WO2010059732A1 - Vaccins antiviraux présentant une immunogénicité cellulaire améliorée - Google Patents

Vaccins antiviraux présentant une immunogénicité cellulaire améliorée Download PDF

Info

Publication number
WO2010059732A1
WO2010059732A1 PCT/US2009/064999 US2009064999W WO2010059732A1 WO 2010059732 A1 WO2010059732 A1 WO 2010059732A1 US 2009064999 W US2009064999 W US 2009064999W WO 2010059732 A1 WO2010059732 A1 WO 2010059732A1
Authority
WO
WIPO (PCT)
Prior art keywords
vaccine
viral
seq
optimized
gag
Prior art date
Application number
PCT/US2009/064999
Other languages
English (en)
Inventor
Dan H. Barouch
Bette T. Korber
William M. Fischer
Original Assignee
Beth Israel Deaconess Medical Center
Los Alamos National Laboratory
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
Priority to BRPI0921588-3A priority Critical patent/BRPI0921588B1/pt
Priority to DK09828172.8T priority patent/DK2358757T3/da
Application filed by Beth Israel Deaconess Medical Center, Los Alamos National Laboratory filed Critical Beth Israel Deaconess Medical Center
Priority to SI200931885T priority patent/SI2358757T1/sl
Priority to AU2009316629A priority patent/AU2009316629B2/en
Priority to NZ593598A priority patent/NZ593598A/xx
Priority to AP2011005767A priority patent/AP3719A/en
Priority to PL09828172T priority patent/PL2358757T3/pl
Priority to BR122021004758-8A priority patent/BR122021004758B1/pt
Priority to US13/130,018 priority patent/US9017691B2/en
Priority to CN200980154787.4A priority patent/CN102282175B/zh
Priority to EP09828172.8A priority patent/EP2358757B1/fr
Priority to EP18191558.8A priority patent/EP3470437A1/fr
Priority to LTEP09828172.8T priority patent/LT2358757T/lt
Priority to JP2011537586A priority patent/JP5694945B2/ja
Priority to ES09828172T priority patent/ES2699685T3/es
Publication of WO2010059732A1 publication Critical patent/WO2010059732A1/fr
Priority to IL212984A priority patent/IL212984A/en
Priority to ZA2011/04538A priority patent/ZA201104538B/en
Priority to HK12105453.5A priority patent/HK1164896A1/xx
Priority to US14/632,869 priority patent/US9670253B2/en
Priority to IL243991A priority patent/IL243991B/en
Priority to US15/489,220 priority patent/US10426831B2/en
Priority to HRP20181776TT priority patent/HRP20181776T1/hr
Priority to CY20181101309T priority patent/CY1121215T1/el
Priority to US16/441,703 priority patent/US10772952B2/en
Priority to US16/948,257 priority patent/US11331386B2/en

