WO2023187407A1 - Human monoclonal antibodies binding to sars-cov-2 and methods of use thereof - Google Patents

Human monoclonal antibodies binding to sars-cov-2 and methods of use thereof Download PDF

Info

Publication number
WO2023187407A1
WO2023187407A1 PCT/GB2023/050858 GB2023050858W WO2023187407A1 WO 2023187407 A1 WO2023187407 A1 WO 2023187407A1 GB 2023050858 W GB2023050858 W GB 2023050858W WO 2023187407 A1 WO2023187407 A1 WO 2023187407A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
fragment
sequences
clone
paired
Prior art date
Application number
PCT/GB2023/050858
Other languages
French (fr)
Inventor
Khelan Pravinkumar PATEL
Original Assignee
Bradcode Limited
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 Bradcode Limited filed Critical Bradcode Limited
Publication of WO2023187407A1 publication Critical patent/WO2023187407A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1002Coronaviridae
    • C07K16/1003Severe acute respiratory syndrome coronavirus 2 [SARS‐CoV‐2 or Covid-19]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/165Coronaviridae, e.g. avian infectious bronchitis virus
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2469/00Immunoassays for the detection of microorganisms
    • G01N2469/10Detection of antigens from microorganism in sample from host

Definitions

  • the present invention provides SARS-CoV-2 antibody molecules and their uses. More particularly, the presently disclosed invention provides human antibody molecules which bind to a novel coronavirus designated and methods of use thereof.
  • SARS-CoV-2 An epidemic of a novel coronavirus (SARS-CoV-2) affected mainland China, along with cases in more than 220 other countries and territories. It was identified in Wuhan, the capital of China's Hubei province, after 41 people developed pneumonia without a clear cause.
  • the virus which causes acute respiratory disease designated coronavirus disease 2019 (COVID-19), is capable of spreading from person to person.
  • the incubation period ranges from 0 to 24 days, with a mean of 3-5 days, but it may be contagious during this period and after recovery. Symptoms include fever, coughing and breathing difficulties. As of January 2022, over 350 million cases have been confirmed worldwide. A larger number of people may have been but not detected (especially mild cases). As of January 2022, over 5.6 million deaths have been attributed to the virus since the first confirmed death on 9 January 2020. The first local transmission outside China occurred in Vietnam between family members, while the first international transmission not involving family occurred in Germany on 22 January 2020. The first death outside China was in the Philippines, where a man from Wuhan died on 1 February 2020.
  • SARS-CoV-2 variants contain an N439K mutation, which has enhanced binding affinity to the human ACE2 receptor (Thomson, E.C., et al., "The circulating SARS-CoV-2 Spike variant N439K maintains fitness while evading antibody-mediated immunity.” bioRxiv, 2020).
  • SARS-CoV-2 variants contain an N501Y mutation, which is associated with increased transmissibility, including the lineages B.1.1.7 (also known as WHO Alpha, 20l/501Y.Vl and VOC 202012/01) and B.1.351 (also known as WHO Beta, 20H/501Y.V2), which were discovered in the United Kingdom and South Africa, respectively (Tegally, H., et al., "Emergence and rapid spread of a new severe acute respiratory syndrome- related coronavirus 2 (SARS-CoV-2) lineage with multiple Spike mutations in South Africa.” medRxiv, 2020: p.2020.12.21.20248640; Leung, K., et al., "Early empirical assessment of the N501Y mutant strains of SARS-CoV-2 in the United Kingdom, October to November 2020.” medRxiv, 2020: p.2020.12.20.20248581).
  • B.1.351 also include two other mutations in the RBD domain of SARS-CoV-2 Spike protein, K417N and E484K (Tegally, H., et al., "Emergence and rapid spread of a new severe acute respiratory syndrome- related coronavirus 2 (SARS-CoV-2) lineage with multiple Spike mutations in South Africa.” medRxiv, 2020: p.2020.12.21.20248640).
  • a further variant of concern is the Gamma variant (P.1) which was identified in Brazil in May 2020 and has substitutions N501Y, E484K and K417T in the SARS-CoV-2 spike protein (Faria, Nuno R.; Claro, Ingra Morales (12 January 2021).
  • the Omicron variant (B.1.1.529) was first reported to the World Health Organization (WHO) from South Africa.
  • the Omicron variant has a total of 60 mutations as compared to the originally identified virus. Thirty-two mutations affect the spike protein, the main antigenic target of antibodies generated by infections and of many vaccines widely administered (Callaway E (December 2021). "Heavily mutated Omicron variant puts scientists on alert”. Nature.600 (7887): 21. Bibcode:2021Natur.600). Many of those mutations had not been observed in other strains. Thus, there remains an urgent need to explore the biology and pathology of SARS-CoV-2 and well as the human immune response to this virus.
  • SARS-CoV-2 is a major health concern with active cases increasing daily. Therefore, understanding the biology of this virus and the nature and extent of the human immune response to the virus is of paramount importance.
  • the inventors have identified the sequences of human antibodies to SARS-CoV-2. Those sequences and uses for such antibodies are disclosed herein.
  • the present invention addresses these and other related needs by providing, inter alia, antibody molecules that are capable of binding to SARS-CoV-2.
  • the present invention provides a monoclonal antibody molecule, wherein the antibody or antibody fragment is characterized by clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
  • the antibody or antibody fragment may be encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having 100% identity to clone-paired sequences as set forth in Table 1.
  • the antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, or light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2.
  • the antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
  • the antibody may be a chimeric antibody, is bispecific antibody, or is an intrabody.
  • the antibody may be an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
  • the antibody or antibody fragment may bind to a SARS-CoV-2 surface spike protein.
  • the antibody or antibody fragment may be encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having 100% identity to clone-paired sequences as set forth in Table 1.
  • the antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, or light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2.
  • the antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
  • the antibody may be a chimeric antibody, a bispecific antibody, or an intrabody.
  • the antibody may bean IgG, or a recombinant IgG antibody or antibody fragment comprising an Fe portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
  • the antibody or antibody fragment maybind to a SARS-CoV-2 surface spike protein.
  • the antibody molecule is a purified or isolated antibody molecule.
  • the antibody molecule may be a polyclonal antibody, a monoclonal antibody, a human antibody, a humanized antibody, a chimeric antibody, fragment of an antibody or monoclonal antibody, in particular a Fab, Fab', or F(ab')2 fragment, a single chain antibody, in particular a single chain variable fragment (scFv), a domain antibody, a nanobody, a diabody, or a DARPin.
  • a polyclonal antibody a monoclonal antibody, a human antibody, a humanized antibody, a chimeric antibody, fragment of an antibody or monoclonal antibody, in particular a Fab, Fab', or F(ab')2 fragment, a single chain antibody, in particular a single chain variable fragment (scFv), a domain antibody, a nanobody, a diabody, or a DARPin.
  • the antibody molecule may be a humanized antibody or a fragment of a humanised antibody, in particular a Fab, Fab', or F(ab')2 fragment, a single chain antibody, in particular a single chain variable fragment (scFv), a Small Modular lmmunopharmaceutical (SMIP), a domain antibody, a nanobody, a diabody, or a Designed Ankyrin Repeat Protein (DARPin).
  • the present invention provides a pharmaceutical composition comprising an antibody molecule of the present invention and a pharmaceutically acceptable carrier. To be used in therapy, the antibody molecule is included into pharmaceutical compositions appropriate to facilitate administration to animals or humans.
  • Suitable formulations of the antibody molecule may be prepared by mixing the antibody molecule with physiologically acceptable carriers, excipients or stabilizers, in the form of lyophilized or otherwise dried formulations or aqueous solutions or aqueous or non-aqueous suspensions.
  • Carriers, excipients, modifiers or stabilizers are nontoxic at the dosages and concentrations employed.
  • buffer systems such as phosphate, citrate, acetate and other inorganic or organic acids and their salts; antioxidants including ascorbic acid and methionine; preservatives such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone or polyethylene glycol (PEG); amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, oligosaccharides,
  • the pharmaceutical composition comprises the antibody molecule of the present invention in an aqueous, buffered solution, or a lyophilisate made from such a solution.
  • a suitable mode of application is parenteral, by infusion or injection (intraveneous, intramuscular, subcutaneous, intraperitoneal, intradermal), but other modes of application such as by inhalation, transdermal, intranasal, buccal, oral, may also be applicable.
  • the present invention provides an antibody molecule of the present invention for use as a medicament.
  • the present invention provides a a method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, comprising delivering to the subject an antibody or antibody fragment having clone- paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
  • the antibody or antibody fragment may be encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having 100% identity to clone-paired sequences as set forth in Table 1.
  • the antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, or light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2.
  • the antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab'h fragment, or Fv fragment.
  • the antibody may be a chimeric antibody or a bispecific antibody.
  • the antibody may be an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
  • the antibody or antibody fragment may bind to a SARS-CoV-2 antigen such as a surface spike protein.
  • the antibody or antibody fragment may be administered prior to infection or after infection.
  • the subject may be of age 60 or older, may be immunocompromised, or may suffer from a respiratory and/or cardiovascular disorder.
  • Delivering may comprise antibody or antibody fragment administration, or genetic delivery with an RNA or DNA sequence or vector encoding the antibody or antibody fragment.
  • the present invention provides an antibody molecule of the invention for use in treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2.
  • the present invention provides the use of an antibody molecule of the invention in the manufacture of a medicament for treating a subject infected with SARS- CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV- 2.
  • the antibody molecule of the invention may be administered either simultaneously with, or before or after, one or more other therapeutic agent.
  • the antibody molecule of the invention may be administered separately, by the same or different route of administration, or together in the same pharmaceutical composition as the other agents.
  • the invention provides a product comprising an antibody molecule of the invention and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy.
  • the therapy is treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2.
  • Products provided as a combined preparation include a composition comprising the antibody molecule of the invention and the other therapeutic agent(s) together in the same pharmaceutical composition, or the antibody molecule of the invention and the other therapeutic agent(s) in separate form, e.g. in the form of a kit.
  • the invention provides a pharmaceutical composition comprising an antibody molecule of the invention and another therapeutic agent(s).
  • the pharmaceutical composition may comprise a pharmaceutically acceptable excipient, as described above.
  • the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains an antibody molecule of the invention.
  • the kit comprises means for separately retaining the compositions, such as a container, divided bottle, or divided foil packet.
  • kits are blister pack, as typically used for the packaging of tablets, capsules and the like.
  • the kit of the invention may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another.
  • the kit of the invention typically comprises directions for administration.
  • the antibody molecule of the invention and the other therapeutic agent may be manufactured and/or formulated by the same or different manufacturers.
  • the antibody molecule of the invention and the other therapeutic may be brought together into a combination therapy: (i) prior to release of the combination product to physicians (e.g.
  • the invention provides the use of an antibody molecule of the invention in the manufacture of a medicament for treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the medicament is prepared for administration with another therapeutic agent.
  • the invention also provides the use of another therapeutic agent for treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the medicament is administered with an antibody molecule of the invention.
  • the invention also provides an antibody molecule of the invention for use in a method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the antibody molecule of the invention is prepared for administration with another therapeutic agent.
  • the invention also provides another therapeutic agent for use in a method of treating a subject infected with SARS- CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV- 2, wherein the other therapeutic agent is prepared for administration with an antibody molecule of the invention.
  • the invention also provides an antibody molecule of the invention for use in a method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2 wherein the antibody molecule of the invention is administered with another therapeutic agent.
  • the invention also provides another therapeutic agent for use in a method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the other therapeutic agent is administered with an antibody molecule of the invention.
  • the invention also provides the use of an antibody molecule of the invention for treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the subject has previously (e.g. within 24 hours) been treated with another therapeutic agent.
  • the invention also provides the use of another therapeutic agent for treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the subject has previously (e.g. within 24 hours) been treated with an antibody molecule of the invention.
  • the other therapeutic agents include a further antibody molecule of the invention.
  • the two or more antibody molecules of the present invention bind to a SARS-CoV-2 surface spike protein.
  • one of the two or more antibody molecules of the invention binds to a SARS- CoV-2 surface spike protein and another of the two or more antibody molecules of the invention binds to a SARS-CoV-2 surface nucleocapsid (N) protein.
  • the invention includes as a further aspect a combination of two or more antibody molecules of the invention.
  • the other therapeutic agents include a further antibody molecule.
  • the antibody molecule of the present invention and the further antibody molecule bind to a SARS-CoV-2 surface spike protein.
  • the antibody molecule of the invention binds to a SARS-CoV-2 surface spike protein and further antibody molecule binds to a SARS-CoV-2 surface nucleocapsid (N) protein.
  • the further antibody molecule is a monoclonal antibody optionally selected from bamlanivimab; bebtelovimab, casirivimab; cilgavimab; etesevimab; imdevimab; sotrovimab; tixagevimab or combinations thereof such as bamlanivimab/ etesevimab, casirivimab/ imdevimab, or tixagevimab/ cilgavimab.
  • the invention includes as a further aspect a combination of an antibody molecule of the invention with one or more further antibody molecules.
  • the other therapeutic agent(s) include an antibody product wherein the antibody product is optionally selected from plasma provided by donors who have recovered from SARS-CoV-2, and SARS-CoV-2 immunoglobulins derived therefrom.
  • the other therapeutic agent(s) include an anti-viral agent optionally selected from the group consisting of remdesivir, molnupiravir (Lagevrio), Ritonavir- Boosted Nirmatrelvir (Paxlovid)..
  • the invention includes as a further aspect a combination of an antibody of the invention with an anti-viral agent optionally selected from the group consisting of remdisivir, molnupiravir (Lagevrio), Ritonavir-Boosted Nirmatrelvir (Paxlovid).
  • the other therapeutic agent(s) include a cell based therapy such as mesenchymal stem cells.
  • the invention includes as a further aspect a combination of an antibody of the invention with a cell based therapy such as mesenchymal stem cells.
  • the other therapeutic agent(s) include an immunomodulator which is optionally selected from the group consisting of corticosteroids, Interleukin-1 inhibitors, Interleukin-6 inhibitors: kinase inhibitors. Granulocyte-Macrophage Colony-Stimulating Factor Inhibitors, colchicine and fluvoxamine.
  • the corticosteroid is an oral systemic corticosteroid such as dexamethasone or an inhaled corticosteroid such as budesonide or ciclesonide.
  • the Interleukin-1 inhibitor is optionally selected from the group consisting of anakinra and canakinumab.
  • the Interleukin-6 inhibitor is optionally selected from the group consisting of sarilumab, tocilizumab and siltuximab.
  • the kinase inhibitor is a Janus Kinase inhibitor optionally selected from the group consisting of baricitinib, tofacitinib and ruxolitinib: Accordingly, the invention includes as a further aspect a combination of an antibody of the invention with an immunomodulator which is optionally selected from the group consisting of corticosteroids, Interleukin-1 inhibitors, Interleukin-6 inhibitors: kinase inhibitors.
  • the other therapeutic agent(s) include an antithrombotic
  • the antithrombotic is an anticoagulant, an antiplatelet or a thrombolytic.
  • the invention includes as a further aspect a combination of an antibody of the invention with an antithrombotic.
  • a vaccine formulation comprising one or more antibodies or antibody fragments characterized by clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
  • the at least one of the antibodies or antibody fragments may be encoded by light and heavy chain variable sequences according to clone- paired sequences from Table 1, by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired sequences from Table 1, or by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 1.
  • the at least one of the antibodies or antibody fragments may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2 or may comprise light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone- paired sequences from Table 2.
  • the at least one of the antibody fragments is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
  • the at least one of the antibodies may a chimeric antibody, a bispecific antibody or an intrabody.
  • the antibody may be an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
  • the antibody or antibody fragment may bind to a SARS-CoV-2 antigen surface spike protein.
  • a vaccine formulation comprising one or more expression vectors encoding a first antibody or antibody fragment as described herein.
  • the expression vector(s) may be Sindbis virus or VEE vector(s).
  • the vaccine may be formulated for delivery by needle injection, jet injection, or electroporation.
  • the vaccine formulation may further comprise one or more expression vectors encoding for a second antibody or antibody fragment, such as a distinct antibody or antibody fragment of claims 26- to34.
  • a method of protecting the health of a subject of age 60 or older, an immunocompromised, subject or a subject suffering from a respiratory and/or cardiovascular disorder that is infected with or at risk of infection with SARS-CoV-2 comprising delivering to the subject an antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
  • the antibody or antibody fragment may be encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having 100% identity to clone- paired sequences as set forth in Table 1.
  • the antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, or light and heavy variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2.
  • the antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab'h fragment, or Fv fragment.
  • the antibody may an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
  • the antibody may be a chimeric antibody or a bispecific antibody.
  • the antibody or antibody fragment may be administered prior to infection or after infection.
  • the antibody or antibody fragment may bind to a SARS-CoV-2 antigen such as a surface spike protein.
  • Delivering may comprise antibody or antibody fragment administration, or genetic delivery with an RNA or DNA sequence or vector encoding the antibody or antibody fragment.
  • the antibody or antibody fragment may improve the subject's respiration as compared to an untreated control and/or may reduce viral load as compared to an untreated control.
  • a method of determining the antigenic integrity, correct conformation and/or correct sequence of a SARS-CoV-2 surface spike protein comprising (a) contacting a sample comprising the antigen with a first antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively; and (b) determining antigenic integrity, correct conformation and/or correct sequence of the antigen by detectable binding of the first antibody or antibody fragment to the antigen.
  • the sample may comprise recombinantly produced antigen or a vaccine formulation or vaccine production batch.
  • Detection may comprise ELISA, RIA, western blot, a biosensor using surface plasmon resonance or biolayer interferometry, or flow cytometric staining.
  • the first antibody or antibody fragment may be encoded by clone- paired variable sequences as set forth in Table 1, by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences as set forth in Table 1.
  • the first antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, may comprise light and heavy chain variable sequences having at least 70%, 80% or 90% identity to clone-paired sequences from Table 2, or may comprise light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 2.
  • the first antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
  • the method may further comprise performing steps (a) and (b) a second time to determine the antigenic stability of the antigen over time.
  • the method may further comprise (c) contacting a sample comprising the antigen with a second antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively; and (d) determining antigenic integrity of the antigen by detectable binding of the second antibody or antibody fragment to the antigen.
  • the second antibody or antibody fragment may be encoded by clone-paired variable sequences as set forth in Table 1, by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences as set forth in Table 1.
  • the second antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, may comprise light and heavy chain variable sequences having at least 70%, 80% or 90% identity to clone-paired sequences from Table 2, or may comprise light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 2.
  • the second antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
  • the method may further comprise performing steps (c) and (d) a second time to determine the antigenic stability of the antigen over time.
  • the present invention provides a kit comprising an antibody molecule of the present invention, or a pharmaceutical composition thereof.
  • the present invention provides a method of manufacturing an antibody molecule of the present invention, comprising: (a) providing a host cell comprising one or more nucleic acids encoding the antibody molecule in functional association with an expression control sequence, (b) cultivating the host cell, and (c) recovering the antibody molecule from the cell culture.
  • the virus has often been referred to in common parlance as “the coronavirus”, “the new coronavirus” and “the Wuhan coronavirus”, while the WHO recommends the designation "SARS-CoV-2".
  • the International Committee on Taxonomy of Viruses (ICTV) announced that the official name for the virus is SARS-CoV-2.
  • Many early cases were linked to a large seafood and animal market in the Chinese city of Wuhan, and the virus is thought to have a zoonotic origin. Comparisons of the genetic sequences of this virus and other virus samples have shown similarities to SARS-Co V (79.5%) and bat coronaviruses (96%).
  • the virus could be a recombinant virus formed from two or more coronaviruses. Human-to-human transmission of the virus has been confirmed. Coronaviruses are primarily spread through close contact, in particular through respiratory droplets from coughs and sneezes within a range of about 6 feet (1.8 m). Viral RNA has also been found in stool samples from infected patients. It is possible that the virus can be infectious even during the incubation period. Animals sold for food were originally suspected to be the reservoir or intermediary hosts of SARS-CoV-2 because many of the first individuals found to be infected by the virus were workers at the Huanan Seafood Market.
  • Rhinolophus sinicus Two genome sequences from Rhinolophus sinicus published in 2015 and 2017 show a resemblance of 80% to SARS-CoV-2.
  • SARS-CoV-2 belongs to the broad family of viruses known as coronaviruses; "nCoV” is the standard term used to refer to novel coronaviruses until the choice of a more specific designation. It is a positive-sense single-stranded RNA (+ssRNA) virus.
  • coronaviruses are capable of causing illnesses ranging from the common cold to more severe diseases such as Middle East respiratory syndrome (MERS) and Severe acute respiratory syndrome (SARS). It is the seventh known coronavirus to infect people, after 229E, NL63, OC43, HKUl, MERS- CoV, and SARS-CoV. Like SARS-CoV, SARS-CoV-2 is a member of the subgenus Sarbecovirus (Beta-CoV lineage B). Its RNA sequence is approximately 30,000 bases in length. By 12 January 2020, five genomes of SARS-CoV-2 had been isolated from Wuhan and reported by the Chinese Center for Disease Control and Prevention (CCDC) and other institutions; the number of genomes increased to 28 by 26 January.
  • CCDC Chinese Center for Disease Control and Prevention
  • the viral 3C-like protease M(pro) from the ORFla polyprotein has also been modelled for drug docking experiments.
  • Innophore has produced two computational models based on SARS protease, and the Chinese Academy of Sciences has produced an unpublished experimental structure of a recombinant SARS- CoV-2 protease.
  • researchers at the University of Michigan have modelled the structures of all mature peptides in the SARS-CoV-2 genome using I-TASSER. The first known human infection occurred in early December 2019. An outbreak of SARS- CoV-2 was first detected in Wuhan, China, in mid-December 2019, likely originating from a single infected animal.
  • SARS-CoV-2 was designated a global health emergency by the WHO. As of January 2022, over 350 million cases have been confirmed worldwide. A larger number of people may have been but not detected (especially mild cases). Initially, nearly all cases outside China occurred in people who either travelled from Wuhan,or were in direct contact with someone who travelled from the area. Later, spread from travellers to other countries resulted in transmission to over 220 countries in the world.
  • COVID-19 acute respiratory disease is a viral respiratory disease caused by SARS- CoV-2. It was first detected during the 2019-20 Wuhan coronavirus outbreak. Symptoms may include fever, dry cough, and shortness of breath. Those infected may either be asymptomatic or have mild to severe symptoms, like fever, cough, shortness of breath. diarrhoea or upper respiratory symptoms (e.g., sneezing, runny nose, sore throat) are less frequent. Cases of severe infection can progress to severe pneumonia, multi-organ failure, and death.
  • the time from exposure to onset of symptoms is estimated at 2 to 10 days by the World Health Organization, and 2 to 14 days by the US Centers for Disease Control and Prevention (CDC). Of those who have died, many had underlying health conditions, exhibiting conditions such as hypertension, diabetes, cardiovascular disease or other diseases or conditions that impaired their immune systems Global health organizations have published preventive measures individuals can take to reduce the chances of SARS-CoV-2 infection. Recommendations are similar to those previously published for other coronaviruses and include: frequent washing of hands with soap and water; not touching the eyes, nose, or mouth with unwashed hands; and practicing good respiratory hygiene. The WHO has published several testing protocols for SARS-CoV-2. The most accurate testing of current infection uses real time reverse transcription-polymerase chain reaction (rRT-PCR).
  • rRT-PCR real time reverse transcription-polymerase chain reaction
  • An "isolated antibody” is one that has been separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
  • the antibody is purified: (1) to greater than 95% by weight of antibody as determined by the Lowry method, and most particularly more than 99% by weight; (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator; or (3) to homogeneity by SDS- PAGE under reducing or non-reducing conditions using Coomassie blue or silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • the basic four-chain antibody unit is a heterotetrameric glycoprotein composed of two identical light (L) chains and two identical heavy (H) chains.
  • An IgM antibody consists of 5 basic heterotetramer units along with an additional polypeptide called J chain, and therefore contain 10 antigen binding sites, while secreted IgA antibodies can polymerize to form polyvalent assemblages comprising 2-5 of the basic 4-chain units along with J chain.
  • the 4-chain unit is generally about 150,000 daltons.
  • Each L chain is linked to an H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype.
  • Each H and L chain also has regularly spaced intrachain disulfide bridges.
  • Each H chain has at the N-terminus, a variable region (VH) followed by three constant domains (CH) for each of the alpha and gamma chains and four CH domains for mu and isotypes.
  • Each L chain has at the N- terminus, a variable region (VL) followed by a constant domain (CL) at its other end.
  • the VL is aligned with the VH and the CL is aligned with the first constant domain of the heavy chain (CH1).
  • Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable regions.
  • the pairing of a VH and VL together forms a single antigen-binding site.
  • L chain from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda based on the amino acid sequences of their constant domains (CL).
  • CL constant domains
  • immunoglobulins can be assigned to different classes or isotypes.
  • immunoglobulins There are five classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, having heavy chains designated alpha, delta, epsilon, gamma and mu, respectively. They gamma and alpha classes are further divided into subclasses on the basis of relatively minor differences in CH sequence and function, humans express the following subclasses: IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2.
  • the term "variable” refers to the fact that certain segments of the V domains differ extensively in sequence among antibodies. The V domain mediates antigen binding and defines specificity of a particular antibody for its particular antigen.
  • variable regions consist of relatively invariant stretches called framework regions (FRs) of 15- 30 amino acids separated by shorter regions of extreme variability called "hypervariable regions” that are each 9-12 amino acids long.
  • FRs framework regions
  • hypervariable regions regions of extreme variability
  • the hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)).
  • the constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody dependent cellular cytotoxicity (ADCC), antibody- dependent cellular phagocytosis (ADCP), antibody-dependent neutrophil phagocytosis (ADNP), and antibody-dependent complement deposition (ADCD).
  • hypervariable region when used herein refers to the amino acid residues of an antibody that are responsible for antigen binding.
  • the hypervariable region generally comprises amino acid residues from a "complementarity determining region" or "CDR" (e.g., around about residues 24-34 (Ll), 50-56 (L2) and 89-97 (L3) in the VL, and around about 31-35 (Hl), 50-65 (H2) and 95-102 (H3) in the VH when numbered in accordance with the Kabat numbering system; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md.
  • CDR complementarity determining region
  • residues from a "hypervariable loop” e.g., residues 24-34 (Ll), 50-56 (L2) and 89-97 (L3) in the VL, and 26-32 (Hl), 52-56 (H2) and 95-101 (H3) in the VH when numbered in accordance with the Chothia numbering system; Chothia and Lesk, J. Mol.
  • the antibody has symmetrical insertions at one or more of the following points 28, in the VsubH when numbered in accordance with AHo; Honneger, A. and Plunkthun, A. J. Mol. Biol.309:657-670 (2001)).
  • germline nucleic acid residue is meant the nucleic acid residue that naturally occurs in a germline gene encoding a constant or variable region.
  • Germline gene is the DNA found in a germ cell (i.e., a cell destined to become an egg or in the sperm).
  • a germline mutation refers to a heritable change in a particular DNA that has occurred in a germ cell or the zygote at the single-cell stage, and when transmitted to offspring, such a mutation is incorporated in every cell of the body.
  • a germline mutation is in contrast to a somatic mutation which is acquired in a single body cell.
  • nucleotides in a germline DNA sequence encoding for a variable region are mutated (i.e., a somatic mutation) and replaced with a different nucleotide.
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations that include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies. The modifier "monoclonal" is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies useful in the present disclosure may be prepared by the hybridoma methodology first described by Kohler et al., Nature, 256:495 (1975), or may be made using recombinant DNA methods in bacterial, eukaryotic animal or plant cells (see, e.g., U.S. Patent 4,816,567) after single cell sorting of an antigen specific B cell, an antigen specific plasmablast responding to an infection or immunization, or capture of linked heavy and light chains from single cells in a bulk sorted antigen specific collection.
  • the "monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991), for example. It will be understood that monoclonal antibodies binding to SARS-CoV-2 will have several applications. These include the production of diagnostic kits for use in detecting and diagnosing SARS-CoV-2 infection, as well as for treating the same. In these contexts, one may link such antibodies to diagnostic or therapeutic agents, use them as capture agents or competitors in competitive assays, or use them individually without additional agents being attached thereto. The antibodies may be mutated or modified, as discussed further below.
  • the methods for generating monoclonal antibodies generally begin along the same lines as those for preparing polyclonal antibodies.
  • the first step for both these methods is immunization of an appropriate host or identification of subjects who are immune due to prior natural infection or vaccination with a licensed or experimental vaccine.
  • a given composition for immunization may vary in its immunogenicity. It is often necessary therefore to boost the host immune system, as may be achieved by coupling a peptide or polypeptide immunogen to a carrier.
  • Exemplary and preferred carriers are keyhole limpet hemocyanin (KLH) and bovine serum albumin (BSA). Other albumins such as ovalbumin, mouse serum albumin or rabbit serum albumin can also be used as carriers.
  • Means for conjugating a polypeptide to a carrier protein are well known in the art and include glutaraldehyde, m-maleimidobenzoyl-N-hydroxysuccinimide ester, carbodiimide and bis-diazotized benzidine.
  • the immunogenicity of a particular immunogen composition can be enhanced by the use of non- specific stimulators of the immune response, known as adjuvants.
  • Exemplary and preferred adjuvants in animals include complete Freund's adjuvant (a non-specific stimulator of the immune response containing killed Mycobacterium tuberculosis), incomplete Freund's adjuvants and aluminium hydroxide adjuvant and in humans include alum, CpG, MFP59 and combinations of immunostimulatory molecules ("Adjuvant Systems", such as AS0l or AS03). Additional experimental forms of inoculation to induce SARS-CoV-2-specific B cells is possible, including nanoparticle vaccines, or gene-encoded antigens delivered as DNA or RNA genes in a physical delivery system (such as lipid nanoparticle or on a gold biolistic bead), and delivered with needle, gene gun, transcutaneous electroporation device.
  • complete Freund's adjuvant a non-specific stimulator of the immune response containing killed Mycobacterium tuberculosis
  • incomplete Freund's adjuvants and aluminium hydroxide adjuvant and in humans include alum, CpG, MFP59
  • the antigen gene also can be carried as encoded by a replication competent or defective viral vector such as adenovirus, adeno-associated virus, poxvirus, herpesvirus, or alphavirus replicon, or alternatively a virus like particle.
  • a suitable approach is to identify subjects that have been exposed to the pathogens, such as those who have been diagnosed as having contracted the disease, or those who have been vaccinated to generate protective immunity against the pathogen or to test the safety or efficacy of an experimental vaccine. Circulating anti-pathogen antibodies can be detected, and antibody encoding or producing B cells from the antibody-positive subject may then be obtained.
  • the amount of immunogen composition used in the production of polyclonal antibodies varies upon the nature of the immunogen as well as the animal used for immunization.
  • a variety of routes can be used to administer the immunogen (subcutaneous, intramuscular, intradermal, intravenous and intraperitoneal).
  • the production of polyclonal antibodies may be monitored by sampling blood of the immunized animal at various points following immunization. A second, booster injection, also may be given. The process of boosting and titering is repeated until a suitable titer is achieved.
  • the immunized animal can be bled and the serum isolated and stored, and/or the animal can be used to generate MAbs.
  • somatic cells with the potential for producing antibodies, specifically B lymphocytes (B cells), are selected for use in the MAb generating protocol. These cells may be obtained from biopsied spleens, lymph nodes, tonsils or adenoids, bone marrow aspirates or biopsies, tissue biopsies from mucosal organs like lung or GI tract, or from circulating blood.
  • the antibody-producing B lymphocytes from the immunized animal or immune human are then fused with cells of an immortal myeloma cell, generally one of the same species as the animal that was immunized or human or human/mouse chimeric cells.
  • Myeloma cell lines suited for use in hybridoma-producing fusion procedures preferably are non-antibody-producing, have high fusion efficiency, and enzyme deficiencies that render then incapable of growing in certain selective media which support the growth of only the desired fused cells (hybridomas). Any one of a number of myeloma cells may be used, as are known to those of skill in the art (Goding, pp.65-66, 1986; Campbell, pp.75-83, 1984). HMMA2.5 cells or MFP-2 cells are particularly useful examples of such cells.
  • Methods for generating hybrids of antibody-producing spleen or lymph node cells and myeloma cells usually comprise mixing somatic cells with myeloma cells in a 2:1 proportion, though the proportion may vary from about 20:1 to about 1:1, respectively, in the presence of an agent or agents (chemical or electrical) that promote the fusion of cell membranes.
  • transformation of human B cells with Epstein Barr virus (EBV) as an initial step increases the size of the B cells, enhancing fusion with the relatively large-sized myeloma cells. Transformation efficiency by EBV is enhanced by using CpG and a Chk2 inhibitor drug in the transforming medium.
  • EBV Epstein Barr virus
  • human B cells can be activated by co-culture with transfected cell lines expressing CD40 Ligand (CD154) in medium containing additional soluble factors, such as IL-21 and human B cell Activating Factor (BAFF), a Type II member of the TNF superfamily.
  • CD40 Ligand CD40 Ligand
  • BAFF human B cell Activating Factor
  • Fusion methods using Sendai virus have been described by Kohler and Milstein (1975; 1976), and those using polyethylene glycol (PEG), such as 37% (v/v) PEG, by Gefter et al. (1977).
  • PEG polyethylene glycol
  • the use of electrically induced fusion methods also is appropriate (Goding, pp.71-74, 1986) and there are processes for better efficiency (Yu et al., 2008).
  • Fusion procedures usually produce viable hybrids at low frequencies, about 1 x 10-6 to 1 x 10-8, but with optimized procedures one can achieve fusion efficiencies close to 1 in 200 (Yu et al., 2008).
  • relatively low efficiency of fusion does not pose a problem, as the viable, fused hybrids are differentiated from the parental, infused cells (particularly the infused myeloma cells that would normally continue to divide indefinitely) by culturing in a selective medium.
  • the selective medium is generally one that contains an agent that blocks the de novo synthesis of nucleotides in the tissue culture medium.
  • Exemplary and preferred agents are aminopterin, methotrexate, and azaserine.
  • Aminopterin and methotrexate block de novo synthesis of both purines and pyrimidines, whereas azaserine blocks only purine synthesis.
  • the medium is supplemented with hypoxanthine and thymidine as a source of nucleotides (HAT medium).
  • HAT medium hypoxanthine and thymidine as a source of nucleotides
  • azaserine the medium is supplemented with hypoxanthine.
  • Ouabain is added if the B cell source is an EBY- transformed human B cell line, in order to eliminate EBY-transformed lines that have not fused to the myeloma.
  • the preferred selection medium is HAT or HAT with ouabain.
  • the myeloma cells are defective in key enzymes of the salvage pathway, e.g., hypoxanthine phosphoribosyl transferase (HPRT), and they cannot survive.
  • HPRT hypoxanthine phosphoribosyl transferase
  • the B cells can operate this pathway, but they have a limited life span in culture and generally die within about two weeks. Therefore, the only cells that can survive in the selective media are those hybrids formed from myeloma and B cells.
  • ouabain may also be used for drug selection of hybrids as EBY-transformed B cells are susceptible to drug killing, whereas the myeloma partner used is chosen to be ouabain resistant.
  • Culturing provides a population of hybridomas from which specific hybridomas are selected.
  • selection of hybridomas is performed by culturing the cells by single-clone dilution in microtiter plates, followed by testing the individual clonal supernatants (after about two to three weeks) for the desired reactivity.
  • the assay should be sensitive, simple and rapid, such as radioimmunoassays, enzyme immunoassays, cytotoxicity assays, plaque assays dot immunobinding assays, and the like.
  • the selected hybridomas are then serially diluted or single-cell sorted by flow cytometric sorting and cloned into individual antibody-producing cell lines, which clones can then be propagated indefinitely to provide mAbs.
  • the cell lines may be exploited for MAb production in two basic ways.
  • a sample of the hybridoma can be injected (often into the peritoneal cavity) into an animal (e.g., a mouse).
  • the animals are primed with a hydrocarbon, especially oils such as pristane (tetramethylpentadecane) prior to injection.
  • a hydrocarbon especially oils such as pristane (tetramethylpentadecane) prior to injection.
  • pristane tetramethylpentadecane
  • SCID mice immunocompromised mice
  • the injected animal develops tumours secreting the specific monoclonal antibody produced by the fused cell hybrid.
  • the body fluids of the animal such as serum or ascites fluid, can then be tapped to provide MAbs in high concentration.
  • the individual cell lines could also be cultured in vitro,where the MAbs are naturally secreted into the culture medium from which they can be readily obtained in high concentrations.
  • human hybridoma cells lines can be used in vitro to produce immunoglobulins in cell supernatant.
  • the cell lines can be adapted for growth in serum-free medium to optimize the ability to recover human monoclonal immunoglobulins of high purity.
  • MAbs produced by either means may be further purified, if desired, using filtration, centrifugation and various chromatographic methods such as FPLC or affinity chromatography. Fragments of the monoclonal antibodies of the disclosure can be obtained from the purified monoclonal antibodies by methods which include digestion with enzymes, such as pepsin or papain, and/or by cleavage of disulfide bonds by chemical reduction.
  • monoclonal antibody fragments encompassed by the present disclosure can be synthesized using an automated peptide synthesizer. It also is contemplated that a molecular cloning approach may be used to generate monoclonal antibodies.
  • Single B cells identified as responding to infection or vaccination because of plasmablast or activated B cell markers, or memory B cells labelled with the antigen of interest can be sorted physically using paramagnetic bead selection or flow cytometric sorting, then RNA can be isolated from the single cells and antibody genes amplified by RT-PCR.
  • RNA-seq methods are available to obtain antibody variable genes from single cells.
  • antigen-specific bulk sorted populations of cells can be segregated into microvesicles and the matched heavy and light chain variable or common barcoding of heavy and light chain genes from a vesicle.
  • Matched heavy and light chain genes from single cells also can be obtained from populations of antigen specific B cells by treating cells with cell-penetrating nanoparticles bearing RT-PCR primers and barcodes for marking transcripts with one barcode per cell.
  • the antibody variable genes also can be isolated by RNA extraction of a hybridoma line and the antibody genes obtained by RT-PCR and cloned into an immunoglobulin expression vector.
  • combinatorial immunoglobulin phagemid libraries are prepared from RNA isolated from the cell lines and phagemids expressing appropriate antibodies are selected by panning using viral antigens.
  • the advantages of this approach over conventional hybridoma techniques are that approximately 104 times as many antibodies can be produced and screened in a single round, and that new specificities are generated by H and L chain combination which further increases the chance of finding appropriate antibodies.
  • Other U.S. patents, each incorporated herein by reference, that teach the production of antibodies useful in the present disclosure include U.S. Patent 5,565,332, which describes the production of chimeric antibodies using a combinatorial approach; U.S. Patent 4,816,567 which describes recombinant immunoglobulin preparations; and U.S.
  • Patent 4,867,973 which describes antibody-therapeutic agent conjugates.
  • Antibodies according to the present disclosure may be defined, in the first instance, by their binding specificity. Those of skill in the art, by assessing the binding specificity/affinity of a given antibody using techniques well known to those of skill in the art, can determine whether such antibodies fall within the scope of the instant claims.
  • the epitope to which a given antibody bind may consist of a single contiguous sequence of 3 or more (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 1112, 13, 14, 15, 16, 17, 18, 19, 20) amino acids located within the antigen molecule (e.g., a linear epitope in a domain).
  • the epitope may consist of a plurality of non-contiguous arnino acids (or amino acid sequences) located within the antigen molecule (e.g., a conformational epitope).
  • S surface spike
  • N nucleocapsid
  • Antibodies to the S protein will be useful for prophylaxis, or therapy, or diagnostics, or for characterizing vaccines.
  • S protein antibodies will have additional binding specificity with that protein, with particular antibodies binding to the full-length ectodomain of the SARS-CoV-2 S protein (presented as a monomer or oligomer such as a timer; with or without conformation stabilizing mutations such as introduction of pralines at critical sites (“2P mutation")) and (a) anti-S protein antibodies that binds to the receptor binding domain (RBD), (b) anti-S protein antibodies that bind to domains other than the RBD.
  • SARS-CoV-2 S protein presented as a monomer or oligomer such as a timer; with or without conformation stabilizing mutations such as introduction of pralines at critical sites (“2P mutation")
  • RBD receptor binding domain
  • NTD N terminal domain
  • S protein antibodies may further be found to neutralize SARS-CoV-2 by blocking binding of the SARS-CoV-2 S protein to its receptor, human angiotensin-converting enzyme 2 (hACE2), with others that neutralize but do not block receptor binding.
  • hACE2 human angiotensin-converting enzyme 2
  • antibodies can cross-react with both SARS-CoV-2 S protein and the S protein of other coronaviruses such as SARS-CoV, MERS-CoV, HCoV-229E, HCoV- OC43, HCoV-NL63 and/or HCoV-HKUl, as well as cross- neutralize both SARS-CoV-2 and these other coronaviruses.
  • SARS-CoV SARS-CoV
  • MERS-CoV HCoV-229E
  • HCoV- OC43 HCoV-NL63 and/or HCoV-HKUl
  • Another specificity will be antibodies that bind to N antibodies (or other internal targets) that will have primarily diagnostics uses.
  • antibodies to Nor other internal proteins of SARS-CoV-2 that specifically recognize SARS-CoV-2 or that cross-reactively recognize SARS-CoV-2 and other coronaviruses such as SARS-CoV, MERS-CoV, HCoV- 229E, HCoV-OC43, HCoV-NL63 and/or HCoV-HKUl.
  • SARS-CoV MERS-CoV
  • HCoV- 229E HCoV-OC43
  • HCoV-NL63 HCoV-NL63
  • HCoV-HKUl HCoV-HKUl.
  • Various techniques known to persons of ordinary skill in the art can be used to determine whether an antibody "interacts with one or more amino acids" within a polypeptide or protein. Exemplary techniques include, for example, routine cross-blocking assays, such as that described in Antibodies, Harlow and Lane (Cold Spring Harbor Press, Cold Spring Harbor, N.Y.).
  • Cross-blocking can be measured in various binding assays such as ELISA, biolayer interferometry, or surface plasmon resonance.
  • Other methods include alanine scanning mutatonal analysis, peptide blot analysis (Reineke, Methods J ⁇ fol. BioL 248: 443- 63, 2004), peptide cleavage analysis, high-resolution electron microscopy techniques using single particle reconstruction, cryoEM, or tomography, crystallographic studies and NMR analysis.
  • methods such as epitope excision, epitope extraction and chemical modification of antigens can be employed (Tomer Prot. Sci. 9: 487--496, 2000).
  • the hydrogen/deuterium exchange method involves deuterium-labelling the protein of interest, followed by binding the antibody to the deuterium-labelled protein. Next, the protein/antibody complex is transferred to water and exchangeable protons within amino acids that are protected by the antibody complex undergo deuterium-to-hydrogen back-exchange at a slower rate than exchangeable protons within amino acids that are not part of the interface. As a result, amino acids that form part of the protein/antibody interface may retain deuterium and therefore exhibit relatively higher mass compared to arnino acids not included in the interface.
  • the target protein After dissociation of the antibody, the target protein is subjected to protease cleavage and mass spectrometry analysis, thereby revealing the deuterium-labelled residues which correspond to the specific amino acids with which the antibody interacts. See, e.g., Ehring, Analytical Biochemistry 267: 252-259 (1999): Engen and Smith, Anal. Chern.73: 256A-265A (2001).
  • antibody escape mutant variant organisms can be isolated by propagating SARS-CoV-2 in vitro or in animal models in the presence of high concentrations of the antibody.
  • epitope refers to a site on an antigen to which B and/or T cells respond.
  • B- cell epitopes can be formed both from contiguous arnino acids or non-contiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epltopes formed by tertiary folding are typically lost on treatment with denaturing solvents.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • Modification-Assisted Profiling also known as Antigen Structure-based Antibody Profiling (ASAP) is a method that categorizes large numbers of monoclonal antibodies (rnAbs) directed against the same antigen according to the similarities of the binding profile of each antibody to chernically or enzymatically modified antigen surfaces (see US. Patent Publication 2004/0101920, herein specifically incorporated by reference in its entirety). Each category may reflect a unique epitope either distinctly different from or partially overlapping with epitope represented by another category.
