment thereof, according to claim 69 or 70, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 46 and a VL comprising an amino acid sequence according to SEQ ID NO: 47. The antibody, or an antigen-binding fragment thereof, according to claim 69 or 70, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 186; and a VL having at least 70% identity to SEQ ID NO: 187. The antibody, or an antigen-binding fragment thereof, according to claim 69 or 70, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 186; and a VL having at least 80% identity to SEQ ID NO: 187. The antibody, or an antigen-binding fragment thereof, according to claim 69 or 70, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 186; and a VL having at least 90% identity to SEQ ID NO: 187. The antibody, or an antigen-binding fragment thereof, according to claim 69 or 70, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 186 and a VL comprising an amino acid sequence according to SEQ ID NO: 187. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 48, a CDRH2 according to SEQ ID NO: 49, a CDRH3 according to SEQ ID NO: 50, a CDRL1 according to SEQ ID NO: 51 , a CDRL2 according to SEQ ID NO: 52, and a CDRL3 according to SEQ ID NO: 53. The antibody, or an antigen-binding fragment thereof, according to claim 79, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 170, an extended CDRH2 according to SEQ ID NO: 49, an extended CDRH3 according to SEQ ID NO: 171 , an extended CDRL1 according to SEQ ID NO: 51 , an extended CDRL2 according to SEQ ID NO: 52, and an extended CDRL3 according to SEQ ID NO: 53. The antibody, or an antigen-binding fragment thereof, according to claim 79 or 80, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 54; and a VL having at least 70% identity to SEQ ID NO: 55. The antibody, or an antigen-binding fragment thereof, according to claim 79 or 80, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 54; and a VL having at least 80% identity to SEQ ID NO: 55. The antibody, or an antigen-binding fragment thereof, according to claim 79 or 80, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 54; and a VL having at least 90% identity to SEQ ID NO: 55. The antibody, or an antigen-binding fragment thereof, according to claim 79 or 80, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 54 and a VL comprising an amino acid sequence according to SEQ ID NO: 55. The antibody, or an antigen-binding fragment thereof, according to claim 79 or 80, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 188; and a VL having at least 70% identity to SEQ ID NO: 189. The antibody, or an antigen-binding fragment thereof, according to claim 79 or 80, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 188; and a VL having at least 80% identity to SEQ ID NO: 189. The antibody, or an antigen-binding fragment thereof, according to claim 79 or 80, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 188; and a VL having at least 90% identity to SEQ ID NO: 189. The antibody, or an antigen-binding fragment thereof, according to claim 79 or 80, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 188 and a VL comprising an amino acid sequence according to SEQ ID NO: 189. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 56, a CDRH2 according to SEQ ID NO: 57, a CDRH3 according to SEQ ID NO: 58, a CDRL1 according to SEQ ID NO: 59, a CDRL2 according to SEQ ID NO: 52, and a CDRL3 according to SEQ ID NO: 60. The antibody, or an antigen-binding fragment thereof, according to claim 89, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 172, an extended CDRH2 according to SEQ ID NO: 57, an extended CDRH3 according to SEQ ID NO: 173, an extended CDRL1 according to SEQ ID NO: 59, an extended CDRL2 according to SEQ ID NO: 52, and an extended CDRL3 according to SEQ ID NO: 60. The antibody, or an antigen-binding fragment thereof, according to claim 89 or 90, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 61 ; and a VL having at least 70% identity to SEQ ID NO: 62. The antibody, or an antigen-binding fragment thereof, according to claim 89 or 90, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 61 ; and a VL having at least 80% identity to SEQ ID NO: 62. The antibody, or an antigen-binding fragment thereof, according to claim 89 or 90, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 61 ; and a VL having at least 90% identity to SEQ ID NO: 62. The antibody, or an antigen-binding fragment thereof, according to claim 89 or 90, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 61 and a VL comprising an amino acid sequence according to SEQ ID NO: 62. The antibody, or an antigen-binding fragment thereof, according to claim 89 or 90, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 190; and a VL having at least 70% identity to SEQ ID NO: 191 . The antibody, or an antigen-binding fragment thereof, according to claim 89 or 90, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 190; and a VL having at least 80% identity to SEQ ID NO: 191 . The antibody, or an antigen-binding fragment thereof, according to claim 89 or 90, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 190; and a VL having at least 90% identity to SEQ ID NO: 191. The antibody, or an antigen-binding fragment thereof, according to claim 89 or 90, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 190 and a VL comprising an amino acid sequence according to SEQ ID NO: 191 . The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 63, a CDRH2 according to SEQ ID NO: 64, a CDRH3 according to SEQ ID NO: 65, a CDRL1 according to SEQ ID NO: 66, a CDRL2 according to SEQ ID NO: 36, and a CDRL3 according to SEQ ID NO: 67. The antibody, or an antigen-binding fragment thereof, according to claim 99, wherein the antibody, or the antigen-binding fragment thereof, comprises an extended CDRH1 according to SEQ ID NO: 174, an extended CDRH2 according to SEQ ID NO: 64, an extended CDRH3 according to SEQ ID NO: 175, an extended CDRL1 according to SEQ ID NO: 66, an extended CDRL2 according to SEQ ID NO: 36, and an extended CDRL3 according to SEQ ID NO: 67. The antibody, or an antigen-binding fragment thereof, according to claim 99 or 100, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 68; and a VL having at least 70% identity to SEQ ID NO: 69. The antibody, or an antigen-binding fragment thereof, according to claim 99 or 100, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 68; and a VL having at least 80% identity to SEQ ID NO: 69. The antibody, or an antigen-binding fragment thereof, according to claim 99 or 100, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 68; and a VL having at least 90% identity to SEQ ID NO: 69. The antibody, or an antigen-binding fragment thereof, according to claim 99 or 100, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 68 and a VL comprising an amino acid sequence according to SEQ ID NO: 69. The antibody, or an antigen-binding fragment thereof, according to claim 99 or 100, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity to SEQ ID NO: 192; and a VL having at least 70% identity to SEQ ID NO: 193. The antibody, or an antigen-binding fragment thereof, according to claim 99 or 100, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 80% identity to SEQ ID NO: 192; and a VL having at least 80% identity to SEQ ID NO: 193.