Links

Classifications

    • 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
    • A61K39/12Viral 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
    • A61K39/235Adenoviridae
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/275Poxviridae, e.g. avipoxvirus
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/24011Poxviridae
    • C12N2710/24041Use of virus, viral particle or viral elements as a vector
    • C12N2710/24043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16171Demonstrated in vivo effect
    • 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
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16222New 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
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16234Use 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16271Demonstrated in vivo effect
    • 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
    • C12N2740/16311Human Immunodeficiency Virus, HIV concerning HIV regulatory proteins
    • C12N2740/16322New 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
    • C12N2740/16311Human Immunodeficiency Virus, HIV concerning HIV regulatory proteins
    • C12N2740/16334Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention provides compositions, methods, and kits for the treatment or prevention of viral infections.
  • the polyvalent (e.g., 2-valent) vaccines described herein incorporate computationally-optimized viral polypeptides that can increase the diversity or breadth and depth of cellular immune response in vaccinated subjects.
  • Vaccines that elicit cellular immune responses against viruses must reflect global viral diversity in order to effectively treat or prevent viral infection. For example, the initiation of intense and diverse HIV-I -specific T cell responses is likely crucial for an effective HIV-I vaccine. Cytotoxic T lymphocyte (CTL) responses are correlated with slow disease progression in humans, and the importance of CTL responses in non-human primate vaccination models is well established. While the highly variable Envelope (Env) is the primary target for neutralizing antibodies against HIV, and vaccine antigens will also need to be tailored to elicit these antibody responses, T cell vaccine components can target more conserved proteins to trigger responses that are more likely to cross-react. But even the most conserved HIV-I proteins are diverse enough that variation will be an issue.
  • the invention features a vaccine for treating or reducing the risk of a viral infection in a mammal, such as a human, that includes at least two distinct optimized viral polypeptides (e.g., 2, 3, 4, 5, or more distinct optimized viral polypeptides), wherein the optimized viral polypeptides correspond to the same viral gene product.
  • a mammal such as a human
  • the optimized viral polypeptides correspond to the same viral gene product.
  • the viral infection is caused by a retrovirus, reovirus, picornavirus, togavirus, orthomyxovirus, paramyxovirus, calicivirus, arenavirus, flavivirus, filovirus, bunyavirus, coronavirus, astrovirus, adenovirus, papillomavirus, parvovirus, herpesvirus, hepadnavirus, poxvirus, or polyomavirus.
  • the retrovirus is human immunodeficiency virus type 1 (HIV-I), and the viral gene products include Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, or Vpu.
  • the vaccine includes no more than two optimized viral polypeptides corresponding to one of the Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, or Vpu viral gene products. In another embodiment, the vaccine does not include optimized viral polypeptides corresponding to Gag and Nef.
  • the vaccine includes at least two distinct optimized viral polypeptides (e.g., 2, 3, 4, 5, or more distinct optimized viral polypeptides) for a first viral gene product selected from Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, and Vpu and one or more distinct optimized viral polypeptides (e.g., 2, 3, 4, 5, or more distinct optimized viral polypeptides) for a second viral gene product different from the first viral gene product selected from Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, and Vpu.
  • first viral gene product selected from Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, and Vpu
  • distinct optimized viral polypeptides e.g., 2, 3, 4, 5, or more distinct optimized viral polypeptides
  • the invention features a vaccine for treating or reducing the risk of human immunodeficiency virus type 1 (HIV-I) infection in a mammal, such as a human, that includes an optimized viral polypeptide that has at least seven contiguous amino acids (e.g., at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 50, 100, 150, 175, 200, 250, 300, 350, 400, 450, 500 or more contiguous amino acids in length) having at least 85% amino acid sequence identity to any one of the sequences set forth in SEQ ID NOS: 1-29.
  • HSV-I human immunodeficiency virus type 1
  • the optimized viral polypeptide has at least seven contiguous amino acids (e.g., at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 50, 100, 150, 175, 200, 250, 300, 350, 400, 450, 500 or more contiguous amino acids in length) having amino acid sequence identity to any one of the sequences set forth in SEQ ID NOS: 1-29.
  • the optimized viral polypeptide has the amino acid sequence of any one of the sequences set forth in SEQ NOS: 1-29.
  • the vaccine includes at least two optimized viral polypeptides selected from any one or more of groups a)-k): a) SEQ ID NOS: 1 and 2; b) SEQ ID NOS:3, 4, and 5; c) SEQ ID NOS:6 and 7; d) SEQ ID NOS:8-12; e) SEQ ID NOS: 13, 14, and 15; f) SEQ ID NOS:16, 17, and 18; g) SEQ ID NOS:19 and 20; h) SEQ ID NOS:21, 22, and 23; i) SEQ ID NOS:24 and 25; j) SEQ ID NOS:26 and 27; k) and SEQ ID NOS:21-22.
  • groups a)-k) selected from any one or more of groups a)-k): a) SEQ ID NOS: 1 and 2; b) SEQ ID NOS:3, 4, and 5; c) SEQ ID NOS:6 and 7; d) SEQ ID NOS:8-12; e)
  • the vaccine can include a pair of optimized viral polypeptides selected from any one of groups a)-k) above and one or more different optimized viral polypeptides from the same or a different group a)-k). In other embodiments, the vaccine can include at least three or four or more optimized viral polypeptides from one or more of groups a)- k).
  • the invention features a vaccine for treating or reducing the risk of a viral infection in a mammal, such as a human, that includes at least two pairs of distinct optimized viral polypeptides, wherein each pair of optimized viral polypeptides corresponds to the same viral gene product, and wherein no more than two optimized viral polypeptides incorporated in the vaccine correspond to the same viral gene product.
  • the vaccine includes at least three pairs of distinct optimized viral polypeptides. In another embodiment, the vaccine includes at least four pairs of distinct optimized viral polypeptides.
  • the viral infection is caused by a retrovirus, reovirus, picornavirus, togavirus, orthomyxovirus, paramyxovirus, calicivirus, arenavirus, flavivirus, f ⁇ lovirus, bunyavirus, coronavirus, astrovirus, adenovirus, papillomavirus, parvovirus, herpesvirus, hepadnavirus, poxvirus, or polyomavirus.
  • the retrovirus is human immunodeficiency virus type 1 (HIV-I), and the viral gene products include Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, or Vpu.
  • the vaccine includes no more than two optimized viral polypeptides corresponding to one of the Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, or Vpu viral gene products. In another embodiment, the vaccine does not include optimized viral polypeptides corresponding to Gag and Nef. In a further embodiment, the vaccine includes at least three pairs of distinct optimized viral polypeptides corresponding to any three of the Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, or Vpu viral gene products.
  • the vaccine includes at least four pairs of distinct optimized viral polypeptides corresponding to any four of the Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, or Vpu viral gene products.
  • the vaccine elicits a cellular immune response against a viral gene product.
  • the vaccine elicits a cellular immune response against HIV-I.
  • the nucleotide sequence of at least one distinct optimized viral polypeptide is encoded by a nucleic acid or vector.
  • the vector is a recombinant adenovirus, such as adenovirus serotype 26 (Ad26), adenovirus serotype 34 (Ad34), adenovirus serotype 35 (Ad35), adenovirus serotype 48 (Ad48), or adenovirus serotype 5 HVR48 (Ad5HVR48).
  • the vaccine is in combination with a pharmaceutically acceptable carrier, excipient, or diluent.
  • the invention features a nucleic acid that includes the nucleotide sequence of an optimized viral polypeptide that has at least seven contiguous amino acids (e.g., at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 50, 100, 150, 175, 200, 250, 300, 350, 400, 450, 500 or more contiguous amino acids in length) having at least 85% amino acid sequence identity to any one of the amino acid sequences set forth in SEQ ID NOS: 1- 29.
  • contiguous amino acids e.g., at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 50, 100, 150, 175, 200, 250, 300, 350, 400, 450, 500 or more contiguous amino acids in length
  • the optimized viral polypeptide has at least seven contiguous amino acids (e.g., at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 50, 100, 150, 175, 200, 250, 300, 350, 400, 450, 500 or more contiguous amino acids in length) having sequence identity to any one of the amino acid sequences set forth in SEQ ID NOS: 1-29.
  • the optimized viral polypeptide has any one of the amino acid sequences set forth in SEQ ID NOS: 1 -29.
  • the nucleic acid includes a vector.
  • the vector is a recombinant adenovirus, such as adenovirus serotype 26 (Ad26), adenovirus serotype 34 (Ad34), adenovirus serotype 35 (Ad35), adenovirus serotype 48 (Ad48), or adenovirus serotype 5 HVR48 (Ad5HVR48).
  • adenovirus serotype 26 Ad26
  • Ad34 adenovirus serotype 34
  • Ad35 adenovirus serotype 35
  • Ad48 adenovirus serotype 48
  • Ad5HVR48 adenovirus serotype 5 HVR48
  • the invention features an optimized viral polypeptide that has at least seven contiguous amino acids (e.g., at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 50, 100, 150, 175, 200, 250, 300, 350, 400, 450, 500 or more contiguous amino acids in length) having at least 85% amino acid sequence identity to any one of the amino acid sequences set forth in SEQ ID NOS: 1-29.
  • contiguous amino acids e.g., at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 50, 100, 150, 175, 200, 250, 300, 350, 400, 450, 500 or more contiguous amino acids in length
  • the optimized viral polypeptide has at least seven contiguous amino acids (e.g., at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 50, 100, 150, 175, 200, 250, 300, 350, 400, 450, 500 or more contiguous amino acids in length) having sequence identity to any one of the amino acid sequences set forth in SEQ ID NOS: 1-29.
  • the optimized viral polypeptide has any one of the amino acid sequences set forth in SEQ ID NOS: 1-29.
  • the invention features a method for treating or reducing the risk of a viral infection in a mammal, such as a human, by administering a vaccine or nucleic acid of the invention,
  • the viral infection is caused by a retrovirus, reovirus, picornavirus, togavirus, orthomyxovirus, paramyxovirus, calicivirus, arenavirus, flavivirus, filovirus, bunyavirus, coronavirus, astrovirus, adenovirus, papillomavirus, parvovirus, herpesvirus, hepadnavirus, poxvirus, or polyomavirus.
  • the retrovirus is human immunodeficiency virus type 1 (HIV-I), and the viral gene products include Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, or Vpu.
  • the vaccine or nucleic acid elicits a cellular immune response against a viral gene product.
  • the invention features a method of manufacturing a vaccine for treating or reducing the risk of a viral infection in a mammal, such as a human, by synthesizing a vaccine of the invention.
  • the invention features a method of manufacturing a vaccine for treating or reducing the risk of a viral infection in a mammal, such as a human, by contacting a nucleic acid of the invention with a cell and isolating a optimized viral polypeptide.
  • the optimized viral polypeptide elicits a cellular immune response when administered to a mammal.
  • the cellular immune response can be against a viral gene product.
  • the viral infection is caused by a retrovirus, reovirus, picornavirus, togavirus, orthomyxovirus, paramyxovirus, calicivirus, arenavirus, flavivirus, filovirus, bunyavirus, coronavirus, astrovirus, adenovirus, papillomavirus, parvovirus, herpesvirus, hepadnavirus, poxvirus, or polyomavirus.
  • the retrovirus is human immunodeficiency virus type 1 (HIV-I), and the viral gene products include Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, or Vpu.
  • the invention features a kit that includes a vaccine of the invention, a pharmaceutically acceptable carrier, excipient, or diluent, and instructions for the use thereof.
  • the kit also includes an adjuvant.
  • the invention features a kit that includes a nucleic acid of the invention, a pharmaceutically acceptable carrier, excipient, or diluent, and instructions for the use thereof.
  • the kit also includes an adjuvant.
  • the optimized viral polypeptide is encoded by a nucleic acid sequence that is optimized for expression in humans (e.g., any one of SEQ ID NOS:5, 10, 11, 12, 15, 18, and 23).
  • Optimized viral polypeptide sequences are initially generated by modifying the amino acid sequence of one or more naturally- occurring viral gene products (e.g., peptides, polypeptides, and proteins) to increase the breadth, intensity, depth, or longevity of the antiviral immune response (e.g., cellular or humoral immune responses) generated upon immunization (e.g., when incorporated into a vaccine of the invention) of a mammal (e.g., a human).
  • a mammal e.g., a human
  • the optimized viral polypeptide may correspond to a "parent" viral gene sequence; alternatively, the optimized viral polypeptide may not correspond to a specific "parent” viral gene sequence but may correspond to analogous sequences from various strains or quasispecies of a virus. Modifications to the viral gene sequence that can be included in an optimized viral polypeptide include amino acid additions, substitutions, and deletions.
  • the optimized viral polypeptide is the composite or merged amino acid sequence of two or more naturally-occurring viral gene products (e.g., natural or clinical viral isolates) in which each potential epitope (e.g., each contiguous or overlapping amino acid sequence of 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more amino acids in length) is analyzed and modified to improve the immunogenicity of the resulting optimized viral polypeptide.
  • Optimized viral polypeptides that correspond to different viral gene products can also be fused to facilitate incorporation in a vaccine of the invention. Methods of generating an optimized viral polypeptides are described in, e.g., Fisher et al.
  • the corresponding polypeptide can be produced or administered by standard techniques (e.g., recombinant viral vectors, such as the adenoviral vectors disclosed in International Patent Application Publications WO 2006/040330 and WO 2007/104792, herein incorporated by reference).
  • pharmaceutically acceptable carrier is meant a carrier which is physiologically acceptable to the treated mammal while retaining the therapeutic properties of the compound with which it is administered.
  • One exemplary pharmaceutically acceptable carrier is physiological saline.
  • Other physiologically acceptable carriers and their formulations are known to one skilled in the art and described, e.g., in Remington 's Pharmaceutical Sciences (18 th edition, ed. A. Gennaro, 1990, Mack Publishing Company, Easton, PA), incorporated herein by reference.
  • vector is meant a DNA construct that contains a promoter operably linked to a downstream gene or coding region (e.g., a cDNA or genomic DNA fragment, which encodes a polypeptide or polypeptide fragment).
  • a recipient cell e.g., a prokaryotic or eukaryotic cell, e.g., a bacterium, yeast, insect cell, or mammalian cell, depending upon the promoter within the expression vector
  • organism including, e.g., a human
  • Vectors for in vitro transcription/translation are also well known in the art and are described further herein.
  • a vector may be a genetically engineered plasmid, virus, or artificial chromosome derived from, e.g., a bacteriophage, adenovirus, retrovirus, poxvirus, or herpesvirus.
  • viral gene product is meant any naturally-occurring viral peptide, polypeptide, or protein, or fragment thereof.
  • the viral gene product is derived from the human immunodeficiency virus type 1 (HIV-I).
  • HIV-I viral gene products include the Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, and Vpu polypeptides.
  • Figure 1 is a chart that illustrates the expanded breadth of computationally- optimized HIV-I Gag, Pol, and Env viral polypeptides against global potential T-cell epitopes (PTE) peptides in Rhesus macaques. Animals immunized with the optimized viral polypeptides (blue) reacted with the greatest number of recall peptide pools.
  • PTE T-cell epitopes
  • Figure 2 is a chart that shows that computationally-modified HIV-I Gag, Pol, and Env viral polypeptides expand the breadth of epitope-specific cellular immune response.
  • Figure 3 illustrates the breadth of cellular immune responses detected in Rhesus macaques following immunization with HIV-I viral gene products Gag, Pol, and Env derived from the computationally-modified viral polypeptides of the invention, as well as animals immunized with consensus HIV-I antigens or HIV-I clade C isolate antigens. Animals immunized with the optimized viral polypeptides (blue) reacted with the greatest number of recall peptide pools. Since the animals are outbred, the pools differ from animal to animal. Gag, Pol, and Env each elicit many cellular immune responses and can have shared patterns of reactivity.
  • Figures 4A-4C are graphs showing the potential epitopes shared between the different vaccines tested (2 valent mosaic (Mos2), M consensus (Mcon), and optimized clade C (OptC)) by viral polypeptide (Pol (Fig. 4A), Gag (Fig. 4B), and Env (Fig. 4C)).
  • Figures 4A-C show the relative coverage of the current HIV database full length genome set and the PTE peptides by the different vaccine candidates.
  • Figure 5 is a graph showing that the number of PTE peptide responses (where each response is considered an independent event regardless of overlap) to the 2 valent mosaic (Mos2) vaccine is greater than the number of responses to the M group consensus (Mcon) vaccine and the natural viral strain vaccine (optimized clade C (C Natural (optimal)), which has been selected to give optimal coverage of the M group collection (OptC) vaccine antigens.
  • Figure 5 shows the number of PTE peptide responses per animal by protein, CD8+ T cell, and CD4+ T cell.
  • Mos2 > Mcon ⁇ OptC (Mcon shows a trend for more response than OptC).
  • the Wilcoxon p- value for Mos2 compared to Mcon: p-value 0.001058.
  • Figure 6 is a chart showing the number of PTE peptides that trigger T cell responses.
  • a median number of 16 (range; 12-29) PTE peptides of the 2 valent mosaic (Mos2) vaccine trigger a response in CD8+ T cells, while only a median number of 6 (range: 0-7) Mcon peptides and only a median number of 3 peptides (range: 0-3) of OptC peptides trigger a response in CD8+ T cells.
  • Mos2 2 valent mosaic
  • the trend for responses is Mos2 > Mcon > OptC.
  • Figure 7 is a schematic summarizing the mapping of all CD8+ T cell Gag PTE peptides that are recognized by T cells from each of the animals studied (see Example 3 below).
  • the animal number, peptide pool and peptide number label the boundaries of each reactive peptide.
  • Mosaics have potential advantages over the monovalent vaccines.
  • Mosaics have a better chance of stimulating a response that reacts with more common variants.
  • Mosaics also stimulate multiple responses to the different forms that are present in the cocktail. Thus, mosaics have the potential to block common escape routes.
  • Figure 8 is a schematic summarizing the mapping of CD4+ T cell Gag PTE peptides that are recognized by T cells from each of the animals studied.
  • Figure 8 discloses SEQ ID NQ: 43.
  • Figure 9 is a chart illustrating typical patterns of PTE responses to the ConM vaccine or to the optimal natural vaccine, aligning peptides that elicit a response with the relevant region of the vaccine. Good matches with solid stretches of identity between vaccine and target PTE peptide are necessary to achieve a reaction to these vaccines.
  • Figure 9 discloses SEQ ID NOS 44-57, respectively, in order of appearance.
