WO2023021319A1 - Monoclonal antibodies against her2/neu and uses thereof - Google Patents

Monoclonal antibodies against her2/neu and uses thereof Download PDF

Info

Publication number
WO2023021319A1
WO2023021319A1 PCT/IB2021/057644 IB2021057644W WO2023021319A1 WO 2023021319 A1 WO2023021319 A1 WO 2023021319A1 IB 2021057644 W IB2021057644 W IB 2021057644W WO 2023021319 A1 WO2023021319 A1 WO 2023021319A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
antigen
binding fragment
cancer
seq
Prior art date
Application number
PCT/IB2021/057644
Other languages
French (fr)
Inventor
Ragupathy Madiyalakan
Thomas Woo
Original Assignee
Oncoquest Pharmaceuticals 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 Oncoquest Pharmaceuticals Inc. filed Critical Oncoquest Pharmaceuticals Inc.
Priority to PCT/IB2021/057644 priority Critical patent/WO2023021319A1/en
Publication of WO2023021319A1 publication Critical patent/WO2023021319A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the subject matter disclosed generally relates to monoclonal antibodies against Her2/Neu. More specifically, the subject matter relates to monoclonal antibodies against Her2/Neu that do not self-aggregate and methods of using the same.
  • Targeted therapy for Her2/neu expressing tumors may be targeted with monoclonal antibodies, such as antibodies comprising the variable regions of the scFv C6MH3-B1 specific for HER2/neu.
  • monoclonal antibodies such as antibodies comprising the variable regions of the scFv C6MH3-B1 specific for HER2/neu.
  • antibodies can suffer from low production yield, and could be further affected by numerous issues such as self- aggregation, oxidation, deamination and isomerization of amino acid chains.
  • only small quantities of these antibodies can be prepared, which precludes large scale preparation for practical therapeutic application developments. [0003] Therefore, there is a need for alternative monoclonal antibodies specific for HER2/neu that at least do not self-aggregate, improve their manufacturability and allow their preparation on a scale that allows their use as therapeutics.
  • an antibody or an antigen-binding fragment that binds specifically to Her2/Neu comprising three variable heavy domain complementarity determining regions (CDR) (CDR H1, H2 and H3) wherein the CDR H1, H2, and H3, comprise an amino acid sequence comprising: CDR H1: GYSFTSYW (SEQ ID NO:1), CDR H2: IYPGX 1 X 2 DT, where X 1 is D, E, or Q, X 2 is S, I, or T, and wherein when X 1 is D, X 2 is different than S (SEQ ID NO:2), and CDR H3: ARHDVGYCTDRTCAKWPEY (SEQ ID NO:3), respectively.
  • the antibody or an antigen-binding fragment may comprise three variable light domain CDR (CDR L1, L2 and L3), wherein the CDR L1, L2, and L3 comprise an amino acid sequence comprising: CDR L1: SSNIGNNY (SEQ ID NO:4), CDR L2: DHT (SEQ ID NO:5), and CDR L3: ASWDYTLSGWV (SEQ ID NO:6), respectively.
  • CDR L1 SSNIGNNY
  • CDR L2 DHT
  • CDR L3 ASWDYTLSGWV
  • X 1 may be D
  • X 2 may be I, or T.
  • X 2 In the antibody or antigen-binding fragment, wherein when X 1 is E, or Q, X 2 is S.
  • X 1 may be E and X 2 may be S.
  • X 1 may be Q and X 2 may be S.
  • X 1 may be D and X 2 may be I.
  • X 1 may be D and X 2 may be T.
  • the antibody or antigen-binding fragment may further comprise four variable heavy domain framework regions (HFR)(HFR 1, 2, 3 and 4), wherein said HFR 1, 2, 3, and 4 comprise an amino acid sequence comprising: HFR 1: VQLVQSGAEVKKPGESLKISCKGS (SEQ ID NO: 15), HFR 2: IAWVRQMPGKGLEYMGL (SEQ ID NO: 16), HFR 3: KYSPSFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFC (SEQ ID NO: 17), and HFR 4: WGQGTLVTV (SEQ ID NO: 18).
  • HFR 1 VQLVQSGAEVKKPGESLKISCKGS
  • HFR 2 IAWVRQMPGKGLEYMGL
  • HFR 3 KYSPSFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFC
  • HFR 4 WGQGTLVTV
  • the antibody or antigen-binding fragment may further comprise four variable light domain framework regions (LFR)(LFR 1, 2, 3 and 4), wherein said LFR 1, 2, 3, and 4 comprise an amino acid sequence comprising: LFR 1: QSVLTQPPSVSAAPGQKVTISCSGS (SEQ ID NO: 84), LFR 2: VSWYQQLPGTAPKLLIY (SEQ ID NO: 85), LFR 3: NRPAGVPDRFSGSKSGTSASLAISGFRSEDEADYYC (SEQ ID NO: 86), and LFR 4: FGGGTKVTVL (SEQ ID NO: 87).
  • LFR 1 QSVLTQPPSVSAAPGQKVTISCSGS
  • LFR 2 VSWYQQLPGTAPKLLIY
  • LFR 3 NRPAGVPDRFSGSKSGTSASLAISGFRSEDEADYYC
  • LFR 4 FGGGTKVTVL (SEQ ID NO: 87).
  • the antibody or antigen-binding fragment may comprise a variable heavy domain (V H ) comprising amino acid sequence comprising: VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSP SFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQ GTLVTV (SEQ ID NO: 23).
  • the antibody or antigen binding fragment may comprise a variable light domain (V L ) comprising amino acid sequence comprising: QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPDRFSGS KSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (SEQ ID NO: 83).
  • V L variable light domain
  • the antibody or antigen-binding fragment may comprise a variable heavy domain (V H ) comprising amino acid sequence comprising: VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSP SFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQ GTLVTV (SEQ ID NO: 23) and a variable light domain (V L ) comprising amino acid sequence comprising: QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPD RFSGSKSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (SEQ ID NO: 83).
  • V H variable heavy domain
  • V L variable light domain
  • the antigen-binding fragment may be a single-domain antibody (sdAb), a fragment antigen binding (Fab), a single-chain variable fragment (scFv), or a single-chain fragment antigen binding (scFab).
  • the antibody or antigen-binding fragment may be an IgA, an IgD, an IgE, an IgG, or an IgM.
  • the antibody or antigen-binding fragment may be an IgE.
  • the antibody or antigen-binding fragment may be humanized or partially humanized.
  • a compound comprising the antibody or antigen-binding fragment of the present invention, and a functional moiety.
  • the antibody or antigen-binding fragment may be linked to the functional moiety via a peptide linker.
  • the antibody or antigen-binding fragment may be functionally linked to the functional moiety via the peptide linker.
  • the peptide linker may comprise about 3 to about 40 amino acid residues.
  • the peptide linker may comprise the amino acid sequence (GGGGS)n or (GGGS)n, wherein n ⁇ 1.
  • the antibody or antigen-binding fragment may be fused to a second antibody or antigen-binding fragment operable to bind a target epitope.
  • the antibody or antigen-binding fragment may be linked to a peptide, a polypeptide, a protein, an enzyme, a second antibody, an antibody fragment, a second antigen-binding fragment or a combination of any two or more thereof; wherein each of the antibody or antigen-binding fragment thereof and the linked peptide, polypeptide, protein, enzyme, second antibody, antibody fragment, second antigen-binding fragment, or the combination of any two or more thereof is functional.
  • the antibody fragment may be a fragment crystallizable (Fc) region.
  • composition comprising the antibody or antigen-binding fragment of the present invention or the compound of the present invention, and a pharmaceutically acceptable diluent, carrier or excipient.
  • a nucleic acid molecule encoding an antibody or antigen-binding fragment of the present invention, or encoding a compound of the present invention.
  • a vector comprising the nucleic acid molecule of the present invention operably linked to one or more regulatory elements to allow expression of the antibody or antigen-binding fragment the present invention, or the compound of the present invention in a host cell.
  • a cell comprising the vector of the present invention for expressing the antibody or antigen-binding fragment thereof of the present invention or the compound of the present invention.
  • a method of treating a Her2/Neu associated disease comprising: administering the antibody or antigen-binding fragment of the present invention, the compound of the present invention, the composition of the present invention, or a combination of any two or more thereof to a subject in need thereof.
  • the Her2/Neu associated disease may be a cancer.
  • the cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
  • the antibody or antigen- binding fragment of the present invention, the compound of the present invention, the composition of the present invention, or a combination of any two or more thereof to a subject in need thereof in the treatment of a Her2/Neu associated disease.
  • the Her2/Neu associated disease may be a cancer.
  • the cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
  • the antibody or antigen-binding fragment, compound, composition, or combination of any two or more thereof of the present invention, wherein the Her2/Neu associated disease may be a cancer.
  • the antibody or antigen-binding fragment, compound, composition, or combination of any two or more thereof the present invention wherein the cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
  • the cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
  • the antibody or antigen-binding fragment of the present invention, the compound of the present invention, the composition of the present invention, or a combination of any two or more thereof to a subject in need thereof in the manufacture of a medicament for the treatment of a Her2/Neu associated disease.
  • the Her2/Neu associated disease may be a cancer.
  • the cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
  • Fig.1A shows an apparent affinity non-linear regression curve comparing the binding on D2F2/E2 cells of parental IgE C6MH3-B1 antibody versus variant 1.
  • V1, P1 Peak 1 antibodies are used at dose -4 (0.0002 nM), -3 (0.002 nM), -2 (0.02 nM), - 1 (0.2 nM), 0 (2 nM), 1 (20 nM), and 2 (200 nM).
  • Fig.1B shows an apparent affinity non-linear regression curve comparing the binding on D2F2/E2 cells of parental IgE C6MH3-B1 antibody versus variant 2.
  • Peak 1 (V2, P1) antibodies are used at dose -4 (0.0002 nM), -3 (0.002 nM), -2 (0.02 nM), - 1 (0.2 nM), 0 (2 nM), 1 (20 nM), and 2 (200 nM).
  • Fig.1C shows an apparent affinity non-linear regression curve comparing the binding on D2F2/E2 cells of parental IgE C6MH3-B1 antibody versus variant 4.
  • Fig.1D shows an apparent affinity non-linear regression curve comparing the binding on D2F2/E2 cells of parental IgE C6MH3-B1 antibody versus variant 5.
  • V5, P1 Peak 1 antibodies are used at dose -4 (0.0001 nM), -3 (0.001 nM), -2 (0.01 nM), - 1 (0.1 nM), 0 (1 nM), 1 (10 nM), and 2 (100 nM).
  • V5, P1 Peak 1 antibodies
  • -4 0.0001 nM
  • -3 0.001 nM
  • -2 0.01 nM
  • - 1 0.1 nM
  • 1 (10 nM) 10 nM
  • 2 100 nM
  • the three variable heavy domain complementarity determining regions may comprise the following amino acid sequences: 1) CDR H1: GYSFTSYW (SEQ ID NO: 1), 2) CDR H2: IYPGX 1 X 2 DT, where X 1 is D, E, or Q, X 2 is S, I, or T, and wherein when X 1 is D, X 2 is different than S (SEQ ID NO: 2), and 3) CDR H3: ARHDVGYCTDRTCAKWPEY (SEQ ID NO: 3).
  • the antibody or an antigen-binding fragment may also comprise three variable light domain CDR (CDR L1, L2 and L3).
  • the three variable light domain CDR may comprise the following amino acid sequences: 1) CDR L1: SSNIGNNY (SEQ ID NO: 4), 2) CDR L2: DHT (SEQ ID NO: 5), and 3) CDR L3: ASWDYTLSGWV (SEQ ID NO: 6).
  • the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein when X 1 is D, X 2 is I, or T (e.g. as per SEQ ID NOs: 65 and 56, respectively).
  • the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein when X 1 is E, or Q, X 2 is S (e.g. as per SEQ ID NOs: 29, and 38, respectively).
  • the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein X 1 is E and X 2 is S (e.g. as per SEQ ID NO: 29).
  • the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein X 1 is Q and X 2 is S (e.g. as per SEQ ID NO: 38).
  • the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein X 1 is D and X 2 is I (e.g. as per SEQ ID NO: 65).
  • the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein X 1 is D and X 2 is T (e.g. as per SEQ ID NO: 56).
  • the antibody or antigen-binding fragment of the present invention may be an antibody comprising four variable heavy domain framework regions (HFR 1 to HFR4) and three variable heavy domain complementarity determining regions (CDR H1, CDR H2 and CDR H3), comprising amino acid sequence comprising : VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 23), VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGQSDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO:
  • the antibody or antigen-binding fragment of the present invention may be an antibody comprising variable heavy domain (V H ) comprising amino acid sequences comprising according to SEQ ID NOs: 22, 31, 49 or 58, respectively, that binds specifically to a Her2/Neu. It is believed to be operable to neutralize Her2/Neu, including, but not limited to a Her2/Neu family member from human, monkey, rat, or mouse.
  • V H variable heavy domain
  • the antibody or antigen-binding fragment of the present invention may be an antibody comprising four variable light domain framework regions (LFR1 to LFR4) and three variable light domain complementarity determining regions (CDR L1, CDR L2 and CDR L3), comprising amino acid sequence comprising : QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPDRFSGS KSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (as per each of SEQ ID NOs: 83, 92, 101, 110 or 119), that binds specifically to a Her2/Neu.
  • the antibody or antigen-binding fragment of the present invention may be an antibody comprising variable light domain (V L ) comprising amino acid sequences according to SEQ ID NOs: 82, 91, 100, 109 or 118, respectively, that binds specifically to a Her2/Neu. It is believed to be operable to neutralize Her2/Neu, including, but not limited to a Her2/Neu family member from human, monkey, rat, or mouse.
  • V L variable light domain
  • the antibody or antigen binding fragment thereof of the present invention may further comprise four variable heavy domain framework regions (HFR)(HFR 1, 2, 3 and 4), which comprise an amino acid sequence comprising: HFR 1: VQLVQSGAEVKKPGESLKISCKGS (SEQ ID NOs: 15, 24, 33, 42 or 51), HFR 2: IAWVRQMPGKGLEYMGL (SEQ ID NOs: 16, 25, 34, 43 or 52), HFR 3: KYSPSFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFC (SEQ ID NOs: 17, 26, 35, 44 or 53), and HFR 4: WGQGTLVTV (SEQ ID NOs: 18, 27, 36, 45 or 54).
  • HFR 1 VQLVQSGAEVKKPGESLKISCKGS
  • HFR 2 IAWVRQMPGKGLEYMGL (SEQ ID NOs: 16, 25, 34, 43 or 52)
  • HFR 3 KYSPSFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFC
  • the antibody or antigen binding fragment thereof of the present invention may further comprise four variable light domain framework regions (LFR)(LFR 1, 2, 3 and 4) which comprise an amino acid sequence comprising: LFR 1: QSVLTQPPSVSAAPGQKVTISCSGS (SEQ ID NOs: 84, 93, 102, 111, or 120), LFR 2: VSWYQQLPGTAPKLLIY (SEQ ID NOs: 85, 94, 103, 112, or 121), LFR 3: NRPAGVPDRFSGSKSGTSASLAISGFRSEDEADYYC (SEQ ID NOs: 86, 95, 104, 113 or 122), and LFR 4: FGGGTKVTVL (SEQ ID NOs: 87, 96, 105, 114, or 123).
  • LFR 1 QSVLTQPPSVSAAPGQKVTISCSGS
  • LFR 2 VSWYQQLPGTAPKLLIY
  • LFR 3 NRPAGVPDRFSGSKSGTSASLAIS
  • the antibody or antigen binding fragment thereof of the present invention may comprise a variable heavy domain (V H ) comprising amino acid sequence comprising: VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 23), and a variable light domain (V L ) comprising amino acid sequence comprising: QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPDRFSGS KSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (SEQ ID NO: 83).
  • V H variable heavy domain
  • V L Her2/Neu specific heavy variable heavy domain
  • FRs Framework regions
  • CDRs complementarity-determining regions
  • Antibody 0 represent the starting antibody and 1 to 5 the mutated sequences.
  • Underlined amino acid sequence of variable region from FR1 to FR4.
  • V L Her2/Neu specific light variable heavy domain
  • FRs Framework regions
  • CDRs complementarity-determining regions
  • Antibody 0 represent the starting antibody and 1 to 5 the mutated sequences.
  • Underlined amino acid sequence of variable region from FR1 to FR4.
  • the dash ( - ) character is only illustrative of a space in the text to identify the CDR2, and not of a CDR2 feature.
  • the antibody or an antigen-binding fragment of the present invention may have sequences substantially identical to the sequences disclosed above, operable to bind to Her2/Neu.
  • a substantially identical sequence may comprise one or more conservative amino acid mutations.
  • one or more conservative amino acid mutations may yield a mutant peptide with no substantial change in physiological, chemical, physico-chemical or functional properties compared to the reference sequence. In such a case, the reference and mutant sequences would be considered “substantially identical” polypeptides.
  • a conservative amino acid substitution is defined herein as the substitution of an amino acid residue for another amino acid residue with similar chemical properties (e.g. size, charge, or polarity).
  • one or more conservative amino acid mutations may be made to one or more framework regions of the sdAb while maintaining both the CDR sequences listed above and the overall structure of the CDR of the antibody or antigen-binding fragment; thus the specificity and binding of the antibody are maintained.
  • one or more conservative amino acid mutations may be made to one or more framework regions of the sdAb and to a CDR sequence listed above while maintaining the antigen- binding function of the overall structure of the CDR of the antibody or antigen-binding fragment; thus the specificity and binding of the antibody are maintained.
  • a conservative mutation may be a conservative amino acid substitution.
  • Such a conservative amino acid substitution may substitute a basic, neutral, hydrophobic, or acidic amino acid for another amino acid of the same group.
  • basic amino acid it is meant a hydrophilic amino acid having a side chain pK value of greater than 7, which is typically positively charged at physiological pH.
  • Basic amino acids include histidine (His or H), arginine (Arg or R), and lysine (Lys or K).
  • neutral amino acid also “polar amino acid”
  • Polar amino acids include serine (Ser or S), threonine (Thr or T), cysteine (Cys or C), tyrosine (Tyr or Y), asparagine (Asn or N), and glutamine (GIn or Q).
