[go: up one dir, main page]

WO2025019730A1 - Engineered ga antibodies for therapy development - Google Patents

Engineered ga antibodies for therapy development Download PDF

Info

Publication number
WO2025019730A1
WO2025019730A1 PCT/US2024/038646 US2024038646W WO2025019730A1 WO 2025019730 A1 WO2025019730 A1 WO 2025019730A1 US 2024038646 W US2024038646 W US 2024038646W WO 2025019730 A1 WO2025019730 A1 WO 2025019730A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
antigen
amino acid
acid sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/US2024/038646
Other languages
French (fr)
Inventor
Laura Ranum
Lisa ROMANO
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.)
University of Florida
University of Florida Research Foundation Inc
Original Assignee
University of Florida
University of Florida Research Foundation Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Florida, University of Florida Research Foundation Inc filed Critical University of Florida
Publication of WO2025019730A1 publication Critical patent/WO2025019730A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • 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

Definitions

  • Microsatellite repeat expansions are known to cause more than forty neurodegenerative disorders. Molecular features common to many of these disorders include the accumulation of RNA foci containing sense and antisense expansion transcripts and the accumulation of proteins from repeat-associated non- AUG (RAN) translation. RAN translation can occur across a broad range of repeat lengths from pre-mutation lengths ( ⁇ 30 - 40 repeats) to full expansions (up to 10,000 repeats).
  • RAN proteins are toxic and contribute to a growing number of diseases and disorders, including, but not limited to, amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), Huntington’s disease (HD), Alzheimer’s disease (AD), and Fragile X Tremor Ataxia Syndrome (FXTAS).
  • ALS amyotrophic lateral sclerosis
  • FTD frontotemporal dementia
  • DM1 myotonic dystrophy type 1
  • DM2 myotonic dystrophy type 2
  • HD Huntington’s disease
  • AD Alzheimer’s disease
  • FXTAS Fragile X Tremor Ataxia Syndrome
  • compositions and methods for the diagnosis and/or treatment of certain neurodegenerative diseases for example those diseases associated with repeat-associated non-ATG (RAN) translation proteins, such as Alzheimer’s disease (AD) and amyotrophic lateral sclerosis (ALS).
  • RAN repeat-associated non-ATG
  • AD Alzheimer’s disease
  • ALS amyotrophic lateral sclerosis
  • the disclosure is based, in part, on antibodies (e.g., monoclonal antibodies) and antigen-binding fragments thereof (e.g., single-chain variable fragments, scFvs) that bind to poly-Glycine- Alanine (poly(GA)) RAN proteins.
  • the disclosure relates to methods of reducing RAN protein (e.g., poly(GA) RAN protein) aggregation in a subject by administering the antibodies or antigen-binding fragments thereof to the subject. In some embodiments, the disclosure relates to methods of treating a RAN protein-associated disease by administering to a subject in need thereof the antibodies or antigen-binding fragments thereof.
  • RAN protein e.g., poly(GA) RAN protein
  • the disclosure provides an antibody or antigen-binding fragment thereof, wherein the antibody or antigen-binding fragment comprises a heavy chain variable region (VH) comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 1; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 2; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 3.
  • VH heavy chain variable region
  • the disclosure provides an antibody or antigen-binding fragment thereof, wherein the antibody or antigen-binding fragment comprises a light chain variable region (VL) comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 4; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 5; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 6.
  • VL light chain variable region
  • an antibody or antigen-binding fragment comprises a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 7.
  • an antibody or antigen-binding fragment comprises a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 12.
  • an antibody of antigen-binding fragment comprises a heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 8 or 10.
  • an antibody of antigen-binding fragment comprises a light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 13 or 15.
  • an antibody or antigen-binding fragment thereof comprises a heavy chain variable region (VH) comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 20; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 21; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 22.
  • VH heavy chain variable region
  • the disclosure provides an antibody or antigen-binding fragment thereof that specifically binds to a RAN protein, wherein the antibody or antigen-binding fragment comprises a light chain variable region (VL) comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 23; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 24; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 25.
  • VL light chain variable region
  • an antibody or antigen-binding fragment comprises a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 26.
  • an antibody or antigen-binding fragment comprises a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 31.
  • an antibody of antigen-binding fragment comprises a heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 27 or 29.
  • an antibody of antigen-binding fragment comprises a light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 32 or 34.
  • the disclosure provides a single chain variable fragment (scFv) comprising a heavy chain variable region comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 1; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 2; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 3.
  • scFv single chain variable fragment
  • the disclosure provides a single chain variable fragment (scFv) comprising a light chain variable region comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 4; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 5; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 6.
  • scFv single chain variable fragment
  • an scFv comprises a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 17.
  • an scFv comprises a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 18.
  • an scFv comprises a linker molecule connecting the heavy chain variable region to the light chain variable region.
  • a linker molecule comprises a poly-GS linker.
  • an antibody or antigen-binding fragment thereof or scFv further comprises a signal peptide.
  • an antibody or antigen-binding fragment thereof or scFv binds poly(GA) RAN protein.
  • the disclosure provides a composition comprising an antibody or antigen-binding fragment thereof, or scFv, as described herein and a pharmaceutically acceptable carrier or buffer.
  • an isolated nucleic acid encoding an antibody or antigen-binding fragment thereof, or scFv, as described herein.
  • an isolated nucleic acid comprises the nucleotide sequence set forth in any one of SEQ ID NOs: 36- 40.
  • the disclosure provides a vector comprising an isolated nucleic acid as described herein.
  • the vector is a plasmid.
  • a plasmid comprises a sequence as set forth in any one of SEQ ID NOs: 42-46.
  • the disclosure provides a cell transformed with an isolated nucleic acid or vector as described herein.
  • the cell is a mammalian cell. In some embodiments, the cell is a human cell.
  • the disclosure provides a method for administering an antibody or antigen-binding fragment to a subject, the method comprising administering the antibody or antigen-binding fragment or scFv as described herein, to the subject.
  • the disclosure provides a method for reducing poly(GA) RAN protein aggregation in a subject, the method comprising administering an antibody or antigen-binding fragment, or scFv, as described herein to the subject.
  • the disclosure provides a method for treating a subject having ALS, the method comprising administering an antibody or antigen-binding fragment, or an scFv, as described herein to the subject.
  • a subject is a mammal. In some embodiments, a subject is a human. In some embodiments, a subject expresses one or more RAN proteins. In some embodiments, a subject expresses a poly(GA) RAN protein.
  • a subject has or is suspected of having Alzheimer’s disease, amyotrophic lateral sclerosis (ALS), frontotemporal dementia, myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), spinocerebellar ataxia (SCA) type 1 (SCA), SCA2, SCA3, SCA6, SCA7, SCA8, SCAIO, SCA12, SCA17, SCA31, SCA36, spinal bulbar muscular atrophy, dentatorubral-pallidoluysian atrophy (DRPLA), Huntington's disease (HD), Fragile X Tremor Ataxia Syndrome (FXTAS), Fuch's endothelial corneal dystrophy (FECD), Huntington's disease-like 2 syndrome (HDL2), Fragile X syndrome (FXS), a disorder related to 7pl 1 .2 folatesensitive fragile site FRA7A, a disorder related to folate-sensitive fragile site 2ql 1 FRA2
  • ALS
  • a subject has or is suspected of having ALS.
  • FIG. 1 shows representative data for immunohistochemistry (IHC) assays for anti- poly(GA) monoclonal antibodies.
  • IHC immunohistochemistry
  • m-GA mouse anti-GA polyclonal antibody
  • EV vehicle- only negative control
  • ScFv 515 27B11 ScFv format
  • IgGl 515A 27B11 monoclonal antibody in IgGl format
  • IgGl 515B 27B11 monoclonal antibody in IgGl format
  • IgG2 515 27B 11 monoclonal antibody in IgG2 format.
  • FIG. 2 shows representative data indicating expression of anti-poly(GA) monoclonal antibody 27B11 in several formats (IgGl, IgG2, ScFv) lowers levels of (GA)eo-GFP RAN protein in cells.
  • compositions and methods for the diagnosis and/or treatment of certain neurodegenerative diseases for example those diseases associated with repeat-associated non-ATG (RAN) translation proteins, such as Alzheimer’s disease (AD) or ALS.
  • the disclosure is based, in part, on antibodies (e.g., monoclonal antibodies) and antigenbinding fragments thereof (e.g., single-chain variable fragments, scFvs) that bind to poly- Glycine-Alanine (poly(GA)) RAN proteins.
  • the disclosure relates to methods of reducing RAN protein (e.g., poly(GA) RAN protein) aggregation in a subject by administering the antibodies or antigen-binding fragments thereof to the subject.
  • the disclosure relates to methods of treating a RAN protein-associated disease by administering to a subject in need thereof the antibodies or antigen-binding fragments thereof.
  • a “RAN protein (repeat- associated non-ATG translated protein)” is a polypeptide that is translated from sense or antisense RNA sequences bidirectionally transcribed from a repeat expansion mutation in the absence of an AUG initiation codon.
  • RAN protein-encoding sequences can be found in the genome at multiple loci, including but not limited to open reading frame 72 of chromosome 9 (C9orf72), open reading frame 80 of chromosome 2 (C2orf80), LRP8, CASP8, CRNDE, EXOC6B, SV2B, PPML1, ADARB2, GREB1, and MSM01.
  • C9orf72 open reading frame 72 of chromosome 9
  • C2orf80 open reading frame 80 of chromosome 2
  • LRP8 CASP8, CRNDE
  • EXOC6B SV2B
  • the protein associated with C9orf72 is currently poorly characterized but known to be abundant in neurons
  • C9orf72 protein is believed to localized in presynaptic termini. C9orf72 protein likely impacts transcription, translation and intra-cellular localization of RNA. C9orf72 gene contains a GGGGCC repeat. This hexanucleotide repeat occurs in variable repeat numbers, and small numbers of repeats are not associated with any pathology.
  • RAN proteins comprise expansion repeats of a single amino acid, di-amino acid, tri-amino acid, or quad-amino acid e.g., tetra-amino acid), termed poly amino acid repeats.
  • poly amino acid repeats For example, “AAAAAAAAAAAAAAAAAAAAAAAAAA” (SEQ ID NO: 47) (poly-Alanine), “LLLLLLLLLLLLLLLLLLLL” (SEQ ID NO: 48) (poly-Leucine), “SSSSSSSSSSSSSSSSSSSSSSSSSSSSSSSSSSSEQ ID NO: 49) (poly-Serine), or “CCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCC” (SEQ ID NO: 50) (poly- Cysteine) are poly amino acid repeats that are each 20 amino acid residues in length.
  • tetra-amino acid repeats examples include LPACLPACLPAC (SEQ ID NO: 56) (e.g., poly-LPAC) and QAGRQAGRQAGR (SEQ ID NO: 57) (e.g., poly-QAGR).
  • RAN proteins can have a poly amino acid repeat of at least 20, at least 25, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, or at least 200 amino acid residues in length.
  • a RAN protein has a poly amino acid repeat more than 200 amino acid residues (e.g., 500, 1000, 5000, 10,000, etc.) in length.
  • RAN proteins are translated from abnormal repeat expansions (e.g., TCT repeats, hexanucleotide repeats such as GGGGCC, etc.) of DNA.
  • the disclosure is based, in part, on the identification of micro satellite repeats in certain subjects having a RAN protein- associated disease that is characterized by expression of one or more (e.g., 2, 3, 4, 5, or more) RAN proteins, for example poly(Glycine- Alanine) [poly(GA)].
  • the disease status of a subject having or suspected of having a RAN protein-associated disease is classified by the number and/or type of micro satellite repeats present (e.g., detected) in the subject (e.g., in the genome of a subject or in a gene of the subject).
  • a “subject having or suspected of having a disease (e.g., neurological diseases) associated with RAN protein expression, translation, and/or accumulation” generally refers to a subject exhibiting one or more signs and symptoms of a neurodegenerative disease, including but not limited to memory deficit (e.g., short term memory loss), confusion, deficiencies of executive functions (e.g., attention, planning, flexibility, abstract thinking, etc.), loss of speech, degeneration or loss of motor skills, etc., or a subject having or being identified as having one or more genetic mutations associated with RAN protein expression, translation, and/or accumulation.
  • memory deficit e.g., short term memory loss
  • confusion e.g., deficiencies of executive functions
  • deficiencies of executive functions e.g., attention, planning, flexibility, abstract thinking, etc.
  • loss of speech e.g., degeneration or loss of motor skills, etc.
  • a subject having or being identified as having one or more genetic mutations associated with RAN protein expression, translation, and/or accumulation e.g., neurological diseases
  • a “subject having or suspected of having amyotrophic lateral sclerosis (ALS)” can be a subject exhibiting one or more signs and symptoms of AD, including but not limited to memory deficit (e.g., short term memory loss), confusion, deficiencies of executive functions (e.g., attention, planning, flexibility, abstract thinking, etc.), loss of speech, degeneration or loss of motor skills, etc., or a subject having or being identified as having one or more genetic mutations associated with ALS, for example mutations in specific genes including C9orf72.
  • a subject has been diagnosed as having ALS by a medical professional.
  • a subject can be a mammal (e.g., human, mouse, rat, dog, cat, or pig).
  • a subject is a non-human animal, for example a mouse, rat, guinea pig, cat dog, horse, camel, etc.
  • the subject is a human.
  • a subject having less than 10 repeat sequences does not exhibit signs or symptoms of a RAN protein-associated disease characterized by RAN protein translation.
  • a subject having between 10 and 40 repeats may or may not exhibit one or more signs or symptoms of a RAN protein-associated disease characterized by RAN protein translation.
  • a subject having more than 40 trinucleotide repeats exhibits one or more signs or symptoms of a RAN protein- associated disease characterized by RAN protein translation.
  • a subject is identified as having a RAN protein-associated disease characterized by large (>100) number of repeats. Micro satellite repeat sequences encoding RAN proteins are generally known.
  • the RAN protein-associated disease is ALS.
  • a subject having or suspected of having a RAN protein-associated disease has one or more micro satellite repeat sequences encoding a poly(GA) RAN protein.
  • a micro satellite repeat sequence encoding poly(GA) proteins comprises the sequence GGGGCC.
  • the disclosure relates to the discovery that RAN protein (e.g., poly(GA), etc.) aggregation patterns are length-dependent.
  • RAN proteins having poly amino acid repeats that are >20, >48, or >80 residues in length aggregate differently in the brain of a subject.
  • the differential aggregation properties of RAN proteins having different lengths can be used to detect RAN proteins in a biological sample. Longer RAN proteins are found at higher levels in biological samples, such as blood, serum, or CSF.
  • RAN proteins having poly amino acid repeats >40, >50, >60, >70, or >80 amino acid residues in length are detectable in a biological sample.
  • the antibody or antigen binding fragment specifically binds to a poly(glycine- alanine) [poly(GA)] RAN protein.
  • the antibody binds directly to the poly(GA) repeat region (e.g., binds to the GAGAGA (SEQ ID NO: 58) motif).
  • the antibody binds to a region of the RAN protein (e.g., poly(GA) RAN protein) that is not the repeat region, for example a unique C-terminal amino acid sequence of the RAN protein.
  • an antibody as used herein, broadly refers to an immunoglobulin molecule or any functional mutant, variant, or derivation thereof. It is desired that functional mutants, variants, and derivations thereof, as well as antigen-binding fragments, retain the essential epitope binding features of an Ig molecule.
  • Antibodies are capable of specific binding to a target through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule.
  • an intact or full-length antibody comprises two heavy chains and two light chains. Each heavy chain contains a heavy chain variable region (VH) and a first, second and third constant regions (CHI, CH2 and CH3). Each light chain contains a light chain variable region (VL) and a constant region (CL).
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • CDR constituents on the heavy chain are referred to as CDRH1, CDRH2, and CDRH3, while CDR constituents on the light chain are referred to as CDRL1, CDRL2, and CDRL3.
  • the CDRs typically refer to the Kabat CDRs, as described in Sequences of Proteins of Immunological Interest, US Department of Health and Human Services (1991), eds. Kabat et al.
  • Another standard for characterizing the antigen binding site is to refer to the hypervariable loops as described by Chothia. See, e.g., Cho thia, D. et al. (1992) J. Mol. Biol. 227:799-817; and Tomlinson et al. (1995) EMBO J. 14:4628-4638.
  • Still another standard is the AbM definition used by Oxford Molecular’s AbM antibody modeling software. See, generally, e.g., Protein Sequence and Structure Analysis of Antibody Variable Domains.
  • Each VH and VL is composed of three CDRs and four FRs, arranged from aminoterminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • a full-length antibody can be an antibody of any class, such as IgD, IgE, IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class.
  • immunoglobulins can be assigned to different classes.
  • immunoglobulins There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • antigen-binding fragment refers to any derivative of an antibody which is less than full-length, and that can bind specifically to a target.
  • antigen-binding fragments provided herein retain the ability to specifically bind to a RAN protein.
  • An antigenbinding fragment may comprise the heavy chain variable region (VH), the light chain variable region (VL), or both.
  • VH heavy chain variable region
  • VL light chain variable region
  • Each of the VH and VL typically contains three complementarity determining regions CDR1, CDR2, and CDR3.
  • antigen binding fragments include, but are not limited to, Fab, Fab’, F(ab’)2, scFv, Fv, dsFv, diabody, affibodies, and Fd fragments.
  • Antigen binding fragments may be produced by any appropriate means. For instance, an antigen binding fragment may be enzymatically or chemically produced by fragmentation of an intact antibody, or it may be recombinantly produced from a gene encoding the partial antibody sequence. Alternatively, an antigen binding fragment may be wholly or partially synthetically produced. An antigen binding fragment may optionally be a single chain antibody fragment. Alternatively, a fragment may comprise multiple chains which are linked together, for instance, by disulfide linkages. An antigen binding fragment may also optionally be a multimolecular complex. A functional antigen binding fragment will typically comprise at least about 50 amino acids and more typically will comprise at least about 200 amino acids.
  • Single-chain Fvs are recombinant antigen binding fragments consisting of only the variable light chain (VE) and variable heavy chain (VH) covalently connected to one another by a polypeptide linker.
  • Either VE or VH may be the NH2-terminal domain.
  • the polypeptide linker may be of variable length and composition so long as the two variable domains are bridged without serious steric interference.
  • the linkers are comprised primarily of stretches of glycine and serine residues with some glutamic acid or lysine residues interspersed for solubility.
  • ScFvs are encompassed within the term “antigen-binding fragment.”
  • Diabodies are dimeric scFvs.
  • the components of diabodies typically have shorter peptide linkers than most scFvs, and they show a preference for associating as dimers (see, e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2: 1121-1123).
  • Diabodies are also encompassed within the term “antigen-binding fragment.”
  • a Fv fragment is an antigen binding fragment which consists of one VH and one VE domain held together by noncovalent interactions.
  • the two domains of the Fv fragment, VE and VH can be coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VE and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • scFv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody.
  • the term dsFv is used herein to refer to an Fv with an engineered intermolecular disulfide bond to stabilize the VH-VE pair. dsFvs are also encompassed within the term “antigen-binding fragment.”
  • a F(ab’)2 fragment is an antigen binding fragment essentially equivalent to that obtained from immunoglobulins (typically IgG) by digestion with an enzyme pepsin at pH 4.0-4.5. The fragment may be recombinantly produced.
  • F(ab’)2 are also encompassed within the term “antigen-binding fragment.”
  • a Fab fragment is an antigen binding fragment essentially equivalent to that obtained by reduction of the disulfide bridge or bridges joining the two heavy chain pieces in the F(ab’)2 fragment.
  • the Fab’ fragment may be recombinantly produced.
  • Fab’ are also encompassed within the term “antigen-binding fragment.”
  • a Fab fragment is an antigen binding fragment essentially equivalent to that obtained by digestion of immunoglobulins (typically IgG) with the enzyme papain.
  • the Fab fragment may be recombinantly produced.
  • the heavy chain segment of the Fab fragment is the Fd piece.
  • Fab fragments are also encompassed within the term “antigen-binding fragment.”
  • An affibody is a small protein comprising a three-helix bundle that functions as an antigen binding molecule (e.g., an antibody mimetic).
  • an antigen binding molecule e.g., an antibody mimetic
  • affibodies are approximately 58 amino acids in length and have a molar mass of approximately 6 kDa.
  • Affibody molecules with unique binding properties are acquired by randomization of 13 amino acids located in two alpha-helices involved in the binding activity of the parent protein domain.
  • Specific affibody molecules binding a desired target protein can be isolated from pools (libraries) containing billions of different variants, using methods such as phage display.
  • Affibodies are also encompassed within the term “antigen-binding fragment.”
  • human antibody refers to antibodies having variable and constant regions corresponding substantially to, or derived from, antibodies obtained from human subjects, e.g., encoded by human germline immunoglobulin sequences or variants thereof.
  • Human antibodies may include one or more amino acid residues not encoded by human germline immunoglobulin sequences e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). Such mutations may present in one or more of the CDRs, particularly CDR3, or in one or more of the framework regions.