  • Figure 10 is a chart illustrating that mosaic vaccines generated many responses that recognized multiple variant overlapping peptides with no apparent antigenic competition and with broad local responses. In particular, four variable PTE peptides were recognized. Moreover, in the region of overlap both mosaic forms were recognized, as well a combination of the two. Finally, a new form (S) was recognized.
  • Figure 10 discloses SEQ ID NOS 58-63, respectively, in order of appearance.
  • Figure 11 is a chart illustrating a typical pattern of CD8+ PTE peptide responses in mosaic vaccinated animal (361-07). 22 PTE peptides were tested and 8 CD8 responsive regions were identified; 5 regions included variable peptides that match amino acids in one or the other of the mosaics. 5 CD4 responsive regions were identified. Thus, T cell responses to Mosaics see more variable peptides in a given region. This seemed to be true of CD8 T cell responses in particular. This could be the result of triggering multiple T cell clones that recognize variants of epitopes, and these may block fit escape routes. Not only are there more responses, they are deeper and cover more variants.
  • Figure 1 1 discloses CD8 responses as SEQ ID NOS 64-101, respectively, in order of appearance. CD4 responses disclosed as SEQ ID NOS 102-1 17, respectively, in order of appearance.
  • Figure 12 is a graph showing the number of overlapping variable PTE peptides that span regions targeted by vaccine elicited T cells.
  • Figure 13 is graph showing that the 2 mosaic antigen vaccine yields more T cell responses, relative to the Mcon and OptC vaccines, to regions that contain one or more overlapping PTE peptides.
  • Figure 13 is similar to Figure 5, monkeys shown in the same order from right to left, but with the scale changed to reflect number of responses to regions that contain one or more overlapping PTE peptides rather than single peptides.
  • FIG 14 is a chart showing the number of T cell responses in animals following administration of 2 valent mosaic (Mos2), Mcon, and OptC vaccines.
  • the 2 valent mosaic (Mos2) vaccine triggers a median number of 8 responses in CD8+ T cells, while only a median number of 3 (range: 0-6) and 1.5 peptides (range: 0-5) CD8+ T cell responses are triggered by Mcon and OptC vaccines, respectively.
  • the 2 valent mosaic (Mos2) vaccine triggers a median number of 3 (range; 2-5) responses in CD4+ T cells, while only a median number of 1 (range: 0-2) and 0.5 (range: 0-2) CD4+ T cell response are triggered by Mcon and OptC vaccines, respectively.
  • the trend for responses is Mos2 > Mcon > OptC.
  • Figure 16 is a graph showing that the mosaic design is robust to changes in viral polypeptides over time (e.g., Gag M).
  • Figure 17 is a graph showing that coverage using 9-mer optimization is robust over near (e.g., 8-12 mers) optimization lengths (Gag is shown).
  • Figure 18 is a graph showing that an increase in the number of variants increases coverage, but has only diminishing returns (Gag is shown).
  • Figures 19A-19B are graphs showing the breadth and magnitude of epitope- specific T lymphocyte responses to PTE peptides.
  • Figure 19A is a graph showing the numbers of epitope-specific CD4+ (top) and CD8+ (bottom) T lymphocyte responses to individual PTE peptides following a single immunization of rAd26 vectors expressing mosaic (blue), M consensus (green), clade B + clade C (purple), or optimal natural clade C (red) HIV-I Gag, Pol, and Env antigens. Individual monkeys are depicted on the x-axis. The different shades of each color reflect responses to the different antigens (Gag, Pol, Env).
  • Figure 19B is a graph showing the numbers of CD4+ (top) and CD8+ (bottom) T lymphocyte response regions.
  • Figures 20A-20C show a schematic showing CD8+ T lymphocyte responses to PTE peptides at week 4 following immunization mapped on HIV-I Gag ( Figure 20A) (SEO ID NO: 1 18). Pol ( Figure 2OB) (SEO ID NO: 1 19). and Env ( Figure 20C) (SEO ID NO: 120) protein sequences. Colors denote monkeys that received the mosaic (blue), M consensus (green), clade B + clade C (purple), or optimal natural clade C (red) HIV-I Gag, Pol, and Env antigens. For each epitope, the monkey number, antigen (G, Gag; P, Pol; E, Env), subpool number, and individual PTE peptide number are indicated.
  • Figures 21A-21C show a schematic showing CD4+ T lymphocyte responses to PTE peptides at week 4 following immunization mapped on HIV-I Gag ( Figure 21 A) (SEQ ID NQ: 121), Pol ( Figure 2I B) (SEO ID NO: 122). and Env ( Figure 21C) (SEQ ID NO; 123) protein sequences. Colors denote monkeys that received the mosaic (blue), M consensus (green), clade B + clade C (purple), or optimal natural clade C (red) HIV-I Gag, Pol, and Env antigens. For each epitope, the monkey number, antigen (G, Gag; P, Pol; E, Env), subpool number, and individual PTE peptide number are indicated.
  • Figure 22 is a schematic showing the alignment of vaccine sequences with reactive PTE peptides in all monkeys at week 4 following immunization with rAd26 vectors expressing mosaic, M consensus, clade B + clade C, or optimal natural clade C HIV-I Gag, PoI, and Env antigens.
  • vaccine sequences are shown on the top, and reactive PTE peptides are shown beneath the vaccine sequences denoted by the antigen (G, Gag; P, Pol; E, Env) and PTE peptide numbers.
  • the minimal overlap region is shown in bold.
  • Sequence polymorphisms between the two mosaic or the two clade B + clade C antigens are shown in blue. Differences between the vaccine sequences and the reactive PTE peptides are shown in red.
  • Figure 22 discloses SEQ ID NOS 124- 640, respectively, in order of appearance.
  • Minimal regions within the peptides that are likely to contain the immune response epitope, based on overlap between reactive peptides when it occurs, are in bold in the vaccines. If there is no overlapping peptide, we assume the epitope can be anywhere in the peptide, so the whole region is bold. We cannot differentiate between different T cell responses targeting epitopes with different boundaries within a peptide, or more promiscuous clonal T cell responses that can tolerate variation when variants are present; either scenario could be advantageous in a vaccine immune response. The number of targeted regions corresponds to the minimum number of T cell responses required to account for the data.
  • Amino acids where the vaccine and the peptides don't match are written in red; if they fall within the region likely to carry the epitope, they are bold red. Amino acid differences outside of the overlapping regions when multiple peptides overlap are marked in red, but not bold.
  • the vaccines are always at the top.
  • the letter for each protein (Gag is G, Pol is P, Envelope is E) and the peptide number are used to label for each reactive PTE peptide.
  • the protein and HXB2 numbers follow each peptide.
  • the mosaic and clade B-"-C vaccines there are 2 antigens each and both are included in the alignment; amino acid differences in the vaccines are noted in blue, and if the reactive peptide carries the variant amino acid in the second mosaic, it is also in blue. In each of the positions where the two vaccine antigens differ, the reactive peptides are also marked in bold to indicate the positions where including two variants may have impacted the vaccine immune response and allowed greater breadth and depth.
  • the first vaccine summarized is the clade B+C vaccine, and animal 287-95 is the first animal for which responses are listed.
  • Two of the CD8 peptides show substantial overlap, E26 and E282, so both may be targets for the same CTL response; thus we also note there are only 2 CD8 responsive regions, and 1 CD4 responsive region.
  • For each responsive region we write out the number of overlapping peptides per region (e.g., CD8: 1 2 CD4: 1) to assess depth of responses; the two is red to indicate that the region of overlap is variable in the reactive peptides.
  • the vaccine differs, like the D/E in the second reactive region, it is marked in blue. Only the region of overlap is bold.
  • the H in E282 was not found in either vaccine so it is marked with red; it is within the region of overlap so it is bold.
  • Each reactive peptide has its protein and corresponding HXB2 numbering noted on the right.
  • Figures 23A-23C are graphs showing the magnitude of all Gag-, PoI-, and Env- specific CD8+ ( Figure 23A and 23B) and CD4+ ( Figure 23C) T lymphocyte responses arranged from lowest to highest.
  • Figures 24A-C show the depth of epitope-specific T lymphocyte responses to PTE peptides.
  • Figure 24A is a schematic showing an example of mapped T lymphocyte responses in monkey 366 that received the optimal natural clade C antigens.
  • Figure 24B is a schematic showing an example of mapped T lymphocyte responses in monkey 361 that received the 2-valent mosaic antigens.
  • vaccine sequences are shown on the top (OptC; Mosl, Mos2), and reactive PTE peptides are shown beneath the vaccine sequences denoted by the antigen (G, Gag; P, Pol; E, Env) and the PTE peptide numbers. The minimal overlap region is shown in bold.
  • FIG. 24C is a graph showing the depth of CD4+ (top) and CD8+ (bottom) T lymphocyte responses following immunization with rAd26 vectors expressing mosaic, M consensus, clade B + clade C, or optimal natural clade C antigens. Individual monkeys are depicted on the x-axis. One response variant (light shade) or >1 response variants (dark shade) arc shown for each cpitopic region.
  • Figure 24A discloses SEQ ID NOS 641- 650, respectively, in order of appearance.
  • Figure 24B discloses SEQ ID NOS 651-685, respectively, in order of appearance.
  • Figure 25 is a graph showing the breadth of epitope-specific T lymphocyte responses to HIV-I Gag peptides from clades A, B, and C. Breadth of cellular immune responses was assessed utilizing subpools of overlapping peptides from the following strains of HIV-I Gag: clade C DU422, clade C ZM651 , consensus C, consensus A, and consensus B. Numbers of positive subpools are shown following a single immunization of rAd26 vectors expressing mosaic (blue), M consensus (green), clade B + clade C (purple), or optimal natural clade C (red) HIV-I Gag, Pol, and Env antigens. Individual monkeys are depicted on the x-axis.
  • Figure 26A-D are graphs showing the cellular and humoral immune responses following the boost immunization. Shown are the magnitude (Fig. 26A) and breadth (Fig. 26B) of individual T lymphocyte responses at week 4 post-prime (left side of each panel) and at week 44 post-boost (right side of each panel) for each monkey. Monkeys were primed at week 0 with rAd26 vectors and were boosted at week 40 with rAd5IIVR48 vectors expressing mosaic, M consensus, or optimal natural clade C HIV-I Gag, Pol, and Env antigens. Individual monkeys are depicted on the x axis. In Fig.
  • FIG. 26A red denotes CD8+ T lymphocyte responses, blue denotes CD4+ T lymphocyte responses, lines depict responses observed at both timepoints, and dots depict responses observed at only one timepoint.
  • Figure 26B different shades of each color reflect responses to the different antigens (Gag, Pol, Env).
  • Figure 26C is a graph showing the Env-specific ELISA endpoint titers at weeks 0, 4, and 44.
  • Figure 26D is a graph showing the neutralizing antibody (NAb) titers to the tier 1 clade A (DJ263.8), clade B (SF162.LS), and clade C (MW965.26) viruses at week 44.
  • NAb titers to murine leukemia virus as a negative control were ⁇ 20 for all samples.
  • Figure 27 is a graph showing the theoretical coverage of PTE peptides by the various vaccine antigens. Percentage of 9 amino acid PTE peptides that are covered by the mosaic (blue), M consensus (green), clade B + clade C (purple), or optimal natural clade C (red) HIV-I Gag, Pol, and Env antigens are shown.
  • the invention features optimized viral polypeptides that are computationally derived from naturally-occurring viral gene products.
  • the optimized viral polypeptides of the invention allow for an increased breadth and depth of virus-specific immunity (e.g., cellular immunity, such as T cell-based immune responses) following immunization of a subject (e.g., a human) with one or more optimized viral polypeptides of the invention or vaccines (e.g., a vector) that incorporate one or more optimized viral polypeptides of the invention.
  • virus-specific immunity e.g., cellular immunity, such as T cell-based immune responses
  • vaccines e.g., a vector
  • the invention provides vaccines that can be administered to a subject (e.g., a human) infected with or at risk of becoming infected with a viral infection.
  • the vaccines of the invention incorporate at least two distinct optimized viral polypeptides for each corresponding viral gene product represented.
  • the incorporation of at least two distinct optimized viral polypeptides allows for increased coverage and representation of immunogenic epitopes in the vaccine, which the inventors have found results in an increase in the total number of virus-specific immune responses following vaccination of a subject.
  • the present invention also provides methods of administering and manufacturing vaccines, vectors, and optimized viral polypeptides that to a subject (e.g., a human).
  • the compositions, methods, and kits described herein can substantially increase the diversity, breadth, and/or depth of the virus-specific cellular immune responses by providing at least two distinct optimized viral polypeptides.
  • the present invention provides for polyvalent (e.g., 2-valent) vaccines that incorporate computationally-optimized viral polypeptides that correspond to and are derived from viral gene products that naturally circulate.
  • Polyvalent mosaic proteins are assembled from natural sequences by in silico recombination and optimized to provide maximal coverage of potential T cell epitopes (PTEs) for a given valency.
  • Mosaic antigens are full-length proteins that are designed to preserve natural antigen expression and processing.
  • a 2-valent vaccine elicits a substantially higher number of cellular immune responses (e.g., T cell responses) than conventional monovalent or polyvalent vaccines that incorporate naturally-occurring polypeptides derived from the same viral gene product (e.g., sequences based on clinical isolates), or a consensus sequence of such naturally-occurring polypeptides derived from the same viral gene product.
  • a vaccine that incorporates computationally-optimized viral polypeptides, the sequences of which provide maximum coverage of non-rare short stretches of circulating viral sequences, can increase the breadth and depth of the immune response.
  • a genetic algorithm is used to create sets of optimized viral polypeptides as "mosaic” blends of fragments of an arbitrary set of naturally-occurring viral gene product sequences provided as inputs.
  • This genetic algorithm strategy uses unaligned protein sequences from a general viral population as an input data set, and thus has the virtue of being “alignment independent.” It creates artificial optimized viral polypeptides that resemble viral proteins found in nature, but are not naturally- occurring.
  • the genetic algorithm can be adjusted to optimize viral polypeptides of different lengths, depending on the intended target or desired immune response.
  • the genetic algorithm utilized to design the optimized viral polypeptides of the invention was based on optimizing each consecutive 9-mer amino acid sequence of a given viral gene product (e.g., HIV-I Gag).
  • a given viral gene product e.g., HIV-I Gag.
  • 9-mers for example
  • that do not exist in nature or that are very rare can be excluded - this is an improvement relative to consensus sequence-based vaccine strategies since the latter can contain some 9-mers (for example) that occur rarely or not at all in nature.
  • the definition of fitness used for the genetic algorithm is that the most "fit" polyvalent cocktail is the combination of input viral sequences that gives the best coverage (highest fraction of perfect matches) of all of the 9 mers in the population and is subject to the constraint that no 9 mer is absent or rare in the population.
  • the genetic algorithm used to generate the optimized viral polypeptides of the invention is further described in International Patent Application Publication WO 2007/024941, herein incorporated by reference.
  • the invention provides polyvalent (e.g., 2-valent) HIV-I vaccines that incorporate single optimized HIV-I polypeptides (e.g., the polypeptides set forth in SEQ ID NOS: 1-29).
  • the invention features a polyvalent vaccine that incorporates two or more optimized HIV-I polypeptides.
  • the optimized HIV-I polypeptides are based on all HIV-I variants in global circulation, known as the HIV-I Main (M) group.
  • the inventors have generated a set of optimized HIV-I polypeptides (SEQ ID NOS: 1-29) that augment the breadth and depth of cellular immunity based on group M mosaic genes that utilize only two variants per gene (e.g., two polypeptide sequences each for Gag, Pol, Env, Nef, Tat, Rev, Vif, Vpr, and Vpu).
  • the invention provides for the fusion of optimized viral polypeptides that correspond to different viral gene products.
  • the genetic algorithm described above can be used to generate fused polypeptides for use in a vaccine of the invention.
  • the optimized HIV-I polypeptide fusions of Gag/Nef (SEQ ID NOS: 19-20), Gag/Pol (SEQ ID NOS:21-27), and Gag/Pol/Nef (SEQ ID NOS:28-29) can be incorporated into a vector of the invention for administration to a subject (e.g., a human) infected with or at risk of being infected with HIV-I.
  • the vaccines of the invention can also include one or more of the non-"mosaic" polypeptides (or sequences encoding them, respectively), such as, e.g., the optimal clade C sequences (SEQ ID NOS: 30-36) or the consensus sequences (SEQ ID NOS: 37-39).
  • the optimized viral polypeptides disclosed in this invention can be prepared conventionally by chemical synthesis techniques, such as described by Merrifield, J. Amer. Chem. Soc. 85:2149 (1963) (see also, e.g., Stemmer et al, 164 Gene 49 (1995)).
  • the vaccines can be readily prepared using solid phase peptide synthesis (SPPS).
  • SPPS solid phase peptide synthesis
  • Automated solid phase synthesis can be performed using any one of a number of well known, commercially available automated synthesizers, such as the Applied Biosystems ABI 433 A peptide synthesizer.
  • the optimized viral polypeptides of the invention can be recombinantly produced by transfecting or transducing a cell or organism with a nucleic acid or vector (e.g., a viral vector, such an adenovirus) that allows for the intracellular expression of the optimized viral polypeptide.
  • a nucleic acid or vector e.g., a viral vector, such an adenovirus
  • Nucleic acids and vectors that encode the nucleotide sequence of optimized viral polypeptides of the invention can be synthesized by well-known recombinant DNA techniques, including those described herein.
  • the invention also features vaccines that can be administered to a patient infected with or at risk of becoming infected with a virus (e.g., HIV-I).
  • a vaccine of the invention contains at least one of the optimized viral polypeptides of the invention, as discussed herein.
  • the vaccine of the invention can be a nucleic acid encoding the nucleotide sequence of two or more optimized viral polypeptides of the invention (e.g., the immunogenic component of a recombinant (e.g., subunit) or whole-organism (e.g., whole- virus) viral vector).
  • Nucleic acids include vectors (e.g., viral vectors, such as adenoviruses) that incorporate the nucleotide sequence of two or more optimized viral polypeptides of the invention.
  • the optimized viral polypeptides of the invention, as well as vaccines, nucleic acids, and vectors that incorporate optimized viral polypeptides, can be recombinantly expressed in a cell or organism, or can be directly administered to subject (e.g., a human) infected with, or at risk of becoming infected with, a virus.
  • the invention also features vectors encoding the nucleotide sequences (e.g., DNA or RNA) of one or more optimized viral polypeptides of the invention.
  • the vector can be a carrier (e.g., a liposome), a plasmid, a cosmid, a yeast artificial chromosome, or a virus that includes a nucleotide sequence encoding one or more optimized viral polypeptides of the invention.
  • the vector can include additional nucleic acid sequences from several sources.
  • Vectors encoding one or more optimized viral polypeptides of the invention can be constructed using any recombinant molecular biology technique known in the art.
  • the vector upon transfection or transduction of a target cell or organism, can be extrachromosomal or it can be integrated into the host cell chromosome.
