WO2020113199A1 - Recombinant hiv env polypeptides and their use - Google Patents

Recombinant hiv env polypeptides and their use Download PDF

Info

Publication number
WO2020113199A1
WO2020113199A1 PCT/US2019/063903 US2019063903W WO2020113199A1 WO 2020113199 A1 WO2020113199 A1 WO 2020113199A1 US 2019063903 W US2019063903 W US 2019063903W WO 2020113199 A1 WO2020113199 A1 WO 2020113199A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
polypeptide
hiv env
amino acid
positions
Prior art date
Application number
PCT/US2019/063903
Other languages
French (fr)
Inventor
Jon M. STEICHEN
William R. Schief
Original Assignee
International Aids Vaccine Initiative
The Scripps Research Institute
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 International Aids Vaccine Initiative, The Scripps Research Institute filed Critical International Aids Vaccine Initiative
Priority to EP19888314.2A priority Critical patent/EP3886898A4/en
Publication of WO2020113199A1 publication Critical patent/WO2020113199A1/en
Priority to US17/335,244 priority patent/US20210363195A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/08RNA viruses
    • C07K14/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus human T-cell leukaemia-lymphoma virus
    • C07K14/155Lentiviridae, e.g. human immunodeficiency virus [HIV], visna-maedi virus or equine infectious anaemia virus
    • C07K14/16HIV-1 ; HIV-2
    • C07K14/162HIV-1 ; HIV-2 env, e.g. gp160, gp110/120, gp41, V3, peptid T, CD4-Binding site
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia 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
    • A61K39/40Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum bacterial
    • 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
    • A61K39/42Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum viral
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5169Proteins, e.g. albumin, gelatin
    • 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
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • C07K16/1036Retroviridae, e.g. leukemia viruses
    • C07K16/1045Lentiviridae, e.g. HIV, FIV, SIV
    • C07K16/1063Lentiviridae, e.g. HIV, FIV, SIV env, e.g. gp41, gp110/120, gp160, V3, PND, CD4 binding site
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16023Virus like particles [VLP]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the field of the invention generally relates to recombinant HIV Env polypeptides and their use in the treatment and prevention of HIV/AIDS.
  • HIV broadly neutralizing antibodies have been isolated from infected individuals, but their induction by vaccination remains a major unmet need for global health.
  • the majority of HIV bnAbs engage the HIV Env trimer with long immunoglobulin heavy chain complementarity determining region 3 (HCDR3) loops that reach through the glycan shield to contact relatively conserved epitopes on the underlying protein surface.
  • HCDR3 immunoglobulin heavy chain complementarity determining region 3
  • a key barrier to inducing long-HCDR3 bnAbs is the priming of appropriate naive B cells that occur at very low frequency in the human repertoire and generally have very low or no detectable affinity for HIV Env.
  • the field lacks methods to design appropriate priming immunogens.
  • isolated polypeptides comprising a variant HIV
  • HIV Env trimers comprising an antigenic polypeptide described herein.
  • nanoparticles comprising an antigenic polypeptide described herein.
  • isolated polynucleotides encoding the isolated polypeptides, HIV ENV trimmers, and nanoparticles described herein.
  • RNA replicons comprising an isolated polynucleotide described herein.
  • viruses comprising an isolated polynucleotide described herein.
  • vectors comprising an isolated polynucleotide described herein.
  • HIV ENV trimmer or nanoparticle described herein.
  • liposomes and VLPs comprising an antigenic polypeptide described herein.
  • compositions comprising an antigenic polypeptide, recombinant HIV Env trimer, nanoparticle, polynucleotide, recombinant virus, VLP, or liposome described herein.
  • immunogenic compositions comprising an antigenic polypeptide, recombinant HIV Env trimer, nanoparticle, polynucleotide, recombinant virus, VLP, or liposome described herein.
  • HIV Env gpl20 in a subject comprising administering to the subject an effective amount of an immunogenic composition described herein.
  • described herein are methods of reducing the likelihood of HIV infection in a subject exposed to HIV comprising administering to the subject an effective amount of an immunogenic composition or pharmaceutical composition described herein.
  • described herein are methods of reducing the risk of a subject becoming infected with HIV comprising administering to the subject in need thereof an effective amount of an immunogenic composition or pharmaceutical composition described herein.
  • described herein are methods of preventing HIV infection comprising administering to a subject in need thereof an effective amount of an immunogenic composition or pharmaceutical composition described herein.
  • described herein are methods of treating HIV/AIDS comprising administering to a subject in need thereof an effective amount of an immunogenic composition or pharmaceutical composition described herein.
  • isolated antibodies comprising a VH and a VK described herein.
  • described herein are methods of identifying a vaccine candidate variant HIV Env polypeptide using an antibody described herein.
  • the disclosure provides:
  • An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises one or more of
  • VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
  • CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • CTNYAPNLLSNMRGEL SEQ ID NO: 72 comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • CTNY APNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
  • CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of (i) TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
  • TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • TGDIIGDIRQAHCNV S (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • FGDIIGDIRMAHCNV S (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
  • FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
  • SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
  • SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of (i) CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
  • CTNYAPNLLSNMRGEL SEQ ID NO: 72 comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • CTNYAPNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
  • CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
  • TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • TGDIIGDIRQAHCNV S (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • FGDIIGDIRMAHCNV S (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
  • FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of (i) SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
  • SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
  • SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises one or more of
  • VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
  • CTNVTNNITDDMRGEL SEQ ID NO: 71
  • CTNY APNLRSDMRGEI SEQ ID NO: 76
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises
  • VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
  • CTNVTNNITDDMRGEL SEQ ID NO: 71
  • CTNY APNLRSDMRGEI SEQ ID NO: 76
  • An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of
  • CTNYAPKLLSNMRGEI SEQ ID NO: 74
  • TRPNNNT SEQ ID NO: 77
  • CTNY APKLRSMMRGEI SEQ ID NO: 75
  • TRPNNNT SEQ ID NO: 77
  • CTNY APKLRSMMRGEI SEQ ID NO: 75
  • TRPSNNT SEQ ID NO: 78
  • CTNY APKLRSMMRGEI SEQ ID NO: 75
  • TRPSNNT SEQ ID NO: 78
  • CTNYAPNLRSDMRGEI SEQ ID NO: 76
  • TRPNNNT SEQ ID NO: 77
  • CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGHVRMAHCNIS SEQ ID NO: 82
  • SLILPCR SEQ ID NO: 86
  • CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGDVRMAHCNIS SEQ ID NO: 81
  • SLILPCR SEQ ID NO: 86
  • CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGHVRMAHCNIS SEQ ID NO: 82
  • SLILPCR SEQ ID NO: 86
  • CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPSNNT SEQ ID NO: 78
  • FGDVLGHVRMAHCNIS SEQ ID NO: 82
  • SLILPCR SEQ ID NO: 86
  • CTNYAPKLRSMMRGEI SEQ ID NO: 75
  • TRPSNNT SEQ ID NO: 78
  • FGDVLGHVRMAHCNIS SEQ ID NO: 82
  • SLILPCW SEQ ID NO: 87
  • CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGDVDMAKCTIS SEQ ID NO: 83
  • SLILPCR SEQ ID NO: 86
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), and SLILPCR (SEQ ID NO: 86), respectively,
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO:
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCW (SEQ ID NO:
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT5, or
  • An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT1, [19.] An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2.
  • An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT3.
  • An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT4.
  • An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT5.
  • An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD64 N332-GT2.
  • An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332-GT1.
  • An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332-GT2.
  • An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332-GT3.
  • An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD64 N332-GT2.
  • a recombinant HIV Env trimer comprising the isolated polypeptide of any one of [1] to [45]
  • a nanoparticle comprising the isolated polypeptide of any one of [1] to [45]
  • SOR sulfur Oxygenase Reductase
  • nanoparticle of [55] wherein the nanoparticle comprises N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5.
  • nanoparticle of [55] wherein the nanoparticle is a ferritin nanoparticle.
  • nanoparticle of [55] wherein the nanoparticle is a ferritin nanoparticle having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of the amino acid sequence of SEQ ID NO.
  • RNA replicon comprising the isolated polynucleotide of [61] .
  • a recombinant virus comprising the polynucleotide of [61]
  • the recombinant virus of [68] which is a recombinant adeno-associated virus (AAV).
  • a host cell comprising the polynucleotide of any one of [61] to [64, or the vector of [66] or [67]
  • Pseudomonas Bacillus, Streptomyces, yeast, CHO, YB/20, NS0, PER-C6, HEK-293T, NIH-3T3, HeLa, BHK, Hep G2, SP2/0, Rl. l, B-W, L-M, COS 1, COS 7, BSC1, BSC40, BMT10 cell, plant cell, insect cell, and human cell in tissue culture.
  • a method of producing the isolated polypeptide of any one of [1] to [45], recombinant HIV Env trimer of any one of [46] to [54], or the nanoparticle of [55] to [60] comprising, culturing the host cell of [71] so that the polynucleotide is expressed and the isolated polypeptide, recombinant HIV Env trimer, or nanoparticle is produced.
  • a VLP comprising the polypeptide of any one of [1] to [45]
  • a liposome comprising the polypeptide of any one of [1] to [45]
  • a pharmaceutical composition comprising the isolated polypeptide of any one of [1] to [45], recombinant HIV Env trimer of any one of [46] to [54], the nanoparticle of any one of [55] to [60], the polynucleotide of any one of [61] to [64], the vector of [66] or [67], the recombinant virus of [68] or [69], VLP of [73], or the liposome of [74] and a pharmaceutically acceptable excipient.
  • An immunogenic composition comprising the isolated polypeptide of any one of [1] to [45], recombinant HIV Env trimer of any one of [46] to [54], the nanoparticle of [any one of [55] to [60], the polynucleotide of any one of [61] to [64], the vector of [66 or [67], the recombinant virus of [68] or [69], VLP of [73], or the liposome of [74] and a pharmaceutically acceptable excipient.
  • a method for eliciting an immune response to HIV Env gpl20 in a subject comprising administering to the subject an effective amount of the immunogenic composition of any one of [76] to [81], thereby generating the immune response.
  • a method of reducing the likelihood of HIV infection in a subject exposed to HIV comprising administering to the subject an effective amount of the immunogenic composition of any one of [76] to [81], or the pharmaceutical composition of [75]
  • a method of reducing the risk of a subject becoming infected with HIV comprising administering to the subject in need thereof an effective amount of the immunogenic composition of any one of [76] to [81], or the pharmaceutical composition of [75]
  • a method of preventing HIV infection comprising administering to a subject in need thereof an effective amount of the immunogenic composition of any one of [76] to [81], or the pharmaceutical composition of [75]
  • a method of treating HIV/AIDS comprising administering to a subject in need thereof an effective amount of the immunogenic composition of any one of [76] to [81], or the
  • An isolated antibody comprising the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3 of BG 18.11, BG 18.6, BG 18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
  • PRE2 PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1-69, VH5-51, VH3-33, or VH3-23; and (b) the VL of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2,
  • BG18 iGLl BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
  • a method for identifying a vaccine candidate variant HIV Env gpl20 polypeptide comprising
  • the plurality of variant HIV Env gp 120 polypeptides comprise variants of a parental HIV Env gpl20 polypeptide comprising one or more amino acid substitutions at a region selected from the VI loop comprising positions 131-154, V3 loop comprising positions 297-334, and ⁇ 19 sheet comprising positions 413-419, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • FIG. 1 Structural and biophysical properties of BG18 germline targeting trimers.
  • BG505 SOSIP is shown as a gray surface and the HCDR3s of BG18 and PGT122 are shown in purple and green, respectively.
  • conserveed residues contacted by the HCDR3 G324, D325, 1326, R327, Q328, H330, T415, and P417) are colored red.
  • MD39 is colored gray with the N332 and N392 glycans shown as green sticks.
  • the BG18 HC and LC are colored purple and cyan, respectively. Red indicates conserved contact residues as in a) c) Cryo-EM derived structure of BG18 iGLO in complex with BG505 N332-GT2 with MD64 stabilizing mutationsl9.
  • N332-GT2 is colored gray with the N332 and N392 glycans shown as green sticks.
  • the BG18 iGLO HC and LC are colored purple and cyan, respectively. The same residues as in a) and c) are colored red.
  • FIG. 1 Naive human B cell sorting with N332-GT trimers.
  • a) Gating strategy for N332-GT epitope-specific sorting of naive human B cells b) HCDR3 length distribution in N332- GT sorted naive B cells compared to control B cells from DeKosky et al38 and Jardine et al39.
  • the BG18 iGLO, HMP1. HMP42, and HMP43 HCDR3 sequences shown are SEQ ID NO: 88-91, respectively e) SPR binding dissociation constants of HMP epitope -specific FAbs from naive B cells isolated with N332-GT1 and -GT2. Dashed line indicates the limit of detection f) Low resolution cryo-EM reconstructions of BG18-like precursors HMP1, HMP42, and HMP43 bound to N332-GT5 trimer with a low-pass filtered reconstruction of BG18-iGL0 (yellow) bound to N332-GT2 (gray) shown for reference.
  • the approximate densities of the HMP Fabs are colored green, purple and blue for HMP1, HMP42, and HMP43, respectively based on comparison to BG18 iGLO.
  • the approximate densities of the trimers are colored gray.
  • FIG. 3 Immunization of BG18 gH B cell adoptive transfer recipient mice with N332-GT NPs.
  • FACS fluorescence-activated cell sorting
  • a) Representative examples of fluorescence-activated cell sorting (FACS) gating to identify epitope-specific (N332-GT2++/N332-GT2-KO-) B cells in BG18gH and WT mice
  • b) Frequency of epitope -specific B cells in non-immunized BG18gH and WT mice
  • d) Representative examples of FACS gating to identify epitope-specific CD45.2+ GC B cells 8 days after imm
  • FIG. 4 Model of BG18 bound to BG505 SOSIP that was used to guide GT design.
  • BG505 SOSIP gray; BG18 HC purple; BG18 LC, cyan; N332 glycan, green sticks.
  • conserveed residues contacted by the HCDR3 G324, D325, 1326, R327, Q328, H330, T415, and P417) are colored red.
  • BG505 SOSIP gray; PGT122 HC, purple; PGT122 LC, cyan; N332 glycan, green sticks.
  • conserveed residues contacted by the HCDR3 are colored red as in a)
  • BG505 SOSIP is not shown in this alignment. MD39, gray; BG18 HC, purple; BG18 LC, cyan; N332 glycan, green sticks.
  • BG18/SOSIP model indicates that the HCDR1 R29 interacts with the N332 glycan.
  • SOSIP gray; BG18 HC, purple; N332 glycan, green sticks e) HCDR1 somatic mutation R29 was reverted to germline Isoleucine and tested for neutralization on a panel of 24 BG18-sensitive pseudoviruses. *IC50 values for BG18 were taken from Freund et all5.
  • FIG. 5 Minimally mutated BG18 variants and alternate VL variants by structural model-guided design, a) Neutralization IC50s on a panel of 24 BG18-sensitive pseudoviruses by less mutated variants of BG18 and variants of BG18 containing alternate LCs as indicated.
  • One variant (BG 18.11, "minBG18") is only 11% mutated in VHVL and was found to be 59% as broad as BG18 and had similar potency to BG18.
  • Another variant with 6% mutation in VHVL (BG18.6) neutralized 26% of the viruses.
  • IC50 values for BG18 were taken from Freund et all5.
  • VHVL somatic mutations were incorporated into each LC and paired with the mature BG18 HC.
  • FIG. 4a The model from Figure 4a is shown with VHVL somatic mutations (red spheres) in BG18, BG18.11 and BG18.6.
  • the essential VH mutations are localized to the HCDR1, whereas the VL mutations are localized to the LCDR2 and LCDR3.
  • VH4-4, BG18 VH, BG18.11 VH, BG18.6 VH, VL3-25, BG18 VL, BG18.11 VL, and BG18.6 VL comprise the amino acid sequences of SEQ ID NO: [VH4-4], 17, 19, 21, [VL3-25], 18, 20, and 22 respectively.
  • FIG. 6 Identifying BG18-like HCDR3s and estimating the BG18 precursor frequency in the human B cell repertoire, a) Table defining key BG18-like features and their frequencies in NGS datasets ofBCR HCs from HIV-uninfected donors. Frequencies for BG18-like feature sets i (HCDR3 main features) and ii (HCDR3 junction features) were measured from NGS data from Briney et al.18 for 10 donors for which sequencing used UIDs to discriminate PCR artifacts from true unique mRNA transcripts.
  • VH CDR3 of BG18, BG18 iGL0-iGL2, and Pre l to 15 have the amino acid sequence of SEQ ID NO: 94, 88, and 95-111, respectively c) Graph of the frequencies for Feature Sets i or (i and ii), as defined in a), for all 14 donors. Each symbol represents one donor, and the total number of sequences for each donor in the NGS dataset is given in the key. Black symbols are for the 10 donors from Briney et al.18 sequenced using UIDs, and red symbols are for the 4 donors sequenced here without the use of UIDs. Bars denote geometric mean and geometric standard deviation measured over the 10 UID donors only. Also see Methods for a detailed explanation of the BG18-like precursor frequency estimate.
  • FIG. 7 Mutations in N332-GT trimers and strategy for designing N332-GT5. a) GT mutations present in N332-GT trimers are localized to the VI, V3, and b 19. Amino acid changes relative to BG505-MD39 are shown in red. Sequences shown are CTNVTNNITDDMRGEL (SEQ ID NO: 71), CTNYAPNLLSNMRGEL (SEQ ID NO: 72),
  • CTNYAPNLLSNMRGEI (SEQ ID NO: 73), CTNYAPKLLSNMRGEI (SEQ ID NO: 74),
  • CTNY APKLRSMMRGEI (SEQ ID NO: 75), CTNYAPNLRSDMRGEI (SEQ ID NO: 76),
  • TRPNNNT SEQ ID NO: 77
  • TRPSNNT SEQ ID NO: 78
  • TGDIIGDIRQAHCNV S SEQ ID NO: 77
  • FIG. 8 Structural comparison of BG18 and BG18 iGLO complexes, a) HCDR3 alignment of BG18 (purple) and BG18 iGLO (pink) shows that there is a large conformational change in the HCDR1 of BG18 that brings it into proximity to the N332 glycan (green sticks). Despite limited resolution, the density map suggests that this rearrangement allows interaction with the N332 glycan through numerous HCDR1 side chain interactions, all of which are somatic mutations b) Effect of reverting HCDR1 residues D32 and H33 to germline in a less mutated variant of BG18 measured by ELISA binding to BG505-MD39.
  • the malleability of the LCDR2 loop is accentuated by the observation that it is fully resolved when in complex with the trimer but disordered in the published ligand-free crystal structure of the Fab alonel5.
  • the VI loop of BG505 MD64 N332-GT2 is shown in tan with the side chains of the most important mutations shown as sticks.
  • the side chain of K137 in VI fits a groove in the LC (dark blue) LCDR1, with the aliphatic portion forming a CH-p interaction with LCDR1 Y32 and the e-amino group a salt bridge with LCDR2 D51, as well as potential hydrogen bonding with nearby backbone carbonyls and the side chain of LFR3 S66.
  • Figure 10 Naive human B cell repertoire screening of llmutB, N332-GT1 and N332-GT2. In total, 57 million B cells were screened from 4 donors using l lmutB as a sorting probe a) Gating strategy for l lmutB epitope-specific sorting of naive human B cells b) HmutB+l lmutB+MD39neg B cells were sorted and clonal B cell lines were generated. Culture supernatants from >1000 clonal B cell lines were tested by ELISA for specificity to l lmutB and MD39.
  • KLH keyhole limpet hemocyanin
  • SA-PE streptavidin- phycoerythrin
  • HIV V3 peptide CRPNNNTRKSIRIGPGQAFYATGDIIGDIRQAHC
  • LCMV peptide DIYKGVYQFKSV
  • FIG. 11 Biophysical characterization of N332-GT2 nanoparticles, a) SPR KDs for N332-GT2 trimer and NP binding to BG18 precursors. The NP was captured as ligand with PGT128, and Fabs were flowed as analytes b) SPR analysis of N332-GT2 NP binding to a panel of non-nAb and bnAbs. The format of the experiment was the same as in a) c) SECMALS analysis of N332-GT2 NPs. The black line indicates the UV trace and the red line indicates the protein molecular weight. [0035] Figure 12. Calibration of transferred CD45.2 B cells into CD45.1 recipient mice.
  • Splenocytes from transferred recipient mice were analyzed by flow cytometry.
  • CountBright Beads (Invitrogen) were added to the samples (5 xl06 splenocytes - quantified by Nucleocounter, ChemoMetec USA) to determine exact cell population numbers a) The raw data acquired at the flow cytometer b) The normalized values in the stained samples, obtained by multiplying the raw data by the beads multiplication factor (starting beads / beads measured) c) The total cell numbers for the mouse spleens, obtained by multiplying the values for 5 xl06 splenocytes by the spleen multiplication factor (total splenocytes / 5).
  • FIG. 13 Overall strategy for designing GT immunogens to prime precursors of HCDR3-dependent antibodies.
  • the BCR HC repertoires of 14 HIV-negative donors were sequenced and queried bioinformatically to identify HCDR3s with key features similar to a known bnAb indicating that such HCDR3s have potential to contribute to bnAb precursors. That process validated the bnAb for germline targeting immunogen design.
  • the human repertoire is shown in green with HCDR3 -dependent bnAb precursors shown in red.
  • the second panel shows the iterative immunogen design process.
  • HIV Env variant polypeptides and immunogenic compositions comprising thereof that are capable of eliciting the formation of antibodies with binding properties similar to the BG18 broadly neutralizing antibody.
  • the inventors used next generation sequencing (NGS) and bioinformatics to identify in HIV-uninfected donors germline encoded heavy chain CDRs that share key properties with the heavy chain CDR3 -dependent broadly neutralizing anti-HIV antibody (bnAb) BG18.
  • NGS next generation sequencing
  • bioinformatics to identify in HIV-uninfected donors germline encoded heavy chain CDRs that share key properties with the heavy chain CDR3 -dependent broadly neutralizing anti-HIV antibody (bnAb) BG18.
  • BG18 inferred-germline (iGL) variants with the NGS-derived HCDR3 loops were used in a structural-model-guided, multitarget mammalian- display selection to identify variant Env polypeptides, including variant Env trimers that bind to diverse BG18 iGL antibodies and potential BG18 precursor antibodies.
  • the N332-GT trimers were used to elicit an immune response.
  • the inventors found that the variant Env polypeptides disclosed herein were capable of inducing the formation of BG18-like antibodies. As such, the HIV Env variant polypeptides and immunogenic compositions disclosed herein have the potential to elicit a protective broadly neutralizing response against HIV.
  • HIV human immunodeficiency virus
  • HIV-Type 1 HIV-1
  • HIV-Type 2 HIV-2
  • retroviruses e.g., simian immunodeficiency virus (SIV)
  • SIV simian immunodeficiency virus
  • an HIV virus is an HIV -Type -1 virus. Previous names for HIV include human T-lymphotropic virus- 111 (HTLV-III), lymphadenopathy-associated virus (LA), and others.
  • AIDS Acquired immune deficiency syndrome
  • envelope glycoprotein refers to the glycoprotein that is expressed on the surface of the envelope of HIV virions and the surface of the plasma membrane of HIV infected cells.
  • envelope glycoprotein encompass, but are not limited to, native Env, an isoform of Env, or a recombinant variant of Env (e.g., SOSIP) derived from an HIV isolate.
  • Env is the sole virally encoded gene product on the surface of the virus and, as such, is the only target of neutralizing antibodies.
  • Env is a trimer of heterodimers composed of two non-covalently associated subunits: the receptor-binding gpl20 and the fusion machinery-containing gp41. Each subunit is derived from a gp 160 precursor glycoprotein following cleavage by cellular ftirins. HIV- 1 gpl20 binds the CD4 molecule on the surface of human target T cells to initiate the viral entry process, and following co-receptor engagement, fusion is mediated by gp41. gpl40 env is the uncleaved ectodomain of gpl60.
  • the Env is a BG505, 92BR020, JR-FL, YU-2, or JR-CSF Env polypeptide. In some embodiments, the Env is a BG505 Env polypeptide. UniProtKB accession number Q2N0S5-1, Q2N0S6-1, and Q2N0S7-1 provide BG505 env gpl60 polypeptide sequences. In some embodiments, the Env is a well-ordered Env trimer. In some embodiments, the BG505 Env polypeptide has the amino acid sequence of SEQ ID NO: 2.
  • well-ordered Env trimer or "well-ordered trimer” as used herein refers to an envelope glycoprotein trimer comprising three cleaved gpl40 polypeptides that closely mimic the quaternary structure of the Env ectodomain on the surface of the envelope of HIV or SIV virions and the surface of the plasma membrane of HIV or SIV infected cells.
  • the gpl40 polypeptides comprise MPER.
  • the well-ordered trimer comprises three MPERs.
  • the gpl20 and gp41 ectodomain is linked by a covalent linkage, for example, a disulfide bond.
  • the gpl40 polypeptide comprises one or more mutations to promote trimer formation. In one embodiment, the gpl40 polypeptide comprises one or more Cys substitutions to promote disulfide formation.
  • the well-ordered trimer is a SOSIP gpl40 trimer. Well-ordered SOSIP trimers have been disclosed in US Patent Appl. Pub. No. 2014/0212458, and Sanders, R. W. et ah, PLoS Pathog. 9, e l003618 (2013), each of which is incorporated by reference herein in its entirety. In one embodiment, a well-ordered trimer is formed from a clade A Env.
  • a well-ordered trimer is formed from a clade B Env. In one embodiment, a well-ordered trimer is formed from a clade C Env. In one embodiment, a well-ordered trimer is formed from a circulating recombinant form Env, wherein 'circulating recombinant form' (CRF) refers to a hybrid virus comprising a combination of genetic material from different subtypes. In one embodiment, a well-ordered trimer is Dul56.12 SOSIP comprising MPER.
  • a well-ordered Env trimer is a native flexibly linked (NFL) trimer as described in Shama, et ak, Cell Reports, l l(4):539-50 (2015).
  • a well- ordered Env trimer is a DS-SOSIP as described in Chuang GY, et al, J. Virology, 91(10). pii: e02268-16 (2017).
  • a well-ordered trimer is formed from an SIV Env.
  • a well-ordered trimer is an SIV Env SOSIP.
  • a well-ordered trimer is formed from an Env comprising a mutation (e.g., substitution or deletion) in the CD4 binding site. In one embodiment, a well-ordered trimer is formed from an Env comprising a mutation (e.g., substitution or deletion) in the CD4 binding site wherein the mutation reduces or disrupts the binding between Env and CD4. In one embodiment, a well-ordered trimer is a CRF or C108 SOSIP. See, e.g., Andrabi R., et al, Immunity 43(5): 959-973 (2015).
  • the gpl20 and gp41 ectodomain is linked by a peptide linker, for example, a Gly-Ser linker, as described in Georgiev IS, et al., J. Virology 89(10): 5318-5329 (2015).
  • the well-ordered Env trimer is stable.
  • antibody means an immunoglobulin molecule (or a group of immunoglobulin molecules) that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule.
  • a target such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule.
  • the terms “antibody” and “antibodies” are terms of art and can be used interchangeably herein and refer to a molecule with an antigen-binding site that specifically binds an antigen.
  • Antibodies can include, for example, monoclonal antibodies, recombinantly produced antibodies, human antibodies, humanized antibodies, resurfaced antibodies, chimeric antibodies, immunoglobulins, synthetic antibodies, tetrameric antibodies comprising two heavy chain and two light chain molecules, an antibody light chain monomer, an antibody heavy chain monomer, an antibody light chain dimer, an antibody heavy chain dimer, an antibody light chain- antibody heavy chain pair, intrabodies, heteroconjugate antibodies, single domain antibodies, monovalent antibodies, single chain antibodies or single-chain Fvs (scFv), affybodies, Fab fragments, F(ab')2 fragments, disulfide-linked Fvs (sdFv), anti-idiotypic (anti-id) antibodies (including, e.g., anti-anti- Id antibodies), bispecific antibodies, and multi-specific antibodies.
  • monoclonal antibodies recombinantly produced antibodies
  • human antibodies humanized antibodies, resurfaced antibodies
  • chimeric antibodies immunoglobulins
  • antibodies described herein refer to polyclonal antibody populations.
  • Antibodies can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, or IgY), any class (e.g., IgGi, IgG 2 , IgG , IgG 4 , IgAi, or IgA 2 ), or any subclasses (isotypes) thereof (e.g. IgGl, IgG2, IgG3, IgG4, IgAl and IgA2), of immunoglobulin molecule, based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu, respectively.
  • the different classes of immunoglobulins have different and well-known subunit structures and three-dimensional configurations.
  • Antibodies can be naked or conjugated or fused to other molecules such as toxins, radioisotopes, other polypeptides etc.
  • antigen-binding domain As used herein, the terms "antigen-binding domain,” “antigen-binding region,” “antigen binding site,” and similar terms refer to the portion of antibody molecules, which comprises the amino acid residues that confer on the antibody molecule its specificity for the antigen (e.g., HIV Env MPER).
  • the antigen-binding region can be derived from any animal species, such as mouse and humans.
  • variable region or “variable domain” are used interchangeably and are common in the art.
  • CDRs complementarity determining regions
  • FR framework regions
  • variable region comprises 3 CDRs (CDR1, CDR2, and CDR3) and 4 framework regions (FR1, FR2, FR3, and FR4) in the order of FR1-CDR1-FR2-CDR2-FR3-CDR3- FR4 from the N terminus to the C terminus.
  • variable region is a human variable region.
  • variable region comprises human CDRs and human framework regions (FRs).
  • the variable region comprises CDRs and framework regions (FRs) wherein one or more of the CDRs were modified by a substitution, deletion, or insertion relative to the CDRs of a parental antibody.
  • variable region comprises CDRs and framework regions (FRs) wherein one or more of the FRs were modified by a substitution, deletion, or insertion relative to the FRs of a parental antibody.
  • variable region comprises CDRs and framework regions (FRs) wherein one or more of the CDRs and one or more of the FRs were modified by a substitution, deletion, or insertion relative to the CDRs and FRs of a parental antibody.
  • the parental antibody is PGZL1.
  • the variable region comprises human CDRs and primate (e.g., non-human primate) framework regions (FRs).
  • CDR sequences are identified according to Rabat.
  • the CDR sequences are identified according to Chothia. It is understood that the identification of CDRs in a variable region also identifies the FRs as the sequences flanking the CDRs.
  • the Kabat numbering system is generally used when referring to a residue in the variable domain (approximately residues 1-107 of the light chain and residues 1-113 of the heavy chain) (e.g., Kabat EA, et al., Sequences of Immunological Interest. (5th Ed., 1991, National Institutes of Health, Bethesda, Md.) ("Kabat").
  • the amino acid position numbering as in Kabat refers to the numbering system used for heavy chain variable domains or light chain variable domains of the compilation of antibodies in Kabat EA, et al. (Sequences of Immunological Interest. (5th Ed., 1991, National Institutes of Health, Bethesda, Md.), "Kabat”).
  • the actual linear amino acid sequence can contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FR or CDR of the variable domain.
  • a heavy chain variable domain can include a single amino acid insert (residue 52a according to Kabat) after residue 52 of H2 and inserted residues (e.g.
  • residues 82a, 82b, and 82c, etc. according to Kabat after heavy chain FR residue 82.
  • the Kabat numbering of residues can be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a "standard” Kabat numbered sequence. Chothia refers instead to the location of the structural loops (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987)).
  • the end of the Chothia CDR-H1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; if neither 35A nor 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35A and 35B are present, the loop ends at 34).
  • the AbM hypervariable regions represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular's AbM antibody modeling software, available, for example, at bioinf.org.uk/abs/software.
  • the CDR sequences are identified according to Kabat.
  • the CDR sequences are identified according to Chothia. In some embodiments, the CDR sequences are identified according to AbM. In some embodiments, the VH CDR3 sequence is identified according to Kabat. In some embodiments, the VH CDR3 sequence is identified according to Chothia. In some embodiments, the VH CDR3 sequence is identified according to AbM. Loop Kabat AbM Chotbia
  • VL and “VL domain” are used interchangeably to refer to the light chain variable region of an antibody.
  • VH and "VH domain” are used interchangeably to refer to the heavy chain variable region of an antibody.
  • antibody fragment refers to a portion of an intact antibody.
  • antigen-binding fragment refers to a portion of an intact antibody that binds to an antigen.
  • An antigen-binding fragment can contain the antigenic determining variable regions of an intact antibody. Examples of antibody fragments include, but are not limited to Fab, Fab', F(ab')2, and Fv fragments, linear antibodies, and single chain antibodies.
  • a "monoclonal” antibody or antigen-binding fragment thereof refers to a homogeneous antibody or antigen-binding fragment population involved in the highly specific recognition and binding of a single antigenic determinant, or epitope. This is in contrast to polyclonal antibodies that typically include different antibodies directed against different antigenic determinants.
  • the term "monoclonal” antibody or antigen-binding fragment thereof encompasses both intact and full- length monoclonal antibodies as well as antibody fragments (such as Fab, Fab', F(ab')2, Fv), single chain (scFv) mutants, fusion proteins comprising an antibody portion, and any other modified immunoglobulin molecule comprising an antigen recognition site.
  • monoclonal antibody or antigen-binding fragment thereof refers to such antibodies and antigen-binding fragments thereof made in any number of manners including but not limited to by hybridoma, phage selection, recombinant expression, and transgenic animals.
  • polyclonal antibody describes a composition of different (diverse) antibody molecules, which are capable of binding to or reacting with several different specific antigenic determinants on the same or on different antigens. Usually, the variability of a polyclonal antibody is primarily located in the so-called variable regions of the polyclonal antibody, in particular in the CDR regions.
  • a mixture of two or more polyclonal antibodies is produced in one mixture from a polyclonal polycomposition cell line, which is produced from two or more parental polyclonal cell lines each expressing antibody molecules, which are capable of binding to a distinct target, but it may also be a mixture of two or more polyclonal antibodies produced separately.
  • a mixture of monoclonal antibodies providing the same antigen/epitope coverage as a polyclonal antibody described herein will be considered as an equivalent of a polyclonal antibody.
  • chimeric antibodies or antigen-binding fragments thereof refers to antibodies or antigen-binding fragments thereof wherein the amino acid sequence is derived from two or more species.
  • the variable region of both light and heavy chains corresponds to the variable region of antibodies or antigen-binding fragments thereof derived from one species of mammals (e.g., mouse) with the desired specificity, affinity, and capability, while the constant regions are homologous to the sequences in antibodies or antigen-binding fragments thereof derived from another (usually human) to avoid eliciting an immune response in that species.
  • epitopes or "antigenic determinant” are used interchangeably herein and refer to that portion of an antigen capable of being recognized and specifically bound by a particular antibody.
  • the antigen is a polypeptide
  • epitopes can be formed both from contiguous amino acids and noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained upon protein denaturing, whereas epitopes formed by tertiary folding are typically lost upon protein denaturing.
  • An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
  • Binding affinity generally refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity” refers to intrinsic binding affinity, which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (K D ). Affinity can be measured by common methods known in the art, including those described herein.
  • an antibody described herein binds to N332- GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 with a K D of at least about 0.1 mM or less, at least about 0.01 mM or less, at least about 1 nM or less, or at least about 0.1 nM or less.
  • an anti-HIV antibody described herein binds to N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 with a K D of at least about 0.01 pM or less.
  • an anti- HIV antibody described herein is capable of binding to cells that express functional, well-ordered HIV-1 membrane Env trimers.
  • an anti -HIV antibody described herein is capable of binding to cells that display N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332- GT5 in a flow cytometry assay.
  • an anti-HIV antibody described herein is capable of binding to N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 in biolayer interferometry (BLI) assay. In one embodiment, an anti-HIV antibody described herein is capable of binding to N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 in ELISA. In one embodiment, an anti-HIV antibody described herein is capable of binding Env trimers from detergent-solubilized HIV-1 virions in an ELISA assay. In one embodiment, an anti-HIV antibody described herein is capable of binding Env trimers from detergent-solubilized HIV-1 virions in a BN-PAGE gel mobility-shift assay.
  • binding affinity refers to a stronger binding between a molecule and its binding partner.
  • “Or better” when used herein refers to a stronger binding, represented by a smaller numerical KD value.
  • an antibody which has an affinity for an antigen of "0.6 nM or better"
  • the antibody's affinity for the antigen is ⁇ 0.6 nM, i.e. 0.59 nM, 0.58 nM, 0.57 nM etc. or any value less than 0.6 nM.
  • immunospecifically binds As used herein, the terms “immunospecifically binds,” “immunospecifically recognizes,” “specifically binds,” and “specifically recognizes” are analogous terms in the context of antibodies and refer to molecules that bind to an antigen (e.g., epitope or immune complex) as such binding is understood by one skilled in the art.
  • an antigen e.g., epitope or immune complex
  • a molecule that specifically binds to an antigen can bind to other peptides or polypeptides, generally with lower affinity as determined by, e.g., immunoassays, BIAcore ® , KinExA 3000 instrument (Sapidyne Instruments, Boise, ID), ELISA, biolayer interferometry (BLI), flow cytometry or other assays known in the art.
  • molecules that immunospecifically bind to an antigen bind to the antigen with a KD that is at least 2 logs, 2.5 logs, 3 logs, or 4 logs lower than the KD when the molecules bind non- specifically to another antigen.
  • the antibody may specifically bind to cells that express functional, well-ordered HIV-1 membrane Env trimers. In one example, the antibody may specifically bind to cells that display MPER-TM peptide. In one example, the antibody may specifically bind to the MPER peptide. In one example, the antibody may specifically bind to the MPER peptide in biolayer interferometry (BLI) assay. In one example, the antibody may specifically bind to the MPER peptide in ELISA assay. In one example, the antibody may specifically bind to Env trimers from detergent-solubilized HIV-1 virions. In one example, the antibody may specifically bind to Env trimers from detergent-solubilized HIV-1 virions in an ELISA assay.
  • BLI biolayer interferometry
  • the antibody may specifically bind to Env trimers from detergent- solubilized HIV-1 virions in a BN-PAGE gel mobility-shift assay.
  • the antibody may bind to N332- GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 with a K D at least 2 logs, 2.5 logs, 3 logs, or 4 logs lower than KD of binding to other viral or non-viral polypeptides.
  • An antibody that specifically binds to Env encompass, but are not limited to, antibodies that specifically bind to native Env, an isoform of Env, or a variant of Env (e.g., SOSIP) derived from an HIV isolate.
  • preferentially binds it is meant that the antibody specifically binds to an epitope more readily than it would bind to a related, similar, homologous, or analogous epitope.
  • an antibody, which "preferentially binds" to a given epitope would more likely bind to that epitope than to a related epitope, even though such an antibody may cross-react with the related epitope.
  • An antibody is said to "competitively inhibit" binding of a reference antibody to a given epitope if it preferentially binds to that epitope or an overlapping epitope to the extent that it blocks, to some degree, binding of the reference antibody to the epitope.
  • Competitive inhibition may be determined by any method known in the art, for example, competition ELISA assays.
  • An antibody may be said to competitively inhibit binding of the reference antibody to a given epitope by at least 90%, at least 80%, at least 70%, at least 60%, or at least 50%.
  • bnAb narrowly neutralizing antibody
  • HIV e.g., HIV-1
  • bnAb broadly neutralizing antibody
  • a broadly neutralizing antibody inhibits infection of a susceptible target cell by HIV.
  • a broadly neutralizing antibody specifically binds an HIV Env and inhibits infection of a susceptible target cell (e.g., TZM-bl) by an HIV pseudovirus comprising an Env polypeptide.
  • HIV pseudovirus neutralization assays have been disclosed in the art, for example, in Walker L.M., et al, Nature 477, 466-470 (2011), Li M., et ak, J. Virol. 79: 10108- 10125 (2005), each of which is incorporated herein by reference in its entirety for all purposes.
  • a broadly neutralizing antibody neutralizes 2, 3, 4, 5, 6, 7, 8, 9, or more HIV strains or pseudoviruses.
  • a broadly neutralizing antibody neutralizes 2, 3, 4, 5, 6, 7, 8, 9, or more HIV strains or pseudoviruses that belong to the same or different clades.
  • a broadly neutralizing antibody is capable of neutralizing HIV strains or pseudoviruses from at least two different clades.
  • a broadly neutralizing antibody is capable of neutralizing at least one clade B strain or pseudovirus and one clade C strain or pseudovirus. In one embodiment, a broadly neutralizing antibody is capable of neutralizing more than one clade B strain or pseudovirus and more than one clade C strain or pseudovirus. In one embodiment, a broadly neutralizing antibody is capable of neutralizing an HIV strain or pseudovirus from at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or at least eleven clades represented in the 130- member indicator virus panel.
  • a broadly neutralizing antibody is capable of neutralizing an HIV strain or pseudovirus from at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, or all fifteen clades selected from the group consisting of clades A, A (T/F), AC, ACD, B, B (T/F), BC, C, C (T/F), CD, CRF01 AE, CRF01 AE (T/F), CRF02 AG, D, and G.
  • a broadly neutralizing antibody is capable of neutralizing an HIV strain or pseudovirus from at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or all eleven clades selected from the group consisting of clades A, AC, ACD, AE, AG, B, BC, C, CD, D, G.
  • the breadth of neutralization is tested on an indicator virus panel comprising cross-clade HIV isolates.
  • the virus panel comprises the 9 cross- clade isolates of 92TH021, JRCSF, C22, 94UG103, 92BR020, Dul56.12, HIV-2, HIV-2C1, and HIV-2C4.
  • the virus panel comprises the 130 cross-clade isolates of NIH-
  • NAC_92TH021 NIH-081_BJOX028000.10.3, NIH-078_BJOX015000.11.5, NIH- 079_BJOX010000.06.2, NIH-074_C4118.c09, NIH-069_C1080.c03, NIH-
  • a broadly neutralizing antibody is capable of neutralizing at least 4, 5, 6, 7, 8 or 9 of the cross-clade HIV isolates in the 9-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least 6 of the cross- clade HIV isolates in the 9-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 50%, 60%, 70%, 80%, 90%, 95%, or 100% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 60% of cross-clade HIV isolates in the 130-member indicator virus panel.
  • a broadly neutralizing antibody is capable of neutralizing at least about 70% of cross-clade HIV isolates in the 130- member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 75% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 80% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 90% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 95% of cross-clade HIV isolates in the 130-member indicator virus panel.
  • a broadly neutralizing antibody is capable of neutralizing at least about 98% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 100% of cross-clade HIV isolates in the 130-member indicator virus panel.
  • the phrase "substantially similar,” or “substantially the same”, as used herein, denotes a sufficiently high degree of similarity between two numeric values (generally one associated with an antibody described herein and the other associated with a reference/comparator antibody) such that one of skill in the art would consider the difference between the two values to be of little or no biological and/or statistical significance within the context of the biological characteristic measured by said values (e.g., KD values).
  • the difference between said two values can be less than about 50%, less than about 40%, less than about 30%, less than about 20%, or less than about 10% as a function of the value for the reference/comparator antibody.
  • a polypeptide, antibody, polynucleotide, vector, cell, or composition, which is "isolated” is a polypeptide, antibody, polynucleotide, vector, cell, or composition, which is in a form not found in nature.
  • Isolated polypeptides, antibodies, polynucleotides, vectors, cell or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature.
  • an antibody, polynucleotide, vector, cell, or composition, which is isolated is substantially pure.
  • substantially pure refers to material, which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.
  • polypeptide polypeptide
  • peptide protein
  • the terms “polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to polymers of amino acids of any length.
  • the polymer can be linear or branched, it can comprise modified amino acids, and it can be interrupted by non-amino acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids, etc.
  • the polypeptides described herein are based upon antibodies, in certain embodiments, the polypeptides can occur as single chains or associated chains.
  • nucleic acids or polypeptides refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity.
  • the percent identity can be measured using sequence comparison software or algorithms or by visual inspection.