  • the human antibodies may have at least one, two, three, four, five, or more positions replaced with an amino acid residue that is not encoded by the human germline immunoglobulin sequence.
  • the term "human antibody”, as used herein is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • recombinant human antibody is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library (Hoogenboom H. R., (1997) TIB Tech. 15:62-70; Azzazy H., and Highsmith W. E., (2002) Clin. Biochem. 35:425-445; Gavilondo J. V., and Larrick J. W. (2002) BioTechniques 29: 128-145;
  • such recombinant human antibodies may be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies may be sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • the antibody or antigen-binding fragment comprises a heavy chain comprising an amino acid sequence represented by any one of SEQ ID NOs: 8, 10, 27, and 29.
  • the anti-RAN antibodies and antigen binding fragments of the disclosure comprise a light chain comprising an amino acid sequence represented by any one of SEQ ID NOs: 13, 15, 32, and 34.
  • the antibody or antigen-binding fragment comprises a heavy chain comprising an amino acid sequence represented by any one of SEQ ID NOs: 8, 10, 27, and 29 and a light chain comprising an amino acid sequence represented by any one of SEQ ID NOs: 13, 15, 32, and 34.
  • the antibody is humanized.
  • the antibody comprises an IgGl, IgG2, IgG3, IgG4, IgAl or IgA2 heavy chain constant region.
  • the anti-RAN antibodies or antigen binding fragments may or may not include the framework region of the antibodies, for example the framework region amino acid sequences.
  • anti-RAN antibodies are murine antibodies.
  • anti-RAN antibodies are chimeric or humanized antibodies.
  • the antibody or antigen binding fragment comprises a VH sequence as set forth in any one of SEQ ID NOs: 7, 17, and 26. In some embodiments, the antibody or antigen binding fragment comprises a VL sequence as set forth in any one of SEQ ID NOs: 12, 18, or 31. In some embodiments, the antibody or antigen binding fragment comprises a VH sequence as set forth in any one of SEQ ID NOs: 7, 17, and 26 and a VL sequence as set forth in any one of SEQ ID NOs: 12, 18, or 31. In some embodiments, the antibody is humanized. In some embodiments, the antibody comprises an IgGl, IgG2, IgG3, IgG4, IgAl or IgA2 heavy chain constant region.
  • the antibody or antigen binding fragment comprises a VH sequence as set forth in SEQ ID NO. 7 and a VL sequence as set forth in SEQ ID NO. 12.
  • the antibody or antigen binding fragment comprises a VH sequence as set forth in SEQ ID NO. 17 and a VL sequence as set forth in SEQ ID NO. 18.
  • the antibody or antigen binding fragment comprises a VH sequence as set forth in SEQ ID NO. 26 and a VL sequence as set forth in SEQ ID NO. 31.
  • antibody or antigen-binding fragment comprises six complementarity determining regions (CDRs): CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3, wherein CDRH1 comprises a sequence as set forth in SEQ ID NO: 1, CDRH2 comprises a sequence as set forth in SEQ ID NO: 2, CDRH3 comprises a sequence as set forth in SEQ ID NO: 3, CDRL1 comprises a sequence as set forth in SEQ ID NO: 4, CDRL2 comprises a sequence as set forth in SEQ ID NO: 5, and CDRL3 comprises a sequence as set forth in SEQ ID NO: 6.
  • the antibody is humanized.
  • the antibody comprises an IgGl, IgG2, IgG3, IgG4, IgAl or IgA2 heavy chain constant region.
  • antibody or antigen-binding fragment comprises six complementarity determining regions (CDRs): CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3, wherein CDRH1 comprises a sequence as set forth in SEQ ID NO: 20, CDRH2 comprises a sequence as set forth in SEQ ID NO: 21, CDRH3 comprises a sequence as set forth in SEQ ID NO: 22, CDRL1 comprises a sequence as set forth in SEQ ID NO: 23, CDRL2 comprises a sequence as set forth in SEQ ID NO: 24, and CDRL3 comprises a sequence as set forth in SEQ ID NO: 25.
  • the antibody is humanized.
  • the antibody comprises an IgGl, IgG2, IgG3, IgG4, IgAl or IgA2 heavy chain constant region.
  • the disclosure contemplates variants (e.g., homologs) of amino acid and nucleic acid sequences for the heavy chain variable region and light chain variable region of the antibodies.
  • “Homology” refers to the percent identity between two polynucleotides or two polypeptide moieties.
  • substantially homology when referring to a nucleic acid, or fragment thereof, indicates that, when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in about 90 to 100% of the aligned sequences.
  • nucleic acid sequences sharing substantial homology are 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% at least 99% sequence identity.
  • substantially homology indicates that, when optimally aligned with appropriate gaps, insertions or deletions with another polypeptide, there is nucleotide sequence identity in about 90 to 100% of the aligned sequences.
  • the term "highly conserved" means at least 80% identity, preferably at least 90% identity, and more preferably, over 97% identity.
  • highly conserved proteins share at least 85%, 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% at least 99% identity.
  • highly conserved may refer to 100% identity. Identity is readily determined by one of skill in the art by, for example, the use of algorithms and computer programs known by those of skill in the art.
  • RAN antibodies of the disclosure can bind to a RAN protein with high affinity, e.g., with a Kd less than 10' 7 M, 10' 8 M, 10' 9 M, 10' 10 M, 10 1 M or lower.
  • anti-RAN antibodies or antigen binding fragments can bind to a RAN protein with an affinity between 5 pM and 500 nM, e.g., between 50 pM and 100 nM, e.g., between 500 pM and 50 nM.
  • the disclosure also includes antibodies or antigen binding fragments that compete with any of the antibodies described herein for binding to RAN proteins and that have an affinity of 50 nM or lower (e.g., 20 nM or lower, 10 nM or lower, 500 pM or lower, 50 pM or lower, or 5 pM or lower).
  • the affinity and binding kinetics of the anti-RAN protein antibody can be tested using any method known in the art including but not limited to biosensor technology (e.g., OCTET or BIACORE).
  • anti-RAN antibodies of the present disclosure include the VH, VL, and CDR, amino acid sequences shown in Table 1 below.
  • Table 1 Representative sequences (listed as SEQ ID NOs) of developed anti-poly(GA) monoclonal antibodies
  • antibody clone 27B11 binds to poly(GA).
  • clone 27B11 is an IgGl antibody (e.g., comprises an IgGl heavy chain constant region).
  • clone 27B11 is an IgG2 antibody (e.g., comprises an IgG2 heavy chain constant region).
  • clone 27B11 is an scFv.
  • antibody clone 23H2 binds to poly(GA).
  • antibody clone 23H2 is an IgGl antibody (e.g., comprises an IgGl heavy chain constant region).
  • antibody clone 23H2 is an IgG2 antibody (e.g., comprises an IgG2 heavy chain constant region).
  • Anti-RAN antibodies may be used to treat, or assist in the treatment of, one or more symptoms of a disease associated with RAN proteins.
  • the disease associated with RAN proteins is selected from the group consisting of: amyotrophic lateral sclerosis (ALS), or frontotemporal dementia; myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2); spinocerebellar ataxia types 1, 2, 3, 6, 7, 8, 10, 12, 17, 31, and 36; spinal bulbar muscular atrophy; dentatorubral-pallidoluysian atrophy (DRPLA); Huntington's disease (HD); Fragile X Tremor Ataxia Syndrome (FXTAS); Fuch's endothelial corneal dystrophy (FECD); Huntington's disease-like 2 syndrome (HDL2); Fragile X syndrome (FXS); disorders related to 7pl 1 .2 folate-sensitive fragile site FRA7A; disorders related to folatesensitive fragile site 2ql 1 FRA2A;
  • the anti-RAN antibodies may be used to treat, or assist in the treatment of, one or more symptoms of a disease associated with RAN proteins, for example by administering a therapeutically effective amount of one or more anti-RAN antibodies to a subject diagnosed as having one or more symptoms of a disease associated with RAN proteins (e.g., the early stages of ALS) or being at risk of developing a disease associated with RAN proteins (e.g., based on one or more assays described in this application).
  • a therapeutically effective amount of one or more anti-RAN antibodies to a subject diagnosed as having one or more symptoms of a disease associated with RAN proteins (e.g., the early stages of ALS) or being at risk of developing a disease associated with RAN proteins (e.g., based on one or more assays described in this application).
  • a disease e.g., a disease associated with poly(GA) RAN translation, for example ALS
  • a disease means to reduce the frequency or severity of at least one sign or symptom of a disease or disorder experienced by a subject.
  • the compositions described above or elsewhere herein are typically administered to a subject in an effective amount, that is, an amount capable of producing a desirable result.
  • the desirable result will depend upon the active agent being administered.
  • a therapeutically acceptable amount of an anti-RAN protein antibody may be an amount that is capable of treating a disease, e.g., ALS, by reducing expression and/or aggregation of RAN proteins and/or appearance or number of RNA foci comprising RAN protein-encoding microsatellite repeat sequences.
  • dosage for any one subject depends on many factors, including the subject's size, body surface area, age, the particular composition to be administered, the active ingredient(s) in the composition, time and route of administration, general health, and other drugs being administered concurrently.
  • one or more of the anti-RAN antibody or antigen binding fragments disclosed herein are administered to a subject, wherein the subject has been characterized as having a disease associated with RAN proteins by the detection of at least one RAN protein in a biological sample obtained from the subject.
  • polyclonal antibodies are produced by inoculation of a suitable mammal, such as a mouse, rabbit or goat.
  • An antigen is injected into the mammal. This induces the B- lymphocytes to produce IgG immunoglobulins specific for the antigen.
  • This polyclonal IgG is purified from the mammal's serum.
  • Monoclonal antibodies are generally produced by a single cell line (e.g., a hybridoma cell line).
  • an anti-RAN antibody is purified (e.g., isolated from serum).
  • an antigen comprises a poly(GA) RAN protein repeat sequence.
  • antibodies can be produced using recombinant DNA methods.
  • Monoclonal antibodies may also be produced by generation of hybridomas (see, e.g., Kohler and Milstein (1975) Nature, 256: 495-499) in accordance with known methods.
  • Hybridomas formed in this manner are then screened using standard methods, such as enzyme-linked immunosorbent assay (ELISA; e.g., RCA-based ELISA or rtPCR-based ELISA) and surface plasmon resonance (e.g., OCTET or BIACORE) analysis, to identify one or more hybridomas that produce an antibody that specifically binds with a specified antigen.
  • ELISA enzyme-linked immunosorbent assay
  • OCTET BIACORE
  • any form of the specified antigen e.g., a RAN protein
  • the immunogen e.g., recombinant antigen, naturally occurring forms, any variants or fragments thereof.
  • One exemplary method of making antibodies includes screening protein expression libraries that express antibodies or fragments thereof (e.g., scFv), e.g., phage or ribosome display libraries. Phage display is described, for example, in Ladner et al., U.S. Pat. No. 5,223,409; Smith (1985) Science 228: 1315-1317; Clackson et al. (1991) Nature, 352: 624-628; Marks et al. (1991) J. Mol.
  • a monoclonal antibody is obtained from the non-human animal, and then modified, e.g., made chimeric, using recombinant DNA techniques known in the art.
  • modified e.g., made chimeric, using recombinant DNA techniques known in the art.
  • a variety of approaches for making chimeric antibodies have been described. See, e.g., Morrison et al., Proc. Natl. Acad. Sci. U.S.A.
  • Antibodies can also be humanized by methods known in the art. For example, monoclonal antibodies with a desired binding specificity can be commercially humanized (Scotgene, Scotland; and Oxford Molecular, Palo Alto, Calif.). Fully humanized antibodies, such as those expressed in transgenic animals are within the scope of the invention (see, e.g., Green et al. (1994) Nature Genetics 7, 13; and U.S. Patent Nos. 5,545,806 and 5,569,825). For additional antibody production techniques, see, Antibodies: A Laboratory Manual, Second Edition. Edited by Edward A. Greenfield, Dana-Farber Cancer Institute, ⁇ 2014. The present disclosure is not necessarily limited to any particular source, method of production, or other special characteristics of an antibody.
  • Host cells may be a prokaryotic or eukaryotic cell.
  • the polynucleotide or vector which is present in the host cell may either be integrated into the genome of the host cell or it may be maintained extrachromosomally.
  • the host cell can be any prokaryotic or eukaryotic cell, such as a bacterial, insect, fungal, plant, animal or human cell.
  • fungal cells are, for example, those of the genus Saccharomyces, in particular those of the species .S'. cerevisiae.
  • prokaryotic includes all bacteria which can be transformed or transfected with a DNA or RNA molecules for the expression of an antibody or the corresponding immunoglobulin chains.
  • Prokaryotic hosts may include gram negative as well as gram positive bacteria such as, for example, E. coli, S. typhimurium, Serratia marcescens and Bacillus subtilis.
  • eukaryotic includes yeast, higher plants, insects and vertebrate cells, e.g., mammalian cells, such as NSO and CHO cells.
  • the antibodies or immunoglobulin chains encoded by the polynucleotide may be glycosylated or may be nonglycosylated.
  • Antibodies or the corresponding immunoglobulin chains may also include an initial methionine amino acid residue.
  • the host may be maintained under conditions suitable for high level expression of the nucleotide sequences, and, as desired, the collection and purification of the immunoglobulin light chains, heavy chains, light/heavy chain dimers or intact antibodies, antigen binding fragments or other immunoglobulin forms may follow; see, Beychok, Cells of Immunoglobulin Synthesis, Academic Press, N.Y., (1979).
  • polynucleotides or vectors are introduced into the cells which in turn produce the antibody or antigen binding fragments.
  • transgenic animals, preferably mammals, comprising the aforementioned host cells may be used for the large scale production of the antibody or antibody fragments.
  • the transformed host cells can be grown in fermenters and cultured according to techniques known in the art to achieve optimal cell growth.
  • the whole antibodies, their dimers, individual light and heavy chains, other immunoglobulin forms, or antigen binding fragments can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like; see, Scopes, "Protein Purification", Springer Verlag, N.Y. (1982).
  • the antibody or antigen binding fragments can then be isolated from the growth medium, cellular lysates, or cellular membrane fractions.
  • the isolation and purification of the, e.g., microbially expressed antibodies or antigen binding fragments may be by any conventional means such as, for example, preparative chromatographic separations and immunological separations, such as those involving the use of monoclonal or polyclonal antibodies directed, e.g., against the constant region of the antibody.
  • hybridoma which provides an indefinitely prolonged source of monoclonal antibodies.
  • hybridoma cell refers to an immortalized cell derived from the fusion of B lymphoblasts with a myeloma fusion partner.
  • monoclonal antibody-producing cells e.g., hybridoma cells
  • an individual animal whose antibody titer has been confirmed e.g., a mouse
  • 2 days to 5 days after the final immunization its spleen or lymph node is harvested and antibody-producing cells contained therein are fused with myeloma cells to prepare the desired monoclonal antibody producer hybridoma.
  • Measurement of the antibody titer in antiserum can be carried out, for example, by reacting the labeled protein, as described hereinafter and antiserum and then measuring the activity of the labeling agent bound to the antibody.
  • the cell fusion can be carried out according to known methods, for example, the method described by Koehler and Milstein (Nature 256:495 (1975)).
  • a fusion promoter for example, polyethylene glycol (PEG) or Sendai virus (HVJ) is used.
  • myeloma cells examples include NS-1, P3U1, SP2/0, AP-1 and the like.
  • the proportion of the number of antibody producer cells (spleen cells) and the number of myeloma cells to be used is preferably about 1:1 to about 20:1.
  • PEG preferably PEG 1000-PEG 6000
  • Cell fusion can be carried out efficiently by incubating a mixture of both cells at about 20°C to about 40 °C, preferably about 30 °C to about 37 °C for about 1 minute to 10 minutes.
  • a hybridoma producing the antibody e.g., against a tumor antigen or autoantibody of the present invention
  • a supernatant of the hybridoma is added to a solid phase (e.g., microplate) to which antibody is adsorbed directly or together with a carrier and then an anti-immunoglobulin antibody (if mouse cells are used in cell fusion, anti-mouse immunoglobulin antibody is used) or Protein A labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase.
  • a solid phase e.g., microplate
  • an anti-immunoglobulin antibody if mouse cells are used in cell fusion, anti-mouse immunoglobulin antibody is used
  • Protein A labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase.
  • a supernatant of the hybridoma is added to a solid phase to which an anti-immunoglobulin antibody or Protein A is adsorbed and then the protein labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase.
  • Selection of the monoclonal antibody can be carried out according to any known method or its modification. Normally, a medium for animal cells to which HAT (hypoxanthine, aminopterin, thymidine) are added is employed. Any selection and growth medium can be employed as long as the hybridoma can grow. For example, RPMI 1640 medium containing 1% to 20%, preferably 10% to 20% fetal bovine serum, GIT medium containing 1% to 10% fetal bovine serum, a serum free medium for cultivation of a hybridoma (SFM-101, Nissui Seiyaku) and the like can be used.
  • HAT hyperxanthine, aminopterin, thymidine
  • the cultivation is carried out at 20°C to 40°C, preferably 37°C for about 5 days to 3 weeks, preferably 1 week to 2 weeks under about 5% CO2 gas.
  • the antibody titer of the supernatant of a hybridoma culture can be measured according to the same manner as described above with respect to the antibody titer of the anti-protein in the antiserum.
  • immortalized hybridoma cells can be used as a source of rearranged heavy chain and light chain loci for subsequent expression and/or genetic manipulation.
  • Rearranged antibody genes can be reverse transcribed from appropriate mRNAs to produce cDNA.
  • the heavy chain constant region can be exchanged for that of a different isotype or eliminated altogether.
  • the variable regions can be linked to encode single chain Fv regions. Multiple Fv regions can be linked to confer binding ability to more than one target or chimeric heavy and light chain combinations can be employed. Any appropriate method may be used for cloning of antibody variable regions and generation of recombinant antibodies.
  • an appropriate nucleic acid that encodes variable regions of a heavy and/or light chain is obtained and inserted into an expression vectors which can be transfected into standard recombinant host cells.
  • a variety of such host cells may be used.
  • mammalian host cells may be advantageous for efficient processing and production. Typical mammalian cell lines useful for this purpose include CHO cells, 293 cells, or NSO cells.
  • the production of the antibody or antigen binding fragment may be undertaken by culturing a modified recombinant host under culture conditions appropriate for the growth of the host cells and the expression of the coding sequences.
  • the antibodies or antigen binding fragments may be recovered by isolating them from the culture.
  • the expression systems may be designed to include signal peptides so that the resulting antibodies are secreted into the medium; however, intracellular production is also possible.
  • the disclosure also includes a polynucleotide encoding at least a variable region of an immunoglobulin chain of the antibodies described herein.
  • the variable region encoded by the polynucleotide comprises at least one complementarity determining region (CDR) of the VH and/or VL of the variable region of the antibody produced by any one of the above described hybridomas.
  • CDR complementarity determining region
  • Polynucleotides encoding antibody or antigen binding fragments may be, e.g., DNA, cDNA, RNA or synthetically produced DNA or RNA or a recombinantly produced chimeric nucleic acid molecule comprising any of those polynucleotides either alone or in combination.
  • a polynucleotide is part of a vector.
  • Such vectors may comprise further genes, such as marker genes which allow for the selection of the vector in a suitable host cell and under suitable conditions.
  • a polynucleotide is operatively linked to expression control sequences allowing expression in prokaryotic or eukaryotic cells.
  • Expression of the polynucleotide comprises transcription of the polynucleotide into a translatable mRNA.
  • Regulatory elements ensuring expression in eukaryotic cells are well known to those skilled in the art. They may include regulatory sequences that facilitate initiation of transcription and optionally poly-A signals that facilitate termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers, and/or naturally associated or heterologous promoter regions.
  • Possible regulatory elements permitting expression in prokaryotic host cells include, e.g., the PL, Lac, Trp or Tac promoter in E. coli, and examples of regulatory elements permitting expression in eukaryotic host cells are the A0X1 or GALI promoter in yeast or the CMV-promoter, SV40-promoter, RSV-promoter (Rous sarcoma virus), CMV-enhancer, SV40- enhancer or a globin intron in mammalian and other animal cells.
  • Such regulatory elements may also include transcription termination signals, such as the SV40-poly-A site or the tk-poly-A site, downstream of the polynucleotide.
  • transcription termination signals such as the SV40-poly-A site or the tk-poly-A site, downstream of the polynucleotide.
  • leader sequences capable of directing the polypeptide to a cellular compartment or secreting it into the medium may be added to the coding sequence of the polynucleotide and are well known in the art.
  • the leader sequence(s) is (are) assembled in appropriate phase with translation, initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein, or a portion thereof, into, for example, the extracellular medium.
  • a heterologous polynucleotide sequence can be used that encode a fusion protein including a C- or N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
  • polynucleotides encoding at least the variable domain of the light and/or heavy chain may encode the variable domains of both immunoglobulin chains or only one.
  • polynucleotides may be under the control of the same promoter or may be separately controlled for expression.
  • vectors, particularly plasmids, cosmids, viruses and bacteriophages used conventionally in genetic engineering that comprise a polynucleotide encoding a variable domain of an immunoglobulin chain of an antibody or antigen binding fragment; optionally in combination with a polynucleotide that encodes the variable domain of the other immunoglobulin chain of the antibody.
  • expression control sequences are provided as eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells, but control sequences for prokaryotic hosts may also be used.
  • Expression vectors derived from viruses such as retroviruses, vaccinia virus, adeno-associated virus, herpes viruses, or bovine papilloma virus, may be used for delivery of the polynucleotides or vector into targeted cell population (e.g., to engineer a cell to express an antibody or antigen binding fragment).
  • viruses such as retroviruses, vaccinia virus, adeno-associated virus, herpes viruses, or bovine papilloma virus