  • the nucleic acid component of a vector can be in single or multiple copy number per target cell, and can be linear, circular, or concatamerized.
  • Vectors of the invention can also include internal ribosome entry site (IRES) sequences to allow for the expression of multiple peptide or polypeptide chains from a single nucleic acid transcript.
  • a vector of the invention can encode one or more optimized viral polypeptides of the invention as well as another polypeptides (e.g., a detectable label, such as green fluorescent protein (GFP)).
  • Vectors of the invention further include gene expression elements that facilitate the expression of optimized viral polypeptides of the invention.
  • Gene expression elements useful for the expression of an vector encoding an optimized viral polypeptide of the invention include, but are not limited to (a) regulatory sequences, such as viral transcription promoters and their enhancer elements, such as the SV40 early promoter, Rous sarcoma virus LTR, and Moloney murine leukemia virus LTR; (b) splice regions and polyadenylation sites such as those derived from the SV40 late region; and (c) polyadenylation sites such as in SV40.
  • regulatory sequences such as viral transcription promoters and their enhancer elements, such as the SV40 early promoter, Rous sarcoma virus LTR, and Moloney murine leukemia virus LTR
  • splice regions and polyadenylation sites such as those derived from the SV40 late region
  • polyadenylation sites such as in SV40.
  • plasmid origins of replication include antibiotic resistance or selection genes, multiple cloning sites (e.g., restriction enzyme cleavage loci), and other viral gene sequences (e.g., sequences encoding viral structural, functional, or regulatory elements, such as the HIV long terminal repeat (LTR)).
  • antibiotic resistance or selection genes include multiple cloning sites (e.g., restriction enzyme cleavage loci), and other viral gene sequences (e.g., sequences encoding viral structural, functional, or regulatory elements, such as the HIV long terminal repeat (LTR)).
  • multiple cloning sites e.g., restriction enzyme cleavage loci
  • other viral gene sequences e.g., sequences encoding viral structural, functional, or regulatory elements, such as the HIV long terminal repeat (LTR)
  • Vectors of the invention can also include optimized viral polypeptides of the invention that have been optimized for expression in humans, such as, e.g., any one of SEQ ID NOS:11, 14-18, and 23.
  • Vectors of the invention can also be engineered to include a multiple cloning site (MCS) having the following enzyme cleavage sites: Xbal-EcoRI-Kozak- Start...Stop-BamHI-NheI; and the following sequence: TCTAGA GAATTC GCCACC [ATG gene TAA TGA] GGATCC GCTAGC.
  • MCS multiple cloning site
  • Vectors having this MCS can be used with optimized viral polypeptides having no internal Xbal, EcoRI, BamHI, Nhel sites and no stretches of 6 or more Cs or G's.
  • the invention features methods for the in vivo administration of one or more vaccines of the invention (e.g., a vector encoding two or more optimized viral polypeptides of the invention) to a subject (e.g., a human) to facilitate the expression of two or more optimized viral polypeptides of the invention.
  • a subject e.g., a human
  • one or more optimized viral polypeptides of the invention will be expressed that can elicit protective or therapeutic immune responses (e.g., cellular or humoral immune responses) directed against the viral immunogens.
  • Vectors of the invention include viruses, naked DNA, oligonucleotides, cationic lipids (e.g., liposomes), cationic polymers (e.g., polysomes), virosomes, and dendrimers.
  • the present invention provides for the ex vivo transfection or transduction of cells (e.g., blood cells) followed by administration of these cells back into the donor subject to allow for the expression of optimized viral polypeptides of the invention that have immunogenic properties.
  • Cells that can be isolated and transfected or transduced ex vivo according to the methods of invention include, but are not limited to, blood cells, skin cells, fibroblasts, endothelial cells, skeletal muscle cells, hepatocytes, prostate epithelial cells, and vascular endothelial cells.
  • Stem cells are also appropriate cells for transduction or transfection with a vector of the invention.
  • Totipotent, pluripotent, multipotent, or unipotent stem cells including bone marrow progenitor cells and hematopoietic stem cells (HSC)
  • HSC hematopoietic stem cells
  • the method of transfection or transduction used to express an optimized viral vector of the invention has a strong influence on the strength and longevity of protein expression in the transfected or transduced cell, and subsequently, in the subject receiving the cell.
  • the present invention provides vectors that are temporal (e.g., adenoviral vectors) or long-lived (e.g., retroviral vectors) in nature. Regulatory sequences (e.g., promoters and enhancers) are known in the art that can be used to regulate protein expression.
  • the type of cell being transfected or transduced also has a strong bearing on the strength and longevity of protein expression. For example, cell types with high rates of turnover can be expected to have shorter periods of protein expression.
  • the invention also features methods for the ex vivo transfection and transduction of cells (e.g., blood cells, such as lymphocytes), followed by administration of these cells to a subject (e.g., a human).
  • the cells are autologous to the treated subject.
  • Cells can be transfected or transduced ex vivo with one or more vectors encoding the nucleotide sequence of one or more optimized viral polypeptides of the invention to allow for the temporal or permanent expression of the optimized viral polypeptides in the treated subject.
  • one or more optimized viral vectors of the invention will be expressed that can elicit protective or therapeutic immune responses (e.g., cellular or humoral immune responses) directed against the viral immunogens.
  • Vectors of the invention include viruses, naked DNA, oligonucleotides, cationic lipids (e.g., liposomes), cationic polymers (e.g., polysomes), virosomes, and dendrimers.
  • the present invention provides for the ex vivo transfection or transduction of cells (e.g., blood cells) followed by administration of these cells back into the donor subject to allow for the expression of optimized viral polypeptides of the invention that have immunogenic properties.
  • Cells that can be isolated and transfected or transduced ex vivo according to the methods of invention include, but are not limited to, blood cells, skin cells, fibroblasts, endothelial cells, skeletal muscle cells, hepatocytes, prostate epithelial cells, and vascular endothelial cells.
  • Stem cells are also appropriate cells for transduction or transfection with a vector of the invention.
  • Totipotent, pluripotent, multipotent, or unipotent stem cells including bone marrow progenitor cells and hematopoietic stem cells (HSC)
  • HSC hematopoietic stem cells
  • the method of transfection or transduction used to express an optimized viral vector of the invention has a strong influence on the strength and longevity of protein expression in the transfected or transduced cell, and subsequently, in the subject receiving the cell.
  • the present invention provides vectors that are temporal (e.g., adenoviral vectors) or long-lived (e.g., retroviral vectors) in nature. Regulatory sequences (e.g., promoters and enhancers) are known in the art that can be used to regulate protein expression.
  • the type of cell being transfected or transduced also has a strong bearing on the strength and longevity of protein expression. For example, cell types with high rates of turnover can be expected to have shorter periods of protein expression.
  • Viral vectors encoding the nucleotide sequence of one or more optimized viral polypeptides of the invention can be used as a vaccine of the invention.
  • the nucleotide sequence of one or more optimized viral polypeptides of the invention can be inserted recombinantly into that of a natural or modified (e.g., attenuated) viral genome suitable for the transduction of a subject (e.g., in vivo administration) or cells isolated from a subject (e.g., for ex vivo transduction followed by administration of the cells back to the subject).
  • a vector of the invention can be expressed by the transduced cell and secreted into the extracellular space or remain with the expressing cell (e.g., as an intracellular molecule or displayed on the cell surface). Chimeric or pseudotyped viral vectors can also be used to transduce a cell to allow for expression of one or more optimized viral polypeptides of the invention. Exemplary vectors are described below.
  • adenoviruses offer several significant advantages for use as vectors for the expression of one or more optimized viral polypeptides of the invention.
  • the viruses can be prepared to high titer, can infect non-replicating cells, and can confer high-efficiency transduction of target cells ex vivo following contact with a target cell population.
  • adenoviruses do not integrate their DNA into the host genome. Thus, their use as expression vectors has a reduced risk of inducing spontaneous proliferative disorders.
  • adenoviral vectors In animal models, adenoviral vectors have generally been found to mediate high-level expression for approximately one week.
  • transgene expression expression of a nucleic acid encoding an optimized viral polypeptide of the invention
  • duration of transgene expression can be prolonged by using cell or tissue- specific promoters.
  • Other improvements in the molecular engineering of the adenoviral vector itself have produced more sustained transgene expression and less inflammation. This is seen with so-called "second generation" vectors harboring specific mutations in additional early adenoviral genes and "gutless” vectors in which virtually all the viral genes are deleted utilizing a Cre-Lox strategy (Engelhardt et al., Proc. Natl. Acad. ScL USA 91 :6196 (1994) and Kochanek et al., Proc. Natl. Acad. ScL USA 93:5731 (1996), each herein incorporated by reference).
  • the rare serotype and chimeric adenoviral vectors disclosed in International Patent Application Publications WO 2006/040330 and WO 2007/104792, each incorporated by reference herein, are particularly useful as vectors of the invention.
  • recombinant adenoviruses rAd26, rAd34, rAd35, rAd48, and rAd5HVR48 can encode one or more optimized viral polypeptides of the invention.
  • One or more recombinant viral vectors encoding optimized viral polypeptides of the invention can be administered to a subject to treat or prevent a viral infection.
  • AAV Adeno-Associated Viruses
  • Adeno-associated viruses derived from non-pathogenic parvoviruses, can also be used to express optimized viral polypeptides of the invention as these vectors evoke almost no anti-vector cellular immune response, and produce transgene expression lasting months in most experimental systems.
  • Retroviruses are useful for the expression of optimized viral polypeptides of the invention. Unlike adenoviruses, the retroviral genome is based in RNA. When a retrovirus infects a cell, it will introduce its RNA together with several enzymes into the cell. The viral RNA molecules from the retrovirus will produce a double-stranded DNA copy, called a provirus, through a process called reverse transcription. Following transport into the cell nucleus, the proviral DNA is integrated in a host cell chromosome, permanently altering the genome of the transduced cell and any progeny cells that may derive from this cell. The ability to permanently introduce a gene into a cell or organism is the defining characteristic of retroviruses used for gene therapy.
  • Retroviruses include lentiviruses, a family of viruses including human immunodeficiency virus (HIV) that includes several accessory proteins to facilitate viral infection and proviral integration.
  • HIV human immunodeficiency virus
  • Current, "third-generation,” lentiviral vectors feature total replication incompetence, broad tropism, and increased gene transfer capacity for mammalian cells (see, e.g., Mangeat and and Trono, Human Gene Therapy 16(8):913 (2005) and Wiznerowicz and Trono, Trends Biotechnol. 23(1):42 (2005), each herein incorporated by reference).
  • viruses include Poxviruses (e.g., vaccinia virus and modified vaccinia virus Ankara or (MVA); see, e.g., U.S. Patent Nos. 4,603,112 and 5,762,938, each incorporated by reference herein), Herpesviruses, Togaviruses (e.g., Venezuelan Equine Encephalitis virus; see, e.g., U.S. Patent No.
  • Poxviruses e.g., vaccinia virus and modified vaccinia virus Ankara or (MVA); see, e.g., U.S. Patent Nos. 4,603,112 and 5,762,938, each incorporated by reference herein
  • Herpesviruses e.g., Venezuelan Equine Encephalitis virus; see, e.g., U.S. Patent No.
  • Picornaviruses e.g., poliovirus; see, e.g., U.S. Patent No. 5,639,649, incorporated by reference herein
  • Baculoviruses and others described by Wattanapitayakul and Bauer (Biomed. Pharmacother, 54:487 (2000), incorporated by reference herein).
  • Naked DNA or oligonucleotides encoding one or more optimized viral polypeptides of the invention can also be used to express these polypeptides in a cell (e.g., a blood cell, such as a lymphocyte) or subject (e.g., a human).
  • a cell e.g., a blood cell, such as a lymphocyte
  • subject e.g., a human
  • a cell e.g., a blood cell, such as a lymphocyte
  • a human e.g., a human
  • This is the simplest method of non- viral transfection.
  • Efficient methods for delivery of naked DNA exist such as electroporation and the use of a "gene gun," which shoots DNA-coated gold particles into a cell using high pressure gas and carrier particles (e.g., gold).
  • lipoplexes e.g., liposomes
  • polyplexes can be used to protect the vector DNA from undesirable degradation during the transfection process.
  • Plasmid DNA can be covered with lipids in an organized structure like a micelle or a liposome. When the organized structure is complexed with DNA it is called a lipoplex.
  • lipids There are three types of lipids, anionic (negatively-charged), neutral, or cationic (positively-charged).
  • Lipoplexes that utilize cationic lipids have proven utility for gene transfer. Cationic lipids, due to their positive charge, naturally complex with the negatively- charged DNA. Also as a result of their charge they interact with the cell membrane, endocytosis of the lipoplex occurs, and the DNA is released into the cytoplasm. The cationic lipids also protect against degradation of the DNA by the cell.
  • polyplexes Complexes of polymers with DNA are called polyplexes. Most polyplexes consist of cationic polymers and their production is regulated by ionic interactions.
  • One large difference between the methods of action of polyplexes and lipoplexes is that polyplexes cannot release their DNA load into the cytoplasm, so to this end, co- transfection with endosome-lytic agents (to lyse the endosome that is made during endocytosis) such as inactivated adenovirus must occur.
  • endosome-lytic agents to lyse the endosome that is made during endocytosis
  • polymers such as polyethylenimine have their own method of endosome disruption as does chitosan and trimethylchitosan.
  • Exemplary cationic lipids and polymers that can be used in combination with an nucleic acid encoding an optimized viral polypeptide of the invention to form lipoplexes, or polyplexes include, but are not limited to, polyethylenimine, lipofectin, lipofectamine, polylysine, chitosan, trimethylchitosan, and alginate.
  • Virosomes for example, combine lipoplexes (e.g., liposomes) with an inactivated virus. This approach has been shown to result in more efficient gene transfer in respiratory epithelial cells than either viral or liposomal methods alone.
  • Other methods involve mixing other viral vectors with cationic lipids or hybridising viruses. Each of these methods can be used to facilitate transfer of an nucleic acid encoding optimized viral polypeptides of the invention into a cell (e.g., a blood cell, such as a lymphocyte) or subject (e.g., a human).
  • Dendrimers e.g., a blood cell, such as a lymphocyte
  • Dendrimers may be also be used to transfer an nucleic acid encoding an optimized viral polypeptide of the invention into a cell (e.g., a blood cell, such as a lymphocyte) or subject (e.g., a human).
  • a dendrimer is a highly branched macromolecule with a spherical shape.
  • the surface of the particle may be functionalized in many ways, and many of the properties of the resulting construct are determined by its surface.
  • a cationic dendrimer i.e. one with a positive surface charge.
  • charge complimentarity leads to a temporary association of the nucleic acid with the cationic dendrimer.
  • the dendrimer- nucleic acid complex On reaching its destination the dendrimer- nucleic acid complex is then taken into the cell via endocytosis.
  • the invention also features in vivo methods for immunizing a subject (e.g., a human) with a vaccine of the invention.
  • a vaccine of the invention can be directly administered to a subject to elicit a protective or therapeutic immune response (e.g., a cellular or humoral immune response) against a virus (e.g., HIV- 1).
  • a vector encoding one or more optimized viral polypeptides of the invention, as described above can be directly administered to a subject to prevent or treat a viral infection.
  • a vector e.g., a viral vector
  • a vector that efficiently transfects or transduces one or more cells in vivo can elicit a broad, durable, and potent immune response in the treated subject.
  • the host cell Upon transfer of the nucleic acid component of the expression vector into a host cell (e.g., a blood cell, such as a lymphocyte), the host cell produces and displays or secretes the vaccine of the invention, which then serves to activate components of the immune system such as antigen-presenting cells (APCs), T cells, and B cells, resulting in the establishment of immunity.
  • a host cell e.g., a blood cell, such as a lymphocyte
  • APCs antigen-presenting cells
  • T cells T cells
  • B cells resulting in the establishment of immunity.
  • the invention features the vaccines, vectors, and optimized viral polypeptides of the invention in combination with one or more pharmaceutically acceptable excipients, diluents, buffers, or other acceptable carriers.
  • the formulation of a vaccine, vector, or optimized viral polypeptides will employ or allow expression of an effective amount of the optimized viral polypeptide immunogen. That is, there will be included an amount of antigen which will cause the treated subject (e.g., a human) to produce a specific and sufficient immunological response so as to impart protection to the subject from subsequent exposure to a virus (e.g., HIV-I) or to treat an existing viral infection.
  • a virus e.g., HIV-I
  • a formulation of a vaccine of the invention can allow for the expression of an amount of antigen which will cause the subject to produce a broad and specific cellular immune response.
  • a subject treated with a vaccine, vector, or optimized viral polypeptide of the invention can also produce anti- viral antibodies (e.g., neutralizing antibodies) which can confer a protective or therapeutic benefit to the subject.
  • a vaccine, vector, or optimized viral polypeptide of the invention can be directly administered to a subject, either alone or in combination with any pharmaceutically acceptable carrier, salt or adjuvant known in the art.
  • Pharmaceutically acceptable salts may include non-toxic acid addition salts or metal complexes that are commonly used in the pharmaceutical industry.
  • acid addition salts include organic acids such as acetic, lactic, pamoic, maleic, citric, malic, ascorbic, succinic, benzoic, palmitic, suberic, salicylic, tartaric, methanesulfonic, toluenesulfonic, or trifluoroacetic acids or the like; polymeric acids such as tannic acid, carboxymethyl cellulose, or the like; and inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid phosphoric acid, or the like.
  • Metal complexes include zinc, iron, and the like.
  • physiological saline One exemplary pharmaceutically acceptable carrier is physiological saline.
  • physiologically acceptable carriers and their formulations are known to one skilled in the art and described, for example, in Remington's Pharmaceutical Sciences, (18 th edition), ed. A. Gennaro, 1990, Mack Publishing Company, Easton, PA.
  • compositions of a prophylactically or therapeutically effective amount of a vaccine, vector, or optimized viral polypeptide of the invention can be administered orally, parenterally (e.g., intramuscular, intraperitoneal, intravenous, or subcutaneous injection, inhalation, intradermally, optical drops, or implant), nasally, vaginally, rectally, sublingually, or topically, in admixture with a pharmaceutically acceptable carrier adapted for the route of administration.