. The antibody, or an antigen-binding fragment thereof, according to claim 99 or 100, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 90% identity to SEQ ID NO: 192; and a VL having at least 90% identity to SEQ ID NO: 193. . The antibody, or an antigen-binding fragment thereof, according to claim 99 or 100, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 192 and a VL comprising an amino acid sequence according to SEQ ID NO: 193. . The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 3, a CDRH3 according to SEQ ID NO: 4, a CDRL1 according to SEQ ID NO: 5 or any one of SEQ ID NOs: 230 - 233, a CDRL2 according to SEQ ID NO: 234, and a CDRL3 according to SEQ ID NO: 7. . The antibody, or an antigen-binding fragment thereof, according to claim 109, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to a sequence according to any one of SEQ ID NO: 200, SEQ ID NO: 201 , and SEQ ID NO: 202; and a VL having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to a sequence according to any one of SEQ ID NO: 203, SEQ ID NO: 204, SEQ ID NO: 205, SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, and SEQ ID NO: 209. 1 . The antibody, or an antigen-binding fragment thereof, according to claim 109 or 110, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to any one of SEQ ID NO: 200, SEQ ID NO: 201 , and SEQ ID NO: 202; and a VL comprising an amino acid sequence according to any one of SEQ ID NO: 203, SEQ ID NO: 204, SEQ ID NO: 205, SEQ ID NO: 206, SEQ ID NO: 207, SEQ ID NO: 208, and SEQ ID NO: 209.
12. The antibody, or an antigen-binding fragment thereof, according to any one of claims 109 to 1 1 1 , wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to SEQ ID NO: 202; and a VL having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to SEQ ID NO:
208. 13. The antibody, or an antigen-binding fragment thereof, according to claim 1 12, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 202; and a VL comprising an amino acid sequence according to SEQ ID NO: 208. 14. The antibody, or an antigen-binding fragment thereof, according to any one of claims 109 to 1 1 1 , wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to SEQ ID NO: 202; and a VL having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to SEQ ID NO:
209. 15. The antibody, or an antigen-binding fragment thereof, according to claim 114, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 202; and a VL comprising an amino acid sequence according to SEQ ID NO: 209. 16. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 13, wherein the antibody, or the antigen-binding fragment thereof, comprises a CDRH1 according to SEQ ID NO: 2, a CDRH2 according to SEQ ID NO: 10, a CDRH3 according to SEQ ID NO: 11 , a CDRL1 according to SEQ ID NO: 12 or SEQ ID NO: 235, a CDRL2 according to SEQ ID NO: 13, SEQ ID NO: 236 or SEQ ID NO: 237, and a CDRL3 according to SEQ ID NO: 14. 17. The antibody, or an antigen-binding fragment thereof, according to claim 1 16, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to a sequence according to SEQ ID NO: 210; and a VL having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to a sequence according to any one of SEQ ID NO: 21 1 , SEQ ID NO: 212, SEQ ID NO:
213, and SEQ ID NO: 214. 18. The antibody, or an antigen-binding fragment thereof, according to claim 116 or 1 17, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 210; and a VL comprising an amino acid sequence according to any one of SEQ ID NO: 211 , SEQ ID NO: 212, SEQ ID NO: 213, and SEQ ID NO: 214. 19. The antibody, or an antigen-binding fragment thereof, according to any one of claims 116 to 118, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to SEQ ID NO: 210; and a VL having at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity to SEQ ID NO:
214. 20. The antibody, or an antigen-binding fragment thereof, according to claim 1 19, wherein the antibody, or the antigen-binding fragment thereof, comprises a VH comprising an amino acid sequence according to SEQ ID NO: 210; and a VL comprising an amino acid sequence according to SEQ ID NO: 214. 21. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, is a humanized antibody. 22. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, is a monoclonal antibody. 23. The antibody according to any one according to the previous claims, wherein the antibody comprises an Fc moiety. The antibody according to any one of the previous claims, wherein the antibody is of the IgG type. The antibody according to claim 124, wherein the antibody is of the IgGI or lgG4 type. The antibody according to any one of the previous claims, wherein the antibody includes the mutations L234A and L235A in its Fc moiety. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, is purified. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims, wherein the antibody, or the antigen-binding fragment thereof, is a single-chain antibody. The antibody, or an antigen-binding fragment thereof, according to claim 128, wherein the antibody, or the antigen-binding fragment thereof, is an scFv. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 127, wherein the antibody, or the antigen-binding fragment thereof, is selected from Fab, Fab', F(ab')2 and Fv. The antibody, or an antigen-binding fragment thereof, according to any one of the previous claims for use as a medicament. The antibody, or an antigen-binding fragment thereof, for use according to claim 131 in prophylaxis or treatment of a neurodegenerative disease, a neuroinflammatory disease, or a peripheral inflammatory disorder. A nucleic acid molecule comprising a polynucleotide encoding the antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130. The nucleic acid molecule according to claim 133 comprising a nucleic acid sequence as set forth in any one of SEQ ID NOs 73 - 140, and SEQ ID NOs 215 - 229; or a sequence variant thereof having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity. A plurality of nucleic acid molecules encoding the antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130, wherein each of the nucleic acid molecules comprises a polynucleotide encoding an immunoglobulin chain of the antibody, or an antigen-binding fragment thereof. The plurality of nucleic acid molecules according to claim 133 comprising a nucleic acid sequence as set forth in any one of SEQ ID NOs 73 - 140, and SEQ ID Nos 215 - 229; or a sequence variant thereof having at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 88%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity. A vector comprising the nucleic acid molecule according to claim 133 or 134. A plurality of vectors comprising the plurality of nucleic acid molecules according to claim 135 or 136. A host cell expressing the antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130, or comprising the vector according to claim 137 or the combination of vectors of claim 138. A method for preparing the antibody, or an antigen-binding fragment thereof, according any one of claims 1 to 130, or immunoglobulin chain(s) thereof, said method comprising