  • hydrophobic amino acid (also “non-polar amino acid”) it is meant an amino acid exhibiting a hydrophobicity of greater than zero according to the normalized consensus hydrophobicity scale of Eisenberg (1984). Hydrophobic amino acids include proline (Pro or P), isoleucine (Ile or I), phenylalanine (Phe or F), valine (Val or V), leucine (Leu or L), tryptophan (Trp or W), methionine (Met or M), alanine (Ala or A), and glycine (Gly or G). “Acidic amino acid” refers to a hydrophilic amino acid having a side chain pK value of less than 7, which is typically negatively charged at physiological pH.
  • Acidic amino acids include glutamate (Glu or E) and aspartate (Asp or D).
  • Sequence identity is used to evaluate the similarity of two sequences. It is determined by calculating the percentage of residues that are the same when the two sequences are aligned for maximum correspondence between residue positions. Any known method may be used to calculate sequence identity; for example, computer software is available to calculate sequence identity. Without wishing to be limiting, sequence identity can be calculated by software such as NCBI BLAST2 service maintained by the Swiss Institute of Bioinformatics (and as found at ca.expasy.org/tools/blast/), BLAST-P, Blast-N, or FASTA-N, or any other appropriate software that is known in the art.
  • the substantially identical sequences of the present invention may be at least 90% identical; in another example, the substantially identical sequences may be at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical, or any percentage therebetween, at the amino acid level to sequences described herein.
  • a substantially identical sequence retains the activity and specificity of the reference sequence.
  • the difference in sequence identity may be due to one or more conservative amino acid mutations.
  • the present invention may be directed to an antibody or antigen-binding fragment comprising a sequence at least 95%, at least 98%, or at least 99% identical to that of one or more of the antibodies or antigen- binding fragments described herein.
  • the antibody or an antigen-binding fragment of the present invention may be used, for example, to bind specifically to a Her2/Neu. It is believed to neutralize the Her2/Neu receptors on the surface of cancer cells and blocking them from receiving growth signals.
  • the expression “bind specifically to a Her2/Neu” or “bind specifically to Her2/Neu” is intended to mean that the antibody or antigen-binding fragment of the present invention is enabled to bind specifically to Her2/Neu and particularly to one or more human, rhesus, mouse or rat serum Her2/Neu.
  • the term “antibody”, also referred to in the art as “immunoglobulin” (Ig), as used herein refers to a protein constructed from paired heavy and light polypeptide chains; various Ig isotypes exist, including IgA, IgD, IgE, IgG, and IgM.
  • each chain fold When an antibody is correctly folded, each chain folds into a number of distinct globular domains joined by more linear polypeptide sequences.
  • V L variable
  • C L constant
  • V H variable
  • C H 1 constant
  • C H 2 constant
  • Fv antigen binding region
  • variable regions show less sequence diversity, and are responsible for binding a number of natural proteins to elicit important biochemical events.
  • the variable region of an antibody contains the antigen-binding determinants of the molecule, and thus determines the specificity of an antibody for its target antigen.
  • the majority of sequence variability occurs in six hypervariable regions, three each per variable heavy (V H ) and light (V L ) chain; the hypervariable regions combine to form the antigen-binding site, and contribute to binding and recognition of an antigenic determinant.
  • the specificity and affinity of an antibody for its antigen is determined by the structure of the hypervariable regions, as well as their size, shape, and chemistry of the surface they present to the antigen.
  • an “antibody fragment” or “antigen-binding fragment” as referred to herein may include any suitable antigen-binding antibody fragment known in the art.
  • the antibody fragment may be a naturally-occurring antibody fragment, or it may be a non-naturally occurring antibody fragment obtained, for example, by manipulation of a naturally-occurring antibody or by recombinant methods.
  • an antibody fragment may include, but is not limited to, a Fv, a single-chain Fv (scFv; a molecule consisting of V L and V H connected with a peptide linker), a Fab, a F(ab’)2, or a single- domain antibody (sdAb; a fragment composed of a single V L or V H or a V H H), or a multivalent presentation of any of these.
  • Antibody fragments such as those just described may require one or more linker sequences, disulfide bonds, or other types of covalent bond to link different portions of the fragments. Those of skill in the art will be familiar with the requirements of the different types of fragments and various approaches for their construction.
  • the antigen-binding fragment of the present invention may be a sdAb derived from a naturally-occurring source.
  • Heavy chain antibodies of camelid origin (Hamers-Casterman et al, 1993) lack light chains and thus their antigen binding sites consist of one domain, termed V H H.
  • SdAbs have also been observed in shark and are termed VNAR (Nuttall et al, 2003).
  • Other sdAbs may be engineered based on human Ig heavy and light chain sequences (Jespers et al, 2004; To et al, 2005).
  • the term “sdAb” includes an sdAb directly isolated from a V H , V H H, V L , or V NAR reservoir of any origin through phage display or other technology, an sdAb derived from the aforementioned sdAb, a recombinantly produced sdAb, as well as an sdAb generated through further modification of such sdAb by humanization, affinity maturation, stabilization, solubilization, camelization, or other methods of antibody engineering. Also encompassed by the present invention are homologues, derivatives, or fragments that retain the antigen-binding function and specificity of the sdAb.
  • SdAbs possess desirable properties for antibody molecules, such as high thermostability, high detergent resistance, relatively high resistance to proteases (Dumoulin et al, 2002) and high production yield (Arbabi-Ghahroudi et al, 1997). They can also be engineered to have very high affinity by isolation from an immune library (Li et al, 2009) or by in vitro affinity maturation (Davies & Riechmann, 1996). Further modifications to increase stability, such as the introduction of one or more non-canonical disulfide bonds (Hussack et al, 2011a,b; Kim et al, 2012), may also be brought to the sdAb.
  • An sdAb comprises a single immunoglobulin domain that retains the immunoglobulin fold; most notably, only three CDR/hypervariable loops form the antigen-binding site.
  • not all CDRs may be required for binding the antigen.
  • one, two, or three of the CDRs may contribute to binding and recognition of the antigen by the sdAb of the present invention.
  • the CDRs of the sdAb or variable domain are referred to herein as CDR1, CDR2, and CDR3.
  • the present invention further encompasses an antibody or an antigen-binding fragment that is “humanized” using any suitable method known in the art, such as, but not limited to, CDR grafting or veneering. Humanization of an antibody or an antigen-binding fragment comprises replacing an amino acid in the antibody or antigen-binding fragment sequence with its human counterpart, as found in the human consensus sequence, without substantial loss of antigen-binding ability or specificity; this approach reduces immunogenicity of the antibody or antigen-binding fragment when introduced into human subjects.
  • one or more than one of the CDRs defined herein may be fused or grafted to a human variable region (V H , or V L ), to a human antibody (IgA, IgD, IgE, IgG, and IgM), to a human antibody fragment framework region (Fv, scFv, Fab) or to another protein of similar size and nature onto which a CDR can be grafted (Nicaise et al, 2004).
  • CDR grafting is known in the art and is described in at least the following: US Patent No.6180370, US Patent No.5693761, US Patent No.6054297, US Patent No. 5859205, and European Patent No.626390.
  • Veneering also referred to in the art as “variable region resurfacing”, involves humanizing solvent-exposed positions of an antibody or antigen-binding fragment; thus, preserving buried non-humanized residues, which may be important for CDR conformation, while minimizing the potential for immunological reaction against solvent-exposed regions. Veneering is known in the art and is described in at least the following: US Patent No. 5869619, US Patent No. 5766886, US Patent No. 5821123, and European Patent No. 519596. Persons of skill in the art would also be amply familiar with methods of preparing such humanized antibody fragments and humanizing amino acid positions.
  • the antibody or antigen-binding fragment according to the present invention may comprise an additional sequence to aid in expression, detection or purification of the antibody or antigen-binding fragment.
  • Any such sequence or tag known to those of skill in the art may be used.
  • the antibody or antigen-binding fragment may comprise a targeting or signal sequence (such as, but not limited to, ompA or pelB), a detection/purification tag (such as, but not limited to, c-Myc, HA, His5, or His6), or a combination of any two or more thereof.
  • the additional sequence may be a biotin recognition site, such as that described by Cronan et al.
  • a linker sequence may be used in conjunction with the additional sequence or tag, or may serve as a detection/purification tag.
  • a linker sequence may be linked to a functional moiety, optionally by a linker sequence.
  • the antibody or antigen-binding fragment may be linked to the functional moiety via a linker (also known as a linker sequence).
  • linker sequence is intended to mean a short (typically 40 amino acids or fewer) peptide sequence that is introduced between protein domains. Linker sequences are often composed of flexible residues such as glycine and serine so that the linked protein domains are free to move relative to one another.
  • the linker sequence can be any linker sequence known in the art that would allow for the antibody and the functional moiety of the present invention to be operably linked for the desired function.
  • the linker may be any sequence known in the art (either a natural or synthetic linker) that allows for an operable fusion comprising an antibody or antigen-binding fragment linked to a polypeptide (e.g. the functional moiety).
  • the linker sequence may be a linker sequence L such as (GGGS)n and (GGGGS)n, wherein n is equal to or greater than 1, or from about 1 to about 5, or from about 1 to 15; or n may be any number that would allow for the operability of the compound of the present invention.
  • the linker may be an amino acid sequence, for example, an amino acid sequence that comprises about 1 to about 40 amino acids, or about 3 to about 40 amino acids, or about 5 to about 40 amino acids, or about 10 to about 40 amino acids, or about 15 to about 40 amino acids, or about 20 to about 40 amino acids, or about 25 to about 40 amino acids, or about 30 to about 40 amino acids, or about 35 to about 40 amino acids, or about 3 to about 35 amino acids, or about 5 to about 35 amino acids, or about 10 to about 35 amino acids, or about 15 to about 35 amino acids, or about 20 to about 35 amino acids, or about 25 to about 35 amino acids, or about 30 to about 35 amino acids, or about 3 to about 30 amino acids, or about 5 to about 30 amino acids, or about 10 to about 30 amino acids, or about 15 to about 30 amino acids, or about 20 to about 30 amino acids, or about 25 to about 30 amino acids, or about 3 to about 25 amino acids, or about 5 to about 25 amino acids, or about 10 to about 25 amino acids, or about 15 to about 25 amino acids, or about
  • the term “functional moiety” is intended to mean a part of the compound having an activity, purpose, or task; relating to the way in which the compound is intended to work or operate.
  • the functional moiety may be linked to the antibody or antigen- binding fragment, for example, through a chemical link pursuant to a chemical reaction, and/or through fusion of the antibody or antigen-binding fragment with the functional moiety, obtained for example using recombinant DNA technology.
  • the antibody or antigen- binding fragment of the compound may be fused to a peptide, a polypeptide (e.g.
  • growth factor CIBP2 an antimicrobial cyclic peptide
  • a protein an enzyme [such as iduronate-2-sulfatase (IDS), acid beta-glucosidase (GCase), a serine protease, a growth factor, etc.], another (or the same) antibody or a fragment operable to bind a target epitope (e.g.
  • IDS iduronate-2-sulfatase
  • GCase acid beta-glucosidase
  • serine protease a growth factor, etc.
  • another (or the same) antibody or a fragment operable to bind a target epitope e.g.
  • an anti-microbial antibody an anti-inflammatory antibody, an intrabody, a BBB-crossing antibody, a neurodegeneration target antibody, an ion channel targeting antibody, a cancer associated antigen antibody, a checkpoint inhibitor targeting antibody, or a GPCR targeting antibody
  • an anti-microbial antibody an anti-microbial antibody, an anti-inflammatory antibody, an intrabody, a BBB-crossing antibody, a neurodegeneration target antibody, an ion channel targeting antibody, a cancer associated antigen antibody, a checkpoint inhibitor targeting antibody, or a GPCR targeting antibody
  • both the antibody or antigen-binding fragment and the rest of the compound i.e. the functional moiety
  • the compound may be fused to a second antibody or antigen-binding fragment, operable to bind a target epitope, which may be the same as, or distinct from the epitope of the antibody or antigen-binding fragment of the present invention.
  • the antibody or antigen-binding fragment of the present invention may also be in a multivalent display format, also referred to herein as multivalent presentation. Multimerization may be achieved by any suitable method known in the art.
  • multimerization may be achieved using self-assembly molecules such as those described in Zhang et al (2004a; 2004b) and W02003/046560, where pentabodies are produced by expressing a fusion protein comprising the antibody or antigen-binding fragment of the present invention and the pentamerization domain of the B-subunit of an AB5 toxin family (Merritt & Hol, 1995).
  • a multimer may also be formed using the multimerization domains described by Zhu et al.
  • antibody form is a fusion of the antibody or fragment of the present invention with a coiled-coil peptide resulting in a multimeric molecule (Zhu et al., 2010).
  • Other forms of multivalent display are also encompassed by the present invention.
  • the antibody or antigen-binding fragment may be presented as a dimer, a trimer, or any other suitable oligomer.
  • Another method known in the art for multimerization is to dimerize the antibody or antigen-binding fragment using an Fc domain, such as, but not limited to a human Fc domain.
  • the Fc domain may be selected from various classes including, but not limited to, IgG, IgM, or various subclasses including, but not limited to IgG1, IgG2, etc.
  • a multivalent display format may encompass a chimeric or humanized format of V H H of the present invention linked to an Fc domain, or bi or tri-specific antibody fusions with two or three V H Hs recognizing unique epitopes.
  • the Fc domain in the multimeric complex as just described may be any suitable Fc fragment known in the art.
  • the Fc fragment may be from any suitable source; for example, the Fc fragment may be of mouse or human origin.
  • the Fc fragment may be a mouse Fc2b fragment or a human Fc1 fragment (Bell et al, 2010; lqbal et al, 2010).
  • the Fc fragment may be fused to the N-terminal or C-terminal end of the V H H or humanized version of the present invention.
  • Each subunit of the multimers described above may comprise the same or different antibodies or antigen-binding fragments of the present invention, which may have the same or different specificity.
  • the multimerization domains may be linked to the antibody or antigen-binding fragment using a linker, as required; such a linker should be of sufficient length and appropriate composition to provide flexible attachment of the two molecules but should not hamper the antigen-binding properties of the antibody or antigen-binding fragment.
  • the linker sequence can be any linker known in the art that would allow for the compound of the present invention to be prepared and be operable for the desired function.
  • the present invention also encompasses a composition comprising one or more than one compound as described herein.
  • the composition may comprise a single antibody and/or compound as described above, or the composition may comprise a mixture of antibody and/or compounds. Furthermore, in a composition comprising a mixture of antibody and/or compounds of the present invention, the antibody and/or compounds may have the same specificity, or they may differ in their specificities; for example, and without wishing to be limiting in any manner, the composition may comprise antibody and/or compounds specific to Her2/Neu (same or different epitope). [0083] A composition according to the invention may also comprise a pharmaceutically acceptable diluent, excipient, or carrier.
  • the diluent, excipient, or carrier may be any suitable diluent, excipient, or carrier known in the art that is compatible with other ingredients in the composition, that is compatible with the method of delivery of the composition, and that is not deleterious to the recipient of the composition.
  • the composition may be in any suitable form; for example, the composition may be provided in suspension form, powder form (such as, but not limited to, lyophilised or encapsulated), capsule form or tablet form.
  • the carrier when the composition is provided in suspension form, may comprise water, saline, or a suitable buffer, and optionally comprise one or more additives to improve solubility and/or stability.
  • Reconstitution to produce a suspension may be effected in a buffer at a suitable pH to ensure the viability of the antibody or antigen-binding fragment.
  • Dry powders may also include additives to improve stability and/or carriers to increase bulk/volume; for example, and without wishing to be limiting, the dry powder composition may comprise sucrose or trehalose.
  • the composition may be formulated for delivery of the antibody or antigen- binding fragment to the gastrointestinal tract of the subject.