  • MAP may facilitate identification of rare hybridoma clones that produce mAbs having the desired characteristics.
  • MAP may be used to sort the antibodies of the disclosure into groups of antibodies binding different epitopes.
  • the present disclosure includes antibodies that may bind to the same epitope, or a portion of the epitope.
  • the present disclosure also includes antibodies that compete for binding to a target or a fragment thereof with any of the specific exemplary antibodies described herein. One can easily determine whether an antibody binds to the same epitope as or competes for binding with, a reference antibody by using routine methods known in the art.
  • test antibody binds to the same epitope as a reference
  • the reference antibody is allowed to bind to target under saturating conditions.
  • the ability of a test antibody to bind to the target molecule is assessed. If the test antibody is able to bind to the target molecule following saturation binding with the reference antibody, it can be concluded that the test antibody binds to a different epitope than the reference antibody. On the other hand, if the test antibody is not able to bind to the target molecule following saturation binding with the reference antibody, then the test antibody may bind to the same epitope as the epitope bound by the reference antibody.
  • the above-described binding rnethodology is perforrned in two orientations: In a first orientation, the reference antibody is allowed to bind to the SARS-CoV-2 antigen under saturating conditions followed by assessment of binding of the test antibody to the SARS- CoV- 2 rnolecu1e. In a second orientation, the test antibody is allowed to bind to the SARS- CoV-2 antigen molecule under saturating conditions followed by assessment of binding of the reference antibody to the SARS-CoV-2 molecule.
  • an antibody that competes for binding with a reference antibody may not necessarily bind to the identical epitope as the reference antibody but may sterically block binding of the reference antibody by binding an overlapping or adjacent epitope. Two antibodies bind to the same or overlapping epitope, if each competitively inhibits (blocks) binding of the other to the antigen.
  • a 1-, 5-, 10-, 20-- or 100-fold excess of one antibody inhibits binding of the other by at least 50%, but preferably 75%, 90% or even 99% as measured in a competitive binding assay (see, e.g._Junghans et aL Cancer Res. 199050:1495-1502).
  • two antibodies have the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • Two antibodies have overlapping epitopes if some arnino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • Additional routine experimentation e.g., peptide mutation and binding analyses
  • peptide mutation and binding analyses can then be carried out to confirm whether the observed lack of binding of the test antibody is in fact due to binding to the same epitope as the reference antibody or if steric blocking (or another phenomenon) is responsible for the lack of observed binding.
  • steric blocking or another phenomenon
  • Structural studies with EM or crystallography also can demonstrate whether or not two antibodies that compete. for binding recognize the same epitope.
  • monoclonal antibodies having clone-paired CDRs from the heavy and light chains as illustrated in Tables 3 and 4, respectively.
  • antibodies may be produced by the clones discussed below in the Examples section using methods described herein.
  • the antibodies may be defined by their variable sequence, which include additional "framework" regions.
  • the antibodies sequences may vary from these sequences, optionally using methods discussed in greater detail below.
  • nucleic acid sequences may vary from those set out above in that (a) the variable regions may be segregated away from the constant domains of the light and heavy chains, (b) the nucleic acids may vary from those set out above while not affecting the residues encoded thereby, (c) the nucleic acids may vary from those set out above by a given percentage, e.g., 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% homology, (d) the nucleic acids may vary from those set out above by virtue of the ability to hybridize under high stringency conditions, as exemplified by low salt and/or high temperature conditions, such as provided by about 0.02 M to about 0.15 M NaCl at temperatures of about 50°C to about 70°C,(e) the amino acids may vary from those set out above by a given percentage, e.g., 80%, 85%, 90%, 91%, 92%, 93%, 9
  • nucleic acid sequences and the amino acid sequences are said to be “identical” if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wis.), using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M.0. (1978) A model of evolutionary change in proteins- - Matrices for detecting distant relationships. In Dayhoff, M. 0. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington D.C. Vol. 5, Suppl.3, pp.345-358; Hein J. (1990) Unified Approach to Alignment and Phylogeny pp. 626-645, Methods in Enzymology vol.
  • optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL. Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol.48:443, by the search for similarity methods of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85: 2444, by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, PASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by inspection.
  • BLAST and BLAST 2.0 are described in Altschul et al. (1977) Nucl. Acids Res.25:3389-3402 and Altschul et al. (1990) J. Mol. Biol.215:403-410, respectively.
  • BLAST and BLAST 2.0 can be used, for example, with the parameters described herein, to determine percent sequence identity for the polynucleotides and polypeptides of the disclosure.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. The rearranged nature of an antibody sequence and the variable length of each gene requires multiple rounds of BLAST searches for a single antibody sequence.
  • IgBLAST (world-wide-web at ncbi.nlm.nih.gov/igblast/) identifies matches to the germline V, D and J genes, details at rearrangement junctions, the delineation of lg V domain framework regions and complementarity determining regions. IgBLAST can analyze nucleotide or protein sequences and can process sequences in batches and allows searches against the germline gene databases and other sequence databases simultaneously to minimize the chance of missing possibly the best matching germline V gene.
  • cumulative scores can be calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0). Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.
  • a scoring matrix can be used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.
  • the "percentage of sequence identity" is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residues occur in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • Yet another way of defining an antibody is as a "derivative" of any of the below- described antibodies and their antigen-binding fragments.
  • derivative refers to an antibody or antigen-binding fragment thereof that immunospecifically binds to an antigen, but which comprises, one, two, three, four, five or more amino acid substitutions, additions, deletions or modifications relative to a "parental" (or wild-type) molecule. Such amino acid substitutions or additions may introduce naturally occurring (i.e., DNA-encoded) or non- naturally occurring amino acid residues.
  • derivative encompasses, for example, as variants having altered CHI, hinge, CH2, CH3 or CH4 regions, so as to form, for example, antibodies, etc., having variant Fc regions that exhibit enhanced or impaired effector or binding characteristics.
  • derivative additionally encompasses non-amino acid modifications, for example, amino acids that may be glycosylated (e.g., have altered mannose, 2-N- acetylglucosamine, galactose, fucose, glucose, sialic acid, 5-N- acetylneuraminic acid, 5-glycolneuraminic acid, etc. content), acetylated, pegylated, phosphorylated, amidated, derivatized by known protecting/blocking groups, proteolytic cleavage, linked to a cellular ligand or other protein, etc.
  • non-amino acid modifications for example, amino acids that may be glycosylated (e.g., have altered mannose, 2-N- acetylglucosamine, galactose, fucose, glucose, sialic acid, 5-N- acetylneuraminic acid, 5-glycolneuraminic acid, etc. content), acetylated, pegy
  • the altered carbohydrate modifications modulate one or more of the following: solubilization of the antibody, facilitation of subcellular transport and secretion of the antibody, promotion of antibody assembly, conformational integrity, and antibody-mediated effector function.
  • the altered carbohydrate modifications enhance antibody mediated effector function relative to the antibody lacking the carbohydrate modification.
  • Carbohydrate modifications that lead to altered antibody mediated effector function are well known in the art (for example, see Shields, R. L. et al. (2002) J. Biol. Chem.277(30): 26733- 26740; Davies J. et al. (2001) Biotechnology & Bioengineering 74(4): 288-294).
  • a derivative antibody or antibody fragment can be generated with an engineered sequence or glycosylation state to confer preferred levels of activity in antibody dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), antibody-dependent neutrophil phagocytosis (ADNP), or antibody-dependent complement deposition (ADCD) functions as measured by bead-based or cell-based assays or in vivo studies in animal models.
  • a derivative antibody or antibody fragment may be modified by chemical modifications using techniques known to those of skill in the art, including, but not limited to, specific chemical cleavage, acetylation, formulation, metabolic synthesis of tunicamycin, etc.
  • an antibody derivative will possess a similar or identical function as the parental antibody.
  • an antibody derivative will exhibit an altered activity relative to the parental antibody.
  • a derivative antibody or fragment thereof
  • Modified antibodies may be made by any technique known to those of skill in the art, including expression through standard molecular biological techniques, or the chemical synthesis of polypeptides. Methods for recombinant expression are addressed elsewhere in this document. The following is a general discussion of relevant goals techniques for antibody engineering. Hybridomas may be cultured, then cells lysed, and total RNA extracted.
  • Random hexamers may be used with RT to generate cDNA copies of RNA, and then PCR performed using a multiplex mixture of PCR primers expected to amplify all human variable gene sequences.
  • PCR product can be cloned into pGEM-T Easy vector, then sequenced by automated DNA sequencing using standard vector primers.
  • Assay of binding and neutralization may be performed using antibodies collected from hybridoma supernatants and purified by FPLC, using Protein G columns.
  • Recombinant full-length IgG antibodies can be generated by subcloning heavy and light chain Fv DNAs from the cloning vector into an IgG plasmid vector, transfected into 293 (e.g., Freestyle) cells or CHO cells, and antibodies can be collected and purified from the 293 or CHO cell supernatant.
  • 293 e.g., Freestyle
  • Other appropriate host cells systems include bacteria, such as E. coli, insect cells (S2, Sf9, Sf29, High Five), plant cells (e.g., tobacco, with or without engineering for human-like glycans), yeast, algae, or in a variety of non-human transgenic contexts, such as mice, rats, goats or cows.
  • the antibody molecules of the invention may be generated using a yeast expression platform.
  • the methylotrophic yeast Pichia pastoris provides an efficient platform and pichia expression systems are commercially available.
  • Antibody coding sequences can be RNA, such as native RNA or modified RNA.
  • Modified RNA contemplates certain chemical modifications that confer increased stability and low immunogenicity to mRNAs, thereby facilitating expression of therapeutically important proteins. For instance, N1-methyl-pseudouridine (N1m ⁇ ) outperforms several other nucleoside modifications and their combinations in terms of translation capacity.
  • RNA may be delivered as naked RNA or in a delivery vehicle, such as a lipid nanoparticle. Alternatively, DNA encoding the antibody may be employed for the same purposes.
  • the DNA is included in an expression cassette comprising a promoter active in the host cell for which it is designed.
  • the expression cassette is advantageously included in a replicable vector, such as a conventional plasmid or minivector.
  • Vectors include viral vectors, such as poxviruses, adenoviruses, herpesviruses, adeno-associated viruses, and lentiviruses are contemplated.
  • Replicons encoding antibody genes such as alphavirus replicons based on VEE virus or Sindbis virus are also contemplated. Delivery of such vectors can be performed by needle through intramuscular, subcutaneous, or intradermal routes, or by transcutaneous electroporation when in vivo expression is desired.
  • Lonza has developed a generic method using pooled transfectants grown in CDACF medium, for the rapid production of small quantities (up to 50 g) of antibodies in CHO cells. Although slightly slower than a true transient system, the advantages include a higher product concentration and use of the same host and process as the production cell line.
  • Antibody molecules will comprise fragments (such as F(ab'), F(ab') 2 ) that are produced, for example, by the proteolytic cleavage of the mAbs, or single-chain immunoglobulins producible, for example, via recombinant means.
  • F(ab') antibody derivatives are monovalent, while F(ab') 2 antibody derivatives are bivalent.
  • fragments can be combined with one another, or with other antibody fragments or receptor ligands to form "chimeric" binding molecules.
  • such chimeric molecules may contain substituents capable of binding to different epitopes of the same molecule.
  • the antibody is a derivative of the disclosed antibodies, e.g.,an antibody comprising the CDR sequences identical to those in the disclosed antibodies (e.g., a chimeric, or CDR-grafted antibody).
  • an antibody comprising the CDR sequences identical to those in the disclosed antibodies (e.g., a chimeric, or CDR-grafted antibody).
  • modifications such as introducing conservative changes into an antibody molecule.
  • the hydropathic index of amino acids may be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982).
  • Patent 4,554,101 the following hydrophilicity values have been assigned to amino acid residues: basic amino acids: arginine (+3.0), lysine (+3.0), and histidine (-0.5); acidic amino acids: aspartate (+3.0 ⁇ 1), glutamate (+3.0 ⁇ 1), asparagine (+0.2), and glutamine (+0.2); hydrophilic, nonionic amino acids: serine (+0.3), asparagine (+0.2), glutamine (+0.2), and threonine (-0.4), sulfur containing amino acids: cysteine (-1.0) and methionine (-1.3); hydrophobic, nonaromatic amino acids: valine (-1.5), leucine (-1.8), isoleucine (-1.8), praline (-0.5 ⁇ 1), alanine (-0.5), and glycine (0); hydrophobic, aromatic amino acids: tryptophan (- 3.4), phenylalanine (-2.5), and tyrosine (-2.3).
  • amino acid can be substituted for another having a similar hydrophilicity and produce a biologically or immunologically modified protein.
  • substitution of amino acids whose hydrophilicity values are within ⁇ 2 is preferred, those that are within ⁇ 1 are particularly preferred, and those within ⁇ 0.5 are even more particularly preferred.
  • amino acid substitutions generally are based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity,hydrophilicity, charge, size, and the like.
  • substitutions that take into consideration the various foregoing characteristics are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
  • the present disclosure also contemplates isotype modification. By modifying the Fc region to have a different isotype, different functionalities can be achieved. For example, changing to IgG1 can increase antibody dependent cell cytotoxicity, switching to class A can improve tissue distribution, and switching to class M can improve valency.
  • binding polypeptide of particular interest may be one that binds to C1q and displays complement dependent cytotoxicity.
  • Polypeptides with pre-existing C1q binding activity, optionally further having the ability to mediate CDC may be modified such that one or both of these activities are enhanced.
  • Amino acid modifications that alter C1q and/or modify its complement dependent cytotoxicity function are described, for example, in WO/0042072, which is hereby incorporated by reference.
  • effector functions are responsible for activating or diminishing a biological activity (e.g., in a subject). Examples of effector functions include, but are not limited to: C1q binding; complement dependent cytotoxicity (CDC); Fc receptor binding; antibody- dependent cell- mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptor; BCR), etc.
  • Such effector functions may require the Fc region to be combined with a binding domain (e.g., an antibody variable domain) and can be assessed using various assays (e.g., Fc binding assays, ADCC assays, CDC assays, etc.).
  • a binding domain e.g., an antibody variable domain
  • assays e.g., Fc binding assays, ADCC assays, CDC assays, etc.
  • Fc binding assays e.g., ADCC assays, CDC assays, etc.
  • Fc binding assays e.g., ADCC assays, CDC assays, etc.
  • a variant Fc region of an antibody with improved C1q binding and improved Fc ⁇ R III binding e.g., having both improved ADCC activity and improved CDC activity.
  • a variant Fc region can be engineered with reduced CDC activity and/or reduced ADCC activity.
  • Fc mutations can also be introduced and engineered to alter their interaction with the neonatal Fc receptor (FcRn) and improve their pharmacokinetic properties.
  • FcRn neonatal Fc receptor
  • a collection of human Fc variants with improved binding to the FcRn have been described (Shields et al., (2001). High resolution mapping of the binding site on human IgG1 for Fc ⁇ R I, Fc ⁇ R II, Fc ⁇ R III, and FcRn and design of IgG1 variants with improved binding to the Fc ⁇ R , (J. Biol.
  • Chem.276:6591-6604 A number of methods are known that can result in increased half- life (Kuo and Aveson, (2011)), including amino acid modifications may be generated through techniques including alanine scanning mutagenesis, random mutagenesis and screening to assess the binding to the neonatal Fc receptor (FcRn) and/or the in vivo behaviour. Computational strategies followed by mutagenesis may also be used to select one of amino acid mutations to mutate. The present disclosure therefore provides a variant of an antigen binding protein with optimized binding to FcRn.
  • the variant of an antigen binding protein comprises at least one amino acid modification in the Fc region of the antigen binding protein, wherein the modification is selected from the group consisting of 226, 227, 228,230,231,233,234,239,241,243,246,250,252,254,256,259,264,265,267,269,270,276,2 84,285,288,289,290,291,292,294,297,298,299,301,302,303,305,307,308,309,311,315,31 7,320,322,325,327,330,332,334,335,338,340,342,343,345,347,350,352,354,355,356,359, 360,361,362,369,370,371,375,378,380,382,384,385,386,387,389,390,392,393,394,395,3 96,397,398,399,400,401403,404,408,411,412,
  • the modifications are M252Y/S254T/T256E. In a further embodiment of the disclosure the modifications are M252Y/S254T/T256E. In a further embodiment of the disclosure, the modifications are T250Q/M428L.
  • various publications describe methods for obtaining physiologically active molecules whose half-lives are modified, see for example Kontermann (2009) either by introducing an FcRn-binding polypeptide into the molecules or by fusing the molecules with antibodies whose FcRn-binding affinities are preserved but affinities for other Fe receptors have been greatly reduced or fusing with FcRn binding domains of antibodies.
  • Derivatized antibodies may be used to alter the half-lives (e.g., serum half-lives) of parental antibodies in a mammal, particularly a human. Such alterations may result in a half- life of greater than 15 days, preferably greater than 20 days, greater than 25 days, greater than 30 days, greater than 35 days, greater than 40 days, greater than 45 days, greater than 2 months, greater than 3 months, greater than 4 months, or greater than 5 months.
  • half-lives e.g., serum half-lives
  • Antibodies or fragments thereof having increased in vivo half-lives can be generated by techniques known to those of skill in the art. For example, antibodies or fragments thereof with increased in vivo half-lives can be generated by modifying (e.g., substituting, deleting or adding) amino acid residues identified as involved in the interaction between the Fe domain and the FcRn receptor. Beltramello et al.
  • LALA mutations of this nature are therefore contemplated in the context of the presently disclosed antibodies.
  • the modifications are L234A /L235A (LALA) or L234A /L235A + P329G (LALA-PG).
  • a particular embodiment of the present disclosure is an isolated monoclonal antibody, or antigen binding fragment thereof, containing a substantially homogeneous glycan without sialic acid, galactose, or fucose.
  • the monoclonal antibody comprises a heavy chain variable region and a light chain variable region, both of which may be attached to heavy chain or light chain constant regions respectively.
  • the aforementioned substantially homogeneous glycan may be covalently attached to the heavy chain constant region.
  • Another embodiment of the present disclosure comprises a mAb with a novel Fc glycosylation pattern.
  • the isolated monoclonal antibody, or antigen binding fragment thereof is present in a substantially homogenous composition represented by the GNGN or G1/G2 glycoform.
  • Fc glycosylation plays a significant role in anti-viral and anti-cancer properties of therapeutic mAbs.
  • the disclosure is in line with a recent study that shows increased anti- lentivirus cell-mediated viral inhibition of a fucose free anti-HIV mAb in vitro.
  • This embodiment of the present disclosure with homogenous glycans lacking a core fucose showed increased protection against specific viruses by a factor greater than two-fold.
  • the isolated monoclonal antibody, or antigen binding fragment thereof, comprising a substantially homogenous composition represented by the GNGN or G1/G2 glycoform exhibits increased binding affinity for Fc gamma RI and Fc gamma RIII compared to the same antibody without the substantially homogeneous GNGN glycoform and with GO, GlF, G2F, GNP, GNGNF or GNGNFX containing glycoforms.
  • the antibody dissociates from Fc gamma RI with a Kd of 1 x 10-3 M or less and from Fc gamma RIII with a Kd of 1 x 10-7 M or less.
  • Glycosylation of an Fc region is typically either N-linked or O-linked.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • O- linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain peptide sequences are asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except praline.
  • X is any amino acid except praline.
  • the glycosylation pattern may be altered, for example, by deleting one or more glycosylation site(s) found in the polypeptide, and/or adding one or more glycosylation site(s) that are not present in the polypeptide.
  • glycosylation sites to the Fc region of an antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • An exemplary glycosylation variant has an amino acid substitution of residue Asn 297 of the heavy chain. The alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original polypeptide (for O-linked glycosylation sites). Additionally, a change of Asn 297 to Ala can remove one of the glycosylation sites.
  • the antibody is expressed in cells that express beta (l,4)-N- acetylglucosaminyltransferase III (GnT III), such that GnT III adds GlcNAc to the IL-23p19 antibody.
  • GnT III beta (l,4)-N- acetylglucosaminyltransferase III
  • Methods for producing antibodies in such a fashion are provided in WO/9954342, WO/03011878, patent publication 20030003097Al, and Umana et al., Nature Biotechnology, 17:176-180, February 1999.
  • Cell lines can be altered to enhance or reduce or eliminate certain post-translational modifications, such as glycosylation, using genome editing technology suchas Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR).
  • CRISPR Clustered Regularly Interspaced Short Palindromic Repeats
  • CRISPR technology can be used to eliminate genes encoding glycosylating enzymes in 293 or CHO cells used to express recombinant monoclonal antibodies. It is possible to engineer the antibody variable gene sequences obtained from human B cells to enhance their manufacturability and safety.
  • Potential protein sequence liabilities can be identified by searching for sequence motifs associated with sites containing: 1) Unpaired Cys residues, 2) N-linked glycosylation, 3) Asn deamidation, 4) Asp isomerization, 5) SYE truncation, 6) Met oxidation, 7) Trp oxidation, 8) N-terminal glutamate, 9) Integrin binding, 10) CD1lc/CD18 binding, or 11) Fragmentation
  • sequence motifs can be eliminated by altering the synthetic gene for the cDNA encoding recombinant antibodies.
  • Elevated temperature may be used to unfold antibodies to determine relative stability, using average apparent melting temperatures.
  • Differential Scanning Calorimetry measures the heat capacity, Cp, of a molecule (the heat required to warm it, per degree) as a function of temperature.
  • Cp heat capacity
  • DSC data for mAbs is particularly interesting because it sometimes resolves the unfolding of individual domains within the mAb structure, producing up to three peaks in the thermogram (from unfolding of the Fab, CH2, and CH3 domains). Typically unfolding of the Fab domain produces the strongest peak.
  • the DSC profiles and relative stability of the Fe portion show characteristic differences for the human IgG1, IgG2, IgG3, and IgG4 subclasses (Garber and Demarest, Biochem. Biophys. Res. Commun. 355, 751-757, 2007).
  • CD circular dichroism
  • Far-UV CD spectra will be measured for antibodies in the range of 200 to 260 nm at increments of 0.5 nm. The final spectra can be determined as averages of 20 accumulations. Residue ellipticity values can be calculated after background subtraction.
  • DLS dynamic light scattering
  • DLS measurements of a sample can show whether the particles aggregate over time or with temperature variation by determining whether the hydrodynamic radius of the particle increases. If particles aggregate, one can see a larger population of particles with a larger radius. Stability depending on temperature can be analyzed by controlling the temperature in situ.
  • Capillary electrophoresis (CE) techniques include proven methodologies for determining features of antibody stability. One can use an iCE approach to resolve antibody protein charge variants due to deamidation, C-terminal lysines, sialylation, oxidation, glycosylation, and any other change to the protein that can result in a change in pH of the protein.
  • Each of the expressed antibody proteins can be evaluated by high throughput, free solution isoelectric focusing (IEF) in a capillary column (cIEF), using a Protein Simple Maurice instrument.
  • IEF free solution isoelectric focusing
  • cIEF capillary column
  • Whole-column UV absorption detection can be performed every 30 seconds for real time monitoring of molecules focusing at the isoelectric points (pls).
  • This approach combines the high resolution of traditional gel IEF with the advantages of quantitation and automation found in column-based separations while eliminating the need for a mobilization step.
  • the technique yields reproducible, quantitative analysis of identity, purity, and heterogeneity profiles for the expressed antibodies.
  • the results identify charge heterogeneity and molecular sizing on the antibodies, with both absorbance and native fluorescence detection modes and with sensitivity of detection down to 0.7 ⁇ g/mL.
  • the intrinsic solubility scores can be calculated using CamSol Intrinsic (Sormanni et al., J Mol Biol 427, 478-490, 2015).
  • the amino acid sequences for residues 95-102 (Kabat numbering) in CDRH3 of each antibody fragment such as a scFv can be evaluated via the online program to calculate the solubility scores.
  • autoreactive clones should be eliminated during ontogeny by negative selection, however it has become clear that many human naturally occurring antibodies with autoreactive properties persist in adult mature repertoires, and the autoreactivity may enhance the antiviral function of many antibodies to pathogens. It has been noted that HCDR3 loops in antibodies during early B cell development are often rich in positive charge and exhibit autoreactive patterns (Wardemann et al., Science 301, 1374- 1377, 2003). A given antibody may be tested for autoreactivity by assessing the level of binding to human origin cells in microscopy (using adherent HeLa or HEp-2 epithelial cells) and flow cytometric cell surface staining (using suspension Jurkat T cells and 293S human embryonic kidney cells).
  • HL Human Likeness
  • rHL Relative Human Likeness
  • Antibodies can be engineered to eliminate residues that are not common in human repertoires.
  • a single chain variable fragment (scFv) is a fusion of the variable regions of the heavy and light chains of immunoglobulins, linked together with a short (usually serine, glycine) linker. This chimeric molecule retains the specificity of the original immunoglobulin, despite removal of the constant regions and the introduction of a linker peptide.
  • scFv can be created directly from subcloned heavy and light chains derived from a hybridoma or B cell.
  • Single chain variable fragments lack the constant Fc region found in complete antibody molecules, and thus, the common binding sites (e.g., protein A/G) used to purify antibodies.
  • These fragments can often be purified/immobilized using Protein L since Protein L interacts with the variable region of kappa light chains.
  • Flexible linkers generally are comprised of helix- and turn-promoting amino acid residues such as alanine, serine and glycine.
  • the recombinant antibodies of the present disclosure may also involve sequences or moieties that permit dimerization or multimerization of the receptors. Such sequences include those derived from IgA, which permit formation of multimers in conjunction with the J-chain. Another multimerization domain is the Gal4 dimerization domain.
  • the chains may be modified with agents such as biotin/avidin, which permit the combination of two antibodies.
  • a single-chain antibody can be created by joining receptor light and heavy chains using a non-peptide linker or chemical unit.
  • the light and heavy chains will be produced in distinct cells, purified, and subsequently linked together in an appropriate fashion (i.e., the N-terminus of the heavy chain being attached to the C-terminus of the light chain via an appropriate chemical bridge).
  • Cross-linking reagents are used to form molecular bridges that tie functional groups of two different molecules, e.g., a stabilizing and coagulating agent.
  • dimers or multimers of the same analog or heteromeric complexes comprised of different analogs can be created.
  • hetero- bifunctional cross-linkers can be used that eliminate unwanted homopolymer formation.
  • An exemplary hetero-bifunctional cross-linker contains two reactive groups: one reacting with primary amine group (e.g., N-hydroxy succinimide) and the other reacting with a thiol group (e.g., pyridyl disulfide, maleimides, halogens, etc.).
  • primary amine group e.g., N-hydroxy succinimide
  • a thiol group e.g., pyridyl disulfide, maleimides, halogens, etc.
  • the cross-linker may react with the lysine residue(s) of one protein (e.g., the selected antibody or fragment) and through the thiol reactive group, the cross-linker, already tied up to the first protein, reacts with the cysteine residue (free sulfhydryl group) of the other protein (e.g., the selective agent).
  • a cross-linker having reasonable stability in blood will be employed.
  • Numerous types of disulfide-bond containing linkers are known that can be successfully employed to conjugate targeting and therapeutic/preventative agents. Linkers that contain a disulfide bond that is sterically hindered may prove to give greater stability in vivo, preventing release of the targeting peptide prior to reaching the site of action. These linkers are thus one group of linking agents.
  • Another cross-linking reagent is SMPT, which is a bifunctional cross-linker containing a disulfide bond that is "sterically hindered" by an adjacent benzene ring and methyl groups.
  • steric hindrance of the disulfide bond serves a function of protecting the bond from attack by thiolate anions such as glutathione which can be present in tissues and blood, and thereby help in preventing decoupling of the conjugate prior to the delivery of the attached agent to the target site.
  • the SMPT cross-linking reagent lends the ability to cross-link functional groups such as the SH of cysteine or primary amines (e.g., the epsilon amino group of lysine).
  • cross-linker includes the hetero- bifunctional photoreactive phenylazides containing a cleavable disulfide bond such as sulfosuccinimidy1-2-(p-azido salicylamido) ethy1-1,3'-dithiopropionate.
  • the N-hydroxy- succinimidy l group reacts with primary amino groups and the phenylazide (upon photolysis) reacts non-selectively with any amino acid residue.
  • non-hindered linkers also can be employed in accordance herewith.
  • cross-linkers not considered to contain or generate a protected disulfide, include SATA, SPDP and 2-iminothiolane (Wawrzynczak & Thorpe, 1987).
  • SATA SATA
  • SPDP 2-iminothiolane
  • Another embodiment includes the use of flexible linkers.
  • U.S. Patents 5,141,648 and 5,563,250 disclose cleavable conjugates containing a labile bond that is cleavable under a variety of mild conditions.
  • This linker is particularly useful in that the agent of interest may be bonded directly to the linker, with cleavage resulting in release of the active agent.
  • Particular uses include adding a free amino or free sulfhydryl group to a protein, such as an antibody, or a drug.
  • U.S. Patent 5,856,456 provides peptide linkers for use in connecting polypeptide constituents to make fusion proteins, e.g., single chain antibodies.
  • the linker is up to about 50 amino acids in length, contains at least one occurrence of a charged amino acid (preferably arginine or lysine) followed by a praline, and is characterized by greater stability and reduced aggregation.
  • Patent 5,880,270 discloses aminooxy-containing linkers useful in a variety of immunodiagnostic and separative techniques.
  • antibodies of the present disclosure are bispecific or multispecific.
  • Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes.
  • Exemplary bispecific antibodies may bind to two different epitopes of a single antigen.
  • Other such antibodies may combine a first antigen binding site with a binding site for a second antigen.
  • an anti-pathogen arm may be combined with an arm that binds to a triggering molecule on a leukocyte, such as a T-cell receptor molecule (e.g., CD3), or Fe receptors for IgG ( Fc ⁇ R ), such as Fc ⁇ R I (CD64), Fc ⁇ R II (CD32) and Fc ⁇ R III (CD16), so as to focus and localize cellular defense mechanisms to the infected cell.
  • a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g., CD3), or Fe receptors for IgG ( Fc ⁇ R ), such as Fc ⁇ R I (CD64), Fc ⁇ R II (CD32) and Fc ⁇ R III (CD16), so as to focus and localize cellular defense mechanisms to the infected cell.
  • Bispecific antibodies may also be used to localize cytotoxic agents to infected cells.
  • bispecific antibodies possess a pathogen-binding arm and an arm that binds the cytotoxic agent (e.g., saporin, anti-interferon-a, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten).
  • cytotoxic agent e.g., saporin, anti-interferon-a, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten.
  • Bispecific antibodies can be prepared as full-length antibodies or antibody fragments (e.g., F(ab') 2 bispecific antibodies).
  • WO 96/16673 describes a bispecific anti- ErbB2/anti-Fc gamma RIII antibody
  • U.S. Patent 5,837,234 discloses a bispecific anti- ErbB2/anti-Fc gamma RI antibody.
  • a bispecific anti-ErbB2/Fc alpha antibody is shown in WO98/02463.
  • U.S. Patent 5,821,337 teaches a bispecific anti-ErbB2/anti-CD3 antibody.
  • Methods for making bispecific antibodies are known in the art.
  • Traditional production of full- length bispecific antibodies is based on the co-expression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305:537-539 (1983)).
  • these hybridomas quadromas
  • these hybridomas produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low.
  • antibody variable regions with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion is with an lg heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light chain bonding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co-transfected into a suitable host cell.
  • This provides for greater flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yield of the desired bispecific antibody. It is, however, possible to insert the coding sequences for two or all three polypeptide chains into a single expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios have no significant effect on the yield of the desired chain combination.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation.
  • This approach is disclosed in WO 94/04690.
  • For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986). According to another approach described in U.S.
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers that are recovered from recombinant cell culture.
  • the preferred interface comprises at least a part of the CH3 domain.
  • one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g., tyrosine or tryptophan).
  • Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g., alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
  • Bispecific antibodies include cross-linked or "heteroconjugate" antibodies.
  • one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin.
  • Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Patent 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089).
  • Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Patent 4,676,980, along with a number of cross-linking techniques. Techniques for generating bispecific antibodies from antibody fragments have also been described in the literature.
  • bispecific antibodies can be prepared using chemical linkage.
  • Brennan et al., Science, 229: 81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab')2 fragments. These fragments are reduced in the presence of the dithiol complexing agent, sodium arsenite, to stabilize vicinal dithiols and prevent intermolecular disulfide formation.
  • the Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives.
  • One of the Fab'-TNB derivatives is then reconverted to the Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody.
  • the bispecific antibodies produced can be used as agents for the selective immobilization of enzymes. Techniques exist that facilitate the direct recovery of Fab'-SH fragments from E. coli, which can be chemically coupled to form bispecific antibodies. Shalaby et al., J. Exp. Med., 175: 217-225 (1992) describe the production of a humanized bispecific antibody F(ab')2 molecule. Each Fab' fragment was separately secreted from E.
  • bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.
  • Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described (Merchant et al., Nat. Bioteclmo!, 16, 677-681 (1998). doi:10.1038/nbt0798-677pmid:9661204).
  • bispecific antibodies have been produced using leucine zippers (Kostelny et al., J. Immunol., 148(5):1547-1553, 1992).
  • the leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion.
  • the antibody homodimers were reduced at the hinge region to form monomers and then re- oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers.
  • the "diabody” technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments.
  • the fragments comprise a VH connected to a VL by a linker that is too short to allow pairing between the two domains on the same chain.
  • a bispecific or multispecific antibody may be formed as a DOCK-AND-LOCK.TM (DNLTM) complex (see, e.g., U.S.
  • DDD dimerization and docking domain
  • R regulatory
  • AD anchor domain
  • the DDD and AD peptides may be attached to any protein, peptide or other rnolecule. Because the DDD sequences spontaneously dimerize, and bind to the AD sequence, the technique allows the formation of complexes between any selected molecules that may be attached to DDD or AD sequences.
  • Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared (Tutt et al., J. Immunol. 147: 60, 1991; Xu et al., Science, 358(6359):85-90, 2017).
  • a multivalent antibody may be internalized (and/or catabolized) faster than a bivalent antibody by a cell expressing an antigen to which the antibodies bind.
  • the antibodies of the present disclosure can be multivalent antibodies with three or more antigen binding sites (e.g., tetravalent antibodies), which can be readily produced by recombinant expression of nucleic acid encoding the polypeptide chains of the antibody.
  • the multivalent antibody can comprise a dimerization domain and three or more antigen binding sites.
  • the preferred dimerization domain comprises (or consists of) an Fc region or a hinge region. In this scenario, the antibody will comprise an Fc region and three or more antigen binding sites amino-terminal to the Fc region.
  • the preferred multivalent antibody herein comprises (or consists of) three to about eight, but preferably four, antigen binding sites.
  • the multivalent antibody comprises at least one polypeptide chain (and preferably two polypeptide chains), wherein the polypeptide chain(s) comprise two or more variable regions.
  • the polypeptide chain(s) may comprise VD1-(X1)n-VD2-(X2)n-Fc, wherein VD1 is a first variable region, VD2 is a second variable region, Fc is one polypeptide chain of an Fc region, X1 and X2 represent an amino acid or polypeptide, and n is 0 or 1.
  • the polypeptide chain(s) may comprise: VH-CH1-flexible linker-VH-CH1-Fc region chain; or VH-CH1-VH- CH1-Fc region chain.
  • the multivalent antibody herein preferably further comprises at least two (and preferably four) light chain variable region polypeptides.
  • the multivalent antibody herein may, for instance, comprise from about two to about eight light chain variable region polypeptides.
  • the light chain variable region polypeptides contemplated here comprise a light chain variable region and, optionally, further comprise a CL domain.
  • Charge modifications are particularly useful in the context of a multispecific antibody, where amino acid substitutions in Fab molecules result in reducing the mispairing of light chains with non-matching heavy chains (Bence-Jones-type side products), which can occur in the production of Fab-based bi-/multispecific antigen binding molecules with a VH/VLexchange in one (or more, in case of molecules comprising more than two antigen-binding Fab molecules) of their binding arms (see also PCT publication no. WO 2015/150447, particularly the examples therein, incorporated herein by reference in its entirety).
  • an antibody comprised in the therapeutic agent comprises (a) a first Fab molecule which specifically binds to a first antigen (b) a second Fab molecule which specifically binds to a second antigen, and wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, wherein the first antigen is an activating T cell antigen, and the second antigen is a target cell antigen, or the first antigen is a target cell antigen, and the second antigen is an activating T cell antigen; and wherein i) in the constant domain CL of the first Fab molecule under a) the amino acid at position 124 is substituted by a positively charged amino acid (numbering according to Kabat), and wherein in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 or the amino acid at position 213 is substituted by a negatively charged amino acid (numbering according to Kabat EU index); or ii) in the constant domain CL of the first
  • the antibody may not comprise both modifications mentioned under i) and ii).
  • the constant domains CL and CH1 of the second Fab molecule are not replaced by each other (i.e., remain unexchanged).
  • the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat) (in one preferred embodiment independently by lysine (K) or arginine (R)), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 or the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index).
  • the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index).
  • the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat) (in one preferred embodiment independently by lysine (K) or arginine (R)) and the amino acid at position 123 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat) (in one preferred embodiment independently by lysine (K) or arginine (R)), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index).
  • the amino acid at position 124 is substituted by lysine (K) (numbering according to Kabat) and the amino acid at position 123 is substituted by lysine (K) or arginine (R) (numbering according to Kabat)
  • the amino acid at position 147 is substituted by glutamic acid (E) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted by glutamic acid (E) (numbering according to Kabat EU index).
  • the amino acid at position 124 is substituted by lysine (K) (numbering according to Kabat) and the amino acid at position 123 is substituted by arginine (R) (numbering according to Kabat), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 is substituted by glutamic acid (E) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted by glutamic acid (E) (numbering according to Kabat EU index).
  • T cell receptors also known as chimeric T cell receptors, chimeric immunoreceptors, chimeric antigen receptors (CARs)
  • CARs chimeric antigen receptors
  • these receptors are used to graft the specificity of a monoclonal antibody onto a T cell, with transfer of their coding sequence facilitated by retroviral vectors. In this way, a large number of target- specific T cells can be generated for adoptive cell transfer. Phase I clinical studies of this approach show efficacy.
  • scFv single-chain variable fragments
  • scFv single-chain variable fragments
  • Such molecules result in the transmission of a zeta signal in response to recognition by the scFv of its target.
  • An example of such a construct is 14g2a-Zeta, which is a fusion of a scFv derived from hybridoma 14g2a (which recognizes disialoganglioside GD2).
  • T cells express this molecule (usually achieved by oncoretroviral vector transduction), they recognize and kill target cells that express GD2 (e.g., neuroblastoma cells).
  • variable portions of an immunoglobulin heavy and light chain are fused by a flexible linker to form a scFv.
  • This scFv is preceded by a signal peptide to direct the nascent protein to the endoplasmic reticulum and subsequent surface expression (this is cleaved).
  • a flexible spacer allows to the scFv to orient in different directions to enable antigen binding.
  • the transmembrane domain is a typical hydrophobic alpha helix usually derived from the original molecule of the signaling endodomain which protrudes into the cell and transmits the desired signal.
  • Type I proteins are in fact two protein domains linked by a transmembrane alpha helix in between.
  • a signal peptide directs the nascent protein into the endoplasmic reticulum. This is essential if the receptor is to be glycosylated and anchored in the cell membrane. Any eukaryotic signal peptide sequence usually works fine. Generally, the signal peptide natively attached to the amino-terminal most component is used (e.g., in a scFv with orientation light chain - linker - heavy chain, the native signal of the light-chain is used The antigen recognition domain is usually an scFv. There are however many alternatives.
  • TCR T-cell receptor alpha and beta single chains
  • simple ectodomains e.g., CD4 ectodomain torecognize HIV infected cells
  • more exotic recognition components such as a linked cytokine (which leads to recognition of cells bearing the cytokine receptor).
  • a spacer region links the antigen binding domain to the transmembrane domain. It should be flexible enough to allow the antigen binding domain to orient in different directions to facilitate antigen recognition.
  • the simplest form is the hinge region from IgG1. Alternatives include the CH2CH3 region of immunoglobulin and portions of CD3.
  • the transmembrane domain is a hydrophobic alpha helix that spans the membrane. Generally, the transmembrane domain from the most membrane proximal component of the endodomain is used. Interestingly, using the CD3-zeta transmembrane domain may result in incorporation of the artificial TCR into the native TCR a factor that is dependent on the presence of the native CD3-zeta transmembrane charged aspartic acid residue. Different transmembrane domains result in different receptor stability. The CD28 transmembrane domain results in a brightly expressed, stable receptor. The endodomain is the "business-end" of the receptor.
  • CD3-zeta which contains 3 ITAMs. This transmits an activation signal to the T cell after antigen is bound. CD3-zeta may not provide a fully competent activation signal and additional co-stimulatory signaling is needed.
  • First-generation CARs typically had the intracellular domain from the CD3E- chain, which is the primary transmitter of signals from endogenous TCRs.
  • Synecond-generation CARs add intracellular signaling domains from various costimulatory protein receptors (e.g., CD28, 41BB, ICOS) to the cytoplasmic tail of the CAR to provide additional signals to the T cell.
  • ADCs are complex molecules composed of an antibody (a whole mAb or an antibody fragment such as a single-chain variable fragment, or scFv) linked, via a stable chemical linker with labile bonds, to a biological active cytotoxic/anti-viral payload or drug.
  • Antibody Drug Conjugates are examples of bioconjugates and immunoconjugates.
  • antibody-drug conjugates allow sensitive discrimination between healthy and diseased tissue. This means that, in contrast to traditional systemic approaches, antibody-drug conjugates target and attack the infected cell so that healthy cells are less severely affected.
  • an anticancer drug e.g., a cell toxin or cytotoxin
  • a certain cell marker e.g., a protein that, ideally, is only to be found in or on infected cells.
  • Antibodies track these proteins down in the body and attach themselves to the surface of cancer cells.
  • the biochemical reaction between the antibody and the target protein (antigen) triggers a signal in the tumour cell, which then absorbs or internalizes the antibody together with the cytotoxin.
  • the cytotoxic drug is released and kills the cell or impairs viral replication. Due to this targeting, ideally the drug has lower side effects and gives a wider therapeutic window than other agents.
  • a stable link between the antibody and cytotoxic/anti-viral agent is a crucial aspect of an ADC.
  • Linkers are based on chemical motifs including disulfides, hydrazones or peptides (cleavable), or thioethers (noncleavable) and control the distribution and delivery of the cytotoxic agent to the target cell. Cleavable and noncleavable types of linkers have been proven to be safe in preclinical and clinical trials.
  • Brentuximab vedotin includes an enzyme- sensitive cleavable linker that delivers the potent and highly toxic antimicrotubule agent Monomethyl auristatin E or MMAE, a synthetic antineoplastic agent, to human specific CD30-positive malignant cells.
  • MMAE which inhibits cell division by blocking the polymerization of tubulin, cannot be used as a single-agent chemotherapeutic drug.
  • cAClO a cell membrane protein of the tumor necrosis factor or TNF receptor
  • Trastuzumab emtansine is a combination of the microtubule-formation inhibitor mertansine (DM- 1), a derivative of the Maytansine, and antibody trastuzumab (Herceptin®/Genentech/Roche) attached by a stable, non-cleavable linker.
  • DM-1 microtubule-formation inhibitor mertansine
  • a derivative of the Maytansine a derivative of the Maytansine
  • antibody trastuzumab Herceptin®/Genentech/Roche
  • the type of linker, cleavable or noncleavable lends specific properties to the cytotoxic (anti-cancer) drug. For example, a non-cleavable linker keeps the drug within the cell.
  • Bi-specific T-cell engagers are a class of artificial bispecific monoclonal antibodies that are investigated for the use as anti-cancer drugs. They direct a host's immune system, more specifically the T cells' cytotoxic activity, against infected cells. BiTE is a registered trademark of Micromet AG.
  • BiTEs are fusion proteins consisting of two single-chain variable fragments (scFvs) of different antibodies, or amino acid sequences from four different genes, on a single peptide chain of about 55 kilodaltons.
  • scFvs single-chain variable fragments
  • One of the scFvs binds to T cells via the CD3 receptor, and the other to an infected cell via a specific molecule.
  • BiTEs form a link between T cells and target cells. This causes T cells to exert cytotoxic/anti-viral activity on infected cells by producing proteins like perforin and granzymes, independently of the presence of MHC I or co-stimulatory molecules. These proteins enter infected cells and initiate the cell's apoptosis.