  • concentration of a vaccine, vector, or optimized viral polypeptide of the invention in the formulation can vary from about 0.1-100 wt. %.
  • Formulations for parenteral administration of compositions containing a vaccine, vector, or optimized viral polypeptide of the invention include sterile aqueous or non-aqueous solutions, suspensions, or emulsions.
  • suitable vehicles include propylene glycol, polyethylene glycol, vegetable oils, gelatin, hydrogenated naphalenes, and injectable organic esters, such as ethyl oleate.
  • Such formulations may also contain adjuvants, such as preserving, wetting, emulsifying, and dispersing agents.
  • Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds.
  • Other potentially useful parenteral delivery systems for compositions containing a vaccine, vector, or optimized viral polypeptide of the invention include ethylene- vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
  • Liquid formulations can be sterilized by, for example, filtration through a bacteria-retaining filter, by incorporating sterilizing agents into the compositions, or by irradiating or heating the compositions. Alternatively, they can also be manufactured in the form of sterile, solid compositions, which can be dissolved in sterile water or some other sterile injectable medium immediately before use.
  • compositions containing vaccine, vector, or optimized viral polypeptide of the invention for rectal or vaginal administration are preferably suppositories which may contain, in addition to active substances, excipients such as coca butter or a suppository wax.
  • Compositions for nasal or sublingual administration are also prepared with standard excipients known in the art.
  • Formulations for inhalation may contain excipients, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate and deoxycholate, or may be oily solutions for administration in the form of nasal drops or spray, or as a gel.
  • the amount of active ingredient in the compositions of the invention can be varied.
  • One skilled in the art will appreciate that the exact individual dosages may be adjusted somewhat depending upon a variety of factors, including the peptide being administered, the time of administration, the route of administration, the nature of the formulation, the rate of excretion, the nature of the subject's conditions, and the age, weight, health, and gender of the patient.
  • the severity of the condition treated by the vaccine, vector, or optimized viral polypeptide will also have an impact on the dosage level.
  • dosage levels of between 0.1 ⁇ g/kg to 100 mg/kg of body weight are administered daily as a single dose or divided into multiple doses.
  • the general dosage range is between 250 ⁇ g/kg to 5.0 mg/kg of body weight per day.
  • the amount of a vaccine, vector, or optimized viral polypeptide of the invention present in each dose given to a patient is selected with regard to consideration of the patient's age, weight, sex, general physical condition and the like.
  • the amount of a vaccine, vector, or optimized viral polypeptide required to induce an immune response (e.g., a cellular immune response) or produce an exogenous effect in the patient without significant adverse side effects varies depending upon the pharmaceutical composition employed and the optional presence of an adjuvant.
  • Initial doses can be optionally followed by repeated boosts, where desirable.
  • the method can involve chronically administering the vaccine, vector, or optimized viral polypeptide of the invention.
  • repeated dosages of the immunizing vaccine, vector, or optimized viral polypeptide can be desirable, such as a yearly booster or a booster at other intervals.
  • the dosage administered will, of course, vary depending upon known factors such as the pharmacodynamic characteristics of the particular vaccine, vector, or optimized viral polypeptide, and its mode and route of administration; age, health, and weight of the recipient; nature and extent of symptoms, kind of concurrent treatment, frequency of treatment, and the effect desired.
  • a vaccine, vector, or optimized viral polypeptide of the invention can be administered in chronic treatments for subjects at risk of acute infection due to needle sticks or maternal infection.
  • a dosage frequency for such "acute" infections may range from daily dosages to once or twice a week i.v.
  • the vaccine, vector, or optimized viral polypeptide can also be employed in chronic treatments for infected patients, or patients with advanced infection with a virus (e.g., HIV-I).
  • a virus e.g., HIV-I
  • the frequency of chronic administration can range from daily dosages to once or twice a week i.v. or i.m., and may depend upon the half-life of immunogen present in the vaccine, vector, or optimized viral polypeptide of the invention.
  • a vaccine of the invention used to vaccinate a mammal (e.g., a human) in need thereof against a virus can be administered concurrent with or in series with one or more pharmaceutically acceptable adjuvants to increase the immunogenicity of the vaccine.
  • adjuvants approved for human use include aluminum salts (alum). These adjuvants have been useful for some vaccines including hepatitis B, diphtheria, polio, rabies, and influenza.
  • CFA Complete Freund's Adjuvant
  • IFA Incomplete Freund's Adjuvant
  • MDP muramyl dipeptide
  • synthetic analogues of MDP N-acetylmuramyl-L-alanyl-D-isoglutamyl-L-alanine-2-[l,2- dipalmitoyl-s-gly- cero-3-(hydroxyphosphoryloxy)]ethylamide (MTP-PE) and compositions containing a metabolizable oil and an emulsifying agent, wherein the oil and emulsifying agent are present in the form of an oil-in-water emulsion having oil droplets substantially all of which are less than one micron in diameter.
  • kits that include a pharmaceutical composition containing a vaccine, vector, or optimized viral polypeptide of the invention, and a pharmaceutically-acceptable carrier, in a therapeutically effective amount for preventing or treating a viral infection.
  • kits include instructions to allow a clinician (e.g., a physician or nurse) to administer the composition contained therein.
  • kits include multiple packages of the single-dose pharmaceutical composition(s) containing an effective amount of a vaccine, vector, or optimized viral polypeptide of the invention.
  • instruments or devices necessary for administering the pharmaceutical composition(s) may be included in the kits.
  • a kit of this invention may provide one or more pre-filled syringes containing an effective amount of a vaccine, vector, or optimized viral polypeptide of the invention.
  • the kits may also include additional components such as instructions or administration schedules for a patient infected with or at risk of being infected with a virus to use the pharmaceutical composition(s) containing a vaccine, vector, or optimized viral polypeptide of the invention.
  • the mosaic antigen Gag, Pol, Nef, and Env sequences were constructed using the genetic algorithm discussed above. These sequences were then modified to make them practical for vaccine development by eliminating cleavage/fusion activity in Env (SEQ ID NOS:9-11), eliminating catalytic activity in Pol (SEQ ID NOS: 12- 14), eliminating myristylation sites in Nef (SEQ ID NOS:16- 18), and constructing fusion constructs including GagNef, GagPol, or GagPolNef (SEQ ID NOS: 19-29).
  • the comparator optimal natural clade C genes are also depicted (SEQ ID NOS:30-36).
  • M consensus (Group 1), 2-valent M mosaic (Group X), or optimal natural clade C (Group 3) sequences.
  • the M consensus sequences represent synthetic sequences that represent the single best "average" of circulating viruses worldwide.
  • the 2-valent M mosaic sequences are described above.
  • the optimal natural clade C sequences are naturally occurring sequences from actual clade C HIV-I viruses that are the most "consensus-like" in character.
  • Cellular immune breadth was assessed by evaluating the number of responding peptides from the global potential T cell epitope (PTE) peptide set.
  • PTE global potential T cell epitope
  • Group II 2-valent M readth Group I: M Consensus Group III: Natural Cla ⁇
  • Cellular immune breadth was assessed by evaluating the number of responding peptides from the global potential T cell epitope (PTE) peptide set.
  • PTE T cell epitope
  • the following table is a tally of the total responses to Gag, Pol, and Env responses to the three vaccines in the 7 animals vaccinated with 2 Mosaic (Mos2) or Mcon, and the 6 animals vaccinated with the Optimal Natural C clade (OptC):
  • the OptC vaccine yielded an average response across all monkeys that was slightly less than the CD 8+ T cell response per protein.
  • the Mcon vaccine exhibited ⁇ 1 response per protein. Only with Mos2 do we observe a difference in the proteins, where Env typically has fewer responses than either Gag or Pol.
  • the 2 mosaic vaccines elicit T cell responses that are capable of recognizing many more epitope-regions than the M consensus or a single optimal natural strain.
  • Mosaic HIV-I vaccines of the invention expand the breadth and depth of cellular immune responses in Rhesus monkeys.
  • a 2-valent mosaic strategy was utilized to balance the competing issues of theoretical coverage and practical utility.
  • 2-valent mosaic HIV-I Gag, Pol, and Env antigens substantially expanded the breadth and magnitude (depth) of epitope-specific CD8+ and CD4+ T lymphocyte responses in rhesus monkeys, relative to the immune response observed using consensus and natural sequence HIV-I antigens in rhesus monkeys.
  • a total dose of 3xlO 10 viral particles of rAd26 vectors expressing these antigens was administered once i.m. to each animal.
  • the optimal clade C antigens were the natural strain sequences selected to provide maximal PTE coverage of clade C sequences in the Los Alamos HIV-I sequence database (discussed in the Materials and Methods below).
  • the median number of CD8+ T lymphocyte responses was highest for the mosaic vaccine, followed by the consensus, the combined B+C, and the natural clade C vaccines (medians of 16, 5, 3, and 2 responses per animal in each group, respectively). Although there were fewer CD4+ T lymphocyte responses overall, the same relative pattern emerged with the highest number of CD4+ T lymphocyte responses to the mosaic vaccine, followed by the consensus, the combined B+C, and the natural clade C vaccines (medians of 4, 1, 1, and 0.5 responses per animal in each group, respectively). The numbers of CD8+ and CD4+ T lymphocyte responses elicited by the consensus, the combined B+C, and the natural clade C vaccines were not statistically distinguishable.
  • PTE peptides include multiple overlapping sequences that reflect naturally occurring HIV-I sequence polymorphisms, and thus the PTE peptide responses encompass both the recognition of a particular epitope (breadth) and the cross- recognition of variants of that epitope (depth).
  • breadth the number of reactive epi topic regions per monkey in which all reactive PTE peptides that overlapped by 8 or more amino acids were counted as one event.
  • Epitopic regions exhibited some clustering across animals, as evidenced by regions of high epitope density (Figs. 20A- 2OC and Figs. 21A-21C). Complete alignments of all positive peptides organized by response regions are shown in Fig. 22.
  • mosaic antigens substantially increased the breadth of cellular immune responses as compared with M consensus and natural clade C antigens.
  • the 2-valent mosaic antigens also proved superior to the 2-valent combination of clade B and clade C antigens (Fig. 19A and 19B), indicating that the enhanced breadth was due to the mosaic sequence design and did not simply reflect the use of two distinct antigenic sequences per protein.
  • mosaic antigens expanded cellular immune breadth without compromising the magnitude of individual epitope-specific responses, indicating that antigenic competition and immunodominance constraints did not limit the immunogenicity of the mosaic antigens in this study.
  • depth As the depth of the cellular immune responses elicited by the various vaccine regimens. We defined depth as the number of simultaneously elicited variant PTE peptides for a particular epitopic region. Inducing responses to multiple common epitope variants may increase immunologic coverage of infecting virus sequences, block common escape routes in vivo, or force the virus down tertiary escape routes that incur high fitness costs.
  • the consensus and natural sequence antigens elicited responses that were characterized by a high degree of sequence identity between the vaccine sequences and the reactive PTE peptides, as exemplified by the responses in monkey 366 that received the natural clade C antigens (Fig. 24A; see also Fig. 22).
  • the mosaic antigens elicited responses that were characterized by multiple reactive PTE peptides in particular epitopic regions. These peptides represented common variants and often reflected the polymorphisms contained in the mosaic vaccine sequences, as exemplified by the responses in monkey 361 (Fig. 24B; see also Fig. 22).
  • the mosaic antigens augmented cellular immune breadth not only to PTE peptides but also to actual Gag peptides from clades A, B, and C.
  • the mosaic antigens even proved superior to the optimal natural clade C antigens for inducing responses against clade C Gag peptides.
  • the mosaic antigens elicited comparable responses to Gag peptides from multiple clades, whereas the natural clade C antigens exhibited diminished responses to clade A and clade B Gag peptides (Fig. 25).
  • Gag-specific cellular immune responses has been shown to be critical for SIV control in rhesus monkeys and for HlV-I control in humans.
  • the rAd5 -based HIV-I vaccine candidate expressing natural clade B Gag, Pol, and Nef antigens elicited only a limited breadth of HIV-I -specific cellular immune responses, and no vaccine benefit was observed.
  • Vaccinees in the STEP study developed a median of only 2-3 epitope-specific T lymphocyte responses, including a median of only 1 epitope-specific response to Gag, and this very narrow breadth of cellular immune responses likely provided insufficient immunologic coverage of the diversity of infecting viruses.
  • Modeling the protective efficacy of mosaic vaccines against SIV challenges in nonhuman primates has intrinsic limitations, since the observed diversity of SIV and HIV-I M group sequences differs substantially and is influenced by different underlying biology. For example, CD8+ T lymphocyte selection pressure in natural hosts such as sooty mangabees appears substantially less than that in humans. Thus, the further evaluation of mosaic antigens as candidate HIV-I vaccines can be benefited by clinical trials.
  • Antigen design and vector production 2-valent mosaic Gag, Pol, and Env antigens were constructed to provide optimal coverage of HIV-I M group sequences in the Los Alamos HIV-I sequence database essentially as described (1, 1).
  • Optimal natural clade C antigens were selected to be the sequences that provide optimal PTE coverage of clade C sequences in the Los Alamos HIV-I sequence database (CIN.- .70177 Gag, C.ZA.04.04ZASK208B1 Pol, C.SN.90.90SE 364 Env).
  • Clade B antigens were selected to be near-consensus or consensus sequences (B.CAM-1 Gag, B.IIIB Pol, B.Con Env) and were used to complement the optimal clade C antigens for the 2-valent clade B + C vaccine approach.
  • Pol antigens contained RT and IN without PR and included point mutations to eliminate catalytic activity as described (Priddy et al., Clinical infectious diseases 46:1769, 2008).
  • Env gpl40 antigens contained point mutations to eliminate cleavage and fusion activity.
  • Vaccine sequences arc depicted in Fig. 27.
  • Recombinant, replication-incompetent adenovirus serotype 26 (rAd26) and hexon-chimeric rAd5HVR48 vectors expressing these antigens were grown in PER.55K cells and purified by double CsCl gradient sedimentation essentially as described (Abbink et al., J. Virol. 81:4654, 2007, and Roberts et al., Nature 441 :239, 2006).
  • IFN- ⁇ ELISPOT assays HIV-I -specific cellular immune responses in vaccinated monkeys were assessed by interferon- ⁇ (IFN- ⁇ ) ELISPOT assays essentially as described (Roberts et al., Nature 441:239, 2006, and Liu et al., Nature 457:87, 2009).
  • HIV-I Gag, Pol, and Env potential T cell epitope (PTE) peptides that included all PTEs found in at least 15% of HIV-I M group sequences as well as HIV- 1 Gag peptides from clade C DU422, clade C ZM651, consensus C, consensus A, and consensus B strains were obtained from the NIH AIDS Research and Reference Reagent Program.
  • 96-well multiscreen plates (Millipore) were coated overnight with 100 ⁇ l/well of 10 ⁇ g/ml anti-human IFN- ⁇ (BD Biosciences) in endotoxin-free Dulbecco's PBS (D-PBS).
  • the plates were then washed three times with D-PBS containing 0.25% Tween-20 (D-PB S/Tween), blocked for 2 h with D-PBS containing 5% FBS at 37 0 C, washed three times with D-PBS/Tween, rinsed with RPMI 1640 containing 10% FBS to remove the Tween-20, and incubated with 2 ⁇ g/ml each peptide and 2 x 10 5 PBMC in triplicate in 100 ⁇ l reaction volumes. Following an 18 h incubation at 37oC, the plates were washed nine times with PBS/Tween and once with distilled water.
  • the plates were then incubated with 2 ⁇ g/ml biotinylated anti- human IFN- ⁇ (BD Biosciences) for 2 h at room temperature, washed six times with PBS/Tween, and incubated for 2 h with a 1:500 dilution of streptavidin-alkaline phosphatase (Southern Biotechnology Associates). Following five washes with PBS/Tween and one with PBS, the plates were developed with nitro blue tetrazolium/5-bromo-4-chloro-3-indolyl -phosphate chromogen (Pierce), stopped by washing with tap water, air dried, and read using an ELISPOT reader (Cellular Technology Ltd). Spot-forming cells (SFC) per 10 6 PBMC were calculated. Media backgrounds were typically ⁇ 15 SFC per 10 6 PBMC. Positive responses were defined as > 55 SFC per 10 6 PBMC and > 4-fold background.
  • SFC spot-forming cells
  • Partial epitope mapping utilizing PTE subpools was also performed 4 weeks following the boost immunization at week 44. All borderline responses were retested and only considered positive if confirmed. Partial epitope mapping utilizing subpools containing 10 overlapping Gag peptides was also performed to assess breadth to HIV-I Gag from various clades.
  • Humoral immune assays Env-specif ⁇ c humoral immune responses were evaluated by direct ELISAs utilizing HIV-I clade C Env g ⁇ l40 and luciferase-based pseudovirus neutralization assays essentially as described (Montefiori, Evaluating neutralizing antibodies against HIV, SIV and SHIV in luciferase reporter gene assays. Current Protocols in Immunology, Coligan, Kruisbeek, Margulies, Shevach, Strober, and Coico, Ed. (John Wiley & Sons, 2004, pp. 1-15).
  • the 3.8-fold enhancement in the number of PTE peptides recognized by monkeys that received the mosaic antigens as compared to those that received the consensus or natural sequence antigens applied equally to PTEs from Gag, Pol, and Env and held for responses by CD8+ as well as CD4+ T lymphocytes.
  • the analysis of the number of reactive epitopic regions also included Poisson regression models with random effects and again fit well (dispersion parameter 0.87) without any significant interactions.
  • Comparisons of the magnitude of CD8+ and CD4+ T lymphocyte responses were performed utilizing 2-sided Kolmogorov- Smimov tests.
  • Non-parametric tests to compare the breadth and depth of responses per monkey between different vaccines were also performed (Figs. 19A and 24C).
  • To analyze the breadth of responses to HIV-I Gag from various clades (Fig. 25), we fit the data to binomial regression models. These models used the vaccine group as an explanatory variable and included random effects to account for animal-to-animal and strain-to- strain variation. The data were slightly underdispersed, but the animals that received the mosaic vaccine still elicited a significantly larger number of responses.
  • PTE coverage assessment was performed using tools available at the Los Alamos HIV-I sequence database.
  • MOSAIC ENVl GP160 (AA SEQUENCE) SEQ ID NO:1
  • MOSAIC ENV2 GP160 (AA SEQUENCE) SEQ ID NO:2
  • MOSAIC GAG2 (AA SEQUENCE) SEQ ID NO:4
  • MOSAIC POLl (AA SEQUENCE) SEQ ID NO:5
  • MOSAIC NEFL (AA SEQUENCE)
  • MOSAIC NEF2 (AA SEQUENCE) SEQ ID NO:8
  • MOSAIC ENVl GP140 (AA SEQUENCE) SEQ ID NO:9
  • MOSAIC ENV2 GP140 (AA SEQUENCE) SEQ ID NO:10
  • MOS3 ENV GP140 (AA SEQUENCE)
  • MOSAIC POLl (AA SEQUENCE) SEQ ID NO:12
  • MOSAIC POL2 (AA SEQUENCE) SEQ ID NO:13
  • MOS3 POL V3 (AA SEQUENCE)
  • MOS3 NEF (208 AA) SEQ ID NO:18