  • the composition may comprise encapsulation, time release, or other suitable technologies for delivery of the antibody and/or compound of the present invention. It would be within the competency of a person of skill in the art to prepare suitable compositions comprising the present antibody and/or compound.
  • the invention also encompasses a nucleic acid molecule comprising a nucleotide sequence encoding an antibody, antigen-binding fragment, or compound of the present invention.
  • the invention further comprises a vector comprising the nucleic acid molecule; a cell comprising the vector, for expressing the antibody, antigen-binding fragment, or compound of the present invention, and a cell for expressing the antibody, antigen-binding fragment, or compound of the present invention.
  • a method of treating a Her2/Neu associated disease comprising administering the antibody or antigen-binding fragment of the present invention, the compound of the present invention, the composition of the present invention or a combination of any two or more thereof to a subject in need thereof.
  • the antibody or antigen- binding fragment of the present invention there is provided a use of the antibody or antigen- binding fragment of the present invention, the compound of the present invention, the composition of the present invention or a combination of any two or more thereof for the treatment of a Her2/Neu associated disease.
  • the expression “Her2/Neu associated disease” is intended to mean that the disease involves, is connected to, Her2/Neu as an etiological factor.
  • the disease is a cancer.
  • Non-limiting examples in which Her2/Neu may be involved include ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
  • the method of the present invention may further comprise administering a second therapeutic agent comprising at least one of a cytotoxic agent, an additional antibody or a therapeutically active fragment thereof, or a chemotherapy regimen.
  • a second therapeutic agent comprising at least one of a cytotoxic agent, an additional antibody or a therapeutically active fragment thereof, or a chemotherapy regimen.
  • the cytotoxic therapeutic agents include, but are not limited to, angiogenesis inhibitors, antiproliferative agents, kinase inhibitors, receptor tyrosine kinase inhibitors, aurora kinase inhibitors, polo-like kinase inhibitors, bcr-abl kinase inhibitors, growth factor inhibitors, COX-2 inhibitors, non-steroidal anti- inflammatory drugs (NSAIDS), antimitotic agents, alkylating agents, antimetabolites, intercalating antibiotics, platinum containing agents, growth factor inhibitors, ionizing radiation, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biologic response modifiers, immunologicals, antibodies, hormonal therapies, retinoids/deltoids plant alkaloids, proteasome inhibitors, HSP-90 inhibitors, histone deacetylase inhibitors (HDAC) inhibitors, purine analogs, pyrimidine analogs, MEK inhibitors, CDK inhibitors, ErbB (such as ErbB2) receptor inhibitors,
  • Angiogenesis inhibitors include, but are not limited to, EGFR inhibitors, PDGFR inhibitors, VEGFR inhibitors, TTE2 inhibitors, IGFlR inhibitors, matrix metalloproteinase 2 (MMP-2) inhibitors, matrix metalloproteinase 9 (MMP-9) inhibitors, thrombospondin analogs such as thrombospondin- 1 and N-Ac-Sar-Gly-Val-D-alloIle-Thr-Nva-He-Arg-Pro- NHCH 2 CH 3 or a salt thereof and analogues of N-Ac-Sar-Gly-Val-D-alloIle-Thr-Nva-Ile-Arg- PrO-NHCH 2 CH 3 such as N-Ac-GlyVal- D-aIle-Ser-Gln-Ile-Arg-ProNHCH2CH3 or a salt thereof.
  • Examples of EGFR inhibitors include, but are not limited to, Iressa (gefitinib), Tarceva (erlotinib or OSI-774), Icotinib, Erbitux (cetuximab), EMD-7200, ABX-EGF, HR3, IgA antibodies, TP- 38 (IVAX), EGFR fusion protein, EGF- vaccine, anti-EGFr immunoliposomes, Tykerb (lapatinib) and AZD-8931 (sapitinib).
  • Examples of PDGFR inhibitors include, but are not limited to, CP-673,451 and CP- 868596.
  • VEGFR inhibitors include, but are not limited to, Avastin (bevacizumab), Sutent (sunitinib, SUl 1248), Nexavar (sorafenib, BAY43-9006), CP-547,632, axitinib (AG13736), Apatinib, cabozantinib, Zactima (vandetanib, ZD-6474), AEE788, AZD-2171, VEGF trap, Vatalanib (PTK-787, ZK-222584), Macugen, M862, Pazopanib (GW786034), ABT-869 and angiozyme.
  • Examples of thrombospondin analogs include, but are not limited to, TSP-I and ABT- 510.
  • aurora kinase inhibitors include, but are not limited to, VX-680, AZD- 1152 and MLN-8054.
  • Example of polo-like kinase inhibitors include, but are not limited to, BI-2536.
  • Examples of bcr-abl kinase inhibitors include, but are not limited to, Gleevec (imatinib) and Dasatinib (BMS354825).
  • platinum containing agents includes, but are not limited to, cisplatin, Paraplatin (carboplatin), eptaplatin, lobaplatin, nedaplatin, Eloxatin (oxaliplatin) or satraplatin.
  • mTOR inhibitors includes, but are not limited to, CCI-779, rapamycin, temsirolimus, everolimus, RAD001, INK-128 and ridaforolimus.
  • HSP-90 inhibitors includes, but are not limited to, geldanamycin, radicicol, 17-AAG, KOS-953, 17-DMAG, CNF-101, CNF-1010, 17-AAG-nab, NCS-683664, Mycograb, CNF-2024, PU3, PU24FC1, VER49009, IPI-504, SNX-2112 and STA-9090.
  • HDAC histone deacetylase inhibitors
  • SAHA Suberoylanilide hydroxamic acid
  • MS-275 valproic acid
  • TSA valproic acid
  • LAQ-824 Trapoxin
  • tubacin tubastatin
  • ACY-1215 Depsipeptide
  • MEK inhibitors include, but are not limited to, PD325901, ARRY-142886, ARRY-438162 and PD98059.
  • CDK inhibitors include, but are not limited to, flavopyridol, MCS-5A, CVT-2584, seliciclib (CYC-202, R-roscovitine), ZK-304709, PHA-690509, BMI-1040, GPC-286199, BMS-387,032, PD0332991 and AZD-5438.
  • COX-2 inhibitors include, but are not limited to, CELEBREXTM (celecoxib), parecoxib, deracoxib, ABT-963, MK-663 (etoricoxib), COX-189 Lumiracoxib), BMS347070, RS 57067, NS-398, Bextra (valdecoxib), paracoxib, Vioxx (rofecoxib), SD- 8381, 4- Methyl-2-(3,4-dimethylphenyl)-l-(4-sulfamoyl- ⁇ henyl-lH-pyrrole, T-614, JTE-522, S-2474, SVT-2016, CT-3, SC-58125 and Arcoxia (etoricoxib).
  • NSAIDs non-steroidal anti-inflammatory drugs
  • examples of non-steroidal anti-inflammatory drugs include, but are not limited to, Salsalate (Amigesic), Diflunisal (Dolobid), Ibuprofen (Motrin), Ketoprofen (Orudis), Nabumetone (Relafen), Piroxicam (Feldene), Naproxen (Aleve, Naprosyn), Diclofenac (Voltaren), Indomethacin (Indocin), Sulindac (Clinoril), Tolmetin (Tolectin), Etodolac (Lodine), Ketorolac (Toradol) and Oxaprozin (Daypro). [00108] Exambles of ErbB (e.g.
  • ErbB2 receptor inhibitors include, but are not limited to, CP- 724-714, CI-1033, (canertinib), Herceptin (trastuzumab), Omitarg (2C4, petuzumab), TAK-165, GW- 572016 (Ionafarnib), GW-282974, EKB-569, PI-166, AZD-8931 (sapitinib), dHER2 (HER2 Vaccine), APC8024 (HER2 Vaccine), anti-HER/2neu bispecific antibody, B7.her2IgG3, AS HER2 trifunctional bispecific antibodies, mAB AR-209 and mAB 2B-1.
  • Exambles of Phosphoinositide 3-kinase inhibitor include, but are not limited to, Wortmannin, LY294002, hibiscone C, Idelalisib, Copanlisib, Duvelisib, Taselisib, Perifosine, Idelalisib, Buparlisib, Duvelisib, Alpelisib, Umbralisib, Copanlisib, PX-866, Dactolisib, CUDC-907, Voxtalisib (also known as SAR245409, XL765), CUDC-907, ME-401, IPI-549, SF1126, RP6530, INK1117, pictilisib, XL147 (also known as SAR245408), Palomid 529, GSK1059615, ZSTK474, PWT33597, IC87114, TG100–115, CAL263, RP6503,
  • alkylating agents include, but are not limited to, nitrogen mustard N- oxide, cyclophosphamide, ifosfamide, trofosfamide, Chlorambucil, melphalan, busulfan, mitobronitol, carboquone, thiotepa, ranimustine, nimustine, temozolomide, AMD-473, altretamine, AP-5280, apaziquone, brostallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, KW- 2170, mafosfamide, and mitolactol, carmustine (BCNU), lomustine (CCNU), Busulfan, Treosulfan, Decarbazine and Temozolomide.
  • antimetabolites include but are not limited to, methotrexate, 6- mercaptopurine riboside, mercaptopurine, uracil analogues such as 5-fluorouracil (5-FU) alone or in combination with leucovorin, tegafur, UFT, doxifluridine, carmofur, cytarabine, cytarabine ocfosfate, enocitabine, S-I, Alimta (premetrexed disodium, LY231514, MTA), Gemzar (gemcitabine), fludarabine, 5-azacitidine, capecitabine, cladribine, clofarabine, decitabine, eflornithine, ethnylcytidine, cytosine arabinoside, hydroxyurea, TS-I, melphalan, nelarabine, nolatrexed, ocfosate, diso
  • 5-FU 5-
  • antibiotics include intercalating antibiotics but are not limited to, aclarubicin, actinomycins such as actinomycin D, amrubicin, annamycin, adriamycin, bleomycin a, bleomycin b, daunorubicin, doxorubicin, elsamitrucin, epirbucin, glarbuicin, idarubicin, mitomycin C, nemorubicin, neocarzinostatin, peplomycin, pirarubicin, rebeccamycin, stimalamer, streptozocin, valrubicin, zinostatin and combinations thereof.
  • actinomycins such as actinomycin D, amrubicin, annamycin, adriamycin, bleomycin a, bleomycin b, daunorubicin, doxorubicin, elsamitrucin, epirbucin, glarbuicin,
  • topoisomerase inhibiting agents include, but are not limited to, one or more agents selected from the group consisting of aclarubicin, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflomotecan, irinotecan HCL (Camptosar), edotecarin, epirubicin (Ellence), etoposide, exatecan, gimatecan, lurtotecan, orathecin (Supergen), BN-80915, mitoxantrone, pirarbucin, pixantrone, rubitecan, sobuzoxane, SN-38, tafluposide and topotecan.
  • Examples of antibodies include, but are not limited to, Rituximab, Cetuximab, Bevacizumab, Trastuzumab, specific CD40 antibodies and specific IGFlR antibodies
  • hormonal therapies include, but are not limited to, exemestane (Aromasin), leuprolide acetate, anastrozole (Arimidex), fosrelin (Zoladex), goserelin, doxercalciferol, fadrozole, formestane, tamoxifen citrate (tamoxifen), Casodex, Abarelix, Trelstar, finasteride, fulvestrant, toremifene, raloxifene, lasofoxifene, letrozole, flutamide, bicalutamide, megesterol, mifepristone, nilutamide, dexamethasone, predisone and other glucocorticoids.
  • retinoids/deltoids examples include, but are not limited to, seocalcitol (EB 1089, CB 1093), lexacalcitrol (KH 1060), fenretinide, Aliretinoin, Bexarotene and LGD-1550.
  • plant alkaloids examples include, but are not limited to, vincristine, vinblastine, vindesine and vinorelbine.
  • proteasome inhibitors include, but are not limited to, bortezomib (Velcade), MGl 32, NPI-0052 and PR-171.
  • immunologicals examples include, but are not limited to, interferons and numerous other immune enhancing agents.
  • Interferons include interferon alpha, interferon alpha-2a, interferon, alpha-2b, interferon beta, interferon gamma- 1a, interferon gamma- 1b (Actimmune), or interferon gamma-nl and combinations thereof.
  • agents include filgrastim, lentinan, sizofilan, TheraCys, ubenimex, WF-10, aldesleukin, alemtuzumab, BAM-002, decarbazine, daclizumab, denileukin, gemtuzumab ozogamicin, ibritumomab, imiquimod, lenograstim, lentinan, melanoma vaccine (Corixa), molgramostim, OncoVAC- CL, sargaramostim, tasonermin, tecleukin, thymalasin, tositumomab, Virulizin, Z-100, epratuzumab, mitumomab, oregovomab, pemtumomab (Y-muHMFGl), Provenge (Dendreon), CTLA4 (cytotoxic lymphocyte antigen 4) antibodies and agents capable of blocking CTLA4 such as MDX-010
  • Examples of biological response modifiers are agents that modify defense mechanisms of living organisms or biological responses, such as survival, growth, or differentiation of tissue cells to direct them to have anti-tumor activity. Such agents include krestin, lentinan, sizofrran, picibanil and ubenimex.
  • Examples of pyrimidine analogs include, but are not limited to, 5-Fluorouracil, Floxuridine, Doxifluridine, Ratitrexed, cytarabine (ara C), Cytosine arabinoside, Fludarabine, and Gemcitabine.
  • Examples of purine analogs include but are not limited to, Mercaptopurine and thioguanine.
  • antimitotic agents include, but are not limited to, ABT-751, paclitaxel, docetaxel, epothilone D (KOS-862) and ZK-EPO.
  • the antibodies or antigen binding fragments thereof of the present invention are also intended to be used as a radiosensitizer that enhances the efficacy of radiotherapy.
  • radiotherapy include but are not limited to, external beam radiotherapy (XBRT), or teletherapy, brachtherapy or sealed source radiotherapy, unsealed source radiotherapy.
  • XBRT external beam radiotherapy
  • the antibodies or antigen binding fragments thereof of the present invention can also be used in combination with a different class of Bcl-2 inhibitors, such as ABT263 or ABT737.
  • the cytotoxic agent may be at least one of gemcitabine and abraxane.
  • the additional antibody or therapeutically fragment thereof may be oregovomab antibody B43.13, AR9.6 antibody, or combinations thereof.
  • the chemotherapy regimen may be Folfirinox.
  • the tumor may be chosen from a pancreatic tumor, a gall bladder tumor, a gastric tumor, a colon tumor, an ovarian tumor, a breast tumor, and a liver tumor, and the method may be for the treatment of a cancer.
  • an antibody or an antigen binding fragment thereof that targets O-glycan mucin-type glycoprotein MUC16 for inhibiting tumor growth of a tumor expressing O-glycan mucin-type glycoprotein MUC16 in a subject in need thereof.
  • an antibody or an antigen binding fragment thereof that targets O-glycan mucin-type glycoprotein MUC16 for use in inhibiting tumor growth of a tumor expressing O-glycan mucin-type glycoprotein MUC16 in a subject in need thereof.
  • an antibody or an antigen binding fragment thereof that targets O-glycan mucin-type glycoprotein MUC16 for use in a method of inhibiting tumor growth of a tumor expressing O-glycan mucin-type glycoprotein MUC16 in a subject in need thereof.
  • a method of detection of a tumor expressing O-glycan mucin-type glycoprotein MUC16 in a subject in need thereof comprising administering to the subject an antibody or an antigen binding fragment thereof specific to O-glycan mucin-type glycoprotein MUC16 according to the present invention and detecting the antibody or antigen binding fragment.
  • the antibody or antigen binding fragment thereof may further comprise a detectable label, for example a fluorescent marker, a radioactive marker, an MRI contrast agent, or combinations thereof, as is known in the art.
  • the invention also encompasses nucleic acid vector comprising a nucleotide sequence encoding the antibody or antigen binding fragment thereof of the present invention, as well as cells comprising the nucleic acid vector, for expressing the the antibody or antigen binding fragment thereof of the present invention, and cells for expressing the the antibody or antigen binding fragment thereof of the present invention.
  • the present invention will be more readily understood by referring to the following examples which are given to illustrate the invention rather than to limit its scope.
  • EXAMPLE 1 Anti-Her2/Neu monoclonal antibody generation
  • the scFv C6MH3-B1 specific for HER2/neu has previously been used in the preparation of monoclonal antibodies, particularly IgE monoclonal antibodies (Daniels, T.R. et al. Cancer Immunol Immunother, 2012 Jul;61(7):991-1003).
  • this antibody has failed to become useful as a therapeutic because low production yield prevent it from being made in sufficient quantities and in a form that would be useable for therapy. This could be caused by numerous issues in addition to self-aggregation, oxidation, deamination and isomerization of amino acid chains.
  • the sequence of the anti-Her2/Neu IgE antibody derived from the C6MH3-B1 scFv comprises a full-length Lambda isotype light chain and a full length IgE heavy chain.
  • the sequence of the entire IgE heavy chain (SEQ ID NO: 13), and particularly the heavy chain variable region from FR1 to FR4 (SEQ ID NO: 14) was point mutated at numerous amino acid residues.
  • the wild type and the mutant variant sequence of the antibody C6MH3-B1 were transiently expressed in Chinese Hamster Ovary cells (CHOK1SV GS- KO) using small scale transient expression, to screen for both improved expression and binding to the antigen.
  • Mutations that caused a decrease in or no improvement on the level of expression were discarded in a first pass. Those that decreased binding of the antibodies to the antigen were also discarded in a first pass.
  • the selected variants were recloned by limiting dilutions to ensure their monoclonality, and sequenced. From the numerous mutants identified, mutations in the CDR2 region were identified and selected for further characterization. Three mutations of the first aspartic acid (D) residue of CDR H2 were identified: D to E (variant 1), D to Q (variant 2), and D to S (variant 3). Two mutations of the first serine (S) residue of CDR H2 were identified: S to T (variant 4) and S to I (variant 5).
  • Antibodies were produced for the wild type and the mutant variants 1 to 5 from transient expression in Chinese Hamster Ovary cells (CHOK1SV GS-KO) using small scale transient expression HiTrap® LambdaFabSelect resin (Sigma®) and buffer exchanged into PBS containing 3 mM EDTA.
  • EXAMPLE 2 Anti-Her2/Neu monoclonal antibody yield measurement
  • During extraction with the LambdaFabSelect resin an estimation of the titre of antibody was made from the obtained chromatograms (no shown).
  • variants 1, 2, 4 and 5 showed an unexpected improvement: variants 1, 4 and 5 eluted as a single peak with no aggregation, while variant 2 could be purified in unaggregated form from the first peak.
  • EXAMPLE 4 Anti-Her2/Neu monoclonal antibody affinity [00143] Next, the binding of variants 1, 2, 4 and 5 was compared to that of the parental antibody by flow cytometry. D2F2/E2 cells [Wei et al. Int J Cancer, 1999 May 31;81(5):748-54] expressing human Her2/Neu were cultured. Cells were detached from tissue culture dishes using 0.5 mM EDTA and incubated with varying concentrations of the various antibodies for 2 h on ice.
  • variants 1, 2, 4 and 5 are all able to bind to Her2/Neu with affinities that are at least as good as that of the wild-type parental antibody. Indeed, variants 1 and 2 have affinities that are close to, or even a little better than the parental antibody (Figs.1A and 1B). Variants 4 and 5, on the other hand, appear to display affinities that are unexpectedly improved over that of the wild-type parental antibody (Figs.1C and 1D). [00144] While preferred embodiments have been described above and illustrated in the accompanying drawings, it will be evident to those skilled in the art that modifications may be made without departing from this disclosure. Such modifications are considered as possible variants comprised in the scope of the disclosure. SEQUENCE TABLE

