WO2021045836A1 - Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments - Google Patents

Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments Download PDF

Info

Publication number
WO2021045836A1
WO2021045836A1 PCT/US2020/039707 US2020039707W WO2021045836A1 WO 2021045836 A1 WO2021045836 A1 WO 2021045836A1 US 2020039707 W US2020039707 W US 2020039707W WO 2021045836 A1 WO2021045836 A1 WO 2021045836A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
antigen
amino acid
acid sequence
seq
Prior art date
Application number
PCT/US2020/039707
Other languages
French (fr)
Inventor
Robert Babb
Alina Baum
Gang Chen
Cindy Gerson
Johanna Hansen
Tammy Huang
Christos Kyratsous
Wen-Yi Lee
Marine Malbec
Andrew Murphy
William Olson
Neil Stahl
George D. Yancopoulos
Original Assignee
Regeneron Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Regeneron Pharmaceuticals, Inc. filed Critical Regeneron Pharmaceuticals, Inc.
Priority to JP2021518800A priority Critical patent/JP7116256B1/en
Priority to BR122023004078-3A priority patent/BR122023004078B1/en
Priority to CR20220552A priority patent/CR20220552A/en
Priority to KR1020217013280A priority patent/KR20210134300A/en
Priority to CA3115553A priority patent/CA3115553C/en
Priority to PE2022002139A priority patent/PE20221893A1/en
Priority to AU2020340881A priority patent/AU2020340881A1/en
Priority to MYPI2021002018A priority patent/MY197648A/en
Priority to SG11202103404PA priority patent/SG11202103404PA/en
Priority to EA202190836A priority patent/EA202190836A1/en
Priority to BR112020015534-9A priority patent/BR112020015534B1/en
Priority to MX2021004130A priority patent/MX2021004130A/en
Priority to CN202080005771.3A priority patent/CN113766928A/en
Priority to ES20186667T priority patent/ES2939895T3/en
Priority to HRP20230338TT priority patent/HRP20230338T1/en
Priority to LTEP20186667.0T priority patent/LT3889177T/en
Priority to SI202030145T priority patent/SI3889177T1/en
Priority to MA54405A priority patent/MA54405B1/en
Priority to HUE20186667A priority patent/HUE061425T2/en
Priority to PT201866670T priority patent/PT3889177T/en
Priority to EP20186667.0A priority patent/EP3889177B1/en
Priority to PL20186667.0T priority patent/PL3889177T3/en
Priority to FIEP20186667.0T priority patent/FI3889177T3/en
Priority to DK20186667.0T priority patent/DK3889177T3/en
Priority to EP22210926.6A priority patent/EP4209507A1/en
Priority to RS20230266A priority patent/RS64105B1/en
Publication of WO2021045836A1 publication Critical patent/WO2021045836A1/en
Priority to IL282060A priority patent/IL282060A/en
Priority to PH12021550793A priority patent/PH12021550793A1/en
Priority to CONC2021/0005072A priority patent/CO2021005072A2/en
Priority to JP2022119389A priority patent/JP7411029B2/en
Priority to JP2023215497A priority patent/JP2024038003A/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/15Reoviridae, e.g. calf diarrhea virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • 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]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • 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
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to antibodies and antigen-binding fragments that bind specifically to coronavirus spike proteins and methods for treating or preventing coronavirus infections with said antibodies and fragments.
  • BACKGROUND OF THE INVENTION [0003] Newly identified viruses, such as coronaviruses, can be difficult to treat because they are not sufficiently characterized. The emergence of these newly identified viruses highlights the need for the development of novel antiviral strategies. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a newly-emergent coronavirus which causes a severe acute respiratory disease, COVID-19.
  • SARS-CoV-2 was first identified from an outbreak in Wuhan, China and as of March 20, 2020, the World Health Organization has reported 209,839 confirmed cases in 168 countries, areas, or territories, resulting in 8,778 deaths. Clinical features of COVID-19 include fever, dry cough, and fatigue, and the disease can cause respiratory failure resulting in death. [0004] Thus far, there has been no vaccine or therapeutic agent to prevent or treat SARS-CoV- 2 infection. In view of the continuing threat to human health, there is an urgent need for preventive and therapeutic antiviral therapies for SARS-CoV-2 control.
  • this virus uses its spike glycoprotein for interaction with the cellular receptor ACE2 and the serine protease TMPRSS2 for entry into a target cell, this spike protein represents an attractive target for antibody therapeutics.
  • fully human antibodies that specifically bind to the SARS-Coactivated virus
  • CoV-2-Spike protein SARS-CoV-2-S
  • SARS-CoV-2-S SARS-CoV-2-S
  • the present disclosure addresses this need, in part, by providing human anti-SARS-CoV-2-S antibodies, such as those of Table 1, and combinations thereof including, for example, combinations with other therapeutics (e.g., anti-inflammatory agents, antimalarial agents, antiviral agents, or other antibodies or antigen-binding fragments), and methods of use thereof for treating viral infections.
  • therapeutics e.g., anti-inflammatory agents, antimalarial agents, antiviral agents, or other antibodies or antigen-binding fragments
  • the present disclosure provides neutralizing human antigen-binding proteins that specifically bind to SARS-CoV-2-S, for example, antibodies or antigen-binding fragments thereof.
  • the present disclosure provides an isolated recombinant antibody or antigen-binding fragment thereof that specifically binds to a coronavirus spike protein (CoV-S), wherein the antibody has one or more of the following characteristics: (a) binds to CoV-S with an EC50 of less than about 10 -9 M; (b) demonstrates an increase in survival in a coronavirus- infected animal after administration to said coronavirus-infected animal, as compared to a comparable coronavirus-infected animal without said administration; and/or (c) comprises three heavy chain complementarity determining regions (CDRs) (CDR-H1, CDR-H2, and CDR-H3) contained within a heavy chain variable region (HCVR) comprising an amino acid sequence having at least about 90% sequence identity to an HCVR of
  • CDRs heavy chain complementar
  • the antibody or antigen-binding fragment comprises: (a) an immunoglobulin heavy chain variable region comprising the CDR-H1, CDR-H2, and CDR-H3 of an antibody of Table 1; and/or (b) an immunoglobulin light chain variable region comprising the CDR-L1, CDR-L2, and CDR-L3 of an antibody of Table 1.
  • the antibody or antigen-binding fragment comprises: (a) a heavy chain immunoglobulin variable region comprising an amino acid sequence having at least 90%
  • the antibody or antigen-binding fragment comprises the CDR- H1, CDR-H2, CDR-H3, CDR-L1, CDR-L2, and CDR-L3 of a single antibody of Table 1.
  • the antibody or antigen-binding fragment comprises an immunoglobulin that comprises the HCVR and the LCVR of a single antibody of Table 1.
  • the present disclosure provides an antigen-binding protein that competes with any one of the antibodies or antigen-binding fragments discussed above or herein for binding to CoV-S.
  • the present disclosure provides an antigen-binding protein that binds to the same epitope as, or to an overlapping epitope on, CoV-S as any one of the antibodies or antigen-binding fragments discussed above or herein.
  • the antibody or antigen-binding fragment may be multispecific.
  • the antibody or antigen-binding fragment may comprise one or more of the following properties: a) inhibits growth of coronavirus; b) binds to the surface of a coronavirus; c) limits spread of coronavirus infection of cells in vitro; and d) protects mice engineered to express the human ACE2 or TMPRSS2 protein from death and/or weight loss caused by coronavirus infection.
  • CoV-S is SARS-CoV-2-S.
  • the present disclosure provides a complex comprising an antibody or antigen-binding fragment as discussed above or herein bound to a CoV-S polypeptide.
  • the CoV-S is SARS-CoV-2-S.
  • the present disclosure provides a method for making an antibody or antigen-binding fragment as discussed above or herein, comprising: (a) introducing into a host cell one or more polynucleotides encoding said antibody or antigen-binding fragment; (b) culturing the host cell under conditions favorable to expression of the one or more polynucleotides; and (c) optionally, isolating the antibody or antigen-binding fragment from the host cell and/or a medium in which the host cell is grown.
  • the host cell is a Chinese hamster ovary cell.
  • the present disclosure provides an antibody or antigen-binding fragment that is a product of the method discussed above.
  • the present disclosure provides a polypeptide comprising: (a) CDR-H1, CDR-H2, and CDR-H3 of an HCVR domain of an antibody or antigen-binding fragment that comprises an HCVR amino acid sequence set forth in Table 1; or (b) CDR-L1, CDR-L2, and CDR-L3 of an LCVR domain of an immunoglobulin chain that comprises an LCVR amino acid sequence set forth in Table 1.
  • the present disclosure provides a polynucleotide encoding the polypeptide discussed above.
  • the present disclosure provides a vector comprising the polynucleotide discussed above.
  • the present disclosure provides a host cell comprising the antibody or antigen-binding fragment or polypeptide or polynucleotide or vector as discussed above or herein.
  • the present disclosure provides a composition or kit comprising the antibody or antigen-binding fragment discussed above or herein in association with a further therapeutic agent.
  • the present disclosure provides a pharmaceutical composition comprising the antigen-binding protein, antibody or antigen-binding fragment discussed above or herein and a pharmaceutically acceptable carrier and, optionally, a further therapeutic agent.
  • the further therapeutic agent is an anti-viral drug or a vaccine.
  • the further therapeutic agent is selected from the group consisting of: an anti- inflammatory agent, an antimalarial agent, an antibody or antigen-binding fragment thereof that specifically binds TMPRSS2, and an antibody or antigen-binding fragment thereof that specifically binds to CoV-S.
  • the antimalarial agent is chloroquine or hydroxychloroquine.
  • the anti-inflammatory agent is an antibody, such as sarilumab, tocilizumab, or gimsilumab.
  • the further therapeutic agent is a second antibody or antigen-binding fragment comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences of Table 1.
  • the present disclosure provides a vessel or injection device comprising the antigen-binding protein, antibody or antigen-binding fragment, or composition as discussed above or herein.
  • the present disclosure provides a method for treating or preventing infection with a coronavirus, in a subject in need thereof, comprising administering a therapeutically effective amount of an antigen-binding protein, antibody or antigen-binding fragment as discussed above or herein.
  • the coronavirus is selected from the group consisting of SARS-CoV-2, SARS-CoV, and MERS-CoV.
  • the subject is administered one or more further therapeutic agents.
  • the one or more further therapeutic agents is an anti-viral drug or a vaccine.
  • the one or more further therapeutic agents is selected from the group consisting of: an anti- inflammatory agent, an antimalarial agent, an antibody or antigen-binding fragment thereof that specifically binds TMPRSS2, and an antibody or antigen-binding fragment thereof that specifically binds to CoV-S.
  • the antimalarial agent is chloroquine or hydroxychloroquine.
  • the anti-inflammatory agent is an antibody, such as for example, sarilumab, tocilizumab, or gimsilumab.
  • the further therapeutic agent is a second antibody or antigen-binding fragment comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences of Table 1.
  • Other antibodies that can be used alone or in combination with one another or with one or more of the antibodies disclosed herein for use in the context of the methods of the present disclosure include, e.g., LY-CoV555 (Eli Lilly); 47D11 (Wang et al Nature Communications Article No.2251); B38, H4, B5 and/or H2 (Wu et al., 10.1126/science.abc2241 (2020); STI-1499 (Sorrento Therapeutics); VIR-7831 and VIR-7832 (Vir Biotherapeutics).
  • the present disclosure provides a method for administering an antibody or antigen-binding fragment discussed above or herein into the body of a subject comprising injecting the antibody or antigen-binding fragment into the body of the subject.
  • the antibody or antigen-binding fragment is injected into the body of the subject subcutaneously, intravenously or intramuscularly.
  • the antibody or antigen- binding binding fragment comprises a VH3-66 or Vk1-33 variable domain sequence.
  • the present disclosure provides an isolated antibody or antigen-binding fragment thereof that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody or antigen-binding fragment comprises three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 210.
  • CDRs heavy chain complementarity determining regions
  • LCVR light chain complementarity determining regions
  • the HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 204
  • the HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 206
  • the HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 208
  • the LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 212
  • the LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 55
  • the LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 214.
  • the isolated antibody or antigen-binding fragment thereof comprises an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202.
  • the isolated antibody or antigen-binding fragment thereof comprises an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210. In some embodiments, the isolated antibody or antigen-binding fragment thereof comprises an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210.
  • the present disclosure provides an isolated antibody that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises an immunoglobulin constant region, three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 210.
  • CDRs heavy chain complementarity determining regions
  • LCVR light chain complementarity determining regions
  • the HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 204
  • the HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 206
  • the HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 208
  • the LCDR1 comprises
  • the isolated antibody comprises an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210.
  • the isolated antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 216 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 218.
  • the immunoglobulin constant region is an IgG1 constant region.
  • the isolated antibody is a recombinant antibody. In some cases, the isolated antibody is multispecific.
  • the present disclosure provides a pharmaceutical composition comprising an isolated antibody as discussed above or herein, and a pharmaceutically acceptable carrier or diluent.
  • the pharmaceutical composition further comprises a second therapeutic agent.
  • the second therapeutic agent is selected from the group consisting of: a second antibody, or an antigen-binding fragment thereof, that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, an anti- inflammatory agent, an antimalarial agent, and an antibody or antigen-binding fragment thereof that binds TMPRSS2.
  • the second therapeutic agent is a second antibody, or an antigen- binding fragment thereof, that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832.
  • the second antibody or antigen-binding fragment thereof comprises three heavy chain CDRs (HCDR1, HCDR2 and HCDR3) contained within an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 640, and three light chain CDRs (LCDR1, LCDR2 and LCDR3) contained within an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 646.
  • the second antibody or antigen-binding fragment thereof comprises: HCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 642; HCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 499; HCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 644; LCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 648; LCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 650; and LCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 652.
  • the second antibody or antigen-binding fragment thereof comprises an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 640 and an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 646. In some cases, the second antibody or antigen-binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 654 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 656.
  • the present disclosure provides an isolated antibody or antigen-binding fragment thereof that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody or antigen-binding fragment comprises three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 640, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 646.
  • CDRs heavy chain complementarity determining regions
  • LCVR light chain complementarity determining regions
  • the HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 642
  • the HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 499
  • the HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 644
  • the LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 648
  • the LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 650
  • the LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 652.
  • the isolated antibody or antigen-binding fragment thereof comprises an HCVR that comprises an amino acid sequence set forth in SEQ ID NO: 640.
  • the isolated antibody or antigen-binding fragment thereof comprises an LCVR that comprises an amino acid sequence set forth in SEQ ID NO: 646. In some embodiments, the isolated antibody or antigen-binding fragment thereof comprises an HCVR that comprises an amino acid sequence set forth in SEQ ID NO: 640 and an LCVR that comprises an amino acid sequence set forth in SEQ ID NO: 646.
  • the present disclosure provides an isolated antibody that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises an immunoglobulin constant region, three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises an immunoglobulin constant region, three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises an immunoglobulin constant region, three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises
  • the HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 642
  • the HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 499
  • the HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 644
  • the LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 648
  • the LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 650
  • the LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 652.
  • the isolated antibody comprises an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 640 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 646.
  • the isolated antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 654 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 656.
  • the immunoglobulin constant region is an IgG1 constant region.
  • the isolated antibody is a recombinant antibody.
  • the isolated antibody is multispecific.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising an isolated antibody, as discussed above or herein, and a pharmaceutically acceptable carrier or diluent.
  • the pharmaceutical composition further comprising a second therapeutic agent.
  • the second therapeutic agent is selected from the group consisting of: a second antibody, or an antigen-binding fragment thereof, that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, an anti- inflammatory agent, an antimalarial agent, and an antibody or antigen-binding fragment thereof that binds TMPRSS2.
  • the second therapeutic agent is a second antibody, or an antigen- binding fragment thereof, that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832.
  • the second antibody or antigen-binding fragment thereof comprises three heavy chain CDRs (HCDR1, HCDR2 and HCDR3) contained within an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain CDRs (LCDR1, LCDR2 and LCDR3) contained within an LCVR comprising the
  • the second antibody or antigen-binding fragment thereof comprises: HCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 204; HCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 206; HCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 208 LCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 212; LCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 55; and LCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 214.
  • the second antibody or antigen-binding fragment thereof comprises an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 210. In some cases, the second antibody or antigen-binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 216 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 218. [00044] In various embodiments, any of the features or components of embodiments discussed above or herein may be combined, and such combinations are encompassed within the scope of the present disclosure.
  • Figure 1 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.
  • Figure 2 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.
  • Figure 3 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.
  • Figure 4 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.
  • Figure 5 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.
  • Figure 6 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.
  • Figure 7 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.
  • Figure 8 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.
  • Figure 10A and Figure 10B display neutralization potency.
  • Figure 10A displays the neutralization potency of anti-SARS-CoV-2 Spike mAbs.
  • Serial dilutions of anti-Spike mAbs, IgG1 isotype control, and recombinant dimeric ACE2 (hACE2.hFc) were added with pVSV- SARS-CoV-2-S-mNeon to Vero cells and mNeon expression was measured 24 hours post- infection as a read-out for virus infectivity.
  • Figure 10B displays neutralization potency of individual anti-Spike mAbs and combinations of mAbs against SARS-CoV-2-S virus in VeroE6 cells.
  • Figure 11 displays epitope bin analysis from a matrix of pre-mix binding assays for different anti-SARS-CoV-2 mAbs. Epitope binning was performed against nine anti-SARS- CoV-2 mAb as described. There were three phases (I, II, and III) for each graph. In phase I anti-
  • FIG. 12 displays a 3D surface model for the structure of the Spike protein RBD domain showing the ACE2 interface and HDX-MS epitope mapping results. RBD residues protected by anti-SARS-CoV2-Spike antibodies are indicated with shading that represent the extent of protection as determined by HDX-MS experiments.
  • FIG. 13A and Figure 13B display a complex of mAb10933 and mAb10987 with the SARS-CoV-2 RBD.
  • Figure 13A displays a 3.9 ⁇ cryoEM map of mAb10933 + RBD + mAb10987 complex, shaded according to the chains in the refined model of Figure 13B.
  • RBD, mAb10933 heavy and light chains, and mAb10987 heavy and light chain are identified.
  • Figure 14 displays cryoEM data statistics. Data collection and refinement statistics are reported for the mAb10987 + mAb10933 + SARS-CoV-2 RBD complex structure shown in Figure 13A and Figure 13B.
  • coronavirus refers to any virus of the coronavirus family, including but not limited to SARS-CoV-2, MERS-CoV, and SARS-CoV.
  • SARS-CoV-2 refers to any virus of the coronavirus family, including but not limited to SARS-CoV-2, MERS-CoV, and SARS-CoV.
  • SARS-CoV-2 refers to any virus of the coronavirus family, including but not limited to SARS-CoV-2, MERS-CoV, and SARS-CoV.
  • SARS-CoV-2 refers to any virus of the coronavirus family, including but not limited to SARS-CoV-2, MERS-CoV, and SARS-CoV.
  • SARS-CoV-2 refers to any virus of the coronavirus family, including but not limited to SARS-CoV-2, MERS-CoV, and SARS-CoV.
  • SARS-CoV-2 refers to any virus of the coronavirus family
  • SARS-CoV-2 has also been known as 2019-nCoV and Wuhan coronavirus. It binds via the viral spike protein to human host cell receptor angiotensin-converting enzyme 2 (ACE2). The spike protein also binds to and is cleaved by TMPRSS2, which activates the spike protein for membrane fusion of the virus.
  • ACE2 human host cell receptor angiotensin-converting enzyme 2
  • CoV-S also called “S” or “S protein” refers to the spike protein of a coronavirus, and can refer to specific S proteins such as SARS-CoV-2-S, MERS-CoV S, and SARS-CoV S.
  • SARS-CoV-2-Spike protein is a 1273 amino acid type I membrane glycoprotein which assembles into trimers that constitute the spikes or peplomers on the surface of the enveloped coronavirus particle.
  • the protein has two essential functions, host receptor binding and membrane fusion, which are attributed to the N-terminal (S1) and C-terminal (S2) halves of the S protein.
  • CoV-S binds to its cognate receptor via a receptor binding domain (RBD) present in the S1 subunit.
  • RBD receptor binding domain
  • the amino acid sequence of full-length SARS-CoV-2 spike protein is exemplified by the amino acid sequence provided in SEQ ID NO: 832.
  • the term “CoV-S” includes protein variants of CoV spike protein isolated from different CoV isolates as well as recombinant CoV spike protein or a fragment thereof. The term also encompasses CoV spike protein or a fragment thereof coupled to, for example, a histidine tag, mouse or human Fc, or a signal sequence such as ROR1.
  • coronavirus infection refers to infection with a coronavirus such as SARS-CoV-2, MERS-CoV, or SARS-CoV.
  • the term includes coronavirus respiratory tract infections, often in the lower respiratory tract. Symptoms can include high fever, dry cough, shortness of breath, pneumonia, gastro-intestinal symptoms such as diarrhea, organ failure (kidney failure and renal dysfunction), septic shock, and death in severe cases.
  • Viruses [00064] The present invention includes methods for treating or preventing a viral infection in a subject.
  • virus includes any virus whose infection in the body of a subject is treatable or preventable by administration of an anti-CoV-S antibody or antigen-binding fragment thereof (e.g., wherein infectivity of the virus is at least partially dependent on CoV-S).
  • an anti-CoV-S antibody or antigen-binding fragment thereof e.g., wherein infectivity of the virus is at least partially dependent on CoV-S.
  • a “virus” is any virus that expresses spike protein (e.g., CoV-S).
  • the term “virus” also includes a CoV-S-dependent respiratory virus which is a virus that infects the respiratory tissue of a subject (e.g., upper and/or lower respiratory tract, trachea, bronchi, lungs) and is treatable or preventable by administration of an anti-CoV-S antibody or antigen- binding fragment thereof.
  • virus includes coronavirus, SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), SARS-CoV (severe acute respiratory syndrome coronavirus), and MERS-CoV (Middle East respiratory syndrome (MERS) coronavirus).
  • Coronaviruses can include the genera of alphacoronaviruses, betacoronaviruses, gammacoronaviruses, and deltacoronaviruses.
  • the antibodies or antigen-binding fragments provided herein can bind to and/or neutralize an alphacoronavirus, a betacoronavirus, a gammacoronavirus, and/or a deltacoronavirus. In certain embodiments, this binding and/or neutralization can be specific for a particular genus of coronavirus or for a particular subgroup of a genus.
  • “Viral infection” refers to the invasion and multiplication of a virus in the body of a subject.
  • Coronavirus virions are spherical with diameters of approximately 125 nm. The most prominent feature of coronaviruses is the club-shape spike projections emanating from the surface of the virion. These spikes are a defining feature of the virion and give them the appearance of a solar corona, prompting the name, coronaviruses. Within the envelope of the virion is the nucleocapsid. Coronaviruses have helically symmetrical nucleocapsids, which is uncommon among positive-sense RNA viruses, but far more common for negative-sense RNA viruses. SARS-CoV-2, MERS-CoV, and SARS-CoV belong to the coronavirus family.
  • the initial attachment of the virion to the host cell is initiated by interactions between the S protein and its receptor.
  • the sites of receptor binding domains (RBD) within the S1 region of a coronavirus S protein vary depending on the virus, with some having the RBD at the C-terminus of S1.
  • the S-protein/receptor interaction is the primary determinant for a coronavirus to infect a host species and also governs the tissue tropism of the virus.
  • Many coronaviruses utilize peptidases as their cellular receptor. Following receptor binding, the virus must next gain access to the host cell cytosol. This is generally accomplished by acid-dependent proteolytic cleavage of S protein by a cathepsin, TMPRRS2 or another protease, followed by fusion of the viral and cellular membranes.
  • the present invention provides antigen-binding proteins, such as antibodies and antigen-binding fragments thereof, that specifically bind to CoV spike protein or an antigenic fragment thereof.
  • antigen-binding proteins such as antibodies and antigen-binding fragments thereof, that specifically bind to CoV spike protein or an antigenic fragment thereof.
  • antibody refers to immunoglobulin molecules comprising four polypeptide chains, two heavy chains (HCs) and two light chains (LCs) inter-connected by disulfide bonds (i.e., "full antibody molecules"), as well as multimers thereof (e.g. IgM).
  • Exemplary antibodies include, for example, those listed in Table 1.
  • Each heavy chain comprises a heavy chain variable region (“HCVR” or “V H ”) and a heavy chain constant region (comprised of domains C H 1, C H 2 and C H 3).
  • Each light chain is comprised of a light chain variable region (“LCVR or “VL”) and a light chain constant region (CL).
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each V H and VL comprises three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • Heavy chain CDRs can also be referred to as HCDRs or CDR-Hs, and numbered as described above (e.g., HCDR1, HCDR2, and HCDR3 or CDR-H1, CDR-H2, and CDR-H3).
  • light chain CDRs can be referred to as LCDRs or CDR-Ls, and numbered LCDR1, LCDR2, and LCDR3, or CDR-L1, CDR-L2, and CDR-L3.
  • the FRs of the antibody (or antigen binding fragment thereof) are identical to the human germline sequences, or are naturally or artificially modified.
  • Exemplary human germline sequences include, but are not limited to, VH3-66 and Vk1-33.
  • anti-CoV-S antibodies or antigen- binding fragments thereof comprising HCDR and LCDR sequences of Table 1 within a VH3-66 or Vk1-33 variable heavy chain or light chain region.
  • the present disclosure further provides anti-CoV-S antibodies or antigen-binding fragments thereof (e.g., anti-SARS-CoV-2-S antibodies or antigen- binding fragments thereof) comprising HCDR and LCDR sequences of Table 1 within a combination of a light chain selected from IgKV4-1, IgKV 1-5, IgKV1-9, IgKV1-12, IgKV3-15, IgKV1-16, IgKV1-17, IgKV3-20, IgLV3-21, IgKV2-24, IgKV1-33, IgKV1-39, IgLV1-40, IgLV1-44, IgLV1-51, IgLV3-1, IgKV1-6, IgLV2-8, IgKV3-11, IgLV2-11, IgLV2-14, IgLV2-23, or IgLV6-57, and a heavy chain selected from IgHV1-69, IgHV3-64, IgHV4-59, IgHV3-53,
  • the present disclosure further provides anti-CoV-S antibodies or antigen-binding fragments thereof (e.g., anti-SARS-CoV-2-S antibodies or antigen-binding fragments thereof) comprising HCVR and LCVR sequences of Table 1 within a combination of a light chain selected from IgKV4-1, IgKV 1-5, IgKV1-9, IgKV1-12, IgKV3-15, IgKV1-16, IgKV1-17, IgKV3-20, IgLV3-21, IgKV2-24, IgKV1-33, IgKV1-39, IgLV1-40, IgLV1-44, IgLV1-51, IgLV3-1, IgKV1-6, IgLV2-8, IgKV3- 11, IgLV2-11, IgLV2-14, IgLV2-23, or IgLV6-57, and a heavy chain selected from IgHV1-69, IgHV3-64, IgHV4-59, IgHV3-53, IgH
  • variable domains of both the heavy and light immunoglobulin chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), located within relatively conserved framework regions (FR).
  • CDRs complementarity determining regions
  • FR framework regions
  • both light and heavy chains variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • the assignment of amino acids to each domain is in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al.; National Institutes of Health, Bethesda, Md.; 5 th ed.; NIH Publ. No.91-3242 (1991); Kabat (1978) Adv. Prot.
  • the present invention includes monoclonal anti-CoV-S antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, as well as monoclonal compositions comprising a plurality of isolated monoclonal antigen-binding proteins.
  • monoclonal antibody refers to a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts.
  • a "plurality" of such monoclonal antibodies and fragments in a composition refers to a concentration of identical (i.e., as discussed above, in amino acid sequence except for possible naturally occurring
  • an anti-CoV-S antigen-binding protein e.g., antibody or antigen-binding fragment comprises a heavy chain constant domain, e.g., of the type IgA (e.g., IgA1 or IgA2), IgD, IgE, IgG (e.g., IgG1, IgG2, IgG3 and IgG4) or IgM.
  • an antigen-binding protein e.g., antibody or antigen-binding fragment comprises a light chain constant domain, e.g., of the type kappa or lambda.
  • the term "human” antigen-binding protein, such as an antibody, as used herein, includes antibodies having variable and constant regions derived from human germline immunoglobulin sequences whether in a human cell or grafted into a non-human cell, e.g., a mouse cell. See e.g., US8502018, US6596541 or US5789215.
  • the human mAbs of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and, in particular, CDR3.
  • human antibody as used herein, is not intended to include mAbs in which CDR sequences derived from the germline of another mammalian species (e.g., mouse) have been grafted onto human FR sequences.
  • the term includes antibodies recombinantly produced in a non-human mammal or in cells of a non-human mammal.
  • the term is not intended to include antibodies isolated from or generated in a human subject. See below.
  • the present invention includes anti-CoV-S chimeric antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, and methods of use thereof.
  • a "chimeric antibody” is an antibody having the variable domain from a first antibody and the constant domain from a second antibody, where the first and second antibodies are from different species. (US4816567; and Morrison et al., (1984) Proc. Natl. Acad. Sci. USA 81: 6851-6855).
  • the present invention includes anti-CoV-S hybrid antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, and methods of use thereof.
  • hybrid antibody is an antibody having the variable domain from a first antibody and the constant domain from a second antibody, wherein the first and second antibodies are from different animals, or wherein the variable domain, but not the constant region, is from a first animal.
  • a variable domain can be taken from an antibody isolated from a human
  • hybrid antibodies are described in Example 1, which refers to antibody heavy chain variable region and light chain variable region derived PCR products that were cloned into expression vectors containing a heavy constant region and a light constant region, respectively.
  • Hybrid antibodies are synthetic and non-natrually occurring because the variable and constant regions they contain are not isolated from a single natural source.
  • the term “recombinant” antigen-binding proteins, such as antibodies or antigen-binding fragments thereof refers to such molecules created, expressed, isolated or obtained by technologies or methods known in the art as recombinant DNA technology which include, e.g., DNA splicing and transgenic expression.
  • the term includes antibodies expressed in a non- human mammal (including transgenic non-human mammals, e.g., transgenic mice), or a cell (e.g., CHO cells) expression system, or a non-human cell expression system, or isolated from a recombinant combinatorial human antibody library.
  • a recombinant antibody shares a sequence with an antibody isolated from an organism (e.g., a mouse or a human), but has been expressed via recombinant DNA technology.
  • Such antibodies may have post-translational modifications (e.g., glycosylation) that differ from the antibody as isolated from the organism.
  • Recombinant anti-CoV-S antigen-binding proteins e.g., antibodies and antigen-binding fragments, disclosed herein may also be produced in an E. coli/T7 expression system.
  • nucleic acids encoding the anti-CoV-S antibody immunoglobulin molecules of the invention e.g., as found in Table 1 may be inserted into a pET-based plasmid and expressed in the E. coli/T7 system.
  • the present invention includes methods for expressing an antibody or antigen-binding fragment thereof or immunoglobulin chain thereof in a host cell (e.g., bacterial host cell such as E.
  • a bacterial host cell such as an E. coli, includes a polynucleotide encoding the T7 RNA polymerase gene operably linked to a lac promoter and expression of the polymerase and the chain is induced by incubation of the host cell with IPTG (isopropyl-beta-D- thiogalactopyranoside). See US4952496 and US5693489 or Studier & Moffatt, Use of
  • Transformation can be by any known method for introducing polynucleotides (e.g., DNA or RNA, including mRNA) into a host cell.
  • polynucleotides e.g., DNA or RNA, including mRNA
  • nucleic acid molecules may be introduced into mammalian cells by viral vectors such as lentivirus or adeno-associated virus. Methods of transforming cells are well known in the art. See, for example, U.S. Pat.
  • an antibody or antigen-binding fragment thereof of the present disclosure can be introduced to a subject in nucleic acid form (e.g, DNA or RNA, including mRNA), such that the subject’s own cells produce the antibody.
  • nucleic acid form e.g, DNA or RNA, including mRNA
  • the present disclosure further provides modifications to nucleotide sequences encoding the anti-CoV-S antibodies described herein that result in increased antibody expression, increased antibody stability, increased nucleic acid (e.g., mRNA) stability, or improved affinity or specificity of the antibodies for the CoV spike protein.
  • the present invention includes recombinant methods for making an anti-CoV-S antigen-binding protein, such as an antibody or antigen-binding fragment thereof of the present invention, or an immunoglobulin chain thereof, comprising (i) introducing one or more polynucleotides (e.g., including the nucleotide sequence of any one or more of the sequences of Table 2) encoding light and/or heavy immunoglobulin chains, or CDRs, of the antigen-binding protein, e.g., of Table 1, for example, wherein the polynucleotide is in a vector; and/or integrated into a host cell chromosome and/or is operably linked to a promoter; (ii) culturing the host cell (e.g., CHO or Pichia or Pichia pastoris) under condition favorable to expression of the polynucleotide and, (iii) optionally, isolating the antigen-binding protein, (e.g.,
  • insertion with a vector such as adeno-associated virus (AAV), e.g., after cleavage of the chromosome using a gene editing system (e.g., CRISPR (for example, CRISPR-Cas9), TALEN, megaTAL, zinc finger, or Argonaute).
  • AAV adeno-associated virus
  • Targeted insertions can take place, for example, at host cell loci such as an albumin or immunoglopbulin genomic locus.
  • insertion can be at a random locus, e.g., using a vector such as lentivirus.
  • an antigen-binding protein e.g., antibody or antigen-binding fragment
  • an immunoglobulin chain e.g., an antibody that comprises two heavy immunoglobulin chains and two light immunoglobulin chains
  • co-expression of the chains in a single host cell leads to association of the chains, e.g., in the cell or on the cell surface or outside the cell if such chains are secreted, so as to form the antigen-binding protein (e.g., antibody or antigen-binding fragment).
  • the methods include those wherein only a heavy immunoglobulin chain or only a light immunoglobulin chain (e.g., any of those discussed herein including mature fragments and/or variable domains thereof) is expressed.
  • the present invention also includes anti-CoV-S antigen-binding proteins, such as antibodies and antigen-binding fragments thereof, comprising a heavy chain immunoglobulin (or variable domain thereof or comprising the CDRs thereof) encoded by a polynucleotide comprising a nucleotide sequence set forth in Table 2 and a light chain immunoglobulin (or variable domain thereof or comprising the CDRs thereof) encoded by a nucleotide sequence set forth in Table 2 which are the product of such production methods, and, optionally, the purification methods set forth herein.
  • anti-CoV-S antigen-binding proteins such as antibodies and antigen-binding fragments thereof, comprising a heavy chain immunoglobulin (or variable domain thereof or comprising the CDRs thereof) encoded by a polynucleotide comprising a nucleotide sequence set forth in Table 2 and a light chain immunoglobulin (or variable domain thereof or comprising the CDRs thereof) encoded by a nucleot
  • the product of the method is an anti-CoV-S antigen-binding protein which is an antibody or fragment comprising an HCVR comprising an amino acid sequence set forth in Table 1 and an LCVR comprising an amino acid sequence set forth in Table 1, wherein the HCVR and LCVR sequences are selected from a single antibody listed in Table 1.
  • the product of the method is an anti-CoV-S antigen-binding protein which is an antibody or fragment comprising HCDR1, HCDR2, and HCDR3 comprising amino acid sequences set forth in Table 1 and LCDR1, LCDR2, and LCDR3 comprising amino acid sequences set forth in Table 1, wherein the six CDR sequences are selected from a single antibody listed in Table 1.
  • the product of the method is an anti-CoV-S antigen-binding protein which is an antibody or fragment
  • Eukaryotic and prokaryotic host cells including mammalian cells, may be used as hosts for expression of an anti-CoV-S antigen-binding protein.
  • host cells are well known in the art and many are available from the American Type Culture Collection (ATCC).
  • These host cells include, inter alia, Chinese hamster ovary (CHO) cells, NS0, SP2 cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells, 3T3 cells, HEK-293 cells and a number of other cell lines.
  • Mammalian host cells include human, mouse, rat, dog, monkey, pig, goat, bovine, horse and hamster cells.
  • Other cell lines that may be used are insect cell lines (e.g., Spodoptera frugiperda or Trichoplusia ni), amphibian cells, bacterial cells, plant cells and fungal cells.
  • Fungal cells include yeast and filamentous fungus cells including, for example, Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia minuta (Ogataea minuta, Pichia lindneri), Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei, Chrysosporium
  • the present invention includes an isolated host cell (e.g., a CHO cell) comprising an antigen-binding protein, such as those of Table 1; or a polynucleotide encoding such a polypeptide thereof.
  • an antigen-binding protein e.g., mAbs
  • mAbs a binding affinity to an antigen
  • an antigen such as a CoV-S protein (e.g., SARS-CoV-2-S)
  • KD e.g., SARS-CoV-2-S
  • the present invention includes antigen-binding proteins that specifically bind to a CoV-S protein.
  • antigen-binding portion or “antigen-binding fragment” of an antibody or antigen-binding protein, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.
  • antigen-binding portion or “antigen-binding fragment” of an antibody or antigen-binding protein, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.
  • antigen-binding portion or “antigen-binding fragment” of an antibody or antigen-binding protein, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.
  • fragments include: (i) Fab fragments; (ii) F(ab')2 fragments; (iii) Fd fragments; (iv) Fv fragments; (v) single-chain Fv (scFv) molecules; (vi) dAb fragments; and (vii) minimal recognition units consisting of the amino acid residues that mimic the hypervariable region of an antibody (e.g., an isolated complementarity determining region (CDR) such as a CDR3 peptide), or a constrained FR3-CDR3-FR4 peptide.
  • CDR complementarity determining region
  • engineered molecules such as domain-specific antibodies, single domain antibodies, domain-deleted antibodies, chimeric antibodies, CDR- grafted antibodies, diabodies, triabodies, tetrabodies, minibodies, nanobodies (e.g., as defined in WO08/020079 or WO09/138519) (e.g., monovalent nanobodies, bivalent nanobodies, etc.), small modular immunopharmaceuticals (SMIPs), and shark variable IgNAR domains, are also encompassed within the expression "antigen-binding fragment," as used herein.
  • SMIPs small modular immunopharmaceuticals
  • the antigen-binding fragment comprises three or more CDRs of an antibody of Table 1 (e.g., CDR-H1, CDR-H2 and CDR-H3; or CDR-L1, CDR-L2 and CDR-L3).
  • An antigen-binding fragment of an antibody will, in an embodiment of the invention, comprise at least one variable domain.
  • the variable domain may be of any size or amino acid composition and will generally comprise at least one CDR, which is adjacent to or in frame with one or more framework sequences.
  • the VH and VL domains may be situated relative to one another in any suitable arrangement.
  • variable region may be dimeric and contain VH - VH, VH - VL or V L - V L dimers.
  • the antigen-binding fragment of an antibody may contain a monomeric VH or VL domain.
  • an antigen-binding fragment of an antibody may contain at least one variable domain covalently linked to at least one constant domain.
  • Non-limiting, exemplary configurations of variable and constant domains that may be found within an antigen- binding fragment of an antibody of the present invention include: (i) VH-CH1; (ii) VH-CH2; (iii) V H -C H 3; (iv) V H -C H 1-C H 2; (v) V H -C H 1-C H 2-C H 3; (vi) V H -C H 2-C H 3; (vii) V H -C L ; (viii) V L -C H 1; (ix) V L -C H 2; (x) V L -C H 3; (xi) V L -C H 1-C H 2; (xii) V L -C H 1-C H 2-C H 3; (xiii) V L -C H 2-C H 3; and (xiv) VL-CL.
  • variable and constant domains may be either directly linked to one another or may be linked by a full or partial hinge or linker region.
  • a hinge region may consist of at least 2 (e.g., 5, 10, 15, 20, 40, 60 or more) amino acids, which result in a flexible or semi-flexible linkage between adjacent variable and/or constant domains in a single polypeptide
  • an antigen-binding fragment of an antibody of the present invention may comprise a homo-dimer or hetero-dimer (or other multimer) of any of the variable and constant domain configurations listed above in non-covalent association with one another and/or with one or more monomeric VH or VL domain (e.g., by disulfide bond(s)).
  • Antigen-binding proteins e.g., antibodies and antigen-binding fragments
  • antibody or antibody fragments of the invention may be conjugated to a moiety such a ligand or a therapeutic moiety (“immunoconjugate”), such as an anti-viral drug, a second anti-influenza antibody, or any other therapeutic moiety useful for treating a viral infection, e.g., influenza viral infection. See below.
  • the present invention also provides a complex comprising an anti-CoV-S antigen- binding protein, e.g., antibody or antigen-binding fragment, discussed herein complexed with CoV-S polypeptide or an antigenic fragment thereof and/or with a secondary antibody or antigen-binding fragment thereof (e.g., detectably labeled secondary antibody) that binds specifically to the anti-CoV-S antibody or fragment.
  • an anti-CoV-S antigen- binding protein e.g., antibody or antigen-binding fragment
  • the antibody or fragment is in vitro (e.g., is immobilized to a solid substrate) or is in the body of a subject.
  • the CoV-S is in vitro (e.g., is immobilized to a solid substrate) or is on the surface of a virus or is in the body of a subject.
  • Immobilized anti-CoV-S antibodies and antigen-binding fragments thereof which are covalently linked to an insoluble matrix material (e.g., glass or polysaccharide such as agarose or sepharose, e.g., a bead or other particle thereof) are also part of the present invention; optionally, wherein the immobilized antibody is complexed with CoV-S or antigenic fragment thereof or a secondary antibody or fragment thereof.
  • isolated antigen-binding proteins antibodies or antigen-binding fragments thereof, polypeptides, polynucleotides and vectors, are at least partially free of other biological molecules from the cells or cell culture from which they are produced.
  • biological molecules include nucleic acids, proteins, other antibodies or antigen-binding fragments, lipids, carbohydrates, or other material such as cellular debris and growth medium.
  • An isolated antibody or antigen- binding fragment may further be at least partially free of expression system components such as biological molecules from a host cell or of the growth medium thereof.
  • expression system components such as biological molecules from a host cell or of the growth medium thereof.
  • epitope refers to an antigenic determinant (e.g., a CoV-S polypeptide) that interacts with a specific antigen-binding site of an antigen-binding protein, e.g., a variable region of an antibody molecule, known as a paratope.
  • a specific antigen-binding site of an antigen-binding protein e.g., a variable region of an antibody molecule, known as a paratope.
  • a single antigen may have more than one epitope.
  • different antibodies may bind to different areas on an antigen and may have different biological effects.
  • epitopes also refers to a site on an antigen to which B and/or T cells respond. It also refers to a region of an antigen that is bound by an antibody.
  • Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction. Epitopes may be linear or conformational, that is, composed of non-linear amino acids.
  • epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.
  • Methods for determining the epitope of an antigen-binding protein include alanine scanning mutational analysis, peptide blot analysis (Reineke (2004) Methods Mol. Biol.248: 443-63), peptide cleavage analysis, crystallographic studies and NMR analysis.
  • methods such as epitope excision, epitope extraction and chemical modification of antigens can be employed (Tomer (2000) Prot. Sci.9: 487-496).
  • Another method that can be used to identify the amino acids within a polypeptide with which an antigen-binding protein e.g., antibody or fragment or polypeptide
  • an antigen-binding protein e.g., antibody or fragment or polypeptide
  • the hydrogen/deuterium exchange method involves deuterium-labeling the protein of interest, followed by binding the antigen-binding protein, e.g., antibody or fragment or polypeptide, to the deuterium-labeled protein.
  • the CoV-S protein/ antigen-binding protein 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.
  • amino acids that form part of the protein/ antigen-binding protein interface may retain deuterium and
  • the target protein After dissociation of the antigen-binding protein (e.g., antibody or fragment or polypeptide), the target protein is subjected to protease cleavage and mass spectrometry analysis, thereby revealing the deuterium-labeled residues which correspond to the specific amino acids with which the antigen-binding protein interacts. See, e.g., Ehring (1999) Analytical Biochemistry 267: 252-259; Engen and Smith (2001) Anal. Chem.73: 256A-265A.
  • the term “competes” as used herein refers to an antigen-binding protein (e.g., antibody or antigen-binding fragment thereof) that binds to an antigen (e.g., CoV-S) and inhibits or blocks the binding of another antigen-binding protein (e.g., antibody or antigen-binding fragment thereof) to the antigen.
  • an antigen e.g., CoV-S
  • another antigen-binding protein e.g., antibody or antigen-binding fragment thereof
  • the term also includes competition between two antigen-binding proteins e.g., antibodies, in both orientations, i.e., a first antibody that binds and blocks binding of second antibody and vice versa.
  • the first antigen-binding protein (e.g., antibody) and second antigen-binding protein (e.g., antibody) may bind to the same epitope.
  • the first and second antigen-binding proteins (e.g., antibodies) may bind to different, but, for example, overlapping epitopes, wherein binding of one inhibits or blocks the binding of the second antibody, e.g., via steric hindrance.
  • Competition between antigen-binding proteins (e.g., antibodies) may be measured by methods known in the art, for example, by a real- time, label-free bio-layer interferometry assay.
  • Epitope mapping (e.g., via alanine scanning or hydrogen-deuterium exchange (HDX)) can be used to determine whether two or more antibodies are non-competing (e.g., on a spike protein receptor binding domain (RBD) monomer), competing for the same epitope, or competing but with diverse micro-epitopes (e.g., identified through HDX).
  • HDX hydrogen-deuterium exchange
  • competition between a first and second anti- CoV-S antigen-binding protein is determined by measuring the ability of an immobilized first anti-CoV-S antigen-binding protein (e.g., antibody) (not initially complexed with CoV-S protein) to bind to soluble CoV-S protein complexed with a second anti-CoV-S antigen-binding protein (e.g., antibody).
  • the degree of competition can be expressed as a percentage of the reduction in binding. Such competition can be measured using a real time, label-free bio-
  • Binding competition between anti-CoV-S antigen-binding proteins can be determined using a real time, label-free bio-layer interferometry assay on an Octet RED384 biosensor (Pall ForteBio Corp.).
  • the anti-CoV-S mAb can be first captured onto anti-hFc antibody coated Octet biosensor tips (Pall ForteBio Corp., # 18-5060) by submerging the tips into a solution of anti-CoV-S mAb (subsequently referred to as “mAb1”).
  • mAb1 anti-CoV-S mAb
  • the antibody captured biosensor tips can then be saturated with a known blocking isotype control mAb (subsequently referred to as “blocking mAb”) by dipping into a solution of blocking mAb.
  • the biosensor tips can then be subsequently dipped into a co-complexed solution of CoV-S polypeptide and a second anti-CoV-S mAb (subsequently referred to as “mAb2”), that had been pre-incubated for a period of time and binding of mAb1 to the CoV-S polypeptide can be determined.
  • the biosensor tips can be washed in buffer in between every step of the experiment. The real-time binding response can be monitored during the course of the experiment and the binding response at the end of every step can be recorded.
  • the competition assay is conducted at 25 o C and pH about 7, e.g., 7.4, e.g., in the presence of buffer, salt, surfactant and a non-specific protein (e.g., bovine serum albumin).
  • a non-specific protein e.g., bovine serum albumin.
  • an antibody or antigen-binding fragment of the invention which is modified in some way retains the ability to specifically bind to CoV-S, e.g., retains at least 10% of its CoV-S binding activity (when compared to the parental antibody) when that activity is expressed on a molar basis.
  • an antibody or antigen-binding fragment of the invention retains at least 20%, 50%, 70%, 80%, 90%, 95% or 100% or more of the CoV-S binding affinity as the parental antibody. It is also intended that an antibody or antigen-binding fragment of the invention can include conservative or non-conservative amino acid substitutions (referred to as “conservative variants” or “function conserved variants” of the antibody) that do not substantially alter its biologic activity.
  • a “variant” of a polypeptide refers to a polypeptide, such as an immunoglobulin chain (e.g., mAb8021 VH, VL, HC, or LC, mAb8028 VH, VL, HC, or LC, or mAb8029 VH, VL, HC, or LC), refers to a polypeptide, such as an immunoglobulin chain (e.g., mAb8021 VH, VL, HC, or LC, mAb8028 VH, VL, HC, or LC, or mAb8029 VH, VL, HC, or LC), refers to a polypeptide, such as an immunoglobulin chain (e.g., mAb8021 VH, VL, HC, or LC, mAb8028 VH, VL, HC, or LC, or mAb8029 VH, VL, HC, or LC), refers to a polypeptide,
  • polypeptide comprising an amino acid sequence that is at least about 70-99.9% (e.g., 70, 72, 74, 75, 76, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.9%) identical or similar to a referenced amino acid sequence that is set forth herein (e.g., SEQ ID NO: 2, 10, 18, 20, 22, 30, 38, 40, 42, 50, 58, or 60); when the comparison is performed by a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences (e.g., expect threshold: 10; word size: 3; max matches in a query range: 0; BLOSUM 62 matrix; gap costs: existence 11, extension 1; conditional compositional score matrix adjustment).
  • a referenced amino acid sequence that is set forth
  • a “variant” of a polynucleotide refers to a polynucleotide comprising a nucleotide sequence that is at least about 70-99.9% (e.g., at least about 70, 72, 74, 75, 76, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or 99.9%) identical to a referenced nucleotide sequence that is set forth herein (e.g., SEQ ID NO: 1, 9, 17, 19, 21, 29, 37, 39, 41, 49, 57, or 59); when the comparison is performed by a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences (e.g., expect threshold: 10; word size: 28; max matches in a query range: 0
  • Anti-CoV-S antigen-binding proteins e.g., antibodies and antigen-binding fragments thereof of the present invention, in an embodiment of the invention, include a heavy chain immunoglobulin variable region having at least 70% (e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater) amino acid sequence identity to the HCVR amino acid sequences set forth in Table 1; and/or a light chain immunoglobulin variable region having at least 70% (e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater) amino acid sequence identity to the LCVR amino acid sequences set forth in Table 1.
  • a heavy chain immunoglobulin variable region having at least 70% (e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%,
  • a variant anti-CoV-S antigen-binding protein may include a polypeptide comprising an amino acid sequence that is set forth herein except for one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10) mutations such as, for example, missense mutations (e.g., conservative substitutions), non-sense mutations, deletions, or insertions.
  • the present invention includes antigen-binding proteins which include an immunoglobulin light chain variant comprising an LCVR amino acid sequence set forth in Table 1 but having one or more of such mutations and/or an immunoglobulin heavy chain variant comprising an HCVR amino acid sequence set forth in Table 1 but having one or more of such mutations.
  • a variant anti-CoV-S antigen-binding protein includes an immunoglobulin light chain variant comprising CDR-L1, CDR-L2 and CDR-L3 wherein one or more (e.g., 1 or 2 or 3) of such CDRs has one or more of such mutations (e.g., conservative substitutions) and/or an immunoglobulin heavy chain variant comprising CDR-H1, CDR-H2 and CDR-H3 wherein one or more (e.g., 1 or 2 or 3) of such CDRs has one or more of such mutations (e.g., conservative substitutions).
  • Substitutions can be in a CDR, framework, or constant region.
  • the invention further provides variant anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments thereof, comprising one or more variant CDRs (e.g., any one or more of CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and/or CDR-H3) that are set forth herein with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.9% sequence identity or similarity to, e.g., the heavy chain and light chain CDRs of Table 1.
  • variant CDRs e.g., any one or more of CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and/or CDR-H3
  • Embodiments of the present invention also include variant antigen-binding proteins, e.g., anti-CoV-S antibodies and antigen-binding fragments thereof, that comprise immunoglobulin VHs and VLs; or HCs and LCs, which comprise an amino acid sequence having 70% or more (e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater) overall amino acid sequence identity or similarity to the amino acid sequences of the corresponding VHs, VLs, HCs or LCs specifically set forth herein, but wherein the CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and CDR-H3 of such immunoglobulins are not variants and comprise CDR amino acid sequence set forth in Table 1.
  • variant antigen-binding proteins e.g., anti-CoV-S antibodies and antigen-binding fragments thereof, that comprise immunoglobulin V
  • variant antigen-binding proteins are not, themselves, variants.
  • Conservatively modified variant anti-CoV-S antibodies and antigen-binding fragments thereof are also part of the present invention.
  • a “conservatively modified variant” or a “conservative substitution” refers to a variant wherein there is one or more substitutions of amino acids in a polypeptide with other amino acids having similar characteristics (e.g. charge, side-chain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc.). Such changes can frequently be made without significantly disrupting the biological activity of the antibody or fragment.
  • Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The
  • substitutions of structurally or functionally similar amino acids are less likely to significantly disrupt biological activity.
  • groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartate and glutamate, and 7) sulfur-containing side chains: cysteine and methionine.
  • Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.
  • a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Science 256: 144345.
  • Function-conservative variants of the anti-CoV-S antibodies and antigen-binding fragments thereof are also part of the present invention.
  • variants of the anti-CoV-S antibodies and antigen-binding fragments thereof may be “function- conservative variants”. Such function-conservative variants may, in some cases, also be characterized as conservatively modified variants.
  • “Function-conservative variants,” as used herein, refers to variants of the anti-CoV-S antibodies or antigen-binding fragments thereof in which one or more amino acid residues have been changed without significantly altering one or more functional properties of the antibody or fragment.
  • a function-conservative variant anti-CoV-S antibody or antigen-binding fragment thereof of the present invention comprises a variant amino acid sequence and exhibits one or more of the following functional properties: ⁇ Inhibits growth of coronavirus (e.g., SARS-CoV-2, SARS-CoV, and/or MERS-CoV) in ACE2- and/or TMPRSS2-expressing cells (e.g., Calu-3 cells); ⁇ Does not significantly bind to MDCK/Tet-on cells which do not express ACE2 and/or TMPRSS2; ⁇ Limits spread of coronavirus infection (e.g., by SARS-CoV-2, SARS-CoV, and/or MERS-CoV) of cells, e.g., Calu-3, in vitro; and/or ⁇ Protects a mouse engineered to express the human TMPRSS2 and/or ACE2 protein from death caused by coronavirus infection (e.g., SARS-Co
  • coronavirus
  • MERS-CoV MERS-CoV
  • the mice are infected with an otherwise lethal dose of the virus, optionally when combined with a second therapeutic agent.
  • Protects a mouse engineered to express the human TMPRSS2 and/or ACE2 protein from weight loss caused by coronavirus infection (e.g., SARS-CoV-2, SARS-CoV, or MERS-CoV), for example, wherein the mice are infected with a dose of the virus that would otherwise cause weighht loss, optionally when combined with a second therapeutic agent.
  • coronavirus infection e.g., SARS-CoV-2, SARS-CoV, or MERS-CoV
  • a “neutralizing” or “antagonist” anti-CoV-S antigen-binding protein refers to a molecule that inhibits an activity of CoV-S to any detectable degree, e.g., inhibits the ability of CoV-S to bind to a receptor such as ACE2, to be cleaved by a protease such as TMPRSS2, or to mediate viral entry into a host cell or viral reproduction in a host cell.
  • Table 1 refers to antigen-binding proteins, such as antibodies and antigen-binding fragments thereof, that comprise the heavy chain or VH (or a variant thereof) and light chain or VL (or a variant thereof) as set forth below; or that comprise a VH that comprises the CDRs thereof (CDR-H1 (or a variant thereof), CDR-H2 (or a variant thereof) and CDR-H3 (or a variant thereof)) and a VL that comprises the CDRs thereof (CDR-L1 (or a variant thereof), CDR-L2 (or a variant thereof) and CDR-L3 (or a variant thereof)), e.g., wherein the immunoglobulin chains, variable regions and/or CDRs comprise the specific amino acid sequences described below.
  • the antibodies described herein also include embodiments wherein the VH is fused to a wild-type IgG4 (e.g., wherein residue 108 is S) or to IgG4 variants (e.g., wherein residue 108 is P).
  • Antibodies and antigen-binding fragments of the present invention comprise immunoglobulin chains including the amino acid sequences set forth herein as well as cellular and in vitro post-translational modifications to the antibody.
  • the present invention includes antibodies and antigen-binding fragments thereof that specifically bind to CoV-S comprising heavy and/or light chain amino acid sequences set forth herein (e.g., CDR-H1, CDR- H2, CDR-H3, CDR-L1, CDR-L2 and/or CDR-L3) as well as antibodies and fragments wherein one or more amino acid residues is glycosylated, one or more Asn residues is deamidated, one or more residues (e.g., Met, Trp and/or His) is oxidized, the N-terminal Gln is pyroglutamate (pyroE) and/or the C-terminal Lysine is missing.
  • CDR-H1, CDR- H2, CDR-H3, CDR-L1, CDR-L2 and/or CDR-L3 as well as antibodies and fragments wherein one or more amino acid residues is glycosylated, one or more Asn residues is deamidated, one or more residues (e.g., Met
  • the present invention provides methods for administering an anti-CoV-S antigen- binding protein of the present invention, e.g., those of Table 1, comprising introducing the antigen-binding protein into the body of a subject (e.g., a human).
  • the method comprises piercing the body of the subject with a needle of a syringe and injecting the antigen- binding protein into the body of the subject, e.g., into the vein, artery, tumor, muscular tissue or subcutis of the subject.
  • the present invention provides a vessel (e.g., a plastic or glass vial, e.g., with a cap or a chromatography column, hollow bore needle or a syringe cylinder) comprising an anti-CoV- S antigen-binding protein of the present invention, e.g., those of Table 1.
  • a vessel e.g., a plastic or glass vial, e.g., with a cap or a chromatography column, hollow bore needle or a syringe cylinder
  • an anti-CoV- S antigen-binding protein of the present invention e.g., those of Table 1.
  • the present invention also provides an injection device comprising one or more antigen-binding proteins (e.g., antibody or antigen-binding fragment) that bind specifically to CoV-S, e.g., those of Table 1, or a pharmaceutical composition thereof.
  • the injection device may be packaged into a kit.
  • An injection device is a device that introduces a substance into the body of a subject via a parenteral route, e.g., intramuscular, subcutaneous or intravenous.
  • an injection device may be a syringe (e.g., pre-filled with the pharmaceutical composition, such as an auto-injector) which, for example, includes a cylinder or barrel for holding fluid to be injected (e.g., comprising the antibody or fragment or a pharmaceutical composition thereof), a needle for piecing skin and/or blood vessels for injection of the fluid; and a plunger for pushing the fluid out of the cylinder and through the needle bore.
  • an injection device that comprises an antigen-binding protein, e.g., an antibody or antigen-binding fragment thereof, from a combination of the present invention, or
  • a pharmaceutical composition thereof is an intravenous (IV) injection device.
  • IV intravenous
  • a device can include the antigen-binding protein or a pharmaceutical composition thereof in a cannula or trocar/needle which may be attached to a tube which may be attached to a bag or reservoir for holding fluid (e.g., saline) introduced into the body of the subject through the cannula or trocar/needle.
  • the antibody or fragment or a pharmaceutical composition thereof may, in an embodiment of the invention, be introduced into the device once the trocar and cannula are inserted into the vein of a subject and the trocar is removed from the inserted cannula.
  • the IV device may, for example, be inserted into a peripheral vein (e.g., in the hand or arm); the superior vena cava or inferior vena cava, or within the right atrium of the heart (e.g., a central IV); or into a subclavian, internal jugular, or a femoral vein and, for example, advanced toward the heart until it reaches the superior vena cava or right atrium (e.g., a central venous line).
  • an injection device is an autoinjector; a jet injector or an external infusion pump.
  • a jet injector uses a high-pressure narrow jet of liquid which penetrate the epidermis to introduce the antibody or fragment or a pharmaceutical composition thereof to a subject’s body.
  • External infusion pumps are medical devices that deliver the antibody or fragment or a pharmaceutical composition thereof into a subject’s body in controlled amounts. External infusion pumps may be powered electrically or mechanically.
  • Different pumps operate in different ways, for example, a syringe pump holds fluid in the reservoir of a syringe, and a moveable piston controls fluid delivery, an elastomeric pump holds fluid in a stretchable balloon reservoir, and pressure from the elastic walls of the balloon drives fluid delivery.
  • a set of rollers pinches down on a length of flexible tubing, pushing fluid forward.
  • a multi-channel pump fluids can be delivered from multiple reservoirs at multiple rates.
  • Preparation of Human Antibodies Methods for generating human antibodies in transgenic mice are known in the art. Any such known methods can be used in the context of the present invention to make human antibodies that specifically bind to CoV-S.
  • An immunogen comprising any one of the following can be used to generate antibodies to CoV-S.
  • the antibodies of the invention are obtained from mice immunized with a full length, native CoV-S, or with a live attenuated or inactivated virus, or with DNA encoding the protein or fragment thereof.
  • the CoV-S protein or a fragment thereof may be produced using standard
  • the immunogen is a recombinantly produced CoV-S protein or fragment thereof.
  • the immunogen may be a CoV-S polypeptide vaccine.
  • one or more booster injections may be administered.
  • the immunogen may be a recombinant CoV-S polypeptide expressed in E. coli or in any other eukaryotic or mammalian cells such as Chinese hamster ovary (CHO) cells.
  • VELOCIMMUNE® technology see, for example, US 6,596,541, Regeneron Pharmaceuticals, VELOCIMMUNE®
  • any other known method for generating monoclonal antibodies high affinity chimeric antibodies to CoV-S can be initially isolated having a human variable region and a mouse constant region.
  • the VELOCIMMUNE® technology involves generation of a transgenic mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antibody comprising a human variable region and a mouse constant region in response to antigenic stimulation.
  • the DNA encoding the variable regions of the heavy and light chains of the antibody are isolated and operably linked to DNA encoding the human heavy and light chain constant regions.
  • the DNA is then expressed in a cell capable of expressing the fully human antibody.
  • lymphatic cells such as B-cells
  • the lymphatic cells may be fused with a myeloma cell line to prepare immortal hybridoma cell lines, and such hybridoma cell lines are screened and selected to identify hybridoma cell lines that produce antibodies specific to the antigen of interest.
  • DNA encoding the variable regions of the heavy chain and light chain may be isolated and linked to desirable isotypic constant regions of the heavy chain and light chain.
  • Such an antibody protein may be produced in a cell, such as a CHO cell.
  • DNA encoding the antigen-specific chimeric antibodies or the variable domains of the light and heavy chains may be isolated directly from antigen-specific lymphocytes.
  • high affinity chimeric antibodies are isolated having a human variable region and a mouse constant region. As in the experimental section below, the antibodies are characterized and selected for desirable characteristics, including affinity, selectivity, epitope, etc. The mouse constant regions are replaced with a desired human constant region to generate
  • anti-CoV-S antigen- binding proteins e.g., antibodies or antigen-binding fragments
  • an Fc domain comprising one or more mutations, which, for example, enhance or diminish antibody binding to the FcRn receptor, e.g., at acidic pH as compared to neutral pH.
  • the present invention includes anti-CoV-S antibodies comprising a mutation in the C H 2 or a C H 3 region of the Fc domain, wherein the mutation(s) increases the affinity of the Fc domain to FcRn in an acidic environment (e.g., in an endosome where pH ranges from about 5.5 to about 6.0).
  • mutations may result in an increase in serum half-life of the antibody when administered to an animal.
  • Non-limiting examples of such Fc modifications include, e.g., a modification at position 250 (e.g., E or Q); 250 and 428 (e.g., L or F); 252 (e.g., L/Y/F/W or T), 254 (e.g., S or T), and 256 (e.g., S/R/Q/E/D or T); or a modification at position 428 and/or 433 (e.g., H/L/R/S/P/Q or K) and/or 434 (e.g., A, W, H, F or Y [N434A, N434W, N434H, N434F or N434Y]); or a modification at position 250 and/or 428; or a modification at position 307 or 308 (e.g., 308F, V308F), and 434.
  • a modification at position 250 e.g., E or Q
  • 250 and 428 e.g., L or F
  • the modification comprises a 428L (e.g., M428L) and 434S (e.g., N434S) modification; a 428L, 259I (e.g., V259I), and 308F (e.g., V308F) modification; a 433K (e.g., H433K) and a 434 (e.g., 434Y) modification; a 252, 254, and 256 (e.g., 252Y, 254T, and 256E) modification; a 250Q and 428L modification (e.g., T250Q and M428L); and a 307 and/or 308 modification (e.g., 308F or 308P).
  • a 428L e.g., M428L
  • 434S e.g., N434S
  • 428L, 259I e.g., V259I
  • 308F e.g., V308F
  • the modification comprises a 265A (e.g., D265A) and/or a 297A (e.g., N297A) modification.
  • the present invention includes anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments, comprising an Fc domain comprising one or more pairs or groups of mutations selected from the group consisting of: 250Q and 248L (e.g., T250Q and M248L); 252Y, 254T and 256E (e.g., M252Y, S254T and T256E); 428L and 434S (e.g., M428L and N434S); 257I and 311I (e.g., P257I and Q311I); 257I and 434H (e.g., P257I and N434H); 376V and 434H (e.g., D376V and N434H); 30
  • Anti-CoV-S antigen-binding proteins e.g., antibodies and antigen-binding fragments thereof, that comprise a V H and/or V L as set forth herein comprising any possible combinations of the foregoing Fc domain mutations, are contemplated within the scope of the present invention.
  • the present invention also includes anti-CoV-S antigen-binding proteins, antibodies or antigen-binding fragments, comprising a V H set forth herein and a chimeric heavy chain constant (CH) region, wherein the chimeric CH region comprises segments derived from the CH regions of more than one immunoglobulin isotype.
  • CH heavy chain constant
  • the antibodies of the invention may comprise a chimeric C H region comprising part or all of a C H 2 domain derived from a human IgG1, human IgG2 or human IgG4 molecule, combined with part or all of a C H 3 domain derived from a human IgG1, human IgG2 or human IgG4 molecule.
  • the antibodies of the invention comprise a chimeric C H region having a chimeric hinge region.
  • a chimeric hinge may comprise an “upper hinge” amino acid sequence (amino acid residues from positions 216 to 227 according to EU numbering) derived from a human IgG1, a human IgG2 or a human IgG4 hinge region, combined with a “lower hinge” sequence (amino acid residues from positions 228 to 236 according to EU numbering) derived from a human IgG1, a human IgG2 or a human IgG4 hinge region.
  • the chimeric hinge region comprises amino acid residues derived from a human IgG1 or a human IgG4 upper hinge and amino acid residues derived from a human IgG2 lower hinge.
  • an antibody comprising a chimeric CH region as described herein may, in certain embodiments, exhibit modified Fc effector functions without adversely affecting the therapeutic or pharmacokinetic properties of the antibody.
  • Immunoconjugates [000119]
  • the invention encompasses an anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments, conjugated to another moiety, e.g., a therapeutic moiety (an “immunoconjugate”), such as a toxoid or an anti-viral drug to treat influenza virus infection.
  • an anti-CoV-S antibody or fragment is conjugated to any of the further therapeutic agents set forth herein.
  • immunoconjugate refers to an antigen-binding protein, e.g., an antibody or antigen-binding fragment, which is chemically or biologically linked to a radioactive agent, a cytokine, an interferon, a target or reporter moiety,
  • the antigen-binding protein may be linked to the radioactive agent, cytokine, interferon, target or reporter moiety, enzyme, peptide or therapeutic agent at any location along the molecule so long as it is able to bind its target (CoV- S).
  • immunoconjugates include antibody-drug conjugates and antibody-toxin fusion proteins.
  • the agent may be a second, different antibody that binds specifically to CoV-S.
  • the type of therapeutic moiety that may be conjugated to the anti- CoV-S antigen-binding protein e.g., antibody or fragment
  • the present invention includes anti-CoV-S antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, as well as methods of use thereof and methods of making such antigen-binding proteins.
  • anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments, includes multispecific (e.g., bispecific or biparatopic) molecules that include at least one first antigen-binding domain that specifically binds to CoV-S (e.g., an antigen-binding domain from an antibody of Table 1) and at least one second antigen-binding domain that binds to a different antigen or to an epitope in CoV-S which is different from that of the first antigen-binding domain.
  • the first antigen-binding domain and the second antigen-binding domain are both selected from the antigen-binding domains of Table 1.
  • the first and second epitopes overlap. In another embodiment of the invention, the first and second epitopes do not overlap. For example, in an
  • a multispecific antibody is a bispecific IgG antibody (e.g., IgG1 or IgG4) that includes a first antigen-binding domain that binds specifically to CoV-S including the heavy and light immunoglobulin chain of an antibody of Table 1, and a second antigen-binding domain that binds specifically to a different epitope of CoV-S.
  • a bispecific IgG antibody e.g., IgG1 or IgG4 includes a first antigen-binding domain that binds specifically to CoV-S and a second binding domain that binds to a host cell protein, e.g., ACE2 or TMPRSS2.
  • the antibodies of Table 1 include multispecific molecules, e.g., antibodies or antigen-binding fragments, that include the CDR-Hs and CDR-Ls, V H and V L , or HC and LC of those antibodies, respectively (including variants thereof as set forth herein).
  • multispecific molecules e.g., antibodies or antigen-binding fragments, that include the CDR-Hs and CDR-Ls, V H and V L , or HC and LC of those antibodies, respectively (including variants thereof as set forth herein).
  • an antigen-binding domain that binds specifically to CoV-S which may be included in a multispecific molecule, comprises: (1) (i) a heavy chain variable domain sequence that comprises CDR-H1, CDR-H2, and CDR-H3 amino acid sequences set forth in Table 1, and (ii) a light chain variable domain sequence that comprises CDR-L1, CDR-L2, and CDR- L3 amino acid sequences set forth in Table 1; or, (2) (i) a heavy chain variable domain sequence comprising an amino acid sequence set forth in Table 1, and (ii) a light chain variable domain sequence comprising an amino acid sequence set forth in Table 1; or, (3) (i) a heavy chain immunoglobulin sequence comprising an amino acid sequence set forth in Table 1, and (ii) a light chain immunoglobulin sequence comprising an amino acid sequence set forth in Table 1.
  • the multispecific antibody or fragment includes more than two different binding specificities (e.g., a trispecific
  • a bispecific antigen-binding fragment comprises a first scFv (e.g., comprising VH and VL sequences of Table 1) having binding specificity for a first epitope (e.g., CoV-S) and a second scFv having binding specificity for a second, different epitope.
  • a first scFv e.g., comprising VH and VL sequences of Table 1 having binding specificity for a first epitope (e.g., CoV-S) and a second scFv having binding specificity for a second, different epitope.
  • the first and second scFv are tethered with a linker, e.g., a peptide linker (e.g., a GS linker such as (GGGGS)n (SEQ ID NO: 834) wherein n is, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10).
  • a linker e.g., a peptide linker (e.g., a GS linker such as (GGGGS)n (SEQ ID NO: 834) wherein n is, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10).
  • a linker e.g., a peptide linker (e.g., a GS linker such as (GGGGS)n (SEQ ID NO: 834) wherein n is, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10).
  • Other bispecific antigen-binding fragments include an F(ab) 2 of a bispecific IgG antibody which comprises the
  • the present invention provides methods for treating or preventing viral infection (e.g., coronavirus infection) by administering a therapeutically effective amount of anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment, (e.g., of Table 1) to a subject (e.g., a human) in need of such treatment or prevention.
  • a therapeutically effective amount of anti-CoV-S antigen-binding protein e.g., antibody or antigen-binding fragment, (e.g., of Table 1)
  • Coronavirus infection may be treated or prevented, in a subject, by administering an anti-CoV-S antigen-binding protein of the present invention to a subject.
  • An effective or therapeutically effective dose of anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment (e.g., of Table 1), for treating or preventing a viral infection refers to the amount of the antibody or fragment sufficient to alleviate one or more signs and/or symptoms of the infection in the treated subject, whether by inducing the regression or elimination of such signs and/or symptoms or by inhibiting the progression of such signs and/or symptoms.
  • the dose amount may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like.
  • an effective or therapeutically effective dose of antibody or antigen-binding fragment thereof of the present invention, for treating or preventing viral infection, e.g., in an adult human subject is about 0.01 to about 200 mg/kg, e.g., up to about 150 mg/kg.
  • the dosage is up to about 10.8 or 11 grams (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11 grams).
  • the frequency and the duration of the treatment can be adjusted.
  • antigen-binding protein of the present invention can be administered at an initial dose, followed by one or more secondary doses.
  • the initial dose may be followed by administration of a second or a plurality of subsequent doses of antibody or antigen-binding fragment thereof in an amount that can be approximately the same or less than that of the initial dose, wherein the subsequent doses are separated by at least 1 day to 3 days; at least one week, at least 2 weeks; at least 3 weeks; at least 4 weeks; at least 5 weeks; at least 6 weeks; at least 7 weeks; at least 8 weeks; at least 9 weeks; at least 10 weeks; at least 12 weeks; or at least 14 weeks.
  • the term “subject” refers to a mammal (e.g., rat, mouse, cat, dog, cow, pig, sheep, horse, goat, rabbit), preferably a human, for example, in need of prevention and/or treatment of a disease or disorder such as viral infection or cancer.
  • the subject may have a viral infection, e.g., an influenza infection, or be predisposed to developing an infection.
  • Subjects predisposed to developing an infection, or subjects who may be at elevated risk for contracting an infection include subjects with compromised immune systems because of autoimmune disease, subjects receiving immunosuppressive therapy (for example, following organ transplant), subjects afflicted with human immunodeficiency syndrome (HIV) or acquired immune deficiency syndrome (AIDS), subjects with forms of anemia that deplete or destroy white blood cells, subjects receiving radiation or chemotherapy, or subjects afflicted with an inflammatory disorder. Additionally, subjects of very young (e.g., 5 years of age or younger) or old age (e.g., 65 years of age or older) are at increased risk.
  • immunosuppressive therapy for example, following organ transplant
  • HIV human immunodeficiency syndrome
  • AIDS acquired immune deficiency syndrome
  • subjects with forms of anemia that deplete or destroy white blood cells subjects receiving radiation or chemotherapy, or subjects afflicted with an inflammatory disorder.
  • subjects of very young e.g., 5 years of age or younger
  • old age e.g., 65 years of age or older
  • a subject may be at risk of contracting a viral infection due to proximity to an outbreak of the disease, e.g. subject resides in a densely-populated city or in close proximity to subjects having confirmed or suspected infections of a virus, or choice of employment, e.g. hospital worker, pharmaceutical researcher, traveler to infected area, or frequent flier.
  • “Treat” or “treating” means to administer an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment of the present invention (e.g., of Table 1), to a subject having one or more signs or symptoms of a disease or infection, e.g., viral infection, for which the antigen-binding protein is effective when administered to the subject at an effective or therapeutically effective amount or dose (as discussed herein).
  • an anti-CoV-S antigen-binding protein e.g., antibody or antigen-binding fragment of the present invention (e.g., of Table 1)
  • a subject having one or more signs or symptoms of a disease or infection e.g., viral infection
  • the present invention also encompasses prophylactically administering an anti-CoV- S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof of the present invention (e.g., of Table 1), to a subject who is at risk of viral infection so as to prevent such infection.
  • an anti-CoV- S antigen-binding protein e.g., antibody or antigen-binding fragment thereof of the present invention (e.g., of Table 1)
  • Passive antibody-based immunoprophylaxis has proven an effective strategy for preventing subject from viral infection. See e.g., Berry et al., Passive broad-spectrum influenza immunoprophylaxis. Influenza Res Treat.2014; 2014:267594.
  • Prevent or “preventing” means to administer an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment of the present invention (e.g., of Table 1), to a subject to inhibit the manifestation of a disease or infection (e.g., viral infection) in the body of a subject, for which the antigen-binding protein is effective when administered to the subject at an effective or therapeutically effective amount or dose (as discussed herein).
  • an anti-CoV-S antigen-binding protein e.g., antibody or antigen-binding fragment of the present invention (e.g., of Table 1)
  • a disease or infection e.g., viral infection
  • a sign or symptom of a viral infection in a subject is survival or proliferation of virus in the body of the subject, e.g., as determined by viral titer assay (e.g., coronavirus propagation in embryonated chicken eggs or coronavirus spike protein assay). Other signs and symptoms of viral infection are discussed herein.
  • viral titer assay e.g., coronavirus propagation in embryonated chicken eggs or coronavirus spike protein assay.
  • Other signs and symptoms of viral infection are discussed herein.
  • the subject may be a non-human animal
  • the antigen-binding proteins e.g., antibodies and antigen-binding fragments
  • the non-human animals e.g., cats, dogs, pigs, cows, horses, goats, rabbits, sheep, and the like.
  • the present invention provides a method for treating or preventing viral infection (e.g., coronavirus infection) or for inducing the regression or elimination or inhibiting the progression of at least one sign or symptom of viral infection such as: ⁇ fever or feeling feverish/chills; ⁇ cough; ⁇ sore throat; ⁇ runny or stuffy nose; ⁇ sneezing;
  • ⁇ muscle or body aches ⁇ headaches; ⁇ fatigue (tiredness); ⁇ vomiting; ⁇ diarrhea; ⁇ respiratory tract infection; ⁇ chest discomfort; ⁇ shortness of breath; ⁇ bronchitis; and/or ⁇ pneumonia, which sign or symptom is secondary to viral infection, in a subject in need thereof (e.g., a human), by administering a therapeutically effective amount of anti-CoV-S antigen-binding protein (e.g., of Table 1) to the subject, for example, by injection of the protein into the body of the subject.
  • a therapeutically effective amount of anti-CoV-S antigen-binding protein e.g., of Table 1
  • antigen-binding protein e.g., antibodies and antigen-binding fragments thereof (e.g., of Table 1)
  • antigen-binding protein is admixed with a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutically acceptable carrier or excipient See, e.g., Remington’s Pharmaceutical Sciences and U.S. Pharmacopeia: National Formulary, Mack Publishing Company, Easton, Pa. (1984); Hardman, et al.
  • the pharmaceutical composition is sterile.
  • Such compositions are part of the present invention.
  • the scope of the present invention includes desiccated, e.g., freeze-dried, compositions comprising an anti-CoV-S antigen-binding proteins, e.g., antibody or antigen-
  • a further therapeutic agent that is administered to a subject in association with an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof (e.g., of Table 1), disclosed herein is administered to the subject in accordance with the Physicians’ Desk Reference 2003 (Thomson Healthcare; 57 th edition (Nov.1, 2002)).
  • the mode of administration can vary.
  • Routes of administration include oral, rectal, transmucosal, intestinal, parenteral; intramuscular, subcutaneous, intradermal, intramedullary, intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, intraocular, inhalation, insufflation, topical, cutaneous, transdermal or intra-arterial.
  • the present invention provides methods for administering an anti-CoV-S antigen- binding protein, e.g., antibody or antigen-binding fragment thereof (e.g., of Table 1), comprising introducing the protein into the body of a subject.
  • the method comprises piercing the body of the subject with a needle of a syringe and injecting the antigen-binding protein into the body of the subject, e.g., into the vein, artery, tumor, muscular tissue or subcutis of the subject.
  • the present invention provides a vessel (e.g., a plastic or glass vial, e.g., with a cap or a chromatography column, hollow bore needle or a syringe cylinder) comprising any of the anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments thereof (e.g., of Table 1), polypeptides (e.g., an HC, LC, VH or VL of Table 1) or polynucleotides (e.g., of Table 2) or vectors set forth herein or a pharmaceutical composition thereof comprising a pharmaceutically acceptable carrier.
  • a vessel e.g., a plastic or glass vial, e.g., with a cap or a chromatography column, hollow bore needle or a syringe cylinder
  • any of the anti-CoV-S antigen-binding proteins e.g., antibodies or antigen-binding fragments thereof (e.g., of Table 1),
  • an anti-CoV-S antigen-binding protein e.g., antibody or antigen-binding fragment thereof of the present invention (e.g., of Table 1)
  • a further therapeutic agent includes, but is not limited to: an anti-inflammatory agent, an antimalarial agent, a second antibody or antigen-binding fragment thereof that specifically binds TMPRSS2, and a second antibody or antigen-binding fragment thereof that specifically binds to CoV-S.
  • an antimalarial agent is chloroquine or hydroxychloroquine.
  • an anti-inflammatory agent is an antibody such as sarilumab, tocilizumab, or gimsilumab.
  • the further therapeutic agent is a second antibody or antigen-binding fragment disclosed herein, e.g., of Table 1.
  • one, two, three, four, or more antibodies, or antigen-binding fragments thereof, of Table 1 can be administered in combination (e.g., concurrently or sequentially).
  • a combination of antibodies can be selected from among those binding to different epitope clusters.
  • certain antibodies described herein belong to epitope clusters as follows: Cluster 1, mAb 10987, mAbl 0922, mAbl0936, and mAbl0934; Cluster 2, mAbl0989, mAb 10977, and mAb 10933; Cluster 3, mAbl0920; Cluster 4, mAbl0954, mAbl0986, and mAbl0964; and Cluster 5, mAbl0984.
  • a combination of two antibodies can be selected from, for example, Cluster I and Cluster 2, Cluster 1 and Cluster 3, Cluster 1 and Cluster 4, Cluster 1 and Cluster 5, Cluster 2 and Cluster 3, Cluster 2 and Cluster 4. Cluster 2 and Cluster 5, Cluster 3 and Cluster 4, Cluster 3 and Cluster 5, and Cluster 4 and Cluster 5.
  • an antibody that specifically binds TMPRSS2 is H1H7017N, as described in International Patent Pub. No. WO/2019/147831.
  • anti-CoV-S antigen-binding proteins e.g., anti-SARS-CoV- 2-S antibodies or antigen-binding fragments thereof
  • the present invention includes a composition comprising two (or more) anti-SARS- CoV-2-S antibodies or antigen-binding fragments comprising variable domains from human subjects, wherein the two (or more) antibodies or antigen-binding fragments are derived from different subjects (e.g., two different human subjects).
  • Antibody variable regions derived from human B cells are discussed, e.g., in Examples 1 and 2 (Table 3), which describes that variable domains cloned from such B cells are combined with a constant region not from those B cells to produce hybrid antibodies.
  • the source (Donor) of such antibody variable regions is shown in the Table of Exemplary Human-Derived Antibody Variable Regions, below.
  • a composition may comprise a combination of an antibody or antigen-binding fragment thereof with variable domains derived from donor 1 and an antibody or antigen-binding fragment thereof with variable domains derived from donor 2.
  • a composition may comprise a combination of an antibody or antigen-binding fragment thereof with variable domains derived from donor 1 and an antibody or antigen-binding fragment thereof with variable domains derived from donor 3. In some embodiments, a composition may comprise a combination of an antibody or antigen-binding fragment thereof with variable domains derived from donor 2 and an antibody or antigen-binding fragment thereof with variable domains derived from donor 3.
  • a composition may comprise a combination of mAb10987 (e.g., an antibody comprising the CDRs, the variable regions, or the heavy and light chain sequences shown in Table 1) from Donor 1, and mAb10989 (e.g., an antibody comprising the CDRs, the variable regions, or the heavy and light chain sequences shown in Table 1) from Donor 3.
  • Table of Exemplary Human-Derived Antibody Variable Regions [000142]
  • the further therapeutic agent is an anti-viral drug and/or a vaccine.
  • anti-viral drug refers to any anti-infective drug or therapy used to treat, prevent, or ameliorate a viral infection in a subject.
  • anti-viral drug includes, but is not limited to a cationic steroid antimicrobial, leupeptin, aprotinin, ribavirin, or interferon-alpha2b.
  • Methods for treating or preventing virus (e.g., coronavirus) infection in a subject in need of said treatment or prevention by administering an antibody or antigen-binding fragment of Table 1 in association with a further therapeutic agent are part of the present invention.
  • the further therapeutic agent is a vaccine, e.g., a coronavirus vaccine.
  • a vaccine is an inactivated/killed virus vaccine, a live attenuated virus vaccine or a virus subunit vaccine.
  • the further therapeutic agent is: (camostat mesylate);
  • the anti-viral drug is an antibody or antigen- binding fragment that binds specifically to coronavirus, e.g., SARS-CoV-2, SARS-CoV, or MERS-CoV.
  • Exemplary anti-CoV-S antibodies include, but are not limited to: H4sH15188P; H1H15188P; H1H15211P; H1H15177P; H4sH15211P; H1H15260P2; H1H15259P2; H1H15203P; H4sH15260P2; H4sH15231P2; H1H15237P2; H1H15208P; H1H15228P2; H1H15233P2; H1H15264P2; H1H15231P2; H1H15253P2; H1H15215P; and H1H15249P2, as set forth in International patent application publication no.
  • the antibody or fragment comprises a light chain immunoglobulin that includes CDR-L1, CDR-L2 and CDR-L3 (e.g., the VL or light chain thereof); and a heavy chain that includes CDR-H1, CDR-H2 and CDR-H3 (e.g., the V H or heavy chain thereof) of any of the foregoing anti-CoV-S antibodies.
  • the further therapeutic agent is not aprotinin, leupeptin, a cationic steroid antimicrobial, an influenza vaccine (e.g., killed, live,
  • an anti-CoV-S antigen- binding protein e.g., antibody or antigen-binding fragment thereof of the present invention
  • an anti-CoV-S antigen- binding protein e.g., antibody or antigen-binding fragment thereof of the present invention
  • another agent e.g., a single composition, e.g., for simultaneous delivery, or formulated separately into two or more compositions (e.g., a kit).
  • Each component can be administered to a subject at a different time than when the other component is administered; for example, each administration may be given non-simultaneously (e.g., separately or sequentially) at intervals over a given period of time.
  • kits comprising one or more components that include, but are not limited to, an anti-CoV-S antigen-binding protein, e.g., an antibody or antigen-binding fragment as discussed herein (e.g., of Table 1), in association with one or more additional components including, but not limited to, a further therapeutic agent, as discussed herein.
  • an anti-CoV-S antigen-binding protein e.g., an antibody or antigen-binding fragment as discussed herein (e.g., of Table 1)
  • additional components including, but not limited to, a further therapeutic agent, as discussed herein.
  • the antigen-binding protein and/or the further therapeutic agent can be formulated as a single composition or separately in two or more compositions, e.g., with a pharmaceutically acceptable carrier, in a pharmaceutical composition.
  • the kit includes an anti-CoV-S antigen-binding protein, e.g., an antibody or antigen-binding fragment thereof of the invention (e.g., of Table 1), or a pharmaceutical composition thereof in one container (e.g., in a sterile glass or plastic vial) and a further therapeutic agent in another container (e.g., in a sterile glass or plastic vial).
  • the kit comprises a combination of the invention, including an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof of the invention (e.g., of Table 1), or pharmaceutical composition thereof in combination with one or more further therapeutic agents formulated together, optionally, in a pharmaceutical composition, in a single, common container.
  • an anti-CoV-S antigen-binding protein e.g., antibody or antigen-binding fragment thereof of the invention (e.g., of Table 1)
  • pharmaceutical composition thereof in combination with one or more further therapeutic agents formulated together, optionally, in a pharmaceutical composition, in a single, common container.
  • the kit can include a device (e.g., an injection device) for performing such