  • a variety of appropriate methods can be used to construct recombinant viral vectors.
  • polynucleotides and vectors can be reconstituted into liposomes for delivery to target cells.
  • the vectors containing the polynucleotides can be transferred into the host cell by suitable methods, which vary depending on the type of cellular host.
  • a vector (e.g., a plasmid) comprises at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% identity to a sequence set forth in any one of SEQ ID NOs: 42-46. In some embodiments, a vector (e.g., a plasmid) comprises a sequence set forth in any one of SEQ ID NOs: 42-46.
  • antibody drug conjugate refers to molecules comprising an antibody, or antigen binding fragment thereof, linked to a targeted molecule (e.g., a biologically active molecule, such as a therapeutic molecule, and/or a detectable label).
  • a targeted molecule e.g., a biologically active molecule, such as a therapeutic molecule, and/or a detectable label.
  • antibodies or antigen binding fragments of the disclosure may be modified with a detectable label, including, but not limited to, an enzyme, prosthetic group, fluorescent material, luminescent material, bioluminescent material, radioactive material, positron emitting metal, nonradioactive paramagnetic metal ion, and affinity label for detection and isolation of one or more RAN proteins.
  • the detectable substance may be coupled or conjugated either directly to the polypeptides of the disclosure or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase,
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include biotin, umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride, or phycoerythrin; an example of a luminescent material includes luminol; non-limiting examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of
  • the detectable substance may be coupled or conjugated either directly to the anti-RAN antibodies or antigen-binding fragments of the disclosure or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art.
  • Anti-RAN antibodies conjugated to a detectable substance may be used for diagnostic assays as described herein.
  • antibodies or antigen binding fragments of the disclosure may be modified with a therapeutic moiety e.g., therapeutic agent).
  • the antibody is coupled to the targeted agent via a linker.
  • linker refers to a molecule or sequence, such as an amino acid sequence, that attaches, as in a bridge, one molecule or sequence to another molecule or sequence.
  • "Linked,” “conjugated,” or “coupled” means attached or bound by covalent bonds, or non-covalent bonds, or other bonds, such as van der Waals forces.
  • Antibodies described by the disclosure can be linked to the targeted agent (e.g., therapeutic moiety or detectable moiety) directly, e.g., as a fusion protein with protein or peptide detectable moieties (with or without an optional linking sequence, e.g., a flexible linker sequence) or via a chemical coupling moiety.
  • a number of such coupling moieties are known in the art, e.g., a peptide linker or a chemical linker, e.g., as described in International Patent Application Publication No. WO 2009/036092.
  • the linker is a flexible amino acid sequence.
  • Examples of flexible amino acid sequences include glycine and serine rich linkers, which comprise a stretch of two or more glycine residues.
  • the linker is a photolinker.
  • Examples of photolinkers include ketyl-reactive benzophenone (BP), anthraquinone (AQ), nitrene-reactive nitrophenyl azide (NPA), and carbene-reactive phenyl- (trifluoromethyl)diazirine (PTD).
  • the disclosure relates to pharmaceutical compositions comprising anti- RAN antibodies or antigen binding fragments.
  • the composition comprises an anti-RAN antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • Pharmaceutical compositions can be prepared as described below. The active ingredients may be admixed or compounded with any conventional, pharmaceutically acceptable carrier or excipient. The compositions may be sterile.
  • compositions are formulated for delivering an effective amount of an agent (e.g., an anti-RAN antibody).
  • an “effective amount” of an active agent refers to an amount sufficient to elicit the desired biological response (e.g., ameliorating one or more symptoms of ALS).
  • An effective amount of an agent may vary depending on such factors as the desired biological endpoint, the pharmacokinetics of the compound, the disease being treated (e.g., ALS, repeat expansion diseases), the mode of administration, and the patient.
  • a composition is said to be a “pharmaceutically acceptable carrier” if its administration can be tolerated by a recipient patient.
  • Sterile phosphate-buffered saline is one example of a pharmaceutically acceptable carrier.
  • suitable carriers are well-known in the art. See, for example, REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Ed. (1990). It will be understood by those skilled in the art that any mode of administration, vehicle or carrier conventionally employed and which is inert with respect to the active agent may be utilized for preparing and administering the pharmaceutical compositions of the present disclosure. Illustrative of such methods, vehicles and carriers are those described, for example, in Remington's Pharmaceutical Sciences, 4th ed.
  • an effective amount, also referred to as a therapeutically effective amount, of a compound is an amount sufficient to ameliorate at least one adverse effect associated with a disease associated with RAN proteins, such as, e.g., memory loss, cognitive impairment, loss of coordination, speech impairment, etc.
  • the neurological disease associated with RAN proteins is selected from the group consisting of: amyotrophic lateral sclerosis (ALS), or frontotemporal dementia; myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2); spinocerebellar ataxia types 1, 2, 3, 6, 7, 8, 10, 12, 17, 31, and 36; spinal bulbar muscular atrophy; dentatorubral-pallidoluysian atrophy (DRPLA); Huntington's disease (HD); Fragile X Tremor Ataxia Syndrome (FXTAS); Fuch's endothelial corneal dystrophy (FECD); Huntington's disease-like 2 syndrome (HDL2); Fragile X syndrome (FXS); disorders related to 7pl 1 .2 folate- sensitive fragile site FRA7A; disorders related to folate- sensitive fragile site 2ql 1 FRA2A; and Fragile XE syndrome (FRAXE).
  • ALS amyotrophic lateral sclerosis
  • DM1
  • the neurological disease associated with RAN proteins is ALS.
  • the therapeutically effective amount to be included in pharmaceutical compositions depends, in each case, upon several factors, e.g., the type, size and condition of the patient to be treated, the intended mode of administration, the capacity of the patient to incorporate the intended dosage form, etc. Generally, an amount of active agent is included in each dosage form to provide from about 0.1 to about 250 mg/kg, and preferably from about 0.1 to about 100 mg/kg. One of ordinary skill in the art would be able to determine empirically an appropriate therapeutically effective amount.
  • an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is entirely effective to treat the particular subject.
  • the effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular therapeutic agent being administered, the size of the subject, or the severity of the disease or condition.
  • One of ordinary skill in the art can empirically determine the effective amount of a particular nucleic acid and/or other therapeutic agent without necessitating undue experimentation .
  • pharmaceutically-acceptable carrier means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other vertebrate animal.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • the components of the pharmaceutical compositions also are capable of being commingled with the compounds of the present disclosure, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency.
  • compositions also may comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • Suitable liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin.
  • the pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above.
  • the pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of methods for drug delivery, see, Langer R (1990) Science 249:1527-1533, which is incorporated herein by reference.
  • the compounds may be administered per se (neat) or in the form of a pharmaceutically acceptable salt.
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof.
  • Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic.
  • such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the compounds into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the compounds into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • Liquid dose units are vials or ampoules. Solid dose units are tablets, capsules and suppositories.
  • a therapeutic agent may be delivered by any suitable modality known in the art.
  • a therapeutically effective amount is an amount effective in reducing repeat expansions in the subject.
  • a therapeutically effective amount is an amount effective in reducing the transcription of RNAs that produce RAN proteins in a subject.
  • a therapeutically effective amount is an amount effective in reducing the translation of RAN proteins in a subject.
  • a therapeutically effective amount is an amount effective for treating ALS associated with repeat expansions. “Reducing” expression of a repeat sequence or RAN protein translation refers to a decrease in the amount or level of repeat sequence expression or RAN protein translation in a subject after administration of a therapeutic agent (and relative to the amount or level in the subject prior to the administration).
  • the effective amount is an amount effective in reducing the level of RAN proteins by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 98% (e.g., the level of RAN proteins relative to the level of RAN proteins in a cell or subject that has not been administered a therapeutic agent).
  • the effective amount is an amount effective in reducing the translation of RAN proteins by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 98% (e.g., the level of RAN proteins relative the level of RAN proteins in a cell or subject that has not been administered a therapeutic agent).
  • compositions described herein can be prepared by any method known in the art of pharmacology.
  • preparatory methods include bringing the compound described herein (z.e., the “active ingredient”) into association with a carrier or excipient, and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping, and/or packaging the product into a desired single- or multi-dose unit.
  • compositions can be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a “unit dose” is a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage, such as one- half or one-third of such a dosage.
  • Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition described herein will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100% (w/w) active ingredient.
  • Therapeutic agents described herein are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions described herein will be decided by a physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the disease being treated and the severity of the disorder; the activity of the specific active ingredient employed; the specific composition employed; the age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific active ingredient employed; the duration of the treatment; drugs used in combination or coincidental with the specific active ingredient employed; and like factors well known in the medical arts.
  • a therapeutic agent can be administered by any route, including enteral (e.g., oral), parenteral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal, buccal, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol.
  • enteral e.g., oral
  • parenteral intravenous, intramuscular, intra-arterial, intramedullary
  • intrathecal subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal
  • topical as by powders, ointments, creams, and/or drops
  • mucosal nasal, buccal, sublingual
  • Specifically contemplated routes are oral administration, intravenous administration (e.g., systemic intravenous injection), regional administration via blood and/or lymph supply, and/or direct administration to an affected site.
  • intravenous administration e.g., systemic intravenous injection
  • regional administration via blood and/or lymph supply e.g., via blood and/or lymph supply
  • direct administration to an affected site.
  • the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), and/or the condition of the subject (e.g., whether the subject is able to tolerate oral administration).
  • the exact amount of a therapeutic agent required to achieve an effective amount will vary from subject to subject, depending, for example, on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular compound, mode of administration, and the like.
  • An effective amount may be included in a single dose (e.g., single oral dose) or multiple doses (e.g., multiple oral doses).
  • a single dose e.g., single oral dose
  • multiple doses e.g., multiple oral doses
  • any two doses of the multiple doses include different or substantially the same amounts of a compound described herein.
  • the frequency of administering the multiple doses to the subject or applying the multiple doses to the biological sample, tissue, or cell is three doses a day, two doses a day, one dose a day, one dose every other day, one dose every third day, one dose every week, one dose every two weeks, one dose every three weeks, or one dose every four weeks.
  • the frequency of administering the multiple doses to the subject or applying the multiple doses to the biological sample, tissue, or cell is one dose per day.
  • the frequency of administering the multiple doses to the subject or applying the multiple doses to the biological sample, tissue, or cell is two doses per day.
  • the frequency of administering the multiple doses to the subject or applying the multiple doses to the biological sample, tissue, or cell is three doses per day.
  • the duration between the first dose and last dose of the multiple doses is one day, two days, four days, one week, two weeks, three weeks, one month, two months, three months, four months, six months, eight months, nine months, one year, two years, three years, four years, five years, seven years, ten years, fifteen years, twenty years, or the lifetime of the subject, tissue, or cell.
  • the duration between the first dose and last dose of the multiple doses is three months, six months, or one year.
  • the duration between the first dose and last dose of the multiple doses is the lifetime of the subject, tissue, or cell.
  • a treatment for a disease associated with RAN protein expression is administered to the central nervous system (CNS) of a subject in need thereof.
  • the “central nervous system (CNS)” refers to all cells and tissues of the brain and spinal cord of a subject, including but not limited to neuronal cells, glial cells, astrocytes, cerebrospinal fluid, etc.
  • Modalities of administering a therapeutic agent to the CNS of a subject include direct injection into the brain (e.g., intracerebral injection, intraventricular injection, intraparenchymal injection, etc.), direct injection into the spinal cord of a subject (e.g., intrathecal injection, lumbar injection, etc.), or any combination thereof.
  • a treatment as described by the disclosure is systemically administered to a subject, for example by intravenous injection.
  • Systemically administered therapeutic molecules can be modified, in some embodiments, in order to improve delivery of the molecules to the CNS of a subject.
  • modifications that improve CNS delivery of therapeutic molecules include but are not limited to co-administration or conjugation to blood brain barrier-targeting agents (e.g., transferrin, melanotransferrin, low-density lipoprotein (LDL), angiopeps, RVG peptide, etc., as disclosed by Georgieva et al. Pharmaceuticals 6(4): 557-583 (2014)), coadministration with BBB disrupting agents (e.g., bradykinins), and physical disruption of the BBB prior to administration (e.g., by MRI-Guided Focused Ultrasound), etc.
  • blood brain barrier-targeting agents e.g., transferrin, melanotransferrin, low-density lipoprotein (LDL), angiopeps, RV
  • a method described herein comprises utilizing an antibody or antigen binding fragment described herein to detect a RAN protein (e.g., a poly(GA) RAN protein).
  • a RAN protein e.g., a poly(GA) RAN protein
  • the RAN protein is comprised or suspected of being comprised in a biological sample obtained from a subject (e.g., a subject characterized as having or suspected of having a disease, such as a neurological disease, associated with RAN protein expression (e.g., poly(GA) RAN protein expression), translation, and/or accumulation).
  • Nonlimiting examples of biological samples include those comprising one or more cells (e.g., brain cells, neuronal cells, skin cells, white blood cells, etc.), and tissue samples, such as blood (e.g., whole blood), serum (e.g., plasma from which the clotting proteins have been removed), cerebrospinal fluid (CSF), and nervous system tissue (e.g., central nervous system (CNS) tissue, such as spinal cord tissue and brain tissues including, but not limited to, hippocampal tissue and cortex tissue) samples.
  • cells e.g., brain cells, neuronal cells, skin cells, white blood cells, etc.
  • tissue samples such as blood (e.g., whole blood), serum (e.g., plasma from which the clotting proteins have been removed), cerebrospinal fluid (CSF), and nervous system tissue (e.g., central nervous system (CNS) tissue, such as spinal cord tissue and brain tissues including, but not limited to, hippocampal tissue and cortex tissue) samples.
  • CNS central nervous system
  • a method comprises contacting one or more antibodies or antigenbinding fragments (e.g., 1, 2, 3, or more than 3 antibodies or antigen-binding fragments) described herein with a biological sample.
  • a method comprises detecting a complex formed between one or more antibodies, or antigen binding fragments, and a RAN protein in a biological sample, such as by measuring a detectable signal in the biological sample that represents the presence of the complex or the amount of the complex and determining the presence and/or amount of the RAN protein in the biological sample.
  • a method comprises contacting an antibody or antigen-binding fragment described herein with a first biological sample and a second biological sample.
  • one of the biological samples is a control sample.
  • the first biological sample is obtained from a subject prior to administration of an agent (e.g., an agent used in the treatment of a disease or disorder described herein) to the subject and the second biological sample is obtained from the subject after administration of the agent.
  • the first biological sample is obtained from a subject after the subject has been administered an agent (e.g., an agent used in the treatment of a disease or disorder described herein) and the second biological sample is obtained from the subject after the subject is no longer being administered the agent.
  • the second biological sample can be collected hours (e.g., 1- 2, 2-6, 6-12, or 12-24 hours), days (e.g., 1, 2, 3, 4, 5, 6, or 7 days), weeks (e.g., 1-2, 2-3, or 3-4 weeks), or months (e.g., 1-2, 2-3, 3-4, 4-5, 5-6, 6-9, 9-12 months, or more than 12 months) after the first biological sample was obtained.
  • hours e.g., 1- 2, 2-6, 6-12, or 12-24 hours
  • days e.g., 1, 2, 3, 4, 5, 6, or 7 days
  • weeks e.g., 1-2, 2-3, or 3-4 weeks
  • months e.g., 1-2, 2-3, 3-4, 4-5, 5-6, 6-9, 9-12 months, or more than 12 months
  • a method described herein comprises detecting a RAN protein (e.g., a poly(GA) RAN protein) in a biological sample obtained from a subject and/or detecting a difference (e.g., an increase or a decrease) in the levels of a RAN protein (e.g., a poly(GA) RAN protein) between two biological samples obtained from a subject, and then administering an agent (e.g., an agent used in the treatment of a disease or disorder described herein) to a subject.
  • a RAN protein e.g., a poly(GA) RAN protein
  • an agent e.g., an agent used in the treatment of a disease or disorder described herein
  • a method for detecting a RAN protein as described herein comprises an immunoassay.
  • immunoassays include immunoblot (e.g., dot blot, 2-D gel electrophoresis, Western Blot, etc.), electrochemiluminescence immunoassay (e.g., Meso-Scale Detection (MSD)), immunohistochemistry (IHC), ELISA (e.g., RCA-based ELISA or RT-PCR-based ELISA), label free immunoassays such as surface plasmon resonance bio layer interferometry, immunoquantitative PCR, bead-based immunoassays, immunoprecipitation, immunostaining, and immunoelectrophoresis.
  • immunoblot e.g., dot blot, 2-D gel electrophoresis, Western Blot, etc.
  • electrochemiluminescence immunoassay e.g., Meso-Scale Detection (MSD)
  • aspects of the RAN protein detection methods described herein may also include a method previously described in International Application No. PCT/US2014/022670, International Application No. PCT/US2018/052745, International Application No. PCT/US2021/032488, International Application No. PCT/US2020/051671, International Application No. PCT/US2020/054976, or International Application No. PCT/US2022/079048, each which is incorporated by reference herein for their disclosures related to RAN protein detection methods.
  • FIG. 1 shows representative data for immunohistochemistry (IHC) assays for anti- poly(GA) monoclonal antibodies.
  • IHC immunohistochemistry
  • m-GA mouse anti-GA polyclonal antibody
  • EV vehicle- only negative control
  • ScFv 515 27B11 ScFv format
  • IgGl 515A 27B11 monoclonal antibody in IgGl format
  • IgGl 515B 27B11 monoclonal antibody in IgGl format
  • IgG2 515 27B 11 monoclonal antibody in IgG2 format.
  • FIG. 2 shows representative data indicating expression of anti-poly(GA) monoclonal antibody 27B11 in several formats (IgGl, IgG2, ScFv) lowers levels of (GA)eo-GFP RAN protein in cells.
  • an antibody or antigen-binding fragment described herein comprises one or more of the amino acid sequences set forth below (see, e.g., any one of the amino acid sequences set forth in SEQ ID NOs: 1-35 and 41).
  • a nucleic acid described herein e.g., a vector, such as a plasmid
  • comprises one or more nucleotide sequences set forth below see, e.g. any one of the nucleotide sequences set forth in SEQ ID NOs: 36-40 and 42-46 and/or encodes one or more of the amino acid sequences set forth below (see, e.g., any one of the amino acid sequences set forth in SEQ ID NOs: 1-35 and 41).
  • one or more of the representative sequences set forth below may be utilized according to the foregoing embodiments of compositions and methods as described herein.
  • Anti-polyGA antibody clone 27B 11 heavy chain variable region amino acid sequence SEQ ID NO: 7
  • Anti-polyGA antibody clone 27B 11 scFv complete sequence with signal peptide and poly-GS linker (SEQ ID NO: 19)
  • Anti-polyGA antibody clone 23H2 heavy chain IgG2 amino acid sequence (SEQ ID NO: 29) EVQLQESGGGSVQPGGALQLSCAASGFTFSSHGMSWVRQTPDKRLEMVATINSNGGST YYPDSVKGRFIISRDNAKNTLYLQMSSLKSEDTAMYYCARVGDNDDFAMGYWGQGTS VTVSSAKTTAPSVYPLAPVCGDTTGSSVTLGCLVKGYFPEPVTLTWNSGSLSSGVHTFP AVLQSDLYTLSSSVTVTSSTWPSQSITCNVAHPASSTKVDKKIEPRGPTIKPCPPCKCPAP
  • Table 2 Representative sequences of vectors encoding anti-polyGA antibodies.
  • inventive embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed.
  • inventive embodiments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein.
  • a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
  • the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
  • This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified.
  • “at least one of A and B” can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Aspects of the disclosure relate to compositions and methods for the diagnosis and/or treatment of certain neurodegenerative diseases, for example those diseases associated with repeat-associated non-ATG (RAN) translation proteins, such as Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). In some embodiments, the disclosure relates to antibodies and antigen-binding fragments thereof that bind to poly (GA) RAN proteins. In some embodiments, the disclosure relates to methods of treating a RAN protein-associated disease by administering to a subject in need thereof the antibodies or antigen-binding fragments thereof.