Abstract

The present document describes an antibody or an antigen-binding fragment comprising three variable heavy domain complementarity determining regions (CDR) (CDR H1, H2 and H3) that binds specifically to Her2/Neu. The present invention also relates to pharmaceutical compositions, nucleic acid molecule, vectors, cells comprising the nucleic acid vectors, and methods of treating Her2/Neu associated diseases.

Description

MONOCLONAL ANTIBODIES AGAINST HER2/NEU AND USES THEREOF BACKGROUND (a) Field [0001] The subject matter disclosed generally relates to monoclonal antibodies against Her2/Neu. More specifically, the subject matter relates to monoclonal antibodies against Her2/Neu that do not self-aggregate and methods of using the same. (b) Related Prior Art [0002] Breast and ovarian cancer are two of the leading causes of cancer deaths among women and overexpression of the Her2/neu oncoprotein has been reported in patients affected with breast and ovarian cancers, and is associated with poor prognosis. Targeted therapy for Her2/neu expressing tumors may be targeted with monoclonal antibodies, such as antibodies comprising the variable regions of the scFv C6MH3-B1 specific for HER2/neu. However, such antibodies can suffer from low production yield, and could be further affected by numerous issues such as self- aggregation, oxidation, deamination and isomerization of amino acid chains. Ultimately, only small quantities of these antibodies can be prepared, which precludes large scale preparation for practical therapeutic application developments. [0003] Therefore, there is a need for alternative monoclonal antibodies specific for HER2/neu that at least do not self-aggregate, improve their manufacturability and allow their preparation on a scale that allows their use as therapeutics. SUMMARY [0004] According to an embodiment, there is provided an antibody or an antigen-binding fragment that binds specifically to Her2/Neu comprising three variable heavy domain complementarity determining regions (CDR) (CDR H1, H2 and H3) wherein the CDR H1, H2, and H3, comprise an amino acid sequence comprising: CDR H1: GYSFTSYW (SEQ ID NO:1), CDR H2: IYPGX1X2DT, where X1 is D, E, or Q, X2 is S, I, or T, and wherein when X1 is D, X2 is different than S (SEQ ID NO:2), and CDR H3: ARHDVGYCTDRTCAKWPEY (SEQ ID NO:3), respectively. [0005] The antibody or an antigen-binding fragment may comprise three variable light domain CDR (CDR L1, L2 and L3), wherein the CDR L1, L2, and L3 comprise an amino acid sequence comprising: CDR L1: SSNIGNNY (SEQ ID NO:4), CDR L2: DHT (SEQ ID NO:5), and CDR L3: ASWDYTLSGWV (SEQ ID NO:6), respectively. [0006] In the antibody or antigen-binding fragment, X1 may be D, X2 may be I, or T. [0007] In the antibody or antigen-binding fragment, wherein when X1 is E, or Q, X2 is S. [0008] In the antibody or antigen-binding fragment, wherein X1 may be E and X2 may be S. [0009] In the antibody or antigen-binding fragment, wherein X1 may be Q and X2 may be S. [0010] In the antibody or antigen-binding fragment, wherein X1 may be D and X2 may be I. [0011] In the antibody or antigen-binding fragment, wherein X1 may be D and X2 may be T. [0012] The antibody or antigen-binding fragment may further comprise four variable heavy domain framework regions (HFR)(HFR 1, 2, 3 and 4), wherein said HFR 1, 2, 3, and 4 comprise an amino acid sequence comprising: HFR 1: VQLVQSGAEVKKPGESLKISCKGS (SEQ ID NO: 15), HFR 2: IAWVRQMPGKGLEYMGL (SEQ ID NO: 16), HFR 3: KYSPSFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFC (SEQ ID NO: 17), and HFR 4: WGQGTLVTV (SEQ ID NO: 18). [0013] The antibody or antigen-binding fragment may further comprise four variable light domain framework regions (LFR)(LFR 1, 2, 3 and 4), wherein said LFR 1, 2, 3, and 4 comprise an amino acid sequence comprising: LFR 1: QSVLTQPPSVSAAPGQKVTISCSGS (SEQ ID NO: 84), LFR 2: VSWYQQLPGTAPKLLIY (SEQ ID NO: 85), LFR 3: NRPAGVPDRFSGSKSGTSASLAISGFRSEDEADYYC (SEQ ID NO: 86), and LFR 4: FGGGTKVTVL (SEQ ID NO: 87). [0014] The antibody or antigen-binding fragment may comprise a variable heavy domain (VH) comprising amino acid sequence comprising: VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSP SFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQ GTLVTV (SEQ ID NO: 23). [0015] The antibody or antigen binding fragment may comprise a variable light domain (VL) comprising amino acid sequence comprising: QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPDRFSGS KSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (SEQ ID NO: 83). [0016] The antibody or antigen-binding fragment may comprise a variable heavy domain (VH) comprising amino acid sequence comprising: VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSP SFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQ GTLVTV (SEQ ID NO: 23) and a variable light domain (VL) comprising amino acid sequence comprising: QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPD RFSGSKSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (SEQ ID NO: 83). [0017] The antigen-binding fragment may be a single-domain antibody (sdAb), a fragment antigen binding (Fab), a single-chain variable fragment (scFv), or a single-chain fragment antigen binding (scFab). [0018] The antibody or antigen-binding fragment may be an IgA, an IgD, an IgE, an IgG, or an IgM. [0019] The antibody or antigen-binding fragment may be an IgE. [0020] The antibody or antigen-binding fragment may be humanized or partially humanized. [0021] According to another embodiment, there is provided a compound comprising the antibody or antigen-binding fragment of the present invention, and a functional moiety. [0022] The antibody or antigen-binding fragment may be linked to the functional moiety via a peptide linker. [0023] The antibody or antigen-binding fragment may be functionally linked to the functional moiety via the peptide linker. [0024] The peptide linker may comprise about 3 to about 40 amino acid residues. [0025] The peptide linker may comprise the amino acid sequence (GGGGS)n or (GGGS)n, wherein n ≥ 1. [0026] The antibody or antigen-binding fragment may be fused to a second antibody or antigen-binding fragment operable to bind a target epitope. [0027] The antibody or antigen-binding fragment may be linked to a peptide, a polypeptide, a protein, an enzyme, a second antibody, an antibody fragment, a second antigen-binding fragment or a combination of any two or more thereof; wherein each of the antibody or antigen-binding fragment thereof and the linked peptide, polypeptide, protein, enzyme, second antibody, antibody fragment, second antigen-binding fragment, or the combination of any two or more thereof is functional. [0028] The antibody fragment may be a fragment crystallizable (Fc) region. [0029] According to another embodiment, there is provided a composition comprising the antibody or antigen-binding fragment of the present invention or the compound of the present invention, and a pharmaceutically acceptable diluent, carrier or excipient. [0030] According to another embodiment, there is provided a nucleic acid molecule encoding an antibody or antigen-binding fragment of the present invention, or encoding a compound of the present invention. [0031] According to another embodiment, there is provided a vector comprising the nucleic acid molecule of the present invention operably linked to one or more regulatory elements to allow expression of the antibody or antigen-binding fragment the present invention, or the compound of the present invention in a host cell. [0032] According to another embodiment, there is provided a cell comprising the vector of the present invention for expressing the antibody or antigen-binding fragment thereof of the present invention or the compound of the present invention. [0033] According to another embodiment, there is provided a method of treating a Her2/Neu associated disease comprising: administering the antibody or antigen-binding fragment of the present invention, the compound of the present invention, the composition of the present invention, or a combination of any two or more thereof to a subject in need thereof. [0034] The Her2/Neu associated disease may be a cancer. [0035] The cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer. [0036] According to another embodiment, there is provided a use of the antibody or antigen- binding fragment of the present invention, the compound of the present invention, the composition of the present invention, or a combination of any two or more thereof to a subject in need thereof in the treatment of a Her2/Neu associated disease. [0037] The Her2/Neu associated disease may be a cancer. [0038] The cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer. [0039] According to another embodiment, there is provided an antibody or antigen-binding fragment of the present invention, the compound of the present invention, the composition of the present invention, or a combination of any two or more thereof to a subject in need thereof, for use in the treatment of a Her2/Neu associated disease. [0040] The antibody or antigen-binding fragment, compound, composition, or combination of any two or more thereof of the present invention, wherein the Her2/Neu associated disease may be a cancer. [0041] The antibody or antigen-binding fragment, compound, composition, or combination of any two or more thereof the present invention, wherein the cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer. [0042] According to another embodiment, there is provided a use of the antibody or antigen- binding fragment of the present invention, the compound of the present invention, the composition of the present invention, or a combination of any two or more thereof to a subject in need thereof in the manufacture of a medicament for the treatment of a Her2/Neu associated disease. [0043] The Her2/Neu associated disease may be a cancer. [0044] The cancer may be ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer. [0045] Features and advantages of the subject matter hereof will become more apparent in light of the following detailed description of selected embodiments, as illustrated in the accompanying figures. As will be realized, the subject matter disclosed and claimed is capable of modifications in various respects, all without departing from the scope of the claims. Accordingly, the drawings and the description are to be regarded as illustrative in nature, and not as restrictive and the full scope of the subject matter is set forth in the claims. BRIEF DESCRIPTION OF THE DRAWINGS [0046] Further features and advantages of the present disclosure will become apparent from the following detailed description, taken in combination with the appended drawings, in which: [0047] Fig.1A shows an apparent affinity non-linear regression curve comparing the binding on D2F2/E2 cells of parental IgE C6MH3-B1 antibody versus variant 1. Each of the wild-type and Variant 1, Peak 1 (V1, P1) antibodies are used at dose -4 (0.0002 nM), -3 (0.002 nM), -2 (0.02 nM), - 1 (0.2 nM), 0 (2 nM), 1 (20 nM), and 2 (200 nM). [0048] Fig.1B shows an apparent affinity non-linear regression curve comparing the binding on D2F2/E2 cells of parental IgE C6MH3-B1 antibody versus variant 2. Each of the wild-type and Variant 2, Peak 1 (V2, P1) antibodies are used at dose -4 (0.0002 nM), -3 (0.002 nM), -2 (0.02 nM), - 1 (0.2 nM), 0 (2 nM), 1 (20 nM), and 2 (200 nM). [0049] Fig.1C shows an apparent affinity non-linear regression curve comparing the binding on D2F2/E2 cells of parental IgE C6MH3-B1 antibody versus variant 4. Each of the wild-type and Variant 4, Peak 1 (V4, P1) antibodies are used at dose -4 (0.0002 nM), -3 (0.002 nM), -2 (0.02 nM), - 1 (0.2 nM), 0 (2 nM), 1 (20 nM), and 2 (200 nM). [0050] Fig.1D shows an apparent affinity non-linear regression curve comparing the binding on D2F2/E2 cells of parental IgE C6MH3-B1 antibody versus variant 5. Each of the wild-type and Variant 5, Peak 1 (V5, P1) antibodies are used at dose -4 (0.0001 nM), -3 (0.001 nM), -2 (0.01 nM), - 1 (0.1 nM), 0 (1 nM), 1 (10 nM), and 2 (100 nM). [0051] It will be noted that throughout the appended drawings, like features are identified by like reference numerals. DETAILED DESCRIPTION [0052] The present invention is directed to a technology for binding specifically to Her2/Neu. In embodiments there is disclosed an antibody or an antigen-binding fragment that specifically binds to Her2/Neu, that comprises three variable heavy domain complementarity determining regions (CDR H1, CDR H2 and CDR H3). [0053] The three variable heavy domain complementarity determining regions (CDR) (CDR H1, H2 and H3) may comprise the following amino acid sequences: 1) CDR H1: GYSFTSYW (SEQ ID NO: 1), 2) CDR H2: IYPGX1X2DT, where X1 is D, E, or Q, X2 is S, I, or T, and wherein when X1 is D, X2 is different than S (SEQ ID NO: 2), and 3) CDR H3: ARHDVGYCTDRTCAKWPEY (SEQ ID NO: 3). [0054] According to an embodiment, the antibody or an antigen-binding fragment, may also comprise three variable light domain CDR (CDR L1, L2 and L3). The three variable light domain CDR (CDR L1, L2 and L3) may comprise the following amino acid sequences: 1) CDR L1: SSNIGNNY (SEQ ID NO: 4), 2) CDR L2: DHT (SEQ ID NO: 5), and 3) CDR L3: ASWDYTLSGWV (SEQ ID NO: 6). [0055] According to embodiments, the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein when X1 is D, X2 is I, or T (e.g. as per SEQ ID NOs: 65 and 56, respectively). According to other embodiments, the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein when X1 is E, or Q, X2 is S (e.g. as per SEQ ID NOs: 29, and 38, respectively). According to another embodiment, the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein X1 is E and X2 is S (e.g. as per SEQ ID NO: 29). According to another embodiment, the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein X1 is Q and X2 is S (e.g. as per SEQ ID NO: 38). According to another embodiment, the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein X1 is D and X2 is I (e.g. as per SEQ ID NO: 65). According to another embodiment, the antibody or an antigen-binding fragment that specifically binds to Her2/Neu may comprise a CDR H2 wherein X1 is D and X2 is T (e.g. as per SEQ ID NO: 56). (See Table A, below) [0056] According to other embodiments, the antibody or antigen-binding fragment of the present invention may be an antibody comprising four variable heavy domain framework regions (HFR 1 to HFR4) and three variable heavy domain complementarity determining regions (CDR H1, CDR H2 and CDR H3), comprising amino acid sequence comprising : VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 23), VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGQSDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 32), QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGSSDTKYSPSFQ GQVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 41), VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGDTDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 50), or VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGDIDTKYSPSFQGQ VTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 59), that binds specifically to a Her2/Neu. It is believed to be operable to neutralize Her2/Neu, including, but not limited to a Her2/Neu family member from human, monkey, rat, or mouse. [0057] According to other embodiments, the antibody or antigen-binding fragment of the present invention may be an antibody comprising variable heavy domain (VH) comprising amino acid sequences comprising according to SEQ ID NOs: 22, 31, 49 or 58, respectively, that binds specifically to a Her2/Neu. It is believed to be operable to neutralize Her2/Neu, including, but not limited to a Her2/Neu family member from human, monkey, rat, or mouse. [0058] According to other embodiments, the antibody or antigen-binding fragment of the present invention may be an antibody comprising four variable light domain framework regions (LFR1 to LFR4) and three variable light domain complementarity determining regions (CDR L1, CDR L2 and CDR L3), comprising amino acid sequence comprising : QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPDRFSGS KSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (as per each of SEQ ID NOs: 83, 92, 101, 110 or 119), that binds specifically to a Her2/Neu. It is believed to be operable to neutralize Her2/Neu, including, but not limited to a Her2/Neu family member from human, monkey, rat, or mouse. [0059] According to other embodiments, the antibody or antigen-binding fragment of the present invention may be an antibody comprising variable light domain (VL) comprising amino acid sequences according to SEQ ID NOs: 82, 91, 100, 109 or 118, respectively, that binds specifically to a Her2/Neu. It is believed to be operable to neutralize Her2/Neu, including, but not limited to a Her2/Neu family member from human, monkey, rat, or mouse. [0060] In embodiments, the antibody or antigen binding fragment thereof of the present invention may further comprise four variable heavy domain framework regions (HFR)(HFR 1, 2, 3 and 4), which comprise an amino acid sequence comprising: HFR 1: VQLVQSGAEVKKPGESLKISCKGS (SEQ ID NOs: 15, 24, 33, 42 or 51), HFR 2: IAWVRQMPGKGLEYMGL (SEQ ID NOs: 16, 25, 34, 43 or 52), HFR 3: KYSPSFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFC (SEQ ID NOs: 17, 26, 35, 44 or 53), and HFR 4: WGQGTLVTV (SEQ ID NOs: 18, 27, 36, 45 or 54). [0061] In other embodiments, the antibody or antigen binding fragment thereof of the present invention may further comprise four variable light domain framework regions (LFR)(LFR 1, 2, 3 and 4) which comprise an amino acid sequence comprising: LFR 1: QSVLTQPPSVSAAPGQKVTISCSGS (SEQ ID NOs: 84, 93, 102, 111, or 120), LFR 2: VSWYQQLPGTAPKLLIY (SEQ ID NOs: 85, 94, 103, 112, or 121), LFR 3: NRPAGVPDRFSGSKSGTSASLAISGFRSEDEADYYC (SEQ ID NOs: 86, 95, 104, 113 or 122), and LFR 4: FGGGTKVTVL (SEQ ID NOs: 87, 96, 105, 114, or 123). [0062] According to another embodiment, the antibody or antigen binding fragment thereof of the present invention may comprise a variable heavy domain (VH) comprising amino acid sequence comprising: VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 23), and a variable light domain (VL) comprising amino acid sequence comprising: QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPDRFSGS KSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (SEQ ID NO: 83).