  • the kit can include one or more hypodermic needles or other injection devices as discussed above containing the anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof of the present invention (e.g., of Table 1).
  • the kit can include a package insert including information concerning the pharmaceutical compositions and dosage forms in the kit. Generally, such information aids patients and physicians in using the enclosed pharmaceutical compositions and dosage forms effectively and safely.
  • the following information regarding a combination of the invention may be supplied in the insert: pharmacokinetics, pharmacodynamics, clinical studies, efficacy parameters, indications and usage, contraindications, warnings, precautions, adverse reactions, overdosage, proper dosage and administration, how supplied, proper storage conditions, references, manufacturer/distributor information and patent information. Diagnostic Uses of the Antibodies [000153]
  • the anti-CoV-S antigen-binding proteins e.g., antibodies or antigen-binding fragments thereof of the present invention (e.g., of Table 1), may be used to detect and/or measure CoV-S in a sample.
  • Exemplary assays for CoV-S may include, e.g., contacting a sample with an anti-CoV-S antigen-binding protein of the invention, wherein the anti-CoV-S antigen-binding protein is labeled with a detectable label or reporter molecule or used as a capture ligand to selectively isolate CoV-S from samples.
  • the presence of an anti-CoV-S antigen-binding protein complexed with CoV-S indicates the presence of CoV-S in the sample.
  • an unlabeled anti-CoV-S antibody can be used in combination with a secondary antibody which is itself detectably labeled.
  • the detectable label or reporter molecule can be a radioisotope, such as 3 H, 14 C, 32 P, 35 S, or 125 I; a fluorescent or chemiluminescent moiety such as fluorescein isothiocyanate, or rhodamine; or an enzyme such as alkaline phosphatase, b- galactosidase, horseradish peroxidase, or luciferase.
  • Specific exemplary assays that can be used to detect or measure CoV-S in a sample include neutralization assays, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence-activated cell sorting (FACS).
  • the present invention includes a method for detecting the presence of spike protein polypeptide in a sample comprising contacting the sample with an anti-CoV-S antigen-binding protein and detecting the presence of a CoV-S/anti-CoV-S antigen-binding protein wherein the presence of the complex indicates the presence of CoV-S.
  • An anti-CoV-S antigen-binding protein of the invention (e.g., of Table 1) may be used in a Western blot or immune-protein blot procedure for detecting the presence of CoV-S or a fragment thereof in a sample.
  • Such a procedure forms part of the present invention and includes the steps of e.g.: (1) providing a membrane or other solid substrate comprising a sample to be tested for the presence of CoV-S, e.g., optionally including the step of transferring proteins from a sample to be tested for the presence of CoV-S (e.g., from a PAGE or SDS-PAGE electrophoretic separation of the proteins in the sample) onto a membrane or other solid substrate using a method known in the art (e.g., semi-dry blotting or tank blotting); and contacting the membrane or other solid substrate to be tested for the presence of CoV-S or a fragment thereof with an anti-CoV-S antigen-binding protein of the invention.
  • a method known in the art e.g., semi-dry blotting or tank blotting
  • Such a membrane may take the form, for example, of a nitrocellulose or vinyl-based (e.g., polyvinylidene fluoride (PVDF)) membrane to which the proteins to be tested for the presence of CoV-S in a non-denaturing PAGE (polyacrylamide gel electrophoresis) gel or SDS- PAGE (sodium dodecyl sulfate polyacrylamide gel electrophoresis) gel have been transferred (e.g., following electrophoretic separation in the gel).
  • PAGE polyacrylamide gel electrophoresis
  • SDS- PAGE sodium dodecyl sulfate polyacrylamide gel electrophoresis
  • Detection of the bound antigen-binding protein indicates that the CoV-S protein is present on the membrane or substrate and in the sample. Detection of the bound antigen-binding protein may be by binding the antigen-binding protein with a secondary antibody (an anti- immunoglobulin antibody) which is detectably labeled and, then, detecting the presence of the secondary antibody label.
  • a secondary antibody an anti- immunoglobulin antibody
  • the anti-CoV-S antigen-binding proteins e.g., antibodies and antigen-binding fragments (e.g., of Table 1)
  • Such a method forms part of the present invention and comprises, e.g., (1) contacting tissue to be tested for the presence of CoV-S protein with an anti-CoV-S antigen-binding protein of the invention; and
  • the antigen-binding protein itself is detectably labeled, it can be detected directly. Alternatively, the antigen-binding protein may be bound by a detectably labeled secondary antibody wherein the label is then detected.
  • EXAMPLES The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the methods and compositions of the invention and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for.
  • SARS-CoV-2-S Human antibodies to SARS-CoV-2 spike protein (SARS-CoV-2-S) were generated in a VELOCIMMUNE ® mouse comprising DNA encoding human immunoglobulin heavy and kappa light chain variable regions or human immunoglobulin heavy and lambda light chain variable regions.
  • Each mouse was immunized with a vector expressing the SARS-CoV-2-S receptor binding domain (RBD) (amino acids 1-1273 of NCBI accession number (MN908947.3), SEQ ID NO: 832), followed by a booster with a SARS-CoV-2-S vector or a SARS-CoV-2-S protein.
  • the antibody immune response was monitored by a SARS-CoV-2-S-specific immunoassay.
  • lymphocytes were harvested and fused with mouse myeloma cells to preserve their viability and form hybridoma cell lines.
  • the hybridoma cell lines were screened and selected to identify cell lines that produce SARS-CoV-2- S-specific antibodies.
  • Anti-SARS-CoV-2-S antibodies were also isolated directly from antigen- positive mouse B cells without fusion to myeloma cells, as described in U.S. Patent 7582298, herein specifically incorporated by reference in its entirety. Using this method, fully human anti- SARS-CoV-2-S antibodies (i.e., antibodies possessing human variable domains and human constant domains) were obtained. [000161] Antibody variable regions were also isolated from human blood samples. Whole
  • One set of the resulting RT products was first amplified by PCR using a 5’ degenerate primer specific for antibody heavy variable region leader sequence or a 5’ degenerate primer specific for antibody light chain variable region leader sequence and a 3’ primer specific for antibody constant region, to form an amplicon.
  • the amplicons were then amplified again by PCR using a 5’ degenerate primer specific for antibody heavy variable region framework 1 or a 5’ degenerate primer specific for antibody light chain variable region framework 1 and a 3’ primer specific for antibody constant region, to generate amplicons for cloning.
  • the antibody heavy chain and light chain derived PCR products were cloned into expression vectors containing heavy constant region and light constant region, respectively, thereby producing expression vectors for hybrid antibodies.
  • Example 2 Heavy and light chain variable region amino acid and nucleotide sequences
  • Table 1 sets forth the amino acid sequence identifiers of the heavy and light chain variable regions and CDRs, as well as the heavy chain and light chain sequences, of exemplary anti-SARS-CoV-2-S antibodies. The corresponding nucleic acid sequence identifiers are set forth in Table 2.
  • Table 1 Amino Acid Sequence Identifiers
  • Antibodies disclosed herein have fully human variable regions but can have mouse constant regions (e.g., a mouse IgG1 Fc or a mouse IgG2 Fc (a or b isotype)) or human constant regions (e.g., a human IgG1 Fc or a human IgG4 Fc).
  • mouse constant regions e.g., a mouse IgG1 Fc or a mouse IgG2 Fc (a or b isotype)
  • human constant regions e.g., a human IgG1 Fc or a human IgG4 Fc
  • variable domains including the CDRs
  • Tables 1 and 2 the variable domains (including the CDRs) – which are indicated by the numerical identifiers shown in Tables 1 and 2 will remain the same, and the binding properties to antigen are expected to be identical or substantially similar regardless of the nature of the constant domain.
  • VI antibodies utilized VH3-53 paired with VK1-9, VK1-33, or VK1-39 while human-derived antibodies utilized VH3-66 paired with VK1-33 or VH2-70 paired with VK1-39.
  • Further analysis of overlaid sequences showed strong overlap in the repertoire of isolated kappa chains between VI and human-derived antibodies.
  • the repertoire of Lambda chains did not overlap well, that may be due to only two lambda mice being included in this trial.
  • the average CDR length for heavy chain was similar between VI and human derived antibodies with an average length of 13 and 14.5 amino acids, respectively.
  • Example 3 Characterization of hybridoma supernatants by binding ELISA [000167] An ELISA binding assay was performed to identify antibody supernatants that bound to the SARS-CoV-2-Spike protein receptor binding domain (RBD). A protein composed of the RBD of SARS-CoV-2 (amino acids 319-541) expressed with a 6X histidine tag and two myc epitope tags at the C-terminus (SARS-CoV-2-S-RBD-mmH; see also NCBI Accession Number MN908947.3) was coated at 1 mg/ml on a 96-well plate in PBS buffer overnight at 4°C. Nonspecific binding sites were subsequently blocked using a 0.5% (w/v) solution of BSA in PBS. Antibody supernatants or media alone were diluted 1:40 or 1 :50 in the PSA+0.5% BSA
  • SARS-CoV-2-S-RBD The ability of anti-SARS-CoV-2-S antibodies to bind the receptor binding domain of SARS-CoV-2-S (SARS-CoV-2-S-RBD) was assessed, as described above, using a binding ELISA with the SARS-CoV-2-S-RBD-mmH protein coated on a microplate. Single point antibody supernatant binding to SARS-COV-2-S-RBD-mmH coated on 96-well microtiter plates was detected with an HRP conjugated anti-hFc or anti-mFc antibody. [000169] The binding results of three trials are summarized in Table 4. The SARS-CoV-2 binding signals (absorbance 450nm) are indicated, with the media only background provided as a negative reference per experiment.
  • Example 4 Antibody binding to SARS-CoV-2-S-expressing virus-like particle [000170] To investigate the ability of a panel of anti-SARS-CoV-2-S monoclonal antibodies to bind the SARS-CoV-2 spike glycoprotein, an in vitro binding assay utilizing SARS-CoV-2 spike protein-expressing viral-like particles (VLPs) in an electrochemiluminescence based detection platform (MSD) was developed.
  • VLPs SARS-CoV-2 spike protein-expressing viral-like particles
  • MSD electrochemiluminescence based detection platform
  • VSV-SARS- CoV-2-S Vesicular stomatitis virus lacking glycoprotein G
  • VSV delta G SARS-CoV-2 spike protein
  • VSV-G VSV G protein
  • VLPs described above were diluted in PBS, seeded into 96-well carbon electrode plates (MULTI-ARRAY high bind plate, MSD), and incubated overnight at 4 °C to allow the VLPs to adhere. Nonspecific binding sites were blocked by 2% BSA (w/v) in PBS for 1 hour at room temperature. Supernatants containing antibodies produced from SARS CoV-2-immunized mice or infected human sera, along with media-only controls which were diluted 1:10 or 1:20 in 1x PBS + 0.5% BSA buffer, were added to the plate-bound particles.
  • the plates were then incubated for 1 hour at room temperature with shaking, after which the plates were washed with 1x PBS to remove the unbound antibodies using an AquaMax2000 plate washer (MDS Analytical Technologies).
  • the plate-bound antibodies were detected with a SULFO-TAGTM- conjugated anti-human IgG antibody (Jackson Immunoresearch) or a SULFO-TAGTM- conjugated anti-mouse IgG antibody (Jackson Immunoresearch) for 1 hour at room temperature.
  • the plates were developed with the Read Buffer (MSD) according to manufacturer’s recommended procedure and the luminescent signals were recorded with a SECTOR Imager 600 (Meso Scale Development) instrument.
  • a signal observed from SARS-COV-2-S-expressing VLPs indicates binding, while comparison with negative VLPs provides a relative background.
  • Media alone samples provide baseline signals of secondary antibody binding to samples with no supernatant.
  • the ratios of SARS- CoV-2-S-expressing VSV: VSV-VLPs (negative control) ranged from 1.1- 22.7, with many having high background on VSV-VLPs.
  • the ratio of mAb11002 of 0.9 is likely due to a low concentration of monoclonal antibody in the supernatant sample.
  • Table 5 SARS-CoV-2-S VLP binding
  • Example 5 Antibody neutralization of VSV-SARS-CoV-2-S pseudovirus infectivity
  • VSV pseudotype viruses were generated by transiently transfecting 293T cells with a plasmid encoding for SARS-CoV-2 spike protein.
  • Cells were seeded in 15 cm plates at 1.2x10 7 cells per plate in DMEM complete media one day prior to transfection with 15 ⁇ g/plate spike protein DNA using 125 ⁇ L Lipofectamine LTX, 30 ⁇ L PLUS reagent, and up to 3 mL Opti-Mem. 24 hours post transfection, the cells were washed with 10 mL PBS, then infected with an MOI of 0.1 VSV DG:mNeon virus in 10 mL Opti-Mem. Virus was incubated on cells for 1 hour, with gentle rocking every 10 minutes.
  • VSV-SARS-CoV-2-S Neutralization assay with VSV-SARS-CoV-2-S [000177] On day 1, Vero cells were seeded at 80% confluency in T225 flasks.
  • the neutralization potency of antibody at each dilution is represented as a percentage compared to mock supernatant control. All antibodies demonstrated neutralization capacity, and particularly for the set of antibodies that were evaluated 1:100, those showing higher neutralization may represent more potent neutralization capacity. Table 6: Neutralization of VLPs
  • Target cells were engineered Jurkat T cells expressing human CD20 (used as a positive control with a CD20-targeting human IgG1 antibody) and the full-length SARS-CoV-2 spike protein controlled by a doxycycline-inducible promoter. Reporter cells were incubated with target cells and engagement of FcgR3a via the Fc domain of human IgG1 antibodies bound to target cells led to the activation of the transcription factor NFAT in the reporter cells and drove the expression of luciferase which was then measured via a luminescence readout.
  • Jurkat T cells were engineered to constitutively express full length human CD20 (amino acids M1-P297 of NCBI accession number NP_690605.1), Tet3G transactivator protein (cloned using a Takara pEF1a-Tet3G Vector, Catalog # 631167), as well as a doxycycline- inducible full-length SARS-CoV-2 spike protein (amino acids M1-T1273 of NCBI accession number YP_009724390.1).
  • Jurkat/Tet3G/hCD20/SARS-CoV2 spike protein- expressing cells were sorted for high expression of the spike protein and subsequently maintained in RPMI + 10% Tet-free FBS + P/S/G + 500 ⁇ g/ml G418 + 1 mg/ml puromycin + 250 ⁇ g/ml hygromycin growth medium.
  • NFAT Nuclear Factor of Activated T- cells
  • luciferase reporter construct along with the high affinity human FcgR3a 176 Val allotype receptor (amino acids M1-K254 of NCBI accession number P08637 VAR_003960).
  • Engineered reporter cells were maintained in RPMI1640 + 10% FBS + P/S/G + 0.5 mg/ml puromycin + 500 mg/ml G418 growth media. [000185] 36 hours prior to the start of the surrogate ADCC assay, 5 x 10 5 target cells/ml were induced in RPMI + 10% Tet-free FBS + P/S/G cell culture media containing 1 ⁇ g/ml doxycycline (Sigma). A day before the experiment, reporter cells were split to a density of 7.5 x 10 5 cells/ml in RPMI 1640 + 10% FBS + P/S/G + 0.5 mg/ml puromycin + 500 mg/ml G418 growth media.
  • the target and reporter cells were transferred into assay media (RPMI + 10% Tet-free FBS + P/S/G) and added at a 3:2 ratio (3 x 10 4 /well target cells and 2 x 10 4 /well reporter cells) to 384-well white microtiter plates, followed by the addition of anti-SARS-CoV-2-S antibody supernatant of varying concentrations.
  • assay media RPMI + 10% Tet-free FBS + P/S/G
  • a positive control CD20 antibody with human IgG1
  • a negative control sample containing no antibody was included on each plate to normalize detected ADCC activities of anti-SARS-CoV- 2-S antibody supernatants.
  • Table 7 summarizes the results, showing the raw luciferase activity and the calculated % of positive control are indicated. A range of % ADCC activity was observed indicating FcgR3a activation by the antibody supernatants. All samples demonstrated some measure of surrogate ADCC activity, and 10 of the antibody supernatants demonstrated surrogate ADCC activity better than observed in positive controls.
  • Table 7 ADCC surrogate activity of anti-SARS-CoV-2-S antibody supernatants.
  • Example 7 Anti-SARS-CoV-2-S antibody binding specificity assay [000189]
  • a Luminex binding assay was performed to determine the binding of anti-SARS- COV-2-S antibodies to a panel of antigens.
  • antigens were amine-coupled or captured by streptavidin to Luminex microspheres as follows: approximately 10 million MagPlex microspheres (Luminex Corp., MagPlex Microspheres, Cat. No. MC10000 and MC12000), were resuspended by vortexing in 500 ⁇ L 0.1M NaPO4, pH 6.2 (activation buffer) and then centrifuged to remove the supernatant. Microspheres were protected from light, as they are light sensitive.
  • microspheres were resuspended in 160 ⁇ L of activation buffer and the carboxylate groups (-COOH) were activated by addition of 20 ⁇ L of 50 mg/mL of N- hydroxysuccinimide (NHS, Thermo Scientific, Cat. No.24525) followed by addition of 20 ⁇ L of 50 mg/mL 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide (EDC, ThermoScientific, Cat. No. 22980) at 25 °C.
  • NHS N- hydroxysuccinimide
  • EDC 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide
  • the pH of the reaction was reduced to 5.0 with the addition of 600 ⁇ L 50 mM MES, pH 5 (coupling buffer), and the microspheres were vortexed and centrifuged to remove supernatant.
  • the activated microspheres were immediately mixed with 500 ⁇ L of 25 ⁇ g/mL of the protein antigen or Streptavidin in coupling buffer and incubated for two hours at 25 °C.
  • the coupling reaction was quenched by addition of 50 ⁇ L of 1M Tris-HCl, pH 8.0 and the microspheres were vortexed, centrifuged, and washed three times with 800 ⁇ L of PBS 0.005% (Tween200.05%), to remove uncoupled proteins and other reaction components.
  • Microspheres were resuspended in 1 mL of PBS 2% BSA 0.05% Na Azide at 10 million microspheres/mL.
  • 500 ⁇ L of 12.5 ⁇ g/mL of biotinylated protein in PBS was added to Streptavidin-coupled microspheres and incubated for one hour at 25 °C.
  • Microspheres were vortexed, centrifuged, and washed three times with 800 ⁇ L of PBS, and then blocked using 500 ⁇ L 30mM Biotin (Millipore-Sigma, Cat. No. B4501) in 0.15M Tris pH 8.0.
  • Microspheres were incubated for 30 minutes then vortexed, centrifuged, and washed three times with 800 ⁇ L of PBS. Microspheres were resuspended in 1 mL of PBS 2% BSA 0.05% Na Azide at 10 million microspheres/mL. [000190] Microspheres for the different proteins and biotinylated proteins were mixed at 2700
  • microspheres were plated per well on a 96 well ProcartaPlex flat bottom plate (ThermoFisher, Cat. No: EPX-44444-000) and mixed with 25 ⁇ L of individual anti-SARS- CoV-2 supernatant containing antibody. Samples and microspheres were incubated for two hours at 25 o C and then washed twice with 200 ⁇ L of DPBS with 0.05% Tween 20.
  • the SARS-CoV-2 proteins used in the assay are as follows: RBD_ (R319-F541).mmh: SEQ ID NO: 829 RBD_ (R319-F541).mFc: SEQ ID NO: 830 RBD_ (R319-F541).hFc): SEQ ID NO: 831 [000191]
  • the results of the Luminex binding are shown in Table 8 and Table 9 as median fluorescence intensity (MFI) signal intensities.
  • MFI median fluorescence intensity
  • Example 8 Anti-SARS-CoV-2-S antibody diversity assay [000192] A binding assay was performed to determine the binding profile of anti-SARS-COV- 2-S antibodies. For this assay, antigens were amine coupled as described for the Luminex binding assay above. Briefly, approximately 9 million MagPlex microspheres for 16 different bead regions (Luminex Corp., MagPLex Microspheres, Cat. No. MagPLex MC10000 and MC12000), were resuspended by vortexing in 500 ⁇ L 0.1M NaPO 4 , pH 6.2 and then centrifuged to remove the supernatant.
  • MagPlex microspheres for 16 different bead regions Luminex Corp., MagPLex Microspheres, Cat. No. MagPLex MC10000 and MC12000
  • microspheres were resuspended in 160 ⁇ L of activation buffer and the carboxylate groups (-COOH) were activated by addition of 20 ⁇ L of 50 mg/mL of N- hydroxysuccinimide (NHS, Thermo Scientific, Cat# 24525) followed by addition of 20 ⁇ L of 50 mg/mL of 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide (EDC, ThermoScientific, Cat# 22980) at 25°C.
  • NHS N- hydroxysuccinimide
  • EDC 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide
  • the pH of the reaction was reduced to 5.0 with the addition of 600 ⁇ L of 50 mM MES, pH 5 (coupling buffer), and the microspheres were vortexed and centrifuged to remove supernatant.
  • the activated microspheres were immediately mixed with 500 ⁇ L of 20 ⁇ g/mL of SARS-CoV-2 Spike Protein (RBD)(R319-F541)-mmH in coupling buffer and incubated for two hours at 25 o C.
  • the coupling reaction was quenched by addition of 50 ⁇ L of 1M Tris-HCl, pH 8.0 and the microspheres were vortexed, centrifuged, and washed three times with 1000 ⁇ L of PBS.
  • Microspheres were resuspended in 250 ⁇ L of PBS at 9 million microspheres/mL. [000193] 15 out of the 16 microsphere regions with amine-coupled protein were modified for the binning assay as follows: microspheres were washed twice with PBS 5% DMSO, and 500 ⁇ l of a chemical or enzyme were dissolved per manufacturing recommendations and added at 10 nM to the amine-coupled microspheres described above. This was subsequently vortexed and incubated for 2 hours at room temperature with rotation. Wash microspheres 3 times with PBS 2% BSA. Microspheres were resuspended in 1 mL of PBS at 9 million microspheres/mL.
  • Protein-modified and protein-unmodified (intact) microspheres were mixed at 2700 beads/ml, and 75 ⁇ L of microspheres were plated per well on a 96 well ProcartaPlex 96 well flat bottom plate (ThermoFisher, Cat. No: EPX-44444-000) and mixed with 25 ⁇ L of individual anti- SARS-CoV-2-S supernatant-containing antibody. Samples and microspheres were incubated for two hours at 25 °C and then washed twice with 200 ⁇ L of DPBS with 0.05% Tween 20.
  • Biacore CM5 sensor chip surface was first derivatized by amine coupling with either mouse anti-human Fc specific mAb or rabbit anti-mouse Fcg monoclonal antibody (GE, Catalog # BR-1008-38) to capture anti-SARS-CoV-2 antibodies.
  • Binding studies were performed on a human SARS-CoV-2 RBD extracellular domain expressed with a C-terminal myc-myc-hexahistidine tag (SARS-COV-2 RBD-MMH), SARS-CoV-2 RBD extracellular domain expressed with a C-terminal mouse IgG2a (SARS-COV-2 RBD-mFc), or SARS-CoV-2 RBD extracellular domain expressed with a C-terminal human IgG1 (SARS-COV- 2 RBD-hFc).
  • the SARS-CoV-2 RBD antibody capture surface was regenerated using either a 10 sec injection of 20mM phosphoric acid for the mouse anti-human Fc specific monoclonal antibody surface or a 40 sec injection of 10mM Glycine, HCl, pH1.5 for the rabbit anti-mouse Fcg specific polyclonal antibody.
  • the association rate (ka) and dissociation rate (kd) were determined by fitting the real-time binding sensorgrams to a 1:1 binding model with mass transport limitation using BiaEvaluation software v3.1 or Biacore Insight Evaluation software v2.0. or curve-fitting software.
  • Binding dissociation equilibrium constant (K D ) and dissociative half-life (t1 ⁇ 2) were calculated from the kinetic rates as: [000197] Binding kinetics parameters for different SARS-CoV-2 monoclonal antibodies binding to different anti-SARS-COV-2 RBD reagents of the invention at 25°C are shown in Tables 11 and 12.
  • Table 11 Binding kinetics of SARS-COV-2 RBD-MMH binding to anti-SARS-CoV-2 monoclonal antibodies at 25 °C
  • Table 12 Binding kinetics of SARS-COV-2 RBD-mFc or SARS-COV-2 RBD-hFc binding to anti-SARS-CoV-2-S monoclonal antibodies at 25 °C
  • Example 10 Characterization of anti-SARS-CoV-2-S monoclonal antibodies by blocking ELISA [000198] An ELISA-based blocking assay was developed to determine the ability of anti- SARS-CoV2-S antibodies to block the binding of the SARS-CoV-2 spike protein receptor binding domain (RBD) to human angiotensin converting enzyme 2 (hACE2).
  • RBD SARS-CoV-2 spike protein receptor binding domain
  • hACE2 human angiotensin converting enzyme 2
  • the SARS-CoV-2 protein used in the experiments was comprised of the receptor binding domain (RBD) portion of the SARS-CoV-2 spike protein (amino acids Arg319 to Phe541) expressed with the Fc portion of the human IgG1 at the c-terminus (SARS-CoV-2 RBD- hFc; see NCBI accession number MN908947.3)
  • the human ACE2 protein used in the experiments was purchased from R&D systems and is comprised of amino acids glutamine 18 to serine 740 with a c-terminal 10X-Histidine tag (hACE2-His; NCBI accession number Q9BYF1). [000200] Experiments were carried out using the following procedure.
  • a monoclonal anti- Penta-His antibody (Qiagen) was coated at 1 ⁇ g/ml in PBS on a 96-well microtiter plate overnight at 4 °C.
  • the hACE2-His receptor was added at 0.2 ⁇ g/ml in PBS and bound for 2 hours at room temperature. Nonspecific binding sites were subsequently blocked using a 0.5% (w/v) solution of BSA in PBS.
  • a constant amount of 10 pM or 15 pM (as indicated in Table 13) of SARS-CoV-2 RBD-hFc protein was bound with antibodies diluted 1:10 or 1:20 in PBS + 0.5% BSA.
  • binding signal of the sample of the constant SARS-CoV-2-S RBD- hFc without the presence of the antibody for each plate was referenced as 100% binding or 0% blocking; and the baseline signal of the sample of media only without the presence of SARS- CoV-2 RBD-hFc was referenced as 0% binding or 100% blocking.
  • the ability of anti-SARS-CoV-2-S antibodies to block SARS-CoV-2-S RBD from binding to human ACE2 was assessed using a blocking ELISA format.
  • Example 11 Epitope mapping of anti-SARS-CoV-2-S monoclonal antibodies to spike glycoprotein by Hydrogen-Deuterium Exchange Mass Spectrometry
  • Hydrogen-Deuterium Exchange Mass Spectrometry was performed to determine the amino acid residues of the SARS-CoV-2 Spike Protein Receptor Binding Domain (RBD (amino acids R319-F541)) that interact with mAb10989, mAb10987, mAb10934, mAb10933, mAb10920, mAb10922, mAb10936, mAb10954, mAb10964, mAb10977, mAb10984, and mAb10986.
  • RBD amino acids R319-F541
  • HDX-MS A general description of the HDX-MS method is set forth in e.g., Ehring (1999) Analytical Biochemistry 267(2):252-259; and Engen and Smith (2001) Anal. Chem.73:256A-265A.
  • the HDX-MS experiments were performed on an integrated HDX-MS platform, consisting of a Leaptec HDX PAL system for the deuterium labeling and quenching, a Waters Acquity I-Class (Binary Solvent Manager) for the sample digestion and loading, a Waters Acquity I-Class (Binary Solvent Manager) for the analytical gradient, and a Thermo Q Exactive HF mass spectrometer for peptide mass measurement.
  • the labeling solution was prepared as PBS buffer in D2O at pD 7.0 (10 mM phosphate buffer, 140 mM NaCl, and 3 mM KCl, equivalent to pH 7.4 at 25°C).
  • pD 7.0 10 mM phosphate buffer, 140 mM NaCl, and 3 mM KCl, equivalent to pH 7.4 at 25°C.
  • 10 mL of the RBD protein or RBD protein premixed with each one of the 12 antibodies listed above were incubated at 20 °C with 90 mL of D 2 O labeling solution for various timepoints, in duplicate.
  • the time points were 0 min (non-deuterated control), 5 min, and 10 min.
  • mAb10920, mAb10922, mAb10936, mAb10954, mAb10964, mAb10977, mAb10984, and mAb10986 the time points were 0 min (non-deuterated control) and 10 min.
  • the deuteration reaction was quenched by adding 90 mL of pre-chilled quench buffer (0.5 M TCEP-HCl, 4 M urea and 0.5% formic acid) to each sample for a 90 second incubation at 20 °C.
  • the quenched samples were then injected into the Leaptec HDX PAL system for online pepsin/protease XIII digestion.
  • the digested peptides were trapped by a C18 column (2.1 mm x 5 mm, Waters) and separated by another C18 column (2.1 mm x 50 mm, Waters) at -5 °C with a 20 minute gradient (for mAb10989, mAb10987, mAb10934, and mAb10933) or a 10 minute gradient (for mAb10920, mAb10922, mAb10936, mAb10954, mAb10956, mAb10964, mAb10977, and mAb10984) from 0% to 90% of movbile phase B solution (mobile phase A solution: 0.5% formic acid and 4.5% acetonitrile in water, mobile phase B solution: 0.5% formic acid in acetonitrile).
  • the eluted peptides were analyzed by a Thermo Q Exactive HF mass spectrometry in LC-MS/MS or LC-MS mode.
  • the LC-MS/MS data from the undeuterated RBD protein sample were searched against a database including amino acid sequences of the RBD protein, pepsin, protease XIII, and their reversed sequences using Byonic search engine (Protein Metrics).
  • the search parameters were set as default using non-specific enzymatic digestion and human glycosylation as common variable modification.
  • Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • Peptides corresponding to amino acids 467-513 (DISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVL) (SEQ ID NO: 835) of the RBD were significantly protected by mAb10989.
  • a total of 187 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10987 samples, representing 86.06% sequence coverage of RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • Peptides corresponding to amino acids 432-452 (CVIAWNSNNLDSKVGGNYNYL) (SEQ ID NO: 836) of the RBD were significantly protected by mAb10987.
  • a total of 188 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10934 samples, representing 86.06% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • Peptides corresponding to amino acids 467-510 (DISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRV) (SEQ ID NO: 839) of the RBD were significantly protected by mAb10933.
  • a total of 75 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10920 samples, representing 83.27% sequence coverage of the RBD.
  • Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • Peptides corresponding to amino acids 471-486 (EIYQAGSTPCNGVEGF) (SEQ ID NO: 840), and 491-515 (PLQSYGFQPTNGVGYQPYRVVVLSF) (SEQ ID NO: 841) of the RBD were significantly protected by mAb10920.
  • a total of 86 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10922 samples, representing 87.25% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less
  • any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • Peptides corresponding to amino acids 400-422 (FVIRGDEVRQIAPGQTGKIADYN) (SEQ ID NO: 845), 453-486 (YRLFRKSNLKPFERDISTEIYQAGSTPCNGVEGF) (SEQ ID NO: 846), and 490-515 (FPLQSYGFQPTNGVGYQPYRVVVLSF) (SEQ ID NO: 847) of the RBD were significantly protected by mAb10954.
  • a total of 109 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10964 samples, representing 83.67% sequence coverage of RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • peptide that exhibited a reduction in deuterium uptake of 5% or greater i.e., a D%D value of less than -5%, such as -6%, -10%, and so on
  • Peptides corresponding to amino acids 351-364 (YAWNRKRISNCVAD) (SEQ ID NO: 850), and 471-486 (EIYQAGSTPCNGVEGF) (SEQ ID NO: 840) of the RBD were significantly protected by mAb10977.
  • a total of 88 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10984 samples, representing 87.25% sequence coverage of RBD.
  • Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • Peptides corresponding to amino acids 400-422 (FVIRGDEVRQIAPGQTGKIADYN) (SEQ ID NO: 845), and 453-486 (YRLFRKSNLKPFERDISTEIYQAGSTPCNGVEGF) (SEQ ID NO: 846) of the RBD were significantly protected by mAb10984.
  • a total of 84 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10986 samples, representing 87.25% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected.
  • Table 18 RBD peptides with significant protection upon formation of RBD-mAb10920 complex comparing to RBD alone
  • Table 19 RBD peptides with significant protection upon formation of RBD-mAb10922 complex comparing to RBD alone
  • Table 20 RBD peptides with significant protection upon formation of RBD-mAb10936 complex comparing to RBD alone
  • Table 21 RBD peptides with significant protection upon formation of RBD-mAb10954 complex comparing to RBD alone
  • Table 23 RBD peptides with significant protection upon formation of RBD-mAb10977 complex comparing to RBD alone
  • Table 24 RBD peptides with significant protection upon formation of RBD-mAb10984 complex comparing to RBD alone
  • Example 12 Neutralization of SARS-CoV-2 wild-type and variant spike proteins [000221] To test whether anti-SARS-CoV-2 spike protein antibodies can neutralize SARS- CoV-2 variants, these antibodies were screened against a panel of VSV pseudotype viruses expressing wild-type and variant spike proteins. VSV pseudotype viruses were generated by transiently transfecting 293T cells with a plasmid encoding the SARS-CoV-2 spike protein or the same plasmid containing nucleotide variations that encode for known variants of the SARS- CoV-2 spike protein amino acid sequence. All plasmids were confirmed by Sanger sequencing.
  • VSV DG:mNeon /Spike pseudovirus or VSV DG:mNeon /Spike_(variant amino acid mutation) (for example, VSV DG:mNeon /Spike_H49Y).
  • Vero cells were seeded to 80% confluency in T225 flasks, the cells were washed with PBS (Gibco: 20012-043), TrypLE was added to detach cells from the flask, and Complete DMEM was added to inactivate trypsin.20,000 Vero cells were plated in in 100 ⁇ L of prewarmed Complete DMEM per well in 96 Well Black Polystyrene Microplate (Corning: 3904). On Day 2, VSV DG:mNeon /Spike pseudovirus was thawed on ice and diluted with Infection media.
  • Antibodies were diluted in a U-bottom 96 well plate, generating a dilution of each antibody in 210 ⁇ l Infection media at 2X assay concentration.120 ⁇ L of diluted antibodies were transferred to a fresh U-bottom plate, and media and an IgG1 control antibody were added to each plate.120 ⁇ l of diluted pseudovirus was added to every well except the media control wells. To those wells, 120 ⁇ L of Infection media was added. Pseudovirus with antibodies were incubated for 30 minutes at room temperature, then media was removed from Vero cells.100 ⁇ L of antibody/pseudovirus mixture were added to the cells, and then incubated at 37 °C, 5% CO2 for 24 hours.
  • Monoclonal antibodies and antibody combinations were serially diluted in DMEM (Quality Biological), supplemented with 10% (v/v) heat inactivated fetal bovine serum (Sigma), 1% (v/v) penicillin/streptomycin (Gemini Bio-products) and 1% (v/v) L-glutamine (2 mM final concentration, Gibco) (VeroE6 media) to a final volume of 250 mL.
  • 250 mL of VeroE6 media containing SARS-CoV-2 (WA-1) 1000 PFU/mL
  • the virus-antibody mixtures were incubated for 60 min at 37 °C.
  • plaque reduction neutralization titer (PRNT50) values the serum dilutions at which plaque formation was reduced by 50% relative to that of the untreated control
  • PRNT50 plaque reduction neutralization titer
  • Amino acid variants in spike (S) protein were identified from over 7000 publicly available SARS-CoV-2 sequences, representing globally circulating isolates, and cloned into VSV pseudoparticles. Neutralization assays with variant-encoding pseudoparticles were performed to assess the impact of each variant on neutralization potency of the monoclonal antibodies.
  • Table 27 illustrates the relative neutralization potency of monoclonal antibodies against variant encoding pseudoparticles relative to SARS-CoV-2 spike (S-wt) at a single concentration of 5 ⁇ g/ml. Percent of neutralization relative to S-wt was captured for each individual antibody and variant.
  • the Biacore CM5 sensor chip surface was first derivatized by amine coupling with either mouse anti-human Fc specific mAb (Regeneron, mAb2567) to capture anti-SARS-COV-2bmAbs. Binding studies were performed on human SARS-COV-2 RBD extracellular domain expressed with a C-terminal
  • SARS-COV-2 RBD-MMH myc-myc-hexahistidine
  • SARS-COV-2 RBD-mFc SARS-COV-2 RBD extracellular domain expressed with a C-terminal mouse IgG2a
  • association rate (ka) and dissociation rate (kd) were determined by fitting the real-time binding sensorgrams to a 1:1 binding model with mass transport limitation using BiaEvaluation software v3.1 or Biacore Insight Evaluation software v2.0. or curve-fitting software. Binding dissociation equilibrium constant (KD) and dissociative half-life (t1 ⁇ 2) were calculated from the kinetic rates as: [000230] Binding kinetics parameters for different SARS-COV-2 mAbs binding to different anti-SARS-COV-2 RBD reagents of the invention at 25 °C and 37 °C are shown in Tables 29 through 32, respectively. Table 29: Binding kinetics parameters of SARS-COV-2 RBD-MMH binding to anti-SARS- COV-2-S monoclonal antibodies at 25 °C.
  • Table 30 Binding kinetics parameters of SARS-COV-2 RBD-MMH binding to anti-SARS- COV-2-S monoclonal antibodies at 37 °C.
  • Table 31 Binding kinetics parameters of SARS-COV-2 RBD-mFc binding to anti-SARS- COV-2-S monoclonal antibodies at 25 °C.
  • Example 14 Anti-SARS-CoV-2 Antibodies block RBD binding to hACE2 as determined by ELISA [000231] An ELISA-based blocking assay was used to determine the ability of anti-SARS- CoV-2 antibodies to block the binding of the SARS-COV-2 Spike protein receptor binding domain (RBD) to its receptor, human angiotensin converting enzyme 2 (hACE2).
  • RBD SARS-COV-2 Spike protein receptor binding domain
  • hACE2 human angiotensin converting enzyme 2
  • the SARS-CoV-2 protein used in this assay was comprised of the receptor binding domain (RBD) portion of the SARS-CoV-2 Spike protein (amino acids Arg319-Phe541) expressed with the Fc portion of the human IgG1 at the c-terminus (SARS-CoV-2 RBD-hFc)
  • the human ACE2 protein used in the experiments was purchased from R&D Systems and was comprised of amino acids Gln18-Ser740 with a C-terminal 10X-Histidine tag (hACE2-His; NCBI Accession No. Q9BYF1). [000233] Experiments were carried out using the following procedure.
  • a monoclonal anti- Penta-His antibody (Qiagen) was coated at 1 ⁇ g/ml in PBS on a 96-well microtiter plate overnight at 4 °C.
  • the hACE2-His receptor was added at 0.2ug/ml in PBS and bound for two hours at room temperature (RT). Nonspecific binding sites were subsequently blocked using a 0.5% (w/v) solution of BSA in PBS.
  • a constant amount of 100pM of SARS-CoV-2 RBD-hFc protein was bound with anti-SARS-COV-2 antibodies and an isotype IgG1 antibody control at dilutions from 0.0008nM to 50nM in PBS +0.5% BSA.
  • the calculated IC50 value defined as the concentration of antibody required to block 50% of SARS-CoV-2 RBD-hFc binding to plate-coated hACE2-His, was used as an indicator of blocking potency. Percent blocking was defined based on the background-corrected binding signal observed at the highest antibody concentration tested using this formula and reported for all tested antibodies: [000235] Antibodies that blocked binding less than or equal to 50% at the highest concentration tested were classified as non-blockers and IC50 values were not reported for those antibodies. [000236] The ability of anti-SARS-CoV-2 antibodies to block SARS-CoV-2 RBD binding to human ACE2 was assessed using a blocking ELISA. In this assay 100pM SARS-COV-2 RBD- hFc was titrated with a wide range of the concentrations of the anti-SARS-CoV-2-S antibody and
  • RBD-hFc at 100 pM was titrated with anti-SARS-COV-2-S antibodies in serial dilutions from 50nM and bound RBD-hFc on immobilized hACE2 with a 10x histidine tag, and detected with HRP-conjugated anti-hFc antibody. NBD; no blocking detected.
  • Example 15 Cross-competition between mAb10987, mAb10989, mAb10933, and mAb10934
  • mAb10987, mAb10989, mAb10933, and mAb10934 were examined in cross- competition binding assays ( Figure 11), identifying several pairs of non-competing mAbs with picomolar neutralization potency that could potentially be combined to form antibody cocktails, e.g., mAb10987 and mAb0933.
  • Epitope binning of the anti-SARS-CoV-2-S mAbs was conducted in a pre-mix sandwich format involving competing mAbs against one another in a pairwise combinatorial manner for binding to SARS-CoV-2 RBD-MMH protein using a ForteBio Octet HTX biolayer interferometry instrument (Molecular Devices ForteBio LLC, Fremont, CA) with running buffer of 10 mM HEPES, 150 mM NaCl, 0.05% (v/v) Tween-20, pH 7.4, 1 mg/mL BSA. Assays were performed at 30 oC with continuous agitation at 1000 rpm.
  • mAb10933 targets the spike-like loop region on one edge of the ACE2 interface. Within that region, the residues that show the most significant HDX protection by mAb10933 face upward, suggesting that the Fab region of mAb10933 binds the RBD from the top direction, where mAb10933 will have significant collisions with ACE2. In order to avoid competition with mAb10933, mAb10987 only binds to the HDX-defined protected regions from the front or the lower left side (in the front view of mAb10987 in Figure 12). This is consistent with the neutralization data described above, as mAb10987 would orient it in a position that has high probability to interfere with ACE2.
  • Example 16 Structure determination of antibody-bound spike protein [000241] To better understand the binding of mAb10933 and mAb10987 to the spike protein RBD, structural analysis was performed via cryo-electron microscopy (cryoEM). Fab fragments of mAb10933 and mAb10987 were isolated using FabALACTICA kit (Genovis).600 ⁇ g of the mAb10933 Fab and 600 ⁇ g of mAb10987 Fab were mixed with 300 ⁇ g of SARS-CoV-2-S RBD and incubated on ice for ⁇ 1 hour then injected into a Superdex 200 increase gel filtration column equilibrated to 50 mM Tris pH 7.5, 150 mM NaCl.
  • Peak fractions containing the mAb10933 Fab - mAb10987 Fab - RBD complex were collected and concentrated using a 10 kDa MWCO centrifugal filter.
  • the protein sample was diluted to 1.5 mg/mL and 0.15% PMAL-C8 amphipol was added.3.5 ⁇ L of protein was deposited onto a freshly plasma cleaned UltrHommeoil grid (1.2/1.3, 300 mesh). Excess solution was blotted away using filter paper and plunge frozen into liquid ethane using a Vitrobot Mark IV.
  • the cryoEM grid was transferred to a Titan Krios (Thermo Fisher) equipped with a K3 detector (Gatan). Movies were collected using EPU (Thermo Fisher) at 105,000x magnification, corresponding to a pixel size of 0.85 ⁇ .
  • cryoSPARC v2.14.2.2,821 movies were aligned using patch motion correction and patch CTF estimation.2,197 aligned micrographs were selected for further processing on the basis of estimated defocus values and CTF fit resolutions.
  • An initial set of particles picked using blob picker were subjected to 2D classification to generate templates for template picking.989,553 particles picked by template picking were subjected to multiple rounds of 2D classification to remove unbound fabs and particles containing an incomplete complex.
  • Example 17 Cross-competition between anti-SARS-CoV-2-S mAbs
  • Binding competition between anti-SARS-CoV-2-S monoclonal antibodies (mAbs) was determined using a real time, label-free bio-layer interferometry (BLI) assay on the Octet HTX biosensor platform (Pall ForteBio Corp.). The entire experiment was performed at 25 °C in 10mM HEPES, 150mM NaCl, 3mM EDTA, and 0.05% v/v Surfactant Tween-20, 1mg/mL BSA,
  • SARS-COV-2 RBD-MMH pH 7.4 (HBS-EBT) buffer with the plate shaking at a speed of 1000rpm.
  • SARS-COV-2 RBD-MMH C-terminal myc-myc-hexahistidine
  • ⁇ 0.51nm of SARS-COV-2-S RBD-MMH was first captured onto anti-Penta-His antibody coated Octet biosensor tips (Fortebio Inc, # 18-5122) by submerging the biosensor tips for 1 minute in wells containing a 10 ⁇ g/mL solution of SARS-COV-2-S RBD- MMH.
  • the SARS-COV-2-S RBD-MMH captured biosensor tips were then saturated with a first anti-SARS-CoV-2-S monoclonal antibody (subsequently referred to as mAb-1) by dipping into wells containing 50 ⁇ g/mL solution of mAb-1 for 5 minutes.
  • the biosensor tips were then subsequently dipped into wells containing 50 ⁇ g/mL solution of a second anti-SARS-CoV-2 monoclonal antibody (subsequently referred to as mAb-2) for 5 minutes.
  • the biosensor tips were washed in HBS-ETB buffer in between every step of the experiment. The real-time binding response was monitored during the entire course of the experiment and the binding response at the end of every step was recorded.
  • Example 18 pH sensitivity of anti-SARS-CoV-2-S monoclonal antibodies binding to monomeric SARS-CoV-2-S RBD reagents measured at 37 oC [000245]
  • the dissociation rate constants (k d ) for different anti-SARS-CoV-2-S monoclonal antibodies in pH 7.4, pH 6.0, and pH5.0 buffers were determined using a real-time surface plasmon resonance (SPR)-based Biacore T200 biosensor.
  • SPR surface plasmon resonance
  • Binding studies were performed on human SARS- COV-2-S RBD extracellular domain expressed with a C-terminal myc-myc-hexahistidine (SARS-COV-2 RBD-MMH), Single concentrations of SARS-COV-2-S RBD-MMH (90nM) prepared in PBS-T-pH 7.4 buffer were injected at a flow rate of 25 ⁇ L/min for 3 minutes followed by the dissociation of bound SARS-COV-2-S RBD-MMH in PBS-T-pH 7.4, PBS-T- pH 6.0 or PBS-T PBS-T-pH 5.0 running buffers for 5 minutes.
  • SARS-COV-2 RBD-MMH Single concentrations of SARS-COV-2-S RBD-MMH (90nM) prepared in PBS-T-pH 7.4 buffer were injected at a flow rate of 25 ⁇ L/min for 3 minutes followed by the dissociation of bound SARS-COV-2-S RBD-MMH in PBS-
  • the dissociation rate constants (k d ) in four pH running buffers were determined by fitting the real-time binding sensorgrams to a 1:1 binding model using Scrubber 2.0c curve- fitting software.
  • the dissociative half-life (t1 ⁇ 2) was calculated from the kd values as: [000247]
  • the k d and t1 ⁇ 2 values for SARS-COV-2-S RBD-MMH binding to different anti- SARS-CoV-2-S monoclonal antibodies in PBS-T-pH 7.4 followed by dissociation in PBS-T-pH 7.4 and PBS-T-pH 6.0 at 37 °C are shown in Table 35.
  • Example 19 Anti-SARS-CoV-2-S antibodies binding to virus-like particles [000248] To investigate the ability of a panel of anti-SARS-CoV-2 monoclonal antibodies to bind SARS-CoV-2 Spike glycoprotein, an in vitro binding assay utilizing vesicular stomatitis virus (VSV) pseudotyped with SARS-CoV-2 Spike protein in an electrochemiluminescence based detection platform (MSD) was developed.
  • VSV vesicular stomatitis virus
  • VLPs viral like particles
  • Nonspecific binding sites were blocked by 2% BSA (w/v) in PBS for 1 hour at room temperature.
  • BSA w/v
  • anti-SARS-CoV-2 antibodies and a non-binding human IgG1 control diluted in PBS + 0.5% BSA at a range of concentrations from 0.0008nM to 50nM, and buffer with no antibody were added in duplicate and the plates incubated for 1 hour at room temperature with shaking.
  • the plates were then washed with 1X PBS to remove the unbound antibodies using an AquaMax2000 plate washer (MDS Analytical Technologies).
  • the plate-bound antibodies were detected with a SULFO-TAGTM-conjugated anti-human IgG antibody (Jackson Immunoresearch) for 1 hour at room temperature.
  • Binding to the immobilized VLPs on 96-well High Bind plates was performed with a series of antibody dilutions and the bound antibodies were detected using SULFO-TAGTM-conjugated anti-human IgG.
  • the binding signals from electrochemiluminescence were recorded on a Sector Imager 600 (MSD). RLU values were determined for the antibody binding to VLPs. All antibodies displayed a concentration-
  • Example 20 Anti-SARS-CoV-2-S antibodies binding to spike protein-expressing cells [000253] To investigate the ability of a panel of anti-SARS-CoV-2-S monoclonal antibodies to bind to SARS-CoV-2-S expressing cells, an in vitro binding assay utilizing SARS-CoV-2-S expressing cells in an electrochemiluminescence based detection platform (MSD) was developed.
  • MSD electrochemiluminescence based detection platform
  • Jurkat/Tet3G/hCD20/Tet-3G inducible cells were engineered to transiently express the SARS-CoV-2 Spike Protein (Accession number MN908947.3, amino acids 16-1211, Jurkat/Tet3G/hCD20/Tet-On 3G Inducible COVID-19 Spike Protein High Sorted), and flow cytometry sorted for selection of high expression of the SARS-CoV-2 protein.
  • Parental Jurkat/Tet3G/hCD20/Tet-3G were also included in the experiments as a negative binding control.
  • Nonspecific binding sites were blocked by 2% BSA (w/v) in PBS for one hour at room temperature.
  • BSA w/v
  • anti-SARS-CoV-2 antibodies and a non-binding human IgG1 control diluted in PBS + 0.5% BSA at a range of concentrations from 0.0008nM to 50nM, and buffer with no antibody were added in duplicate and the plates incubated for one hour at room temperature with shaking.
  • the plates were then washed with 1X PBS to remove the unbound antibodies using an AquaMax2000 plate washer (MDS Analytical Technologies).
  • the plate-bound antibodies were detected with a SULFO-TAGTM-conjugated anti-human IgG antibody (Jackson Immunoresearch) for one hour at room temperature.
  • Binding to the immobilized cells on 96-well high bind plates was performed with a series of antibody dilutions and the bound antibodies were detected using SULFO-TAGTM-conjugated anti-human IgG.
  • the binding signals from electrochemiluminescence were recorded on a Sector Imager 600 (MSD). All antibodies displayed a concentration-dependent binding and the ratio of the binding on spike expressing cells to the parental cells were analyzed at the concentration of 5.5nM and 0.20nM.
  • the binding results of the anti-SARS-COV-2-S mAbs at the two concentrations to Spike protein expressing and parental Jurkat cells are summarized in Table 38.
  • Table 38 Specificity of anti-SARS-CoV-2-S antibodies binding to spike protein-expressing Jurkat cells vs parental cells by electrochemiluminescence