(i) culturing the host cell according to claim 139; and
(ii) isolating the antibody or immunoglobulin chain(s) thereof from the culture. A composition comprising the antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130, the nucleic acid molecule or the plurality of nucleic acid molecules according to any one of claims 133 to 136, the vector or the plurality of vectors according to claim 137 or 138, or the cell according to claim 139. The composition according to claim 141 further comprising a pharmaceutically acceptable excipient, diluent or carrier. A kit comprising one or more of
(i) the antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130;
(ii) the nucleic acid molecule(s) according to any one of claims 133 to 136;
(iii) the vector(s) according to claim 137 or 138;
(iv) the cell according to claim 139; and/or
(v) the composition according to claim 141 or 142. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130, the nucleic acid molecule or the plurality of nucleic acid molecules according to any one of claims 133 to 136, the vector or the plurality of vectors according to claim 137 or 138, the cell according to claim 139, the composition according to claim 141 or 142, or the kit according to claim 143 for use as a medicament. The antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130, the nucleic acid molecule or the plurality of nucleic acid molecules according to any one of claims 133 to 136, the vector or the plurality of vectors according to claim 137 or 138, the cell according to claim 139, the composition according to claim 141 or 142, or the kit according to claim 143 for use in the prophylaxis or treatment of a neurodegenerative or neuroinflammatory disease, or a peripheral inflammatory disorder. The antibody, or the antigen-binding fragment thereof, the nucleic acid or the plurality of nucleic acids, the vector or the plurality of vectors, the cell, or the composition for use according to claim 145, wherein the neurodegenerative or neuroinflammatory disease is associated with the formation of ASC aggregates and/or amyloid-P aggregates. The antibody, or the antigen-binding fragment thereof, the nucleic acid or the plurality of nucleic acids, the vector or the plurality of vectors, the cell, or the composition for use according to claim 145 or 146, wherein the neurodegenerative or neuroinflammatory disease is characterized and/or accompanied by dementia. The antibody, or the antigen-binding fragment thereof, the nucleic acid or the plurality of nucleic acids, the vector or the plurality of vectors, the cell, or the composition for use according to any one of claims 145 to 147, wherein the neurodegenerative or neuroinflammatory disease is selected from Alzheimer's Disease, Parkinsons's Disease, Huntington's disease, Multiple System Atrophy, Multiple Sclerosis, traumatic brain injury, Amyotrophic Lateral Sclerosis, Sinocerebellar ataxia, Frontotemporal Dementia, Frontotemporal Lobar Degeneration, Mild Cognitive Impairment, Parkinson-plus syndromes, Pick disease, Progressive isolated aphasia, Grey-matter degeneration [Alpers], Subacute necrotizing encephalopathy, and Lewy body dementia. The antibody, or the antigen-binding fragment thereof, the nucleic acid or the plurality of nucleic acids, the vector or the plurality of vectors, the cell, or the composition for use according to any one of claims 145 to 148, wherein the neurodegenerative disease is Alzheimer's Disease. The antibody, or the antigen-binding fragment thereof, the nucleic acid or the plurality of nucleic acids, the vector or the plurality of vectors, the cell, or the composition for use according to any one of claims 145 to 148, wherein the neurodegenerative disease is Mild Cognitive Impairment. The antibody, or the antigen-binding fragment thereof, the nucleic acid or the plurality of nucleic acids, the vector or the plurality of vectors, the cell, or the composition for use according to claim 145, wherein the peripheral inflammatory disorder is an inflammatory bowel disease. Use of the antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130 or the composition according to claim 141 or 142 in Un-vitrd} diagnosis of a neurodegenerative or neuroinflammatory disease, or a peripheral inflammatory disorder. Use of the antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130, the nucleic acid molecule or the plurality of nucleic acid molecules according to any one of claims 133 to 136, the vector or the plurality of vectors according to claim 137 or 138, the cell according to claim 139, the composition according to claim 141 or 142, or the kit according to claim 143 in the manufacture of a medicament for prophylaxis, treatment or attenuation of a neurodegenerative or neuroinflammatory disease, or a peripheral inflammatory disorder. A method of treating, ameliorating or reducing a neurodegenerative or neuroinflammatory disease, or a peripheral inflammatory disorder, or lowering the risk of a neurodegenerative or neuroinflammatory disease, or a peripheral inflammatory disorder, comprising: administering to a subject in need thereof, the antibody, or an antigen-binding fragment thereof, according to any one of claims 1 to 130, the nucleic acid molecule or the plurality of nucleic acid molecules according to any one of claims 133 to 136, the vector or the plurality of vectors according to claim 137 or 138, the cell according to claim 139, the composition according to claim 141 or 142, or the kit according to claim 143.
PCT/EP2023/083828 2022-11-30 2023-11-30 Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof Ceased WO2024115700A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
AU2023401113A AU2023401113A1 (en) 2022-11-30 2023-11-30 Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof
KR1020257021663A KR20250134595A (en) 2022-11-30 2023-11-30 Antibodies to apoptosis-associated speck-like protein (ASC) containing CARD and uses thereof
CN202380091643.9A CN120897927A (en) 2022-11-30 2023-11-30 Antibodies against CARD-containing apoptosis-associated speckle-like protein (ASC) and their applications
EP23814496.8A EP4626917A1 (en) 2022-11-30 2023-11-30 Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof
IL320880A IL320880A (en) 2022-11-30 2025-05-13 Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EPPCT/EP2022/083921 2022-11-30
PCT/EP2022/083921 WO2024114906A1 (en) 2022-11-30 2022-11-30 Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof

Publications (1)

Publication Number Publication Date
WO2024115700A1 true WO2024115700A1 (en) 2024-06-06

Family

ID=84688322

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2022/083921 Ceased WO2024114906A1 (en) 2022-11-30 2022-11-30 Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof
PCT/EP2023/083828 Ceased WO2024115700A1 (en) 2022-11-30 2023-11-30 Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/083921 Ceased WO2024114906A1 (en) 2022-11-30 2022-11-30 Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof

Country Status (6)

Country Link
EP (1) EP4626917A1 (en)
KR (1) KR20250134595A (en)
CN (1) CN120897927A (en)
AU (1) AU2023401113A1 (en)
IL (1) IL320880A (en)
WO (2) WO2024114906A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008143954A2 (en) 2007-05-14 2008-11-27 Biogen Idec Ma Inc. Single-chain fc (scfc) regions, binding polypeptides comprising same, and methods related thereto
US20090104200A1 (en) * 2007-07-30 2009-04-23 University Of Miami Modulating inflammasome activity and inflammation in the central nervous system
WO2009155726A2 (en) 2008-06-25 2009-12-30 Esbatech, An Alcon Biomedical Research Unit Llc Humanization of rabbit antibodies using a universal antibody framework
US20110081708A1 (en) 2009-10-07 2011-04-07 Genscript Holdings (Hong Kong) Limited Method of Sequence Optimization for Improved Recombinant Protein Expression using a Particle Swarm Optimization Algorithm
WO2019122270A1 (en) * 2017-12-20 2019-06-27 Michael Heneka Novel means and methods for treating neurodegenerative diseases
US10961306B2 (en) 2016-12-29 2021-03-30 University Of Miami Methods for treating lung inflammation with an anti-ASC antibody
US20210284723A1 (en) * 2018-07-03 2021-09-16 University Of Miami Compositions and methods for treating inflammasome related diseases or conditions

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008143954A2 (en) 2007-05-14 2008-11-27 Biogen Idec Ma Inc. Single-chain fc (scfc) regions, binding polypeptides comprising same, and methods related thereto
US20090104200A1 (en) * 2007-07-30 2009-04-23 University Of Miami Modulating inflammasome activity and inflammation in the central nervous system
WO2009155726A2 (en) 2008-06-25 2009-12-30 Esbatech, An Alcon Biomedical Research Unit Llc Humanization of rabbit antibodies using a universal antibody framework
US20110081708A1 (en) 2009-10-07 2011-04-07 Genscript Holdings (Hong Kong) Limited Method of Sequence Optimization for Improved Recombinant Protein Expression using a Particle Swarm Optimization Algorithm
US10961306B2 (en) 2016-12-29 2021-03-30 University Of Miami Methods for treating lung inflammation with an anti-ASC antibody
WO2019122270A1 (en) * 2017-12-20 2019-06-27 Michael Heneka Novel means and methods for treating neurodegenerative diseases
US20210284723A1 (en) * 2018-07-03 2021-09-16 University Of Miami Compositions and methods for treating inflammasome related diseases or conditions