Figure imgf000011_0001
Figure imgf000012_0001
Table A. Amino acid sequence of Her2/Neu specific heavy variable heavy domain (VL). Framework regions (FRs) and complementarity-determining regions (CDRs) are classified and numbered according to IMGT. Antibody 0 represent the starting antibody and 1 to 5 the mutated sequences. Underlined = amino acid sequence of variable region from FR1 to FR4.
Figure imgf000013_0001
Table B. Amino acid sequence of Her2/Neu specific light variable heavy domain (VL). Framework regions (FRs) and complementarity-determining regions (CDRs) are classified and numbered according to IMGT. Antibody 0 represent the starting antibody and 1 to 5 the mutated sequences. Underlined = amino acid sequence of variable region from FR1 to FR4. The dash ( - ) character is only illustrative of a space in the text to identify the CDR2, and not of a CDR2 feature. [0063] According to other embodiments, the antibody or an antigen-binding fragment of the present invention may have sequences substantially identical to the sequences disclosed above, operable to bind to Her2/Neu. A substantially identical sequence may comprise one or more conservative amino acid mutations. It is known in the art that the introduction of one or more conservative amino acid mutations to a reference sequence may yield a mutant peptide with no substantial change in physiological, chemical, physico-chemical or functional properties compared to the reference sequence. In such a case, the reference and mutant sequences would be considered “substantially identical” polypeptides. A conservative amino acid substitution is defined herein as the substitution of an amino acid residue for another amino acid residue with similar chemical properties (e.g. size, charge, or polarity). According to one embodiment, one or more conservative amino acid mutations may be made to one or more framework regions of the sdAb while maintaining both the CDR sequences listed above and the overall structure of the CDR of the antibody or antigen-binding fragment; thus the specificity and binding of the antibody are maintained. According to another embodiment, one or more conservative amino acid mutations may be made to one or more framework regions of the sdAb and to a CDR sequence listed above while maintaining the antigen- binding function of the overall structure of the CDR of the antibody or antigen-binding fragment; thus the specificity and binding of the antibody are maintained. [0064] In a non-limiting example, a conservative mutation may be a conservative amino acid substitution. Such a conservative amino acid substitution may substitute a basic, neutral, hydrophobic, or acidic amino acid for another amino acid of the same group. By the term “basic amino acid” it is meant a hydrophilic amino acid having a side chain pK value of greater than 7, which is typically positively charged at physiological pH. Basic amino acids include histidine (His or H), arginine (Arg or R), and lysine (Lys or K). By the term “neutral amino acid” (also “polar amino acid”), it is meant a hydrophilic amino acid having a side chain that is uncharged at physiological pH, but which has at least one bond in which the pair of electrons shared in common by two atoms is held more closely by one of the atoms. Polar amino acids include serine (Ser or S), threonine (Thr or T), cysteine (Cys or C), tyrosine (Tyr or Y), asparagine (Asn or N), and glutamine (GIn or Q). By the term “hydrophobic amino acid” (also “non-polar amino acid”) it is meant an amino acid exhibiting a hydrophobicity of greater than zero according to the normalized consensus hydrophobicity scale of Eisenberg (1984). Hydrophobic amino acids include proline (Pro or P), isoleucine (Ile or I), phenylalanine (Phe or F), valine (Val or V), leucine (Leu or L), tryptophan (Trp or W), methionine (Met or M), alanine (Ala or A), and glycine (Gly or G). “Acidic amino acid” refers to a hydrophilic amino acid having a side chain pK value of less than 7, which is typically negatively charged at physiological pH. Acidic amino acids include glutamate (Glu or E) and aspartate (Asp or D). [0065] Sequence identity is used to evaluate the similarity of two sequences. It is determined by calculating the percentage of residues that are the same when the two sequences are aligned for maximum correspondence between residue positions. Any known method may be used to calculate sequence identity; for example, computer software is available to calculate sequence identity. Without wishing to be limiting, sequence identity can be calculated by software such as NCBI BLAST2 service maintained by the Swiss Institute of Bioinformatics (and as found at ca.expasy.org/tools/blast/), BLAST-P, Blast-N, or FASTA-N, or any other appropriate software that is known in the art. [0066] The substantially identical sequences of the present invention may be at least 90% identical; in another example, the substantially identical sequences may be at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical, or any percentage therebetween, at the amino acid level to sequences described herein. Importantly, a substantially identical sequence retains the activity and specificity of the reference sequence. In a non-limiting embodiment, the difference in sequence identity may be due to one or more conservative amino acid mutations. In a non-limiting example, the present invention may be directed to an antibody or antigen-binding fragment comprising a sequence at least 95%, at least 98%, or at least 99% identical to that of one or more of the antibodies or antigen- binding fragments described herein. [0067] The antibody or an antigen-binding fragment of the present invention may be used, for example, to bind specifically to a Her2/Neu. It is believed to neutralize the Her2/Neu receptors on the surface of cancer cells and blocking them from receiving growth signals. As used herein, the expression “bind specifically to a Her2/Neu” or “bind specifically to Her2/Neu” is intended to mean that the antibody or antigen-binding fragment of the present invention is enabled to bind specifically to Her2/Neu and particularly to one or more human, rhesus, mouse or rat serum Her2/Neu. [0068] The term “antibody”, also referred to in the art as “immunoglobulin” (Ig), as used herein refers to a protein constructed from paired heavy and light polypeptide chains; various Ig isotypes exist, including IgA, IgD, IgE, IgG, and IgM. When an antibody is correctly folded, each chain folds into a number of distinct globular domains joined by more linear polypeptide sequences. For example, the immunoglobulin light chain folds into a variable (VL) and a constant (CL) domain, while the heavy chain folds into a variable (VH) and three constant (CH1, CH2, CH3) domains. Interaction of the heavy and light chain variable domains (VH and VL) results in the formation of an antigen binding region (Fv). Each domain has a well-established structure familiar to those of skill in the art. [0069] The light and heavy chain variable regions are responsible for binding the target antigen and can therefore show significant sequence diversity between antibodies. The constant regions show less sequence diversity, and are responsible for binding a number of natural proteins to elicit important biochemical events. The variable region of an antibody contains the antigen-binding determinants of the molecule, and thus determines the specificity of an antibody for its target antigen. The majority of sequence variability occurs in six hypervariable regions, three each per variable heavy (VH) and light (VL) chain; the hypervariable regions combine to form the antigen-binding site, and contribute to binding and recognition of an antigenic determinant. The specificity and affinity of an antibody for its antigen is determined by the structure of the hypervariable regions, as well as their size, shape, and chemistry of the surface they present to the antigen. Various schemes exist for identification of the regions of hypervariability, the two most common being those of Kabat and of Chothia and Lesk. Kabat and Wu (1991) define the “complementarity-determining regions” (CDRs) based on sequence variability at the antigen-binding regions of the VH and VL domains. Chothia and Lesk (1987) define the “hypervariable loops” (H or L) based on the location of the structural loop regions in the VH and VL domains. These individual schemes define CDR and hypervariable loop regions that are adjacent or overlapping. Those of skill in the antibody art often utilize the terms “CDR” and “hypervariable loop” interchangeably, and they may be so used herein. The CDRs/loops are identified herein according to the IMGT nomenclature scheme (i.e., CDR1, 2 and 3, for each variable region). [0070] An “antibody fragment” or “antigen-binding fragment” as referred to herein may include any suitable antigen-binding antibody fragment known in the art. The antibody fragment may be a naturally-occurring antibody fragment, or it may be a non-naturally occurring antibody fragment obtained, for example, by manipulation of a naturally-occurring antibody or by recombinant methods. For example, an antibody fragment may include, but is not limited to, a Fv, a single-chain Fv (scFv; a molecule consisting of VL and VH connected with a peptide linker), a Fab, a F(ab’)2, or a single- domain antibody (sdAb; a fragment composed of a single VL or VH or a VHH), or a multivalent presentation of any of these. Antibody fragments such as those just described may require one or more linker sequences, disulfide bonds, or other types of covalent bond to link different portions of the fragments. Those of skill in the art will be familiar with the requirements of the different types of fragments and various approaches for their construction. [0071] In a non-limiting example, the antigen-binding fragment of the present invention may be a sdAb derived from a naturally-occurring source. Heavy chain antibodies of camelid origin (Hamers-Casterman et al, 1993) lack light chains and thus their antigen binding sites consist of one domain, termed VHH. SdAbs have also been observed in shark and are termed VNAR (Nuttall et al, 2003). Other sdAbs may be engineered based on human Ig heavy and light chain sequences (Jespers et al, 2004; To et al, 2005). As used herein, the term “sdAb” includes an sdAb directly isolated from a VH, VHH, VL, or VNAR reservoir of any origin through phage display or other technology, an sdAb derived from the aforementioned sdAb, a recombinantly produced sdAb, as well as an sdAb generated through further modification of such sdAb by humanization, affinity maturation, stabilization, solubilization, camelization, or other methods of antibody engineering. Also encompassed by the present invention are homologues, derivatives, or fragments that retain the antigen-binding function and specificity of the sdAb. [0072] SdAbs possess desirable properties for antibody molecules, such as high thermostability, high detergent resistance, relatively high resistance to proteases (Dumoulin et al, 2002) and high production yield (Arbabi-Ghahroudi et al, 1997). They can also be engineered to have very high affinity by isolation from an immune library (Li et al, 2009) or by in vitro affinity maturation (Davies & Riechmann, 1996). Further modifications to increase stability, such as the introduction of one or more non-canonical disulfide bonds (Hussack et al, 2011a,b; Kim et al, 2012), may also be brought to the sdAb. [0073] A person of skill in the art would be well-acquainted with the structure of a single- domain antibody (see, for example, 3DWT, 2P42 in Protein Data Bank). An sdAb comprises a single immunoglobulin domain that retains the immunoglobulin fold; most notably, only three CDR/hypervariable loops form the antigen-binding site. However, and as would be understood by those of skill in the art, not all CDRs may be required for binding the antigen. For example, and without wishing to be limiting, one, two, or three of the CDRs may contribute to binding and recognition of the antigen by the sdAb of the present invention. The CDRs of the sdAb or variable domain are referred to herein as CDR1, CDR2, and CDR3. [0074] The present invention further encompasses an antibody or an antigen-binding fragment that is “humanized” using any suitable method known in the art, such as, but not limited to, CDR grafting or veneering. Humanization of an antibody or an antigen-binding fragment comprises replacing an amino acid in the antibody or antigen-binding fragment sequence with its human counterpart, as found in the human consensus sequence, without substantial loss of antigen-binding ability or specificity; this approach reduces immunogenicity of the antibody or antigen-binding fragment when introduced into human subjects. In the process of CDR grafting, one or more than one of the CDRs defined herein may be fused or grafted to a human variable region (VH, or VL), to a human antibody (IgA, IgD, IgE, IgG, and IgM), to a human antibody fragment framework region (Fv, scFv, Fab) or to another protein of similar size and nature onto which a CDR can be grafted (Nicaise et al, 2004). In such a case, the conformation of the one or more than one hypervariable loop is likely preserved, and the affinity and specificity of the antibody or antigen-binding fragment for its target (i.e., a human/rhesus/rat/mouse Her2/Neu family member, collectively referred to as Her2/Neu family member) is likely minimally affected. CDR grafting is known in the art and is described in at least the following: US Patent No.6180370, US Patent No.5693761, US Patent No.6054297, US Patent No. 5859205, and European Patent No.626390. Veneering, also referred to in the art as “variable region resurfacing”, involves humanizing solvent-exposed positions of an antibody or antigen-binding fragment; thus, preserving buried non-humanized residues, which may be important for CDR conformation, while minimizing the potential for immunological reaction against solvent-exposed regions. Veneering is known in the art and is described in at least the following: US Patent No. 5869619, US Patent No. 5766886, US Patent No. 5821123, and European Patent No. 519596. Persons of skill in the art would also be amply familiar with methods of preparing such humanized antibody fragments and humanizing amino acid positions. [0075] The antibody or antigen-binding fragment according to the present invention may comprise an additional sequence to aid in expression, detection or purification of the antibody or antigen-binding fragment. Any such sequence or tag known to those of skill in the art may be used. For example, and without wishing to be limiting, the antibody or antigen-binding fragment may comprise a targeting or signal sequence (such as, but not limited to, ompA or pelB), a detection/purification tag (such as, but not limited to, c-Myc, HA, His5, or His6), or a combination of any two or more thereof. In another example, the additional sequence may be a biotin recognition site, such as that described by Cronan et al. in WO 95/04069 or by Voges et