  • the antibody is a recombinant antibody that is suitable for action inside of a cell - such antibodies are known as "intrabodies.”
  • Intrabodies these antibodies may interfere with target function by a variety of mechanism, such as by altering intracellular protein trafficking, interfering with enzymatic function, and blocking protein-protein or protein-DNA interactions.
  • their structures mimic or parallel those of single chain and single domain antibodies, discussed above. Indeed, single-transcript/single-chain is an important feature that permits intracellular expression in a target cell, and also makes protein transit across cell membranes more feasible. However, additional features are required.
  • intrabody therapeutic The two major issues impacting the implementation of intrabody therapeutic are delivery, including cell/tissue targeting, and stability.
  • delivery a variety of approaches have been employed, such as tissue-directed delivery, use of cell-type specific promoters, viral-based delivery and use of cell-permeability/membrane translocating peptides.
  • the approach is generally to either screen by brute force, including methods that involve phage display and may include sequence maturation or development of consensus sequences, or more directed modifications such as insertion stabilizing sequences (e.g., Fc regions, chaperone protein sequences, leucine zippers) and disulfide replacement/modification.
  • insertion stabilizing sequences e.g., Fc regions, chaperone protein sequences, leucine zippers
  • disulfide replacement/modification An additional feature that intrabodies may require is a signal for intracellular targeting.
  • Intrabodies or other proteins
  • intrabodies have been designed and are commercially available (Invitrogen Corp.; Persic et al., 1997). By virtue of their ability to enter cells, intrabodies have additional uses that other types of antibodies may not achieve.
  • the ability to interact with the MUCl cytoplasmic domain in a living cell may interfere with functions associated with the MUCl CD, such as signalling functions (binding to other molecules) or oligomer formation.
  • signalling functions binding to other molecules
  • oligomer formation such antibodies can be used to inhibit MUC1 dimer formation.
  • the antibodies of the present disclosure may be purified.
  • purified is intended to refer to a composition, isolatable from other components, wherein the protein is purified to any degree relative to its naturally-obtainable state.
  • a purified protein therefore also refers to a protein, free from the environment in which it may naturally occur.
  • substantially purified this designation will refer to a composition in which the protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the proteins in the composition.
  • Protein purification techniques are well known to those of skill in the art. These techniques involve, at one level, the crude fractionation of the cellular milieu to polypeptide and non- polypeptide fractions.
  • the polypeptide of interest may be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or purification to homogeneity).
  • Analytical methods particularly suited to the preparation of a pure peptide are ion-exchange chromatography, exclusion chromatography; polyacrylamide gel electrophoresis; isoelectric focusing.
  • Other methods for protein purification include, precipitation with ammonium sulfate, PEG, antibodies and the like or by heat denaturation, followed by centrifugation; gel filtration, reverse phase, hydroxylapatite and affinity chromatography; and combinations of such and other techniques.
  • an antibody of the present disclosure it may be desirable to express the polypeptide in a prokaryotic or eukaryotic expression system and extract the protein using denaturing conditions.
  • the polypeptide may be purified from other cellular components using an affinity column, which binds to a tagged portion of the polypeptide.
  • affinity column which binds to a tagged portion of the polypeptide.
  • the order of conducting the various purification steps may be changed, or that certain steps may be omitted, and still result in a suitable method for the preparation of a substantially purified protein or peptide.
  • complete antibodies are fractionated utilizing agents (i.e., protein A) that bind the Fc portion of the antibody.
  • antigens may be used to simultaneously purify and select appropriate antibodies.
  • Such methods often utilize the selection agent bound to a support, such as a column, filter or bead.
  • the antibodies are bound to a support, contaminants removed (e.g., washed away), and the antibodies released by applying conditions (salt, heat, etc.).
  • Various methods for quantifying the degree of purification of the protein or peptide will be known to those of skill in the art in light of the present disclosure. These include, for example, determining the specific activity of an active fraction, or assessing the amount of polypeptides within a fraction by SDS/PAGE analysis. Another method for assessing the purity of a fraction is to calculate the specific activity of the fraction, to compare it to the specific activity of the initial extract, and to thus calculate the degree of purity.
  • the actual units used to represent the amount of activity will, of course, be dependent upon the particular assay technique chosen to follow the purification and whether or not the expressed protein or peptide exhibits a detectable activity. It is known that the migration of a polypeptide can vary, sometimes significantly, with different conditions of SDS/PAGE (Capaldi et al., 1977). It will therefore be appreciated that under differing electrophoresis conditions, the apparent molecular weights of purified or partially purified expression products may vary.
  • the present disclosure provides pharmaceutical compositions comprising anti-SARS- CoV- 2 virus antibodies and antigens for generating the same.
  • compositions comprise a prophylactically or therapeutically effective amount of an antibody or a fragment thereof, or a peptide immunogen, and a pharmaceutically acceptable carrier.
  • the compositions of the invention may include an "effective amount” or “therapeutically effective amount” or a “prophylactically effective amount” of an antibody or antigen-binding portion of the invention. These terms are used interchangeably.
  • a “therapeutically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
  • a therapeutically effective amount of the antibody or antibody portion may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the antibody or antibody portion to elicit a desired response in the subject.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the term “treat”, “treating” or “treatment” of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment “treat”, “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • “treat”, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
  • “treat”, “treating” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder.
  • "Prevention" of a condition or disorder refers to delaying or preventing the onset of a condition or disorder or reducing its severity, as assessed by the appearance or extent of one or more symptoms of the condition or disorder.
  • the term “subject” refers to an animal. Typically the animal is a mammal.
  • a subject also refers to for example, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like.
  • the subject is a primate.
  • the subject is a human.
  • a subject is “in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
  • the term "pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavouring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp.1289- 1329).
  • carrier refers to a diluent, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a particular carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • compositions include starch, glucose, lactose, sucrose, gelatine, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical agents are described in "Remington's Pharmaceutical Sciences.” Such compositions will contain a prophylactically or therapeutically effective amount of the antibody or fragment thereof, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration, which can be oral, intravenous, intraarterial, intrabuccal, intranasal, nebulized, bronchial inhalation, intra-rectal, vaginal, topical or delivered by mechanical ventilation.
  • Active vaccines are also envisioned where antibodies like those disclosed are produced in vivo in a subject at risk of SARS-CoV-2 infection.
  • Such vaccines can be formulated for parenteral administration, e.g., formulated for injection via the intradermal, intravenous, intramuscular, subcutaneous, or even intraperitoneal routes. Administration by intradermal and intramuscular routes are contemplated.
  • the vaccine could alternatively be administered by a topical route directly to the mucosa, for example, by nasal drops, inhalation, by nebulizer, or via intrarectal or vaginal delivery.
  • Pharmaceutically acceptable salts include the acid salts and those which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like. Passive transfer of antibodies, known as artificially acquired passive immunity, generally will involve the use of intravenous or intramuscular injections.
  • the forms of antibody can be human or animal blood plasma or serum, as pooled human immunoglobulin for intravenous (IVIG) or intramuscular (IG) use, as high-titre human IVIG or IG from immunized or from donors recovering from disease, and as monoclonal antibodies (MAb).
  • IVIG intravenous
  • IG intramuscular
  • MAb monoclonal antibodies
  • Such immunity generally lasts for only a short period of time, and there is also a potential risk for hypersensitivity reactions, and serum sickness, especially from gamma globulin of non- human origin.
  • passive immunity provides immediate protection.
  • the antibodies will be formulated in a carrier suitable for injection, i.e., sterile and syringeable.
  • compositions of the disclosure are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water- free concentrate in a hermetically sealed container such as an ampoule or sachet indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachet indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the compositions of the disclosure can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the target cells are cells to which antibodies or fragments thereof comprising an Fc region specifically bind, generally via the protein part that is N-terminal to the Fc region.
  • ADCC antibody having increased/reduced antibody dependent cell-mediated cytotoxicity
  • ADCC antibody having increased/reduced ADCC as determined by any suitable method known to those of ordinary skill in the art.
  • the term “increased/reduced ADCC” is defined as either an increase/reduction in the number of target cells that are lysed in a given time, at a given concentration of antibody in the medium surrounding the target cells, by the mechanism of ADCC defined above, and/or a reduction/increase in the concentration of antibody, in the medium surrounding the target cells, required to achieve the lysis of a given number of target cells in a given time, by the mechanism of ADCC.
  • ADCC increase/reduction in ADCC is relative to the ADCC mediated by the same antibody produced by the same type of host cells, using the same standard production, purification, formulation and storage methods (which are known to those skilled in the art), but that has not been engineered.
  • ADCC mediated by an antibody produced by host cells engineered to have an altered pattern of glycosylation e.g., to express the glycosyltransferase, GnTIII, or other glycosyltransferases
  • CDC Complement-dependent cytotoxicity
  • Antibodies of the present disclosure may be linked to at least one agent to form an antibody conjugate. In order to increase the efficacy of antibody molecules as diagnostic or therapeutic agents, it is conventional to link or covalently bind or complex at least one desired molecule or moiety. Such a molecule or moiety may be, but is not limited to, at least one effector or reporter molecule.
  • Effector molecules comprise molecules having a desired activity, e.g., cytotoxic activity.
  • Non-limiting examples of effector molecules which have been attached to antibodies include toxins, anti-tumour agents, therapeutic enzymes, radionuclides, antiviral agents, chelating agents, cytokines, growth factors, and oligo- or polynucleotides.
  • a reporter molecule is defined as any moiety which may be detected using an assay.
  • Non- limiting examples of reporter molecules which have been conjugated to antibodies include enzymes, radiolabels, haptens, fluorescent labels, phosphorescent molecules chemiluminescent molecules, chromophores, photoaffinity molecules, colored particles or ligands, such as biotin.
  • Antibody conjugates are generally preferred for use as diagnostic agents.
  • Antibody diagnostics generally fall within two classes, those for use in in vitro diagnostics, such as in a variety of immunoassays, and those for use in vivo diagnostic protocols, generally known as "antibody-directed imaging.”
  • Many appropriate imaging agents are known in the art, as are methods for their attachment to antibodies (see, for e.g., U.S. Patents 5,021,236, 4,938,948, and 4,472,509).
  • the imaging moieties used can be paramagnetic ions, radioactive isotopes, fluorochromes, NMR-detectable substances, and X-ray imaging agents.
  • paramagnetic ions such as chromium (III), manganese (II), iron (III), iron (II), cobalt (II), nickel (II), copper (II), neodymium (III), samarium (III), ytterbium (III), gadolinium (III), vanadium (II), terbium (III), dysprosium (III), holmium (III) and/or erbium (III), with gadolinium being particularly preferred.
  • Ions useful in other contexts, such as X-ray imaging include but are not limited to lanthanum (III), gold (III), lead (II), and especially bismuth (III).
  • radioactive isotopes for therapeutic and/or diagnostic application, one might mention astatine211, 14carbon, 51chromium, 36chlorine, 57cobalt, 58cobalt, copper67, 152Eu, gallium67, 3hydrogen, iodine123, iodine125, iodine131, indium111, 59iron, 32phosphorus, rhenium186, rhenium188, 75selenium, 35sulphur, technicium99m and/or yttrium90.125I is often being preferred for use in certain embodiments, and technicium99m and/or indium111 are also often preferred due to their low energy and suitability for long range detection.
  • Radioactively labelled monoclonal antibodies of the present disclosure may be produced according to well-known methods in the art.
  • monoclonal antibodies can be iodinated by contact with sodium and/or potassium iodide and a chemical oxidizing agent such as sodium hypochlorite, or an enzymatic oxidizing agent, such as lactoperoxidase.
  • Monoclonal antibodies according to the disclosure may be labelled with technetium99m by ligand exchange process, for example, by reducing pertechnate with stannous solution, chelating the reduced technetium onto a Sephadex column and applying the antibody to this column.
  • direct labelling techniques may be used, e.g., by incubating pertechnate, a reducing agent such as SnCl 2 , a buffer solution such as sodium- potassium phthalate solution, and the antibody.
  • Intermediary functional groups which are often used to bind radioisotopes which exist as metallic ions to antibody are diethylenetriaminepentaacetic acid (DTPA) or ethylene diaminetetracetic acid (EDTA).
  • fluorescent labels contemplated for use as conjugates include Alexa 350, Alexa 430, AMCA, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, Cascade Blue, Cy3, Cy5,6-FAM, Fluorescein Isothiocyanate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rhodamine Green, Rhodamine Red, Renographin, ROX, TAMRA, TET, Tetramethylrhodamine, and/or Texas Red.
  • antibodies contemplated in the present disclosure are those intended primarily for use in vitro, where the antibody is linked to a secondary binding ligand and/or to an enzyme (an enzyme tag) that will generate a coloured product upon contact with a chromogenic substrate.
  • suitable enzymes include urease, alkaline phosphatase, (horseradish) hydrogen peroxidase or glucose oxidase.
  • Preferred secondary binding ligands are biotin and avidin and streptavidin compounds. The use of such labels is well known to those of skill in the art and are described, for example, in U.S.
  • Yet another known method of site-specific attachment of molecules to antibodies comprises the reaction of antibodies with hapten-based affinity labels.
  • hapten-based affinity labels react with amino acids in the antigen binding site, thereby destroying this site and blocking specific antigen reaction.
  • Molecules containing azido groups may also be used to form covalent bonds to proteins through reactive nitrene intermediates that are generated by low intensity ultraviolet light (Potter and Haley, 1983).
  • 2- and 8-azido analogues of purine nucleotides have been used as site-directed photoprobes to identify nucleotide binding proteins in crude cell extracts (Owens & Haley, 1987; Atherton et al., 1985).
  • the 2- and 8-azido nucleotides have also been used to map nucleotide binding domains of purified proteins (Khatoon et al., 1989; King et al., 1989; Dholakia et al., 1989) and may be used as antibody binding agents.
  • Several methods are known in the art for the attachment or conjugation of an antibody to its conjugate moiety.
  • Some attachment methods involve the use of a metal chelate complex employing, for example, an organic chelating agent such a diethylenetriaminepentaacetic acid anhydride (DTPA); ethylenetriaminetetraacetic acid; N-chloro-p-toluenesulfonamide; and/or tetrachloro-3a-6a-diphenylglycouril-3 attached to the antibody (U.S. Patents 4,472,509 and 4,938,948).
  • DTPA diethylenetriaminepentaacetic acid anhydride
  • ethylenetriaminetetraacetic acid N-chloro-p-toluenesulfonamide
  • tetrachloro-3a-6a-diphenylglycouril-3 attached to the antibody
  • Monoclonal antibodies may also be reacted with an enzyme in the presence of a coupling agent such as glutaraldehyde or periodate.
  • Conjugates with fluorescein markers are prepared in
  • U.S. Patent 4,938,948 imaging of breast tumours is achieved using monoclonal antibodies and thedetectable imaging moieties are bound to the antibody using linkers such as methyl-p- hydroxybenzimidate or N-succinimidy1-3-(4- hydroxypheny1)propionate.
  • linkers such as methyl-p- hydroxybenzimidate or N-succinimidy1-3-(4- hydroxypheny1)propionate.
  • derivatization of immunoglobulins by selectively introducing sulfhydryl groups in the Fc region of an immunoglobulin, using reaction conditions that do not alter the antibody combining site are contemplated.
  • Antibody conjugates produced according to this methodology are disclosed to exhibit improved longevity, specificity and sensitivity (U.S. Patent 5,196,066, incorporated herein by reference).
  • the present disclosure concerns immunodetection methods for binding, purifying, removing, quantifying and otherwise generally detecting SARS-CoV- 2 and its associated antigens. While such methods can be applied in a traditional sense, another use will be in quality control and monitoring of vaccine and other virus stocks, where antibodies according to the present disclosure can be used to assess the amount or integrity (i.e., long term stability) of antigens in viruses.
  • the methods may be used to screenmvarious antibodies for appropriate/desired reactivity profiles.
  • Other immunodetection methods include specific assays for determining the presence of SARS-CoV-2 in a subject.
  • assay formats are contemplated, but specifically those that would be used to detect SARS-CoV-2 in a fluid obtained from a subject, such as saliva, blood, plasma, sputum, semen or urine.
  • semen has been demonstrated as a viable sample for detecting SARS-CoV-2 (Purpura et al., 2016; Mansuy et al., 2016; Barzon et al., 2016; Garnet et al., 2016; Duffy et al., 2009; CDC, 2016; Halfon et al., 2010; Elder et al. 2005).
  • the assays may be advantageously formatted for non- healthcare (home) use, including lateral flow assays (see below) analogous to home pregnancy tests.
  • These assays may be packaged in the form of a kit with appropriate reagents and instructions to permit use by the subject of a family member.
  • Some immunodetection methods include enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoradiometric assay, fluoroimmunoassay, chemiluminescent assay, bioluminescent assay, and Western blot to mention a few.
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • immunoradiometric assay fluoroimmunoassay
  • fluoroimmunoassay fluoroimmunoassay
  • chemiluminescent assay chemiluminescent assay
  • bioluminescent assay bioluminescent assay
  • Western blot to mention a few.
  • a competitive assay for the detection and quantitation of SARS-CoV-2 antibodies directed to specific parasite epitopes in samples also is provided.
  • the steps of various useful immunodetection methods have been described in the scientific literature, such as, e.g., Doolittle and
  • the immunobinding methods include obtaining a sample suspected of containing SARS-CoV-2, and contacting the sample with a first antibody in accordance with the present disclosure, as the case may be, under conditions effective to allow the formation of immunocomplexes.
  • These methods include methods for purifying SARS-CoV-2 or related antigens from a sample.
  • the antibody will preferably be linked to a solid support, such as in the form of a column matrix, and the sample suspected of containing the SARS-CoV-2 or antigenic component will be applied to the immobilized antibody. The unwanted components will be washed from the column, leaving the SARS-CoV-2 antigen immunocomplexed to the immobilized antibody, which is then collected by removing the organism or antigen from the column.
  • the immunobinding methods also include methods for detecting and quantifying the amount of SARS-CoV-2 or related components in a sample and the detection and quantification of any immune complexes formed during the binding process.
  • a sample suspected of containing SARS-CoV-2 or its antigens and contact the sample with an antibody that binds SARS-CoV-2 or components thereof, followed by detecting and quantifying the amount of immune complexes formed under the specific conditions.
  • the biological sample analyzed may be any sample that is suspected of containing SARS-CoV-2 or SARS-CoV-2 antigen, such as a tissue section or specimen, a homogenized tissue extract, a biological fluid, including blood and serum, or a secretion, such as faeces or urine.
  • a tissue section or specimen such as a tissue section or specimen, a homogenized tissue extract, a biological fluid, including blood and serum, or a secretion, such as faeces or urine.
  • Contacting the chosen biological sample with the antibody under effective conditions and for a period of time sufficient to allow the formation of immune complexes (primary immune complexes) is generally a matter of simply adding the antibody composition to the sample and incubating the mixture for a period of time long enough for the antibodies to form immune complexes with, i.e., to bind to SARS-CoV-2 or antigens present.
  • sample-antibody composition such as a tissue section, ELISA plate, dot blot or Western blot
  • sample-antibody composition will generally be washed to remove any non-specifically bound antibody species, allowing only those antibodies specifically bound within the primary immune complexes to be detected.
  • detection of immunocomplex formation is well known in the art and may be achieved through the application of numerous approaches. These methods are based upon the detection of a label or marker, such as any of those radioactive, fluorescent, biological and enzymatic tags.
  • Patents concerning the use of such labels include U.S. Patents 3,817,837, 3,850,752, 3,939,350, 3,996,345, 4,277,437, 4,275,149 and 4,366,241.
  • a secondary binding ligand such as a second antibody and/or a biotin/avidin ligand binding arrangement, as is known in the art.
  • the antibody employed in the detection may itself be linked to a detectable label, wherein one would then simply detect this label, thereby allowing the amount of the primary immune complexes in the composition to be determined.
  • the first antibody that becomes bound within the primary immune complexes may be detected by means of a second binding ligand that has binding affinity for the antibody.
  • the second binding ligand may be linked to a detectable label.
  • the second binding ligand is itself often an antibody, which may thus be termed a "secondary" antibody.
  • the primary immune complexes are contacted with the labelled, secondary binding ligand, or antibody, under effective conditions and for a period of time sufficient to allow the formation of secondary immune complexes.
  • the secondary immune complexes are then generally washed to remove any non-specifically bound labelled secondary antibodies or ligands, and the remaining label in the secondary immune complexes is then detected. Further methods include the detection of primary immune complexes by a two-step approach.
  • a second binding ligand such as an antibody that has binding affinity for the antibody, is used to form secondary immune complexes, as described above.
  • the secondary immune complexes are contacted with a third binding ligand or antibody that has binding affinity for the second antibody, again under effective conditions and for a period of time sufficient to allow the formation of immune complexes (tertiary immune complexes).
  • the third ligand or antibody is linked to a detectable label, allowing detection of the tertiary immune complexes thus formed.
  • This system may provide for signal amplification if this is desired.
  • One method of immunodetection uses two different antibodies. A first biotinylated antibody is used to detect the target antigen, and a second antibody is then used to detect the biotin attached to the complexed biotin. In that method, the sample to be tested is first incubated in a solution containing the first step antibody.
  • the antibody/antigen complex is then amplified by incubation in successive solutions of streptavidin (or avidin), biotinylated DNA, and/or complementary biotinylated DNA, with each step adding additional biotin sites to the antibody/antigen complex.
  • streptavidin or avidin
  • biotinylated DNA or complementary biotinylated DNA
  • the amplification steps are repeated until a suitable level of amplification is achieved, at which point the sample is incubated in a solution containing the second step antibody against biotin.
  • This second step antibody is labelled, for example, with an enzyme that can be used to detect the presence of the antibody/antigen complex by histoenzymology using a chromogen substrate.
  • a conjugate can be produced which is macroscopically visible.
  • Another known method of immunodetection takes advantage of the immuno-PCR (Polymerase Chain Reaction) methodology.
  • the PCR method is similar to the Cantor method up to the incubation with biotinylated DNA, however, instead of using multiple rounds of streptavidin and biotinylated DNA incubation, the DNA/biotin/streptavidin/antibody complex is washed out with a low pH or high salt buffer that releases the antibody.
  • the resulting wash solution is then used to carry out a PCR reaction with suitable primers with appropriate controls.
  • the enormous amplification capability and specificity of PCR can be utilized to detect a single antigen molecule.
  • Immunoassays in their most simple and direct sense, are binding assays. Certain preferred immunoassays are the various types of enzyme linked immunosorbent assays (ELISAs) and radioimmunoassays (RIA) known in the art. Immunohistochemical detection using tissue sections is also particularly useful. However, it will be readily appreciated that detection is not limited to such techniques, and western blotting, dot blotting, FACS analyses, and the like may also be used. In one exemplary ELISA, the antibodies of the disclosure are immobilized onto a selected surface exhibiting protein affinity, such as a well in a polystyrene microtiter plate.
  • a test composition suspected of containing the SARS-CoV-2 or SARS-CoV-2 antigen is added to the wells. After binding and washing to remove non-specifically bound immune complexes, the bound antigen may be detected. Detection may be achieved by the addition of another anti-SARS-CoV-2 antibody that is linked to a detectable label. This type of ELISA is a simple "sandwich ELISA.” Detection may also be achieved by the addition of a second anti-SARS-CoV-2 antibody, followed by the addition of a third antibody that has binding affinity for the second antibody, with the third antibody being linked to a detectable label.
  • the samples suspected of containing the SARS-Co V-2 or SARS-CoV-2 antigen are immobilized onto the well surface and then contacted with the anti- SARS-CoV-2 antibodies of the disclosure. After binding and washing to remove non- specifically bound immune complexes, the bound anti-SARS-CoV-2 antibodies are detected. Where the initial anti-SARS-CoV-2 antibodies are linked to a detectable label, the immune complexes may be detected directly. Again, the immune complexes may be detected using a second antibody that has binding affinity for the first anti-SARS-CoV-2 antibody, with the second antibody being linked to a detectable label.
  • ELISAs have certain features in common, such as coating, incubating and binding, washing to remove non-specifically bound species, and detecting the bound immune complexes. These are described below.
  • coating a plate with either antigen or antibody one will generally incubate the wells of the plate with a solution of the antigen or antibody, either overnight or for a specified period of hours. The wells of the plate will then be washed to remove incompletely adsorbed material. Any remaining available surfaces of the wells are then "coated" with a nonspecific protein that is antigenically neutral with regard to the test antisera.
  • BSA bovine serum albumin
  • casein solutions of milk powder.
  • the coating allows for blocking of nonspecific adsorption sites on the immobilizing surface and thus reduces the background caused by nonspecific binding of antisera onto the surface.
  • ELISAs it is probably more customary to use a secondary or tertiary detection means rather than a direct procedure.
  • the immobilizing surface is contacted with the biological sample to be tested under conditions effective to allow immune complex (antigen/antibody) formation. Detection of the immune complex then requires a labelled secondary binding ligand or antibody, and a secondary binding ligand or antibody in conjunction with a labelled tertiary antibody or a third binding ligand.
  • Under conditions effective to allow immune complex (antigen/antibody) formation means that the conditions preferably include diluting the antigens and/or antibodies with solutions such as BSA, bovine gamma globulin (BGG) or phosphate buffered saline (PBS)/Tween. These added agents also tend to assist in the reduction of nonspecific background.
  • the "suitable” conditions also mean that the incubation is at a temperature or for a period of time sufficient to allow effective binding. Incubation steps are typically from about 1 to 2 to 4 hours or so, at temperatures preferably on the order of 25°C to 27°C, or may be overnight at about 4°C or so.
  • the contacted surface is washed so as to remove non-complexed material.
  • a preferred washing procedure includes washing with a solution such as PBS/Tween, or borate buffer. Following the formation of specific immune complexes between the test sample and the originally bound material, and subsequent washing, the occurrence of even minute amounts of immune complexes may be determined.
  • the second or third antibody will have an associated label to allow detection. Preferably, this will be an enzyme that will generate colour development upon incubating with an appropriate chromogenic substrate.
  • a urease glucose oxidase
  • alkaline phosphatase or hydrogen peroxidase-conjugated antibody
  • the amount of label is quantified, e.g., by incubation with a chromogenic substrate such as urea, or bromocresol purple, or 2,2'-azino-di-(3-ethyl-benzthiazoline-6- sulfonic acid (ABTS), or H 2 O 2 , in the case of peroxidase as the enzyme label. Quantification is then achieved by measuring the degree of color generated, e.g., using a visible spectra spectrophotometer. In another embodiment, the present disclosure contemplates the use of competitive formats. This is particularly useful in the detection of SARS-CoV-2 antibodies in sample.
  • an unknown amount of analyte or antibody is determined by its ability to displace a known amount of labelled antibody or analyte.
  • the quantifiable loss of a signal is an indication of the amount of unknown antibody or analyte in a sample.
  • the inventor proposes the use of labelled SARS-CoV-2 monoclonal antibodies to determine the amount of SARS-CoV-2 antibodies in a sample.
  • the basic format would include contacting a known amount of SARS-CoV-2 monoclonal antibody (linked to a detectable label) with SARS-CoV-2 antigen or particle.
  • the SARS-CoV-2 antigen or organism is preferably attached to a support.
  • the Western blot is an analytical technique used to detect specific proteins in a given sample of tissue homogenate or extract. It uses gel electrophoresis to separate native or denatured proteins by the length of the polypeptide (denaturing conditions) or by the 3-D structure of the protein (native/ non-denaturing conditions).
  • the proteins are then transferred to a membrane (typically nitrocellulose or PVDF), where they are probed (detected) using antibodies specific to the target protein.
  • Samples may be taken from whole tissue or from cell culture. In most cases, solid tissues are first broken down mechanically using a blender (for larger sample volumes), using a homogenizer (smaller volumes), or by sonication. Cells may also be broken open by one of the above mechanical methods. However, it should be noted that bacteria, virus or environmental samples can be the source of protein and thus Western blotting is not restricted to cellular studies only. Assorted detergents, salts, and buffers may be employed to encourage lysis of cells and to solubilize proteins.
  • Protease and phosphatase inhibitors are often added to prevent the digestion of the sample by its own enzymes. Tissue preparation is often done at cold temperatures to avoid protein denaturing.
  • the proteins of the sample are separated using gel electrophoresis. Separation of proteins may be by isoelectric point (pl), molecular weight, electric charge, or a combination of these factors. The nature of the separation depends on the treatment of the sample and the nature of the gel. This is a very useful way to determine a protein. It is also possible to use a two- dimensional (2-D) gel which spreads the proteins from a single sample out in two dimensions.
  • Proteins are separated according to isoelectric point (pH at which they have neutral net charge) in the first dimension, and according to their molecular weight in the second dimension.
  • they are moved from within the gel onto a membrane made of nitrocellulose or polyvinylidene difluoride (PVDF).
  • PVDF polyvinylidene difluoride
  • the membrane is placed on top of the gel, and a stack of filter papers placed on top of that. The entire stack is placed in a buffer solution which moves up the paper by capillary action, bringing the proteins with it.
  • Another method for transferring the proteins is called electroblotting and uses an electric current to pull proteins from the gel into the PVDF or nitrocellulose membrane. The proteins move from within the gel onto the membrane while maintaining the organization they had within the gel.
  • the proteins are exposed on a thin surface layer for detection (see below).
  • Both varieties of membrane are chosen for their non-specific protein binding properties (i.e., binds all proteins equally well). Protein binding is based upon hydrophobic interactions, as well as charged interactions between the membrane and protein. Nitrocellulose membranes are cheaper than PVDF but are far more fragile and do not stand up well to repeated probing. The uniformity and overall effectiveness of transfer of protein from the gel to the membrane can be checked by staining the membrane with Coomassie Brilliant Blue or Ponceau S dyes. Once transferred, proteins are detected using labelled primary antibodies, or unlabelled primary antibodies followed by indirect detection using labelled protein A or secondary labelled antibodies binding to the Fc region of the primary antibodies.
  • Lateral flow assays also known as lateral flow immunochromatographic assays, are simple devices intended to detect the presence (or absence) of a target analyte in sample (matrix) without the need for specialized and costly equipment, though many laboratory-based applications exist that are supported by reading equipment. Typically, these tests are used as low resources medical diagnostics, either for home testing, point of care testing, or laboratory use. A widely spread and well-known application is the home pregnancy test.
  • the technology is based on a series of capillary beds, such as pieces of porous paper or sintered polymer. Each of these elements has the capacity to transport fluid (e.g., urine) spontaneously.
  • the first element (the sample pad) acts as a sponge and holds an excess of sample fluid.
  • the fluid migrates to the second element (conjugate pad) in which the manufacturer has stored the so-called conjugate, a dried format of bio-active particles (see below) in a salt-sugar matrix that contains everything to guarantee an optimized chemical reaction between the target molecule (e.g., an antigen) and its chemical partner (e.g., antibody) that has been immobilized on the particle's surface.
  • the sample fluid dissolves the salt- sugar matrix, it also dissolves the particles and in one combined transport action the sample and conjugate mix while flowing through the porous structure. In this way, the analyte binds to the particles while migrating further through the third capillary bed.
  • This material has one or more areas (often called stripes) where a third molecule has been immobilized by the manufacturer. By the time the sample-conjugate mix reaches these strips, analyte has been bound on the particle and the third 'capture' molecule binds the complex. After a while, when more and more fluid has passed the stripes, particles accumulate and the stripe-area changes colour. Typically, there are at least two stripes: one (the control) that captures any particle and thereby shows that reaction conditions and technology worked fine, the second contains a specific capture molecule and only captures those particles onto which an analyte molecule has been immobilized. After passing these reaction zones, the fluid enters the final porous material- the wick - that simply acts as a waste container.
  • Lateral Flow Tests can operate as either competitive or sandwich assays. Lateral flow assays are disclosed in U.S. Patent 6,485,982.
  • the antibodies of the present disclosure may also be used in conjunction with both fresh- frozen and/or formalin-fixed, paraffin-embedded tissue blocks prepared for study by immunohistochemistry (IHC).
  • IHC immunohistochemistry
  • the method of preparing tissue blocks from these particulate specimens has been successfully used in previous IHC studies of various prognostic factors and is well known to those of skill in the art (Brown et al., 1990; Abbondanzo et al., 1990; Allred et al., 1990).
  • frozen-sections may be prepared by rehydrating 50 mg of frozen "pulverized” tissue at room temperature in phosphate buffered saline (PBS) in small plastic capsules; pelleting the particles by centrifugation; resuspending them in a viscous embedding medium (OCT); inverting the capsule and/or pelleting again by centrifugation; snap-freezing in -70°C isopentane; cutting the plastic capsule and/or removing the frozen cylinder of tissue; securing the tissue cylinder on a cryostat microtome chuck; and/or cutting 25-50 serial sections from the capsule.
  • whole frozen tissue samples may be used for serial section cuttings.
  • Permanent-sections may be prepared by a similar method involving rehydration of the 50 mg sample in a plastic microfuge tube; pelleting; resuspending in 10% formalin for 4 hours fixation; washing/pelleting; resuspending in warm 2.5% agar; pelleting; cooling in ice water to harden the agar; removing the tissue/agar block from the tube; infiltrating and/or embedding the block in paraffin; and/or cutting up to 50 serial permanent sections. Again, whole tissue samples may be substituted.
  • the present disclosure concerns immunodetection kits for use with the immunodetection methods described above.
  • the antibodies may be used to detect SARS-CoV-2 or SARS-CoV-2 antigens
  • the antibodies may be included in the kit.
  • the immunodetection kits will thus comprise, in suitable container means, a first antibody that binds to SARS-CoV-2 or SARS-CoV-2 antigen, and optionally an immunodetection reagent.
  • the SARS-CoV-2 antibody may be pre-bound to a solid support, such as a column matrix and/or well of a microtiter plate.
  • the immunodetection reagents of the kit may take any one of a variety of forms, including those detectable labels that are associated with or linked to the given antibody.
  • Detectable labels that are associated with or attached to a secondary binding ligand are also contemplated.
  • Exemplary secondary ligands are those secondary antibodies that have binding affinity for the first antibody.
  • Further suitable immunodetection reagents for use in the present kits include the two- component reagent that comprises a secondary antibody that has binding affinity for the first antibody, along with a third antibody that has binding affinity for the second antibody, the third antibody being linked to a detectable label.
  • a number of exemplary labels are known in the art and all such labels may be employed in connection with the present disclosure.
  • kits may further comprise a suitably aliquoted composition of the SARS-CoV-2 or SARS-CoV-2 antigens, whether labelled or unlabelled, as may be used to prepare a standard curve for a detection assay.
  • the kits may contain antibody-label conjugates either in fully conjugated form, in the form of intermediates, or as separate moieties to be conjugated by the user of the kit.
  • the components of the kits may be packaged either in aqueous media or in lyophilized form.
  • the container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which the antibody may be placed, or preferably, suitably aliquoted.
  • kits of the present disclosure will also typically include a means for containing the antibody, antigen, and any other reagent containers in close confinement for commercial sale.
  • Such containers may include injection or blow-moulded plastic containers into which the desired vials are retained.
  • the present disclosure also contemplates the use of antibodies and antibody fragments as described herein for use in assessing the antigenic integrity of a viral antigen in a sample.
  • Biological medicinal products like vaccines differ from chemical drugs in that they cannot normally be characterized molecularly; antibodies are large molecules of significant complexity and have the capacity to vary widely from preparation to preparation.
  • the quality control processes may therefore begin with preparing a sample for an immunoassay that identifies binding of an antibody or fragment disclosed herein to a viral antigen.
  • immunoassays are disclosed elsewhere in this document, and any of these may be used to assess the structural/antigenic integrity of the antigen.
  • Standards for finding the sample to contain acceptable amounts of antigenically correct and intact antigen may be established by regulatory agencies. Another important embodiment where antigen integrity is assessed is in determining shelf- life and storage stability. Most medicines, including vaccines, can deteriorate over time. Therefore, it is critical to determine whether, over time, the degree to which an antigen, such as in a vaccine, degrades or destabilizes such that is it no longer antigenic and/or capable of generating an immune response when administered to a subject. Again, standards for finding the sample to contain acceptable amounts of antigenically intact antigen may be established by regulatory agencies. In certain embodiments, viral antigens may contain more than one protective epitope.
  • antibodies may prove useful to employ assays that look at the binding of more than one antibody, such as 2, 3, 4, 5 or even more antibodies.
  • These antibodies bind to closely related epitopes, such that they are adjacent or even overlap each other. On the other hand, they may represent distinct epitopes from disparate parts of the antigen. By examining the integrity of multiple epitopes, a more complete picture of the antigen's overall integrity, and hence ability to generate a protective immune response, may be determined.
  • Antibodies and fragments thereof as described in the present disclosure may also be used in a kit for monitoring the efficacy of vaccination procedures by detecting the presence of protective SARS-CoV-2 antibodies.
  • Antibodies, antibody fragment, or variants and derivatives thereof, as described in the present disclosure may also be used in a kit for monitoring vaccine manufacture with the desired immunogenicity.
  • Example 1 ELISA Binding Assay Binding of the antibodies to the following 7 RBD variants was determined using ELISA : - Wild-Type - UK Variant - B 1.1.7 lineage - Alpha variant - South African variant - B.1.351 lineage - Beta variant - Brazilian variant - B.1.1.248 lineage - Gamma variant - Californian variant - B.1.429 lineage - Epsilon variant - Indian variant - B.1.617 lineage - Kappa variant - Indian variant - B.1.617.2 lineage - Delta variant Binding of the antibodies to Omicron variant (B.1.1.529 lineage – Omicron variant) was similarly detemined Methodology 1.
  • Table 1D ELISA results of antibody B9 against RBD variants
  • Table 1E ELISA results of antibody D8 against RBD variants
  • Table 1F ELISA results of antibody A9 against RBD variants
  • Table 1G ELISA results of antibody A11 against RBD variants
  • Table 1H ELISA results of antibody B7 against RBD variants
  • Table 1J ELISA results against Omicron variant
  • Antibodies B7, A9 and A11 show a clear binding to RBD Omicron.
  • the affinities of these antibodies for this variant are however lower than what was observed against the other RBDs, with estimated KDs of 0.8 ⁇ g/mL, 6.25 ⁇ g/mL and 0.78 ⁇ g/mL for A9, A11 and B7, respectively.
  • Antibodies G4, B4, A5, D8, A9, A11 and B7 all bind very strongly to the remaining 7 RBD variants, as shown by the high OD values that are still obtained at an antibody concentration as low as 0.015 ⁇ g/mL.
  • the only exception is B7, which binds with a slightly lower affinity to the Indian kappa variant.
  • Antibody B9 binds strongly to the wild- type and UK variants, does not appear to bind to RBD omicron, and mildly to all the other RBD variants tested.
  • Example 2 Recombinant antibody neutralization assay test The aim of this study was to determine the neutralising properties of the antibodies when pre-incubated with different SARS-CoV2 isolates (Early isolate England 02/2020, Alpha, Beta, Gamma, Delta, and Omicron) for 1 hour before infection. Methodology The assay consists in treating a fixed concentration of virus with different concentrations of the antibody molecules of the invention and assessing the ability of such of inhibiting infection over the range of dilutions tested, once the mix is added to the assay cells.
  • An immunofluorescence-based assay where antiviral activity is measured as the ability of an antibody to reduce the percentage of infected cells, determined by immunostaining of a viral antigen.
  • a schematic diagram of the assay is shown in Figure 1.
  • the immunofluorescence assay it is possible to stop the assay after a single round of replication, infection is stopped before synthesis of new viral progeny and further viral spread, which allows to assess the neutralising effect of the antibodies on the starting amount of virus.
  • the cells are fixed and immuno-stained for a viral antigen, and antiviral activity is determined by quantifying the percentage of infected cells by comparison with infected, untreated cells.
  • SARS-CoV2 variants were incubated with a 2-fold serial dilution of each antibody for 1 hour, after which the mix was added to Vero cells. Antiviral activity was determined 6h later using an immunofluorescence-based assay. A positive and negative control antibody were included as controls.
  • Controls Uninfected untreated cells - Infected untreated cells - Positive control antibody: Anti-SARS-CoV2 spike protein (SinoBiologicals, 40591-MM43) - Negative control antibody: Mouse IgG1 Isotype control (Invitrogen, 02-6100) Test system - Cells: Vero E6, p3-p5, (VRS stock: ADp3) - Viruses: SARS-CoV2/England/02/2020, from BEI Resources – Early isolate SARS-CoV2 Isolate USA/CA_CDC_5574/2020 (B.1.1.7 - UK variant) - Alpha SARS-CoV2 isolate USA hCoV-19/South African/KRISP-EC-K 005321/2020 (Lineage B.1.351) – Beta SARS-CoV2 hCoV19/Japan/TY7-503/2021 P.1 variant, TY7-503 (Calu3) – Gamma SARS-CoV-2 VOC-202104
  • Virus and formulations were left on the cells for the entire duration of the experiment (6h) before fixation and immunostaining.
  • Cell plating Cells were detached and counted following SOP-RA 003 and SOP-RA 004 before seeding in complete media at 8,000cells/100 ⁇ l/well. After seeding, the plates were incubated at room temperature (RT) for 5 minutes for even distribution, and then at 37°C, 5% CO2 until the following day.
  • RT room temperature
  • any residual formaldehyde was quenched with 50 mM ammonium chloride, after which cells were permeabilised (0.1% Triton X100) and stained with an antibody recognising SARS-CoV2 nucleoprotein (Invitrogen, MA5-36271). The primary antibody was detected with an Alexa-488 conjugate secondary antibody (Life Technologies, A21244), and nuclei were stained with Hoechst. Images were acquired on an CellInsight CX5 high content platform (Thermo Scientific) using a 4X objective, and percentage infection calculated using CellInsight CX5 software (infected cells/total cells x 100).
  • EC50 values Normalised percentages of inhibition were calculated EC50 values were extrapolated from the curves representing the best fit (non-linear regression analysis, variable slope) of the logarithm of antibody dilutions vs. the normalised percentages of inhibition, using GraphPad Prism (version 9). Results Table 2A displays the EC50 values for each antibody sample. The graphs in Figure 2 display the percentage of inhibition of SARS-CoV2 infection at different antibody dilutions. Sample values in each plate are normalised to the plate internal controls, where 100% inhibition is derived from the average of the negative control (untreated uninfected) and 0% inhibition is derived from the average of the positive control (untreated infected).
  • the x axes show the dilution factor (log scale). The curves represent the best fit of the logarithm of antibody dilution vs. the normalised percentage of inhibition (variable slope).
  • A5 neutralises all variants except Omicron, with an EC50 lower than ⁇ 0.2 ⁇ g/ml.
  • A9 displays no inhibition against Omicron and modest neutralising activity against the other variants, with EC50 up to 18.7 ⁇ g/ml.
  • A11 neutralises all variants, with EC50 lower than ⁇ 0.1 ⁇ g/ml.
  • B4 neutralises all variants, with EC50 lower than ⁇ 0.05 ⁇ g/ml.
  • B7 neutralises all variants, with EC50 lower than ⁇ 3 ⁇ g/ml for Delta and lower than ⁇ 0.05 ⁇ g/ml for all other variants.
  • B9 only neutralises the early isolate England/02/2020 and the Alpha variants, with IC50 ⁇ 0.2 ⁇ g/ml.
  • D8 neutralises all variants, and displayed higher potency against England/02/2020, Alpha, and Delta (EC50 lower than ⁇ 0.02) than Beta, Gamma, and Omicron (EC50 between ⁇ 0.31 ⁇ g/ml and 1 ⁇ g/ml) G4 neutralises all variants except for Omicron, with EC50 lower than ⁇ 0.4 ⁇ g/ml.
  • the EC50 value is either extrapolated from the rest of the curve (shaded cells in Table 1) or could not be determined due to maximal inhibition (close to 100%) maintained at the. lowest concentration tested (0.08 ⁇ g/ml). (>0.08* in Table 2A).
  • the selected positive control antibody neutralised all variants with similar potency ( ⁇ 0.3 ⁇ g/ml), except for Omicron. No inhibition was observed when the viruses were pre-incubated with the negative control antibody.
  • Table 2A EC50 values for each tested antibody, expressed as the concentration ( ⁇ g/ml) at which 50% of infection is neutralised. > 0.08*: The EC50 values could not be determined or extrapolated, due to maximal inhibition (close to 100%) maintained at the.
  • antibodies A11, B4, B7, and D8 neutralised all variants including Omicron with potency in the ng/ml range.
  • A5, A9, and G4 inhibited all variants except Omicron.
  • A9 neutralised all variants except Omicron at higher concentrations only; B9 only inhibited the early isolate England/02/2020 and the Alpha variant.
  • Conclusion Under the conditions tested, four antibodies - A11, B4, B7, and D8 – neutralised all variants with potency in the ng/ml range.
  • the Omicron variant showed higher resistance to neutralisation but was inhibited with similar potency by these four antibodies.
  • A5, A9, and G4 inhibited all variants except Omicron.
  • A9 and B9 were the least potent antibodies, with A9 neutralising all variants except Omicron in the ⁇ g/ml range, and B9 only inhibiting the early isolate England/02/2020 and the Alpha variant.