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
AGRAWAL ISHAN ET AL: "Comprehensive review of ASC structure and function in immune homeostasis and disease", MOLECULAR BIOLOGY REPORTS, SPRINGER NETHERLANDS, NL, vol. 47, no. 4, 2 March 2020 (2020-03-02), pages 3077 - 3096, XP037094785, ISSN: 0301-4851, [retrieved on 20200302], DOI: 10.1007/S11033-020-05345-2 *
AMOR SPUENTES FBAKER DVAN DER VALK P: "Inflammation in neurodegenerative diseases", IMMUNOLOGY, vol. 129, no. 2, February 2010 (2010-02-01), pages 154 - 69, XP055269656, DOI: 10.1111/j.1365-2567.2009.03225.x
BAROJA-MAZO AMARTIN-SANCHEZ FGOMEZ ALMARTINEZ CMAMORES-INIESTA JCOMPAN VBARBERA-CREMADES MYAGUE JRUIZ-ORTIZ EANTON J: "The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response", NAT IMMUNOL, vol. 15, no. 8, August 2014 (2014-08-01), pages 738 - 48
BERNARDO S FRANKLIN ET AL: "The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation", NATURE IMMULOGY, vol. 15, no. 8, 22 June 2014 (2014-06-22), New York, pages 727 - 737, XP055473542, ISSN: 1529-2908, DOI: 10.1038/ni.2913 *
BERTHELOOT DAMIEN ET AL: "Nanobodies dismantle post-pyroptotic ASC specks and counteract inflammation in vivo", vol. 14, no. 6, 19 April 2022 (2022-04-19), US, XP093024077, ISSN: 1757-4676, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.15252/emmm.202115415> DOI: 10.15252/emmm.202115415 *
BORRAS LGUNDE TTIETZ JBAUER UHULMANN-COTTIER VGRIMSHAW JPURECH DM: "Generic approach for the generation of stable humanized single-chain Fv fragments from rabbit monoclonal antibodies", J BIOL CHEM., vol. 285, no. 12, 19 March 2010 (2010-03-19), pages 9054 - 66, XP002668298, DOI: 10.1074/JBC.M109.072876
CARMEN VENEGAS ET AL: "Nanobodies dismantle post-pyroptotic ASC specks and counteract inflammation in vivo", NATURE, vol. 552, no. 7685, 21 December 2017 (2017-12-21), London, pages 355 - 361, XP055473472, ISSN: 0028-0836, DOI: 10.1038/nature25158 *
CHITNIS TWEINER HL: "CNS inflammation and neurodegeneration", J CLIN INVEST, vol. 127, no. 10, 2 October 2017 (2017-10-02), pages 3577 - 3587
CHOTHIA CLESK AM: "Canonical structures for the hypervariable regions of immunoglobulins", J MOL BIOL., vol. 196, 1987, pages 901 - 17, XP024010426, DOI: 10.1016/0022-2836(87)90412-8
DESU HARITHA L. ET AL: "IC100: a novel anti-ASC monoclonal antibody improves functional outcomes in an animal model of multiple sclerosis", vol. 17, no. 1, 4 May 2020 (2020-05-04), XP093024070, Retrieved from the Internet <URL:https://link.springer.com/article/10.1186/s12974-020-01826-0/fulltext.html> DOI: 10.1186/s12974-020-01826-0 *
DESU HLPLASTINI MILLIANO PBRAMLETT HMDIETRICH WDDE RIVERO VACCARI JPBRAMBILLA RKEANE RW: "IC100: a novel anti-ASC monoclonal antibody improves functional outcomes in an animal model of multiple sclerosis", J NEUROINFLAMMATION, vol. 17, no. 1, 4 May 2020 (2020-05-04), pages 143
EDELMAN, G.M.CUNNINGHAM, B.A.GALL, W.E.GOTTLIEB, P.D.RUTISHAUSER, U.WAXDAL, M.J: "The covalent structure of an entire gammaG immunoglobulin molecule", PROC. NATL. ACAD. SCI. USA, vol. 63, 1969, pages 78 - 85, XP055122293, DOI: 10.1073/pnas.63.1.78
FRANKLIN BSBOSSALLER LDE NARDO DRATTER JMSTUTZ AENGELS GBRENKER CNORDHOFF MMIRANDOLA SRAL-AMOUDI A: "The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation", NAT IMMUNOL, vol. 15, no. 8, August 2014 (2014-08-01), pages 727 - 37, XP055473542, DOI: 10.1038/ni.2913