Abstract

The present disclosure provides antibodies and antigen-binding fragments thereof that bind specifically to a coronavirus spike protein and methods of using such antibodies and fragments for treating or preventing viral infections (e.g., coronavirus infections). The present disclosure provides neutralizing human antigen-binding proteins that specifically bind to SARS-CoV-2-S, for example, antibodies or antigen-binding fragments thereof.

Description

ANTI-SARS-COV-2-SPIKE GLYCOPROTEIN ANTIBODIES AND ANTIGEN-BINDING FRAGMENTS SEQUENCE LISTING [0001] An official copy of the sequence listing is submitted concurrently with the specification electronically via EFS-Web as an ASCII formatted sequence listing with a file name of “10753WO01-Sequence.txt”, created on June 25, 2020, and having a size of 922,462 bytes. The sequence listing contained in this ASCII formatted document is part of the specification and is herein incorporated by reference in its entirety. FIELD OF THE INVENTION [0002] The present invention relates to antibodies and antigen-binding fragments that bind specifically to coronavirus spike proteins and methods for treating or preventing coronavirus infections with said antibodies and fragments. BACKGROUND OF THE INVENTION [0003] Newly identified viruses, such as coronaviruses, can be difficult to treat because they are not sufficiently characterized. The emergence of these newly identified viruses highlights the need for the development of novel antiviral strategies. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a newly-emergent coronavirus which causes a severe acute respiratory disease, COVID-19. SARS-CoV-2 was first identified from an outbreak in Wuhan, China and as of March 20, 2020, the World Health Organization has reported 209,839 confirmed cases in 168 countries, areas, or territories, resulting in 8,778 deaths. Clinical features of COVID-19 include fever, dry cough, and fatigue, and the disease can cause respiratory failure resulting in death. [0004] Thus far, there has been no vaccine or therapeutic agent to prevent or treat SARS-CoV- 2 infection. In view of the continuing threat to human health, there is an urgent need for preventive and therapeutic antiviral therapies for SARS-CoV-2 control. Because this virus uses its spike glycoprotein for interaction with the cellular receptor ACE2 and the serine protease TMPRSS2 for entry into a target cell, this spike protein represents an attractive target for antibody therapeutics. In particular, fully human antibodies that specifically bind to the SARS-  
CoV-2-Spike protein (SARS-CoV-2-S) with high affinity and that inhibit virus infectivity could be important in the prevention and treatment of COVID-19. SUMMARY OF THE INVENTION [0005] There is a need for neutralizing therapeutic anti-SARS-CoV-2-Spike protein (SARS- CoV-2-S) antibodies and their use for treating or preventing viral infection. The present disclosure addresses this need, in part, by providing human anti-SARS-CoV-2-S antibodies, such as those of Table 1, and combinations thereof including, for example, combinations with other therapeutics (e.g., anti-inflammatory agents, antimalarial agents, antiviral agents, or other antibodies or antigen-binding fragments), and methods of use thereof for treating viral infections. [0006] The present disclosure provides neutralizing human antigen-binding proteins that specifically bind to SARS-CoV-2-S, for example, antibodies or antigen-binding fragments thereof. [0007] In one aspect, the present disclosure provides an isolated recombinant antibody or antigen-binding fragment thereof that specifically binds to a coronavirus spike protein (CoV-S), wherein the antibody has one or more of the following characteristics: (a) binds to CoV-S with an EC50 of less than about 10-9 M; (b) demonstrates an increase in survival in a coronavirus- infected animal after administration to said coronavirus-infected animal, as compared to a comparable coronavirus-infected animal without said administration; and/or (c) comprises three heavy chain complementarity determining regions (CDRs) (CDR-H1, CDR-H2, and CDR-H3) contained within a heavy chain variable region (HCVR) comprising an amino acid sequence having at least about 90% sequence identity to an HCVR of Table 1; and three light chain CDRs (CDR-L1, CDR-L2, and CDR-L3) contained within a light chain variable region (LCVR) comprising an amino acid sequence having at least about 90% sequence identity to an LCVR Table 1. [0008] In some embodiments, the antibody or antigen-binding fragment comprises: (a) an immunoglobulin heavy chain variable region comprising the CDR-H1, CDR-H2, and CDR-H3 of an antibody of Table 1; and/or (b) an immunoglobulin light chain variable region comprising the CDR-L1, CDR-L2, and CDR-L3 of an antibody of Table 1. [0009] In some embodiments, the antibody or antigen-binding fragment comprises: (a) a heavy chain immunoglobulin variable region comprising an amino acid sequence having at least 90%  
amino acid sequence identity to an HCVR sequence of Table 1; and/or (b) a light chain immunoglobulin variable region comprising an amino acid sequence having at least 90% amino acid sequence identity to an LCVR sequence of Table 1. [00010] In some embodiments, the antibody or antigen-binding fragment comprises the CDR- H1, CDR-H2, CDR-H3, CDR-L1, CDR-L2, and CDR-L3 of a single antibody of Table 1. In some embodiments, the antibody or antigen-binding fragment comprises an immunoglobulin that comprises the HCVR and the LCVR of a single antibody of Table 1. [00011] In one aspect, the present disclosure provides an antigen-binding protein that competes with any one of the antibodies or antigen-binding fragments discussed above or herein for binding to CoV-S. [00012] In one aspect, the present disclosure provides an antigen-binding protein that binds to the same epitope as, or to an overlapping epitope on, CoV-S as any one of the antibodies or antigen-binding fragments discussed above or herein. [00013] In any of the various embodiments, the antibody or antigen-binding fragment may be multispecific. [00014] In any of the various embodiments, the antibody or antigen-binding fragment may comprise one or more of the following properties: a) inhibits growth of coronavirus; b) binds to the surface of a coronavirus; c) limits spread of coronavirus infection of cells in vitro; and d) protects mice engineered to express the human ACE2 or TMPRSS2 protein from death and/or weight loss caused by coronavirus infection. [00015] In any of the various embodiments, CoV-S is SARS-CoV-2-S. [00016] In one aspect, the present disclosure provides a complex comprising an antibody or antigen-binding fragment as discussed above or herein bound to a CoV-S polypeptide. In some embodiments, the CoV-S is SARS-CoV-2-S. [00017] In one aspect, the present disclosure provides a method for making an antibody or antigen-binding fragment as discussed above or herein, comprising: (a) introducing into a host cell one or more polynucleotides encoding said antibody or antigen-binding fragment; (b) culturing the host cell under conditions favorable to expression of the one or more polynucleotides; and (c) optionally, isolating the antibody or antigen-binding fragment from the host cell and/or a medium in which the host cell is grown. In some embodiments, the host cell is a Chinese hamster ovary cell.  
[00018] In one aspect, the present disclosure provides an antibody or antigen-binding fragment that is a product of the method discussed above. [00019] In one aspect, the present disclosure provides a polypeptide comprising: (a) CDR-H1, CDR-H2, and CDR-H3 of an HCVR domain of an antibody or antigen-binding fragment that comprises an HCVR amino acid sequence set forth in Table 1; or (b) CDR-L1, CDR-L2, and CDR-L3 of an LCVR domain of an immunoglobulin chain that comprises an LCVR amino acid sequence set forth in Table 1. [00020] In one aspect, the present disclosure provides a polynucleotide encoding the polypeptide discussed above. [00021] In one aspect, the present disclosure provides a vector comprising the polynucleotide discussed above. [00022] In one aspect, the present disclosure provides a host cell comprising the antibody or antigen-binding fragment or polypeptide or polynucleotide or vector as discussed above or herein. [00023] In one aspect, the present disclosure provides a composition or kit comprising the antibody or antigen-binding fragment discussed above or herein in association with a further therapeutic agent. [00024] In one aspect, the present disclosure provides a pharmaceutical composition comprising the antigen-binding protein, antibody or antigen-binding fragment discussed above or herein and a pharmaceutically acceptable carrier and, optionally, a further therapeutic agent. In some embodiments, the further therapeutic agent is an anti-viral drug or a vaccine. In some embodiments, the further therapeutic agent is selected from the group consisting of: an anti- inflammatory agent, an antimalarial agent, an antibody or antigen-binding fragment thereof that specifically binds TMPRSS2, and an antibody or antigen-binding fragment thereof that specifically binds to CoV-S. In some cases, the antimalarial agent is chloroquine or hydroxychloroquine. In some cases, the anti-inflammatory agent is an antibody, such as sarilumab, tocilizumab, or gimsilumab. In some embodiments, the further therapeutic agent is a second antibody or antigen-binding fragment comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences of Table 1.  
[00025] In one aspect, the present disclosure provides a vessel or injection device comprising the antigen-binding protein, antibody or antigen-binding fragment, or composition as discussed above or herein. [00026] In one aspect, the present disclosure provides a method for treating or preventing infection with a coronavirus, in a subject in need thereof, comprising administering a therapeutically effective amount of an antigen-binding protein, antibody or antigen-binding fragment as discussed above or herein. In some embodiments, the coronavirus is selected from the group consisting of SARS-CoV-2, SARS-CoV, and MERS-CoV. [00027] In some embodiments of the method for treating or preventing infection with a coronavirus, the subject is administered one or more further therapeutic agents. In some cases, the one or more further therapeutic agents is an anti-viral drug or a vaccine. In some cases, the one or more further therapeutic agents is selected from the group consisting of: an anti- inflammatory agent, an antimalarial agent, an antibody or antigen-binding fragment thereof that specifically binds TMPRSS2, and an antibody or antigen-binding fragment thereof that specifically binds to CoV-S. In some cases, the antimalarial agent is chloroquine or hydroxychloroquine. In some cases, the anti-inflammatory agent is an antibody, such as for example, sarilumab, tocilizumab, or gimsilumab. In some embodiments, the further therapeutic agent is a second antibody or antigen-binding fragment comprising HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences of Table 1. Other antibodies that can be used alone or in combination with one another or with one or more of the antibodies disclosed herein for use in the context of the methods of the present disclosure include, e.g., LY-CoV555 (Eli Lilly); 47D11 (Wang et al Nature Communications Article No.2251); B38, H4, B5 and/or H2 (Wu et al., 10.1126/science.abc2241 (2020); STI-1499 (Sorrento Therapeutics); VIR-7831 and VIR-7832 (Vir Biotherapeutics). [00028] In one aspect, the present disclosure provides a method for administering an antibody or antigen-binding fragment discussed above or herein into the body of a subject comprising injecting the antibody or antigen-binding fragment into the body of the subject. In some embodiments, the antibody or antigen-binding fragment is injected into the body of the subject subcutaneously, intravenously or intramuscularly. [00029] In any of the various embodiments discussed above or herein, the antibody or antigen- binding binding fragment comprises a VH3-66 or Vk1-33 variable domain sequence.  
[00030] In one aspect, the present disclosure provides an isolated antibody or antigen-binding fragment thereof that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody or antigen-binding fragment comprises three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 210. [00031] In some embodiments, the HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 204, the HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 206, the HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 208, the LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 212, the LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 55, and the LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 214. In some embodiments, the isolated antibody or antigen-binding fragment thereof comprises an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202. In some embodiments, the isolated antibody or antigen-binding fragment thereof comprises an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210. In some embodiments, the isolated antibody or antigen-binding fragment thereof comprises an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210. [00032] In one aspect, the present disclosure provides an isolated antibody that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises an immunoglobulin constant region, three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 210. [00033] In some embodiments, the HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 204, the HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 206, the HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 208, the LCDR1 comprises  
the amino acid sequence set forth in SEQ ID NO: 212, the LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 55, and the LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 214. In some embodiments, the isolated antibody comprises an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210. In some embodiments, the isolated antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 216 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 218. In some cases, the immunoglobulin constant region is an IgG1 constant region. In some cases, the isolated antibody is a recombinant antibody. In some cases, the isolated antibody is multispecific. [00034] In one aspect, the present disclosure provides a pharmaceutical composition comprising an isolated antibody as discussed above or herein, and a pharmaceutically acceptable carrier or diluent. [00035] In some embodiments, the pharmaceutical composition further comprises a second therapeutic agent. In some cases, the second therapeutic agent is selected from the group consisting of: a second antibody, or an antigen-binding fragment thereof, that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, an anti- inflammatory agent, an antimalarial agent, and an antibody or antigen-binding fragment thereof that binds TMPRSS2. [00036] In some embodiments, the second therapeutic agent is a second antibody, or an antigen- binding fragment thereof, that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832. In some cases, the second antibody or antigen-binding fragment thereof comprises three heavy chain CDRs (HCDR1, HCDR2 and HCDR3) contained within an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 640, and three light chain CDRs (LCDR1, LCDR2 and LCDR3) contained within an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 646. In some cases, the second antibody or antigen-binding fragment thereof comprises: HCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 642; HCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 499; HCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 644; LCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 648; LCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 650; and LCDR3, comprising the amino acid   sequence set forth in SEQ ID NO: 652. In some cases, the second antibody or antigen-binding fragment thereof comprises an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 640 and an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 646. In some cases, the second antibody or antigen-binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 654 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 656. [00037] In one aspect, the present disclosure provides an isolated antibody or antigen-binding fragment thereof that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody or antigen-binding fragment comprises three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 640, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 646. [00038] In some embodiments, the HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 642, the HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 499, the HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 644, the LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 648, the LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 650, and the LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 652. In some embodiments, the isolated antibody or antigen-binding fragment thereof comprises an HCVR that comprises an amino acid sequence set forth in SEQ ID NO: 640. In some embodiments, the isolated antibody or antigen-binding fragment thereof comprises an LCVR that comprises an amino acid sequence set forth in SEQ ID NO: 646. In some embodiments, the isolated antibody or antigen-binding fragment thereof comprises an HCVR that comprises an amino acid sequence set forth in SEQ ID NO: 640 and an LCVR that comprises an amino acid sequence set forth in SEQ ID NO: 646. [00039] In one aspect, the present disclosure provides an isolated antibody that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises an immunoglobulin constant region, three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID  
NO: 640, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 646. [00040] In some embodiments, the HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 642, the HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 499, the HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 644, the LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 648, the LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 650, and the LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 652. In some embodiments, the isolated antibody comprises an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 640 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 646. In some embodiments, the isolated antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 654 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 656. In some cases, the immunoglobulin constant region is an IgG1 constant region. In some cases, the isolated antibody is a recombinant antibody. In some cases, the isolated antibody is multispecific. [00041] In one aspect, the present disclosure provides a pharmaceutical composition comprising an isolated antibody, as discussed above or herein, and a pharmaceutically acceptable carrier or diluent. [00042] In some embodiments, the pharmaceutical composition further comprising a second therapeutic agent. In some cases, the second therapeutic agent is selected from the group consisting of: a second antibody, or an antigen-binding fragment thereof, that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, an anti- inflammatory agent, an antimalarial agent, and an antibody or antigen-binding fragment thereof that binds TMPRSS2. [00043] In some embodiments, the second therapeutic agent is a second antibody, or an antigen- binding fragment thereof, that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832. In some cases, the second antibody or antigen-binding fragment thereof comprises three heavy chain CDRs (HCDR1, HCDR2 and HCDR3) contained within an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain CDRs (LCDR1, LCDR2 and LCDR3) contained within an LCVR comprising the  
amino acid sequence set forth in SEQ ID NO: 210. In some cases, the second antibody or antigen-binding fragment thereof comprises: HCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 204; HCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 206; HCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 208 LCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 212; LCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 55; and LCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 214. In some cases, the second antibody or antigen-binding fragment thereof comprises an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 210. In some cases, the second antibody or antigen-binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 216 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 218. [00044] In various embodiments, any of the features or components of embodiments discussed above or herein may be combined, and such combinations are encompassed within the scope of the present disclosure. Any specific value discussed above or herein may be combined with another related value discussed above or herein to recite a range with the values representing the upper and lower ends of the range, and such ranges are encompassed within the scope of the present disclosure. BRIEF DESCRIPTION OF THE FIGURES [00045] Figure 1 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2. [00046] Figure 2 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2. [00047] Figure 3 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2.  
[00048] Figure 4 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2. [00049] Figure 5 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2. [00050] Figure 6 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2. [00051] Figure 7 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2. [00052] Figure 8 shows ELISA blocking data for selected anti-SARS-CoV-2-S antibodies against SARS-CoV-2 spike protein, preventing the spike protein from binding to its receptor ACE2. [00053] Figure 9A and Figure 9B display V gene frequencies for paired Heavy (X-axis) and Light (Y-axis) chains of isolated neutralizing antibodies to SARS-CoV-2 for VelocImmune® mice (Figure 9A; N=185) and convalescent human donors (Figure 9B; N=68). The shade and size of the circle corresponds to the number of Heavy and Light chain pairs present in the repertoires of isolated neutralizing antibodies. Neutralization is defined as >70% with 1:4 dilution of antibody (~2 µg/ml) in VSV pseudoparticle neutralization assay. [00054] Figure 10A and Figure 10B display neutralization potency. Figure 10A displays the neutralization potency of anti-SARS-CoV-2 Spike mAbs. Serial dilutions of anti-Spike mAbs, IgG1 isotype control, and recombinant dimeric ACE2 (hACE2.hFc) were added with pVSV- SARS-CoV-2-S-mNeon to Vero cells and mNeon expression was measured 24 hours post- infection as a read-out for virus infectivity. Data is graphed as percent neutralization relative to virus only infection control. Figure 10B displays neutralization potency of individual anti-Spike mAbs and combinations of mAbs against SARS-CoV-2-S virus in VeroE6 cells. [00055] Figure 11 displays epitope bin analysis from a matrix of pre-mix binding assays for different anti-SARS-CoV-2 mAbs. Epitope binning was performed against nine anti-SARS- CoV-2 mAb as described. There were three phases (I, II, and III) for each graph. In phase I anti-  
SARS-CoV-2 mAb (20ug/ml) was loaded to the anti-human Fc probe. In phase II human IgG1 blocking mAb solution (100ug/ml). In phase III a solution of 100nM SARS CoV-2 RBD-MMH pre-mix complex of each 600 nM anti-SARS-CoV-2 mAb binding site flowed over the mAb capture probe. [00056] Figure 12 displays a 3D surface model for the structure of the Spike protein RBD domain showing the ACE2 interface and HDX-MS epitope mapping results. RBD residues protected by anti-SARS-CoV2-Spike antibodies are indicated with shading that represent the extent of protection as determined by HDX-MS experiments. The RBD structure is reproduced from PDB 6M17. [00057] Figure 13A and Figure 13B display a complex of mAb10933 and mAb10987 with the SARS-CoV-2 RBD. Figure 13A displays a 3.9 Å cryoEM map of mAb10933 + RBD + mAb10987 complex, shaded according to the chains in the refined model of Figure 13B.RBD, mAb10933 heavy and light chains, and mAb10987 heavy and light chain are identified. [00058] Figure 14 displays cryoEM data statistics. Data collection and refinement statistics are reported for the mAb10987 + mAb10933 + SARS-CoV-2 RBD complex structure shown in Figure 13A and Figure 13B. DETAILED DESCRIPTION OF THE INVENTION [00059] Before the present methods are described, it is to be understood that this invention is not limited to particular methods, and experimental conditions described, as such methods and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims. [00060] 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. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference in their entirety. [00061] The term “coronavirus” or “CoV” refers to any virus of the coronavirus family, including but not limited to SARS-CoV-2, MERS-CoV, and SARS-CoV. SARS-CoV-2 refers to  
the newly-emerged coronavirus which was identified as the cause of a serious outbreak starting in Wuhan, China, and which is rapidly spreading to other areas of the globe. SARS-CoV-2 has also been known as 2019-nCoV and Wuhan coronavirus. It binds via the viral spike protein to human host cell receptor angiotensin-converting enzyme 2 (ACE2). The spike protein also binds to and is cleaved by TMPRSS2, which activates the spike protein for membrane fusion of the virus. [00062] The term “CoV-S”, also called “S” or “S protein” refers to the spike protein of a coronavirus, and can refer to specific S proteins such as SARS-CoV-2-S, MERS-CoV S, and SARS-CoV S. The SARS-CoV-2-Spike protein is a 1273 amino acid type I membrane glycoprotein which assembles into trimers that constitute the spikes or peplomers on the surface of the enveloped coronavirus particle. The protein has two essential functions, host receptor binding and membrane fusion, which are attributed to the N-terminal (S1) and C-terminal (S2) halves of the S protein. CoV-S binds to its cognate receptor via a receptor binding domain (RBD) present in the S1 subunit. The amino acid sequence of full-length SARS-CoV-2 spike protein is exemplified by the amino acid sequence provided in SEQ ID NO: 832. The term “CoV-S” includes protein variants of CoV spike protein isolated from different CoV isolates as well as recombinant CoV spike protein or a fragment thereof. The term also encompasses CoV spike protein or a fragment thereof coupled to, for example, a histidine tag, mouse or human Fc, or a signal sequence such as ROR1. [00063] The term “coronavirus infection” or “CoV infection,” as used herein, refers to infection with a coronavirus such as SARS-CoV-2, MERS-CoV, or SARS-CoV. The term includes coronavirus respiratory tract infections, often in the lower respiratory tract. Symptoms can include high fever, dry cough, shortness of breath, pneumonia, gastro-intestinal symptoms such as diarrhea, organ failure (kidney failure and renal dysfunction), septic shock, and death in severe cases. Viruses [00064] The present invention includes methods for treating or preventing a viral infection in a subject. The term “virus” includes any virus whose infection in the body of a subject is treatable or preventable by administration of an anti-CoV-S antibody or antigen-binding fragment thereof (e.g., wherein infectivity of the virus is at least partially dependent on CoV-S). In an  
embodiment of the invention, a “virus” is any virus that expresses spike protein (e.g., CoV-S). The term “virus” also includes a CoV-S-dependent respiratory virus which is a virus that infects the respiratory tissue of a subject (e.g., upper and/or lower respiratory tract, trachea, bronchi, lungs) and is treatable or preventable by administration of an anti-CoV-S antibody or antigen- binding fragment thereof. For example, in an embodiment of the invention, virus includes coronavirus, SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), SARS-CoV (severe acute respiratory syndrome coronavirus), and MERS-CoV (Middle East respiratory syndrome (MERS) coronavirus). Coronaviruses can include the genera of alphacoronaviruses, betacoronaviruses, gammacoronaviruses, and deltacoronaviruses. In some embodiments, the antibodies or antigen-binding fragments provided herein can bind to and/or neutralize an alphacoronavirus, a betacoronavirus, a gammacoronavirus, and/or a deltacoronavirus. In certain embodiments, this binding and/or neutralization can be specific for a particular genus of coronavirus or for a particular subgroup of a genus. “Viral infection” refers to the invasion and multiplication of a virus in the body of a subject. [00065] Coronavirus virions are spherical with diameters of approximately 125 nm. The most prominent feature of coronaviruses is the club-shape spike projections emanating from the surface of the virion. These spikes are a defining feature of the virion and give them the appearance of a solar corona, prompting the name, coronaviruses. Within the envelope of the virion is the nucleocapsid. Coronaviruses have helically symmetrical nucleocapsids, which is uncommon among positive-sense RNA viruses, but far more common for negative-sense RNA viruses. SARS-CoV-2, MERS-CoV, and SARS-CoV belong to the coronavirus family. The initial attachment of the virion to the host cell is initiated by interactions between the S protein and its receptor. The sites of receptor binding domains (RBD) within the S1 region of a coronavirus S protein vary depending on the virus, with some having the RBD at the C-terminus of S1. The S-protein/receptor interaction is the primary determinant for a coronavirus to infect a host species and also governs the tissue tropism of the virus. Many coronaviruses utilize peptidases as their cellular receptor. Following receptor binding, the virus must next gain access to the host cell cytosol. This is generally accomplished by acid-dependent proteolytic cleavage of S protein by a cathepsin, TMPRRS2 or another protease, followed by fusion of the viral and cellular membranes.  
Anti-CoV-S Antibodies and Antigen-Binding Fragments [00066] The present invention provides antigen-binding proteins, such as antibodies and antigen-binding fragments thereof, that specifically bind to CoV spike protein or an antigenic fragment thereof. [00067] The term "antibody", as used herein, refers to immunoglobulin molecules comprising four polypeptide chains, two heavy chains (HCs) and two light chains (LCs) inter-connected by disulfide bonds (i.e., "full antibody molecules"), as well as multimers thereof (e.g. IgM). Exemplary antibodies include, for example, those listed in Table 1. Each heavy chain comprises a heavy chain variable region (“HCVR” or “VH”) and a heavy chain constant region (comprised of domains CH1, CH2 and CH3). Each light chain is comprised of a light chain variable region (“LCVR or “VL”) and a light chain constant region (CL). The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL comprises three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. Heavy chain CDRs can also be referred to as HCDRs or CDR-Hs, and numbered as described above (e.g., HCDR1, HCDR2, and HCDR3 or CDR-H1, CDR-H2, and CDR-H3). Likewise, light chain CDRs can be referred to as LCDRs or CDR-Ls, and numbered LCDR1, LCDR2, and LCDR3, or CDR-L1, CDR-L2, and CDR-L3. In certain embodiments of the invention, the FRs of the antibody (or antigen binding fragment thereof) are identical to the human germline sequences, or are naturally or artificially modified. Exemplary human germline sequences include, but are not limited to, VH3-66 and Vk1-33. Thus, the present disclosure provides anti-CoV-S antibodies or antigen- binding fragments thereof (e.g., anti-SARS-CoV-2-S antibodies or antigen-binding fragments thereof) comprising HCDR and LCDR sequences of Table 1 within a VH3-66 or Vk1-33 variable heavy chain or light chain region. The present disclosure further provides anti-CoV-S antibodies or antigen-binding fragments thereof (e.g., anti-SARS-CoV-2-S antibodies or antigen- binding fragments thereof) comprising HCDR and LCDR sequences of Table 1 within a combination of a light chain selected from IgKV4-1, IgKV 1-5, IgKV1-9, IgKV1-12, IgKV3-15, IgKV1-16, IgKV1-17, IgKV3-20, IgLV3-21, IgKV2-24, IgKV1-33, IgKV1-39, IgLV1-40, IgLV1-44, IgLV1-51, IgLV3-1, IgKV1-6, IgLV2-8, IgKV3-11, IgLV2-11, IgLV2-14, IgLV2-23, or IgLV6-57, and a heavy chain selected from IgHV1-69, IgHV3-64, IgHV4-59, IgHV3-53,  
IgHV3-48, IgHV4-34, IgHV3-33, IgHV3-30, IgHV3-23, IgHV3-20, IgHV1-18, IgHV3-15, IgHV3-11, IgHV3-9, IgHV1-8, IgHV3-7, IgHV2-5, IgHV1-2, IgHV2-70, IgHV3-66, IgHV5-51, IgHV1-46, IgHV4-39, IgHV4-31, IgHV3-30-3, IgHV2-26, or IgHV7-4-1. The present disclosure further provides anti-CoV-S antibodies or antigen-binding fragments thereof (e.g., anti-SARS-CoV-2-S antibodies or antigen-binding fragments thereof) comprising HCVR and LCVR sequences of Table 1 within a combination of a light chain selected from IgKV4-1, IgKV 1-5, IgKV1-9, IgKV1-12, IgKV3-15, IgKV1-16, IgKV1-17, IgKV3-20, IgLV3-21, IgKV2-24, IgKV1-33, IgKV1-39, IgLV1-40, IgLV1-44, IgLV1-51, IgLV3-1, IgKV1-6, IgLV2-8, IgKV3- 11, IgLV2-11, IgLV2-14, IgLV2-23, or IgLV6-57, and a heavy chain selected from IgHV1-69, IgHV3-64, IgHV4-59, IgHV3-53, IgHV3-48, IgHV4-34, IgHV3-33, IgHV3-30, IgHV3-23, IgHV3-20, IgHV1-18, IgHV3-15, IgHV3-11, IgHV3-9, IgHV1-8, IgHV3-7, IgHV2-5, IgHV1-2, IgHV2-70, IgHV3-66, IgHV5-51, IgHV1-46, IgHV4-39, IgHV4-31, IgHV3-30-3, IgHV2-26, or IgHV7-4-1. [00068] Typically, the variable domains of both the heavy and light immunoglobulin chains comprise three hypervariable regions, also called complementarity determining regions (CDRs), located within relatively conserved framework regions (FR). In general, from N-terminal to C- terminal, both light and heavy chains variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. In an embodiment of the invention, the assignment of amino acids to each domain is in accordance with the definitions of Sequences of Proteins of Immunological Interest, Kabat, et al.; National Institutes of Health, Bethesda, Md.; 5th ed.; NIH Publ. No.91-3242 (1991); Kabat (1978) Adv. Prot. Chem.32:1-75; Kabat, et al., (1977) J. Biol. Chem.252:6609- 6616; Chothia, et al., (1987) J Mol. Biol.196:901-917 or Chothia, et al., (1989) Nature 342:878- 883. [00069] The present invention includes monoclonal anti-CoV-S antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, as well as monoclonal compositions comprising a plurality of isolated monoclonal antigen-binding proteins. The term "monoclonal antibody", as used herein, refers to a population of substantially homogeneous antibodies, i.e., the antibody molecules comprising the population are identical in amino acid sequence except for possible naturally occurring mutations that may be present in minor amounts. A "plurality" of such monoclonal antibodies and fragments in a composition refers to a concentration of identical (i.e., as discussed above, in amino acid sequence except for possible naturally occurring  
mutations that may be present in minor amounts) antibodies and fragments which is above that which would normally occur in nature, e.g., in the blood of a host organism such as a mouse or a human. [00070] In an embodiment of the invention, an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment comprises a heavy chain constant domain, e.g., of the type IgA (e.g., IgA1 or IgA2), IgD, IgE, IgG (e.g., IgG1, IgG2, IgG3 and IgG4) or IgM. In an embodiment of the invention, an antigen-binding protein, e.g., antibody or antigen-binding fragment comprises a light chain constant domain, e.g., of the type kappa or lambda. [00071] The term "human” antigen-binding protein, such as an antibody, as used herein, includes antibodies having variable and constant regions derived from human germline immunoglobulin sequences whether in a human cell or grafted into a non-human cell, e.g., a mouse cell. See e.g., US8502018, US6596541 or US5789215. The human mAbs of the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example in the CDRs and, in particular, CDR3. However, the term "human antibody", as used herein, is not intended to include mAbs in which CDR sequences derived from the germline of another mammalian species (e.g., mouse) have been grafted onto human FR sequences. The term includes antibodies recombinantly produced in a non-human mammal or in cells of a non-human mammal. The term is not intended to include antibodies isolated from or generated in a human subject. See below. [00072] The present invention includes anti-CoV-S chimeric antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, and methods of use thereof. As used herein, a "chimeric antibody" is an antibody having the variable domain from a first antibody and the constant domain from a second antibody, where the first and second antibodies are from different species. (US4816567; and Morrison et al., (1984) Proc. Natl. Acad. Sci. USA 81: 6851-6855). [00073] The present invention includes anti-CoV-S hybrid antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, and methods of use thereof. As used herein, a "hybrid antibody" is an antibody having the variable domain from a first antibody and the constant domain from a second antibody, wherein the first and second antibodies are from different animals, or wherein the variable domain, but not the constant region, is from a first animal. For example, a variable domain can be taken from an antibody isolated from a human  
and expressed with a fixed constant region not isolated from that antibody. Exemplary hybrid antibodies are described in Example 1, which refers to antibody heavy chain variable region and light chain variable region derived PCR products that were cloned into expression vectors containing a heavy constant region and a light constant region, respectively. Hybrid antibodies are synthetic and non-natrually occurring because the variable and constant regions they contain are not isolated from a single natural source. [00074] The term “recombinant” antigen-binding proteins, such as antibodies or antigen-binding fragments thereof, refers to such molecules created, expressed, isolated or obtained by technologies or methods known in the art as recombinant DNA technology which include, e.g., DNA splicing and transgenic expression. The term includes antibodies expressed in a non- human mammal (including transgenic non-human mammals, e.g., transgenic mice), or a cell (e.g., CHO cells) expression system, or a non-human cell expression system, or isolated from a recombinant combinatorial human antibody library. In some embodiments, a recombinant antibody shares a sequence with an antibody isolated from an organism (e.g., a mouse or a human), but has been expressed via recombinant DNA technology. Such antibodies may have post-translational modifications (e.g., glycosylation) that differ from the antibody as isolated from the organism. [00075] Recombinant anti-CoV-S antigen-binding proteins, e.g., antibodies and antigen-binding fragments, disclosed herein may also be produced in an E. coli/T7 expression system. In this embodiment, nucleic acids encoding the anti-CoV-S antibody immunoglobulin molecules of the invention (e.g., as found in Table 1) may be inserted into a pET-based plasmid and expressed in the E. coli/T7 system. For example, the present invention includes methods for expressing an antibody or antigen-binding fragment thereof or immunoglobulin chain thereof in a host cell (e.g., bacterial host cell such as E. coli such as BL21 or BL21DE3) comprising expressing T7 RNA polymerase in the cell which also includes a polynucleotide encoding an immunoglobulin chain that is operably linked to a T7 promoter. For example, in an embodiment of the invention, a bacterial host cell, such as an E. coli, includes a polynucleotide encoding the T7 RNA polymerase gene operably linked to a lac promoter and expression of the polymerase and the chain is induced by incubation of the host cell with IPTG (isopropyl-beta-D- thiogalactopyranoside). See US4952496 and US5693489 or Studier & Moffatt, Use of  
bacteriophage T7 RNA polymerase to direct selective high-level expression of cloned genes, J. Mol. Biol.1986 May 5;189(1): 113-30. [00076] There are several methods by which to produce recombinant antibodies which are known in the art. One example of a method for recombinant production of antibodies is disclosed in US4816567. [00077] Transformation can be by any known method for introducing polynucleotides (e.g., DNA or RNA, including mRNA) into a host cell. Methods for introduction of heterologous polynucleotides into mammalian cells are well known in the art and include dextran-mediated transfection, calcium phosphate precipitation, polybrene-mediated transfection, protoplast fusion, electroporation, encapsulation of the polynucleotide(s) in liposomes, lipid nanoparticle technology, biolistic injection and direct microinjection of the DNA into nuclei. In addition, nucleic acid molecules may be introduced into mammalian cells by viral vectors such as lentivirus or adeno-associated virus. Methods of transforming cells are well known in the art. See, for example, U.S. Pat. Nos.4,399,216; 4,912,040; 4,740,461 and 4,959,455. In some embodiments, an antibody or antigen-binding fragment thereof of the present disclosure can be introduced to a subject in nucleic acid form (e.g, DNA or RNA, including mRNA), such that the subject’s own cells produce the antibody. The present disclosure further provides modifications to nucleotide sequences encoding the anti-CoV-S antibodies described herein that result in increased antibody expression, increased antibody stability, increased nucleic acid (e.g., mRNA) stability, or improved affinity or specificity of the antibodies for the CoV spike protein. [00078] Thus, the present invention includes recombinant methods for making an anti-CoV-S antigen-binding protein, such as an antibody or antigen-binding fragment thereof of the present invention, or an immunoglobulin chain thereof, comprising (i) introducing one or more polynucleotides (e.g., including the nucleotide sequence of any one or more of the sequences of Table 2) encoding light and/or heavy immunoglobulin chains, or CDRs, of the antigen-binding protein, e.g., of Table 1, for example, wherein the polynucleotide is in a vector; and/or integrated into a host cell chromosome and/or is operably linked to a promoter; (ii) culturing the host cell (e.g., CHO or Pichia or Pichia pastoris) under condition favorable to expression of the polynucleotide and, (iii) optionally, isolating the antigen-binding protein, (e.g., antibody or fragment) or chain from the host cell and/or medium in which the host cell is grown. For example, a polynucleotide can be integrated into a host cell chromosome through targeted  
insertion with a vector such as adeno-associated virus (AAV), e.g., after cleavage of the chromosome using a gene editing system (e.g., CRISPR (for example, CRISPR-Cas9), TALEN, megaTAL, zinc finger, or Argonaute). Targeted insertions can take place, for example, at host cell loci such as an albumin or immunoglopbulin genomic locus. Alternatively, insertion can be at a random locus, e.g., using a vector such as lentivirus. When making an antigen-binding protein (e.g., antibody or antigen-binding fragment) comprising more than one immunoglobulin chain, e.g., an antibody that comprises two heavy immunoglobulin chains and two light immunoglobulin chains, co-expression of the chains in a single host cell leads to association of the chains, e.g., in the cell or on the cell surface or outside the cell if such chains are secreted, so as to form the antigen-binding protein (e.g., antibody or antigen-binding fragment). The methods include those wherein only a heavy immunoglobulin chain or only a light immunoglobulin chain (e.g., any of those discussed herein including mature fragments and/or variable domains thereof) is expressed. Such chains are useful, for example, as intermediates in the expression of an antibody or antigen-binding fragment that includes such a chain. For example, the present invention also includes anti-CoV-S antigen-binding proteins, such as antibodies and antigen-binding fragments thereof, comprising a heavy chain immunoglobulin (or variable domain thereof or comprising the CDRs thereof) encoded by a polynucleotide comprising a nucleotide sequence set forth in Table 2 and a light chain immunoglobulin (or variable domain thereof or comprising the CDRs thereof) encoded by a nucleotide sequence set forth in Table 2 which are the product of such production methods, and, optionally, the purification methods set forth herein. For example, in some embodiments, the product of the method is an anti-CoV-S antigen-binding protein which is an antibody or fragment comprising an HCVR comprising an amino acid sequence set forth in Table 1 and an LCVR comprising an amino acid sequence set forth in Table 1, wherein the HCVR and LCVR sequences are selected from a single antibody listed in Table 1. In some embodiments, the product of the method is an anti-CoV-S antigen-binding protein which is an antibody or fragment comprising HCDR1, HCDR2, and HCDR3 comprising amino acid sequences set forth in Table 1 and LCDR1, LCDR2, and LCDR3 comprising amino acid sequences set forth in Table 1, wherein the six CDR sequences are selected from a single antibody listed in Table 1. In some embodiments, the product of the method is an anti-CoV-S antigen-binding protein which is an antibody or fragment  
comprising a heavy chain comprising an HC amino acid sequence set forth in Table 1 and a light chain comprising an LC amino acid sequence set forth in Table 1. [00079] Eukaryotic and prokaryotic host cells, including mammalian cells, may be used as hosts for expression of an anti-CoV-S antigen-binding protein. Such host cells are well known in the art and many are available from the American Type Culture Collection (ATCC). These host cells include, inter alia, Chinese hamster ovary (CHO) cells, NS0, SP2 cells, HeLa cells, baby hamster kidney (BHK) cells, monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells, 3T3 cells, HEK-293 cells and a number of other cell lines. Mammalian host cells include human, mouse, rat, dog, monkey, pig, goat, bovine, horse and hamster cells. Other cell lines that may be used are insect cell lines (e.g., Spodoptera frugiperda or Trichoplusia ni), amphibian cells, bacterial cells, plant cells and fungal cells. Fungal cells include yeast and filamentous fungus cells including, for example, Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia minuta (Ogataea minuta, Pichia lindneri), Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei, Chrysosporium lucknowense, Fusarium sp., Fusarium gramineum, Fusarium venenatum, Physcomitrella patens and Neurospora crassa. The present invention includes an isolated host cell (e.g., a CHO cell) comprising an antigen-binding protein, such as those of Table 1; or a polynucleotide encoding such a polypeptide thereof. [00080] The term “specifically binds” refers to those antigen-binding proteins (e.g., mAbs) having a binding affinity to an antigen, such as a CoV-S protein (e.g., SARS-CoV-2-S), expressed as KD, of at least about 10-8 M, as measured by real-time, label free bio-layer interferometry assay, for example, at 25oC or 37oC, e.g., an Octet® HTX biosensor, or by surface plasmon resonance, e.g., BIACORE™, or by solution-affinity ELISA. The present invention includes antigen-binding proteins that specifically bind to a CoV-S protein. [00081] The terms "antigen-binding portion" or "antigen-binding fragment" of an antibody or antigen-binding protein, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex. Non-limiting examples of antigen-binding  
fragments include: (i) Fab fragments; (ii) F(ab')2 fragments; (iii) Fd fragments; (iv) Fv fragments; (v) single-chain Fv (scFv) molecules; (vi) dAb fragments; and (vii) minimal recognition units consisting of the amino acid residues that mimic the hypervariable region of an antibody (e.g., an isolated complementarity determining region (CDR) such as a CDR3 peptide), or a constrained FR3-CDR3-FR4 peptide. Other engineered molecules, such as domain-specific antibodies, single domain antibodies, domain-deleted antibodies, chimeric antibodies, CDR- grafted antibodies, diabodies, triabodies, tetrabodies, minibodies, nanobodies (e.g., as defined in WO08/020079 or WO09/138519) (e.g., monovalent nanobodies, bivalent nanobodies, etc.), small modular immunopharmaceuticals (SMIPs), and shark variable IgNAR domains, are also encompassed within the expression "antigen-binding fragment," as used herein. In an embodiment of the invention, the antigen-binding fragment comprises three or more CDRs of an antibody of Table 1 (e.g., CDR-H1, CDR-H2 and CDR-H3; or CDR-L1, CDR-L2 and CDR-L3). [00082] An antigen-binding fragment of an antibody will, in an embodiment of the invention, comprise at least one variable domain. The variable domain may be of any size or amino acid composition and will generally comprise at least one CDR, which is adjacent to or in frame with one or more framework sequences. In antigen-binding fragments having a VH domain associated with a VL domain, the VH and VL domains may be situated relative to one another in any suitable arrangement. For example, the variable region may be dimeric and contain VH - VH, VH - VL or VL - VL dimers. Alternatively, the antigen-binding fragment of an antibody may contain a monomeric VH or VL domain. [00083] In certain embodiments, an antigen-binding fragment of an antibody may contain at least one variable domain covalently linked to at least one constant domain. Non-limiting, exemplary configurations of variable and constant domains that may be found within an antigen- binding fragment of an antibody of the present invention include: (i) VH-CH1; (ii) VH-CH2; (iii) VH-CH3; (iv) VH-CH1-CH2; (v) VH-CH1-CH2-CH3; (vi) VH-CH2-CH3; (vii) VH-CL; (viii) VL-CH1; (ix) VL-CH2; (x) VL-CH3; (xi) VL-CH1-CH2; (xii) VL-CH1-CH2-CH3; (xiii) VL-CH2-CH3; and (xiv) VL-CL. In any configuration of variable and constant domains, including any of the exemplary configurations listed above, the variable and constant domains may be either directly linked to one another or may be linked by a full or partial hinge or linker region. A hinge region may consist of at least 2 (e.g., 5, 10, 15, 20, 40, 60 or more) amino acids, which result in a flexible or semi-flexible linkage between adjacent variable and/or constant domains in a single polypeptide  
molecule. Moreover, an antigen-binding fragment of an antibody of the present invention may comprise a homo-dimer or hetero-dimer (or other multimer) of any of the variable and constant domain configurations listed above in non-covalent association with one another and/or with one or more monomeric VH or VL domain (e.g., by disulfide bond(s)). [00084] Antigen-binding proteins (e.g., antibodies and antigen-binding fragments) may be mono-specific or multi-specific (e.g., bi-specific). Multispecific antigen-binding proteins are discussed further herein. [00085] In specific embodiments, antibody or antibody fragments of the invention may be conjugated to a moiety such a ligand or a therapeutic moiety (“immunoconjugate”), such as an anti-viral drug, a second anti-influenza antibody, or any other therapeutic moiety useful for treating a viral infection, e.g., influenza viral infection. See below. [00086] The present invention also provides a complex comprising an anti-CoV-S antigen- binding protein, e.g., antibody or antigen-binding fragment, discussed herein complexed with CoV-S polypeptide or an antigenic fragment thereof and/or with a secondary antibody or antigen-binding fragment thereof (e.g., detectably labeled secondary antibody) that binds specifically to the anti-CoV-S antibody or fragment. In an embodiment of the invention, the antibody or fragment is in vitro (e.g., is immobilized to a solid substrate) or is in the body of a subject. In an embodiment of the invention, the CoV-S is in vitro (e.g., is immobilized to a solid substrate) or is on the surface of a virus or is in the body of a subject. Immobilized anti-CoV-S antibodies and antigen-binding fragments thereof which are covalently linked to an insoluble matrix material (e.g., glass or polysaccharide such as agarose or sepharose, e.g., a bead or other particle thereof) are also part of the present invention; optionally, wherein the immobilized antibody is complexed with CoV-S or antigenic fragment thereof or a secondary antibody or fragment thereof. [00087] "Isolated" antigen-binding proteins, antibodies or antigen-binding fragments thereof, polypeptides, polynucleotides and vectors, are at least partially free of other biological molecules from the cells or cell culture from which they are produced. Such biological molecules include nucleic acids, proteins, other antibodies or antigen-binding fragments, lipids, carbohydrates, or other material such as cellular debris and growth medium. An isolated antibody or antigen- binding fragment may further be at least partially free of expression system components such as biological molecules from a host cell or of the growth medium thereof. Generally, the term  
"isolated" is not intended to refer to a complete absence of such biological molecules or to an absence of water, buffers, or salts or to components of a pharmaceutical formulation that includes the antibodies or fragments. [00088] The term “epitope” refers to an antigenic determinant (e.g., a CoV-S polypeptide) that interacts with a specific antigen-binding site of an antigen-binding protein, e.g., a variable region of an antibody molecule, known as a paratope. A single antigen may have more than one epitope. Thus, different antibodies may bind to different areas on an antigen and may have different biological effects. The term “epitope” also refers to a site on an antigen to which B and/or T cells respond. It also refers to a region of an antigen that is bound by an antibody. Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction. Epitopes may be linear or conformational, that is, composed of non-linear amino acids. In certain embodiments, epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics. [00089] Methods for determining the epitope of an antigen-binding protein, e.g., antibody or fragment or polypeptide, include alanine scanning mutational analysis, peptide blot analysis (Reineke (2004) Methods Mol. Biol.248: 443-63), peptide cleavage analysis, crystallographic studies and NMR analysis. In addition, methods such as epitope excision, epitope extraction and chemical modification of antigens can be employed (Tomer (2000) Prot. Sci.9: 487-496). Another method that can be used to identify the amino acids within a polypeptide with which an antigen-binding protein (e.g., antibody or fragment or polypeptide) (e.g., coversin) interacts is hydrogen/deuterium exchange detected by mass spectrometry. In general terms, the hydrogen/deuterium exchange method involves deuterium-labeling the protein of interest, followed by binding the antigen-binding protein, e.g., antibody or fragment or polypeptide, to the deuterium-labeled protein. Next, the CoV-S protein/ antigen-binding protein 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/ antigen-binding protein interface may retain deuterium and  
therefore exhibit relatively higher mass compared to amino acids not included in the interface. After dissociation of the antigen-binding protein (e.g., antibody or fragment or polypeptide), the target protein is subjected to protease cleavage and mass spectrometry analysis, thereby revealing the deuterium-labeled residues which correspond to the specific amino acids with which the antigen-binding protein interacts. See, e.g., Ehring (1999) Analytical Biochemistry 267: 252-259; Engen and Smith (2001) Anal. Chem.73: 256A-265A. [00090] The term “competes” as used herein, refers to an antigen-binding protein (e.g., antibody or antigen-binding fragment thereof) that binds to an antigen (e.g., CoV-S) and inhibits or blocks the binding of another antigen-binding protein (e.g., antibody or antigen-binding fragment thereof) to the antigen. The term also includes competition between two antigen-binding proteins e.g., antibodies, in both orientations, i.e., a first antibody that binds and blocks binding of second antibody and vice versa. In certain embodiments, the first antigen-binding protein (e.g., antibody) and second antigen-binding protein (e.g., antibody) may bind to the same epitope. Alternatively, the first and second antigen-binding proteins (e.g., antibodies) may bind to different, but, for example, overlapping epitopes, wherein binding of one inhibits or blocks the binding of the second antibody, e.g., via steric hindrance. Competition between antigen-binding proteins (e.g., antibodies) may be measured by methods known in the art, for example, by a real- time, label-free bio-layer interferometry assay. Epitope mapping (e.g., via alanine scanning or hydrogen-deuterium exchange (HDX)) can be used to determine whether two or more antibodies are non-competing (e.g., on a spike protein receptor binding domain (RBD) monomer), competing for the same epitope, or competing but with diverse micro-epitopes (e.g., identified through HDX). In an embodiment of the invention, competition between a first and second anti- CoV-S antigen-binding protein (e.g., antibody) is determined by measuring the ability of an immobilized first anti-CoV-S antigen-binding protein (e.g., antibody) (not initially complexed with CoV-S protein) to bind to soluble CoV-S protein complexed with a second anti-CoV-S antigen-binding protein (e.g., antibody). A reduction in the ability of the first anti-CoV-S antigen-binding protein (e.g., antibody) to bind to the complexed CoV-S protein, relative to uncomplexed CoV-S protein, indicates that the first and second anti-CoV-S antigen-binding proteins (e.g., antibodies) compete. The degree of competition can be expressed as a percentage of the reduction in binding. Such competition can be measured using a real time, label-free bio-  
layer interferometry assay, e.g., on an Octet RED384 biosensor (Pall ForteBio Corp.), ELISA (enzyme-linked immunosorbent assays) or SPR (surface plasmon resonance). [00091] Binding competition between anti-CoV-S antigen-binding proteins (e.g., monoclonal antibodies (mAbs)) can be determined using a real time, label-free bio-layer interferometry assay on an Octet RED384 biosensor (Pall ForteBio Corp.). For example, to determine competition between two anti-CoV-S monoclonal antibodies, the anti-CoV-S mAb can be first captured onto anti-hFc antibody coated Octet biosensor tips (Pall ForteBio Corp., # 18-5060) by submerging the tips into a solution of anti-CoV-S mAb (subsequently referred to as “mAb1”). As a positive- control for blocking, the antibody captured biosensor tips can then be saturated with a known blocking isotype control mAb (subsequently referred to as “blocking mAb”) by dipping into a solution of blocking mAb. To determine if mAb2 competes with mAb1, the biosensor tips can then be subsequently dipped into a co-complexed solution of CoV-S polypeptide and a second anti-CoV-S mAb (subsequently referred to as “mAb2”), that had been pre-incubated for a period of time and binding of mAb1 to the CoV-S polypeptide can be determined. The biosensor tips can be washed in buffer in between every step of the experiment. The real-time binding response can be monitored during the course of the experiment and the binding response at the end of every step can be recorded. [00092] For example, in an embodiment of the invention, the competition assay is conducted at 25 oC and pH about 7, e.g., 7.4, e.g., in the presence of buffer, salt, surfactant and a non-specific protein (e.g., bovine serum albumin). [00093] Typically, an antibody or antigen-binding fragment of the invention which is modified in some way retains the ability to specifically bind to CoV-S, e.g., retains at least 10% of its CoV-S binding activity (when compared to the parental antibody) when that activity is expressed on a molar basis. Preferably, an antibody or antigen-binding fragment of the invention retains at least 20%, 50%, 70%, 80%, 90%, 95% or 100% or more of the CoV-S binding affinity as the parental antibody. It is also intended that an antibody or antigen-binding fragment of the invention can include conservative or non-conservative amino acid substitutions (referred to as “conservative variants” or “function conserved variants” of the antibody) that do not substantially alter its biologic activity. [00094] A “variant” of a polypeptide, such as an immunoglobulin chain (e.g., mAb8021 VH, VL, HC, or LC, mAb8028 VH, VL, HC, or LC, or mAb8029 VH, VL, HC, or LC), refers to a  
polypeptide comprising an amino acid sequence that is at least about 70-99.9% (e.g., 70, 72, 74, 75, 76, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, 99.9%) identical or similar to a referenced amino acid sequence that is set forth herein (e.g., SEQ ID NO: 2, 10, 18, 20, 22, 30, 38, 40, 42, 50, 58, or 60); when the comparison is performed by a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences (e.g., expect threshold: 10; word size: 3; max matches in a query range: 0; BLOSUM 62 matrix; gap costs: existence 11, extension 1; conditional compositional score matrix adjustment). [00095] A “variant” of a polynucleotide refers to a polynucleotide comprising a nucleotide sequence that is at least about 70-99.9% (e.g., at least about 70, 72, 74, 75, 76, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or 99.9%) identical to a referenced nucleotide sequence that is set forth herein (e.g., SEQ ID NO: 1, 9, 17, 19, 21, 29, 37, 39, 41, 49, 57, or 59); when the comparison is performed by a BLAST algorithm wherein the parameters of the algorithm are selected to give the largest match between the respective sequences over the entire length of the respective reference sequences (e.g., expect threshold: 10; word size: 28; max matches in a query range: 0; match/mismatch scores: 1, -2; gap costs: linear). [00096] Anti-CoV-S antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof of the present invention, in an embodiment of the invention, include a heavy chain immunoglobulin variable region having at least 70% (e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater) amino acid sequence identity to the HCVR amino acid sequences set forth in Table 1; and/or a light chain immunoglobulin variable region having at least 70% (e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater) amino acid sequence identity to the LCVR amino acid sequences set forth in Table 1. [00097] In addition, a variant anti-CoV-S antigen-binding protein may include a polypeptide comprising an amino acid sequence that is set forth herein except for one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10) mutations such as, for example, missense mutations (e.g., conservative substitutions), non-sense mutations, deletions, or insertions. For example, the present invention includes antigen-binding proteins which include an immunoglobulin light chain variant comprising an LCVR amino acid sequence set forth in Table 1 but having one or more of such mutations and/or an immunoglobulin heavy chain variant comprising an HCVR amino acid sequence set forth in Table 1 but having one or more of such mutations. In an embodiment of  
the invention, a variant anti-CoV-S antigen-binding protein includes an immunoglobulin light chain variant comprising CDR-L1, CDR-L2 and CDR-L3 wherein one or more (e.g., 1 or 2 or 3) of such CDRs has one or more of such mutations (e.g., conservative substitutions) and/or an immunoglobulin heavy chain variant comprising CDR-H1, CDR-H2 and CDR-H3 wherein one or more (e.g., 1 or 2 or 3) of such CDRs has one or more of such mutations (e.g., conservative substitutions). Substitutions can be in a CDR, framework, or constant region. [00098] The invention further provides variant anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments thereof, comprising one or more variant CDRs (e.g., any one or more of CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and/or CDR-H3) that are set forth herein with at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 99.9% sequence identity or similarity to, e.g., the heavy chain and light chain CDRs of Table 1. [00099] Embodiments of the present invention also include variant antigen-binding proteins, e.g., anti-CoV-S antibodies and antigen-binding fragments thereof, that comprise immunoglobulin VHs and VLs; or HCs and LCs, which comprise an amino acid sequence having 70% or more (e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or greater) overall amino acid sequence identity or similarity to the amino acid sequences of the corresponding VHs, VLs, HCs or LCs specifically set forth herein, but wherein the CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and CDR-H3 of such immunoglobulins are not variants and comprise CDR amino acid sequence set forth in Table 1. Thus, in such embodiments, the CDRs within variant antigen-binding proteins are not, themselves, variants. [000100] Conservatively modified variant anti-CoV-S antibodies and antigen-binding fragments thereof are also part of the present invention. A “conservatively modified variant” or a “conservative substitution” refers to a variant wherein there is one or more substitutions of amino acids in a polypeptide with other amino acids having similar characteristics (e.g. charge, side-chain size, hydrophobicity/hydrophilicity, backbone conformation and rigidity, etc.). Such changes can frequently be made without significantly disrupting the biological activity of the antibody or fragment. Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the Gene, The  
Benjamin/Cummings Pub. Co., p.224 (4th Ed.)). In addition, substitutions of structurally or functionally similar amino acids are less likely to significantly disrupt biological activity. [000101] Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartate and glutamate, and 7) sulfur-containing side chains: cysteine and methionine. Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine. Alternatively, a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Science 256: 144345. [000102] Function-conservative variants of the anti-CoV-S antibodies and antigen-binding fragments thereof are also part of the present invention. Any of the variants of the anti-CoV-S antibodies and antigen-binding fragments thereof (as discussed herein) may be “function- conservative variants”. Such function-conservative variants may, in some cases, also be characterized as conservatively modified variants. “Function-conservative variants,” as used herein, refers to variants of the anti-CoV-S antibodies or antigen-binding fragments thereof in which one or more amino acid residues have been changed without significantly altering one or more functional properties of the antibody or fragment. In an embodiment of the invention, a function-conservative variant anti-CoV-S antibody or antigen-binding fragment thereof of the present invention comprises a variant amino acid sequence and exhibits one or more of the following functional properties: ^ Inhibits growth of coronavirus (e.g., SARS-CoV-2, SARS-CoV, and/or MERS-CoV) in ACE2- and/or TMPRSS2-expressing cells (e.g., Calu-3 cells); ^ Does not significantly bind to MDCK/Tet-on cells which do not express ACE2 and/or TMPRSS2; ^ Limits spread of coronavirus infection (e.g., by SARS-CoV-2, SARS-CoV, and/or MERS-CoV) of cells, e.g., Calu-3, in vitro; and/or ^ Protects a mouse engineered to express the human TMPRSS2 and/or ACE2 protein from death caused by coronavirus infection (e.g., SARS-CoV-2, SARS-CoV, or  
MERS-CoV), for example, wherein the mice are infected with an otherwise lethal dose of the virus, optionally when combined with a second therapeutic agent. ^ Protects a mouse engineered to express the human TMPRSS2 and/or ACE2 protein from weight loss caused by coronavirus infection (e.g., SARS-CoV-2, SARS-CoV, or MERS-CoV), for example, wherein the mice are infected with a dose of the virus that would otherwise cause weighht loss, optionally when combined with a second therapeutic agent. [000103] A “neutralizing” or “antagonist” anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment, refers to a molecule that inhibits an activity of CoV-S to any detectable degree, e.g., inhibits the ability of CoV-S to bind to a receptor such as ACE2, to be cleaved by a protease such as TMPRSS2, or to mediate viral entry into a host cell or viral reproduction in a host cell. [000104] Table 1 refers to antigen-binding proteins, such as antibodies and antigen-binding fragments thereof, that comprise the heavy chain or VH (or a variant thereof) and light chain or VL (or a variant thereof) as set forth below; or that comprise a VH that comprises the CDRs thereof (CDR-H1 (or a variant thereof), CDR-H2 (or a variant thereof) and CDR-H3 (or a variant thereof)) and a VL that comprises the CDRs thereof (CDR-L1 (or a variant thereof), CDR-L2 (or a variant thereof) and CDR-L3 (or a variant thereof)), e.g., wherein the immunoglobulin chains, variable regions and/or CDRs comprise the specific amino acid sequences described below. [000105] The antibodies described herein also include embodiments wherein the VH is fused to a wild-type IgG4 (e.g., wherein residue 108 is S) or to IgG4 variants (e.g., wherein residue 108 is P). [000106] Antibodies and antigen-binding fragments of the present invention comprise immunoglobulin chains including the amino acid sequences set forth herein as well as cellular and in vitro post-translational modifications to the antibody. For example, the present invention includes antibodies and antigen-binding fragments thereof that specifically bind to CoV-S comprising heavy and/or light chain amino acid sequences set forth herein (e.g., CDR-H1, CDR- H2, CDR-H3, CDR-L1, CDR-L2 and/or CDR-L3) as well as antibodies and fragments wherein one or more amino acid residues is glycosylated, one or more Asn residues is deamidated, one or more residues (e.g., Met, Trp and/or His) is oxidized, the N-terminal Gln is pyroglutamate (pyroE) and/or the C-terminal Lysine is missing.  
[000107] The amino acid and nucleotide sequences of exemplary anti-SARS-CoV-2-Spike protein (SARS-CoV-2-S) antibodies are shown in the Table of Exemparly Sequences, below. Table of Exemplary Sequences
Figure imgf000033_0001
 