  • sequence comparison software or algorithms or by visual inspection.
  • Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences.
  • One such non-limiting example of a sequence alignment algorithm is the algorithm described in Karlin S., et al, Proc. Natl. Acad.
  • Gapped BLAST can be used as described in Altschul SF, et al., Nucleic Acids Res. 25:3389-3402 (1997).
  • BLAST-2 Altschul SF, et al., Methods in Enzymology, 266:460-480 (1996)), ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or Megalign (DNASTAR) are additional publicly available software programs that can be used to align sequences.
  • the percent identity between two nucleotide sequences is determined using the GAP program in GCG software (e.g., using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 90 and a length weight of 1, 2, 3, 4, 5, or 6).
  • the GAP program in the GCG software package which incorporates the algorithm ofNeedleman and Wunsch (J. Mol. Biol. (48):444-453 (1970)) can be used to determine the percent identity between two amino acid sequences (e.g., using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5).
  • the percent identity between nucleotide or amino acid sequences is determined using the algorithm of Myers and Miller (CABIOS, 4: 11-17 (1989)).
  • the percent identity can be determined using the ALIGN program (version 2.0) and using a PAM120 with residue table, a gap length penalty of 12 and a gap penalty of 4. Appropriate parameters for maximal alignment by particular alignment software can be determined by one skilled in the art. In certain embodiments, the default parameters of the alignment software are used. In certain embodiments, the percentage identity "X" of a first amino acid sequence to a second sequence amino acid is calculated as 100 x (Y/Z), where Y is the number of amino acid residues scored as identical matches in the alignment of the first and second sequences (as aligned by visual inspection or a particular sequence alignment program) and Z is the total number of residues in the second sequence. If the length of a first sequence is longer than the second sequence, the percent identity of the first sequence to the second sequence will be higher than the percent identity of the second sequence to the first sequence.
  • whether any particular polynucleotide has a certain percentage sequence identity can, in certain embodiments, be determined using the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711). Bestfit uses the local homology algorithm of Smith and Waterman (Advances in Applied Mathematics 2: 482 489 (1981)) to find the best segment of homology between two sequences.
  • the parameters are set such that the percentage of identity is calculated over the full length of the reference nucleotide sequence and that gaps in identity of up to 5% of the total number of nucleotides in the reference sequence are allowed.
  • two nucleic acids or polypeptides described herein are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection.
  • Identity can exist over a region of the sequences that is at least about 10, about 20, about 40-60 residues in length or any integral value there between, and can be over a longer region than 60-80 residues, for example, at least about 90-100 residues, and in some embodiments, the sequences are substantially identical over the full length of the sequences being compared, such as the coding region of a nucleotide sequence for example.
  • a "conservative amino acid substitution" is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • substitution of a phenylalanine for a tyrosine is a conservative substitution.
  • conservative substitutions in the sequences of the polypeptides and antibodies described herein do not abrogate the binding of the polypeptide or antibody containing the amino acid sequence, to the antigen(s).
  • Methods of identifying nucleotide and amino acid conservative substitutions, which do not eliminate antigen binding are well-known in the art (see, e.g., Brummell DA, et al., Biochem. 32: 1180-1 187 (1993); Kobayashi et al., Protein Eng. 12(10):879-884 (1999); and Burks EA, et ak, Proc. Natl. Acad. Sci. USA 94: 412-417 (1997)).
  • treatment refers to treatment of an infected person.
  • treating includes alleviating or reducing at least one adverse or negative effect or symptom of a condition, disease or disorder. This condition, disease or disorder can be HIV infection.
  • Terms such as “treating” or “treatment” or “to treat” or “alleviating” or “to alleviate” refer to therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder, such as HIV or AIDS. Thus, those in need of treatment include those already diagnosed with or suspected of having the disorder.
  • a subject is successfully "treated” for the disorder according to the methods described herein if the patient shows one or more of the following: a reduction in the number of or complete absence of viral load; a reduction in the viral burden; inhibition of or an absence of the virus into peripheral organs; relief of one or more symptoms associated with the disorder; reduced morbidity and mortality; improvement in quality of life; increased progression-free survival (PFS), disease-free survival (DFS), or overall survival (OS), complete response (CR), partial response (PR), stable disease (SD), a decrease in progressive disease (PD), a reduced time to progression (TTP), or any combination thereof.
  • PFS progression-free survival
  • DFS disease-free survival
  • OS overall survival
  • C complete response
  • PR partial response
  • SD stable disease
  • PD progressive disease
  • TTP time to progression
  • prevention refers to preventing a subject from becoming infected with, or reducing the risk of a subject from becoming infected with, or halting transmission of, or the reducing the risk of transmission of a virus.
  • Prophylactic or preventative measures refer to measures that prevent and/or slow the development of a targeted pathological condition or disorder.
  • those in need of prophylactic or preventative measures include those prone to have the disorder and those in whom the disorder is to be prevented.
  • prevention encompasses passive immunization of a subject in need thereof comprising administering an effective amount of an antibody described herein.
  • an “effective amount” refers to an amount effective, at dosages, and for periods of time necessary, to achieve the desired result with respect to the treatment of the relevant disorder, condition, or side effect.
  • An “effective amount” can be determined empirically and in a routine manner, in relation to the stated purpose.
  • the effective amount of components of the present invention will vary from patient to patient not only with the particular vaccine, component or composition selected, the route of administration, and the ability of the components to elicit a desired result in the individual, but also with factors such as the disease state or severity of the condition to be alleviated, hormone levels, age, sex, weight of the individual, the state of being of the patient, and the severity of the pathological condition being treated, concurrent medication or special diets then being followed by the particular patient, and other factors, which those skilled in the art will recognize, with the appropriate dosage being at the discretion of the attending physician. Dosage regimes may be adjusted to provide the improved therapeutic response. An effective amount is also one in which any toxic or detrimental effects of the components are outweighed by the therapeutically beneficial effects.
  • the term "therapeutically effective amount” refers to an amount of an antibody, recombinant virus, immunoconjugate, or other drug effective to "treat” a disease or disorder in a subject or mammal. To the extent an antibody can prevent growth and/or kill existing cells, it can be cytostatic and/or cytotoxic.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • subject refers to an animal, for example a human, to whom treatment, including prophylactic treatment, with the antibody or pharmaceutical composition according to the present disclosure, is provided.
  • the subject, individual, or patient has been infected with HIV.
  • the subject, individual, or patient suffers from AIDS.
  • the subject, individual, or patient has been exposed to HIV.
  • the subject, individual, or patient is at risk of being exposed to HIV.
  • Administration "in combination with” one or more further therapeutic agents includes simultaneous (concurrent) or consecutive administration in any order.
  • composition refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem complications commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable” or “pharmaceutical formulation” refers to a preparation, which is in such form as to permit the biological activity of the active ingredient to be effective, and which contains no additional components, which are unacceptably toxic to a subject to which the formulation would be administered.
  • the formulation can be sterile.
  • ART antiretroviral therapy
  • NRTIs nucleoside reverse transcriptase inhibitors
  • NRTIs non-nucleoside reverse transcriptase inhibitors
  • Pro protease inhibitors
  • fusion inhibitors entry inhibitors, maturation inhibitors, cellular inhibitors, integrase strand transfer inhibitors, and multi-class combinations.
  • Such drugs include, but are not limited to, lamivudine and zidovudine, emtricitabine (FTC), zidovudine (ZDV), azidothymidine (AZT), lamivudine (3TC), zalcitabine, dideoxycytidine (ddC), tenofovir disoproxil fumarate (TDF), didanosine (ddl), stavudine (d4T), abacavir sulfate (ABC), etravirine (ETR), delavirdine (DLV), efavirenz (EFV), nevirapine (NVP), amprenavir (APV), tipranavir (TPV), indinavir (IDV), saquinavir, saquinavir mesylate (SQV), lopinavir (LPV), ritonavir (RTV), fosamprenavir calcium (FOS-APV), ritona
  • ART drugs can also include antibodies that target HIV proteins or cellular proteins associated with disease progression. Also included are immune-based therapies, such as IL-2, IL-12, and alpha- epibromide. Each of these drugs can be administered alone or in combination with any other ART drug or any HIV-specific neutralizing antibody, such as a broadly neutralizing antibody, which is incorporated by reference herein in its entirety for all purposes.
  • a reservoir activator comprises a histone deacytelase (HDAC) inhibitor (e.g., romidepsin, vorinostat, and panobinostat), immunologic activator (e.g., cytokines and TLR agonists), or a dedicated small molecule drug.
  • HDAC histone deacytelase
  • immunomodulator refers to an agent, such as an antibody or peptide, which is capable of increasing, inducing, or extending an immune response (e.g., a cell- mediated immune response and/or a humoral immune response) when administered to a subject (e.g., a human, e.g., a human infected with HIV or at risk of an HIV infection or transmission).
  • Immunomodulators include, but are not limited to immune checkpoint inhibitors, for example, a PD-1, PD-L1, LAG-3, or TIG IT antagonist.
  • an immunomodulator used in the methods described herein comprises an anti -PD-1 antibody, anti-PD-Ll antibody, anti-LAG3 antibody, or an anti-TIGIT antibody.
  • An immunomodulator can be administered in conjunction with (e.g., prior to, concurrently with, or subsequent to, or within the context of a treatment regimen that includes the administration of a broadly neutralizing antibody described herein.
  • each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.
  • an isolated polypeptide described herein comprises a variant gpl60 polypeptide. In some embodiments, an isolated polypeptide described herein comprises a variant gpl40 polypeptide. In some embodiments, an isolated polypeptide described herein comprises a variant gp 120 polypeptide. It is understood that in some embodiments, a variant gp 160 or gpl40 polypeptide described herein comprises a variant gpl20 polypeptide described herein.
  • an isolated polypeptide described herein is capable of binding to a BG18 inferred-germline (iGL) variant antibody or a BG18-like native antibody described herein.
  • the BG18 iGL is BG18 iGLO, BG18 iGLl, BG18 iGL2.
  • a variant HIV Env gpl20 polypeptide described herein comprises one or more of
  • a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPNLLSNMRGEL (SEQ ID NO: 72) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of TGDIIGDIRQAHCNVS (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDIIGDIRMAHCNVS (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
  • FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; and
  • a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • a variant HIV Env gpl20 polypeptide described herein comprises
  • a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPNLLSNMRGEL (SEQ ID NO: 72) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprise an amino acid sequence of TGDIIGDIRQAHCNVS (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDIIGDIRMAHCNVS (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
  • FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; and
  • a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • a variant HIV Env gpl20 polypeptide described herein comprises one or more of
  • a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of CTNVTNNITDDMRGEL (SEQ ID NO: 71), CTNYAPNLLSNMRGEL (SEQ ID NO: 72), CTNYAPNLLSNMRGEI (SEQ ID NO: 73), CTNYAPKLLSNMRGEI (SEQ ID NO: 74), CTNY APKLRSMMRGEI (SEQ ID NO: 75), or CTNYAPNLRSDMRGEI (SEQ ID NO: 76);
  • a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of TRPNNNT (SEQ ID NO: 77), or TRPSNNT (SEQ ID NO: 78);
  • a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of TGDIIGDIRQAHCNVS (SEQ ID NO: 79), FGDIIGDIRMAHCNVS (SEQ ID NO: 80), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), or FGDVLGDVDMAKCTIS (SEQ ID NO: 83); and
  • b 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of SITLPCR (SEQ ID NO: 84), SIVLPCR (SEQ ID NO: 85), SLILPCR (SEQ ID NO: 86), or SLILPCW (SEQ ID NO: 87); wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • a variant HIV Env gpl20 polypeptide described herein comprises
  • a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of CTNVTNNITDDMRGEL (SEQ ID NO: 71), CTNYAPNLLSNMRGEL (SEQ ID NO: 72), CTNYAPNLLSNMRGEI (SEQ ID NO: 73), CTNYAPKLLSNMRGEI (SEQ ID NO: 74), CTNY APKLRSMMRGEI (SEQ ID NO: 75), or CTNYAPNLRSDMRGEI (SEQ ID NO: 76);
  • a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of TRPNNNT (SEQ ID NO: 77), or TRPSNNT (SEQ ID NO: 78);
  • a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of TGDIIGDIRQAHCNVS (SEQ ID NO: 79), FGDIIGDIRMAHCNV S (SEQ ID NO: 80), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), or FGDVLGDVDMAKCTIS (SEQ ID NO: 83); and
  • a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of SITLPCR (SEQ ID NO: 84), SIVLPCR (SEQ ID NO: 85), SLILPCR (SEQ ID NO: 86), or SLILPCW (SEQ ID NO: 87);
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • a variant HIV Env gp 120 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of
  • CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77),
  • CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77),
  • CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77),
  • CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78),
  • FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCW (SEQ ID NO: 87), respectively,
  • CTNYAPNLRSDMRGEI SEQ ID NO: 76
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGDVDMAKCTIS SEQ ID NO: 83
  • SLILPCR SEQ ID NO: 86
  • CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGHVRMAHCNIS SEQ ID NO: 82
  • SLILPCR SEQ ID NO: 86
  • CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGDVRMAHCNIS SEQ ID NO: 81
  • SLILPCR SEQ ID NO: 86
  • CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGHVRMAHCNIS SEQ ID NO: 82
  • SLILPCR SEQ ID NO: 86
  • CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPSNNT SEQ ID NO: 78
  • FGDVLGHVRMAHCNIS SEQ ID NO: 82
  • SLILPCR SEQ ID NO: 86
  • CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPSNNT SEQ ID NO: 78
  • FGDVLGHVRMAHCNIS SEQ ID NO: 82
  • SLILPCW SEQ ID NO: 87
  • CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution
  • TRPNNNT SEQ ID NO: 77
  • FGDVLGDVDMAKCTIS SEQ ID NO: 83
  • SLILPCR SEQ ID NO: 86
  • HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • a variant HIV Env gp 120 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77),
  • FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • a variant HIV Env gpl20 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77),
  • a variant HIV Env gpl20 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • a variant HIV Env gpl20 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • a variant HIV Env gpl20 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCW (SEQ ID NO: 87), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
  • a variant HIV Env gpl20 polypeptide described herein comprises N332.
  • a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env positions 1-35. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env positions 1-10. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env positions 10-20. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env positions 20-35.
  • a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env position 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, and 35. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N- terminal residue of HIV Env position 30. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of HIV Env position 31. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of HIV Env position 32. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • a variant HIV Env gp 120 polypeptide described herein comprises a C-terminal residue of one of HIV Env positions 503-512. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a C-terminal residue of one of HIV Env positions 503-508. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a C-terminal residue of one of HIV Env positions 508-512. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a C-terminal residue of HIV Env position 512. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • a variant HIV Env gpl20 polypeptide described herein is a variant of the BG505, BG505 N332, BG505 MD39, BG505 MD64, BG505 MD39 N332, BG505 MD39 1 ImutB, or BG505 MD39 17mutE gpl20 polypeptide.
  • a variant HIV Env gpl20 polypeptide described herein is a variant of the BG505 MD39 N332 polypeptide.
  • a variant HIV Env gpl20 polypeptide described herein is a variant of the BG505 MD64 N332 polypeptide.
  • a variant HIV Env gpl20 polypeptide described herein comprises K137 and H325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137 and P325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137, M141 and H325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137, R139, M141, and H325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137 and P325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137, M141, and P325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137, R139, M141, and P325. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • an isolated polypeptide described herein comprises
  • amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT1,
  • amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2,
  • amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT3,
  • amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT4,
  • amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT5, or
  • an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2-KO.
  • an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT1.
  • an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT2. In some embodiments, an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT3.
  • an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT4. In some embodiments, an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT5.
  • an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD64 N332- GT2.
  • an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT1, BG505 MD39 N332-GT2, BG505 MD39 N332-GT3, BG505 MD39 N332-GT4, BG505 MD39 N332-GT5, or BG505 MD39 N332-GT2-KO.
  • an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT1.
  • an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT2.
  • an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT3.
  • an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT4.
  • an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT5.
  • an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT1. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT2. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332- GT3. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT4. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT5. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD64 N332- GT2.
  • an isolated polypeptide described herein comprises K137 and H325. In some embodiments, an isolated polypeptide described herein comprises K137 and P325. In some embodiments, an isolated polypeptide described herein comprises K137, M141 and H325. In some embodiments, an isolated polypeptide described herein comprises K137, R139, M141, and H325. In some embodiments, an isolated polypeptide described herein comprises K137 and P325. In some embodiments, an isolated polypeptide described herein comprises K137, M141, and P325. In some embodiments, an isolated polypeptide described herein comprises K137, R139, M141, and P325. In some embodiments, an isolated polypeptide described herein comprises N332. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • an isolated polypeptide described herein specifically binds to the BG18 antibody. In some embodiments, an isolated polypeptide described herein specifically binds to an antibody selected from the group consisting of BG18.11 and BG18.6. In some embodiments, an isolated polypeptide described herein specifically binds to an antibody selected from the group consisting of BG18 iGLO, BG18 iGLl, and BG18 iGL2. In some embodiments, an isolated polypeptide described herein specifically binds to BG18 iGLO. In some embodiments, an isolated polypeptide described herein specifically binds to BG18 iGLl .
  • an isolated polypeptide described herein specifically binds to BG18 iGL2. In some embodiments, an isolated polypeptide described herein specifically binds to an antibody selected from the group consisting of PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, and PRE15. In some embodiments, an isolated polypeptide described herein specifically binds to the PG121 iGL antibody.
  • an isolated polypeptide described herein specifically binds to an antibody comprising the VH and VL of
  • the affinity of an isolated polypeptide disclosed for an antibody described herein can be determined experimentally using any suitable method well-known in the art, e.g., flow cytometry, enzyme-linked immunoabsorbent assay (ELISA), biolayer interferometry (BLI) assay, radioimmunoassay (RIA), or kinetics (e.g., BIACORETM analysis).
  • ELISA enzyme-linked immunoabsorbent assay
  • BLI biolayer interferometry
  • RIA radioimmunoassay
  • kinetics e.g., BIACORETM analysis.
  • Direct binding assays as well as competitive binding assay formats can be readily employed. (See, for example, Berzofsky, et ak, "Antibody-Antigen Interactions," In Fundamental Immunology, Paul, W. E., Ed., Raven Press: New York, N.Y.
  • the measured affinity of a particular antibody-antigen interaction can vary if measured under different conditions (e.g., salt concentration, pH, temperature).
  • affinity and other antigen-binding parameters e.g., KD or Kd, K on , K 0h
  • KD or Kd, K on , K 0h are made with standardized solutions of antibody and antigen, and a standardized buffer, as known in the art and such as the buffer described herein.
  • an isolated polypeptide described herein comprises a gpl40. In some embodiments, an isolated polypeptide described herein comprises a gpl20.
  • recombinant HIV Env trimers comprising an isolated polypeptide described herein.
  • a recombinant HIV Env trimer described herein is a homotrimer.
  • a recombinant HIV Env trimer described herein a heterotrimer.
  • a recombinant HIV Env trimer described herein comprises gpl20-gp41 heterodimers.
  • a recombinant HIV Env trimer described herein comprises gpl20-gp41 heterodimers wherein the heterodimers are covalently linked.
  • a recombinant HIV Env trimer described herein comprises gpl20-gp41 fusions. In some embodiments, a recombinant HIV Env trimer described herein is a stabilized trimer. In some embodiments, a recombinant HIV Env trimer described herein is an SOSIP, NFL or UFO trimer. In some embodiments, a recombinant HIV Env trimer described herein is an SOSIP trimer. In some embodiments, a recombinant HIV Env trimer described herein comprises a variant gpl20 polypeptide described herein.
  • a recombinant HIV Env trimer described herein comprises the amino acid sequence ofBG505 MD39 N332-GT1, BG505 MD39 N332-GT2, BG505 MD39 N332- GT3, BG505 MD39 N332-GT4, BG505 MD39 N332-GT5, or BG505 MD39 N332-GT2-KO.
  • a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT1.
  • a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT2.
  • a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT3. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT4. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT5. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD64 N332-GT2. In some embodiments, a recombinant HIV Env trimer described herein comprises K137 and H325.
  • a recombinant HIV Env trimer described herein comprises K137 and P325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137, M141 and H325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137, R139, M141, and H325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137 and P325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137, M141, and P325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137, R139, M141, and P325. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • Env-based trimeric candidate immunogens are engineered to eliminate cleavage between gpl20 and gp41 (so called uncleaved gpl40 trimers), usually generating imperfect mimetics of the functional spike based on antigenic profding or EM analysis (Tran K, et al. (2014) Proc Natl Acad Sci US All l(7):E738-747; Ringe RP, et al. (2013) Proc Natl Acad Sci U S A 110(45): 18256-18261). As agroup, the defined, or presumed to be, disordered trimers (in adjuvant) generate high self-binding antibody titers.
  • Antibodies elicited by these immunogens target epitopes exposed only on the free gpl20 and trimeric post-fusion forms of gp41 or disordered gpl40s and thus are ineffective at accessing their epitopes buried within the ordered, quaternary structure achieved in the native Env spike.
  • Applicants recently described the limitations of two CD4bs-directed non-bnAbs, (GE148 and GE136) generated following immunization of uncleaved gpl40 trimers (YU2 gpl40-foldon) in non-human primates (NHP).
  • Non- bnAbs represented by GE136 and 148, can only neutralize the sensitive so-called "tier I viruses" that are not representative of the more neutralization resistant tier 2-like primary isolates circulating in the human population.
  • tier I viruses a virus that are not representative of the more neutralization resistant tier 2-like primary isolates circulating in the human population.
  • EM reconstructions, paratope scanning and molecular modeling Applicants determined that these vaccine-elicited antibodies fail to reach the CD4bs due to steric barriers imposed by quaternary packing of the native Env on neutralization resistant primary isolates, a property that Applicants use to Applicants' advantage in the negative -selection strategy presented here (Tran K, et al. (2014) Proc Natl Acad Sci US A 11 l(7):E738-747).
  • the BG505 SOSIP and KNHI 144 SOSIP trimers are derived from the Env sequences of the subtype A BG505 and KNHI 144 strains. These soluble trimers possess an engineered disulfide linkage between the gpl20 and gp41 (at residues 501C and 605C, respectively) and an additional mutation in the heptad repeat I (HRI) of gp41 (1559P) that facilitates trimerization (Binley JM, et al. (2000) J Virol 74(2):627-643; Sanders RW, et al. (2002) J Virol 76(17):8875-8889).
  • HRI heptad repeat I
  • BG505 SOSIP.664 trimers Sanders RW, et al. (2013) PLoS Pathog 9(9):el003618; Sanders RW, et al. (2002) J Virol 76(17):8875-8889.
  • SOSIP molecules based on other HIV-I primary strains were attempted over the past decade, the BG505- and KNHI 144-derived SOSIP trimers are the two limited examples of SOSIPs that yield homogeneous trimers suitable for high resolution biophysical and structural analysis.
  • the structural explanation for the difficulty to readily transfer the SOSIP design to other HIV-I strain-derived sequences is not yet fully understood and would be valuable information to broaden the trimer design horizon.
  • tier 2 neutralizing Abs One approach to elicit tier 2 neutralizing Abs has been to immunize the existing well-ordered trimers using prime:boosting in selected animal models. For BG505 and 16055 native-like trimers this approach does elicit tier 2 neutralizing antibodies, but of limited cross-reactive breadth.
  • PLoS Pathog 9:el003618 forming a prefusion native-like conformation (Lyumkis et al., 2013. Cryo-EM structure of a fully glycosylated soluble cleaved HIV-1 envelope trimer. Science 342: 1484-1490; Julien et al, 2013. Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science 342: 1477- 1483; Garces et al., 2015. Affinity Maturation of a Potent Family of HIV Antibodies Is Primarily Focused on Accommodating or Avoiding Glycans. Immunity 43: 1053-1063; Pancera et al., 2014. Structure and immune recognition of trimeric pre-fusion HIV-1 Env.
  • the SOSIP gpl40 trimer is proteolytically cleaved by cellular furins to gpl20 and gp41 subunits and covalently linked by an engineered intra-protomer disulfide bond A501C-T605C (SOS). These trimers also require mutation (I559P) in the gp41 heptad repeat 1 (HR1) to maintain well-ordered oligomers, as well as expression of exogenous ftirin for full conformational integrity (Sanders et al., 2013.
  • a next-generation cleaved, soluble HIV-1 Env trimer BG505 SOSIP.664 gpl40, expresses multiple epitopes for broadly neutralizing but not non- neutralizing antibodies.
  • PLoS Pathog 9:el003618 Both the original SOSIP and NFL designs do not form a high percentage of well-ordered trimers in all Env contexts. In the original NFL design, it is relatively inefficient in generating high yields of trimers derived from clade C strains, such as 16055 (Guenaga et al. 2015. Structure-Guided Redesign Increases the Propensity of HIV Env To Generate Highly Stable Soluble Trimers. J Virol 90:2806-2817).
  • trimer-derived (TD) residues residues from BG505 (called trimer-derived (TD) residues) into 16055 NFLs, substantially improving the propensity to form native-like trimers (Guenaga et al. 2015. Structure-Guided Redesign Increases the Propensity of HIV Env To Generate Highly Stable Soluble Trimers. J Virol 90:2806-2817) and the elicitation of tier 2 clade C neutralizing antibodies (Martinez-Murillo et al, GB. 2017. Particulate Array of Well-Ordered HIV Clade C Env Trimers Elicits Neutralizing Antibodies that Display a Unique V2 Cap Approach. Immunity 46:804-817 e807; Dubrovskaya et al.
  • nanoparticles comprising an isolated polypeptide described herein.
  • a nanoparticle described herein is a ferritin nanoparticle, an encapsulin nanoparticle, a Sulfur Oxygenase Reductase (SOR) nanoparticle, a lumazine synthase nanoparticle or a pyruvate dehydrogenase nanoparticle.
  • a nanoparticle described herein is a ferritin nanoparticle.
  • a nanoparticle described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5.
  • a nanoparticle described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of N332-GT1, N332-GT2, N332-GT3, N332-GT4, orN332-GT5.
  • a nanoparticle described herein comprises N332-GT1, N332- GT2, N332-GT3, N332-GT4, orN332-GT5. In some embodiments, a nanoparticle described herein comprises N332-GT2. In some embodiments, a nanoparticle described herein comprises N332- GT5. In some embodiments, a nanoparticle described herein has at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 13 or 15. In some embodiments, a nanoparticle described herein is a ferritin nanoparticle comprising the amino acid sequence of SEQ ID NO. 13 or 15.
  • virus-like particles comprising an isolated polypeptide described herein.
  • polypeptides e.g., Env trimers, nanoparticles, and other antigens
  • the invention contemplates deletions, additions and substitutions to the sequences shown, so long as the sequences function in accordance with the methods described herein.
  • substitutions are conservative in nature, i.e., those substitutions that take place within a family of amino acids.
  • amino acids are generally divided into four families: (1) acidic— aspartate and glutamate; (2) basic— lysine, arginine, histidine; (3) non-polar— alanine, valine, leucine, isoleucine, praline, phenylalanine, methionine, tryptophan; and (4) uncharged polar— glycine, asparagine, glutamine, cysteine, serine threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine are sometimes classified as aromatic amino acids.
  • germline reverted antibodies can be designed by comparing the heavy and light chains of an isolated bnAb (e.g., BG18) that binds to an epitope of interest to nucleotide sequences and/or the amino acid sequences potentially encoded by germline
  • an isolated bnAb e.g., BG18
  • V gene segments For example, one can obtain a germline reverted antibody starting from the amino acid sequence of an antibody directed to an antigen or epitope of choice and identifying germline encoded antibody segments (i.e., heavy chain V, D, and J gene segments and light chain
  • Germline variable genes are cataloged, for example at VBASE2 (Retter I, Althaus HH, Miinch R, Miiller W: VBASE2, an integrative V gene database. Nucleic Acids Res. 2005 Jan l;33(Database issue):D671-4.). Starting from a known or preselected antibody and moving to such a database can be advantageous as the paired heavy and light chains will be compatible and likely to be well represented in the repertoire of expressed germline genes.
  • Germline antibody genes or inferred germline genes can be identified by analyzing
  • Germline antibody genes or inferred germline genes that bind to an epitope of interest can also be identified by screening libraries of human heavy and light chain genes, for example by phage display. Another method is by immunization and preparation of monoclonal antibodies from a mouse that expresses human antibody genes. Preferably hybridomas are prepared before there is substantial somatic mutation and selection. It can be advantageous to select germline antibody gene segments that are well represented in the repertoire of expressed human antibodies, also taking into account compatibility of various heavy chain - light chain pairings. In certain embodiments, it is advantageous to select V-D, D-J, V-D-J (heavy chain) or V-J (light chain) combinations that are well represented.
  • germline reverted and inferred-germline (iGL) variant heavy and light chain variable domains including heavy and light chain CDR regions of BG 18 that bind to an HIV Env polypeptide disclosed herein (e.g., N332-GT1, N332-GT2, N332-GT3, N332- GT4, or N332-GT5).
  • iGL inferred-germline
  • Germline reverted and inferred-germline antibodies disclosed herein are suitable for the selection of candidate vaccine molecules according to methods disclosed herein.
  • isolated antibodies comprising the VHCDR1, VH CDR2, and VH CDR3 of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1-69, VH5-51, VH3-33, or VH3-23; and the VL CDR1, VL CDR2, and VL CDR3 of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10,
  • an isolated antibody described herein comprises the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3 of BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
  • an isolated antibody described herein comprises the 3 VH CDRs and 3 VL CDRs of
  • the CDR sequences are identified according to Rabat.
  • an isolated antibody described herein comprises the VH of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1-69, VH5-51, VH3-33, or VH3-23; and the VL of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VL3-10, VL3-19, VL3-1, VL3-21, or VL2-8.
  • an isolated antibody described herein comprises the VH and VL of BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
  • an isolated antibody described herein comprises the VH and VL of VH4-59 and BG18 iGL, respectively;
  • an isolated polynucleotide described herein is a DNA.
  • an isolated polynucleotide described herein is an mRNA.
  • an isolated polynucleotide described herein is an mRNA comprising a modified nucleotide.
  • vectors comprising a polynucleotide described herein.
  • a vector described herein can be used for recombinant expression of an isolated polypeptide described herein, a variant HIV Env gpl20 polypeptide described herein, a recombinant HIV Env trimer described herein, a VLP described herein, or a nanoparticle described herein.
  • a vector described herein can be used for administration of an isolated polypeptide described herein, a variant HIV Env gpl20 polypeptide described herein, a recombinant HIV Env trimer described herein, or a nanoparticle described herein to a patient in need thereof.
  • RNA replicons comprising an isolated polynucleotide described herein.
  • an isolated polynucleotide described herein encodes an isolated polypeptide described herein.
  • the isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT1, BG505 MD39 N332-GT2, BG505 MD39 N332-GT3, BG505 MD39 N332-GT4, BG505 MD39 N332-GT5, or BG505 MD39 N332-GT2-KO.
  • an isolated polynucleotide described herein encodes a nanoparticle described herein.
  • a nanoparticle described herein comprises N332-GT1, N332-GT2, N332-GT3, N332-GT4, orN332-GT5.
  • a nanoparticle described herein comprises N332-GT2.
  • a nanoparticle described herein comprises N332-GT5.
  • a nanoparticle described herein is a ferritin nanoparticle comprising the SEQ ID NO. of 13 or 15.
  • an isolated polynucleotide described herein encodes a recombinant HIV Env trimer described herein.
  • the recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT1, BG505 MD39 N332-GT2, BG505 MD39 N332-GT3, BG505 MD39 N332-GT4, BG505 MD39 N332-GT5, or BG505 MD39 N332-GT2-KO.
  • an isolated polynucleotide described herein encodes an antibody described herein. In some embodiments, an isolated polynucleotide described herein encodes BG18.11, BG18.6. In some embodiments, an isolated polynucleotide described herein encodes BG18 iGLO, BG18 iGLl, or BG18 iGL2. In some embodiments, an isolated polynucleotide described herein encodes PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
  • an isolated polynucleotide described herein encodes VH4-595, VH1-69, VH5-51, VH3-33, VH3-23, VL3-10, VL3-19, VL3- 1, VL3-21, or VL2-8.
  • an isolated polynucleotide described herein encodes a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein and comprises an mRNA.
  • the mRNA comprises at least one modified nucleotide.
  • a modified mRNA encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein is for administering to a subject to treat or prevent HIV infection.
  • an "isolated" polynucleotide or nucleic acid molecule is one, which is separated from other nucleic acid molecules, which are present in the natural source (e.g., in a mouse or a human) of the nucleic acid molecule.
  • an "isolated" nucleic acid molecule such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • the language "substantially free” includes preparations of polynucleotide or nucleic acid molecule having less than about 15%, 10%, 5%, 2%, 1%, 0.5%, or 0.1% (in particular less than about 10%) of other material, e.g., cellular material, culture medium, other nucleic acid molecules, chemical precursors and/or other chemicals.
  • a nucleic acid molecule(s) encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein is isolated or purified.
  • a polynucleotide provided herein comprises a nucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein that is optimized, e.g., by codon/RNA optimization, replacement with heterologous signal sequences, and elimination of mRNA instability elements.
  • Methods to generate optimized nucleic acids for recombinant expression by introducing codon changes and/or eliminating inhibitory regions in the mRNA can be carried out by adapting the optimization methods described in, e.g., U.S. Patent Nos. 5,965,726; 6, 174,666; 6,291,664; 6,414,132; and 6,794,498, accordingly.
  • potential splice sites and instability elements within the RNA can be mutated without altering the amino acids encoded by the nucleic acid sequences to increase stability of the RNA for recombinant expression.
  • the alterations utilize the degeneracy of the genetic code, e.g., using an alternative codon for an identical amino acid.
  • an optimized polynucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein can hybridize to an antisense (e.g., complementary) polynucleotide of an unoptimized polynucleotide sequence encoding polypeptide described herein or a fragment thereof (e.g., VL domain or VH domain).
  • an antisense e.g., complementary polynucleotide of an unoptimized polynucleotide sequence encoding polypeptide described herein or a fragment thereof (e.g., VL domain or VH domain).
  • an optimized nucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein or a fragment hybridizes under high stringency conditions to antisense polynucleotide of an unoptimized polynucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein or a fragment thereof.
  • an optimized nucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein hybridizes under high stringency, intermediate or lower stringency hybridization conditions to an antisense polynucleotide of an unoptimized nucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein or a fragment thereof.
  • Information regarding hybridization conditions has been described, see, e.g., U.S. Patent Application Publication No. US 2005/0048549 (e.g., paragraphs 72-73), which is incorporated herein by reference.
  • the polynucleotides can be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art. Nucleotide sequences encoding antibodies described herein, and modified versions of these antibodies can be determined using methods well-known in the art, i.e., nucleotide codons known to encode particular amino acids are assembled in such a way to generate a nucleic acid that encodes the antibody.
  • Such a polynucleotide encoding the antibody can be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier G et ah, (1994), BioTechniques 17: 242-246), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.
  • chemically synthesized oligonucleotides e.g., as described in Kutmeier G et ah, (1994), BioTechniques 17: 242-246
  • polynucleotides that hybridize under high stringency, intermediate or lower stringency hybridization conditions to polynucleotides that encode an antibody described herein.
  • polynucleotides described herein hybridize under high stringency, intermediate or lower stringency hybridization conditions to polynucleotides encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) provided herein.
  • Hybridization conditions have been described in the art and are known to one of skill in the art.
  • hybridization under stringent conditions can involve hybridization to filter-bound DNA in 6x sodium chloride/sodium citrate (SSC) at about 45°C followed by one or more washes in 0.2xSSC/0.1% SDS at about 50-65°C;
  • hybridization under highly stringent conditions can involve hybridization to filter-bound nucleic acid in 6xSSC at about 45°C followed by one or more washes in O. lxSSC/O.2% SDS at about 68°C.
  • Hybridization under other stringent hybridization conditions are known to those of skill in the art and have been described, see, for example, Ausubel FM et ah, eds., (1989) Current Protocols in Molecular Biology, Vol. I, Green Publishing Associates, Inc. and John Wiley & Sons, Inc., New York at pages 6.3.1-6.3.6 and 2.10.3.
  • isolated vectors comprising a polynucleotide described herein.
  • an isolated vector described herein is a viral vector.
  • recombinant viruses comprising a polynucleotide described herein.
  • a recombinant virus described herein is a recombinant adeno-associated virus (AAV).
  • AAV adeno-associated virus
  • host cells comprising a polynucleotide described herein, or a vector described herein.
  • a host cell described herein is selected from the group consisting of E.
  • coli Pseudomonas, Bacillus, Streptomyces, yeast, CHO, YB/20, NSO, PER-C6, HEK-293T, NIH-3T3, HeLa, BHK, Hep G2, SP2/0, Rl . l, B-W, L-M, COS 1, COS 7, BSC1, BSC40, BMT10 cell, plant cell, insect cell, and human cell in tissue culture.
  • kits for producing an isolated polypeptide described herein, a recombinant HIV Env trimer described herein, or a nanoparticle described herein comprising, culturing the host cell described herein so that the polynucleotide is expressed and the isolated polypeptide, recombinant HIV Env trimer, or the nanoparticle is produced.
  • cells e.g., host cells
  • vectors e.g., expression vectors
  • the vectors can be used for recombinant expression of a polypeptide described herein in host cells (e.g., mammalian cells).
  • the vectors can be used for administration of a polypeptide described herein to a patient in need thereof.
  • host cells comprising such vectors for recombinantly expressing polypeptides described herein.
  • methods for producing a polypeptide described herein, comprising expressing such polypeptides in a host cell.
  • an isolated vector comprising a polynucleotide described herein.
  • the vector is a viral vector.
  • a recombinant virus comprising a polynucleotide described herein.
  • the recombinant virus encodes a polypeptide described herein.
  • the recombinant virus encodes a variant HIV Env polypeptide described herein.
  • the recombinant virus is a replication defective virus. Suitable replication defective viral vectors are known to those skilled in the art, for example, as disclosed in U.S. Pat. Nos. 7198784, 9408905, 9862931, 8067156, U.S. Pat. Appl. Pub. Nos.
  • the recombinant virus is a retrovirus or retroviral vector, for example, a lentivirus or lentiviral vector.
  • the recombinant virus is an adenovirus or adenoviral vector, HSV or HSV vector, or influenza virus or viral vector.
  • the recombinant virus is an adeno-associated virus (AAV).
  • the recombinant virus is for administration to a subject to prevent or treat HIV infection.
  • the recombinant virus is an adeno-associated virus (AAV) for administration to a subject to prevent or treat HIV infection.
  • AAV adeno-associated virus
  • Recombinant AAV particles encoding a polypeptide described herein and methods for producing thereof are known to one skilled in the art, for example, as disclosed in US Patent 8,865,881 and US20190031740, each of which is incorporated by reference herein in its entirety for all purposes. See also, Lin and Balazs, Retrovirology 15:66 (2016) and van den Berg et al, Molecular Therapy: Methods & Clinical Development 14: 100-112 (2019), each of which is incorporated by reference herein in its entirety for all purposes.
  • a host cell comprising a polynucleotide described herein, or a vector described herein.
  • the vector encodes a polypeptide described herein.
  • the host cell is selected from the group consisting of E. coli, Pseudomonas, Bacillus, Streptomyces, yeast, CHO, YB/20, NS0, PER-C6, HEK-293T, NIH-3T3, Helga, BHK, Hep G2, SP2/0, Rl . l, B-W, L-M, COS 1, COS 7, BSC1, BSC40, BMT10 cell, plant cell, insect cell, and human cell in tissue culture.
  • the host cell is CHO.
  • a method of producing a polypeptide described herein comprising culturing a host cell described herein so that the polynucleotide is expressed and the polypeptide is produced. In one embodiment, the method further comprises recovering the polypeptide.
  • the isolated polypeptides (e.g., recombinant HIV Env trimer or nanoparticle) described herein can be produced by any suitable method known in the art. Such methods range from direct protein synthetic methods to constructing a DNA sequence encoding isolated polypeptide sequences and expressing those sequences in a suitable transformed host.
  • a DNA sequence is constructed using recombinant technology by isolating or synthesizing a DNA sequence encoding a wild-type protein of interest.
  • the sequence can be mutagenized by site-specific mutagenesis to provide functional analogs thereof. See, e.g. Zoeller et al., Proc. Nat'l. Acad. Sci. USA 81 :5662-5066 (1984) and U.S. Pat. No. 4,588,585.
  • a DNA sequence encoding a polypeptide of interest would be constructed by chemical synthesis using an oligonucleotide synthesizer.
  • Such oligonucleotides can be designed based on the amino acid sequence of the desired polypeptide and selecting those codons that are favored in the host cell in which the recombinant polypeptide of interest will be produced. Standard methods can be applied to synthesize an isolated polynucleotide sequence encoding an isolated polypeptide of interest. For example, a complete amino acid sequence can be used to construct a back-translated gene.