FRANKLIN BSBOSSALLER LDE NARDO DRATTER JMSTUTZ AENGELS GBRENKER CNORDHOFF MMIRANDOLA SRAL-AMOUDI A: "The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation", NAT IMMUNOL., vol. 15, no. 8, August 2014 (2014-08-01), pages 727 - 37, XP055473542, DOI: 10.1038/ni.2913
FRIKER LEA L. ET AL: "[beta]-Amyloid Clustering around ASC Fibrils Boosts Its Toxicity in Microglia", CELL REPORTS, vol. 30, no. 11, 1 March 2020 (2020-03-01), US, pages 3743 - 3754.e6, XP093052983, ISSN: 2211-1247, DOI: 10.1016/j.celrep.2020.02.025 *
FRIKER LLSCHEIBLICH HHOCHHEISER IVBRINKSCHULTE RRIEDEL DLATZ EGEYER MHENEKA MT: "β-Amyloid Clustering around ASC Fibrils Boosts Its Toxicity in Microglia", CELL REP, vol. 30, no. 11, 17 March 2020 (2020-03-17), pages 3743 - 3754, XP093052983, DOI: 10.1016/j.celrep.2020.02.025
GENNARO, REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY, 2000
GEORGE JOHNSONTAI TE WU: "Kabat Database and its applications: 30 years after the first variability plot", NUCLEIC ACIDS RESEARCH, vol. 28, 1 January 2000 (2000-01-01), pages 214 - 218, XP009132432
GREVYS ABERN MFOSS SBRATLIE DBMOEN AGUNNARSEN KSAASE AMICHAELSEN TESANDLIE IANDERSEN JT, FC ENGINEERING OF HUMAN !GG1 FOR ALTERED BINDING TO THE NEONATAL FC RECEPTOR AFFECTS FC EFFECTOR FUNCTIONS, vol. 194, 2015, pages 5497 - 5508
GROTE AHILLER KSCHEER MMUNCH RNORTEMANN BHEMPEL DCJAHN D: "JCat: a novel tool to adapt codon usage of a target gene to its potential expression host", NUCLEIC ACIDS RES., vol. 33, 1 July 2005 (2005-07-01), pages 526 - 31
GUZMAN-MARTINEZ LMACCIONI RBANDRADE VNAVARRETE LPPASTOR MGRAMOS-ESCOBAR N: "Neuroinflammation as a Common Feature of Neurodegenerative Disorders", FRONT PHARMACOL, vol. 10, 12 September 2019 (2019-09-12), pages 1008
HENEKA MICHAEL T ET AL: "Inflammasome signalling in brain function and neurodegenerative disease", NATURE REVIEWS. NEUROSCIENCE, NATURE PUBLISHING GROUP, GB, vol. 19, no. 10, 11 September 2018 (2018-09-11), pages 610 - 621, XP036699534, ISSN: 1471-003X, [retrieved on 20180911], DOI: 10.1038/S41583-018-0055-7 *
HESSELL AJHANGARTNER LHUNTER MHAVENITH CEGBEURSKENS FJBAKKER JMLANIGAN CMSLANDUCCI GFORTHAL DNPARREN PWHI ET AL.: "Fc receptor but not complement binding is important in antibody protection against HIV", NATURE, vol. 449, 2007, pages 101 - 104, XP055077687, DOI: 10.1038/nature06106
HONEGGER APLUCKTHUN A: "Yet another numbering scheme for immunoglobulin variable domains: an automatic modeling and analysis tool", J MOL BIOL., vol. 309, no. 3, 8 June 2001 (2001-06-08), pages 657 - 70, XP004626893, DOI: 10.1006/jmbi.2001.4662
HULSE JBHASKAR K: "Crosstalk Between the NLRP3 Inflammasome/ASC Speck and Amyloid Protein Aggregates Drives Disease Progression in Alzheimer's and Parkinson's Disease", FRONT MOL NEUROSCI, vol. 15, 3 February 2022 (2022-02-03), pages 805169
JU XIN CHINBEVAN KAI-SHENG CHUNGDONG-YUP LEE: "Codon Optimization OnLine (COOL): a web-based multi-objective optimization platform for synthetic gene design", BIOINFORMATICS, vol. 30, 1 August 2014 (2014-08-01), pages 2210 - 2212, XP055640263, DOI: 10.1093/bioinformatics/btu192
KABAT, E.A.: "Sequences of Proteins of Immunological Interest", 1991, DIANE PUBLISHING, article "National Institutes of Health (U.S.) Office of the Director"
KUNIK VASHKENAZI SOFRAN Y: "Paratome: an online tool for systematic identification of antigen-binding regions in antibodies based on sequence or structure", NUCLEIC ACIDS RES., vol. 40, July 2012 (2012-07-01), pages W521 - 4, XP055246927, DOI: 10.1093/nar/gks480