Figure imgf000034_0001
 
Figure imgf000035_0001
 
Figure imgf000036_0001
 
Figure imgf000037_0001
 
Figure imgf000038_0001
 
Figure imgf000039_0001
 
Figure imgf000040_0001
 
Figure imgf000041_0001
 
Figure imgf000042_0001
 
Figure imgf000043_0001
 
Figure imgf000044_0001
Administration of Antibodies [000108] The present invention provides methods for administering an anti-CoV-S antigen- binding protein of the present invention, e.g., those of Table 1, comprising introducing the antigen-binding protein into the body of a subject (e.g., a human). For example, the method comprises piercing the body of the subject with a needle of a syringe and injecting the antigen- binding protein into the body of the subject, e.g., into the vein, artery, tumor, muscular tissue or subcutis of the subject. [000109] The present invention provides a vessel (e.g., a plastic or glass vial, e.g., with a cap or a chromatography column, hollow bore needle or a syringe cylinder) comprising an anti-CoV- S antigen-binding protein of the present invention, e.g., those of Table 1. [000110] The present invention also provides an injection device comprising one or more antigen-binding proteins (e.g., antibody or antigen-binding fragment) that bind specifically to CoV-S, e.g., those of Table 1, or a pharmaceutical composition thereof. The injection device may be packaged into a kit. An injection device is a device that introduces a substance into the body of a subject via a parenteral route, e.g., intramuscular, subcutaneous or intravenous. For example, an injection device may be a syringe (e.g., pre-filled with the pharmaceutical composition, such as an auto-injector) which, for example, includes a cylinder or barrel for holding fluid to be injected (e.g., comprising the antibody or fragment or a pharmaceutical composition thereof), a needle for piecing skin and/or blood vessels for injection of the fluid; and a plunger for pushing the fluid out of the cylinder and through the needle bore. In an embodiment of the invention, an injection device that comprises an antigen-binding protein, e.g., an antibody or antigen-binding fragment thereof, from a combination of the present invention, or  
a pharmaceutical composition thereof is an intravenous (IV) injection device. Such a device can include the antigen-binding protein or a pharmaceutical composition thereof in a cannula or trocar/needle which may be attached to a tube which may be attached to a bag or reservoir for holding fluid (e.g., saline) introduced into the body of the subject through the cannula or trocar/needle. The antibody or fragment or a pharmaceutical composition thereof may, in an embodiment of the invention, be introduced into the device once the trocar and cannula are inserted into the vein of a subject and the trocar is removed from the inserted cannula. The IV device may, for example, be inserted into a peripheral vein (e.g., in the hand or arm); the superior vena cava or inferior vena cava, or within the right atrium of the heart (e.g., a central IV); or into a subclavian, internal jugular, or a femoral vein and, for example, advanced toward the heart until it reaches the superior vena cava or right atrium (e.g., a central venous line). In an embodiment of the invention, an injection device is an autoinjector; a jet injector or an external infusion pump. A jet injector uses a high-pressure narrow jet of liquid which penetrate the epidermis to introduce the antibody or fragment or a pharmaceutical composition thereof to a subject’s body. External infusion pumps are medical devices that deliver the antibody or fragment or a pharmaceutical composition thereof into a subject’s body in controlled amounts. External infusion pumps may be powered electrically or mechanically. Different pumps operate in different ways, for example, a syringe pump holds fluid in the reservoir of a syringe, and a moveable piston controls fluid delivery, an elastomeric pump holds fluid in a stretchable balloon reservoir, and pressure from the elastic walls of the balloon drives fluid delivery. In a peristaltic pump, a set of rollers pinches down on a length of flexible tubing, pushing fluid forward. In a multi-channel pump, fluids can be delivered from multiple reservoirs at multiple rates. Preparation of Human Antibodies [000111] Methods for generating human antibodies in transgenic mice are known in the art. Any such known methods can be used in the context of the present invention to make human antibodies that specifically bind to CoV-S. An immunogen comprising any one of the following can be used to generate antibodies to CoV-S. In certain embodiments of the invention, the antibodies of the invention are obtained from mice immunized with a full length, native CoV-S, or with a live attenuated or inactivated virus, or with DNA encoding the protein or fragment thereof. Alternatively, the CoV-S protein or a fragment thereof may be produced using standard  
biochemical techniques and modified and used as immunogen. In one embodiment of the invention, the immunogen is a recombinantly produced CoV-S protein or fragment thereof. In certain embodiments of the invention, the immunogen may be a CoV-S polypeptide vaccine. In certain embodiments, one or more booster injections may be administered. In certain embodiments, the immunogen may be a recombinant CoV-S polypeptide expressed in E. coli or in any other eukaryotic or mammalian cells such as Chinese hamster ovary (CHO) cells. [000112] Using VELOCIMMUNE® technology (see, for example, US 6,596,541, Regeneron Pharmaceuticals, VELOCIMMUNE®) or any other known method for generating monoclonal antibodies, high affinity chimeric antibodies to CoV-S can be initially isolated having a human variable region and a mouse constant region. The VELOCIMMUNE® technology involves generation of a transgenic mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antibody comprising a human variable region and a mouse constant region in response to antigenic stimulation. The DNA encoding the variable regions of the heavy and light chains of the antibody are isolated and operably linked to DNA encoding the human heavy and light chain constant regions. The DNA is then expressed in a cell capable of expressing the fully human antibody. [000113] Generally, a VELOCIMMUNE® mouse is challenged with the antigen of interest, and lymphatic cells (such as B-cells) are recovered from the mice that express antibodies. The lymphatic cells may be fused with a myeloma cell line to prepare immortal hybridoma cell lines, and such hybridoma cell lines are screened and selected to identify hybridoma cell lines that produce antibodies specific to the antigen of interest. DNA encoding the variable regions of the heavy chain and light chain may be isolated and linked to desirable isotypic constant regions of the heavy chain and light chain. Such an antibody protein may be produced in a cell, such as a CHO cell. Alternatively, DNA encoding the antigen-specific chimeric antibodies or the variable domains of the light and heavy chains may be isolated directly from antigen-specific lymphocytes. [000114] Initially, high affinity chimeric antibodies are isolated having a human variable region and a mouse constant region. As in the experimental section below, the antibodies are characterized and selected for desirable characteristics, including affinity, selectivity, epitope, etc. The mouse constant regions are replaced with a desired human constant region to generate  
the fully human antibody of the invention, for example wild-type or modified IgG1 or IgG4. While the constant region selected may vary according to specific use, high affinity antigen- binding and target specificity characteristics reside in the variable region. Anti-Coronavirus Spike Protein Antibodies Comprising Fc Variants [000115] According to certain embodiments of the present invention, anti-CoV-S antigen- binding proteins, e.g., antibodies or antigen-binding fragments, are provided comprising an Fc domain comprising one or more mutations, which, for example, enhance or diminish antibody binding to the FcRn receptor, e.g., at acidic pH as compared to neutral pH. For example, the present invention includes anti-CoV-S antibodies comprising a mutation in the CH2 or a CH3 region of the Fc domain, wherein the mutation(s) increases the affinity of the Fc domain to FcRn in an acidic environment (e.g., in an endosome where pH ranges from about 5.5 to about 6.0). Such mutations may result in an increase in serum half-life of the antibody when administered to an animal. Non-limiting examples of such Fc modifications include, e.g., a modification at position 250 (e.g., E or Q); 250 and 428 (e.g., L or F); 252 (e.g., L/Y/F/W or T), 254 (e.g., S or T), and 256 (e.g., S/R/Q/E/D or T); or a modification at position 428 and/or 433 (e.g., H/L/R/S/P/Q or K) and/or 434 (e.g., A, W, H, F or Y [N434A, N434W, N434H, N434F or N434Y]); or a modification at position 250 and/or 428; or a modification at position 307 or 308 (e.g., 308F, V308F), and 434. In one embodiment, the modification comprises a 428L (e.g., M428L) and 434S (e.g., N434S) modification; a 428L, 259I (e.g., V259I), and 308F (e.g., V308F) modification; a 433K (e.g., H433K) and a 434 (e.g., 434Y) modification; a 252, 254, and 256 (e.g., 252Y, 254T, and 256E) modification; a 250Q and 428L modification (e.g., T250Q and M428L); and a 307 and/or 308 modification (e.g., 308F or 308P). In yet another embodiment, the modification comprises a 265A (e.g., D265A) and/or a 297A (e.g., N297A) modification. [000116] For example, the present invention includes anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments, comprising an Fc domain comprising one or more pairs or groups of mutations selected from the group consisting of: 250Q and 248L (e.g., T250Q and M248L); 252Y, 254T and 256E (e.g., M252Y, S254T and T256E); 428L and 434S (e.g., M428L and N434S); 257I and 311I (e.g., P257I and Q311I); 257I and 434H (e.g., P257I and N434H); 376V and 434H (e.g., D376V and N434H); 307A, 380A and 434A (e.g., T307A, E380A and N434A); and 433K and 434F (e.g., H433K and N434F).  
[000117] Anti-CoV-S antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, that comprise a VH and/or VL as set forth herein comprising any possible combinations of the foregoing Fc domain mutations, are contemplated within the scope of the present invention. [000118] The present invention also includes anti-CoV-S antigen-binding proteins, antibodies or antigen-binding fragments, comprising a VH set forth herein and a chimeric heavy chain constant (CH) region, wherein the chimeric CH region comprises segments derived from the CH regions of more than one immunoglobulin isotype. For example, the antibodies of the invention may comprise a chimeric CH region comprising part or all of a CH2 domain derived from a human IgG1, human IgG2 or human IgG4 molecule, combined with part or all of a CH3 domain derived from a human IgG1, human IgG2 or human IgG4 molecule. According to certain embodiments, the antibodies of the invention comprise a chimeric CH region having a chimeric hinge region. For example, a chimeric hinge may comprise an “upper hinge” amino acid sequence (amino acid residues from positions 216 to 227 according to EU numbering) derived from a human IgG1, a human IgG2 or a human IgG4 hinge region, combined with a “lower hinge” sequence (amino acid residues from positions 228 to 236 according to EU numbering) derived from a human IgG1, a human IgG2 or a human IgG4 hinge region. According to certain embodiments, the chimeric hinge region comprises amino acid residues derived from a human IgG1 or a human IgG4 upper hinge and amino acid residues derived from a human IgG2 lower hinge. An antibody comprising a chimeric CH region as described herein may, in certain embodiments, exhibit modified Fc effector functions without adversely affecting the therapeutic or pharmacokinetic properties of the antibody. (See, e.g., WO2014/022540). Immunoconjugates [000119] The invention encompasses an anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments, conjugated to another moiety, e.g., a therapeutic moiety (an “immunoconjugate”), such as a toxoid or an anti-viral drug to treat influenza virus infection. In an embodiment of the invention, an anti-CoV-S antibody or fragment is conjugated to any of the further therapeutic agents set forth herein. As used herein, the term “immunoconjugate” refers to an antigen-binding protein, e.g., an antibody or antigen-binding fragment, which is chemically or biologically linked to a radioactive agent, a cytokine, an interferon, a target or reporter moiety,  
an enzyme, a peptide or protein or a therapeutic agent. The antigen-binding protein may be linked to the radioactive agent, cytokine, interferon, target or reporter moiety, enzyme, peptide or therapeutic agent at any location along the molecule so long as it is able to bind its target (CoV- S). Examples of immunoconjugates include antibody-drug conjugates and antibody-toxin fusion proteins. In one embodiment of the invention, the agent may be a second, different antibody that binds specifically to CoV-S. The type of therapeutic moiety that may be conjugated to the anti- CoV-S antigen-binding protein (e.g., antibody or fragment) will take into account the condition to be treated and the desired therapeutic effect to be achieved. See, e.g., Arnon et al., “Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy”, Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp.243-56 (Alan R. Liss, Inc.1985); Hellstrom et al., “Antibodies For Drug Delivery”, Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp.623-53 (Marcel Dekker, Inc.1987); Thorpe, “Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review”, Monoclonal Antibodies 1984: Biological And Clinical Applications, Pinchera et al. (eds.), pp.475-506 (1985); “Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy”, Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp.303-16 (Academic Press 1985), and Thorpe et al., “The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates”, Immunol. Rev., 62: 119-58 (1982). Multi-specific Antibodies [000120] The present invention includes anti-CoV-S antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof, as well as methods of use thereof and methods of making such antigen-binding proteins. The term “anti-CoV-S” antigen-binding proteins, e.g., antibodies or antigen-binding fragments, includes multispecific (e.g., bispecific or biparatopic) molecules that include at least one first antigen-binding domain that specifically binds to CoV-S (e.g., an antigen-binding domain from an antibody of Table 1) and at least one second antigen-binding domain that binds to a different antigen or to an epitope in CoV-S which is different from that of the first antigen-binding domain. In some embodiments, the first antigen-binding domain and the second antigen-binding domain are both selected from the antigen-binding domains of Table 1. In an embodiment of the invention, the first and second epitopes overlap. In another embodiment of the invention, the first and second epitopes do not overlap. For example, in an  
embodiment of the invention, a multispecific antibody is a bispecific IgG antibody (e.g., IgG1 or IgG4) that includes a first antigen-binding domain that binds specifically to CoV-S including the heavy and light immunoglobulin chain of an antibody of Table 1, and a second antigen-binding domain that binds specifically to a different epitope of CoV-S. In some embodiments, a bispecific IgG antibody (e.g., IgG1 or IgG4) includes a first antigen-binding domain that binds specifically to CoV-S and a second binding domain that binds to a host cell protein, e.g., ACE2 or TMPRSS2. [000121] The antibodies of Table 1 include multispecific molecules, e.g., antibodies or antigen-binding fragments, that include the CDR-Hs and CDR-Ls, VH and VL, or HC and LC of those antibodies, respectively (including variants thereof as set forth herein). [000122] In an embodiment of the invention, an antigen-binding domain that binds specifically to CoV-S, which may be included in a multispecific molecule, comprises: (1) (i) a heavy chain variable domain sequence that comprises CDR-H1, CDR-H2, and CDR-H3 amino acid sequences set forth in Table 1, and (ii) a light chain variable domain sequence that comprises CDR-L1, CDR-L2, and CDR- L3 amino acid sequences set forth in Table 1; or, (2) (i) a heavy chain variable domain sequence comprising an amino acid sequence set forth in Table 1, and (ii) a light chain variable domain sequence comprising an amino acid sequence set forth in Table 1; or, (3) (i) a heavy chain immunoglobulin sequence comprising an amino acid sequence set forth in Table 1, and (ii) a light chain immunoglobulin sequence comprising an amino acid sequence set forth in Table 1. [000123] In an embodiment of the invention, the multispecific antibody or fragment includes more than two different binding specificities (e.g., a trispecific molecule), for example, one or  
more additional antigen-binding domains which are the same or different from the first and/or second antigen-binding domain. [000124] In one embodiment of the invention, a bispecific antigen-binding fragment comprises a first scFv (e.g., comprising VH and VL sequences of Table 1) having binding specificity for a first epitope (e.g., CoV-S) and a second scFv having binding specificity for a second, different epitope. For example, in an embodiment of the invention, the first and second scFv are tethered with a linker, e.g., a peptide linker (e.g., a GS linker such as (GGGGS)n (SEQ ID NO: 834) wherein n is, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10). Other bispecific antigen-binding fragments include an F(ab)2 of a bispecific IgG antibody which comprises the heavy and light chain CDRs of Table 1 and of another antibody that binds to a different epitope. Therapeutic Methods [000125] The present invention provides methods for treating or preventing viral infection (e.g., coronavirus infection) by administering a therapeutically effective amount of anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment, (e.g., of Table 1) to a subject (e.g., a human) in need of such treatment or prevention. [000126] Coronavirus infection may be treated or prevented, in a subject, by administering an anti-CoV-S antigen-binding protein of the present invention to a subject. [000127] An effective or therapeutically effective dose of anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment (e.g., of Table 1), for treating or preventing a viral infection refers to the amount of the antibody or fragment sufficient to alleviate one or more signs and/or symptoms of the infection in the treated subject, whether by inducing the regression or elimination of such signs and/or symptoms or by inhibiting the progression of such signs and/or symptoms. The dose amount may vary depending upon the age and the size of a subject to be administered, target disease, conditions, route of administration, and the like. In an embodiment of the invention, an effective or therapeutically effective dose of antibody or antigen-binding fragment thereof of the present invention, for treating or preventing viral infection, e.g., in an adult human subject, is about 0.01 to about 200 mg/kg, e.g., up to about 150 mg/kg. In an embodiment of the invention, the dosage is up to about 10.8 or 11 grams (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11 grams). Depending on the severity of the infection, the frequency and the duration of the treatment can be adjusted. In certain embodiments, the  
antigen-binding protein of the present invention can be administered at an initial dose, followed by one or more secondary doses. In certain embodiments, the initial dose may be followed by administration of a second or a plurality of subsequent doses of antibody or antigen-binding fragment thereof in an amount that can be approximately the same or less than that of the initial dose, wherein the subsequent doses are separated by at least 1 day to 3 days; at least one week, at least 2 weeks; at least 3 weeks; at least 4 weeks; at least 5 weeks; at least 6 weeks; at least 7 weeks; at least 8 weeks; at least 9 weeks; at least 10 weeks; at least 12 weeks; or at least 14 weeks. [000128] As used herein, the term “subject” refers to a mammal (e.g., rat, mouse, cat, dog, cow, pig, sheep, horse, goat, rabbit), preferably a human, for example, in need of prevention and/or treatment of a disease or disorder such as viral infection or cancer. The subject may have a viral infection, e.g., an influenza infection, or be predisposed to developing an infection. Subjects predisposed to developing an infection, or subjects who may be at elevated risk for contracting an infection (e.g., of coronavirus or influenza virus), include subjects with compromised immune systems because of autoimmune disease, subjects receiving immunosuppressive therapy (for example, following organ transplant), subjects afflicted with human immunodeficiency syndrome (HIV) or acquired immune deficiency syndrome (AIDS), subjects with forms of anemia that deplete or destroy white blood cells, subjects receiving radiation or chemotherapy, or subjects afflicted with an inflammatory disorder. Additionally, subjects of very young (e.g., 5 years of age or younger) or old age (e.g., 65 years of age or older) are at increased risk. Moreover, a subject may be at risk of contracting a viral infection due to proximity to an outbreak of the disease, e.g. subject resides in a densely-populated city or in close proximity to subjects having confirmed or suspected infections of a virus, or choice of employment, e.g. hospital worker, pharmaceutical researcher, traveler to infected area, or frequent flier. [000129] “Treat” or “treating” means to administer an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment of the present invention (e.g., of Table 1), to a subject having one or more signs or symptoms of a disease or infection, e.g., viral infection, for which the antigen-binding protein is effective when administered to the subject at an effective or therapeutically effective amount or dose (as discussed herein).  
[000130] The present invention also encompasses prophylactically administering an anti-CoV- S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof of the present invention (e.g., of Table 1), to a subject who is at risk of viral infection so as to prevent such infection. Passive antibody-based immunoprophylaxis has proven an effective strategy for preventing subject from viral infection. See e.g., Berry et al., Passive broad-spectrum influenza immunoprophylaxis. Influenza Res Treat.2014; 2014:267594. Epub 2014 Sep 22; and Jianqiang et al., Passive immune neutralization strategies for prevention and control of influenza A infections, Immunotherapy.2012 February; 4(2): 175–186; Prabhu et al., Antivir Ther. 2009;14(7):911-21, Prophylactic and therapeutic efficacy of a chimeric monoclonal antibody specific for H5 hemagglutinin against lethal H5N1 influenza. “Prevent” or “preventing” means to administer an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment of the present invention (e.g., of Table 1), to a subject to inhibit the manifestation of a disease or infection (e.g., viral infection) in the body of a subject, for which the antigen-binding protein is effective when administered to the subject at an effective or therapeutically effective amount or dose (as discussed herein). [000131] In an embodiment of the invention, a sign or symptom of a viral infection in a subject is survival or proliferation of virus in the body of the subject, e.g., as determined by viral titer assay (e.g., coronavirus propagation in embryonated chicken eggs or coronavirus spike protein assay). Other signs and symptoms of viral infection are discussed herein. [000132] As noted above, in some embodiments the subject may be a non-human animal, and the antigen-binding proteins (e.g., antibodies and antigen-binding fragments) discussed herein may be used in a veterinary context to treat and/or prevent disease in the non-human animals (e.g., cats, dogs, pigs, cows, horses, goats, rabbits, sheep, and the like). [000133] The present invention provides a method for treating or preventing viral infection (e.g., coronavirus infection) or for inducing the regression or elimination or inhibiting the progression of at least one sign or symptom of viral infection such as: ^ fever or feeling feverish/chills; ^ cough; ^ sore throat; ^ runny or stuffy nose; ^ sneezing;  
^ muscle or body aches; ^ headaches; ^ fatigue (tiredness); ^ vomiting; ^ diarrhea; ^ respiratory tract infection; ^ chest discomfort; ^ shortness of breath; ^ bronchitis; and/or ^ pneumonia, which sign or symptom is secondary to viral infection, in a subject in need thereof (e.g., a human), by administering a therapeutically effective amount of anti-CoV-S antigen-binding protein (e.g., of Table 1) to the subject, for example, by injection of the protein into the body of the subject. Combinations and Pharmaceutical Compositions [000134] To prepare pharmaceutical compositions of the anti-CoV-S antigen-binding proteins, e.g., antibodies and antigen-binding fragments thereof (e.g., of Table 1), antigen-binding protein is admixed with a pharmaceutically acceptable carrier or excipient. See, e.g., Remington’s Pharmaceutical Sciences and U.S. Pharmacopeia: National Formulary, Mack Publishing Company, Easton, Pa. (1984); Hardman, et al. (2001) Goodman and Gilman’s The Pharmacological Basis of Therapeutics, McGraw-Hill, New York, N.Y.; Gennaro (2000) Remington: The Science and Practice of Pharmacy, Lippincott, Williams, and Wilkins, New York, N.Y.; Avis, et al. (eds.) (1993) Pharmaceutical Dosage Forms: Parenteral Medications, Marcel Dekker, NY; Lieberman, et al. (eds.) (1990) Pharmaceutical Dosage Forms: Tablets, Marcel Dekker, NY; Lieberman, et al. (eds.) (1990) Pharmaceutical Dosage Forms: Disperse Systems, Marcel Dekker, NY; Weiner and Kotkoskie (2000) Excipient Toxicity and Safety, Marcel Dekker, Inc., New York, N.Y. In an embodiment of the invention, the pharmaceutical composition is sterile. Such compositions are part of the present invention. [000135] The scope of the present invention includes desiccated, e.g., freeze-dried, compositions comprising an anti-CoV-S antigen-binding proteins, e.g., antibody or antigen-  
binding fragment thereof (e.g., of Table 1), or a pharmaceutical composition thereof that includes a pharmaceutically acceptable carrier but substantially lacks water. [000136] In a further embodiment of the invention, a further therapeutic agent that is administered to a subject in association with an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof (e.g., of Table 1), disclosed herein is administered to the subject in accordance with the Physicians’ Desk Reference 2003 (Thomson Healthcare; 57th edition (Nov.1, 2002)). [000137] The mode of administration can vary. Routes of administration include oral, rectal, transmucosal, intestinal, parenteral; intramuscular, subcutaneous, intradermal, intramedullary, intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, intraocular, inhalation, insufflation, topical, cutaneous, transdermal or intra-arterial. [000138] The present invention provides methods for administering an anti-CoV-S antigen- binding protein, e.g., antibody or antigen-binding fragment thereof (e.g., of Table 1), comprising introducing the protein into the body of a subject. For example, the method comprises piercing the body of the subject with a needle of a syringe and injecting the antigen-binding protein into the body of the subject, e.g., into the vein, artery, tumor, muscular tissue or subcutis of the subject. [000139] The present invention provides a vessel (e.g., a plastic or glass vial, e.g., with a cap or a chromatography column, hollow bore needle or a syringe cylinder) comprising any of the anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments thereof (e.g., of Table 1), polypeptides (e.g., an HC, LC, VH or VL of Table 1) or polynucleotides (e.g., of Table 2) or vectors set forth herein or a pharmaceutical composition thereof comprising a pharmaceutically acceptable carrier. [000140] In an embodiment of the present disclosure, an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof of the present invention (e.g., of Table 1), is administered in association with one or more further therapeutic agents. A further therapeutic agent includes, but is not limited to: an anti-inflammatory agent, an antimalarial agent, a second antibody or antigen-binding fragment thereof that specifically binds TMPRSS2, and a second antibody or antigen-binding fragment thereof that specifically binds to CoV-S. In some embodiments, an antimalarial agent is chloroquine or hydroxychloroquine. In some embodiments, an anti-inflammatory agent is an antibody such as sarilumab, tocilizumab, or   gimsilumab. In some embodiments, the further therapeutic agent is a second antibody or antigen-binding fragment disclosed herein, e.g., of Table 1. In certain embodiments, one, two, three, four, or more antibodies, or antigen-binding fragments thereof, of Table 1 can be administered in combination (e.g., concurrently or sequentially). Particular combinations of antibodies of Table 1 are listed in the Table of Exemplary' Antibody Combinations, below (each number representing a specific combination, e.g., mAbl0989 and mAbl0987 is Combination 1, m Ab10989 and mAb10934 is Combination 2, and so on). In some embodiments, a combination of antibodies can be selected from among those binding to different epitope clusters. For example, certain antibodies described herein belong to epitope clusters as follows: Cluster 1, mAb 10987, mAbl 0922, mAbl0936, and mAbl0934; Cluster 2, mAbl0989, mAb 10977, and mAb 10933; Cluster 3, mAbl0920; Cluster 4, mAbl0954, mAbl0986, and mAbl0964; and Cluster 5, mAbl0984. Thus, a combination of two antibodies can be selected from, for example, Cluster I and Cluster 2, Cluster 1 and Cluster 3, Cluster 1 and Cluster 4, Cluster 1 and Cluster 5, Cluster 2 and Cluster 3, Cluster 2 and Cluster 4. Cluster 2 and Cluster 5, Cluster 3 and Cluster 4, Cluster 3 and Cluster 5, and Cluster 4 and Cluster 5. In some embodiments, an antibody that specifically binds TMPRSS2 is H1H7017N, as described in International Patent Pub. No. WO/2019/147831.
55  
Figure imgf000057_0001
[000141] In some embodiments, anti-CoV-S antigen-binding proteins (e.g., anti-SARS-CoV- 2-S antibodies or antigen-binding fragments thereof) from different human donors may be combined. The present invention includes a composition comprising two (or more) anti-SARS- CoV-2-S antibodies or antigen-binding fragments comprising variable domains from human subjects, wherein the two (or more) antibodies or antigen-binding fragments are derived from different subjects (e.g., two different human subjects). Antibody variable regions derived from human B cells are discussed, e.g., in Examples 1 and 2 (Table 3), which describes that variable domains cloned from such B cells are combined with a constant region not from those B cells to produce hybrid antibodies. The source (Donor) of such antibody variable regions is shown in the Table of Exemplary Human-Derived Antibody Variable Regions, below. In some embodiments, a composition may comprise a combination of an antibody or antigen-binding fragment thereof with variable domains derived from donor 1 and an antibody or antigen-binding fragment thereof with variable domains derived from donor 2. In some embodiments, a composition may comprise a combination of an antibody or antigen-binding fragment thereof with variable domains derived from donor 1 and an antibody or antigen-binding fragment thereof with variable domains derived from donor 3. In some embodiments, a composition may comprise a combination of an antibody or antigen-binding fragment thereof with variable domains derived from donor 2 and an antibody or antigen-binding fragment thereof with variable domains derived from donor 3. In some embodiments, a composition may comprise a combination of mAb10987 (e.g., an antibody comprising the CDRs, the variable regions, or the heavy and light chain sequences shown in Table 1) from Donor 1, and mAb10989 (e.g., an antibody comprising the CDRs, the variable regions, or the heavy and light chain sequences shown in Table 1) from Donor 3. Table of Exemplary Human-Derived Antibody Variable Regions
Figure imgf000058_0001
 
Figure imgf000059_0002
[000142] In some embodiments, the further therapeutic agent is an anti-viral drug and/or a vaccine. As used herein, the term “anti-viral drug” refers to any anti-infective drug or therapy used to treat, prevent, or ameliorate a viral infection in a subject. The term “anti-viral drug” includes, but is not limited to a cationic steroid antimicrobial, leupeptin, aprotinin, ribavirin, or interferon-alpha2b. Methods for treating or preventing virus (e.g., coronavirus) infection in a subject in need of said treatment or prevention by administering an antibody or antigen-binding fragment of Table 1 in association with a further therapeutic agent are part of the present invention. [000143] For example, in an embodiment of the invention, the further therapeutic agent is a vaccine, e.g., a coronavirus vaccine. In an embodiment of the invention, a vaccine is an inactivated/killed virus vaccine, a live attenuated virus vaccine or a virus subunit vaccine. [000144] For example, in an embodiment of the invention, the further therapeutic agent is:
Figure imgf000059_0001
(camostat mesylate);  
Figure imgf000060_0001
(nafamostat mesylate);
Figure imgf000060_0002
(bromhexine hydrochloride (BHH));
Figure imgf000060_0003
(4-(2-aminomethyl)benzenesulfonyl fluoride hydrochloride (AEBSF));
Figure imgf000060_0004
 
Figure imgf000061_0001
(polyamide). See Shen et al. Biochimie 142: 1-10 (2017). [000145] In an embodiment of the invention, the anti-viral drug is an antibody or antigen- binding fragment that binds specifically to coronavirus, e.g., SARS-CoV-2, SARS-CoV, or MERS-CoV. Exemplary anti-CoV-S antibodies include, but are not limited to: H4sH15188P; H1H15188P; H1H15211P; H1H15177P; H4sH15211P; H1H15260P2; H1H15259P2; H1H15203P; H4sH15260P2; H4sH15231P2; H1H15237P2; H1H15208P; H1H15228P2; H1H15233P2; H1H15264P2; H1H15231P2; H1H15253P2; H1H15215P; and H1H15249P2, as set forth in International patent application publication no. WO/2015/179535, or an antigen- binding fragment thereof, e.g., wherein the antibody or fragment comprises a light chain immunoglobulin that includes CDR-L1, CDR-L2 and CDR-L3 (e.g., the VL or light chain thereof); and a heavy chain that includes CDR-H1, CDR-H2 and CDR-H3 (e.g., the VH or heavy chain thereof) of any of the foregoing anti-CoV-S antibodies. [000146] In a certain embodiment of the invention, the further therapeutic agent is not aprotinin, leupeptin, a cationic steroid antimicrobial, an influenza vaccine (e.g., killed, live,  
attenuated whole virus or subunit vaccine), or an antibody against influenza virus (e.g., an anti- hemagglutinin antibody). [000147] The term "in association with" indicates that the components, an anti-CoV-S antigen- binding protein, e.g., antibody or antigen-binding fragment thereof of the present invention, along with another agent, can be formulated into a single composition, e.g., for simultaneous delivery, or formulated separately into two or more compositions (e.g., a kit). Each component can be administered to a subject at a different time than when the other component is administered; for example, each administration may be given non-simultaneously (e.g., separately or sequentially) at intervals over a given period of time. Moreover, the separate components may be administered to a subject by the same or by a different route (e.g., wherein an anti-CoV-S antibody or antigen-binding fragment thereof. Kits [000148] Further provided are kits comprising one or more components that include, but are not limited to, an anti-CoV-S antigen-binding protein, e.g., an antibody or antigen-binding fragment as discussed herein (e.g., of Table 1), in association with one or more additional components including, but not limited to, a further therapeutic agent, as discussed herein. The antigen-binding protein and/or the further therapeutic agent can be formulated as a single composition or separately in two or more compositions, e.g., with a pharmaceutically acceptable carrier, in a pharmaceutical composition. [000149] In one embodiment of the invention, the kit includes an anti-CoV-S antigen-binding protein, e.g., an antibody or antigen-binding fragment thereof of the invention (e.g., of Table 1), or a pharmaceutical composition thereof in one container (e.g., in a sterile glass or plastic vial) and a further therapeutic agent in another container (e.g., in a sterile glass or plastic vial). [000150] In another embodiment, the kit comprises a combination of the invention, including an anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof of the invention (e.g., of Table 1), or pharmaceutical composition thereof in combination with one or more further therapeutic agents formulated together, optionally, in a pharmaceutical composition, in a single, common container. [000151] If the kit includes a pharmaceutical composition for parenteral administration to a subject, the kit can include a device (e.g., an injection device) for performing such  
administration. For example, the kit can include one or more hypodermic needles or other injection devices as discussed above containing the anti-CoV-S antigen-binding protein, e.g., antibody or antigen-binding fragment thereof of the present invention (e.g., of Table 1). [000152] The kit can include a package insert including information concerning the pharmaceutical compositions and dosage forms in the kit. Generally, such information aids patients and physicians in using the enclosed pharmaceutical compositions and dosage forms effectively and safely. For example, the following information regarding a combination of the invention may be supplied in the insert: pharmacokinetics, pharmacodynamics, clinical studies, efficacy parameters, indications and usage, contraindications, warnings, precautions, adverse reactions, overdosage, proper dosage and administration, how supplied, proper storage conditions, references, manufacturer/distributor information and patent information. Diagnostic Uses of the Antibodies [000153] The anti-CoV-S antigen-binding proteins, e.g., antibodies or antigen-binding fragments thereof of the present invention (e.g., of Table 1), may be used to detect and/or measure CoV-S in a sample. Exemplary assays for CoV-S may include, e.g., contacting a sample with an anti-CoV-S antigen-binding protein of the invention, wherein the anti-CoV-S antigen-binding protein is labeled with a detectable label or reporter molecule or used as a capture ligand to selectively isolate CoV-S from samples. The presence of an anti-CoV-S antigen-binding protein complexed with CoV-S indicates the presence of CoV-S in the sample. Alternatively, an unlabeled anti-CoV-S antibody can be used in combination with a secondary antibody which is itself detectably labeled. The detectable label or reporter molecule can be a radioisotope, such as 3H, 14C, 32P, 35S, or 125I; a fluorescent or chemiluminescent moiety such as fluorescein isothiocyanate, or rhodamine; or an enzyme such as alkaline phosphatase, b- galactosidase, horseradish peroxidase, or luciferase. Specific exemplary assays that can be used to detect or measure CoV-S in a sample include neutralization assays, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and fluorescence-activated cell sorting (FACS). Thus, the present invention includes a method for detecting the presence of spike protein polypeptide in a sample comprising contacting the sample with an anti-CoV-S antigen-binding protein and detecting the presence of a CoV-S/anti-CoV-S antigen-binding protein wherein the presence of the complex indicates the presence of CoV-S.  
[000154] An anti-CoV-S antigen-binding protein of the invention (e.g., of Table 1) may be used in a Western blot or immune-protein blot procedure for detecting the presence of CoV-S or a fragment thereof in a sample. Such a procedure forms part of the present invention and includes the steps of e.g.: (1) providing a membrane or other solid substrate comprising a sample to be tested for the presence of CoV-S, e.g., optionally including the step of transferring proteins from a sample to be tested for the presence of CoV-S (e.g., from a PAGE or SDS-PAGE electrophoretic separation of the proteins in the sample) onto a membrane or other solid substrate using a method known in the art (e.g., semi-dry blotting or tank blotting); and contacting the membrane or other solid substrate to be tested for the presence of CoV-S or a fragment thereof with an anti-CoV-S antigen-binding protein of the invention. [000155] Such a membrane may take the form, for example, of a nitrocellulose or vinyl-based (e.g., polyvinylidene fluoride (PVDF)) membrane to which the proteins to be tested for the presence of CoV-S in a non-denaturing PAGE (polyacrylamide gel electrophoresis) gel or SDS- PAGE (sodium dodecyl sulfate polyacrylamide gel electrophoresis) gel have been transferred (e.g., following electrophoretic separation in the gel). Before contacting the membrane with the anti-CoV-S antigen-binding protein, the membrane is optionally blocked, e.g., with non-fat dry milk or the like so as to bind non-specific protein binding sites on the membrane. (2) washing the membrane one or more times to remove unbound anti-CoV-S antigen- binding protein and other unbound substances; and (3) detecting the bound anti-CoV-S antigen-binding protein. [000156] Detection of the bound antigen-binding protein indicates that the CoV-S protein is present on the membrane or substrate and in the sample. Detection of the bound antigen-binding protein may be by binding the antigen-binding protein with a secondary antibody (an anti- immunoglobulin antibody) which is detectably labeled and, then, detecting the presence of the secondary antibody label. [000157] The anti-CoV-S antigen-binding proteins (e.g., antibodies and antigen-binding fragments (e.g., of Table 1)) disclosed herein may also be used for immunohistochemistry. Such a method forms part of the present invention and comprises, e.g., (1) contacting tissue to be tested for the presence of CoV-S protein with an anti-CoV-S antigen-binding protein of the invention; and  
(2) detecting the antigen-binding protein on or in the tissue. [000158] If the antigen-binding protein itself is detectably labeled, it can be detected directly. Alternatively, the antigen-binding protein may be bound by a detectably labeled secondary antibody wherein the label is then detected. EXAMPLES [000159] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the methods and compositions of the invention and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is average molecular weight, temperature is in degrees Centigrade, room temperature is about 25oC, and pressure is at or near atmospheric. Example 1: Generation of human antibodies to SARS-CoV-2 spike protein (SARS-CoV-2-S) [000160] Human antibodies to SARS-CoV-2-Spike protein (SARS-CoV-2-S) were generated in a VELOCIMMUNE® mouse comprising DNA encoding human immunoglobulin heavy and kappa light chain variable regions or human immunoglobulin heavy and lambda light chain variable regions. Each mouse was immunized with a vector expressing the SARS-CoV-2-S receptor binding domain (RBD) (amino acids 1-1273 of NCBI accession number (MN908947.3), SEQ ID NO: 832), followed by a booster with a SARS-CoV-2-S vector or a SARS-CoV-2-S protein. The antibody immune response was monitored by a SARS-CoV-2-S-specific immunoassay. When a desired immune response was achieved, lymphocytes were harvested and fused with mouse myeloma cells to preserve their viability and form hybridoma cell lines. The hybridoma cell lines were screened and selected to identify cell lines that produce SARS-CoV-2- S-specific antibodies. Anti-SARS-CoV-2-S antibodies were also isolated directly from antigen- positive mouse B cells without fusion to myeloma cells, as described in U.S. Patent 7582298, herein specifically incorporated by reference in its entirety. Using this method, fully human anti- SARS-CoV-2-S antibodies (i.e., antibodies possessing human variable domains and human constant domains) were obtained. [000161] Antibody variable regions were also isolated from human blood samples. Whole  
blood was received from patients 3-4 weeks after a laboratory-confirmed PCR positive test for SARS-CoV- 2 and symptomatic COVID-19 disease. Red blood cells were lysed using an ammonium chloride based lysis buffer (Life Technologies) and B cells were enriched by negative selection. Single B cells that bound the SARS-CoV-2 spike protein were isolated by fluorescent-activated cell sorting (FACS). Isolated B cells were single-well plated and mixed with antibody light and heavy variable region-specific PCR primers. cDNAs for each single B cell were synthesized via a reverse transcriptase (RT) reaction. Each resulting RT product was then split and transferred into two corresponding wells for subsequent antibody heavy and light chain PCRs. One set of the resulting RT products was first amplified by PCR using a 5’ degenerate primer specific for antibody heavy variable region leader sequence or a 5’ degenerate primer specific for antibody light chain variable region leader sequence and a 3’ primer specific for antibody constant region, to form an amplicon. The amplicons were then amplified again by PCR using a 5’ degenerate primer specific for antibody heavy variable region framework 1 or a 5’ degenerate primer specific for antibody light chain variable region framework 1 and a 3’ primer specific for antibody constant region, to generate amplicons for cloning. The antibody heavy chain and light chain derived PCR products were cloned into expression vectors containing heavy constant region and light constant region, respectively, thereby producing expression vectors for hybrid antibodies. The expression vectors expressing full-length heavy and light chain pairs were transfected into CHO cells to produce antibody proteins for testing. [000162] The biological properties of exemplary antibodies generated in accordance with the methods of this Example are described in detail in the Examples set forth below. Example 2: Heavy and light chain variable region amino acid and nucleotide sequences [000163] Table 1 sets forth the amino acid sequence identifiers of the heavy and light chain variable regions and CDRs, as well as the heavy chain and light chain sequences, of exemplary anti-SARS-CoV-2-S antibodies. The corresponding nucleic acid sequence identifiers are set forth in Table 2. Table 1: Amino Acid Sequence Identifiers
Figure imgf000066_0001
 
Figure imgf000067_0001
 
Figure imgf000068_0001
 
Figure imgf000069_0001
Table 2: Nucleic Acid Sequence Identifiers
Figure imgf000069_0002
 
Figure imgf000070_0001
 
Figure imgf000071_0001
[000164] Antibodies disclosed herein have fully human variable regions but can have mouse constant regions (e.g., a mouse IgG1 Fc or a mouse IgG2 Fc (a or b isotype)) or human constant regions (e.g., a human IgG1 Fc or a human IgG4 Fc). As will be appreciated by a person of  
ordinary skill in the art, an antibody having a particular Fc isotype can be converted to an antibody with a different Fc isotype (e.g., an antibody with a mouse IgG1 Fc can be converted to an antibody with a human IgG4, etc.), but in any event, the variable domains (including the CDRs) – which are indicated by the numerical identifiers shown in Tables 1 and 2 will remain the same, and the binding properties to antigen are expected to be identical or substantially similar regardless of the nature of the constant domain. [000165] The variable regions of antibodies derived from VELOCIMMUNE® mice and from human samples were sequenced by Next Generation Sequencing and the repertoire for heavy and light chain pairs was identified (Figure 10A and Figure 10B). The predominant lineage of VI antibodies utilized VH3-53 paired with VK1-9, VK1-33, or VK1-39 while human-derived antibodies utilized VH3-66 paired with VK1-33 or VH2-70 paired with VK1-39. Further analysis of overlaid sequences showed strong overlap in the repertoire of isolated kappa chains between VI and human-derived antibodies. Although the repertoire of Lambda chains did not overlap well, that may be due to only two lambda mice being included in this trial. The average CDR length for heavy chain was similar between VI and human derived antibodies with an average length of 13 and 14.5 amino acids, respectively. Average kappa CDR length was the same for VI and human derived antibodies at 9 amino acids and was close for lambda chains with an average length of 11.1 and 10.6 amino acids, respectively. Availability of humanized mouse and human-derived antibodies allowed for more diversity of V genes and enabled the later identification of noncompeting antibodies. [000166] As described above, the antibodies were obtained from hybridomas generated from VELOCIMMUNE® mice, by direct isolation from antigen-positive VELOCIMMUNE® mouse B cells, or derived from variable regions cloned from antigen-positive human B cells. A summary of these sources is shown in Table 3. Table 3: Antibody/Variable Region sources
Figure imgf000072_0001
 
Figure imgf000073_0001
 
Figure imgf000074_0001
 
Figure imgf000075_0001
 
Figure imgf000076_0001
Example 3: Characterization of hybridoma supernatants by binding ELISA [000167] An ELISA binding assay was performed to identify antibody supernatants that bound to the SARS-CoV-2-Spike protein receptor binding domain (RBD). A protein composed of the RBD of SARS-CoV-2 (amino acids 319-541) expressed with a 6X histidine tag and two myc epitope tags at the C-terminus (SARS-CoV-2-S-RBD-mmH; see also NCBI Accession Number MN908947.3) was coated at 1 mg/ml on a 96-well plate in PBS buffer overnight at 4°C. Nonspecific binding sites were subsequently blocked using a 0.5% (w/v) solution of BSA in PBS. Antibody supernatants or media alone were diluted 1:40 or 1 :50 in the PSA+0.5% BSA  
blocking buffer and transferred to the washed microtiter plates. After one hour of incubation at room temperature, the wells were washed, and plate-bound supernatant was detected with either goat-anti-human IgG antibody conjugated with horseradish peroxidase (HRP) (Jackson Immunoresearch), or anti-mouse IgG antibody conjugated with horseradish peroxidase (HRP) (Jackson Immunoresearch). The plates were then developed using TMB substrate solution (BD Biosciences) according to manufacturer’s recommendation and absorbance at 450nm was measured on a Victor X5 plate reader. [000168] The ability of anti-SARS-CoV-2-S antibodies to bind the receptor binding domain of SARS-CoV-2-S (SARS-CoV-2-S-RBD) was assessed, as described above, using a binding ELISA with the SARS-CoV-2-S-RBD-mmH protein coated on a microplate. Single point antibody supernatant binding to SARS-COV-2-S-RBD-mmH coated on 96-well microtiter plates was detected with an HRP conjugated anti-hFc or anti-mFc antibody. [000169] The binding results of three trials are summarized in Table 4. The SARS-CoV-2 binding signals (absorbance 450nm) are indicated, with the media only background provided as a negative reference per experiment. A sample marked IC (Inconclusive) had an experimental anomaly to the plate and is therefore reported without a value. As shown in comparison to the media only control, the supernatants tested showed substantial binding to the SARS-CoV-2-S- RBD. Table 4: Supernatant binding to SARS-CoV-2 spike protein receptor binding domain
Figure imgf000077_0001
 
Figure imgf000078_0001
 
Figure imgf000079_0001
Example 4: Antibody binding to SARS-CoV-2-S-expressing virus-like particle [000170] To investigate the ability of a panel of anti-SARS-CoV-2-S monoclonal antibodies to bind the SARS-CoV-2 spike glycoprotein, an in vitro binding assay utilizing SARS-CoV-2 spike protein-expressing viral-like particles (VLPs) in an electrochemiluminescence based detection platform (MSD) was developed. [000171] To transiently express the SARS-CoV-2 spike protein (NCBI Accession number MN908947.3, amino acids 16-1211; SEQ ID NO: 833), Vesicular stomatitis virus (VSV) lacking glycoprotein G (VSV delta G) was pseudotyped with SARS-CoV-2 spike protein (VSV-SARS- CoV-2-S) and generated in HEK293T cells. As a negative binding control, VSV delta G was pseudotyped with VSV G protein (VSV-G). [000172] Experiments were carried out according to following procedure. The two types of VLPs described above were diluted in PBS, seeded into 96-well carbon electrode plates (MULTI-ARRAY high bind plate, MSD), and incubated overnight at 4 °C to allow the VLPs to adhere. Nonspecific binding sites were blocked by 2% BSA (w/v) in PBS for 1 hour at room temperature. Supernatants containing antibodies produced from SARS CoV-2-immunized mice or infected human sera, along with media-only controls which were diluted 1:10 or 1:20 in 1x PBS + 0.5% BSA buffer, were added to the plate-bound particles. The plates were then incubated for 1 hour at room temperature with shaking, after which the plates were washed with 1x PBS to remove the unbound antibodies using an AquaMax2000 plate washer (MDS Analytical Technologies). The plate-bound antibodies were detected with a SULFO-TAGTM- conjugated anti-human IgG antibody (Jackson Immunoresearch) or a SULFO-TAGTM- conjugated anti-mouse IgG antibody (Jackson Immunoresearch) for 1 hour at room temperature. After washes, the plates were developed with the Read Buffer (MSD) according to manufacturer’s recommended procedure and the luminescent signals were recorded with a SECTOR Imager 600 (Meso Scale Development) instrument. Direct binding signals (in RLU) were captured, and a ratio of SARS-CoV-2-S-expressing VLPs to the irrelevant VLP was calculated. [000173] The ability of the anti-SARS-CoV-2-S monoclonal antibodies to bind to SARS-CoV- 2-S-expressing VLPs compared with binding to irrelevant VSV-expressing VLPs was assessed  
using an immunobinding assay, as described above. Single-point binding to the immobilized VLPs on 96-well High Bind plates (MSD) was performed with an antibody supernatant dilution of 1:10 or 1:20, bound for 1 hour, and detected using SULFO-TAGTM-conjugated anti-human IgG or anti-mouse IgG antibody. The binding signals from electrochemiluminescence were recorded on a Sector Imager 600 (MSD). RLU values were determined for the antibody binding to VLPs. Ratios were calculated comparing the SARS-CoV-2-S-expressing VLP binding signals to control VLPs. [000174] The binding results from three experiments are summarized in Table 5. A signal observed from SARS-COV-2-S-expressing VLPs indicates binding, while comparison with negative VLPs provides a relative background. Media alone samples provide baseline signals of secondary antibody binding to samples with no supernatant. The 46 antibodies bound specifically at >4-fold higher than the media-only samples (20-35 RLU) on the SARS-CoV-2-S- expressing VLPs, with a range of binding signals from 85-13,600 RLU. The ratios of SARS- CoV-2-S-expressing VSV: VSV-VLPs (negative control) ranged from 1.1- 22.7, with many having high background on VSV-VLPs. The ratio of mAb11002 of 0.9 is likely due to a low concentration of monoclonal antibody in the supernatant sample. Table 5: SARS-CoV-2-S VLP binding
Figure imgf000080_0001
 
Figure imgf000081_0001
 
Figure imgf000082_0001
 
Figure imgf000083_0001
Example 5: Antibody neutralization of VSV-SARS-CoV-2-S pseudovirus infectivity [000175] To investigate the ability of a panel of anti-SARS-CoV-2-S monoclonal antibodies to neutralize SARS-CoV-2, an in vitro neutralization assay utilizing VSV-SARS-CoV-2-S pseudovirus was developed. [000176] As described above, VSV pseudotype viruses were generated by transiently transfecting 293T cells with a plasmid encoding for SARS-CoV-2 spike protein. Cells were seeded in 15 cm plates at 1.2x107 cells per plate in DMEM complete media one day prior to transfection with 15 µg/plate spike protein DNA using 125 µL Lipofectamine LTX, 30 µL PLUS reagent, and up to 3 mL Opti-Mem. 24 hours post transfection, the cells were washed with 10 mL PBS, then infected with an MOI of 0.1 VSVDG:mNeon virus in 10 mL Opti-Mem. Virus was incubated on cells for 1 hour, with gentle rocking every 10 minutes. Cells were washed 3 times with 10 mL PBS, then overlaid with 20 mL Infection media before incubation at 37 C, 5% CO2 for 24 hours. Supernatant was collected into 250 mL centrifuge tubes on ice, then centrifuged at 3000 rpm for 5 minutes to pellet any cellular debris, aliquoted on ice, then frozen to -80 °C. Infectivity was tested on Vero cells prior to use in neutralization assays. This material will be referred to as VSV-SARS-CoV-2-S. Neutralization assay with VSV-SARS-CoV-2-S [000177] On day 1, Vero cells were seeded at 80% confluency in T225 flasks. To seed cells, media was removed from the cells, the cells were washed with 20mL PBS (Gibco: 20012-043), and 5mL TrypLE was added and incubated for ~5 minutes at 37 °C until the cells dislodged.5 mL of complete DMEM was added to inactivate the trypsin, and pipetted up and down to distribute the cells. To count the resuspended cells, 20,000 Vero cells were plated in 100 µL prewarmed Complete DMEM per well in a 96 Well Black Polystyrene Microplate (Corning: 3904). [000178] On day 2, VSV-SARS-CoV-2-S was thawed on ice and diluted 1:1 with infection media.  
[000179] In a V-bottom 96 well plate, a dilution of each supernatant was generated in 60ul infection media. For media (negative) controls, 60 µl of diluted conditioned media was added to the wells. 60 µL of diluted VSV-SARS-CoV-2-S were added to every well except the media control wells. To those wells, 60 µL of infection media was added. Pseudoviruses were then incubated with supernatant dilutions for 30 minutes at room temperature. Media was removed from the Vero cell plates, 100 µL of supernatant/pseudovirus mixtures were transferred to the cells, and the plate was incubated at 37 °C, 5% CO2 for 24 hours. The final supernatant dilutions of 1:4 and 1:20, and for some samples 1:100, were used to assess neutralization of VSV-SARS- CoV-2-S pseudoviruses. [000180] On day 3, after the 24 hr incubation, supernatant was removed from the cell wells and replaced with 100 µL of PBS. The plates were then read on a SpectraMax i3 with MiniMax imaging cytometer. [000181] The ability of the anti-SARS-CoV-2-S antibodies to neutralize VSV-based SARS- CoV-2-S-expressing pseudotyped virus was assessed using a neutralization fluorescence focus assay. The binding results of three assays are summarized below. The neutralization potency of antibody at each dilution is represented as a percentage compared to mock supernatant control. All antibodies demonstrated neutralization capacity, and particularly for the set of antibodies that were evaluated 1:100, those showing higher neutralization may represent more potent neutralization capacity. Table 6: Neutralization of VLPs
Figure imgf000084_0001
 
Figure imgf000085_0001
 
Figure imgf000086_0001
 
Figure imgf000087_0001
*NT: not tested Example 6: Characterization of antibodies in an antibody-dependent cell-mediated toxicity surrogate assay [000182] The ability of antibodies targeting the spike protein of SARS-CoV-2 to interact with FcgR3a, an Fc-receptor prominently expressed on natural killer (NK) cells that induces antibody dependent cell-mediated cytotoxicity (ADCC), was measured in a surrogate bioassay using reporter cells and target cells bound to antibodies. This assay used Jurkat T cells that were engineered to express the reporter gene luciferase under the control of the transcription factor NFAT (NFAT-Luc) along with the high affinity human FcgR3a 176Val allotype receptor (Jurkat/NFAT-Luc/hFcgR3a 176Val). Target cells were engineered Jurkat T cells expressing human CD20 (used as a positive control with a CD20-targeting human IgG1 antibody) and the full-length SARS-CoV-2 spike protein controlled by a doxycycline-inducible promoter. Reporter cells were incubated with target cells and engagement of FcgR3a via the Fc domain of human IgG1 antibodies bound to target cells led to the activation of the transcription factor NFAT in the reporter cells and drove the expression of luciferase which was then measured via a luminescence readout. [000183] Jurkat T cells were engineered to constitutively express full length human CD20 (amino acids M1-P297 of NCBI accession number NP_690605.1), Tet3G transactivator protein (cloned using a Takara pEF1a-Tet3G Vector, Catalog # 631167), as well as a doxycycline- inducible full-length SARS-CoV-2 spike protein (amino acids M1-T1273 of NCBI accession number YP_009724390.1). Engineered Jurkat/Tet3G/hCD20/SARS-CoV2 spike protein- expressing cells were sorted for high expression of the spike protein and subsequently maintained in RPMI + 10% Tet-free FBS + P/S/G + 500 µg/ml G418 + 1 mg/ml puromycin + 250 µg/ml hygromycin growth medium. [000184] Jurkat T cells were engineered to stably express a Nuclear Factor of Activated T- cells (NFAT) luciferase reporter construct along with the high affinity human FcgR3a 176Val   allotype receptor (amino acids M1-K254 of NCBI accession number P08637 VAR_003960). Engineered reporter cells were maintained in RPMI1640 + 10% FBS + P/S/G + 0.5 mg/ml puromycin + 500 mg/ml G418 growth media. [000185] 36 hours prior to the start of the surrogate ADCC assay, 5 x 105 target cells/ml were induced in RPMI + 10% Tet-free FBS + P/S/G cell culture media containing 1 µg/ml doxycycline (Sigma). A day before the experiment, reporter cells were split to a density of 7.5 x 105 cells/ml in RPMI 1640 + 10% FBS + P/S/G + 0.5 mg/ml puromycin + 500 mg/ml G418 growth media. [000186] Briefly, on the day of the experiment, the target and reporter cells were transferred into assay media (RPMI + 10% Tet-free FBS + P/S/G) and added at a 3:2 ratio (3 x 104/well target cells and 2 x 104/well reporter cells) to 384-well white microtiter plates, followed by the addition of anti-SARS-CoV-2-S antibody supernatant of varying concentrations. A positive control (CD20 antibody with human IgG1) sample and a negative control sample containing no antibody was included on each plate to normalize detected ADCC activities of anti-SARS-CoV- 2-S antibody supernatants. Plates were incubated at 37 °C/5% CO2 for 5 h followed by the addition of an equal volume of ONE-Glo™ (Promega) reagent to lyse cells and detect luciferase activity. The emitted light was captured in Relative Light Units (RLU) on a multi-label plate reader Envision (PerkinElmer), and data was analyzed and normalized using the following equation:
Figure imgf000088_0001
[000187] The ability of anti-SARS-COV-2-S antibodies to activate FcgR3a receptors was evaluated in a surrogate ADCC assay using Jurkat/NFAT-Luc/FcgR3a 176Val) as reporter cells and Jurkat/hCD20/SARS-CoV2 Spike as target cells. Each antibody tested contained an IgG1 domain. [000188] Table 7 summarizes the results, showing the raw luciferase activity and the calculated % of positive control are indicated. A range of % ADCC activity was observed indicating FcgR3a activation by the antibody supernatants. All samples demonstrated some measure of surrogate ADCC activity, and 10 of the antibody supernatants demonstrated surrogate ADCC activity better than observed in positive controls.  
Table 7: ADCC surrogate activity of anti-SARS-CoV-2-S antibody supernatants.
Figure imgf000089_0001
 