  • a DNA oligomer containing a nucleotide sequence coding for the particular isolated polypeptide can be synthesized. For example, several small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated. The individual oligonucleotides typically contain 5' or 3' overhangs for complementary assembly.
  • the polynucleotide sequences encoding a particular isolated polypeptide of interest will be inserted into an expression vector and operatively linked to an expression control sequence appropriate for expression of the protein in a desired host. Proper assembly can be confirmed by nucleotide sequencing, restriction mapping, and expression of a biologically active polypeptide in a suitable host. As is well-known in the art, in order to obtain high expression levels of a transfected gene in a host, the gene must be operatively linked to transcriptional and translational expression control sequences that are functional in the chosen expression host.
  • recombinant expression vectors are used to amplify and express DNA encoding polypeptides of interest.
  • Recombinant expression vectors are replicable DNA constructs, which have synthetic or cDNA-derived DNA fragments encoding a polypeptide operatively linked to suitable transcriptional or translational regulatory elements derived from mammalian, microbial, viral or insect genes.
  • a transcriptional unit generally comprises an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, transcriptional promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription and translation initiation and termination sequences.
  • Such regulatory elements can include an operator sequence to control transcription.
  • DNA regions are operatively linked when they are functionally related to each other.
  • DNA for a signal peptide secretory leader
  • DNA for a polypeptide is operatively linked to DNA for a polypeptide if it is expressed as a precursor, which participates in the secretion of the polypeptide
  • a promoter is operatively linked to a coding sequence if it controls the transcription of the sequence
  • a ribosome binding site is operatively linked to a coding sequence if it is positioned so as to permit translation.
  • Structural elements intended for use in yeast expression systems include a leader sequence enabling extracellular secretion of translated protein by a host cell.
  • recombinant protein is expressed without a leader or transport sequence, it can include an N- terminal methionine residue. This residue can optionally be subsequently cleaved from the expressed recombinant protein to provide a final product.
  • expression control sequence and expression vector will depend upon the choice of host.
  • a variety of host-expression vector systems can be utilized to express polypeptide molecules described herein (see, e.g., U.S. Patent No. 5,807,715).
  • host-expression systems represent vehicles by which the coding sequences of interest can be produced and subsequently purified, but also represent cells which can, when transformed or transfected with the appropriate nucleotide coding sequences, express a polypeptide molecule described herein in situ. These include but are not limited to microorganisms such as bacteria (e.g., E. coli and B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing polypeptide coding sequences; yeast (e.g., Saccharomyces pichia) transformed with recombinant yeast expression vectors containing polypeptide coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing polypeptide coding sequences; plant cell systems (e.g., green algae such as Chlamydomonas reinhardtii) infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing polypeptide coding sequences; or mammalian cell systems (e.g., COS (e.g., COS1 or COS), CHO, BHK, MDCK, HEK 293, NS
  • cells for expressing polypeptides described herein are CHO cells, for example CHO cells from the CHO GS SystemTM (Lonza).
  • cells for expressing polypeptides described herein are human cells, e.g., human cell lines.
  • a mammalian expression vector is pOptiVECTM or pcDNA3.3.
  • bacterial cells such as E. coli, or eukaryotic cells (e.g., mammalian cells) are used for the expression of a recombinant polypeptide molecule.
  • mammalian cells such as Chinese hamster ovary (CHO) cells in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for polypeptides.
  • the expression of nucleotide sequence encoding polypeptides described herein is regulated by a constitutive promoter, inducible promoter or tissue specific promoter.
  • any vector that allows for the expression of the polypeptides and is safe for use in vivo can be used.
  • the vector is a viral vector.
  • Viral vectors can include poxvirus (vaccinia), including vaccinia Ankara and canarypox; adenoviruses, including adenovirus type 5 (Ad5); rubella; sendai virus; rhabdovirus; alphaviruses; and adeno-associated viruses.
  • the viral vector is an adeno-associated virus.
  • a polynucleotide encoding the polypeptide could be delivered as DNA or RNA to the subject for in vivo expression of the polypeptide.
  • Suitable host cells for expression of a polypeptide of interest such as a polypeptide described herein include prokaryotes, yeast, insect or higher eukaryotic cells under the control of appropriate promoters.
  • Prokaryotes include gram negative or gram-positive organisms, for example E. coli or bacilli.
  • Higher eukaryotic cells include established cell lines of mammalian origin. Cell- free translation systems could also be employed.
  • Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. (Cloning Vectors: A Laboratory Manual, Elsevier, N.Y., 1985), the relevant disclosure of which is hereby incorporated by reference.
  • recombinant protein such as a polypeptide described herein.
  • Expression of recombinant proteins in mammalian cells can be performed because such proteins are generally correctly folded, appropriately modified and completely functional.
  • suitable mammalian host cell lines include but are not limited to CHO, VERO, BHK, Hela, MDCK, HEK 293, NIH 3T3, W138, BT483, Hs578T, HTB2, BT20 and T47D, NS0 (a murine myeloma cell line that does not endogenously produce any immunoglobulin chains), CRL7030, COS (e.g., COS1 or COS), PER.C6, VERO, HsS78Bst, HEK-293T, HepG2, SP210, Rl . l, B-W, L-M, BSC1, BSC40, YB/20, BMT10 and HsS78Bst cells.
  • COS e.g., COS1 or COS
  • PER.C6 VERO
  • HsS78Bst HEK-293T
  • HepG2 SP210
  • Rl . l B-W, L-M
  • Mammalian expression vectors can comprise non-transcribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5' or 3' flanking non-transcribed sequences, and 5' or 3' non-translated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
  • Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Bio/Technology 6:47 (1988).
  • the proteins produced by a transformed host can be purified according to any suitable method.
  • Such standard methods include chromatography (e.g., ion exchange, affinity and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification.
  • Affinity tags such as hexahistidine, maltose binding domain, influenza HA peptide sequence and glutathione-S-transferase can be attached to the protein to allow easy purification by passage over an appropriate affinity column.
  • Isolated proteins can also be physically characterized using such techniques as proteolysis, nuclear magnetic resonance and x-ray crystallography.
  • supernatants from systems, which secrete recombinant protein, e.g., a polypeptide, into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. Following the concentration step, the concentrate can be applied to a suitable purification matrix.
  • a suitable purification matrix for example, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups.
  • the matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification.
  • a cation exchange step can be employed.
  • Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups.
  • one or more reversed-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, e.g. , silica gel having pendant methyl or other aliphatic groups, can be employed to further an agent.
  • RP-HPLC reversed-phase high performance liquid chromatography
  • Recombinant protein produced in bacterial culture can be isolated, for example, by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange or size exclusion chromatography steps. High performance liquid chromatography (HPLC) can be employed for final purification steps.
  • Microbial cells employed in expression of a recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
  • a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein is isolated or purified.
  • an isolated polypeptide is one that is substantially free of other polypeptides.
  • a preparation of a polypeptide described herein is substantially free of cellular material and/or chemical precursors.
  • substantially free of cellular material includes preparations of a polypeptide in which the polypeptide is separated from cellular components of the cells from which it is isolated or recombinantly produced.
  • a polypeptide that is substantially free of cellular material includes preparations having less than about 30%, 20%, 10%, 5%, 2%, 1%, 0.5%, or 0.1% (by dry weight) of heterologous protein (also referred to herein as a "contaminating protein") and/or variants of a polypeptide, for example, different post-translational modified forms of a polypeptide.
  • heterologous protein also referred to herein as a "contaminating protein”
  • variants of a polypeptide for example, different post-translational modified forms of a polypeptide.
  • the polypeptide e.g., recombinant HIV Env trimer or nanoparticle described herein
  • culture medium represents less than about 20%, 10%, 2%, 1%, 0.5%, or 0.1% of the volume of the protein preparation.
  • polypeptide e.g., recombinant HIV Env trimer or nanoparticle described herein
  • polypeptides described herein are isolated or purified.
  • compositions comprising polypeptides described herein (e.g., recombinant HIV Env trimer or nanoparticle) are also provided. Further provided herein are compositions comprising a polynucleotide encoding a polypeptide described herein. In some embodiments, the polynucleotide comprises mRNA. In some embodiments, the composition is a pharmaceutical composition. In some embodiments, the composition is an immunogenic composition.
  • compositions comprising an isolated polypeptide described herein, recombinant HIV Env trimer described herein, nanoparticle described herein, polynucleotide described herein, recombinant virus described herein, VLP described herein, or liposome described herein and a pharmaceutically acceptable excipient.
  • the composition is an immunogenic composition.
  • the composition is a vaccine.
  • an immunogenic composition or vaccine further comprises an adjuvant.
  • the composition e.g., immunogenic composition
  • the composition is formulated for subcutaneous administration.
  • the composition e.g., immunogenic composition
  • the composition is formulated for intramuscular administration.
  • the composition e.g., immunogenic composition
  • the composition is formulated for topical administration, and in certain embodiments the composition is formulated for vaginal or rectal administration.
  • immunogenic compositions comprising an isolated polypeptide described herein, recombinant HIV Env trimer described herein, nanoparticle described herein, polynucleotide described herein, recombinant virus described herein, VLP described herein, or liposome described herein and a pharmaceutically acceptable excipient.
  • an immunogenic composition described herein further comprises an adjuvant.
  • the adjuvant comprises lecithin.
  • the adjuvant comprises alum.
  • the adjuvant comprises saponin, cholesterol and phospholipid.
  • the adjuvant comprises ISCOMATRIXTM.
  • the adjuvant comprises carbomer homopolymer and lecithin.
  • the adjuvant comprises AdjuplexTM.
  • an immunogenic composition described herein is capable of eliciting a BG18 like response. In some embodiments, an immunogenic composition described herein is capable of eliciting a BG18 like response in a human subject. In some embodiments, an immunogenic composition described herein is capable of eliciting a BG18 like response in BG18 gH B cell adoptive transfer recipient mice. In some embodiments, an immunogenic composition described herein is capable of eliciting the production of an antibody that binds to N332-GT2. In some embodiments, an immunogenic composition described herein is capable of eliciting the production of an antibody that binds to N332-GT2 with a higher affinity than to N332-GT2-KO. In some embodiments, an immunogenic composition described herein is capable of eliciting the production of a broadly neutralizing antibody in a subject.
  • liposomes comprising an isolated polypeptide described herein.
  • compositions described herein comprising a polypeptide described herein are intended for prevention and treatment of HIV infection.
  • compositions described herein e.g., immunogenic composition
  • compositions described herein are for eliciting an immune reaction in a subject against HIV.
  • compositions described herein are for eliciting a protective immune reaction in a subject against HIV.
  • a composition comprising a polypeptide described herein can additionally be combined with other compositions for the treatment of HIV infection or the prevention of HIV transmission.
  • a polypeptide described herein can be administered within a pharmaceutically acceptable diluent, carrier, or excipient, in unit dose form.
  • Conventional pharmaceutical practice can be employed to provide suitable formulations or compositions for administration to individuals.
  • the administration is prophylactic. Any appropriate route of administration can be employed, for example, administration can be parenteral, intravenous, intra-arterial, subcutaneous, intramuscular, intraperitoneal, intranasal, aerosol, suppository, oral administration, vaginal, or anal.
  • the polypeptides and polynucleotides described herein are administered as a component of an immunogenic composition comprising the polypeptides and/or polynucleotides described herein in admixture with a pharmaceutically acceptable carrier.
  • the immunogenic compositions described herein are useful to stimulate an immune response against HIV- 1 and can be used as one or more components of a prophylactic or therapeutic vaccine against HIV-1 for the prevention, amelioration or treatment of AIDS.
  • the polynucleotides and vectors described herein are particularly useful for providing genetic vaccines, i.e. vaccines for delivering the polynucleotides encoding the antigenic polypeptides described herein to a subject, such as a human, such that the antigenic polypeptides are then expressed in the subject to elicit an immune response.
  • immunogenic compositions described herein comprise injectable suspensions, solutions, sprays, lyophilized powders, syrups, elixirs and the like. Any suitable form of composition can be used.
  • a polypeptide and/or polynucleotide described herein, having the desired degree of purity is mixed with one or more pharmaceutically acceptable carriers and/or excipients.
  • the carriers and excipients must be "acceptable" in the sense of being compatible with the other ingredients of the composition.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, or combinations thereof, buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobul
  • immunogenic compositions described herein are formulated in the form of an oil-in-water emulsion.
  • the oil-in-water emulsion is based, for example, on light liquid paraffin oil (European Pharmacopeatype); isoprenoid oil such as squalane, squalene, EICOSANETM or tetratetracontane; oil resulting from the oligomerization of alkene(s), e.g., isobutene or decene; esters of acids or of alcohols containing a linear alkyl group, such as plant oils, ethyl oleate, propylene glycol di(caprylate/caprate), glyceryl tri(caprylate/caprate) or propylene glycol dioleate; esters of branched fatty acids or alcohols, e.g., isostearic acid esters.
  • light liquid paraffin oil European Pharmacopeatype
  • isoprenoid oil such as squa
  • the oil is used in combination with an emulsifier to form the emulsion.
  • the emulsifier comprises a nonionic surfactant, such as esters of sorbitan, mannide (e.g., anhydromannitol oleate), glycerol, polyglycerol, propylene glycol, and oleic, isostearic, ricinoleic, or hydroxy stearic acid, which are optionally ethoxylated, and polyoxypropylene- polyoxyethylene copolymer blocks, such as the Pluronic® products, e.g., L121.
  • the adjuvant can be a mixture of emulsifier(s), micelle-forming agent, and oil such as that which is commercially available under the name Provax® (IDEC Pharmaceuticals, San Diego, CA). (PEG).
  • an immunogenic composition described herein comprises additional substances, such as wetting or emulsifying agents, buffering agents, or adjuvants to enhance the effectiveness of the vaccines (Remington's Pharmaceutical Sciences, 18th edition, Mack Publishing Company, (ed.) 1980).
  • an immunogenic composition described herein comprises an adjuvant.
  • Suitable adjuvants include, but are not limited to, mineral salts (e.g., AIK ⁇ SCEK AlNa(S04)2, A1NH(S04)2, silica, alum, Al(OH)3, Ca3(PC>4)2, kaolin, or carbon), polynucleotides with or without immune stimulating complexes (ISCOMs) (e.g., CpG oligonucleotides, such as those described in Chuang, T.H. et al, (2002) J. Leuk. Biol. 71(3): 538- 44; Ahmad-Nejad, P. et al (2002) Eur. J. Immunol.
  • mineral salts e.g., AIK ⁇ SCEK AlNa(S04)2, A1NH(S04)2, silica, alum, Al(OH)3, Ca3(PC>4)2, kaolin, or carbon
  • ISCOMs immune stimulating complexes
  • monophosphoryl lipid A in particular, 3-de-0- acylated monophosphoryl lipid A (3D-MPL), imiquimod (also known in the art as IQM and commercially available as Aldara®; U.S. Patent Nos. 4,689,338; 5,238,944; Zuber, AK. et al (2004) 22(13-14): 1791-8), and the CCR5 inhibitor CMPD 167 (see Veazey, RS. et al (2003) J. Exp. Med. 198: 1551-1562).
  • 3D-MPL 3-de-0- acylated monophosphoryl lipid A
  • imiquimod also known in the art as IQM and commercially available as Aldara®
  • U.S. Patent Nos. 4,689,338; 5,238,944 Zuber, AK. et al (2004) 22(13-14): 1791-8
  • CMPD 167 see Veazey, RS. et al (2003) J. Exp. Med. 198:
  • Aluminum hydroxide or phosphate (alum) are commonly used at 0.05 to 0.1% solution in phosphate buffered saline.
  • Other adjuvants that can be used, especially with DNA vaccines, are cholera toxin, especially CTAl-DD/ISCOMs (see Mowat, AM. et al (2001) J. Immunol. 167(6): 3398-405), polyphosphazenes (Allcock, H.R. (1998) App. Organometallic Chem. 12(10-11): 659- 666; Payne, L.G. et al (1995) Pharm. Biotechnol.
  • cytokines such as, but not limited to, IL-2, IL-4, GM-CSF, IL-12, IL-15 IGF-1, IFN-a, IFN- , and IFN-y
  • immunoregulatory proteins such as CD40F (ADX40; see, for example, W003/063899)
  • CDla ligand of natural killer cells also known as CRONY or a- galactosyl ceramide; see Green, T.D. et al, (2003) J. Virol.
  • immunostimulatory fusion proteins such as IF-2 fused to the Fe fragment of immunoglobulins (Barouch et al., Science 290:486-492, 2000) and co-stimulatory molecules B7. 1 and B7.2 (Boyer), all of which can be administered either as proteins or in the form of DNA, on the same expression vectors as those encoding the antigens described herein or on separate expression vectors.
  • the adjuvant comprises lecithin combined with an acrylic polymer (Adjuplex-FAP), lecithin coated oil droplets in an oil-in-water emulsion (Adjuplex-FE) or lecithin and acrylic polymer in an oil-in water emulsion (Adjuplex-FAO) (Advanced BioAdjuvants (ABA)).
  • Adjuplex-FAP acrylic polymer
  • Adjuplex-FE lecithin coated oil droplets in an oil-in-water emulsion
  • Adjuplex-FAO Advanced BioAdjuvants
  • an immunogenic composition described herein is formulated to introduce the polypeptides and polynucleotides disclosed herein (collectively, the immunogens) to a desired site of action and release it at an appropriate and controllable rate.
  • Methods of preparing controlled-release formulations are known in the art.
  • controlled release preparations can be produced by the use of polymers to complex or absorb the immunogen and/or immunogenic composition.
  • a controlled-release formulation can be prepared using appropriate macromolecules (for example, polyesters, polyamino acids, polyvinyl, pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate) known to provide the desired controlled release characteristics or release profile.
  • Another possible method to control the duration of action by a controlled-release preparation is to incorporate the active ingredients into particles of a polymeric material such as, for example, polyesters, polyamino acids, hydrogels, polylactic acid, polygly colic acid, copolymers of these acids, or ethylene vinylacetate copolymers.
  • a polymeric material such as, for example, polyesters, polyamino acids, hydrogels, polylactic acid, polygly colic acid, copolymers of these acids, or ethylene vinylacetate copolymers.
  • microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin- microcapsule and poly-(methylmethacrylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • compositions described herein can be, for example, in unit dose form, such as in the form of ampoules, vials, suppositories, tablets, pills, or capsules.
  • the formulations can be administered to human individuals in therapeutically or prophylactic effective amounts (e.g., amounts which prevent, eliminate, or reduce a pathological condition) to provide therapy or prophylaxis for a disease or condition (e.g., HIV infection).
  • a disease or condition e.g., HIV infection
  • the preferred dosage of therapeutic or prophylactic agent to be administered is likely to depend on such variables as the type and extent of the disorder, the overall health status of the particular patient, the formulation of the compound excipients, and its route of administration.
  • Suitable dosages of the polypeptides and polynucleotides disclosed herein (collectively, the immunogens) in the immunogenic composition disclosed herein can be readily determined by those of skill in the art.
  • the dosage of the immunogens can vary depending on the route of administration and the size of the subject.
  • Suitable doses can be determined by those of skill in the art, for example by measuring the immune response of a subject, such as a laboratory animal, using conventional immunological techniques, and adjusting the dosages as appropriate.
  • Such techniques for measuring the immune response of the subject include but are not limited to, chromium release assays, tetramer binding assays, IFN-y ELISPOT assays, IL-2 ELISPOT assays, intracellular cytokine assays, and other immunological detection assays, e.g., as detailed in the text "Antibodies: A Laboratory Manual” by Ed Harlow and David Lane.
  • HIV Env gpl20 in a subject comprising administering to the subject an effective amount of an immunogenic composition described herein, thereby generating the immune response.
  • the subject is a human.
  • the subject has been exposed to HIV.
  • the subject is at risk of being exposed to HIV.
  • the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker.
  • the subject is a non-human primate.
  • the subject is a BG18 gH B cell adoptive transfer recipient mouse.
  • the subject is a mouse.
  • the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
  • kits for reducing the likelihood of HIV infection in a subject exposed to HIV comprising administering to the subject an effective amount of an immunogenic composition described herein, or the pharmaceutical composition described herein.
  • the subject is a human.
  • the subject has been exposed to HIV.
  • the subject is at risk of being exposed to HIV.
  • the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker.
  • the subject is a non-human primate.
  • the immunogenic composition comprises an isolated polypeptide described herein.
  • the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
  • kits for reducing the risk of a subject becoming infected with HIV comprising administering to the subject in need thereof an effective amount of an immunogenic composition described herein, or the pharmaceutical composition described herein.
  • the subject is a human.
  • the subject has been exposed to HIV.
  • the subject is at risk of being exposed to HIV.
  • the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker.
  • the subject is a non-human primate.
  • the immunogenic composition comprises an isolated polypeptide described herein.
  • the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
  • kits for preventing HIV infection comprising administering to a subject in need thereof an effective amount of the immunogenic composition described herein, or the pharmaceutical composition described herein.
  • the subject is a human.
  • the subject has been exposed to HIV.
  • the subject is at risk of being exposed to HIV.
  • the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker.
  • the subject is a non-human primate.
  • the immunogenic composition comprises an isolated polypeptide described herein.
  • the immunogenic composition comprises a recombinant HIV Env trimer described herein.
  • the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
  • kits for treating HIV/AIDS comprising administering to a subject in need thereof an effective amount of the immunogenic composition described herein, or the pharmaceutical composition described herein.
  • the subject is a human.
  • the subject has been exposed to HIV.
  • the subject is at risk of being exposed to HIV.
  • the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker.
  • the subject is a non-human primate.
  • the immunogenic composition comprises an isolated polypeptide described herein.
  • the immunogenic composition comprises a recombinant HIV Env trimer described herein.
  • the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
  • the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein.
  • the immunogenic composition comprises a nucleic acid described herein.
  • the pharmaceutical composition comprises a recombinant virus described herein.
  • an immunogenic composition further comprises an adjuvant.
  • the immunogenic composition is formulated for subcutaneous administration.
  • the immunogenic composition is formulated for intramuscular administration.
  • a method described herein further comprises administering at least one additional therapeutic agent.
  • the additional therapeutic agent is an antiretroviral agent.
  • the additional therapeutic agent comprises an antiretroviral therapy (ART) agent, a reservoir activator, an immunomodulator, a second antibody, or a broadly neutralizing anti-BIV antibody.
  • ART antiretroviral therapy
  • the administering to the subject is by at least one mode selected from oral, parenteral, subcutaneous, intramuscular, intravenous, vaginal, rectal, buccal, sublingual, and transdermal.
  • the subject is at risk for exposure to HIV.
  • the subject is infected with HIV.
  • the subject is diagnosed with AIDS.
  • the subject at risk for exposure to HIV is a health care worker.
  • the subject at risk for exposure to HIV is a sex worker.
  • the subject at risk for exposure to HIV is a sexual partner of an HIV infected individual.
  • the subject at risk for exposure to HIV is a newborn.
  • the polypeptides and/or polynucleotides described herein are administered in vivo, for example where the aim is to produce an immunogenic response in a subject.
  • the "subject" may be any animal.
  • the subject is a human, for example a human that is infected with, or is at risk of infection with, HIV-1.
  • the polypeptides and/or polynucleotides described herein can be administered as a component of an immunogenic composition comprising the polypeptides and/or polynucleotides described herein in admixture with a pharmaceutically acceptable carrier.
  • the immunogenic compositions described herein are useful to stimulate an immune response against HIV- 1 and can be used as one or more components of a prophylactic or therapeutic vaccine against HIV-1 for the prevention, amelioration or treatment of AIDS.
  • the polynucleotides described herein are particularly useful for providing genetic vaccines, i.e. vaccines for delivering the polynucleotides encoding the polypeptides described herein to a subject, such as a human, such that the polypeptides are then expressed in the subject to elicit an immune response.
  • the immunogenic compositions described herein can be administered to a subject in advance of HIV infection, or evidence of HIV infection, or in advance of any symptom due to AIDS, especially in high-risk subjects.
  • the prophylactic administration of the immunogenic compositions described herein can serve to provide protective immunity of a subject against HIV-1 infection or to prevent or attenuate the progression of AIDS in a subject already infected with HIV-1.
  • the immunogenic compositions described herein can serve to ameliorate and treat AIDS symptoms and can be used as soon after infection as possible, for example, before appearance of any symptoms of AIDS, but can also be used at (or after) the onset of the disease symptoms.
  • Immunization schedules are well known for animals (including humans) and can be readily determined for the particular subject and immunogenic composition. Hence, the immunogens can be administered one or more times to the subject. In some embodiments, there is a set time interval between separate administrations of the immunogenic composition. While this interval varies for every subject, typically it ranges from 10 days to several weeks, and is often 2, 4, 6 or 8 weeks. For humans, the interval is typically from 2 to 6 weeks.
  • the immunization regimes typically have from 1 to 6 administrations of the immunogenic composition, but can have as few as one or two or four.
  • the methods of inducing an immune response described herein can also include administration of an adjuvant with the immunogens. In some instances, annual, biannual or other long interval (5-10 years) booster immunization can supplement the initial immunization protocol.
  • the methods described herein also include a variety of prime-boost regimens.
  • one or more priming immunizations are followed by one or more boosting immunizations.
  • the actual immunogenic composition can be the same or different for each immunization and the type of immunogenic composition (e.g., containing protein or expression vector), the route, and formulation of the immunogens can also be varied.
  • an expression vector e.g., DNA or bacterial or viral expression vector.
  • the prime-boost regimen provides for two priming immunizations, four weeks apart, followed by two boosting immunizations at 4 and 8 weeks after the last priming immunization. It should also be readily apparent to one of skill in the art that there are several permutations and combinations that are encompassed using a polypeptide (e.g., Env trimer or nanoparticle) described herein to provide priming and boosting regimens.
  • a polypeptide e.g., Env trimer or nanoparticle
  • a method of inducing an immune response against HIV in a subject described herein comprises administering an immunogenic composition of the invention comprising an adenovirus vector containing DNA encoding one or more of the polypeptides described herein, one or more times to a subject wherein the polypeptides are expressed at a level sufficient to induce a specific immune response in the subject.
  • Such immunizations can be repeated multiple times at time intervals of at least 2, 4 or 6 weeks (or more) in accordance with a desired immunization regime.
  • the immunogenic compositions of the invention can be administered alone, or can be co-administered, or sequentially administered, with other HIV immunogens and/or HIV immunogenic compositions, e.g., with "other" immunological, antigenic or vaccine or therapeutic compositions thereby providing multivalent or "cocktail” or combination compositions.
  • the ingredients and manner (sequential or co-administration) of administration, as well as dosages can be determined taking into consideration such factors as the age, sex, weight, species and condition of the particular subject, and the route of administration.
  • the other HIV immunogens can be administered at the same time or at different times as part of an overall immunization regime, e.g., as part of a prime- boost regimen or other immunization protocol.
  • the other HIV immunogen is Env, for example, an HIV Env trimer.
  • bnAb broadly neutralizing antibody
  • a vaccine candidate variant HIV Env polypeptide e.g., gpl20 polypeptide or HIV Env trimer
  • the method comprising providing a library comprising a plurality of variant HIV Env polypeptides; contacting the library with an antibody described herein; and identifying a variant HIV Env polypeptide that specifically binds to the antibody.
  • the plurality of variant HIV Env polypeptides comprise variants of a parental HIV Env polypeptide comprising one or more amino acid substitutions at a region selected from the VI loop comprising positions 131-154, V3 loop comprising positions 297-334, and b19 sheet comprising positions 413-419, wherein HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
  • the parental HIV Env polypeptide comprises BG505, BG505 N332, BG505 MD39, BG505 MD39 N332, BG505 MD39 l lmutB, or BG505 MD39 17mutE Env polypeptide.
  • the Env polypeptide is a gpl60 polypeptide, a gpl20 polypeptide, or an Env trimer polypeptide.
  • NGS Next generation sequencing
  • bioinformatics was used to identify HCDR3s sharing key properties with the HCDR3 -dependent bnAb BG18 in 14 of 14 HIV-uninfected donors.
  • BG18 inferred-germline (iGL) variants with NGS-derived HCDR3 loops were used as targets for structural-model-guided, multitarget mammalian-display selection of a series of Env trimers (N332-GT trimers) that bind to diverse BG18 potential precursors.
  • Sorting of human B cells from HIV-uninfected donors using N332-GT trimer probes revealed BG18-like naive B cells with long HCDR3 loops and light chains similar to those in BG18.
  • N332-GT trimer-based nanoparticles were constructed. It was found that nanoparticle immunization initiated responses from rare BG18-like B cells in mice hosting transferred BG18 iGL heavy chain B cells. This study demonstrated the potential for vaccine targeting of rare HCDR3 -dependent bnAb precursors and suggested that N332-GT-trimer-based nanoparticles have promise for priming BG18-like bnAb responses in humans.
  • Germline -targeting (GT) HIV vaccine design is a strategy that aims to induce bnAbs by first priming the precursor B cells of known bnAbs and then shepherding B cell affinity maturation with a series of rationally designed boosting immunogens.
  • GT germline -targeting
  • VRC01 -class bnAbs represent a specialized case in which non-HCDR3 features dominate the binding modality.
  • GT Env trimers with affinity for iGL precursors of the HCDR3 -dependent bnAb PGT121 11 that binds to the N332 glycan supersite, also known as the high mannose patch or V3- glycan epitope, on the trimer 12 13 were previously engineered. These GT trimers (lOmut, 1 limits) were able to prime B cells in a PGT121 iGL homozygous B cell receptor (BCR) knock-in mouse.
  • BCR B cell receptor
  • BG18 a more recently described N332 glycan-dependent bnAb 15
  • PGT21 activity requires indels.
  • BG18 iGL does not bind to WT BG505 Env (T332N), indicating that protein design would be required to develop an effective priming immunogen.
  • T332N WT BG505 Env
  • GT immunogen design was initiated using our directed evolution methodology for engineering trimers on the surface of mammalian cells that previously proved capable of producing stabilized native-like trimers with affinity for PGT121 iGL variants 11 19 .
  • this case was an attempt to overcome the limitations of using only iGL variants to represent the pool of potential bnAb precursors in the human repertoire.
  • Inferred-germline Abs contain bnAb HCDR3 junctions that have been selected and possibly somatically mutated for high affinity Env binding during bnAb development, hence such iGL Abs under-represent both the diversity of potential precursors and the difficulty of engineering high affinity antigens to target those precursors.
  • BG18 iGL three iGL variants (BG18 1GL0-2), that could be used as selection reagents for multitarget optimization 4 .
  • Structure-guided libraries were designed to screen by mammalian cell surface display using 1 Imut B as the starting construct. Five libraries were screened iteratively with selection against the least challenging antibody targets first (i.e. BG18 iGL) followed by selection against more difficult targets (i.e. NGS-derived and alternate VH/VL precursors) in the last three libraries ( Figure Id).
  • N332-GT1 and -GT2 bound with a KD of 213 pM to BG18 iGLi, which represented a -130, 000-fold improvement over 1 Imut B .
  • N332-GT1 and N332-GT2 were used as sorting reagents and either BG505- MD39 (containing a native N332 epitope) or N332-GT2-KO (an epitope knockout) as negative sorting probes (Figure 2a).
  • BG505- MD39 containing a native N332 epitope
  • N332-GT2-KO an epitope knockout
  • Epitope-specific naive B cells isolated at a frequency of 0.001% (Figure 10), were enriched for long HCDR3s (Figure 2b) and highly enriched for VL3- 25 and VL3-1 LCS (Figure 2c) corresponding to the BG18 LC and a LC with similar binding properties, respectively ( Figure le and Figure 5).
  • Figure 10 epitope-specific naive B cells, isolated at a frequency of 0.001%
  • ferritin nanoparticles (NPs) 22,23 were constructed that displayed up to 8 copies of MD39 native-like trimers, and it was found that in mouse immunization studies that such NPs were superior to MD39 trimers in trafficking to follicular dendritic cell networks, concentrating in germinal centers, and eliciting IgG responses.
  • similar ferritin NPs displaying N332-GT2 trimers were engineered (Figure 11).
  • Example 6 BG18 gH knock-in mouse
  • BG18 1GL2 had detectable affinity to the l lmut B (PGT121 germline targeting) trimer but no detectable affinity to BG505 MD39 containing a native N332 epitope, therefore 1 Imut B was used as a base construct to begin the BG18 iGL GT design process.
  • the following libraries were screened using a previously described mammalian cell surface display method 11 .
  • Library 1 was an NNK codon scan of positions 294, 297, 298, 299, 300, 302, 304, 305, 326, 329, 330, 333, 386, 413, 414, 415, 416, 417, 419, 420.
  • NNK codons were introduced into BG505-1 lmut B -gpl20 using the QuikChange Site-Directed Mutagensis Kit (Agilent).
  • Library 1 was screened for binding to BG18 1GL2 and PGT121-GLc DR3rev4 -
  • Library 2 was a combinatorial library with amino acids F/I/L/V introduced at positions 154, 322, 323, 326, 333, 414, 415, and 416.
  • the library insert was assembled with overlapping ultramers (IDT DNA) followed by Gibson cloning (NEB) into BG505-1 lmut B - gpl20.
  • Library 2 was screened for binding to BG18 1GL2 and PGT121-GLCDR 3IEV4 .
  • Library 3 was a combinatorial library with NNK codons introduced at positions 137, 325, and (F/I/L/V) at position 326.
  • the library insert was assembled with overlapping ultramers (IDT DNA) followed by Gibson cloning (NEB) into BG505-MD39-17mut E .
  • Library 3 was screened for binding to the following 24 Abs: BG18 iGL 0 , BG18 iGLi, prel - pre6, pre8, prelO - pre l5, V L 2-8, V L 2-14, V L 3-21, V H l-69, VH3-33, VH4-59, VH5-5 1, PGT121-GLcDR3m at , PGT121-GLcDR3 revi .
  • Library 4 was a combinatorial library with NNK codons introduced at positions 138 and 141 and (P/H/A/D) at position 325.
  • the library insert was assembled with overlapping ultramers (IDT DNA) followed by Gibson cloning (NEB) into BG505-MD39-17mut E -N137K.
  • Library 4 was screened for binding to BG18 iGLo, pre3, pre l4, VL3-21, VL2-8, VL2-14, VH3-33.
  • Library 5 was a combinatorial library with NNK codons introduced at positions 138 and 139 and (P/H/A/D) at position 325.
  • the library insert was assembled with overlapping ultramers (IDT DNA) followed by Gibson cloning (NEB) into BG505- MD39-17mut E -N137K.
  • Library 5 was screened for binding to BG18 iGLo, VL3-21, VL2-8, VL2-14, VH1-69, VH5-5 1.
  • Library 6 was an NNK scan of positions 167 to 308.
  • the insert was synthesized at SGI-DNA and Gibson cloned (NEB) into BG505-MD39-N332-GT3.
  • Library 6 was screened for binding to BG18 iGLo, VL2-8.
  • Library 7 was an NNK scan of positions 309 to 443.
  • the insert was synthesized at SGI-DNA and Gibson cloned (NEB) into BG505-MD39-N332-GT3.
  • Library 7 was screened for binding to BG18 iGLo, pre l, pre2, pre4, prelO, prel5, VL2-8. All constructs contained a C-terminal myc tag and were anchored to the cell membrane via a C-terminal PDGLR transmembrane domain. Staining of the cell populations was typically done with IgG until saturated binding was obtained at low nanomolar IgG concentrations, and then Labs were used for staining to maintain selection pressure. Typically, libraries were sorted 3 to 5 times, and the enriched cell populations were frozen until sequencing could be carried out as described previously 11 . The most enriched clones were synthesized at Genscript either as C-terminal His tagged gpl20s or MD39/MD64 trimers in the pHLsec vector and expressed and purified as described previously 11 . Nanoparticle design and purification
  • trimers were genetically fused to ferritin from Helicobacter pylori using a short flexible linker. Genes were codon optimized for HEK293 cells and cloned into the pHLsec plasmid (GenScript). MD39-NP DNA was co-transfected with a plasmid encoding human Turin protease into LreeStyle 293L cells (Invitrogen, Cat no. R79007) using 293Lectin (ThermoPisher) and proteins were expressed at 37 ° C for four days.
  • N332-GT2 NP has the Furin cleavage site removed and was not co-transfected with Furin.
  • NP -assembly was assessed by negative-stain EM and SEC + multi-angle light scattering (SEC-MALS) using a Superose 6 10/300 column (GE Healthcare) at a flow rate of 0.5 mL/min followed by DAWN HELEOS II and Optilab T-rEX detectors (Wyatt Technology), correcting for the glycan molecular mass by applying the built-in protein-conjugate analysis (ASTRA).
  • Atomic models were built and refined into the high-resolution reconstructions by creating homology models based off deposited coordinates of BG505 SOSIP.664 (PDB 5cez) and 354BG18 fab (PDB 5ud9), followed by an iterative cycle of manual building in COOT 31 and real space refinement in Phenix 1.13 32 .
  • Glycans were validated by CARP 33 and Privateer 34 , and overall structures were evaluated using EMRinger 35 and Molprobity 36 .
  • N332-GT2-specific antibody titers were detected by ELISA, using anti-His Ab (2 pg/ml) to capture N332-GT2 or N332-GT2-KO antigen (2 pg/ml) on the plate.
  • Mouse sera were incubated for two hours and alkaline phosphatase conjugated anti-mouse IgG (Jackson ImmunoResearch, #115-055-071) was incubate another hour. Titers were determined from the dilution curve in the linear range of absorbance. All non-commercial ELISA plates were developed with p-Nitrophenyl Phosphate (Sigma, # N2770). Absorbance at 405 nm was determined with a plate reader (BioTek).
  • PGT128 IgG was captured at 1300 RU level in all channels including reference followed by NP (as ligand) capture at 1600 RU. All other steps were the same as in Human Antibody Capture protocol. Analyte concentrations were measured on a NanoDrop 2000c Spectrophotometer using Absorption signal at 280 nm 3 .
  • This work utilized a large NGS dataset of 1.1 c 10 9 unique amino acid sequences of BCR HCs from 14 healthy, HIV-uninfected donors.
  • 255 million sequences from 10 donors were obtained from ref. 18 which used the HiSeq sequencing platform and an amplification strategy including unique identifiers (UIDs) to enable discrimination of unique mRNA transcripts from PCR artifacts.
  • UIDs unique identifiers
  • These sequences were collapsed by UID, assigned to VDJ gene segments with Abstar 18 , and then rendered unique by clustering at the 99% amino acid identity level within each of six biological replicates per donor.
  • JSON output files from Abstar were converted to parquet format and uploaded to the Amazon S3 storage cloud.
  • PBMCs peripheral blood mononuclear cells
  • junctional regions of antibody heavy chain libraries were amplified as in Willis et ah, 2015 37 .
  • SPRI-purified sequencing libraries were initially quantified using fluorometry (Qubit, Thermo Fisher Scientific) before size determination using a bioanalyzer (Agilent 2100). Libraries were re-quantified using qPCR (KAPA Biosystems) before sequencing on either an Illumina HiSeq (2xl50bp chemistry) or NextSeq (2xl50bp chemistry). Sequences were merged with PANDAseq using the default (symple_bayesian) merging algorithm before annotation with Abstar 18 .
  • BG18-like feature set ii (HCDR3 junction features) in Figure 6a characterized amino acids present in the non-templated junction regions of BG18 and its somatic variants 15 and in the 11 precursors that bound to N332-GT2.
  • the frequency of HCDR3s meeting the definitions for both feature sets i and ii was found to be lower than those within set i by a factor of 104.
  • V L gene usage was incorporated into the frequency estimate, as feature set iii (“V L gene family”).
  • the conservative assumption was that only V L 3 LCS can support the BG18-class binding mode, as all V L 3 LCS tested bound with high affinity to N332-GT2.
  • the frequency of all V L 3-derived Abs in the HC-LC paired sequences in DeKosky et al 38 was 1 in 9 (13845 V L 3S in 127701 sequences).
  • LRS (leukoreduction) tubes were obtained from the San Diego Blood Bank from healthy, HIV-seronegative human donors. These studies do not constitute human subjects research, as determined by the Institutional Review Boards of both La Jolla Institute and The Scripps Research Institute. More than 1 billion peripheral blood mononuclear cells were regularly recovered. CD19 + B cells were isolated using a positive selection magnetic bead separation kit (Miltenyi Biotec) and resuspended in complete RPMI media with 10% FBS.
  • Avi-tagged protein immunogens were biotinylated using the Bulk BirA kit (Avidity, LLC).
  • N332-GT2 and N332-GT2KO probes were used in N332-GT2 sorting experiments.
  • N332- GT1 and MD39 probes were used in N332-GT1 sorting experiments.
  • l lmut B and MD39 probes were used in l lmut B sorting experiments.
  • Biotinylated protein immunogens were individually premixed with fluorescently labeled streptavidin to form tetramer probes.
  • N332-GT2 sorting experiments used the follow probes: N332-GT2-Histag-biotin + streptavidin Alexa Fluor 647, N332-GT2- streptag-biotin + streptavidin Brilliant Violet 421, and N332-GT2KO biotin + streptavidin phycoerythrin.
  • a BD FACSAria was used for all cell sorting. Cells were sorted at a flow rate of 1500 events/second using an 85 -pm nozzle. Sorting stringency was set to a strict setting to obtain one cell per well. Single B cells were sorted directly into cold lysis buffer or N332-specific clonal B cell lines were generated and interrogated as in 4 . cDNA synthesis, nested BCR PCR, Sanger sequencing and sequence analysis was carried out as in 10 . Sorting was done with FACSDiva (BD) software and post-sort analyses were done with FlowJo (FlowJo, LLC).
  • B cells were isolated from CD45.2 C57BL/6J ("WT") or B ⁇ d ®41 KI mice of 8-10 weeks of age, and cells were resuspended in 150 pi of PBS and injected i.v. into CD45.1 B6.
  • SJL-Ptprc a Pepc b /BoyJ recipient animals (5 x 10 3 cells per mouse for BG 18" H transfers and 50 x 10 3 cells per mouse for C57BL/6 transfers).
  • recipient mice were injected i.p. with 10 pg GT2- or MD39-NPs with Sigma adjuvant (Sigma, # S6322 SIGMA). After eight days, mice were sacrificed to harvest spleen samples.
  • Antigen tetramers were prepared by conjugating for 1 hour (room temp.) biotinylated N332-GT2 and N332-GT2-KO trimers with fluorescently labeled streptavidins (Alexa Fluor 488, Alexa Fluor 647, eBioscience; Alexa Fluor 568, Thermo Fisher Scientific) in a 4: 1 molar ratio.
  • streptavidins conjugated with biotinylated Fab anti-IgM and biotinylated BSA were used as positive and negative staining controls, respectively (data not shown).
  • Single cell suspensions generated from spleen samples were depleted of red blood cells by ACK lysis, Fc blocked (BD Biosciences) and stained in FACS buffer (2% FCS/PBS) with antigen tetramers for 30 min at 4C, 50 nM concentration. Next, a cocktail of mAbs was added for 30 minutes at 4C.
  • the cocktail was: B220 PerCP-Cy5.5 (Clone RA3-6B2, Biolegend), IgD PE-Cy7 (Clone l l-26c.2a, Biolegend), CD4 APC-eFluor780 (Clone RM4-5, eBioscience), CD8a APC-eFluor780 (Clone 53-6.7, eBioscience), F4/80 APC-eFluor780 (Clone BM8, eBioscience), Ly-6G APC-eFluor780 (Clone RB6-8C5, eBioscience).
  • the cocktail was: CD38 Alexa Fluor 700 (Clone 90, Invitrogen), CD45.2 PE (Clone 104, Biolegend), CD45.1 PerCP-Cy5.5 (Clone A20, Biolegend), B220 PB (Clone RA3-6B2, Biolegend), CD95 PE-Cy7 (Clone Jo2, BD Bioscience), CD4 APC- eFluor780 (Clone RM4-5, eBioscience), CD8a APC-eFluor780 (Clone 53-6.7, eBioscience), F4/80 APC-eFluor780 (Clone BM8, eBioscience), Ly-6G APC-eFluor780 (Clone RB6-8C5, eBioscience).
  • VL BG18 iGL2 (Same as VL BG18 iGLO)
  • VL PRE8 (Same as VL BG18 iGLO)