LEFRANC, M.P.GIUDICELLI, V.GINESTOUX, C.BODMER, J.MULLER, W.BONTROP, R.LEMAITRE, M.MALIK, A.BARBIE, V.CHAUME, D: "Imgt, the international immunogenetics database", NUCLEIC ACIDS RES., vol. 27, 1999, pages 209 - 212, XP002637025, DOI: 10.1093/nar/27.1.209
LUND JWINTER GJONES PTPOUND JDTANAKA TWALKER MR ET AL.: "Human Fc gamma Rl and Fc gamma RII interact with distinct but overlapping sites on human IgG", J IMMUNOL., vol. 147, 1991, pages 2657 - 2662, XP009546324, DOI: 10.4049/jimmunol.147.8.2657
MARTIN-SANCHEZ FGOMEZ ALPELEGRIN P: "Isolation of Particles of Recombinant ASC and NLRP3", BIO PROTOC, vol. 5, no. 10, 20 May 2015 (2015-05-20), pages 1480
PEREZ LGCOSTA MRTODD CAHAYNES BFMONTEFIORI DC: "Utilization of immunoglobulin G Fc receptors by human immunodeficiency virus type 1: a specific role for antibodies against the membrane-proximal external region of gp41", J VIROL, vol. 83, 2009, pages 7397 - 7410
PICCOLI LCAMPO IFREGNI CSRODRIGUEZ BMFMINOLA ASALLUSTO FLUISETTI MCORTI DLANZAVECCHIA A: "Neutralization and clearance of GM-CSF by autoantibodies in pulmonary alveolar proteinosis", NAT COMMUN, vol. 6, 2015, pages 1 - 9, XP093023065, DOI: 10.1038/ncomms8375
S. ANGALD.J. KINGM.W. BODMERA. TURNERA.D.G. LAWSONG. ROBERTSB. PEDLEYJ.R. ADAIR: "A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (lgG4) antibody", MOLECULAR IMMUNOLOGY, vol. 30, 1993, pages 105 - 108, Retrieved from the Internet <URL:https://doi.org/10.1016/0161-5890(93)90432-B>
SAUNDERS KO: "Conceptual Approaches to Modulating Antibody Effector Functions and Circulation Half-Life", FRONT IMMUNOL, vol. 10, 7 June 2019 (2019-06-07), pages 1296, XP055654839, DOI: 10.3389/fimmu.2019.01296
SCHMIDT FLLU ACHEN JWRUAN JTANG CWU HPLOEGH HL: "A single domain antibody fragment that recognizes the adaptor ASC defines the role of ASC domains in inflammasome assembly", J EXP MED., vol. 213, no. 5, 2 May 2016 (2016-05-02), pages 771 - 90, XP093024079, DOI: 10.1084/jem.20151790
TAI TE WUELVIN A. KABAT: "An analysis of the sequences of the variable regions of Bence Jones proteins and myeloma light chains and their implications for antibody complementarity", J EXP MED, vol. 132, no. 2, 1 August 1970 (1970-08-01), pages 211 - 250
VENEGAS CKUMAR SFRANKLIN BSDIERKES TBRINKSCHULTE RTEJERA DVIEIRA-SAECKER ASCHWARTZ SSANTARELLI FKUMMER MP: "Microglia-derived ASC specks cross-seed amyloid-(3 in Alzheimer's disease", NATURE, vol. 552, no. 7685, 20 December 2017 (2017-12-20), pages 355 - 361, XP055473472, DOI: 10.1038/nature25158
VENEGAS CKUMAR SFRANKLIN BSDIERKES TBRINKSCHULTE RTEJERA DVIEIRA-SAECKER ASCHWARTZ SSANTARELLI FKUMMER MP: "Microglia-derived ASC specks cross-seed amyloid-P in Alzheimer's disease", NATURE, vol. 552, no. 7685, 20 December 2017 (2017-12-20), pages 355 - 361, XP055473472, DOI: 10.1038/nature25158
VOET SSRINIVASAN SLAML<ANFI MVAN LOO G: "Inflammasomes in neuroinflammatory and neurodegenerative diseases", EMBO MOL MED, vol. 11, no. 6, June 2019 (2019-06-01), pages 10248
ZHANG YFHO M: "Humanization of rabbit monoclonal antibodies via grafting combined Kabat/IMGT/Paratome complementarity-determining regions: Rationale and examples", MABS, vol. 9, no. 3, April 2017 (2017-04-01), pages 419 - 429, XP055982961, DOI: 10.1080/19420862.2017.1289302