Abstract

The present disclosure provides SARS-CoV-2 antibody molecules and their uses. More particularly, the presently disclosed disclosure provides human antibody molecules which bind to a novel coronavirus designated and methods of use thereof.

Description

HUMAN MONOCLONAL ANTIBODIES AND METHODS OF USE THEREOF The present invention provides SARS-CoV-2 antibody molecules and their uses. More particularly, the presently disclosed invention provides human antibody molecules which bind to a novel coronavirus designated and methods of use thereof. An epidemic of a novel coronavirus (SARS-CoV-2) affected mainland China, along with cases in more than 220 other countries and territories. It was identified in Wuhan, the capital of China's Hubei province, after 41 people developed pneumonia without a clear cause. The virus, which causes acute respiratory disease designated coronavirus disease 2019 (COVID-19), is capable of spreading from person to person. The incubation period (time from exposure to onset of symptoms) ranges from 0 to 24 days, with a mean of 3-5 days, but it may be contagious during this period and after recovery. Symptoms include fever, coughing and breathing difficulties. As of January 2022, over 350 million cases have been confirmed worldwide. A larger number of people may have been but not detected (especially mild cases). As of January 2022, over 5.6 million deaths have been attributed to the virus since the first confirmed death on 9 January 2020. The first local transmission outside China occurred in Vietnam between family members, while the first international transmission not involving family occurred in Germany on 22 January 2020. The first death outside China was in the Philippines, where a man from Wuhan died on 1 February 2020. As of 10 February 2020, the death toll from this virus had already surpassed the global SARS outbreak in 2003. The outbreak was understandably declared a Public Health Emergency of International Concern (PHEIC) by the World Health Organization (WHO) As of early 2022, despite the development of certain licensed vaccines, new variants of concern of the SARS-CoV-2 virus continue to emerge and cases and deaths continue to rise resulting in significant detriment to health and the global economy. There have been a number of emerging SARS-CoV-2 variants. Some SARS-CoV-2 variants contain an N439K mutation, which has enhanced binding affinity to the human ACE2 receptor (Thomson, E.C., et al., "The circulating SARS-CoV-2 Spike variant N439K maintains fitness while evading antibody-mediated immunity." bioRxiv, 2020). Some SARS-CoV-2 variants contain an N501Y mutation, which is associated with increased transmissibility, including the lineages B.1.1.7 (also known as WHO Alpha, 20l/501Y.Vl and VOC 202012/01) and B.1.351 (also known as WHO Beta, 20H/501Y.V2), which were discovered in the United Kingdom and South Africa, respectively (Tegally, H., et al., "Emergence and rapid spread of a new severe acute respiratory syndrome- related coronavirus 2 (SARS-CoV-2) lineage with multiple Spike mutations in South Africa." medRxiv, 2020: p.2020.12.21.20248640; Leung, K., et al., "Early empirical assessment of the N501Y mutant strains of SARS-CoV-2 in the United Kingdom, October to November 2020." medRxiv, 2020: p.2020.12.20.20248581). B.1.351 also include two other mutations in the RBD domain of SARS-CoV-2 Spike protein, K417N and E484K (Tegally, H., et al., "Emergence and rapid spread of a new severe acute respiratory syndrome- related coronavirus 2 (SARS-CoV-2) lineage with multiple Spike mutations in South Africa." medRxiv, 2020: p.2020.12.21.20248640).. A further variant of concern is the Gamma variant (P.1) which was identified in Brazil in May 2020 and has substitutions N501Y, E484K and K417T in the SARS-CoV-2 spike protein (Faria, Nuno R.; Claro, Ingra Morales (12 January 2021). "Genomic characterisation of an emergent SARS-CoV-2 lineage in Manaus: preliminary findings". Virological.).A further variant of concern is the Delta variant (B.1.617.2) which was identified in India in October 2020 and has mutations in the gene encoding the SARS-CoV-2 spike protein causing the substitutions T478K, P681R and L452R (Starr TN, Greaney AJ, Dingens AS, Bloom JD. Complete map of SARS-CoV-2 RBD mutations that escape the monoclonal antibody LY-CoV555 and its cocktail with LY-CoV016. Cell Rep Med.2021;2(4):100255. doi:10.1016/j.xcrm.2021.100255). More recently on 24 November 2021, the Omicron variant (B.1.1.529) was first reported to the World Health Organization (WHO) from South Africa. The Omicron variant has a total of 60 mutations as compared to the originally identified virus. Thirty-two mutations affect the spike protein, the main antigenic target of antibodies generated by infections and of many vaccines widely administered (Callaway E (December 2021). "Heavily mutated Omicron variant puts scientists on alert". Nature.600 (7887): 21. Bibcode:2021Natur.600). Many of those mutations had not been observed in other strains. Thus, there remains an urgent need to explore the biology and pathology of SARS-CoV-2 and well as the human immune response to this virus. As discussed above, SARS-CoV-2 is a major health concern with active cases increasing daily. Therefore, understanding the biology of this virus and the nature and extent of the human immune response to the virus is of paramount importance. The inventors have identified the sequences of human antibodies to SARS-CoV-2. Those sequences and uses for such antibodies are disclosed herein. The present invention addresses these and other related needs by providing, inter alia, antibody molecules that are capable of binding to SARS-CoV-2. The present invention provides a monoclonal antibody molecule, wherein the antibody or antibody fragment is characterized by clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively. The antibody or antibody fragment may be encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having 100% identity to clone-paired sequences as set forth in Table 1. The antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, or light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2. The antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment. The antibody may be a chimeric antibody, is bispecific antibody, or is an intrabody. The antibody may be an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern. The antibody or antibody fragment may bind to a SARS-CoV-2 surface spike protein. A hybridoma or engineered cell encoding an antibody or antibody fragment wherein the antibody or antibody fragment is characterized by clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively. The antibody or antibody fragment may be encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having 100% identity to clone-paired sequences as set forth in Table 1. The antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, or light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2. The antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment. The antibody may be a chimeric antibody, a bispecific antibody, or an intrabody. The antibody may bean IgG, or a recombinant IgG antibody or antibody fragment comprising an Fe portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern. The antibody or antibody fragment maybind to a SARS-CoV-2 surface spike protein. Typically, the antibody molecule is a purified or isolated antibody molecule. The antibody molecule may be a polyclonal antibody, a monoclonal antibody, a human antibody, a humanized antibody, a chimeric antibody, fragment of an antibody or monoclonal antibody, in particular a Fab, Fab', or F(ab')2 fragment, a single chain antibody, in particular a single chain variable fragment (scFv), a domain antibody, a nanobody, a diabody, or a DARPin. More specifically, the antibody molecule may be a humanized antibody or a fragment of a humanised antibody, in particular a Fab, Fab', or F(ab')2 fragment, a single chain antibody, in particular a single chain variable fragment (scFv), a Small Modular lmmunopharmaceutical (SMIP), a domain antibody, a nanobody, a diabody, or a Designed Ankyrin Repeat Protein (DARPin). In one aspect, the present invention provides a pharmaceutical composition comprising an antibody molecule of the present invention and a pharmaceutically acceptable carrier. To be used in therapy, the antibody molecule is included into pharmaceutical compositions appropriate to facilitate administration to animals or humans. Suitable formulations of the antibody molecule may be prepared by mixing the antibody molecule with physiologically acceptable carriers, excipients or stabilizers, in the form of lyophilized or otherwise dried formulations or aqueous solutions or aqueous or non-aqueous suspensions. Carriers, excipients, modifiers or stabilizers are nontoxic at the dosages and concentrations employed. They include buffer systems such as phosphate, citrate, acetate and other inorganic or organic acids and their salts; antioxidants including ascorbic acid and methionine; preservatives such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone or polyethylene glycol (PEG); amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, oligosaccharides or polysaccharides and other carbohydrates including glucose, man nose, sucrose, trehalose, dextrins or dextrans; chelating agents such as EDTA; sugar alcohols such as, mannitol or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or ionic or non-ionic surfactants such as TWEEN™ (polysorbates), PLURONICS ™ or fatty acid esters, fatty acid ethers or sugar esters. Also organic solvents may be contained in the antibody formulation such as ethanol or isopropanol. The excipients may also have a release-modifying or absorption-modifying function. In one aspect, the pharmaceutical composition comprises the antibody molecule of the present invention in an aqueous, buffered solution, or a lyophilisate made from such a solution. A suitable mode of application is parenteral, by infusion or injection (intraveneous, intramuscular, subcutaneous, intraperitoneal, intradermal), but other modes of application such as by inhalation, transdermal, intranasal, buccal, oral, may also be applicable. In a further aspect, the present invention provides an antibody molecule of the present invention for use as a medicament. In one aspect, the present invention provides a a method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, comprising delivering to the subject an antibody or antibody fragment having clone- paired heavy and light chain CDR sequences from Tables 3 and 4, respectively. The antibody or antibody fragment may be encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having 100% identity to clone-paired sequences as set forth in Table 1. The antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, or light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2. The antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab'h fragment, or Fv fragment. The antibody may be a chimeric antibody or a bispecific antibody. The antibody may be an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern. The antibody or antibody fragment may bind to a SARS-CoV-2 antigen such as a surface spike protein. The antibody or antibody fragment may be administered prior to infection or after infection. The subject may be of age 60 or older, may be immunocompromised, or may suffer from a respiratory and/or cardiovascular disorder. Delivering may comprise antibody or antibody fragment administration, or genetic delivery with an RNA or DNA sequence or vector encoding the antibody or antibody fragment. In a further aspect, the present invention provides an antibody molecule of the invention for use in treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2. In a further aspect, the present invention provides the use of an antibody molecule of the invention in the manufacture of a medicament for treating a subject infected with SARS- CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV- 2. The antibody molecule of the invention may be administered either simultaneously with, or before or after, one or more other therapeutic agent. The antibody molecule of the invention may be administered separately, by the same or different route of administration, or together in the same pharmaceutical composition as the other agents. In one embodiment, the invention provides a product comprising an antibody molecule of the invention and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy. In one embodiment, the therapy is treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2. Products provided as a combined preparation include a composition comprising the antibody molecule of the invention and the other therapeutic agent(s) together in the same pharmaceutical composition, or the antibody molecule of the invention and the other therapeutic agent(s) in separate form, e.g. in the form of a kit. In one embodiment, the invention provides a pharmaceutical composition comprising an antibody molecule of the invention and another therapeutic agent(s). Optionally, the pharmaceutical composition may comprise a pharmaceutically acceptable excipient, as described above. In one embodiment, the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains an antibody molecule of the invention. In one embodiment, the kit comprises means for separately retaining the compositions, such as a container, divided bottle, or divided foil packet. An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like. The kit of the invention may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another. To assist compliance, the kit of the invention typically comprises directions for administration. In the combination therapies of the invention, the antibody molecule of the invention and the other therapeutic agent may be manufactured and/or formulated by the same or different manufacturers. Moreover, the antibody molecule of the invention and the other therapeutic may be brought together into a combination therapy: (i) prior to release of the combination product to physicians (e.g. in the case of a kit comprising the antibody molecule of the invention and the other therapeutic agent); (ii) by the physician themselves (or under the guidance of the physician) shortly before administration; (iii) in the patient themselves, e.g. during sequential administration of the antibody molecule of the invention and the other therapeutic agent. Accordingly, the invention provides the use of an antibody molecule of the invention in the manufacture of a medicament for treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the medicament is prepared for administration with another therapeutic agent. The invention also provides the use of another therapeutic agent for treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the medicament is administered with an antibody molecule of the invention. The invention also provides an antibody molecule of the invention for use in a method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the antibody molecule of the invention is prepared for administration with another therapeutic agent. The invention also provides another therapeutic agent for use in a method of treating a subject infected with SARS- CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV- 2, wherein the other therapeutic agent is prepared for administration with an antibody molecule of the invention. The invention also provides an antibody molecule of the invention for use in a method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2 wherein the antibody molecule of the invention is administered with another therapeutic agent. The invention also provides another therapeutic agent for use in a method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the other therapeutic agent is administered with an antibody molecule of the invention. The invention also provides the use of an antibody molecule of the invention for treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the subject has previously (e.g. within 24 hours) been treated with another therapeutic agent. The invention also provides the use of another therapeutic agent for treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, wherein the subject has previously (e.g. within 24 hours) been treated with an antibody molecule of the invention. In one embodiment, the other therapeutic agents) include a further antibody molecule of the invention. In a further embodiment, the two or more antibody molecules of the present invention bind to a SARS-CoV-2 surface spike protein. In an alternative further embodiment, one of the two or more antibody molecules of the invention binds to a SARS- CoV-2 surface spike protein and another of the two or more antibody molecules of the invention binds to a SARS-CoV-2 surface nucleocapsid (N) protein. Accordingly, the invention includes as a further aspect a combination of two or more antibody molecules of the invention. In one embodiment, the other therapeutic agents) include a further antibody molecule. In a further embodiment, the antibody molecule of the present invention and the further antibody molecule bind to a SARS-CoV-2 surface spike protein. In an alternative further embodiment, the antibody molecule of the invention binds to a SARS-CoV-2 surface spike protein and further antibody molecule binds to a SARS-CoV-2 surface nucleocapsid (N) protein. In one embodiment, the further antibody molecule is a monoclonal antibody optionally selected from bamlanivimab; bebtelovimab, casirivimab; cilgavimab; etesevimab; imdevimab; sotrovimab; tixagevimab or combinations thereof such as bamlanivimab/ etesevimab, casirivimab/ imdevimab, or tixagevimab/ cilgavimab. Accordingly, the invention includes as a further aspect a combination of an antibody molecule of the invention with one or more further antibody molecules. In one embodiment, the other therapeutic agent(s) include an antibody product wherein the antibody product is optionally selected from plasma provided by donors who have recovered from SARS-CoV-2, and SARS-CoV-2 immunoglobulins derived therefrom. In one embodiment, the other therapeutic agent(s) include an anti-viral agent optionally selected from the group consisting of remdesivir, molnupiravir (Lagevrio), Ritonavir- Boosted Nirmatrelvir (Paxlovid).. Accordingly, the invention includes as a further aspect a combination of an antibody of the invention with an anti-viral agent optionally selected from the group consisting of remdisivir, molnupiravir (Lagevrio), Ritonavir-Boosted Nirmatrelvir (Paxlovid).. In one embodiment, the other therapeutic agent(s) include a cell based therapy such as mesenchymal stem cells. Accordingly, the invention includes as a further aspect a combination of an antibody of the invention with a cell based therapy such as mesenchymal stem cells. In one embodiment, the other therapeutic agent(s) include an immunomodulator which is optionally selected from the group consisting of corticosteroids, Interleukin-1 inhibitors, Interleukin-6 inhibitors: kinase inhibitors. Granulocyte-Macrophage Colony-Stimulating Factor Inhibitors, colchicine and fluvoxamine. In a further embodiment the corticosteroid is an oral systemic corticosteroid such as dexamethasone or an inhaled corticosteroid such as budesonide or ciclesonide. In a further embodiment, the Interleukin-1 inhibitor is optionally selected from the group consisting of anakinra and canakinumab. In a further embodiment, the Interleukin-6 inhibitor is optionally selected from the group consisting of sarilumab, tocilizumab and siltuximab. In a further embodiment, the kinase inhibitor is a Janus Kinase inhibitor optionally selected from the group consisting of baricitinib, tofacitinib and ruxolitinib: Accordingly, the invention includes as a further aspect a combination of an antibody of the invention with an immunomodulator which is optionally selected from the group consisting of corticosteroids, Interleukin-1 inhibitors, Interleukin-6 inhibitors: kinase inhibitors. Granulocyte-Macrophage Colony-Stimulating Factor Inhibitors, colchicine and fluvoxamine. In one embodiment, the other therapeutic agent(s) include an antithrombotic In a further embodiment the antithrombotic is an anticoagulant, an antiplatelet or a thrombolytic. Accordingly, the invention includes as a further aspect a combination of an antibody of the invention with an antithrombotic. In still yet another aspect, there is provided a vaccine formulation comprising one or more antibodies or antibody fragments characterized by clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively. The at least one of the antibodies or antibody fragments may be encoded by light and heavy chain variable sequences according to clone- paired sequences from Table 1, by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired sequences from Table 1, or by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 1. The at least one of the antibodies or antibody fragments may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2 or may comprise light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone- paired sequences from Table 2. The at least one of the antibody fragments is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment. The at least one of the antibodies may a chimeric antibody, a bispecific antibody or an intrabody. The antibody may be an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern. The antibody or antibody fragment may bind to a SARS-CoV-2 antigen surface spike protein. In a further embodiment, there is provided a vaccine formulation comprising one or more expression vectors encoding a first antibody or antibody fragment as described herein. The expression vector(s) may be Sindbis virus or VEE vector(s). The vaccine may be formulated for delivery by needle injection, jet injection, or electroporation. The vaccine formulation may further comprise one or more expression vectors encoding for a second antibody or antibody fragment, such as a distinct antibody or antibody fragment of claims 26- to34. In yet a further embodiment, there is provided a method of protecting the health of a subject of age 60 or older, an immunocompromised, subject or a subject suffering from a respiratory and/or cardiovascular disorder that is infected with or at risk of infection with SARS-CoV-2 comprising delivering to the subject an antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively. The antibody or antibody fragment may be encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having 100% identity to clone- paired sequences as set forth in Table 1. The antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, or light and heavy variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2. The antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab'h fragment, or Fv fragment. The antibody may an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern. The antibody may be a chimeric antibody or a bispecific antibody. The antibody or antibody fragment may be administered prior to infection or after infection. The antibody or antibody fragment may bind to a SARS-CoV-2 antigen such as a surface spike protein. Delivering may comprise antibody or antibody fragment administration, or genetic delivery with an RNA or DNA sequence or vector encoding the antibody or antibody fragment. The antibody or antibody fragment may improve the subject's respiration as compared to an untreated control and/or may reduce viral load as compared to an untreated control. In still yet a further embodiment, there is provided a method of determining the antigenic integrity, correct conformation and/or correct sequence of a SARS-CoV-2 surface spike protein comprising (a) contacting a sample comprising the antigen with a first antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively; and (b) determining antigenic integrity, correct conformation and/or correct sequence of the antigen by detectable binding of the first antibody or antibody fragment to the antigen. The sample may comprise recombinantly produced antigen or a vaccine formulation or vaccine production batch. Detection may comprise ELISA, RIA, western blot, a biosensor using surface plasmon resonance or biolayer interferometry, or flow cytometric staining. The first antibody or antibody fragment may be encoded by clone- paired variable sequences as set forth in Table 1, by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences as set forth in Table 1. The first antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, may comprise light and heavy chain variable sequences having at least 70%, 80% or 90% identity to clone-paired sequences from Table 2, or may comprise light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 2. The first antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment. The method may further comprise performing steps (a) and (b) a second time to determine the antigenic stability of the antigen over time. The method may further comprise (c) contacting a sample comprising the antigen with a second antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively; and (d) determining antigenic integrity of the antigen by detectable binding of the second antibody or antibody fragment to the antigen. The second antibody or antibody fragment may be encoded by clone-paired variable sequences as set forth in Table 1, by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired variable sequences as set forth in Table 1, or by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences as set forth in Table 1. The second antibody or antibody fragment may comprise light and heavy chain variable sequences according to clone-paired sequences from Table 2, may comprise light and heavy chain variable sequences having at least 70%, 80% or 90% identity to clone-paired sequences from Table 2, or may comprise light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 2. The second antibody fragment may be a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment. The method may further comprise performing steps (c) and (d) a second time to determine the antigenic stability of the antigen over time. Also provided is human monoclonal antibody or antibody fragment, or hybridoma or engineered cell producing the same, wherein the antibody binds to a SARS-CoV-2 antigen surface spike protein. In a further aspect, the present invention provides a kit comprising an antibody molecule of the present invention, or a pharmaceutical composition thereof. In one aspect, the present invention provides a method of manufacturing an antibody molecule of the present invention, comprising: (a) providing a host cell comprising one or more nucleic acids encoding the antibody molecule in functional association with an expression control sequence, (b) cultivating the host cell, and (c) recovering the antibody molecule from the cell culture. Advantages of the presently disclosed subject matter will become evident to those of ordinary skill in the art after a study of the description, Figures, and non-limiting Examples in this document. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1: Principle of the Neutralization assay (Example 2) Figure 2: Percentages of inhibition: regression curves (Example 2) DETAILED DESCRIPTION OF THE INVENTION: SARS-CoV-2 is a contagious virus that causes the acute respiratory disease designated coronavirus disease 2019 (COVID-19), a respiratory infection. It is the cause of the ongoing 2019-20 coronavirus outbreak, a global health emergency. Genomic sequencing has shown that it is a positive-sense, single-stranded RNA coronavirus. During the ongoing outbreak, the virus has often been referred to in common parlance as "the coronavirus", "the new coronavirus" and "the Wuhan coronavirus", while the WHO recommends the designation "SARS-CoV-2". The International Committee on Taxonomy of Viruses (ICTV) announced that the official name for the virus is SARS-CoV-2. Many early cases were linked to a large seafood and animal market in the Chinese city of Wuhan, and the virus is thought to have a zoonotic origin. Comparisons of the genetic sequences of this virus and other virus samples have shown similarities to SARS-Co V (79.5%) and bat coronaviruses (96%). This finding makes an ultimate origin in bats likely, although an intermediate host, such as a pangolin, cannot be ruled out. The virus could be a recombinant virus formed from two or more coronaviruses. Human-to-human transmission of the virus has been confirmed. Coronaviruses are primarily spread through close contact, in particular through respiratory droplets from coughs and sneezes within a range of about 6 feet (1.8 m). Viral RNA has also been found in stool samples from infected patients. It is possible that the virus can be infectious even during the incubation period. Animals sold for food were originally suspected to be the reservoir or intermediary hosts of SARS-CoV-2 because many of the first individuals found to be infected by the virus were workers at the Huanan Seafood Market. A market selling live animals for food was also blamed in the SARS outbreak in 2003; such markets are considered to be incubators for novel pathogens. The outbreak has prompted a temporary ban on the trade and consumption of wild animals in China. However, some researchers have suggested that the Huanan Seafood Market may not be the original source of viral transmission to humans. With a sufficient number of sequenced genomes, it is possible to reconstruct a phylogenetic tree of the mutation history of a family of viruses. Research into the origin of the 2003 SARS outbreak has resulted in the discovery of many SARS-like bat coronaviruses, most originating in the Rhinolophus genus of horseshoe bats. SARS-CoV-2 falls into this category of SARS-related coronaviruses. Two genome sequences from Rhinolophus sinicus published in 2015 and 2017 show a resemblance of 80% to SARS-CoV-2. A third virus genome from Rhinolophus affinis, "RaTG13" collected in Yunnan province, has a 96% resemblance to SARS-CoV-2. For comparison, this amount of variation among viruses is similar to the amount of mutation observed over ten years in the H3N2 human influenza virus strain. SARS-CoV-2 belongs to the broad family of viruses known as coronaviruses; "nCoV" is the standard term used to refer to novel coronaviruses until the choice of a more specific designation. It is a positive-sense single-stranded RNA (+ssRNA) virus. Other coronaviruses are capable of causing illnesses ranging from the common cold to more severe diseases such as Middle East respiratory syndrome (MERS) and Severe acute respiratory syndrome (SARS). It is the seventh known coronavirus to infect people, after 229E, NL63, OC43, HKUl, MERS- CoV, and SARS-CoV. Like SARS-CoV, SARS-CoV-2 is a member of the subgenus Sarbecovirus (Beta-CoV lineage B). Its RNA sequence is approximately 30,000 bases in length. By 12 January 2020, five genomes of SARS-CoV-2 had been isolated from Wuhan and reported by the Chinese Center for Disease Control and Prevention (CCDC) and other institutions; the number of genomes increased to 28 by 26 January. Except for the earliest GenBank genome, the genomes are under an embargo at GISAID. A phylogenic analysis for the samples is available through Nextstrain. Publication of the SARS-CoV-2 genome led to several protein modeling experiments on the receptor binding protein (RBD) of the spike (S) protein of the virus. Results suggest that the S protein retains sufficient affinity to the Angiotensin converting enzyme 2 (ACE2) receptor to use it as a mechanism of cell entry. On 22 January, a group in China working with the full virus and a group in the U.S. working with reverse genetics independently and experimentally demonstrated human ACE2 as the receptor for SARS-CoV-2. To look for potential protease inhibitors, the viral 3C-like protease M(pro) from the ORFla polyprotein has also been modelled for drug docking experiments. Innophore has produced two computational models based on SARS protease, and the Chinese Academy of Sciences has produced an unpublished experimental structure of a recombinant SARS- CoV-2 protease. In addition, researchers at the University of Michigan have modelled the structures of all mature peptides in the SARS-CoV-2 genome using I-TASSER. The first known human infection occurred in early December 2019. An outbreak of SARS- CoV-2 was first detected in Wuhan, China, in mid-December 2019, likely originating from a single infected animal. The virus subsequently spread to all provinces of China and to more than two dozen other countries in Asia, Europe, North America, and Oceania. Human- to- human spread of the virus has been confirmed in all of these regions. On 30 January 2020, SARS-CoV-2 was designated a global health emergency by the WHO. As of January 2022, over 350 million cases have been confirmed worldwide. A larger number of people may have been but not detected (especially mild cases). Initially, nearly all cases outside China occurred in people who either travelled from Wuhan,or were in direct contact with someone who travelled from the area. Later, spread from travellers to other countries resulted in transmission to over 220 countries in the world. While the proportion of infections that result in confirmed infection or progress to diagnosable SARS-CoV-2 acute respiratory disease remains unclear, the total number of deaths attributed to the virus was over 5.6 million as of January 2022. COVID-19 acute respiratory disease is a viral respiratory disease caused by SARS- CoV-2. It was first detected during the 2019-20 Wuhan coronavirus outbreak. Symptoms may include fever, dry cough, and shortness of breath. Those infected may either be asymptomatic or have mild to severe symptoms, like fever, cough, shortness of breath. diarrhoea or upper respiratory symptoms (e.g., sneezing, runny nose, sore throat) are less frequent. Cases of severe infection can progress to severe pneumonia, multi-organ failure, and death. The time from exposure to onset of symptoms is estimated at 2 to 10 days by the World Health Organization, and 2 to 14 days by the US Centers for Disease Control and Prevention (CDC). Of those who have died, many had underlying health conditions, exhibiting conditions such as hypertension, diabetes, cardiovascular disease or other diseases or conditions that impaired their immune systems Global health organizations have published preventive measures individuals can take to reduce the chances of SARS-CoV-2 infection. Recommendations are similar to those previously published for other coronaviruses and include: frequent washing of hands with soap and water; not touching the eyes, nose, or mouth with unwashed hands; and practicing good respiratory hygiene. The WHO has published several testing protocols for SARS-CoV-2. The most accurate testing of current infection uses real time reverse transcription-polymerase chain reaction (rRT-PCR). The test can be done on respiratory or blood samples and results are generally available within a few hours to days. An "isolated antibody" is one that has been separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes. In particular embodiments, the antibody is purified: (1) to greater than 95% by weight of antibody as determined by the Lowry method, and most particularly more than 99% by weight; (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator; or (3) to homogeneity by SDS- PAGE under reducing or non-reducing conditions using Coomassie blue or silver stain. Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step. The basic four-chain antibody unit is a heterotetrameric glycoprotein composed of two identical light (L) chains and two identical heavy (H) chains. An IgM antibody consists of 5 basic heterotetramer units along with an additional polypeptide called J chain, and therefore contain 10 antigen binding sites, while secreted IgA antibodies can polymerize to form polyvalent assemblages comprising 2-5 of the basic 4-chain units along with J chain. In the case of IgGs, the 4-chain unit is generally about 150,000 daltons. Each L chain is linked to an H chain by one covalent disulfide bond, while the two H chains are linked to each other by one or more disulfide bonds depending on the H chain isotype. Each H and L chain also has regularly spaced intrachain disulfide bridges. Each H chain has at the N-terminus, a variable region (VH) followed by three constant domains (CH) for each of the alpha and gamma chains and four CH domains for mu and isotypes. Each L chain has at the N- terminus, a variable region (VL) followed by a constant domain (CL) at its other end. The VL is aligned with the VH and the CL is aligned with the first constant domain of the heavy chain (CH1). Particular amino acid residues are believed to form an interface between the light chain and heavy chain variable regions. The pairing of a VH and VL together forms a single antigen-binding site. For the structure and properties of the different classes of antibodies, see, e.g., Basic and Clinical Immunology, 8th edition, Daniel P. Stites, Abba I. Terr and Tristram G. Parslow (eds.), Appleton & Lange, Norwalk, Conn., 1994, page 71, and Chapter 6. The L chain from any vertebrate species can be assigned to one of two clearly distinct types, called kappa and lambda based on the amino acid sequences of their constant domains (CL). Depending on the amino acid sequence of the constant domain of their heavy chains (CH), immunoglobulins can be assigned to different classes or isotypes. There are five classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, having heavy chains designated alpha, delta, epsilon, gamma and mu, respectively. They gamma and alpha classes are further divided into subclasses on the basis of relatively minor differences in CH sequence and function, humans express the following subclasses: IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2. The term "variable" refers to the fact that certain segments of the V domains differ extensively in sequence among antibodies. The V domain mediates antigen binding and defines specificity of a particular antibody for its particular antigen. However, the variability is not evenly distributed across the 110-amino acid span of the variable regions. Instead, the V regions consist of relatively invariant stretches called framework regions (FRs) of 15- 30 amino acids separated by shorter regions of extreme variability called "hypervariable regions" that are each 9-12 amino acids long. The variable regions of native heavy and light chains each comprise four FRs, largely adopting a beta-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some cases forming part of, the beta-sheet structure. The hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)). The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody dependent cellular cytotoxicity (ADCC), antibody- dependent cellular phagocytosis (ADCP), antibody-dependent neutrophil phagocytosis (ADNP), and antibody-dependent complement deposition (ADCD). The term "hypervariable region" when used herein refers to the amino acid residues of an antibody that are responsible for antigen binding. The hypervariable region generally comprises amino acid residues from a "complementarity determining region" or "CDR" (e.g., around about residues 24-34 (Ll), 50-56 (L2) and 89-97 (L3) in the VL, and around about 31-35 (Hl), 50-65 (H2) and 95-102 (H3) in the VH when numbered in accordance with the Kabat numbering system; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)); and/or those residues from a "hypervariable loop" (e.g., residues 24-34 (Ll), 50-56 (L2) and 89-97 (L3) in the VL, and 26-32 (Hl), 52-56 (H2) and 95-101 (H3) in the VH when numbered in accordance with the Chothia numbering system; Chothia and Lesk, J. Mol. Biol.196:901- 917 (1987)); and/or those residues from a "hypervariable loop"/CDR (e.g., residues 27-38 (Ll), 56-65 (L2) and 105-120 (L3) in the VL, and 27-38 (Hl), 56-65 (H2) and 105-120 (H3) in the VH when numbered in accordance with the IMGT numbering system; Lefranc, M. P. et al. Nucl. Acids Res.27:209-212 (1999), Ruiz, M. et al. Nucl. Acids Res.28:219-221 (2000)). Optionally the antibody has symmetrical insertions at one or more of the following points 28, in the VsubH when numbered in accordance with AHo; Honneger, A. and Plunkthun, A. J. Mol. Biol.309:657-670 (2001)). By "germline nucleic acid residue" is meant the nucleic acid residue that naturally occurs in a germline gene encoding a constant or variable region. "Germline gene" is the DNA found in a germ cell (i.e., a cell destined to become an egg or in the sperm). A "germline mutation" refers to a heritable change in a particular DNA that has occurred in a germ cell or the zygote at the single-cell stage, and when transmitted to offspring, such a mutation is incorporated in every cell of the body. A germline mutation is in contrast to a somatic mutation which is acquired in a single body cell. In some cases, nucleotides in a germline DNA sequence encoding for a variable region are mutated (i.e., a somatic mutation) and replaced with a different nucleotide. The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations that include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies. The modifier "monoclonal" is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies useful in the present disclosure may be prepared by the hybridoma methodology first described by Kohler et al., Nature, 256:495 (1975), or may be made using recombinant DNA methods in bacterial, eukaryotic animal or plant cells (see, e.g., U.S. Patent 4,816,567) after single cell sorting of an antigen specific B cell, an antigen specific plasmablast responding to an infection or immunization, or capture of linked heavy and light chains from single cells in a bulk sorted antigen specific collection. The "monoclonal antibodies" may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991), for example. It will be understood that monoclonal antibodies binding to SARS-CoV-2 will have several applications. These include the production of diagnostic kits for use in detecting and diagnosing SARS-CoV-2 infection, as well as for treating the same. In these contexts, one may link such antibodies to diagnostic or therapeutic agents, use them as capture agents or competitors in competitive assays, or use them individually without additional agents being attached thereto. The antibodies may be mutated or modified, as discussed further below. Methods for preparing and characterizing antibodies are well known in the art (see, e.g.,Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988; U.S. Patent 4,196,265). The methods for generating monoclonal antibodies (MAbs) generally begin along the same lines as those for preparing polyclonal antibodies. The first step for both these methods is immunization of an appropriate host or identification of subjects who are immune due to prior natural infection or vaccination with a licensed or experimental vaccine. As is well known in the art, a given composition for immunization may vary in its immunogenicity. It is often necessary therefore to boost the host immune system, as may be achieved by coupling a peptide or polypeptide immunogen to a carrier. Exemplary and preferred carriers are keyhole limpet hemocyanin (KLH) and bovine serum albumin (BSA). Other albumins such as ovalbumin, mouse serum albumin or rabbit serum albumin can also be used as carriers. Means for conjugating a polypeptide to a carrier protein are well known in the art and include glutaraldehyde, m-maleimidobenzoyl-N-hydroxysuccinimide ester, carbodiimide and bis-diazotized benzidine. As also is well known in the art, the immunogenicity of a particular immunogen composition can be enhanced by the use of non- specific stimulators of the immune response, known as adjuvants. Exemplary and preferred adjuvants in animals include complete Freund's adjuvant (a non-specific stimulator of the immune response containing killed Mycobacterium tuberculosis), incomplete Freund's adjuvants and aluminium hydroxide adjuvant and in humans include alum, CpG, MFP59 and combinations of immunostimulatory molecules ("Adjuvant Systems", such as AS0l or AS03). Additional experimental forms of inoculation to induce SARS-CoV-2-specific B cells is possible, including nanoparticle vaccines, or gene-encoded antigens delivered as DNA or RNA genes in a physical delivery system (such as lipid nanoparticle or on a gold biolistic bead), and delivered with needle, gene gun, transcutaneous electroporation device. The antigen gene also can be carried as encoded by a replication competent or defective viral vector such as adenovirus, adeno-associated virus, poxvirus, herpesvirus, or alphavirus replicon, or alternatively a virus like particle. In the case of human antibodies against natural pathogens, a suitable approach is to identify subjects that have been exposed to the pathogens, such as those who have been diagnosed as having contracted the disease, or those who have been vaccinated to generate protective immunity against the pathogen or to test the safety or efficacy of an experimental vaccine. Circulating anti-pathogen antibodies can be detected, and antibody encoding or producing B cells from the antibody-positive subject may then be obtained. The amount of immunogen composition used in the production of polyclonal antibodies varies upon the nature of the immunogen as well as the animal used for immunization. A variety of routes can be used to administer the immunogen (subcutaneous, intramuscular, intradermal, intravenous and intraperitoneal). The production of polyclonal antibodies may be monitored by sampling blood of the immunized animal at various points following immunization. A second, booster injection, also may be given. The process of boosting and titering is repeated until a suitable titer is achieved. When a desired level of immunogenicity is obtained, the immunized animal can be bled and the serum isolated and stored, and/or the animal can be used to generate MAbs. Following immunization, somatic cells with the potential for producing antibodies, specifically B lymphocytes (B cells), are selected for use in the MAb generating protocol. These cells may be obtained from biopsied spleens, lymph nodes, tonsils or adenoids, bone marrow aspirates or biopsies, tissue biopsies from mucosal organs like lung or GI tract, or from circulating blood. The antibody-producing B lymphocytes from the immunized animal or immune human are then fused with cells of an immortal myeloma cell, generally one of the same species as the animal that was immunized or human or human/mouse chimeric cells. Myeloma cell lines suited for use in hybridoma-producing fusion procedures preferably are non-antibody-producing, have high fusion efficiency, and enzyme deficiencies that render then incapable of growing in certain selective media which support the growth of only the desired fused cells (hybridomas). Any one of a number of myeloma cells may be used, as are known to those of skill in the art (Goding, pp.65-66, 1986; Campbell, pp.75-83, 1984). HMMA2.5 cells or MFP-2 cells are particularly useful examples of such cells. Methods for generating hybrids of antibody-producing spleen or lymph node cells and myeloma cells usually comprise mixing somatic cells with myeloma cells in a 2:1 proportion, though the proportion may vary from about 20:1 to about 1:1, respectively, in the presence of an agent or agents (chemical or electrical) that promote the fusion of cell membranes. In some cases, transformation of human B cells with Epstein Barr virus (EBV) as an initial step increases the size of the B cells, enhancing fusion with the relatively large-sized myeloma cells. Transformation efficiency by EBV is enhanced by using CpG and a Chk2 inhibitor drug in the transforming medium. Alternatively, human B cells can be activated by co-culture with transfected cell lines expressing CD40 Ligand (CD154) in medium containing additional soluble factors, such as IL-21 and human B cell Activating Factor (BAFF), a Type II member of the TNF superfamily. Fusion methods using Sendai virus have been described by Kohler and Milstein (1975; 1976), and those using polyethylene glycol (PEG), such as 37% (v/v) PEG, by Gefter et al. (1977). The use of electrically induced fusion methods also is appropriate (Goding, pp.71-74, 1986) and there are processes for better efficiency (Yu et al., 2008). Fusion procedures usually produce viable hybrids at low frequencies, about 1 x 10-6 to 1 x 10-8, but with optimized procedures one can achieve fusion efficiencies close to 1 in 200 (Yu et al., 2008). However, relatively low efficiency of fusion does not pose a problem, as the viable, fused hybrids are differentiated from the parental, infused cells (particularly the infused myeloma cells that would normally continue to divide indefinitely) by culturing in a selective medium. The selective medium is generally one that contains an agent that blocks the de novo synthesis of nucleotides in the tissue culture medium. Exemplary and preferred agents are aminopterin, methotrexate, and azaserine. Aminopterin and methotrexate block de novo synthesis of both purines and pyrimidines, whereas azaserine blocks only purine synthesis. Where aminopterin or methotrexate is used, the medium is supplemented with hypoxanthine and thymidine as a source of nucleotides (HAT medium). Where azaserine is used, the medium is supplemented with hypoxanthine. Ouabain is added if the B cell source is an EBY- transformed human B cell line, in order to eliminate EBY-transformed lines that have not fused to the myeloma. The preferred selection medium is HAT or HAT with ouabain. Only cells capable of operating nucleotide salvage pathways are able to survive in HAT medium. The myeloma cells are defective in key enzymes of the salvage pathway, e.g., hypoxanthine phosphoribosyl transferase (HPRT), and they cannot survive. The B cells can operate this pathway, but they have a limited life span in culture and generally die within about two weeks. Therefore, the only cells that can survive in the selective media are those hybrids formed from myeloma and B cells. When the source of B cells used for fusion is a line of EBY-transformed B cells, as here, ouabain may also be used for drug selection of hybrids as EBY-transformed B cells are susceptible to drug killing, whereas the myeloma partner used is chosen to be ouabain resistant. Culturing provides a population of hybridomas from which specific hybridomas are selected. Typically, selection of hybridomas is performed by culturing the cells by single-clone dilution in microtiter plates, followed by testing the individual clonal supernatants (after about two to three weeks) for the desired reactivity. The assay should be sensitive, simple and rapid, such as radioimmunoassays, enzyme immunoassays, cytotoxicity assays, plaque assays dot immunobinding assays, and the like. The