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Genetics & Genomics (AREA)
  • Mycology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • AIDS & HIV (AREA)
  • Oncology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Nanotechnology (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • General Engineering & Computer Science (AREA)
  • Optics & Photonics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present disclosure relates to recombinant HIV Env polypeptides and their use in the treatment and prevention of HIV/AIDS.

Description

RECOMBINANT HIV ENV POLYPEPTIDES AND THEIR USE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority of U.S. Provisional Application No. 62/774,178 filed December 1, 2018, which is incorporated herein in its entirety.
FIELD OF THE INVENTION
[0002] The field of the invention generally relates to recombinant HIV Env polypeptides and their use in the treatment and prevention of HIV/AIDS.
GOVERNMENT INTEREST
[0003] The invention was made with government support. The U.S. government has certain rights in the invention.
BACKGROUND
[0004] HIV broadly neutralizing antibodies (bnAbs) have been isolated from infected individuals, but their induction by vaccination remains a major unmet need for global health. The majority of HIV bnAbs engage the HIV Env trimer with long immunoglobulin heavy chain complementarity determining region 3 (HCDR3) loops that reach through the glycan shield to contact relatively conserved epitopes on the underlying protein surface. A key barrier to inducing long-HCDR3 bnAbs is the priming of appropriate naive B cells that occur at very low frequency in the human repertoire and generally have very low or no detectable affinity for HIV Env. Furthermore, the field lacks methods to design appropriate priming immunogens. Thus, there remains a need for the development of immunogenic compositions capable of eliciting the production of broadly neutralizing antibodies. BRIEF SUMMARY
[0005] In one aspect, described herein are isolated polypeptides comprising a variant HIV
Env gpl20 polypeptide.
[0006] In one aspect, described herein are recombinant HIV Env trimers comprising an antigenic polypeptide described herein.
[0007] In one aspect, described herein are nanoparticles comprising an antigenic polypeptide described herein.
[0008] In one aspect, described herein are isolated polynucleotides encoding the isolated polypeptides, HIV ENV trimmers, and nanoparticles described herein.
[0009] In one aspect, described herein are RNA replicons comprising an isolated polynucleotide described herein.
[0010] In one aspect, described herein are recombinant viruses comprising an isolated polynucleotide described herein.
[0011] In one aspect, described herein are vectors comprising an isolated polynucleotide described herein.
[0012] In one aspect, described herein are methods of producing an antigenic polypeptide,
HIV ENV trimmer, or nanoparticle described herein.
[0013] In one aspect, described herein are liposomes and VLPs comprising an antigenic polypeptide described herein.
[0014] In one aspect, described herein are pharmaceutical compositions comprising an antigenic polypeptide, recombinant HIV Env trimer, nanoparticle, polynucleotide, recombinant virus, VLP, or liposome described herein.
[0015] In one aspect, described herein are immunogenic compositions comprising an antigenic polypeptide, recombinant HIV Env trimer, nanoparticle, polynucleotide, recombinant virus, VLP, or liposome described herein.
[0016] In one aspect, described herein are methods for generating an immune response to
HIV Env gpl20 in a subject, comprising administering to the subject an effective amount of an immunogenic composition described herein.
[0017] In one aspect, described herein are methods of reducing the likelihood of HIV infection in a subject exposed to HIV comprising administering to the subject an effective amount of an immunogenic composition or pharmaceutical composition described herein. [0018] In one aspect, described herein are methods of reducing the risk of a subject becoming infected with HIV comprising administering to the subject in need thereof an effective amount of an immunogenic composition or pharmaceutical composition described herein.
[0019] In one aspect, described herein are methods of preventing HIV infection comprising administering to a subject in need thereof an effective amount of an immunogenic composition or pharmaceutical composition described herein.
[0020] In one aspect, described herein are methods of treating HIV/AIDS comprising administering to a subject in need thereof an effective amount of an immunogenic composition or pharmaceutical composition described herein.
[0021] In one aspect, described herein are isolated antibodies comprising a VH and a VK described herein.
[0022] In one aspect, described herein are methods of identifying a vaccine candidate variant HIV Env polypeptide using an antibody described herein.
[0023] In some embodiments, the disclosure provides:
[1.] An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises one or more of
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
(i) CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) CTNYAPNLLSNMRGEL (SEQ ID NO: 72) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) CTNY APNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iv) CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(v) CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(vi) CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of (i) TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(ii) TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of
(i) TGDIIGDIRQAHCNV S (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) FGDIIGDIRMAHCNV S (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iv) FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(v) FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of
(i) SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(iv) SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[2.] An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of (i) CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) CTNYAPNLLSNMRGEL (SEQ ID NO: 72) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) CTNYAPNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iv) CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(v) CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(vi) CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of
(i) TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(ii) TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of
(i) TGDIIGDIRQAHCNV S (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) FGDIIGDIRMAHCNV S (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iv) FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(v) FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of (i) SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(iv) SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[3.] An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises one or more of
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
(i) CTNVTNNITDDMRGEL (SEQ ID NO: 71),
(ii) CTNY APNLLSNMRGEL (SEQ ID NO: 72),
(iii) CTNYAPNLLSNMRGEI (SEQ ID NO: 73),
(iv) CTNYAPKLLSNMRGEI (SEQ ID NO: 74),
(v) CTNY APKLRSMMRGEI (SEQ ID NO: 75), or
(vi) CTNY APNLRSDMRGEI (SEQ ID NO: 76);
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of
(i) TRPNNNT (SEQ ID NO: 77), or
(ii) TRPSNNT (SEQ ID NO: 78);
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of
(i) TGDIIGDIRQAHCNV S (SEQ ID NO: 79),
(ii) FGDIIGDIRMAHCNVS (SEQ ID NO: 80),
(iii) FGDVLGDVRMAHCNIS (SEQ ID NO : 81 ),
(iv) FGDVLGHVRMAHCNIS (SEQ ID NO: 82), or
(v) FGDVLGDVDMAKCTIS (SEQ ID NO: 83); and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of (i) SITLPCR (SEQ ID NO: 84),
(ii) SIVLPCR (SEQ ID NO: 85),
(iii) SLILPCR (SEQ ID NO: 86), or
(iv) SLILPCW (SEQ ID NO: 87);
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[4.] An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
(i) CTNVTNNITDDMRGEL (SEQ ID NO: 71),
(ii) CTNY APNLLSNMRGEL (SEQ ID NO: 72),
(iii) CTNYAPNLLSNMRGEI (SEQ ID NO: 73),
(iv) CTNYAPKLLSNMRGEI (SEQ ID NO: 74),
(v) CTNY APKLRSMMRGEI (SEQ ID NO: 75), or
(vi) CTNY APNLRSDMRGEI (SEQ ID NO: 76);
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of
(i) TRPNNNT (SEQ ID NO: 77), or
(ii) TRPSNNT (SEQ ID NO: 78);
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of
(i) TGDIIGDIRQAHCNV S (SEQ ID NO: 79),
(ii) FGDIIGDIRMAHCNVS (SEQ ID NO: 80),
(iii) FGDVLGDVRMAHCNIS (SEQ ID NO : 81 ),
(iv) FGDVLGHVRMAHCNIS (SEQ ID NO: 82), or
(v) FGDVLGDVDMAKCTIS (SEQ ID NO: 83); and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of
(i) SITLPCR (SEQ ID NO: 84),
(ii) SIVLPCR (SEQ ID NO: 85),
(iii) SLILPCR (SEQ ID NO: 86), or
(iv) SLILPCW (SEQ ID NO: 87); wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[5.] An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of
(a) CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77),
FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
(b) CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77),
FGDVLGDVRMAHCNIS (SEQ ID NO: 81), and SLILPCR (SEQ ID NO: 86), respectively,
(c) CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77),
FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
(d) CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78),
FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
(e) CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78),
FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCW (SEQ ID NO: 87), respectively,
(f) CTNYAPNLRSDMRGEI (SEQ ID NO: 76), TRPNNNT (SEQ ID NO: 77),
FGDVLGDVDMAKCTIS (SEQ ID NO: 83), and SLILPCR (SEQ ID NO: 86), respectively,
(g) CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
(h) CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
(i) CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
(j) CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
(k) CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively, or
(l) CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[6.] An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[7.] An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), and SLILPCR (SEQ ID NO: 86), respectively,
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[8.] An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[9.] An isolated polypeptide comprising a variant HIV Env gp 120 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO:
86), respectively,
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[10.] An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCW (SEQ ID NO:
87), respectively,
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[I T] The isolated polypeptide of any one of [1] to [10], wherein the variant HIV Env gpl20 polypeptide comprises N332.
[12.] The isolated polypeptide of any one of [1] to [11], wherein the N-terminal residue of the variant HIV Env gpl20 polypeptide is one of HIV Env positions 1-35. [13.] The isolated polypeptide of any one of [1] to [12], wherein the C-terminal residue of the variant HIV Env gpl20 polypeptide is one of HIV Env positions 503-512.
[14.] The isolated polypeptide of any one of [1] to [13], wherein the variant HIV Env gpl20 polypeptide is a variant of the BG505, BG505 N332, BG505 MD39, BG505 MD64, BG505 MD39 N332, BG505 MD39 1 ImutB, or BG505 MD39 17mutE gpl20 polypeptide.
[15.] The isolated polypeptide of any one of [1] to [13], wherein the variant HIV Env gpl20 polypeptide is a variant of the BG505 MD39 N332 gpl20 polypeptide.
[16.] The isolated of any one of [1] to [13 wherein the variant HIV Env gpl20 polypeptide is a variant of the BG505 MD64 N332 gpl20 polypeptide.
[17.] An isolated polypeptide comprising
(a) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT1,
(b) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2,
(c) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT3,
(d) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT4,
(e) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT5, or
(f) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2-KO.
[18.] An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT1, [19.] An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2.
[20.] An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT3.
[21.] An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT4.
[22.] An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT5.
[23.] An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD64 N332-GT2.
[24.] An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env polypeptide comprises
(a) the amino acid sequence of BG505 MD39 N332-GT1,
(b) the amino acid sequence of BG505 MD39 N332-GT2,
(c) the amino acid sequence of BG505 MD39 N332-GT3,
(d) the amino acid sequence of BG505 MD39 N332-GT4,
(e) the amino acid sequence of BG505 MD39 N332-GT5, or
(f) the amino acid sequence of BG505 MD39 N332-GT2-KO.
[25.] An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332-GT1.
[26.] An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332-GT2.
[27.] An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332-GT3.
[28.] An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332-GT4. [29.] An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332-GT5.
[30.] An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD64 N332-GT2.
[31.] The isolated polypeptide of any one of [1] to [30], wherein the variant HIV Env polypeptide comprises K137 and H325.
[32.] The isolated polypeptide of any one of [1] to [30], wherein the variant HIV Env polypeptide comprises K137 and P325.
[33.] The isolated polypeptide of any one of [1] to [30], wherein the variant HIV Env polypeptide comprises K137, M141 and H325.
[34.] The isolated polypeptide of any one of [1] to [30], wherein the variant HIV Env polypeptide comprises K137, R139, M141, and H325.
[35.] The isolated polypeptide of any one of [1] to [30], wherein the variant HIV Env polypeptide comprises K137 and P325.
[36.] The isolated polypeptide of any one of [1] to [30], wherein the variant HIV Env polypeptide comprises K137, M141, and P325.
[37.] The isolated polypeptide of any one of [1] to [30], wherein the variant HIV Env polypeptide comprises K137, R139, M141, and P325.
[38.] The isolated polypeptide of any one of [1] to [37] that specifically binds to the BG18 antibody.
[39.] The isolated polypeptide of any one of [1] to [38] that specifically binds to an antibody selected from the group consisting of BG18.11 and BG18.6.
[40.] The isolated polypeptide of any one of [1] to [39] that specifically binds to an antibody selected from the group consisting of BG18 iGLO, BG18 iGLl, and BG18 iGL2.
[41.] The isolated polypeptide of any one of [ 1 ] to [40] that specifically binds to an antibody selected from the group consisting of PREl, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PREIO, PREl 1, PREl 2, PREl 3, PRE14, and PRE15.
[42.] The isolated polypeptide of any one of [1] to [41] that specifically binds to the PG121 iGL antibody.
[43.] The isolated polypeptide of any one of [1] to [42] that specifically binds to an antibody comprising the VH and VL of
(a) VH4-59 and BG18 iGL, respectively; (b) VH1-69 and BG18 iGL, respectively;
(c) VH5-51 and BG18 iGL, respectively;
(d) VH3-33 and BG18 iGL, respectively;
(e) VH3-23 and BG18 iGL, respectively;
(f) BG18 iGLl and VL3-19, respectively;
(g) BG18 iGLl and VL3-10, respectively;
(h) BG18 iGLl and VL3-1, respectively;
(i) BG18 iGLl and VL3-21, respectively; or
(j) BG18 iGLl and VL2-8, respectively.
[44.] The isolated polypeptide of any one of [1] to [43] that comprises a gpl40.
[45.] The isolated polypeptide of any one of [1] to [43] that comprises a gpl60.
[46.] A recombinant HIV Env trimer comprising the isolated polypeptide of any one of [1] to [45]
[47.] The recombinant HIV Env trimer of [46] that is a homotrimer.
[48.] The recombinant HIV Env trimer of [46] that is a heterotrimer.
[49.] The recombinant HIV Env trimer of any one of [46] to [48] that comprises gpl20-gp41 heterodimers.
[50.] The recombinant HIV Env trimer of any one of [46] to [48] that comprises gpl20-gp41 heterodimers wherein the heterodimers are covalently linked.
[51.] The recombinant HIV Env trimer of any one of [46] to [48] that comprises gp 120-gp41 fusions.
[52.] The recombinant HIV Env trimer of any one of [46] to [48] that is a stabilized trimer.
[53.] The recombinant HIV Env trimer of any one of [46] to [48] that is an SOSIP, NFL or
UFO trimer.
[54.] The recombinant HIV Env trimer of any one of [46] to [48] that is an SOSIP trimer.
[55.] A nanoparticle comprising the isolated polypeptide of any one of [1] to [45]
[56.] The nanoparticle of [55], wherein the nanoparticle is a ferritin nanoparticle, an encapsulin nanoparticle, a Sulfur Oxygenase Reductase (SOR) nanoparticle, a lumazine synthase nanoparticle or a pyruvate dehydrogenase nanoparticle.
[57.] The nanoparticle of [55], wherein the nanoparticle comprises N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5.
[58.] The nanoparticle of [55], wherein the nanoparticle is a ferritin nanoparticle. [59.] The nanoparticle of [55], wherein the nanoparticle is a ferritin nanoparticle having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of the amino acid sequence of SEQ ID NO.
13 or 15.
[60.] The nanoparticle of [55], wherein the nanoparticle is a ferritin nanoparticle comprising the amino acid sequence of SEQ ID NO. 13 or 15.
[61.] An isolated polynucleotide encoding the isolated polypeptide of any one of [1] to [45], recombinant HIV Env trimer of any one of [46] to [54], or the nanoparticle of [55] to [60]
[62.] The isolated polynucleotide of [61], which is a DNA.
[63.] The isolated polynucleotide of [61], which is an mRNA.
[64.] The isolated polynucleotide of [63], wherein the mRNA comprises a modified nucleotide.
[65.] An RNA replicon comprising the isolated polynucleotide of [61] .
[66.] An isolated vector comprising the polynucleotide of [61] .
[67.] The isolated vector of [66], wherein the vector is a viral vector.
[68.] A recombinant virus comprising the polynucleotide of [61]
[69.] The recombinant virus of [68], which is a recombinant adeno-associated virus (AAV).
[70.] A host cell comprising the polynucleotide of any one of [61] to [64, or the vector of [66] or [67]
[71.] The host cell of [70], which is selected from the group consisting of E. coli,
Pseudomonas, Bacillus, Streptomyces, yeast, CHO, YB/20, NS0, PER-C6, HEK-293T, NIH-3T3, HeLa, BHK, Hep G2, SP2/0, Rl. l, B-W, L-M, COS 1, COS 7, BSC1, BSC40, BMT10 cell, plant cell, insect cell, and human cell in tissue culture.
[72.] A method of producing the isolated polypeptide of any one of [1] to [45], recombinant HIV Env trimer of any one of [46] to [54], or the nanoparticle of [55] to [60] comprising, culturing the host cell of [71] so that the polynucleotide is expressed and the isolated polypeptide, recombinant HIV Env trimer, or nanoparticle is produced.
[73.] A VLP comprising the polypeptide of any one of [1] to [45]
[74.] A liposome comprising the polypeptide of any one of [1] to [45]
[75.] A pharmaceutical composition comprising the isolated polypeptide of any one of [1] to [45], recombinant HIV Env trimer of any one of [46] to [54], the nanoparticle of any one of [55] to [60], the polynucleotide of any one of [61] to [64], the vector of [66] or [67], the recombinant virus of [68] or [69], VLP of [73], or the liposome of [74] and a pharmaceutically acceptable excipient.
[76.] An immunogenic composition comprising the isolated polypeptide of any one of [1] to [45], recombinant HIV Env trimer of any one of [46] to [54], the nanoparticle of [any one of [55] to [60], the polynucleotide of any one of [61] to [64], the vector of [66 or [67], the recombinant virus of [68] or [69], VLP of [73], or the liposome of [74] and a pharmaceutically acceptable excipient.
[77.] The immunogenic composition of [76], further comprising an adjuvant.
[78.] The immunogenic composition of [77], wherein the adjuvant comprises lecithin.
[79.] The immunogenic composition of [77], wherein the adjuvant comprises alum.
[80.] The immunogenic composition of [77], wherein the adjuvant comprises saponin, cholesterol and phospholipid.
[81.] The immunogenic composition of [7], wherein the adjuvant comprises carbomer homopolymer and lecithin.
[82.] The immunogenic composition of any one of [76] to [81] that is capable of eliciting a BG18 like response.
[83.] The immunogenic composition of any one of [76] to [81] that is capable of eliciting a BG18 like response in a human subject.
[84.] The immunogenic composition of any one of [76] to [81] that is capable of eliciting a BG18 like response in BG18gH B cell adoptive transfer recipient mice.
[85.] The immunogenic composition of any one of [76] to [81] that is capable of eliciting the production of an antibody that binds to N332-GT2.
[86.] The immunogenic composition of any one of [76] to [81] that is capable of eliciting the production of an antibody that binds to N332-GT2 with a higher affinity than to N332-GT2-KO.
[87.] The immunogenic composition of any one of [76] to [81] that is capable of eliciting the production of a broadly neutralizing antibody in a subject.
[88.] A method for eliciting an immune response to HIV Env gpl20 in a subject, comprising administering to the subject an effective amount of the immunogenic composition of any one of [76] to [81], thereby generating the immune response.
[89.] The method of [88], wherein the subject is a human.
[90.] The method of [88], wherein the subject is a non-human primate. [91.] The method of [88], wherein the subject is a BG18gH B cell adoptive transfer recipient mouse.
[92.] The method of [88], wherein the subject is a mouse.
[93.] A method of reducing the likelihood of HIV infection in a subject exposed to HIV comprising administering to the subject an effective amount of the immunogenic composition of any one of [76] to [81], or the pharmaceutical composition of [75]
[94.] A method of reducing the risk of a subject becoming infected with HIV comprising administering to the subject in need thereof an effective amount of the immunogenic composition of any one of [76] to [81], or the pharmaceutical composition of [75]
[95.] A method of preventing HIV infection comprising administering to a subject in need thereof an effective amount of the immunogenic composition of any one of [76] to [81], or the pharmaceutical composition of [75]
[96.] A method of treating HIV/AIDS comprising administering to a subject in need thereof an effective amount of the immunogenic composition of any one of [76] to [81], or the
pharmaceutical composition of [75]
[97.] The method of any one of [88] to [96], wherein the immunogenic composition comprises the isolated polypeptide of any one of [1] to [45]
[98.] The method of any one of [88] to [96], wherein the immunogenic composition comprises the recombinant HIV Env trimer of any one of [46] to [54]
[99.] The method of any one of [88] to [96], wherein the immunogenic composition comprises the nanoparticle of any one of [55] to [60]
[100.] The method of any one of [88] to [96], wherein the immunogenic composition comprises the polynucleotide of any one of [61] to [64]
[101.] The method of any one of [88] to [96], wherein the immunogenic composition comprises the vector of any one of [66] to [67] .
[102.] The method of any one of [88] to [96], wherein the pharmaceutical composition comprises the recombinant virus of any one of [68] to [69]
[103.] The method of any one of [88] to [102], further comprising administering at least one additional therapeutic agent.
[104.] The method of [103], wherein the additional therapeutic agent comprises an antiretroviral agent. [105.] The method of [103] or [104], wherein the additional therapeutic agent comprises a broadly neutralizing anti-HIV antibody.
[106.] The method of any one of [88] to [105], wherein the subject is a human.
[107.] The method of any one of [88] to [105], wherein the subject is a non-human primate.
[108.] An isolated antibody comprising
(a) the VHCDR1, VH CDR2, and VH CDR3 of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1-69, VH5-51, VH3-33, or VH3-23; and
(b) the VL CDR1, VL CDR2, and VL CDR3 of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VL3-10, VL3-19, VL3-1, VL3-21, or VL2-8.
[109.] An isolated antibody comprising the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3 of BG 18.11, BG 18.6, BG 18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
[110.] An isolated antibody comprising the 3 VH CDRs and 3 VL CDRs of
(a) VH4-59 and BG18 iGL, respectively;
(b) VH1-69 and BG18 iGL, respectively;
(c) VH5-51 and BG18 iGL, respectively;
(d) VH3-33 and BG18 iGL, respectively;
(e) VH3-23 and BG18 iGL, respectively;
(f) BG18 iGLl and VL3-19, respectively;
(g) BG18 iGLl and VL3-10, respectively;
(h) BG18 iGLl and VL3-1, respectively;
(i) BG18 iGLl and VL3-21, respectively; or
(j) BG18 iGLl and VL2-8, respectively.
[111.] An isolated antibody comprising
(a) the VH of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1,
PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1-69, VH5-51, VH3-33, or VH3-23; and (b) the VL of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2,
PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VL3-10, VL3-19, VL3-1, VL3-21, or VL2-8.
[112.] An isolated antibody comprising the VH and VL of BG18.11, BG18.6, BG18 iGLO,
BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
[113.] An isolated antibody comprising the VH and VL of
(a) VH4-59 and BG18 iGL, respectively;
(b) VH1-69 and BG18 iGL, respectively;
(c) VH5-51 and BG18 iGL, respectively;
(d) VH3-33 and BG18 iGL, respectively;
(e) VH3-23 and BG18 iGL, respectively;
(f) BG18 iGLl and VL3-19, respectively;
(g) BG18 iGLl and VL3-10, respectively;
(h) BG18 iGLl and VL3-1, respectively;
(i) BG18 iGLl and VL3-21, respectively; or
(j) BG18 iGLl and VL2-8, respectively.
[114.] A method for identifying a vaccine candidate variant HIV Env gpl20 polypeptide, the method comprising
(a) providing a library comprising a plurality of variant HIV Env gp 120 polypeptides;
(b) contacting the library with an antibody according to any one of [107] to [109]; and
(c) identifying a variant HIV Env gpl20 polypeptide that specifically binds to the antibody.
[115.] The method of [ 110] , wherein the plurality of variant HIV Env gp 120 polypeptides comprise variants of a parental HIV Env gpl20 polypeptide comprising one or more amino acid substitutions at a region selected from the VI loop comprising positions 131-154, V3 loop comprising positions 297-334, and□ 19 sheet comprising positions 413-419, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[116.] The method of [111], wherein the parental HIV Env gpl20 polypeptide comprises BG505, BG505 N332, BG505 MD39, BG505 MD39 N332, BG505 MD39 1 ImutB, or BG505 MD39 17mutE gpl20 polypeptide. BRIEF DESCRIPTION OF THE DRAWINGS
[0024] Figure 1. Structural and biophysical properties of BG18 germline targeting trimers. a) HCDR3 structural alignment of unliganded BG18 (PDB:5UD9) to PGT122 bound to BG505 SOSIP (PDB:4TVP). BG505 SOSIP is shown as a gray surface and the HCDR3s of BG18 and PGT122 are shown in purple and green, respectively. Conserved residues contacted by the HCDR3 (G324, D325, 1326, R327, Q328, H330, T415, and P417) are colored red. b) Cryo-EM derived structure of BG18 bound to BG505 MD39. MD39 is colored gray with the N332 and N392 glycans shown as green sticks. The BG18 HC and LC are colored purple and cyan, respectively. Red indicates conserved contact residues as in a) c) Cryo-EM derived structure of BG18 iGLO in complex with BG505 N332-GT2 with MD64 stabilizing mutationsl9. N332-GT2 is colored gray with the N332 and N392 glycans shown as green sticks. The BG18 iGLO HC and LC are colored purple and cyan, respectively. The same residues as in a) and c) are colored red. d) Schematic of the N332-GT1 and -GT2 directed evolution design process e) BG505 MD39 N332-GT binding affinities (KD) for BG18 iGLO-2 (red), BG18 iGLl with alternate germline VL (blue open symbols) or VH genes (blue filled symbols), and BG18 iGL variants containing NGS-derived HCDR3s (pre l-15) (black). Dashed line indicates the limit of detection.
[0025] Figure 2. Naive human B cell sorting with N332-GT trimers. a) Gating strategy for N332-GT epitope-specific sorting of naive human B cells b) HCDR3 length distribution in N332- GT sorted naive B cells compared to control B cells from DeKosky et al38 and Jardine et al39. c) Frequency of VL3-25/VL3-1 LCs from N332-GT2 sorted naive B cells relative to control B cells from DeKosky et al.38 and Jardine et al39. d) HCDR3 sequence and gene usage of three BG18- like precursors for which cryo-EM structures were determined. The BG18 iGLO, HMP1. HMP42, and HMP43 HCDR3 sequences shown are SEQ ID NO: 88-91, respectively e) SPR binding dissociation constants of HMP epitope -specific FAbs from naive B cells isolated with N332-GT1 and -GT2. Dashed line indicates the limit of detection f) Low resolution cryo-EM reconstructions of BG18-like precursors HMP1, HMP42, and HMP43 bound to N332-GT5 trimer with a low-pass filtered reconstruction of BG18-iGL0 (yellow) bound to N332-GT2 (gray) shown for reference. The approximate densities of the HMP Fabs are colored green, purple and blue for HMP1, HMP42, and HMP43, respectively based on comparison to BG18 iGLO. The approximate densities of the trimers are colored gray.
[0026] Figure 3. Immunization of BG18gH B cell adoptive transfer recipient mice with N332-GT NPs. a) Representative examples of fluorescence-activated cell sorting (FACS) gating to identify epitope-specific (N332-GT2++/N332-GT2-KO-) B cells in BG18gH and WT mice b) Frequency of epitope -specific B cells in non-immunized BG18gH and WT mice c) Pie charts showing the distribution of VH and VL genes in epitope -specific naive B cells in non-immunized BG18gH mice (left) and epitope -specific GC (CD381owCD95+) B cells 8 days after immunization of BG18gH B cell adoptive transfer recipient mice (right) d) Representative examples of FACS gating to identify epitope-specific CD45.2+ GC B cells 8 days after immunization of mouse recipients of either BG18gH (top) or WT B cells (bottom) e) Frequency of GC B cells (left) or CD45.2+ GC B cells (right) in four immunization conditions f) Frequency of CD45.2+ (left) or CD45.1+ (right) epitope-specific B cells in four immunization conditions g) Serum ELISA 50% equilibrium dilution values for GT2 and GT2-KO at day 14 after immunization for four immunization conditions h) Dissociation constants measured by SPR for the interaction of N332- GT2 trimer with Fabs derived from epitope-specific naive B cells in non-immunized BG18gH mice (left) and epitope-specific GC B cells 8 days after immunization of BG18gH B cell adoptive transfer recipient mice (right). Statistical significance tests in e), f), and g) were unpaired t tests, with * indicating p=0.024 and *** indicating p<0.001.
[0027] Figure 4. Model of BG18 bound to BG505 SOSIP that was used to guide GT design. a) Model of BG18 bound to BG505 SOSIP obtained by HCDR3 alignment of unliganded BG18 (PDB:5UD9) to PGT122 bound to BG505 SOSIP (PDB:4TVP). BG505 SOSIP, gray; BG18 HC purple; BG18 LC, cyan; N332 glycan, green sticks. Conserved residues contacted by the HCDR3 (G324, D325, 1326, R327, Q328, H330, T415, and P417) are colored red. b) PGT122 complexed with BG505 SOSIP shown for comparison. BG505 SOSIP, gray; PGT122 HC, purple; PGT122 LC, cyan; N332 glycan, green sticks. Conserved residues contacted by the HCDR3 are colored red as in a) c) Model of BG18 bound to BG505 SOSIP aligned to the cryo-EM derived structure of BG18 bound to BG505 MD39 from Fig lb. BG505 SOSIP is not shown in this alignment. MD39, gray; BG18 HC, purple; BG18 LC, cyan; N332 glycan, green sticks. Conserved residues contacted by the HCDR3 are colored red as in a) d) BG18/SOSIP model indicates that the HCDR1 R29 interacts with the N332 glycan. SOSIP, gray; BG18 HC, purple; N332 glycan, green sticks e) HCDR1 somatic mutation R29 was reverted to germline Isoleucine and tested for neutralization on a panel of 24 BG18-sensitive pseudoviruses. *IC50 values for BG18 were taken from Freund et all5.
[0028] Figure 5. Minimally mutated BG18 variants and alternate VL variants by structural model-guided design, a) Neutralization IC50s on a panel of 24 BG18-sensitive pseudoviruses by less mutated variants of BG18 and variants of BG18 containing alternate LCs as indicated. One variant (BG 18.11, "minBG18") is only 11% mutated in VHVL and was found to be 59% as broad as BG18 and had similar potency to BG18. Another variant with 6% mutation in VHVL (BG18.6) neutralized 26% of the viruses. IC50 values for BG18 were taken from Freund et all5. For testing alternate LCs (VL3-1, VL3-21, VL2-8), BG18 somatic mutations were incorporated into each LC and paired with the mature BG18 HC. b) The model from Figure 4a is shown with VHVL somatic mutations (red spheres) in BG18, BG18.11 and BG18.6. The essential VH mutations are localized to the HCDR1, whereas the VL mutations are localized to the LCDR2 and LCDR3. c) VH/VL sequences of less mutated variants of BG18. VH4-4, BG18 VH, BG18.11 VH, BG18.6 VH, VL3-25, BG18 VL, BG18.11 VL, and BG18.6 VL comprise the amino acid sequences of SEQ ID NO: [VH4-4], 17, 19, 21, [VL3-25], 18, 20, and 22 respectively.
[0029] Figure 6. Identifying BG18-like HCDR3s and estimating the BG18 precursor frequency in the human B cell repertoire, a) Table defining key BG18-like features and their frequencies in NGS datasets ofBCR HCs from HIV-uninfected donors. Frequencies for BG18-like feature sets i (HCDR3 main features) and ii (HCDR3 junction features) were measured from NGS data from Briney et al.18 for 10 donors for which sequencing used UIDs to discriminate PCR artifacts from true unique mRNA transcripts. On average, -80% of the sequences from these 10 donors were IgM, and of those, -50% had two or fewer nucleotide mutations in the VH gene indicating that they were enriched for naive B cells 18. Sequences of BG18 somatic variants are described in Freund et al.15 The frequency for feature set iii (VL family) was measured from the HC-LC paired sequences in DeKosky et al38. b) HCDR3 amino acid sequence alignment for BG18, BG18 iGLO-2, and 15 NGS-derived BG18-like HCDR3s. VH CDR3 of BG18, BG18 iGL0-iGL2, and Pre l to 15 have the amino acid sequence of SEQ ID NO: 94, 88, and 95-111, respectively c) Graph of the frequencies for Feature Sets i or (i and ii), as defined in a), for all 14 donors. Each symbol represents one donor, and the total number of sequences for each donor in the NGS dataset is given in the key. Black symbols are for the 10 donors from Briney et al.18 sequenced using UIDs, and red symbols are for the 4 donors sequenced here without the use of UIDs. Bars denote geometric mean and geometric standard deviation measured over the 10 UID donors only. Also see Methods for a detailed explanation of the BG18-like precursor frequency estimate.
[0030] Figure 7. Mutations in N332-GT trimers and strategy for designing N332-GT5. a) GT mutations present in N332-GT trimers are localized to the VI, V3, and b 19. Amino acid changes relative to BG505-MD39 are shown in red. Sequences shown are CTNVTNNITDDMRGEL (SEQ ID NO: 71), CTNYAPNLLSNMRGEL (SEQ ID NO: 72),
CTNYAPNLLSNMRGEI (SEQ ID NO: 73), CTNYAPKLLSNMRGEI (SEQ ID NO: 74),
CTNY APKLRSMMRGEI (SEQ ID NO: 75), CTNYAPNLRSDMRGEI (SEQ ID NO: 76),
TRPNNNT (SEQ ID NO: 77), TRPSNNT (SEQ ID NO: 78), TGDIIGDIRQAHCNV S (SEQ ID
NO: 79), F GDIIGDIRMAHCNV S (SEQ ID NO: 80), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), F GD VLGHVRM AHCNI S (SEQ ID NO: 82), F GD VLGD VDMAKCTI S (SEQ ID NO: 83), SITLPCR (SEQ ID NO: 84), SIVLPCR (SEQ ID NO: 85), SLILPCR (SEQ ID NO: 86), SLILPCW (SEQ ID NO: 87). b) SPR binding KDs demonstrating that mutations in the VI loop provide large gains in affinity for binding to BG18 iGLl . c) Schematic showing the flow of iterative directed evolution for the design ofN332-GT5.
[0031] Figure 8. Structural comparison of BG18 and BG18 iGLO complexes, a) HCDR3 alignment of BG18 (purple) and BG18 iGLO (pink) shows that there is a large conformational change in the HCDR1 of BG18 that brings it into proximity to the N332 glycan (green sticks). Despite limited resolution, the density map suggests that this rearrangement allows interaction with the N332 glycan through numerous HCDR1 side chain interactions, all of which are somatic mutations b) Effect of reverting HCDR1 residues D32 and H33 to germline in a less mutated variant of BG18 measured by ELISA binding to BG505-MD39. c) The LC of BG18 (cyan) straddles the VI loop (tan), but residues 142-152 of VI are disordered, suggesting that this loop is utilized differently by mature BG18 compared to BG18 iGLO. Notably, the LCDR2 R54 is inserted into a cleft formed by the GDIR motif at the base of V3 (yellow), and the N-acetylglucosamine residues at N137 and N 156 (tan sticks) d) There is a rearrangement of the LCDR2 in BG18 relative to BG18 iGLO, which allows the mature BG18 LC to accommodate the topology of the VI loop. The malleability of the LCDR2 loop is accentuated by the observation that it is fully resolved when in complex with the trimer but disordered in the published ligand-free crystal structure of the Fab alonel5. e) The VI loop of BG505 MD64 N332-GT2 is shown in tan with the side chains of the most important mutations shown as sticks. The side chain of K137 in VI fits a groove in the LC (dark blue) LCDR1, with the aliphatic portion forming a CH-p interaction with LCDR1 Y32 and the e-amino group a salt bridge with LCDR2 D51, as well as potential hydrogen bonding with nearby backbone carbonyls and the side chain of LFR3 S66. Additionally, the HCDR3 (pink) packs against VI, with the side chain of R139 situated between the variable light and heavy chains of the antibody. The mutation of the VI residue D141 to methionine further stabilizes this region, as it interacts favorably with HCDR3 IlOOe. [0032] Figure 9. Cryo-EM statistics, a) Data collection, processing, building and validation statistics b) Summary of buried solvent accessible surface area between Fabs and Env trimer in high resolution cryo-EM datasets. Area values are totals buried on both Fab and Env and are stated in units of A2. No glycan, gene lacks PNGS consensus sequence.
[0033] Figure 10. Naive human B cell repertoire screening of llmutB, N332-GT1 and N332-GT2. In total, 57 million B cells were screened from 4 donors using l lmutB as a sorting probe a) Gating strategy for l lmutB epitope-specific sorting of naive human B cells b) HmutB+l lmutB+MD39neg B cells were sorted and clonal B cell lines were generated. Culture supernatants from >1000 clonal B cell lines were tested by ELISA for specificity to l lmutB and MD39. c) Many clonal lines did not show binding to MD39 with a strep tag, but did show specificity MD39 with a His tag, implying that many of the clonal lines were not MD39-specific. d) Summary ELISA results for eight representative B cell clonal lines with differing polyreactivity profiles, but all positive for l lmutB and MD39. KLH (keyhole limpet hemocyanin), SA-PE (streptavidin- phycoerythrin), HIV V3 peptide (CTRPNNNTRKSIRIGPGQAFYATGDIIGDIRQAHC), and LCMV peptide (DIYKGVYQFKSV) were used to test for polyreactivity.e) B cell lines were sequenced for HC and LC. e) Frequency of N332-GT probe double positive (N332-GT+N332- GT+) of total IgG negative B cells as shown in Figure 2a left f) Frequency of N332 epitope-specific B cells among N332-GT probe double positive B cells (MD39neg of N332-GT1+N332-GT1+ or N332-GT2KOneg of N332-GT2+N332-GT2+) as shown in Figure 2a right g) Frequency of N332 epitope-specific B cells of total IgG negative B cells (N332-GTl+N332-GTl+MD39neg or N332- GT2+N332-GT2+N332-GT2KOneg) as shown in Figure 2a right h) Epitope classification among 45 HMP Fabs from naive human B cells. 17 HMP Fabs showed epitope-specific binding to N332- GT trimers (red). Three Fabs showed epitope-specificity to non-N332 epitopes on N332-GT trimers (white). The remaining Fabs had weak/undetectable binding to N332-GT trimers (gray) i) Genetic features of Ab H-L paired sequences and expressed Fabs isolated from N332-GT1 and N332-GT2 B cell sorting.
[0034] Figure 11. Biophysical characterization of N332-GT2 nanoparticles, a) SPR KDs for N332-GT2 trimer and NP binding to BG18 precursors. The NP was captured as ligand with PGT128, and Fabs were flowed as analytes b) SPR analysis of N332-GT2 NP binding to a panel of non-nAb and bnAbs. The format of the experiment was the same as in a) c) SECMALS analysis of N332-GT2 NPs. The black line indicates the UV trace and the red line indicates the protein molecular weight. [0035] Figure 12. Calibration of transferred CD45.2 B cells into CD45.1 recipient mice.
Splenocytes from transferred recipient mice were analyzed by flow cytometry. CountBright Beads (Invitrogen) were added to the samples (5 xl06 splenocytes - quantified by Nucleocounter, ChemoMetec USA) to determine exact cell population numbers a) The raw data acquired at the flow cytometer b) The normalized values in the stained samples, obtained by multiplying the raw data by the beads multiplication factor (starting beads / beads measured) c) The total cell numbers for the mouse spleens, obtained by multiplying the values for 5 xl06 splenocytes by the spleen multiplication factor (total splenocytes / 5). d+e) Histograms of HC amino acid mutations d) and LC VL amino acid mutations e) for the 136 Abs derived from epitope -specific GC B cells 8 days after immunization of BG18gH B cell adoptive transfer recipient mice.
[0036] Figure 13. Overall strategy for designing GT immunogens to prime precursors of HCDR3-dependent antibodies. The BCR HC repertoires of 14 HIV-negative donors were sequenced and queried bioinformatically to identify HCDR3s with key features similar to a known bnAb indicating that such HCDR3s have potential to contribute to bnAb precursors. That process validated the bnAb for germline targeting immunogen design. In the top panel, the human repertoire is shown in green with HCDR3 -dependent bnAb precursors shown in red. The second panel shows the iterative immunogen design process. To design a GT immunogen capable of binding to human naive potential precursor B cells, potential bnAb precursor antibodies containing NGS-derived HCDR3s were used for multi-target iterative immunogen library screening. Key aspects of preclinical immunogen validation are summarized in the third panel. Structural analyses confirmed the expected binding orientation between GT immunogen and iGL Ab. Sorting naive human B cells with the GT immunogens was employed to isolate bona fide potential precursors that have features matching the target precursors. Cryo-EM and sequence analysis of isolated epitope-specific precursors can serve to refine and broaden the definition of potential bnAb precursors, enabling iterative design improvements. Adoptive transfer immunization experiments with inferred- germline bnAb knock-in B cells demonstrated that a GT immunogen can prime rare target precursor B cells.
DETAILED DESCRIPTION
[0037] In one aspect, provided herein are HIV Env variant polypeptides and immunogenic compositions comprising thereof that are capable of eliciting the formation of antibodies with binding properties similar to the BG18 broadly neutralizing antibody. The inventors used next generation sequencing (NGS) and bioinformatics to identify in HIV-uninfected donors germline encoded heavy chain CDRs that share key properties with the heavy chain CDR3 -dependent broadly neutralizing anti-HIV antibody (bnAb) BG18. BG18 inferred-germline (iGL) variants with the NGS-derived HCDR3 loops were used in a structural-model-guided, multitarget mammalian- display selection to identify variant Env polypeptides, including variant Env trimers that bind to diverse BG18 iGL antibodies and potential BG18 precursor antibodies. The N332-GT trimers were used to elicit an immune response. The inventors found that the variant Env polypeptides disclosed herein were capable of inducing the formation of BG18-like antibodies. As such, the HIV Env variant polypeptides and immunogenic compositions disclosed herein have the potential to elicit a protective broadly neutralizing response against HIV.
I. Definitions
[0038] To facilitate an understanding of the present invention, a number of terms and phrases are defined below.
[0039] The terms "human immunodeficiency virus" or "HIV," as used herein, refer generally to a retrovirus that is the causative agent for acquired immunodeficiency syndrome (AIDS), variants thereof (e.g., simian acquired immunodeficiency syndrome, SAIDS), and diseases, conditions, or opportunistic infections associated with AIDS or its variants, and includes HIV-Type 1 (HIV-1) and HIV-Type 2 (HIV-2) of any clade or strain therein, related retroviruses (e.g., simian immunodeficiency virus (SIV)), and variants thereof (e.g., engineered retroviruses, e.g., chimeric HIV viruses, e.g., simian-human immunodeficiency viruses (SHIVs)). In some embodiments, an HIV virus is an HIV -Type -1 virus. Previous names for HIV include human T-lymphotropic virus- 111 (HTLV-III), lymphadenopathy-associated virus (LAV), and AIDS-associated retrovirus (ARV).
[0040] Acquired immune deficiency syndrome ("AIDS") is a disease caused by the human immunodeficiency virus, or HIV.
[0041] As used herein, the term "envelope glycoprotein" or "Env" refers to the glycoprotein that is expressed on the surface of the envelope of HIV virions and the surface of the plasma membrane of HIV infected cells. "Envelope glycoprotein" or "Env" encompass, but are not limited to, native Env, an isoform of Env, or a recombinant variant of Env (e.g., SOSIP) derived from an HIV isolate. Env is the sole virally encoded gene product on the surface of the virus and, as such, is the only target of neutralizing antibodies. Env is a trimer of heterodimers composed of two non-covalently associated subunits: the receptor-binding gpl20 and the fusion machinery-containing gp41. Each subunit is derived from a gp 160 precursor glycoprotein following cleavage by cellular ftirins. HIV- 1 gpl20 binds the CD4 molecule on the surface of human target T cells to initiate the viral entry process, and following co-receptor engagement, fusion is mediated by gp41. gpl40 env is the uncleaved ectodomain of gpl60. In some embodiments, the Env is a BG505, 92BR020, JR-FL, YU-2, or JR-CSF Env polypeptide. In some embodiments, the Env is a BG505 Env polypeptide. UniProtKB accession number Q2N0S5-1, Q2N0S6-1, and Q2N0S7-1 provide BG505 env gpl60 polypeptide sequences. In some embodiments, the Env is a well-ordered Env trimer. In some embodiments, the BG505 Env polypeptide has the amino acid sequence of SEQ ID NO: 2.
[0042] The term "well-ordered Env trimer" or "well-ordered trimer" as used herein refers to an envelope glycoprotein trimer comprising three cleaved gpl40 polypeptides that closely mimic the quaternary structure of the Env ectodomain on the surface of the envelope of HIV or SIV virions and the surface of the plasma membrane of HIV or SIV infected cells. In one embodiment, the gpl40 polypeptides comprise MPER. In one embodiment, the well-ordered trimer comprises three MPERs. In one embodiment, the gpl20 and gp41 ectodomain is linked by a covalent linkage, for example, a disulfide bond. In one embodiment, the gpl40 polypeptide comprises one or more mutations to promote trimer formation. In one embodiment, the gpl40 polypeptide comprises one or more Cys substitutions to promote disulfide formation. In one embodiment, the well-ordered trimer is a SOSIP gpl40 trimer. Well-ordered SOSIP trimers have been disclosed in US Patent Appl. Pub. No. 2014/0212458, and Sanders, R. W. et ah, PLoS Pathog. 9, e l003618 (2013), each of which is incorporated by reference herein in its entirety. In one embodiment, a well-ordered trimer is formed from a clade A Env. In one embodiment, a well-ordered trimer is formed from a clade B Env. In one embodiment, a well-ordered trimer is formed from a clade C Env. In one embodiment, a well-ordered trimer is formed from a circulating recombinant form Env, wherein 'circulating recombinant form' (CRF) refers to a hybrid virus comprising a combination of genetic material from different subtypes. In one embodiment, a well-ordered trimer is Dul56.12 SOSIP comprising MPER. In one embodiment, a well-ordered Env trimer is a native flexibly linked (NFL) trimer as described in Shama, et ak, Cell Reports, l l(4):539-50 (2015). In one embodiment, a well- ordered Env trimer is a DS-SOSIP as described in Chuang GY, et al, J. Virology, 91(10). pii: e02268-16 (2017). In one embodiment, a well-ordered trimer is formed from an SIV Env. In one embodiment, a well-ordered trimer is an SIV Env SOSIP. In one embodiment, a well-ordered trimer is formed from an Env comprising a mutation (e.g., substitution or deletion) in the CD4 binding site. In one embodiment, a well-ordered trimer is formed from an Env comprising a mutation (e.g., substitution or deletion) in the CD4 binding site wherein the mutation reduces or disrupts the binding between Env and CD4. In one embodiment, a well-ordered trimer is a CRF or C108 SOSIP. See, e.g., Andrabi R., et al, Immunity 43(5): 959-973 (2015). In some embodiments, the gpl20 and gp41 ectodomain is linked by a peptide linker, for example, a Gly-Ser linker, as described in Georgiev IS, et al., J. Virology 89(10): 5318-5329 (2015). In some embodiments, the well-ordered Env trimer is stable.
[0043] The term "antibody" means an immunoglobulin molecule (or a group of immunoglobulin molecules) that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule. As used herein, the terms "antibody" and "antibodies" are terms of art and can be used interchangeably herein and refer to a molecule with an antigen-binding site that specifically binds an antigen.
[0044] Antibodies can include, for example, monoclonal antibodies, recombinantly produced antibodies, human antibodies, humanized antibodies, resurfaced antibodies, chimeric antibodies, immunoglobulins, synthetic antibodies, tetrameric antibodies comprising two heavy chain and two light chain molecules, an antibody light chain monomer, an antibody heavy chain monomer, an antibody light chain dimer, an antibody heavy chain dimer, an antibody light chain- antibody heavy chain pair, intrabodies, heteroconjugate antibodies, single domain antibodies, monovalent antibodies, single chain antibodies or single-chain Fvs (scFv), affybodies, Fab fragments, F(ab')2 fragments, disulfide-linked Fvs (sdFv), anti-idiotypic (anti-id) antibodies (including, e.g., anti-anti- Id antibodies), bispecific antibodies, and multi-specific antibodies. In certain embodiments, antibodies described herein refer to polyclonal antibody populations. Antibodies can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, or IgY), any class (e.g., IgGi, IgG2, IgG , IgG4, IgAi, or IgA2), or any subclasses (isotypes) thereof (e.g. IgGl, IgG2, IgG3, IgG4, IgAl and IgA2), of immunoglobulin molecule, based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu, respectively. The different classes of immunoglobulins have different and well-known subunit structures and three-dimensional configurations. Antibodies can be naked or conjugated or fused to other molecules such as toxins, radioisotopes, other polypeptides etc.
[0045] As used herein, the terms "antigen-binding domain," "antigen-binding region," "antigen binding site," and similar terms refer to the portion of antibody molecules, which comprises the amino acid residues that confer on the antibody molecule its specificity for the antigen (e.g., HIV Env MPER). The antigen-binding region can be derived from any animal species, such as mouse and humans.
[0046] As used herein, the terms "variable region" or "variable domain" are used interchangeably and are common in the art. The variability in sequence is concentrated in those regions called complementarity determining regions (CDRs) while the more highly conserved regions in the variable domain are called framework regions (FR). Without wishing to be bound by any particular mechanism or theory, it is believed that the CDRs of the light and heavy chains are primarily responsible for the interaction and specificity of the antibody with antigen (e.g., HIV Env MPER). In certain embodiments, the variable region comprises 3 CDRs (CDR1, CDR2, and CDR3) and 4 framework regions (FR1, FR2, FR3, and FR4) in the order of FR1-CDR1-FR2-CDR2-FR3-CDR3- FR4 from the N terminus to the C terminus. In certain embodiments, the variable region is a human variable region. In certain embodiments, the variable region comprises human CDRs and human framework regions (FRs). In certain embodiments, the variable region comprises CDRs and framework regions (FRs) wherein one or more of the CDRs were modified by a substitution, deletion, or insertion relative to the CDRs of a parental antibody. In certain embodiments, the variable region comprises CDRs and framework regions (FRs) wherein one or more of the FRs were modified by a substitution, deletion, or insertion relative to the FRs of a parental antibody. In certain embodiments, the variable region comprises CDRs and framework regions (FRs) wherein one or more of the CDRs and one or more of the FRs were modified by a substitution, deletion, or insertion relative to the CDRs and FRs of a parental antibody. In certain embodiments, the parental antibody is PGZL1. In certain embodiments, the variable region comprises human CDRs and primate (e.g., non-human primate) framework regions (FRs).
[0047] A skilled artisan understands that there are several methods for determining CDRs. One approach is based on cross-species sequence variability (i.e., Rabat EA, et al, Sequences of Proteins of Immunological Interest, (5th ed., 1991, National Institutes of Health, Bethesda Md.) ("Rabat"). Another approach is based on crystallographic studies of antigen-antibody complexes (Al-lazikani B., et al, J. Mol. Biol. 273:927-948 (1997)) ("Chothia"). In addition, combinations of these two approaches are sometimes used in the art to determine CDRs. In some embodiments, the CDR sequences are identified according to Rabat. In some embodiments, the CDR sequences are identified according to Chothia. It is understood that the identification of CDRs in a variable region also identifies the FRs as the sequences flanking the CDRs. [0048] The Kabat numbering system is generally used when referring to a residue in the variable domain (approximately residues 1-107 of the light chain and residues 1-113 of the heavy chain) (e.g., Kabat EA, et al., Sequences of Immunological Interest. (5th Ed., 1991, National Institutes of Health, Bethesda, Md.) ("Kabat").
[0049] The amino acid position numbering as in Kabat, refers to the numbering system used for heavy chain variable domains or light chain variable domains of the compilation of antibodies in Kabat EA, et al. (Sequences of Immunological Interest. (5th Ed., 1991, National Institutes of Health, Bethesda, Md.), "Kabat"). Using this numbering system, the actual linear amino acid sequence can contain fewer or additional amino acids corresponding to a shortening of, or insertion into, a FR or CDR of the variable domain. For example, a heavy chain variable domain can include a single amino acid insert (residue 52a according to Kabat) after residue 52 of H2 and inserted residues (e.g. residues 82a, 82b, and 82c, etc. according to Kabat) after heavy chain FR residue 82. The Kabat numbering of residues can be determined for a given antibody by alignment at regions of homology of the sequence of the antibody with a "standard" Kabat numbered sequence. Chothia refers instead to the location of the structural loops (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987)). The end of the Chothia CDR-H1 loop when numbered using the Kabat numbering convention varies between H32 and H34 depending on the length of the loop (this is because the Kabat numbering scheme places the insertions at H35A and H35B; if neither 35A nor 35B is present, the loop ends at 32; if only 35A is present, the loop ends at 33; if both 35A and 35B are present, the loop ends at 34). The AbM hypervariable regions represent a compromise between the Kabat CDRs and Chothia structural loops, and are used by Oxford Molecular's AbM antibody modeling software, available, for example, at bioinf.org.uk/abs/software. In some embodiments, the CDR sequences are identified according to Kabat. In some embodiments, the CDR sequences are identified according to Chothia. In some embodiments, the CDR sequences are identified according to AbM. In some embodiments, the VH CDR3 sequence is identified according to Kabat. In some embodiments, the VH CDR3 sequence is identified according to Chothia. In some embodiments, the VH CDR3 sequence is identified according to AbM. Loop Kabat AbM Chotbia
LI L24-L34 L24-L34 L24-L34
1,2 L50-L56 L50-L56 1,50-1 56
L3 L89-L97 L89-L97 1,894.97
I-Il I-I31-H35B H26-H35B H26-H32..34
(Kabat Numbering)
HI H31-H35 H26-H35 H26-H32
(Chotbia Numbering)
H2 H50-H65 H50-H58 H52-H56
H3 H95-H ! 02 H95-H102 H95-H102
[0050] The terms "VL" and "VL domain" are used interchangeably to refer to the light chain variable region of an antibody.
[0051] The terms "VH" and "VH domain" are used interchangeably to refer to the heavy chain variable region of an antibody.
[0052] The term "antibody fragment" refers to a portion of an intact antibody. An "antigen-binding fragment" refers to a portion of an intact antibody that binds to an antigen. An antigen-binding fragment can contain the antigenic determining variable regions of an intact antibody. Examples of antibody fragments include, but are not limited to Fab, Fab', F(ab')2, and Fv fragments, linear antibodies, and single chain antibodies.
[0053] A "monoclonal" antibody or antigen-binding fragment thereof refers to a homogeneous antibody or antigen-binding fragment population involved in the highly specific recognition and binding of a single antigenic determinant, or epitope. This is in contrast to polyclonal antibodies that typically include different antibodies directed against different antigenic determinants. The term "monoclonal" antibody or antigen-binding fragment thereof encompasses both intact and full- length monoclonal antibodies as well as antibody fragments (such as Fab, Fab', F(ab')2, Fv), single chain (scFv) mutants, fusion proteins comprising an antibody portion, and any other modified immunoglobulin molecule comprising an antigen recognition site. Furthermore, "monoclonal" antibody or antigen-binding fragment thereof refers to such antibodies and antigen-binding fragments thereof made in any number of manners including but not limited to by hybridoma, phage selection, recombinant expression, and transgenic animals.
[0054] The term "polyclonal antibody" describes a composition of different (diverse) antibody molecules, which are capable of binding to or reacting with several different specific antigenic determinants on the same or on different antigens. Usually, the variability of a polyclonal antibody is primarily located in the so-called variable regions of the polyclonal antibody, in particular in the CDR regions. In the present disclosure, a mixture of two or more polyclonal antibodies (a polycomposition) is produced in one mixture from a polyclonal polycomposition cell line, which is produced from two or more parental polyclonal cell lines each expressing antibody molecules, which are capable of binding to a distinct target, but it may also be a mixture of two or more polyclonal antibodies produced separately. A mixture of monoclonal antibodies providing the same antigen/epitope coverage as a polyclonal antibody described herein will be considered as an equivalent of a polyclonal antibody.