Description

ENGINEERED GA ANTIBODIES FOR THERAPY DEVELOPMENT
RELATED APPLICATIONS
The application claims the benefit under 35 U.S.C. 119(e) of U.S. Provisional Application number 63/514,643 filed July 20, 2023, which is incorporated by reference in its entirety.
BACKGROUND
Microsatellite repeat expansions are known to cause more than forty neurodegenerative disorders. Molecular features common to many of these disorders include the accumulation of RNA foci containing sense and antisense expansion transcripts and the accumulation of proteins from repeat-associated non- AUG (RAN) translation. RAN translation can occur across a broad range of repeat lengths from pre-mutation lengths (~30 - 40 repeats) to full expansions (up to 10,000 repeats). There is growing evidence that RAN proteins are toxic and contribute to a growing number of diseases and disorders, including, but not limited to, amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), Huntington’s disease (HD), Alzheimer’s disease (AD), and Fragile X Tremor Ataxia Syndrome (FXTAS).
SUMMARY
Aspects of the disclosure relate to compositions and methods for the diagnosis and/or treatment of certain neurodegenerative diseases, for example those diseases associated with repeat-associated non-ATG (RAN) translation proteins, such as Alzheimer’s disease (AD) and amyotrophic lateral sclerosis (ALS). The disclosure is based, in part, on antibodies (e.g., monoclonal antibodies) and antigen-binding fragments thereof (e.g., single-chain variable fragments, scFvs) that bind to poly-Glycine- Alanine (poly(GA)) RAN proteins. In some embodiments, the disclosure relates to methods of reducing RAN protein (e.g., poly(GA) RAN protein) aggregation in a subject by administering the antibodies or antigen-binding fragments thereof to the subject. In some embodiments, the disclosure relates to methods of treating a RAN protein-associated disease by administering to a subject in need thereof the antibodies or antigen-binding fragments thereof.
Accordingly, in some aspects, the disclosure provides an antibody or antigen-binding fragment thereof, wherein the antibody or antigen-binding fragment comprises a heavy chain variable region (VH) comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 1; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 2; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 3.
In some aspects, the disclosure provides an antibody or antigen-binding fragment thereof, wherein the antibody or antigen-binding fragment comprises a light chain variable region (VL) comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 4; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 5; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 6.
In some embodiments, an antibody or antigen-binding fragment comprises a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 7.
In some embodiments, an antibody or antigen-binding fragment comprises a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 12.
In some embodiments, an antibody of antigen-binding fragment comprises a heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 8 or 10.
In some embodiments, an antibody of antigen-binding fragment comprises a light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 13 or 15.
In some embodiments, an antibody or antigen-binding fragment thereof comprises a heavy chain variable region (VH) comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 20; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 21; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 22.
In some aspects, the disclosure provides an antibody or antigen-binding fragment thereof that specifically binds to a RAN protein, wherein the antibody or antigen-binding fragment comprises a light chain variable region (VL) comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 23; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 24; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 25.
In some embodiments, an antibody or antigen-binding fragment comprises a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 26.
In some embodiments, an antibody or antigen-binding fragment comprises a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 31.
In some embodiments, an antibody of antigen-binding fragment comprises a heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 27 or 29.
In some embodiments, an antibody of antigen-binding fragment comprises a light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 32 or 34. In some aspects, the disclosure provides a single chain variable fragment (scFv) comprising a heavy chain variable region comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 1; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 2; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 3.
In some aspects, the disclosure provides a single chain variable fragment (scFv) comprising a light chain variable region comprising a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 4; a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 5; and/or a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 6.
In some embodiments, an scFv comprises a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 17.
In some embodiments, an scFv comprises a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 18.
In some embodiments, an scFv comprises a linker molecule connecting the heavy chain variable region to the light chain variable region. In some embodiments, a linker molecule comprises a poly-GS linker.
In some embodiments, an antibody or antigen-binding fragment thereof or scFv further comprises a signal peptide.
In some embodiments, an antibody or antigen-binding fragment thereof or scFv binds poly(GA) RAN protein.
In some aspects, the disclosure provides a composition comprising an antibody or antigen-binding fragment thereof, or scFv, as described herein and a pharmaceutically acceptable carrier or buffer.
In some aspects, the disclosure provides an isolated nucleic acid encoding an antibody or antigen-binding fragment thereof, or scFv, as described herein. In some embodiments, an isolated nucleic acid comprises the nucleotide sequence set forth in any one of SEQ ID NOs: 36- 40.
In some aspects, the disclosure provides a vector comprising an isolated nucleic acid as described herein. In some embodiments, the vector is a plasmid. In some embodiments, a plasmid comprises a sequence as set forth in any one of SEQ ID NOs: 42-46.
In some aspects, the disclosure provides a cell transformed with an isolated nucleic acid or vector as described herein. In some embodiments, the cell is a mammalian cell. In some embodiments, the cell is a human cell.
In some aspects, the disclosure provides a method for administering an antibody or antigen-binding fragment to a subject, the method comprising administering the antibody or antigen-binding fragment or scFv as described herein, to the subject.
In some aspects, the disclosure provides a method for reducing poly(GA) RAN protein aggregation in a subject, the method comprising administering an antibody or antigen-binding fragment, or scFv, as described herein to the subject.
In some aspects, the disclosure provides a method for treating a subject having ALS, the method comprising administering an antibody or antigen-binding fragment, or an scFv, as described herein to the subject.
In some embodiments, a subject is a mammal. In some embodiments, a subject is a human. In some embodiments, a subject expresses one or more RAN proteins. In some embodiments, a subject expresses a poly(GA) RAN protein.
In some embodiments, a subject has or is suspected of having Alzheimer’s disease, amyotrophic lateral sclerosis (ALS), frontotemporal dementia, myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), spinocerebellar ataxia (SCA) type 1 (SCA), SCA2, SCA3, SCA6, SCA7, SCA8, SCAIO, SCA12, SCA17, SCA31, SCA36, spinal bulbar muscular atrophy, dentatorubral-pallidoluysian atrophy (DRPLA), Huntington's disease (HD), Fragile X Tremor Ataxia Syndrome (FXTAS), Fuch's endothelial corneal dystrophy (FECD), Huntington's disease-like 2 syndrome (HDL2), Fragile X syndrome (FXS), a disorder related to 7pl 1 .2 folatesensitive fragile site FRA7A, a disorder related to folate-sensitive fragile site 2ql 1 FRA2A, or Fragile XE syndrome (FRAXE).
In some embodiments, a subject has or is suspected of having ALS.
BRIEF DESCRIPTION OF DRAWINGS
FIG. 1 shows representative data for immunohistochemistry (IHC) assays for anti- poly(GA) monoclonal antibodies. Supernatants from cells expressing anti-poly(GA) monoclonal antibody 27B11 in several formats (IgGl, IgG2, ScFv) recognize poly-GA RAN protein aggregates in C9Bac mouse cells. m-GA= mouse anti-GA polyclonal antibody; EV= vehicle- only negative control; ScFv 515 = 27B11 ScFv format; IgGl 515A = 27B11 monoclonal antibody in IgGl format; IgGl 515B = 27B11 monoclonal antibody in IgGl format; IgG2 515 = 27B 11 monoclonal antibody in IgG2 format.
FIG. 2 shows representative data indicating expression of anti-poly(GA) monoclonal antibody 27B11 in several formats (IgGl, IgG2, ScFv) lowers levels of (GA)eo-GFP RAN protein in cells. DETAILED DESCRIPTION
Aspects of the disclosure relate to compositions and methods for the diagnosis and/or treatment of certain neurodegenerative diseases, for example those diseases associated with repeat-associated non-ATG (RAN) translation proteins, such as Alzheimer’s disease (AD) or ALS. The disclosure is based, in part, on antibodies (e.g., monoclonal antibodies) and antigenbinding fragments thereof (e.g., single-chain variable fragments, scFvs) that bind to poly- Glycine-Alanine (poly(GA)) RAN proteins. In some embodiments, the disclosure relates to methods of reducing RAN protein (e.g., poly(GA) RAN protein) aggregation in a subject by administering the antibodies or antigen-binding fragments thereof to the subject. In some embodiments, the disclosure relates to methods of treating a RAN protein-associated disease by administering to a subject in need thereof the antibodies or antigen-binding fragments thereof.
RAN Proteins
A “RAN protein (repeat- associated non-ATG translated protein)” is a polypeptide that is translated from sense or antisense RNA sequences bidirectionally transcribed from a repeat expansion mutation in the absence of an AUG initiation codon. RAN protein-encoding sequences can be found in the genome at multiple loci, including but not limited to open reading frame 72 of chromosome 9 (C9orf72), open reading frame 80 of chromosome 2 (C2orf80), LRP8, CASP8, CRNDE, EXOC6B, SV2B, PPML1, ADARB2, GREB1, and MSM01. The protein associated with C9orf72 is currently poorly characterized but known to be abundant in neurons, especially in the cerebral cortex and motor neurons. C9orf72 protein is believed to localized in presynaptic termini. C9orf72 protein likely impacts transcription, translation and intra-cellular localization of RNA. C9orf72 gene contains a GGGGCC repeat. This hexanucleotide repeat occurs in variable repeat numbers, and small numbers of repeats are not associated with any pathology.
Generally, RAN proteins comprise expansion repeats of a single amino acid, di-amino acid, tri-amino acid, or quad-amino acid e.g., tetra-amino acid), termed poly amino acid repeats. For example, “AAAAAAAAAAAAAAAAAAAA” (SEQ ID NO: 47) (poly-Alanine), “LLLLLLLLLLLLLLLLLL” (SEQ ID NO: 48) (poly-Leucine), “SSSSSSSSSSSSSSSSSSSS” (SEQ ID NO: 49) (poly-Serine), or “CCCCCCCCCCCCCCCCCCCC” (SEQ ID NO: 50) (poly- Cysteine) are poly amino acid repeats that are each 20 amino acid residues in length. Examples of di-amino acid RAN proteins include GPGPGPGPGPGPGPGPGPGP (SEQ ID NO: 51) (poly- GP), GAGAGAGAGAGAGAGAGAGA (SEQ ID NO: 52) (poly-GA), GRGRGRGRGRGRGRGRGRGR (SEQ ID NO: 53) (poly-GR), PAPAPAPAPAPAPAPAPAPA (SEQ ID NO: 54) (poly-PA), and PRPRPRPRPRPRPRPRPRPR (SEQ ID NO: 55) (poly-PR). Examples of tetra-amino acid repeats include LPACLPACLPAC (SEQ ID NO: 56) (e.g., poly-LPAC) and QAGRQAGRQAGR (SEQ ID NO: 57) (e.g., poly-QAGR). RAN proteins can have a poly amino acid repeat of at least 20, at least 25, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, or at least 200 amino acid residues in length. In some embodiments, a RAN protein has a poly amino acid repeat more than 200 amino acid residues (e.g., 500, 1000, 5000, 10,000, etc.) in length.
Generally, RAN proteins are translated from abnormal repeat expansions (e.g., TCT repeats, hexanucleotide repeats such as GGGGCC, etc.) of DNA. The disclosure is based, in part, on the identification of micro satellite repeats in certain subjects having a RAN protein- associated disease that is characterized by expression of one or more (e.g., 2, 3, 4, 5, or more) RAN proteins, for example poly(Glycine- Alanine) [poly(GA)]. In some embodiments, the disease status of a subject having or suspected of having a RAN protein-associated disease is classified by the number and/or type of micro satellite repeats present (e.g., detected) in the subject (e.g., in the genome of a subject or in a gene of the subject).
A “subject having or suspected of having a disease (e.g., neurological diseases) associated with RAN protein expression, translation, and/or accumulation” generally refers to a subject exhibiting one or more signs and symptoms of a neurodegenerative disease, including but not limited to memory deficit (e.g., short term memory loss), confusion, deficiencies of executive functions (e.g., attention, planning, flexibility, abstract thinking, etc.), loss of speech, degeneration or loss of motor skills, etc., or a subject having or being identified as having one or more genetic mutations associated with RAN protein expression, translation, and/or accumulation.
A “subject having or suspected of having amyotrophic lateral sclerosis (ALS)” can be a subject exhibiting one or more signs and symptoms of AD, including but not limited to memory deficit (e.g., short term memory loss), confusion, deficiencies of executive functions (e.g., attention, planning, flexibility, abstract thinking, etc.), loss of speech, degeneration or loss of motor skills, etc., or a subject having or being identified as having one or more genetic mutations associated with ALS, for example mutations in specific genes including C9orf72. In some embodiments, a subject has been diagnosed as having ALS by a medical professional. A subject can be a mammal (e.g., human, mouse, rat, dog, cat, or pig). In some embodiments, a subject is a non-human animal, for example a mouse, rat, guinea pig, cat dog, horse, camel, etc. In some embodiments, the subject is a human.
In some embodiments, a subject having less than 10 repeat sequences does not exhibit signs or symptoms of a RAN protein-associated disease characterized by RAN protein translation. In some embodiments, a subject having between 10 and 40 repeats may or may not exhibit one or more signs or symptoms of a RAN protein-associated disease characterized by RAN protein translation. In some embodiments, a subject having more than 40 trinucleotide repeats exhibits one or more signs or symptoms of a RAN protein- associated disease characterized by RAN protein translation. In certain cases, a subject is identified as having a RAN protein-associated disease characterized by large (>100) number of repeats. Micro satellite repeat sequences encoding RAN proteins are generally known. In some embodiments, the RAN protein-associated disease is ALS.
In some embodiments, a subject having or suspected of having a RAN protein-associated disease has one or more micro satellite repeat sequences encoding a poly(GA) RAN protein. In some embodiments, a micro satellite repeat sequence encoding poly(GA) proteins comprises the sequence GGGGCC.
In some aspects, the disclosure relates to the discovery that RAN protein (e.g., poly(GA), etc.) aggregation patterns are length-dependent. For example, RAN proteins having poly amino acid repeats that are >20, >48, or >80 residues in length aggregate differently in the brain of a subject. Generally, the differential aggregation properties of RAN proteins having different lengths can be used to detect RAN proteins in a biological sample. Longer RAN proteins are found at higher levels in biological samples, such as blood, serum, or CSF. In some embodiments, RAN proteins having poly amino acid repeats >40, >50, >60, >70, or >80 amino acid residues in length are detectable in a biological sample.
Monoclonal Antibodies
Various aspects of the disclosure relate to antibodies and antigen-binding fragments that specifically bind to RAN proteins, and methods of making and using the same. In some embodiments, the antibody or antigen binding fragment specifically binds to a poly(glycine- alanine) [poly(GA)] RAN protein. In some embodiments, the antibody binds directly to the poly(GA) repeat region (e.g., binds to the GAGAGA (SEQ ID NO: 58) motif). In some embodiments, the antibody binds to a region of the RAN protein (e.g., poly(GA) RAN protein) that is not the repeat region, for example a unique C-terminal amino acid sequence of the RAN protein.
An antibody, as used herein, broadly refers to an immunoglobulin molecule or any functional mutant, variant, or derivation thereof. It is desired that functional mutants, variants, and derivations thereof, as well as antigen-binding fragments, retain the essential epitope binding features of an Ig molecule. Antibodies are capable of specific binding to a target through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule. Generally, an intact or full-length antibody comprises two heavy chains and two light chains. Each heavy chain contains a heavy chain variable region (VH) and a first, second and third constant regions (CHI, CH2 and CH3). Each light chain contains a light chain variable region (VL) and a constant region (CL). The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FRs). CDR constituents on the heavy chain are referred to as CDRH1, CDRH2, and CDRH3, while CDR constituents on the light chain are referred to as CDRL1, CDRL2, and CDRL3.
The CDRs typically refer to the Kabat CDRs, as described in Sequences of Proteins of Immunological Interest, US Department of Health and Human Services (1991), eds. Kabat et al. Another standard for characterizing the antigen binding site is to refer to the hypervariable loops as described by Chothia. See, e.g., Cho thia, D. et al. (1992) J. Mol. Biol. 227:799-817; and Tomlinson et al. (1995) EMBO J. 14:4628-4638. Still another standard is the AbM definition used by Oxford Molecular’s AbM antibody modeling software. See, generally, e.g., Protein Sequence and Structure Analysis of Antibody Variable Domains. In: Antibody Engineering Lab Manual (Ed.: Duebel, S, and Kontermann, R., Springer- Verlag, Heidelberg). Embodiments described with respect to Kabat CDRs can alternatively be implemented using similar described relationships with respect to Chothia hypervariable loops or to the AbM-defined loops, or combinations of any of these methods.
Each VH and VL is composed of three CDRs and four FRs, arranged from aminoterminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. A full-length antibody can be an antibody of any class, such as IgD, IgE, IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant domain of its heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
The term “antigen-binding fragment” refers to any derivative of an antibody which is less than full-length, and that can bind specifically to a target. Preferably, antigen-binding fragments provided herein retain the ability to specifically bind to a RAN protein. An antigenbinding fragment may comprise the heavy chain variable region (VH), the light chain variable region (VL), or both. Each of the VH and VL typically contains three complementarity determining regions CDR1, CDR2, and CDR3.
Examples of antigen binding fragments include, but are not limited to, Fab, Fab’, F(ab’)2, scFv, Fv, dsFv, diabody, affibodies, and Fd fragments. Antigen binding fragments may be produced by any appropriate means. For instance, an antigen binding fragment may be enzymatically or chemically produced by fragmentation of an intact antibody, or it may be recombinantly produced from a gene encoding the partial antibody sequence. Alternatively, an antigen binding fragment may be wholly or partially synthetically produced. An antigen binding fragment may optionally be a single chain antibody fragment. Alternatively, a fragment may comprise multiple chains which are linked together, for instance, by disulfide linkages. An antigen binding fragment may also optionally be a multimolecular complex. A functional antigen binding fragment will typically comprise at least about 50 amino acids and more typically will comprise at least about 200 amino acids.
Single-chain Fvs (scFvs) are recombinant antigen binding fragments consisting of only the variable light chain (VE) and variable heavy chain (VH) covalently connected to one another by a polypeptide linker. Either VE or VH may be the NH2-terminal domain. The polypeptide linker may be of variable length and composition so long as the two variable domains are bridged without serious steric interference. Typically, the linkers are comprised primarily of stretches of glycine and serine residues with some glutamic acid or lysine residues interspersed for solubility. ScFvs are encompassed within the term “antigen-binding fragment.”
Diabodies are dimeric scFvs. The components of diabodies typically have shorter peptide linkers than most scFvs, and they show a preference for associating as dimers (see, e.g., Holliger, P., et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448; Poljak, R. J., et al. (1994) Structure 2: 1121-1123). Diabodies are also encompassed within the term “antigen-binding fragment.”
A Fv fragment is an antigen binding fragment which consists of one VH and one VE domain held together by noncovalent interactions. Although the two domains of the Fv fragment, VE and VH, can be coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VE and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody. The term dsFv is used herein to refer to an Fv with an engineered intermolecular disulfide bond to stabilize the VH-VE pair. dsFvs are also encompassed within the term “antigen-binding fragment.” A F(ab’)2 fragment is an antigen binding fragment essentially equivalent to that obtained from immunoglobulins (typically IgG) by digestion with an enzyme pepsin at pH 4.0-4.5. The fragment may be recombinantly produced. F(ab’)2 are also encompassed within the term “antigen-binding fragment.”
A Fab fragment is an antigen binding fragment essentially equivalent to that obtained by reduction of the disulfide bridge or bridges joining the two heavy chain pieces in the F(ab’)2 fragment. The Fab’ fragment may be recombinantly produced. Fab’ are also encompassed within the term “antigen-binding fragment.”
A Fab fragment is an antigen binding fragment essentially equivalent to that obtained by digestion of immunoglobulins (typically IgG) with the enzyme papain. The Fab fragment may be recombinantly produced. The heavy chain segment of the Fab fragment is the Fd piece. Fab fragments are also encompassed within the term “antigen-binding fragment.”
An affibody is a small protein comprising a three-helix bundle that functions as an antigen binding molecule (e.g., an antibody mimetic). Generally, affibodies are approximately 58 amino acids in length and have a molar mass of approximately 6 kDa. Affibody molecules with unique binding properties are acquired by randomization of 13 amino acids located in two alpha-helices involved in the binding activity of the parent protein domain. Specific affibody molecules binding a desired target protein can be isolated from pools (libraries) containing billions of different variants, using methods such as phage display. Affibodies are also encompassed within the term “antigen-binding fragment.”
The term “human antibody” refers to antibodies having variable and constant regions corresponding substantially to, or derived from, antibodies obtained from human subjects, e.g., encoded by human germline immunoglobulin sequences or variants thereof. Human antibodies may include one or more amino acid residues not encoded by human germline immunoglobulin sequences e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). Such mutations may present in one or more of the CDRs, particularly CDR3, or in one or more of the framework regions. In some embodiments, the human antibodies may have at least one, two, three, four, five, or more positions replaced with an amino acid residue that is not encoded by the human germline immunoglobulin sequence. However, the term "human antibody", as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
The term "recombinant human antibody", as used herein, is intended to include all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library (Hoogenboom H. R., (1997) TIB Tech. 15:62-70; Azzazy H., and Highsmith W. E., (2002) Clin. Biochem. 35:425-445; Gavilondo J. V., and Larrick J. W. (2002) BioTechniques 29: 128-145;