Figure imgf000090_0001
 
Figure imgf000091_0001
Example 7: Anti-SARS-CoV-2-S antibody binding specificity assay [000189] A Luminex binding assay was performed to determine the binding of anti-SARS- COV-2-S antibodies to a panel of antigens. For this assay, antigens were amine-coupled or captured by streptavidin to Luminex microspheres as follows: approximately 10 million MagPlex microspheres (Luminex Corp., MagPlex Microspheres, Cat. No. MC10000 and MC12000), were resuspended by vortexing in 500 µL 0.1M NaPO4, pH 6.2 (activation buffer) and then centrifuged to remove the supernatant. Microspheres were protected from light, as they are light sensitive. The microspheres were resuspended in 160 µL of activation buffer and the carboxylate groups (-COOH) were activated by addition of 20 µL of 50 mg/mL of N- hydroxysuccinimide (NHS, Thermo Scientific, Cat. No.24525) followed by addition of 20 µL of 50 mg/mL 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide (EDC, ThermoScientific, Cat. No. 22980) at 25 °C. After 10 minutes, the pH of the reaction was reduced to 5.0 with the addition of 600 µL 50 mM MES, pH 5 (coupling buffer), and the microspheres were vortexed and centrifuged to remove supernatant. The activated microspheres were immediately mixed with 500 µL of 25 µg/mL of the protein antigen or Streptavidin in coupling buffer and incubated for two hours at 25 °C. The coupling reaction was quenched by addition of 50 µL of 1M Tris-HCl, pH 8.0 and the microspheres were vortexed, centrifuged, and washed three times with 800 µL of PBS 0.005% (Tween200.05%), to remove uncoupled proteins and other reaction components. Microspheres were resuspended in 1 mL of PBS 2% BSA 0.05% Na Azide at 10 million microspheres/mL. For Streptavidin capture of antigens, 500 µL of 12.5 µg/mL of biotinylated protein in PBS was added to Streptavidin-coupled microspheres and incubated for one hour at 25 °C. Microspheres were vortexed, centrifuged, and washed three times with 800 µL of PBS, and then blocked using 500 µL 30mM Biotin (Millipore-Sigma, Cat. No. B4501) in 0.15M Tris pH 8.0. Microspheres were incubated for 30 minutes then vortexed, centrifuged, and washed three times with 800 µL of PBS. Microspheres were resuspended in 1 mL of PBS 2% BSA 0.05% Na Azide at 10 million microspheres/mL. [000190] Microspheres for the different proteins and biotinylated proteins were mixed at 2700  
beads/ml, and 75 µL of microspheres were plated per well on a 96 well ProcartaPlex flat bottom plate (ThermoFisher, Cat. No: EPX-44444-000) and mixed with 25 µL of individual anti-SARS- CoV-2 supernatant containing antibody. Samples and microspheres were incubated for two hours at 25oC and then washed twice with 200 µL of DPBS with 0.05% Tween 20. To detect bound antibody levels to individual microspheres, 100 µL of 2.5 µg/mL R-Phycoerythrin conjugated goat F(ab')2 anti-human kappa (Southern Biotech, Cat# 2063-09) in blocking buffer (for antibodies with murine Fc regions) or 100 µL of 1.25 µg/mL R-Phycoerythrin AffiniPure F(ab')₂ Fragment Goat Anti-Mouse IgG, F(ab')₂ Fragment Specific (Jackson Immunoresearch, Cat. No: 115-116-072) in blocking buffer (for antibodies with human Fc regions), was added and incubated for 30 minutes at 25 °C. After 30 minutes, the samples were washed twice with 200 µl of washing buffer and resuspended in 150 µL of wash buffer. The plates were read in a Luminex FlexMap 3D ^ (Luminex Corp.) and Luminex xPonent ^ software version 4.3 (Luminex Corp.). The SARS-CoV-2 proteins used in the assay are as follows: RBD_ (R319-F541).mmh: SEQ ID NO: 829 RBD_ (R319-F541).mFc: SEQ ID NO: 830 RBD_ (R319-F541).hFc): SEQ ID NO: 831 [000191] The results of the Luminex binding are shown in Table 8 and Table 9 as median fluorescence intensity (MFI) signal intensities. The results show that the 46 anti-SARS-CoV-2-S antibody supernatants bound specifically to SARS-CoV-2-S RBD proteins. These results also show that five of these antibodies cross-react with SARS Coronavirus spike RBD proteins with binding signal greater than 1000 MFI.   1 0 O W 3 5 7 0 1 19  
Figure imgf000093_0001
29  
Figure imgf000094_0001
39  
Figure imgf000095_0001
1 0 O W 3 5 7 0 1 49  
Figure imgf000096_0001
1 0 O W 3 5 7 0 1 59  
Figure imgf000097_0001
1 0 O W 3 5 7 0 1 69  
Figure imgf000098_0001
Example 8: Anti-SARS-CoV-2-S antibody diversity assay [000192] A binding assay was performed to determine the binding profile of anti-SARS-COV- 2-S antibodies. For this assay, antigens were amine coupled as described for the Luminex binding assay above. Briefly, approximately 9 million MagPlex microspheres for 16 different bead regions (Luminex Corp., MagPLex Microspheres, Cat. No. MagPLex MC10000 and MC12000), were resuspended by vortexing in 500 µL 0.1M NaPO4, pH 6.2 and then centrifuged to remove the supernatant. The microspheres were resuspended in 160 µL of activation buffer and the carboxylate groups (-COOH) were activated by addition of 20 µL of 50 mg/mL of N- hydroxysuccinimide (NHS, Thermo Scientific, Cat# 24525) followed by addition of 20 µL of 50 mg/mL of 1-ethyl-3-[3-dimethylaminopropyl]carbodiimide (EDC, ThermoScientific, Cat# 22980) at 25°C. After 10 minutes, the pH of the reaction was reduced to 5.0 with the addition of 600 µL of 50 mM MES, pH 5 (coupling buffer), and the microspheres were vortexed and centrifuged to remove supernatant. The activated microspheres were immediately mixed with 500 µL of 20 µg/mL of SARS-CoV-2 Spike Protein (RBD)(R319-F541)-mmH in coupling buffer and incubated for two hours at 25oC. The coupling reaction was quenched by addition of 50 µL of 1M Tris-HCl, pH 8.0 and the microspheres were vortexed, centrifuged, and washed three times with 1000 µL of PBS. Microspheres were resuspended in 250 µL of PBS at 9 million microspheres/mL. [000193] 15 out of the 16 microsphere regions with amine-coupled protein were modified for the binning assay as follows: microspheres were washed twice with PBS 5% DMSO, and 500 µl of a chemical or enzyme were dissolved per manufacturing recommendations and added at 10 nM to the amine-coupled microspheres described above. This was subsequently vortexed and incubated for 2 hours at room temperature with rotation. Wash microspheres 3 times with PBS 2% BSA. Microspheres were resuspended in 1 mL of PBS at 9 million microspheres/mL. [000194] Protein-modified and protein-unmodified (intact) microspheres were mixed at 2700 beads/ml, and 75 µL of microspheres were plated per well on a 96 well ProcartaPlex 96 well flat bottom plate (ThermoFisher, Cat. No: EPX-44444-000) and mixed with 25 µL of individual anti- SARS-CoV-2-S supernatant-containing antibody. Samples and microspheres were incubated for two hours at 25 °C and then washed twice with 200 µL of DPBS with 0.05% Tween 20. To detect bound antibody levels to individual microspheres, 100 µL of 2.5 µg/mL R-Phycoerythrin conjugated goat F(ab')2 anti-human kappa (Southern Biotech, Cat# 2063-09) in blocking buffer  
(for antibodies with hFc), or 100 µL of 1.25 µg/mL R-Phycoerythrin AffiniPure F(ab')₂ Fragment Goat Anti-Mouse IgG, F(ab')₂ Fragment Specific (Jackson Immunoresearch, Cat. No: 115-116- 072) in blocking buffer (for antibodies with mFc), or 100 µL of 1.25 µg/mL R-Phycoerythrin Anti-His (Biolegend, Cat. No: 362603) in blocking buffer (for ACE-2 control, R&D, Cat. No. 933-ZN), was added and incubated for 30 minutes at 25 °C. After 30 minutes, the samples were washed twice with 200 µl of washing buffer and resuspended in 150 µL of wash buffer. The plates were read in FlexMap 3D ^ (Luminex Corp.) and Luminex xPonent ^ software version 4.3 (Luminex Corp.). [000195] The results of the Luminex binning results are shown in Table 10 as median fluorescence intensity (MFI) signal intensities. To determine clusters, data was normalized to the intact protein (unmodified microspheres) and clustered. The 46 anti-SARS-CoV-2 antibodies were classified in 9 clusters with 2 or more antibodies, and 11 antibodies were classified as single nodes. Clusters were assigned by based on these results of the hierarchical clustering and dendrogram. These results show that the 46 anti-SARS-CoV-2-S antibody supernatants had diverse binding characteristics and profiles, suggesting that the collection of antibodies bound to different epitopes on the SARS-CoV-2 spike protein.   1 0 O W 3 5 7 0 1 99  
Figure imgf000101_0001
1 0 O W 3 5 7 0 1 001  
Figure imgf000102_0001
94 7 3 8 9 702 38 3 2 9 2 4 2 42 72 7 2 7 3 6 2 4 1 2 0 O W 3 5 7 3 6 4 9 0 3 1 6 2 7 2 8 4 3 5 2 6 2 4 2 101 5 6 2 0 7 2 5 2 3 2 2 2 2 4 0 6 2 3 2 7 2 1 5 2 3 86 6 6 4 8 5 9 8 0 9 1 0 b 1 A b m A m
Figure imgf000103_0001
  1 0 O W 3 5 7 0 1 201  
Figure imgf000104_0001
Example 9: Biacore binding kinetics of anti-SARS-CoV-2-S monoclonal antibodies [000196] Equilibrium dissociation constants (KD) for different SARS-CoV-2-S antibodies from primary supernatants from CHOt cells or from hybridomas were determined using a real-time surface plasmon resonance-based Biacore T200/Biacore 8K biosensor. All binding studies were performed in 10mM HEPES, 150mM NaCl, 3mM EDTA, and 0.05% v/v Surfactant Tween-20, pH 7.4 (HBS-ET) running buffer at 25 °C. The Biacore CM5 sensor chip surface was first derivatized by amine coupling with either mouse anti-human Fc specific mAb or rabbit anti-mouse Fcg monoclonal antibody (GE, Catalog # BR-1008-38) to capture anti-SARS-CoV-2 antibodies. Binding studies were performed on a human SARS-CoV-2 RBD extracellular domain expressed with a C-terminal myc-myc-hexahistidine tag (SARS-COV-2 RBD-MMH), SARS-CoV-2 RBD extracellular domain expressed with a C-terminal mouse IgG2a (SARS-COV-2 RBD-mFc), or SARS-CoV-2 RBD extracellular domain expressed with a C-terminal human IgG1 (SARS-COV- 2 RBD-hFc). Single concentrations of SARS-COV-2 RBD-MMH, (100nM); SARS-COV-2 RBD- mFc (50nM), or SARS-COV-2 RBD-hFc (50nM), prepared in HBS-ET running buffer, were injected for 1.5 minutes at a flow rate of 30µL/min while the dissociation of antibody-bound different SARS-CoV-2 RBD reagents was monitored for 2 minutes in HBS-ET running buffer. At the end of each cycle, the SARS-CoV-2 RBD antibody capture surface was regenerated using either a 10 sec injection of 20mM phosphoric acid for the mouse anti-human Fc specific monoclonal antibody surface or a 40 sec injection of 10mM Glycine, HCl, pH1.5 for the rabbit anti-mouse Fcg specific polyclonal antibody. The association rate (ka) and dissociation rate (kd) were determined by fitting the real-time binding sensorgrams to a 1:1 binding model with mass transport limitation using BiaEvaluation software v3.1 or Biacore Insight Evaluation software v2.0. or curve-fitting software. Binding dissociation equilibrium constant (KD) and dissociative half-life (t½) were calculated from the kinetic rates as:
Figure imgf000105_0001
[000197] Binding kinetics parameters for different SARS-CoV-2 monoclonal antibodies binding to different anti-SARS-COV-2 RBD reagents of the invention at 25°C are shown in Tables 11 and 12.  
Table 11: Binding kinetics of SARS-COV-2 RBD-MMH binding to anti-SARS-CoV-2 monoclonal antibodies at 25 °C
Figure imgf000106_0001
 
Figure imgf000107_0001
 
Figure imgf000108_0001
Table 12: Binding kinetics of SARS-COV-2 RBD-mFc or SARS-COV-2 RBD-hFc binding to anti-SARS-CoV-2-S monoclonal antibodies at 25 °C
Figure imgf000108_0002
 
Figure imgf000109_0001
 
Figure imgf000110_0001
 
Figure imgf000111_0001
*: Estimated value based on the limit of measurement of the dissociative rate constant and dissociative half-life under the experimental conditions. Example 10: Characterization of anti-SARS-CoV-2-S monoclonal antibodies by blocking ELISA [000198] An ELISA-based blocking assay was developed to determine the ability of anti- SARS-CoV2-S antibodies to block the binding of the SARS-CoV-2 spike protein receptor binding domain (RBD) to human angiotensin converting enzyme 2 (hACE2). [000199] The SARS-CoV-2 protein used in the experiments was comprised of the receptor binding domain (RBD) portion of the SARS-CoV-2 spike protein (amino acids Arg319 to Phe541) expressed with the Fc portion of the human IgG1 at the c-terminus (SARS-CoV-2 RBD- hFc; see NCBI accession number MN908947.3) The human ACE2 protein used in the experiments was purchased from R&D systems and is comprised of amino acids glutamine 18 to serine 740 with a c-terminal 10X-Histidine tag (hACE2-His; NCBI accession number Q9BYF1). [000200] Experiments were carried out using the following procedure. A monoclonal anti- Penta-His antibody (Qiagen) was coated at 1 µg/ml in PBS on a 96-well microtiter plate overnight at 4 °C. The hACE2-His receptor was added at 0.2 µg/ml in PBS and bound for 2 hours at room temperature. Nonspecific binding sites were subsequently blocked using a 0.5% (w/v) solution of BSA in PBS. In other microtiter plates, a constant amount of 10 pM or 15 pM (as indicated in Table 13) of SARS-CoV-2 RBD-hFc protein was bound with antibodies diluted 1:10 or 1:20 in PBS + 0.5% BSA. These antibody-protein complexes, after a one-hour incubation, were transferred to the microtiter plate coated with hACE2-His. After 1.5 hours of  
incubation at RT, the wells were washed, and plate-bound SARS-CoV-2 RBD-hFc protein was detected with goat-anti-human IgG antibody conjugated with horseradish peroxidase (HRP) (Jackson). The plates were then developed using TMB substrate solution (BD Biosciences, catalog #555214) according to manufacturer’s recommendation and absorbance at 450nm was measured on a Victor X5 plate reader. [000201] Data analysis was performed by calculating the % reduction of signal of the fixed SARS-CoV-2-S RBD-hFc concentration in the presence of the antibody vs in the absence of the antibody. In the calculation, binding signal of the sample of the constant SARS-CoV-2-S RBD- hFc without the presence of the antibody for each plate was referenced as 100% binding or 0% blocking; and the baseline signal of the sample of media only without the presence of SARS- CoV-2 RBD-hFc was referenced as 0% binding or 100% blocking. [000202] The ability of anti-SARS-CoV-2-S antibodies to block SARS-CoV-2-S RBD from binding to human ACE2 was assessed using a blocking ELISA format. Single point test antibody supernatant blocking of either 10 pM or 15 pM SARS-CoV-2-S RBD-hFc binding to hACE2-His, which was presented on anti-His antibody coated on 96-well microtiter plates, was detected with an HRP conjugated anti-hFc antibody. [000203] The blocking results of three assays are summarized in Table 13. The SARS-CoV-2- S binding signal (450 nm) and theG calculated % blocking are indicated. A range of blocking is observed for the test samples. For samples where an NA is indicated in columns 6 and 7, a plate- corrected value is included in columns 4 and 5, as data was consistent with a single plate switch occurring for those samples.43 of 46 antibody supernatants blocked greater than 50% of the SARS-CoV-2-S RBD-hFc binding to plate-coated human ACE2, with 16 of them blocking >90% of the signal. Table 13: Blocking ELISA Results
Figure imgf000112_0001
 
Figure imgf000113_0001
 
Figure imgf000114_0001
 
Figure imgf000115_0001
Example 11: Epitope mapping of anti-SARS-CoV-2-S monoclonal antibodies to spike glycoprotein by Hydrogen-Deuterium Exchange Mass Spectrometry [000204] Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS) was performed to determine the amino acid residues of the SARS-CoV-2 Spike Protein Receptor Binding Domain (RBD (amino acids R319-F541)) that interact with mAb10989, mAb10987, mAb10934, mAb10933, mAb10920, mAb10922, mAb10936, mAb10954, mAb10964, mAb10977, mAb10984, and mAb10986. A general description of the HDX-MS method is set forth in e.g., Ehring (1999) Analytical Biochemistry 267(2):252-259; and Engen and Smith (2001) Anal. Chem.73:256A-265A. [000205] The HDX-MS experiments were performed on an integrated HDX-MS platform, consisting of a Leaptec HDX PAL system for the deuterium labeling and quenching, a Waters Acquity I-Class (Binary Solvent Manager) for the sample digestion and loading, a Waters Acquity I-Class (Binary Solvent Manager) for the analytical gradient, and a Thermo Q Exactive HF mass spectrometer for peptide mass measurement. [000206] The labeling solution was prepared as PBS buffer in D2O at pD 7.0 (10 mM phosphate buffer, 140 mM NaCl, and 3 mM KCl, equivalent to pH 7.4 at 25°C). For deuterium labeling, 10 mL of the RBD protein or RBD protein premixed with each one of the 12 antibodies listed above were incubated at 20 °C with 90 mL of D2O labeling solution for various timepoints, in duplicate. For mAb10989, mAb10987, mAb10934, and mAb10933, the time points were 0   min (non-deuterated control), 5 min, and 10 min. For mAb10920, mAb10922, mAb10936, mAb10954, mAb10964, mAb10977, mAb10984, and mAb10986, the time points were 0 min (non-deuterated control) and 10 min. The deuteration reaction was quenched by adding 90 mL of pre-chilled quench buffer (0.5 M TCEP-HCl, 4 M urea and 0.5% formic acid) to each sample for a 90 second incubation at 20 °C. The quenched samples were then injected into the Leaptec HDX PAL system for online pepsin/protease XIII digestion. The digested peptides were trapped by a C18 column (2.1 mm x 5 mm, Waters) and separated by another C18 column (2.1 mm x 50 mm, Waters) at -5 °C with a 20 minute gradient (for mAb10989, mAb10987, mAb10934, and mAb10933) or a 10 minute gradient (for mAb10920, mAb10922, mAb10936, mAb10954, mAb10956, mAb10964, mAb10977, and mAb10984) from 0% to 90% of movbile phase B solution (mobile phase A solution: 0.5% formic acid and 4.5% acetonitrile in water, mobile phase B solution: 0.5% formic acid in acetonitrile). The eluted peptides were analyzed by a Thermo Q Exactive HF mass spectrometry in LC-MS/MS or LC-MS mode. [000207] The LC-MS/MS data from the undeuterated RBD protein sample were searched against a database including amino acid sequences of the RBD protein, pepsin, protease XIII, and their reversed sequences using Byonic search engine (Protein Metrics). The search parameters were set as default using non-specific enzymatic digestion and human glycosylation as common variable modification. The list of identified peptides was then imported into HDExaminer software (version 3.1) to calculate the deuterium uptake (D-uptake) and differences in deuterium uptake percentage (D%D) for all deuterated samples. Difference in deuterium uptake percentage (D%D) was calculated as follows. Difference in deuterium uptake (DD) = D-uptake (RBD-mAb) - D-uptake (RBD alone) Difference^in^deuterium^uptake^percentage
Figure imgf000116_0001
[000208] A total of 190 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10989 samples, representing 86.06% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 467-513 (DISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVL) (SEQ ID NO: 835) of the RBD were significantly protected by mAb10989.  
[000209] A total of 187 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10987 samples, representing 86.06% sequence coverage of RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 432-452 (CVIAWNSNNLDSKVGGNYNYL) (SEQ ID NO: 836) of the RBD were significantly protected by mAb10987. [000210] A total of 188 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10934 samples, representing 86.06% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 432-452 (CVIAWNSNNLDSKVGGNYNYL) (SEQ ID NO: 836), 467-474 (DISTEIYQ) (SEQ ID NO: 837), and 480-513 (CNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVL) (SEQ ID NO: 838) of the RBD were significantly protected by mAb10934. [000211] A total of 188 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10933 samples, representing 86.06% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 467-510 (DISTEIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRV) (SEQ ID NO: 839) of the RBD were significantly protected by mAb10933. [000212] A total of 75 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10920 samples, representing 83.27% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 471-486 (EIYQAGSTPCNGVEGF) (SEQ ID NO: 840), and 491-515 (PLQSYGFQPTNGVGYQPYRVVVLSF) (SEQ ID NO: 841) of the RBD were significantly protected by mAb10920. [000213] A total of 86 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10922 samples, representing 87.25% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less  
than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 432-452 (CVIAWNSNNLDSKVGGNYNYL) (SEQ ID NO: 836) of the RBD were significantly protected by mAb10922. [000214] A total of 81 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10936 samples, representing 82.07% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 351-360 (YAWNRKRISN) (SEQ ID NO: 842), 432-452 (CVIAWNSNNLDSKVGGNYNYL) (SEQ ID NO: 836), 467-486 (DISTEIYQAGSTPCNGVEGF) (SEQ ID NO: 843), and 491-513 (PLQSYGFQPTNGVGYQPYRVVVL) (SEQ ID NO: 844) of the RBD were significantly protected by mAb10936. [000215] A total of 84 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10954 samples, representing 87.25% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 400-422 (FVIRGDEVRQIAPGQTGKIADYN) (SEQ ID NO: 845), 453-486 (YRLFRKSNLKPFERDISTEIYQAGSTPCNGVEGF) (SEQ ID NO: 846), and 490-515 (FPLQSYGFQPTNGVGYQPYRVVVLSF) (SEQ ID NO: 847) of the RBD were significantly protected by mAb10954. [000216] A total of 109 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10964 samples, representing 83.67% sequence coverage of RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 401-424 (VIRGDEVRQIAPGQTGKIADYNYK) (SEQ ID NO: 848), and 471-513 (EIYQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVVVL) (SEQ ID NO: 849) of the RBD were significantly protected by mAb10964. [000217] A total of 78 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10977 samples, representing 87.25% sequence coverage of the RBD. Any  
peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 351-364 (YAWNRKRISNCVAD) (SEQ ID NO: 850), and 471-486 (EIYQAGSTPCNGVEGF) (SEQ ID NO: 840) of the RBD were significantly protected by mAb10977. [000218] A total of 88 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10984 samples, representing 87.25% sequence coverage of RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 400-422 (FVIRGDEVRQIAPGQTGKIADYN) (SEQ ID NO: 845), and 453-486 (YRLFRKSNLKPFERDISTEIYQAGSTPCNGVEGF) (SEQ ID NO: 846) of the RBD were significantly protected by mAb10984. [000219] A total of 84 peptides from the RBD were identified from both RBD alone and RBD in complex with mAb10986 samples, representing 87.25% sequence coverage of the RBD. Any peptide that exhibited a reduction in deuterium uptake of 5% or greater (i.e., a D%D value of less than -5%, such as -6%, -10%, and so on) upon mAb binding was defined as significantly protected. Peptides corresponding to amino acids 400-422 (FVIRGDEVRQIAPGQTGKIADYN) (SEQ ID NO: 845), 453-486 (YRLFRKSNLKPFERDISTEIYQAGSTPCNGVEGF) (SEQ ID NO: 846), and 490-515 (FPLQSYGFQPTNGVGYQPYRVVVLSF) (SEQ ID NO: 847) of the RBD were significantly protected by mAb10986. [000220] In sum, the majority of the neutralizing antibodies tested contact the RBD in a manner that overlaps the RBD residues that comprise the ACE2 interface; furthermore, the antibodies can be grouped based on their pattern of contacting the RBD surface, as shown in Figure 15. The above data are also summarized in Tables 14-25. Table 14: Spike protein receptor binding domain (RBD) peptides with significant protection upon formation of RBD-mAb compared to RBD alone
Figure imgf000119_0001
 
Figure imgf000120_0002
Table 15: Spike protein RBD peptides with significant protection upon formation of RBD- mAb10987 complex comparing to RBD alone
Figure imgf000120_0001
 
Figure imgf000121_0002
Table 16: RBD peptides with significant protection upon formation of RBD-mAb10934 complex comparing to RBD alone
Figure imgf000121_0001
 
Figure imgf000122_0002
Table 17: RBD peptides with significant protection upon formation of RBD-mAb10933 complex comparing to RBD alone
Figure imgf000122_0001
 
Figure imgf000123_0003
Table 18: RBD peptides with significant protection upon formation of RBD-mAb10920 complex comparing to RBD alone
Figure imgf000123_0001
Table 19: RBD peptides with significant protection upon formation of RBD-mAb10922 complex comparing to RBD alone
Figure imgf000123_0002
 
Figure imgf000124_0002
Table 20: RBD peptides with significant protection upon formation of RBD-mAb10936 complex comparing to RBD alone
Figure imgf000124_0003
Table 21: RBD peptides with significant protection upon formation of RBD-mAb10954 complex comparing to RBD alone
Figure imgf000124_0001
 
Figure imgf000125_0002
Table 22: RBD peptides with significant protection upon formation of RBD-mAb10964 complex comparing to RBD alone
Figure imgf000125_0001
 
Figure imgf000126_0003
Table 23: RBD peptides with significant protection upon formation of RBD-mAb10977 complex comparing to RBD alone
Figure imgf000126_0001
Table 24: RBD peptides with significant protection upon formation of RBD-mAb10984 complex comparing to RBD alone
Figure imgf000126_0002
 
Figure imgf000127_0001
Table 25: RBD peptides with significant protection upon formation of RBD-mAb10986 complex comparing to RBD alone
Figure imgf000127_0002
 
Figure imgf000128_0001
Example 12: Neutralization of SARS-CoV-2 wild-type and variant spike proteins [000221] To test whether anti-SARS-CoV-2 spike protein antibodies can neutralize SARS- CoV-2 variants, these antibodies were screened against a panel of VSV pseudotype viruses expressing wild-type and variant spike proteins. VSV pseudotype viruses were generated by transiently transfecting 293T cells with a plasmid encoding the SARS-CoV-2 spike protein or the same plasmid containing nucleotide variations that encode for known variants of the SARS- CoV-2 spike protein amino acid sequence. All plasmids were confirmed by Sanger sequencing. Cells were seeded in 15 cm plates at 1.2x107 cells per plate in DMEM Complete Media (1000 mL DMEM, Gibco; 100 mL FBS, Gibco; 10 mL PSG, Gibco) one day prior to transfection with 15 µg/plate Spike DNA using 125 µL Lipofectamine LTX, 30 µL PLUS reagent, and up to 3 mL Opti-Mem. 24 hours post transfection, the cells were washed with 10 mL PBS, then infected with an MOI of 0.1 VSVDG:mNeon virus in 10 mL of Opti-Mem. Virus was incubated on cells for 1 hour, with gentle rocking every 10 minutes. Cells were washed 3 times with 10 mL PBS, then overlaid with 20 mL Infection media (1000 mL DMEM, Gibco; 10 mL Sodium Pyruvate, Gibco; 7 mL BSA, Sigma; 5 mL Gentamicin, Gibco) before incubation at 37 °C, 5% CO2 for 24 hours.  
Pseudovirus supernatant was collected into 250 mL centrifuge tubes on ice, then centrifuged at 3000 rpm for 5 minutes to pellet any cellular debris, aliquoted on ice, then frozen to -80 °C. Infectivity was tested on Vero cells prior to use in neutralization assays. This material will be referred to as VSVDG:mNeon/Spike pseudovirus, or VSVDG:mNeon/Spike_(variant amino acid mutation) (for example, VSVDG:mNeon/Spike_H49Y). [000222] On Day 1, Vero cells were seeded to 80% confluency in T225 flasks, the cells were washed with PBS (Gibco: 20012-043), TrypLE was added to detach cells from the flask, and Complete DMEM was added to inactivate trypsin.20,000 Vero cells were plated in in 100 µL of prewarmed Complete DMEM per well in 96 Well Black Polystyrene Microplate (Corning: 3904). On Day 2, VSVDG:mNeon/Spike pseudovirus was thawed on ice and diluted with Infection media. Antibodies were diluted in a U-bottom 96 well plate, generating a dilution of each antibody in 210 µl Infection media at 2X assay concentration.120 µL of diluted antibodies were transferred to a fresh U-bottom plate, and media and an IgG1 control antibody were added to each plate.120 µl of diluted pseudovirus was added to every well except the media control wells. To those wells, 120 µL of Infection media was added. Pseudovirus with antibodies were incubated for 30 minutes at room temperature, then media was removed from Vero cells.100 µL of antibody/pseudovirus mixture were added to the cells, and then incubated at 37 °C, 5% CO2 for 24 hours. On day 3, supernatant was removed from cell wells and replaced with 100 µL of PBS. Plates were read on a SpectraMax i3 with MiniMax imaging cytometer. [000223] In addition to testing neutralization capacity with non-replicating VSV-SARS-CoV- 2-S virus, antibodies also were tested with SARS-CoV-2 virus. Monoclonal antibodies and antibody combinations were serially diluted in DMEM (Quality Biological), supplemented with 10% (v/v) heat inactivated fetal bovine serum (Sigma), 1% (v/v) penicillin/streptomycin (Gemini Bio-products) and 1% (v/v) L-glutamine (2 mM final concentration, Gibco) (VeroE6 media) to a final volume of 250 mL. Next, 250 mL of VeroE6 media containing SARS-CoV-2 (WA-1) (1000 PFU/mL) was added to each serum dilution and to 250 mL media as an untreated control. The virus-antibody mixtures were incubated for 60 min at 37 °C. Following incubation, virus titers of the mixtures were determined by plaque assay. Finally, 50% plaque reduction neutralization titer (PRNT50) values (the serum dilutions at which plaque formation was reduced by 50% relative to that of the untreated control) were calculated using a 4-parameter logistic curve fit to the percent neutralization data (GraphPad Software, La Jolla, CA).  
[000224] Individual monoclonal antibody half maximal inhibitory concentration (IC50) against VSV-SARS-CoV-2 spike protein (S)-expressing pseudovirus encoding the Wuhan-Hu-1 (NCBI Accession Number MN908947.3) sequence of spike protein (S-wt) were determined in Vero cells (Table 26). The majority of antibodies displayed neutralization potency in the picomolar range (pM), with some exhibiting neutralization potency in nanomolar (nM) range. [000225] While recombinant ACE2 was able to mediate neutralization of the VSV-spike pseudoparticles, as previously reported, its potency was far inferior to that of the monoclonal antibodies, with more than 1000-fold decrease in potency seen relative to the best neutralizing mAbs (Figure 10A). In addition, the potent neutralizing activity of mAb10987, mAb10989, mAb10933, and mAb10934 was confirmed in neutralization assays, including neutralization of SARS-CoV-2 in VeroE6 cells (Figure 10B). All neutralization assays generated similar potency across the four mAbs (mAb10987, mAb10989, mAb10933, and mAb10934) and no combinations demonstrated synergistic neutralization activity (Figure 10B). Table 26: mAb neutralization potency (IC50 (M)) against wild-type strain of VSV-SARS- CoV-2-S pseudoparticles in Vero cells
Figure imgf000130_0001
 
Figure imgf000131_0001
 
Figure imgf000132_0001
[000226] Amino acid variants in spike (S) protein were identified from over 7000 publicly available SARS-CoV-2 sequences, representing globally circulating isolates, and cloned into VSV pseudoparticles. Neutralization assays with variant-encoding pseudoparticles were performed to assess the impact of each variant on neutralization potency of the monoclonal antibodies. Table 27 illustrates the relative neutralization potency of monoclonal antibodies against variant encoding pseudoparticles relative to SARS-CoV-2 spike (S-wt) at a single concentration of 5 µg/ml. Percent of neutralization relative to S-wt was captured for each individual antibody and variant. None of the antibodies demonstrated loss of neutralization potency at the 5 µg/ml concentration with the exception of mAb10985 and the R408I variant. These data demonstrate broad functional neutralization coverage of monoclonal antibodies against globally circulating SARS-CoV-2 spike variants. [000227] To further interrogate the impact of the S protein variants on neutralization potency of the monoclonal antibodies, full neutralization curves were run to determine the IC50 value of the most potent neutralizing antibodies against a subset of variants localized within the receptor binding domain (RBD) of the S protein. Table 28 shows the IC50 neutralization values for each  
variant psuedoparticle. Intrinsic variability of up to 3-fold can be observed between pseudoparticle neutralization assays and does not indicate a change in neutralization potency. These data demonstrate that the antibodies retained their neutralization potency against a diverse panel of S protein RBD variants. Table 27. Relative neutralization of VSV-SARS-CoV-2 variants encoding S protein at 5 µg/ml antibody concentration in Vero cells
Figure imgf000133_0001
 
Figure imgf000134_0001
 
Figure imgf000135_0001
Table 28. Neutralization IC50 (M) of VSV-SARS-CoV-2-S RBD variants in Vero cells
Figure imgf000135_0002
 
Figure imgf000136_0001
Table 28 (cont’d)
Figure imgf000136_0002
Example 13: Biacore binding kinetics of purified anti-SARS-CoV-2-S monoclonal antibodies [000228] Equilibrium dissociation constant (KD) for different SARS-COV-2 RBD reagents binding to purified CHOt anti-SARS-COV-2 monoclonal antibodies (mAbs) were determined using a real-time surface plasmon resonance based Biacore T200/Biacore 8K biosensor. All binding studies were performed in 10mM HEPES, 150mM NaCl, 3mM EDTA, and 0.05% v/v Surfactant Tween-20, pH 7.4 (HBS-ET) running buffer at 25 °C and 37 °C. The Biacore CM5 sensor chip surface was first derivatized by amine coupling with either mouse anti-human Fc specific mAb (Regeneron, mAb2567) to capture anti-SARS-COV-2bmAbs. Binding studies were performed on human SARS-COV-2 RBD extracellular domain expressed with a C-terminal  
myc-myc-hexahistidine (SARS-COV-2 RBD-MMH) and SARS-COV-2 RBD extracellular domain expressed with a C-terminal mouse IgG2a (SARS-COV-2 RBD-mFc). Use of these reagents allowed for the testing of the antibodies’ ability to bind monomeric and dimeric RBD peptides, respectively. [000229] Different concentrations of hSARS-COV-2 RBD-MMH, (90nM-3.33nM, 3-fold dilution ) and SARS-COV-2 RBD-mFc (30nM-1.11nM 3-fold dilution) prepared in HBS-ET running buffer, were injected for 3 minutes at a flow rate of 50µL/min while the dissociation of mAb bound different SARS-COV-2 RBD reagents was monitored for 6-10 minutes in HBS-ET running buffer. At the end of each cycle, the SARS-COV-2 RBD mAb capture surface was regenerated using either 12sec injection of 20mM phosphoric acid for mouse anti-human Fc specific mAb surface. The association rate (ka) and dissociation rate (kd) were determined by fitting the real-time binding sensorgrams to a 1:1 binding model with mass transport limitation using BiaEvaluation software v3.1 or Biacore Insight Evaluation software v2.0. or curve-fitting software. Binding dissociation equilibrium constant (KD) and dissociative half-life (t½) were calculated from the kinetic rates as:
Figure imgf000137_0001
[000230] Binding kinetics parameters for different SARS-COV-2 mAbs binding to different anti-SARS-COV-2 RBD reagents of the invention at 25 °C and 37 °C are shown in Tables 29 through 32, respectively. Table 29: Binding kinetics parameters of SARS-COV-2 RBD-MMH binding to anti-SARS- COV-2-S monoclonal antibodies at 25 °C.
Figure imgf000137_0002
 
Figure imgf000138_0001
Table 30: Binding kinetics parameters of SARS-COV-2 RBD-MMH binding to anti-SARS- COV-2-S monoclonal antibodies at 37 °C.  
Figure imgf000139_0001
 
Figure imgf000140_0001
Table 31: Binding kinetics parameters of SARS-COV-2 RBD-mFc binding to anti-SARS- COV-2-S monoclonal antibodies at 25 °C.
Figure imgf000140_0002
 
Figure imgf000141_0001
Table 32: Binding kinetics parameters of SARS-COV-2 RBD-mFc binding to anti-SARS- COV-2-S monoclonal antibodies at 37 °C
Figure imgf000141_0002
 
Figure imgf000142_0001
Example 14: Anti-SARS-CoV-2 Antibodies block RBD binding to hACE2 as determined by ELISA [000231] An ELISA-based blocking assay was used to determine the ability of anti-SARS- CoV-2 antibodies to block the binding of the SARS-COV-2 Spike protein receptor binding domain (RBD) to its receptor, human angiotensin converting enzyme 2 (hACE2). [000232] The SARS-CoV-2 protein used in this assay was comprised of the receptor binding domain (RBD) portion of the SARS-CoV-2 Spike protein (amino acids Arg319-Phe541) expressed with the Fc portion of the human IgG1 at the c-terminus (SARS-CoV-2 RBD-hFc) The human ACE2 protein used in the experiments was purchased from R&D Systems and was   comprised of amino acids Gln18-Ser740 with a C-terminal 10X-Histidine tag (hACE2-His; NCBI Accession No. Q9BYF1). [000233] Experiments were carried out using the following procedure. A monoclonal anti- Penta-His antibody (Qiagen) was coated at 1µg/ml in PBS on a 96-well microtiter plate overnight at 4 °C. The hACE2-His receptor was added at 0.2ug/ml in PBS and bound for two hours at room temperature (RT). Nonspecific binding sites were subsequently blocked using a 0.5% (w/v) solution of BSA in PBS. In other microtiter plates, a constant amount of 100pM of SARS-CoV-2 RBD-hFc protein was bound with anti-SARS-COV-2 antibodies and an isotype IgG1 antibody control at dilutions from 0.0008nM to 50nM in PBS +0.5% BSA. After a one- hour incubation, the mixture solutions were transferred to the microtiter plate coated hACE2-His. After 1.5 hours of incubation at RT, the wells were washed, and plate-bound SARS-COV2 was detected with goat-anti-human IgG antibody conjugated with horseradish peroxidase (HRP) (Jackson). The plates were then developed using TMB substrate solution (BD Biosciences, #555214) according to manufacturer’s recommendation and absorbance at 450nm was measured on a Victor X5 plate reader. [000234] Binding data were analyzed using a sigmoidal dose-response model within Prism™ software (GraphPad). The calculated IC50 value, defined as the concentration of antibody required to block 50% of SARS-CoV-2 RBD-hFc binding to plate-coated hACE2-His, was used as an indicator of blocking potency. Percent blocking was defined based on the background-corrected binding signal observed at the highest antibody concentration tested using this formula and reported for all tested antibodies:
Figure imgf000143_0001
[000235] Antibodies that blocked binding less than or equal to 50% at the highest concentration tested were classified as non-blockers and IC50 values were not reported for those antibodies. [000236] The ability of anti-SARS-CoV-2 antibodies to block SARS-CoV-2 RBD binding to human ACE2 was assessed using a blocking ELISA. In this assay 100pM SARS-COV-2 RBD- hFc was titrated with a wide range of the concentrations of the anti-SARS-CoV-2-S antibody and  
the inhibition of the presence of the antibody on RBD binding to hACE2-His was evaluated. The plate-bound RBD-hFc was detected with an HRP conjugated anti-hFc antibody. [000237] The blocking IC50s and maximum blocking at the highest tested concentrations of the anti-SARS-CoV-2-S antibodies are summarized in Table 33, and the blocking curves shown in Figures 1-8. Of the 46 antibodies tested, 44 displayed antibody concentration-dependent blocking of RBD.hFc binding to hACE-2. IC50 values ranged from 41pM to 4.5nM and maximum blocking ranging from 55% to about 100% at the highest antibody concentration tested. Two antibodies out of 46 tested showed no blocking activities under the assay conditions. The irrelevant isotype control antibody showed no blocking activity, as expected. Table 33: Blocking potency of Anti-SAR-COV-2 Antibodies on Spike RBD-hFc Binding to Immobilized Human ACE-2
Figure imgf000144_0001
 
Figure imgf000145_0001
 
Figure imgf000146_0001
 
Figure imgf000147_0001
Note: RBD-hFc at 100 pM was titrated with anti-SARS-COV-2-S antibodies in serial dilutions from 50nM and bound RBD-hFc on immobilized hACE2 with a 10x histidine tag, and detected with HRP-conjugated anti-hFc antibody. NBD; no blocking detected. Example 15: Cross-competition between mAb10987, mAb10989, mAb10933, and mAb10934 [000238] mAb10987, mAb10989, mAb10933, and mAb10934 were examined in cross- competition binding assays (Figure 11), identifying several pairs of non-competing mAbs with picomolar neutralization potency that could potentially be combined to form antibody cocktails, e.g., mAb10987 and mAb0933. [000239] Epitope binning of the anti-SARS-CoV-2-S mAbs was conducted in a pre-mix sandwich format involving competing mAbs against one another in a pairwise combinatorial manner for binding to SARS-CoV-2 RBD-MMH protein using a ForteBio Octet HTX biolayer interferometry instrument (Molecular Devices ForteBio LLC, Fremont, CA) with running buffer of 10 mM HEPES, 150 mM NaCl, 0.05% (v/v) Tween-20, pH 7.4, 1 mg/mL BSA. Assays were performed at 30 ºC with continuous agitation at 1000 rpm. After obtaining an initial baseline in running buffer 20 mg/mL of anti-COVID19 mAbs was captured onto anti-human Fc (AHC) biosensor tips for 300 s. To block remaining free unsaturated binding sites on AHC biosensor tips, all sensors were exposed for 240 s to blocking solution well containing 100 mg/mL irrelevant IgG1. Following this process, biosensors were immersed into wells containing pre-mix  
solution of 100nM SARS CoV-2 RBD-MMH protein and 600 nM of anti-COVID19 mAb binding site of a second mAbs for 300 s. Binding response at each step was recorded and specific signal was normalized by subtracting self-blocking mAb competing control from dataset. Data analysis was performed with Octet Data Analysis HT 10.0 software using the Epitope Binning. [000240] Comparing the cross-competition binding assays with the HDX-MS results described above provides structural insights into the mechanism by which non-competing pairs of antibodies can simultaneously bind the RBD, and can thus be ideal partners for a therapeutic antibody cocktail. mAb10987 and mAb10933 represent such a pair of antibodies. mAb10933 targets the spike-like loop region on one edge of the ACE2 interface. Within that region, the residues that show the most significant HDX protection by mAb10933 face upward, suggesting that the Fab region of mAb10933 binds the RBD from the top direction, where mAb10933 will have significant collisions with ACE2. In order to avoid competition with mAb10933, mAb10987 only binds to the HDX-defined protected regions from the front or the lower left side (in the front view of mAb10987 in Figure 12). This is consistent with the neutralization data described above, as mAb10987 would orient it in a position that has high probability to interfere with ACE2. Example 16: Structure determination of antibody-bound spike protein [000241] To better understand the binding of mAb10933 and mAb10987 to the spike protein RBD, structural analysis was performed via cryo-electron microscopy (cryoEM). Fab fragments of mAb10933 and mAb10987 were isolated using FabALACTICA kit (Genovis).600 µg of the mAb10933 Fab and 600 µg of mAb10987 Fab were mixed with 300 µg of SARS-CoV-2-S RBD and incubated on ice for ~1 hour then injected into a Superdex 200 increase gel filtration column equilibrated to 50 mM Tris pH 7.5, 150 mM NaCl. Peak fractions containing the mAb10933 Fab - mAb10987 Fab - RBD complex were collected and concentrated using a 10 kDa MWCO centrifugal filter. For cryoEM grid preparation, the protein sample was diluted to 1.5 mg/mL and 0.15% PMAL-C8 amphipol was added.3.5 µL of protein was deposited onto a freshly plasma cleaned UltrAufoil grid (1.2/1.3, 300 mesh). Excess solution was blotted away using filter paper and plunge frozen into liquid ethane using a Vitrobot Mark IV. The cryoEM grid was transferred to a Titan Krios (Thermo Fisher) equipped with a K3 detector (Gatan). Movies were collected using EPU (Thermo Fisher) at 105,000x magnification, corresponding to a pixel size of 0.85 Å.  
A dose rate of 15 electrons per pixel per second was used and each movie was 2 seconds, corresponding to a total dose of ~40 electrons per Å2. [000242] All cryoEM data processing was carried out using cryoSPARC v2.14.2.2,821 movies were aligned using patch motion correction and patch CTF estimation.2,197 aligned micrographs were selected for further processing on the basis of estimated defocus values and CTF fit resolutions. An initial set of particles picked using blob picker were subjected to 2D classification to generate templates for template picking.989,553 particles picked by template picking were subjected to multiple rounds of 2D classification to remove unbound fabs and particles containing an incomplete complex. Ab initio reconstruction with three classes generated a single class containing 61,707 particles that corresponded to the mAb10933 Fab - mAb10987 Fab - RBD complex. Heterogenous refinement of the particles in this class followed by non- uniform refinement resulted in a 3.9 Å resolution (FSC=0.143) map containing 48,140 particles that was used for model building. Into this map, models of the RBD (taken from PDB code 6M17) and the two Fabs (taken from prior antibody structures, except for the lambda light chain of mAb10987 which came from PDB code 5U15), were manually placed. These models were then manually rebuilt using Coot and real-space refined against the map using Phenix. [000243] Confirming the above-described data, single-particle cryoEM of the complex of SARS-CoV-2 spike RBD bound to Fab fragments of mAb10933 and mAb10987 shows that the two antibodies in this cocktail can simultaneously bind to distinct regions of the RBD (Figure 13A, Figure 13B, and Figure 14). A 3D reconstructed map of the complex with nominal resolution of 3.9Å shows that the both Fab fragments bind at different epitopes on the RBD, confirming that they are non-competing antibodies. mAb10933 binds at the top of the RBD, extensively overlapping the binding site for ACE2. On the other hand, the epitope for mAb10987 is located on the side of the RBD, well away from the mAb10933 epitope, and has little to no overlap with the ACE2 binding site. Example 17: Cross-competition between anti-SARS-CoV-2-S mAbs [000244] Binding competition between anti-SARS-CoV-2-S monoclonal antibodies (mAbs) was determined using a real time, label-free bio-layer interferometry (BLI) assay on the Octet HTX biosensor platform (Pall ForteBio Corp.). The entire experiment was performed at 25 °C in 10mM HEPES, 150mM NaCl, 3mM EDTA, and 0.05% v/v Surfactant Tween-20, 1mg/mL BSA,  
pH 7.4 (HBS-EBT) buffer with the plate shaking at a speed of 1000rpm. To assess whether two mAbs were able to compete with one another for binding to their respective epitopes on the SARS-COV-2-S RBD extracellular domain expressed with a C-terminal myc-myc-hexahistidine (SARS-COV-2 RBD-MMH), ~0.51nm of SARS-COV-2-S RBD-MMH was first captured onto anti-Penta-His antibody coated Octet biosensor tips (Fortebio Inc, # 18-5122) by submerging the biosensor tips for 1 minute in wells containing a 10 µg/mL solution of SARS-COV-2-S RBD- MMH. The SARS-COV-2-S RBD-MMH captured biosensor tips were then saturated with a first anti-SARS-CoV-2-S monoclonal antibody (subsequently referred to as mAb-1) by dipping into wells containing 50µg/mL solution of mAb-1 for 5 minutes. The biosensor tips were then subsequently dipped into wells containing 50µg/mL solution of a second anti-SARS-CoV-2 monoclonal antibody (subsequently referred to as mAb-2) for 5 minutes. The biosensor tips were washed in HBS-ETB buffer in between every step of the experiment. The real-time binding response was monitored during the entire course of the experiment and the binding response at the end of every step was recorded. The response of mAb-2 binding to SARS-COV-2 RBD- MMH pre-complexed with mAb-1 was compared and competitive/non-competitive behavior of different anti-SARS-CoV-2 monoclonal antibodies was determined as shown in Table 34. Table 34: Cross-competition between anti-SARS-CoV-2-S antibodies
Figure imgf000150_0001
 
mAb10964
Figure imgf000151_0002
Figure imgf000151_0001
 
Figure imgf000152_0001
Figure imgf000152_0002
Figure imgf000152_0003
Figure imgf000152_0004
 
Figure imgf000153_0001
Figure imgf000153_0002
Figure imgf000153_0003
 
mAb10984
Figure imgf000154_0002
Figure imgf000154_0001
 
Figure imgf000155_0001
Figure imgf000155_0002
Figure imgf000155_0003
Figure imgf000155_0004
 
Figure imgf000156_0001
Figure imgf000156_0002
Figure imgf000156_0003
 
mAb10988
Figure imgf000157_0002
Figure imgf000157_0001
 
Figure imgf000158_0001
Figure imgf000158_0002
Figure imgf000158_0003
Figure imgf000158_0004
 
Figure imgf000159_0001
Figure imgf000159_0002
Figure imgf000159_0003
 
mAb10988
Figure imgf000160_0002
Figure imgf000160_0001
 
Figure imgf000161_0001
Figure imgf000161_0002
Figure imgf000161_0003
Figure imgf000161_0004
 
Figure imgf000162_0001
Figure imgf000162_0002
Figure imgf000162_0003
 
mAb10996
Figure imgf000163_0002
Figure imgf000163_0001
 
Figure imgf000164_0001
Figure imgf000164_0002
Figure imgf000164_0003
Figure imgf000164_0004
 
Figure imgf000165_0001
Figure imgf000165_0002
Figure imgf000165_0003
 
Figure imgf000166_0001
Figure imgf000166_0002
Figure imgf000166_0003
 
mAb10920
Figure imgf000167_0002
Figure imgf000167_0001
 
Figure imgf000168_0001
Figure imgf000168_0002
Figure imgf000168_0003
Figure imgf000168_0004
 
Figure imgf000169_0001
Figure imgf000169_0002
Figure imgf000169_0003
 
mAb10988
Figure imgf000170_0002
Figure imgf000170_0001
 
Figure imgf000171_0001
Figure imgf000171_0002
Figure imgf000171_0003
Figure imgf000171_0004
 
Figure imgf000172_0001
Figure imgf000172_0002
Figure imgf000172_0003
 
Figure imgf000173_0001
Figure imgf000173_0002
Figure imgf000173_0003
 
mAb10930
Figure imgf000174_0002
Figure imgf000174_0001
 
Figure imgf000175_0001
Figure imgf000175_0002
Figure imgf000175_0003
Figure imgf000175_0004
 
Figure imgf000176_0001
Figure imgf000176_0002
Figure imgf000176_0003
 
Figure imgf000177_0001
Figure imgf000177_0002
Figure imgf000177_0003
 
mAb10923
Figure imgf000178_0002
Figure imgf000178_0001
 
Figure imgf000179_0001
Figure imgf000179_0002
Figure imgf000179_0003
Figure imgf000179_0004
 
Figure imgf000180_0001
Figure imgf000180_0002
Figure imgf000180_0003
 
Figure imgf000181_0001
Figure imgf000181_0002
Figure imgf000181_0003
 
mAb10920
Figure imgf000182_0002
Figure imgf000182_0001
 
Figure imgf000183_0001
Figure imgf000183_0002
Figure imgf000183_0003
Figure imgf000183_0004
 
Figure imgf000184_0001
Figure imgf000184_0002
Figure imgf000184_0003
 
Figure imgf000185_0001
Figure imgf000185_0002
Figure imgf000185_0003
 
mAb10920
Figure imgf000186_0002
Figure imgf000186_0001
 
Figure imgf000187_0001
Figure imgf000187_0002
Figure imgf000187_0003
Figure imgf000187_0004
 
Figure imgf000188_0001
Figure imgf000188_0002
Figure imgf000188_0003
 
mAb10996
Figure imgf000189_0002
Figure imgf000189_0001
 
Figure imgf000190_0001
Figure imgf000190_0002
Figure imgf000190_0003
 
Figure imgf000191_0001
Figure imgf000191_0002
Figure imgf000191_0003
Figure imgf000191_0004
 
Figure imgf000192_0001
Figure imgf000192_0002
Figure imgf000192_0003
 
Figure imgf000193_0001
Figure imgf000193_0002
Figure imgf000193_0003
 
mAb10920
Figure imgf000194_0002
Figure imgf000194_0001
 
Figure imgf000195_0001
Figure imgf000195_0002
Figure imgf000195_0003
Figure imgf000195_0004
 
Figure imgf000196_0001
Figure imgf000196_0002
Figure imgf000196_0003
 
Figure imgf000197_0001
Figure imgf000197_0002
Figure imgf000197_0003
 
Figure imgf000198_0001
 
Figure imgf000199_0001
Figure imgf000199_0002
Figure imgf000199_0003
Figure imgf000199_0004
 
Figure imgf000200_0001
Figure imgf000200_0002
Figure imgf000200_0003
Figure imgf000200_0004
 
Figure imgf000201_0001
 
Figure imgf000202_0001
 
Figure imgf000203_0001
Figure imgf000203_0002
Figure imgf000203_0003
Figure imgf000203_0004
 
Figure imgf000204_0001
Example 18: pH sensitivity of anti-SARS-CoV-2-S monoclonal antibodies binding to monomeric SARS-CoV-2-S RBD reagents measured at 37 ºC [000245] The dissociation rate constants (kd) for different anti-SARS-CoV-2-S monoclonal antibodies in pH 7.4, pH 6.0, and pH5.0 buffers were determined using a real-time surface plasmon resonance (SPR)-based Biacore T200 biosensor. All binding studies were performed at 37 °C using three running buffers, (i) PBS, 0.05% v/v Surfactant Tween-20, pH7.4 (PBS-T- pH7.4) (ii) PBS, 0.05% v/v Surfactant Tween-20, pH6.0 (PBS-T-pH6.0), and (iii) PBS, 0.05%   v/v Surfactant Tween-20, pH5.0 (PBS-T-pH5.0). The Biacore CM5 sensor chip surface was first derivatized by amine coupling with a mouse anti-human Fc specific mAb (Regeneron) to capture anti-SARS-CoV-2-S monoclonal antibodies. Binding studies were performed on human SARS- COV-2-S RBD extracellular domain expressed with a C-terminal myc-myc-hexahistidine (SARS-COV-2 RBD-MMH), Single concentrations of SARS-COV-2-S RBD-MMH (90nM) prepared in PBS-T-pH 7.4 buffer were injected at a flow rate of 25µL/min for 3 minutes followed by the dissociation of bound SARS-COV-2-S RBD-MMH in PBS-T-pH 7.4, PBS-T- pH 6.0 or PBS-T PBS-T-pH 5.0 running buffers for 5 minutes. [000246] The dissociation rate constants (kd) in four pH running buffers were determined by fitting the real-time binding sensorgrams to a 1:1 binding model using Scrubber 2.0c curve- fitting software. The dissociative half-life (t½) was calculated from the kd values as:
Figure imgf000205_0001
[000247] The kd and t½ values for SARS-COV-2-S RBD-MMH binding to different anti- SARS-CoV-2-S monoclonal antibodies in PBS-T-pH 7.4 followed by dissociation in PBS-T-pH 7.4 and PBS-T-pH 6.0 at 37 °C are shown in Table 35. The kd and t½ values for SARS-COV-2- S RBD-MMH binding to different anti-SARS-CoV-2-S monoclonal antibodies in PBS-T-pH 7.4 followed by dissociation in PBS-T-pH 7.4 and PBS-T-pH 5.0 at 37 °C are shown in Table 36. The comparison of the dissociative half-life (t½) of SARS-COV-2 RBD-MMH in pH 7.4, pH 6.0 and pH 5.0 buffers.   1 0 O W 3 5 7 0 1 402  
Figure imgf000206_0001
3. 1 6. 4 - E - 4 E 0 3 . 1 5 0 2 . 2 731 1 0 O W 3 5 7 0 1 9 7 3 5 6 02 - E - 5 E 6. 4 9 3 . 0 1 1 0 4 1 083 4 690 1 b A m
Figure imgf000207_0001
 
9. 1 5. 1 - E - 7 E 4 3 . 1 5 0 9 . 7 651 1 0 O W 3 5 7 0 1 1 3 4 6 3 02 - E - 6 E 6. 0 9 3 . 0 8 3 7 5 1 234 0 101 1 b A m
Figure imgf000208_0001
 
702  
Figure imgf000209_0001
8. 0 9. 1 - E - 1 E 6 3 . 2 0 0 7 . 6 201 1 0 O W 3 5 7 0 1 1 4 3 5 8 . 0 1 2 - E - 5 E 0 3 . 5 6 0 1 . 7 39 043 0 790 1 b A m
Figure imgf000210_0001
 
4. 1 5. 5 - E - 0 E 7 3 . 0 1 0 1 . 2 39 1 0 O W 3 5 7 0 1 1 1 4 9 9 . 0 7 2 - E - 4 E 4. 6 4 3 . 0 9 1 5 0 1 893 6 001 1 b A m
Figure imgf000211_0001
 
- E0 1. 1 1 0 O W 3 5 7 0 1 012 - E3 3. 1
Figure imgf000212_0001
 
Example 19: Anti-SARS-CoV-2-S antibodies binding to virus-like particles [000248] To investigate the ability of a panel of anti-SARS-CoV-2 monoclonal antibodies to bind SARS-CoV-2 Spike glycoprotein, an in vitro binding assay utilizing vesicular stomatitis virus (VSV) pseudotyped with SARS-CoV-2 Spike protein in an electrochemiluminescence based detection platform (MSD) was developed. [000249] Pseudotyped vesicular stomatitis virus (VSV) viral like particles (VLPs) were generated from HEK293T cells to transiently express the SARS-CoV-2 Spike Protein (Accession number MN908947.3, amino acids 16-1211). VLPs expressing VSV only were also generated as a negative binding control. [000250] Experiments were carried out according to following procedure. VLPs from the two sources described above were diluted in PBS, seeded into the 96-well carbon electrode plates (MULTI-ARRAY high bind plate, MSD) and incubated overnight at 4 °C to allow the VLPs to adhere. Nonspecific binding sites were blocked by 2% BSA (w/v) in PBS for 1 hour at room temperature. To the plate-bound particles, anti-SARS-CoV-2 antibodies and a non-binding human IgG1 control, diluted in PBS + 0.5% BSA at a range of concentrations from 0.0008nM to 50nM, and buffer with no antibody were added in duplicate and the plates incubated for 1 hour at room temperature with shaking. The plates were then washed with 1X PBS to remove the unbound antibodies using an AquaMax2000 plate washer (MDS Analytical Technologies). The plate-bound antibodies were detected with a SULFO-TAGTM-conjugated anti-human IgG antibody (Jackson Immunoresearch) for 1 hour at room temperature. After washes, the plates were developed with the Read Buffer (MSD) according to manufacturer’s recommended procedure and the luminescent signals were recorded with a SECTOR Imager 600 (Meso Scale Development) instrument. The direct binding signals (in RLU) were captured for SARS-CoV-2- expressing VLPs and VSV only VLPs. [000251] The ability of the anti-SARS-CoV-2-S monoclonal antibodies to bind to SARS-CoV- 2-S expressing VLPs compared with binding to irrelevant VSV expressing VLPs was assessed using an immunobinding assay. Binding to the immobilized VLPs on 96-well High Bind plates (MSD) was performed with a series of antibody dilutions and the bound antibodies were detected using SULFO-TAGTM-conjugated anti-human IgG. The binding signals from electrochemiluminescence were recorded on a Sector Imager 600 (MSD). RLU values were determined for the antibody binding to VLPs. All antibodies displayed a concentration-  
dependent binding and the ratios of binding on the SARS-COV-2-S expressing VLPs to VSV only were analyzed at 5.5nM and 0.20nM. [000252] The binding results of anti-SARS-CoV-2-S mAbs at the two concentrations to VSV/spike and VSV-only VLPs are summarized in Table 37. Of 46 antibodies tested, 44 antibodies bound specifically to VSV/spike with a ratio to VSV of 3 or higher at either concentration. At 0.2nM antibody, the ratio of VSV/spike to VSV ranged from 3 to 56, and at 5nM the ratio ranged from 3 to 303. Although two antibodies (mAb10998 and mAb11002) displayed weak binding to the VSV/Spike VLPs, with ratios of less than 3 to the VSV VLPs, the signals at 5nM were higher on the VSV/spike than the VSV. An irrelevant IgG1 isotype antibody showed minimal binding, as expected. Table 37: Specificity of anti-SARS-COV-2-S antibodies binding to spike protein-expressing VSV VLPs vs VSV by Electrochemiluminescence
Figure imgf000214_0001
 
Figure imgf000215_0001
 
Figure imgf000216_0001
Example 20: Anti-SARS-CoV-2-S antibodies binding to spike protein-expressing cells [000253] To investigate the ability of a panel of anti-SARS-CoV-2-S monoclonal antibodies to bind to SARS-CoV-2-S expressing cells, an in vitro binding assay utilizing SARS-CoV-2-S expressing cells in an electrochemiluminescence based detection platform (MSD) was developed. [000254] Jurkat/Tet3G/hCD20/Tet-3G inducible cells were engineered to transiently express the SARS-CoV-2 Spike Protein (Accession number MN908947.3, amino acids 16-1211, Jurkat/Tet3G/hCD20/Tet-On 3G Inducible COVID-19 Spike Protein High Sorted), and flow cytometry sorted for selection of high expression of the SARS-CoV-2 protein. Parental Jurkat/Tet3G/hCD20/Tet-3G were also included in the experiments as a negative binding control. [000255] Experiments were carried out according to following procedure. Cells from the two lines described above were induced with 1 µg/ml doxycycline at 37 °C for 36 hours prior to harvest, spun down, washed with PBS, then diluted in PBS, seeded into the 96-well carbon electrode plates (MULTI-ARRAY high bind plate, MSD), and incubated overnight at 4 °C to  
allow the cells to adhere. Nonspecific binding sites were blocked by 2% BSA (w/v) in PBS for one hour at room temperature. To the plate-bound cells, anti-SARS-CoV-2 antibodies and a non-binding human IgG1 control, diluted in PBS + 0.5% BSA at a range of concentrations from 0.0008nM to 50nM, and buffer with no antibody were added in duplicate and the plates incubated for one hour at room temperature with shaking. The plates were then washed with 1X PBS to remove the unbound antibodies using an AquaMax2000 plate washer (MDS Analytical Technologies). The plate-bound antibodies were detected with a SULFO-TAGTM-conjugated anti-human IgG antibody (Jackson Immunoresearch) for one hour at room temperature. After washes, the plates were developed with the Read Buffer (MSD) according to manufacturer’s recommended procedure and the luminescent signals were recorded with a SECTOR Imager 600 (Meso Scale Development) instrument. The direct binding signals (in RLU) were captured for SARS-CoV-2-S expressing cells and a negative control cell line. [000256] The ability of the anti-SARS-CoV-2 monoclonal antibodies to bind to SARS-CoV-2 Spike Protein expressing cells compared with binding to parental cells was assessed using an immunobinding assay. Binding to the immobilized cells on 96-well high bind plates (MSD) was performed with a series of antibody dilutions and the bound antibodies were detected using SULFO-TAGTM-conjugated anti-human IgG. The binding signals from electrochemiluminescence were recorded on a Sector Imager 600 (MSD). All antibodies displayed a concentration-dependent binding and the ratio of the binding on spike expressing cells to the parental cells were analyzed at the concentration of 5.5nM and 0.20nM. [000257] The binding results of the anti-SARS-COV-2-S mAbs at the two concentrations to Spike protein expressing and parental Jurkat cells are summarized in Table 38. Of the 46 antibodies tested, 44 antibodies bound specifically to Jurkat/spike cells (Jurkat/Tet3G/hCD20/Tet-On 3G Inducible SARS-CoV-2 Spike Protein High Sorted cells) with a ratio to the parental cells of 4 or higher at either concentration. At 0.2nM, the ratios of the binding signals on Jurkat/spike cells to the parental cells ranged from 4 to 36, and at 5 nM the ratio ranged from 4 to 63. Although the two antibodies (mAb10998 and mAb11002) displayed weak binding to Jurkat/spike cells with binding ratio to the parental cells less than 4, at 5 nM the binding signals were higher on Jurket/spike than on the parental cells. An irrelevant IgG1 isotype antibody showed minimal binding, as expected.  
Table 38: Specificity of anti-SARS-CoV-2-S antibodies binding to spike protein-expressing Jurkat cells vs parental cells by electrochemiluminescence
Figure imgf000218_0001
 
Figure imgf000219_0001
 
Figure imgf000220_0001
***************** [000258] All references cited herein are incorporated by reference to the same extent as if each individual publication, database entry (e.g., Genbank sequences or GeneID entries), patent application, or patent, was specifically and individually indicated to be incorporated by reference. This statement of incorporation by reference is intended by Applicants to relate to each and every individual publication, database entry (e.g., Genbank sequences or GeneID entries), patent application, or patent identified even if such citation is not immediately adjacent to a dedicated statement of incorporation by reference. The inclusion of dedicated statements of incorporation by reference, if any, within the specification does not in any way weaken this general statement of incorporation by reference. Citation of the references herein is not intended as an admission that the reference is pertinent prior art, nor does it constitute any admission as to the contents or date of these publications or documents.  