[0055] The term "chimeric" antibodies or antigen-binding fragments thereof refers to antibodies or antigen-binding fragments thereof wherein the amino acid sequence is derived from two or more species. Typically, the variable region of both light and heavy chains corresponds to the variable region of antibodies or antigen-binding fragments thereof derived from one species of mammals (e.g., mouse) with the desired specificity, affinity, and capability, while the constant regions are homologous to the sequences in antibodies or antigen-binding fragments thereof derived from another (usually human) to avoid eliciting an immune response in that species.
[0056] The term "epitope" or "antigenic determinant" are used interchangeably herein and refer to that portion of an antigen capable of being recognized and specifically bound by a particular antibody. When the antigen is a polypeptide, epitopes can be formed both from contiguous amino acids and noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained upon protein denaturing, whereas epitopes formed by tertiary folding are typically lost upon protein denaturing. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation.
[0057] "Binding affinity" generally refers to the strength of the sum total of non-covalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity" refers to intrinsic binding affinity, which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including those described herein. Uow-affinity antibodies generally bind antigen slowly and tend to dissociate readily, whereas high-affinity antibodies generally bind antigen faster and tend to remain bound longer. A variety of methods of measuring binding affinity are known in the art, any of which can be used for purposes of the present invention. Specific illustrative embodiments are described in the following. In certain embodiments, an antibody described herein binds to N332- GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 with a KD of at least about 0.1 mM or less, at least about 0.01 mM or less, at least about 1 nM or less, or at least about 0.1 nM or less. In certain embodiments, an anti-HIV antibody described herein binds to N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 with a KD of at least about 0.01 pM or less. In one embodiment, an anti- HIV antibody described herein is capable of binding to cells that express functional, well-ordered HIV-1 membrane Env trimers. In one embodiment, an anti -HIV antibody described herein is capable of binding to cells that display N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332- GT5 in a flow cytometry assay. In one embodiment, an anti-HIV antibody described herein is capable of binding to N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 in biolayer interferometry (BLI) assay. In one embodiment, an anti-HIV antibody described herein is capable of binding to N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 in ELISA. In one embodiment, an anti-HIV antibody described herein is capable of binding Env trimers from detergent-solubilized HIV-1 virions in an ELISA assay. In one embodiment, an anti-HIV antibody described herein is capable of binding Env trimers from detergent-solubilized HIV-1 virions in a BN-PAGE gel mobility-shift assay.
[0058] "Or better" when used herein to refer to binding affinity refers to a stronger binding between a molecule and its binding partner. "Or better" when used herein refers to a stronger binding, represented by a smaller numerical KD value. For example, an antibody, which has an affinity for an antigen of "0.6 nM or better", the antibody's affinity for the antigen is <0.6 nM, i.e. 0.59 nM, 0.58 nM, 0.57 nM etc. or any value less than 0.6 nM.
[0059] As used herein, the terms "immunospecifically binds," "immunospecifically recognizes," "specifically binds," and "specifically recognizes" are analogous terms in the context of antibodies and refer to molecules that bind to an antigen (e.g., epitope or immune complex) as such binding is understood by one skilled in the art. For example, a molecule that specifically binds to an antigen can bind to other peptides or polypeptides, generally with lower affinity as determined by, e.g., immunoassays, BIAcore®, KinExA 3000 instrument (Sapidyne Instruments, Boise, ID), ELISA, biolayer interferometry (BLI), flow cytometry or other assays known in the art. In a specific embodiment, molecules that immunospecifically bind to an antigen bind to the antigen with a KD that is at least 2 logs, 2.5 logs, 3 logs, or 4 logs lower than the KD when the molecules bind non- specifically to another antigen. In one example, the antibody may specifically bind to cells that express functional, well-ordered HIV-1 membrane Env trimers. In one example, the antibody may specifically bind to cells that display MPER-TM peptide. In one example, the antibody may specifically bind to the MPER peptide. In one example, the antibody may specifically bind to the MPER peptide in biolayer interferometry (BLI) assay. In one example, the antibody may specifically bind to the MPER peptide in ELISA assay. In one example, the antibody may specifically bind to Env trimers from detergent-solubilized HIV-1 virions. In one example, the antibody may specifically bind to Env trimers from detergent-solubilized HIV-1 virions in an ELISA assay. In one example, the antibody may specifically bind to Env trimers from detergent- solubilized HIV-1 virions in a BN-PAGE gel mobility-shift assay. The antibody may bind to N332- GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5 with a KD at least 2 logs, 2.5 logs, 3 logs, or 4 logs lower than KD of binding to other viral or non-viral polypeptides. An antibody that specifically binds to Env encompass, but are not limited to, antibodies that specifically bind to native Env, an isoform of Env, or a variant of Env (e.g., SOSIP) derived from an HIV isolate.
[0060] By "preferentially binds," it is meant that the antibody specifically binds to an epitope more readily than it would bind to a related, similar, homologous, or analogous epitope. Thus, an antibody, which "preferentially binds" to a given epitope would more likely bind to that epitope than to a related epitope, even though such an antibody may cross-react with the related epitope.
[0061] An antibody is said to "competitively inhibit" binding of a reference antibody to a given epitope if it preferentially binds to that epitope or an overlapping epitope to the extent that it blocks, to some degree, binding of the reference antibody to the epitope. Competitive inhibition may be determined by any method known in the art, for example, competition ELISA assays. An antibody may be said to competitively inhibit binding of the reference antibody to a given epitope by at least 90%, at least 80%, at least 70%, at least 60%, or at least 50%.
[0062] The term "broadly neutralizing antibody" or "bnAb," as used herein, with respect to HIV (e.g., HIV-1), refers to an antibody that recognizes HIV Env of more than one isolate or strain of HIV and inhibits or prevents receptor binding of target cells as evaluated in an in vitro neutralization assay. In one embodiment, a broadly neutralizing antibody inhibits infection of a susceptible target cell by HIV. In one embodiment, a broadly neutralizing antibody specifically binds an HIV Env and inhibits infection of a susceptible target cell (e.g., TZM-bl) by an HIV pseudovirus comprising an Env polypeptide. HIV pseudovirus neutralization assays have been disclosed in the art, for example, in Walker L.M., et al, Nature 477, 466-470 (2011), Li M., et ak, J. Virol. 79: 10108- 10125 (2005), each of which is incorporated herein by reference in its entirety for all purposes. In one embodiment, a broadly neutralizing antibody neutralizes 2, 3, 4, 5, 6, 7, 8, 9, or more HIV strains or pseudoviruses. In one embodiment, a broadly neutralizing antibody neutralizes 2, 3, 4, 5, 6, 7, 8, 9, or more HIV strains or pseudoviruses that belong to the same or different clades. In one embodiment, a broadly neutralizing antibody is capable of neutralizing HIV strains or pseudoviruses from at least two different clades. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least one clade B strain or pseudovirus and one clade C strain or pseudovirus. In one embodiment, a broadly neutralizing antibody is capable of neutralizing more than one clade B strain or pseudovirus and more than one clade C strain or pseudovirus. In one embodiment, a broadly neutralizing antibody is capable of neutralizing an HIV strain or pseudovirus from at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or at least eleven clades represented in the 130- member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing an HIV strain or pseudovirus from at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least thirteen, at least fourteen, or all fifteen clades selected from the group consisting of clades A, A (T/F), AC, ACD, B, B (T/F), BC, C, C (T/F), CD, CRF01 AE, CRF01 AE (T/F), CRF02 AG, D, and G. In one embodiment, a broadly neutralizing antibody is capable of neutralizing an HIV strain or pseudovirus from at least one, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or all eleven clades selected from the group consisting of clades A, AC, ACD, AE, AG, B, BC, C, CD, D, G.
[0063] In one embodiment, the breadth of neutralization is tested on an indicator virus panel comprising cross-clade HIV isolates. In one embodiment, the virus panel comprises the 9 cross- clade isolates of 92TH021, JRCSF, C22, 94UG103, 92BR020, Dul56.12, HIV-2, HIV-2C1, and HIV-2C4. In one embodiment, the virus panel comprises the 130 cross-clade isolates of NIH-
065 _ 191955 Al l, NIH-063_0330.v4.c3, NIH-066_191084 B7.19, NAC_BG505, NIH-
057_MS208.A1, NIH-064_0260.v5.cl, NAC_KNH1144, NAC_94UG103, NAC_92RW020, NIH-060_Q769ENVd22, NIH-058_Q23ENV17, NIH-056_Q168ENVa2, NIH-
061_Q259ENVd2.17, NIH-059_Q461ENVe2, NIH-117_T278.5, NIH-100_6041.v3.c23, NIH- 099_3301.vl .c24, NIH-102_6545.v4.cl, NIH-101_6540.v4.cl, NIH-103_0815.v3.c3, NIH- 104_3103.v3.cl0, NIH-073_C3347.cl 1, NIH-080_BJOX025000.01.1, NIH-075_CNE8,
NAC_92TH021 , NIH-081_BJOX028000.10.3, NIH-078_BJOX015000.11.5, NIH- 079_BJOX010000.06.2, NIH-074_C4118.c09, NIH-069_C1080.c03, NIH-
077_BJOX009000.02.4, NIH-068_620345.c01, NIH-076_CNE5, NIH-070_R2184.c04, NIH- 072_R3265.c06, NIH-071 R1166.c01, NIH-115_928.28, NIH-118_T255.34, NIH-116_T263.8, NIH- 108_T235.47, NIH-114_T257.31, NIH-107_T251.18, NIH-106_T250.4, NIH-119 T211.9, NAC MN, NAC_HxB2, NIH-012_1012.11.TC21.3257, NAC ADA, NAC WIT0.33, NIH- 006_AC10.0.29, NIH-003_SC422661.8, NAC_89.6, NAC_BaL.26, NAC_RHPA4.7, NIH- 014_6244.13.B5.4567, NAC_TRJ0.58, NAC_92BR020, NIH-008_WEAU.dl5.410.787,
NAC REJ0.67, NIH-009_1006.11.C3.1601, NAC_SS 1196.1, NAC_YU2, NAC JRCSF, NAC JRFL, NIH-004 PV0.4, NAC DH12, NIH-005_TRO.11, NAC_SF162, NIH-
011_1056.10.TA11.1826, NIH-016_SC05.8C 11.2344, NIH-013_6240.08.TA5.4622, NIH-
007_CAAN5342.A2, NIH-010_1054.07.TC4.1499, NIH-001_6535.3, NIH-
015_62357.14.D3.4589, NIH-002_QH0692 .42, NIH-050_CNE21, NIH-054_CNE53, NIH- 055_CNE58, NIH-048_CNE19, NIH-053_CNE52, NIH-051_CNE17, NIH-052_CNE30, NIH- 049_CNE20, NIH-030_HIV-0013095.2.11, NIH-026_ZM135M.PL10a, NIH-017_Dul56.12, NIH-019_Du422.1 , NIH-018_Dul72.17, NIH-032_HIV-16845.2.22, NIH-039_Ce2060 G9, NAC_93IN905, NIH-029_HIV-001428.2.42, NIH-041 BF 1266.43 la, NIH-023_ZM249M.PL1, NIH-031 HIV-16055.2.3, NIH-043 ZM249M.B 10, NIH-025 ZM109F.PB4, NIH-
040_Ce703010054 2A2, NIH-036_Ce2010 F5, NIH-020_ZM197M.PB7, NIH-044_ZM247F.F7, NIH-046_1394C9Gl(Rev.), NIH-047_Ce704809221 1B3, NAC IAVIC22, NIH-034_Ce0393 C3, NIH-045_7030102001E5(Rev.), NIH-021_ZM214M.PL15, NIH-024_ZM53M.PB12, NIH-
035_Ce l 176 A3, NIH-022 ZM233M.PB6, NIH-027 CAP45.G3, NIH-042 ZM246F.D5, NHI- 028_CAP210.E8, NIH-038_Cel 172 HI, NIH-095_6480.v4.c25, NIH-096_6952.vl .c20, NIH- 097_6811.v7.cl8, NIH-094_3817.v2.c59, NIH-098_89.F1 2 25, NIH-089_3016.v5.c45, NIH- 090_A07412M 1.vrc 12, NIH-091_231965 01, NIH-086_X2131 Cl B5, NIH-082_X1193 cl, NIH-084_X1254 c3, NIH-083_P0402 c2 11, NIH-087_P1981 C5 3, NIH-088_X1632 S2 B IO, and NIH-085_X2088 c9. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least 4, 5, 6, 7, 8 or 9 of the cross-clade HIV isolates in the 9-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least 6 of the cross- clade HIV isolates in the 9-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 50%, 60%, 70%, 80%, 90%, 95%, or 100% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 60% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 70% of cross-clade HIV isolates in the 130- member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 75% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 80% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 90% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 95% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 98% of cross-clade HIV isolates in the 130-member indicator virus panel. In one embodiment, a broadly neutralizing antibody is capable of neutralizing at least about 100% of cross-clade HIV isolates in the 130-member indicator virus panel.
[0064] The phrase "substantially similar," or "substantially the same", as used herein, denotes a sufficiently high degree of similarity between two numeric values (generally one associated with an antibody described herein and the other associated with a reference/comparator antibody) such that one of skill in the art would consider the difference between the two values to be of little or no biological and/or statistical significance within the context of the biological characteristic measured by said values (e.g., KD values). The difference between said two values can be less than about 50%, less than about 40%, less than about 30%, less than about 20%, or less than about 10% as a function of the value for the reference/comparator antibody.
[0065] A polypeptide, antibody, polynucleotide, vector, cell, or composition, which is "isolated" is a polypeptide, antibody, polynucleotide, vector, cell, or composition, which is in a form not found in nature. Isolated polypeptides, antibodies, polynucleotides, vectors, cell or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature. In some embodiments, an antibody, polynucleotide, vector, cell, or composition, which is isolated is substantially pure.
[0066] As used herein, "substantially pure" refers to material, which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.
[0067] The terms "polypeptide," "peptide," and "protein" are used interchangeably herein to refer to polymers of amino acids of any length. The polymer can be linear or branched, it can comprise modified amino acids, and it can be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. It is understood that, because the polypeptides described herein are based upon antibodies, in certain embodiments, the polypeptides can occur as single chains or associated chains.
[0068] The terms "identical" or percent "identity" in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity can be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software are known in the art that can be used to obtain alignments of amino acid or nucleotide sequences. One such non-limiting example of a sequence alignment algorithm is the algorithm described in Karlin S., et al, Proc. Natl. Acad. Sci., 87:2264-2268 (1990), as modified in Karlin S., et al, Proc. Natl. Acad. Sci., 90:5873-5877 (1993), and incorporated into the NBLAST and XBLAST programs (Altschul SF, et al, Nucleic Acids Res., 25:3389-3402 (1991)). In certain embodiments, Gapped BLAST can be used as described in Altschul SF, et al., Nucleic Acids Res. 25:3389-3402 (1997). BLAST-2, WU-BLAST-2 (Altschul SF, et al., Methods in Enzymology, 266:460-480 (1996)), ALIGN, ALIGN-2 (Genentech, South San Francisco, California) or Megalign (DNASTAR) are additional publicly available software programs that can be used to align sequences. In certain embodiments, the percent identity between two nucleotide sequences is determined using the GAP program in GCG software (e.g., using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 90 and a length weight of 1, 2, 3, 4, 5, or 6). In certain alternative embodiments, the GAP program in the GCG software package, which incorporates the algorithm ofNeedleman and Wunsch (J. Mol. Biol. (48):444-453 (1970)) can be used to determine the percent identity between two amino acid sequences (e.g., using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5). Alternatively, in certain embodiments, the percent identity between nucleotide or amino acid sequences is determined using the algorithm of Myers and Miller (CABIOS, 4: 11-17 (1989)). For example, the percent identity can be determined using the ALIGN program (version 2.0) and using a PAM120 with residue table, a gap length penalty of 12 and a gap penalty of 4. Appropriate parameters for maximal alignment by particular alignment software can be determined by one skilled in the art. In certain embodiments, the default parameters of the alignment software are used. In certain embodiments, the percentage identity "X" of a first amino acid sequence to a second sequence amino acid is calculated as 100 x (Y/Z), where Y is the number of amino acid residues scored as identical matches in the alignment of the first and second sequences (as aligned by visual inspection or a particular sequence alignment program) and Z is the total number of residues in the second sequence. If the length of a first sequence is longer than the second sequence, the percent identity of the first sequence to the second sequence will be higher than the percent identity of the second sequence to the first sequence.
[0069] As a non-limiting example, whether any particular polynucleotide has a certain percentage sequence identity (e.g., is at least 80% identical, at least 85% identical, at least 90% identical, and in some embodiments, at least 95%, 96%, 97%, 98%, or 99% identical) to a reference sequence can, in certain embodiments, be determined using the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711). Bestfit uses the local homology algorithm of Smith and Waterman (Advances in Applied Mathematics 2: 482 489 (1981)) to find the best segment of homology between two sequences. When using Bestfit or any other sequence alignment program to determine whether a particular sequence is, for instance, 95% identical to a reference sequence described herein, the parameters are set such that the percentage of identity is calculated over the full length of the reference nucleotide sequence and that gaps in identity of up to 5% of the total number of nucleotides in the reference sequence are allowed.
[0070] In some embodiments, two nucleic acids or polypeptides described herein are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection. Identity can exist over a region of the sequences that is at least about 10, about 20, about 40-60 residues in length or any integral value there between, and can be over a longer region than 60-80 residues, for example, at least about 90-100 residues, and in some embodiments, the sequences are substantially identical over the full length of the sequences being compared, such as the coding region of a nucleotide sequence for example. [0071] A "conservative amino acid substitution" is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). For example, substitution of a phenylalanine for a tyrosine is a conservative substitution. In some embodiments, conservative substitutions in the sequences of the polypeptides and antibodies described herein do not abrogate the binding of the polypeptide or antibody containing the amino acid sequence, to the antigen(s). Methods of identifying nucleotide and amino acid conservative substitutions, which do not eliminate antigen binding are well-known in the art (see, e.g., Brummell DA, et al., Biochem. 32: 1180-1 187 (1993); Kobayashi et al., Protein Eng. 12(10):879-884 (1999); and Burks EA, et ak, Proc. Natl. Acad. Sci. USA 94: 412-417 (1997)).
[0072] As used herein, the terms "treatment" or "therapy" (as well as different forms thereof, including curative or palliative) refer to treatment of an infected person. As used herein, the term "treating" includes alleviating or reducing at least one adverse or negative effect or symptom of a condition, disease or disorder. This condition, disease or disorder can be HIV infection.
[0073] Terms such as "treating" or "treatment" or "to treat" or "alleviating" or "to alleviate" refer to therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder, such as HIV or AIDS. Thus, those in need of treatment include those already diagnosed with or suspected of having the disorder. In certain embodiments, a subject is successfully "treated" for the disorder according to the methods described herein if the patient shows one or more of the following: a reduction in the number of or complete absence of viral load; a reduction in the viral burden; inhibition of or an absence of the virus into peripheral organs; relief of one or more symptoms associated with the disorder; reduced morbidity and mortality; improvement in quality of life; increased progression-free survival (PFS), disease-free survival (DFS), or overall survival (OS), complete response (CR), partial response (PR), stable disease (SD), a decrease in progressive disease (PD), a reduced time to progression (TTP), or any combination thereof.
[0074] As used herein, the terms "prevention" or "prophylaxis" refer to preventing a subject from becoming infected with, or reducing the risk of a subject from becoming infected with, or halting transmission of, or the reducing the risk of transmission of a virus. Prophylactic or preventative measures refer to measures that prevent and/or slow the development of a targeted pathological condition or disorder. Thus, those in need of prophylactic or preventative measures include those prone to have the disorder and those in whom the disorder is to be prevented. In one embodiment, prevention encompasses passive immunization of a subject in need thereof comprising administering an effective amount of an antibody described herein.
[0075] As employed above and throughout the disclosure the term "effective amount" refers to an amount effective, at dosages, and for periods of time necessary, to achieve the desired result with respect to the treatment of the relevant disorder, condition, or side effect. An "effective amount" can be determined empirically and in a routine manner, in relation to the stated purpose. It will be appreciated that the effective amount of components of the present invention will vary from patient to patient not only with the particular vaccine, component or composition selected, the route of administration, and the ability of the components to elicit a desired result in the individual, but also with factors such as the disease state or severity of the condition to be alleviated, hormone levels, age, sex, weight of the individual, the state of being of the patient, and the severity of the pathological condition being treated, concurrent medication or special diets then being followed by the particular patient, and other factors, which those skilled in the art will recognize, with the appropriate dosage being at the discretion of the attending physician. Dosage regimes may be adjusted to provide the improved therapeutic response. An effective amount is also one in which any toxic or detrimental effects of the components are outweighed by the therapeutically beneficial effects.
[0076] The term "therapeutically effective amount" refers to an amount of an antibody, recombinant virus, immunoconjugate, or other drug effective to "treat" a disease or disorder in a subject or mammal. To the extent an antibody can prevent growth and/or kill existing cells, it can be cytostatic and/or cytotoxic. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
[0077] The terms "subject," "individual," and "patient" are used interchangeably herein, and refer to an animal, for example a human, to whom treatment, including prophylactic treatment, with the antibody or pharmaceutical composition according to the present disclosure, is provided. In one embodiment, the subject, individual, or patient has been infected with HIV. In one embodiment, the subject, individual, or patient suffers from AIDS. In one embodiment, the subject, individual, or patient has been exposed to HIV. In one embodiment, the subject, individual, or patient is at risk of being exposed to HIV.
[0078] Administration "in combination with" one or more further therapeutic agents includes simultaneous (concurrent) or consecutive administration in any order.
[0079] The terms "pharmaceutically composition," "pharmaceutical formulation," "pharmaceutically acceptable formulation," or "pharmaceutically acceptable composition" all of which are used interchangeably, refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem complications commensurate with a reasonable benefit/risk ratio. "Pharmaceutically acceptable" or "pharmaceutical formulation" refers to a preparation, which is in such form as to permit the biological activity of the active ingredient to be effective, and which contains no additional components, which are unacceptably toxic to a subject to which the formulation would be administered. The formulation can be sterile.
[0080] The term "antiretroviral therapy" or "ART," as used herein, refers to any of the therapies used to manage progression of a retrovirus (e.g., HIV) infection in a subject (e.g., a human), including, for example, nucleoside reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (Pis), fusion inhibitors, entry inhibitors, maturation inhibitors, cellular inhibitors, integrase strand transfer inhibitors, and multi-class combinations. Such drugs include, but are not limited to, lamivudine and zidovudine, emtricitabine (FTC), zidovudine (ZDV), azidothymidine (AZT), lamivudine (3TC), zalcitabine, dideoxycytidine (ddC), tenofovir disoproxil fumarate (TDF), didanosine (ddl), stavudine (d4T), abacavir sulfate (ABC), etravirine (ETR), delavirdine (DLV), efavirenz (EFV), nevirapine (NVP), amprenavir (APV), tipranavir (TPV), indinavir (IDV), saquinavir, saquinavir mesylate (SQV), lopinavir (LPV), ritonavir (RTV), fosamprenavir calcium (FOS-APV), ritonavir (RTV), darunavir (DRV), atazanavir sulfate (ATV), nelfmavir mesylate (NFV), enfuvirtide (T-20), maraviroc and raltegravir. ART drugs can also include antibodies that target HIV proteins or cellular proteins associated with disease progression. Also included are immune-based therapies, such as IL-2, IL-12, and alpha- epibromide. Each of these drugs can be administered alone or in combination with any other ART drug or any HIV-specific neutralizing antibody, such as a broadly neutralizing antibody, which is incorporated by reference herein in its entirety for all purposes. [0081] The term "reservoir activator," as used herein, refers to an agent capable of activating a viral reservoir (e.g., an HIV reservoir). In one embodiment, a reservoir activator comprises a histone deacytelase (HDAC) inhibitor (e.g., romidepsin, vorinostat, and panobinostat), immunologic activator (e.g., cytokines and TLR agonists), or a dedicated small molecule drug.
[0082] The term "immunomodulator," as used herein, refers to an agent, such as an antibody or peptide, which is capable of increasing, inducing, or extending an immune response (e.g., a cell- mediated immune response and/or a humoral immune response) when administered to a subject (e.g., a human, e.g., a human infected with HIV or at risk of an HIV infection or transmission). Immunomodulators include, but are not limited to immune checkpoint inhibitors, for example, a PD-1, PD-L1, LAG-3, or TIG IT antagonist. In one embodiment, an immunomodulator used in the methods described herein comprises an anti -PD-1 antibody, anti-PD-Ll antibody, anti-LAG3 antibody, or an anti-TIGIT antibody. An immunomodulator can be administered in conjunction with (e.g., prior to, concurrently with, or subsequent to, or within the context of a treatment regimen that includes the administration of a broadly neutralizing antibody described herein.
[0083] As used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the content clearly dictates otherwise. Thus, for example, reference to "a cell" includes a combination of two or more cells, and the like.
[0084] The term "and/or" as used in a phrase such as "A and/or B" herein is intended to include both "A and B," "A or B," "A," and "B." Likewise, the term "and/or" as used in a phrase such as "A, B, and/or C" is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
[0085] The term "about" as used herein when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of up to ±20% from the specified value, as such variations are appropriate to perform the disclosed methods. Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term "about." Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained by the present invention. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. [0086] Notwithstanding that the numerical ranges and parameters setting forth the broad scope described herein are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements.
[0087] It is understood that wherever embodiments are described herein with the language "comprising," otherwise analogous embodiments described in terms of "consisting of and/or "consisting essentially of' are also provided.
II. Polypeptides
[0088] In one aspect, provided herein are isolated polypeptides derived from an HIV Env polypeptide. In some embodiments, an isolated polypeptide described herein comprises a variant gpl60 polypeptide. In some embodiments, an isolated polypeptide described herein comprises a variant gpl40 polypeptide. In some embodiments, an isolated polypeptide described herein comprises a variant gp 120 polypeptide. It is understood that in some embodiments, a variant gp 160 or gpl40 polypeptide described herein comprises a variant gpl20 polypeptide described herein.
[0089] In some embodiments, an isolated polypeptide described herein is capable of binding to a BG18 inferred-germline (iGL) variant antibody or a BG18-like native antibody described herein. In some embodiments, the BG18 iGL is BG18 iGLO, BG18 iGLl, BG18 iGL2.
[0090] In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises one or more of
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPNLLSNMRGEL (SEQ ID NO: 72) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of TGDIIGDIRQAHCNVS (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDIIGDIRMAHCNVS (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[0091] In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPNLLSNMRGEL (SEQ ID NO: 72) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; (c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprise an amino acid sequence of TGDIIGDIRQAHCNVS (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDIIGDIRMAHCNVS (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[0092] In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises one or more of
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of CTNVTNNITDDMRGEL (SEQ ID NO: 71), CTNYAPNLLSNMRGEL (SEQ ID NO: 72), CTNYAPNLLSNMRGEI (SEQ ID NO: 73), CTNYAPKLLSNMRGEI (SEQ ID NO: 74), CTNY APKLRSMMRGEI (SEQ ID NO: 75), or CTNYAPNLRSDMRGEI (SEQ ID NO: 76);
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of TRPNNNT (SEQ ID NO: 77), or TRPSNNT (SEQ ID NO: 78);
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of TGDIIGDIRQAHCNVS (SEQ ID NO: 79), FGDIIGDIRMAHCNVS (SEQ ID NO: 80), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), or FGDVLGDVDMAKCTIS (SEQ ID NO: 83); and
(d) a b 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of SITLPCR (SEQ ID NO: 84), SIVLPCR (SEQ ID NO: 85), SLILPCR (SEQ ID NO: 86), or SLILPCW (SEQ ID NO: 87); wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[0093] In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of CTNVTNNITDDMRGEL (SEQ ID NO: 71), CTNYAPNLLSNMRGEL (SEQ ID NO: 72), CTNYAPNLLSNMRGEI (SEQ ID NO: 73), CTNYAPKLLSNMRGEI (SEQ ID NO: 74), CTNY APKLRSMMRGEI (SEQ ID NO: 75), or CTNYAPNLRSDMRGEI (SEQ ID NO: 76);
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of TRPNNNT (SEQ ID NO: 77), or TRPSNNT (SEQ ID NO: 78);
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of TGDIIGDIRQAHCNVS (SEQ ID NO: 79), FGDIIGDIRMAHCNV S (SEQ ID NO: 80), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), or FGDVLGDVDMAKCTIS (SEQ ID NO: 83); and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of SITLPCR (SEQ ID NO: 84), SIVLPCR (SEQ ID NO: 85), SLILPCR (SEQ ID NO: 86), or SLILPCW (SEQ ID NO: 87);
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[0094] In some embodiments, a variant HIV Env gp 120 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of
CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77),
FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77),
FGDVLGDVRMAHCNIS (SEQ ID NO: 81), and SLILPCR (SEQ ID NO: 86), respectively,
CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77),
FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78),
FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCW (SEQ ID NO: 87), respectively,
CTNYAPNLRSDMRGEI (SEQ ID NO: 76), TRPNNNT (SEQ ID NO: 77), FGDVLGDVDMAKCTIS (SEQ ID NO: 83), and SLILPCR (SEQ ID NO: 86), respectively,
CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively, or
CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[0095] In some embodiments, a variant HIV Env gp 120 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77),
FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1). In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77),
FGDVLGDVRMAHCNIS (SEQ ID NO: 81), and SLILPCR (SEQ ID NO: 86), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the b 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCW (SEQ ID NO: 87), respectively, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
[0096] In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises N332.
[0097] In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env positions 1-35. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env positions 1-10. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env positions 10-20. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env positions 20-35. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of one of HIV Env position 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, and 35. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N- terminal residue of HIV Env position 30. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of HIV Env position 31. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises an N-terminal residue of HIV Env position 32. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[0098] In some embodiments, a variant HIV Env gp 120 polypeptide described herein comprises a C-terminal residue of one of HIV Env positions 503-512. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a C-terminal residue of one of HIV Env positions 503-508. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a C-terminal residue of one of HIV Env positions 508-512. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises a C-terminal residue of HIV Env position 512. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[0099] In some embodiments, a variant HIV Env gpl20 polypeptide described herein is a variant of the BG505, BG505 N332, BG505 MD39, BG505 MD64, BG505 MD39 N332, BG505 MD39 1 ImutB, or BG505 MD39 17mutE gpl20 polypeptide. In some embodiments, a variant HIV Env gpl20 polypeptide described herein is a variant of the BG505 MD39 N332 polypeptide. In some embodiments, a variant HIV Env gpl20 polypeptide described herein is a variant of the BG505 MD64 N332 polypeptide.
[00100] In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137 and H325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137 and P325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137, M141 and H325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137, R139, M141, and H325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137 and P325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137, M141, and P325. In some embodiments, a variant HIV Env gpl20 polypeptide described herein comprises K137, R139, M141, and P325. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[00101] In some embodiments, an isolated polypeptide described herein comprises
an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT1,
an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2,
an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT3,
an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT4,
an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT5, or
an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2-KO. [00102] In some embodiments, an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT1. In some embodiments, an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT2. In some embodiments, an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT3. In some embodiments, an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT4. In some embodiments, an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332- GT5. In some embodiments, an isolated polypeptide described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD64 N332- GT2.
[00103] In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT1, BG505 MD39 N332-GT2, BG505 MD39 N332-GT3, BG505 MD39 N332-GT4, BG505 MD39 N332-GT5, or BG505 MD39 N332-GT2-KO. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT1. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT2. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT3. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT4. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT5.
[00104] In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT1. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT2. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332- GT3. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT4. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT5. In some embodiments, an isolated polypeptide described herein comprises the amino acid sequence of BG505 MD64 N332- GT2. In some embodiments, an isolated polypeptide described herein comprises K137 and H325. In some embodiments, an isolated polypeptide described herein comprises K137 and P325. In some embodiments, an isolated polypeptide described herein comprises K137, M141 and H325. In some embodiments, an isolated polypeptide described herein comprises K137, R139, M141, and H325. In some embodiments, an isolated polypeptide described herein comprises K137 and P325. In some embodiments, an isolated polypeptide described herein comprises K137, M141, and P325. In some embodiments, an isolated polypeptide described herein comprises K137, R139, M141, and P325. In some embodiments, an isolated polypeptide described herein comprises N332. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[00105] In some embodiments, an isolated polypeptide described herein specifically binds to the BG18 antibody. In some embodiments, an isolated polypeptide described herein specifically binds to an antibody selected from the group consisting of BG18.11 and BG18.6. In some embodiments, an isolated polypeptide described herein specifically binds to an antibody selected from the group consisting of BG18 iGLO, BG18 iGLl, and BG18 iGL2. In some embodiments, an isolated polypeptide described herein specifically binds to BG18 iGLO. In some embodiments, an isolated polypeptide described herein specifically binds to BG18 iGLl . In some embodiments, an isolated polypeptide described herein specifically binds to BG18 iGL2. In some embodiments, an isolated polypeptide described herein specifically binds to an antibody selected from the group consisting of PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, and PRE15. In some embodiments, an isolated polypeptide described herein specifically binds to the PG121 iGL antibody.
[00106] In some embodiments, an isolated polypeptide described herein specifically binds to an antibody comprising the VH and VL of
VH4-59 and BG18 iGL, respectively;
VH1-69 and BG18 iGL, respectively;
VH5-51 and BG18 iGL, respectively;
VH3-33 and BG18 iGL, respectively; VH3-23 and BG18 iGL, respectively;
BG18 iGLl and VL3-19, respectively;
BG18 iGLl and VL3-10, respectively;
BG18 iGLl and VL3-1, respectively;
BG18 iGLl and VL3-21, respectively; or
BG18 iGLl and VL2-8, respectively.
[00107] The affinity of an isolated polypeptide disclosed for an antibody described herein (e.g., BG18 iGLO) can be determined experimentally using any suitable method well-known in the art, e.g., flow cytometry, enzyme-linked immunoabsorbent assay (ELISA), biolayer interferometry (BLI) assay, radioimmunoassay (RIA), or kinetics (e.g., BIACORE™ analysis). Direct binding assays as well as competitive binding assay formats can be readily employed. (See, for example, Berzofsky, et ak, "Antibody-Antigen Interactions," In Fundamental Immunology, Paul, W. E., Ed., Raven Press: New York, N.Y. (1984); Kuby, Janis Immunology, W. H. Freeman and Company: New York, N.Y. (1992); and methods described herein. The measured affinity of a particular antibody-antigen interaction can vary if measured under different conditions (e.g., salt concentration, pH, temperature). Thus, measurements of affinity and other antigen-binding parameters (e.g., KD or Kd, Kon, K0h) are made with standardized solutions of antibody and antigen, and a standardized buffer, as known in the art and such as the buffer described herein.
[00108] In some embodiments, an isolated polypeptide described herein comprises a gpl40. In some embodiments, an isolated polypeptide described herein comprises a gpl20.
[00109] In one aspect, provided herein are recombinant HIV Env trimers comprising an isolated polypeptide described herein. In some embodiments, a recombinant HIV Env trimer described herein is a homotrimer. In some embodiments, a recombinant HIV Env trimer described herein a heterotrimer. In some embodiments, a recombinant HIV Env trimer described herein comprises gpl20-gp41 heterodimers. In some embodiments, a recombinant HIV Env trimer described herein comprises gpl20-gp41 heterodimers wherein the heterodimers are covalently linked. In some embodiments, a recombinant HIV Env trimer described herein comprises gpl20-gp41 fusions. In some embodiments, a recombinant HIV Env trimer described herein is a stabilized trimer. In some embodiments, a recombinant HIV Env trimer described herein is an SOSIP, NFL or UFO trimer. In some embodiments, a recombinant HIV Env trimer described herein is an SOSIP trimer. In some embodiments, a recombinant HIV Env trimer described herein comprises a variant gpl20 polypeptide described herein. [00110] In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence ofBG505 MD39 N332-GT1, BG505 MD39 N332-GT2, BG505 MD39 N332- GT3, BG505 MD39 N332-GT4, BG505 MD39 N332-GT5, or BG505 MD39 N332-GT2-KO. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT1. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT2. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT3. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT4. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT5. In some embodiments, a recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD64 N332-GT2. In some embodiments, a recombinant HIV Env trimer described herein comprises K137 and H325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137 and P325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137, M141 and H325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137, R139, M141, and H325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137 and P325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137, M141, and P325. In some embodiments, a recombinant HIV Env trimer described herein comprises K137, R139, M141, and P325. HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1).
[00111] Many Env-based trimeric candidate immunogens are engineered to eliminate cleavage between gpl20 and gp41 (so called uncleaved gpl40 trimers), usually generating imperfect mimetics of the functional spike based on antigenic profding or EM analysis (Tran K, et al. (2014) Proc Natl Acad Sci US All l(7):E738-747; Ringe RP, et al. (2013) Proc Natl Acad Sci U S A 110(45): 18256-18261). As agroup, the defined, or presumed to be, disordered trimers (in adjuvant) generate high self-binding antibody titers. However, these vaccine-elicited antibodies do not efficiently neutralize most HIV -I primary isolates, that is, strains representative of those circulating in the human population (Sundling C, et al. (2012) Science translational medicine 4(142): 142ral96; Chakrabarti BK, et al. (2013) J Virol 87(24): 13239- 13251; Kovacs JM, et al. (2012) Proc Natl Acad Sci US A 109(30): 12111-12116; Nkolola JP, et al. (2014) Comparison of multiple adjuvants on the stability and immunogenicity of a clade C HIV-I gpl40 trimer. Vaccine 32(18):2109-2116). Antibodies elicited by these immunogens target epitopes exposed only on the free gpl20 and trimeric post-fusion forms of gp41 or disordered gpl40s and thus are ineffective at accessing their epitopes buried within the ordered, quaternary structure achieved in the native Env spike. Applicants recently described the limitations of two CD4bs-directed non-bnAbs, (GE148 and GE136) generated following immunization of uncleaved gpl40 trimers (YU2 gpl40-foldon) in non-human primates (NHP). Non- bnAbs, represented by GE136 and 148, can only neutralize the sensitive so-called "tier I viruses" that are not representative of the more neutralization resistant tier 2-like primary isolates circulating in the human population. Using crystallography, EM reconstructions, paratope scanning and molecular modeling, Applicants determined that these vaccine-elicited antibodies fail to reach the CD4bs due to steric barriers imposed by quaternary packing of the native Env on neutralization resistant primary isolates, a property that Applicants use to Applicants' advantage in the negative -selection strategy presented here (Tran K, et al. (2014) Proc Natl Acad Sci US A 11 l(7):E738-747).
[00112] The cumulative historical data have led to the hypothesis that a more faithful mimic of the HIV-I spike that better recapitulates the native, pre-fusion form of Env, selectively displaying neutralizing determinants while occluding non-neutralizing determinants, may better elicit antibodies capable of accessing the native spike. A soluble Env mimetic, containing a disulfide linkage between gpl20 and gp41 (SOS), first described in the 2000s, and further developed over the next decade, displays many of these properties, culminating in the determination of the high resolution structures of the well-ordered BG505 SOSIP trimers by crystallography and EM (Lyumkis D, et al. (2013) Science 342(6165): 1484-1490; Julien JP, et al. (2013) Science 342(6165): 1477-1483; Sanders RW, et al. (2013) PLoS Pathog 9(9):el003618; Depetris RS, et al. (2012) J Biol Chem 287(29):24239-24254). A sub-nanometer EM reconstruction of KNHI 144 SOSIP is also available but does not provide atomic level details (Bartesaghi A, Merk A, Borgnia MJ, Milne JL, & Subramaniam S (2013) Nat Struct Mol Biol 20(12): 1352-1357). The BG505 SOSIP and KNHI 144 SOSIP trimers are derived from the Env sequences of the subtype A BG505 and KNHI 144 strains. These soluble trimers possess an engineered disulfide linkage between the gpl20 and gp41 (at residues 501C and 605C, respectively) and an additional mutation in the heptad repeat I (HRI) of gp41 (1559P) that facilitates trimerization (Binley JM, et al. (2000) J Virol 74(2):627-643; Sanders RW, et al. (2002) J Virol 76(17):8875-8889). A truncation of the membrane proximal external region (MPER) at residue 664 enhances expression while decreasing aggregation is incorporated into the so-called BG505 SOSIP.664 trimers (Sanders RW, et al. (2013) PLoS Pathog 9(9):el003618; Sanders RW, et al. (2002) J Virol 76(17):8875-8889). Although SOSIP molecules based on other HIV-I primary strains were attempted over the past decade, the BG505- and KNHI 144-derived SOSIP trimers are the two limited examples of SOSIPs that yield homogeneous trimers suitable for high resolution biophysical and structural analysis. The structural explanation for the difficulty to readily transfer the SOSIP design to other HIV-I strain-derived sequences is not yet fully understood and would be valuable information to broaden the trimer design horizon.
[00113] Since the initial soluble native-like BG505 SOPIP.664 Env trimer was confirmed to adopt a near-native conformation by high-resolution structural analysis, multiple efforts to produce stable, soluble Env mimetics derived from multiple HIV-1 strains were pursued. Multiple solutions to this objective include the improved cleavage-independent NFL trimers, UFOs and modified SOSIPs. Both the SOSIP and NFL well-ordered trimers are efficiently recognized by broadly neutralizing antibodies (bnAbs) which arise sporadically during the course of natural infection. In some cases, including the important advances described here, have been used to isolate such bnAbs. One approach to elicit tier 2 neutralizing Abs has been to immunize the existing well-ordered trimers using prime:boosting in selected animal models. For BG505 and 16055 native-like trimers this approach does elicit tier 2 neutralizing antibodies, but of limited cross-reactive breadth.
[00114] Most cross-conserved sites on the HIV Env spike are occluded by evolved, incorporated self-N-glycans, limiting naive B cell recognition of the underlying polypeptide surface. The exceptions are the protein surfaces of the primary receptor CD4 binding site (CD4bs) and the ftirin cleavage site (proximal to the gpl20:41 interface). Infrequently, during the course of the natural HIV infection process, bnAbs are elicited to these aforementioned sites of vulnerability. In addition, other bnAbs directed to the V2 apex, the 332N-glycan supersite and to the fusion peptide or the high-mannose patch are elicited during the course of chronic HIV infection.
[00115] After decades of development, advances in soluble HIV-1 Env mimics design permits the generation of a diverse array of native-like trimers (Ward and Wilson, 2017. The HIV-1 envelope glycoprotein structure: nailing down a moving target. Immunol Rev 275:21-32; Karlssonet ah, 2017. Evolution of B cell analysis and Env trimer redesign. Immunol Rev 275: 183-202). The successful development of the soluble SOSIP trimers provided proof-of-principle (Sanders et al, 2013. A next-generation cleaved, soluble HIV-1 Env trimer, BG505 SOSIP.664 gpl40, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS Pathog 9:el003618) forming a prefusion native-like conformation (Lyumkis et al., 2013. Cryo-EM structure of a fully glycosylated soluble cleaved HIV-1 envelope trimer. Science 342: 1484-1490; Julien et al, 2013. Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science 342: 1477- 1483; Garces et al., 2015. Affinity Maturation of a Potent Family of HIV Antibodies Is Primarily Focused on Accommodating or Avoiding Glycans. Immunity 43: 1053-1063; Pancera et al., 2014. Structure and immune recognition of trimeric pre-fusion HIV-1 Env. Nature 514:455-461). The SOSIP gpl40 trimer is proteolytically cleaved by cellular furins to gpl20 and gp41 subunits and covalently linked by an engineered intra-protomer disulfide bond A501C-T605C (SOS). These trimers also require mutation (I559P) in the gp41 heptad repeat 1 (HR1) to maintain well-ordered oligomers, as well as expression of exogenous ftirin for full conformational integrity (Sanders et al., 2013. A next-generation cleaved, soluble HIV-1 Env trimer, BG505 SOSIP.664 gpl40, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS Pathog 9:el003618; Guenaga et al., 2015. Well-Ordered Trimeric HIV-1 Subtype B and C Soluble Spike Mimetics Generated by Negative Selection Display Native-like Properties. PLoS Pathog l l :el004570; Julien et al., 2015. Design and structure of two HIV-1 clade C SOSIP.664 trimers that increase the arsenal of native-like Env immunogens. Proc Natl Acad Sci U S A 112: 11947- 11952; de Taeye et al. 2015. Immunogenicity of Stabilized HIV-1 Envelope Trimers with Reduced Exposure of Non-neutralizing Epitopes. Cell 163: 1702-1715; Pugach et al. 2015. A native-like SOSIP.664 trimer based on an HIV-1 subtype B env gene. J Virol 89:3380-3395; Ringe et al. 2013. Cleavage strongly influences whether soluble HIV-1 envelope glycoprotein trimers adopt a native like conformation. Proc Natl Acad Sci U S A 110: 18256-18261; Ringe et al. 2015. Influences on the Design and Purification of Soluble, Recombinant Native-Like HIV-1 Envelope Glycoprotein Trimers. J Virol 89: 12189-12210; Ringe et al. 2017. Reducing V3 Antigenicity and Immunogenicity on Soluble, Native-Like HIV-1 Env SOSIP Trimers. J Virol 91; Ahmedet al. 2017. Stabilization of a soluble, native-like trimeric form of an efficiently cleaved Indian HIV-1 clade C envelope glycoprotein. J Biol Chem 292:8236-8243; Sanders et al. 2002. Stabilization of the soluble, cleaved, trimeric form of the envelope glycoprotein complex of human immunodeficiency virus type 1. J Virol 76:8875-8889; Binley et al. 2000. A recombinant human immunodeficiency virus type 1 envelope glycoprotein complex stabilized by an intermolecular disulfide bond between the gpl20 and gp41 subunits is an antigenic mimic of the trimeric virion-associated structure. J Virol 74:627-643). In the past years, Applicants developed an improved native-like trimer design, generating well-ordered soluble Env mimics that are fully cleavage-independent, termed native flexibly linked (NFL) trimers. This design uses a flexible linker (two copies of Gly4-Ser,“G4S”) to replace the natural cleavage site and sequence (Sharma et al. 2015. Cleavage -independent HIV- 1 Env trimers engineered as soluble native spike mimetics for vaccine design. Cell Rep 11 :539- 550). The flexible linker between the natural C-terminus of gpl20 and N-terminus of gp41, allows the un-cleaved trimers to achieve a native-like conformation without the need of ftirin for precursor processing. However, the original NFL trimer design contains the I559P mutation (Sharma et al. 2015. Cleavage-independent HIV-1 Env trimers engineered as soluble native spike mimetics for vaccine design. Cell Rep 11 :539-550) that was initially identified in the SOSIP context to disfavor the post fusion state (Sanders et al. 2013. A next-generation cleaved, soluble HIV-1 Env trimer, BG505 SOSIP.664 gpl40, expresses multiple epitopes for broadly neutralizing but not non- neutralizing antibodies. PLoS Pathog 9:el003618). Both the original SOSIP and NFL designs do not form a high percentage of well-ordered trimers in all Env contexts. In the original NFL design, it is relatively inefficient in generating high yields of trimers derived from clade C strains, such as 16055 (Guenaga et al. 2015. Structure-Guided Redesign Increases the Propensity of HIV Env To Generate Highly Stable Soluble Trimers. J Virol 90:2806-2817). To improve trimer design, Applicants incorporated residues from BG505 (called trimer-derived (TD) residues) into 16055 NFLs, substantially improving the propensity to form native-like trimers (Guenaga et al. 2015. Structure-Guided Redesign Increases the Propensity of HIV Env To Generate Highly Stable Soluble Trimers. J Virol 90:2806-2817) and the elicitation of tier 2 clade C neutralizing antibodies (Martinez-Murillo et al, GB. 2017. Particulate Array of Well-Ordered HIV Clade C Env Trimers Elicits Neutralizing Antibodies that Display a Unique V2 Cap Approach. Immunity 46:804-817 e807; Dubrovskaya et al. 2017. Targeted N-glycan deletion at the receptor-binding site retains HIV Env NFL trimer integrity and accelerates the elicited antibody response. PLoS Pathog 13:el006614). Further improvements on the TD design by targeted glycine substitutions at helix- to-coil transitions that disfavor the post-fusion state of Env (TD CC+, namely“TD+”), significantly improve trimer homogeneity, yield, stability and antigenicity, resulting in the first high-resolution clade C Env structure (Guenaga et al. 2017. Glycine Substitution at Helix-to-Coil Transitions Facilitates the Structural Determination of a Stabilized Subtype C HIV Envelope Glycoprotein. Immunity 46:792-803 e793).
[00116] In one aspect, provided herein are nanoparticles comprising an isolated polypeptide described herein. In some embodiments, a nanoparticle described herein is a ferritin nanoparticle, an encapsulin nanoparticle, a Sulfur Oxygenase Reductase (SOR) nanoparticle, a lumazine synthase nanoparticle or a pyruvate dehydrogenase nanoparticle. In some embodiments, a nanoparticle described herein is a ferritin nanoparticle. In some embodiments, a nanoparticle described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5. In some embodiments, a nanoparticle described herein comprises an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of N332-GT1, N332-GT2, N332-GT3, N332-GT4, orN332-GT5. In some embodiments, a nanoparticle described herein comprises N332-GT1, N332- GT2, N332-GT3, N332-GT4, orN332-GT5. In some embodiments, a nanoparticle described herein comprises N332-GT2. In some embodiments, a nanoparticle described herein comprises N332- GT5. In some embodiments, a nanoparticle described herein has at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO: 13 or 15. In some embodiments, a nanoparticle described herein is a ferritin nanoparticle comprising the amino acid sequence of SEQ ID NO. 13 or 15.
[00117] In one aspect, provided herein are virus-like particles (VLPs) comprising an isolated polypeptide described herein.
[00118] It should be understood that in some embodiments, the polypeptides (e.g., Env trimers, nanoparticles, and other antigens) described herein can differ from the exact sequences illustrated and described herein. Thus, the invention contemplates deletions, additions and substitutions to the sequences shown, so long as the sequences function in accordance with the methods described herein. In this regard, in some embodiments, substitutions are conservative in nature, i.e., those substitutions that take place within a family of amino acids. For example, amino acids are generally divided into four families: (1) acidic— aspartate and glutamate; (2) basic— lysine, arginine, histidine; (3) non-polar— alanine, valine, leucine, isoleucine, praline, phenylalanine, methionine, tryptophan; and (4) uncharged polar— glycine, asparagine, glutamine, cysteine, serine threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine are sometimes classified as aromatic amino acids. It is reasonably predictable that an isolated replacement of leucine with isoleucine or valine, or vice versa; an aspartate with a glutamate or vice versa; a threonine with a serine or vice versa; or a similar conservative replacement of an amino acid with a structurally related amino acid, will not have a major effect on the biological activity. Proteins having substantially the same amino acid sequence as the sequences illustrated and described but possessing minor amino acid substitutions that do not substantially affect the immunogenicity of the protein are, therefore, within the scope of the invention III. Antibodies
[00119] As disclosed and exemplified herein, germline reverted antibodies can be designed by comparing the heavy and light chains of an isolated bnAb (e.g., BG18) that binds to an epitope of interest to nucleotide sequences and/or the amino acid sequences potentially encoded by germline
V gene segments. For example, one can obtain a germline reverted antibody starting from the amino acid sequence of an antibody directed to an antigen or epitope of choice and identifying germline encoded antibody segments (i.e., heavy chain V, D, and J gene segments and light chain
V and J gene segments) close in sequence to the starting antibody. Germline variable genes are cataloged, for example at VBASE2 (Retter I, Althaus HH, Miinch R, Miiller W: VBASE2, an integrative V gene database. Nucleic Acids Res. 2005 Jan l;33(Database issue):D671-4.). Starting from a known or preselected antibody and moving to such a database can be advantageous as the paired heavy and light chains will be compatible and likely to be well represented in the repertoire of expressed germline genes.