Hoogenboom H., and Chames P. (2000) Immunology Today 21:371-378), antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see, e.g., Taylor, L. D., et al. (1992) Nucl. Acids Res. 20:6287-6295; Kellermann S-A., and Green L. L. (2002) Current Opinion in Biotechnology 13:593-597; Little M. et al (2000) Immunology Today 21:364-370) or antibodies prepared, expressed, created or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions as defined above. In certain embodiments, however, such recombinant human antibodies may be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies may be sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
In some embodiments, the antibody or antigen-binding fragment comprises a heavy chain comprising an amino acid sequence represented by any one of SEQ ID NOs: 8, 10, 27, and 29. In some embodiments, the anti-RAN antibodies and antigen binding fragments of the disclosure comprise a light chain comprising an amino acid sequence represented by any one of SEQ ID NOs: 13, 15, 32, and 34. In some embodiments, the antibody or antigen-binding fragment comprises a heavy chain comprising an amino acid sequence represented by any one of SEQ ID NOs: 8, 10, 27, and 29 and a light chain comprising an amino acid sequence represented by any one of SEQ ID NOs: 13, 15, 32, and 34. In some embodiments, the antibody is humanized. In some embodiments, the antibody comprises an IgGl, IgG2, IgG3, IgG4, IgAl or IgA2 heavy chain constant region.
In some embodiments, the anti-RAN antibodies or antigen binding fragments may or may not include the framework region of the antibodies, for example the framework region amino acid sequences. In some embodiments, anti-RAN antibodies are murine antibodies. In some embodiments, anti-RAN antibodies are chimeric or humanized antibodies.
In some embodiments, the antibody or antigen binding fragment comprises a VH sequence as set forth in any one of SEQ ID NOs: 7, 17, and 26. In some embodiments, the antibody or antigen binding fragment comprises a VL sequence as set forth in any one of SEQ ID NOs: 12, 18, or 31. In some embodiments, the antibody or antigen binding fragment comprises a VH sequence as set forth in any one of SEQ ID NOs: 7, 17, and 26 and a VL sequence as set forth in any one of SEQ ID NOs: 12, 18, or 31. In some embodiments, the antibody is humanized. In some embodiments, the antibody comprises an IgGl, IgG2, IgG3, IgG4, IgAl or IgA2 heavy chain constant region.
In some embodiments, the antibody or antigen binding fragment comprises a VH sequence as set forth in SEQ ID NO. 7 and a VL sequence as set forth in SEQ ID NO. 12.
In some embodiments, the antibody or antigen binding fragment comprises a VH sequence as set forth in SEQ ID NO. 17 and a VL sequence as set forth in SEQ ID NO. 18.
In some embodiments, the antibody or antigen binding fragment comprises a VH sequence as set forth in SEQ ID NO. 26 and a VL sequence as set forth in SEQ ID NO. 31.
In some embodiments, antibody or antigen-binding fragment comprises six complementarity determining regions (CDRs): CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3, wherein CDRH1 comprises a sequence as set forth in SEQ ID NO: 1, CDRH2 comprises a sequence as set forth in SEQ ID NO: 2, CDRH3 comprises a sequence as set forth in SEQ ID NO: 3, CDRL1 comprises a sequence as set forth in SEQ ID NO: 4, CDRL2 comprises a sequence as set forth in SEQ ID NO: 5, and CDRL3 comprises a sequence as set forth in SEQ ID NO: 6. In some embodiments, the antibody is humanized. In some embodiments, the antibody comprises an IgGl, IgG2, IgG3, IgG4, IgAl or IgA2 heavy chain constant region.
In some embodiments, antibody or antigen-binding fragment comprises six complementarity determining regions (CDRs): CDRH1, CDRH2, CDRH3, CDRL1, CDRL2, and CDRL3, wherein CDRH1 comprises a sequence as set forth in SEQ ID NO: 20, CDRH2 comprises a sequence as set forth in SEQ ID NO: 21, CDRH3 comprises a sequence as set forth in SEQ ID NO: 22, CDRL1 comprises a sequence as set forth in SEQ ID NO: 23, CDRL2 comprises a sequence as set forth in SEQ ID NO: 24, and CDRL3 comprises a sequence as set forth in SEQ ID NO: 25. In some embodiments, the antibody is humanized. In some embodiments, the antibody comprises an IgGl, IgG2, IgG3, IgG4, IgAl or IgA2 heavy chain constant region.
It should be appreciated that, in some embodiments, the disclosure contemplates variants (e.g., homologs) of amino acid and nucleic acid sequences for the heavy chain variable region and light chain variable region of the antibodies. “Homology” refers to the percent identity between two polynucleotides or two polypeptide moieties. The term "substantial homology", when referring to a nucleic acid, or fragment thereof, indicates that, when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in about 90 to 100% of the aligned sequences. For example, in some embodiments, nucleic acid sequences sharing substantial homology are 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% at least 99% sequence identity. When referring to a polypeptide, or fragment thereof, the term “substantial homology” indicates that, when optimally aligned with appropriate gaps, insertions or deletions with another polypeptide, there is nucleotide sequence identity in about 90 to 100% of the aligned sequences. The term "highly conserved" means at least 80% identity, preferably at least 90% identity, and more preferably, over 97% identity. For example, in some embodiments, highly conserved proteins share at least 85%, 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% at least 99% identity. In some cases, highly conserved may refer to 100% identity. Identity is readily determined by one of skill in the art by, for example, the use of algorithms and computer programs known by those of skill in the art.
In some embodiments, RAN antibodies of the disclosure can bind to a RAN protein with high affinity, e.g., with a Kd less than 10'7 M, 10'8 M, 10'9 M, 10'10 M, 10 1 M or lower. For example, anti-RAN antibodies or antigen binding fragments can bind to a RAN protein with an affinity between 5 pM and 500 nM, e.g., between 50 pM and 100 nM, e.g., between 500 pM and 50 nM. The disclosure also includes antibodies or antigen binding fragments that compete with any of the antibodies described herein for binding to RAN proteins and that have an affinity of 50 nM or lower (e.g., 20 nM or lower, 10 nM or lower, 500 pM or lower, 50 pM or lower, or 5 pM or lower). The affinity and binding kinetics of the anti-RAN protein antibody can be tested using any method known in the art including but not limited to biosensor technology (e.g., OCTET or BIACORE).
In some embodiments, anti-RAN antibodies of the present disclosure include the VH, VL, and CDR, amino acid sequences shown in Table 1 below.
Table 1: Representative sequences (listed as SEQ ID NOs) of developed anti-poly(GA) monoclonal antibodies
Figure imgf000015_0001
Figure imgf000016_0001
In some embodiments, antibody clone 27B11 binds to poly(GA). In some embodiments, clone 27B11 is an IgGl antibody (e.g., comprises an IgGl heavy chain constant region). In some embodiments, clone 27B11 is an IgG2 antibody (e.g., comprises an IgG2 heavy chain constant region). In some embodiments, clone 27B11 is an scFv. In some embodiments, antibody clone 23H2 binds to poly(GA). In some embodiments, antibody clone 23H2 is an IgGl antibody (e.g., comprises an IgGl heavy chain constant region). In some embodiments, antibody clone 23H2 is an IgG2 antibody (e.g., comprises an IgG2 heavy chain constant region).
Anti-RAN antibodies may be used to treat, or assist in the treatment of, one or more symptoms of a disease associated with RAN proteins. In some embodiments, the disease associated with RAN proteins is selected from the group consisting of: amyotrophic lateral sclerosis (ALS), or frontotemporal dementia; myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2); spinocerebellar ataxia types 1, 2, 3, 6, 7, 8, 10, 12, 17, 31, and 36; spinal bulbar muscular atrophy; dentatorubral-pallidoluysian atrophy (DRPLA); Huntington's disease (HD); Fragile X Tremor Ataxia Syndrome (FXTAS); Fuch's endothelial corneal dystrophy (FECD); Huntington's disease-like 2 syndrome (HDL2); Fragile X syndrome (FXS); disorders related to 7pl 1 .2 folate-sensitive fragile site FRA7A; disorders related to folatesensitive fragile site 2ql 1 FRA2A; and Fragile XE syndrome (FRAXE). In a specific embodiment, the neurological disease associated with RAN proteins is ALS.
In some embodiments, the anti-RAN antibodies may be used to treat, or assist in the treatment of, one or more symptoms of a disease associated with RAN proteins, for example by administering a therapeutically effective amount of one or more anti-RAN antibodies to a subject diagnosed as having one or more symptoms of a disease associated with RAN proteins (e.g., the early stages of ALS) or being at risk of developing a disease associated with RAN proteins (e.g., based on one or more assays described in this application).
To “treat” a disease (e.g., a disease associated with poly(GA) RAN translation, for example ALS) as the term is used herein, means to reduce the frequency or severity of at least one sign or symptom of a disease or disorder experienced by a subject. The compositions described above or elsewhere herein are typically administered to a subject in an effective amount, that is, an amount capable of producing a desirable result. The desirable result will depend upon the active agent being administered. A therapeutically acceptable amount of an anti-RAN protein antibody may be an amount that is capable of treating a disease, e.g., ALS, by reducing expression and/or aggregation of RAN proteins and/or appearance or number of RNA foci comprising RAN protein-encoding microsatellite repeat sequences. As is well known in the medical and veterinary arts, dosage for any one subject depends on many factors, including the subject's size, body surface area, age, the particular composition to be administered, the active ingredient(s) in the composition, time and route of administration, general health, and other drugs being administered concurrently.
In some embodiments, one or more of the anti-RAN antibody or antigen binding fragments disclosed herein are administered to a subject, wherein the subject has been characterized as having a disease associated with RAN proteins by the detection of at least one RAN protein in a biological sample obtained from the subject.
Anti-RAN antibody production
Typically, polyclonal antibodies are produced by inoculation of a suitable mammal, such as a mouse, rabbit or goat. An antigen is injected into the mammal. This induces the B- lymphocytes to produce IgG immunoglobulins specific for the antigen. This polyclonal IgG is purified from the mammal's serum. Monoclonal antibodies are generally produced by a single cell line (e.g., a hybridoma cell line). In some embodiments, an anti-RAN antibody is purified (e.g., isolated from serum). In some embodiments, an antigen comprises a poly(GA) RAN protein repeat sequence.
Numerous methods may be used for obtaining anti-RAN antibodies. For example, antibodies can be produced using recombinant DNA methods. Monoclonal antibodies may also be produced by generation of hybridomas (see, e.g., Kohler and Milstein (1975) Nature, 256: 495-499) in accordance with known methods. Hybridomas formed in this manner are then screened using standard methods, such as enzyme-linked immunosorbent assay (ELISA; e.g., RCA-based ELISA or rtPCR-based ELISA) and surface plasmon resonance (e.g., OCTET or BIACORE) analysis, to identify one or more hybridomas that produce an antibody that specifically binds with a specified antigen. Any form of the specified antigen (e.g., a RAN protein) may be used as the immunogen, e.g., recombinant antigen, naturally occurring forms, any variants or fragments thereof. One exemplary method of making antibodies includes screening protein expression libraries that express antibodies or fragments thereof (e.g., scFv), e.g., phage or ribosome display libraries. Phage display is described, for example, in Ladner et al., U.S. Pat. No. 5,223,409; Smith (1985) Science 228: 1315-1317; Clackson et al. (1991) Nature, 352: 624-628; Marks et al. (1991) J. Mol. Biol., 222: 581-597W092/18619; WO 91/17271 ; WO 92/20791; WO 92/15679; WO 93/01288; WO 92/01047; WO 92/09690; and WO 90/02809. In another embodiment, a monoclonal antibody is obtained from the non-human animal, and then modified, e.g., made chimeric, using recombinant DNA techniques known in the art. A variety of approaches for making chimeric antibodies have been described. See, e.g., Morrison et al., Proc. Natl. Acad. Sci. U.S.A. 81:6851, 1985; Takeda et al., Nature 314:452, 1985, Cabilly at al., U.S. Pat. No. 4,816,567; Boss et al., U.S. Pat. No. 4,816,397; Tanaguchi et al., European Patent Publication EP 171496; European Patent Publication 0173494, United Kingdom Patent GB 2177096B.
Antibodies can also be humanized by methods known in the art. For example, monoclonal antibodies with a desired binding specificity can be commercially humanized (Scotgene, Scotland; and Oxford Molecular, Palo Alto, Calif.). Fully humanized antibodies, such as those expressed in transgenic animals are within the scope of the invention (see, e.g., Green et al. (1994) Nature Genetics 7, 13; and U.S. Patent Nos. 5,545,806 and 5,569,825). For additional antibody production techniques, see, Antibodies: A Laboratory Manual, Second Edition. Edited by Edward A. Greenfield, Dana-Farber Cancer Institute, ©2014. The present disclosure is not necessarily limited to any particular source, method of production, or other special characteristics of an antibody.
Some aspects of the present disclosure relate to isolated cells (e.g., host cells) transformed with a polynucleotide or vector. Host cells may be a prokaryotic or eukaryotic cell. The polynucleotide or vector which is present in the host cell may either be integrated into the genome of the host cell or it may be maintained extrachromosomally. The host cell can be any prokaryotic or eukaryotic cell, such as a bacterial, insect, fungal, plant, animal or human cell. In some embodiments, fungal cells are, for example, those of the genus Saccharomyces, in particular those of the species .S'. cerevisiae. The term "prokaryotic" includes all bacteria which can be transformed or transfected with a DNA or RNA molecules for the expression of an antibody or the corresponding immunoglobulin chains. Prokaryotic hosts may include gram negative as well as gram positive bacteria such as, for example, E. coli, S. typhimurium, Serratia marcescens and Bacillus subtilis. The term "eukaryotic" includes yeast, higher plants, insects and vertebrate cells, e.g., mammalian cells, such as NSO and CHO cells. Depending upon the host employed in a recombinant production procedure, the antibodies or immunoglobulin chains encoded by the polynucleotide may be glycosylated or may be nonglycosylated. Antibodies or the corresponding immunoglobulin chains may also include an initial methionine amino acid residue.
In some embodiments, once a vector has been incorporated into an appropriate host, the host may be maintained under conditions suitable for high level expression of the nucleotide sequences, and, as desired, the collection and purification of the immunoglobulin light chains, heavy chains, light/heavy chain dimers or intact antibodies, antigen binding fragments or other immunoglobulin forms may follow; see, Beychok, Cells of Immunoglobulin Synthesis, Academic Press, N.Y., (1979). Thus, polynucleotides or vectors are introduced into the cells which in turn produce the antibody or antigen binding fragments. Furthermore, transgenic animals, preferably mammals, comprising the aforementioned host cells may be used for the large scale production of the antibody or antibody fragments.
The transformed host cells can be grown in fermenters and cultured according to techniques known in the art to achieve optimal cell growth. Once expressed, the whole antibodies, their dimers, individual light and heavy chains, other immunoglobulin forms, or antigen binding fragments, can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like; see, Scopes, "Protein Purification", Springer Verlag, N.Y. (1982). The antibody or antigen binding fragments can then be isolated from the growth medium, cellular lysates, or cellular membrane fractions. The isolation and purification of the, e.g., microbially expressed antibodies or antigen binding fragments may be by any conventional means such as, for example, preparative chromatographic separations and immunological separations, such as those involving the use of monoclonal or polyclonal antibodies directed, e.g., against the constant region of the antibody.
Aspects of the disclosure relate to a hybridoma, which provides an indefinitely prolonged source of monoclonal antibodies. As used herein, “hybridoma cell” refers to an immortalized cell derived from the fusion of B lymphoblasts with a myeloma fusion partner. For preparing monoclonal antibody-producing cells (e.g., hybridoma cells), an individual animal whose antibody titer has been confirmed (e.g., a mouse) is selected, and 2 days to 5 days after the final immunization, its spleen or lymph node is harvested and antibody-producing cells contained therein are fused with myeloma cells to prepare the desired monoclonal antibody producer hybridoma. Measurement of the antibody titer in antiserum can be carried out, for example, by reacting the labeled protein, as described hereinafter and antiserum and then measuring the activity of the labeling agent bound to the antibody. The cell fusion can be carried out according to known methods, for example, the method described by Koehler and Milstein (Nature 256:495 (1975)). As a fusion promoter, for example, polyethylene glycol (PEG) or Sendai virus (HVJ) is used.
Examples of myeloma cells include NS-1, P3U1, SP2/0, AP-1 and the like. The proportion of the number of antibody producer cells (spleen cells) and the number of myeloma cells to be used is preferably about 1:1 to about 20:1. PEG (preferably PEG 1000-PEG 6000) is preferably added in concentration of about 10% to about 80%. Cell fusion can be carried out efficiently by incubating a mixture of both cells at about 20°C to about 40 °C, preferably about 30 °C to about 37 °C for about 1 minute to 10 minutes.
Various methods may be used for screening for a hybridoma producing the antibody (e.g., against a tumor antigen or autoantibody of the present invention). For example, where a supernatant of the hybridoma is added to a solid phase (e.g., microplate) to which antibody is adsorbed directly or together with a carrier and then an anti-immunoglobulin antibody (if mouse cells are used in cell fusion, anti-mouse immunoglobulin antibody is used) or Protein A labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase. Alternately, a supernatant of the hybridoma is added to a solid phase to which an anti-immunoglobulin antibody or Protein A is adsorbed and then the protein labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase.
Selection of the monoclonal antibody can be carried out according to any known method or its modification. Normally, a medium for animal cells to which HAT (hypoxanthine, aminopterin, thymidine) are added is employed. Any selection and growth medium can be employed as long as the hybridoma can grow. For example, RPMI 1640 medium containing 1% to 20%, preferably 10% to 20% fetal bovine serum, GIT medium containing 1% to 10% fetal bovine serum, a serum free medium for cultivation of a hybridoma (SFM-101, Nissui Seiyaku) and the like can be used. Normally, the cultivation is carried out at 20°C to 40°C, preferably 37°C for about 5 days to 3 weeks, preferably 1 week to 2 weeks under about 5% CO2 gas. The antibody titer of the supernatant of a hybridoma culture can be measured according to the same manner as described above with respect to the antibody titer of the anti-protein in the antiserum.
As an alternative to obtaining immunoglobulins directly from the culture of hybridomas, immortalized hybridoma cells can be used as a source of rearranged heavy chain and light chain loci for subsequent expression and/or genetic manipulation. Rearranged antibody genes can be reverse transcribed from appropriate mRNAs to produce cDNA. If desired, the heavy chain constant region can be exchanged for that of a different isotype or eliminated altogether. The variable regions can be linked to encode single chain Fv regions. Multiple Fv regions can be linked to confer binding ability to more than one target or chimeric heavy and light chain combinations can be employed. Any appropriate method may be used for cloning of antibody variable regions and generation of recombinant antibodies.
In some embodiments, an appropriate nucleic acid that encodes variable regions of a heavy and/or light chain is obtained and inserted into an expression vectors which can be transfected into standard recombinant host cells. A variety of such host cells may be used. In some embodiments, mammalian host cells may be advantageous for efficient processing and production. Typical mammalian cell lines useful for this purpose include CHO cells, 293 cells, or NSO cells. The production of the antibody or antigen binding fragment may be undertaken by culturing a modified recombinant host under culture conditions appropriate for the growth of the host cells and the expression of the coding sequences. The antibodies or antigen binding fragments may be recovered by isolating them from the culture. The expression systems may be designed to include signal peptides so that the resulting antibodies are secreted into the medium; however, intracellular production is also possible.
The disclosure also includes a polynucleotide encoding at least a variable region of an immunoglobulin chain of the antibodies described herein. In some embodiments, the variable region encoded by the polynucleotide comprises at least one complementarity determining region (CDR) of the VH and/or VL of the variable region of the antibody produced by any one of the above described hybridomas.
Polynucleotides encoding antibody or antigen binding fragments may be, e.g., DNA, cDNA, RNA or synthetically produced DNA or RNA or a recombinantly produced chimeric nucleic acid molecule comprising any of those polynucleotides either alone or in combination. In some embodiments, a polynucleotide is part of a vector. Such vectors may comprise further genes, such as marker genes which allow for the selection of the vector in a suitable host cell and under suitable conditions.
In some embodiments, a polynucleotide is operatively linked to expression control sequences allowing expression in prokaryotic or eukaryotic cells. Expression of the polynucleotide comprises transcription of the polynucleotide into a translatable mRNA. Regulatory elements ensuring expression in eukaryotic cells, preferably mammalian cells, are well known to those skilled in the art. They may include regulatory sequences that facilitate initiation of transcription and optionally poly-A signals that facilitate termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers, and/or naturally associated or heterologous promoter regions. Possible regulatory elements permitting expression in prokaryotic host cells include, e.g., the PL, Lac, Trp or Tac promoter in E. coli, and examples of regulatory elements permitting expression in eukaryotic host cells are the A0X1 or GALI promoter in yeast or the CMV-promoter, SV40-promoter, RSV-promoter (Rous sarcoma virus), CMV-enhancer, SV40- enhancer or a globin intron in mammalian and other animal cells.
Beside elements which are responsible for the initiation of transcription, such regulatory elements may also include transcription termination signals, such as the SV40-poly-A site or the tk-poly-A site, downstream of the polynucleotide. Furthermore, depending on the expression system employed, leader sequences capable of directing the polypeptide to a cellular compartment or secreting it into the medium may be added to the coding sequence of the polynucleotide and are well known in the art. The leader sequence(s) is (are) assembled in appropriate phase with translation, initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein, or a portion thereof, into, for example, the extracellular medium. Optionally, a heterologous polynucleotide sequence can be used that encode a fusion protein including a C- or N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
In some embodiments, polynucleotides encoding at least the variable domain of the light and/or heavy chain may encode the variable domains of both immunoglobulin chains or only one. Likewise, polynucleotides may be under the control of the same promoter or may be separately controlled for expression. Furthermore, some aspects relate to vectors, particularly plasmids, cosmids, viruses and bacteriophages used conventionally in genetic engineering that comprise a polynucleotide encoding a variable domain of an immunoglobulin chain of an antibody or antigen binding fragment; optionally in combination with a polynucleotide that encodes the variable domain of the other immunoglobulin chain of the antibody.