Claims

We claim: 1. An isolated antibody or antigen-binding fragment thereof that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody or antigen-binding fragment comprises three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 210.
2. The isolated antibody or antigen-binding fragment thereof of claim 1, wherein HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 204, HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 206, HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 208, LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 212, LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 55, and LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 214.
3. The isolated antibody or antigen-binding fragment thereof of claim 2, comprising an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202.
4. The isolated antibody or antigen-binding fragment thereof of claim 2, comprising an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210.
5. The isolated antibody or antigen-binding fragment thereof of claim 2, comprising an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210.
6. An isolated antibody that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises an immunoglobulin constant region, three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR)  
comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 210.
7. The isolated antibody of claim 6, wherein HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 204, HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 206, HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 208, LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 212, LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 55, and LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 214.
8. The isolated antibody of claim 6, comprising an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 210.
9. The isolated antibody of claim 6, wherein said isolated antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 216 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 218.
10. The isolated antibody of claim 6, wherein said immunoglobulin constant region is an IgG1 constant region.
11. The isolated antibody of claim 6 which is a recombinant antibody.
12. The isolated antibody of claim 6 which is multispecific.
13. A pharmaceutical composition comprising the isolated antibody of claim 6 and a pharmaceutically acceptable carrier or diluent.  
14. The pharmaceutical composition of claim 13, further comprising a second therapeutic agent.
15. The pharmaceutical composition of claim 14, wherein the second therapeutic agent is selected from the group consisting of: a second antibody, or an antigen-binding fragment thereof, that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, an anti-inflammatory agent, an antimalarial agent, and an antibody or antigen- binding fragment thereof that binds TMPRSS2.
16. The pharmaceutical composition of claim 14, wherein the second therapeutic agent is a second antibody, or an antigen-binding fragment thereof, that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832.
17. The pharmaceutical composition of claim 16, wherein the second antibody or antigen-binding fragment thereof comprises three heavy chain CDRs (HCDR1, HCDR2 and HCDR3) contained within an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 640, and three light chain CDRs (LCDR1, LCDR2 and LCDR3) contained within an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 646.
18. The pharmaceutical composition of claim 17, wherein the second antibody or antigen-binding fragment thereof comprises: HCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 642; HCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 499; HCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 644; LCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 648; LCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 650; and LCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 652.
19. The pharmaceutical composition of claim 18, wherein the second antibody or antigen-binding fragment thereof comprises an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 640 and an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 646.  
20. The pharmaceutical composition of claim 19, wherein the second antibody or antigen-binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 654 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 656.
21. An isolated antibody or antigen-binding fragment thereof that binds a SARS- CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody or antigen-binding fragment comprises three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 640, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 646.
22. The isolated antibody or antigen-binding fragment thereof of claim 21, wherein HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 642, HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 499, HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 644, LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 648, LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 650, and LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 652.
23. The isolated antibody or antigen-binding fragment thereof of claim 21, comprising an HCVR that comprises an amino acid sequence set forth in SEQ ID NO: 640.
24. The isolated antibody or antigen-binding fragment thereof of claim 21, comprising an LCVR that comprises an amino acid sequence set forth in SEQ ID NO: 646.
25. The isolated antibody or antigen-binding fragment thereof of claim 21, comprising an HCVR that comprises an amino acid sequence set forth in SEQ ID NO: 640 and an LCVR that comprises an amino acid sequence set forth in SEQ ID NO: 646.  
26. An isolated antibody that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, wherein said isolated antibody comprises an immunoglobulin constant region, three heavy chain complementarity determining regions (CDRs) (HCDR1, HCDR2 and HCDR3) contained within a heavy chain variable region (HCVR) comprising the amino acid sequence set forth in SEQ ID NO: 640, and three light chain complementarity determining regions (CDRs) (LCDR1, LCDR2 and LCDR3) contained within a light chain variable region (LCVR) comprising the amino acid sequence set forth in SEQ ID NO: 646.
27. The isolated antibody of claim 26, wherein HCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 642, HCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 499, HCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 644, LCDR1 comprises the amino acid sequence set forth in SEQ ID NO: 648, LCDR2 comprises the amino acid sequence set forth in SEQ ID NO: 650, and LCDR3 comprises the amino acid sequence set forth in SEQ ID NO: 652.
28. The isolated antibody of claim 26, comprising an HCVR that comprises the amino acid sequence set forth in SEQ ID NO: 640 and an LCVR that comprises the amino acid sequence set forth in SEQ ID NO: 646.
29. The isolated antibody of claim 26, wherein said isolated antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 654 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 656.
30. The isolated antibody of claim 26, wherein said immunoglobulin constant region is an IgG1 constant region.
31. The isolated antibody of claim 26 which is a recombinant antibody.
32. The isolated antibody of claim 26 which is multispecific.  
33. A pharmaceutical composition comprising the isolated antibody of claim 26 and a pharmaceutically acceptable carrier or diluent.
34. The pharmaceutical composition of claim 33, further comprising a second therapeutic agent.
35. The pharmaceutical composition of claim 34, wherein the second therapeutic agent is selected from the group consisting of: a second antibody, or an antigen-binding fragment thereof, that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832, an anti-inflammatory agent, an antimalarial agent, and an antibody or antigen- binding fragment thereof that binds TMPRSS2.
36. The pharmaceutical composition of claim 34, wherein the second therapeutic agent is a second antibody, or an antigen-binding fragment thereof, that binds a SARS-CoV-2 spike protein comprising the amino acid sequence set forth in SEQ ID NO: 832.
37. The pharmaceutical composition of claim 36, wherein the second antibody or antigen-binding fragment thereof comprises three heavy chain CDRs (HCDR1, HCDR2 and HCDR3) contained within an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 202, and three light chain CDRs (LCDR1, LCDR2 and LCDR3) contained within an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 210.
38. The pharmaceutical composition of claim 37, wherein the second antibody or antigen-binding fragment thereof comprises: HCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 204; HCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 206; HCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 208 LCDR1, comprising the amino acid sequence set forth in SEQ ID NO: 212; LCDR2, comprising the amino acid sequence set forth in SEQ ID NO: 55; and LCDR3, comprising the amino acid sequence set forth in SEQ ID NO: 214.  
39. The pharmaceutical composition of claim 38, wherein the second antibody or antigen-binding fragment thereof comprises an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 202 and an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 210.
40. The pharmaceutical composition of claim 39, wherein the second antibody or antigen-binding fragment thereof comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 216 and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 218.
41. An isolated recombinant antibody or antigen-binding fragment thereof that specifically binds to a coronavirus spike protein (CoV-S), wherein the antibody has one or more of the following characteristics: (a) binds to CoV-S with an EC50 of less than about 10-9 M; (b) demonstrates an increase in survival in a coronavirus-infected animal after administration to said coronavirus-infected animal, as compared to a comparable coronavirus- infected animal without said administration; and/or (c) comprises three heavy chain complementarity determining regions (CDRs) (CDR- H1, CDR-H2, and CDR-H3) contained within a heavy chain variable region (HCVR) comprising an amino acid sequence having at least about 90% sequence identity to an HCVR of Table 1; and three light chain CDRs (CDR-L1, CDR-L2, and CDR-L3) contained within a light chain variable region (LCVR) comprising an amino acid sequence having at least about 90% sequence identity to an LCVR Table 1.
42. The antibody or antigen-binding fragment of claim 41, comprising: (a) an immunoglobulin heavy chain variable region comprising the CDR-H1, CDR- H2, and CDR-H3 of an antibody of Table 1; and/or (b) an immunoglobulin light chain variable region comprising the CDR-L1, CDR-L2, and CDR-L3 of an antibody of Table 1.
43. The antibody or antigen-binding fragment of claim 41 or 42, comprising:  
(a) a heavy chain immunoglobulin variable region comprising an amino acid sequence having at least 90% amino acid sequence identity to an HCVR sequence of Table 1; and/or (b) a light chain immunoglobulin variable region comprising an amino acid sequence having at least 90% amino acid sequence identity to an LCVR sequence of Table 1.
44. The antibody or antigen-binding fragment of any one of claims 41-43, wherein said antibody or antigen-binding fragment comprises the CDR-H1, CDR-H2, CDR-H3, CDR-L1, CDR-L2, and CDR-L3 of a single antibody of Table 1.
45. The antibody or antigen-binding fragment of any one of claims 41-44, comprising an immunoglobulin that comprises the HCVR and the LCVR of a single antibody of Table 1.
46. An antigen-binding protein that competes with the antibody or antigen- binding fragment of any one of claims 41-45 for binding to CoV-S.
47. An antigen-binding protein that binds to the same epitope as, or to an overlapping epitope on, CoV-S as the antibody or antigen-binding fragment of any one of claims 41-46.
48. The antibody or antigen-binding fragment of any one of claims 41-47 which is multispecific.
49. The antibody or antigen-binding fragment of any one of claims 41-48 which comprises one or more of the following properties: (a) inhibits growth of coronavirus; (b) binds to the surface of a coronavirus; (c) limits spread of coronavirus infection of cells in vitro; and (d) protects mice engineered to express the human ACE2 or TMPRSS2 protein from death and/or weight loss caused by coronavirus infection.  
50. The antibody or antigen-binding fragment of any one of claims 41-49, wherein said CoV-S is SARS-CoV-2-S.
51. A complex comprising an antibody or antigen-binding fragment of any one of claims 41-50 bound to a CoV-S polypeptide.
52. The complex of claim 51, wherein said CoV-S is SARS-CoV-2-S.
53. A method for making an antibody or antigen-binding fragment of any one of claims 41-50, comprising: (a) introducing into a host cell one or more polynucleotides encoding said antibody or antigen-binding fragment; (b) culturing the host cell under conditions favorable to expression of the one or more polynucleotides; and (c) optionally, isolating the antibody or antigen-binding fragment from the host cell and/or a medium in which the host cell is grown.
54. The method of claim 53, wherein the host cell is a Chinese hamster ovary cell.
55. An antibody or antigen-binding fragment which is a product of the method of claim 53 or 54.
56. A polypeptide comprising: (a) CDR-H1, CDR-H2, and CDR-H3 of an HCVR domain of an antibody or antigen- binding fragment that comprises an HCVR amino acid sequence set forth in Table 1; or (b) CDR-L1, CDR-L2, and CDR-L3 of an LCVR domain of an immunoglobulin chain that comprises an LCVR amino acid sequence set forth in Table 1.
57. A polynucleotide encoding the polypeptide of claim 56.
58. A vector comprising the polynucleotide of claim 57.  
59. A host cell comprising the antibody or antigen-binding fragment of any one of claims 41-50, or polypeptide or polynucleotide or vector of any one of claims 55-58.
60. A composition or kit comprising the antibody or antigen-binding fragment of any one of claims 41-50 and 55 in association with a further therapeutic agent.
61. A pharmaceutical composition comprising the antigen-binding protein of any one of claims 41-50 and 55 and pharmaceutically acceptable carrier and, optionally, a further therapeutic agent.
62. The composition or kit of claim 60 or 61 in association with a further therapeutic agent which is an anti-viral drug or a vaccine.
63. The composition or kit of any one of claims 60-62 wherein the further therapeutic agent is selected from the group consisting of: an anti-inflammatory agent, an antimalarial agent, an antibody or antigen-binding fragment thereof that specifically binds TMPRSS2, and an antibody or antigen-binding fragment thereof that specifically binds to CoV- S.
64. The composition or kit of claim 63, wherein said antimalarial agent is chloroquine or hydroxychloroquine.
65. The composition or kit of claim 63, wherein said anti-inflammatory agent is an antibody.
66. The composition or kit of claim 65, wherein said antibody is sarilumab, tocilizumab, or gimsilumab.  
67. The composition or kit of claim 63, wherein the antibody or antigen-binding fragment comprises the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences of a second antibody of Table 1.
68. A vessel or injection device comprising the antigen-binding protein or composition of any one of claims 41-51, 55, and 60-63.
69. A method for treating or preventing infection with a coronavirus, in a subject in need thereof, comprising administering a therapeutically effective amount of antigen-binding protein of any one of claims 41-50 and 55.
70. The method of claim 68, wherein said coronavirus is selected from the group consisting of SARS-CoV-2, SARS-CoV, and MERS-CoV.
71. The method of claim 69 or 70, wherein the subject is administered one or more further therapeutic agents.
72. The method of claim 71, wherein the one or more further therapeutic agents is an anti-viral drug or a vaccine.
73. The method of claim 71, wherein the one or more further therapeutic agents is selected from the group consisting of: an anti-inflammatory agent, an antimalarial agent, an antibody or antigen-binding fragment thereof that specifically binds TMPRSS2, and an antibody or antigen-binding fragment thereof that specifically binds to CoV-S.
74. The method of claim 73, wherein said antimalarial agent is chloroquine or hydroxychloroquine.
75. The method of claim 73, wherein said anti-inflammatory agent is an antibody.  
76. The method of claim 75, wherein said antibody is sarilumab, tocilizumab, or gimsilumab.
77. The method of claim 73, wherein the antibody or antigen-binding fragment comprises the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences of a second antibody of Table 1.
78. A method for administering an antibody or antigen-binding fragment of any one of claims 41-50 and 55 into the body of a subject comprising injecting the antibody or antigen-binding fragment into the body of the subject.
79. The method of claim 78, wherein the antibody or antigen-binding fragment is injected into the body of the subject subcutaneously, intravenously or intramuscularly.
80. The antibody or antigen-binding fragment, composition, kit, complex, polypeptide, polynucleotide, vector, cell, or method of any one of claims 41-79, wherein said antibody or antigen-binding binding fragment comprises a VH3-66 or Vk1-33 variable domain sequence.  
PCT/US2020/039707 2020-04-02 2020-06-25 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments WO2021045836A1 (en)

Priority Applications (31)

Application Number Priority Date Filing Date Title
JP2021518800A JP7116256B1 (en) 2020-04-02 2020-06-25 Anti-SARS-COV-2-Spike Glycoprotein Antibodies and Antigen-Binding Fragments
BR122023004078-3A BR122023004078B1 (en) 2020-04-02 2020-06-25 PHARMACEUTICAL COMPOSITION, ITS USE AND METHOD FOR MANUFACTURING AN ANTIBODY OR ANTIGEN BINDING FRAGMENT THEREOF
CR20220552A CR20220552A (en) 2020-04-02 2020-06-25 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
KR1020217013280A KR20210134300A (en) 2020-04-02 2020-06-25 Anti-SARS-COV-2 Spike Glycoprotein Antibodies and Antigen-Binding Fragments
CA3115553A CA3115553C (en) 2020-04-02 2020-06-25 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
PE2022002139A PE20221893A1 (en) 2020-04-02 2020-06-25 ANTIBODIES AGAINST SARS-COV-2 SPICLE GLYCOPROTEIN AND ANTIGEN-BINDING FRAGMENTS
AU2020340881A AU2020340881A1 (en) 2020-04-02 2020-06-25 Anti-SARS-CoV-2-spike glycoprotein antibodies and antigen-binding fragments
MYPI2021002018A MY197648A (en) 2020-04-02 2020-06-25 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
SG11202103404PA SG11202103404PA (en) 2020-04-02 2020-06-25 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
EA202190836A EA202190836A1 (en) 2020-06-04 2020-06-25 ANTIBODIES AND ANTIGEN-BINDING FRAGMENTS AGAINST SARS-COV-2 SPIKE GLYCOPROTEIN
BR112020015534-9A BR112020015534B1 (en) 2020-04-02 2020-06-25 ISOLATED ANTIBODY OR ANTIGEN-BINDING FRAGMENT THEREOF, ITS MANUFACTURING METHOD, ITS USES, AS WELL AS PHARMACEUTICAL COMPOSITION
MX2021004130A MX2021004130A (en) 2020-04-02 2020-06-25 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments.
CN202080005771.3A CN113766928A (en) 2020-04-02 2020-06-25 anti-SARS-COV-2 fiber process glycoprotein antibody and antigen binding fragment
LTEP20186667.0T LT3889177T (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
FIEP20186667.0T FI3889177T3 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
ES20186667T ES2939895T3 (en) 2020-04-02 2020-07-20 Anti-SARS-CoV-2 spike glycoprotein antibodies and antigen-binding fragments
SI202030145T SI3889177T1 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
MA54405A MA54405B1 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2 spike glycoprotein antibodies and antigen-binding fragments
HUE20186667A HUE061425T2 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
PT201866670T PT3889177T (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
EP20186667.0A EP3889177B1 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
PL20186667.0T PL3889177T3 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
HRP20230338TT HRP20230338T1 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
DK20186667.0T DK3889177T3 (en) 2020-04-02 2020-07-20 Anti-SARS-CoV-2 spike glycoprotein antibodies and antigen-binding fragments
EP22210926.6A EP4209507A1 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
RS20230266A RS64105B1 (en) 2020-04-02 2020-07-20 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
IL282060A IL282060A (en) 2020-04-02 2021-04-05 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
PH12021550793A PH12021550793A1 (en) 2020-04-02 2021-04-11 Anti-SARS-CoV-2-Spike Glycoprotein Antibodies and Antigen-Binding Fragments
CONC2021/0005072A CO2021005072A2 (en) 2020-04-02 2021-04-21 Anti-sars-cov-2 spike glycoprotein antibodies and antigen-binding fragments
JP2022119389A JP7411029B2 (en) 2020-04-02 2022-07-27 Anti-SARS-COV-2-spike glycoprotein antibody and antigen-binding fragment
JP2023215497A JP2024038003A (en) 2020-04-02 2023-12-21 Anti-SARS-COV-2-spike glycoprotein antibodies and antigen-binding fragments

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US202063004312P 2020-04-02 2020-04-02
US63/004,312 2020-04-02
US202063014687P 2020-04-23 2020-04-23
US63/014,687 2020-04-23
US202063025949P 2020-05-15 2020-05-15
US63/025,949 2020-05-15
US202063034865P 2020-06-04 2020-06-04
US63/034,865 2020-06-04

Publications (1)

Publication Number Publication Date
WO2021045836A1 true WO2021045836A1 (en) 2021-03-11

Family

ID=72614999

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/039707 WO2021045836A1 (en) 2020-04-02 2020-06-25 Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments

Country Status (29)

Country Link
US (5) US10787501B1 (en)
EP (2) EP4209507A1 (en)
JP (3) JP7116256B1 (en)
KR (1) KR20210134300A (en)
CN (1) CN113766928A (en)
AU (1) AU2020340881A1 (en)
CA (2) CA3192925A1 (en)
CL (2) CL2021000997A1 (en)
CO (1) CO2021005072A2 (en)
CR (1) CR20220552A (en)
DK (1) DK3889177T3 (en)
ES (1) ES2939895T3 (en)
FI (1) FI3889177T3 (en)
HR (1) HRP20230338T1 (en)
HU (1) HUE061425T2 (en)
IL (1) IL282060A (en)
LT (1) LT3889177T (en)
MA (1) MA54405B1 (en)
MX (1) MX2021004130A (en)
MY (1) MY197648A (en)
PE (1) PE20221893A1 (en)
PH (1) PH12021550793A1 (en)
PL (1) PL3889177T3 (en)
PT (1) PT3889177T (en)
RS (1) RS64105B1 (en)
SG (1) SG11202103404PA (en)
SI (1) SI3889177T1 (en)
TW (2) TWI785350B (en)
WO (1) WO2021045836A1 (en)

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3872091A1 (en) * 2020-02-26 2021-09-01 VIR Biotechnology, Inc. Antibodies against sars-cov-2 and methods of using the same
WO2021194965A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Superhuman anti-sars-cov-2 antibodies and uses thereof
WO2021194891A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from cr3022
WO2021190980A1 (en) * 2020-03-22 2021-09-30 Quadrucept Bio Limited Multimers for viral strain evolution
WO2021194896A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from 2ghw
WO2021194886A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from 3bgf
WO2021194985A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from 6nb6
WO2021194951A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from 2dd8
WO2021221136A1 (en) * 2020-05-01 2021-11-04 花王株式会社 Anti-sars-cov-2 antibody
WO2021226405A1 (en) * 2020-05-06 2021-11-11 International Aids Vaccine Initiative Inc. Covid-19 antibodies and uses thereof
WO2021242815A1 (en) * 2020-05-26 2021-12-02 Regeneron Pharmaceuticals, Inc. Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022054068A1 (en) * 2020-09-14 2022-03-17 Ramot At Tel-Aviv University Ltd. Antibodies for the prevention, treatment and detection of coronavirus infection
WO2022060916A1 (en) 2020-09-15 2022-03-24 Regenxbio Inc. Vectorized antibodies for anti-viral therapy
WO2022162587A1 (en) * 2021-01-27 2022-08-04 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
WO2022210830A1 (en) * 2021-03-30 2022-10-06 国立大学法人 富山大学 Anti-sars-cov-2 antibody
WO2022226079A1 (en) * 2021-04-20 2022-10-27 Inbios International, Inc. Neutralizing antibodies against sars-cov-2
WO2022232262A3 (en) * 2021-04-28 2022-12-01 The Trustees Of Columbia University In The City Of New York Antibodies for the treatment and prevention of covid-19 and emerging variants
WO2022263638A1 (en) * 2021-06-17 2022-12-22 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
WO2022197720A3 (en) * 2021-03-15 2022-12-22 Beth Israel Deaconess Medical Center, Inc. Compositions and methods for treating coronavirus infection
WO2022271258A1 (en) * 2021-06-25 2022-12-29 La Jolla Institute For Immunology Chimeric anti-sars-cov2 nucleoprotein antibodies
WO2023275538A1 (en) * 2021-06-28 2023-01-05 Diosynvax Ltd Beta-coronavirus vaccines
WO2023287875A1 (en) * 2021-07-14 2023-01-19 Regeneron Pharmaceuticals, Inc. Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
WO2023002944A1 (en) * 2021-07-19 2023-01-26 公立大学法人福島県立医科大学 Antibody capable of binding to novel coronavirus (sars-cov-2) and sars coronavirus (sars-cov)
WO2022271884A3 (en) * 2021-06-22 2023-01-26 Twist Bioscience Corporation Methods and compositions relating to covid antibody epitopes
WO2023004477A1 (en) * 2021-07-30 2023-02-02 The University Of Melbourne Neutralising antibodies and uses thereof
WO2023015232A1 (en) * 2021-08-04 2023-02-09 The Regents Of The University Of California Sars-cov-2 virus-like particles
WO2023285620A3 (en) * 2021-07-14 2023-03-09 Alchemab Therapeutics Ltd. Antibodies targeting sars-cov-2
WO2023035016A1 (en) * 2021-09-03 2023-03-09 The Uab Research Foundation Human neutralizing antibodies against sars-cov-2 spike s2 domain and uses thereof
WO2023033363A1 (en) * 2021-09-06 2023-03-09 국민대학교산학협력단 Bispecific antibody specifically binding to sars-cov-2
WO2023056482A1 (en) * 2021-10-01 2023-04-06 Academia Sinica Antibody specific to spike protein of sars-cov-2 and uses thereof
WO2023004431A3 (en) * 2021-07-23 2023-04-20 The Trustees Of Columbia University In The City Of New York Characterization of potent and broadly neutralizing monoclonal antibodies against sars-cov-2, its variants, and related coronaviruses and methods of use
WO2023084055A1 (en) * 2021-11-12 2023-05-19 Rq Biotechnology Limited Compositions
WO2023048656A3 (en) * 2021-09-24 2023-06-08 Chulalongkorn University Human monoclonal antibodies against the receptor-binding domain of sars-cov-2 spike protein
WO2023081434A3 (en) * 2021-11-07 2023-06-15 Regeneron Pharmaceuticals, Inc. Methods for treating or preventing sars-cov-2 infections and covid-19 with anti-sars-cov-2 spike glycoprotein antibodies
WO2023109419A1 (en) * 2021-12-14 2023-06-22 Beijing Acrobiosystems Biotechnology Co., Ltd A neutralizing antibody that can bind sars-cov-2 virus and its application
WO2023113094A1 (en) * 2021-12-16 2023-06-22 주식회사 씨티씨백 Covid-19 vaccine composition with increased immunogenicity
WO2023104904A3 (en) * 2021-12-08 2023-07-20 Genclis The sars-cov-2 and variants use two independent cell receptors to replicate
WO2023064435A3 (en) * 2021-10-15 2023-07-27 The Children's Medical Center Corporation Compositions and methods relating to sars-cov-2 neutralizing antibodies
WO2023122535A3 (en) * 2021-12-20 2023-08-03 DNARx Nucleic acid expression using subcutaneous administration
WO2023145859A1 (en) * 2022-01-28 2023-08-03 株式会社イーベック HUMAN NEUTRALIZING ANTIBODY AGAINST SARS-CoV-2 HAVING BREADTH TO VARIANT STRAINS, AND ANTIGEN-BINDING FRAGMENT THEREOF
WO2023023183A3 (en) * 2021-08-17 2023-08-17 Twist Bioscience Corporation Sars-cov-2 antibodies and related compositions and methods of use
US11732030B2 (en) 2020-04-02 2023-08-22 Regeneron Pharmaceuticals, Inc. Anti-SARS-CoV-2-spike glycoprotein antibodies and antigen-binding fragments
WO2023156187A1 (en) * 2022-02-16 2023-08-24 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts High affinity antibodies against the sars-cov-2 receptor binding domain
JP7343739B2 (en) 2021-06-04 2023-09-13 武漢生物制品研究所有限責任公司 Monoclonal antibody 20D8 against SARS-CoV-2 epidemic variant
WO2023064717A3 (en) * 2021-10-08 2023-09-14 Augmenta Bioworks, Inc. Antibodies for sars-cov-2 and uses thereof
WO2023178182A1 (en) * 2022-03-16 2023-09-21 10X Genomics, Inc. Compositions and methods for detection and treatment of coronavirus infection
WO2023190851A1 (en) * 2022-03-31 2023-10-05 公立大学法人福島県立医科大学 Antibody binding to novel coronavirus (sars-cov-2)
WO2023190852A1 (en) * 2022-03-31 2023-10-05 公立大学法人福島県立医科大学 Antibody capable of binding to new coronavirus (sars-cov-2)
US11918641B2 (en) 2020-05-08 2024-03-05 Academia Sinica Chimeric influenza vaccines
RU2817697C1 (en) * 2023-12-08 2024-04-18 Федеральное государственное бюджетное учреждение науки Институт молекулярной и клеточной биологии Сибирского отделения Российской академии наук (ИМКБ СО РАН) MONOCLONAL ANTIBODY IC2 AND ITS ANTIGEN-BINDING FRAGMENT, SELECTIVELY BINDING RECEPTOR-BINDING DOMAIN OF SPIKE PROTEIN OF SARS-CoV-2 VIRUS, HAVING VIRUS-NEUTRALIZING ACTIVITY

Families Citing this family (133)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL292464A (en) 2019-11-12 2022-06-01 Regeneron Pharma Methods and systems for identifying, classifying, and/or ranking genetic sequences
JP2023510289A (en) * 2020-01-09 2023-03-13 ザ ユニバーシティー オブ クイーンズランド Self-assembling self-adjuvant system for vaccine delivery
AU2021230334A1 (en) * 2020-03-05 2022-09-29 New York Blood Center, Inc. Immunogenic compositions against severe acute respiratory syndrome coronavirus 2
US11365239B2 (en) * 2020-03-20 2022-06-21 Tsb Therapeutics (Beijing) Co., Ltd. Anti-SARS-COV-2 antibodies and uses thereof
CN111349150B (en) * 2020-03-24 2021-02-09 北京中科微盾生物科技有限责任公司 Polypeptide for inhibiting novel coronavirus and application thereof
WO2021198999A1 (en) * 2020-04-03 2021-10-07 Axon Neuroscience Se Epitope-based vaccines for treatment of coronavirus associated diseases
WO2021205022A1 (en) * 2020-04-09 2021-10-14 Valneva Austria Gmbh Improved methods of producing a lipidated protein
US20210331150A1 (en) * 2020-04-24 2021-10-28 Kevin D. Fialko Sars-cov-2 susceptibility detection
WO2021218947A1 (en) * 2020-04-28 2021-11-04 北京大学 Anti-novel coronavirus monoclonal antibody and application thereof
US20230168249A1 (en) * 2020-05-06 2023-06-01 Academia Sinica Recombinant antibodies, kits comprising the same, and uses thereof
WO2021222988A1 (en) * 2020-05-07 2021-11-11 Griffith University Cell entry-modulating agents and uses therefor
WO2021231659A1 (en) * 2020-05-12 2021-11-18 Sapphire Biotech, Inc. Neutralizing antibody testing and treatment
KR20230019166A (en) 2020-06-03 2023-02-07 리제너론 파아마슈티컬스, 인크. Methods for treating or preventing SARS-CoV-2 infection and COVID-19 using anti-SARS-CoV-2 spike glycoprotein antibodies
EP4176262A1 (en) 2020-07-02 2023-05-10 King Abdullah University Of Science And Technology Nanobody functionalized electrochemical transistors and methods of making and using thereof
US11740240B2 (en) * 2020-07-20 2023-08-29 Bio-Rad Laboratories, Inc. Immunoassay for SARS-CoV-2 neutralizing antibodies and materials therefor
WO2022027037A1 (en) 2020-07-27 2022-02-03 Single Cell Technology, Inc. Anti-sars corona virus-2 spike protein antibodies
CN111978378B (en) * 2020-08-10 2022-02-01 武汉大学 SARS-CoV-2 antigen polypeptide and its application
EP4206224A1 (en) * 2020-08-26 2023-07-05 National University Corporation Kumamoto University Human antibody or antigen-binding fragment thereof against coronavirus spike protein
EP4211158A1 (en) * 2020-09-14 2023-07-19 Nanjing Vazyme Biotech Co., Ltd. Neutralizing antibodies against sars-cov-2
TWI796597B (en) * 2020-09-21 2023-03-21 長庚大學 Method against coronavirus infection with water-extracted product of perilla frutescens
CN116023478A (en) * 2020-09-30 2023-04-28 上海市公共卫生临床中心 Neutralizing antibodies or antigen binding fragments thereof for coronaviruses
CN116419971A (en) * 2020-09-30 2023-07-11 南京金斯瑞生物科技有限公司 Monoclonal antibody against SARS-CoV-2 spike protein outer region and application thereof
WO2022068844A1 (en) * 2020-09-30 2022-04-07 Vazyme Biotech Co., Ltd. Neutralizing antibody against sars-cov-2
EP4221749A1 (en) * 2020-09-30 2023-08-09 The Research Foundation for The State University of New York Particle based formulations of sars-cov-2 receptor binding domain
CN112159469B (en) * 2020-09-30 2022-08-02 上海市公共卫生临床中心 Antibodies or antigen-binding fragments thereof to coronaviruses
CN112125973B (en) * 2020-09-30 2022-02-01 上海市公共卫生临床中心 Specific antibodies or antigen-binding fragments thereof for coronaviruses
KR20230061462A (en) * 2020-10-02 2023-05-08 일라이 릴리 앤드 캄파니 Method for reducing host cell protein content in protein purification process
WO2022075921A1 (en) * 2020-10-08 2022-04-14 Hummingbird Bioscience Holdings Limited Sars-cov-2 spike protein antigen-binding molecules
WO2022079606A1 (en) * 2020-10-12 2022-04-21 Icosagen Cell Factory Oü Sars cov-2 neutralizing antibodies
CN112251414A (en) * 2020-10-12 2021-01-22 中国科学院苏州纳米技术与纳米仿生研究所 Hybridoma cell strain, preparation method and application thereof
CN112194711A (en) * 2020-10-15 2021-01-08 深圳市疾病预防控制中心(深圳市卫生检验中心、深圳市预防医学研究所) B cell linear epitope of novel coronavirus S protein, antibody, identification method and application
EP4229081A1 (en) * 2020-10-15 2023-08-23 The United States of America, as represented by The Secretary, Department of Health and Human Services Antibody specific for sars-cov-2 receptor binding domain and therapeutic methods
AU2021362007A1 (en) 2020-10-16 2023-06-22 Invisishield Technologies Ltd. Compositions for preventing or treating viral and other microbial infections
WO2022084817A1 (en) * 2020-10-19 2022-04-28 Lay Sciences Inc. Protein drugs for prevention and treatment of covid-19
US20230390383A1 (en) * 2020-10-20 2023-12-07 Duke University Replication incompetent influenza vaccine platform for foreign protein delivery
GB202016650D0 (en) * 2020-10-20 2020-12-02 Primer Design Ltd Kit and method
US20230398204A1 (en) * 2020-10-21 2023-12-14 La Jolla Institute For Immunology Coronavirus Spike Glycoprotein With Improved Expression and Stability
EP4232470A1 (en) * 2020-10-23 2023-08-30 Atreca, Inc. Antibodies to coronavirus sars-cov-2
KR20220054080A (en) * 2020-10-23 2022-05-02 주식회사 와이바이오로직스 ANTIBODY SPECIFICALLY BINDING TO SARS-CoV-2 SPIKE PROTEIN AND USES THEREOF
CN112409488B (en) * 2020-10-23 2022-07-01 中国科学院上海药物研究所 Monoclonal antibody aiming at various coronaviruses and application
WO2022093745A1 (en) * 2020-10-26 2022-05-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Single domain antibodies targeting sars coronavirus spike protein and uses thereof
US20240018191A1 (en) * 2020-10-27 2024-01-18 The University Of North Carolina At Chapel Hill Chimeric coronavirus s protein compositions and methods of use
CN112358533B (en) * 2020-10-30 2023-07-14 上海泽润生物科技有限公司 Recombinant spike protein and preparation method and application thereof
US11453705B2 (en) 2020-10-30 2022-09-27 Achelois Biopharma, Inc. Multivalent particles compositions and methods of use
CA3200263A1 (en) * 2020-10-30 2022-05-05 Adaptive Biotechnologies Corporation Anti-sars-cov-2 antigen antibodies and related compositions and methods
US11739137B2 (en) * 2020-10-30 2023-08-29 The Government Of The United States Of America, As Represented By The Secretary Of The Navy Single domain antibodies to SARS-CoV-2 nucleocapsid protein
US11872279B2 (en) 2020-10-30 2024-01-16 Massachusetts Institute Of Technology SARS-CoV-2 antigens and uses thereof
WO2022094622A1 (en) * 2020-11-02 2022-05-05 The University Of Chicago Polypeptides for detection and treatment of sars-cov-2
CN112098660B (en) * 2020-11-03 2021-02-09 北京百普赛斯生物科技股份有限公司 Novel coronavirus neutralizing antibody detection kit
IL302645A (en) * 2020-11-03 2023-07-01 The Wistrar Inst Of Anatomy And Biology Dna encoded nanoparticles and method of use thereof as a coronavirus disease 2019 (covid-19) vaccine
AR123997A1 (en) * 2020-11-04 2023-02-01 Univ Rockefeller NEUTRALIZING ANTIBODIES AGAINST SARS-CoV-2
TW202233233A (en) * 2020-11-06 2022-09-01 醣基生醫股份有限公司 Immune composition comprising antigen and glycoengineered antibody thereof
US20230417748A1 (en) * 2020-11-06 2023-12-28 Green Cross Corporation Coronavirus Spike Protein-Specific Antibody and Use Thereof
WO2022095045A1 (en) * 2020-11-09 2022-05-12 上海济煜医药科技有限公司 Sars-cov-2 antibody and application thereof
WO2022101839A1 (en) * 2020-11-12 2022-05-19 Cadila Healthcare Limited Anti-sars-cov-2 monoclonal antibodies and cocktail thereof
WO2022102744A1 (en) * 2020-11-13 2022-05-19 株式会社ハカレル Antibody against spike protein of sars-cov-2
CN112625136B (en) * 2020-11-18 2022-02-11 三优生物医药(上海)有限公司 Bispecific antibodies having neutralizing activity against coronaviruses and uses thereof
CA3200885A1 (en) * 2020-11-23 2022-05-27 Genentech, Inc. Methods for modulating host cell surface interactions with sars-cov-2
CN112409479B (en) * 2020-11-23 2022-04-26 中国疾病预防控制中心病毒病预防控制所 Humanized anti-neocoronavirus neutralizing antibody nCoV-121 and application thereof
CN112439058A (en) * 2020-11-25 2021-03-05 深圳市第二人民医院(深圳市转化医学研究院) Recombinant novel coronavirus nano vaccine method based on exosome as vector
WO2022115558A1 (en) * 2020-11-25 2022-06-02 Deciphera Pharmaceuticals, Llc Morpholino derivatives as vsp34 inhibitors for use in the treatment of a viral infection
CA3199988A1 (en) * 2020-11-25 2022-06-02 Daniel L. Flynn Pyridylpyridone derivatives as vsp34 inhibitors for use in the treatment of a viral infection
CN112442120A (en) * 2020-11-25 2021-03-05 苏州大学 Neutralizing antibody against SARS-COV-2 of severe acute respiratory syndrome type II coronavirus
WO2022115562A1 (en) * 2020-11-25 2022-06-02 Deciphera Pharmaceuticals, Llc Morpholino derivatives as vsp34 inhibitors for use in the treatment of a viral infection
WO2022112392A1 (en) 2020-11-26 2022-06-02 Memo Therapeutics Ag Anti-sars-cov-2 antibody molecules
EP4256080A1 (en) 2020-12-03 2023-10-11 Regeneron Pharmaceuticals, Inc. Methods of identifying subjects having an increased risk of developing a coronavirus infection and treatment thereof
US20240044896A1 (en) * 2020-12-09 2024-02-08 The University Of Chicago Methods and systems for detection and analysis of angiotensin binding antibodies
US20240035035A1 (en) * 2020-12-09 2024-02-01 Yale University Compositions and methods for treating, ameliorating, and/or preventing viral infections
WO2022122788A1 (en) * 2020-12-09 2022-06-16 Institute For Research In Biomedicine Multispecific antibodies against severe acute respiratory syndrome coronavirus 2
US11693011B2 (en) 2020-12-14 2023-07-04 Inbios International, Inc. Neutralizing antibody immunoassays
WO2022132887A1 (en) * 2020-12-15 2022-06-23 Board Of Regents, The University Of Texas System Human monoclonal antibodies targeting the sars-cov-2 spike protein
WO2022132904A1 (en) * 2020-12-17 2022-06-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Human monoclonal antibodies targeting sars-cov-2
WO2022139680A1 (en) * 2020-12-21 2022-06-30 Chugai Seiyaku Kabushiki Kaisha Sars-cov-2 binding molecules and uses thereof
US20240043506A1 (en) * 2020-12-23 2024-02-08 Garvan Institute Of Medical Research Sars-cov-2 antibodies
CN112794884B (en) * 2020-12-28 2022-02-18 中国科学院微生物研究所 Novel coronavirus protein, preparation method and neutralizing antibody detection kit
CN112326973B (en) * 2020-12-31 2021-03-23 北京金沃夫生物工程科技有限公司 Kit for detecting novel coronavirus antibody and application thereof
WO2022147290A1 (en) * 2020-12-31 2022-07-07 The Broad Institute, Inc. Cross-neutralizing sars-cov2 antibodies
WO2022150809A1 (en) * 2021-01-05 2022-07-14 Immunome, Inc. Antibody cocktail against sars-cov-2 spike protein
WO2022150660A1 (en) * 2021-01-08 2022-07-14 10X Genomics, Inc. Antigen-binding polypeptides specific for coronaviruses and uses thereof
WO2022159834A1 (en) * 2021-01-22 2022-07-28 La Jolla Institute For Immunology Chimeric anti-coronavirus spike protein antibodies
WO2022158497A1 (en) * 2021-01-22 2022-07-28 株式会社mAbProtein ANTIBODY AGAINST SPIKE PROTEIN OF SARS-CoV-2
CN113264998B (en) * 2021-01-28 2023-02-28 四川大学华西医院 Single-chain antibody of S1 protein on surface of anti-new coronavirus SARS-CoV-2 and application thereof
CN117440966A (en) * 2021-01-28 2024-01-23 塔勒姆治疗有限责任公司 anti-SARS-CoV-2 spike glycoprotein antibody and therapeutic use thereof
CN115109151A (en) * 2021-01-31 2022-09-27 中南大学湘雅医院 Novel coronavirus monoclonal antibody XY6 and application thereof
RU2771288C1 (en) * 2021-02-02 2022-04-29 федеральное государственное бюджетное учреждение "Национальный исследовательский центр эпидемиологии и микробиологии имени почетного академика Н.Ф. Гамалеи" Министерства здравоохранения Российской Федерации STRAIN OF HYBRID ANIMAL CELLS MUS MUSCULUS 2E1B5 - PRODUCER OF A MONOCLONAL ANTIBODY AGAINST THE RECEPROT-BINDING DOMAIN OF PROTEIN S OF THE SARS-CoV-2 VIRUS
WO2022167816A2 (en) * 2021-02-04 2022-08-11 Oxford University Innovation Limited Antibodies
US20240117011A1 (en) * 2021-02-09 2024-04-11 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Antibodies targeting the spike protein of coronaviruses
EP4291212A1 (en) 2021-02-15 2023-12-20 LivingMed Biotech S.R.L. Genetically clostridium modifiedstrains expressing recombinant antigens and uses thereof
EP4297786A1 (en) * 2021-02-23 2024-01-03 Pandion Operations, Inc. Pd-1 antibodies, polypeptides and uses thereof
WO2022182662A1 (en) 2021-02-23 2022-09-01 10X Genomics, Inc. Compositions and methods for mapping antigen-binding molecule affinity to antigen regions of interest
CN113009153B (en) * 2021-02-25 2023-12-15 山东莱博生物科技有限公司 New coronavirus neutralizing antibody detection kit based on magnetic particle chemiluminescence and application thereof
CN112679605B (en) * 2021-03-15 2021-07-09 安源医药科技(上海)有限公司 Antibodies or antigen binding fragments thereof against novel coronavirus nucleocapsid proteins and uses thereof
EP4063384A1 (en) * 2021-03-23 2022-09-28 Ceinge Biotecnologie Avanzate SCarl Human neutralizing antigen specific proteins for spike-rbd of sars-cov-2
CN113092776A (en) * 2021-03-24 2021-07-09 苏州携创生物技术有限公司 SARS-CoV-2 neutralizing antibody detection kit
WO2022204713A2 (en) * 2021-03-26 2022-09-29 Atreca, Inc. Antibodies to sars-cov-2
KR20220136945A (en) * 2021-04-01 2022-10-11 (주)셀트리온 SARS-CoV-2 NEUTRALIZING BINDING MOLECULE BINDING TO EPITOPES OF SARS-CoV-2 SPIKE PROTEIN
EP4321178A1 (en) * 2021-04-05 2024-02-14 Lg Chem, Ltd. Vaccine composition against coronavirus
EP4324848A1 (en) * 2021-04-16 2024-02-21 Korea University Research and Business Foundation Human antibody targeting covid-19 virus
AR125373A1 (en) * 2021-04-19 2023-07-12 Bharat Serums & Vaccines Ltd POLYCLONAL ANTIBODIES AGAINST SARS-CoV-2 AND ITS APPLICATIONS
EP4326320A1 (en) * 2021-04-20 2024-02-28 D4 Labs, Llc Adjuvanted vaccine composition and methods
KR102426782B1 (en) * 2021-04-27 2022-07-28 국민대학교산학협력단 Diagnostic composition or Kit comprising SARS-CoV-2 RBD antigen specific antibody
WO2022231320A1 (en) * 2021-04-27 2022-11-03 국민대학교산학협력단 Antibody binding specifically to sars-cov-2 s protein or antigen-binding fragment thereof, and uses thereof
WO2022235678A1 (en) * 2021-05-03 2022-11-10 Ohio State Innovation Foundation Human anti-coronavirus peptide vaccines and methods of their use
WO2022235960A1 (en) * 2021-05-06 2022-11-10 Sorrento Therapeutics, Inc. Neutralizing antibodies that bind variant sars-cov-2 spike proteins
CN112981011B (en) * 2021-05-12 2021-08-17 广东凯普生物科技股份有限公司 Primer composition for detecting SARS-CoV-2 and its application
WO2022241057A1 (en) * 2021-05-12 2022-11-17 Applied Biomedical Science Institute Binding polypeptides against sars cov-2 and uses thereof
EP4089112A1 (en) * 2021-05-14 2022-11-16 Ustav organicke chemie a biochemie AV CR, v.v.i. Antibody binding to rbd of spike protein of sars-cov-2 and a method for quantifying protective antibodies against sars-cov-2
GB202107057D0 (en) 2021-05-18 2021-06-30 Univ York Glycosylation method
WO2022251403A1 (en) * 2021-05-28 2022-12-01 Icahn School Of Medicine At Mount Sinai Sars-cov-2 antibodies and uses thereof
CN113416245A (en) * 2021-06-15 2021-09-21 北京华大蛋白质研发中心有限公司 Neutralizing antibody capable of combining SARS-CoV-2 virus RBD protein and application thereof
CN113584223B (en) * 2021-06-28 2024-01-12 中国人民解放军疾病预防控制中心 Identification method of D614G mutation in SARS-CoV-2 based on CRISPR-Cas12a
WO2023283134A1 (en) 2021-07-05 2023-01-12 Regeneron Pharmaceuticals, Inc. Utilization of antibodies to shape antibody responses to an antigen
CN113621651A (en) * 2021-07-19 2021-11-09 南昌五元生物科技有限公司 Cell syncytium lesion model based on S-TET protein expression system and preparation method thereof
GB202110391D0 (en) 2021-07-19 2021-09-01 Tissue Click Ltd A detection kit and methods of detection of infectious agents
US11724077B2 (en) * 2021-07-28 2023-08-15 Subhash Dhawan Therapeutic swabs for treating upper respiratory infections
WO2023008463A1 (en) * 2021-07-28 2023-02-02 株式会社イーベック Neutralizing human antibody against wild type strain and mutant strain of sars-cov-2 and antigen-binding fragment thereof
CN113512113A (en) * 2021-08-03 2021-10-19 浙江大学医学院附属第一医院 Humanized broad-spectrum high-neutralization-activity anti-novel coronavirus monoclonal antibody and application thereof
WO2023026207A1 (en) * 2021-08-25 2023-03-02 Translational Health Science And Technology Institute Potently neutralizing novel human monoclonal antibodies against sars-cov-2 (covid-19)
WO2023070029A1 (en) * 2021-10-20 2023-04-27 Arizona Board Of Regents On Behalf Of Arizona State University DNA ORIGAMI SUBUNIT VACCINE FOR PREVENTION OF SARS-CoV-2 VARIANT INFECTION
WO2023091920A1 (en) * 2021-11-16 2023-05-25 The University Of Chicago Polypeptides for detection and treatment of coronavirus infection
WO2023099688A1 (en) * 2021-12-01 2023-06-08 Universität Zu Köln Neutralizing antibodies against sars-related coronavirus
EP4190810A1 (en) * 2021-12-01 2023-06-07 Universität zu Köln Neutralizing antibodies against sars-related coronavirus
CN113862286B (en) * 2021-12-03 2022-03-04 艾棣维欣(苏州)生物制药有限公司 DNA molecule for coding SARS-COV-2 virus C.37 mutant strain antigen, DNA vaccine and application
US20230279080A1 (en) * 2021-12-27 2023-09-07 Academia Sinica Antibody specific to spike protein of sars-cov-2 and uses thereof
WO2023137443A1 (en) 2022-01-14 2023-07-20 Regeneron Pharmaceuticals, Inc. Verrucarin a derivatives and antibody drug conjugates thereof
TW202346329A (en) * 2022-01-26 2023-12-01 中央研究院 Antibody specific to coronaviruses and uses thereof
WO2023152188A1 (en) * 2022-02-11 2023-08-17 Attana Ab Analytical method
TW202342510A (en) * 2022-02-18 2023-11-01 英商Rq生物科技有限公司 Antibodies
CN114231497B (en) * 2022-02-24 2022-05-20 广州伯尼兹生物科技有限公司 Monoclonal antibody hybridoma cell line expressing SARS-CoV-2S 1 protein and neutralizing active antibody
CN115287265B (en) * 2022-07-12 2023-05-23 四川大学华西医院 Immune control model for inducing rhesus monkey resistant new crown mutant by using multipotential active preparation
EP4335870A1 (en) * 2022-09-06 2024-03-13 NantCell, Inc. Peptide therapeutics against sars-cov-2 spike protein
EP4349860A1 (en) 2022-10-06 2024-04-10 Fundació Institut Hospital Del Mar D'Investigacions Mèdiques (IMIM) Anti-sars-cov-2 antibodies
WO2024077288A1 (en) * 2022-10-07 2024-04-11 Medigen Vaccine Biologics Corporation Immunogenic compositions against the omicron variant of severe acute respiratory syndrome coronavirus 2 (sars-cov-2)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005060520A2 (en) * 2003-11-25 2005-07-07 Dana-Farber Cancer Institute, Inc. ANTIBODIES AGAINST SARS-CoV AND METHODS OF USE THEREOF
US20060240551A1 (en) * 2004-06-02 2006-10-26 Shibo Jiang Neutralizing monoclonal antibodies against severe acute respiratory syndrome-associated coronavirus
US20170096455A1 (en) * 2014-03-20 2017-04-06 The University Of North Carolina At Chapel Hill Methods and compositions for chimeric coronavirus spike proteins
CN111088283A (en) * 2020-03-20 2020-05-01 苏州奥特铭医药科技有限公司 mVSV viral vector, viral vector vaccine thereof and mVSV-mediated novel coronary pneumonia vaccine