[00120] Germline antibody genes or inferred germline genes can be identified by analyzing
NGS dataset obtained from HIV-uninfected human subjects, as discussed in the Examples below. Germline antibody genes or inferred germline genes that bind to an epitope of interest can also be identified by screening libraries of human heavy and light chain genes, for example by phage display. Another method is by immunization and preparation of monoclonal antibodies from a mouse that expresses human antibody genes. Preferably hybridomas are prepared before there is substantial somatic mutation and selection. It can be advantageous to select germline antibody gene segments that are well represented in the repertoire of expressed human antibodies, also taking into account compatibility of various heavy chain - light chain pairings. In certain embodiments, it is advantageous to select V-D, D-J, V-D-J (heavy chain) or V-J (light chain) combinations that are well represented.
[00121] In one aspect, provided herein are germline reverted and inferred-germline (iGL) variant heavy and light chain variable domains, including heavy and light chain CDR regions of BG 18 that bind to an HIV Env polypeptide disclosed herein (e.g., N332-GT1, N332-GT2, N332-GT3, N332- GT4, or N332-GT5).
[00122] Germline reverted and inferred-germline antibodies disclosed herein are suitable for the selection of candidate vaccine molecules according to methods disclosed herein. [00123] In one aspect, provided herein are isolated antibodies comprising the VHCDR1, VH CDR2, and VH CDR3 of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1-69, VH5-51, VH3-33, or VH3-23; and the VL CDR1, VL CDR2, and VL CDR3 of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VL3-10, VL3- 19, VL3-1, VL3-21, or VL2-8. In some embodiments, an isolated antibody described herein comprises the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3 of BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15. In some embodiments, an isolated antibody described herein comprises the 3 VH CDRs and 3 VL CDRs of
VH4-59 and BG18 iGL, respectively;
VH1-69 and BG18 iGL, respectively;
VH5-51 and BG18 iGL, respectively;
VH3-33 and BG18 iGL, respectively;
VH3-23 and BG18 iGL, respectively;
BG18 iGLl and VL3-19, respectively;
BG18 iGLl and VL3-10, respectively;
BG18 iGLl and VL3-1, respectively;
BG18 iGLl and VL3-21, respectively; or
BG18 iGLl and VL2-8, respectively.
In some embodiments, the CDR sequences are identified according to Rabat.
[00124] In some embodiments, an isolated antibody described herein comprises the VH of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1-69, VH5-51, VH3-33, or VH3-23; and the VL of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VL3-10, VL3-19, VL3-1, VL3-21, or VL2-8. In some embodiments, an isolated antibody described herein comprises the VH and VL of BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15. In some embodiments, an isolated antibody described herein comprises the VH and VL of VH4-59 and BG18 iGL, respectively;
VH1-69 and BG18 iGL, respectively;
VH5-51 and BG18 iGL, respectively;
VH3-33 and BG18 iGL, respectively;
VH3-23 and BG18 iGL, respectively;
BG18 iGLl and VL3-19, respectively;
BG18 iGLl and VL3-10, respectively;
BG18 iGLl and VL3-1, respectively;
BG18 iGLl and VL3-21, respectively; or
BG18 iGLl and VL2-8, respectively.
IV. Polynucleotides
[00125] In one aspect, provided herein are isolated polynucleotides encoding an isolated polypeptide described herein, a variant HIV Env gpl20 polypeptide described herein, a recombinant HIV Env trimer described herein, a VLP described herein, or a nanoparticle described herein. In some embodiments, an isolated polynucleotide described herein is a DNA. In some embodiments, an isolated polynucleotide described herein is an mRNA. In some embodiments, an isolated polynucleotide described herein is an mRNA comprising a modified nucleotide.
[00126] Further provided herein are vectors comprising a polynucleotide described herein. In some embodiments, a vector described herein can be used for recombinant expression of an isolated polypeptide described herein, a variant HIV Env gpl20 polypeptide described herein, a recombinant HIV Env trimer described herein, a VLP described herein, or a nanoparticle described herein. In one embodiment, a vector described herein can be used for administration of an isolated polypeptide described herein, a variant HIV Env gpl20 polypeptide described herein, a recombinant HIV Env trimer described herein, or a nanoparticle described herein to a patient in need thereof.
[00127] In one aspect, provided herein are RNA replicons comprising an isolated polynucleotide described herein.
[00128] In some embodiments, an isolated polynucleotide described herein encodes an isolated polypeptide described herein. In some embodiments, the isolated polypeptide described herein comprises the amino acid sequence of BG505 MD39 N332-GT1, BG505 MD39 N332-GT2, BG505 MD39 N332-GT3, BG505 MD39 N332-GT4, BG505 MD39 N332-GT5, or BG505 MD39 N332-GT2-KO.
[00129] In some embodiments, an isolated polynucleotide described herein encodes a nanoparticle described herein. In some embodiments, a nanoparticle described herein comprises N332-GT1, N332-GT2, N332-GT3, N332-GT4, orN332-GT5. In some embodiments, a nanoparticle described herein comprises N332-GT2. In some embodiments, a nanoparticle described herein comprises N332-GT5. In some embodiments, a nanoparticle described herein is a ferritin nanoparticle comprising the SEQ ID NO. of 13 or 15.
[00130] In some embodiments, an isolated polynucleotide described herein encodes a recombinant HIV Env trimer described herein. In some embodiments, the recombinant HIV Env trimer described herein comprises the amino acid sequence of BG505 MD39 N332-GT1, BG505 MD39 N332-GT2, BG505 MD39 N332-GT3, BG505 MD39 N332-GT4, BG505 MD39 N332-GT5, or BG505 MD39 N332-GT2-KO.
[00131] In some embodiments, an isolated polynucleotide described herein encodes an antibody described herein. In some embodiments, an isolated polynucleotide described herein encodes BG18.11, BG18.6. In some embodiments, an isolated polynucleotide described herein encodes BG18 iGLO, BG18 iGLl, or BG18 iGL2. In some embodiments, an isolated polynucleotide described herein encodes PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15. In some embodiments, an isolated polynucleotide described herein encodes VH4-595, VH1-69, VH5-51, VH3-33, VH3-23, VL3-10, VL3-19, VL3- 1, VL3-21, or VL2-8.
[00132] In one embodiment, an isolated polynucleotide described herein encodes a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein and comprises an mRNA. In one embodiment, the mRNA comprises at least one modified nucleotide. In one embodiment, a modified mRNA encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein is for administering to a subject to treat or prevent HIV infection.
[00133] As used herein, an "isolated" polynucleotide or nucleic acid molecule is one, which is separated from other nucleic acid molecules, which are present in the natural source (e.g., in a mouse or a human) of the nucleic acid molecule. Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized. For example, the language "substantially free" includes preparations of polynucleotide or nucleic acid molecule having less than about 15%, 10%, 5%, 2%, 1%, 0.5%, or 0.1% (in particular less than about 10%) of other material, e.g., cellular material, culture medium, other nucleic acid molecules, chemical precursors and/or other chemicals. In a specific embodiment, a nucleic acid molecule(s) encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein is isolated or purified.
[00134] In yet another specific embodiment, a polynucleotide provided herein comprises a nucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein that is optimized, e.g., by codon/RNA optimization, replacement with heterologous signal sequences, and elimination of mRNA instability elements. Methods to generate optimized nucleic acids for recombinant expression by introducing codon changes and/or eliminating inhibitory regions in the mRNA can be carried out by adapting the optimization methods described in, e.g., U.S. Patent Nos. 5,965,726; 6, 174,666; 6,291,664; 6,414,132; and 6,794,498, accordingly. For example, potential splice sites and instability elements (e.g., A/T or A/U rich elements) within the RNA can be mutated without altering the amino acids encoded by the nucleic acid sequences to increase stability of the RNA for recombinant expression. The alterations utilize the degeneracy of the genetic code, e.g., using an alternative codon for an identical amino acid. In some embodiments, it can be desirable to alter one or more codons to encode a conservative mutation, e.g., a similar amino acid with similar chemical structure and properties and/or function as the original amino acid.
[00135] In certain embodiments, an optimized polynucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein can hybridize to an antisense (e.g., complementary) polynucleotide of an unoptimized polynucleotide sequence encoding polypeptide described herein or a fragment thereof (e.g., VL domain or VH domain). In specific embodiments, an optimized nucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein or a fragment hybridizes under high stringency conditions to antisense polynucleotide of an unoptimized polynucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein or a fragment thereof. In a specific embodiment, an optimized nucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein hybridizes under high stringency, intermediate or lower stringency hybridization conditions to an antisense polynucleotide of an unoptimized nucleotide sequence encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein or a fragment thereof. Information regarding hybridization conditions has been described, see, e.g., U.S. Patent Application Publication No. US 2005/0048549 (e.g., paragraphs 72-73), which is incorporated herein by reference.
[00136] The polynucleotides can be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art. Nucleotide sequences encoding antibodies described herein, and modified versions of these antibodies can be determined using methods well-known in the art, i.e., nucleotide codons known to encode particular amino acids are assembled in such a way to generate a nucleic acid that encodes the antibody. Such a polynucleotide encoding the antibody can be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier G et ah, (1994), BioTechniques 17: 242-246), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.
[00137] Also provided are polynucleotides that hybridize under high stringency, intermediate or lower stringency hybridization conditions to polynucleotides that encode an antibody described herein. In specific embodiments, polynucleotides described herein hybridize under high stringency, intermediate or lower stringency hybridization conditions to polynucleotides encoding a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) provided herein.
[00138] Hybridization conditions have been described in the art and are known to one of skill in the art. For example, hybridization under stringent conditions can involve hybridization to filter- bound DNA in 6x sodium chloride/sodium citrate (SSC) at about 45°C followed by one or more washes in 0.2xSSC/0.1% SDS at about 50-65°C; hybridization under highly stringent conditions can involve hybridization to filter-bound nucleic acid in 6xSSC at about 45°C followed by one or more washes in O. lxSSC/O.2% SDS at about 68°C. Hybridization under other stringent hybridization conditions are known to those of skill in the art and have been described, see, for example, Ausubel FM et ah, eds., (1989) Current Protocols in Molecular Biology, Vol. I, Green Publishing Associates, Inc. and John Wiley & Sons, Inc., New York at pages 6.3.1-6.3.6 and 2.10.3.
V. Vectors, Cells, and Methods of Producing a Polypeptide
[00139] In one aspect, provided herein are isolated vectors comprising a polynucleotide described herein. In some embodiments, an isolated vector described herein is a viral vector.
[00140] In one aspect, provided herein are recombinant viruses comprising a polynucleotide described herein. In some embodiments, a recombinant virus described herein is a recombinant adeno-associated virus (AAV). [00141] In one aspect, provided herein are host cells comprising a polynucleotide described herein, or a vector described herein. In some embodiments, a host cell described herein is selected from the group consisting of E. coli, Pseudomonas, Bacillus, Streptomyces, yeast, CHO, YB/20, NSO, PER-C6, HEK-293T, NIH-3T3, HeLa, BHK, Hep G2, SP2/0, Rl . l, B-W, L-M, COS 1, COS 7, BSC1, BSC40, BMT10 cell, plant cell, insect cell, and human cell in tissue culture.
[00142] In one aspect, provided herein are methods of producing an isolated polypeptide described herein, a recombinant HIV Env trimer described herein, or a nanoparticle described herein comprising, culturing the host cell described herein so that the polynucleotide is expressed and the isolated polypeptide, recombinant HIV Env trimer, or the nanoparticle is produced.
[00143] In certain aspects, provided herein are cells (e.g., host cells) expressing (e.g., recombinantly) polypeptides described herein and related polynucleotides and expression vectors. Provided herein are vectors (e.g., expression vectors) comprising polynucleotides comprising nucleotide sequences encoding polypeptides described herein. In one embodiment, the vectors can be used for recombinant expression of a polypeptide described herein in host cells (e.g., mammalian cells). In one embodiment, the vectors can be used for administration of a polypeptide described herein to a patient in need thereof. Also provided herein are host cells comprising such vectors for recombinantly expressing polypeptides described herein. In a particular aspect, provided herein are methods for producing a polypeptide described herein, comprising expressing such polypeptides in a host cell.
[00144] In certain aspects, provided herein is an isolated vector comprising a polynucleotide described herein. In one embodiment, the vector is a viral vector.
[00145] In certain aspects, provided herein is a recombinant virus comprising a polynucleotide described herein. In one embodiment, the recombinant virus encodes a polypeptide described herein. In one embodiment, the recombinant virus encodes a variant HIV Env polypeptide described herein. In one embodiment, the recombinant virus is a replication defective virus. Suitable replication defective viral vectors are known to those skilled in the art, for example, as disclosed in U.S. Pat. Nos. 7198784, 9408905, 9862931, 8067156, U.S. Pat. Appl. Pub. Nos. 20150291935, 20120220492, 20180291351, and 20170175137, each of which is incorporated herein by reference in its entirety. In one embodiment, the recombinant virus is a retrovirus or retroviral vector, for example, a lentivirus or lentiviral vector. In one embodiment, the recombinant virus is an adenovirus or adenoviral vector, HSV or HSV vector, or influenza virus or viral vector. In one embodiment, the recombinant virus is an adeno-associated virus (AAV). In one embodiment, the recombinant virus is for administration to a subject to prevent or treat HIV infection. In one embodiment, the recombinant virus is an adeno-associated virus (AAV) for administration to a subject to prevent or treat HIV infection. Recombinant AAV particles encoding a polypeptide described herein and methods for producing thereof are known to one skilled in the art, for example, as disclosed in US Patent 8,865,881 and US20190031740, each of which is incorporated by reference herein in its entirety for all purposes. See also, Lin and Balazs, Retrovirology 15:66 (2018) and van den Berg et al, Molecular Therapy: Methods & Clinical Development 14: 100-112 (2019), each of which is incorporated by reference herein in its entirety for all purposes.
[00146] In certain aspects, provided herein is a host cell comprising a polynucleotide described herein, or a vector described herein. In one embodiment, the vector encodes a polypeptide described herein.
[00147] In one embodiment, the host cell is selected from the group consisting of E. coli, Pseudomonas, Bacillus, Streptomyces, yeast, CHO, YB/20, NS0, PER-C6, HEK-293T, NIH-3T3, Helga, BHK, Hep G2, SP2/0, Rl . l, B-W, L-M, COS 1, COS 7, BSC1, BSC40, BMT10 cell, plant cell, insect cell, and human cell in tissue culture. In one embodiment, the host cell is CHO.
[00148] In certain aspects, provided herein is a method of producing a polypeptide described herein comprising culturing a host cell described herein so that the polynucleotide is expressed and the polypeptide is produced. In one embodiment, the method further comprises recovering the polypeptide.
[00149] The isolated polypeptides (e.g., recombinant HIV Env trimer or nanoparticle) described herein can be produced by any suitable method known in the art. Such methods range from direct protein synthetic methods to constructing a DNA sequence encoding isolated polypeptide sequences and expressing those sequences in a suitable transformed host. In some embodiments, a DNA sequence is constructed using recombinant technology by isolating or synthesizing a DNA sequence encoding a wild-type protein of interest. Optionally, the sequence can be mutagenized by site-specific mutagenesis to provide functional analogs thereof. See, e.g. Zoeller et al., Proc. Nat'l. Acad. Sci. USA 81 :5662-5066 (1984) and U.S. Pat. No. 4,588,585.
[00150] In some embodiments, a DNA sequence encoding a polypeptide of interest would be constructed by chemical synthesis using an oligonucleotide synthesizer. Such oligonucleotides can be designed based on the amino acid sequence of the desired polypeptide and selecting those codons that are favored in the host cell in which the recombinant polypeptide of interest will be produced. Standard methods can be applied to synthesize an isolated polynucleotide sequence encoding an isolated polypeptide of interest. For example, a complete amino acid sequence can be used to construct a back-translated gene. Further, a DNA oligomer containing a nucleotide sequence coding for the particular isolated polypeptide can be synthesized. For example, several small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated. The individual oligonucleotides typically contain 5' or 3' overhangs for complementary assembly.
[00151] Once assembled (by synthesis, site-directed mutagenesis or another method), the polynucleotide sequences encoding a particular isolated polypeptide of interest will be inserted into an expression vector and operatively linked to an expression control sequence appropriate for expression of the protein in a desired host. Proper assembly can be confirmed by nucleotide sequencing, restriction mapping, and expression of a biologically active polypeptide in a suitable host. As is well-known in the art, in order to obtain high expression levels of a transfected gene in a host, the gene must be operatively linked to transcriptional and translational expression control sequences that are functional in the chosen expression host.
[00152] In certain embodiments, recombinant expression vectors are used to amplify and express DNA encoding polypeptides of interest. Recombinant expression vectors are replicable DNA constructs, which have synthetic or cDNA-derived DNA fragments encoding a polypeptide operatively linked to suitable transcriptional or translational regulatory elements derived from mammalian, microbial, viral or insect genes. A transcriptional unit generally comprises an assembly of (1) a genetic element or elements having a regulatory role in gene expression, for example, transcriptional promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription and translation initiation and termination sequences. Such regulatory elements can include an operator sequence to control transcription. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants can additionally be incorporated. DNA regions are operatively linked when they are functionally related to each other. For example, DNA for a signal peptide (secretory leader) is operatively linked to DNA for a polypeptide if it is expressed as a precursor, which participates in the secretion of the polypeptide; a promoter is operatively linked to a coding sequence if it controls the transcription of the sequence; or a ribosome binding site is operatively linked to a coding sequence if it is positioned so as to permit translation. Structural elements intended for use in yeast expression systems include a leader sequence enabling extracellular secretion of translated protein by a host cell. Alternatively, where recombinant protein is expressed without a leader or transport sequence, it can include an N- terminal methionine residue. This residue can optionally be subsequently cleaved from the expressed recombinant protein to provide a final product.
[00153] The choice of expression control sequence and expression vector will depend upon the choice of host. A variety of host-expression vector systems can be utilized to express polypeptide molecules described herein (see, e.g., U.S. Patent No. 5,807,715). Such host-expression systems represent vehicles by which the coding sequences of interest can be produced and subsequently purified, but also represent cells which can, when transformed or transfected with the appropriate nucleotide coding sequences, express a polypeptide molecule described herein in situ. These include but are not limited to microorganisms such as bacteria (e.g., E. coli and B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing polypeptide coding sequences; yeast (e.g., Saccharomyces pichia) transformed with recombinant yeast expression vectors containing polypeptide coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing polypeptide coding sequences; plant cell systems (e.g., green algae such as Chlamydomonas reinhardtii) infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing polypeptide coding sequences; or mammalian cell systems (e.g., COS (e.g., COS1 or COS), CHO, BHK, MDCK, HEK 293, NS0, PER.C6, VERO, CRL7030, HsS78Bst, Helga, and NIH 3T3, HEK-293T, HepG2, SP210, Rl.l, B-W, L-M, BSC1, BSC40, YB/20 and BMT10 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). In a specific embodiment, cells for expressing polypeptides described herein are CHO cells, for example CHO cells from the CHO GS System™ (Lonza). In a particular embodiment, cells for expressing polypeptides described herein are human cells, e.g., human cell lines. In a specific embodiment, a mammalian expression vector is pOptiVEC™ or pcDNA3.3. In a particular embodiment, bacterial cells such as E. coli, or eukaryotic cells (e.g., mammalian cells) are used for the expression of a recombinant polypeptide molecule. For example, mammalian cells such as Chinese hamster ovary (CHO) cells in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for polypeptides. In a specific embodiment, the expression of nucleotide sequence encoding polypeptides described herein is regulated by a constitutive promoter, inducible promoter or tissue specific promoter. [00154] For applications where it is desired that the polypeptides described herein be expressed in vivo, for example in a subject in need of treatment with a polypeptide described herein, any vector that allows for the expression of the polypeptides and is safe for use in vivo can be used. In one embodiment, the vector is a viral vector. Viral vectors can include poxvirus (vaccinia), including vaccinia Ankara and canarypox; adenoviruses, including adenovirus type 5 (Ad5); rubella; sendai virus; rhabdovirus; alphaviruses; and adeno-associated viruses. In one embodiment, the viral vector is an adeno-associated virus. Alternatively, a polynucleotide encoding the polypeptide could be delivered as DNA or RNA to the subject for in vivo expression of the polypeptide.
[00155] Suitable host cells for expression of a polypeptide of interest such as a polypeptide described herein include prokaryotes, yeast, insect or higher eukaryotic cells under the control of appropriate promoters. Prokaryotes include gram negative or gram-positive organisms, for example E. coli or bacilli. Higher eukaryotic cells include established cell lines of mammalian origin. Cell- free translation systems could also be employed. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. (Cloning Vectors: A Laboratory Manual, Elsevier, N.Y., 1985), the relevant disclosure of which is hereby incorporated by reference.
[00156] Various mammalian or insect cell culture systems are also advantageously employed to express a recombinant protein such as a polypeptide described herein. Expression of recombinant proteins in mammalian cells can be performed because such proteins are generally correctly folded, appropriately modified and completely functional. Examples of suitable mammalian host cell lines include but are not limited to CHO, VERO, BHK, Hela, MDCK, HEK 293, NIH 3T3, W138, BT483, Hs578T, HTB2, BT20 and T47D, NS0 (a murine myeloma cell line that does not endogenously produce any immunoglobulin chains), CRL7030, COS (e.g., COS1 or COS), PER.C6, VERO, HsS78Bst, HEK-293T, HepG2, SP210, Rl . l, B-W, L-M, BSC1, BSC40, YB/20, BMT10 and HsS78Bst cells. Mammalian expression vectors can comprise non-transcribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5' or 3' flanking non-transcribed sequences, and 5' or 3' non-translated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Bio/Technology 6:47 (1988). [00157] The proteins produced by a transformed host can be purified according to any suitable method. Such standard methods include chromatography (e.g., ion exchange, affinity and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for protein purification. Affinity tags such as hexahistidine, maltose binding domain, influenza HA peptide sequence and glutathione-S-transferase can be attached to the protein to allow easy purification by passage over an appropriate affinity column. Isolated proteins can also be physically characterized using such techniques as proteolysis, nuclear magnetic resonance and x-ray crystallography.
[00158] For example, supernatants from systems, which secrete recombinant protein, e.g., a polypeptide, into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. Following the concentration step, the concentrate can be applied to a suitable purification matrix. Alternatively, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification. Alternatively, a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Finally, one or more reversed-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, e.g. , silica gel having pendant methyl or other aliphatic groups, can be employed to further an agent. Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a homogeneous recombinant protein.
[00159] Recombinant protein produced in bacterial culture can be isolated, for example, by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange or size exclusion chromatography steps. High performance liquid chromatography (HPLC) can be employed for final purification steps. Microbial cells employed in expression of a recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
[00160] In specific embodiments, a polypeptide (e.g., recombinant HIV Env trimer or nanoparticle) described herein is isolated or purified. Generally, an isolated polypeptide is one that is substantially free of other polypeptides. For example, in a particular embodiment, a preparation of a polypeptide described herein is substantially free of cellular material and/or chemical precursors. The language "substantially free of cellular material" includes preparations of a polypeptide in which the polypeptide is separated from cellular components of the cells from which it is isolated or recombinantly produced. Thus, a polypeptide that is substantially free of cellular material includes preparations having less than about 30%, 20%, 10%, 5%, 2%, 1%, 0.5%, or 0.1% (by dry weight) of heterologous protein (also referred to herein as a "contaminating protein") and/or variants of a polypeptide, for example, different post-translational modified forms of a polypeptide. When the polypeptide (e.g., recombinant HIV Env trimer or nanoparticle described herein) is recombinantly produced, it is also generally substantially free of culture medium, i.e., culture medium represents less than about 20%, 10%, 2%, 1%, 0.5%, or 0.1% of the volume of the protein preparation. When the polypeptide (e.g., recombinant HIV Env trimer or nanoparticle described herein) is produced by chemical synthesis, it is generally substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals, which are involved in the synthesis of the protein. Accordingly, such preparations of the polypeptide (e.g., recombinant HIV Env trimer or nanoparticle described herein) have less than about 30%, 20%, 10%, or 5% (by dry weight) of chemical precursors or compounds other than the polypeptide of interest. In one embodiment, polypeptides described herein are isolated or purified.
VI. Pharmaceutical and Immunogenic Compositions
[00161] Compositions comprising polypeptides described herein (e.g., recombinant HIV Env trimer or nanoparticle) are also provided. Further provided herein are compositions comprising a polynucleotide encoding a polypeptide described herein. In some embodiments, the polynucleotide comprises mRNA. In some embodiments, the composition is a pharmaceutical composition. In some embodiments, the composition is an immunogenic composition.
[00162] In one aspect, provided herein are pharmaceutical compositions comprising an isolated polypeptide described herein, recombinant HIV Env trimer described herein, nanoparticle described herein, polynucleotide described herein, recombinant virus described herein, VLP described herein, or liposome described herein and a pharmaceutically acceptable excipient. In some embodiments, the composition is an immunogenic composition. In some embodiments, the composition is a vaccine. In some embodiments, an immunogenic composition or vaccine further comprises an adjuvant. In some embodiments, the composition (e.g., immunogenic composition) is formulated for subcutaneous administration. In some embodiments, the composition (e.g., immunogenic composition) is formulated for intramuscular administration. In some embodiments, the composition (e.g., immunogenic composition) is formulated for topical administration, and in certain embodiments the composition is formulated for vaginal or rectal administration.
[00163] In one aspect, provided herein are immunogenic compositions comprising an isolated polypeptide described herein, recombinant HIV Env trimer described herein, nanoparticle described herein, polynucleotide described herein, recombinant virus described herein, VLP described herein, or liposome described herein and a pharmaceutically acceptable excipient.
[00164] In some embodiments, an immunogenic composition described herein further comprises an adjuvant. In some embodiments, the adjuvant comprises lecithin. In some embodiments, the adjuvant comprises alum. In some embodiments, the adjuvant comprises saponin, cholesterol and phospholipid. In some embodiments, the adjuvant comprises ISCOMATRIX™. In some embodiments, the adjuvant comprises carbomer homopolymer and lecithin. In some embodiments, the adjuvant comprises Adjuplex™.
[00165] In some embodiments, an immunogenic composition described herein is capable of eliciting a BG18 like response. In some embodiments, an immunogenic composition described herein is capable of eliciting a BG18 like response in a human subject. In some embodiments, an immunogenic composition described herein is capable of eliciting a BG18 like response in BG18gH B cell adoptive transfer recipient mice. In some embodiments, an immunogenic composition described herein is capable of eliciting the production of an antibody that binds to N332-GT2. In some embodiments, an immunogenic composition described herein is capable of eliciting the production of an antibody that binds to N332-GT2 with a higher affinity than to N332-GT2-KO. In some embodiments, an immunogenic composition described herein is capable of eliciting the production of a broadly neutralizing antibody in a subject.
[00166] In one aspect, provided herein are liposomes comprising an isolated polypeptide described herein.
[00167] Compositions described herein comprising a polypeptide described herein are intended for prevention and treatment of HIV infection. In some embodiments, compositions described herein (e.g., immunogenic composition) are for eliciting an immune reaction in a subject against HIV. In some embodiments, compositions described herein (e.g., immunogenic composition) are for eliciting a protective immune reaction in a subject against HIV.
[00168] In further embodiments of the present disclosure, a composition comprising a polypeptide described herein can additionally be combined with other compositions for the treatment of HIV infection or the prevention of HIV transmission. [00169] In some embodiments, a polypeptide described herein can be administered within a pharmaceutically acceptable diluent, carrier, or excipient, in unit dose form. Conventional pharmaceutical practice can be employed to provide suitable formulations or compositions for administration to individuals. In some embodiments, the administration is prophylactic. Any appropriate route of administration can be employed, for example, administration can be parenteral, intravenous, intra-arterial, subcutaneous, intramuscular, intraperitoneal, intranasal, aerosol, suppository, oral administration, vaginal, or anal.
[00170] In some embodiments, the polypeptides and polynucleotides described herein are administered as a component of an immunogenic composition comprising the polypeptides and/or polynucleotides described herein in admixture with a pharmaceutically acceptable carrier. The immunogenic compositions described herein are useful to stimulate an immune response against HIV- 1 and can be used as one or more components of a prophylactic or therapeutic vaccine against HIV-1 for the prevention, amelioration or treatment of AIDS. The polynucleotides and vectors described herein are particularly useful for providing genetic vaccines, i.e. vaccines for delivering the polynucleotides encoding the antigenic polypeptides described herein to a subject, such as a human, such that the antigenic polypeptides are then expressed in the subject to elicit an immune response.
[00171] In some embodiments, immunogenic compositions described herein comprise injectable suspensions, solutions, sprays, lyophilized powders, syrups, elixirs and the like. Any suitable form of composition can be used. To prepare such a composition, a polypeptide and/or polynucleotide described herein, having the desired degree of purity, is mixed with one or more pharmaceutically acceptable carriers and/or excipients. The carriers and excipients must be "acceptable" in the sense of being compatible with the other ingredients of the composition. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to, water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, or combinations thereof, buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn- protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol. In some embodiments, immunogenic compositions described herein are formulated in the form of an oil-in-water emulsion. In some embodiments, the oil-in-water emulsion is based, for example, on light liquid paraffin oil (European Pharmacopeatype); isoprenoid oil such as squalane, squalene, EICOSANE™ or tetratetracontane; oil resulting from the oligomerization of alkene(s), e.g., isobutene or decene; esters of acids or of alcohols containing a linear alkyl group, such as plant oils, ethyl oleate, propylene glycol di(caprylate/caprate), glyceryl tri(caprylate/caprate) or propylene glycol dioleate; esters of branched fatty acids or alcohols, e.g., isostearic acid esters. In some embodiments, the oil is used in combination with an emulsifier to form the emulsion. In some embodiments, the emulsifier comprises a nonionic surfactant, such as esters of sorbitan, mannide (e.g., anhydromannitol oleate), glycerol, polyglycerol, propylene glycol, and oleic, isostearic, ricinoleic, or hydroxy stearic acid, which are optionally ethoxylated, and polyoxypropylene- polyoxyethylene copolymer blocks, such as the Pluronic® products, e.g., L121. In some embodiments, the adjuvant can be a mixture of emulsifier(s), micelle-forming agent, and oil such as that which is commercially available under the name Provax® (IDEC Pharmaceuticals, San Diego, CA). (PEG).
[00172] In some embodiments, an immunogenic composition described herein comprises additional substances, such as wetting or emulsifying agents, buffering agents, or adjuvants to enhance the effectiveness of the vaccines (Remington's Pharmaceutical Sciences, 18th edition, Mack Publishing Company, (ed.) 1980).
[00173] In some embodiments, an immunogenic composition described herein comprises an adjuvant. Suitable adjuvants include, but are not limited to, mineral salts (e.g., AIK^SCEK AlNa(S04)2, A1NH(S04)2, silica, alum, Al(OH)3, Ca3(PC>4)2, kaolin, or carbon), polynucleotides with or without immune stimulating complexes (ISCOMs) (e.g., CpG oligonucleotides, such as those described in Chuang, T.H. et al, (2002) J. Leuk. Biol. 71(3): 538- 44; Ahmad-Nejad, P. et al (2002) Eur. J. Immunol. 32(7): 1958-68; poly IC or poly AU acids, polyarginine with or without CpG (also known in the art as IC3 1; see Schellack, C. et al (2003) Proceedings of the 34th Annual Meeting of the German Society of Immunology; Lingnau, K. et al (2002) Vaccine 20(29-30): 3498- 508), JuvaVax™ (U.S. Patent No. 6,693,086), certain natural substances (e.g., wax D fromMycobacterium tuberculosis, substances found in Comyebacterium parvum, Bordetella pertussis, or members of the genus Brucella), flagellin (Toll-like receptor 5 ligand; see McSorley, S.J. et al (2002) J. Immunol. 169(7): 3914-9), saponins such as QS21, QS17, and QS7 (U.S. Patent Nos. 5,057,540; 5,650,398; 6,524,584; 6,645,495), monophosphoryl lipid A, in particular, 3-de-0- acylated monophosphoryl lipid A (3D-MPL), imiquimod (also known in the art as IQM and commercially available as Aldara®; U.S. Patent Nos. 4,689,338; 5,238,944; Zuber, AK. et al (2004) 22(13-14): 1791-8), and the CCR5 inhibitor CMPD 167 (see Veazey, RS. et al (2003) J. Exp. Med. 198: 1551-1562).
[00174] Aluminum hydroxide or phosphate (alum) are commonly used at 0.05 to 0.1% solution in phosphate buffered saline. Other adjuvants that can be used, especially with DNA vaccines, are cholera toxin, especially CTAl-DD/ISCOMs (see Mowat, AM. et al (2001) J. Immunol. 167(6): 3398-405), polyphosphazenes (Allcock, H.R. (1998) App. Organometallic Chem. 12(10-11): 659- 666; Payne, L.G. et al (1995) Pharm. Biotechnol. 6: 473-93), cytokines such as, but not limited to, IL-2, IL-4, GM-CSF, IL-12, IL-15 IGF-1, IFN-a, IFN- , and IFN-y (Boyer et al, (2002) J. Fiposome Res. 121 : 137-142; WOO1/095919), immunoregulatory proteins such as CD40F (ADX40; see, for example, W003/063899), and the CDla ligand of natural killer cells (also known as CRONY or a- galactosyl ceramide; see Green, T.D. et al, (2003) J. Virol. 77(3): 2046-2055), immunostimulatory fusion proteins such as IF-2 fused to the Fe fragment of immunoglobulins (Barouch et al., Science 290:486-492, 2000) and co-stimulatory molecules B7. 1 and B7.2 (Boyer), all of which can be administered either as proteins or in the form of DNA, on the same expression vectors as those encoding the antigens described herein or on separate expression vectors. In some embodiments, the adjuvant comprises lecithin combined with an acrylic polymer (Adjuplex-FAP), lecithin coated oil droplets in an oil-in-water emulsion (Adjuplex-FE) or lecithin and acrylic polymer in an oil-in water emulsion (Adjuplex-FAO) (Advanced BioAdjuvants (ABA)).
[00175] In some embodiments, an immunogenic composition described herein is formulated to introduce the polypeptides and polynucleotides disclosed herein (collectively, the immunogens) to a desired site of action and release it at an appropriate and controllable rate. Methods of preparing controlled-release formulations are known in the art. For example, controlled release preparations can be produced by the use of polymers to complex or absorb the immunogen and/or immunogenic composition. A controlled-release formulation can be prepared using appropriate macromolecules (for example, polyesters, polyamino acids, polyvinyl, pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate) known to provide the desired controlled release characteristics or release profile. Another possible method to control the duration of action by a controlled-release preparation is to incorporate the active ingredients into particles of a polymeric material such as, for example, polyesters, polyamino acids, hydrogels, polylactic acid, polygly colic acid, copolymers of these acids, or ethylene vinylacetate copolymers. Alternatively, instead of incorporating these active ingredients into polymeric particles, it is possible to entrap these materials into microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin- microcapsule and poly-(methylmethacrylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in New Trends and Developments in Vaccines, Voller et al. (eds.), University Park Press, Baltimore, Md., 1978 and Remington's Pharmaceutical Sciences, 16th edition.
[00176] Pharmaceutical compositions described herein can be, for example, in unit dose form, such as in the form of ampoules, vials, suppositories, tablets, pills, or capsules. The formulations can be administered to human individuals in therapeutically or prophylactic effective amounts (e.g., amounts which prevent, eliminate, or reduce a pathological condition) to provide therapy or prophylaxis for a disease or condition (e.g., HIV infection). The preferred dosage of therapeutic or prophylactic agent to be administered is likely to depend on such variables as the type and extent of the disorder, the overall health status of the particular patient, the formulation of the compound excipients, and its route of administration. Suitable dosages of the polypeptides and polynucleotides disclosed herein (collectively, the immunogens) in the immunogenic composition disclosed herein can be readily determined by those of skill in the art. For example, the dosage of the immunogens can vary depending on the route of administration and the size of the subject. Suitable doses can be determined by those of skill in the art, for example by measuring the immune response of a subject, such as a laboratory animal, using conventional immunological techniques, and adjusting the dosages as appropriate. Such techniques for measuring the immune response of the subject include but are not limited to, chromium release assays, tetramer binding assays, IFN-y ELISPOT assays, IL-2 ELISPOT assays, intracellular cytokine assays, and other immunological detection assays, e.g., as detailed in the text "Antibodies: A Laboratory Manual" by Ed Harlow and David Lane.
VII. Uses and Methods
Therapeutic Uses and Methods: [00177] In one aspect, provided herein are methods for eliciting an immune response to
HIV Env gpl20 in a subject, comprising administering to the subject an effective amount of an immunogenic composition described herein, thereby generating the immune response. In some embodiments, the subject is a human. In some embodiments, the subject has been exposed to HIV. In some embodiments, the subject is at risk of being exposed to HIV. In some embodiments, the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker. In some embodiments, the subject is a non-human primate. In some embodiments, the subject is a BG18gH B cell adoptive transfer recipient mouse. In some embodiments, the subject is a mouse. In some embodiments, the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
[00178] In one aspect, provided herein are methods of reducing the likelihood of HIV infection in a subject exposed to HIV comprising administering to the subject an effective amount of an immunogenic composition described herein, or the pharmaceutical composition described herein. In some embodiments, the subject is a human. In some embodiments, the subject has been exposed to HIV. In some embodiments, the subject is at risk of being exposed to HIV. In some embodiments, the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker. In some embodiments, the subject is a non-human primate. In some embodiments, the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
[00179] In one aspect, provided herein are methods of reducing the risk of a subject becoming infected with HIV comprising administering to the subject in need thereof an effective amount of an immunogenic composition described herein, or the pharmaceutical composition described herein. In some embodiments, the subject is a human. In some embodiments, the subject has been exposed to HIV. In some embodiments, the subject is at risk of being exposed to HIV. In some embodiments, the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker. In some embodiments, the subject is a non-human primate. In some embodiments, the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
[00180] In one aspect, provided herein are methods of preventing HIV infection comprising administering to a subject in need thereof an effective amount of the immunogenic composition described herein, or the pharmaceutical composition described herein. In some embodiments, the subject is a human. In some embodiments, the subject has been exposed to HIV. In some embodiments, the subject is at risk of being exposed to HIV. In some embodiments, the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker. In some embodiments, the subject is a non-human primate. In some embodiments, the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
[00181] In one aspect, provided herein are methods of treating HIV/AIDS comprising administering to a subject in need thereof an effective amount of the immunogenic composition described herein, or the pharmaceutical composition described herein. In some embodiments, the subject is a human. In some embodiments, the subject has been exposed to HIV. In some embodiments, the subject is at risk of being exposed to HIV. In some embodiments, the subject at risk of being exposed to HIV is a health care worker, a sexual partner of an HIV infected individual, or a sex worker. In some embodiments, the subject is a non-human primate. In some embodiments, the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration.
[00182] In some embodiments, the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, the immunogenic composition comprises an isolated polypeptide described herein. In some embodiments, the immunogenic composition comprises a recombinant HIV Env trimer described herein. In some embodiments, the immunogenic composition comprises a nanoparticle described herein. In some embodiments, the immunogenic composition comprises a nucleic acid described herein. In some embodiments, the pharmaceutical composition comprises a recombinant virus described herein. In some embodiments, an immunogenic composition further comprises an adjuvant. In some embodiments, the immunogenic composition is formulated for subcutaneous administration. In some embodiments, the immunogenic composition is formulated for intramuscular administration. [00183] In some embodiments, a method described herein further comprises administering at least one additional therapeutic agent. In some embodiments, the additional therapeutic agent is an antiretroviral agent. In some embodiments, the additional therapeutic agent comprises an antiretroviral therapy (ART) agent, a reservoir activator, an immunomodulator, a second antibody, or a broadly neutralizing anti-BIV antibody.
[00184] In one embodiment, the administering to the subject is by at least one mode selected from oral, parenteral, subcutaneous, intramuscular, intravenous, vaginal, rectal, buccal, sublingual, and transdermal.
[00185] In certain embodiments, the subject is at risk for exposure to HIV. In some embodiments, the subject is infected with HIV. In some embodiments, the subject is diagnosed with AIDS. In certain embodiments, the subject at risk for exposure to HIV is a health care worker. In certain embodiments, the subject at risk for exposure to HIV is a sex worker. In certain embodiments, the subject at risk for exposure to HIV is a sexual partner of an HIV infected individual. In certain embodiments, the subject at risk for exposure to HIV is a newborn.
[00186] In some embodiments, the polypeptides and/or polynucleotides described herein are administered in vivo, for example where the aim is to produce an immunogenic response in a subject. In some embodiments, the "subject" may be any animal. In some embodiments, it will be desirable to express the antigens described herein in human subjects, such as in clinical trials and for actual clinical use of the immunogenic compositions and vaccine of the invention. In some embodiments, the subject is a human, for example a human that is infected with, or is at risk of infection with, HIV-1.
[00187] For such in vivo applications the polypeptides and/or polynucleotides described herein can be administered as a component of an immunogenic composition comprising the polypeptides and/or polynucleotides described herein in admixture with a pharmaceutically acceptable carrier. The immunogenic compositions described herein are useful to stimulate an immune response against HIV- 1 and can be used as one or more components of a prophylactic or therapeutic vaccine against HIV-1 for the prevention, amelioration or treatment of AIDS. The polynucleotides described herein are particularly useful for providing genetic vaccines, i.e. vaccines for delivering the polynucleotides encoding the polypeptides described herein to a subject, such as a human, such that the polypeptides are then expressed in the subject to elicit an immune response.
[00188] When provided prophylactically, the immunogenic compositions described herein can be administered to a subject in advance of HIV infection, or evidence of HIV infection, or in advance of any symptom due to AIDS, especially in high-risk subjects. The prophylactic administration of the immunogenic compositions described herein can serve to provide protective immunity of a subject against HIV-1 infection or to prevent or attenuate the progression of AIDS in a subject already infected with HIV-1. When provided therapeutically, the immunogenic compositions described herein can serve to ameliorate and treat AIDS symptoms and can be used as soon after infection as possible, for example, before appearance of any symptoms of AIDS, but can also be used at (or after) the onset of the disease symptoms.
[00189] Immunization schedules (or regimens) are well known for animals (including humans) and can be readily determined for the particular subject and immunogenic composition. Hence, the immunogens can be administered one or more times to the subject. In some embodiments, there is a set time interval between separate administrations of the immunogenic composition. While this interval varies for every subject, typically it ranges from 10 days to several weeks, and is often 2, 4, 6 or 8 weeks. For humans, the interval is typically from 2 to 6 weeks. The immunization regimes typically have from 1 to 6 administrations of the immunogenic composition, but can have as few as one or two or four. The methods of inducing an immune response described herein can also include administration of an adjuvant with the immunogens. In some instances, annual, biannual or other long interval (5-10 years) booster immunization can supplement the initial immunization protocol.
[00190] The methods described herein also include a variety of prime-boost regimens. In these methods, one or more priming immunizations are followed by one or more boosting immunizations. The actual immunogenic composition can be the same or different for each immunization and the type of immunogenic composition (e.g., containing protein or expression vector), the route, and formulation of the immunogens can also be varied. For example, if an expression vector is used for the priming and boosting steps, it can either be of the same or different type (e.g., DNA or bacterial or viral expression vector). In some embodiments, the prime-boost regimen provides for two priming immunizations, four weeks apart, followed by two boosting immunizations at 4 and 8 weeks after the last priming immunization. It should also be readily apparent to one of skill in the art that there are several permutations and combinations that are encompassed using a polypeptide (e.g., Env trimer or nanoparticle) described herein to provide priming and boosting regimens.
[00191] In some embodiments, a method of inducing an immune response against HIV in a subject described herein comprises administering an immunogenic composition of the invention comprising an adenovirus vector containing DNA encoding one or more of the polypeptides described herein, one or more times to a subject wherein the polypeptides are expressed at a level sufficient to induce a specific immune response in the subject. Such immunizations can be repeated multiple times at time intervals of at least 2, 4 or 6 weeks (or more) in accordance with a desired immunization regime.
[00192] The immunogenic compositions of the invention can be administered alone, or can be co-administered, or sequentially administered, with other HIV immunogens and/or HIV immunogenic compositions, e.g., with "other" immunological, antigenic or vaccine or therapeutic compositions thereby providing multivalent or "cocktail" or combination compositions. Again, the ingredients and manner (sequential or co-administration) of administration, as well as dosages can be determined taking into consideration such factors as the age, sex, weight, species and condition of the particular subject, and the route of administration.
[00193] When used in combination, the other HIV immunogens can be administered at the same time or at different times as part of an overall immunization regime, e.g., as part of a prime- boost regimen or other immunization protocol. In some embodiments, the other HIV immunogen is Env, for example, an HIV Env trimer.
Vaccine design
[00194] Provided herein are methods for identifying vaccines capable of eliciting a broadly neutralizing antibody (bnAb) response in a subject (e.g., a human subject).
[00195] In one aspect, provided herein are methods for identifying a vaccine candidate variant HIV Env polypeptide (e.g., gpl20 polypeptide or HIV Env trimer), the method comprising providing a library comprising a plurality of variant HIV Env polypeptides; contacting the library with an antibody described herein; and identifying a variant HIV Env polypeptide that specifically binds to the antibody. In some embodiments of a method for identifying a vaccine candidate variant HIV Env polypeptide described herein the plurality of variant HIV Env polypeptides comprise variants of a parental HIV Env polypeptide comprising one or more amino acid substitutions at a region selected from the VI loop comprising positions 131-154, V3 loop comprising positions 297-334, and b19 sheet comprising positions 413-419, wherein HIV Env polypeptide positions correspond to the HXB2 reference (SEQ ID NO: 1). In some embodiments of a method for identifying a vaccine candidate variant HIV Env polypeptide described herein the parental HIV Env polypeptide comprises BG505, BG505 N332, BG505 MD39, BG505 MD39 N332, BG505 MD39 l lmutB, or BG505 MD39 17mutE Env polypeptide. In some embodiments, the Env polypeptide is a gpl60 polypeptide, a gpl20 polypeptide, or an Env trimer polypeptide.
[00196] Embodiments of the present disclosure can be further defined by reference to the following non-limiting examples, which describe in detail materials and methods of the present disclosure. It will be apparent to those skilled in the art that many modifications, both to materials and methods, can be practiced without departing from the scope of the present disclosure.
[00197] All documents, patent, and patent applications cited herein are hereby incorporated by reference, and may be employed in the practice described herein.
EXAMPLES
Experimental design
[00198] Next generation sequencing (NGS) and bioinformatics was used to identify HCDR3s sharing key properties with the HCDR3 -dependent bnAb BG18 in 14 of 14 HIV-uninfected donors. Then, BG18 inferred-germline (iGL) variants with NGS-derived HCDR3 loops were used as targets for structural-model-guided, multitarget mammalian-display selection of a series of Env trimers (N332-GT trimers) that bind to diverse BG18 potential precursors. Sorting of human B cells from HIV-uninfected donors using N332-GT trimer probes revealed BG18-like naive B cells with long HCDR3 loops and light chains similar to those in BG18. Cryo-electron microscopy complexes of a native-like trimer bound to BG18, and of N332-GT trimers bound to either BG18 iGL or human naive B cell sorting-derived Abs, all showed a similar binding epitope and orientation, supporting the bnAb precursor potential of the sorting-derived Abs. N332-GT trimer-based nanoparticles were constructed. It was found that nanoparticle immunization initiated responses from rare BG18-like B cells in mice hosting transferred BG18 iGL heavy chain B cells. This study demonstrated the potential for vaccine targeting of rare HCDR3 -dependent bnAb precursors and suggested that N332-GT-trimer-based nanoparticles have promise for priming BG18-like bnAb responses in humans.
[00199] Germline -targeting (GT) HIV vaccine design is a strategy that aims to induce bnAbs by first priming the precursor B cells of known bnAbs and then shepherding B cell affinity maturation with a series of rationally designed boosting immunogens. There have been many advances in developing and testing germline -targeting immunogens to prime precursors of VRC01- class bnAbs with an Env-binding mode that is dependent on the human VH1-2 gene but permissive for a wide variety of HCDR3 loops1 10, and at least one such immunogen is entering human clinical testing. However, VRC01 -class bnAbs represent a specialized case in which non-HCDR3 features dominate the binding modality. In contrast, most HIV bnAbs and most Abs in general employ the HCDR3 as a major determinant for epitope interaction. Hence, an optimally effective HIV vaccine that induces multiple bnAbs for optimal coverage, and a general solution to germline -targeting vaccine design, will need to include HCDR3 -dependent bnAbs. The enormous junctional diversity within long HCDR3s (at junctions between VH and DH, and DH and JH genes) presents a key difficulty for developing GT immunogens to initiate HCDR3 -dependent bnAb responses consistently. This diversity in the human B cell repertoire makes any single HCDR3 sequence an impractical vaccine target and requires that a pool of potential precursors sharing a minimal set of bnAb-associated genetic features should be identified and targeted.