In some embodiments, expression control sequences are provided as eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells, but control sequences for prokaryotic hosts may also be used. Expression vectors derived from viruses, such as retroviruses, vaccinia virus, adeno-associated virus, herpes viruses, or bovine papilloma virus, may be used for delivery of the polynucleotides or vector into targeted cell population (e.g., to engineer a cell to express an antibody or antigen binding fragment). A variety of appropriate methods can be used to construct recombinant viral vectors. In some embodiments, polynucleotides and vectors can be reconstituted into liposomes for delivery to target cells. The vectors containing the polynucleotides (e.g., the heavy and/or light variable domain(s) of the immunoglobulin chains encoding sequences and expression control sequences) can be transferred into the host cell by suitable methods, which vary depending on the type of cellular host.
In some embodiments, a vector (e.g., a plasmid) comprises at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% identity to a sequence set forth in any one of SEQ ID NOs: 42-46. In some embodiments, a vector (e.g., a plasmid) comprises a sequence set forth in any one of SEQ ID NOs: 42-46.
Modifications Some aspects of the disclosure relate to antibody-drug conjugates targeted against one or more RAN proteins. As used herein, “antibody drug conjugate” refers to molecules comprising an antibody, or antigen binding fragment thereof, linked to a targeted molecule (e.g., a biologically active molecule, such as a therapeutic molecule, and/or a detectable label). Accordingly, in some embodiments, antibodies or antigen binding fragments of the disclosure may be modified with a detectable label, including, but not limited to, an enzyme, prosthetic group, fluorescent material, luminescent material, bioluminescent material, radioactive material, positron emitting metal, nonradioactive paramagnetic metal ion, and affinity label for detection and isolation of one or more RAN proteins. The detectable substance may be coupled or conjugated either directly to the polypeptides of the disclosure or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. Non-limiting examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, |3-galactosidase, glucose oxidase, or acetylcholinesterase; non-limiting examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; non-limiting examples of suitable fluorescent materials include biotin, umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride, or phycoerythrin; an example of a luminescent material includes luminol; non-limiting examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include a radioactive metal ion, e.g., alpha-emitters or other radioisotopes such as, for example, iodine (131I, 125I, 123I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (115mln, 113mln, 112In, U 1ln), and technetium ("Tc, "mTc), thallium (201Ti), gallium (68Ga, 67Ga), palladium (103Pd), molybdenum ("Mo), xenon (133Xe), fluorine (18F), 153Sm, Lu, 159Gd, 149Pm, 140La, 175Yb, 166HO, 90Y, 47SC, 86R, 188Re, 142Pr, 105Rh, 97Ru, 68Ge, 57Co, 65Zn, 85Sr, 32P, 153Gd, 169Yb, 51Cr, 54Mn, 75Se, and tin (113Sn, 117Sn). The detectable substance may be coupled or conjugated either directly to the anti-RAN antibodies or antigen-binding fragments of the disclosure or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. Anti-RAN antibodies conjugated to a detectable substance may be used for diagnostic assays as described herein.
In some embodiments, antibodies or antigen binding fragments of the disclosure may be modified with a therapeutic moiety e.g., therapeutic agent). In some embodiments, the antibody is coupled to the targeted agent via a linker. As used herein, the term "linker" refers to a molecule or sequence, such as an amino acid sequence, that attaches, as in a bridge, one molecule or sequence to another molecule or sequence. "Linked," "conjugated," or "coupled" means attached or bound by covalent bonds, or non-covalent bonds, or other bonds, such as van der Waals forces. Antibodies described by the disclosure can be linked to the targeted agent (e.g., therapeutic moiety or detectable moiety) directly, e.g., as a fusion protein with protein or peptide detectable moieties (with or without an optional linking sequence, e.g., a flexible linker sequence) or via a chemical coupling moiety. A number of such coupling moieties are known in the art, e.g., a peptide linker or a chemical linker, e.g., as described in International Patent Application Publication No. WO 2009/036092. In some embodiments, the linker is a flexible amino acid sequence. Examples of flexible amino acid sequences include glycine and serine rich linkers, which comprise a stretch of two or more glycine residues. In some embodiments, the linker is a photolinker. Examples of photolinkers include ketyl-reactive benzophenone (BP), anthraquinone (AQ), nitrene-reactive nitrophenyl azide (NPA), and carbene-reactive phenyl- (trifluoromethyl)diazirine (PTD).
Pharmaceutical Compositions
In some aspects, the disclosure relates to pharmaceutical compositions comprising anti- RAN antibodies or antigen binding fragments. In some embodiments, the composition comprises an anti-RAN antibody and a pharmaceutically acceptable carrier. As used herein the term “pharmaceutically acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions. Pharmaceutical compositions can be prepared as described below. The active ingredients may be admixed or compounded with any conventional, pharmaceutically acceptable carrier or excipient. The compositions may be sterile.
Typically, pharmaceutical compositions are formulated for delivering an effective amount of an agent (e.g., an anti-RAN antibody). In general, an “effective amount” of an active agent refers to an amount sufficient to elicit the desired biological response (e.g., ameliorating one or more symptoms of ALS). An effective amount of an agent may vary depending on such factors as the desired biological endpoint, the pharmacokinetics of the compound, the disease being treated (e.g., ALS, repeat expansion diseases), the mode of administration, and the patient.
A composition is said to be a “pharmaceutically acceptable carrier” if its administration can be tolerated by a recipient patient. Sterile phosphate-buffered saline is one example of a pharmaceutically acceptable carrier. Other suitable carriers are well-known in the art. See, for example, REMINGTON'S PHARMACEUTICAL SCIENCES, 18th Ed. (1990). It will be understood by those skilled in the art that any mode of administration, vehicle or carrier conventionally employed and which is inert with respect to the active agent may be utilized for preparing and administering the pharmaceutical compositions of the present disclosure. Illustrative of such methods, vehicles and carriers are those described, for example, in Remington's Pharmaceutical Sciences, 4th ed. (1970), the disclosure of which is incorporated herein by reference. Those skilled in the art, having been exposed to the principles of the disclosure, will experience no difficulty in determining suitable and appropriate vehicles, excipients and carriers or in compounding the active ingredients therewith to form the pharmaceutical compositions of the disclosure.
An effective amount, also referred to as a therapeutically effective amount, of a compound (e.g., an anti-RAN antibody) is an amount sufficient to ameliorate at least one adverse effect associated with a disease associated with RAN proteins, such as, e.g., memory loss, cognitive impairment, loss of coordination, speech impairment, etc. In some embodiments, the neurological disease associated with RAN proteins is selected from the group consisting of: amyotrophic lateral sclerosis (ALS), or frontotemporal dementia; myotonic dystrophy type 1 (DM1) and myotonic dystrophy type 2 (DM2); spinocerebellar ataxia types 1, 2, 3, 6, 7, 8, 10, 12, 17, 31, and 36; spinal bulbar muscular atrophy; dentatorubral-pallidoluysian atrophy (DRPLA); Huntington's disease (HD); Fragile X Tremor Ataxia Syndrome (FXTAS); Fuch's endothelial corneal dystrophy (FECD); Huntington's disease-like 2 syndrome (HDL2); Fragile X syndrome (FXS); disorders related to 7pl 1 .2 folate- sensitive fragile site FRA7A; disorders related to folate- sensitive fragile site 2ql 1 FRA2A; and Fragile XE syndrome (FRAXE). In a specific embodiment, the neurological disease associated with RAN proteins is ALS. The therapeutically effective amount to be included in pharmaceutical compositions depends, in each case, upon several factors, e.g., the type, size and condition of the patient to be treated, the intended mode of administration, the capacity of the patient to incorporate the intended dosage form, etc. Generally, an amount of active agent is included in each dosage form to provide from about 0.1 to about 250 mg/kg, and preferably from about 0.1 to about 100 mg/kg. One of ordinary skill in the art would be able to determine empirically an appropriate therapeutically effective amount.
Combined with the teachings provided herein, by choosing among the various active compounds and weighing factors such as potency, relative bioavailability, patient body weight, severity of adverse side-effects and selected mode of administration, an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial toxicity and yet is entirely effective to treat the particular subject. The effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular therapeutic agent being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art can empirically determine the effective amount of a particular nucleic acid and/or other therapeutic agent without necessitating undue experimentation .
The term pharmaceutically-acceptable carrier means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other vertebrate animal. The term carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being commingled with the compounds of the present disclosure, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency.
The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
Suitable liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin. The pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above. The pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of methods for drug delivery, see, Langer R (1990) Science 249:1527-1533, which is incorporated herein by reference.
The compounds may be administered per se (neat) or in the form of a pharmaceutically acceptable salt. When used in medicine the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically acceptable salts thereof. Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p-toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthalene-2-sulphonic, and benzene sulphonic. Also, such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
The compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the compounds into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the compounds into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product. Liquid dose units are vials or ampoules. Solid dose units are tablets, capsules and suppositories.
Administration
A therapeutic agent may be delivered by any suitable modality known in the art.
Aspects of the disclosure relate to the delivery of a therapeutically effective amount of a therapeutic agent to a subject. In some embodiments, a therapeutically effective amount is an amount effective in reducing repeat expansions in the subject. In some embodiments, a therapeutically effective amount is an amount effective in reducing the transcription of RNAs that produce RAN proteins in a subject. In certain embodiments, a therapeutically effective amount is an amount effective in reducing the translation of RAN proteins in a subject. In some embodiments, a therapeutically effective amount is an amount effective for treating ALS associated with repeat expansions. “Reducing” expression of a repeat sequence or RAN protein translation refers to a decrease in the amount or level of repeat sequence expression or RAN protein translation in a subject after administration of a therapeutic agent (and relative to the amount or level in the subject prior to the administration).
In certain embodiments, the effective amount is an amount effective in reducing the level of RAN proteins by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 98% (e.g., the level of RAN proteins relative to the level of RAN proteins in a cell or subject that has not been administered a therapeutic agent). In certain embodiments, the effective amount is an amount effective in reducing the translation of RAN proteins by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 98% (e.g., the level of RAN proteins relative the level of RAN proteins in a cell or subject that has not been administered a therapeutic agent).
Pharmaceutical compositions described herein can be prepared by any method known in the art of pharmacology. In general, such preparatory methods include bringing the compound described herein (z.e., the “active ingredient”) into association with a carrier or excipient, and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping, and/or packaging the product into a desired single- or multi-dose unit.
Pharmaceutical compositions can be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. A “unit dose” is a discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient. The amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage, such as one- half or one-third of such a dosage.
Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition described herein will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered. The composition may comprise between 0.1% and 100% (w/w) active ingredient.
Therapeutic agents described herein are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions described herein will be decided by a physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the disease being treated and the severity of the disorder; the activity of the specific active ingredient employed; the specific composition employed; the age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific active ingredient employed; the duration of the treatment; drugs used in combination or coincidental with the specific active ingredient employed; and like factors well known in the medical arts.
A therapeutic agent can be administered by any route, including enteral (e.g., oral), parenteral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal, buccal, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol. Specifically contemplated routes are oral administration, intravenous administration (e.g., systemic intravenous injection), regional administration via blood and/or lymph supply, and/or direct administration to an affected site. In general, the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), and/or the condition of the subject (e.g., whether the subject is able to tolerate oral administration). The exact amount of a therapeutic agent required to achieve an effective amount will vary from subject to subject, depending, for example, on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular compound, mode of administration, and the like. An effective amount may be included in a single dose (e.g., single oral dose) or multiple doses (e.g., multiple oral doses). In certain embodiments, when multiple doses are administered to a subject or applied to a biological sample, tissue, or cell, any two doses of the multiple doses include different or substantially the same amounts of a compound described herein. In certain embodiments, when multiple doses are administered to a subject or applied to a biological sample, tissue, or cell, the frequency of administering the multiple doses to the subject or applying the multiple doses to the biological sample, tissue, or cell is three doses a day, two doses a day, one dose a day, one dose every other day, one dose every third day, one dose every week, one dose every two weeks, one dose every three weeks, or one dose every four weeks. In certain embodiments, the frequency of administering the multiple doses to the subject or applying the multiple doses to the biological sample, tissue, or cell is one dose per day. In certain embodiments, the frequency of administering the multiple doses to the subject or applying the multiple doses to the biological sample, tissue, or cell is two doses per day. In certain embodiments, the frequency of administering the multiple doses to the subject or applying the multiple doses to the biological sample, tissue, or cell is three doses per day. In certain embodiments, when multiple doses are administered to a subject or applied to a biological sample, tissue, or cell, the duration between the first dose and last dose of the multiple doses is one day, two days, four days, one week, two weeks, three weeks, one month, two months, three months, four months, six months, eight months, nine months, one year, two years, three years, four years, five years, seven years, ten years, fifteen years, twenty years, or the lifetime of the subject, tissue, or cell. In certain embodiments, the duration between the first dose and last dose of the multiple doses is three months, six months, or one year. In certain embodiments, the duration between the first dose and last dose of the multiple doses is the lifetime of the subject, tissue, or cell.
In some embodiments, a treatment for a disease associated with RAN protein expression is administered to the central nervous system (CNS) of a subject in need thereof. As used herein, the “central nervous system (CNS)” refers to all cells and tissues of the brain and spinal cord of a subject, including but not limited to neuronal cells, glial cells, astrocytes, cerebrospinal fluid, etc. Modalities of administering a therapeutic agent to the CNS of a subject include direct injection into the brain (e.g., intracerebral injection, intraventricular injection, intraparenchymal injection, etc.), direct injection into the spinal cord of a subject (e.g., intrathecal injection, lumbar injection, etc.), or any combination thereof. In some embodiments, a treatment as described by the disclosure is systemically administered to a subject, for example by intravenous injection. Systemically administered therapeutic molecules can be modified, in some embodiments, in order to improve delivery of the molecules to the CNS of a subject. Examples of modifications that improve CNS delivery of therapeutic molecules include but are not limited to co-administration or conjugation to blood brain barrier-targeting agents (e.g., transferrin, melanotransferrin, low-density lipoprotein (LDL), angiopeps, RVG peptide, etc., as disclosed by Georgieva et al. Pharmaceuticals 6(4): 557-583 (2014)), coadministration with BBB disrupting agents (e.g., bradykinins), and physical disruption of the BBB prior to administration (e.g., by MRI-Guided Focused Ultrasound), etc.
Detection Methods
In some embodiments, a method described herein comprises utilizing an antibody or antigen binding fragment described herein to detect a RAN protein (e.g., a poly(GA) RAN protein). In some embodiments, the RAN protein is comprised or suspected of being comprised in a biological sample obtained from a subject (e.g., a subject characterized as having or suspected of having a disease, such as a neurological disease, associated with RAN protein expression (e.g., poly(GA) RAN protein expression), translation, and/or accumulation). Nonlimiting examples of biological samples include those comprising one or more cells (e.g., brain cells, neuronal cells, skin cells, white blood cells, etc.), and tissue samples, such as blood (e.g., whole blood), serum (e.g., plasma from which the clotting proteins have been removed), cerebrospinal fluid (CSF), and nervous system tissue (e.g., central nervous system (CNS) tissue, such as spinal cord tissue and brain tissues including, but not limited to, hippocampal tissue and cortex tissue) samples.
In some embodiments, a method comprises contacting one or more antibodies or antigenbinding fragments (e.g., 1, 2, 3, or more than 3 antibodies or antigen-binding fragments) described herein with a biological sample. In some embodiments, a method comprises detecting a complex formed between one or more antibodies, or antigen binding fragments, and a RAN protein in a biological sample, such as by measuring a detectable signal in the biological sample that represents the presence of the complex or the amount of the complex and determining the presence and/or amount of the RAN protein in the biological sample. In some embodiments, a method comprises contacting an antibody or antigen-binding fragment described herein with a first biological sample and a second biological sample. In some embodiments, one of the biological samples is a control sample. In some embodiments, the first biological sample is obtained from a subject prior to administration of an agent (e.g., an agent used in the treatment of a disease or disorder described herein) to the subject and the second biological sample is obtained from the subject after administration of the agent. In some embodiments, the first biological sample is obtained from a subject after the subject has been administered an agent (e.g., an agent used in the treatment of a disease or disorder described herein) and the second biological sample is obtained from the subject after the subject is no longer being administered the agent. In some embodiments, wherein a method comprises contacting a first biological sample and a second biological sample obtained from a subject with an antibody or antigenbinding fragment described herein, the second biological sample can be collected hours (e.g., 1- 2, 2-6, 6-12, or 12-24 hours), days (e.g., 1, 2, 3, 4, 5, 6, or 7 days), weeks (e.g., 1-2, 2-3, or 3-4 weeks), or months (e.g., 1-2, 2-3, 3-4, 4-5, 5-6, 6-9, 9-12 months, or more than 12 months) after the first biological sample was obtained. In some embodiments, a method described herein comprises detecting a RAN protein (e.g., a poly(GA) RAN protein) in a biological sample obtained from a subject and/or detecting a difference (e.g., an increase or a decrease) in the levels of a RAN protein (e.g., a poly(GA) RAN protein) between two biological samples obtained from a subject, and then administering an agent (e.g., an agent used in the treatment of a disease or disorder described herein) to a subject.
In some embodiments, a method for detecting a RAN protein as described herein comprises an immunoassay. Non-limiting examples of immunoassays include immunoblot (e.g., dot blot, 2-D gel electrophoresis, Western Blot, etc.), electrochemiluminescence immunoassay (e.g., Meso-Scale Detection (MSD)), immunohistochemistry (IHC), ELISA (e.g., RCA-based ELISA or RT-PCR-based ELISA), label free immunoassays such as surface plasmon resonance bio layer interferometry, immunoquantitative PCR, bead-based immunoassays, immunoprecipitation, immunostaining, and immunoelectrophoresis. Aspects of the RAN protein detection methods described herein may also include a method previously described in International Application No. PCT/US2014/022670, International Application No. PCT/US2018/052745, International Application No. PCT/US2021/032488, International Application No. PCT/US2020/051671, International Application No. PCT/US2020/054976, or International Application No. PCT/US2022/079048, each which is incorporated by reference herein for their disclosures related to RAN protein detection methods.
The following Examples are intended to illustrate the benefits of the present invention and to describe particular embodiments, but are not intended to exemplify the full scope of the invention. Accordingly, it will be understood that the Examples are not meant to limit the scope of the invention.
EXAMPLES This Example describes production and testing of monoclonal antibodies that bind to poly (GA) RAN proteins.
FIG. 1 shows representative data for immunohistochemistry (IHC) assays for anti- poly(GA) monoclonal antibodies. Supernatants from cells expressing anti-poly(GA) monoclonal antibody 27B11 in several formats (IgGl, IgG2, ScFv) recognize poly-GA RAN protein aggregates in C9Bac mouse cells. m-GA= mouse anti-GA polyclonal antibody; EV= vehicle- only negative control; ScFv 515 = 27B11 ScFv format; IgGl 515A = 27B11 monoclonal antibody in IgGl format; IgGl 515B = 27B11 monoclonal antibody in IgGl format; IgG2 515 = 27B 11 monoclonal antibody in IgG2 format.
FIG. 2 shows representative data indicating expression of anti-poly(GA) monoclonal antibody 27B11 in several formats (IgGl, IgG2, ScFv) lowers levels of (GA)eo-GFP RAN protein in cells.
REPRESENTATIVE SEQUENCES
In some embodiments, an antibody or antigen-binding fragment described herein comprises one or more of the amino acid sequences set forth below (see, e.g., any one of the amino acid sequences set forth in SEQ ID NOs: 1-35 and 41). In some embodiments, a nucleic acid described herein (e.g., a vector, such as a plasmid) comprises one or more nucleotide sequences set forth below (see, e.g. any one of the nucleotide sequences set forth in SEQ ID NOs: 36-40 and 42-46) and/or encodes one or more of the amino acid sequences set forth below (see, e.g., any one of the amino acid sequences set forth in SEQ ID NOs: 1-35 and 41). It should be appreciated that one or more of the representative sequences set forth below may be utilized according to the foregoing embodiments of compositions and methods as described herein.