selected hybridomas are then serially diluted or single-cell sorted by flow cytometric sorting and cloned into individual antibody-producing cell lines, which clones can then be propagated indefinitely to provide mAbs. The cell lines may be exploited for MAb production in two basic ways. A sample of the hybridoma can be injected (often into the peritoneal cavity) into an animal (e.g., a mouse). Optionally, the animals are primed with a hydrocarbon, especially oils such as pristane (tetramethylpentadecane) prior to injection. When human hybridomas are used in this way, it is optimal to inject immunocompromised mice, such as SCID mice, to prevent tumour rejection. The injected animal develops tumours secreting the specific monoclonal antibody produced by the fused cell hybrid. The body fluids of the animal, such as serum or ascites fluid, can then be tapped to provide MAbs in high concentration. The individual cell lines could also be cultured in vitro,where the MAbs are naturally secreted into the culture medium from which they can be readily obtained in high concentrations. Alternatively, human hybridoma cells lines can be used in vitro to produce immunoglobulins in cell supernatant. The cell lines can be adapted for growth in serum-free medium to optimize the ability to recover human monoclonal immunoglobulins of high purity. MAbs produced by either means may be further purified, if desired, using filtration, centrifugation and various chromatographic methods such as FPLC or affinity chromatography. Fragments of the monoclonal antibodies of the disclosure can be obtained from the purified monoclonal antibodies by methods which include digestion with enzymes, such as pepsin or papain, and/or by cleavage of disulfide bonds by chemical reduction. Alternatively, monoclonal antibody fragments encompassed by the present disclosure can be synthesized using an automated peptide synthesizer. It also is contemplated that a molecular cloning approach may be used to generate monoclonal antibodies. Single B cells identified as responding to infection or vaccination because of plasmablast or activated B cell markers, or memory B cells labelled with the antigen of interest, can be sorted physically using paramagnetic bead selection or flow cytometric sorting, then RNA can be isolated from the single cells and antibody genes amplified by RT-PCR. Various single-cell RNA-seq methods are available to obtain antibody variable genes from single cells. Alternatively, antigen-specific bulk sorted populations of cells can be segregated into microvesicles and the matched heavy and light chain variable or common barcoding of heavy and light chain genes from a vesicle. Matched heavy and light chain genes from single cells also can be obtained from populations of antigen specific B cells by treating cells with cell-penetrating nanoparticles bearing RT-PCR primers and barcodes for marking transcripts with one barcode per cell. The antibody variable genes also can be isolated by RNA extraction of a hybridoma line and the antibody genes obtained by RT-PCR and cloned into an immunoglobulin expression vector. Alternatively, combinatorial immunoglobulin phagemid libraries are prepared from RNA isolated from the cell lines and phagemids expressing appropriate antibodies are selected by panning using viral antigens. The advantages of this approach over conventional hybridoma techniques are that approximately 104 times as many antibodies can be produced and screened in a single round, and that new specificities are generated by H and L chain combination which further increases the chance of finding appropriate antibodies. Other U.S. patents, each incorporated herein by reference, that teach the production of antibodies useful in the present disclosure include U.S. Patent 5,565,332, which describes the production of chimeric antibodies using a combinatorial approach; U.S. Patent 4,816,567 which describes recombinant immunoglobulin preparations; and U.S. Patent 4,867,973 which describes antibody-therapeutic agent conjugates. Antibodies according to the present disclosure may be defined, in the first instance, by their binding specificity. Those of skill in the art, by assessing the binding specificity/affinity of a given antibody using techniques well known to those of skill in the art, can determine whether such antibodies fall within the scope of the instant claims. For example, the epitope to which a given antibody bind may consist of a single contiguous sequence of 3 or more (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 1112, 13, 14, 15, 16, 17, 18, 19, 20) amino acids located within the antigen molecule (e.g., a linear epitope in a domain). Alternatively, the epitope may consist of a plurality of non-contiguous arnino acids (or amino acid sequences) located within the antigen molecule (e.g., a conformational epitope). Two main categories of SARS - CoV -2 antigens are the surface spike (S) protein and the internal proteins, especially the nucleocapsid (N) protein. Antibodies to the S protein will be useful for prophylaxis, or therapy, or diagnostics, or for characterizing vaccines. S protein antibodies will have additional binding specificity with that protein, with particular antibodies binding to the full-length ectodomain of the SARS-CoV-2 S protein (presented as a monomer or oligomer such as a timer; with or without conformation stabilizing mutations such as introduction of pralines at critical sites ("2P mutation")) and (a) anti-S protein antibodies that binds to the receptor binding domain (RBD), (b) anti-S protein antibodies that bind to domains other than the RBD. Some of the subset that bind to domains other than the RBD bind to the N terminal domain (NTD), while others bind to an epitope other than the NTD or RBD), and (c) S protein antibodies may further be found to neutralize SARS-CoV-2 by blocking binding of the SARS-CoV-2 S protein to its receptor, human angiotensin-converting enzyme 2 (hACE2), with others that neutralize but do not block receptor binding. Finally, antibodies can cross-react with both SARS-CoV-2 S protein and the S protein of other coronaviruses such as SARS-CoV, MERS-CoV, HCoV-229E, HCoV- OC43, HCoV-NL63 and/or HCoV-HKUl, as well as cross- neutralize both SARS-CoV-2 and these other coronaviruses. Another specificity will be antibodies that bind to N antibodies (or other internal targets) that will have primarily diagnostics uses. For example, antibodies to Nor other internal proteins of SARS-CoV-2 that specifically recognize SARS-CoV-2 or that cross-reactively recognize SARS-CoV-2 and other coronaviruses such as SARS-CoV, MERS-CoV, HCoV- 229E, HCoV-OC43, HCoV-NL63 and/or HCoV-HKUl. Various techniques known to persons of ordinary skill in the art can be used to determine whether an antibody "interacts with one or more amino acids" within a polypeptide or protein. Exemplary techniques include, for example, routine cross-blocking assays, such as that described in Antibodies, Harlow and Lane (Cold Spring Harbor Press, Cold Spring Harbor, N.Y.). Cross-blocking can be measured in various binding assays such as ELISA, biolayer interferometry, or surface plasmon resonance. Other methods include alanine scanning mutatonal analysis, peptide blot analysis (Reineke, Methods J\fol. BioL 248: 443- 63, 2004), peptide cleavage analysis, high-resolution electron microscopy techniques using single particle reconstruction, cryoEM, or tomography, crystallographic studies and NMR analysis. In addition, methods such as epitope excision, epitope extraction and chemical modification of antigens can be employed (Tomer Prot. Sci. 9: 487--496, 2000). Another method that can be used to identify the amino acids within a polypeptide with which an antibody interacts is hydrogen/deuterium exchange detected by mass spectrometry. In general terms, the hydrogen/deuterium exchange method involves deuterium-labelling the protein of interest, followed by binding the antibody to the deuterium-labelled protein. Next, the protein/antibody complex is transferred to water and exchangeable protons within amino acids that are protected by the antibody complex undergo deuterium-to-hydrogen back-exchange at a slower rate than exchangeable protons within amino acids that are not part of the interface. As a result, amino acids that form part of the protein/antibody interface may retain deuterium and therefore exhibit relatively higher mass compared to arnino acids not included in the interface. After dissociation of the antibody, the target protein is subjected to protease cleavage and mass spectrometry analysis, thereby revealing the deuterium-labelled residues which correspond to the specific amino acids with which the antibody interacts. See, e.g., Ehring, Analytical Biochemistry 267: 252-259 (1999): Engen and Smith, Anal. Chern.73: 256A-265A (2001). When the antibody neutralizes SARS-CoV- 2, antibody escape mutant variant organisms can be isolated by propagating SARS-CoV-2 in vitro or in animal models in the presence of high concentrations of the antibody. Sequence analysis of the SARS-CoV-2 gene encoding the antigen targeted by the antibody reveals the mutation(s) conferring antibody escape, indicating residues in the epitope or that affect the structure of the epitope allosterically. The term "epitope" refers to a site on an antigen to which B and/or T cells respond. B- cell epitopes can be formed both from contiguous arnino acids or non-contiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epltopes formed by tertiary folding are typically lost on treatment with denaturing solvents. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Modification-Assisted Profiling (MAP), also known as Antigen Structure-based Antibody Profiling (ASAP) is a method that categorizes large numbers of monoclonal antibodies (rnAbs) directed against the same antigen according to the similarities of the binding profile of each antibody to chernically or enzymatically modified antigen surfaces (see US. Patent Publication 2004/0101920, herein specifically incorporated by reference in its entirety). Each category may reflect a unique epitope either distinctly different from or partially overlapping with epitope represented by another category. This technology allows rapid filtering of genetically identical antibodies, such that characterization can be focused on genetically distinct antibodies, When applied to hybridoma screening, MAP may facilitate identification of rare hybridoma clones that produce mAbs having the desired characteristics. MAP may be used to sort the antibodies of the disclosure into groups of antibodies binding different epitopes. The present disclosure includes antibodies that may bind to the same epitope, or a portion of the epitope. Likewise, the present disclosure also includes antibodies that compete for binding to a target or a fragment thereof with any of the specific exemplary antibodies described herein. One can easily determine whether an antibody binds to the same epitope as or competes for binding with, a reference antibody by using routine methods known in the art. For example, to determine if a test antibody binds to the same epitope as a reference, the reference antibody is allowed to bind to target under saturating conditions. Next, the ability of a test antibody to bind to the target molecule is assessed. If the test antibody is able to bind to the target molecule following saturation binding with the reference antibody, it can be concluded that the test antibody binds to a different epitope than the reference antibody. On the other hand, if the test antibody is not able to bind to the target molecule following saturation binding with the reference antibody, then the test antibody may bind to the same epitope as the epitope bound by the reference antibody. To determine if an antibody competes for binding with a reference anti-SARS-CoV-2 antibody, the above-described binding rnethodology is perforrned in two orientations: In a first orientation, the reference antibody is allowed to bind to the SARS-CoV-2 antigen under saturating conditions followed by assessment of binding of the test antibody to the SARS- CoV- 2 rnolecu1e. In a second orientation, the test antibody is allowed to bind to the SARS- CoV-2 antigen molecule under saturating conditions followed by assessment of binding of the reference antibody to the SARS-CoV-2 molecule. If, in both orientations, only the first (saturating) antibody is capable of binding to SARS-CoV-2, then it is concluded that the test antibody and the reference antibody compete for binding to SARS-CoV-2. As will be appreciated by a person of ordinary skill in the art, an antibody that competes for binding with a reference antibody may not necessarily bind to the identical epitope as the reference antibody but may sterically block binding of the reference antibody by binding an overlapping or adjacent epitope. Two antibodies bind to the same or overlapping epitope, if each competitively inhibits (blocks) binding of the other to the antigen. That is, a 1-, 5-, 10-, 20-- or 100-fold excess of one antibody inhibits binding of the other by at least 50%, but preferably 75%, 90% or even 99% as measured in a competitive binding assay (see, e.g._Junghans et aL Cancer Res. 199050:1495-1502). Alternatively, two antibodies have the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of one antibody reduce or eliminate binding of the other. Two antibodies have overlapping epitopes if some arnino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other. Additional routine experimentation (e.g., peptide mutation and binding analyses) can then be carried out to confirm whether the observed lack of binding of the test antibody is in fact due to binding to the same epitope as the reference antibody or if steric blocking (or another phenomenon) is responsible for the lack of observed binding. Experiments of this sort can be performed using ELISA, RIA, surface plasmon resonance, flow cytometry or any other quantitative or qualitative antibody-binding assay available in the art. Structural studies with EM or crystallography also can demonstrate whether or not two antibodies that compete. for binding recognize the same epitope. In another aspect, there are provided monoclonal antibodies having clone-paired CDRs from the heavy and light chains as illustrated in Tables 3 and 4, respectively. Such antibodies may be produced by the clones discussed below in the Examples section using methods described herein. In another aspect, the antibodies may be defined by their variable sequence, which include additional "framework" regions. Furthermore, the antibodies sequences may vary from these sequences, optionally using methods discussed in greater detail below. For example, nucleic acid sequences may vary from those set out above in that (a) the variable regions may be segregated away from the constant domains of the light and heavy chains, (b) the nucleic acids may vary from those set out above while not affecting the residues encoded thereby, (c) the nucleic acids may vary from those set out above by a given percentage, e.g., 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% homology, (d) the nucleic acids may vary from those set out above by virtue of the ability to hybridize under high stringency conditions, as exemplified by low salt and/or high temperature conditions, such as provided by about 0.02 M to about 0.15 M NaCl at temperatures of about 50°C to about 70°C,(e) the amino acids may vary from those set out above by a given percentage, e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% homology, or (f) the amino acids may vary from those set out above by permitting conservative substitutions (discussed below). Each of the foregoing applies to the nucleic acid sequences and the amino acid sequences. When comparing polynucleotide and polypeptide sequences, two sequences are said to be "identical" if the sequence of nucleotides or amino acids in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A "comparison window" as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wis.), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M.0. (1978) A model of evolutionary change in proteins- - Matrices for detecting distant relationships. In Dayhoff, M. 0. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington D.C. Vol. 5, Suppl.3, pp.345-358; Hein J. (1990) Unified Approach to Alignment and Phylogeny pp. 626-645, Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, Calif.; Higgins, D. G. and Sharp, P. M. (1989) CABIOS 5:151-153; Myers, E.W. and Muller W. (1988) CABIOS 4:11- 17; Robinson, E. D. (1971) Comb. Theor 11:105; Santou, N. Nes, M. (1987) Mol. Biol. Evol. 4:406-425; Sneath, P.H. A. and Sokal, R.R. (1973) Numerical Taxonomy--the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, Calif.; Wilbur, W. J. and Lipman, D. J. (1983) Proc. Natl. Acad., Sci. USA 80:726- 730. Alternatively, optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL. Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol.48:443, by the search for similarity methods of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85: 2444, by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, PASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by inspection. One particular example of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nucl. Acids Res.25:3389-3402 and Altschul et al. (1990) J. Mol. Biol.215:403-410, respectively. BLAST and BLAST 2.0 can be used, for example, with the parameters described herein, to determine percent sequence identity for the polynucleotides and polypeptides of the disclosure. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. The rearranged nature of an antibody sequence and the variable length of each gene requires multiple rounds of BLAST searches for a single antibody sequence. Also, manual assembly of different genes is difficult and error-prone. The sequence analysis tool IgBLAST (world-wide-web at ncbi.nlm.nih.gov/igblast/) identifies matches to the germline V, D and J genes, details at rearrangement junctions, the delineation of lg V domain framework regions and complementarity determining regions. IgBLAST can analyze nucleotide or protein sequences and can process sequences in batches and allows searches against the germline gene databases and other sequence databases simultaneously to minimize the chance of missing possibly the best matching germline V gene. In one illustrative example, cumulative scores can be calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915) alignments, (B) of 50, expectation (E) of 10, M=5, N=-4 and a comparison of both strands. For amino acid sequences, a scoring matrix can be used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. In one approach, the "percentage of sequence identity" is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases or amino acid residues occur in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity. Yet another way of defining an antibody is as a "derivative" of any of the below- described antibodies and their antigen-binding fragments. The term "derivative" refers to an antibody or antigen-binding fragment thereof that immunospecifically binds to an antigen, but which comprises, one, two, three, four, five or more amino acid substitutions, additions, deletions or modifications relative to a "parental" (or wild-type) molecule. Such amino acid substitutions or additions may introduce naturally occurring (i.e., DNA-encoded) or non- naturally occurring amino acid residues. The term "derivative" encompasses, for example, as variants having altered CHI, hinge, CH2, CH3 or CH4 regions, so as to form, for example, antibodies, etc., having variant Fc regions that exhibit enhanced or impaired effector or binding characteristics. The term "derivative" additionally encompasses non-amino acid modifications, for example, amino acids that may be glycosylated (e.g., have altered mannose, 2-N- acetylglucosamine, galactose, fucose, glucose, sialic acid, 5-N- acetylneuraminic acid, 5-glycolneuraminic acid, etc. content), acetylated, pegylated, phosphorylated, amidated, derivatized by known protecting/blocking groups, proteolytic cleavage, linked to a cellular ligand or other protein, etc. In some embodiments, the altered carbohydrate modifications modulate one or more of the following: solubilization of the antibody, facilitation of subcellular transport and secretion of the antibody, promotion of antibody assembly, conformational integrity, and antibody-mediated effector function. In a specific embodiment, the altered carbohydrate modifications enhance antibody mediated effector function relative to the antibody lacking the carbohydrate modification. Carbohydrate modifications that lead to altered antibody mediated effector function are well known in the art (for example, see Shields, R. L. et al. (2002) J. Biol. Chem.277(30): 26733- 26740; Davies J. et al. (2001) Biotechnology & Bioengineering 74(4): 288-294). Methods of altering carbohydrate contents are known to those skilled in the art, see, e.g., Wallick, S. C. et al. (1988) J. Exp. Med.168(3): 1099-1109; Tao, M. H. et al. (1989) J. Immunol.143(8): 2595-2601; Routledge, E. G. et al. (1995) Transplantation 60(8):847-53; Elliott, S. et al. (2003) Nature Biotechnol. 21:414-21; Shields, R. L. et al. (2002) J. Biol. Chem.277(30): 26733-26740). A derivative antibody or antibody fragment can be generated with an engineered sequence or glycosylation state to confer preferred levels of activity in antibody dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), antibody- dependent neutrophil phagocytosis (ADNP), or antibody-dependent complement deposition (ADCD) functions as measured by bead-based or cell-based assays or in vivo studies in animal models. A derivative antibody or antibody fragment may be modified by chemical modifications using techniques known to those of skill in the art, including, but not limited to, specific chemical cleavage, acetylation, formulation, metabolic synthesis of tunicamycin, etc. In one embodiment, an antibody derivative will possess a similar or identical function as the parental antibody. In another embodiment, an antibody derivative will exhibit an altered activity relative to the parental antibody. For example, a derivative antibody (or fragment thereof) can bind to its epitope more tightly or be more resistant to proteolysis than the parental antibody. In various embodiments, one may choose to engineer sequences of the identified antibodies for a variety of reasons, such as improved expression, improved cross-reactivity or diminished off-target binding. Modified antibodies may be made by any technique known to those of skill in the art, including expression through standard molecular biological techniques, or the chemical synthesis of polypeptides. Methods for recombinant expression are addressed elsewhere in this document. The following is a general discussion of relevant goals techniques for antibody engineering. Hybridomas may be cultured, then cells lysed, and total RNA extracted. Random hexamers may be used with RT to generate cDNA copies of RNA, and then PCR performed using a multiplex mixture of PCR primers expected to amplify all human variable gene sequences. PCR product can be cloned into pGEM-T Easy vector, then sequenced by automated DNA sequencing using standard vector primers. Assay of binding and neutralization may be performed using antibodies collected from hybridoma supernatants and purified by FPLC, using Protein G columns. Recombinant full-length IgG antibodies can be generated by subcloning heavy and light chain Fv DNAs from the cloning vector into an IgG plasmid vector, transfected into 293 (e.g., Freestyle) cells or CHO cells, and antibodies can be collected and purified from the 293 or CHO cell supernatant. Other appropriate host cells systems include bacteria, such as E. coli, insect cells (S2, Sf9, Sf29, High Five), plant cells (e.g., tobacco, with or without engineering for human-like glycans), yeast, algae, or in a variety of non-human transgenic contexts, such as mice, rats, goats or cows. In particular, the antibody molecules of the invention may be generated using a yeast expression platform. The methylotrophic yeast Pichia pastoris provides an efficient platform and pichia expression systems are commercially available. Expression of nucleic acids encoding antibodies, both for the purpose of subsequent antibody purification, and for immunization of a host, is also contemplated. Antibody coding sequences can be RNA, such as native RNA or modified RNA. Modified RNA contemplates certain chemical modifications that confer increased stability and low immunogenicity to mRNAs, thereby facilitating expression of therapeutically important proteins. For instance, N1-methyl-pseudouridine (N1mΨ) outperforms several other nucleoside modifications and their combinations in terms of translation capacity. In addition to turning off the immune/eIF2α phosphorylation-dependent inhibition of translation, incorporated N1mΨ nucleotides dramatically alter the dynamics of the translation process by increasing ribosome pausing and density on the mRNA. Increased ribosome loading of modified mRNAs renders them more permissive for initiation by favouring either ribosome recycling on the same mRNA or de novo ribosome recruitment. Such modifications could be used to enhance antibody expression in vivo following inoculation with RNA. The RNA, whether native or modified, may be delivered as naked RNA or in a delivery vehicle, such as a lipid nanoparticle. Alternatively, DNA encoding the antibody may be employed for the same purposes. The DNA is included in an expression cassette comprising a promoter active in the host cell for which it is designed. The expression cassette is advantageously included in a replicable vector, such as a conventional plasmid or minivector. Vectors include viral vectors, such as poxviruses, adenoviruses, herpesviruses, adeno-associated viruses, and lentiviruses are contemplated. Replicons encoding antibody genes such as alphavirus replicons based on VEE virus or Sindbis virus are also contemplated. Delivery of such vectors can be performed by needle through intramuscular, subcutaneous, or intradermal routes, or by transcutaneous electroporation when in vivo expression is desired. The rapid availability of antibody produced in the same host cell and cell culture process as the final cGMP manufacturing process has the potential to reduce the duration of process development programs. Lonza has developed a generic method using pooled transfectants grown in CDACF medium, for the rapid production of small quantities (up to 50 g) of antibodies in CHO cells. Although slightly slower than a true transient system, the advantages include a higher product concentration and use of the same host and process as the production cell line. Example of growth and productivity of GS-CHO pools, expressing a model antibody, in a disposable bioreactor: in a disposable bag bioreactor culture (5 L working volume) operated in fed-batch mode, a harvest antibody concentration of 2 g/L was achieved within 9 weeks of transfection. Antibody molecules will comprise fragments (such as F(ab'), F(ab')2) that are produced, for example, by the proteolytic cleavage of the mAbs, or single-chain immunoglobulins producible, for example, via recombinant means. F(ab') antibody derivatives are monovalent, while F(ab')2 antibody derivatives are bivalent. In one embodiment, such fragments can be combined with one another, or with other antibody fragments or receptor ligands to form "chimeric" binding molecules. Significantly, such chimeric molecules may contain substituents capable of binding to different epitopes of the same molecule. In related embodiments, the antibody is a derivative of the disclosed antibodies, e.g.,an antibody comprising the CDR sequences identical to those in the disclosed antibodies (e.g., a chimeric, or CDR-grafted antibody). Alternatively, one may wish to make modifications, such as introducing conservative changes into an antibody molecule. In making such changes, the hydropathic index of amino acids may be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like. It also is understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. U.S. Patent 4,554,101, incorporated herein by reference, states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with a biological property of the protein. As detailed in U.S. Patent 4,554,101, the following hydrophilicity values have been assigned to amino acid residues: basic amino acids: arginine (+3.0), lysine (+3.0), and histidine (-0.5); acidic amino acids: aspartate (+3.0 ± 1), glutamate (+3.0 ± 1), asparagine (+0.2), and glutamine (+0.2); hydrophilic, nonionic amino acids: serine (+0.3), asparagine (+0.2), glutamine (+0.2), and threonine (-0.4), sulfur containing amino acids: cysteine (-1.0) and methionine (-1.3); hydrophobic, nonaromatic amino acids: valine (-1.5), leucine (-1.8), isoleucine (-1.8), praline (-0.5 ± 1), alanine (-0.5), and glycine (0); hydrophobic, aromatic amino acids: tryptophan (- 3.4), phenylalanine (-2.5), and tyrosine (-2.3). It is understood that an amino acid can be substituted for another having a similar hydrophilicity and produce a biologically or immunologically modified protein. In such changes, the substitution of amino acids whose hydrophilicity values are within± 2 is preferred, those that are within ± 1 are particularly preferred, and those within ± 0.5 are even more particularly preferred. As outlined above, amino acid substitutions generally are based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity,hydrophilicity, charge, size, and the like. Exemplary substitutions that take into consideration the various foregoing characteristics are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine. The present disclosure also contemplates isotype modification. By modifying the Fc region to have a different isotype, different functionalities can be achieved. For example, changing to IgG1 can increase antibody dependent cell cytotoxicity, switching to class A can improve tissue distribution, and switching to class M can improve valency. Alternatively or additionally, it may be useful to combine amino acid modifications with one or more further amino acid modifications that alter C1q binding and/or the complement dependent cytotoxicity (CDC) function of the Fc region of an IL-23pl9 binding molecule. The binding polypeptide of particular interest may be one that binds to C1q and displays complement dependent cytotoxicity. Polypeptides with pre-existing C1q binding activity, optionally further having the ability to mediate CDC may be modified such that one or both of these activities are enhanced. Amino acid modifications that alter C1q and/or modify its complement dependent cytotoxicity function are described, for example, in WO/0042072, which is hereby incorporated by reference. One can design an Fc region of an antibody with altered effector function, e.g., by modifying C1q binding and/or FcγR binding and thereby changing CDC activity and/or ADCC activity. "Effector functions" are responsible for activating or diminishing a biological activity (e.g., in a subject). Examples of effector functions include, but are not limited to: C1q binding; complement dependent cytotoxicity (CDC); Fc receptor binding; antibody- dependent cell- mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptor; BCR), etc. Such effector functions may require the Fc region to be combined with a binding domain (e.g., an antibody variable domain) and can be assessed using various assays (e.g., Fc binding assays, ADCC assays, CDC assays, etc.). For example, one can generate a variant Fc region of an antibody with improved C1q binding and improved FcγR III binding (e.g., having both improved ADCC activity and improved CDC activity). Alternatively, if it is desired that effector function be reduced or ablated, a variant Fc region can be engineered with reduced CDC activity and/or reduced ADCC activity. In other embodiments, only one of these activities may be increased, and, optionally, also the other activity reduced (e.g., to generate an Fc region variant with improved ADCC activity, but reduced CDC activity and vice versa). Fc mutations can also be introduced and engineered to alter their interaction with the neonatal Fc receptor (FcRn) and improve their pharmacokinetic properties. A collection of human Fc variants with improved binding to the FcRn have been described (Shields et al., (2001). High resolution mapping of the binding site on human IgG1 for FcγR I, FcγR II, FcγR III, and FcRn and design of IgG1 variants with improved binding to the FcγR , (J. Biol. Chem.276:6591-6604). A number of methods are known that can result in increased half- life (Kuo and Aveson, (2011)), including amino acid modifications may be generated through techniques including alanine scanning mutagenesis, random mutagenesis and screening to assess the binding to the neonatal Fc receptor (FcRn) and/or the in vivo behaviour. Computational strategies followed by mutagenesis may also be used to select one of amino acid mutations to mutate. The present disclosure therefore provides a variant of an antigen binding protein with optimized binding to FcRn. In a particular embodiment, the variant of an antigen binding protein comprises at least one amino acid modification in the Fc region of the antigen binding protein, wherein the modification is selected from the group consisting of 226, 227, 228,230,231,233,234,239,241,243,246,250,252,254,256,259,264,265,267,269,270,276,2 84,285,288,289,290,291,292,294,297,298,299,301,302,303,305,307,308,309,311,315,31 7,320,322,325,327,330,332,334,335,338,340,342,343,345,347,350,352,354,355,356,359, 360,361,362,369,370,371,375,378,380,382,384,385,386,387,389,390,392,393,394,395,3 96,397,398,399,400,401403,404,408,411,412,414,415,416,418,419,420,421, 422, 424, 426, 428, 433, 434, 438, 439, 440, 443, 444, 445, 446 and 447 of the Fc region as compared to the parent polypeptide, wherein the numbering of the amino acids in the Fc region is that of the EU index in Kabat. In a further embodiment of the disclosure the modifications are M252Y/S254T/T256E. In a further embodiment of the disclosure the modifications are M252Y/S254T/T256E. In a further embodiment of the disclosure, the modifications are T250Q/M428L Additionally, various publications describe methods for obtaining physiologically active molecules whose half-lives are modified, see for example Kontermann (2009) either by introducing an FcRn-binding polypeptide into the molecules or by fusing the molecules with antibodies whose FcRn-binding affinities are preserved but affinities for other Fe receptors have been greatly reduced or fusing with FcRn binding domains of antibodies. Derivatized antibodies may be used to alter the half-lives (e.g., serum half-lives) of parental antibodies in a mammal, particularly a human. Such alterations may result in a half- life of greater than 15 days, preferably greater than 20 days, greater than 25 days, greater than 30 days, greater than 35 days, greater than 40 days, greater than 45 days, greater than 2 months, greater than 3 months, greater than 4 months, or greater than 5 months. The increased half- lives of the antibodies of the present disclosure or fragments thereof in a mammal, preferably in a human, results in a higher serum titre of the antibodies or antibody fragments in the mammal, and thus reduces the frequency of the administration of the antibodies or antibody fragments and/or reduces the concentration of the antibodies or antibody fragments to be administered. Antibodies or fragments thereof having increased in vivo half-lives can be generated by techniques known to those of skill in the art. For example, antibodies or fragments thereof with increased in vivo half-lives can be generated by modifying (e.g., substituting, deleting or adding) amino acid residues identified as involved in the interaction between the Fe domain and the FcRn receptor. Beltramello et al. (2010) previously reported the modification of neutralizing mAbs, due to their tendency to enhance dengue virus infection, by generating in which leucine residues at positions 1.3 and 1.2 of CH2 domain (according to the IMGT unique numbering for C-domain) were substituted with alanine residues. This modification, also known as "LALA" mutation, abolishes antibody binding to FcγR I, FcγR II and FcγR IIIa, as described by Hessell et al. (2007). The variant and unmodified recombinant mAbs were compared for their capacity to neutralize and enhance infection by the four dengue virus serotypes. LALA variants retained the same neutralizing activity as unmodified mAb but were completely devoid of enhancing activity. LALA mutations of this nature are therefore contemplated in the context of the presently disclosed antibodies. In a further embodiment of the disclosure the modifications are L234A /L235A (LALA) or L234A /L235A + P329G (LALA-PG). A particular embodiment of the present disclosure is an isolated monoclonal antibody, or antigen binding fragment thereof, containing a substantially homogeneous glycan without sialic acid, galactose, or fucose. The monoclonal antibody comprises a heavy chain variable region and a light chain variable region, both of which may be attached to heavy chain or light chain constant regions respectively. The aforementioned substantially homogeneous glycan may be covalently attached to the heavy chain constant region. Another embodiment of the present disclosure comprises a mAb with a novel Fc glycosylation pattern. The isolated monoclonal antibody, or antigen binding fragment thereof, is present in a substantially homogenous composition represented by the GNGN or G1/G2 glycoform. Fc glycosylation plays a significant role in anti-viral and anti-cancer properties of therapeutic mAbs. The disclosure is in line with a recent study that shows increased anti- lentivirus cell-mediated viral inhibition of a fucose free anti-HIV mAb in vitro. This embodiment of the present disclosure with homogenous glycans lacking a core fucose, showed increased protection against specific viruses by a factor greater than two-fold. Elimination of core fucose dramatically improves the ADCC activity of mAbs mediated by natural killer (NK) cells but appears to have the opposite effect on the ADCC activity of polymorphonuclear cells (PMNs). The isolated monoclonal antibody, or antigen binding fragment thereof, comprising a substantially homogenous composition represented by the GNGN or G1/G2 glycoform exhibits increased binding affinity for Fc gamma RI and Fc gamma RIII compared to the same antibody without the substantially homogeneous GNGN glycoform and with GO, GlF, G2F, GNP, GNGNF or GNGNFX containing glycoforms. In one embodiment of the present disclosure, the antibody dissociates from Fc gamma RI with a Kd of 1 x 10-3 M or less and from Fc gamma RIII with a Kd of 1 x 10-7 M or less. Glycosylation of an Fc region is typically either N-linked or O-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue. O- linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used. The recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain peptide sequences are asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except praline. Thus, the presence of either of these peptide sequences in a polypeptide creates a potential glycosylation site. The glycosylation pattern may be altered, for example, by deleting one or more glycosylation site(s) found in the polypeptide, and/or adding one or more glycosylation site(s) that are not present in the polypeptide. Addition of glycosylation sites to the Fc region of an antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites). An exemplary glycosylation variant has an amino acid substitution of residue Asn 297 of the heavy chain. The alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original polypeptide (for O-linked glycosylation sites). Additionally, a change of Asn 297 to Ala can remove one of the glycosylation sites. In certain embodiments, the antibody is expressed in cells that express beta (l,4)-N- acetylglucosaminyltransferase III (GnT III), such that GnT III adds GlcNAc to the IL-23p19 antibody. Methods for producing antibodies in such a fashion are provided in WO/9954342, WO/03011878, patent publication 20030003097Al, and Umana et al., Nature Biotechnology, 17:176-180, February 1999. Cell lines can be altered to enhance or reduce or eliminate certain post-translational modifications, such as glycosylation, using genome editing technology suchas Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR). For example, CRISPR technology can be used to eliminate genes encoding glycosylating enzymes in 293 or CHO cells used to express recombinant monoclonal antibodies. It is possible to engineer the antibody variable gene sequences obtained from human B cells to enhance their manufacturability and safety. Potential protein sequence liabilities can be identified by searching for sequence motifs associated with sites containing: 1) Unpaired Cys residues, 2) N-linked glycosylation, 3) Asn deamidation, 4) Asp isomerization, 5) SYE truncation, 6) Met oxidation, 7) Trp oxidation, 8) N-terminal glutamate, 9) Integrin binding, 10) CD1lc/CD18 binding, or 11) Fragmentation Such motifs can be eliminated by altering the synthetic gene for the cDNA encoding recombinant antibodies. Protein engineering efforts in the field of development of therapeutic antibodies clearly reveal that certain sequences or residues are associated with solubility differences (Fernandez- Escamilla et al., Nature Biotech., 22 (10), 1302-1306, 2004; Chennamsetty et al., PNAS, 106 (29), 11937-11942, 2009; Voynov et al., Biocon. Chem., 21 (2), 385-392, 2010) Evidence from solubility-altering mutations in the literature indicate that some hydrophilic residues such as aspartic acid, glutamic acid, and serine contribute significantly more favourably to protein solubility than other hydrophilic residues, such as asparagine, glutamine, threonine, lysine, and arginine. Elevated temperature may be used to unfold antibodies to determine relative stability, using average apparent melting temperatures. Differential Scanning Calorimetry (DSC) measures the heat capacity, Cp, of a molecule (the heat required to warm it, per degree) as a function of temperature. One can use DSC to study the thermal stability of antibodies. DSC data for mAbs is particularly interesting because it sometimes resolves the unfolding of individual domains within the mAb structure, producing up to three peaks in the thermogram (from unfolding of the Fab, CH2, and CH3 domains). Typically unfolding of the Fab domain produces the strongest peak. The DSC profiles and relative stability of the Fe portion show characteristic differences for the human IgG1, IgG2, IgG3, and IgG4 subclasses (Garber and Demarest, Biochem. Biophys. Res. Commun. 355, 751-757, 2007). One also can determine average apparent melting temperature using circular dichroism (CD), performed with a CD spectrometer. Far-UV CD spectra will be measured for antibodies in the range of 200 to 260 nm at increments of 0.5 nm. The final spectra can be determined as averages of 20 accumulations. Residue ellipticity values can be calculated after background subtraction. Thermal unfolding of antibodies (0.1 mg/mL) can be monitored at 235 nm from 25-95 °C and a heating rate of 1 °C/min. One can use dynamic light scattering (DLS) to assess for propensity for aggregation. DLS is used to characterize size of various particles including proteins. If the system is not diverse in size, the mean effective diameter of the particles can be determined. This measurement depends on the size of the particle core, the size of surface structures, and particle concentration. Since DLS essentially measures fluctuations in scattered light intensity due to particles, the diffusion coefficient of the particles can be determined. DLS software in commercial DLA instruments displays the particle population at different diameters. Stability studies can be done conveniently using DLS. DLS measurements of a sample can show whether the particles aggregate over time or with temperature variation by determining whether the hydrodynamic radius of the particle increases. If particles aggregate, one can see a larger population of particles with a larger radius. Stability depending on temperature can be analyzed by controlling the temperature in situ. Capillary electrophoresis (CE) techniques include proven methodologies for determining features of antibody stability. One can use an iCE approach to resolve antibody protein charge variants due to deamidation, C-terminal lysines, sialylation, oxidation, glycosylation, and any other change to the protein that can result in a change in pH of the protein. Each of the expressed antibody proteins can be evaluated by high throughput, free solution isoelectric focusing (IEF) in a capillary column (cIEF), using a Protein Simple Maurice instrument. Whole-column UV absorption detection can be performed every 30 seconds for real time monitoring of molecules focusing at the isoelectric points (pls). This approach combines the high resolution of traditional gel IEF with the advantages of quantitation and automation found in column-based separations while eliminating the need for a mobilization step. The technique yields reproducible, quantitative analysis of identity, purity, and heterogeneity profiles for the expressed antibodies. The results identify charge heterogeneity and molecular sizing on the antibodies, with both absorbance and native fluorescence detection modes and with sensitivity of detection down to 0.7 µg/mL. One can determine the intrinsic solubility score of antibody sequences. The intrinsic solubility scores can be calculated using CamSol Intrinsic (Sormanni et al., J Mol Biol 427, 478-490, 2015). The amino acid sequences for residues 95-102 (Kabat numbering) in CDRH3 of each antibody fragment such as a scFv can be evaluated via the online program to calculate the solubility scores. One also can determine solubility using laboratory techniques. Various techniques exist, including addition of lyophilized protein to a solution until the solution becomes saturated and the solubility limit is reached, or concentration by ultrafiltration in a microconcentrator with a suitable molecular weight cut-off. The most straightforward method is induction of amorphous precipitation, which measures protein solubility using a method involving protein precipitation using ammonium sulfate (Trevino et al., J Mol Biol, 366: 449-460, 2007). Ammonium sulfate precipitation gives quick and accurate information on relative solubility values. Ammonium sulfate precipitation produces precipitated solutions with well-defined aqueous and solid phases and requires relatively small amounts of protein. Solubility measurements performed using induction of amorphous precipitation by ammonium sulfate also can be done easily at different pH values. Protein solubility is highly pH dependent, and pH is considered the most important extrinsic factor that affects solubility. Generally, it is thought that autoreactive clones should be eliminated during ontogeny by negative selection, however it has become clear that many human naturally occurring antibodies with autoreactive properties persist in adult mature repertoires, and the autoreactivity may enhance the antiviral function of many antibodies to pathogens. It has been noted that HCDR3 loops in antibodies during early B cell development are often rich in positive charge and exhibit autoreactive patterns (Wardemann et al., Science 301, 1374- 1377, 2003). A given antibody may be tested for autoreactivity by assessing the level of binding to human origin cells in microscopy (using adherent HeLa or HEp-2 epithelial cells) and flow cytometric cell surface staining (using suspension Jurkat T cells and 293S human embryonic kidney cells). Autoreactivity also can be surveyed using assessment of binding to tissues in tissue arrays. B cell repertoire deep sequencing of human B cells from blood donors is being performed on a wide scale in many recent studies. Sequence information about a significant portion of the human antibody repertoire facilitates statistical assessment of antibody sequence features common in healthy humans. With knowledge about the antibody sequence features in a human recombined antibody variable gene reference database, the position specific degree of "Human Likeness" (HL) of an antibody sequence can be estimated. HL has been shown to be useful for the development of antibodies in clinical use, like therapeutic antibodies or antibodies as vaccines. The goal is to increase the human likeness of antibodies to reduce potential adverse effects and anti-antibody immune responses that will lead to significantly decreased efficacy of the antibody drug or can induce serious health implications. It is possible to assess antibody characteristics of the combined antibody repertoire of healthy human blood donors of about 400 million sequences in total and create a novel "relative Human Likeness" (rHL) score that focuses on the hypervariable region of the antibody. The rHL score may easily distinguish between human (positive score) and non-human sequences (negative score). Antibodies can be engineered to eliminate residues that are not common in human repertoires. A single chain variable fragment (scFv) is a fusion of the variable regions of the heavy and light chains of immunoglobulins, linked together with a short (usually serine, glycine) linker. This chimeric molecule retains the specificity of the original immunoglobulin, despite removal of the constant regions and the introduction of a linker peptide. This modification usually leaves the specificity unaltered. These molecules were created historically to facilitate phage display where it is highly convenient to express the antigen binding domain as a single peptide. Alternatively, scFv can be created directly from subcloned heavy and light chains derived from a hybridoma or B cell. Single chain variable fragments lack the constant Fc region found in complete antibody molecules, and thus, the common binding sites (e.g., protein A/G) used to purify antibodies. These fragments can often be purified/immobilized using Protein L since Protein L interacts with the variable region of kappa light chains. Flexible linkers generally are comprised of helix- and turn-promoting amino acid residues such as alanine, serine and glycine. However, other residues can function as well. Tang et al. (1996) used phage display as a means of rapidly selecting tailored linkers for single- chain antibodies (scFvs) from protein linker libraries. A random linker library was constructed in which the genes for the heavy and light chain variable domains were linked by a segment encoding an 18-amino acid polypeptide of variable composition. The scFv repertoire (approx. 