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Communicable Diseases (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • AIDS & HIV (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention porte sur des compositions, des procédés et des trousses pour le traitement ou la prévention d'infections virales. Les vaccins polyvalents (c'est-à-dire divalents) décrits incorporent des polypeptides viraux optimisés par ordinateur et qui peuvent augmenter la diversité ou la largeur et la profondeur de la réponse immunitaire cellulaire chez des sujets vaccinés.
PCT/US2009/064999 2008-11-18 2009-11-18 Vaccins antiviraux présentant une immunogénicité cellulaire améliorée WO2010059732A1 (fr)

Priority Applications (25)

Application Number Priority Date Filing Date Title
LTEP09828172.8T LT2358757T (lt) 2008-11-18 2009-11-18 Pagerinto ląstelinio imunogeniškumo priešvirusinės vakcinos
EP18191558.8A EP3470437A1 (fr) 2008-11-18 2009-11-18 Vaccins antiviraux présentant une immunogénicité cellulaire améliorée
SI200931885T SI2358757T1 (sl) 2008-11-18 2009-11-18 Antivirusna cepiva z izboljšano celularno imunogenostjo
AU2009316629A AU2009316629B2 (en) 2008-11-18 2009-11-18 Antiviral vaccines with improved cellular immunogenicity
NZ593598A NZ593598A (en) 2008-11-18 2009-11-18 Optimized antiviral vaccines with improved cellular immunogenicity
AP2011005767A AP3719A (en) 2008-11-18 2009-11-18 Antiviral vaccines with improved cellular immunogenicity
DK09828172.8T DK2358757T3 (da) 2008-11-18 2009-11-18 Antivirale vacciner med forbedret cellulær immunogenicitet
BR122021004758-8A BR122021004758B1 (pt) 2008-11-18 2009-11-18 Uso de um peptídeo viral otimizado, vacina e seu método de fabricação, vetor e kit
US13/130,018 US9017691B2 (en) 2008-11-18 2009-11-18 Antiviral vaccines with improved cellular immunogenicity
CN200980154787.4A CN102282175B (zh) 2008-11-18 2009-11-18 具有改进的细胞免疫原性的抗病毒疫苗
EP09828172.8A EP2358757B1 (fr) 2008-11-18 2009-11-18 Vaccins antiviraux présentant une immunogénicité cellulaire améliorée
BRPI0921588-3A BRPI0921588B1 (pt) 2008-11-18 2009-11-18 Vacina compreendendo um polipeptídeo viral e seu método de preparação, kit e uso do polipeptídeo viral
JP2011537586A JP5694945B2 (ja) 2008-11-18 2009-11-18 細胞性免疫原性が向上した抗ウイルスワクチン
PL09828172T PL2358757T3 (pl) 2008-11-18 2009-11-18 Szczepionki antywirusowe o ulepszonej immunogenności komórkowej
ES09828172T ES2699685T3 (es) 2008-11-18 2009-11-18 Vacunas antivíricas con inmunogenicidad celular mejorada
IL212984A IL212984A (en) 2008-11-18 2011-05-18 Vaccines to treat or reduce the risk of mammalian viral infection containing at least two differentiated optimized HIV-1 viral polypeptides and drugs containing them
ZA2011/04538A ZA201104538B (en) 2008-11-18 2011-06-20 Antiviral vaccines with improved cellular immunogenicity
HK12105453.5A HK1164896A1 (en) 2008-11-18 2012-06-05 Antiviral vaccines with improved cellular immunogenicity
US14/632,869 US9670253B2 (en) 2008-11-18 2015-02-26 Antiviral vaccines with improved cellular immunogenicity
IL243991A IL243991B (en) 2008-11-18 2016-02-07 Antiviral vaccines with enhanced cellular immunogenicity
US15/489,220 US10426831B2 (en) 2008-11-18 2017-04-17 Antiviral vaccines with improved cellular immunogenicity
HRP20181776TT HRP20181776T1 (hr) 2008-11-18 2018-10-29 Antivirusna cjepiva sa poboljšanom staničnom imunogenosti
CY20181101309T CY1121215T1 (el) 2008-11-18 2018-12-10 Αντιικα εμβολια με βελτιωμενη κυτταρικη ανοσογονικοτητα
US16/441,703 US10772952B2 (en) 2008-11-18 2019-06-14 Antiviral vaccines with improved cellular immunogenicity
US16/948,257 US11331386B2 (en) 2008-11-18 2020-09-10 Antiviral vaccines with improved cellular immunogenicity

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US11570308P 2008-11-18 2008-11-18
US61/115,703 2008-11-18
US15218409P 2009-02-12 2009-02-12
US61/152,184 2009-02-12
US24818809P 2009-10-02 2009-10-02
US61/248,188 2009-10-02

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/130,018 A-371-Of-International US9017691B2 (en) 2008-11-18 2009-11-18 Antiviral vaccines with improved cellular immunogenicity
US14/632,869 Continuation US9670253B2 (en) 2008-11-18 2015-02-26 Antiviral vaccines with improved cellular immunogenicity

Publications (1)

Publication Number Publication Date
WO2010059732A1 true WO2010059732A1 (fr) 2010-05-27

Family

ID=42198483

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/064999 WO2010059732A1 (fr) 2008-11-18 2009-11-18 Vaccins antiviraux présentant une immunogénicité cellulaire améliorée

Country Status (22)

Country Link
US (5) US9017691B2 (fr)
EP (2) EP3470437A1 (fr)
JP (4) JP5694945B2 (fr)
CN (1) CN102282175B (fr)
AP (1) AP3719A (fr)
AU (1) AU2009316629B2 (fr)
BR (2) BR122021004758B1 (fr)
CY (1) CY1121215T1 (fr)
DK (1) DK2358757T3 (fr)
ES (1) ES2699685T3 (fr)
HK (1) HK1164896A1 (fr)
HR (1) HRP20181776T1 (fr)
HU (1) HUE042397T2 (fr)
IL (2) IL212984A (fr)
LT (1) LT2358757T (fr)
NZ (2) NZ593598A (fr)
PL (1) PL2358757T3 (fr)
PT (1) PT2358757T (fr)
SG (1) SG10201408784SA (fr)
SI (1) SI2358757T1 (fr)
WO (1) WO2010059732A1 (fr)
ZA (2) ZA201104538B (fr)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014107744A1 (fr) 2013-01-07 2014-07-10 Beth Israel Deaconess Medical Center, Inc. Vaccins de trimère d'enveloppe (env) de virus d'immunodéficience humaine (vih) stabilisés et procédés d'utilisation de ceux-ci
WO2015051270A1 (fr) 2013-10-04 2015-04-09 Beth Israel Deaconess Medical Center, Inc. Vaccins de trimères d'enveloppe (env) de clade c du virus de l'immunodéficience humaine (vih) stabilisés et procédés d'utilisation de ceux-ci
US20150104790A1 (en) * 2013-10-16 2015-04-16 New England Biolabs, Inc. Reverse Transcriptase with Enhanced Properties
US9670253B2 (en) 2008-11-18 2017-06-06 Beth Israel Deaconess Medical Center, Inc. Antiviral vaccines with improved cellular immunogenicity
WO2017102929A1 (fr) * 2015-12-15 2017-06-22 Janssen Vaccines & Prevention B.V. Antigènes du virus de l'immunodéficience humaine, vecteurs, compositions, et leurs procédés d'utilisation
US9707289B2 (en) 2008-10-10 2017-07-18 Children's Medical Center Corporation Biochemically stabilized HIV-1 ENV trimer vaccine
WO2017216288A1 (fr) 2016-06-16 2017-12-21 Janssen Vaccines & Prevention B.V. Formulation de vaccin contre le vih
WO2018050747A1 (fr) 2016-09-15 2018-03-22 Janssen Vaccines & Prevention B.V. Mutations de protéine d'enveloppe du vih stabilisant la forme trimère
US10130705B2 (en) 2013-10-07 2018-11-20 The Trustees Of The University Of Pennsylvania Vaccines with interleukin-33 as an adjuvant
US10137191B2 (en) 2014-09-26 2018-11-27 Janssen Vaccines & Prevention B.V. Methods and compositions for inducing protective immunity against human immunodeficiency virus infection
WO2018229711A1 (fr) 2017-06-15 2018-12-20 Janssen Vaccines & Prevention B.V. Vecteurs à poxvirus codant pour des antigènes du vih, et leurs procédés d'utilisation
WO2019016062A1 (fr) 2017-07-19 2019-01-24 Janssen Vaccines & Prevention B.V. Mutations de protéine d'enveloppe du vih stabilisant la forme trimère
US10307477B2 (en) 2016-09-02 2019-06-04 Janssen Vaccines & Prevention B.V. Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
US10894078B2 (en) 2014-10-03 2021-01-19 Vir Biotechnology, Inc. HIV vaccines comprising one or more population episensus antigens
WO2021228816A1 (fr) 2020-05-12 2021-11-18 Janssen Vaccines & Prevention B.V. Administration de vecteurs adénoviraux homologues
US11230572B2 (en) 2016-10-17 2022-01-25 Beth Israel Deaconess Medical Center, Inc. Signature-based human immunodeficiency virus (HIV) envelope (Env) trimer vaccines and methods of using the same
WO2022084333A1 (fr) 2020-10-20 2022-04-28 Janssen Vaccines & Prevention B.V. Régimes de vaccin contre le vih
WO2022180007A1 (fr) 2021-02-23 2022-09-01 Janssen Vaccines & Prevention B.V. Mutation de protéine d'enveloppe du vih stabilisant un trimère
WO2023156505A1 (fr) 2022-02-17 2023-08-24 Janssen Vaccines & Prevention B.V. Trimère stabilisant les mutations de protéines d'enveloppe du vih r304v, n302m et t320l
WO2023198815A1 (fr) 2022-04-14 2023-10-19 Janssen Vaccines & Prevention B.V. Administration séquentielle d'adénovirus
US11795210B2 (en) 2019-07-16 2023-10-24 Gilead Sciences, Inc. HIV vaccines and methods of making and using
US11919926B2 (en) 2012-01-27 2024-03-05 Esteve Pharmaceuticals, S.A. Method of treating HIV-1 infection utilizing a multiepitope T cell immunogen comprising Gag, Pol, Vif, and Nef epitopes