> Anti-polyGA antibody clone 27B11 heavy chain CDR1 amino acid sequence (SEQ ID NO: 1) GFAFSNYG
> Anti-polyGA antibody clone 27B11 heavy chain CDR2 amino acid sequence (SEQ ID NO: 2) INSDGDST
> Anti-polyGA antibody clone 27B11 heavy chain CDR3 amino acid sequence (SEQ ID NO: 3) ARVGGNYDFAMDY
> Anti-polyGA antibody clone 27B11 light chain CDR1 amino acid sequence (SEQ ID NO: 4) QSLLNSRTRKNY
> Anti-polyGA antibody clone 27B11 light chain CDR2 amino acid sequence (SEQ ID NO: 5) WTS > Anti-polyGA antibody clone 27B11 light chain CDR3 amino acid sequence (SEQ ID NO: 6) KQSYNNPWT
> Anti-polyGA antibody clone 27B 11 heavy chain variable region amino acid sequence (SEQ ID NO: 7)
EVQLQESGGGSVQPGGSLKLSCAASGFAFSNYGMSWVRQTPDKRLELVTTINSDGDSTF YPDSVKGRFTISRDNAKNAEYEQMSSEKSDDTAMYYCARVGGNYDFAMDYWGQGTS VIVSS
> Anti-polyGA antibody clone 27B11 heavy chain IgGl amino acid sequence (SEQ ID NO: 8) EVQEQESGGGSVQPGGSEKESCAASGFAFSNYGMSWVRQTPDKREEEVTTINSDGDSTF YPDSVKGRFTISRDNAKNAEYEQMSSEKSDDTAMYYCARVGGNYDFAMDYWGQGTS VIVSSAKTTPPSVYPEAPGSAAQTNSMVTEGCEVKGYFPEPVTVTWNSGSESSGVHTFP AVEQSDEYTESSSVTVPSSTWPSETVTCNVAHPASSTKVDKKIVPRDCGCKPCICTVPEV SSVFIFPPKPKDVETITETPKVTCVVVDISKDDPEVQFSWFVDDVEVHTAQTQPREEQFN STFRSVSEEPIMHQDWENGKEFKCRVNSAAFPAPIEKTISKTKGRPKAPQVYTIPPPKEQ MAKDKVSETCMITDFFPEDITVEWQWNGQPAENYKNTQPIMDTDGSYFVYSKENVQK SNWEAGNTFTCSVEHEGEHNHHTEKSESHSPGK
> Anti-polyGA antibody clone 27B11 heavy chain IgGl amino acid sequence with signal peptide (SEQ ID NO: 9)
MDWTWRVFCLLAVAPGAHSEVQLQESGGGSVQPGGSLKLSCAASGFAFSNYGMSWV RQTPDKRLELVTTINSDGDSTFYPDSVKGRFTISRDNAKNALYLQMSSLKSDDTAMYYC ARVGGNYDFAMDYWGQGTSVIVSSAKTTPPSVYPLAPGSAAQTNSMVTLGCLVKGYF PEPVTVTWNSGSLSSGVHTFPAVLQSDLYTLSSSVTVPSSTWPSETVTCNVAHPASSTKV DKKIVPRDCGCKPCICTVPEVSSVFIFPPKPKDVLTITLTPKVTCVVVDISKDDPEVQFSW FVDDVEVHTAQTQPREEQFNSTFRSVSELPIMHQDWLNGKEFKCRVNSAAFPAPIEKTIS KTKGRPKAPQVYTIPPPKEQMAKDKVSLTCMITDFFPEDITVEWQWNGQPAENYKNTQ PIMDTDGSYFVYSKLNVQKSNWEAGNTFTCSVLHEGLHNHHTEKSLSHSPGK
> Anti-polyGA antibody clone 27B11 heavy chain IgG2 amino acid sequence (SEQ ID NO: 10) EVQLQESGGGSVQPGGSLKLSCAASGFAFSNYGMSWVRQTPDKRLELVTTINSDGDSTF YPDSVKGRFTISRDNAKNALYLQMSSLKSDDTAMYYCARVGGNYDFAMDYWGQGTS VIVSSAKTTAPSVYPLAPVCGDTTGSSVTLGCLVKGYFPEPVTLTWNSGSLSSGVHTFPA VLQSDLYTLSSSVTVTSSTWPSQSITCNVAHPASSTKVDKKIEPRGPTIKPCPPCKCPAPN LLGGPSVFIFPPKIKDVLMISLSPIVTCVVVDVSEDDPDVQISWFVNNVEVHTAQTQTHR EDYNSTLRVVSALPIQHQDWMSGKEFKCKVNNKDLPAPIERTISKPKGSVRAPQVYVLP PPEEEMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPVLDSDGSYFMYSK LRVEKKNWVERNSYSCSVVHEGLHNHHTTKSFSRTPGK
> Anti-polyGA antibody clone 27B 11 heavy chain IgG2 amino acid sequence with signal peptide (SEQ ID NO: 11)
MKCSWVIFFLMAVVIGINSEVQLQESGGGSVQPGGSLKLSCAASGFAFSNYGMSWVRQ TPDKRLELVTTINSDGDSTFYPDSVKGRFTISRDNAKNALYLQMSSLKSDDTAMYYCAR VGGNYDFAMDYWGQGTSVIVSSAKTTAPSVYPLAPVCGDTTGSSVTLGCLVKGYFPEP VTLTWNSGSLSSGVHTFPAVLQSDLYTLSSSVTVTSSTWPSQSITCNVAHPASSTKVDKK IEPRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLMISLSPIVTCVVVDVSEDDPDVQIS WFVNNVEVHTAQTQTHREDYNSTLRVVSALPIQHQDWMSGKEFKCKVNNKDLPAPIE RTISKPKGSVRAPQVYVLPPPEEEMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNY
KNTEPVLDSDGSYFMYSKLRVEKKNWVERNSYSCSVVHEGLHNHHTTKSFSRTPGK
> Anti-polyGA antibody clone 27B 11 light chain variable region amino acid sequence (SEQ ID NO: 12)
DIVMSQFPSSLAVSAGDKVTMSCKSSQSLLNSRTRKNYLAWYQQKPGQSPKLLIYWTS TRESGVPDRFTGSRSGTDFTLTISSVQAEDLAVYYCKQSYNNPWTFGGGTKLEIK
> Anti-polyGA antibody clone 27B11 light chain IgGl amino acid sequence (SEQ ID NO: 13) DIVMSQFPSSLAVSAGDKVTMSCKSSQSLLNSRTRKNYLAWYQQKPGQSPKLLIYWTS TRESGVPDRFTGSRSGTDFTLTISSVQAEDLAVYYCKQSYNNPWTFGGGTKLEIKRADA APTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGSERQNGVLNSWTDQDSKD STYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRNEC
> Anti-polyGA antibody clone 27B11 light chain IgGl amino acid sequence with signal peptide (SEQ ID NO: 14)
MKLPVRLLVLMFWIPASSSDIVMSQFPSSLAVSAGDKVTMSCKSSQSLLNSRTRKNYLA WYQQKPGQSPKLLIYWTSTRESGVPDRFTGSRSGTDFTLTISSVQAEDLAVYYCKQSYN NPWTFGGGTKLEIKRADAAPTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGS ERQNGVLNSWTDQDSKDSTYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRN EC
> Anti-polyGA antibody clone 27B11 light chain IgG2 amino acid sequence (SEQ ID NO: 15) DIVMSQFPSSLAVSAGDKVTMSCKSSQSLLNSRTRKNYLAWYQQKPGQSPKLLIYWTS TRESGVPDRFTGSRSGTDFTLTISSVQAEDLAVYYCKQSYNNPWTFGGGTKLEIKRADA APTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGSERQNGVLNSWTDQDSKD STYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRNEC
> Anti-polyGA antibody clone 27B 11 light chain IgG2 amino acid sequence with signal peptide (SEQ ID NO: 16)
MKLPVRLLVLMFWIPASSSDIVMSQFPSSLAVSAGDKVTMSCKSSQSLLNSRTRKNYLA WYQQKPGQSPKLLIYWTSTRESGVPDRFTGSRSGTDFTLTISSVQAEDLAVYYCKQSYN NPWTFGGGTKLEIKRADAAPTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGS ERQNGVLNSWTDQDSKDSTYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRN EC
> Anti-polyGA antibody clone 27B11 scFv heavy chain (SEQ ID NO: 17)
EVQLQESGGGSVQPGGSLKLSCAASGFAFSNYGMSWVRQTPDKRLELVTTINSDGDSTF YPDSVKGRFTISRDNAKNALYLQMSSLKSDDTAMYYCARVGGNYDFAMDYWGQGTS VIVSS > Anti-polyGA antibody clone 27B11 scFv light chain (SEQ ID NO: 18)
DIVMSQFPSSLAVSAGDKVTMSCKSSQSLLNSRTRKNYLAWYQQKPGQSPKLLIYWTS
TRESGVPDRFTGSRSGTDFTLTISSVQAEDLAVYYCKQSYNNPWTFGGGTKLEIK
> Anti-polyGA antibody clone 27B 11 scFv complete sequence with signal peptide and poly-GS linker (SEQ ID NO: 19)
MDWTWRVFCLLAVAPGAHSEVQLQESGGGSVQPGGSLKLSCAASGFAFSNYGMSWV
RQTPDKRLELVTTINSDGDSTFYPDSVKGRFTISRDNAKNALYLQMSSLKSDDTAMYYC
ARVGGNYDFAMDYWGQGTSVIVSSGGGGSGGGGSGGGGSDIVMSQFPSSLAVSAGDK VTMSCKSSQSLLNSRTRKNYLAWYQQKPGQSPKLLIYWTSTRESGVPDRFTGSRSGTDF TLTISSVQAEDLAVYYCKQSYNNPWTFGGGTKLEIK
> Anti-polyGA antibody clone 23H2 heavy chain CDR1 amino acid sequence (SEQ ID NO: 20) GFTFSSHG
> Anti-polyGA antibody clone 23H2heavy chain CDR2 amino acid sequence (SEQ ID NO: 21) INSNGGST
> Anti-polyGA antibody clone 23H2heavy chain CDR3 amino acid sequence (SEQ ID NO: 22) ARVGDNDDFAMGY
> Anti-polyGA antibody clone 23H2 light chain CDR1 amino acid sequence (SEQ ID NO: 23) QSLFNSRTRKNY
> Anti-polyGA antibody clone 23H2 light chain CDR2 amino acid sequence (SEQ ID NO: 24) WTS
> Anti-polyGA antibody clone 23H2 light chain CDR3 amino acid sequence (SEQ ID NO: 25) KQSYNNPWT
> Anti-polyGA antibody clone 23H2 heavy chain variable region amino acid sequence (SEQ ID
NO: 26)
EVQLQESGGGSVQPGGALQLSCAASGFTFSSHGMSWVRQTPDKRLEMVATINSNGGST YYPDSVKGRFIISRDNAKNTLYLQMSSLKSEDTAMYYCARVGDNDDFAMGYWGQGTS VTVSS
> Anti-polyGA antibody clone 23H2 heavy chain IgGl amino acid sequence (SEQ ID NO: 27)
EVQLQESGGGSVQPGGALQLSCAASGFTFSSHGMSWVRQTPDKRLEMVATINSNGGST
YYPDSVKGRFIISRDNAKNTLYLQMSSLKSEDTAMYYCARVGDNDDFAMGYWGQGTS
VTVSSAKTTPPSVYPLAPGSAAQTNSMVTLGCLVKGYFPEPVTVTWNSGSLSSGVHTFP
AVLQSDLYTLSSSVTVPSSTWPSETVTCNVAHPASSTKVDKKIVPRDCGCKPCICTVPEV
SSVFIFPPKPKDVLTITLTPKVTCVVVDISKDDPEVQFSWFVDDVEVHTAQTQPREEQFN
STFRSVSELPIMHQDWLNGKEFKCRVNSAAFPAPIEKTISKTKGRPKAPQVYTIPPPKEQ MAKDKVSLTCMITDFFPEDITVEWQWNGQPAENYKNTQPIMDTDGSYFVYSKLNVQK SNWEAGNTFTCSVLHEGLHNHHTEKSLSHSPGK > Anti-poly GA antibody clone 23H2 heavy chain IgGl amino acid sequence with signal peptide (SEQ ID NO: 28)
MDWTWRVFCLLAVAPGAHSEVQLQESGGGSVQPGGALQLSCAASGFTFSSHGMSWVR QTPDKRLEMVATINSNGGSTYYPDSVKGRFIISRDNAKNTLYLQMSSLKSEDTAMYYC ARVGDNDDFAMGYWGQGTSVTVSSAKTTPPSVYPLAPGSAAQTNSMVTLGCLVKGYF PEPVTVTWNSGSLSSGVHTFPAVLQSDLYTLSSSVTVPSSTWPSETVTCNVAHPASSTKV DKKIVPRDCGCKPCICTVPEVSSVFIFPPKPKDVLTITLTPKVTCVVVDISKDDPEVQFSW
FVDDVEVHTAQTQPREEQFNSTFRSVSELPIMHQDWLNGKEFKCRVNSAAFPAPIEKTIS KTKGRPKAPQVYTIPPPKEQMAKDKVSLTCMITDFFPEDITVEWQWNGQPAENYKNTQ PIMDTDGSYFVYSKLNVQKSNWEAGNTFTCSVLHEGLHNHHTEKSLSHSPGK
> Anti-polyGA antibody clone 23H2 heavy chain IgG2 amino acid sequence (SEQ ID NO: 29) EVQLQESGGGSVQPGGALQLSCAASGFTFSSHGMSWVRQTPDKRLEMVATINSNGGST YYPDSVKGRFIISRDNAKNTLYLQMSSLKSEDTAMYYCARVGDNDDFAMGYWGQGTS VTVSSAKTTAPSVYPLAPVCGDTTGSSVTLGCLVKGYFPEPVTLTWNSGSLSSGVHTFP AVLQSDLYTLSSSVTVTSSTWPSQSITCNVAHPASSTKVDKKIEPRGPTIKPCPPCKCPAP
NLLGGPSVFIFPPKIKDVLMISLSPIVTCVVVDVSEDDPDVQISWFVNNVEVHTAQTQTH REDYNSTLRVVSALPIQHQDWMSGKEFKCKVNNKDLPAPIERTISKPKGSVRAPQVYVL PPPEEEMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPVLDSDGSYFMYSK LRVEKKNWVERNSYSCSVVHEGLHNHHTTKSFSRTPGK
> Anti-polyGA antibody clone 23H2 heavy chain IgG2 amino acid sequence with signal peptide (SEQ ID NO: 30)
MDWTWRVFCLLAVAPGAHSEVQLQESGGGSVQPGGALQLSCAASGFTFSSHGMSWVR QTPDKRLEMVATINSNGGSTYYPDSVKGRFIISRDNAKNTLYLQMSSLKSEDTAMYYC ARVGDNDDFAMGYWGQGTSVTVSSAKTTAPSVYPLAPVCGDTTGSSVTLGCLVKGYF PEPVTLTWNSGSLSSGVHTFPAVLQSDLYTLSSSVTVTSSTWPSQSITCNVAHPASSTKV DKKIEPRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLMISLSPIVTCVVVDVSEDDPD
VQISWFVNNVEVHTAQTQTHREDYNSTLRVVSALPIQHQDWMSGKEFKCKVNNKDLP APIERTISKPKGSVRAPQVYVLPPPEEEMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTE LNYKNTEPVLDSDGSYFMYSKLRVEKKNWVERNSYSCSVVHEGLHNHHTTKSFSRTPG K
> Anti-polyGA antibody clone 23H2 light chain variable region amino acid sequence (SEQ ID NO: 31)
DIVMSQSPSSLAVSEGEKVTLTCKSSQSLFNSRTRKNYLAWYQQKPGQPPKLLIYWTST RESGVPDRFTGSGYGTDFTLTISSVQAEDLAVYYCKQSYNNPWTFGGGTKLEIK
> Anti-polyGA antibody clone 23H2 light chain IgGl amino acid sequence (SEQ ID NO: 32) DIVMSQSPSSLAVSEGEKVTLTCKSSQSLFNSRTRKNYLAWYQQKPGQPPKLLIYWTST RESGVPDRFTGSGYGTDFTLTISSVQAEDLAVYYCKQSYNNPWTFGGGTKLEIKRADA APTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGSERQNGVLNSWTDQDSKD STYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRNEC > Anti-poly GA antibody clone 23H2 light chain IgGl amino acid sequence with signal peptide (SEQ ID NO: 33)
MKLPVRLLVLMFWIPASSSDIVMSQSPSSLAVSEGEKVTLTCKSSQSLFNSRTRKNYLA WYQQKPGQPPKLLIYWTSTRESGVPDRFTGSGYGTDFTLTISSVQAEDLAVYYCKQSYN NPWTFGGGTKLEIKRADAAPTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGS ERQNGVLNSWTDQDSKDSTYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRN EC
> Anti-polyGA antibody clone 23H2 light chain IgG2 amino acid sequence (SEQ ID NO: 34) DIVMSQSPSSLAVSEGEKVTLTCKSSQSLFNSRTRKNYLAWYQQKPGQPPKLLIYWTST RESGVPDRFTGSGYGTDFTLTISSVQAEDLAVYYCKQSYNNPWTFGGGTKLEIKRADA APTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGSERQNGVLNSWTDQDSKD STYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRNEC
> Anti-polyGA antibody clone 23H2 light chain IgG2 amino acid sequence with signal peptide (SEQ ID NO: 35)
MKLPVRLLVLMFWIPASSSDIVMSQSPSSLAVSEGEKVTLTCKSSQSLFNSRTRKNYLA WYQQKPGQPPKLLIYWTSTRESGVPDRFTGSGYGTDFTLTISSVQAEDLAVYYCKQSYN NPWTFGGGTKLEIKRADAAPTVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGS ERQNGVLNSWTDQDSKDSTYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRN EC
> Anti-polyGA antibody clone 27B11 IgGl nucleic acid sequence (with signal peptide and bGH poly(A) signal) (SEQ ID NO: 36) atggattggacttggagagtgttttgcctgctggctgtcgcacctggagctcatagtgaggtgcagctgcaggagtctgggggaggctcag tgcagcctggagggtccctgaaactctcctgcgcagcctctggattcgctttcagtaactatggcatgtcttgggttcgccagactccagaca agaggctggagttggtcacaaccattaatagtgatggtgatagtaccttttatccagacagtgtgaagggccgattcaccatctccagagaca atgccaagaacgccctgtacctgcaaatgagcagtctgaagtcagacgacacagccatgtattactgtgcaagagtgggaggtaactacg actttgctatggactactggggtcagggaacctcagtcatcgtGtcctcaGCTAAAACGACACCCCCATCTGTCTA TCCACTGGCCCCTGGATCTGCTGCCCAAACTAACTCgATGGTGACCCTGGGATGCCT GGTCAAGGGCTATTTCCCTGAGCCAGTGACAGTGACCTGGAACTCTGGTTCCCTGTC CAGCGGTGTGCACACCTTCCCAGCTGTCCTcCAGTCTGACCTCTACACTCTGAGCAG CTCAGTGACTGTCCCCTCCAGCACCTGGCCCAGCGAGACCGTCACCTGCAACGTTGC CCACCCGGCCAGCAGCACCAAGGTGGACAAGAAAATTGTGCCCAGGGATTGTGGTT GTAAGCCTTGCATATGcACAGTCCCAGAAGTATCATCTGTCTTCATCTTCCCCCCAA AGCCCAAGGATGTGCTCACCATTACTCTGACTCCTAAGGTCACGTGTGTTGTGGTAG ACATCAGCAAGGATGATCCCGAGGTCCAGTTCAGCTGGTTTGTAGATGATGTGGAG GTGCACACAGCTCAGACGCAACCCCGGGAGGAGCAGTTCAACAGCACTTTCCGCTC AGTCAGTGAACTTCCCATCATGCACCAGGACTGGCTCAATGGCAAGGAGTTCAAAT GCAGGGTCAACAGTGCAGCTTTCCCTGCCCCCATCGAGAAAACCATCTCCAAAACC AAAGGCAGACCGAAGGCTCCGCAGGTcTACACCATTCCACCTCCCAAGGAGCAGAT GGCCAAGGATAAAGTCAGTCTGACCTGCATGATAACAGACTTCTTCCCTGAAGACA TTACTGTGGAGTGGCAGTGGAATGGGCAGCCAGCGGAGAACTACAAGAACACTCA GCCCATCATGGACACAGATGGCTCTTACTTCGTCTACAGCAAGCTCAATGTGCAGA AGAGCAACTGGGAGGCAGGAAATACTTTCACCTGCTCTGTGTTACATGAGGGCCTG CACAACCACCATACTGAGAAGAGCCTCTCCCACTCTCCTGGTAAAgtgaaacagactttgaattt tgaccttctcaagttggcgggaGACGTCgagtccaaccctggacccATGAAGTTGCCTGTTAGGCTGTTGGT GCTGATGTTCTGGATCCCTGCTTCCAGCAGTGACATTGTGATGTCACAGTTTCCATC CTCCCTGGCTGTGTCAGCAGGAGATAAGGTCACTATGAGCTGCAAATCCAGTCAGA GTCTGCTCAACAGTAGGACCCGAAAGAACTACTTGGCTTGGTAtCAGCAGAAACCA GGGCAGTCTCCTAAACTACTGATCTACTGGACATCCACTCGGGAATCTGGcGTCCCT GATCGCTTCACAGGCAGTCGATCTGGGACAGATTTCACTCTCACCATCAGCAGTGTa CAGGCTGAAGACCTGGCAGTTTATTACTGCAAGCAATCTTATAATAATCCGTGGAC GTTCGGTGGAGGCACCAAGCTtGAAATAAAACGGGCAGATGCTGCAcCAACTGTATC gATCTTCCCACCATCCAGTGAGCAGTTAACATCcGGAGGTGCCTCAGTCGTGTGCTTC TTGAACAACTTCTACCCCAAAGACATCAATGTCAAGTGGAAGATTGATGGCAGTGA ACGACAAAATGGCGTCCTGAACAGTTGGACTGATCAGGACAGCAAAGACAGCACCT ACAGCATGAGCAGCACCCTCACGTTGACCAAGGACGAGTATGAACGACATAACAGC TATACCTGTGAGGCCACTCACAAGACATCAACTTCACCCATTGTCAAaAGCTTCAAC AGGAATGAGTGTTAGgcGGCCGCCACTGTGCTGGATATCGTTTAAACCGCTGATCAG CCTCGACTGTGCCTTCTAGTTGCCAGCCATCTGTTGTTTGCCCCTCCCCCGTGCCTTC CTTGACCCTGGAAGGTGCCACTCCCACTGTCCTTTCCTAATAAAATGAGGAAATTGC ATCGCATTGTCTGAGTAGGTGTCATTCTATTCTGGGGGGTGGGGTGGGGCAGGACA GCAAGGGGGAGGATTGGGAAGACAATAGCAGGCATGCTGGGGATGCGGTGGGCTC TATGG
> Anti-polyGA antibody clone 27B 11 IgG2 nucleic acid sequence (with signal peptide and bGH poly(A) signal) (SEQ ID NO: 37)
ATGAAATGCAGCTGGGTCATCTTCTTCCTGATGGCAGTGGTTATAGGAATCAATTCA gaggtgcagctgcaggagtctgggggaggctcagtgcagcctggagggtccctgaaactctcctgcgcagcctctggattcgctttcagt aactatggcatgtcttgggttcgccagactccagacaagaggctggagttggtcacaaccattaatagtgatggtgatagtaccttttatccag acagtgtgaagggccgattcaccatctccagagacaatgccaagaacgccctgtacctgcaaatgagcagtctgaagtcagacgacaca gccatgtattactgtgcaagagtgggaggtaactacgactttgctatggactactggggtcagggaacctcagtcatcgtgtcctcaGCC AAAACAACAGCCCCATCGGTCTATCCACTGGCCCCTGTGTGTGGAGATACAACTGG CTCCTCGGTGACcCTAGGATGCCTGGTCAAGGGTTATTTCCCTGAGCCAGTGACCTT GACCTGGAACTCTGGATCgCTGTCCAGTGGTGTGCACACCTTCCCAGCTGTCCTGCA GTCTGACCTCTACACCCTgAGCAGCTCAGTGACTGTAACCTCcAGCACCTGGCCCAG CCAGTCCATCACCTGCAATGTGGCCCACCCGGCAAGCAGCACCAAGGTGGACAAGA AAATTGAGCCCAGAGGGCCCACAATCAAGCCCTGTCCTCCATGCAAATGCCCAGCA CCTAACCTCTTGGGTGGACCATCCGTCTTCATCTTCCCTCCAAAGATCAAGGATGTA CTCATGATCTCCCTGAGCCCCATAGTCACATGTGTGGTGGTGGATGTGAGCGAGGAT GACCCAGATGTCCAGATCAGCTGGTTTGTGAACAACGTGGAAGTACACACAGCTCA GACACAAACCCATAGAGAGGATTACAACAGTACTCTCCGGGTGGTCAGTGCCCTCC CCATCCAGCACCAGGACTGGATGAGTGGCAAGGAGTTCAAATGCAAGGTCAACAAC AAAGACCTCCCAGCGCCCATCGAGAGAACCATCTCAAAACCCAAAGGGTCAGTAAG AGCTCCACAGGTATATGTCTTGCCTCCACCAGAAGAAGAGATGACTAAGAAACAGG TCACTCTGACCTGCATGGTCACAGACTTCATGCCTGAAGACATTTACGTGGAGTGGA CCAACAACGGGAAAACAGAGCTAAACTACAAGAACACTGAACCAGTCCTGGACTCT GATGGTTCTTACTTCATGTACAGCAAGCTGAGAGTGGAAAAGAAGAACTGGGTGGA AAGAAATAGCTACTCCTGTTCAGTGGTCCACGAGGGTCTGCACAATCACCACACGA CTAAGAGCTTCTCCCGGACTCCGGGTAAAgtgaaacagactttgaattttgaccttctcaagttggcgggagacg tggagtccaaccctggacctATGAAGTTGCCTGTTAGGCTGTTGGTGCTGATGTTCTGGATcCCT GCTTCCAGCAGTgacattgtgatgtcacagtttccatcctccctggctgtgtcagcaggagataaggtcactatgagctgcaaatc cagtcagagtctgctcaacagtaggacccgaaagaactacttggcttggtaccagcagaaaccagggcagtctcctaaactactgatctact ggacatccactcgggaatctggcgtccctgatcgcttcacaggcagtcgatctgggacagatttcactctcaccatcagcagtgtgcaggct gaagacctggcagtttattactgcaagcaatcttataataatccgtggacgttcggtggaggcaccaagcttgaaataaaaCGGGCTG ATGCTGCACCAACTGTATCgATCTTCCCACCATCCAGTGAGCAGTTAACATCcGGAG GTGCCTCAGTCGTGTGCTTCTTGAACAACTTCTACCCCAAAGACATCAATGTCAAGT GGAAGATTGATGGCAGTGAACGACAAAATGGCGTCCTGAACAGTTGGACTGATCAG GACAGCAAAGACAGCACCTACAGCATGAGCAGCACCCTCACGTTGACCAAGGACG AGTATGAACGACATAACAGCTATACCTGTGAGGCCACTCACAAGACATCAACTTCA CCCATTGTCAAaAGCTTCAACAGGAATGAGTGTTAGgcGGCCGCCACTGTGCTGGAT ATCGTTTAAACCGCTGATCAGCCTCGACTGTGCCTTCTAGTTGCCAGCCATCTGTTG TTTGCCCCTCCCCCGTGCCTTCCTTGACCCTGGAAGGTGCCACTCCCACTGTCCTTTC CTAATAAAATGAGGAAATTGCATCGCATTGTCTGAGTAGGTGTCATTCTATTCTGGG GGGTGGGGTGGGGCAGGACAGCAAGGGGGAGGATTGGGAAGACAATAGCAGGCAT GCTGGGGATGCGGTGGGCTCTATGG
> Anti-polyGA antibody clone 27B 11 ScFv nucleic acid sequence (with signal peptide, FLAG, and bGH poly(A) signal) (SEQ ID NO: 38)
ATGGATTGGACTTGGAGAGTGTTTTGCCTGCTGGCTGTCGCACCTGGGGCTCATAGT GAGGTGCAGCTGCAGGAGTCTGGGGGAGGCTCAGTGCAGCCTGGAGGGTCCCTGAA ACTCTCCTGCGCAGCCTCTGGATTCGCTTTCAGTAACTATGGCATGTCTTGGGTTCG CCAGACTCCAGACAAGAGGCTGGAGTTGGTCACAACCATTAATAGTGATGGTGATA GTACCTTTTATCCAGACAGTGTGAAGGGCCGATTCACCATCTCCAGAGACAATGCC AAGAACGCCCTGTACCTGCAAATGAGCAGTCTGAAGTCAGACGACACAGCCATGTA TTACTGTGCAAGAGTGGGAGGTAACTACGACTTTGCTATGGACTACTGGGGTCAGG GAACCTCAGTCATCGTCTCCTCAGGTGGAGGCGGTTCAGGCGGAGGTGGCAGCGGC GGTGGCGGGTCGGACATTGTGATGTCACAGTTTCCATCCTCCCTGGCTGTGTCAGCA GGAGATAAGGTCACTATGAGCTGCAAATCCAGTCAGAGTCTGCTCAACAGTAGGAC CCGAAAGAACTACTTGGCTTGGTACCAGCAGAAACCAGGGCAGTCTCCTAAACTAC TGATCTACTGGACATCCACTCGGGAATCTGGGGTCCCTGATCGCTTCACAGGCAGTC GATCTGGGACAGATTTCACTCTCACCATCAGCAGTGTGCAGGCTGAAGACCTGGCA GTTTATTACTGCAAGCAATCTTATAATAATCCGTGGACGTTCGGTGGAGGCACCAAG CTGGAAATAAAAgattataaagatcatgatggcgattataaagatcatgatattgattataaagatgatgatgataaataagcGG CCGCCACTGTGCTGGATATCGTTTAAACCGCTGATCAGCCTCGACTGTGCCTTCTAG TTGCCAGCCATCTGTTGTTTGCCCCTCCCCCGTGCCTTCCTTGACCCTGGAAGGTGCC ACTCCCACTGTCCTTTCCTAATAAAATGAGGAAATTGCATCGCATTGTCTGAGTAGG
TGTCATTCTATTCTGGGGGGTGGGGTGGGGCAGGACAGCAAGGGGGAGGATTGGGA AGACAATAGCAGGCATGCTGGGGATGCGGTGGGCTCTATGG
> Anti-polyGA antibody clone 23H2 IgGl nucleic acid sequence (with signal peptide and bGH poly(A) signal) (SEQ ID NO: 39) atggattggacttggagagtgttttgcctgctggctgtcgcacctggagctcatagtgaggtgcagctgcaggagtctgggggaggctcag tgcagcctggaggggccctgcaactctcctgtgcagcctctggattcactttcagtagtcatggcatgtcttgggttcgccagactccagaca agaggctggaaatggtcgcaaccattaatagtaatggtgggagtacctattacccagacagtgtgaagggccgattcatcatctccagaga caatgccaaaaacaccctgtacctgcaaatgagcagtctgaagtctgaggacacagccatgtattactgtgcaagagtgggagataacgac gactttgctatgggctactggggtcaaggaacctcagtcaccgtgtcctcaGCTAAAACGACACCCCCATCTGTCT ATCCACTGGCCCCTGGATCTGCTGCCCAAACTAACTCgATGGTGACCCTGGGATGCC TGGTCAAGGGCTATTTCCCTGAGCCAGTGACAGTGACCTGGAACTCTGGTTCCCTGT CCAGCGGTGTGCACACCTTCCCAGCTGTCCTcCAGTCTGACCTCTACACTCTGAGCA GCTCAGTGACTGTCCCCTCCAGCACCTGGCCCAGCGAGACCGTCACCTGCAACGTTG CCCACCCGGCCAGCAGCACCAAGGTGGACAAGAAAATTGTGCCCAGGGATTGTGGT TGTAAGCCTTGCATATGcACAGTCCCAGAAGTATCATCTGTCTTCATCTTCCCCCCAA AGCCCAAGGATGTGCTCACCATTACTCTGACTCCTAAGGTCACGTGTGTTGTGGTAG ACATCAGCAAGGATGATCCCGAGGTCCAGTTCAGCTGGTTTGTAGATGATGTGGAG GTGCACACAGCTCAGACGCAACCCCGGGAGGAGCAGTTCAACAGCACTTTCCGCTC AGTCAGTGAACTTCCCATCATGCACCAGGACTGGCTCAATGGCAAGGAGTTCAAAT GCAGGGTCAACAGTGCAGCTTTCCCTGCCCCCATCGAGAAAACCATCTCCAAAACC AAAGGCAGACCGAAGGCTCCGCAGGTcTACACCATTCCACCTCCCAAGGAGCAGAT GGCCAAGGATAAAGTCAGTCTGACCTGCATGATAACAGACTTCTTCCCTGAAGACA TTACTGTGGAGTGGCAGTGGAATGGGCAGCCAGCGGAGAACTACAAGAACACTCA GCCCATCATGGACACAGATGGCTCTTACTTCGTCTACAGCAAGCTCAATGTGCAGA AGAGCAACTGGGAGGCAGGAAATACTTTCACCTGCTCTGTGTTACATGAGGGCCTG CACAACCACCATACTGAGAAGAGCCTCTCCCACTCTCCTGGTAAAgtgaaacagactttgaattt tgaccttctcaagttggcgggaGACGTCgagtccaaccctggacccATGAAGTTGCCTGTTAGGCTGTTGGT GCTGATGTTCTGGATCCCTGCTTCCAGCAGTgacattgtgatgtcacagtctccatcctccctggctgtgtcaga aggagagaaggtcactttaacctgcaaatccagtcagagtttgttcaacagtagaacccgaaagaactacttggcttggtaccagcagaaa ccagggcagcctcctaaactgttgatctactggacatccactagggaatctggggtccctgatcgcttcacaggcagtggatatgggacag atttcactctcaccatcagcagtgtgcaggctgaagacctggcagtttattactgcaaacaatcttataataatccgtggacgttcggtggagg caccaagcttgaaataaaaCGGGCAGATGCTGCAcCAACTGTATCgATCTTCCCACCATCCAGT GAGCAGTTAACATCcGGAGGTGCCTCAGTCGTGTGCTTCTTGAACAACTTCTACCCC AAAGACATCAATGTCAAGTGGAAGATTGATGGCAGTGAACGACAAAATGGCGTCCT GAACAGTTGGACTGATCAGGACAGCAAAGACAGCACCTACAGCATGAGCAGCACC CTCACGTTGACCAAGGACGAGTATGAACGACATAACAGCTATACCTGTGAGGCCAC TCACAAGACATCAACTTCACCCATTGTCAAaAGCTTCAACAGGAATGAGTGTTAGgc GGCCGCCACTGTGCTGGATATCGTTTAAACCGCTGATCAGCCTCGACTGTGCCTTCT AGTTGCCAGCCATCTGTTGTTTGCCCCTCCCCCGTGCCTTCCTTGACCCTGGAAGGT GCCACTCCCACTGTCCTTTCCTAATAAAATGAGGAAATTGCATCGCATTGTCTGAGT AGGTGTCATTCTATTCTGGGGGGTGGGGTGGGGCAGGACAGCAAGGGGGAGGATTG GGAAGACAATAGCAGGCATGCTGGGGATGCGGTGGGCTCTATGG
> Anti-polyGA antibody clone 23H2 IgG2 nucleic acid sequence (with signal peptide and bGH poly(A) signal) (SEQ ID NO: 40) atggattggacttggagagtgttttgcctgctggctgtcgcacctggagctcatagtgaggtgcagctgcaggagtctgggggaggctcag tgcagcctggaggggccctgcaactctcctgtgcagcctctggattcactttcagtagtcatggcatgtcttgggttcgccagactccagaca agaggctggaaatggtcgcaaccattaatagtaatggtgggagtacctattacccagacagtgtgaagggccgattcatcatctccagaga caatgccaaaaacaccctgtacctgcaaatgagcagtctgaagtctgaggacacagccatgtattactgtgcaagagtgggagataacgac gactttgctatgggctactggggtcaaggaacctcagtcaccgtgtcctcaGCCAAAACAACAGCCCCATCGGTCT ATCCACTGGCCCCTGTGTGTGGAGATACAACTGGCTCCTCGGTGACcCTAGGATGCC TGGTCAAGGGTTATTTCCCTGAGCCAGTGACCTTGACCTGGAACTCTGGATCgCTGT CCAGTGGTGTGCACACCTTCCCAGCTGTCCTGCAGTCTGACCTCTACACCCTgAGCA GCTCAGTGACTGTAACCTCcAGCACCTGGCCCAGCCAGTCCATCACCTGCAATGTGG CCCACCCGGCAAGCAGCACCAAGGTGGACAAGAAAATTGAGCCCAGAGGGCCCAC AATCAAGCCCTGTCCTCCATGCAAATGCCCAGCACCTAACCTCTTGGGTGGACCATC CGTCTTCATCTTCCCTCCAAAGATCAAGGATGTACTCATGATCTCCCTGAGCCCCAT AGTCACATGTGTGGTGGTGGATGTGAGCGAGGATGACCCAGATGTCCAGATCAGCT GGTTTGTGAACAACGTGGAAGTACACACAGCTCAGACACAAACCCATAGAGAGGAT TACAACAGTACTCTCCGGGTGGTCAGTGCCCTCCCCATCCAGCACCAGGACTGGAT GAGTGGCAAGGAGTTCAAATGCAAGGTCAACAACAAAGACCTCCCAGCGCCCATCG AGAGAACCATCTCAAAACCCAAAGGGTCAGTAAGAGCTCCACAGGTATATGTCTTG CCTCCACCAGAAGAAGAGATGACTAAGAAACAGGTCACTCTGACCTGCATGGTCAC AGACTTCATGCCTGAAGACATTTACGTGGAGTGGACCAACAACGGGAAAACAGAGC TAAACTACAAGAACACTGAACCAGTCCTGGACTCTGATGGTTCTTACTTCATGTACA GCAAGCTGAGAGTGGAAAAGAAGAACTGGGTGGAAAGAAATAGCTACTCCTGTTC AGTGGTCCACGAGGGTCTGCACAATCACCACACGACTAAGAGCTTCTCCCGGACTC CGGGTAAAgtgaaacagactttgaattttgaccttctcaagttggcgggagacgtggagtccaaccctggacctATGAAGTT GCCTGTTAGGCTGTTGGTGCTGATGTTCTGGATCCCTGCTTCCAGCAGTgacattgtgatgtc acagtctccatcctccctggctgtgtcagaaggagagaaggtcactttaacctgcaaatccagtcagagtttgttcaacagtagaacccgaa agaactacttggcttggtaccagcagaaaccagggcagcctcctaaactgttgatctactggacatccactagggaatctggggtccctgat cgcttcacaggcagtggatatgggacagatttcactctcaccatcagcagtgtgcaggctgaagacctggcagtttattactgcaaacaatct tataataatccgtggacgttcggtggaggcaccaagcttgaaataaaaCGGGCTGATGCTGCACCAACTGTATCg ATCTTCCCACCATCCAGTGAGCAGTTAACATCcGGAGGTGCCTCAGTCGTGTGCTTC TTGAACAACTTCTACCCCAAAGACATCAATGTCAAGTGGAAGATTGATGGCAGTGA ACGACAAAATGGCGTCCTGAACAGTTGGACTGATCAGGACAGCAAAGACAGCACCT ACAGCATGAGCAGCACCCTCACGTTGACCAAGGACGAGTATGAACGACATAACAGC TATACCTGTGAGGCCACTCACAAGACATCAACTTCACCCATTGTCAAaAGCTTCAAC AGGAATGAGTGTTAGgcGGCCGCCACTGTGCTGGATATCGTTTAAACCGCTGATCAG CCTCGACTGTGCCTTCTAGTTGCCAGCCATCTGTTGTTTGCCCCTCCCCCGTGCCTTC CTTGACCCTGGAAGGTGCCACTCCCACTGTCCTTTCCTAATAAAATGAGGAAATTGC ATCGCATTGTCTGAGTAGGTGTCATTCTATTCTGGGGGGTGGGGTGGGGCAGGACA GCAAGGGGGAGGATTGGGAAGACAATAGCAGGCATGCTGGGGATGCGGTGGGCTC TATGG
> F2A Self-Cleaving Peptide (SEQ ID NO: 41)
VKQTLNFDLLKLAGDVESNPGP
Table 2. Representative sequences of vectors encoding anti-polyGA antibodies.
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
EQUIVALENTS
While several inventive embodiments have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the function and/or obtaining the results and/or one or more of the advantages described herein, and each of such variations and/or modifications is deemed to be within the scope of the inventive embodiments described herein. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, dimensions, materials, and/or configurations will depend upon the specific application or applications for which the inventive teachings is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific inventive embodiments described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed. Inventive embodiments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, kits, and/or methods, if such features, systems, articles, materials, kits, and/or methods are not mutually inconsistent, is included within the inventive scope of the present disclosure.
All definitions, as defined and used herein, should be understood to control over dictionary definitions, definitions in documents incorporated by reference, and/or ordinary meanings of the defined terms.
All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document. The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.”
The phrase “and/or,” as used herein in the specification and in the claims, should be understood to mean “either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with “and/or” should be construed in the same fashion, i.e., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
As used herein in the specification and in the claims, “or” should be understood to have the same meaning as “and/or” as defined above. For example, when separating items in a list, “or” or “and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of’ or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e., “one or the other but not both”) when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or “exactly one of.” “Consisting essentially of,” when used in the claims, shall have its ordinary meaning as used in the field of patent law.
As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.
It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of’ and “consisting essentially of’ shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03. It should be appreciated that embodiments described in this document using an open-ended transitional phrase (e.g., “comprising”) are also contemplated, in alternative embodiments, as “consisting of’ and “consisting essentially of’ the feature described by the open-ended transitional phrase. For example, if the disclosure describes “a composition comprising A and B”, the disclosure also contemplates the alternative embodiments “a composition consisting of A and B” and “a composition consisting essentially of A and B”.