5 x 106 different members) was displayed on filamentous phage and subjected to affinity selection with hapten. The population of selected variants exhibited significant increases in binding activity but retained considerable sequence diversity. Screening 1054 individual variants subsequently yielded a catalytically active scFv that was produced efficiently in soluble form. Sequence analysis revealed a conserved praline in the linker two residues after the VH C terminus and an abundance of arginines and pralines at other positions as the only common features of the selected tethers. The recombinant antibodies of the present disclosure may also involve sequences or moieties that permit dimerization or multimerization of the receptors. Such sequences include those derived from IgA, which permit formation of multimers in conjunction with the J-chain. Another multimerization domain is the Gal4 dimerization domain. In other embodiments, the chains may be modified with agents such as biotin/avidin, which permit the combination of two antibodies. In a separate embodiment, a single-chain antibody can be created by joining receptor light and heavy chains using a non-peptide linker or chemical unit. Generally, the light and heavy chains will be produced in distinct cells, purified, and subsequently linked together in an appropriate fashion (i.e., the N-terminus of the heavy chain being attached to the C-terminus of the light chain via an appropriate chemical bridge). Cross-linking reagents are used to form molecular bridges that tie functional groups of two different molecules, e.g., a stabilizing and coagulating agent. However, it is contemplated that dimers or multimers of the same analog or heteromeric complexes comprised of different analogs can be created. To link two different compounds in a step-wise manner, hetero- bifunctional cross-linkers can be used that eliminate unwanted homopolymer formation. An exemplary hetero-bifunctional cross-linker contains two reactive groups: one reacting with primary amine group (e.g., N-hydroxy succinimide) and the other reacting with a thiol group (e.g., pyridyl disulfide, maleimides, halogens, etc.). Through the primary amine reactive group, the cross-linker may react with the lysine residue(s) of one protein (e.g., the selected antibody or fragment) and through the thiol reactive group, the cross-linker, already tied up to the first protein, reacts with the cysteine residue (free sulfhydryl group) of the other protein (e.g., the selective agent). Typically, a cross-linker having reasonable stability in blood will be employed. Numerous types of disulfide-bond containing linkers are known that can be successfully employed to conjugate targeting and therapeutic/preventative agents. Linkers that contain a disulfide bond that is sterically hindered may prove to give greater stability in vivo, preventing release of the targeting peptide prior to reaching the site of action. These linkers are thus one group of linking agents. Another cross-linking reagent is SMPT, which is a bifunctional cross-linker containing a disulfide bond that is "sterically hindered" by an adjacent benzene ring and methyl groups. It is believed that steric hindrance of the disulfide bond serves a function of protecting the bond from attack by thiolate anions such as glutathione which can be present in tissues and blood, and thereby help in preventing decoupling of the conjugate prior to the delivery of the attached agent to the target site. The SMPT cross-linking reagent, as with many other known cross-linking reagents, lends the ability to cross-link functional groups such as the SH of cysteine or primary amines (e.g., the epsilon amino group of lysine). Another possible type of cross-linker includes the hetero- bifunctional photoreactive phenylazides containing a cleavable disulfide bond such as sulfosuccinimidy1-2-(p-azido salicylamido) ethy1-1,3'-dithiopropionate. The N-hydroxy- succinimidy l group reacts with primary amino groups and the phenylazide (upon photolysis) reacts non-selectively with any amino acid residue. In addition to hindered cross-linkers, non-hindered linkers also can be employed in accordance herewith. Other useful cross-linkers, not considered to contain or generate a protected disulfide, include SATA, SPDP and 2-iminothiolane (Wawrzynczak & Thorpe, 1987). The use of such cross-linkers is well understood in the art. Another embodiment includes the use of flexible linkers. U.S. Patent 4,680,338, describes bifunctional linkers useful for producing conjugates of ligands with amine-containing polymers and/or proteins, especially for forming antibody conjugates with chelators, drugs, enzymes, detectable labels and the like. U.S. Patents 5,141,648 and 5,563,250 disclose cleavable conjugates containing a labile bond that is cleavable under a variety of mild conditions. This linker is particularly useful in that the agent of interest may be bonded directly to the linker, with cleavage resulting in release of the active agent. Particular uses include adding a free amino or free sulfhydryl group to a protein, such as an antibody, or a drug. U.S. Patent 5,856,456 provides peptide linkers for use in connecting polypeptide constituents to make fusion proteins, e.g., single chain antibodies. The linker is up to about 50 amino acids in length, contains at least one occurrence of a charged amino acid (preferably arginine or lysine) followed by a praline, and is characterized by greater stability and reduced aggregation. U.S. Patent 5,880,270 discloses aminooxy-containing linkers useful in a variety of immunodiagnostic and separative techniques. In certain embodiments, antibodies of the present disclosure are bispecific or multispecific. Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Exemplary bispecific antibodies may bind to two different epitopes of a single antigen. Other such antibodies may combine a first antigen binding site with a binding site for a second antigen. Alternatively, an anti-pathogen arm may be combined with an arm that binds to a triggering molecule on a leukocyte, such as a T-cell receptor molecule (e.g., CD3), or Fe receptors for IgG ( FcγR ), such as FcγR I (CD64), FcγR II (CD32) and FcγR III (CD16), so as to focus and localize cellular defense mechanisms to the infected cell. Bispecific antibodies may also be used to localize cytotoxic agents to infected cells. These antibodies possess a pathogen-binding arm and an arm that binds the cytotoxic agent (e.g., saporin, anti-interferon-a, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten). Bispecific antibodies can be prepared as full-length antibodies or antibody fragments (e.g., F(ab')2 bispecific antibodies). WO 96/16673 describes a bispecific anti- ErbB2/anti-Fc gamma RIII antibody and U.S. Patent 5,837,234 discloses a bispecific anti- ErbB2/anti-Fc gamma RI antibody. A bispecific anti-ErbB2/Fc alpha antibody is shown in WO98/02463. U.S. Patent 5,821,337 teaches a bispecific anti-ErbB2/anti-CD3 antibody. Methods for making bispecific antibodies are known in the art. Traditional production of full- length bispecific antibodies is based on the co-expression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al., EMBO J., 10:3655-3659 (1991). According to a different approach, antibody variable regions with the desired binding specificities (antibody-antigen combining sites) are fused to immunoglobulin constant domain sequences. Preferably, the fusion is with an lg heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CH1) containing the site necessary for light chain bonding, present in at least one of the fusions. DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host cell. This provides for greater flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of the three polypeptide chains used in the construction provide the optimum yield of the desired bispecific antibody. It is, however, possible to insert the coding sequences for two or all three polypeptide chains into a single expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios have no significant effect on the yield of the desired chain combination. In a particular embodiment of this approach, the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986). According to another approach described in U.S. Patent 5,731,168, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers that are recovered from recombinant cell culture. The preferred interface comprises at least a part of the CH3 domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g., tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g., alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers. Bispecific antibodies include cross-linked or "heteroconjugate" antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Patent 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Patent 4,676,980, along with a number of cross-linking techniques. Techniques for generating bispecific antibodies from antibody fragments have also been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science, 229: 81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab')2 fragments. These fragments are reduced in the presence of the dithiol complexing agent, sodium arsenite, to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes. Techniques exist that facilitate the direct recovery of Fab'-SH fragments from E. coli, which can be chemically coupled to form bispecific antibodies. Shalaby et al., J. Exp. Med., 175: 217-225 (1992) describe the production of a humanized bispecific antibody F(ab')2 molecule. Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets. Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described (Merchant et al., Nat. Bioteclmo!, 16, 677-681 (1998). doi:10.1038/nbt0798-677pmid:9661204). For example, bispecific antibodies have been produced using leucine zippers (Kostelny et al., J. Immunol., 148(5):1547-1553, 1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re- oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a VH connected to a VL by a linker that is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen- binding sites. Another strategy for making bispecific antibody fragments by the use of single- chain Fv (sFv) dimers has also been reported. See Gruber et al., J. Immunol., 152:5368 (1994). In a particular embodiment, a bispecific or multispecific antibody may be formed as a DOCK-AND-LOCK.™ (DNL™) complex (see, e.g., U.S. Patents 7,521,056: 7,527,787: 7,534,866; 7,550,143 and 7,666,400, the Examples section of each of which is incorporated herein by reference.) Generally, the technique takes advantage of the specific and high affinity binding interactions that occur between a dimerization and docking domain (DDD) sequence of the regulatory (R) subunits of cAMP-dependent protein kinase (PKA) and an anchor domain (AD) sequence derived from any of a variety of AKAP proteins (Baillie er aL FEES Letters.2005; 579: 3264; Wong and Scott, Nat. Rev. Mol. Cell Biol.2004; 5: 959). The DDD and AD peptides may be attached to any protein, peptide or other rnolecule. Because the DDD sequences spontaneously dimerize, and bind to the AD sequence, the technique allows the formation of complexes between any selected molecules that may be attached to DDD or AD sequences. Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared (Tutt et al., J. Immunol. 147: 60, 1991; Xu et al., Science, 358(6359):85-90, 2017). A multivalent antibody may be internalized (and/or catabolized) faster than a bivalent antibody by a cell expressing an antigen to which the antibodies bind. The antibodies of the present disclosure can be multivalent antibodies with three or more antigen binding sites (e.g., tetravalent antibodies), which can be readily produced by recombinant expression of nucleic acid encoding the polypeptide chains of the antibody. The multivalent antibody can comprise a dimerization domain and three or more antigen binding sites. The preferred dimerization domain comprises (or consists of) an Fc region or a hinge region. In this scenario, the antibody will comprise an Fc region and three or more antigen binding sites amino-terminal to the Fc region. The preferred multivalent antibody herein comprises (or consists of) three to about eight, but preferably four, antigen binding sites. The multivalent antibody comprises at least one polypeptide chain (and preferably two polypeptide chains), wherein the polypeptide chain(s) comprise two or more variable regions. For instance, the polypeptide chain(s) may comprise VD1-(X1)n-VD2-(X2)n-Fc, wherein VD1 is a first variable region, VD2 is a second variable region, Fc is one polypeptide chain of an Fc region, X1 and X2 represent an amino acid or polypeptide, and n is 0 or 1. For instance, the polypeptide chain(s) may comprise: VH-CH1-flexible linker-VH-CH1-Fc region chain; or VH-CH1-VH- CH1-Fc region chain. The multivalent antibody herein preferably further comprises at least two (and preferably four) light chain variable region polypeptides. The multivalent antibody herein may, for instance, comprise from about two to about eight light chain variable region polypeptides. The light chain variable region polypeptides contemplated here comprise a light chain variable region and, optionally, further comprise a CL domain. Charge modifications are particularly useful in the context of a multispecific antibody, where amino acid substitutions in Fab molecules result in reducing the mispairing of light chains with non-matching heavy chains (Bence-Jones-type side products), which can occur in the production of Fab-based bi-/multispecific antigen binding molecules with a VH/VLexchange in one (or more, in case of molecules comprising more than two antigen-binding Fab molecules) of their binding arms (see also PCT publication no. WO 2015/150447, particularly the examples therein, incorporated herein by reference in its entirety). Accordingly, in particular embodiments, an antibody comprised in the therapeutic agent comprises (a) a first Fab molecule which specifically binds to a first antigen (b) a second Fab molecule which specifically binds to a second antigen, and wherein the variable domains VL and VH of the Fab light chain and the Fab heavy chain are replaced by each other, wherein the first antigen is an activating T cell antigen, and the second antigen is a target cell antigen, or the first antigen is a target cell antigen, and the second antigen is an activating T cell antigen; and wherein i) in the constant domain CL of the first Fab molecule under a) the amino acid at position 124 is substituted by a positively charged amino acid (numbering according to Kabat), and wherein in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 or the amino acid at position 213 is substituted by a negatively charged amino acid (numbering according to Kabat EU index); or ii) in the constant domain CL of the second Fab molecule under b) the amino acid at position 124 is substituted by a positively charged amino acid (numbering according to Kabat), and wherein in the constant domain CH1 of the second Fab molecule under b) the amino acid at position 147 or the amino acid at position 213 is substituted by a negatively charged amino acid (numbering according to Kabat EU index). The antibody may not comprise both modifications mentioned under i) and ii). The constant domains CL and CH1 of the second Fab molecule are not replaced by each other (i.e., remain unexchanged). In another embodiment of the antibody, in the constant domain CL of the first Fab molecule under a) the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat) (in one preferred embodiment independently by lysine (K) or arginine (R)), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 or the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index). In a further embodiment, in the constant domain CL of the first Fab molecule under a) the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index). In a particular embodiment, in the constant domain CL of the first Fab molecule under a) the amino acid at position 124 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat) (in one preferred embodiment independently by lysine (K) or arginine (R)) and the amino acid at position 123 is substituted independently by lysine (K), arginine (R) or histidine (H) (numbering according to Kabat) (in one preferred embodiment independently by lysine (K) or arginine (R)), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted independently by glutamic acid (E), or aspartic acid (D) (numbering according to Kabat EU index). In a more particular embodiment, in the constant domain CL of the first Fab molecule under a) the amino acid at position 124 is substituted by lysine (K) (numbering according to Kabat) and the amino acid at position 123 is substituted by lysine (K) or arginine (R) (numbering according to Kabat), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 is substituted by glutamic acid (E) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted by glutamic acid (E) (numbering according to Kabat EU index). In an even more particular embodiment, in the constant domain CL of the first Fab molecule under a) the amino acid at position 124 is substituted by lysine (K) (numbering according to Kabat) and the amino acid at position 123 is substituted by arginine (R) (numbering according to Kabat), and in the constant domain CH1 of the first Fab molecule under a) the amino acid at position 147 is substituted by glutamic acid (E) (numbering according to Kabat EU index) and the amino acid at position 213 is substituted by glutamic acid (E) (numbering according to Kabat EU index). Artificial T cell receptors (also known as chimeric T cell receptors, chimeric immunoreceptors, chimeric antigen receptors (CARs)) are engineered receptors, which graft an arbitrary specificity onto an immune effector cell. Typically, these receptors are used to graft the specificity of a monoclonal antibody onto a T cell, with transfer of their coding sequence facilitated by retroviral vectors. In this way, a large number of target- specific T cells can be generated for adoptive cell transfer. Phase I clinical studies of this approach show efficacy. The most common form of these molecules are fusions of single-chain variable fragments (scFv) derived from monoclonal antibodies, fused to CD3-zeta transmembrane and endodomain. Such molecules result in the transmission of a zeta signal in response to recognition by the scFv of its target. An example of such a construct is 14g2a-Zeta, which is a fusion of a scFv derived from hybridoma 14g2a (which recognizes disialoganglioside GD2). When T cells express this molecule (usually achieved by oncoretroviral vector transduction), they recognize and kill target cells that express GD2 (e.g., neuroblastoma cells). To target malignant B cells, investigators have redirected the specificity of T cells using a chimeric immunoreceptor specific for the B-lineage molecule, CD19. The variable portions of an immunoglobulin heavy and light chain are fused by a flexible linker to form a scFv. This scFv is preceded by a signal peptide to direct the nascent protein to the endoplasmic reticulum and subsequent surface expression (this is cleaved). A flexible spacer allows to the scFv to orient in different directions to enable antigen binding. The transmembrane domain is a typical hydrophobic alpha helix usually derived from the original molecule of the signaling endodomain which protrudes into the cell and transmits the desired signal. Type I proteins are in fact two protein domains linked by a transmembrane alpha helix in between. The cell membrane lipid bilayer, through which the transmembrane domain passes, acts to isolate the inside portion (endodomain) from the external portion (ectodomain). It is not so surprising that attaching an ectodomain from one protein to an endodomain of another protein results in a molecule that combines the recognition of the former to the signal of the latter. With regards the ectodomain, a signal peptide directs the nascent protein into the endoplasmic reticulum. This is essential if the receptor is to be glycosylated and anchored in the cell membrane. Any eukaryotic signal peptide sequence usually works fine. Generally, the signal peptide natively attached to the amino-terminal most component is used (e.g., in a scFv with orientation light chain - linker - heavy chain, the native signal of the light-chain is used The antigen recognition domain is usually an scFv. There are however many alternatives. An antigen recognition domain from native T-cell receptor (TCR) alpha and beta single chains have been described, as have simple ectodomains (e.g., CD4 ectodomain torecognize HIV infected cells) and more exotic recognition components such as a linked cytokine (which leads to recognition of cells bearing the cytokine receptor). In fact, almost anything that binds a given target with high affinity can be used as an antigen recognition region. A spacer region links the antigen binding domain to the transmembrane domain. It should be flexible enough to allow the antigen binding domain to orient in different directions to facilitate antigen recognition. The simplest form is the hinge region from IgG1. Alternatives include the CH2CH3 region of immunoglobulin and portions of CD3. For most scFv based constructs, the IgG1 hinge suffices. The transmembrane domain is a hydrophobic alpha helix that spans the membrane. Generally, the transmembrane domain from the most membrane proximal component of the endodomain is used. Interestingly, using the CD3-zeta transmembrane domain may result in incorporation of the artificial TCR into the native TCR a factor that is dependent on the presence of the native CD3-zeta transmembrane charged aspartic acid residue. Different transmembrane domains result in different receptor stability. The CD28 transmembrane domain results in a brightly expressed, stable receptor. The endodomain is the "business-end" of the receptor. After antigen recognition, receptors cluster and a signal is transmitted to the cell. The most commonly used endodomain component is CD3-zeta which contains 3 ITAMs. This transmits an activation signal to the T cell after antigen is bound. CD3-zeta may not provide a fully competent activation signal and additional co-stimulatory signaling is needed. "First-generation" CARs typically had the intracellular domain from the CD3E- chain, which is the primary transmitter of signals from endogenous TCRs. "Second-generation" CARs add intracellular signaling domains from various costimulatory protein receptors (e.g., CD28, 41BB, ICOS) to the cytoplasmic tail of the CAR to provide additional signals to the T cell. Preclinical studies have indicated that the second generation of CAR designs improves the antitumor activity of T cells. More recent, "third-generation" CARs combine multiple signaling domains, such as CD3z-CD28-41BB or CD3z-CD28-OX40, to further augment Antibody Drug Conjugates or ADCs are a new class of highly potent biopharmaceutical drugs designed as a targeted therapy for the treatment of people with infectious disease. ADCs are complex molecules composed of an antibody (a whole mAb or an antibody fragment such as a single-chain variable fragment, or scFv) linked, via a stable chemical linker with labile bonds, to a biological active cytotoxic/anti-viral payload or drug. Antibody Drug Conjugates are examples of bioconjugates and immunoconjugates. By combining the unique targeting capabilities of monoclonal antibodies with the cancer- killing ability of cytotoxic drugs, antibody-drug conjugates allow sensitive discrimination between healthy and diseased tissue. This means that, in contrast to traditional systemic approaches, antibody-drug conjugates target and attack the infected cell so that healthy cells are less severely affected. In the development ADC-based anti-tumour therapies, an anticancer drug (e.g., a cell toxin or cytotoxin) is coupled to an antibody that specifically targets a certain cell marker (e.g., a protein that, ideally, is only to be found in or on infected cells). Antibodies track these proteins down in the body and attach themselves to the surface of cancer cells. The biochemical reaction between the antibody and the target protein (antigen) triggers a signal in the tumour cell, which then absorbs or internalizes the antibody together with the cytotoxin. After the ADC is internalized, the cytotoxic drug is released and kills the cell or impairs viral replication. Due to this targeting, ideally the drug has lower side effects and gives a wider therapeutic window than other agents. A stable link between the antibody and cytotoxic/anti-viral agent is a crucial aspect of an ADC. Linkers are based on chemical motifs including disulfides, hydrazones or peptides (cleavable), or thioethers (noncleavable) and control the distribution and delivery of the cytotoxic agent to the target cell. Cleavable and noncleavable types of linkers have been proven to be safe in preclinical and clinical trials. Brentuximab vedotin includes an enzyme- sensitive cleavable linker that delivers the potent and highly toxic antimicrotubule agent Monomethyl auristatin E or MMAE, a synthetic antineoplastic agent, to human specific CD30-positive malignant cells. Because of its high toxicity MMAE, which inhibits cell division by blocking the polymerization of tubulin, cannot be used as a single-agent chemotherapeutic drug. However, the combination of MMAE linked to an anti-CD30 monoclonal antibody (cAClO, a cell membrane protein of the tumor necrosis factor or TNF receptor) proved to be stable in extracellular fluid, cleavable by cathepsin and safe for therapy. Trastuzumab emtansine, the other approved ADC, is a combination of the microtubule-formation inhibitor mertansine (DM- 1), a derivative of the Maytansine, and antibody trastuzumab (Herceptin®/Genentech/Roche) attached by a stable, non-cleavable linker. The availability of better and more stable linkers has changed the function of the chemical bond. The type of linker, cleavable or noncleavable, lends specific properties to the cytotoxic (anti-cancer) drug. For example, a non-cleavable linker keeps the drug within the cell. As a result, the entire antibody, linker and cytotoxic agent enter the targeted cancer cell where the antibody is degraded to the level of an amino acid. The resulting complex - amino acid, linker and cytotoxic agent - now becomes the active drug. In contrast, cleavable linkers are catalyzed by enzymes in the host cell where it releases the cytotoxic agent. Another type of cleavable linker, currently in development, adds an extra molecule between the cytotoxic/anti-viral drug and the cleavage site. This linker technology allows researchers to create ADCs with more flexibility without worrying about changing cleavage kinetics. Researchers are also developing a new method of peptide cleavage based on Edman degradation, a method of sequencing amino acids in a peptide. Future direction in the development of ADCs also include the development of site-specific conjugation (TDCs) to further improve stability and therapeutic index and a emitting immunoconjugates and antibody-conjugated nanoparticles. Bi-specific T-cell engagers (BiTEs) are a class of artificial bispecific monoclonal antibodies that are investigated for the use as anti-cancer drugs. They direct a host's immune system, more specifically the T cells' cytotoxic activity, against infected cells. BiTE is a registered trademark of Micromet AG. BiTEs are fusion proteins consisting of two single-chain variable fragments (scFvs) of different antibodies, or amino acid sequences from four different genes, on a single peptide chain of about 55 kilodaltons. One of the scFvs binds to T cells via the CD3 receptor, and the other to an infected cell via a specific molecule. Like other bispecific antibodies, and unlike ordinary monoclonal antibodies, BiTEs form a link between T cells and target cells. This causes T cells to exert cytotoxic/anti-viral activity on infected cells by producing proteins like perforin and granzymes, independently of the presence of MHC I or co-stimulatory molecules. These proteins enter infected cells and initiate the cell's apoptosis. This action mimics physiological processes observed during T cell attacks against infected cells. In a particular embodiment, the antibody is a recombinant antibody that is suitable for action inside of a cell - such antibodies are known as "intrabodies." These antibodies may interfere with target function by a variety of mechanism, such as by altering intracellular protein trafficking, interfering with enzymatic function, and blocking protein-protein or protein-DNA interactions. In many ways, their structures mimic or parallel those of single chain and single domain antibodies, discussed above. Indeed, single-transcript/single-chain is an important feature that permits intracellular expression in a target cell, and also makes protein transit across cell membranes more feasible. However, additional features are required. The two major issues impacting the implementation of intrabody therapeutic are delivery, including cell/tissue targeting, and stability. With respect to delivery, a variety of approaches have been employed, such as tissue-directed delivery, use of cell-type specific promoters, viral-based delivery and use of cell-permeability/membrane translocating peptides. With respect to the stability, the approach is generally to either screen by brute force, including methods that involve phage display and may include sequence maturation or development of consensus sequences, or more directed modifications such as insertion stabilizing sequences (e.g., Fc regions, chaperone protein sequences, leucine zippers) and disulfide replacement/modification. An additional feature that intrabodies may require is a signal for intracellular targeting. Vectors that can target intrabodies (or other proteins) to subcellular regions such as the cytoplasm, nucleus, mitochondria and ER have been designed and are commercially available (Invitrogen Corp.; Persic et al., 1997). By virtue of their ability to enter cells, intrabodies have additional uses that other types of antibodies may not achieve. In the case of the present antibodies, the ability to interact with the MUCl cytoplasmic domain in a living cell may interfere with functions associated with the MUCl CD, such as signalling functions (binding to other molecules) or oligomer formation. In particular, it is contemplated that such antibodies can be used to inhibit MUC1 dimer formation. In certain embodiments, the antibodies of the present disclosure may be purified. The term "purified," as used herein, is intended to refer to a composition, isolatable from other components, wherein the protein is purified to any degree relative to its naturally-obtainable state. A purified protein therefore also refers to a protein, free from the environment in which it may naturally occur. Where the term "substantially purified" is used, this designation will refer to a composition in which the protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the proteins in the composition. Protein purification techniques are well known to those of skill in the art. These techniques involve, at one level, the crude fractionation of the cellular milieu to polypeptide and non- polypeptide fractions. Having separated the polypeptide from other proteins, the polypeptide of interest may be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or purification to homogeneity). Analytical methods particularly suited to the preparation of a pure peptide are ion-exchange chromatography, exclusion chromatography; polyacrylamide gel electrophoresis; isoelectric focusing. Other methods for protein purification include, precipitation with ammonium sulfate, PEG, antibodies and the like or by heat denaturation, followed by centrifugation; gel filtration, reverse phase, hydroxylapatite and affinity chromatography; and combinations of such and other techniques. In purifying an antibody of the present disclosure, it may be desirable to express the polypeptide in a prokaryotic or eukaryotic expression system and extract the protein using denaturing conditions. The polypeptide may be purified from other cellular components using an affinity column, which binds to a tagged portion of the polypeptide. As is generally known in the art, it is believed that the order of conducting the various purification steps may be changed, or that certain steps may be omitted, and still result in a suitable method for the preparation of a substantially purified protein or peptide. Commonly, complete antibodies are fractionated utilizing agents (i.e., protein A) that bind the Fc portion of the antibody. Alternatively, antigens may be used to simultaneously purify and select appropriate antibodies. Such methods often utilize the selection agent bound to a support, such as a column, filter or bead. The antibodies are bound to a support, contaminants removed (e.g., washed away), and the antibodies released by applying conditions (salt, heat, etc.). Various methods for quantifying the degree of purification of the protein or peptide will be known to those of skill in the art in light of the present disclosure. These include, for example, determining the specific activity of an active fraction, or assessing the amount of polypeptides within a fraction by SDS/PAGE analysis. Another method for assessing the purity of a fraction is to calculate the specific activity of the fraction, to compare it to the specific activity of the initial extract, and to thus calculate the degree of purity. The actual units used to represent the amount of activity will, of course, be dependent upon the particular assay technique chosen to follow the purification and whether or not the expressed protein or peptide exhibits a detectable activity. It is known that the migration of a polypeptide can vary, sometimes significantly, with different conditions of SDS/PAGE (Capaldi et al., 1977). It will therefore be appreciated that under differing electrophoresis conditions, the apparent molecular weights of purified or partially purified expression products may vary. The present disclosure provides pharmaceutical compositions comprising anti-SARS- CoV- 2 virus antibodies and antigens for generating the same. Such compositions comprise a prophylactically or therapeutically effective amount of an antibody or a fragment thereof, or a peptide immunogen, and a pharmaceutically acceptable carrier.The compositions of the invention may include an "effective amount" or "therapeutically effective amount" or a "prophylactically effective amount" of an antibody or antigen-binding portion of the invention. These terms are used interchangeably. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result. A therapeutically effective amount of the antibody or antibody portion may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the antibody or antibody portion to elicit a desired response in the subject. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. . As used herein, the term “treat”, “treating" or "treatment" of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment “treat”, "treating" or "treatment" refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient. In yet another embodiment, “treat”, "treating" or "treatment" refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In yet another embodiment, “treat”, "treating" or "treatment" refers to preventing or delaying the onset or development or progression of the disease or disorder. "Prevention" of a condition or disorder refers to delaying or preventing the onset of a condition or disorder or reducing its severity, as assessed by the appearance or extent of one or more symptoms of the condition or disorder. As used herein, the term “subject” refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human. As used herein, a subject is “in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment. As used herein, the term "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavouring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp.1289- 1329). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.. The term "carrier" refers to a diluent, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a particular carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Other suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatine, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical agents are described in "Remington's Pharmaceutical Sciences." Such compositions will contain a prophylactically or therapeutically effective amount of the antibody or fragment thereof, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration, which can be oral, intravenous, intraarterial, intrabuccal, intranasal, nebulized, bronchial inhalation, intra-rectal, vaginal, topical or delivered by mechanical ventilation. Active vaccines are also envisioned where antibodies like those disclosed are produced in vivo in a subject at risk of SARS-CoV-2 infection. Such vaccines can be formulated for parenteral administration, e.g., formulated for injection via the intradermal, intravenous, intramuscular, subcutaneous, or even intraperitoneal routes. Administration by intradermal and intramuscular routes are contemplated. The vaccine could alternatively be administered by a topical route directly to the mucosa, for example, by nasal drops, inhalation, by nebulizer, or via intrarectal or vaginal delivery. Pharmaceutically acceptable salts include the acid salts and those which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like. Passive transfer of antibodies, known as artificially acquired passive immunity, generally will involve the use of intravenous or intramuscular injections. The forms of antibody can be human or animal blood plasma or serum, as pooled human immunoglobulin for intravenous (IVIG) or intramuscular (IG) use, as high-titre human IVIG or IG from immunized or from donors recovering from disease, and as monoclonal antibodies (MAb). Such immunity generally lasts for only a short period of time, and there is also a potential risk for hypersensitivity reactions, and serum sickness, especially from gamma globulin of non- human origin. However, passive immunity provides immediate protection. The antibodies will be formulated in a carrier suitable for injection, i.e., sterile and syringeable. Generally, the ingredients of compositions of the disclosure are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water- free concentrate in a hermetically sealed container such as an ampoule or sachet indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration. The compositions of the disclosure can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc. Antibody-dependent cell-mediated cytotoxicity (ADCC) is an immune mechanism leading to the lysis of antibody-coated target cells by immune effector cells. The target cells are cells to which antibodies or fragments thereof comprising an Fc region specifically bind, generally via the protein part that is N-terminal to the Fc region. By "antibody having increased/reduced antibody dependent cell-mediated cytotoxicity (ADCC)" is meant an antibody having increased/reduced ADCC as determined by any suitable method known to those of ordinary skill in the art. As used herein, the term "increased/reduced ADCC" is defined as either an increase/reduction in the number of target cells that are lysed in a given time, at a given concentration of antibody in the medium surrounding the target cells, by the mechanism of ADCC defined above, and/or a reduction/increase in the concentration of antibody, in the medium surrounding the target cells, required to achieve the lysis of a given number of target cells in a given time, by the mechanism of ADCC. The increase/reduction in ADCC is relative to the ADCC mediated by the same antibody produced by the same type of host cells, using the same standard production, purification, formulation and storage methods (which are known to those skilled in the art), but that has not been engineered. For example, the increase in ADCC mediated by an antibody produced by host cells engineered to have an altered pattern of glycosylation (e.g., to express the glycosyltransferase, GnTIII, or other glycosyltransferases) by the methods described herein, is relative to the ADCC mediated by the same antibody produced by the same type of non-engineered host cells. Complement-dependent cytotoxicity (CDC) is a function of the complement system. It is the processes in the immune system that kill pathogens by damaging their membranes without the involvement of antibodies or cells of the immune system. There are three main processes. All three insert one or more membrane attack complexes (MAC) into the pathogen which cause lethal colloid-osmotic swelling, i.e., CDC. It is one of the mechanisms by which antibodies or antibody fragments have an anti-viral effect. Antibodies of the present disclosure may be linked to at least one agent to form an antibody conjugate. In order to increase the efficacy of antibody molecules as diagnostic or therapeutic agents, it is conventional to link or covalently bind or complex at least one desired molecule or moiety. Such a molecule or moiety may be, but is not limited to, at least one effector or reporter molecule. Effector molecules comprise molecules having a desired activity, e.g., cytotoxic activity. Non-limiting examples of effector molecules which have been attached to antibodies include toxins, anti-tumour agents, therapeutic enzymes, radionuclides, antiviral agents, chelating agents, cytokines, growth factors, and oligo- or polynucleotides. By contrast, a reporter molecule is defined as any moiety which may be detected using an assay. Non- limiting examples of reporter molecules which have been conjugated to antibodies include enzymes, radiolabels, haptens, fluorescent labels, phosphorescent molecules chemiluminescent molecules, chromophores, photoaffinity molecules, colored particles or ligands, such as biotin. Antibody conjugates are generally preferred for use as diagnostic agents. Antibody diagnostics generally fall within two classes, those for use in in vitro diagnostics, such as in a variety of immunoassays, and those for use in vivo diagnostic protocols, generally known as "antibody-directed imaging." Many appropriate imaging agents are known in the art, as are methods for their attachment to antibodies (see, for e.g., U.S. Patents 5,021,236, 4,938,948, and 4,472,509). The imaging moieties used can be paramagnetic ions, radioactive isotopes, fluorochromes, NMR-detectable substances, and X-ray imaging agents. In the case of paramagnetic ions, one might mention by way of example ions such as chromium (III), manganese (II), iron (III), iron (II), cobalt (II), nickel (II), copper (II), neodymium (III), samarium (III), ytterbium (III), gadolinium (III), vanadium (II), terbium (III), dysprosium (III), holmium (III) and/or erbium (III), with gadolinium being particularly preferred. Ions useful in other contexts, such as X-ray imaging, include but are not limited to lanthanum (III), gold (III), lead (II), and especially bismuth (III). In the case of radioactive isotopes for therapeutic and/or diagnostic application, one might mention astatine211, 14carbon, 51chromium, 36chlorine, 57cobalt, 58cobalt, copper67, 152Eu, gallium67, 3hydrogen, iodine123, iodine125, iodine131, indium111, 59iron, 32phosphorus, rhenium186, rhenium188, 75selenium, 35sulphur, technicium99m and/or yttrium90.125I is often being preferred for use in certain embodiments, and technicium99m and/or indium111 are also often preferred due to their low energy and suitability for long range detection. Radioactively labelled monoclonal antibodies of the present disclosure may be produced according to well-known methods in the art. For instance, monoclonal antibodies can be iodinated by contact with sodium and/or potassium iodide and a chemical oxidizing agent such as sodium hypochlorite, or an enzymatic oxidizing agent, such as lactoperoxidase. Monoclonal antibodies according to the disclosure may be labelled with technetium99m by ligand exchange process, for example, by reducing pertechnate with stannous solution, chelating the reduced technetium onto a Sephadex column and applying the antibody to this column. Alternatively, direct labelling techniques may be used, e.g., by incubating pertechnate, a reducing agent such as SnCl2, a buffer solution such as sodium- potassium phthalate solution, and the antibody. Intermediary functional groups which are often used to bind radioisotopes which exist as metallic ions to antibody are diethylenetriaminepentaacetic acid (DTPA) or ethylene diaminetetracetic acid (EDTA). Among the fluorescent labels contemplated for use as conjugates include Alexa 350, Alexa 430, AMCA, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, Cascade Blue, Cy3, Cy5,6-FAM, Fluorescein Isothiocyanate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rhodamine Green, Rhodamine Red, Renographin, ROX, TAMRA, TET, Tetramethylrhodamine, and/or Texas Red. Additional types of antibodies contemplated in the present disclosure are those intended primarily for use in vitro, where the antibody is linked to a secondary binding ligand and/or to an enzyme (an enzyme tag) that will generate a coloured product upon contact with a chromogenic substrate. Examples of suitable enzymes include urease, alkaline phosphatase, (horseradish) hydrogen peroxidase or glucose oxidase. Preferred secondary binding ligands are biotin and avidin and streptavidin compounds. The use of such labels is well known to those of skill in the art and are described, for example, in U.S. Patents 3,817,837, 3,850,752, 3,939,350, 3,996,345, 4,277,437, 4,275,149 and 4,366,241. Yet another known method of site-specific attachment of molecules to antibodies comprises the reaction of antibodies with hapten-based affinity labels. Essentially, hapten-based affinity labels react with amino acids in the antigen binding site, thereby destroying this site and blocking specific antigen reaction. However, this may not be advantageous since it results in loss of antigen binding by the antibody conjugate. Molecules containing azido groups may also be used to form covalent bonds to proteins through reactive nitrene intermediates that are generated by low intensity ultraviolet light (Potter and Haley, 1983). In particular, 2- and 8-azido analogues of purine nucleotides have been used as site-directed photoprobes to identify nucleotide binding proteins in crude cell extracts (Owens & Haley, 1987; Atherton et al., 1985). The 2- and 8-azido nucleotides have also been used to map nucleotide binding domains of purified proteins (Khatoon et al., 1989; King et al., 1989; Dholakia et al., 1989) and may be used as antibody binding agents. Several methods are known in the art for the attachment or conjugation of an antibody to its conjugate moiety. Some attachment methods involve the use of a metal chelate complex employing, for example, an organic chelating agent such a diethylenetriaminepentaacetic acid anhydride (DTPA); ethylenetriaminetetraacetic acid; N-chloro-p-toluenesulfonamide; and/or tetrachloro-3a-6a-diphenylglycouril-3 attached to the antibody (U.S. Patents 4,472,509 and 4,938,948). Monoclonal antibodies may also be reacted with an enzyme in the presence of a coupling agent such as glutaraldehyde or periodate. Conjugates with fluorescein markers are prepared in the presence of these coupling agents or by reaction with an isothiocyanate. In U.S. Patent 4,938,948, imaging of breast tumours is achieved using monoclonal antibodies and thedetectable imaging moieties are bound to the antibody using linkers such as methyl-p- hydroxybenzimidate or N-succinimidy1-3-(4- hydroxypheny1)propionate. In other embodiments, derivatization of immunoglobulins by selectively introducing sulfhydryl groups in the Fc region of an immunoglobulin, using reaction conditions that do not alter the antibody combining site are contemplated. Antibody conjugates produced according to this methodology are disclosed to exhibit improved longevity, specificity and sensitivity (U.S. Patent 5,196,066, incorporated herein by reference). Site-specific attachment of effector or reporter molecules, wherein the reporter or effector molecule is conjugated to a carbohydrate residue in the Fc region have also been disclosed in the literature (0' Shannessy et al., 1987). This approach has been reported to produce diagnostically and therapeutically promising antibodies which are currently in clinical evaluation. In still further embodiments, the present disclosure concerns immunodetection methods for binding, purifying, removing, quantifying and otherwise generally detecting SARS-CoV- 2 and its associated antigens. While such methods can be applied in a traditional sense, another use will be in quality control and monitoring of vaccine and other virus stocks, where antibodies according to the present disclosure can be used to assess the amount or integrity (i.e., long term stability) of antigens in viruses. Alternatively, the methods may be used to screenmvarious antibodies for appropriate/desired reactivity profiles. Other immunodetection methods include specific assays for determining the presence of SARS-CoV-2 in a subject. A wide variety of assay formats are contemplated, but specifically those that would be used to detect SARS-CoV-2 in a fluid obtained from a subject, such as saliva, blood, plasma, sputum, semen or urine. In particular, semen has been demonstrated as a viable sample for detecting SARS-CoV-2 (Purpura et al., 2016; Mansuy et al., 2016; Barzon et al., 2016; Garnet et al., 2016; Duffy et al., 2009; CDC, 2016; Halfon et al., 2010; Elder et al. 2005). The assays may be advantageously formatted for non- healthcare (home) use, including lateral flow assays (see below) analogous to home pregnancy tests. These assays may be packaged in the form of a kit with appropriate reagents and instructions to permit use by the subject of a family member. Some immunodetection methods include enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoradiometric assay, fluoroimmunoassay, chemiluminescent assay, bioluminescent assay, and Western blot to mention a few. In particular, a competitive assay for the detection and quantitation of SARS-CoV-2 antibodies directed to specific parasite epitopes in samples also is provided. The steps of various useful immunodetection methods have been described in the scientific literature, such as, e.g., Doolittle and Ben-Zeev (1999), Gulbis and Galand (1993), De Jager et al. (1993), and Nakamura et al. (1987). In general, the immunobinding methods include obtaining a sample suspected of containing SARS-CoV-2, and contacting the sample with a first antibody in accordance with the present