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3200425A1 (fr) 2012-11-16 2014-05-22 Peter ABBINK Adenovirus recombinants et leur utilisation
US10376583B2 (en) 2013-09-30 2019-08-13 Beth Israel Deaconess Medical Center, Inc. Human immunodeficiency virus therapies utilizing N332-glycan-dependent antibodies and a reservoir activator
WO2015128421A1 (fr) * 2014-02-28 2015-09-03 Crucell Holland B.V. Vecteurs adénoviaux recombinés se répliquant, compositions et procédés pour les utiliser
EP3212209A4 (fr) * 2014-10-27 2018-10-03 Academia Sinica Peptides de signalisation de défense des plantes et leurs applications
WO2016196471A1 (fr) 2015-06-02 2016-12-08 Cooper Human Systems Llc Procédés et compositions pour le traitement d'une infection par le vih
US10542961B2 (en) 2015-06-15 2020-01-28 The Research Foundation For The State University Of New York System and method for infrasonic cardiac monitoring
WO2019018724A1 (fr) 2017-07-21 2019-01-24 Janssen Vaccines & Prevention B.V. Procédés d'induction sûre d'une immunité croisée multiclades contre une infection par le virus de l'immunodéficience humaine chez l'être humain
US20190083620A1 (en) 2017-09-18 2019-03-21 Janssen Vaccines & Prevention B.V. Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
EP3723771A4 (fr) 2017-12-11 2022-04-06 Beth Israel Deaconess Medical Center, Inc. Adénovirus recombinés et leurs utilisations
US11845788B2 (en) 2018-05-22 2023-12-19 Beth Israel Deaconess Medical Center, Inc. Antibody therapies for human immunodeficiency virus (HIV)
US11517618B2 (en) 2018-09-25 2022-12-06 Janssen Vaccines & Prevention B.V. Method of inducing an immune response against human immunodeficiency virus by co-localized administration of vaccine components
WO2020237052A1 (fr) 2019-05-22 2020-11-26 Janssen Vaccines & Prevention B.V. Procédés pour induire une réponse immunitaire contre une infection par le virus de l'immunodéficience humaine chez des sujets suivant un traitement antirétroviral
US11666651B2 (en) 2019-11-14 2023-06-06 Aelix Therapeutics, S.L. Prime/boost immunization regimen against HIV-1 utilizing a multiepitope T cell immunogen comprising Gag, Pol, Vif, and Nef epitopes
WO2021155323A1 (fr) 2020-01-31 2021-08-05 Beth Israel Deaconess Medical Center, Inc. Compositions et procédés de prévention et de traitement d'une infection par le coronavirus - vaccins contre le sars-cov-2
WO2021211748A1 (fr) * 2020-04-14 2021-10-21 The Regents Of The University Of California Compositions de vaccin universel contre tous les coronavirus

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4603112A (en) 1981-12-24 1986-07-29 Health Research, Incorporated Modified vaccinia virus
US5639649A (en) 1989-01-18 1997-06-17 Brtish Technology Group Limited Attenuated viruses
US5643576A (en) 1994-05-27 1997-07-01 The University Of North Carolina At Chapel Hill Method of inducing an immune response with a live Venezuelan Equine Encephalitis virus expressing a heterologous immunogen
US5762938A (en) 1991-03-07 1998-06-09 Virogenetics Corporation Modified recombinant vaccinia virus and expression vectors thereof
US20030207287A1 (en) * 1995-12-07 2003-11-06 Short Jay M. Non-stochastic generation of genetic vaccines
WO2006040330A2 (fr) 2004-10-13 2006-04-20 Crucell Holland B.V. Vecteurs adenoviraux ameliores et leurs utilisations
WO2007024941A2 (fr) 2005-08-23 2007-03-01 The Regents Of The University Of California Vaccin polyvalent
WO2007104792A2 (fr) 2006-03-16 2007-09-20 Crucell Holland B.V. Adénovirus recombinés basés sur les sérotypes 26 et 48 et utilisation de ceux-ci
US20080279879A1 (en) * 2006-11-17 2008-11-13 New York University INDUCTION OF BROADLY REACTIVE NEUTRALIZING ANTIBODIES BY FOCUSING THE IMMUNE RESPONSE ON V3 EPITOPES OF THE HIV-1 gp120 ENVELOPE

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050232900A1 (en) 1999-05-18 2005-10-20 Crucell Holland B.V. Serotype of adenovirus and uses thereof
US6913922B1 (en) 1999-05-18 2005-07-05 Crucell Holland B.V. Serotype of adenovirus and uses thereof
WO2002022080A2 (fr) * 2000-09-15 2002-03-21 Merck & Co., Inc. Vaccins adenoviraux de premiere generation evolues, exprimant les proteines gag, pol et nef du vih-1 a optimisation des codons et leurs modifications
US20040101957A1 (en) * 2001-09-14 2004-05-27 Emini Emilio A. Enhanced first generation adenovirus vaccines expressing codon optimized hiv1-gag, pol.nef and modifications
EP1461077A4 (fr) 2001-11-07 2006-01-25 Univ Duke Immunogene polyvalent
US7172761B2 (en) 2001-11-07 2007-02-06 Duke University Polyvalent immunogen
US7195768B2 (en) 2001-11-07 2007-03-27 Duke University Polyvalent immunogen
US20030219452A1 (en) 2001-11-27 2003-11-27 Haynes Barton F. HIV envelope V3-CCR5 binding site immunogen
PL208588B1 (pl) 2002-04-25 2011-05-31 Crucell Holland Bv Rekombinowane adenowirusy, wyizolowane kwasy nukleinowe, komórki pakujące oraz sposoby zwiększania stabilności i/lub pojemności upakowania rekombinowanego adenowirusa
AU2003288273A1 (en) 2002-10-23 2004-05-13 Crucell Holland B.V. New settings for recombinant adenoviral-based vaccines
US20080153083A1 (en) 2003-10-23 2008-06-26 Crucell Holland B.V. Settings for recombinant adenoviral-based vaccines
US20050221493A1 (en) 2002-12-04 2005-10-06 Crucell Holland B.V. Recombinant virus production for the manufacturing of vaccines
KR20120086379A (ko) 2003-09-17 2012-08-02 듀크 유니버시티 컨센서스/원형 면역원
US20070298051A1 (en) 2003-11-19 2007-12-27 Beth Israel Deaconess Medical Center Adjuvants Of Immune Response
BRPI0518933A2 (pt) 2004-11-16 2008-12-16 Crucell Holland B V E Aeras Gl adenovÍrus recombinante defeituoso de replicaÇço, vetor de polinucleotÍdeo recombinante, vacina multivalente da tuberculose, e, uso do antÍgeno tb10.4 de mycobacterium
US7951377B2 (en) 2005-08-23 2011-05-31 Los Alamos National Security, Llc Mosaic clade M human immunodeficiency virus type 1 (HIV-1) envelope immunogens
JP2010530356A (ja) 2007-03-27 2010-09-09 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 急性伝染hivエンベロープ・サイン
WO2010042817A1 (fr) 2008-10-10 2010-04-15 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Utilisation d'une mosaïque de protéines d'enveloppe du virus du vih en vue de l'optimisation de la réponse des lymphocytes t contre l'enveloppe du vih
DK2358757T3 (da) 2008-11-18 2019-01-02 Beth Israel Deaconess Medical Ct Inc Antivirale vacciner med forbedret cellulær immunogenicitet
WO2010096561A1 (fr) * 2009-02-18 2010-08-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Protéines gag du vih/vis de synthèse et leurs utilisations
US9017961B2 (en) 2012-03-05 2015-04-28 E.I. Du Pont De Nemours And Company Recombinant bacteria comprising novel sucrose transporters

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4603112A (en) 1981-12-24 1986-07-29 Health Research, Incorporated Modified vaccinia virus
US5639649A (en) 1989-01-18 1997-06-17 Brtish Technology Group Limited Attenuated viruses
US5762938A (en) 1991-03-07 1998-06-09 Virogenetics Corporation Modified recombinant vaccinia virus and expression vectors thereof
US5643576A (en) 1994-05-27 1997-07-01 The University Of North Carolina At Chapel Hill Method of inducing an immune response with a live Venezuelan Equine Encephalitis virus expressing a heterologous immunogen
US20030207287A1 (en) * 1995-12-07 2003-11-06 Short Jay M. Non-stochastic generation of genetic vaccines
WO2006040330A2 (fr) 2004-10-13 2006-04-20 Crucell Holland B.V. Vecteurs adenoviraux ameliores et leurs utilisations
US20080199939A1 (en) * 2004-10-13 2008-08-21 Crucell Holland B.V. Adenoviral Vectors and Uses Thereof
WO2007024941A2 (fr) 2005-08-23 2007-03-01 The Regents Of The University Of California Vaccin polyvalent
WO2007104792A2 (fr) 2006-03-16 2007-09-20 Crucell Holland B.V. Adénovirus recombinés basés sur les sérotypes 26 et 48 et utilisation de ceux-ci
US20080279879A1 (en) * 2006-11-17 2008-11-13 New York University INDUCTION OF BROADLY REACTIVE NEUTRALIZING ANTIBODIES BY FOCUSING THE IMMUNE RESPONSE ON V3 EPITOPES OF THE HIV-1 gp120 ENVELOPE

Non-Patent Citations (22)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Immunology", 2004, JOHN WILEY & SONS, pages: 1 - 15
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY
ABBINK ET AL., J. VIROL., vol. 81, 2007, pages 4654
BAROUCH ET AL., NAT. IINIIII NOL., vol. 6, 2005, pages 247
BLONDELLE ET AL.: "Immunogenically optimized peptides derived from natural mutants of HIV CTL epitopes and peptide combinatorial libraries.", PEPTIDE SCIENCE, vol. 90, no. 5, 15 May 2008 (2008-05-15), pages 683 - 694, XP008148830 *
COHEN, SCIENCE, vol. 259, 1993, pages 1691 - 1692
DATABASE GENBANK [online] 28 December 2005 (2005-12-28), "Envelope glycoprotein [Human immunodeficiency virus 1].", XP008149163, Database accession no. AAY23526 *
ENGELHARDT ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 6196
FISHER ET AL.: "Polyvalent Vaccine for Optimal Coverage of Potential T-Cell Epitopes in Global HIV-1 Variants", NAT. MED., vol. 13, no. 1, 2007, pages 100 - 106, XP007911386, DOI: doi:10.1038/nm1461
FISHER ET AL.: "Polyvalent Vaccine for Optimal Coverage of Potential T-Cell Epitopes in Global HΣV-I Variants", NAT. MED., vol. 13, no. 1, 2007, pages 100 - 106
FYNAN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 11478
KOCHANEK ET AL., PROC. NATL. ACAD. SCI. USA, vol. 93, 1996, pages 5731
LIU ET AL., NATURE, vol. 457, 2009, pages 87
MANGEAT; TRONO, HUMAN GENE THERAPY, vol. 16, no. 8, 2005, pages 913
MERRIFIELD, J. AMER. CHEM. SOC., vol. 85, 1963, pages 2149
PRIDDY ET AL., CLINICAL INFECTIOUS DISEASES, vol. 46, 2008, pages 1769
ROBERTS ET AL., NATURE, vol. 441, 2006, pages 239
See also references of EP2358757A4
STEMMER ET AL., GENE, vol. 164, 1995, pages 49
WATTANAPITAYAKUL; BAUER, BIOMED. PHARMACOTHER, vol. 54, 2000, pages 487
WIZNEROWICZ; TRONO, TRENDS BIOTECHNOL., vol. 23, no. 1, 2005, pages 42
WOLFF ET AL., BIOTECHNIQUES, vol. 11, 1991, pages 474485