Family Cites Families (199)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4740461A (en) 1983-12-27 1988-04-26 Genetics Institute, Inc. Vectors and methods for transformation of eucaryotic cells
US5693489A (en) 1984-03-30 1997-12-02 Associated Universities, Inc. Cloning and expression of the gene for bacteriophage T7 RNA polymerase
US4952496A (en) 1984-03-30 1990-08-28 Associated Universities, Inc. Cloning and expression of the gene for bacteriophage T7 RNA polymerase
US4959455A (en) 1986-07-14 1990-09-25 Genetics Institute, Inc. Primate hematopoietic growth factors IL-3 and pharmaceutical compositions
US4912040A (en) 1986-11-14 1990-03-27 Genetics Institute, Inc. Eucaryotic expression system
AU2515992A (en) 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
MX2008014804A (en) 2006-06-02 2009-01-27 Regeneron Pharma High affinity antibodies to human il-6 receptor.
US8629244B2 (en) 2006-08-18 2014-01-14 Ablynx N.V. Interleukin-6 receptor binding polypeptides
JP6034023B2 (en) 2008-05-16 2016-11-30 アブリンクス エン.ヴェー. Amino acid sequences directed to CXCR4 and other GPCRs and compounds containing the same
US20150203591A1 (en) 2012-08-02 2015-07-23 Regeneron Pharmaceuticals, Inc. Mutivalent antigen-binding proteins
JO3701B1 (en) 2014-05-23 2021-01-31 Regeneron Pharma Human antibodies to middle east respiratory syndrome – coronavirus spike protein
KR102369014B1 (en) 2016-08-16 2022-03-02 리제너론 파아마슈티컬스, 인크. Methods for quantifying individual antibodies from mixtures
LT3638698T (en) 2018-01-26 2021-04-12 Regeneron Pharmaceuticals, Inc. Anti-tmprss2 antibodies and antigen-binding fragments
CN114096560A (en) 2019-07-03 2022-02-25 默克专利股份公司 Purification of sugar forms
US20220273718A1 (en) 2019-08-02 2022-09-01 University Of Virginia Patent Foundation Bispecific antibody targeting of t regulatory cells for treatment of inflammatory conditions
CA3152009A1 (en) 2019-08-22 2021-02-25 Cidara Therapeutics, Inc. Variant fc domains and uses thereof
EP3795163A1 (en) 2019-09-23 2021-03-24 The Ceutics Company GesmbH A topical plant extract formulation comprising urtica dioica and vitamin a
US20210093709A1 (en) 2019-09-27 2021-04-01 George Mason University Use of shrek proteins for inactivating viral infectivity and to produce live-attenuated vaccines against viruses
CN111748571B (en) 2019-10-11 2022-05-03 浙江禾霖生物科技有限公司 Method for engineering bacillus subtilis into multifunctional stable platform for producing nano antibody
IL292464A (en) 2019-11-12 2022-06-01 Regeneron Pharma Methods and systems for identifying, classifying, and/or ranking genetic sequences
WO2021148884A1 (en) 2020-01-24 2021-07-29 Tychan Pte. Ltd. Anti-wuhan coronavirus antibodies
NL2030835B1 (en) * 2020-01-24 2022-12-29 Aim Immunotech Inc Methods, compositions, and vaccinces for treating a virus infection
CN113201068A (en) 2020-02-03 2021-08-03 深圳市雅臣智能生物工程有限公司 Gene recombination anti 2019-nCoV and other coronavirus IgY and small molecule antibody and application thereof
IL295310A (en) 2020-02-11 2022-10-01 Univ Vanderbilt Human monoclonal antibodies to severe acute respiratory syndrome coronavirus 2 (sars-cov- 2)
CN113248579B (en) 2020-02-12 2022-10-18 重庆医科大学 Novel coronavirus (2019-ncov) epitope, antibody and application thereof
CN111303254A (en) 2020-02-20 2020-06-19 北京新创生物工程有限公司 Novel coronavirus (SARS-CoV-2) antigen detection kit
DE202020105116U1 (en) 2020-02-20 2020-10-06 Charité - Universitätsmedizin Berlin Reagents and uses in diagnosing SARS-CoV-2 infection
US20210260201A1 (en) 2020-02-21 2021-08-26 Physis Biotechnologies, Llc Extracellular vesicles for the treatment and prevention of infections and other diseases
WO2021168483A2 (en) * 2020-02-21 2021-08-26 Florida State University Research Foundation, Inc. Treatment of human coronavirus infections using alpha-glucosidase glycoprotein processing inhibitors
CN113292650B (en) 2020-02-24 2022-08-12 中国科学院微生物研究所 Human monoclonal antibodies to novel coronaviruses and uses thereof
CN113292649B (en) 2020-02-24 2022-08-12 中国科学院微生物研究所 Human monoclonal antibodies to novel coronaviruses and uses thereof
CN111333704B (en) 2020-02-24 2021-01-12 军事科学院军事医学研究院微生物流行病研究所 Novel coronavirus COVID-19 vaccine, preparation method and application thereof
CA3172878A1 (en) 2020-02-25 2021-09-02 Ligandal, Inc. Identification of biomimetic viral peptides and uses thereof
HUE062777T2 (en) 2020-02-26 2023-12-28 Vir Biotechnology Inc Antibodies against sars-cov-2
CN113307865B (en) 2020-02-26 2022-12-13 复旦大学 Fully human single domain antibody of novel coronavirus and application
EP3885361A1 (en) 2020-02-27 2021-09-29 Macrofarm Srl Monoclonal-type synthetic antibodies obtained by molecular imprinting and their use for the treatment and preventive medicine of covid-19
CN113354731A (en) 2020-03-02 2021-09-07 中国科学院微生物研究所 Human monoclonal antibodies to coronaviruses and uses thereof
CN111333722A (en) 2020-03-03 2020-06-26 江苏省疾病预防控制中心(江苏省公共卫生研究院) SARS-CoV-2 inhibitor and its application
WO2021178637A1 (en) 2020-03-05 2021-09-10 Iowa State University Research Foundation, Inc. IMMUNOGENIC AND VACCINE COMPOSITIONS AGAINST SARS-CoV-2
CN111285933A (en) 2020-03-09 2020-06-16 四川省人民医院 Novel coronavirus antigen colloidal gold diagnostic kit
CN116096742A (en) 2020-03-09 2023-05-09 雅伯希勒拉生物公司 Anti-coronavirus antibodies and methods of use
US20210277093A1 (en) 2020-03-09 2021-09-09 Adma Biologics, Inc. Immunotherapeutic compositions and methods of production for coronavirus
CN111420048B (en) 2020-03-11 2023-09-19 中国人民解放军第四军医大学 Application of anti-BASIGAN humanized antibody in preparation of medicine for treating novel coronavirus pneumonia
WO2021183790A1 (en) 2020-03-12 2021-09-16 Rigel Pharmaceuticals, Inc. Method for treatment of covid-19-associated conditions
US10822379B1 (en) 2020-03-12 2020-11-03 University of Pittsburgh—of the Commonwealth System of Higher Education Molecules that bind to SARS-CoV-2
GB202003632D0 (en) 2020-03-12 2020-04-29 Harbour Antibodies Bv SARS-Cov-2 (SARS2, COVID-19) antibodies
CN112557645B (en) 2020-03-13 2022-03-08 珠海碳云智能科技有限公司 Method and device for screening epitope polypeptides
CN111592594B (en) 2020-03-13 2022-05-10 北京大学 Monoclonal antibody for resisting novel coronavirus and application thereof
CN114163523B (en) 2020-03-17 2023-07-18 北京凯因科技股份有限公司 Single-domain antibody for novel coronavirus and application thereof
CN111303280B (en) 2020-03-22 2022-01-07 中国人民解放军军事科学院军事医学研究院 High-neutralization-activity anti-SARS-CoV-2 fully human monoclonal antibody and application
WO2021190980A1 (en) 2020-03-22 2021-09-30 Quadrucept Bio Limited Multimers for viral strain evolution
KR102205028B1 (en) 2020-03-22 2021-01-20 (주)셀트리온 A binding molecules able to neutralize SARS-CoV-2
US11021531B1 (en) 2020-03-23 2021-06-01 Centivax, Inc. Anti-SARS-Cov-2 antibodies derived from 2GHW
US11021532B1 (en) 2020-03-23 2021-06-01 Centivax, Inc. Superhuman anti-SARS-CoV-2 antibodies and uses thereof
US11028150B1 (en) 2020-03-23 2021-06-08 Centivax, Inc. Anti-SARS-CoV-2 antibodies derived from 2DD8
US11028167B1 (en) 2020-03-23 2021-06-08 Centivax, Inc. Anti-SARS-Cov-2 antibodies derived from 3bgf
CN111690058B (en) 2020-03-30 2021-02-05 三优生物医药(上海)有限公司 Antibodies with neutralizing activity against coronaviruses and uses thereof
CN111423508A (en) 2020-03-31 2020-07-17 江苏省疾病预防控制中心(江苏省公共卫生研究院) Separated SARS-CoV-2 protein binding molecule for resisting virus infection
CN111848750B (en) 2020-03-31 2023-09-12 浙江聚康生物工程有限公司 Method and kit for rapidly enriching and detecting 2019-nCoV
SG11202103404PA (en) 2020-04-02 2021-04-29 Regeneron Pharma Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
CA3174212A1 (en) * 2020-04-03 2021-10-07 Blis Technologies Limited Antiviral treatment comprising blis containing probiotic products
WO2021203053A1 (en) 2020-04-03 2021-10-07 Vir Biotechnology, Inc. Immunotherapy targeting a conserved region in sars coronaviruses
CN111499765B (en) 2020-04-08 2022-04-08 四川携光生物技术有限公司 Coronavirus fusion protein and preparation method and application thereof
CN111440229B (en) 2020-04-13 2021-08-03 中国人民解放军军事科学院军事医学研究院 Novel coronavirus T cell epitope and application thereof
CN111647053A (en) 2020-04-16 2020-09-11 军事科学院军事医学研究院生命组学研究所 Polypeptide and application thereof in novel coronavirus detection and antibody or vaccine screening
CN111592595B (en) 2020-04-27 2021-02-19 南京医科大学 Neutralizing antibody against novel coronavirus SARS-Cov-2 and application thereof
CN111647076B (en) 2020-04-27 2021-02-26 南京医科大学 Neutralizing single-domain antibody for resisting novel coronavirus SARS-Cov-2 and application thereof
US11634477B2 (en) 2020-04-28 2023-04-25 The Rockefeller University Neutralizing anti-SARS-CoV-2 antibodies and methods of use thereof
CN111471105A (en) 2020-05-05 2020-07-31 广西医科大学 Preparation and application of novel coronavirus silver therapeutic neutralizing antibody
JP2023525039A (en) 2020-05-08 2023-06-14 ヴィア・バイオテクノロジー・インコーポレイテッド Antibodies against SARS-COV-2
CN111620946B (en) 2020-05-09 2020-12-22 江苏省疾病预防控制中心(江苏省公共卫生研究院) Isolated novel coronavirus monoclonal antibodies or antigen binding portions thereof
CN111518773B (en) 2020-05-09 2023-01-03 山东兴瑞生物科技有限公司 CAR-T cell for resisting novel coronavirus S protein, preparation method and application thereof
CN111662379B (en) 2020-05-09 2021-03-02 江苏省疾病预防控制中心(江苏省公共卫生研究院) Antibody for resisting novel coronavirus, preparation method and application
CN111620945B (en) 2020-05-09 2021-01-15 江苏省疾病预防控制中心(江苏省公共卫生研究院) Monoclonal antibody or derivative thereof for resisting novel coronavirus
CN112010966B (en) 2020-05-15 2021-03-19 潍坊医学院 Monoclonal antibody aiming at non-RBD (radial basis function) region of new coronavirus spike protein and application thereof
JP2023528235A (en) 2020-05-17 2023-07-04 アストラゼネカ・ユーケイ・リミテッド SARS-CoV-2 Antibodies and Methods of Selecting and Using The Same
CN111607003B (en) 2020-05-21 2022-08-23 上海百英生物科技有限公司 SARS-CoV-2N/S1 (RBD) recombinant protein and its preparation method and application
CN111560399B (en) 2020-05-22 2021-07-06 苏州君盟生物医药科技有限公司 Large-scale transient transfection method for cells
CN111574614A (en) 2020-05-23 2020-08-25 湖南源品细胞生物科技有限公司 TCR enrichment clone type and acquisition method and application thereof
IL298194A (en) 2020-05-26 2023-01-01 Regeneron Pharma Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
CN111748032B (en) 2020-05-27 2021-03-16 江苏省疾病预防控制中心(江苏省公共卫生研究院) Antibody against novel coronavirus and immunoassay using the same
TW202210504A (en) 2020-05-29 2022-03-16 德國科隆大學 Neutralizing antibodies against sars-related coronavirus
WO2021245184A1 (en) 2020-06-02 2021-12-09 Neurimmune Ag HUMAN ANTIBODIES AGAINST SEVERE ACUTE RESPIRATORY SYNDROME CORONAVIRUS-2 (SARS-CoV-2)
CN111647077B (en) 2020-06-02 2021-02-09 深圳市因诺赛生物科技有限公司 Novel coronavirus (SARS-COV-2) spike protein binding molecule and application thereof
KR20230019166A (en) 2020-06-03 2023-02-07 리제너론 파아마슈티컬스, 인크. Methods for treating or preventing SARS-CoV-2 infection and COVID-19 using anti-SARS-CoV-2 spike glycoprotein antibodies
CN113336846B (en) 2020-06-04 2023-11-10 山东宽和正生物医药有限公司 Monoclonal antibody E11 against novel coronavirus SARS-CoV-2
WO2021249547A1 (en) 2020-06-12 2021-12-16 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Anti-coronavirus antibodies and uses thereof
CN113151184B (en) 2020-06-15 2023-03-21 上海市公共卫生临床中心 Method for cell membrane-based display of coronavirus immunogens to induce neutralizing antibodies
CN111499692B (en) 2020-06-16 2020-12-04 国家纳米科学中心 Polypeptide of targeting novel coronavirus COVID-19 and application thereof
CN111825762A (en) 2020-06-17 2020-10-27 武汉华美生物工程有限公司 Nano antibody of S protein RBD structure domain of anti SARS-COV-2 virus and its use
CN111848751B (en) 2020-06-18 2023-09-29 河南省生物工程技术研究中心 Novel coronavirus S protein dominant antigen epitope peptide and application thereof
CN111690059B (en) 2020-06-19 2022-03-08 武汉生物制品研究所有限责任公司 Monoclonal antibody 1D7 for resisting SARS-CoV-2
CN111732654B (en) 2020-06-19 2021-05-25 武汉生物制品研究所有限责任公司 Monoclonal antibody 1E10 for resisting SARS-CoV-2
CN111718411B (en) 2020-06-19 2022-03-08 武汉生物制品研究所有限责任公司 Monoclonal antibody 1F2 for resisting SARS-CoV-2
CN112521494B (en) 2020-06-19 2021-06-25 武汉生物制品研究所有限责任公司 Monoclonal antibody 2B11 for resisting SARS-CoV-2
US11020474B1 (en) 2020-06-25 2021-06-01 Abclonal Science, Inc. Producing recombinant SARS-CoV-2 spike protein in a pre-fusion state
CN111714621B (en) 2020-06-29 2021-04-27 中国科学院昆明动物研究所 Application of transferrin, transferrin receptor and antibody thereof in preparing medicine for resisting SARS-CoV-2 virus
CN111732655B (en) 2020-07-01 2021-10-22 中国人民解放军军事科学院军事医学研究院 RBD-targeted high-neutralization-activity anti-SARS-CoV-2 fully-humanized monoclonal antibody and application thereof
CN111848789B (en) 2020-07-02 2022-04-22 武汉华美生物工程有限公司 Single chain antibody for resisting SARS-COV-2 virus S protein and its use
CN111732664B (en) 2020-07-06 2020-12-01 北京金智准科技有限公司 Novel coronavirus recombinant protein, rabbit-human chimeric antibody, preparation method and application thereof
CN111690060A (en) 2020-07-06 2020-09-22 深圳市亚辉龙生物科技股份有限公司 IgA antibody capable of specifically recognizing RBD protein and kit
CN111978377B (en) 2020-07-09 2022-05-31 苏州和锐生物科技有限公司 COVID-19 antigen, preparation method and application
CN113072640A (en) 2020-07-16 2021-07-06 深圳市雅臣智能生物工程有限公司 Anti-mutation SARS-CoV-2 multi-binding site IgY and its preparation and antibody combination therapy and application
CN112500480B (en) 2020-07-17 2022-04-01 上海洛启生物医药技术有限公司 Nanobodies against novel coronaviruses and uses thereof
CN111978399B (en) 2020-07-20 2022-05-20 江苏集萃医学免疫技术研究所有限公司 Antibody specifically binding SARS-COV-2 antigen protein and its use
CN111995672B (en) 2020-07-20 2022-05-20 江苏集萃医学免疫技术研究所有限公司 Specific antibody of coronavirus SARS-COV-2S protein and its use
CN112010963B (en) 2020-07-20 2022-05-20 江苏集萃医学免疫技术研究所有限公司 SARS-COV-2 antibody and use thereof
CN112210004B (en) 2020-07-20 2021-06-29 江苏集萃医学免疫技术研究所有限公司 Coronavirus COVID-19 detection antibody and application thereof
CN112010962B (en) 2020-07-20 2022-05-20 江苏集萃医学免疫技术研究所有限公司 Antibody for detecting COVID-19 and medical application thereof
CN111978396B (en) 2020-07-20 2022-05-20 江苏集萃医学免疫技术研究所有限公司 Antibody specifically binding SARS-COV-2 NP protein and its use
CN111978398B (en) 2020-07-20 2022-05-20 江苏集萃医学免疫技术研究所有限公司 Antibody against coronavirus SARS-COV-2 and medical use thereof
CN111978395B (en) 2020-07-20 2022-06-10 四川大学 Monoclonal antibody against novel coronavirus RBD domain antigen
CN111978397B (en) 2020-07-20 2022-05-20 江苏集萃医学免疫技术研究所有限公司 Antibody specifically binding SARS-COV-2S protein and its use
CN111825771A (en) 2020-07-23 2020-10-27 白涛 Antiviral antibacterial biological missile
CN112062859B (en) 2020-07-24 2022-08-09 沣潮医药科技(上海)有限公司 Chimeric antigen receptor for pathogen clearance and uses thereof
CN111793129B (en) 2020-07-28 2021-09-24 上海市公共卫生临床中心 Antibody or antigen binding fragment thereof specifically binding to coronavirus
CN112094340B (en) 2020-07-31 2023-06-20 王跃驹 Application of plant as host in expression of novel coronavirus pneumonia neutralizing antibody B38 antibody and/or H4 antibody
CN112300274B (en) 2020-08-10 2022-05-31 苏州方科生物科技有限公司 Human source antibody of novel coronavirus specific antigen peptide, preparation method and use
CN111875701A (en) 2020-08-14 2020-11-03 江苏中慧元通生物科技有限公司 Single-chain antibody of SARS-CoV-2 virus and its use
CN111925439A (en) 2020-08-19 2020-11-13 重庆医科大学 Method for rapidly screening new coronavirus RBD (radial basis function) specific fully human neutralizing monoclonal antibody
CN111925442B (en) 2020-08-19 2022-10-11 重庆医科大学 New coronavirus RBD specific monoclonal antibody and application
CN111925444B (en) 2020-08-19 2022-10-11 重庆医科大学 New coronavirus RBD specific monoclonal antibody and application
CN111925441B (en) 2020-08-19 2022-10-04 重庆医科大学 New coronavirus RBD specific monoclonal antibody and application
CN111909261B (en) 2020-08-19 2022-10-04 重庆医科大学 New coronavirus RBD specific monoclonal antibody and application
CN111909262B (en) 2020-08-19 2022-10-04 重庆医科大学 New coronavirus RBD specific monoclonal antibody and application
CN115477698A (en) 2020-08-19 2022-12-16 重庆医科大学 New coronavirus RBD specific monoclonal antibody and application
CN114920832B (en) 2020-08-19 2023-10-13 重庆医科大学 New coronavirus RBD specific monoclonal antibody and application
CN111944026B (en) 2020-08-19 2023-10-20 重庆医科大学 Linear antigen epitope of novel coronavirus RBD specific monoclonal antibody and application
CN111909260B (en) 2020-08-19 2022-06-21 重庆医科大学 New coronavirus RBD specific monoclonal antibody and application
CN115925898A (en) 2020-08-19 2023-04-07 重庆医科大学 Novel coronavirus RBD specific monoclonal antibody and application
CN112062838B (en) 2020-08-25 2021-03-23 南京医科大学 Neutralizing single-domain antibody for resisting novel coronavirus SARS-Cov-2 and application thereof
CN112010964A (en) 2020-09-02 2020-12-01 安第斯抗体生物技术衡水有限公司 Novel coronavirus alpaca antibody and preparation method and application thereof
CN111995674B (en) 2020-09-03 2022-02-11 中国人民解放军军事科学院军事医学研究院 anti-COVID-19 virus neutralizing antibody mhC3, humanized antibody and application thereof
KR102229225B1 (en) 2020-09-04 2021-03-19 (주)셀트리온 Binding Molecules Binding To A Spike Protein Of SARS-CoV-2 For Diagnosis Of COVID-19
CN112010967B (en) 2020-09-07 2021-03-23 康维众和(中山)生物药业有限公司 Novel nano antibody for neutralizing toxicity of coronavirus SARS-Cov-2 and preparation method and application thereof
CN112076316B (en) 2020-09-21 2022-10-11 中国人民解放军军事科学院军事医学研究院 Double-antibody composition and application thereof in preparation of COVID-19 therapeutic drugs
CN112062840A (en) 2020-09-22 2020-12-11 石河子大学 Nano antibody based on novel coronavirus S protein and application thereof
CN112175071A (en) 2020-09-22 2021-01-05 通用生物系统(安徽)有限公司 Preparation method of novel coronavirus spike protein monoclonal antibody
CN112062839A (en) 2020-09-22 2020-12-11 石河子大学 Nano antibody based on novel coronavirus S protein S1 subunit and application thereof
CN114437206B (en) 2020-09-23 2023-05-16 深圳市因诺赛生物科技有限公司 Novel coronavirus (SARS-COV-2) spike protein binding molecule and application thereof
CN112094342B (en) 2020-09-25 2022-05-13 中国科学技术大学 Alpaca source nano antibody combined with SARS-CoV-2RBD
CN112094343B (en) 2020-09-25 2022-05-13 中国科学技术大学 Alpaca source nano antibody combined with SARS-CoV-2 RBD
CN112094327A (en) 2020-09-25 2020-12-18 军事科学院军事医学研究院军事兽医研究所 Truncation body based on novel coronavirus RBD-SD1 protein and application thereof
CN112094326B (en) 2020-09-25 2023-06-02 河南省生物工程技术研究中心 New coronavirus antigen and application thereof
CN112159469B (en) 2020-09-30 2022-08-02 上海市公共卫生临床中心 Antibodies or antigen-binding fragments thereof to coronaviruses
CN116023478A (en) 2020-09-30 2023-04-28 上海市公共卫生临床中心 Neutralizing antibodies or antigen binding fragments thereof for coronaviruses
CN112125973B (en) 2020-09-30 2022-02-01 上海市公共卫生临床中心 Specific antibodies or antigen-binding fragments thereof for coronaviruses
CN112251414A (en) 2020-10-12 2021-01-22 中国科学院苏州纳米技术与纳米仿生研究所 Hybridoma cell strain, preparation method and application thereof
CN112194711A (en) 2020-10-15 2021-01-08 深圳市疾病预防控制中心(深圳市卫生检验中心、深圳市预防医学研究所) B cell linear epitope of novel coronavirus S protein, antibody, identification method and application
CN112409488B (en) 2020-10-23 2022-07-01 中国科学院上海药物研究所 Monoclonal antibody aiming at various coronaviruses and application
GB202017058D0 (en) 2020-10-27 2020-12-09 Kymab Ltd Antibodies and uses thereof
CN112225806B (en) 2020-11-13 2021-04-13 李亚峰 Neutralizing active monoclonal antibody of human source for resisting novel coronavirus (SARS-CoV-2)
CN112625136B (en) 2020-11-18 2022-02-11 三优生物医药(上海)有限公司 Bispecific antibodies having neutralizing activity against coronaviruses and uses thereof
RU2744274C1 (en) 2020-11-20 2021-03-04 Федеральное государственное бюджетное учреждение науки институт биоорганической химии им. академиков М.М. Шемякина и Ю.А. Овчинникова Российской академии наук (ИБХ РАН) Monoclonal antibody to rdb fragment in composition of sars-cov-2 s protein
CN112341541B (en) 2020-11-23 2022-05-06 中国疾病预防控制中心病毒病预防控制所 Humanized anti-neocoronavirus neutralizing antibody nCoV-163 and application thereof
CN112409479B (en) 2020-11-23 2022-04-26 中国疾病预防控制中心病毒病预防控制所 Humanized anti-neocoronavirus neutralizing antibody nCoV-121 and application thereof
CN112430265B (en) 2020-11-23 2022-04-12 中国疾病预防控制中心病毒病预防控制所 Humanized anti-neocoronavirus neutralizing antibody nCoV-61 and application thereof
CN112522203B (en) 2020-11-23 2023-08-11 中山大学附属第五医院 Cell vesicle for expressing chimeric antigen receptor, and preparation method and application thereof
CN112442120A (en) 2020-11-25 2021-03-05 苏州大学 Neutralizing antibody against SARS-COV-2 of severe acute respiratory syndrome type II coronavirus
CN112574299B (en) 2020-11-25 2023-03-21 苏州方科生物科技有限公司 Human source antibody of novel coronavirus specific antigen peptide, preparation method and use
KR102233689B1 (en) 2020-11-26 2021-03-30 재단법인 오송첨단의료산업진흥재단 Antibodies specifically binding to receptor-binding domain of SARS-CoV-2 spike protein and use thereof
CN112500481B (en) 2020-11-29 2021-08-20 山西省人民医院 Human source neutralizing active monoclonal antibody for resisting novel coronavirus
CN112574300B (en) 2020-12-02 2022-03-08 深圳先进技术研究院 anti-SAR-COV-2 fully human monoclonal antibody and preparation method and application thereof
CN112661841B (en) 2020-12-04 2022-10-14 广州市第八人民医院 Fully human monoclonal antibody 17-2 for neutralizing neoepitope of new coronavirus and application thereof
CN112626089B (en) 2020-12-08 2022-03-11 杭州百凌生物科技有限公司 SARS-CoV-2 virus S protein receptor binding region coding gene, antibody and application
CN112538111B (en) 2020-12-09 2022-04-29 深圳市亚辉龙生物科技股份有限公司 New coronavirus single-chain antibody, quality control product and preparation method
CN112485455B (en) 2020-12-09 2022-11-01 深圳市亚辉龙生物科技股份有限公司 New coronavirus antibody quality control product and preparation method thereof
CN112250763B (en) 2020-12-21 2021-03-26 三优生物医药(上海)有限公司 Antibody targeting SARS-CoV-2 coronavirus and its diagnosis and detection use
CN112724247A (en) 2020-12-25 2021-04-30 江苏中方基因生物医学科技有限公司 SARS-CoV-2 coronavirus IgA antibody and its application in ELISA detection kit
CN112626030A (en) 2020-12-31 2021-04-09 深圳市第二人民医院 Production method for neutralizing new coronavirus by using nano antibody of spinous process protein Spike displayed on surface of exosome
CN112341542B (en) 2021-01-08 2021-05-14 清华大学 3-hydroxybutyrylated modified protein medicine and preparation method and application thereof
CN112625125B (en) 2021-01-18 2021-12-14 中国人民解放军军事科学院军事医学研究院 Monoclonal antibody for neutralizing novel coronavirus infection
CN112390879B (en) 2021-01-21 2021-04-02 上海科技大学 Antibody targeting SARS-CoV-2 and its preparation method and use
WO2022162587A1 (en) 2021-01-27 2022-08-04 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
CN113264998B (en) 2021-01-28 2023-02-28 四川大学华西医院 Single-chain antibody of S1 protein on surface of anti-new coronavirus SARS-CoV-2 and application thereof
CN113150129B (en) 2021-01-28 2023-02-28 四川大学华西医院 Single-chain antibody for resisting S2 protein on surface of new coronavirus SARS-CoV-2 and application thereof
CN112724248A (en) 2021-01-28 2021-04-30 南京拓峰生物科技有限公司 Nano antibody capable of combining SARS-CoV-2 and application thereof
CN112521496B (en) 2021-01-29 2022-09-06 中国人民解放军空军军医大学 Monoclonal antibody specifically binding SARS-CoV-2 Spike RBD and application thereof
CN115109151A (en) 2021-01-31 2022-09-27 中南大学湘雅医院 Novel coronavirus monoclonal antibody XY6 and application thereof
CN112552399B (en) 2021-02-24 2021-07-20 恒翼生物医药科技(上海)有限公司 anti-SARS-COV-2 neutralizing antibody
CN113150132B (en) 2021-03-11 2022-05-31 苏州携创生物技术有限公司 anti-SARS-CoV-2 recombinant antibody and its application
CN112794899B (en) 2021-03-16 2021-09-24 易康生物(苏州)有限公司 Fully human monoclonal neutralizing antibody for resisting novel coronavirus and application thereof
CN112794898B (en) 2021-03-16 2021-08-03 易康生物(苏州)有限公司 Fully human monoclonal antibody for resisting novel coronavirus and application thereof
CN112980885B (en) 2021-03-18 2022-04-15 恒翼生物医药科技(上海)有限公司 Expression vector of anti-SARS-COV-2 neutralizing antibody
CN113045647B (en) 2021-03-22 2022-04-26 南京传奇生物科技有限公司 Neutralizing antibody of novel coronavirus SARS-CoV-2 and application thereof
CN113150135B (en) 2021-04-14 2022-09-20 中山大学 Neutralizing antibodies against novel coronavirus receptor binding domains and uses thereof
CN113173995B (en) 2021-04-30 2022-09-23 上海市公共卫生临床中心 Bispecific antibody combined with coronavirus
CN113234149B (en) 2021-05-19 2023-06-16 武汉菲沙基因组医学有限公司 New fully human coronal IgA single-chain antibody and application thereof
CN113234150A (en) 2021-05-19 2021-08-10 武汉菲沙基因组医学有限公司 Fully human novel crown IgG1 single-chain antibody and application thereof
CN113234148A (en) 2021-05-19 2021-08-10 武汉菲沙基因组医学有限公司 Fully human-derived novel crown IgK single-chain antibody and application thereof
CN113185609A (en) 2021-05-19 2021-07-30 武汉菲沙基因组医学有限公司 Fully human novel crown IgG2 single-chain antibody and application thereof
CN113214389A (en) 2021-05-19 2021-08-06 武汉菲沙基因组医学有限公司 Fully human-derived novel crown IgL single-chain antibody and application thereof
CN113215106B (en) 2021-05-20 2022-04-01 广东省公共卫生研究院 Hybridoma cell, monoclonal antibody for resisting SARS-CoV-2 and application thereof
CN113354733B (en) 2021-06-04 2022-02-18 武汉生物制品研究所有限责任公司 Monoclonal antibody 20D8 for resisting SARS-CoV-2 epidemic mutant strain
CN113234151B (en) 2021-06-08 2022-02-15 皖南医学院第一附属医院(皖南医学院弋矶山医院) Development of CAR-NK (Carcar-natural killer) based on trimer nano antibody of SARS-CoV2 virus S protein
CN113248581A (en) 2021-06-15 2021-08-13 江西浩然生物制药有限公司 Novel corona S antigen for generating neutralizing antibody of novel corona virus and preparation method thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005060520A2 (en) * 2003-11-25 2005-07-07 Dana-Farber Cancer Institute, Inc. ANTIBODIES AGAINST SARS-CoV AND METHODS OF USE THEREOF
US20060240551A1 (en) * 2004-06-02 2006-10-26 Shibo Jiang Neutralizing monoclonal antibodies against severe acute respiratory syndrome-associated coronavirus
US20170096455A1 (en) * 2014-03-20 2017-04-06 The University Of North Carolina At Chapel Hill Methods and compositions for chimeric coronavirus spike proteins
CN111088283A (en) * 2020-03-20 2020-05-01 苏州奥特铭医药科技有限公司 mVSV viral vector, viral vector vaccine thereof and mVSV-mediated novel coronary pneumonia vaccine

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHRISTOPHER O.BARNES, ET AL.,: "Structures of Human Antibodies Bound to SARS-CoV-2 Spike Reveal Common Epitopes and Recurrent Features of Antibodies", CELL, vol. 182, no. 4, 23 June 2020 (2020-06-23), pages 828 - 842, XP055802279 *
QIANG XIAOLING, ZHU SHU, LI JIANHUA, WANG PING, TRACEY KEVIN J, WANG HAICHAO: "Monoclonal Antibodies Capable of Binding SARS-CoV-2 Spike Protein Receptor Binding Motif Specifically Prevent GM-CSF Induction", BIORXIV, 4 September 2020 (2020-09-04), pages 1 - 27, XP055802280 *
XIAOLONG TIAN; LI CHENG; HUANG AILING; XIA SHUAI; LU SICONG; SHI ZHENGLI; LU LU; JIANG SHIBO; YANG ZHENLIN; WU YANLING; YING TIANL: "Potent binding of 2019 novel coronavirus spike protein by a SARS coronavirus-specific human monoclonal antibody", EMERGING MICROBES & INFECTIONS, vol. 17, 17 February 2020 (2020-02-17), pages 647 - 649, XP055736759 *

Cited By (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11168128B2 (en) 2020-02-26 2021-11-09 Vir Biotechnology, Inc. Antibodies against SARS-CoV-2 and methods of using the same
WO2021173753A1 (en) * 2020-02-26 2021-09-02 Vir Biotechnology, Inc. Antibodies against sars-cov-2 and methods of using the same
US11479599B2 (en) 2020-02-26 2022-10-25 Vir Biotechnology, Inc. Antibodies against SARS-CoV-2 and methods of using the same
EP4245373A3 (en) * 2020-02-26 2023-12-20 VIR Biotechnology, Inc. Antibodies against sars-cov-2
EP3872091A1 (en) * 2020-02-26 2021-09-01 VIR Biotechnology, Inc. Antibodies against sars-cov-2 and methods of using the same
WO2021190980A1 (en) * 2020-03-22 2021-09-30 Quadrucept Bio Limited Multimers for viral strain evolution
WO2021194896A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from 2ghw
WO2021194985A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from 6nb6
WO2021194951A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from 2dd8
WO2021194965A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Superhuman anti-sars-cov-2 antibodies and uses thereof
WO2021194886A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from 3bgf
WO2021194891A1 (en) * 2020-03-23 2021-09-30 Centivax, Inc. Anti-sars-cov-2 antibodies derived from cr3022
US11732030B2 (en) 2020-04-02 2023-08-22 Regeneron Pharmaceuticals, Inc. Anti-SARS-CoV-2-spike glycoprotein antibodies and antigen-binding fragments
JP2022008067A (en) * 2020-05-01 2022-01-13 花王株式会社 Anti-SARS-CoV-2 antibody
JP7038367B2 (en) 2020-05-01 2022-03-18 花王株式会社 Anti-SARS-CoV-2 antibody
WO2021221136A1 (en) * 2020-05-01 2021-11-04 花王株式会社 Anti-sars-cov-2 antibody
WO2021226405A1 (en) * 2020-05-06 2021-11-11 International Aids Vaccine Initiative Inc. Covid-19 antibodies and uses thereof
US11918641B2 (en) 2020-05-08 2024-03-05 Academia Sinica Chimeric influenza vaccines
WO2021242815A1 (en) * 2020-05-26 2021-12-02 Regeneron Pharmaceuticals, Inc. Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
WO2022026592A2 (en) 2020-07-28 2022-02-03 Celltas Bio, Inc. Antibody molecules to coronavirus and uses thereof
WO2022054068A1 (en) * 2020-09-14 2022-03-17 Ramot At Tel-Aviv University Ltd. Antibodies for the prevention, treatment and detection of coronavirus infection
WO2022060916A1 (en) 2020-09-15 2022-03-24 Regenxbio Inc. Vectorized antibodies for anti-viral therapy
WO2022162587A1 (en) * 2021-01-27 2022-08-04 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
WO2022197720A3 (en) * 2021-03-15 2022-12-22 Beth Israel Deaconess Medical Center, Inc. Compositions and methods for treating coronavirus infection
WO2022210830A1 (en) * 2021-03-30 2022-10-06 国立大学法人 富山大学 Anti-sars-cov-2 antibody
WO2022226079A1 (en) * 2021-04-20 2022-10-27 Inbios International, Inc. Neutralizing antibodies against sars-cov-2
WO2022232262A3 (en) * 2021-04-28 2022-12-01 The Trustees Of Columbia University In The City Of New York Antibodies for the treatment and prevention of covid-19 and emerging variants
JP7343739B2 (en) 2021-06-04 2023-09-13 武漢生物制品研究所有限責任公司 Monoclonal antibody 20D8 against SARS-CoV-2 epidemic variant
WO2022263638A1 (en) * 2021-06-17 2022-12-22 Centre Hospitalier Universitaire Vaudois (C.H.U.V.) Anti-sars-cov-2 antibodies and use thereof in the treatment of sars-cov-2 infection
WO2022271884A3 (en) * 2021-06-22 2023-01-26 Twist Bioscience Corporation Methods and compositions relating to covid antibody epitopes
WO2022271258A1 (en) * 2021-06-25 2022-12-29 La Jolla Institute For Immunology Chimeric anti-sars-cov2 nucleoprotein antibodies
WO2023275538A1 (en) * 2021-06-28 2023-01-05 Diosynvax Ltd Beta-coronavirus vaccines
WO2023287875A1 (en) * 2021-07-14 2023-01-19 Regeneron Pharmaceuticals, Inc. Anti-sars-cov-2-spike glycoprotein antibodies and antigen-binding fragments
WO2023285620A3 (en) * 2021-07-14 2023-03-09 Alchemab Therapeutics Ltd. Antibodies targeting sars-cov-2
WO2023002944A1 (en) * 2021-07-19 2023-01-26 公立大学法人福島県立医科大学 Antibody capable of binding to novel coronavirus (sars-cov-2) and sars coronavirus (sars-cov)
WO2023004431A3 (en) * 2021-07-23 2023-04-20 The Trustees Of Columbia University In The City Of New York Characterization of potent and broadly neutralizing monoclonal antibodies against sars-cov-2, its variants, and related coronaviruses and methods of use
WO2023004477A1 (en) * 2021-07-30 2023-02-02 The University Of Melbourne Neutralising antibodies and uses thereof
WO2023015231A1 (en) * 2021-08-04 2023-02-09 The Regents Of The University Of California Sars-cov-2 virus-like particles
WO2023015232A1 (en) * 2021-08-04 2023-02-09 The Regents Of The University Of California Sars-cov-2 virus-like particles
WO2023023183A3 (en) * 2021-08-17 2023-08-17 Twist Bioscience Corporation Sars-cov-2 antibodies and related compositions and methods of use
WO2023035016A1 (en) * 2021-09-03 2023-03-09 The Uab Research Foundation Human neutralizing antibodies against sars-cov-2 spike s2 domain and uses thereof
WO2023033363A1 (en) * 2021-09-06 2023-03-09 국민대학교산학협력단 Bispecific antibody specifically binding to sars-cov-2
WO2023048656A3 (en) * 2021-09-24 2023-06-08 Chulalongkorn University Human monoclonal antibodies against the receptor-binding domain of sars-cov-2 spike protein
WO2023056482A1 (en) * 2021-10-01 2023-04-06 Academia Sinica Antibody specific to spike protein of sars-cov-2 and uses thereof
US11866485B2 (en) 2021-10-01 2024-01-09 Academia Sinica Antibody specific to spike protein of SARS-CoV-2 and uses thereof
WO2023064717A3 (en) * 2021-10-08 2023-09-14 Augmenta Bioworks, Inc. Antibodies for sars-cov-2 and uses thereof
WO2023064435A3 (en) * 2021-10-15 2023-07-27 The Children's Medical Center Corporation Compositions and methods relating to sars-cov-2 neutralizing antibodies
WO2023081434A3 (en) * 2021-11-07 2023-06-15 Regeneron Pharmaceuticals, Inc. Methods for treating or preventing sars-cov-2 infections and covid-19 with anti-sars-cov-2 spike glycoprotein antibodies
WO2023084055A1 (en) * 2021-11-12 2023-05-19 Rq Biotechnology Limited Compositions
WO2023104904A3 (en) * 2021-12-08 2023-07-20 Genclis The sars-cov-2 and variants use two independent cell receptors to replicate
WO2023109419A1 (en) * 2021-12-14 2023-06-22 Beijing Acrobiosystems Biotechnology Co., Ltd A neutralizing antibody that can bind sars-cov-2 virus and its application
WO2023113094A1 (en) * 2021-12-16 2023-06-22 주식회사 씨티씨백 Covid-19 vaccine composition with increased immunogenicity
WO2023122535A3 (en) * 2021-12-20 2023-08-03 DNARx Nucleic acid expression using subcutaneous administration
WO2023145859A1 (en) * 2022-01-28 2023-08-03 株式会社イーベック HUMAN NEUTRALIZING ANTIBODY AGAINST SARS-CoV-2 HAVING BREADTH TO VARIANT STRAINS, AND ANTIGEN-BINDING FRAGMENT THEREOF
WO2023156187A1 (en) * 2022-02-16 2023-08-24 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts High affinity antibodies against the sars-cov-2 receptor binding domain
WO2023178182A1 (en) * 2022-03-16 2023-09-21 10X Genomics, Inc. Compositions and methods for detection and treatment of coronavirus infection
WO2023190851A1 (en) * 2022-03-31 2023-10-05 公立大学法人福島県立医科大学 Antibody binding to novel coronavirus (sars-cov-2)
WO2023190852A1 (en) * 2022-03-31 2023-10-05 公立大学法人福島県立医科大学 Antibody capable of binding to new coronavirus (sars-cov-2)
RU2817696C1 (en) * 2023-11-22 2024-04-18 Общество с ограниченной ответственностью "ИМГЕН+" (ООО "ИМГЕН+") MONOCLONAL ANTIBODY IC1 AND ITS ANTIGEN-BINDING FRAGMENT, SELECTIVELY BINDING RECEPTOR-BINDING DOMAIN OF SPIKE PROTEIN OF SARS-CoV-2 VIRUS, HAVING VIRUS-NEUTRALIZING ACTIVITY
RU2817697C1 (en) * 2023-12-08 2024-04-18 Федеральное государственное бюджетное учреждение науки Институт молекулярной и клеточной биологии Сибирского отделения Российской академии наук (ИМКБ СО РАН) MONOCLONAL ANTIBODY IC2 AND ITS ANTIGEN-BINDING FRAGMENT, SELECTIVELY BINDING RECEPTOR-BINDING DOMAIN OF SPIKE PROTEIN OF SARS-CoV-2 VIRUS, HAVING VIRUS-NEUTRALIZING ACTIVITY

Also Published As

Publication number Publication date
PL3889177T3 (en) 2023-05-08
HUE061425T2 (en) 2023-06-28
US20220356230A1 (en) 2022-11-10
CA3192925A1 (en) 2021-03-11
US10787501B1 (en) 2020-09-29
CL2021000997A1 (en) 2021-09-10
MA54405B1 (en) 2023-05-31
US10954289B1 (en) 2021-03-23
PE20221893A1 (en) 2022-12-13
PH12021550793A1 (en) 2022-02-28
MY197648A (en) 2023-06-30
CA3115553C (en) 2023-04-25
US20230348569A1 (en) 2023-11-02
IL282060A (en) 2021-05-31
HRP20230338T1 (en) 2023-05-26
JP2022536429A (en) 2022-08-17
JP7116256B1 (en) 2022-08-09
PT3889177T (en) 2023-02-02
TW202308686A (en) 2023-03-01
JP7411029B2 (en) 2024-01-10
US11732030B2 (en) 2023-08-22
TW202138004A (en) 2021-10-16
JP2022160520A (en) 2022-10-19
SG11202103404PA (en) 2021-04-29
CA3115553A1 (en) 2021-03-11
BR112020015534A2 (en) 2021-10-05
MX2021004130A (en) 2021-06-15
US10975139B1 (en) 2021-04-13
EP4209507A1 (en) 2023-07-12
CL2023000705A1 (en) 2023-08-25
EP3889177B1 (en) 2023-01-11
MA54405A (en) 2022-01-26
FI3889177T3 (en) 2023-03-19
SI3889177T1 (en) 2023-03-31
JP2024038003A (en) 2024-03-19
CN113766928A (en) 2021-12-07
LT3889177T (en) 2023-02-10
CR20220552A (en) 2023-01-17
AU2020340881A1 (en) 2021-10-21
KR20210134300A (en) 2021-11-09
CO2021005072A2 (en) 2021-04-30
DK3889177T3 (en) 2023-02-13
TWI785350B (en) 2022-12-01
RS64105B1 (en) 2023-04-28
EP3889177A1 (en) 2021-10-06
ES2939895T3 (en) 2023-04-27

Similar Documents

Publication Publication Date Title
US11732030B2 (en) Anti-SARS-CoV-2-spike glycoprotein antibodies and antigen-binding fragments
US20230265216A1 (en) Anti-tmprss2 antibodies and antigen-binding fragments
US20240043504A1 (en) Anti-SARS-CoV-2-Spike Glycoprotein Antibodies and Antigen-Binding Fragments
AU2021219671A1 (en) Anti-Tmprss2 Antibodies and Antigen-Binding Fragments
WO2022187626A1 (en) Anti-sars-cov-2-variant-spike glycoprotein antibodies and antigen-binding fragments
BR122023004078B1 (en) PHARMACEUTICAL COMPOSITION, ITS USE AND METHOD FOR MANUFACTURING AN ANTIBODY OR ANTIGEN BINDING FRAGMENT THEREOF
BR112020015534B1 (en) ISOLATED ANTIBODY OR ANTIGEN-BINDING FRAGMENT THEREOF, ITS MANUFACTURING METHOD, ITS USES, AS WELL AS PHARMACEUTICAL COMPOSITION
US20230125469A1 (en) Anti-SARS-CoV-2-Spike Glycoprotein Antibodies and Antigen-Binding Fragments

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 122023004078

Country of ref document: BR

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112020015534

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2021518800

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3115553

Country of ref document: CA

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

Ref document number: 20860660

Country of ref document: EP

Kind code of ref document: A1

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112020015534

Country of ref document: BR

Free format text: COM BASE NA PORTARIA 405 DE 21/12/2020, SOLICITA-SE QUE SEJA APRESENTADO, EM ATE 60 (SESSENTA) DIAS, NOVO CONTEUDO DE LISTAGEM DE SEQUENCIA CONTENDO TODOS OS CAMPOS OBRIGATORIOS, UMA VEZ QUE A LISTAGEM APRESENTADA NA PETICAO NO 870200114221 DE 09/09/2020 ESTA INCOMPLETA (FALTAM AS PRIORIDADES)

ENP Entry into the national phase

Ref document number: 112020015534

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20200730

ENP Entry into the national phase

Ref document number: 2020340881

Country of ref document: AU

Date of ref document: 20200625

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 522440749

Country of ref document: SA

122 Ep: pct application non-entry in european phase

Ref document number: 20860660

Country of ref document: EP

Kind code of ref document: A1