disclosure, as the case may be, under conditions effective to allow the formation of immunocomplexes. These methods include methods for purifying SARS-CoV-2 or related antigens from a sample. The antibody will preferably be linked to a solid support, such as in the form of a column matrix, and the sample suspected of containing the SARS-CoV-2 or antigenic component will be applied to the immobilized antibody. The unwanted components will be washed from the column, leaving the SARS-CoV-2 antigen immunocomplexed to the immobilized antibody, which is then collected by removing the organism or antigen from the column. The immunobinding methods also include methods for detecting and quantifying the amount of SARS-CoV-2 or related components in a sample and the detection and quantification of any immune complexes formed during the binding process. Here, one would obtain a sample suspected of containing SARS-CoV-2 or its antigens and contact the sample with an antibody that binds SARS-CoV-2 or components thereof, followed by detecting and quantifying the amount of immune complexes formed under the specific conditions. In terms of antigen detection, the biological sample analyzed may be any sample that is suspected of containing SARS-CoV-2 or SARS-CoV-2 antigen, such as a tissue section or specimen, a homogenized tissue extract, a biological fluid, including blood and serum, or a secretion, such as faeces or urine. Contacting the chosen biological sample with the antibody under effective conditions and for a period of time sufficient to allow the formation of immune complexes (primary immune complexes) is generally a matter of simply adding the antibody composition to the sample and incubating the mixture for a period of time long enough for the antibodies to form immune complexes with, i.e., to bind to SARS-CoV-2 or antigens present. After this time, the sample-antibody composition, such as a tissue section, ELISA plate, dot blot or Western blot, will generally be washed to remove any non-specifically bound antibody species, allowing only those antibodies specifically bound within the primary immune complexes to be detected. In general, the detection of immunocomplex formation is well known in the art and may be achieved through the application of numerous approaches. These methods are based upon the detection of a label or marker, such as any of those radioactive, fluorescent, biological and enzymatic tags. Patents concerning the use of such labels include U.S. Patents 3,817,837, 3,850,752, 3,939,350, 3,996,345, 4,277,437, 4,275,149 and 4,366,241. Of course one may find additional advantages through the use of a secondary binding ligand such as a second antibody and/or a biotin/avidin ligand binding arrangement, as is known in the art. The antibody employed in the detection may itself be linked to a detectable label, wherein one would then simply detect this label, thereby allowing the amount of the primary immune complexes in the composition to be determined. Alternatively, the first antibody that becomes bound within the primary immune complexes may be detected by means of a second binding ligand that has binding affinity for the antibody. In these cases, the second binding ligand may be linked to a detectable label. The second binding ligand is itself often an antibody, which may thus be termed a "secondary" antibody. The primary immune complexes are contacted with the labelled, secondary binding ligand, or antibody, under effective conditions and for a period of time sufficient to allow the formation of secondary immune complexes. The secondary immune complexes are then generally washed to remove any non-specifically bound labelled secondary antibodies or ligands, and the remaining label in the secondary immune complexes is then detected. Further methods include the detection of primary immune complexes by a two-step approach. A second binding ligand, such as an antibody that has binding affinity for the antibody, is used to form secondary immune complexes, as described above. After washing, the secondary immune complexes are contacted with a third binding ligand or antibody that has binding affinity for the second antibody, again under effective conditions and for a period of time sufficient to allow the formation of immune complexes (tertiary immune complexes). The third ligand or antibody is linked to a detectable label, allowing detection of the tertiary immune complexes thus formed. This system may provide for signal amplification if this is desired. One method of immunodetection uses two different antibodies. A first biotinylated antibody is used to detect the target antigen, and a second antibody is then used to detect the biotin attached to the complexed biotin. In that method, the sample to be tested is first incubated in a solution containing the first step antibody. If the target antigen is present, some of the antibody binds to the antigen to form a biotinylated antibody/antigen complex. The antibody/antigen complex is then amplified by incubation in successive solutions of streptavidin (or avidin), biotinylated DNA, and/or complementary biotinylated DNA, with each step adding additional biotin sites to the antibody/antigen complex. The amplification steps are repeated until a suitable level of amplification is achieved, at which point the sample is incubated in a solution containing the second step antibody against biotin. This second step antibody is labelled, for example, with an enzyme that can be used to detect the presence of the antibody/antigen complex by histoenzymology using a chromogen substrate. With suitable amplification, a conjugate can be produced which is macroscopically visible. Another known method of immunodetection takes advantage of the immuno-PCR (Polymerase Chain Reaction) methodology. The PCR method is similar to the Cantor method up to the incubation with biotinylated DNA, however, instead of using multiple rounds of streptavidin and biotinylated DNA incubation, the DNA/biotin/streptavidin/antibody complex is washed out with a low pH or high salt buffer that releases the antibody. The resulting wash solution is then used to carry out a PCR reaction with suitable primers with appropriate controls. At least in theory, the enormous amplification capability and specificity of PCR can be utilized to detect a single antigen molecule. Immunoassays, in their most simple and direct sense, are binding assays. Certain preferred immunoassays are the various types of enzyme linked immunosorbent assays (ELISAs) and radioimmunoassays (RIA) known in the art. Immunohistochemical detection using tissue sections is also particularly useful. However, it will be readily appreciated that detection is not limited to such techniques, and western blotting, dot blotting, FACS analyses, and the like may also be used. In one exemplary ELISA, the antibodies of the disclosure are immobilized onto a selected surface exhibiting protein affinity, such as a well in a polystyrene microtiter plate. Then, a test composition suspected of containing the SARS-CoV-2 or SARS-CoV-2 antigen is added to the wells. After binding and washing to remove non-specifically bound immune complexes, the bound antigen may be detected. Detection may be achieved by the addition of another anti-SARS-CoV-2 antibody that is linked to a detectable label. This type of ELISA is a simple "sandwich ELISA." Detection may also be achieved by the addition of a second anti-SARS-CoV-2 antibody, followed by the addition of a third antibody that has binding affinity for the second antibody, with the third antibody being linked to a detectable label. In another exemplary ELISA, the samples suspected of containing the SARS-Co V-2 or SARS-CoV-2 antigen are immobilized onto the well surface and then contacted with the anti- SARS-CoV-2 antibodies of the disclosure. After binding and washing to remove non- specifically bound immune complexes, the bound anti-SARS-CoV-2 antibodies are detected. Where the initial anti-SARS-CoV-2 antibodies are linked to a detectable label, the immune complexes may be detected directly. Again, the immune complexes may be detected using a second antibody that has binding affinity for the first anti-SARS-CoV-2 antibody, with the second antibody being linked to a detectable label. Irrespective of the format employed, ELISAs have certain features in common, such as coating, incubating and binding, washing to remove non-specifically bound species, and detecting the bound immune complexes. These are described below. In coating a plate with either antigen or antibody, one will generally incubate the wells of the plate with a solution of the antigen or antibody, either overnight or for a specified period of hours. The wells of the plate will then be washed to remove incompletely adsorbed material. Any remaining available surfaces of the wells are then "coated" with a nonspecific protein that is antigenically neutral with regard to the test antisera. These include bovine serum albumin (BSA), casein or solutions of milk powder. The coating allows for blocking of nonspecific adsorption sites on the immobilizing surface and thus reduces the background caused by nonspecific binding of antisera onto the surface. In ELISAs, it is probably more customary to use a secondary or tertiary detection means rather than a direct procedure. Thus, after binding of a protein or antibody to the well, coating with a non-reactive material to reduce background, and washing to remove unbound material, the immobilizing surface is contacted with the biological sample to be tested under conditions effective to allow immune complex (antigen/antibody) formation. Detection of the immune complex then requires a labelled secondary binding ligand or antibody, and a secondary binding ligand or antibody in conjunction with a labelled tertiary antibody or a third binding ligand. "Under conditions effective to allow immune complex (antigen/antibody) formation" means that the conditions preferably include diluting the antigens and/or antibodies with solutions such as BSA, bovine gamma globulin (BGG) or phosphate buffered saline (PBS)/Tween. These added agents also tend to assist in the reduction of nonspecific background. The "suitable" conditions also mean that the incubation is at a temperature or for a period of time sufficient to allow effective binding. Incubation steps are typically from about 1 to 2 to 4 hours or so, at temperatures preferably on the order of 25°C to 27°C, or may be overnight at about 4°C or so. Following all incubation steps in an ELISA, the contacted surface is washed so as to remove non-complexed material. A preferred washing procedure includes washing with a solution such as PBS/Tween, or borate buffer. Following the formation of specific immune complexes between the test sample and the originally bound material, and subsequent washing, the occurrence of even minute amounts of immune complexes may be determined. To provide a detecting means, the second or third antibody will have an associated label to allow detection. Preferably, this will be an enzyme that will generate colour development upon incubating with an appropriate chromogenic substrate. Thus, for example, one will desire to contact or incubate the first and second immune complex with a urease, glucose oxidase, alkaline phosphatase or hydrogen peroxidase-conjugated antibody for a period of time and under conditions that favour the development of further immune complex formation (e.g., incubation for 2 hours at room temperature in a PBS-containing solution such as PBS- Tween). After incubation with the labelled antibody, and subsequent to washing to remove unbound material, the amount of label is quantified, e.g., by incubation with a chromogenic substrate such as urea, or bromocresol purple, or 2,2'-azino-di-(3-ethyl-benzthiazoline-6- sulfonic acid (ABTS), or H2O2, in the case of peroxidase as the enzyme label. Quantification is then achieved by measuring the degree of color generated, e.g., using a visible spectra spectrophotometer. In another embodiment, the present disclosure contemplates the use of competitive formats. This is particularly useful in the detection of SARS-CoV-2 antibodies in sample. In competition-based assays, an unknown amount of analyte or antibody is determined by its ability to displace a known amount of labelled antibody or analyte. Thus, the quantifiable loss of a signal is an indication of the amount of unknown antibody or analyte in a sample. Here, the inventor proposes the use of labelled SARS-CoV-2 monoclonal antibodies to determine the amount of SARS-CoV-2 antibodies in a sample. The basic format would include contacting a known amount of SARS-CoV-2 monoclonal antibody (linked to a detectable label) with SARS-CoV-2 antigen or particle. The SARS-CoV-2 antigen or organism is preferably attached to a support. After binding of the labelled monoclonal antibody to the support, the sample is added and incubated under conditions permitting any unlabelled antibody in the sample to compete with, and hence displace, the labelled monoclonal antibody. By measuring either the lost label or the label remaining (and subtracting that from the original amount of bound label), one can determine how much non-labelled antibody is bound to the support, and thus how much antibody was present in the sample. The Western blot (alternatively, protein immunoblot) is an analytical technique used to detect specific proteins in a given sample of tissue homogenate or extract. It uses gel electrophoresis to separate native or denatured proteins by the length of the polypeptide (denaturing conditions) or by the 3-D structure of the protein (native/ non-denaturing conditions). The proteins are then transferred to a membrane (typically nitrocellulose or PVDF), where they are probed (detected) using antibodies specific to the target protein. Samples may be taken from whole tissue or from cell culture. In most cases, solid tissues are first broken down mechanically using a blender (for larger sample volumes), using a homogenizer (smaller volumes), or by sonication. Cells may also be broken open by one of the above mechanical methods. However, it should be noted that bacteria, virus or environmental samples can be the source of protein and thus Western blotting is not restricted to cellular studies only. Assorted detergents, salts, and buffers may be employed to encourage lysis of cells and to solubilize proteins. Protease and phosphatase inhibitors are often added to prevent the digestion of the sample by its own enzymes. Tissue preparation is often done at cold temperatures to avoid protein denaturing. The proteins of the sample are separated using gel electrophoresis. Separation of proteins may be by isoelectric point (pl), molecular weight, electric charge, or a combination of these factors. The nature of the separation depends on the treatment of the sample and the nature of the gel. This is a very useful way to determine a protein. It is also possible to use a two- dimensional (2-D) gel which spreads the proteins from a single sample out in two dimensions. Proteins are separated according to isoelectric point (pH at which they have neutral net charge) in the first dimension, and according to their molecular weight in the second dimension. In order to make the proteins accessible to antibody detection, they are moved from within the gel onto a membrane made of nitrocellulose or polyvinylidene difluoride (PVDF). The membrane is placed on top of the gel, and a stack of filter papers placed on top of that. The entire stack is placed in a buffer solution which moves up the paper by capillary action, bringing the proteins with it. Another method for transferring the proteins is called electroblotting and uses an electric current to pull proteins from the gel into the PVDF or nitrocellulose membrane. The proteins move from within the gel onto the membrane while maintaining the organization they had within the gel. As a result of this blotting process, the proteins are exposed on a thin surface layer for detection (see below). Both varieties of membrane are chosen for their non- specific protein binding properties (i.e., binds all proteins equally well). Protein binding is based upon hydrophobic interactions, as well as charged interactions between the membrane and protein. Nitrocellulose membranes are cheaper than PVDF but are far more fragile and do not stand up well to repeated probing. The uniformity and overall effectiveness of transfer of protein from the gel to the membrane can be checked by staining the membrane with Coomassie Brilliant Blue or Ponceau S dyes. Once transferred, proteins are detected using labelled primary antibodies, or unlabelled primary antibodies followed by indirect detection using labelled protein A or secondary labelled antibodies binding to the Fc region of the primary antibodies. Lateral flow assays, also known as lateral flow immunochromatographic assays, are simple devices intended to detect the presence (or absence) of a target analyte in sample (matrix) without the need for specialized and costly equipment, though many laboratory-based applications exist that are supported by reading equipment. Typically, these tests are used as low resources medical diagnostics, either for home testing, point of care testing, or laboratory use. A widely spread and well-known application is the home pregnancy test. The technology is based on a series of capillary beds, such as pieces of porous paper or sintered polymer. Each of these elements has the capacity to transport fluid (e.g., urine) spontaneously. The first element (the sample pad) acts as a sponge and holds an excess of sample fluid. Once soaked, the fluid migrates to the second element (conjugate pad) in which the manufacturer has stored the so-called conjugate, a dried format of bio-active particles (see below) in a salt-sugar matrix that contains everything to guarantee an optimized chemical reaction between the target molecule (e.g., an antigen) and its chemical partner (e.g., antibody) that has been immobilized on the particle's surface. While the sample fluid dissolves the salt- sugar matrix, it also dissolves the particles and in one combined transport action the sample and conjugate mix while flowing through the porous structure. In this way, the analyte binds to the particles while migrating further through the third capillary bed. This material has one or more areas (often called stripes) where a third molecule has been immobilized by the manufacturer. By the time the sample-conjugate mix reaches these strips, analyte has been bound on the particle and the third 'capture' molecule binds the complex. After a while, when more and more fluid has passed the stripes, particles accumulate and the stripe-area changes colour. Typically, there are at least two stripes: one (the control) that captures any particle and thereby shows that reaction conditions and technology worked fine, the second contains a specific capture molecule and only captures those particles onto which an analyte molecule has been immobilized. After passing these reaction zones, the fluid enters the final porous material- the wick - that simply acts as a waste container. Lateral Flow Tests can operate as either competitive or sandwich assays. Lateral flow assays are disclosed in U.S. Patent 6,485,982. The antibodies of the present disclosure may also be used in conjunction with both fresh- frozen and/or formalin-fixed, paraffin-embedded tissue blocks prepared for study by immunohistochemistry (IHC). The method of preparing tissue blocks from these particulate specimens has been successfully used in previous IHC studies of various prognostic factors and is well known to those of skill in the art (Brown et al., 1990; Abbondanzo et al., 1990; Allred et al., 1990). Briefly, frozen-sections may be prepared by rehydrating 50 mg of frozen "pulverized" tissue at room temperature in phosphate buffered saline (PBS) in small plastic capsules; pelleting the particles by centrifugation; resuspending them in a viscous embedding medium (OCT); inverting the capsule and/or pelleting again by centrifugation; snap-freezing in -70°C isopentane; cutting the plastic capsule and/or removing the frozen cylinder of tissue; securing the tissue cylinder on a cryostat microtome chuck; and/or cutting 25-50 serial sections from the capsule. Alternatively, whole frozen tissue samples may be used for serial section cuttings. Permanent-sections may be prepared by a similar method involving rehydration of the 50 mg sample in a plastic microfuge tube; pelleting; resuspending in 10% formalin for 4 hours fixation; washing/pelleting; resuspending in warm 2.5% agar; pelleting; cooling in ice water to harden the agar; removing the tissue/agar block from the tube; infiltrating and/or embedding the block in paraffin; and/or cutting up to 50 serial permanent sections. Again, whole tissue samples may be substituted. In still further embodiments, the present disclosure concerns immunodetection kits for use with the immunodetection methods described above. As the antibodies may be used to detect SARS-CoV-2 or SARS-CoV-2 antigens, the antibodies may be included in the kit. The immunodetection kits will thus comprise, in suitable container means, a first antibody that binds to SARS-CoV-2 or SARS-CoV-2 antigen, and optionally an immunodetection reagent. In certain embodiments, the SARS-CoV-2 antibody may be pre-bound to a solid support, such as a column matrix and/or well of a microtiter plate. The immunodetection reagents of the kit may take any one of a variety of forms, including those detectable labels that are associated with or linked to the given antibody. Detectable labels that are associated with or attached to a secondary binding ligand are also contemplated. Exemplary secondary ligands are those secondary antibodies that have binding affinity for the first antibody. Further suitable immunodetection reagents for use in the present kits include the two- component reagent that comprises a secondary antibody that has binding affinity for the first antibody, along with a third antibody that has binding affinity for the second antibody, the third antibody being linked to a detectable label. As noted above, a number of exemplary labels are known in the art and all such labels may be employed in connection with the present disclosure. The kits may further comprise a suitably aliquoted composition of the SARS-CoV-2 or SARS-CoV-2 antigens, whether labelled or unlabelled, as may be used to prepare a standard curve for a detection assay. The kits may contain antibody-label conjugates either in fully conjugated form, in the form of intermediates, or as separate moieties to be conjugated by the user of the kit. The components of the kits may be packaged either in aqueous media or in lyophilized form. The container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which the antibody may be placed, or preferably, suitably aliquoted. The kits of the present disclosure will also typically include a means for containing the antibody, antigen, and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow-moulded plastic containers into which the desired vials are retained. The present disclosure also contemplates the use of antibodies and antibody fragments as described herein for use in assessing the antigenic integrity of a viral antigen in a sample. Biological medicinal products like vaccines differ from chemical drugs in that they cannot normally be characterized molecularly; antibodies are large molecules of significant complexity and have the capacity to vary widely from preparation to preparation. They are also administered to healthy individuals, including children at the start of their lives, and thus a strong emphasis must be placed on their quality to ensure, to the greatest extent possible, that they are efficacious in preventing or treating life-threatening disease, without themselves causing harm. The increasing globalization in the production and distribution of vaccines has opened new possibilities to better manage public health concerns but has also raised questions about the equivalence and interchangeability of vaccines procured across a variety of sources. International standardization of starting materials, of production and quality control testing, and the setting of high expectations for regulatory oversight on the way these products are manufactured and used, have thus been the cornerstone for continued success. But it remains a field in constant change, and continuous technical advances in the field offer a promise of developing potent new weapons against the oldest public health threats, as well as new ones - malaria, pandemic influenza, and HIV, to name a few - but also put a great pressure on manufacturers, regulatory authorities, and the wider medical community to ensure that products continue to meet the highest standards of quality attainable. Thus, one may obtain an antigen or vaccine from any source or at any point during a manufacturing process. The quality control processes may therefore begin with preparing a sample for an immunoassay that identifies binding of an antibody or fragment disclosed herein to a viral antigen. Such immunoassays are disclosed elsewhere in this document, and any of these may be used to assess the structural/antigenic integrity of the antigen. Standards for finding the sample to contain acceptable amounts of antigenically correct and intact antigen may be established by regulatory agencies. Another important embodiment where antigen integrity is assessed is in determining shelf- life and storage stability. Most medicines, including vaccines, can deteriorate over time. Therefore, it is critical to determine whether, over time, the degree to which an antigen, such as in a vaccine, degrades or destabilizes such that is it no longer antigenic and/or capable of generating an immune response when administered to a subject. Again, standards for finding the sample to contain acceptable amounts of antigenically intact antigen may be established by regulatory agencies. In certain embodiments, viral antigens may contain more than one protective epitope. In these cases, it may prove useful to employ assays that look at the binding of more than one antibody, such as 2, 3, 4, 5 or even more antibodies. These antibodies bind to closely related epitopes, such that they are adjacent or even overlap each other. On the other hand, they may represent distinct epitopes from disparate parts of the antigen. By examining the integrity of multiple epitopes, a more complete picture of the antigen's overall integrity, and hence ability to generate a protective immune response, may be determined. Antibodies and fragments thereof as described in the present disclosure may also be used in a kit for monitoring the efficacy of vaccination procedures by detecting the presence of protective SARS-CoV-2 antibodies. Antibodies, antibody fragment, or variants and derivatives thereof, as described in the present disclosure may also be used in a kit for monitoring vaccine manufacture with the desired immunogenicity. TABLE 1 - NUCLEOTIDE SEQUENCES FOR ANTIBODY VARIABLE REGION
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
TABLE 2 - PROTEIN SEQUENCES FOR ANTIBODY VARIABLE REGION
Figure imgf000085_0002
Figure imgf000086_0001
Figure imgf000087_0001
TABLE 3 - HEAVY CHAIN SEQUENCES
Figure imgf000087_0002
TABLE 4 - LIGHT CHAIN SEQUENCES
Figure imgf000087_0003
Figure imgf000088_0001
The following examples are intended to illustrate the invention and are not to be construed as being limitations thereon. Abbreviations used are those conventional in the art. If not defined, the terms have their generally accepted meanings. Abbreviations and acronyms used herein include the following: Expi293 Human Embryonic Kidney (HEK293 High density/serum free) cells bp base pairs °C Centigrade MEM Minimal Essential Medium DLS Dynamic light scattering DNA Deoxyribonucleic acid ELISA Enzyme linked immuno-adsorbent assay EC50 Concentration of antibody providing half-maximal response ECD extracellular domain g grams HRP Horseradish peroxidase IgG Immunoglobulin-G LIC Ligase independent cloning min minute MALS Multi-angle light scattering nm nanometre OD optical density PBS Phosphate Buffered Saline PCR Polymerase chain reaction RT Room Temperature s second SEC Size exclusion chromatography TMB 3,3',5,5'- tetramethylbenzidine UV Ultra Violet Nucleotides :
Figure imgf000090_0001
Amino acids :
Figure imgf000090_0002
Figure imgf000091_0001
All publications, patents and patent applications herein are incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference. The foregoing detailed description has been given for clearness of understanding only and no unnecessary limitations should be understood therefrom as modifications will be obvious to those skilled in the art. It is not an admission that any of the information provided herein is prior art or relevant to the presently claimed inventions, or that any publication specifically or implicitly referenced is prior art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth and as follows in the scope of the appended claims. Example 1 ELISA Binding Assay Binding of the antibodies to the following 7 RBD variants was determined using ELISA : - Wild-Type - UK Variant - B 1.1.7 lineage - Alpha variant - South African variant - B.1.351 lineage - Beta variant - Brazilian variant - B.1.1.248 lineage - Gamma variant - Californian variant - B.1.429 lineage - Epsilon variant - Indian variant - B.1.617 lineage - Kappa variant - Indian variant - B.1.617.2 lineage - Delta variant Binding of the antibodies to Omicron variant (B.1.1.529 lineage – Omicron variant) was similarly detemined Methodology 1. Coating of RBD variant at 2μg/ml in 0.58M carbonate-bicarbonate buffer pH 9.5, 100μl/well, 4°C, O/N 2. Washing: PBS + Tween-200.05% v/v (0.05% PBST), 300μl/well 2 times, PBS 1 time 3. Blocking: 3% BSA-PBS, 300μl/well, 30°C, 2H 4. Washing: 0.05% PBST, 300μl/well 2 times, PBS 1 time 5. Incubation of primary antibody: dilutions from 1 to 0.015 μg/mL*, 100μl/well, 37°C, 1H 6. Washing: 0.05% PBST, 300μl/well 2 times, PBS 1 time 7. Incubation of secondary antibody (@human IgG-HRP, ref: https://www.jacksonimmuno.com/catalog/products/109-035-170) 100μl/well, 37°C, 1H 8. Washing: 0.05% PBST 300μl/well 2 times, PBS 1 time 9. Incubation of TMB substrate: 100μl/well, 37°C, 10min 10. Stop: 2M HCl, 50μl/well 11. Reading: OD 450 * Incubation of primary antibody dilutions from 50 to 0.78 μg/mL for assay with Omicron variant Results The tables below represent the average OD values (n=2) obtained for each antibody against each RBD variant. Table 1A : ELISA results of antibody G4 against RBD variants
Figure imgf000093_0001
Table 1B: ELISA results of antibody A5 against RBD variants
Figure imgf000093_0002
Figure imgf000093_0003
Figure imgf000093_0004
Table 1C: ELISA results of antibody B4 against RBD variants
Figure imgf000094_0003
Table 1D: ELISA results of antibody B9 against RBD variants
Figure imgf000094_0004
Table 1E: ELISA results of antibody D8 against RBD variants
Figure imgf000094_0001
Figure imgf000094_0002
Figure imgf000094_0005
Table 1F: ELISA results of antibody A9 against RBD variants
Figure imgf000095_0001
Figure imgf000095_0002
Figure imgf000095_0005
Table 1G: ELISA results of antibody A11 against RBD variants
Figure imgf000095_0006
Table 1H: ELISA results of antibody B7 against RBD variants
Figure imgf000095_0003
Figure imgf000095_0004
Figure imgf000095_0007
Table 1J: ELISA results against Omicron variant
Figure imgf000096_0001
Figure imgf000097_0002
Figure imgf000097_0001
Antibodies B7, A9 and A11 show a clear binding to RBD Omicron. The affinities of these antibodies for this variant are however lower than what was observed against the other RBDs, with estimated KDs of 0.8 μg/mL, 6.25 μg/mL and 0.78 μg/mL for A9, A11 and B7, respectively. Antibodies G4, B4, A5, D8, A9, A11 and B7 all bind very strongly to the remaining 7 RBD variants, as shown by the high OD values that are still obtained at an antibody concentration as low as 0.015 μg/mL. The only exception is B7, which binds with a slightly lower affinity to the Indian kappa variant. Antibody B9 binds strongly to the wild- type and UK variants, does not appear to bind to RBD omicron, and mildly to all the other RBD variants tested. Example 2 Recombinant antibody neutralization assay test The aim of this study was to determine the neutralising properties of the antibodies when pre-incubated with different SARS-CoV2 isolates (Early isolate England 02/2020, Alpha, Beta, Gamma, Delta, and Omicron) for 1 hour before infection. Methodology The assay consists in treating a fixed concentration of virus with different concentrations of the antibody molecules of the invention and assessing the ability of such of inhibiting infection over the range of dilutions tested, once the mix is added to the assay cells. An immunofluorescence-based assay, where antiviral activity is measured as the ability of an antibody to reduce the percentage of infected cells, determined by immunostaining of a viral antigen. A schematic diagram of the assay is shown in Figure 1. In the immunofluorescence assay it is possible to stop the assay after a single round of replication, infection is stopped before synthesis of new viral progeny and further viral spread, which allows to assess the neutralising effect of the antibodies on the starting amount of virus. At the end of the incubation time, the cells are fixed and immuno-stained for a viral antigen, and antiviral activity is determined by quantifying the percentage of infected cells by comparison with infected, untreated cells. This is achieved by high content imaging, using a CellInsight CX5 (Thermo Fisher Scientific) instrument. An Orbitor plate handling robotic system allows for processing of up to 60 plates. SARS-CoV2 variants were incubated with a 2-fold serial dilution of each antibody for 1 hour, after which the mix was added to Vero cells. Antiviral activity was determined 6h later using an immunofluorescence-based assay. A positive and negative control antibody were included as controls. Controls - Uninfected untreated cells - Infected untreated cells - Positive control antibody: Anti-SARS-CoV2 spike protein (SinoBiologicals, 40591-MM43) - Negative control antibody: Mouse IgG1 Isotype control (Invitrogen, 02-6100) Test system - Cells: Vero E6, p3-p5, (VRS stock: ADp3) - Viruses: SARS-CoV2/England/02/2020, from BEI Resources – Early isolate SARS-CoV2 Isolate USA/CA_CDC_5574/2020 (B.1.1.7 - UK variant) - Alpha SARS-CoV2 isolate USA hCoV-19/South African/KRISP-EC-K 005321/2020 (Lineage B.1.351) – Beta SARS-CoV2 hCoV19/Japan/TY7-503/2021 P.1 variant, TY7-503 (Calu3) – Gamma SARS-CoV-2 VOC-202104/02 (B.1.617.2 lineage) – Delta SARS-CoV2 hCoV19/USA/MD-HP20874/2021 B.1.1.529 (P1 in V/T/A) - Omicron Key reagents - Complete media: M199 medium (Gibco, 41150087) supplemented with 5% FBS (Gibco 10500064) and 1X p/s (Gibco 15070063) - Supplemented media: M199 medium (Gibco, 41150087) supplemented with 0.4% BSA (Gibco 15260037) and1X p/s (Gibco 15070063) The antiviral activity of 8 dilutions of each antibody was explored by pre-incubation with SARS-CoV2 for 1 hour before addition to the cells. Virus and formulations were left on the cells for the entire duration of the experiment (6h) before fixation and immunostaining. Cell plating Cells were detached and counted following SOP-RA 003 and SOP-RA 004 before seeding in complete media at 8,000cells/100μl/well. After seeding, the plates were incubated at room temperature (RT) for 5 minutes for even distribution, and then at 37°C, 5% CO2 until the following day. Antibody dilutions Each test antibody was diluted to twice the highest test concentration (20μg/ml) in 750μl: A5 (1,110 μg/ml) à 13.5 μl stock Ab in 736.5 μl infection medium A9 (420 μg/ml) à 35.7 μl stock Ab in 714.3 μl infection medium A11 (900 μg/ml) à 16.7 μl stock Ab in 733.3 μl infection medium B4 (620 μg/ml) à 24.2 μl stock Ab in 725.8 μl infection medium B7 (440 μg/ml) à 34 μl stock Ab in 716 μl infection medium B9 (980 μg/ml) à 15.3 μl stock Ab in 734.7 μl infection medium D8 (530 μg/ml) à 28.3 μl stock Ab in 721.7 μl infection medium G4 (1,100 μg/ml) à 13.6 μl stock Ab in 736.4 μl infection medium Positive control: 8 μl of stock (6,090μg/ml) in 482 μl infection medium Negative control: 49μl of stock (1,000μg/ml) in 441μl infection medium 70μl of test antibodies (in triplicate) or control antibodies (in duplicate) were added to row A of a 96 well plate. For untreated infected and untreated uninfected controls 70μl of infection medium were added.35μl of infection medium were added to all wells of the plates except raw A.35μl were next transferred from the top row down, in a 2-fold dilution series. Virus addition 8,000 infectious units of SARS-CoV2 (MOI 0.5) were added to each well (except the uninfected control) in 35μl of supplemented media, halving the concentrations of the antibodies to the final desired dilution.35μl of supplemented media were added to column 11 (untreated uninfected control). Next, virus and plasma were incubated for 1 hour at 37ºC, 5% CO2. Cell infection After 1 hour, media was removed from the cells and replaced with the virus + antibody dilution mixtures. Plates were incubated for 6h at 37ºC and 5% CO2, and the virus + antibody were left on the cells for the entire duration of the experiment. Fixation and development After 6h, the infection plates were washed with PBS, fixed for 30 mins with 4% formaldehyde, washed again with PBS, and stored in PBS at 4°C until staining. Infectivity readout Cells were immunostained following SOP-RA 005. Briefly, any residual formaldehyde was quenched with 50 mM ammonium chloride, after which cells were permeabilised (0.1% Triton X100) and stained with an antibody recognising SARS-CoV2 nucleoprotein (Invitrogen, MA5-36271). The primary antibody was detected with an Alexa-488 conjugate secondary antibody (Life Technologies, A21244), and nuclei were stained with Hoechst. Images were acquired on an CellInsight CX5 high content platform (Thermo Scientific) using a 4X objective, and percentage infection calculated using CellInsight CX5 software (infected cells/total cells x 100). Determination of EC50 values Normalised percentages of inhibition were calculated EC50 values were extrapolated from the curves representing the best fit (non-linear regression analysis, variable slope) of the logarithm of antibody dilutions vs. the normalised percentages of inhibition, using GraphPad Prism (version 9). Results Table 2A displays the EC50 values for each antibody sample. The graphs in Figure 2 display the percentage of inhibition of SARS-CoV2 infection at different antibody dilutions. Sample values in each plate are normalised to the plate internal controls, where 100% inhibition is derived from the average of the negative control (untreated uninfected) and 0% inhibition is derived from the average of the positive control (untreated infected). The x axes show the dilution factor (log scale). The curves represent the best fit of the logarithm of antibody dilution vs. the normalised percentage of inhibition (variable slope). A5 neutralises all variants except Omicron, with an EC50 lower than ~0.2μg/ml. A9 displays no inhibition against Omicron and modest neutralising activity against the other variants, with EC50 up to 18.7μg/ml. A11 neutralises all variants, with EC50 lower than ~0.1μg/ml. B4 neutralises all variants, with EC50 lower than ~0.05μg/ml. B7 neutralises all variants, with EC50 lower than ~3μg/ml for Delta and lower than ~0.05μg/ml for all other variants. B9 only neutralises the early isolate England/02/2020 and the Alpha variants, with IC50 ~0.2μg/ml. D8 neutralises all variants, and displayed higher potency against England/02/2020, Alpha, and Delta (EC50 lower than ~0.02) than Beta, Gamma, and Omicron (EC50 between ~0.31μg/ml and 1μg/ml) G4 neutralises all variants except for Omicron, with EC50 lower than ~0.4μg/ml. For several antibodies, more than 50% neutralisation was observed even at the highest dilutions. Consequently, for some antibody/variant combinations the EC50 value is either extrapolated from the rest of the curve (shaded cells in Table 1) or could not be determined due to maximal inhibition (close to 100%) maintained at the. lowest concentration tested (0.08μg/ml). (>0.08* in Table 2A). The selected positive control antibody neutralised all variants with similar potency (~0.3μg/ml), except for Omicron. No inhibition was observed when the viruses were pre-incubated with the negative control antibody. Table 2A. EC50 values for each tested antibody, expressed as the concentration (μg/ml) at which 50% of infection is neutralised. > 0.08*: The EC50 values could not be determined or extrapolated, due to maximal inhibition (close to 100%) maintained at the. lowest concentration tested (0.08μg/ml). Shaded cells: More than 50% inhibition observed even at the highest dilution.
Figure imgf000103_0001
Using an immunofluorescence-based microneutralisation assay 6h after infection, antibodies A11, B4, B7, and D8 neutralised all variants including Omicron with potency in the ng/ml range. A5, A9, and G4 inhibited all variants except Omicron. A9 neutralised all variants except Omicron at higher concentrations only; B9 only inhibited the early isolate England/02/2020 and the Alpha variant. Conclusion Under the conditions tested, four antibodies - A11, B4, B7, and D8 – neutralised all variants with potency in the ng/ml range. The Omicron variant showed higher resistance to neutralisation but was inhibited with similar potency by these four antibodies. A5, A9, and G4 inhibited all variants except Omicron. A9 and B9 were the least potent antibodies, with A9 neutralising all variants except Omicron in the μg/ml range, and B9 only inhibiting the early isolate England/02/2020 and the Alpha variant.
SEQUENCE LISTINGS SEQ ID NO:1 GAGGTGCAGCTGGTGCAGTCTGGAGGAGGCTTGGTCCAGCCTGGGGGGTCCCTGAGACTCT CCTGTGCAGCCTCGGGCTTAACCGTCAGTAGCAACTACATGACCTGGGTCCGCCAGGCTCCA GGGAAGGGGCTGGAGTGGGTCTCACTTATTTATGCCGGTGGTAGCACATTCTACGCAGACTC CGTGAAGGGCCGATTCACCATCTCCAGACACGACTCCAAGAACACTCTGTATCTTCAAATGAA CAGCCTGAGACCTGAAGACACGGCCGTTTATTACTGTGCGCGAGATCTGGTCACCTATGGTAT GGACGTCTGGGGCCAAGGGACCCTGGTCACCGTCTCGAGT SEQ ID NO:2 GAAATTGTGCTGACTCAGTCTCCAGGCACCCTGTCTTTGTCTCCAGGGGACAGAGCCACCCT CTCCTGCAGGGCCAGTCAGAGTGTTAGCAGCAGGTACTTAGCCTGGTACCAGCAGAAACCTG GCCAGGCTCCCAGGCTCCTCATCTATGGTGCATCCAGCAGGGCCACTGGCATCCCAGACAG GTTCAGTGGCAGTGGGTCTGGGACAGACTTCACTCTTACCATCAGTAGACTGGAGCCTGAAG ACTTTGCAGTGTATTACTGTCAGCAGTATGGTAGCTCACCGCTGCATTTTGGCCAAGGGACAC GACTGGAGATTAAA SEQ ID NO:3 CAGGTGCAGCTGGTGCAATCTGGGGGAGGCTTGGTCCAGCGGGGGGGGTCCCTGAGACTCT CCTGTGCAGCCTCTGGAATCACCGTCAGGAACAACTACATGACCTGGGTCCGCCAGGCTCCA GGGAAGGGGCTGGAGTGGGTCTCACTTATTTATGCCGGTGGTAGCACATTCTTCGCAGACTC CGTGAAGGGCCGATTCACCCTCTCCAGAGACAATTCCCAGAACATGCTGCATCTTCAAATGAA CAGCCTGAGAGAAGAGGACACGGCTGTGTATTATTGTGCGAGAGATCTCTTGGAGAGGGGGG GTATGGACGTCTGGGGCCAAGGAACCCTGGTCACCGTCTCGAGT SEQ ID NO:4 CAGCCTGTGCTGACTCAGCCACCTTCAGCGTCTGGGACCCCCGGGCAGAGGGTCACCATCT CTTGTTCTGGAGGCAGCTCCAACGTCGGAAGTAATTATGTATTCTGGTACCAGCAGTTCCCAG GAATGGCCCCCAAACTCCTCATTTATAGTGATAATCAGCGGCCCTCAGGGGTCCCTGACCGAT TCTCTGGCTCCAAGTCTGGCACCTCAGCCTCCCTGGCCATCAGTGGGCTCCGGTCCGACGAT GAGGCTGATTACTATTGCGCGGCTTGGGATGGCACCCTGAGCGGCCCGCTATTCGGCGGAG GCACCCAGCTGACCGTCCTC SEQ ID NO;5 GAGGTGCAGCTGGTGGAGTCTGGGGGAGGCTTGGTCCAGCCTGGGGGGTCCCTGAGACTCT CCTGTGCAGCCTCTGGAGTCACCGTCAGTAGCAACTACATGAGGTGGGTCCGCCAGGCTCCA GGGAAGGGGCTGGAGTGGGTCTCACTCATTTATAGCGGTGGTAGCACATTCTACGCAGACTC CGTGAAGGGCAGATTCACCATCTCCAGAGACGATTCCAAGAACACGCTGTATCTTCAAATGAA CAGCCTGAGAGCCGAGGACACGGCTGTGTATCACTGTGCGAGAGATCTTATGGTCTACGGTA TGGACGTCTGGGGCCAAGGGACCCTGGTCACCGTCTCGAGT SEQ ID NO:6 CAGCCTGTTCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGACAGACGGCCAGGATTAC CTGTGGGGGAAACAACATTGGAAGTAAAAGTGTGCACTGGTACCAGCAGAAGCCAGGCCAGG CCCCTGTGCTGGTCGTCTATGATGATAGCGACCGGCCCTCAGGGATCCCTGAGCGATTCTCT GGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAAGCCGGGGATGAGG CCGACTATTACTGTCAGGTGTGGGATAGTAGTAGTGATCATGAGGTGTTCGGCGGAGGGACC AAGGTGACCGTCCTC SEQ ID NO:7 CAGGTGCAGCTGGTGCAATCTGGGGCTGAGGTGAAGAAGCCTGGGTCCTCGGTGAAGGTCT CCTGCAAGGCTTCTGGAGGCACCTTCAGCAGTCATACTTTCAGCTGGGTGCGACAGGCCCCT GGACAAGGGCTTGAGTGGATGGGAAGGATCATCCCTCTCGTTGGTATAGCAAACTACGCACA GAAGTTCCAGGGCAGAGTCACGATTACCGCGGACAAGTCCACGCGCACAGCCTACATGGATC TGAGCAGCCTGAGATCTGAGGATACGGCCGTGTATTACTGTGCGAGAGGCGAGCTTGACAGC TATGGTCACTGGGGTAACTACTTTGTCGACTGGGGCCAGGGGACCCTGGTCACCGTCTCGAG T SEQ ID NO:8 CAGGCAGGGCTGACTCAGCCACCCTCGGTGTCAGTGGCCCCAGGACAGACGGCCAGGATTA CCTGTGGGGGAAACAACATTGGAAGTAAAAGTGTGCACTGGTACCAGCAGAAGCCAGGCCAG GCCCCTGTGCTGGTCGTCTATGATGATAGCGACCGGTCCTCAGGGATCCCTGAGCGATTCTC TGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAAGCCGGGGATGAG GCCGACTATTACTGTCAGGTGTGGGATAGTAGTAGTGATCATGTGGTATTCGGCGGAGGCAC CCAGCTGACCGTCCTC SEQ ID NO:9 CAGGTGCAGCTGGTGCAATCTGGGGCTGAGGTGAAGAAGCCTGGGTCCTCGGTGAAGGTCT CCTGCAAGTCTTTTGGAGGCACCTTCAGCAGCTATACTTTCAGCTGGGTGCGACAGGCCCCT GGGCAAGGTCTTAAATGGATGGGAAGGATCATCCCTTTGGTTGGTGTAACAAACTACGCACAG GACTTCCAGGGCAGAGTCACAATAACCGCGGACAAATCCACGAGCACAGCCTACATGGAGCT GAGCAGCCTGAGATCTGAGGACACGGCCGTCTATTATTGCGCGAGAGGCGAGCTTGACAGTT ATGGTCACTGGGGTACCTACTTTAACTCCTGGGGCCAGGGGACCCTGGTC ACCGTCTCGAGT SEQ ID NO:10 GAAATTGTGCTGACTCAGTCTCCACTCTCCCTGCCCGTCATTCTTGGACAGCCGGCCTCCATC TCCTGCAGGTCTAGTCAAAGCCTCGTTCACAGTGATGGAAACACCTACTTGACTTGGTTTCAC CAGAGGCCAGGCCAATCTCCAAGGCGCCTAATTTATAAGGTTTCTGACCGGGACTCTGGGGT CCCAGACAGATTTAGCGGCAGTGGGTCAGGCACTGATTTCACACTGAAAATCAGCAGGGTGG AGGCTGAGGATGTTGGAGTTTATTACTGCATGCAAGGTACACACCCGCCTTACTACACTTTTG GCCAAGGGACCAAGGTGGAAATCAAA SEQ ID NO:11 GAGGTGCAGCTGTTGGAGTCTGGGGGAGACTTGGTTCACCCGGGGGGATCCCTGAGACTCT CCTGCGCAGCCTCTGGAATCACCGTCAGTAGGAATTACATGACGTGGGTCCGCCAGGCTCCA GGGAAGGGGCTGGAGTGGGTCTCAACTCTTTATGCCGGTGGTAGCACATTTTACTCAGACTC CGTGAAGGGCCGATTCACCATCTCCAGAGACAATTCCAAGAACACTCTTTATCTTCAAATGAA CAGCCTGAGAACTGAAGACACGGCTGTGTATTACTGTGTGAGAGATCTTCAGGACTTCGGCAT GGACGTCTGGGGCCAAGGGACCACGGTCACCGTCTCGAGT SEQ ID NO:12 CAGTCTGCCCTGACTCAGCCTCCCTCAGCGTCTGGGACCCCCGGGCAGACGATCACGATCTC TTGTTCTGGAGACAGCTCTAACATCGGGCCTAATCTTGTCAACTGGTACCAGTTGCTCCCGGG AATGGCCCCCAGACAGGTCATTTATAGTAATTATGTGCGGCCGTCGGGGGTGCCTGAACGAT TCTCTGGCTCCAAGTCTGGCACCTCAGCCTCCCTGGCCGTCAGTGGGCTCCAGTCTGAGGAT GAGGGTGAATATTTCTGTGCAGCCTGGGATGACTCCGTGAACTCTTGGGTGTTCGGCGGAGG CACCCAGCTGACCGCCCTC SEQ ID NO:13 CAGGTGCAGCTGGTGCAGTCTGGGGGTGAGGTGAGGAAGCCTGGGGCCTCAGTGAAGATTT CCTGCAAGGCATCTGAAGACACCTTCACCAACCACTATATACACTGGGTGCGACAGGCCCCT GGACAAGGACTTGAGTGGATAGGGATAATCAACCCTCATGCTGGCAGTATAACCTACGCACA GAAGTTCCAGGGCAGAGTCACCATGACCAGCGACACGGCCACGAGCACAGTCTACATGGAG CTGAGGAGCCTAAGATCTGAGGACACGGCCGTGTATTACTGTGCGCGAGATGGGAATTGGAT ACCGAGTCGGGGATACTTTGACTACTGGGGCCAGGGCACCCTGGTCACTGTCTCGAGT SEQ ID NO:14 CAGTCTGTCGTGACGCAGCCGCCCTCAGTGTCTGCGGCCCCAGGACAGAAGGTCACCATCTC CTGCTCTGGAAGCAGCTCGAACATTGGAAATAATTATGTATCCTGGTACCAGCAGCTCCCAGG AACAGCCCCCCAACTCCTCATTTATGAAAATAATAAGCGATCCTCAGGGATTCCTGACCGATT CTCTGGCTCCAAGTCTGGCACGTCAGCCACCCTGGCCATCACCGGACTCCAGACTGGGGAC GAGGCCGATTATTACTGCGCAACATGGGATATCAGCTTGACTCCTGGCGGGGTGTTCGGCGG AGGCACCCAGCTGACCGTCCTC SEQ ID NO:15 GAGGTGCAGCTGGTGCAGTCTGGAGGAGGCTTGGTCCAGCCTGGGGGGTCCCTGAGACTCT CCTGTGCAGCCTCGGGCTTAACCGTCAGTAGCAACTACATGACCTGGGTCCGCCAGGCTCCA GGGAAGGGGCTGGAGTGGGTCTCACTTATTTATGCCGGTGGTAGCACATTCTACGCAGACTC CGTGAAGGGCCGATTCACCATCTCCAGACACGACTCCAAGAACACTCTGTATCTTCAAATGAA CAGCCTGAGACCTGAAGACACGGCCGTTTATTACTGTGCGCGAGATCTGGTCACCTATGGTAT GGACGTCTGGGGCCAAGGGACAATGGTCACCGTCTCGAGT SEQ ID NO:16 CAGCCTGTGCTGACTCAGCCACCTTCGGTGTCAGCGGCCCCAGGACAGACGGCCAGGATTA CCTGTGGGGGAAACAACATTGGAAGTAAAAGTGTGCACTGGTTCCAGCAGAAGCCAGGCCAG GCCCCTGTGCTGGTCGTCTATGATGATAGCGACCGGCCCTCAGGGATCCCTGAGCGATTCTC TGGCTCCAACTCTGGGAACACGGCCACCCTGACCATCAGCAGGGTCGAAGCCGGGGATGAG GCCGACTATTACTGTCAGGTGTGGGATAGTAGTAGTGATCAAGGGGTGTTCGGCGGAGGCAC CCAGCTGACCGCCCTC SEQ ID NO:17 EVQLVQSGGGLVQPGGSLRLSCAASGLTVSSNYMTWVRQAPGKGLEWVSLIYAGGSTFYADSVK GRFTISRHDS KNTLYLQMNSLRPEDTAVYYCARDLVTYGMDVWGQGTLVTVSS SEQ ID NO:18 EIVLTQSPGTLSLSPGDRATLSCRASQSVSSRYLAWYQQKPGQAPRLLIYGASSRATGIPDRFSGS GSGTDFTLTISR LEPEDFAVYYCQQYGSSPLHFGQGTRLEIK SEQ ID NO:19 QVQLVQSGGGLVQRGGSLRLSCAASGITVRNNYMTWVRQAPGKGLEWVSLIYAGGSTFFADSVK GRFTLSRDNS QNMLHLQMNSLREEDTAVYYCARDLLERGGMDVWGQGTLVTVSS SEQ ID NO.20 QPVLTQPPSASGTPGQRVTISCSGGSSNVGSNYVFWYQQFPGMAPKLLIYSDNQRPSGVPDRFS GSKSGTSASLAIS GLRSDDEADYYCAAWDGTLSGPLFGGGTQLTVL SEQ ID NO:21 EVQLVESGGGLVQPGGSLRLSCAASGVTVSSNYMRWVRQAPGKGLEWVSLIYSGGSTFYADSV KGRFTISRDDSK NTLYLQMNSLRAEDTAVYHCARDLMVYGMDVWGQGTLVTVSS SEQ ID NO:22 QPVLTQPPSVSVAPGQTARITCGGNNIGSKSVHWYQQKPGQAPVLVVYDDSDRPSGIPERFSGS NSGNTATLTISRV EAGDEADYYCQVWDSSSDHEVFGGGTKVTVL SEQ ID NO:23 QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSHTFSWVRQAPGQGLEWMGRIIPLVGIANYAQKF QGRVTITADKS TRTAYMDLSSLRSEDTAVYYCARGELDSYGHWGNYFVDWGQGTLVTVSS SEQ ID NO:24 QAGLTQPPSVSVAPGQTARITCGGNNIGSKSVHWYQQKPGQAPVLVVYDDSDRSSGIPERFSGS NSGNTATLTISR VEAGDEADYYCQVWDSSSDHVVFGGGTQLTVL SED ID NO:25 QVQLVQSGAEVKKPGSSVKVSCKSFGGTFSSYTFSWVRQAPGQGLKWMGRIIPLVGVTNYAQDF QGRVTITADKS TSTAYMELSSLRSEDTAVYYCARGELDSYGHWGTYFNSWGQGTLVTVSS SEQ ID NO:26 EIVLTQSPLSLPVILGQPASISCRSSQSLVHSDGNTYLTWFHQRPGQSPRRLIYKVSDRDSGVPDR FSGSGSGTDFTL KISRVEAEDVGVYYCMQGTHPPYYTFGQGTKVEIK SEQ ID NO:27 EVQLLESGGDLVHPGGSLRLSCAASGITVSRNYMTWVRQAPGKGLEWVSTLYAGGSTFYSDSVK GRFTISRDNSK NTLYLQMNSLRTEDTAVYYCVRDLQDFGMDVWGQGTTVTVSS SEQ ID NO:28 QSALTQPPSASGTPGQTITISCSGDSSNIGPNLVNWYQLLPGMAPRQVIYSNYVRPSGVPERFSG SKSGTSASLAVSG LQSEDEGEYFCAAWDDSVNSWVFGGGTQLTAL SEQ ID NO:29 QVQLVQSGGEVRKPGASVKISCKASEDTFTNHYIHWVRQAPGQGLEWIGIINPHAGSITYAQKFQG RVTMTSDTA TSTVYMELRSLRSEDTAVYYCARDGNWIPSRGYFDYWGQGTLVTVSS SEQ ID NO:30 QSVVTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPQLLIYENNKRSSGIPDRFSG SKSGTSATLAITG LQTGDEADYYCATWDISLTPGGVFGGGTQLTVL SEQ ID NO:31 EVQLVQSGGGLVQPGGSLRLSCAASGLTVSSNYMTWVRQAPGKGLEWVSLIYAGGSTFYADSVK GRFTISRHDS KNTLYLQMNSLRPEDTAVYYCARDLVTYGMDVWGQGTMVTVSS SEQ ID NO:32 QPVLTQPPSVSAAPGQTARITCGGNNIGSKSVHWFQQKPGQAPVLVVYDDSDRPSGIPERFSGS NSGNTATLTISRV EAGDEADYYCQVWDSSSDQGVFGGGTQLTAL SEQ ID NO:33 GLTVSSNY SEQ ID NO:34 IYAGGST SEQ ID NO:35 ARDLVTYGMDV SEQ ID NO:36 GITVRNNY SEQ ID NO:37 IYAGGST SEQ ID NO:38 ARDLLERGGMDV SEQ ID NO:39 GVTVSSNY SEQ ID NO:40 IYSGGST SEQ ID NO:41 ARDLMVYGMDV SEQ ID NO:42 GGTFSSHT SEQ ID NO:43 IIPLVGIA SEQ ID NO:44 ARGELDSYGHWGNYFVD SEQ ID NO:45 GGTFSSYT SEQ ID NO:46 IIPLVGVT SEQ ID NO:47 ARGELDSYGHWGTYFNS SEQ ID NO:48 GITVSRNY SEQ ID NO:49 LYAGGST SEQ ID NO:50 VRDLQDFGMDV SEQ ID NO:51 EDTFTNHY SEQ ID NO:52 INPHAGSI SEQ ID NO:53 ARDGNWIPSRGYFDY SEQ ID NO:54 GLTVSSNY SEQ ID NO:55 IYAGGST SEQ ID NO:56 ARDLVTYGMDV SEQ ID NO:57 QSVSSRY SEQ ID NO:58 GAS SEQ ID NO:59 QQYGSSPLH SEQ ID NO:60 SSNVGSNY SEQ ID NO:61 SDN SEQ ID NO:62 AAWDGTLSGPL SEQ ID NO:63 NIGSKS SEQ ID NO:64 DDS SEQ ID NO:65 QVWDSSSDHEV SEQ ID NO:66 NIGSKS SEQ ID NO:67 DDS SEQ ID NO:68 QVWDSSSDHVV SEQ ID NO:69 QSLVHSDGNTY SEQ ID NO:70 KVS SEQ ID NO:71 MQGTHPPYYT SEQ ID NO:72 SSNIGPNL SEQ ID NO:73 SNY SEQ ID NO:74 AAWDDSVNSWV SEQ ID NO:75 SSNIGNNY SEQ ID NO:76 ENN SEQ ID NO:77 ATWDISLTPGGV SEQ ID NO:78 NIGSKS SEQ ID NO:79 DDS SEQ ID NO:80 QVWDSSSDQGV