al. in WO/2004/076670. As is also known to those of skill in the art, a linker sequence may be used in conjunction with the additional sequence or tag, or may serve as a detection/purification tag. [0076] In another embodiment, there is disclosed a compound comprising an antibody or an antigen-binding fragment according to the present invention, linked to a functional moiety, optionally by a linker sequence. In embodiments of the compound, the antibody or antigen-binding fragment may be linked to the functional moiety via a linker (also known as a linker sequence). As used herein, the term “linker sequence” is intended to mean a short (typically 40 amino acids or fewer) peptide sequence that is introduced between protein domains. Linker sequences are often composed of flexible residues such as glycine and serine so that the linked protein domains are free to move relative to one another. The linker sequence can be any linker sequence known in the art that would allow for the antibody and the functional moiety of the present invention to be operably linked for the desired function. The linker may be any sequence known in the art (either a natural or synthetic linker) that allows for an operable fusion comprising an antibody or antigen-binding fragment linked to a polypeptide (e.g. the functional moiety). For example, the linker sequence may be a linker sequence L such as (GGGS)n and (GGGGS)n, wherein n is equal to or greater than 1, or from about 1 to about 5, or from about 1 to 15; or n may be any number that would allow for the operability of the compound of the present invention. In another example, the linker may be an amino acid sequence, for example, an amino acid sequence that comprises about 1 to about 40 amino acids, or about 3 to about 40 amino acids, or about 5 to about 40 amino acids, or about 10 to about 40 amino acids, or about 15 to about 40 amino acids, or about 20 to about 40 amino acids, or about 25 to about 40 amino acids, or about 30 to about 40 amino acids, or about 35 to about 40 amino acids, or about 3 to about 35 amino acids, or about 5 to about 35 amino acids, or about 10 to about 35 amino acids, or about 15 to about 35 amino acids, or about 20 to about 35 amino acids, or about 25 to about 35 amino acids, or about 30 to about 35 amino acids, or about 3 to about 30 amino acids, or about 5 to about 30 amino acids, or about 10 to about 30 amino acids, or about 15 to about 30 amino acids, or about 20 to about 30 amino acids, or about 25 to about 30 amino acids, or about 3 to about 25 amino acids, or about 5 to about 25 amino acids, or about 10 to about 25 amino acids, or about 15 to about 25 amino acids, or about 20 to about 25 amino acids, or about 3 to about 20 amino acids, or about 5 to about 20 amino acids, or about 10 to about 20 amino acids, or about 15 to about 20 amino acids, or about 3 to about 15 amino acids, or about 5 to about 15 amino acids, or about 10 to about 15 amino acids, or about 15 to about 20 amino acids, or about 3 to about 10 amino acids, or about 5 to about 10 amino acids, or about 3 to about 5 amino acids, or up to 3, up to 5, up to 10, up to 15, up to 20, up to 25, up to 30, up to 35, or up to 40 amino acids. [0077] As used herein, the term “functional moiety” is intended to mean a part of the compound having an activity, purpose, or task; relating to the way in which the compound is intended to work or operate. In embodiments, the functional moiety may be linked to the antibody or antigen- binding fragment, for example, through a chemical link pursuant to a chemical reaction, and/or through fusion of the antibody or antigen-binding fragment with the functional moiety, obtained for example using recombinant DNA technology. According to an embodiment, the antibody or antigen- binding fragment of the compound may be fused to a peptide, a polypeptide (e.g. growth factor CIBP2, an antimicrobial cyclic peptide), a protein, an enzyme [such as iduronate-2-sulfatase (IDS), acid beta-glucosidase (GCase), a serine protease, a growth factor, etc.], another (or the same) antibody or a fragment operable to bind a target epitope (e.g. an anti-microbial antibody, an anti- inflammatory antibody, an intrabody, a BBB-crossing antibody, a neurodegeneration target antibody, an ion channel targeting antibody, a cancer associated antigen antibody, a checkpoint inhibitor targeting antibody, or a GPCR targeting antibody)(for any use and for example for use in imaging, diagnostic, affinity purification, etc.), or a combination of any two or more thereof, in which both the antibody or antigen-binding fragment and the rest of the compound (i.e. the functional moiety) remain functional for their intended purpose. In a preferred embodiment, the compound may be fused to a second antibody or antigen-binding fragment, operable to bind a target epitope, which may be the same as, or distinct from the epitope of the antibody or antigen-binding fragment of the present invention. [0078] The antibody or antigen-binding fragment of the present invention may also be in a multivalent display format, also referred to herein as multivalent presentation. Multimerization may be achieved by any suitable method known in the art. For example, and without wishing to be limiting in any manner, multimerization may be achieved using self-assembly molecules such as those described in Zhang et al (2004a; 2004b) and W02003/046560, where pentabodies are produced by expressing a fusion protein comprising the antibody or antigen-binding fragment of the present invention and the pentamerization domain of the B-subunit of an AB5 toxin family (Merritt & Hol, 1995). A multimer may also be formed using the multimerization domains described by Zhu et al. (2010); this form, referred to herein as a “combody” form, is a fusion of the antibody or fragment of the present invention with a coiled-coil peptide resulting in a multimeric molecule (Zhu et al., 2010). Other forms of multivalent display are also encompassed by the present invention. For example, and without wishing to be limiting, the antibody or antigen-binding fragment may be presented as a dimer, a trimer, or any other suitable oligomer. This may be achieved by methods known in the art (Spiess et al, 2015), for example by direct linking connection (Nielsen et al, 2000), c-jun/Fos interaction (de Kruif & Logtenberg, 1996), or “Knob into holes” interaction (Ridgway et al, 1996). [0079] Another method known in the art for multimerization is to dimerize the antibody or antigen-binding fragment using an Fc domain, such as, but not limited to a human Fc domain. The Fc domain may be selected from various classes including, but not limited to, IgG, IgM, or various subclasses including, but not limited to IgG1, IgG2, etc. In this approach, the Fc gene is inserted into a vector along with the sdAb gene to generate a sdAb-Fc fusion protein (Bell et al, 2010; lqbal et al, 2010); the fusion protein is recombinantly expressed, then purified. For example, and without wishing to be limiting in any manner, a multivalent display format may encompass a chimeric or humanized format of VHH of the present invention linked to an Fc domain, or bi or tri-specific antibody fusions with two or three VHHs recognizing unique epitopes. Such antibodies are easy to engineer and produce, can greatly extend the serum half-life of a sdAb, and may be excellent tumor imaging reagents (Bell et al., 2010). [0080] The Fc domain in the multimeric complex as just described may be any suitable Fc fragment known in the art. The Fc fragment may be from any suitable source; for example, the Fc fragment may be of mouse or human origin. In a specific, non-limiting example, the Fc fragment may be a mouse Fc2b fragment or a human Fc1 fragment (Bell et al, 2010; lqbal et al, 2010). The Fc fragment may be fused to the N-terminal or C-terminal end of the VHH or humanized version of the present invention. [0081] Each subunit of the multimers described above may comprise the same or different antibodies or antigen-binding fragments of the present invention, which may have the same or different specificity. Additionally, the multimerization domains may be linked to the antibody or antigen-binding fragment using a linker, as required; such a linker should be of sufficient length and appropriate composition to provide flexible attachment of the two molecules but should not hamper the antigen-binding properties of the antibody or antigen-binding fragment. As defined above, the linker sequence can be any linker known in the art that would allow for the compound of the present invention to be prepared and be operable for the desired function. [0082] According to another embodiment, the present invention also encompasses a composition comprising one or more than one compound as described herein. The composition may comprise a single antibody and/or compound as described above, or the composition may comprise a mixture of antibody and/or compounds. Furthermore, in a composition comprising a mixture of antibody and/or compounds of the present invention, the antibody and/or compounds may have the same specificity, or they may differ in their specificities; for example, and without wishing to be limiting in any manner, the composition may comprise antibody and/or compounds specific to Her2/Neu (same or different epitope). [0083] A composition according to the invention may also comprise a pharmaceutically acceptable diluent, excipient, or carrier. The diluent, excipient, or carrier may be any suitable diluent, excipient, or carrier known in the art that is compatible with other ingredients in the composition, that is compatible with the method of delivery of the composition, and that is not deleterious to the recipient of the composition. The composition may be in any suitable form; for example, the composition may be provided in suspension form, powder form (such as, but not limited to, lyophilised or encapsulated), capsule form or tablet form. For example, and without wishing to be limiting, when the composition is provided in suspension form, the carrier may comprise water, saline, or a suitable buffer, and optionally comprise one or more additives to improve solubility and/or stability. Reconstitution to produce a suspension may be effected in a buffer at a suitable pH to ensure the viability of the antibody or antigen-binding fragment. Dry powders may also include additives to improve stability and/or carriers to increase bulk/volume; for example, and without wishing to be limiting, the dry powder composition may comprise sucrose or trehalose. In a specific, non-limiting example, the composition may be formulated for delivery of the antibody or antigen- binding fragment to the gastrointestinal tract of the subject. Thus, the composition may comprise encapsulation, time release, or other suitable technologies for delivery of the antibody and/or compound of the present invention. It would be within the competency of a person of skill in the art to prepare suitable compositions comprising the present antibody and/or compound. [0084] The invention also encompasses a nucleic acid molecule comprising a nucleotide sequence encoding an antibody, antigen-binding fragment, or compound of the present invention. The invention further comprises a vector comprising the nucleic acid molecule; a cell comprising the vector, for expressing the antibody, antigen-binding fragment, or compound of the present invention, and a cell for expressing the antibody, antigen-binding fragment, or compound of the present invention. [0085] According to another embodiment, there is provided a method of treating a Her2/Neu associated disease comprising administering the antibody or antigen-binding fragment of the present invention, the compound of the present invention, the composition of the present invention or a combination of any two or more thereof to a subject in need thereof. [0086] According to another embodiment, there is provided a use of the antibody or antigen- binding fragment of the present invention, the compound of the present invention, the composition of the present invention or a combination of any two or more thereof for the treatment of a Her2/Neu associated disease. [0087] According to another embodiment, there is provided a use of the antibody or antigen- binding fragment of the present invention, the compound of the present invention, the composition of the present invention or a combination of any two or more thereof in the manufacture of a medicament for the treatment of a Her2/Neu associated disease. [0088] According to another embodiment, there is provided an antibody or antigen-binding fragment of the present invention, the compound of the present invention, the composition of the present invention or a combination of any two or more thereof for use in the treatment of a Her2/Neu associated disease. [0089] As used herein, the expression “Her2/Neu associated disease” is intended to mean that the disease involves, is connected to, Her2/Neu as an etiological factor. [0090] According to an embodiment, the disease is a cancer. Non-limiting examples in which Her2/Neu may be involved include ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer. [0091] According to another embodiment, the method of the present invention may further comprise administering a second therapeutic agent comprising at least one of a cytotoxic agent, an additional antibody or a therapeutically active fragment thereof, or a chemotherapy regimen. [0092] Indeed, it is contemplated that the present method and therapeutic strategies may be used alone or in combination with cytotoxic agents to increase overall patient survival. The cytotoxic therapeutic agents include, but are not limited to, angiogenesis inhibitors, antiproliferative agents, kinase inhibitors, receptor tyrosine kinase inhibitors, aurora kinase inhibitors, polo-like kinase inhibitors, bcr-abl kinase inhibitors, growth factor inhibitors, COX-2 inhibitors, non-steroidal anti- inflammatory drugs (NSAIDS), antimitotic agents, alkylating agents, antimetabolites, intercalating antibiotics, platinum containing agents, growth factor inhibitors, ionizing radiation, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biologic response modifiers, immunologicals, antibodies, hormonal therapies, retinoids/deltoids plant alkaloids, proteasome inhibitors, HSP-90 inhibitors, histone deacetylase inhibitors (HDAC) inhibitors, purine analogs, pyrimidine analogs, MEK inhibitors, CDK inhibitors, ErbB (such as ErbB2) receptor inhibitors, phosphoinositide 3-kinases (PI3Ks) / Akt signaling inhibitors, mTOR inhibitors and combinations thereof as well as other antitumor agents. [0093] Angiogenesis inhibitors include, but are not limited to, EGFR inhibitors, PDGFR inhibitors, VEGFR inhibitors, TTE2 inhibitors, IGFlR inhibitors, matrix metalloproteinase 2 (MMP-2) inhibitors, matrix metalloproteinase 9 (MMP-9) inhibitors, thrombospondin analogs such as thrombospondin- 1 and N-Ac-Sar-Gly-Val-D-alloIle-Thr-Nva-He-Arg-Pro- NHCH2CH3 or a salt thereof and analogues of N-Ac-Sar-Gly-Val-D-alloIle-Thr-Nva-Ile-Arg- PrO-NHCH2CH3 such as N-Ac-GlyVal- D-aIle-Ser-Gln-Ile-Arg-ProNHCH2CH3 or a salt thereof. [0094] Examples of EGFR inhibitors include, but are not limited to, Iressa (gefitinib), Tarceva (erlotinib or OSI-774), Icotinib, Erbitux (cetuximab), EMD-7200, ABX-EGF, HR3, IgA antibodies, TP- 38 (IVAX), EGFR fusion protein, EGF- vaccine, anti-EGFr immunoliposomes, Tykerb (lapatinib) and AZD-8931 (sapitinib). [0095] Examples of PDGFR inhibitors include, but are not limited to, CP-673,451 and CP- 868596. [0096] Examples of VEGFR inhibitors include, but are not limited to, Avastin (bevacizumab), Sutent (sunitinib, SUl 1248), Nexavar (sorafenib, BAY43-9006), CP-547,632, axitinib (AG13736), Apatinib, cabozantinib, Zactima (vandetanib, ZD-6474), AEE788, AZD-2171, VEGF trap, Vatalanib (PTK-787, ZK-222584), Macugen, M862, Pazopanib (GW786034), ABT-869 and angiozyme. [0097] Examples of thrombospondin analogs include, but are not limited to, TSP-I and ABT- 510. [0098] Examples of aurora kinase inhibitors include, but are not limited to, VX-680, AZD- 1152 and MLN-8054. Example of polo-like kinase inhibitors include, but are not limited to, BI-2536. [0099] Examples of bcr-abl kinase inhibitors include, but are not limited to, Gleevec (imatinib) and Dasatinib (BMS354825). [00100] Examples of platinum containing agents includes, but are not limited to, cisplatin, Paraplatin (carboplatin), eptaplatin, lobaplatin, nedaplatin, Eloxatin (oxaliplatin) or satraplatin. [00101] Examples of mTOR inhibitors includes, but are not limited to, CCI-779, rapamycin, temsirolimus, everolimus, RAD001, INK-128 and ridaforolimus. [00102] Examples of HSP-90 inhibitors includes, but