[00200] GT Env trimers with affinity for iGL precursors of the HCDR3 -dependent bnAb PGT12111 that binds to the N332 glycan supersite, also known as the high mannose patch or V3- glycan epitope, on the trimer12 13 were previously engineered. These GT trimers (lOmut, 1 limits) were able to prime B cells in a PGT121 iGL homozygous B cell receptor (BCR) knock-in mouse. Immunization with a GT prime followed by a series of our shepherding boosts elicited PGT121- class bnAbs in this mouse model, providing proof of principle that HIV bnAbs can be induced starting from B cells bearing human iGL BCRs11 14. Despite this success, human clinical testing of this immunization regimen was not pursued, primarily because lOmut and 1 ImutB trimers had low affinity for PGT121 iGL variants, suggesting that bona fide naive precursors lacking bnAb junctional regions may have very low or no detectable affinity. Correspondingly, attempts to isolate PGT121 precursors from naive human B cells by using 1 ImutB trimers as bait did not succeed (Ligure 10).
[00201] It was considered that BG18, a more recently described N332 glycan-dependent bnAb15, may be a superior vaccine target to PGT 121. BG 18 has higher potency and similar breadth, a shorter HCDR3 suggesting a higher precursor frequency, and a potentially simpler maturation pathway lacking insertions or deletions (indels), whereas PGT21 activity requires indels. As with PGT121, BG18 iGL does not bind to WT BG505 Env (T332N), indicating that protein design would be required to develop an effective priming immunogen. To enable structure-guided immunogen design, how BG18 engages the Env protein was studied. Alignment of the HCDR3 from the crystal structure of unliganded BG1815 to the structure of PGT 122 (a somatic variant of PGT121) bound to BG505 SOSIP16, revealed that the HCDR3s are in a highly similar conformation with identical or chemically similar side chains at epitope contact positions (Figure la). The structural alignment suggested a BG18 binding mode in which the HCDR3 engages the conserved GDIR motif like PGT121, while the HCDR1 contacts the relatively conserved N332 glycan, and the light chain (LC) straddles the VI loop of gpl20, unlike PGT121 (Figure 4a-c). Mutagenesis studies (Figure 4d-e), structural model-guided design of a minimally mutated BG18 bnAb (minBG18) (Figure 5), and a 4.4 A resolution cryo-electron microscopy (cryo-EM) derived structure of BG18 bound to the BG505 MD39 trimer11 (Figure lb), confirmed this HCDR3 binding mode, in agreement with recently published crystal structures of BG18 bound to BG505 and B41 SOSIP trimers17.
Example 1. Identification of BG18-like HCDR3s
[00202] To assess whether BG18-like HCDR3s are present in the HIV-uninfected population, a bioinformatics approach was used to search an NGS dataset of 1.1 c 109 unique sequences of human BCR heavy chains (HCs) from 14 healthy, HIV-uninfected donors (from ref. 18 and this work). Informed by our structural model for the BG18-Env interaction, HCDR3s with the same length as BG18, the same D gene in the same reading frame and position within the HCDR3, and the same JH gene were searched for. Such HCDR3s were identified in all 14 donors (Figure 6), encouraging us to proceed with vaccine design.
Example 2. GT immunogen design
[00203] GT immunogen design was initiated using our directed evolution methodology for engineering trimers on the surface of mammalian cells that previously proved capable of producing stabilized native-like trimers with affinity for PGT121 iGL variants11 19. However, this case was an attempt to overcome the limitations of using only iGL variants to represent the pool of potential bnAb precursors in the human repertoire. Inferred-germline Abs contain bnAb HCDR3 junctions that have been selected and possibly somatically mutated for high affinity Env binding during bnAb development, hence such iGL Abs under-represent both the diversity of potential precursors and the difficulty of engineering high affinity antigens to target those precursors. A set of 15 Abs were designed that use BG18 germline -reverted genes but contain naive human BG18-like HCDR3s with diverse junction regions identified in our search of HC NGS data described above (Figure 6b). It was also hypothesized that BG18-like bnAbs that interact with Env in a mode similar to that of BG18 can utilize alternate VH or VL genes. This idea was supported by our finding that BG18-like bnAbs can derive from at least 3 alternate LCs (Figure 5a), as well as the fact that the key VH contact residues in BG18 are somatic mutations (Figures 5b-c and 8a-b). Therefore, 12 other variants of BG18 iGL were produced that use common human germline VH and VL genes. This gave 28 potential BG18-like precursors, including three iGL variants (BG18 1GL0-2), that could be used as selection reagents for multitarget optimization4. Structure-guided libraries were designed to screen by mammalian cell surface display using 1 ImutB as the starting construct. Five libraries were screened iteratively with selection against the least challenging antibody targets first (i.e. BG18 iGL) followed by selection against more difficult targets (i.e. NGS-derived and alternate VH/VL precursors) in the last three libraries (Figure Id). This directed evolution design process resulted in two GT trimers (N332-GT1 and -GT2; Figure Id and Figure 7). N332-GT2 bound with a KD of 213 pM to BG18 iGLi, which represented a -130, 000-fold improvement over 1 ImutB. 11 of 15 NGS-derived precursors bound to N332-GT2 with a geomean KD of 519 nM, and all VH/VL variants bound to N332-GT2. Overall, 17 of 28 precursors had submicromolar KDS for N332-GT2 (Fig le), a potential benchmark for generating robust germinal center responses from rare B cell precursors in vivo20.
Example 3. Cryo-EM derived structure of GT immunogen-antibody complexes
[00204] Our 3.9 A resolution cryo-EM derived structure of BG18 iGLo complexed with N332- GT2 (Figure lc) showed that the HCDR3 makes a similar interaction to the base of the V3 as mature BG18 (Figure lb) and PGT121, and most of the additional interactions come from VI mutations that fill a groove in the light chain and contact the HCDR3 (Figure 8). The HCDR3 dominated the interaction in the BG18 iGLo complex, burying 64% of the total buried surface area. In the mature BG18 complex, the HCDR3 maintained the same key interactions and contributed 35% of the total buried area as the Ab made substantially increased contacts to glycans (Figure 9b). Overall, our cryo-EM structures showed that N332-GT2 binds to BG18 iGLo with a similar HCDR3 -dependent, VI -straddling binding mode as BG505 MD39 binds to BG18.
Example 4. Identification of BG18-like precursors in the human naive B cell repertoire
[00205] To identify BG18-like precursors in the human naive B cell repertoire that recognize N332-GT trimers, N332-GT1 and N332-GT2 were used as sorting reagents and either BG505- MD39 (containing a native N332 epitope) or N332-GT2-KO (an epitope knockout) as negative sorting probes (Figure 2a). Approximately 47 million naive B cells from 6 donors were probed with N332-GT1, and 112 million naive B cells from 10 donors were probed with N332-GT2, after accounting for PCR and sorting efficiencies. Epitope-specific naive B cells, isolated at a frequency of 0.001% (Figure 10), were enriched for long HCDR3s (Figure 2b) and highly enriched for VL3- 25 and VL3-1 LCS (Figure 2c) corresponding to the BG18 LC and a LC with similar binding properties, respectively (Figure le and Figure 5). Of 146 HC-LC pairs sequenced, one precursor, high mannose patch- 1 (HMP1) was a near perfect match to BG18 iGL, with the same HCDR3 length, D gene, D gene position within the HCDR3, and JH, VL and JL genes (Figure 2d). The isolation of one ideal BG18-like precursor in 159 million B cells is approximately consistent (within a factor of 3) with our bioinformatic estimate of 1 in 54 million human B cells (see Methods and Figure 6). Fabs of 45 HMP naive B cell clones were expressed and purified (Figure lOi) and binding affinities to native-like and GT Env trimers were measured. 17 HMP Fabs bound to N332- GT1 and/or GT2 and did not bind detectably to the MD39 native-like trimer, indicate proper epitope specificity (Figure 2e). HMP1 bound to N332-GT2 with a KD of 220 nM and was the highest affinity HMP Fab tested. Four of the five highest affinity naive Abs isolated (HMP1, HMP30, HMP42, and HMP43), used VL3-25 or VL3- 1 LCS and have long HCDR3 loops (>20 AA) (Figure 2d-e). Initial attempts to determine low resolution EM structures of HMP Fabs bound to N332-GT2 were not successful. However, in the late stages of this study, a higher affinity GT trimer (N332- GT5, Figure 2e and Figure 7) was developed, and this trimer proved capable of forming stable complexes with HMP1, HMP42, and HMP43 that were amenable to cryo-EM structural analysis. At ~15 A resolution, the binding orientations of all three HMP precursors were indistinguishable from BG18 iGLo complexed with N332-GT2 (Figure 2f), suggesting that these naive Abs use a BG18-like binding mode to engage the N332-GT trimer.
Example 5. Ferritin nanoparticle design
[00206] In separate work21, ferritin nanoparticles (NPs)22,23 were constructed that displayed up to 8 copies of MD39 native-like trimers, and it was found that in mouse immunization studies that such NPs were superior to MD39 trimers in trafficking to follicular dendritic cell networks, concentrating in germinal centers, and eliciting IgG responses. To gain such immunogenicity enhancements here, similar ferritin NPs displaying N332-GT2 trimers were engineered (Figure 11). Example 6. BG18gH knock-in mouse
[00207] To test the immunogenicity of these N332-GT2-NPs, a BG18gH knock-in mouse engineered via a CRISPR Cas9 rapid targeting strategy was employed, in which -30% of B cells express the BG18 1GL2 HC variable region and mouse constant region paired with mouse LCs24. N332-GT2 but not MD39 bound to 12±1% of naive B cells in this mouse compared to 0.06±0.01% in WT (C57BL/6) mice, demonstrating N332-GT2 specificity for BG 18sH naive B cells (Figure 3a, b). Antigen-specific single B cell sorting and BCR sequencing demonstrated that the N332-GT2- specific naive BG18gH B cells carry a variety of mouse LCs paired with BG18gH (Figure 3c). Example 7. Immunization studies
[00208] To generate a mouse model with rare bnAb precursor B cells, adoptive transfer experiments were carried out in which 5,000 CD45.2 BG 18gH B cells were transferred to CD45.1 WT mice on day -1, establishing a frequency of ~7 GT2++/KO BG18gH CD45.2 B cells per million CD45.1 B cells by day 0 (Figure 12a-c). Control transfers were 50,000 CD45.2 WT B cells. Recipient mice were immunized at day 0 with either N332-GT2-NPs or control NPs displaying MD39 trimers lacking GT mutations, for a total of four immunization conditions (BG18gH or WT B cells transferred, N332-GT2- or MD39-NPs immunized). Splenocytes were analyzed by cytometry at day 8 (Figure 3d-f). Germinal center (GC) B cells (CD38lowCD95+) were detected in all four immunization conditions, but CD45.2 GC B cells were detected only in the case of N332- GT2-NP immunization of BG 18gH B cell recipients, demonstrating that N332-GT2-NPs activated rare BG18gH B cells in vivo but MD39-NPs did not (Figure 3e). N332-GT2-NP-induced CD45.2 GC B cells bound to N332-GT2 and not to N332-GT2-KO (Fig 3f), consistent with a BG18-like response. In contrast, host CD45.1 GC B cells showed negligible binding to N332-GT2 (Figure 3f), suggesting that the day 8 CD45.1 GC responses were directed either to the ferritin nanoparticle base or to the adjuvant. In day 14 serum binding analyses, GT2-NPs induced strong epitope-specific responses (strong binding to N332-GT2 and weak binding to N332-GT2-KO) in BG 18gH B cell recipients and 15-fold weaker epitope-specific responses in WT B cell recipients (Figure 3g), demonstrating that activation of rare BG 18gH precursor B cells led to potent serum responses but also showing that WT B cells responded to the BG18 epitope on N332-GT2. In contrast, MD39- NPs induced negligible epitope-specific serum responses in either BG18gH or WT B cell recipients (Figure 3g). Together these results demonstrated that N332-GT2-NPs elicited GC and antibody responses from rare BG18gH B cells.
[00209] By single cell sorting and BCR sequencing CD45.2+/N332-GT2++/KO GC B cells from 3 BG18gH recipient mice immunized with N332-GT2-NPs, 134 HC-LC pairs were obtained. 100% of the HCs were derived from BG18gH, formally proving that these GC responses utilized the knockin HC. In contrast to the wide variety of mouse IGKV genes used in LCs of GT2-specific naive BG18gH B cells, the LCs from day 8 GC B cells were highly enriched for two mouse kappa genes: IGKV12-46 and IGKV12-44 (Figure 3c). The amino-acid mutation levels in the day 8 GC BCRs were 1.1±1.2 in the HCs and 1.8±1.3 in the VL genes (Figure 12d-e), whereas naive B cells from unimmunized mice showed no BCR mutations. 14 of the most mutated GT2-NP -induced Fabs were expressed and it was found that they had KDS for N332-GT2 ranging from 15 to 580 nM (geomean 97 nM), whereas 8 Fabs produced from N332-GT2+KO naive B cells in un-immunized BG 18"H mice bound more weakly by an average factor of 6.8, with geomean 655 nM (Fig 3h). This showed that N332-GT-NPs can induce affinity maturation of rare BG18-like precursors.
Discussion
[00210] Most antibodies recognize their target in a strongly HCDR3 -dependent manner. This includes most HIV and influenza bnAbs. A central challenge of GT vaccine design is the large paratope sequence and structural complexity possible for any set of Abs targeting a conserved epitope via a shared HCDR3 -dependent binding modality. Here the successful design of a GT immunogen for this general class of Ab recognition was demonstrated (Figure 13). The strategy was to use the human repertoire as a guide to identify potential bnAb precursors and then to design an immunogen that has affinity for a representative set of those precursors. This procedure was validated by the isolation of an authentic BG18-class precursor (HMP1) from naive human B cells with N332-GT trimers and the demonstration that N332-GT nanoparticles drive a robust BG18- class B cell and GC response in an animal model with rare BG18 precursors. Furthermore, our findings that N332-GT trimer-sorted human naive B cells are enriched for VL3-25 and VL3- 1 light chains with long HCDR3s, and that high affinity clones of this type exhibit a BG18-like binding mode, indicate that the pool of potential BG18 human naive precursors is larger and more diverse than originally expected. This study lays out a pathway for the development and pre-clinical validation of HCDR3 -dependent GT immunogens, a pathway that could potentially be expanded to other HIV epitopes or other vaccine targets.
Methods
BG18/BG505 SOSIP structural model
[00211] The HCDR3 loop of unliganded BG18 was aligned to PGT122 (PDB: 4TVP) and several features suggested this as a plausible binding mode. First, ArgL54 in the LCDR2 would be positioned in a similar space as the LCDR3 ArgL94 in PGT122, a known critical contact residue for PGT122. Second, ArgH29 was positioned close to the N332 glycan, and it was confirmed that ArgH29 was important for neutralization, by mutagenesis (Figure 4d-e). Finally, analysis of the V 1 conformation suggested that the LC could plausibly avoid clashing with the N 137 glycan.
Design of minimally mutated versions of BG18 and alternate VL variants
[00212] Design of minimally mutated versions of BG18 was guided by analysis ofthe structural model of BG18 bound to BG505 SOSIP (Figure 4a). Framework mutations and CDR mutations were reverted to germline if structural inspection indicated they were not contributing to the binding interaction. Several HC and LC variants were tested. BG18.11, which is referred to as "minBG18", was the least mutated variant that showed >50% breadth compared to BG18 and maintained similar potency as BG18. BG18.6 was the least mutated variant that showed any neutralization. To make the VL variants shown in Figure 5, the indicated VL gene was substituted for the BG18 VL gene and BG18 mutations were incorporated.
Immunogen design
[00213] BG18 1GL2 had detectable affinity to the l lmutB (PGT121 germline targeting) trimer but no detectable affinity to BG505 MD39 containing a native N332 epitope, therefore 1 ImutB was used as a base construct to begin the BG18 iGL GT design process. The following libraries were screened using a previously described mammalian cell surface display method11. Library 1 was an NNK codon scan of positions 294, 297, 298, 299, 300, 302, 304, 305, 326, 329, 330, 333, 386, 413, 414, 415, 416, 417, 419, 420. NNK codons were introduced into BG505-1 lmutB-gpl20 using the QuikChange Site-Directed Mutagensis Kit (Agilent). Library 1 was screened for binding to BG18 1GL2 and PGT121-GLcDR3rev4- Library 2 was a combinatorial library with amino acids F/I/L/V introduced at positions 154, 322, 323, 326, 333, 414, 415, and 416. The library insertwas assembled with overlapping ultramers (IDT DNA) followed by Gibson cloning (NEB) into BG505-1 lmutB- gpl20. Library 2 was screened for binding to BG18 1GL2 and PGT121-GLCDR3IEV4. Library 3 was a combinatorial library with NNK codons introduced at positions 137, 325, and (F/I/L/V) at position 326. The library insert was assembled with overlapping ultramers (IDT DNA) followed by Gibson cloning (NEB) into BG505-MD39-17mutE. Library 3 was screened for binding to the following 24 Abs: BG18 iGL0, BG18 iGLi, prel - pre6, pre8, prelO - pre l5, VL2-8, VL2-14, VL3-21, VHl-69, VH3-33, VH4-59, VH5-5 1, PGT121-GLcDR3mat, PGT121-GLcDR3revi. Library 4 was a combinatorial library with NNK codons introduced at positions 138 and 141 and (P/H/A/D) at position 325. The library insert was assembled with overlapping ultramers (IDT DNA) followed by Gibson cloning (NEB) into BG505-MD39-17mutE-N137K. Library 4 was screened for binding to BG18 iGLo, pre3, pre l4, VL3-21, VL2-8, VL2-14, VH3-33. Library 5 was a combinatorial library with NNK codons introduced at positions 138 and 139 and (P/H/A/D) at position 325. The library insert was assembled with overlapping ultramers (IDT DNA) followed by Gibson cloning (NEB) into BG505- MD39-17mutE-N137K. Library 5 was screened for binding to BG18 iGLo, VL3-21, VL2-8, VL2-14, VH1-69, VH5-5 1. Library 6 was an NNK scan of positions 167 to 308. The insert was synthesized at SGI-DNA and Gibson cloned (NEB) into BG505-MD39-N332-GT3. Library 6 was screened for binding to BG18 iGLo, VL2-8. Library 7 was an NNK scan of positions 309 to 443. The insert was synthesized at SGI-DNA and Gibson cloned (NEB) into BG505-MD39-N332-GT3. Library 7 was screened for binding to BG18 iGLo, pre l, pre2, pre4, prelO, prel5, VL2-8. All constructs contained a C-terminal myc tag and were anchored to the cell membrane via a C-terminal PDGLR transmembrane domain. Staining of the cell populations was typically done with IgG until saturated binding was obtained at low nanomolar IgG concentrations, and then Labs were used for staining to maintain selection pressure. Typically, libraries were sorted 3 to 5 times, and the enriched cell populations were frozen until sequencing could be carried out as described previously11. The most enriched clones were synthesized at Genscript either as C-terminal His tagged gpl20s or MD39/MD64 trimers in the pHLsec vector and expressed and purified as described previously11. Nanoparticle design and purification
[00214] To obtain multivalent immunogens, trimers were genetically fused to ferritin from Helicobacter pylori using a short flexible linker. Genes were codon optimized for HEK293 cells and cloned into the pHLsec plasmid (GenScript). MD39-NP DNA was co-transfected with a plasmid encoding human Turin protease into LreeStyle 293L cells (Invitrogen, Cat no. R79007) using 293Lectin (ThermoPisher) and proteins were expressed at 37°C for four days. NPs were purified either using snow drop lectin-conjugated agarose beads (Vector laboratories) or HiTrap NHS-Activated HP affinity columns (GE Healthcare) conjugated with PGT145 (kindly provided by the Dennis Burton lab), each followed by gel-filtration using a Superose 6 sixe-exclusion chromatography column (GE Healthcare). N332-GT2 NP has the Furin cleavage site removed and was not co-transfected with Furin. NP -assembly was assessed by negative-stain EM and SEC + multi-angle light scattering (SEC-MALS) using a Superose 6 10/300 column (GE Healthcare) at a flow rate of 0.5 mL/min followed by DAWN HELEOS II and Optilab T-rEX detectors (Wyatt Technology), correcting for the glycan molecular mass by applying the built-in protein-conjugate analysis (ASTRA).
Neutralization Activity
[00215] Neutralizing activity of monoclonal antibodies (mAbs) was assessed using single round of replication in TZM-bl target cells, in the absence of DEAE-dextran as described previously25. Briefly, pseudoviruses were generated by co-transfection of HEK293T cells with an Env- expressing plasmid and an Env-deficient genomic backbone plasmid (pSG3AEnv).
Cryo-EM structure determination [00216] Trimers were incubated with a 10X molar excess of Fab overnight at room temperature. The following morning, each complex was purified using a HiLoad 16/600 Superdex 200pg size exclusion column (GE Healthcare) with Tris-buffered saline (50 mM Tris pH 7.4, 150 mM NaCl) as the running buffer, and the peak corresponding to trimer-fab complex was pooled and concentrated to ~8 mg/mL. 18 pL of complex were mixed with 3 pL of 0.42 mM n-dodecyl b-D- maltoside (DDM; Anatrace), such that final DDM concentration (0.06 mM) is below the critical micellar concentration (CMC). A 3 pL aliquot of the complex was applied to a C-Flat grid (CF- 2/2-4C, Electron Microscopy Sciences, Protochips, Inc.) which had been plasma cleaned for 10 s using a mixture of Ar/CE (Gatan Solarus 950 Plasma system), and following a 10 s incubation, the grid was blotted between 4-5 s and plunged into liquid ethane using the FEI Vitrobot system (100% relative humidity, 10°C).
[00217] The samples were imaged using an FEI Titan Krios electron microscope (Thermo Fisher) operating at 300 kV and a Gatan K2 Summit direct electron director operating in counting mode. Automated data collection was performed using the Leginon software suite26. Each micrograph movie (250 ms exposure per frame) was collected at a magnification of 29,000, which resulted in a pixel size of 1.03 A in the specimen plane. Data collection statistics for each sample are summarized in Figure 9a. Micrograph movie frames were aligned and dose-weighted using MotionCor227, and CTF models were calculated using GCTF28.
[00218] Initial negative-stain EM attempts of HMP-GT5 Fab complexes suffered from severe preferred orientation, so samples were instead prepared for low resolution cryoEM in a similar fashion as above except that the size exclusion step was omitted to prevent antibody dissociation due to faster off-rates. The samples were imaged using an FEI Talos Arctica electron microscope (Thermo Fisher) operating at 200 kV and an FEI Ceta 16M CMOS camera (Thermo Fisher). Single frame exposures were collected with atotal dose of ~50 eVA2 at a magnification of 73,000, resulting in a pixel size of 1.98 A at the specimen plane. CTF models were calculated as above.
[00219] Single particles were selected using DoGPicker29 from the whole-frame aligned and summed micrographs, and particles extracted using Relion 2.130 using a box size of 288 pixels. After numerous rounds of 2D and 3D classification, final reconstructions were performed in Relion 2.1 with C3 symmetry imposed, and after post-processing, the final resolution estimates (FSC 0.143) are ~3.9 A for BG505-MD64-N332-GT2 in complex with BG18 iGL0 and ~4.4 A for BG505 -MD39 in complex with mature BG 18. Additional data processing statistics are summarized in Figure 9a. The low resolution cryoEM datasets of HMP complexes were processed in a similar manner except that the final combined map did not undergo post-processing and final resolution estimates are ~15 A for each complex.
[00220] Atomic models were built and refined into the high-resolution reconstructions by creating homology models based off deposited coordinates of BG505 SOSIP.664 (PDB 5cez) and 354BG18 fab (PDB 5ud9), followed by an iterative cycle of manual building in COOT31 and real space refinement in Phenix 1.1332. Glycans were validated by CARP33 and Privateer34, and overall structures were evaluated using EMRinger35 and Molprobity36.
ELISA
[00221] N332-GT2-specific antibody titers were detected by ELISA, using anti-His Ab (2 pg/ml) to capture N332-GT2 or N332-GT2-KO antigen (2 pg/ml) on the plate. Mouse sera were incubated for two hours and alkaline phosphatase conjugated anti-mouse IgG (Jackson ImmunoResearch, #115-055-071) was incubate another hour. Titers were determined from the dilution curve in the linear range of absorbance. All non-commercial ELISA plates were developed with p-Nitrophenyl Phosphate (Sigma, # N2770). Absorbance at 405 nm was determined with a plate reader (BioTek).
Surface plasmon resonance (SPR)
[00222] Kinetics and affinities of antibody/antigen interactions were measured on a ProteOn XPR36 (Bio-Rad) using GLC Sensor Chip (Bio-Rad) or Biacore4000 (GE) with Series S Sensor Chip CM5 (GE). lx HBS-EP+ pH 7.4 running buffer (20x stock from Teknova, Cat. No H8022) supplemented with BSA at lmg/ml was used. Following manufacturer’s instructions for Human Antibody Capture Kit (Cat. No BR-1008-39 from GE) about six thousand RUs of capture mAb were immobilized onto each flow cell of GLC Sensor Chip or about ten thousand RUs in case of CM5 Sensor Chip. In atypical experiment on ProteOn XPR36 system, approximately 300-400 RUs of mAbs were captured onto each flow cell and analytes were passed over the flow cell at 50 pL/min for 3 min followed by a 5 min dissociation time. Regeneration was accomplished using 3M Magnesium Chloride with 180 seconds contact time and injected four times per cycle. Raw sensograms were analyzed using ProteOn Manager software (Bio-Rad), including interspot and column double referencing, and either Equilibrium fits or Kinetic fits with Langmuir model, or both, were employed when applicable. For Biacore4000 instrument similar conditions were used but lower ligand capture levels. In the case of Fab-fragment/antigen kinetic and affinity measurements on ProteOn XPR36 or Biacore4000 similar ligand-capture technique with several modifications was used. The capture reagent was His-tag Rabbit pAb (GenScript Cat. No. A00174). It was amine coupled to Sensor Chip surface using the same protocol from GE Human Antibody Capture Kit referenced above. Our regeneration solution was Phosphoric Acid 0.85% with 30 seconds contact time, four injections per cycle. In the case of ferritin nanoparticle experiment ProteOn XPR36 system and Human Antibody Capture protocol described above was used with one additional step. PGT128 IgG was captured at 1300 RU level in all channels including reference followed by NP (as ligand) capture at 1600 RU. All other steps were the same as in Human Antibody Capture protocol. Analyte concentrations were measured on a NanoDrop 2000c Spectrophotometer using Absorption signal at 280 nm3.
NGS dataset of human BCR HCs
[00223] This work utilized a large NGS dataset of 1.1 c 109 unique amino acid sequences of BCR HCs from 14 healthy, HIV-uninfected donors. In this dataset, 255 million sequences from 10 donors were obtained from ref.18 which used the HiSeq sequencing platform and an amplification strategy including unique identifiers (UIDs) to enable discrimination of unique mRNA transcripts from PCR artifacts. These sequences were collapsed by UID, assigned to VDJ gene segments with Abstar18, and then rendered unique by clustering at the 99% amino acid identity level within each of six biological replicates per donor. JSON output files from Abstar were converted to parquet format and uploaded to the Amazon S3 storage cloud. To query databases, Amazon Elastic Map Reduce (EMR) 5.15.5 was used to configure a Spark cluster, and PySpark was used to query the database with custom Python scripts. Sequences for 4 additional donors were obtained here by both HiSeq and NextSeq sequencing platforms without the use of UIDs, as described below.
BCR HC sequencing for 4 donors
[00224] Full leukopaks (3 blood volumes) were obtained from four human subjects (AllCells LLC or Hemacare, Inc) under a protocol approved by the Institutional Review Board of the respective commercial provider. All subjects were healthy, HIV-negative adults with no reported acute illness in the 14 days prior to leukapheresis, and samples were de-identified prior to shipment. The Institutional Review Board of The Scripps Research Institute determined that research with these samples did not constitute human subjects research. Immediately upon receipt of the leukopak, peripheral blood mononuclear cells (PBMCs) were purified by gradient centrifugation and cryopreserved in aliquots of approximately 5xl08 PBMCs. The junctional regions of antibody heavy chain libraries were amplified as in Willis et ah, 201537. SPRI-purified sequencing libraries were initially quantified using fluorometry (Qubit, Thermo Fisher Scientific) before size determination using a bioanalyzer (Agilent 2100). Libraries were re-quantified using qPCR (KAPA Biosystems) before sequencing on either an Illumina HiSeq (2xl50bp chemistry) or NextSeq (2xl50bp chemistry). Sequences were merged with PANDAseq using the default (symple_bayesian) merging algorithm before annotation with Abstar18.
BG18 precursor frequency estimate
[00225] The NGS dataset of human BCR HCs was queried by bioinformatic searches to gain information on the frequency of BG18-like HCDR3s in the human B cell repertoire ( Figure 6). HCDR3s meeting the definition of BG18-like feature set i in Figure 6a, constituting a broad set of potential BG18-like HCDR3 precursors, were identified in 14 of 14 donors (Figure 6c). The geomean frequency was 1 in 58,000 among the 10 donors sequenced by Briney et al.18 using UIDs. To refine this frequency estimate, it was considered that only 11 of 14 BG18 iGL variants with NGS-derived HCDR3s differing in the HCDR3 junctions (Figure 6b) exhibited binding to N332- GT2 (Figure le). BG18-like feature set ii (HCDR3 junction features) in Figure 6a characterized amino acids present in the non-templated junction regions of BG18 and its somatic variants15 and in the 11 precursors that bound to N332-GT2. The frequency of HCDR3s meeting the definitions for both feature sets i and ii was found to be lower than those within set i by a factor of 104. Because the VL gene plays an important role In the BG18 VI -loop straddling binding mode, VL gene usage was incorporated into the frequency estimate, as feature set iii (“VL gene family”). The conservative assumption was that only VL3 LCS can support the BG18-class binding mode, as all VL3 LCS tested bound with high affinity to N332-GT2. The frequency of all VL3-derived Abs in the HC-LC paired sequences in DeKosky et al38 was 1 in 9 (13845 VL3S in 127701 sequences). It was also assumed that any VH gene can support this binding mode, because when five of the most common human VH genes were substituted into BG18 iGLi, all five variants showed low nanomolar binding to N332-GT2 (Figure le). Therefore no frequency factorwas imposed for VH gene usage. Multiplying the frequencies of all three feature sets together gave our best estimate for the frequency in the human B cell repertoire of BG18-like precursors that could be targeted by N332-GT2: 1 in 54 million.
N332-GT specific naive human B cell sorting and BCR sequencing
[00226] LRS (leukoreduction) tubes were obtained from the San Diego Blood Bank from healthy, HIV-seronegative human donors. These studies do not constitute human subjects research, as determined by the Institutional Review Boards of both La Jolla Institute and The Scripps Research Institute. More than 1 billion peripheral blood mononuclear cells were regularly recovered. CD19+ B cells were isolated using a positive selection magnetic bead separation kit (Miltenyi Biotec) and resuspended in complete RPMI media with 10% FBS.
[00227] Avi-tagged protein immunogens were biotinylated using the Bulk BirA kit (Avidity, LLC). N332-GT2 and N332-GT2KO probes were used in N332-GT2 sorting experiments. N332- GT1 and MD39 probes were used in N332-GT1 sorting experiments. l lmutB and MD39 probes were used in l lmutB sorting experiments. Biotinylated protein immunogens were individually premixed with fluorescently labeled streptavidin to form tetramer probes. Multiple tactics were used to avoid false positives: 1) used two“positive” probes, 2) each“positive” probe used a different protein tag (His-tag or Strep-tag) to avoid tag specific B cells, 3) used a“negative” probe to identify N332-epitope specific B cells, 4) choose independent (no tandems) fluorochromes for all probes to avoid fluorochrome specific B cells. For example, N332-GT2 sorting experiments used the follow probes: N332-GT2-Histag-biotin + streptavidin Alexa Fluor 647, N332-GT2- streptag-biotin + streptavidin Brilliant Violet 421, and N332-GT2KO biotin + streptavidin phycoerythrin.
[00228] Cells were incubated with N332-GT probes for 20 min at 4°C. Without washing, anti- CD^ (PE-Cy7,ThermoFisher, clone HIB19) and anti-CD20 (PE-Cy7, ThermoFisher, clone 2H7), in addition to anti-IgG (APC-Cy7, Biolegend, clone HP6017), anti-CD3 (APC eFluor®780, ThermoFisher, clone UCHT1), anti-CD 14 (APC eFluor®780, ThermoFisher, clone 61D3), anti- CD16 (APC eFluor®780, ThermoFisher, clone eBioCB 16), and Live/Dead (APC eFluor®780, ThermoFisher) for exclusion, were added for an additional 20 min. A BD FACSAria was used for all cell sorting. Cells were sorted at a flow rate of 1500 events/second using an 85 -pm nozzle. Sorting stringency was set to a strict setting to obtain one cell per well. Single B cells were sorted directly into cold lysis buffer or N332-specific clonal B cell lines were generated and interrogated as in4. cDNA synthesis, nested BCR PCR, Sanger sequencing and sequence analysis was carried out as in10. Sorting was done with FACSDiva (BD) software and post-sort analyses were done with FlowJo (FlowJo, LLC).
Adoptive transfer experiments and immunization
[00229] For adoptive transfer experiments, B cells were isolated from CD45.2 C57BL/6J ("WT") or Bϋΐd®41 KI mice of 8-10 weeks of age, and cells were resuspended in 150 pi of PBS and injected i.v. into CD45.1 B6. SJL-Ptprca Pepcb/BoyJ recipient animals (5 x 103 cells per mouse for BG 18"H transfers and 50 x 103 cells per mouse for C57BL/6 transfers). One day later, recipient mice were injected i.p. with 10 pg GT2- or MD39-NPs with Sigma adjuvant (Sigma, # S6322 SIGMA). After eight days, mice were sacrificed to harvest spleen samples. Blood samples were taken from the submandibular vein on day 0 and 14 after immunization. Four immunization conditions were tested in two independent experiments, with the following total number of mice in each condition: (1) BG 18sH B cell transfer, N332-GT2 NP immunization (N=6 for day 8 GC analysis, N=5 for day 14 ELISA); (2) BG18gH B cell transfer, MD39 NP immunization (N=3 for day 8 GC analysis, N=3 for day 14 ELISA); (3) WT B cell transfer, N332-GT2 NP immunization (N=5 for day 8 GC analysis, N=5 for day 14 ELISA); (4) WT B cell transfer, MD39 NP immunization (N=3 for day 8 GC analysis, N=3 for day 14 ELISA).
Antigen specific single cell sorting for BCR sequencing in mouse experiments
[00230] Antigen tetramers were prepared by conjugating for 1 hour (room temp.) biotinylated N332-GT2 and N332-GT2-KO trimers with fluorescently labeled streptavidins (Alexa Fluor 488, Alexa Fluor 647, eBioscience; Alexa Fluor 568, Thermo Fisher Scientific) in a 4: 1 molar ratio. The same streptavidins conjugated with biotinylated Fab anti-IgM and biotinylated BSA were used as positive and negative staining controls, respectively (data not shown). Single cell suspensions generated from spleen samples were depleted of red blood cells by ACK lysis, Fc blocked (BD Biosciences) and stained in FACS buffer (2% FCS/PBS) with antigen tetramers for 30 min at 4C, 50 nM concentration. Next, a cocktail of mAbs was added for 30 minutes at 4C. For staining of splenocytes from naive mice (Figure 3a), the cocktail was: B220 PerCP-Cy5.5 (Clone RA3-6B2, Biolegend), IgD PE-Cy7 (Clone l l-26c.2a, Biolegend), CD4 APC-eFluor780 (Clone RM4-5, eBioscience), CD8a APC-eFluor780 (Clone 53-6.7, eBioscience), F4/80 APC-eFluor780 (Clone BM8, eBioscience), Ly-6G APC-eFluor780 (Clone RB6-8C5, eBioscience). For staining of splenocytes from immunized mice (Fig 3d), the cocktail was: CD38 Alexa Fluor 700 (Clone 90, Invitrogen), CD45.2 PE (Clone 104, Biolegend), CD45.1 PerCP-Cy5.5 (Clone A20, Biolegend), B220 PB (Clone RA3-6B2, Biolegend), CD95 PE-Cy7 (Clone Jo2, BD Bioscience), CD4 APC- eFluor780 (Clone RM4-5, eBioscience), CD8a APC-eFluor780 (Clone 53-6.7, eBioscience), F4/80 APC-eFluor780 (Clone BM8, eBioscience), Ly-6G APC-eFluor780 (Clone RB6-8C5, eBioscience). Live-Dead staining kits (Thermo Scientific) were used to identify dead cells for exclusion from the analysis. Data acquisition and single cell sorting were performed on FACS ARIA II (BD Bioscience) and analyzed with Flowlo v. 10 (Tree Star). Single cell sorting and single cell PCR was carried out as described previously24. [00231] While the invention has been described in connection with what is presently considered to be the most practical and preferred embodiments, it is to be understood that the invention is not to be limited to the disclosed embodiments, but on the contrary, is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.
[00232] All publications, patents, patent applications, internet sites, and accession numbers/database sequences including both polynucleotide and polypeptide sequences cited herein are hereby incorporated by reference herein in their entirety for all purposes to the same extent as if each individual publication, patent, patent application, internet site, or accession number/database sequence were specifically and individually indicated to be so incorporated by reference.
References
1 Jardine, J. et al. Rational HIV immunogen design to target specific germline B cell
receptors. Science 340, 711-716, doi: 10.1126/science.1234150 (2013).
2 McGuire, A. T. et al. Engineering HIV envelope protein to activate germline B cell receptors of broadly neutralizing anti-CD4 binding site antibodies. J Exp Med 210, 655- 663, doi: 10.1084/jem.20122824 (2013).
3 Jardine, J. G. et al. HIV-1 VACCINES. Priming a broadly neutralizing antibody response to HIV-1 using a germline -targeting immunogen. Science 349, 156-161,
doi: 10.1126/science. aac5894 (2015).
4 Jardine, J. G. et al. HIV-1 broadly neutralizing antibody precursor B cells revealed by germline-targeting immunogen. Science 351, 1458-1463, doi: 10.1126/science. aad9195 (2016).
5 McGuire, A. T. et al. Specifically modified Env immunogens activate B-cell precursors of broadly neutralizing HIV-1 antibodies in transgenic mice. Nat Commun 7, 10618, doi : 10.1038/ncomms 10618 (2016).
6 Briney, B. et al. Tailored Immunogens Direct Affinity Maturation toward HIV
Neutralizing Antibodies. Cell 166, 1459-1470 el411, doi: 10.1016/j.cell.2016.08.005 (2016).
7 Tian, M. et al. Induction of HIV Neutralizing Antibody Lineages in Mice with Diverse Precursor Repertoires. Cell 166, 1471-1484 e l418, doi: 10.1016/j .cell.2016.07.029 (2016).
8 Sok, D. et al. Priming HIV-1 broadly neutralizing antibody precursors in human Ig loci transgenic mice. Science 353, 1557-1560, doi: 10.1126/science. aah3945 (2016).
9 Medina-Ramirez, M. et al. Design and crystal structure of a native-like HIV-1 envelope trimer that engages multiple broadly neutralizing antibody precursors in vivo. J Exp Med 214, 2573-2590, doi: 10.1084/jem.20161160 (2017).
10 Havenar-Daughton, C. et al. The human naive B cell repertoire contains distinct
subclasses for a germline-targeting HIV-1 vaccine immunogen. Sci Transl Med 10, doi : 10.1126/scitranslmed.aat0381 (2018). Steichen, J. M. et al. HIV Vaccine Design to Target Germline Precursors of Glycan- Dependent Broadly Neutralizing Antibodies. Immunity 45, 483-496,
doi: 10.1016/j.immuni.2016.08.016 (2016).
Julien, J. P. et al. Broadly neutralizing antibody PGT121 allosterically modulates CD4 binding via recognition of the HIV-1 gpl20 V3 base and multiple surrounding glycans. PLoS pathogens 9, el003342, doi: 10.1371/joumal.ppat.1003342 (2013).
Julien, J. P. et al. Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science 342, 1477-1483, doi: 10.1126/science.1245625 (2013).
Escolano, A. et al. Sequential Immunization Elicits Broadly Neutralizing Anti-HIV-1 Antibodies in Ig Knockin Mice. Cell 166, 1445-1458 el412,
doi: 10.1016/j.cell.2016.07.030 (2016).
Freund, N. T. et al. Coexistence of potent HIV-1 broadly neutralizing antibodies and antibody-sensitive viruses in a viremic controller. Sci Transl Med 9,
doi : 10.1126/scitranslmed.aal2144 (2017) .
Pancera, M. et al. Structure and immune recognition of trimeric pre -fusion HIV-1 Env. Nature 514, 455-461, doi: 10.1038/naturel3808 (2014).
Bames, C. O. et al. Structural characterization of a highly-potent V3-glycan broadly neutralizing antibody bound to natively-glycosylated HIV-1 envelope. Nat Commun 9, 1251, doi: 10.1038/s41467-018-03632-y (2018).
Briney, B. I., A.; Joyce, C.; Burton, D. R. Uniqueness, commonality and exceptional diversity in the baseline human antibody repertoire (submitted)
Kulp, D. W . et al. Structure-based design of native-like HIV-1 envelope trimers to silence non-neutralizing epitopes and eliminate CD4 binding. Nat Commun 8, 1655, doi: 10.1038/s41467-017-01549-6 (2017).
Abbott, R. K. et al. Precursor Frequency and Affinity Determine B Cell Competitive Fitness in Germinal Centers, Tested with Germline-Targeting HIV Vaccine Immunogens. Immunity 48, 133-146 el36, doi: 10.1016/j.immuni.2017.11.023 (2018).
Tokatlian, T. et al. Innate immune recognition of glycans targets nanoparticle immunogens to germinal centers (submitted).
Kanekiyo, M. et al. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature 499, 102-106, doi: 10.1038/nature 12202 (2013). Sliepen, K. et al. Presenting native-like HIV-1 envelope trimers on ferritin nanoparticles improves their immunogenicity. Retrovirology 12, 82, doi: 10.1186/sl2977-015-0210-4 (2015).
Lin, Y.-C. P., S.; Steichen, J. M.; Kratochvil, S.; Melzi, E.; Arnold, J., Dougan, S. K.;
Wu, L.; Hirsch, K. H.; Nair, U.; Schief, W. R.; Batista, F. D. . One-step CRISPR Cas9 method for the rapid generation of human antibody heavy chain knock-in mice. The EMBO Journal, doi: 10.15252 (2018).
Landais, E. et al. Broadly Neutralizing Antibody Responses in a Large Longitudinal Sub- Saharan HIV Primary Infection Cohort. PLoS pathogens 12, el005369,
doi : 10.1371/j oumal .ppat.1005369 (2016).
Suloway, C. et al. Automated molecular microscopy: the new Leginon system. Journal of structural biology 151, 41-60, doi: 10.1016/j.jsb.2005.03.010 (2005).
Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat Methods 14, 331-332, doi: 10.1038/nmeth.4193 (2017).
Zhang, K. Gctf: Real-time CTF determination and correction. Journal of structural biology 193, 1-12, doi: 10.1016/j jsb.2015.11.003 (2016). 29 Voss, N. R., Yoshioka, C. K., Radermacher, M., Potter, C. S. & Carragher, B. DoG Picker and TiltPicker: software tools to facilitate particle selection in single particle electron microscopy. Journal of structural biology 166, 205-213 (2009).
30 Kimanius, D., Forsberg, B. O., Scheres, S. H. & Lindahl, E. Accelerated cryo-EM
structure determination with parallelisation using GPUs in RELION-2. Elife 5, doi: 10.7554/eLife.18722 (2016).
31 Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot.
Acta Crystallogr D Biol Crystallogr 66, 486-501, doi: 10.1107/S0907444910007493 (2010).
32 Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr D Biol Crystallogr 66, 213-221,
doi: 10.1107/S0907444909052925 (2010).
33 Lutteke, T., Frank, M. & von der Lieth, C. W. Carbohydrate Structure Suite (CSS): analysis of carbohydrate 3D structures derived from the PDB. Nucleic Acids Res 33, D242-246, doi: 10.1093/nar/gki013 (2005).
34 Winn, M. D. et al. Overview of the CCP4 suite and current developments. Acta
Crystallogr D Biol Crystallogr 67, 235-242, doi: 10.1107/S0907444910045749 (2011).
35 Barad, B. A . et al. EMRinger: side chain-directed model and map validation for 3D cryo- electron microscopy. Nat Methods 12, 943-946, doi: 10.1038/nmeth.3541 (2015).
36 Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular
crystallography. Acta Crystallogr D Biol Crystallogr 66, 12-21,
doi: 10.1107/S0907444909042073 (2010).
37 Willis, J. R. et al. Redesigned HIV antibodies exhibit enhanced neutralizing potency and breadth. JClin Invest 125, 2523-2531, doi: 10.1172/JCI80693 (2015).
38 DeKosky, B. J . et al. High-throughput sequencing of the paired human immunoglobulin heavy and light chain repertoire. Nat Biotechnol 31, 166-169, doi: 10.1038/nbt.2492 (2013).
39 Jardine, J. G. et al. Minimally Mutated HIV-1 Broadly Neutralizing Antibodies to Guide Reductionist Vaccine Design. PLoS pathogens 12, el005815,
doi : 10.1371 /j oumal .ppat.1005815 (2016) .
SEQUENCES
SEQ ID NO: 1; HXB2 Env
MRVKEKYQHLWRWGWRWGTMLLGMLMICSATEKLWVTVYYGVPVWKEATTTLFCASDAK AYDTEVHNVWATHACVPTDPNPQEVVLVNVTENFNMWKNDMVEQMHEDI ISLWDQSLKP CVKLTPLCVSLKCTDLKNDTNTNSSSGRMIMEKGEIKNCSFNISTS IRGKVQKEYAFFY KLDI IPIDNDTTSYKLTSCNTSVITQACPKVSFEPIPIHYCAPAGFAILKCNNKTFNGT GPCTNVSTVQCTHGIRPWSTQLLLNGSLAEEEWIRSVNFTDNAKTI IVQLNTSVEIN CTRPNNNTRKRIRIQRGPGRAFVTIGKIGNMRQAHCNISRAKWNNTLKQIASKLREQFG NNKTI I FKQSSGGDPEIVTHSFNCGGEFFYCNSTQLFNSTWFNSTWSTEGSNNTEGSDT ITLPCRIKQI INMWQKVGKAMYAPPISGQIRCSSNITGLLLTRDGGNSNNESEI FRPGG GDMRDNWRSELYKYKWKIEPLGVAPTKAKRRWQREKRAVGIGALFLGFLGAAGSTMG AASMTLTVQARQLLSGIVQQQNNLLRAIEAQQHLLQLTVWGIKQLQARILAVERYLKDQ QLLGIWGCSGKLICTTAVPWNASWSNKSLEQIWNHTTWMEWDREINNYTSLIHSLIEES QNQQEKNEQELLELDKWASLWNWFNITNWLWYIKLFIMIVGGLVGLRIVFAVLS IVNRV RQGYSPLSFQTHLPTPRGPDRPEGIEEEGGERDRDRS IRLVNGSLALIWDDLRSLCLFS YHRLRDLLLIVTRIVELLGRRGWEALKYWWNLLQYWSQELKNSAVSLLNATAIAVAEGT DRVIEWQGACRAIRHIPRRIRQGLERILL
SEQ ID NO: 2; BG505 Env
MRVMGIQRNCQHLFRWGTMILGM111CSAAENLWVTVYYGVPVWKDAETTLFCASDAKA YETEKHNVWATHACVPTDPNPQEIHLENVTEEFNMWKNNMVEQMHTDI ISLWDQSLKPC VKLTPLCVTLQCTNVTNNITDDMRGELKNCSFNMTTELRDKKQKVYSLFYRLDWQINE NQGNRSNNSNKEYRLINCNTSAITQACPKVSFEPIPIHYCAPAGFAILKCKDKKFNGTG PCPSVSTVQCTHGIKPWSTQLLLNGSLAEEEVMIRSENITNNAKNILVQFNTPVQINC TRPNNNTRKS IRIGPGQAFYATGDI IGDIRQAHCTVSKATWNETLGKVVKQLRKHFGNN TI IRFANSSGGDLEVTTHSFNCGGEFFYCNTSGLFNSTWISNTSVQGSNSTGSNDS ITL PCRIKQI INMWQRIGQAMYAPPIQGVIRCVSNITGLILTRDGGSTNSTTETFRPGGGDM RDNWRSELYKYKWKIEPLGVAPTRAKRRWGREKRAVGIGAVFLGFLGAAGSTMGAAS MTLTVQARNLLSGIVQQQSNLLRAIEAQQHLLKLTVWGIKQLQARVLAVERYLRDQQLL GIWGCSGKLICTTNVPWNSSWSNRNLSEIWDNMTWLQWDKEISNYTQI IYGLLEESQNQ QEKNEQDLLALDKWASLWNWFDISNWLWYIKI FIMIVGGLIGLRIVFAVLSVIHRVRQG YSPLSFQTHTPNPRGLDRPERIEEEDGEQDRGRSTRLVSGFLALAWDDLRSLCLFCYHR LRDFILIAARIVELLGHSSLKGLRLGWEGLKYLWNLLAYWGRELKISAINLFDTIAIAV AEWTDRVIEIGQRLCRAFLHIPRRIRQGLERALL
SEQ ID NO: 3; BG505 MD39
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNVTNNITDDMRGELKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKS IRIGPGQAFYYTGDI IGDI RQAHCNVSKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSITLPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 4; BG505 MD39 llmutB
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI I SLWDQSLKPCVKLTPLCVTLQCTNYAPNLLSNMRGELKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKS IRIGPGQAFYYFGDI IGDI RMAHCNVSKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSIVLPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 5; BG505 MD39 17mutE
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPNLLSNMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKSIRIGPGQAFYYFGDVLGDV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 6; BG505 MD39 N332 GT1
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPKLLSNMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKSIRIGPGQAFYYFGDVLGHV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 7; BG505 MD39 N332 GT2
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPKLRSMMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKSIRIGPGQAFYYFGDVLGDV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 8; BG505 MD39 N332 GT3 AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPKLRSMMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKSIRIGPGQAFYYFGDVLGHV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 9; BG505 MD39 N332 GT4
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPKLRSMMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPSNNTVKSIRIGPGQAFYYFGDVLGHV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 10; BG505 MD39 N332 GT5
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPKLRSMMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPSNNTVKSIRIGPGQAFYYFGDVLGHV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCWIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 11; BG505 MD39 N332 GT2 KO
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPNLRSDMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKSIRIGPGQAFYYFGDVLGDV DMAKCTISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 12; BG505 MD64 N332 GT2
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHECVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHTDI IELWDQSLKPCVKLTPLCVTLQCTNYAPKLRSMMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKSIRIGPGQAFYYFGDVLGDV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKLTVWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 13; N332-GT2 NP (ferritin)
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPKLRSMMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKSIRIGPGQAFYYFGDVLGDV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGSHSGSGGSGSGGHAAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLPSGIVQQQS NLLRAPEPQQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNS SWSNRNLSEIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALDGSGGLSE RMLKALNDQLNRELYSAYLYFAMAAYFEDLGLEGFANWMKAQAEEEIGHALRFYNYIYD KNGRVELDEIPKPPKEWESPLKAFEAAYEHEKFISKSIYELAALAEEEKDYSTRAFLEW FINEQVEEEASVKKILDKLKFAKDSPQILEMLDKELSARAPKLPGLLMQGGE
SEQ ID NO: 14; MD39 NP (ferritin)
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNVTNNITDDMRGELKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPNNNTVKSIRIGPGQAFYYTGDI IGDI RQAHCNVSKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSITLPCRIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALDGSGGLSERMLKALND QLNRELYSAYLYFAMAAYFEDLGLEGFANWMKAQAEEEIGHALRFYNYIYDKNGRVELD EIPKPPKEWESPLKAFEAAYEHEKFISKSIYELAALAEEEKDYSTRAFLEWFINEQVEE EASVKKILDKLKFAKDSPQILEMLDKELSARAPKLPGLLMQGGE
SEQ ID NO: 15; N332-GT5 NP (ferritin 3bve)
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPKLRSMMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCPSVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTPVQINCTRPSNNTVKSIRIGPGQAFYYFGDVLGHV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCWIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRE WGRRRRRRAVGIGAVSLGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALDGSGGLSKDI IKLLNE QVNKEMQSSNLYMSMSSWCYTHSLDGAGLFLFDHAAEEYEHAKKLI IFLNENNVPVQLT SISAPEHKFEGLTQIFQKAYEHEQHISESINNIVDHAIKSKDHATFNFLQWYVAEQHEE EVLFKDILDKIELIGNENHGLYLADQYVKGIAKSRKS
SEQ ID NO: 16; BG505 MD65 congly N332-GT5
AENLWVTVYYGVPVWKDAETTLFCASDAKAYETEKHNVWATHACVPTDPNPQEIHLENV TEEFNMWKNNMVEQMHEDI ISLWDQSLKPCVKLTPLCVTLQCTNYAPKLRSMMRGEIKN CSFNMTTELRDKKQKVYSLFYRLDWQINENQGNRSNNSNKEYRLINCNTSAITQACPK VSFEPIPIHYCAPAGFAILKCKDKKFNGTGPCQNVSTVQCTHGIKPWSTQLLLNGSLA EEEVI IRSENITNNAKNILVQLNTSVQINCTRPSNNTVKSIRIGPGQAFYYFGDVLGHV RMAHCNISKATWNETLGKWKQLRKHFGNNTIIRFAQSSGGDLEVTTHSFNCGGEFFYC NTSGLFNSTWISNTSVQGSNSTGSNDSLILPCWIKQI INMWQRIGQAMYAPPIQGVIRC VSNITGLILTRDGGSTNSTTETFRPGGGDMRDNWRSELYKYKWKIEPLGVAPTRCKRR TVGRRRRRRAAGIGASSDGFLGAAGSTMGAASMTLTVQARNLLSGIVQQQSNLLRAPEP QQHLLKDTHWGIKQLQARVLAVEHYLRDQQLLGIWGCSGKLICCTNVPWNSSWSNRNLS EIWDNMTWLQWDKEISNYTQI IYGLLEESQNQQEKNEQDLLALD
SEQ ID NO: 17; VH BG18
QVQLRESGPGLVKPSETLSLSCTVSNDSRPSDHSWTWVRQSPGKALEWIGDIHYNGATT YNPSLRSRVRIELDQSIPRFSLKMTSMTAADTGMYYCARNAIRIYGWALGEWFHYGMD VWGQGTAVTVSS
SEQ ID NO: 18; VL BG18
SSELTQPPSVSVSPGQTARITCSGAPLTSRFTYWYRQKPGQAPVLI ISRSSQRSSGWSG RFSASWSGTTVTLTIRGVQADDEADYYCQSSDTSDSYKMFGGGTKLTVL
SEQ ID NO: 19; VH BG18.11 QVQLQESGPGLVKPSGTLSLTCAVSNDSRPSDHSWSWVRQPPGKGLEWIGDIHYSGSTT YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARNAIRIYGVVALGEWFHYGMD VWGQGTTVTVSS
SEQ ID NO: 20; VL BG18.11
SYELTQPPSVSVSPGQTARITCSGDALPSRYAYWYQQKPGQAPVLVISRDSQRSSGISG
RFSGSWSGTTVTLTISGVQAEDEADYYCQSSDSSDTYKVFGGGTKLTVL
SEQ ID NO: 21; VH BG18.6
QVQLQESGPGLVKPSGTLSLTCAVSGDSRPSDHSWSWVRQPPGKGLEWIGEIYYSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARNAIRIYGVVALGEWFHYGMD VWGQGTTVTVSS
SEQ ID NO: 22; VL BG18.6
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYRDSQRPSGISG
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSDTYKVFGGGTKLTVL
SEQ ID NO: 23; VH BG18 iGLO
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARNAITIFGWI IGEYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 24; VL BG18 iGLO
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 25; VH BG18 iGLl
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARNAITIFGWILGEYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 26; VL BG18 iGLl (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 27; VH BG18 iGL2
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARNAITIFGWALGEYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 28; VL BG18 iGL2 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 29; VH VH4-59 QVQLQESGPGLVKPSETLSLTCTVSGGS ISSYYWSWIRQPPGKGLEWIGYIYYSGSTNY NPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARNAITI FGWALGEYYYYGMDV WGQGTTVTVSS
SEQ ID NO: 30; VH VH1-69
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMGGI IPIFGTAN YAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARNAITI FGWALGEYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 31; VH VH5-51
EVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMGI IYPGDSDTR YSPSFQGQVTISADKS ISTAYLQWSSLKASDTAMYYCARNAITI FGWALGEYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 32; VH VH3-33
QVQLVESGGGWQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVAVIWYDGSNKY YADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARNAITI FGWALGEYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 33; VH VH3-23
EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSAISGSGGSTY YADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKNAITI FGWALGEYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 34; VL VL3-19
SSELTQDPAVSVALGQTVRITCQGDSLRSYYASWYQQKPGQAPVLVIYGKNNRPSGIPD
RFSGSSSGNTASLTITGAQAEDEADYYCNSRDSSGNHWVFGGGTKLTVL
SEQ ID NO: 35; VL VL3-10
SYELTQPPSVSVSPGQTARITCSGDALPKKYAYWYQQKSGQAPVLVIYKDSKRPSGIPE RFSGSSSGTMATLTISGAQVEDEDDYYCYSADYSGNHWVFGGGTKLTVL
SEQ ID NO: 36; VL VL3-1
SYELTQPPSVSVSPGQTASITCSGDKLGDKYACWYQQKPGQSPVLVIYQDSKRPSGIPE
RFSGSNSGNTATLTISGTQAMDEADYYCQAWDSSTAWVFGGGTKLTVL
SEQ ID NO: 37; VL VL3-21
SYVLTQPPSVSVAPGQTARITCGGNNIGSKSVHWYQQKPGQAPVLWYDDSDRPSGIPE
RFSGSNSGNTATLTISRVEAGDEADYYCQVWDSSSDHWVFGGGTKLTVL
SEQ ID NO: 38; VL VL2-8
QSALTQPPSASGSPGQSVTISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYEVSKRPSG
VPDRFSGSKSGNTASLTVSGLQAEDEADYYCSSYAGSNNFWVFGGGTKLTVL
SEQ ID NO: 39; VH PRE1 QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARGQRTIFGVVILFNYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 40; VL PRE1 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 41; VH PRE2
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN
YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARDLLTSFGWIFSQHKSYGMD
VWGQGTTVTVSS
SEQ ID NO: 42; VL PRE2 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 43; VH PRE3
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARDSITIFGWILQDYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 44; VL PRE3 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 45; VH PRE4
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARERDTIFGWSLRVYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 46; VL PRE4 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 47; VH PRE5
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARDKDTIFGWI IGEYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 48; VL PRE5 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 49; VH PRE6 QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARGSITIFGWI ILEDYYYGMD VWGQGTTVTVSS
SEQ ID NO: 50; VL PRE6 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 51; VH PRE7
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCAREQRAIFGWI IREDYYYGMD VWGQGTTVTVSS
SEQ ID NO: 52; VL PRE7 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 53; VH PRE8
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARARLRVFGWIFLYGYYYGMD VWGQGTTVTVSS
SEQ ID NO: 54; VL PRE8 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 55; VH PRE9
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARDRKRFFGWIMDESYYYGMD VWGQGTTVTVSS
SEQ ID NO: 56; VL PRE9 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 57; VH PRE10
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARDGVKIFGWI IREAYYYGMD VWGQGTTVTVSS
SEQ ID NO: 58; VL PRE10 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 59; VH PRE11 QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN
YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARDGIKIFGWIMGEAHYYGMD
VWGQGTTVTVSS
SEQ ID NO: 60; VL PRE11 (Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 61; VH PRE12
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN
YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCAKGRGTIFGWRFYENYYYGMD
VWGQGTTVTVSS
SEQ ID NO: 62; VL PRE12(Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 63; VH PRE13
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN
YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCAKALNTIFGWTLTQYYYYGMD
VWGQGTTVTVSS
SEQ ID NO: 64; VL PRE13(Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 65; VH PRE14
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARGRITIFGWTLRGYYYYGMD VWGQGTTVTVSS
SEQ ID NO: 66; VL PRE14(Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 67; VH PRE15
QVQLQESGPGLVKPSGTLSLTCAVSGGSISSSNWWSWVRQPPGKGLEWIGEIYHSGSTN YNPSLKSRVTISVDKSKNQFSLKLSSVTAADTAVYYCARDIHNSFGWSLDQNYYYGMD VWGQGTTVTVSS
SEQ ID NO: 68; VL PRE15(Same as VL BG18 iGLO)
SYELTQPPSVSVSPGQTARITCSGDALPKQYAYWYQQKPGQAPVLVIYKDSERPSGIPE
RFSGSSSGTTVTLTISGVQAEDEADYYCQSADSSGTYWVFGGGTKLTVL
SEQ ID NO: 69; VH PG121 iGL QVQLQESGPGLVKPSETLSLTCTVSGGSISSYYWSWIRQPPGKGLEWIGYIYYSGSTNY NPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARTQQGKRIYGVVSFGDYYYYYY MDVWGKGTTVTVSS
SEQ ID NO: 70; VL PG121 iGL
SYVLTQPPSVSVAPGQTARITCGGNNIGSKSVHWYQQKPGQAPVLWYDDSDRPSGIPE
RFSGSNSGNTATLTISRVEAGDEADYYCQVWDSSSDHPWVFGGGTKLTVL
SEQ ID NO: 71
CTNVTNNITDDMRGEL
SEQ ID NO: 72
CTNYAPNLLSNMRGEL
SEQ ID NO: 73
CTNYAPNLLSNMRGEI
SEQ ID NO: 74
CTNYAPKLLSNMRGEI
SEQ ID NO: 75
CTNYAPKLRSMMRGEI
SEQ ID NO: 76
CTNYAPNLRSDMRGEI
SEQ ID NO: 77
TRPNNNT
SEQ ID NO: 78
TRPSNNT
SEQ ID NO: 79
TGDIIGDIRQAHCNVS
SEQ ID NO: 80
FGDI IGDIRMAHCNVS
SEQ ID NO: 81
FGDVLGDVRMAHCNIS
SEQ ID NO: 82
FGDVLGHVRMAHCNIS
SEQ ID NO: 83
FGDVLGDVDMAKCTIS SEQ ID NO: 84
SITLPCR
SEQ ID NO: 85
SIVLPCR
SEQ ID NO: 86
SLILPCR
SEQ ID NO: 87
SLILPCW
SEQ ID NO: 88; HCDR3 of BG18 iGLO ARNAI T I FGWI IGEYYYYGMDV SEQ ID NO: 89; HCDR3 of HMP1 AREGFT I FGWTFSEGYYYGMDV SEQ ID NO: 90; HCDR3 of HMP42 ARDRGREWELESYYYYYYMDV SEQ ID NO: 91; HCDR3 of HMP43 AKDIESRYFDWDNYYYYGMDV SEQ ID NO: 92; VH4-4
SEQ ID NO: 93; VL3-25
SEQ ID NO: 94; BG18 VH CDR3
ARNAIRIYGWALGEWFHYGMDV
SEQ ID NO: 95; BG18 iGLl VH CDR3
ARNAI T I FGWILGEYYYYGMDV
SEQ ID NO: 96; BG18 iGL2 VH CDR3
ARNAI T I FGWALGEYYYYGMDV
SEQ ID NO: 97; Prel VH CDR3
ARGQRT I FGWILFNYYYYGMDV
SEQ ID NO: 98; Pre2 VH CDR3
ARDLLTSFGWI FSQHKSYGMDV
SEQ ID NO: 99; Pre3 VH CDR3 ARDSITIFGWILQDYYYYGMDV
SEQ ID NO: 100; Pre4 VH CDR3
ARERDTIFGWSLRVYYYYGMDV
SEQ ID NO: 101; Pre5 VH CDR3
ARDKDTIFGWI IGEYYYYGMDV
SEQ ID NO: 102; Pre6 VH CDR3
ARGSITIFGWI ILEDYYYGMDV
SEQ ID NO: 103; Pre7 VH CDR3
AREQRAIFGWI IREDYYYGMDV
SEQ ID NO: 104; Pre8 VH CDR3
ARARLRVFGWIFLYGYYYGMDV
SEQ ID NO: 105; Pre9 VH CDR3
ARDRKRFFGWIMDESYYYGMDV
SEQ ID NO: 106; PrelO VH CDR3
ARDGVKIFGWI IREAYYYGMDV
SEQ ID NO: 107; Prell VH CDR3
ARDGIKIFGWIMGEAHYYGMDV
SEQ ID NO: 108; Prel2 VH CDR3
AKGRGTIFGWRFYENYYYGMDV
SEQ ID NO: 109; Prel3 VH CDR3
AKALNTIFGWTLTQYYYYGMDV
SEQ ID NO: 110; Prel4 VH CDR3
ARGRITIFGWTLRGYYYYGMDV
SEQ ID NO: 111; Prel5 VH CDR3
ARDIHNSFGWSLDQNYYYGMDV