Claims

CLAIMS What is claimed is: 1. A method of detecting COVID-19 infection with SARS-CoV-2 in a subject comprising: (a) contacting a sample from the subject with an antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively; and (b) detecting SARS-CoV-2 in the sample by binding of the antibody or antibody fragment to a SARS-CoV-2 antigen in the sample.
2. The method of claim 1, wherein the sample is a body fluid.
3. The method of claims 1 or 2, wherein the sample is blood, sputum, tears, saliva, mucous or serum, semen, cervical or vaginal secretions, amniotic fluid, placental tissues, urine, exudate, transudate, tissue scrapings or faeces.
4. The method of any of claims 1 to 3, wherein detection comprises ELISA, RIA, lateral flow assay or western blot.
5. The method of any of claims 1 to 4, further comprising performing steps (a) and (b) a second time and determining a change in SARS-CoV-2 antigen levels as compared to the first assay.
6. The method of any of claims 1 to 5, wherein the antibody or antibody fragment is encoded by clone-paired variable sequences as set forth in Table 1.
7. The method of any of claims 1 to 5, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired variable sequences as set forth in Table 1.
8. The method of any of claims 1 to 5, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences having 100% identity to clone- paired sequences as set forth in Table 1.
9. The method of any of claims 1 to 5, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences according to clone-paired sequences from Table 2.
10. The method of any of claims 1 to 5, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2.
11. The method of any of claims 1 to 10, wherein the antibody or antibody fragment binds to a SARS-CoV-2 surface spike protein.
12. The method of any of claims 1 to11, wherein the antibody fragment is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab'h fragment, or Fv fragment.
13. A method of treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2, comprising delivering to the subject an antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
14. The method of claim 13, the antibody or antibody fragment is encoded by clone- paired light and heavy chain variable sequences as set forth in Table 1.
15. The method of claims 13 or 14, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 1.
16. The method of claim 13, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences according to clone-paired sequences from Table 2.
17. The method of claim 13, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences having at least 70%, 80% or 90% identity to clone- paired sequences from Table 2.
18. The method of claim 13, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 2.
19. The method of claims 13-18, wherein the antibody fragment is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
20. The method of any of claims 13 to19, wherein the antibody is an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
21. The method of any of claims 13 to 18, wherein the antibody is a chimeric antibody or a bispecific antibody.
22. The method of any of claims 13 to 21, wherein the antibody or antibody fragment binds to a SARS-CoV-2 surface spike protein.
23. The method of any of claims 13 to 22, wherein the antibody or antibody fragment is administered prior to infection or after infection.
24. The method of any of claims 13 to 23, wherein the subject is of age 60 or older, is immunocompromised, or suffers from a respiratory and/or cardiovascular disorder.
25. The method of any of claims 13 to 24, wherein delivering comprises antibody or antibody fragment administration, or genetic delivery with an RNA or DNA sequence or vector encoding the antibody or antibody fragment.
26. A monoclonal antibody molecule, wherein the antibody or antibody fragment is characterized by clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
27. The monoclonal antibody molecule of claim 26, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences according to clone- paired sequences from Table 1.
28. The monoclonal antibody molecule of claim 26, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 70%, 80%, 90%, or 95% identity to clone-paired sequences from Table 1.
29. The monoclonal antibody molecule of claim 26, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences according to clone-paired sequences from Table 2.
30. The monoclonal antibody molecule of claim 26, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences having at least 70%, 80%, 90%, or 95% identity to clone-paired sequences from Table 2.
31. The monoclonal antibody molecule of any of claims 26 to 30, wherein the antibody fragment is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
32. The monoclonal antibody molecule of any of claims 26 to 30, wherein the antibody is a chimeric antibody, or is a bispecific antibody.
33. The monoclonal antibody molecule of any of claims 26 to 32, wherein the antibody is an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
34. The monoclonal antibody molecule of any of claims 26 to 33, wherein the antibody or antibody fragment binds to a SARS-CoV-2 antigen such as a surface spike protein.
35. The monoclonal antibody molecule of any of claims 26 to 34, wherein the antibody is an intrabody.
36. A pharmaceutical composition comprising a monoclonal antibody molecule of any of claims 26 to 34 and one or more pharmaceutically acceptable excipients, diluents and/or carriers.
37. A pharmaceutical composition according to claim 36 in combination with one or more further therapeutic agents
38. A pharmaceutical composition according to claim 36 or 37 comprising two or more monoclonal antibody molecules of any of claims 26 to 35.
39. The monoclonal antibody molecule of any of claims 26 to 35 for use as a medicament.
40. The monoclonal antibody molecule of any of claims 26 to 34 for use in treating a subject infected with SARS-CoV-2 or reducing the likelihood of infection of a subject at risk of contracting SARS-CoV-2
41. The monoclonal antibody molecule according to claim 40 in combination with one or more further therapeutic agents for use according to claim 40.
42. A hybridoma or engineered cell encoding an antibody or antibody fragment wherein the antibody or antibody fragment is characterized by clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
43. The hybridoma or engineered cell of claim 42, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences according to clone- paired sequences from Table 1.
44. The hybridoma or engineered cell of claim 42, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired variable sequences from Table 1.
45. The hybridoma or engineered cell of claim 42, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 95% identity to clone-paired variable sequences from Table 1.
46. The hybridoma or engineered cell of claim 42, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences according to clone-paired sequences from Table 2.
47. The hybridoma or engineered cell of claim 42, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired variable sequences from Table 2.
48. The hybridoma or engineered cell of claim 42, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 2.
49. The hybridoma or engineered cell of any of claims 42 to 48, wherein the antibody fragment is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
50. The hybridoma or engineered cell of any of claims 42 to 49, wherein the antibody is a chimeric antibody, a bispecific antibody, or an intrabody.
51. The hybridoma or engineered cell of any of claims 42 to 49, wherein the antibody is an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
52. The hybridoma or engineered cell of any of claims 42 to 51, wherein the antibody or antibody fragment binds to a SARS-CoV-2 surface spike protein.
53. A vaccine formulation comprising one or more antibodies or antibody fragments characterized by clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
54. The vaccine formulation of claim 53, wherein at least one of the antibodies or antibody fragments is encoded by light and heavy chain variable sequences according to clone- paired sequences from Table 1.
55. The vaccine formulation of claim 53, wherein at least one of the antibodies or antibody fragments is encoded by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired sequences from Table 1.
56. The vaccine formulation of claim 53, wherein at least one of the antibodies or antibody fragments is encoded by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 1.
57. The vaccine formulation of claim 53, wherein at least one of the antibodies or antibody fragments comprises light and heavy chain variable sequences according to clone- paired sequences from Table 2.
58. The vaccine formulation of claim 53, wherein at least one of the antibodies or antibody fragments comprises light and heavy chain variable sequences having at least 70%, 80%, 90% or 95% identity to clone-paired sequences from Table 2.
59. The vaccine formulation of claims 53-58, wherein at least one of the antibody fragments is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
60. The vaccine formulation of claims 53-58, wherein at least one of the antibodies is a chimeric antibody, is bispecific antibody or an intrabody.
61. The vaccine formulation of claims 53-60, wherein the antibody is an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
62. The vaccine formulation of claims 53-61, wherein the antibody or antibody fragment binds to a SARS-CoV-2 surface spike protein.
63. A vaccine formulation comprising one or more expression vectors encoding a first antibody or antibody fragment according to claims 26-34.
64. The vaccine formulation of claim 63 wherein the expression vector(s) is/are Sindbis virus or VEE vector(s).
65. The vaccine formulation of claims 63 or 64, formulated for delivery by needle injection, jet injection, or electroporation.
66. The vaccine formulation of claim 63, further comprising one or more expression vectors encoding for a second antibody or antibody fragment, such as a distinct antibody or antibody fragment of claims 26-34.
67. A method of protecting the health of a subject of age 60 or older, an immunocompromised, subject or a subject suffering from a respiratory and/or cardiovascular disorder that is infected with or at risk of infection with SARS-CoV-2 comprising delivering to the subject an antibody or antibody fragment having clone- paired heavy and light chain CDR sequences from Tables 3 and 4, respectively.
68. The method of claim 67, the antibody or antibody fragment is encoded by clone- paired light and heavy chain variable sequences as set forth in Table 1.
69. The method of claims 67 or 68, wherein the antibody or antibody fragment is encoded by clone- paired light and heavy chain variable sequences having at least 95% identity to as set forth in Table 1.
70. The method of claim 61-62, wherein the antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired sequences from Table 1.
71. The method of claim 67, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences according to clone-paired sequences from Table 2.
72. The method of claim 67, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences having at least 70%, 80% or 90% identity to clone- paired sequences from Table 2.
73. The method of claim 67, wherein the antibody or antibody fragment comprises light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 2.
74 The method of any of claims 67 to 73, wherein the antibody fragment is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
75. The method of any of claims 67to 73 wherein the antibody is a chimeric antibody or a bispecific antibody.
76. The method of any of claims 67 to 75, wherein the antibody is an IgG, or a recombinant IgG antibody or antibody fragment comprising an Fc portion mutated to alter (eliminate or enhance) FcR interactions, to increase half-life and/or increase therapeutic efficacy, such as a LALA, LALA PG, N297, GASD/ALIE, DHS, YTE or LS mutation or glycan modified to alter (eliminate or enhance) FcR interactions such as enzymatic or chemical addition or removal of glycans or expression in a cell line engineered with a defined glycosylating pattern.
77. The method of any of claims 67 to 76, wherein the antibody or antibody fragment is administered prior to infection or after infection.
78. The method of any of claims 67 to77, wherein the antibody or antibody fragment binds to a SARS-CoV-2 surface spike protein.
79. The method of any of claims 67 to 78, wherein delivering comprises antibody or antibody fragment administration, or genetic delivery with an RNA or DNA sequence or vector encoding the antibody or antibody fragment.
80 The method of claim 67, wherein the antibody or antibody fragment improves the subject's respiration as compared to an untreated control.
81. The method of claim 67, wherein the antibody or antibody fragment reduces viral load as compared to an untreated control.
82. A method of determining the antigenic integrity, correct conformation and/or correct sequence of a SARS-CoV-2 surface spike protein comprising: (a) contacting a sample comprising the antigen with a first antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively; and (b) determining antigenic integrity, correct conformation and/or correct sequence of the antigen by detectable binding of the first antibody or antibody fragment to the antigen.
83. The method of claim 82, wherein the sample comprises recombinantly produced antigen.
84. The method of claim 82, wherein the sample comprises a vaccine formulation or vaccine production batch.
85. The method of any of claims 82 to 84, wherein detection comprises ELISA, RIA, western blot, a biosensor using surface plasmon resonance or biolayer interferometry, or flow cytometric staining.
86. The method of any of claims 82 to 85, wherein the first antibody or antibody fragment is encoded by clone-paired variable sequences as set forth in Table 1.
87. The method of any of claims 82 to 85, wherein the first antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired variable sequences as set forth in Table 1.
88. The method of any of claims 82 to 85 wherein the first antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences as set forth in Table 1.
89. The method of any of claims 82 to 85, wherein the first antibody or antibody fragment comprises light and heavy chain variable sequences according to clone-paired sequences from Table 2.
90 The method of any of claims 82 to 85, wherein the first antibody or antibody fragment comprises light and heavy chain variable sequences having at least 70%, 80% or 90% identity to clone-paired sequences from Table 2.
91. The method of any of claims 82 to 85, wherein the first antibody or antibody fragment comprises light and heavy chain variable sequences having at least 95% identity to clone-paired sequences from Table 2.
92. The method of any of claims 82 to 91, wherein the first antibody fragment is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
93. The method of any of claims 82 to 92, further comprising performing steps (a) and (b) a second time to determine the antigenic stability of the antigen over time.
94. The method of any of claims 82 to 93, further comprising: (c) contacting a sample comprising the antigen with a second antibody or antibody fragment having clone-paired heavy and light chain CDR sequences from Tables 3 and 4, respectively; and (d) determining antigenic integrity of the antigen by detectable binding of the second antibody or antibody fragment to the antigen.
95. The method of claim 94, wherein the second antibody or antibody fragment is encoded by clone-paired variable sequences as set forth in Table 1.
96. The method of claim 95, wherein the second antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 70%, 80%, or 90% identity to clone-paired variable sequences as set forth in Table 1.
97. The method of claim 96, wherein the second antibody or antibody fragment is encoded by light and heavy chain variable sequences having at least 95% identity to clone-paired sequences as set forth in Table 1.
98. The method of claims 95, wherein the second antibody or antibody fragment comprises light and heavy chain variable sequences according to clone-paired sequences from Table 2.
99. The method of claim 95, wherein the second antibody or antibody fragment comprises light and heavy chain variable sequences having at least 70%, 80% or 90% identity to clone-paired sequences from Table 2.
100. The method of claim 95, wherein the second antibody or antibody fragment comprises light and heavy chain variable sequences having at least 95% identity to clone- paired sequences from Table 2.
101. The method of claim 95, wherein the second antibody fragment is a recombinant scFv (single chain fragment variable) antibody, Fab fragment, F(ab')2 fragment, or Fv fragment.
102. The method of claim 95, further comprising performing steps (c) and (d) a second time to determine the antigenic stability of the antigen over time.
PCT/GB2023/050858 2022-04-01 2023-03-31 Human monoclonal antibodies binding to sars-cov-2 and methods of use thereof WO2023187407A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB2204813.6A GB202204813D0 (en) 2022-04-01 2022-04-01 Human monoclonal antibodies and methods of use thereof
GB2204813.6 2022-04-01

Publications (1)

Publication Number Publication Date
WO2023187407A1 true WO2023187407A1 (en) 2023-10-05

Family

ID=81581478

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2023/050858 WO2023187407A1 (en) 2022-04-01 2023-03-31 Human monoclonal antibodies binding to sars-cov-2 and methods of use thereof

Country Status (2)

Country Link
GB (1) GB202204813D0 (en)
WO (1) WO2023187407A1 (en)

Citations (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
EP0003089A1 (en) 1978-01-06 1979-07-25 Bernard David Drier for silkscreen printed sheets
US4196265A (en) 1977-06-15 1980-04-01 The Wistar Institute Method of producing antibodies
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4472509A (en) 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4554101A (en) 1981-01-09 1985-11-19 New York Blood Center, Inc. Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4680338A (en) 1985-10-17 1987-07-14 Immunomedics, Inc. Bifunctional linker
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4867973A (en) 1984-08-31 1989-09-19 Cytogen Corporation Antibody-therapeutic agent conjugates
US4938948A (en) 1985-10-07 1990-07-03 Cetus Corporation Method for imaging breast tumors using labeled monoclonal anti-human breast cancer antibodies
WO1991000360A1 (en) 1989-06-29 1991-01-10 Medarex, Inc. Bispecific reagents for aids therapy
US5021236A (en) 1981-07-24 1991-06-04 Schering Aktiengesellschaft Method of enhancing NMR imaging using chelated paramagnetic ions bound to biomolecules
WO1992000373A1 (en) 1990-06-29 1992-01-09 Biosource Genetics Corporation Melanin production by transformed microorganisms
US5141648A (en) 1987-12-02 1992-08-25 Neorx Corporation Methods for isolating compounds using cleavable linker bound matrices
US5196066A (en) 1990-04-27 1993-03-23 Kusuda Company Limited Facing material spray apparatus
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1994004690A1 (en) 1992-08-17 1994-03-03 Genentech, Inc. Bispecific immunoadhesins
WO1996016673A1 (en) 1994-12-02 1996-06-06 Chiron Corporation Method of promoting an immune response with a bispecific antibody
US5563250A (en) 1987-12-02 1996-10-08 Neorx Corporation Cleavable conjugates for the delivery and release of agents in native form
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1998002463A1 (en) 1996-07-11 1998-01-22 Medarex, Inc. THERAPEUTIC MULTISPECIFIC COMPOUNDS COMPRISED OF ANTI-FCα RECEPTOR ANTIBODIES
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US5837234A (en) 1995-06-07 1998-11-17 Cytotherapeutics, Inc. Bioartificial organ containing cells encapsulated in a permselective polyether suflfone membrane
US5856456A (en) 1992-11-20 1999-01-05 Enzon, Inc. Linker for linked fusion polypeptides
US5880270A (en) 1995-06-07 1999-03-09 Cellpro, Incorporated Aminooxy-containing linker compounds for formation of stably-linked conjugates and methods related thereto
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US6485982B1 (en) 1988-06-27 2002-11-26 Armkel, Llc Test device and method for colored particle immunoassay
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20040101920A1 (en) 2002-11-01 2004-05-27 Czeslaw Radziejewski Modification assisted profiling (MAP) methodology
US7521056B2 (en) 2005-04-06 2009-04-21 Ibc Pharmaceuticals, Inc. Stably tethered structures of defined compositions with multiple functions or binding specificities
US7527787B2 (en) 2005-10-19 2009-05-05 Ibc Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
US7534866B2 (en) 2005-10-19 2009-05-19 Ibc Pharmaceuticals, Inc. Methods and compositions for generating bioactive assemblies of increased complexity and uses
US7550143B2 (en) 2005-04-06 2009-06-23 Ibc Pharmaceuticals, Inc. Methods for generating stably linked complexes composed of homodimers, homotetramers or dimers of dimers and uses
US7666400B2 (en) 2005-04-06 2010-02-23 Ibc Pharmaceuticals, Inc. PEGylation by the dock and lock (DNL) technique
WO2015150447A1 (en) 2014-04-02 2015-10-08 F. Hoffmann-La Roche Ag Multispecific antibodies
WO2021195326A1 (en) * 2020-03-26 2021-09-30 Vanderbilt University Human monoclonal antibodies to severe acute respiratory syndrome coronavirus 2 (sars-cov-2)
CN114031685A (en) * 2022-01-10 2022-02-11 中国人民解放军军事科学院军事医学研究院 Fully human anti-new coronavirus broad-spectrum neutralizing antibody ZW2G10 and application thereof

Patent Citations (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4196265A (en) 1977-06-15 1980-04-01 The Wistar Institute Method of producing antibodies
EP0003089A1 (en) 1978-01-06 1979-07-25 Bernard David Drier for silkscreen printed sheets
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4366241B1 (en) 1980-08-07 1988-10-18
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4554101A (en) 1981-01-09 1985-11-19 New York Blood Center, Inc. Identification and preparation of epitopes on antigens and allergens on the basis of hydrophilicity
US5021236A (en) 1981-07-24 1991-06-04 Schering Aktiengesellschaft Method of enhancing NMR imaging using chelated paramagnetic ions bound to biomolecules
US4472509A (en) 1982-06-07 1984-09-18 Gansow Otto A Metal chelate conjugated monoclonal antibodies
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4867973A (en) 1984-08-31 1989-09-19 Cytogen Corporation Antibody-therapeutic agent conjugates
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4938948A (en) 1985-10-07 1990-07-03 Cetus Corporation Method for imaging breast tumors using labeled monoclonal anti-human breast cancer antibodies
US4680338A (en) 1985-10-17 1987-07-14 Immunomedics, Inc. Bifunctional linker
US5563250A (en) 1987-12-02 1996-10-08 Neorx Corporation Cleavable conjugates for the delivery and release of agents in native form
US5141648A (en) 1987-12-02 1992-08-25 Neorx Corporation Methods for isolating compounds using cleavable linker bound matrices
US6485982B1 (en) 1988-06-27 2002-11-26 Armkel, Llc Test device and method for colored particle immunoassay
WO1991000360A1 (en) 1989-06-29 1991-01-10 Medarex, Inc. Bispecific reagents for aids therapy
US5196066A (en) 1990-04-27 1993-03-23 Kusuda Company Limited Facing material spray apparatus
WO1992000373A1 (en) 1990-06-29 1992-01-09 Biosource Genetics Corporation Melanin production by transformed microorganisms
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1994004690A1 (en) 1992-08-17 1994-03-03 Genentech, Inc. Bispecific immunoadhesins
US5856456A (en) 1992-11-20 1999-01-05 Enzon, Inc. Linker for linked fusion polypeptides
WO1996016673A1 (en) 1994-12-02 1996-06-06 Chiron Corporation Method of promoting an immune response with a bispecific antibody
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5837234A (en) 1995-06-07 1998-11-17 Cytotherapeutics, Inc. Bioartificial organ containing cells encapsulated in a permselective polyether suflfone membrane
US5880270A (en) 1995-06-07 1999-03-09 Cellpro, Incorporated Aminooxy-containing linker compounds for formation of stably-linked conjugates and methods related thereto
WO1998002463A1 (en) 1996-07-11 1998-01-22 Medarex, Inc. THERAPEUTIC MULTISPECIFIC COMPOUNDS COMPRISED OF ANTI-FCα RECEPTOR ANTIBODIES
WO1999054342A1 (en) 1998-04-20 1999-10-28 Pablo Umana Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20040101920A1 (en) 2002-11-01 2004-05-27 Czeslaw Radziejewski Modification assisted profiling (MAP) methodology
US7521056B2 (en) 2005-04-06 2009-04-21 Ibc Pharmaceuticals, Inc. Stably tethered structures of defined compositions with multiple functions or binding specificities
US7550143B2 (en) 2005-04-06 2009-06-23 Ibc Pharmaceuticals, Inc. Methods for generating stably linked complexes composed of homodimers, homotetramers or dimers of dimers and uses
US7666400B2 (en) 2005-04-06 2010-02-23 Ibc Pharmaceuticals, Inc. PEGylation by the dock and lock (DNL) technique
US7527787B2 (en) 2005-10-19 2009-05-05 Ibc Pharmaceuticals, Inc. Multivalent immunoglobulin-based bioactive assemblies
US7534866B2 (en) 2005-10-19 2009-05-19 Ibc Pharmaceuticals, Inc. Methods and compositions for generating bioactive assemblies of increased complexity and uses
WO2015150447A1 (en) 2014-04-02 2015-10-08 F. Hoffmann-La Roche Ag Multispecific antibodies
WO2021195326A1 (en) * 2020-03-26 2021-09-30 Vanderbilt University Human monoclonal antibodies to severe acute respiratory syndrome coronavirus 2 (sars-cov-2)
CN114031685A (en) * 2022-01-10 2022-02-11 中国人民解放军军事科学院军事医学研究院 Fully human anti-new coronavirus broad-spectrum neutralizing antibody ZW2G10 and application thereof

Non-Patent Citations (63)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1990, MACK PRINTING COMPANY, pages: 1289 - 1329
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., NUCL. ACIDS RES., vol. 25, 1977, pages 3389 - 3402
BAILLIE, FEES LETTERS, vol. 579, 2005, pages 3264
BRENNAN ET AL., SCIENCE, vol. 229, 1985, pages 81
CALLAWAY E: "Heavily mutated Omicron variant puts scientists on alert", NATURE, vol. 600, no. 7887, December 2021 (2021-12-01), pages 21, XP037632537, DOI: 10.1038/d41586-021-03552-w
CHENNAMSETTY ET AL., PNAS, vol. 106, no. 29, 2009, pages 11937 - 11942
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
DAVIES J ET AL., BIOTECHNOLOGY & BIOENGINEERING, vol. 74, no. 4, 2001, pages 288 - 294
DUAN XUEJUN ET AL: "Dual-detection fluorescent immunochromatographic assay for quantitative detection of SARS-CoV-2 spike RBD-ACE2 blocking neutralizing antibody", BIOSENSORS AND BIOELECTRONICS, ELSEVIER SCIENCE LTD, UK, AMSTERDAM , NL, vol. 199, 15 December 2021 (2021-12-15), XP086913700, ISSN: 0956-5663, [retrieved on 20211215], DOI: 10.1016/J.BIOS.2021.113883 *
EHRING, ANALYTICAL BIOCHEMISTRY, vol. 267, 1999, pages 252 - 259
ELLIOTT, S., NATURE BIOTECHNOL., vol. 21, 2003, pages 414 - 21
FARIA, NUNO R.CLARO, INGRA MORALES: "Genomic characterisation of an emergent SARS-CoV-2 lineage in Manaus: preliminary findings", VIROLOGICAL, 12 January 2021 (2021-01-12)
FERNANDEZ- ESCAMILLA ET AL., NATURE BIOTECH., vol. 22, no. 10, 2004, pages 1302 - 1306
GARBERDEMAREST, BIOCHEM. BIOPHYS. RES. COMMUN., vol. 355, 2007, pages 751 - 757
GRUBER ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368
HEIN J: "Atlas of Protein Sequence and Structure, National Biomedical Research Foundation", vol. 183, 1990, ACADEMIC PRESS, INC., article "Unified Approach to Alignment and Phylogeny", pages: 626 - 645
HENIKOFFHENIKOFF, PROC. NATL. ACAD. SCI. USA, vol. 89, 1989, pages 10915
HIGGINS, D. GSHARP, P. M., CABIOS, vol. 5, 1989, pages 151 - 153
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
J. BIOL. CHEM., vol. 276, pages 6591 - 6604
JUNGHANS ET AL., CANCER RES., vol. 50, 1990, pages 1495 - 1502
KOHLER ET AL., NATURE, vol. 256, 1975, pages 495
KOSTELNY ET AL., J. IMMUNOL., vol. 148, no. 5, 1992, pages 1547 - 1553
LEFRANC, M. P. ET AL., NUCL. ACIDS RES., vol. 27, 1999, pages 209 - 212
LEUNG, K. ET AL.: "Early empirical assessment of the N501Y mutant strains of SARS-CoV-2 in the United Kingdom, October to November 2020.", MEDRXIV, 2020
MARKS ET AL., J. MOL. BIOL., vol. 222, 1991, pages 581 - 597
MERCHANT ET AL., NAT. BIOTECLMO!, vol. 16, 1998, pages 677 - 681
MILLSTEIN ET AL., NATURE, vol. 305, 1983, pages 537 - 539
MYERS, E.W., MULLER W., CABIOS, vol. 4, 1988, pages 11 - 17
NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
PEARSONLIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
PLANAS DELPHINE ET AL: "Considerable escape of SARS-CoV-2 Omicron to antibody neutralization", NATURE, NATURE PUBLISHING GROUP UK, LONDON, vol. 602, no. 7898, 23 December 2021 (2021-12-23), pages 671 - 675, XP037700793, ISSN: 0028-0836, [retrieved on 20211223], DOI: 10.1038/S41586-021-04389-Z *
REINEKE, METHODS J\FOL. BIOL, vol. 248, 2004, pages 443 - 63
ROBINSON, E. D., COMB. THEOR, vol. 11, 1971, pages 105
ROUTLEDGE, E. G. ET AL., TRANSPLANTATION, vol. 60, no. 8, 1995, pages 847 - 53
RUIZ, M. ET AL., NUCL. ACIDS RES., vol. 28, 2000, pages 219 - 221
SANTOU, N. NES, M., MOL. BIOL. EVOL., vol. 4, 1987, pages 406 - 425
SHALABY ET AL., J. EXP. MED., vol. 175, 1992, pages 217 - 225
SHIELDS, R. L. ET AL., J. BIOL. CHEM., vol. 277, no. 30, 2002, pages 26733 - 26740
SMITHWATERMAN, ADD. APL. MATH, vol. 2, 1981, pages 482
SNEATH, P.H. ASOKAL, R.R.: "Numerical Taxonomy--the Principles and Practice of Numerical Taxonomy", 1973, FREEMAN PRESS
SORMANNI ET AL., J MOL BIOL, vol. 427, 2015, pages 478 - 490
STARR TNGREANEY AJDINGENS ASBLOOM JD: "Complete map of SARS-CoV-2 RBD mutations that escape the monoclonal antibody LY-CoV555 and its cocktail with LY-CoV016", CELL REP MED, vol. 2, no. 4, 2021, pages 100255
SURESH ET AL., METHODS IN ENZYMOLOGY, vol. 121, 1986, pages 210
TAO, M. H. ET AL., J. IMMUNOL., vol. 143, no. 8, 1989, pages 2595 - 2601
TEGALLY, H. ET AL.: "Emergence and rapid spread of a new severe acute respiratory syndrome- related coronavirus 2 (SARS-CoV-2) lineage with multiple Spike mutations in South Africa", MEDRXIV, 2020
TEGALLY, H. ET AL.: "Emergence and rapid spread of a new severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) lineage with multiple Spike mutations in South Africa", MEDRXIV, 2020
THOMSON, E.C. ET AL.: "The circulating SARS-CoV-2 Spike variant N439K maintains fitness while evading antibody-mediated immunity", BIORXIV, 2020
TORTORICI M ALEJANDRA ET AL: "Ultrapotent human antibodies protect against SARS-CoV-2 challenge via multiple mechanisms", SCIENCE, 20 November 2020 (2020-11-20), United States, pages 950 - 957, XP055794998, Retrieved from the Internet <URL:https://science.sciencemag.org/content/sci/370/6519/950.full.pdf> [retrieved on 20210414], DOI: 10.1126/science.abe3354 *
TRAUNECKER ET AL., EMBO J., vol. 10, 1991, pages 3655 - 3659
TREVINO ET AL., J MOL BIOL, vol. 366, 2007, pages 449 - 460
TUTT ET AL., J. IMMUNOL., vol. 147, 1991, pages 60
UENO MIKAKO ET AL: "Isolation of human monoclonal antibodies with neutralizing activity to a broad spectrum of SARS-CoV-2 viruses including the Omicron variants", ANTIVIRAL RESEARCH, ELSEVIER BV, NL, vol. 201, 24 March 2022 (2022-03-24), XP087014276, ISSN: 0166-3542, [retrieved on 20220324], DOI: 10.1016/J.ANTIVIRAL.2022.105297 *
UMANA ET AL., NATURE BIOTECHNOLOGY, vol. 17, February 1999 (1999-02-01), pages 176 - 180
VOYNOV ET AL., BIOCON. CHEM., vol. 21, no. 2, 2010, pages 385 - 392
WALLICK, S. C. ET AL., J. EXP. MED., vol. 168, no. 3, 1988, pages 1099 - 1109
WARDEMANN ET AL., SCIENCE, vol. 301, 2003, pages 1374 - 1377
WESTENDORF KATHRYN ET AL: "LY-CoV1404 (bebtelovimab) potently neutralizes SARS-CoV-2 variants", BIORXIV, 7 January 2022 (2022-01-07), XP055916038, Retrieved from the Internet <URL:https://www.biorxiv.org/content/10.1101/2021.04.30.442182v5.full.pdf> [retrieved on 20220427], DOI: 10.1101/2021.04.30.442182 *
WILBUR, W. J.LIPMAN, D. J., PROC. NATL. ACAD., SCI. USA, vol. 80, 1983, pages 726 - 730
WONGSCOTT, NAT. REV. MOL. CELL BIOL, vol. 5, 2004, pages 959
XU ET AL., SCIENCE, vol. 358, no. 6359, 2017, pages 85 - 90

Also Published As

Publication number Publication date
GB202204813D0 (en) 2022-05-18

Similar Documents

Publication Publication Date Title
US20210277092A1 (en) HUMAN MONOCLONAL ANTIBODIES TO SEVERE ACUTE RESPIRATORY SYNDROME CORONAVIRUS 2 (SARS-CoV-2)
US11345741B2 (en) Human monoclonal antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)
WO2021195326A1 (en) Human monoclonal antibodies to severe acute respiratory syndrome coronavirus 2 (sars-cov-2)
US20220289828A1 (en) Human monoclonal antibodies to enterovirus d68
US20230073075A1 (en) Human hendra virus and nipah virus antibodies and methods of use therefor
WO2020061159A1 (en) Human antibodies to zika virus
US20230122364A1 (en) HUMAN MONOCLONAL ANTIBODIES TO SEVERE ACUTE RESPIRATORY SYNDROME CORONAVIRUS 2 (SARS-CoV-2)
WO2019210144A1 (en) Broadly neutralizing antibodies against hepatitis c virus
US20230063625A1 (en) Human antibodies to rift valley fever virus
US20220380442A1 (en) Human monoclonal antibodies to hantavirus and methods of use therefore
WO2021195385A1 (en) HUMAN MONOCLONAL ANTIBODIES TO SEVERE ACUTE RESPIRATORY SYNDROME CORONAVIRUS 2 (SARS-GoV-2)
WO2023187407A1 (en) Human monoclonal antibodies binding to sars-cov-2 and methods of use thereof
US20230085393A1 (en) Human antibodies that neutralize zika virus and methods of use therefor
US20230181714A1 (en) Human monoclonal antibodies to venezuelan equine encephalitis virus and uses therefor
US20230072640A1 (en) Human monoclonal antibodies against yellow fever virus and uses therefor
US20240026035A1 (en) Human ige monoclonal antibodies to antibodies to alpha-gal (galactose-a-1,3-galactose) and uses therefor
WO2024015760A2 (en) Human monoclonal antibodies to omicron variant of severe acute respiratory syndrome coronavirus 2 (sars-cov- 2)
EP4356924A2 (en) Human monoclonal antibodies to severe acute respiratory syndrome coronavirus 2 (sars-cov-2)
WO2021142413A1 (en) Human antibodies to crimean congo hemorrhagic fever virus
WO2023235666A2 (en) Human antibodies to bordetella pertussis and uses therefor
AU2020273365A1 (en) Human antibodies to Ross River virus and methods of use therefor
CN116529259A (en) Human monoclonal antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)

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: 23720651

Country of ref document: EP

Kind code of ref document: A1