are not limited to, geldanamycin, radicicol, 17-AAG, KOS-953, 17-DMAG, CNF-101, CNF-1010, 17-AAG-nab, NCS-683664, Mycograb, CNF-2024, PU3, PU24FC1, VER49009, IPI-504, SNX-2112 and STA-9090. [00103] Examples of histone deacetylase inhibitors (HDAC) includes, but are not limited to, Suberoylanilide hydroxamic acid (SAHA), MS-275, valproic acid, TSA, LAQ-824, Trapoxin, tubacin, tubastatin, ACY-1215 and Depsipeptide. [00104] Examples of MEK inhibitors include, but are not limited to, PD325901, ARRY-142886, ARRY-438162 and PD98059. [00105] Examples of CDK inhibitors include, but are not limited to, flavopyridol, MCS-5A, CVT-2584, seliciclib (CYC-202, R-roscovitine), ZK-304709, PHA-690509, BMI-1040, GPC-286199, BMS-387,032, PD0332991 and AZD-5438. [00106] Examples of COX-2 inhibitors include, but are not limited to, CELEBREX™ (celecoxib), parecoxib, deracoxib, ABT-963, MK-663 (etoricoxib), COX-189 Lumiracoxib), BMS347070, RS 57067, NS-398, Bextra (valdecoxib), paracoxib, Vioxx (rofecoxib), SD- 8381, 4- Methyl-2-(3,4-dimethylphenyl)-l-(4-sulfamoyl-ρhenyl-lH-pyrrole, T-614, JTE-522, S-2474, SVT-2016, CT-3, SC-58125 and Arcoxia (etoricoxib). [00107] Examples of non-steroidal anti-inflammatory drugs (NSAIDs) include, but are not limited to, Salsalate (Amigesic), Diflunisal (Dolobid), Ibuprofen (Motrin), Ketoprofen (Orudis), Nabumetone (Relafen), Piroxicam (Feldene), Naproxen (Aleve, Naprosyn), Diclofenac (Voltaren), Indomethacin (Indocin), Sulindac (Clinoril), Tolmetin (Tolectin), Etodolac (Lodine), Ketorolac (Toradol) and Oxaprozin (Daypro). [00108] Exambles of ErbB (e.g. ErbB2) receptor inhibitors include, but are not limited to, CP- 724-714, CI-1033, (canertinib), Herceptin (trastuzumab), Omitarg (2C4, petuzumab), TAK-165, GW- 572016 (Ionafarnib), GW-282974, EKB-569, PI-166, AZD-8931 (sapitinib), dHER2 (HER2 Vaccine), APC8024 (HER2 Vaccine), anti-HER/2neu bispecific antibody, B7.her2IgG3, AS HER2 trifunctional bispecific antibodies, mAB AR-209 and mAB 2B-1. [00109] Exambles of Phosphoinositide 3-kinase inhibitor include, but are not limited to, Wortmannin, LY294002, hibiscone C, Idelalisib, Copanlisib, Duvelisib, Taselisib, Perifosine, Idelalisib, Buparlisib, Duvelisib, Alpelisib, Umbralisib, Copanlisib, PX-866, Dactolisib, CUDC-907, Voxtalisib (also known as SAR245409, XL765), CUDC-907, ME-401, IPI-549, SF1126, RP6530, INK1117, pictilisib, XL147 (also known as SAR245408), Palomid 529, GSK1059615, ZSTK474, PWT33597, IC87114, TG100–115, CAL263, RP6503, PI-103, GNE-477, and AEZS-136. [00110] Examples of alkylating agents include, but are not limited to, nitrogen mustard N- oxide, cyclophosphamide, ifosfamide, trofosfamide, Chlorambucil, melphalan, busulfan, mitobronitol, carboquone, thiotepa, ranimustine, nimustine, temozolomide, AMD-473, altretamine, AP-5280, apaziquone, brostallicin, bendamustine, carmustine, estramustine, fotemustine, glufosfamide, KW- 2170, mafosfamide, and mitolactol, carmustine (BCNU), lomustine (CCNU), Busulfan, Treosulfan, Decarbazine and Temozolomide. [00111] Examples of antimetabolites include but are not limited to, methotrexate, 6- mercaptopurine riboside, mercaptopurine, uracil analogues such as 5-fluorouracil (5-FU) alone or in combination with leucovorin, tegafur, UFT, doxifluridine, carmofur, cytarabine, cytarabine ocfosfate, enocitabine, S-I, Alimta (premetrexed disodium, LY231514, MTA), Gemzar (gemcitabine), fludarabine, 5-azacitidine, capecitabine, cladribine, clofarabine, decitabine, eflornithine, ethnylcytidine, cytosine arabinoside, hydroxyurea, TS-I, melphalan, nelarabine, nolatrexed, ocfosate, disodium premetrexed, pentostatin, pelitrexol, raltitrexed, triapine, trimetrexate, vidarabine, vincristine, vinorelbine, mycophenolic acid, tiazofurin, Ribavirin, EICAR, hydroxyurea and deferoxamine. [00112] Examples of antibiotics include intercalating antibiotics but are not limited to, aclarubicin, actinomycins such as actinomycin D, amrubicin, annamycin, adriamycin, bleomycin a, bleomycin b, daunorubicin, doxorubicin, elsamitrucin, epirbucin, glarbuicin, idarubicin, mitomycin C, nemorubicin, neocarzinostatin, peplomycin, pirarubicin, rebeccamycin, stimalamer, streptozocin, valrubicin, zinostatin and combinations thereof. [00113] Examples of topoisomerase inhibiting agents include, but are not limited to, one or more agents selected from the group consisting of aclarubicin, amonafide, belotecan, camptothecin, 10-hydroxycamptothecin, 9-aminocamptothecin, diflomotecan, irinotecan HCL (Camptosar), edotecarin, epirubicin (Ellence), etoposide, exatecan, gimatecan, lurtotecan, orathecin (Supergen), BN-80915, mitoxantrone, pirarbucin, pixantrone, rubitecan, sobuzoxane, SN-38, tafluposide and topotecan. [00114] Examples of antibodies include, but are not limited to, Rituximab, Cetuximab, Bevacizumab, Trastuzumab, specific CD40 antibodies and specific IGFlR antibodies, [00115] Examples of hormonal therapies include, but are not limited to, exemestane (Aromasin), leuprolide acetate, anastrozole (Arimidex), fosrelin (Zoladex), goserelin, doxercalciferol, fadrozole, formestane, tamoxifen citrate (tamoxifen), Casodex, Abarelix, Trelstar, finasteride, fulvestrant, toremifene, raloxifene, lasofoxifene, letrozole, flutamide, bicalutamide, megesterol, mifepristone, nilutamide, dexamethasone, predisone and other glucocorticoids. [00116] Examples of retinoids/deltoids include, but are not limited to, seocalcitol (EB 1089, CB 1093), lexacalcitrol (KH 1060), fenretinide, Aliretinoin, Bexarotene and LGD-1550. [00117] Examples of plant alkaloids include, but are not limited to, vincristine, vinblastine, vindesine and vinorelbine. [00118] Examples of proteasome inhibitors include, but are not limited to, bortezomib (Velcade), MGl 32, NPI-0052 and PR-171. [00119] Examples of immunologicals include, but are not limited to, interferons and numerous other immune enhancing agents. Interferons include interferon alpha, interferon alpha-2a, interferon, alpha-2b, interferon beta, interferon gamma- 1a, interferon gamma- 1b (Actimmune), or interferon gamma-nl and combinations thereof. Other agents include filgrastim, lentinan, sizofilan, TheraCys, ubenimex, WF-10, aldesleukin, alemtuzumab, BAM-002, decarbazine, daclizumab, denileukin, gemtuzumab ozogamicin, ibritumomab, imiquimod, lenograstim, lentinan, melanoma vaccine (Corixa), molgramostim, OncoVAC- CL, sargaramostim, tasonermin, tecleukin, thymalasin, tositumomab, Virulizin, Z-100, epratuzumab, mitumomab, oregovomab, pemtumomab (Y-muHMFGl), Provenge (Dendreon), CTLA4 (cytotoxic lymphocyte antigen 4) antibodies and agents capable of blocking CTLA4 such as MDX-010. [00120] Examples of biological response modifiers are agents that modify defense mechanisms of living organisms or biological responses, such as survival, growth, or differentiation of tissue cells to direct them to have anti-tumor activity. Such agents include krestin, lentinan, sizofrran, picibanil and ubenimex. [00121] Examples of pyrimidine analogs include, but are not limited to, 5-Fluorouracil, Floxuridine, Doxifluridine, Ratitrexed, cytarabine (ara C), Cytosine arabinoside, Fludarabine, and Gemcitabine. [00122] Examples of purine analogs include but are not limited to, Mercaptopurine and thioguanine. [00123] Examples of antimitotic agents include, but are not limited to, ABT-751, paclitaxel, docetaxel, epothilone D (KOS-862) and ZK-EPO. [00124] The antibodies or antigen binding fragments thereof of the present invention are also intended to be used as a radiosensitizer that enhances the efficacy of radiotherapy. Examples of radiotherapy include but are not limited to, external beam radiotherapy (XBRT), or teletherapy, brachtherapy or sealed source radiotherapy, unsealed source radiotherapy. [00125] The antibodies or antigen binding fragments thereof of the present invention can also be used in combination with a different class of Bcl-2 inhibitors, such as ABT263 or ABT737. [00126] According to some embodiments, the cytotoxic agent may be at least one of gemcitabine and abraxane. [00127] According to yet another embodiment, the additional antibody or therapeutically fragment thereof may be oregovomab antibody B43.13, AR9.6 antibody, or combinations thereof. [00128] According to an embodiment, the chemotherapy regimen may be Folfirinox. [00129] In embodiments of the present invention, the tumor may be chosen from a pancreatic tumor, a gall bladder tumor, a gastric tumor, a colon tumor, an ovarian tumor, a breast tumor, and a liver tumor, and the method may be for the treatment of a cancer. [00130] In another embodiment, there is disclosed a use of an antibody or an antigen binding fragment thereof that targets O-glycan mucin-type glycoprotein MUC16, according to the present invention, or of a composition according to the present invention, for inhibiting tumor growth of a tumor expressing O-glycan mucin-type glycoprotein MUC16 in a subject in need thereof. [00131] In another embodiment, there is disclosed an antibody or an antigen binding fragment thereof that targets O-glycan mucin-type glycoprotein MUC16, according to the present invention for use in inhibiting tumor growth of a tumor expressing O-glycan mucin-type glycoprotein MUC16 in a subject in need thereof. [00132] In another embodiment, there is disclosed an antibody or an antigen binding fragment thereof that targets O-glycan mucin-type glycoprotein MUC16, according to the present invention for use in a method of inhibiting tumor growth of a tumor expressing O-glycan mucin-type glycoprotein MUC16 in a subject in need thereof. [00133] In another embodiment, there is disclosed a method of detection of a tumor expressing O-glycan mucin-type glycoprotein MUC16 in a subject in need thereof, comprising administering to the subject an antibody or an antigen binding fragment thereof specific to O-glycan mucin-type glycoprotein MUC16 according to the present invention and detecting the antibody or antigen binding fragment. According to an embodiment, the antibody or antigen binding fragment thereof may further comprise a detectable label, for example a fluorescent marker, a radioactive marker, an MRI contrast agent, or combinations thereof, as is known in the art. [00134] The invention also encompasses nucleic acid vector comprising a nucleotide sequence encoding the antibody or antigen binding fragment thereof of the present invention, as well as cells comprising the nucleic acid vector, for expressing the the antibody or antigen binding fragment thereof of the present invention, and cells for expressing the the antibody or antigen binding fragment thereof of the present invention. [00135] The present invention will be more readily understood by referring to the following examples which are given to illustrate the invention rather than to limit its scope. EXAMPLE 1 Anti-Her2/Neu monoclonal antibody generation [00136] The scFv C6MH3-B1 specific for HER2/neu has previously been used in the preparation of monoclonal antibodies, particularly IgE monoclonal antibodies (Daniels, T.R. et al. Cancer Immunol Immunother, 2012 Jul;61(7):991-1003). However, this antibody has failed to become useful as a therapeutic because low production yield prevent it from being made in sufficient quantities and in a form that would be useable for therapy. This could be caused by numerous issues in addition to self-aggregation, oxidation, deamination and isomerization of amino acid chains. [00137] The sequence of the anti-Her2/Neu IgE antibody derived from the C6MH3-B1 scFv comprises a full-length Lambda isotype light chain and a full length IgE heavy chain. As a first step in modifying the structure to improve production yield, the sequence of the entire IgE heavy chain (SEQ ID NO: 13), and particularly the heavy chain variable region from FR1 to FR4 (SEQ ID NO: 14) was point mutated at numerous amino acid residues. The wild type and the mutant variant sequence of the antibody C6MH3-B1 were transiently expressed in Chinese Hamster Ovary cells (CHOK1SV GS- KO) using small scale transient expression, to screen for both improved expression and binding to the antigen. Mutations that caused a decrease in or no improvement on the level of expression were discarded in a first pass. Those that decreased binding of the antibodies to the antigen were also discarded in a first pass. The selected variants were recloned by limiting dilutions to ensure their monoclonality, and sequenced. From the numerous mutants identified, mutations in the CDR2 region were identified and selected for further characterization. Three mutations of the first aspartic acid (D) residue of CDR H2 were identified: D to E (variant 1), D to Q (variant 2), and D to S (variant 3). Two mutations of the first serine (S) residue of CDR H2 were identified: S to T (variant 4) and S to I (variant 5). [00138] Antibodies were produced for the wild type and the mutant variants 1 to 5 from transient expression in Chinese Hamster Ovary cells (CHOK1SV GS-KO) using small scale transient expression HiTrap® LambdaFabSelect resin (Sigma®) and buffer exchanged into PBS containing 3 mM EDTA. EXAMPLE 2 Anti-Her2/Neu monoclonal antibody yield measurement [00139] During extraction with the LambdaFabSelect resin, an estimation of the titre of antibody was made from the obtained chromatograms (no shown). The results are summarized in Table 1, below:
Figure imgf000029_0001
Table 1 - Estimation of titre from LambdaFabSelect chromatogram [00140] The results show that unexpectedly, cells expressing the variants 1 to 3 and 5 were able to increase expression and provide higher yields of the antibodies than the parental C6MH3-B1 IgE antibody. Variant 4 performed only slightly better than the parental antibody. EXAMPLE 3 Anti-Her2/Neu monoclonal antibody purification and aggregation [00141] Pursuant to elution of the purified antibodies from the LambdaFabSelect resin, they were subjected to size exclusion chromatography (SEC) using a Superdex® S7510/300 GL column. The results are summarized in Table 2, below:
Figure imgf000030_0001
Table 2 - SEC analysis of the eluted fractions from LambdaFabSelect Column containing antibodies [00142] The results show that unexpectedly, variants 1, 4 and 5 eluted as monomeric antibodies and showed no aggregates and no or only small amount of low molecular weight (LMW) products. Variant 2 eluted as two peaks, the first of which unexpectedly comprised of monomers, while the second comprised a significant portion of undesirable aggregates (31.03%) and LMW products (13.36). Variant 3 eluted as one peak comprised of undesirable aggregates (52.23%), monomers (32.80) and low molecular weight (LMW) products (14.97%). Therefore, variants 1, 2, 4 and 5 showed an unexpected improvement: variants 1, 4 and 5 eluted as a single peak with no aggregation, while variant 2 could be purified in unaggregated form from the first peak. EXAMPLE 4 Anti-Her2/Neu monoclonal antibody affinity [00143] Next, the binding of variants 1, 2, 4 and 5 was compared to that of the parental antibody by flow cytometry. D2F2/E2 cells [Wei et al. Int J Cancer, 1999 May 31;81(5):748-54] expressing human Her2/Neu were cultured. Cells were detached from tissue culture dishes using 0.5 mM EDTA and incubated with varying concentrations of the various antibodies for 2 h on ice. Cells were then washed in buffer (2 mM EDTA, 0.5% BSA in PBS) and binding was detected using an anti- human ^ fluorescein isothiocyanate (FITC)-conjugated secondary antibody (BD Biosciences, San Jose, CA). Cells were also stained with an anti-Fc ^RI phycoerythrin (PE)-conjugate (eBioscience, San Diego, CA) to ensure expression of the receptor on the surface of the cells. All cells were washed, fixed with 2% paraformaldehyde in PBS, and analyzed on a Becton–Dickinson FACScan Analytic Flow Cytometer. Now referring to Fig. 1, the results show that unexpectedly, despite the mutations introduced into the CDR H2 region, variants 1, 2, 4 and 5 are all able to bind to Her2/Neu with affinities that are at least as good as that of the wild-type parental antibody. Indeed, variants 1 and 2 have affinities that are close to, or even a little better than the parental antibody (Figs.1A and 1B). Variants 4 and 5, on the other hand, appear to display affinities that are unexpectedly improved over that of the wild-type parental antibody (Figs.1C and 1D). [00144] While preferred embodiments have been described above and illustrated in the accompanying drawings, it will be evident to those skilled in the art that modifications may be made without departing from this disclosure. Such modifications are considered as possible variants comprised in the scope of the disclosure. SEQUENCE TABLE
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
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