Claims

CLAIMS What is claimed is:
1. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises one or more of
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
(i) CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) CTNYAPNLLSNMRGEL (SEQ ID NO: 72) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) CTNYAPNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iv) CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(v) CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(vi) CTNY APNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of
(i) TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(ii) TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of
(i) TGDIIGDIRQAHCNV S (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) FGDIIGDIRMAHCNV S (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, (iv) FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(v) FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of
(i) SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(iv) SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
2. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
(i) CTNVTNNITDDMRGEL (SEQ ID NO: 71) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) CTNYAPNLLSNMRGEL (SEQ ID NO: 72) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) CTNYAPNLLSNMRGEI (SEQ ID NO: 73) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iv) CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(v) CTNYAPKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(vi) CTNY APNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of (i) TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(ii) TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of
(i) TGDIIGDIRQAHCNV S (SEQ ID NO: 79) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) FGDIIGDIRMAHCNV S (SEQ ID NO: 80) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iv) FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(v) FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution; and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of
(i) SITLPCR (SEQ ID NO: 84) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(ii) SIVLPCR (SEQ ID NO: 85) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution,
(iii) SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, or
(iv) SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution;
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
3. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises one or more of
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of
(i) CTNVTNNITDDMRGEL (SEQ ID NO: 71), (ii) CTNY APNLLSNMRGEL (SEQ ID NO: 72),
(iii) CTNY APNLLSNMRGEI (SEQ ID NO: 73),
(iv) CTNYAPKLLSNMRGEI (SEQ ID NO: 74),
(v) CTNYAPKLRSMMRGEI (SEQ ID NO: 75), or
(vi) CTNY APNLRSDMRGEI (SEQ ID NO: 76);
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of
(i) TRPNNNT (SEQ ID NO: 77), or
(ii) TRPSNNT (SEQ ID NO: 78);
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of
(i) TGDIIGDIRQAHCNV S (SEQ ID NO: 79),
(ii) FGDIIGDIRMAHCNVS (SEQ ID NO: 80),
(iii) FGDVLGDVRMAHCNIS (SEQ ID NO: 81),
(iv) FGDVLGHVRMAHCNIS (SEQ ID NO: 82), or
(v) FGDVLGDVDMAKCTIS (SEQ ID NO: 83); and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of
(i) SITLPCR (SEQ ID NO: 84),
(ii) SIVLPCR (SEQ ID NO: 85),
(iii) SLILPCR (SEQ ID NO: 86), or
(iv) SLILPCW (SEQ ID NO: 87);
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
4. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises
(a) a VI loop comprising positions 131-154, wherein the VI loop comprises an amino acid sequence of (1) CTNVTNNITDDMRGEL (SEQ ID NO: 71),
(ii) CTNY APNLLSNMRGEL (SEQ ID NO: 72),
(iii) CTNY APNLLSNMRGEI (SEQ ID NO: 73),
(iv) CTNYAPKLLSNMRGEI (SEQ ID NO: 74),
(v) CTNYAPKLRSMMRGEI (SEQ ID NO: 75), or
(vi) CTNY APNLRSDMRGEI (SEQ ID NO: 76);
(b) a V3 loop comprising positions 297-334, wherein positions 297-303 of the V3 loop comprises an amino acid sequence of
(i) TRPNNNT (SEQ ID NO: 77), or
(ii) TRPSNNT (SEQ ID NO: 78);
(c) a V3 loop comprising positions 297-334, wherein positions 319-334 of the V3 loop comprises an amino acid sequence of
(i) TGDIIGDIRQAHCNV S (SEQ ID NO: 79),
(ii) FGDIIGDIRMAHCNVS (SEQ ID NO: 80),
(iii) FGDVLGDVRMAHCNIS (SEQ ID NO: 81),
(iv) FGDVLGHVRMAHCNIS (SEQ ID NO: 82), or
(v) FGDVLGDVDMAKCTIS (SEQ ID NO: 83); and
(d) a P 19 sheet comprising positions 413-419, wherein the b 19 sheet comprises an amino acid sequence of
(i) SITLPCR (SEQ ID NO: 84),
(ii) SIVLPCR (SEQ ID NO: 85),
(iii) SLILPCR (SEQ ID NO: 86), or
(iv) SLILPCW (SEQ ID NO: 87);
wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
5. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the p 19 sheet comprises the amino acid sequence of
(a) CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
(b) CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), and SLILPCR (SEQ ID NO: 86), respectively,
(c) CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
(d) CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively,
(e) CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCW (SEQ ID NO: 87), respectively,
(f) CTNY APNLRSDMRGEI (SEQ ID NO: 76), TRPNNNT (SEQ ID NO: 77), FGDVLGDVDMAKCTIS (SEQ ID NO: 83), and SLILPCR (SEQ ID NO: 86), respectively,
(g) CTNYAPKLLSNMRGEI (SEQ ID NO: 74) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
(h) CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVRMAHCNIS (SEQ ID NO: 81) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
(i) CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively, (j) CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
(k) CTNY APKLRSMMRGEI (SEQ ID NO: 75) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPSNNT (SEQ ID NO: 78) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGHVRMAHCNIS (SEQ ID NO: 82) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCW (SEQ ID NO: 87) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively, or
(l) CTNYAPNLRSDMRGEI (SEQ ID NO: 76) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, TRPNNNT (SEQ ID NO: 77) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, FGDVLGDVDMAKCTIS (SEQ ID NO: 83) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, and SLILPCR (SEQ ID NO: 86) comprising 0, 1, 2, 3, 4, or 5 insertion, deletion, or substitution, respectively,
and wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
6. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the p 19 sheet comprises the amino acid sequence of CTNYAPKLLSNMRGEI (SEQ ID NO: 74), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, and wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
7. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the P 19 sheet comprises the amino acid sequence of CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGDVRMAHCNIS (SEQ ID NO: 81), and SLILPCR (SEQ ID NO: 86), respectively, and wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
8. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the P 19 sheet comprises the amino acid sequence of CTNY APKLRSMMRGEI (SEQ ID NO: 75), TRPNNNT (SEQ ID NO: 77), FGDVLGHVRMAHCNIS (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, and wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
9. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the p 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), F GD VLGHVRMAHCNI S (SEQ ID NO: 82), and SLILPCR (SEQ ID NO: 86), respectively, and wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
10. An isolated polypeptide comprising a variant HIV Env gpl20 polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises a VI loop comprising positions 131-154, a V3 loop comprising positions 297-334, and a b 19 sheet comprising positions 413-419, wherein the VI loop, positions 297-303 of the V3 loop, positions 319-334 of the V3 loop, and the P 19 sheet comprises the amino acid sequence of CTNYAPKLRSMMRGEI (SEQ ID NO: 75), TRPSNNT (SEQ ID NO: 78), FGD VLGHVRMAHCNI S (SEQ ID NO: 82), and SLILPCW (SEQ ID NO: 87), respectively, and wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
11. The isolated polypeptide of any one of claims 1 to 10, wherein the variant HIV Env gp 120 polypeptide comprises N332.
12. The isolated polypeptide of any one of claims 1 to 11, wherein the N-terminal residue of the variant HIV Env gpl20 polypeptide is one of HIV Env positions 1-35.
13. The isolated polypeptide of any one of claims 1 to 12, wherein the C-terminal residue of the variant HIV Env gpl20 polypeptide is one of HIV Env positions 503-512.
14. The isolated polypeptide of any one of claims 1 to 13, wherein the variant HIV Env gpl20 polypeptide is a variant of the BG505, BG505 N332, BG505 MD39, BG505 MD64, BG505 MD39 N332, BG505 MD39 1 ImutB, or BG505 MD39 17mutE gpl20 polypeptide.
15. The isolated polypeptide of any one of claims 1 to 13, wherein the variant HIV Env gpl20 polypeptide is a variant of the BG505 MD39 N332 gpl20 polypeptide.
16. The isolated of any one of claims 1 to 13 wherein the variant HIV Env gpl20 polypeptide is a variant of the BG505 MD64 N332 gpl20 polypeptide.
17. An isolated polypeptide comprising
(a) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT1, (b) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2,
(c) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT3,
(d) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT4,
(e) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT5, or
(f) an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2-KO.
18. An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT1,
19. An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT2.
20. An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT3.
21. An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT4.
22. An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD39 N332-GT5.
23. An isolated polypeptide comprising an amino acid sequence having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of BG505 MD64 N332-GT2.
24. An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env polypeptide comprises
(a) the amino acid sequence of BG505 MD39 N332-GT1, (b) the amino acid sequence of BG505 MD39 N332-GT2,
(c) the amino acid sequence of BG505 MD39 N332-GT3,
(d) the amino acid sequence of BG505 MD39 N332-GT4,
(e) the amino acid sequence of BG505 MD39 N332-GT5, or
(f) the amino acid sequence of BG505 MD39 N332-GT2-KO.
25. An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332- GT1.
26. An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332- GT2.
27. An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332- GT3.
28. An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332- GT4.
29. An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD39 N332- GT5.
30. An isolated polypeptide comprising a variant HIV Env polypeptide, wherein the variant HIV Env gpl20 polypeptide comprises the amino acid sequence of BG505 MD64 N332- GT2.
31. The isolated polypeptide of any one of claims 1 to 30, wherein the variant HIV Env polypeptide comprises K137 and H325.
32. The isolated polypeptide of any one of claims 1 to 30, wherein the variant HIV Env polypeptide comprises K137 and P325.
33. The isolated polypeptide of any one of claims 1 to 30, wherein the variant HIV Env polypeptide comprises K137, M141 and H325.
34. The isolated polypeptide of any one of claims 1 to 30, wherein the variant HIV Env polypeptide comprises K137, R139, M141, and H325.
35. The isolated polypeptide of any one of claims 1 to 30, wherein the variant HIV Env polypeptide comprises K137 and P325.
36. The isolated polypeptide of any one of claims 1 to 30, wherein the variant HIV Env polypeptide comprises K137, M141, and P325.
37. The isolated polypeptide of any one of claims 1 to 30, wherein the variant HIV Env polypeptide comprises K137, R139, M141, and P325.
38. The isolated polypeptide of any one of claims 1 to 37 that specifically binds to the BG18 antibody.
39. The isolated polypeptide of any one of claims 1 to 38 that specifically binds to an antibody selected from the group consisting of BG18.11 and BG18.6.
40. The isolated polypeptide of any one of claims 1 to 39 that specifically binds to an antibody selected from the group consisting of BG18 iGLO, BG18 iGLl, and BG18 iGL2.
41. The isolated polypeptide of any one of claims 1 to 40 that specifically binds to an antibody selected from the group consisting of PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, and PRE15.
42. The isolated polypeptide of any one of claims 1 to 41 that specifically binds to the PG121 iGL antibody.
43. The isolated polypeptide of any one of claims 1 to 42 that specifically binds to an antibody comprising the VH and VL of
(a) VH4-59 and BG18 iGL, respectively;
(b) VH1-69 and BG18 iGL, respectively;
(c) VH5-51 and BG18 iGL, respectively;
(d) VH3-33 and BG18 iGL, respectively;
(e) VH3-23 and BG18 iGL, respectively;
(f) BG18 iGLl and VL3-19, respectively;
(g) BG18 iGLl and VL3-10, respectively;
(h) BG18 iGLl and VL3-1, respectively;
(i) BG18 iGLl and VL3-21, respectively; or
(j) BG18 iGLl and VL2-8, respectively.
44. The isolated polypeptide of any one of claims 1 to 43 that comprises a gpl40.
45. The isolated polypeptide of any one of claims 1 to 43 that comprises a gpl60.
46. A recombinant HIV Env trimer comprising the isolated polypeptide of any one of claims 1 to 45.
47. The recombinant HIV Env trimer of claim 46 that is a homotrimer.
48. The recombinant HIV Env trimer of claim 46 that is a heterotrimer.
49. The recombinant HIV Env trimer of any one of claims 46 to 48 that comprises gp 120-gp41 heterodimers.
50. The recombinant HIV Env trimer of any one of claims 46 to 48 that comprises gp 120-gp41 heterodimers wherein the heterodimers are covalently linked.
51. The recombinant HIV Env trimer of any one of claims 46 to 48 that comprises gp 120-gp41 fusions.
52. The recombinant HIV Env trimer of any one of claims 46 to 48 that is a stabilized trimer.
53. The recombinant HIV Env trimer of any one of claims 46 to 48 that is an SOSIP, NFL or
UFO trimer.
54. The recombinant HIV Env trimer of any one of claims 46 to 48 that is an SOSIP trimer.
55. A nanoparticle comprising the isolated polypeptide of any one of claims 1 to 45.
56. The nanoparticle of claim 55, wherein the nanoparticle is a ferritin nanoparticle, an encapsulin nanoparticle, a Sulfur Oxygenase Reductase (SOR) nanoparticle, a lumazine synthase nanoparticle or a pyruvate dehydrogenase nanoparticle.
57. The nanoparticle of claim 55, wherein the nanoparticle comprises N332-GT1, N332-GT2, N332-GT3, N332-GT4, or N332-GT5.
58. The nanoparticle of claim 55, wherein the nanoparticle is a ferritin nanoparticle.
59. The nanoparticle of claim 55, wherein the nanoparticle is a ferritin nanoparticle having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 99%, or 100% sequence identity to the amino acid sequence of the amino acid sequence of SEQ ID NO. 13 or 15.
60. The nanoparticle of claim 55, wherein the nanoparticle is a ferritin nanoparticle comprising the amino acid sequence of SEQ ID NO. 13 or 15.
61. An isolated polynucleotide encoding the isolated polypeptide of any one of claims 1 to 45, recombinant HIV Env trimer of any one of claims 46 to 54, or the nanoparticle of claim 55 to 60.
62. The isolated polynucleotide of claim 61, which is a DNA.
63. The isolated polynucleotide of claim 61, which is an mRNA.
64. The isolated polynucleotide of claim 63, wherein the mRNA comprises a modified nucleotide.
65. An RNA replicon comprising the isolated polynucleotide of claim 61.
66. An isolated vector comprising the polynucleotide of claim 61.
67. The isolated vector of claim 66, wherein the vector is a viral vector.
68. A recombinant virus comprising the polynucleotide of claim 61.
69. The recombinant virus of claim 68, which is a recombinant adeno-associated virus (AAV).
70. A host cell comprising the polynucleotide of any one of claims 61 to 64, or the vector of claim 66 or claim 67.
71. The host cell of claim 70, which is selected from the group consisting of E. coli, Pseudomonas, Bacillus, Streptomyces, yeast, CHO, YB/20, NSO, PER-C6, HEK-293T, NIH-3T3, HeLa, BHK, Hep G2, SP2/0, Rl.l, B-W, L-M, COS 1, COS 7, BSC1, BSC40, BMT10 cell, plant cell, insect cell, and human cell in tissue culture.
72. A method of producing the isolated polypeptide of any one of claims 1 to 45, recombinant HIV Env trimer of any one of claims 46 to 54, or the nanoparticle of claim 55 to 60 comprising, culturing the host cell of claim 71 so that the polynucleotide is expressed and the isolated polypeptide, recombinant HIV Env trimer, or nanoparticle is produced.
73. A VLP comprising the polypeptide of any one of claims 1 to 45.
74. A liposome comprising the polypeptide of any one of claims 1 to 45.
75. A pharmaceutical composition comprising the isolated polypeptide of any one of claims 1 to 45, recombinant HIV Env trimer of any one of claims 46 to 54, the nanoparticle of claim any one of claims 55 to 60, the polynucleotide of any one of claims 61 to 64, the vector of claim 66 or claim 67, the recombinant virus of claim 68 or claim 69, VLP of claim 73, or the liposome of claim 74 and a pharmaceutically acceptable excipient.
76. An immunogenic composition comprising the isolated polypeptide of any one of claims 1 to 45, recombinant HIV Env trimer of any one of claims 46 to 54, the nanoparticle of claim any one of claims 55 to 60, the polynucleotide of any one of claims 61 to 64, the vector of claim 66 or claim 67, the recombinant virus of claim 68 or claim 69, VLP of claim 73, or the liposome of claim 74 and a pharmaceutically acceptable excipient.
77. The immunogenic composition of claim 76, further comprising an adjuvant.
78. The immunogenic composition of claim 77, wherein the adjuvant comprises lecithin.
79. The immunogenic composition of claim 77, wherein the adjuvant comprises alum.
80. The immunogenic composition of claim 77, wherein the adjuvant comprises saponin, cholesterol and phospholipid.
81. The immunogenic composition of claim 7, wherein the adjuvant comprises carbomer homopolymer and lecithin.
82. The immunogenic composition of any one of claims 76 to 81 that is capable of eliciting a BG18 like response.
83. The immunogenic composition of any one of claims 76 to 81 that is capable of eliciting a BG18 like response in a human subject.
84. The immunogenic composition of any one of claims 76 to 81 that is capable of eliciting a BG18 like response in BG 18sH B cell adoptive transfer recipient mice.
85. The immunogenic composition of any one of claims 76 to 81 that is capable of eliciting the production of an antibody that binds to N332-GT2.
86. The immunogenic composition of any one of claims 76 to 81 that is capable of eliciting the production of an antibody that binds to N332-GT2 with a higher affinity than to N332- GT2-KO.
87. The immunogenic composition of any one of claims 76 to 81 that is capable of eliciting the production of a broadly neutralizing antibody in a subject.
88. A method for eliciting an immune response to HIV Env gpl20 in a subject, comprising administering to the subject an effective amount of the immunogenic composition of any one of claims 76 to 81, thereby generating the immune response.
89. The method of claim 88, wherein the subject is a human.
90. The method of claim 88, wherein the subject is a non-human primate.
91. The method of claim 88, wherein the subject is a BG 18"H B cell adoptive transfer recipient mouse.
92. The method of claim 88, wherein the subject is a mouse.
93. A method of reducing the likelihood of HIV infection in a subject exposed to HIV comprising administering to the subject an effective amount of the immunogenic composition of any one of claims 76 to 81, or the pharmaceutical composition of claim 75.
94. A method of reducing the risk of a subject becoming infected with HIV comprising administering to the subject in need thereof an effective amount of the immunogenic composition of any one of claims 76 to 81, or the pharmaceutical composition of claim 75.
95. A method of preventing HIV infection comprising administering to a subject in need thereof an effective amount of the immunogenic composition of any one of claims 76 to 81, or the pharmaceutical composition of claim 75.
96. A method of treating HIV/AIDS comprising administering to a subject in need thereof an effective amount of the immunogenic composition of any one of claims 76 to 81, or the pharmaceutical composition of claim 75.
97. The method of any one of claims 88 to 96, wherein the immunogenic composition comprises the isolated polypeptide of any one of claims 1 to 45.
98. The method of any one of claims 88 to 96, wherein the immunogenic composition comprises the recombinant HIV Env trimer of any one of claims 46 to 54.
99. The method of any one of claims 88 to 96, wherein the immunogenic composition comprises the nanoparticle of any one of claims 55 to 60.
100 The method of any one of claims 88 to 96, wherein the immunogenic composition comprises the polynucleotide of any one of claims 61 to 64.
101 The method of any one of claims 88 to 96, wherein the immunogenic composition comprises the vector of any one of claims 66 to 67.
102 The method of any one of claims 88 to 96, wherein the pharmaceutical composition comprises the recombinant virus of any one of claims 68 to 69.
103. The method of any one of claims 88 to 102, further comprising administering at least one additional therapeutic agent.
104. The method of claim 103, wherein the additional therapeutic agent comprises an antiretroviral agent.
105. The method of claim 103 or 104, wherein the additional therapeutic agent comprises a broadly neutralizing anti-HIV antibody.
106. The method of any one of claims 88 to 105, wherein the subject is a human.
107. The method of any one of claims 88 to 105, wherein the subject is a non-human primate.
108. An isolated antibody comprising
(a) the VHCDR1, VH CDR2, and VH CDR3 of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1- 69, VH5-51, VH3-33, or VH3-23; and
(b) the VL CDR1, VL CDR2, and VL CDR3 of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VL3-10, VL3- 19, VL3-1, VL3-21, or VL2-8.
109. An isolated antibody comprising the VH CDR1, VH CDR2, VH CDR3, VL CDR1, VL CDR2, and VL CDR3 ofBG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
110. An isolated antibody comprising the 3 VH CDRs and 3 VL CDRs of
(a) VH4-59 and BG18 iGL, respectively;
(b) VH1-69 and BG18 iGL, respectively;
(c) VH5-51 and BG18 iGL, respectively;
(d) VH3-33 and BG18 iGL, respectively;
(e) VH3-23 and BG18 iGL, respectively;
(f) BG18 iGLl and VL3-19, respectively;
(g) BG18 iGLl and VL3-10, respectively;
(h) BG18 iGLl and VL3-1, respectively;
(i) BG18 iGLl and VL3-21, respectively; or
(j) BG18 iGLl and VL2-8, respectively.
111. An isolated antibody comprising
(a) the VH of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VH4-595, VH1-69, VH5-51, VH3-33, or VH3- 23; and
(b) the VL of BG18, BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, PRE15, VL3-10, VL3-19, VL3-1, VL3-21, or VL2-8.
112. An isolated antibody comprising the VH and VL of BG18.11, BG18.6, BG18 iGLO, BG18 iGLl, BG18 iGL2, PRE1, PRE2, PRE3, PRE4, PRE5, PRE6, PRE7, PRE8, PRE9, PRE10, PRE11, PRE12, PRE13, PRE14, or PRE15.
113. An isolated antibody comprising the VH and VL of
(a) VH4-59 and BG18 iGL, respectively;
(b) VH1-69 and BG18 iGL, respectively;
(c) VH5-51 and BG18 iGL, respectively;
(d) VH3-33 and BG18 iGL, respectively; (e) VH3-23 and BG18 iGL, respectively;
(f) BG18 iGLl and VL3-19, respectively;
(g) BG18 iGLl and VL3-10, respectively;
(h) BG18 iGLl and VL3-1, respectively;
(i) BG18 iGLl and VL3-21, respectively; or
(j) BG18 iGLl and VL2-8, respectively.
114. A method for identifying a vaccine candidate variant HIV Env gpl20 polypeptide, the method comprising
(a) providing a library comprising a plurality of variant HIV Env gp 120 polypeptides;
(b) contacting the library with an antibody according to any one of claims 107 to 109; and
(c) identifying a variant HIV Env gpl20 polypeptide that specifically binds to the antibody.
115. The method of claim 110, wherein the plurality of variant HIV Env gpl20 polypeptides comprise variants of a parental HIV Env gp 120 polypeptide comprising one or more amino acid substitutions at a region selected from the VI loop comprising positions 131-154, V3 loop comprising positions 297-334, and b 19 sheet comprising positions 413-419, wherein HIV Env gpl20 polypeptide positions correspond to the HXB2 reference.
116. The method of claim 111, wherein the parental HIV Env gpl20 polypeptide comprises BG505, BG505 N332, BG505 MD39, BG505 MD39 N332, BG505 MD39 l lmutB, or BG505 MD39 17mutE gpl20 polypeptide.
PCT/US2019/063903 2018-12-01 2019-12-01 Recombinant hiv env polypeptides and their use WO2020113199A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP19888314.2A EP3886898A4 (en) 2018-12-01 2019-12-01 Recombinant hiv env polypeptides and their use
US17/335,244 US20210363195A1 (en) 2018-12-01 2021-06-01 Recombinant hiv env polypeptides and their use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862774178P 2018-12-01 2018-12-01
US62/774,178 2018-12-01

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/335,244 Continuation-In-Part US20210363195A1 (en) 2018-12-01 2021-06-01 Recombinant hiv env polypeptides and their use

Publications (1)

Publication Number Publication Date
WO2020113199A1 true WO2020113199A1 (en) 2020-06-04

Family

ID=70853695

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/063903 WO2020113199A1 (en) 2018-12-01 2019-12-01 Recombinant hiv env polypeptides and their use

Country Status (3)

Country Link
US (1) US20210363195A1 (en)
EP (1) EP3886898A4 (en)
WO (1) WO2020113199A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050064392A1 (en) * 2003-01-29 2005-03-24 The Trustees Of The University Of Pennylvania Compositions, methods and kits relating to deletion mutations of immunodeficiency virus gp120 hypervariable regions
WO2017165674A1 (en) * 2016-03-23 2017-09-28 International Aids Vaccine Initiative Immunogenic trimers

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016037154A1 (en) * 2014-09-04 2016-03-10 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Recombinant hiv-1 envelope proteins and their use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050064392A1 (en) * 2003-01-29 2005-03-24 The Trustees Of The University Of Pennylvania Compositions, methods and kits relating to deletion mutations of immunodeficiency virus gp120 hypervariable regions
WO2017165674A1 (en) * 2016-03-23 2017-09-28 International Aids Vaccine Initiative Immunogenic trimers

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3886898A4 *

Also Published As

Publication number Publication date
US20210363195A1 (en) 2021-11-25
EP3886898A4 (en) 2022-08-24
EP3886898A1 (en) 2021-10-06

Similar Documents

Publication Publication Date Title
US20230060304A1 (en) Neutralizing antibodies to gp120 and their use
US11897940B2 (en) Broadly neutralizing anti-HIV-1 antibodies and methods of use thereof
AU2020201993A1 (en) Human immunodeficiency virus neutralising antibodies and methods of use thereof
EP4001304A1 (en) Broadly neutralizing antibody and uses thereof
EP3334446A2 (en) Engineered outer domain (eod) of hiv gp120, mutants and use thereof
Schiffner et al. Structural and immunologic correlates of chemically stabilized HIV-1 envelope glycoproteins
US20240109956A1 (en) Rapid elicitation of broadly neutralizing bovine antibodies to hiv env
US20210355197A1 (en) Anti-hiv antibodies
US20230272048A1 (en) Hiv-1 antibodies
US20240124560A1 (en) Human broadly neutralizing antibodies against the membrane-proximal external region of hiv env for vaccine design and intervention
US20210363195A1 (en) Recombinant hiv env polypeptides and their use
Escolano et al. Immunization expands HIV-1 V3-glycan specific B-cells in mice and macaques
WO2020117740A1 (en) Recombinant hiv env polypeptides and their uses
Cottrell Structure-Based HIV Immunogen Design
CA3229447A1 (en) Antibodies that target hla-e-host peptide complexes and uses thereof
IL300355A (en) Broadly neutralizing anti-hiv-1 antibodies and methods of use thereof

Legal Events

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

Ref document number: 19888314

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2019888314

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2019888314

Country of ref document: EP

Effective date: 20210701