Claims

CLAIMS What is claimed is:
1. An antibody or antigen-binding fragment thereof, wherein the antibody or antigenbinding fragment comprises a heavy chain variable region (VH) comprising:
(i) a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 1;
(ii) a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 2; and/or
(iii) a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 3.
2. An antibody or antigen-binding fragment thereof, wherein the antibody or antigenbinding fragment comprises a light chain variable region (VL) comprising:
(i) a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 4;
(ii) a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 5; and/or
(iii) a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 6.
3. The antibody or antigen-binding fragment thereof of claim 1 or 2, wherein the antibody or antigen-binding fragment comprises a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 7.
4. The antibody or antigen-binding fragment thereof of any one of claims 1 to 3, wherein the antibody or antigen-binding fragment comprises a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 12.
5. The antibody of antigen-binding fragment thereof of any one of claims 1 to 4, wherein the antibody of antigen-binding fragment comprises a heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 8 or 10.
6. The antibody of antigen-binding fragment thereof of any one of claims 1 to 5, wherein the antibody of antigen-binding fragment comprises a light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 13 or 15.
7. An antibody or antigen-binding fragment thereof, wherein the antibody or antigenbinding fragment comprises a heavy chain variable region (VH) comprising:
(i) a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 20;
(ii) a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 21; and/or
(iii) a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 22.
8. An antibody or antigen-binding fragment thereof, wherein the antibody or antigenbinding fragment comprises a light chain variable region (VL) comprising:
(i) a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 23;
(ii) a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 24; and/or
(iii) a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 25.
9. The antibody or antigen-binding fragment thereof of claim 7 or 8, wherein the antibody or antigen-binding fragment comprises a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 26.
10. The antibody or antigen-binding fragment thereof of any one of claims 7 to 9, wherein the antibody or antigen-binding fragment comprises a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 31.
11. The antibody of antigen-binding fragment thereof of any one of claims 7 to 10, wherein the antibody of antigen-binding fragment comprises a heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 27 or 29.
12. The antibody of antigen-binding fragment thereof of any one of claims 7 to 11, wherein the antibody of antigen-binding fragment comprises a light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 32 or 34.
13. A single chain variable fragment (scFv) comprising a heavy chain variable region comprising:
(i) a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 1;
(ii) a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 2; and/or
(iii) a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 3.
14. A single chain variable fragment (scFv) comprising a light chain variable region comprising:
(i) a CDR1 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 4;
(ii) a CDR2 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 5; and/or
(iii) a CDR3 region comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 6.
15. The scFv of claim 13 or 14, comprising a variable heavy chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 17.
16. The scFv of any one of claims 13 to 15, comprising a variable light chain comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 18.
17. The scFv of any one of claims 13 to 16, comprising a linker molecule connecting the heavy chain variable region to the light chain variable region, optionally wherein the linker molecule comprises a poly-GS linker.
18. The antibody or antigen-binding fragment thereof of any one of claims 1 to 12, or the scFv of any one of claims 13 to 17, further comprising a signal peptide.
19. The antibody or antigen-binding fragment thereof of any one of claims 1 to 12, or the scFv of any one of claims 13 to 17, wherein the antibody or antigen-binding fragment or scFv binds poly(GA) RAN protein.
20. A composition comprising the antibody or antigen-binding fragment thereof of any one of claims 1-12 or 18-19, or the scFv of any one of claims 13 to 17, and a pharmaceutically acceptable carrier or buffer.
21. An isolated nucleic acid encoding the antibody or antigen-binding fragment thereof of any one of claims 1 to 12 or 18 to 19, or the scFv of any one of claims 13 to 17.
22. The isolated nucleic acid of claim 21 comprising the nucleotide sequence set forth in any one of SEQ ID NOs: 36-40.
23. A vector comprising the isolated nucleic acid of claim 21 or 22.
24. The vector of claim 23, wherein the vector is a plasmid.
25. A cell transformed with the isolated nucleic acid of claim 21 or 22, or the vector of claim 23 or 24.
26. The cell of claim 25, wherein the cell is a mammalian cell, optionally wherein the cell is a human cell.
27. A method for administering an antibody or antigen -binding fragment to a subject, the method comprising administering the antibody or antigen-binding fragment of any one of claims 1 to 12 or 18 to 19, or the scFv of any one of claims 13 to 17, to the subject.
28. The method of claim 27, wherein the subject is a mammal, optionally wherein the subject is a human.
29. The method of claim 27 or 28, wherein the subject expresses one or more RAN proteins.
30. The method of any one of claims 27 to 29, wherein the subject expresses a poly(GA) RAN protein.
31. The method of any one of claims 27 to 30, wherein the subject has or is suspected of having Alzheimer’s disease, amyotrophic lateral sclerosis (ALS), frontotemporal dementia, myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), spinocerebellar ataxia (SCA) type 1 (SCA1), SCA2, SCA3, SCA6, SCA7, SCA8, SCAIO, SCA12, SCA17, SCA31, SCA36, spinal bulbar muscular atrophy, dentatorubral-pallidoluysian atrophy (DRPLA), Huntington's disease (HD), Fragile X Tremor Ataxia Syndrome (FXTAS), Fuch's endothelial corneal dystrophy (FECD), Huntington's disease-like 2 syndrome (HDL2), Fragile X syndrome (FXS), a disorder related to 7pl 1 .2 folate- sensitive fragile site FRA7A, a disorder related to folate- sensitive fragile site 2ql 1 FRA2A, or Fragile XE syndrome (FRAXE).
32. The method of claim 31, wherein the subject has or is suspected of having ALS.
33. A method for reducing poly(GA) RAN protein aggregation in a subject, the method comprising administering the antibody or antigen-binding fragment of any one of claims 1 to 12 or 18 to 19, or the scFv of any one of claims 13 to 17, to the subject.
34. The method of claim 33, wherein the subject is a mammal, optionally wherein the subject is a human.
35. The method of claim 33 or 34, wherein the subject expresses one or more RAN proteins.
36. The method of any one of claims 33 to 35, wherein the subject expresses a poly(GA) RAN protein.
37. The method of any one of claims 33 to 36, wherein the subject has or is suspected of having Alzheimer’s disease, amyotrophic lateral sclerosis (ALS), frontotemporal dementia, myotonic dystrophy type 1 (DM1), myotonic dystrophy type 2 (DM2), spinocerebellar ataxia (SCA) type 1 (SCA1), SCA2, SCA3, SCA6, SCA7, SCA8, SCAIO, SCA12, SCA17, SCA31, SCA36, spinal bulbar muscular atrophy, dentatorubral-pallidoluysian atrophy (DRPLA), Huntington's disease (HD), Fragile X Tremor Ataxia Syndrome (FXTAS), Fuch's endothelial corneal dystrophy (FECD), Huntington's disease-like 2 syndrome (HDL2), Fragile X syndrome (FXS), a disorder related to 7pl 1 .2 folate- sensitive fragile site FRA7A, a disorder related to folate- sensitive fragile site 2ql 1 FRA2A, or Fragile XE syndrome (FRAXE).
38. The method of claim 37, wherein the subject has or is suspected of having ALS.
39. A method for treating a subject having ALS, the method comprising administering the antibody or antigen-binding fragment of any one of claims 1 to 12 or 18 to 19, or the scFv of any one of claims 13 to 17, to the subject.
40. The method of claim 39, wherein the subject is a human.
PCT/US2024/038646 2023-07-20 2024-07-19 Engineered ga antibodies for therapy development Pending WO2025019730A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363514643P 2023-07-20 2023-07-20
US63/514,643 2023-07-20

Publications (1)

Publication Number Publication Date
WO2025019730A1 true WO2025019730A1 (en) 2025-01-23

Family

ID=94282678

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/038646 Pending WO2025019730A1 (en) 2023-07-20 2024-07-19 Engineered ga antibodies for therapy development

Country Status (1)

Country Link
WO (1) WO2025019730A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220267776A1 (en) * 2019-07-05 2022-08-25 University Of Florida Research Foundation, Incorporated Methods for treating ran protein-associated neurological diseases
WO2023077153A1 (en) * 2021-11-01 2023-05-04 University Of Florida Research Foundation, Incorporated Poly-ga proteins in alzheimer's disease

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220267776A1 (en) * 2019-07-05 2022-08-25 University Of Florida Research Foundation, Incorporated Methods for treating ran protein-associated neurological diseases
WO2023077153A1 (en) * 2021-11-01 2023-05-04 University Of Florida Research Foundation, Incorporated Poly-ga proteins in alzheimer's disease

Similar Documents

Publication Publication Date Title
US20230381308A1 (en) Neutralizing anti-tl1a monoclonal antibodies
CN113321731B (en) Antibodies that bind to human programmed death ligand 1 (PD-L1)
US20220267776A1 (en) Methods for treating ran protein-associated neurological diseases
KR101462307B1 (en) Anti-hepcidin-25 selective antibodies and uses thereof
JP2020531045A (en) Anti-CD166 antibody and its use
CA3007115A1 (en) Antibodies targeting fc receptor-like 5 and methods of use
TW201417830A (en) Human IL-23 antigen binding protein
US20220119546A1 (en) Plectin-1 binding antibodies and uses thereof
US20220153863A1 (en) Prame binding molecules and uses thereof
JP2021521896A (en) Human-derived anti- (polyGA) dipeptide repeat (DPR) antibody
WO2025019730A1 (en) Engineered ga antibodies for therapy development
EP4427049A1 (en) Poly-ga proteins in alzheimer's disease
US20230250191A1 (en) Anti-ige engineered antibody and application thereof
WO2025146161A1 (en) Antibody, antigen-binding fragment and medical use thereof
EP4137512A1 (en) Anti-gm2ap antibody and applications thereof
WO2025128551A1 (en) Aav-based anti-ga therapy
HK40088459A (en) Anti-gm2ap antibody and applications thereof
EP4210751A1 (en) Anti-ceruloplasmin antibodies and uses thereof

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: 24843997

Country of ref document: EP

Kind code of ref document: A1