Claims

CLAIMS: 1. An antibody or an antigen-binding fragment that binds specifically to Her2/Neu comprising three variable heavy domain complementarity determining regions (CDR) (CDR H1, H2 and H3) wherein the CDR H1, H2, and H3, comprise an amino acid sequence comprising: CDR H1: GYSFTSYW (SEQ ID NO:1), CDR H2: IYPGX1X2DT, where X1 is D, E, or Q, X2 is S, I, or T, and wherein when X1 is D, X2 is different than S (SEQ ID NO:2), and CDR H3: ARHDVGYCTDRTCAKWPEY (SEQ ID NO:3), respectively.
2. The antibody or an antigen-binding fragment of claim 1, comprising three variable light domain CDR (CDR L1, L2 and L3), wherein the CDR L1, L2, and L3 comprise an amino acid sequence comprising: CDR L1: SSNIGNNY (SEQ ID NO:4), CDR L2: DHT (SEQ ID NO:5), and CDR L3: ASWDYTLSGWV (SEQ ID NO:6), respectively.
3. The antibody or antigen-binding fragment of claim 1 or 2, wherein when X1 is D, X2 is I, or T.
4. The antibody or antigen-binding fragment of claim 1 or 2, wherein when X1 is E, or Q, X2 is S.
5. The antibody or antigen-binding fragment of claim 1 or 2, wherein X1 is E and X2 is S.
6. The antibody or antigen-binding fragment of claim 1 or 2, wherein X1 is Q and X2 is S.
7. The antibody or antigen-binding fragment of claim 1 or 2, wherein X1 is D and X2 is I.
8. The antibody or antigen-binding fragment of claim 1 or 2, wherein X1 is D and X2 is T.
9. The antibody or antigen-binding fragment of any one of claims 1 - 8, further comprising four variable heavy domain framework regions (HFR)(HFR 1, 2, 3 and 4), wherein said HFR 1, 2, 3, and 4 comprise an amino acid sequence comprising: HFR 1: VQLVQSGAEVKKPGESLKISCKGS (SEQ ID NO: 15), HFR 2: IAWVRQMPGKGLEYMGL (SEQ ID NO: 16), HFR 3: KYSPSFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFC (SEQ ID NO: 17), and HFR 4: WGQGTLVTV (SEQ ID NO: 18).
10. The antibody or antigen-binding fragment of any one of claims 1 - 8, further comprising four variable light domain framework regions (LFR)(LFR 1, 2, 3 and 4), wherein said LFR 1, 2, 3, and 4 comprise an amino acid sequence comprising: LFR 1: QSVLTQPPSVSAAPGQKVTISCSGS (SEQ ID NO: 84), LFR 2: VSWYQQLPGTAPKLLIY (SEQ ID NO: 85), LFR 3: NRPAGVPDRFSGSKSGTSASLAISGFRSEDEADYYC (SEQ ID NO: 86), and LFR 4: FGGGTKVTVL (SEQ ID NO: 87).
11. The antibody or antigen-binding fragment of claim 1 or 2, comprising a variable heavy domain (VH) comprising amino acid sequence comprising: VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 23).
12. The antibody or antigen binding fragment of claim 1 or 2, comprising a variable light domain (VL) comprising amino acid sequence comprising: QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPDRFSGS KSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (SEQ ID NO: 83).
13. The antibody or antigen-binding fragment of claim 1 or 2, comprising a variable heavy domain (VH) comprising amino acid sequence comprising: VQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYPGESDTKYSPSFQG QVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEYFQHWGQGTLVTV (SEQ ID NO: 23) and a variable light domain (VL) comprising amino acid sequence comprising: QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDHTNRPAGVPDRFSGS KSGTSASLAISGFRSEDEADYYCASWDYTLSGWVFGGGTKVTVL (SEQ ID NO: 83).
14. The antibody or antigen-binding fragment of any one of claims 1 to 12, wherein the antigen- binding fragment is a single-domain antibody (sdAb), a fragment antigen binding (Fab), a single- chain variable fragment (scFv), or a single-chain fragment antigen binding (scFab).
15. The antibody or antigen-binding fragment of claim 1, wherein the antibody is an IgA, an IgD, an IgE, an IgG, or an IgM.
16. The antibody or antigen-binding fragment of claim 15, wherein the antibody is an IgE.
17. The antibody or antigen-binding fragment of any one of claims 1 to 16, wherein the antibody or antigen-binding fragment is humanized or partially humanized.
18. A compound comprising the antibody or antigen-binding fragment of any one of claims 1 to 17, and a functional moiety.
19. The compound of claim 18, wherein the antibody or antigen-binding fragment is linked to the functional moiety via a peptide linker.
20. The compound of claim 19, wherein the antibody or antigen-binding fragment is functionally linked to the functional moiety via the peptide linker.
21. The compound of claim 20, wherein the peptide linker comprises about 3 to about 40 amino acid residues.
22. The compound of any one of claims 20 to 21, wherein the peptide linker comprises the amino acid sequence (GGGGS)n or (GGGS)n, wherein n ≥ 1.
23. The compound of any one of claims 18 to 22, wherein the antibody or antigen-binding fragment is fused to a second antibody or antigen-binding fragment operable to bind a target epitope.
24. The compound of any one of claims 18 to 23, wherein the antibody or antigen-binding fragment is linked to a peptide, a polypeptide, a protein, an enzyme, a second antibody, an antibody fragment, a second antigen-binding fragment or a combination of any two or more thereof; wherein each of the antibody or antigen-binding fragment thereof and the linked peptide, polypeptide, protein, enzyme, second antibody, antibody fragment, second antigen-binding fragment, or the combination of any two or more thereof is functional.
25. The compound of claim 24, wherein the antibody fragment is a fragment crystallizable (Fc) region.
26. A composition comprising the antibody or antigen-binding fragment of any one of claims 1 to 17 or the compound of any one of claims 18 to 25, and a pharmaceutically acceptable diluent, carrier or excipient.
27. A nucleic acid molecule encoding an antibody or antigen-binding fragment of any one of claims 1 to 17, or encoding a compound of any one of claims 18 to 25.
28. A vector comprising the nucleic acid molecule of claim 27 operably linked to one or more regulatory elements to allow expression of the antibody or antigen-binding fragment of any one of claims 1 to 17, or the compound of any one of claims 18 to 25 in a host cell.
29. A cell comprising the vector of claim 28 for expressing the antibody or antigen-binding fragment thereof of any one of claims 1 to 17 or the compound of any one of claims 18 to 25.
30. A method of treating a Her2/Neu associated disease comprising: administering the antibody or antigen-binding fragment of any one of claims 1 to 17, the compound of any one of claims 18 to 25, the composition of claim 26, or a combination of any two or more thereof to a subject in need thereof.
31. The method of claim 30, wherein the Her2/Neu associated disease is a cancer.
32. The method of claim 31, wherein the cancer is ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
33. The use of the antibody or antigen-binding fragment of any one of claims 1 to 17, the compound of any one of claims 18 to 25, the composition of claim 26, or a combination of any two or more thereof to a subject in need thereof in the treatment of a Her2/Neu associated disease.
34. The use of claim 33, wherein the Her2/Neu associated disease is a cancer.
35. The use of claim 34, wherein the cancer is ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
36. The antibody or antigen-binding fragment of any one of claims 1 to 17, the compound of any one of claims 18 to 25, the composition of claim 26, or a combination of any two or more thereof to a subject in need thereof, for use in the treatment of a Her2/Neu associated disease.
37. The antibody or antigen-binding fragment, compound, composition, or combination of any two or more thereof of claim 36, wherein the Her2/Neu associated disease is a cancer.
38. The antibody or antigen-binding fragment, compound, composition, or combination of any two or more thereof of claim 37, wherein the cancer is ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
39. The use of the antibody or antigen-binding fragment of any one of claims 1 to 17, the compound of any one of claims 18 to 25, the composition of claim 26, or a combination of any two or more thereof to a subject in need thereof in the manufacture of a medicament for the treatment of a Her2/Neu associated disease.
40. The use of claim 39, wherein the Her2/Neu associated disease is a cancer.
41. The use of claim 40, wherein the cancer is ovarian cancer, breast cancer, stomach cancer, lung cancer, uterine cancer, salivary gland cancer, testicular germ cell cancer, bladder cancer, pancreatic cancer, and esophageal cancer.
PCT/IB2021/057644 2021-08-19 2021-08-19 Monoclonal antibodies against her2/neu and uses thereof WO2023021319A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/IB2021/057644 WO2023021319A1 (en) 2021-08-19 2021-08-19 Monoclonal antibodies against her2/neu and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IB2021/057644 WO2023021319A1 (en) 2021-08-19 2021-08-19 Monoclonal antibodies against her2/neu and uses thereof

Publications (1)

Publication Number Publication Date
WO2023021319A1 true WO2023021319A1 (en) 2023-02-23

Family

ID=85240108

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/057644 WO2023021319A1 (en) 2021-08-19 2021-08-19 Monoclonal antibodies against her2/neu and uses thereof

Country Status (1)

Country Link
WO (1) WO2023021319A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060099205A1 (en) * 2002-04-05 2006-05-11 The Regents Of The University Of California Bispecific single chain FV antibody molecules and methods of use thereof
EP1210372B1 (en) * 1999-07-29 2008-01-23 Medarex, Inc. Human monoclonal antibodies to her2/neu
WO2009123894A2 (en) * 2008-04-02 2009-10-08 Macrogenics, Inc. Her2/neu-specific antibodies and methods of using same
KR20140103135A (en) * 2011-12-05 2014-08-25 노파르티스 아게 Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
US20160130359A1 (en) * 2013-06-11 2016-05-12 The United States of America, as represented by The Secretary, Dep of Health and Human Services Her2-specific monoclonal antibodies and conjugates thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1210372B1 (en) * 1999-07-29 2008-01-23 Medarex, Inc. Human monoclonal antibodies to her2/neu
US20060099205A1 (en) * 2002-04-05 2006-05-11 The Regents Of The University Of California Bispecific single chain FV antibody molecules and methods of use thereof
WO2009123894A2 (en) * 2008-04-02 2009-10-08 Macrogenics, Inc. Her2/neu-specific antibodies and methods of using same
KR20140103135A (en) * 2011-12-05 2014-08-25 노파르티스 아게 Antibodies for epidermal growth factor receptor 3 (her3) directed to domain ii of her3
US20160130359A1 (en) * 2013-06-11 2016-05-12 The United States of America, as represented by The Secretary, Dep of Health and Human Services Her2-specific monoclonal antibodies and conjugates thereof

Similar Documents

Publication Publication Date Title
JP6870161B1 (en) Anti-cMet antibody drug conjugate and how to use it
US10730953B2 (en) Anti-PD-1 antibodies and their uses
JP2019500327A (en) Anti-huLRRC15 antibody drug conjugate and method of use thereof
JP2019501124A (en) Anti-huLRRC15 antibody drug conjugate and method of use thereof
CN104254544A (en) Cdim binding proteins and uses thereof
US11773183B2 (en) MUC16 monoclonal antibody and uses thereof
US20200101159A1 (en) Treatment of cancer with therapeutic monoclonal antibody specific for a tumor associated antigen and an immune adjuvant
US20230070306A1 (en) Monoclonal antibodies against her2/neu and uses thereof
WO2023021319A1 (en) Monoclonal antibodies against her2/neu and uses thereof
EP4357362A1 (en) Bispecific antibody combination and use thereof
US10023652B2 (en) Method of targeting glycoproteins to treat cancer
WO2023023871A1 (en) Combination of an ige monoclonal antibody specific to a cancer antigen and nk cell for the treatment of cancer

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: 21954107

Country of ref document: EP

Kind code of ref document: A1