Also Published As

Publication number Publication date
WO2024114906A1 (en) 2024-06-06
AU2023401113A1 (en) 2025-05-22
EP4626917A1 (en) 2025-10-08
IL320880A (en) 2025-07-01
KR20250134595A (en) 2025-09-11
CN120897927A (en) 2025-11-04

Similar Documents

Publication Publication Date Title
AU2022252753B2 (en) ANTIBODIES TO a-SYNUCLEIN AND USES THEREOF
RU2571224C2 (en) Humanised anti-axl antibodies
CN107406500B (en) Anti-transthyretin antibodies
CA3077361A1 (en) Anti-transthyretin antibodies
CN107406499A (en) Antithyroidin transporter antibody
US11267878B2 (en) Anti-transthyretin antibodies
EA036048B1 (en) Anti-transthyretin antibodies
EP3478716A2 (en) Anti-transthyretin antibodies
TW201942135A (en) Anti-CD27 antibody, antigen binding fragment thereof, and pharmaceutical use thereof
KR20200138726A (en) Therapeutic anti-SPLA2-GIB antibodies and uses thereof
JP2025083342A (en) TGF-βRII BINDING PROTEINS
EP2270053A1 (en) Humanized AXL antibodies
JP2025517760A (en) Anti-allergen antibodies and uses thereof
EP4626917A1 (en) Antibodies against apoptosis-associated speck-like protein containing a card (asc) and uses thereof
KR20250027598A (en) KLRB1 binder and method of use thereof
JP2025503707A (en) Galectin-10 Antibody
JP2025540752A (en) Antibodies against CARD-containing apoptosis-associated speck-like protein (ASC) and uses thereof
CN120225554A (en) VEGF antibodies
WO2022124247A1 (en) Agent for preventing or treating frontotemporal lobar degeneration
CN113164601A (en) Isolated antigen binding protein and uses thereof
HK1240247B (en) Anti‐transthyretin antibodies
HK1240247A1 (en) Anti‐transthyretin antibodies

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: 23814496

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: AU2023401113

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 320880

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2023401113

Country of ref document: AU

Date of ref document: 20231130

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2025531054

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2025531054

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2023814496

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2023814496

Country of ref document: EP

Effective date: 20250630

WWE Wipo information: entry into national phase

Ref document number: 202380091643.9

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2023814496

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 202380091643.9

Country of ref document: CN