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9950060B2 (en) 2008-10-10 2018-04-24 Beth Israel Deaconess Medical Center Biochemically stabilized HIV-1 Env trimer vaccine
US10653770B2 (en) 2008-10-10 2020-05-19 Beth Israel Deaconess Medical Center, Inc. Biochemically stabilized HIV-1 Env trimer vaccine
US10463729B2 (en) 2008-10-10 2019-11-05 Beth Israel Deaconess Medical Center, Inc. Biochemically stabilized HIV-1 env trimer vaccine
US10307478B2 (en) 2008-10-10 2019-06-04 Beth Israel Deaconess Medical Center, Inc. Biochemically stabilized HIV-1 Env trimer vaccine
US11229696B2 (en) 2008-10-10 2022-01-25 Beth Israel Deaconess Medical Center, Inc. Biochemically stabilized HIV-1 Env trimer vaccine
US9707289B2 (en) 2008-10-10 2017-07-18 Children's Medical Center Corporation Biochemically stabilized HIV-1 ENV trimer vaccine
US10772952B2 (en) 2008-11-18 2020-09-15 Beth Israel Deaconess Medical Center, Inc. Antiviral vaccines with improved cellular immunogenicity
US10426831B2 (en) 2008-11-18 2019-10-01 Beth Israel Deaconess Medical Center, Inc. Antiviral vaccines with improved cellular immunogenicity
US9670253B2 (en) 2008-11-18 2017-06-06 Beth Israel Deaconess Medical Center, Inc. Antiviral vaccines with improved cellular immunogenicity
US11331386B2 (en) 2008-11-18 2022-05-17 Beth Israel Deaconess Medical Center, Inc. Antiviral vaccines with improved cellular immunogenicity
US11919926B2 (en) 2012-01-27 2024-03-05 Esteve Pharmaceuticals, S.A. Method of treating HIV-1 infection utilizing a multiepitope T cell immunogen comprising Gag, Pol, Vif, and Nef epitopes
US10160788B2 (en) 2013-01-07 2018-12-25 Beth Israel Deaconess Medical Center, Inc. Stabilized human immunodeficiency virus (HIV) envelope (ENV) trimer vaccines and methods of using same
US20140302080A1 (en) * 2013-01-07 2014-10-09 Beth Israel Deaconess Medical Center, Inc. Stabilized human immunodeficiency virus (hiv) envelope (env) trimer vaccines and methods of using same
US9932370B2 (en) 2013-01-07 2018-04-03 Beth Israel Deaconess Medical Center, Inc. Stabilized mosaic human immunodeficiency virus type 1 (HIV-1) GP140 envelope (ENV) trimers
AU2014203886B2 (en) * 2013-01-07 2017-11-02 Beth Israel Deaconess Medical Center, Inc. Stabilized human immunodeficiency virus (HIV) envelope (Env) trimer vaccines and methods of using the same
EA030983B1 (ru) * 2013-01-07 2018-10-31 Бет Изрейэл Диконисс Медикал Сентер, Инк. ВАКЦИНЫ, СОДЕРЖАЩИЕ СТАБИЛИЗИРОВАННЫЕ ТРИМЕРЫ ОБОЛОЧКИ (Env) ВИРУСА ИММУНОДЕФИЦИТА ЧЕЛОВЕКА (ВИЧ), И СПОСОБЫ ИХ ПРИМЕНЕНИЯ
AU2018200696B2 (en) * 2013-01-07 2019-09-12 Beth Israel Deaconess Medical Center, Inc. Stabilized human immunodeficiency virus (hiv) envelope (env) trimer vaccines and methods of using the same
WO2014107744A1 (fr) 2013-01-07 2014-07-10 Beth Israel Deaconess Medical Center, Inc. Vaccins de trimère d'enveloppe (env) de virus d'immunodéficience humaine (vih) stabilisés et procédés d'utilisation de ceux-ci
KR20150118131A (ko) * 2013-01-07 2015-10-21 베쓰 이스라엘 디코니스 메디칼 센터 인크 안정화된 사람 면역결핍 바이러스 (hiv) 외피 (env) 삼량체 백신 및 이의 사용 방법
EP2983686A4 (fr) * 2013-01-07 2016-09-14 Beth Israel Hospital Vaccins de trimère d'enveloppe (env) de virus d'immunodéficience humaine (vih) stabilisés et procédés d'utilisation de ceux-ci
KR102020758B1 (ko) 2013-01-07 2019-09-11 베쓰 이스라엘 디코니스 메디칼 센터 인크 안정화된 사람 면역결핍 바이러스 (hiv) 외피 (env) 삼량체 백신 및 이의 사용 방법
US11059863B2 (en) 2013-01-07 2021-07-13 Beth Israel Deaconess Medical Center, Inc. Stabilized human immunodeficiency virus (HIV) envelope (ENV) trimer vaccines and methods of using same
US10716845B2 (en) 2013-10-04 2020-07-21 Beth Israel Deaconess Medical Center, Inc. Stabilized human immunodeficiency virus (HIV) clade C envelope (Env) trimer vaccines and methods of using same
WO2015051270A1 (fr) 2013-10-04 2015-04-09 Beth Israel Deaconess Medical Center, Inc. Vaccins de trimères d'enveloppe (env) de clade c du virus de l'immunodéficience humaine (vih) stabilisés et procédés d'utilisation de ceux-ci
US10130705B2 (en) 2013-10-07 2018-11-20 The Trustees Of The University Of Pennsylvania Vaccines with interleukin-33 as an adjuvant
US10933130B2 (en) 2013-10-07 2021-03-02 The Trustees Of The University Of Pennsylvania Vaccines with interleukin-33 as an adjuvant
WO2015057950A1 (fr) * 2013-10-16 2015-04-23 New England Biolabs, Inc. Transcriptase inverse ayant des propriétés améliorées
US20150104790A1 (en) * 2013-10-16 2015-04-16 New England Biolabs, Inc. Reverse Transcriptase with Enhanced Properties
US9920305B2 (en) 2013-10-16 2018-03-20 New England Biolabs, Inc. Reverse transcriptase with enhanced properties
US11207400B2 (en) 2014-09-26 2021-12-28 Janssen Vaccines & Prevention B.V. Compositions and methods for inducing protective immunity against human immunodeficiency virus infection
US10137191B2 (en) 2014-09-26 2018-11-27 Janssen Vaccines & Prevention B.V. Methods and compositions for inducing protective immunity against human immunodeficiency virus infection
US10471137B2 (en) 2014-09-26 2019-11-12 Janssen Vaccines & Prevention B.V. Methods for inducing protective immunity against human immunodeficiency virus infection
US10894078B2 (en) 2014-10-03 2021-01-19 Vir Biotechnology, Inc. HIV vaccines comprising one or more population episensus antigens
US11628215B2 (en) 2014-10-03 2023-04-18 Vir Biotechnology, Inc. HIV vaccines comprising one or more population episensus antigens
US11554168B2 (en) 2014-10-03 2023-01-17 Vir Biotechnology, Inc. HIV vaccines comprising one or more population episensus antigens
AU2019203468B2 (en) * 2015-12-15 2020-05-21 Janssen Vaccines & Prevention B.V. Human immunodeficiency virus antigens, vectors, compositions, and methods of use thereof
EP3964569A1 (fr) 2015-12-15 2022-03-09 Janssen Vaccines & Prevention B.V. Antigènes, vecteurs, compositions du virus de l'immunodéficience humaine et leurs procédés d'utilisation
EP3584252A1 (fr) 2015-12-15 2019-12-25 Janssen Vaccines & Prevention B.V. Antigènes, vecteurs, compositions du virus de l'immunodéficience humaine et leurs procédés d'utilisation
WO2017102929A1 (fr) * 2015-12-15 2017-06-22 Janssen Vaccines & Prevention B.V. Antigènes du virus de l'immunodéficience humaine, vecteurs, compositions, et leurs procédés d'utilisation
US11896663B2 (en) 2015-12-15 2024-02-13 Janssen Vaccines & Prevention B.V. Immunodeficiency virus type 1 (HIV-1) mutant envelope proteins
US10369214B2 (en) 2015-12-15 2019-08-06 Janssen Vaccines & Prevention B.V. Synthetic human immunodeficiency virus (HIV) envelope antigen, vectors, and compositions thereof
TWI792091B (zh) * 2015-12-15 2023-02-11 荷蘭商傑森疫苗防護公司 人類免疫缺陷病毒抗原、載體、組成物、及其使用方法
AU2016369326B2 (en) * 2015-12-15 2019-02-21 Janssen Vaccines & Prevention B.V. Human immunodeficiency virus antigens, vectors, compositions, and methods of use thereof
EA038974B1 (ru) * 2015-12-15 2021-11-17 Янссен Вэксинс Энд Превеншн Б.В. Антигены вируса иммунодефицита человека, векторы, композиции и способы их применения
US10973907B2 (en) 2015-12-15 2021-04-13 Janssen Vaccines & Prevention B.V. Recombinant adenoviruses encoding mosaic human immunodeficiency virus (HIV) Env, Gag, and Pol antigens
US10273268B2 (en) 2016-06-16 2019-04-30 Janssen Vaccines & Prevention B.V. HIV vaccine formulation
US10934328B2 (en) 2016-06-16 2021-03-02 Janssen Vaccines & Prevention B.V. HIV vaccine formulation
WO2017216288A1 (fr) 2016-06-16 2017-12-21 Janssen Vaccines & Prevention B.V. Formulation de vaccin contre le vih
US11603389B2 (en) 2016-06-16 2023-03-14 Janssen Vaccines & Prevention B.V. HIV vaccine formulation
US10525123B2 (en) 2016-09-02 2020-01-07 Janssen Vaccines & Prevention B.V. Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
US10307477B2 (en) 2016-09-02 2019-06-04 Janssen Vaccines & Prevention B.V. Methods for inducing an immune response against human immunodeficiency virus infection in subjects undergoing antiretroviral treatment
US10793607B2 (en) 2016-09-15 2020-10-06 Janssen Vaccines & Prevention B.V. Trimer stabilizing HIV envelope protein mutations
EP3747463A1 (fr) 2016-09-15 2020-12-09 Janssen Vaccines & Prevention B.V. Trimère de stabilisation de mutations de protéine d'enveloppe de vih
US11820796B2 (en) 2016-09-15 2023-11-21 Janssen Vaccines & Prevention B.V. Trimer stabilizing HIV envelope protein mutations
US11365222B2 (en) 2016-09-15 2022-06-21 Janssen Vaccines & Prevention B.V. Trimer stabilizing HIV envelope protein mutations
WO2018050747A1 (fr) 2016-09-15 2018-03-22 Janssen Vaccines & Prevention B.V. Mutations de protéine d'enveloppe du vih stabilisant la forme trimère
US11230572B2 (en) 2016-10-17 2022-01-25 Beth Israel Deaconess Medical Center, Inc. Signature-based human immunodeficiency virus (HIV) envelope (Env) trimer vaccines and methods of using the same
AU2018283811B2 (en) * 2017-06-15 2022-10-20 Bavarian Nordic A/S Poxvirus vectors encoding HIV antigens, and methods of use thereof
WO2018229711A1 (fr) 2017-06-15 2018-12-20 Janssen Vaccines & Prevention B.V. Vecteurs à poxvirus codant pour des antigènes du vih, et leurs procédés d'utilisation
US11229693B2 (en) 2017-06-15 2022-01-25 Janssen Vaccines & Prevention B.V. Poxvirus vectors encoding HIV antigens, and methods of use thereof
US11723970B2 (en) 2017-06-15 2023-08-15 Janssen Vaccines & Prevention B. V. Poxvirus vectors encoding HIV antigens, and methods of use thereof
US11732010B2 (en) 2017-07-19 2023-08-22 Janssen Vaccines & Prevention B.V. Trimer stabilizing HIV envelope protein mutations
WO2019016062A1 (fr) 2017-07-19 2019-01-24 Janssen Vaccines & Prevention B.V. Mutations de protéine d'enveloppe du vih stabilisant la forme trimère
US10968254B2 (en) 2017-07-19 2021-04-06 Janssen Vaccines & Prevention B.V. Trimer stabilizing HIV envelope protein mutations
US11795210B2 (en) 2019-07-16 2023-10-24 Gilead Sciences, Inc. HIV vaccines and methods of making and using
WO2021228816A1 (fr) 2020-05-12 2021-11-18 Janssen Vaccines & Prevention B.V. Administration de vecteurs adénoviraux homologues
WO2022084333A1 (fr) 2020-10-20 2022-04-28 Janssen Vaccines & Prevention B.V. Régimes de vaccin contre le vih
WO2022180007A1 (fr) 2021-02-23 2022-09-01 Janssen Vaccines & Prevention B.V. Mutation de protéine d'enveloppe du vih stabilisant un trimère
WO2023156505A1 (fr) 2022-02-17 2023-08-24 Janssen Vaccines & Prevention B.V. Trimère stabilisant les mutations de protéines d'enveloppe du vih r304v, n302m et t320l
WO2023198815A1 (fr) 2022-04-14 2023-10-19 Janssen Vaccines & Prevention B.V. Administration séquentielle d'adénovirus

Also Published As

Publication number Publication date
US20120076812A1 (en) 2012-03-29
IL243991A0 (en) 2016-04-21
EP2358757B1 (fr) 2018-09-12
PL2358757T3 (pl) 2019-02-28
CN102282175B (zh) 2015-09-30
IL243991B (en) 2019-12-31
PT2358757T (pt) 2018-12-04
HRP20181776T1 (hr) 2019-02-08
IL212984A (en) 2016-12-29
SI2358757T1 (sl) 2018-12-31
EP2358757A1 (fr) 2011-08-24
CN102282175A (zh) 2011-12-14
US11331386B2 (en) 2022-05-17
US9670253B2 (en) 2017-06-06
HK1164896A1 (en) 2012-09-28
US20170239344A1 (en) 2017-08-24
NZ593598A (en) 2013-02-22
US10426831B2 (en) 2019-10-01
JP2015134775A (ja) 2015-07-27
JP2012509340A (ja) 2012-04-19
EP2358757A4 (fr) 2013-07-31
US20200397890A1 (en) 2020-12-24
JP2018035177A (ja) 2018-03-08
AP2011005767A0 (en) 2011-06-30
ES2699685T3 (es) 2019-02-12
JP5694945B2 (ja) 2015-04-01
BR122021004758B1 (pt) 2022-04-19
BRPI0921588A2 (pt) 2017-06-06
NZ602504A (en) 2014-01-31
JP6368416B2 (ja) 2018-08-01
JP6023233B2 (ja) 2016-11-09
US20160024156A1 (en) 2016-01-28
HUE042397T2 (hu) 2019-06-28
CY1121215T1 (el) 2020-05-29
ZA201104538B (en) 2012-10-31
SG10201408784SA (en) 2015-02-27
AU2009316629A1 (en) 2011-07-07
EP3470437A1 (fr) 2019-04-17
US9017691B2 (en) 2015-04-28
DK2358757T3 (da) 2019-01-02
ZA201204999B (en) 2014-08-27
JP2017052762A (ja) 2017-03-16
AU2009316629B2 (en) 2016-02-04
AP3719A (en) 2016-06-30
IL212984A0 (en) 2011-07-31
US10772952B2 (en) 2020-09-15
BRPI0921588B1 (pt) 2021-12-28
LT2358757T (lt) 2018-11-26
JP6224197B2 (ja) 2017-11-01
US20190314492A1 (en) 2019-10-17

Similar Documents

Publication Publication Date Title
US11331386B2 (en) Antiviral vaccines with improved cellular immunogenicity
US8454972B2 (en) Method for inducing a multiclade immune response against HIV utilizing a multigene and multiclade immunogen
Mascola et al. Vaccines for the prevention of HIV-1 disease
US7323557B2 (en) Genome of the HIV-1 inter-subtype (C/B') and use thereof
ZA200504021B (en) Methods and compositions for immunization against HIV
EP2266602A2 (fr) Approches combinatoires destinées à produire des réponses immunitaires
EP1371730B1 (fr) Poxvirus recombine pour proteines chimeres du virus de l'immunodeficience humaine
US20090142373A1 (en) Immunizing Against HIV Infection
Ellenberger et al. Comparative immunogenicity in rhesus monkeys of multi-protein HIV-1 (CRF02_AG) DNA/MVA vaccines expressing mature and immature VLPs
WO2005047483A2 (fr) Renta: un immunogene du vih, et utilisations de renta
Singh et al. HIV vaccine development
EP1776961A1 (fr) Vaccination contre infections de VIH
Wang et al. Polyvalent DNA prime and envelope protein boost HIV‐1 vaccine elicits humoral and cellular responses and controls...
against Simian et al. A Replication-Competent
ZA200603029B (en) Combination approaches for generating immune responses

Legal Events

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

Ref document number: 200980154787.4

Country of ref document: CN

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

Ref document number: 09828172

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2011537586

Country of ref document: JP

Ref document number: 212984

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 593598

Country of ref document: NZ

Ref document number: 2009316629

Country of ref document: AU

Ref document number: 4698/DELNP/2011

Country of ref document: IN

Ref document number: 2009828172

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2009316629

Country of ref document: AU

Date of ref document: 20091118

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13130018

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 243991

Country of ref document: IL

ENP Entry into the national phase

Ref document number: PI0921588

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110518