US20040146528A1 - MVA expressing modified HIV envelope, gag, and pol genes - Google Patents

MVA expressing modified HIV envelope, gag, and pol genes Download PDF

Info

Publication number
US20040146528A1
US20040146528A1 US10/646,628 US64662803A US2004146528A1 US 20040146528 A1 US20040146528 A1 US 20040146528A1 US 64662803 A US64662803 A US 64662803A US 2004146528 A1 US2004146528 A1 US 2004146528A1
Authority
US
United States
Prior art keywords
gag
gene
hiv
pol
hiv env
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/646,628
Other languages
English (en)
Inventor
Bernard Moss
Linda Wyatt
Patricia Earl
Harriet Robinson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Health and Human Services
Emory University
Original Assignee
US Department of Health and Human Services
Emory University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by US Department of Health and Human Services, Emory University filed Critical US Department of Health and Human Services
Priority to US10/646,628 priority Critical patent/US20040146528A1/en
Assigned to GOVERNMENT OF THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment GOVERNMENT OF THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EARL, PATRICIA, MOSS, BERNARD, WYATT, LINDA
Assigned to EMORY UNIVERSITY reassignment EMORY UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROBINSON, HARRIET L.
Publication of US20040146528A1 publication Critical patent/US20040146528A1/en
Priority to US12/018,150 priority patent/US7867982B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: EMORY UNIVERSITY
Priority to US12/987,791 priority patent/US8916172B2/en
Priority to US14/566,066 priority patent/US20150238593A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH -DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH -DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: EMORY UNIVERSITY
Abandoned legal-status Critical Current

Links

Images

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
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • A61K2039/525Virus
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • 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
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24143Use 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
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24171Demonstrated 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
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15022New 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
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15034Use 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
    • C12N2740/16034Use 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
    • C12N2740/16122New 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
    • C12N2740/16211Human 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/15Vector systems having a special element relevant for transcription chimeric enhancer/promoter combination
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/60Vector systems having a special element relevant for transcription from viruses

Definitions

  • the invention provides modified vaccinia Ankara (MVA), a replication-deficient strain of vaccinia virus, expressing human immunodeficiency virus (HIV) env, gag, and pol genes.
  • VAA modified vaccinia Ankara
  • HAV human immunodeficiency virus
  • FIG. 1 Phylogenetic relationships of HIV-1 and HIV-2 based on identity of pol gene sequences.
  • SIV cpz and SIV smm are subhuman primate lentiviruses recovered from a chimpanzee and sooty mangabey monkey, respectively.
  • FIG. 2 Phylogenetic relationships of HIV-1 groups M, N and O with four different SIV cpz isolates based on full-length pol gene sequences.
  • the bar indicates a genetic distance of 0.1 (10% nucleotide divergence) and the asterisk positions group N HIV-1 isolates based on env sequences.
  • FIG. 3 Tropic and biologic properties of HIV-1 isolates.
  • FIG. 4 HIV-encoded proteins. The location of the HIV genes, the sizes of primary translation products (in some cases polyproteins), and the processed mature viral proteins are indicated.
  • FIG. 5 Schematic representation of a mature HIV-1 virion.
  • FIG. 6 Linear representation of the HIV-1 Env glycoprotein. The arrow indicates the site of gp160 cleavage to gp120 and gp41. In gp120, cross-hatched areas represent variable domains (V 1 to V 5 ) and open boxes depict conserved sequences (C 1 to C 5 ). In the gp41 ectodomain, several domains are indicated: the N-terminal fusion peptide, and the two ectodomain helices (N- and C-helix). The membrane-spanning domain is represented by a black box.
  • FIG. 7 Temporal frequencies of Gag-specific T cells.
  • A Gag-specific CD8 T cell responses raised by DNA priming and rMVA booster immunizations. The schematic presents mean Gag-CM9-tetramer data generated in the high-dose i.d. DNA-immunized animals.
  • B Gag-specific IFN- ⁇ ELISPOTs in A*01 (open bars) and non-A*01 (filled bars) macaques at various times before challenge and at two weeks after challenge. Three pools of 10 to 13 Gag peptides (22-mers overlapping by 12) were used for the analyses. The numbers above data bars represent the arithmetic mean ⁇ SD for the ELISPOTs within each group.
  • FIG. 8 Temporal viral loads, CD4 counts, and survival after challenge of vaccinated and control animals.
  • A Geometric mean viral loads and
  • B geometric mean CD4 counts.
  • C Survival curve for vaccinated and control animals. The dotted line represents all 24 vaccinated animals.
  • D Viral loads and
  • E CD4 counts for individual animals in the vaccine and control groups. The key to animal numbers is presented in (E). Assays for the first 12 weeks after challenge had a detection level of 1000 copies of RNA per milliliter of plasma. Animals with loads below 1000 were scored with a load of 500. For weeks 16 and 20, the detection level was 300 copies of RNA per milliliter. Animals with levels of virus below 300 were scored at 300.
  • FIG. 9 Postchallenge T cell responses in vaccine and control groups.
  • A Temporal tetramer + cells (dashed line) and viral loads (solid line).
  • B Intracellular cytokine assays for IFN- ⁇ production in response to stimulation with the Gag-CM9 peptide at two weeks after challenge. This ex vivo assay allows evaluation of the functional status of the peak postchallenge tetramer + cells displayed in FIG. 7A.
  • C Proliferation assay at 12 weeks after challenge. Gag-Pol-Env (open bars) and Gag-Pol (hatched bars) produced by transient transfections were used for stimulation. Supernatants from mock-transfected cultures served as control antigen. Stimulation indices are the growth of cultures in the presence of viral antigens divided by the growth of cultures in the presence of mock antigen.
  • FIG. 10 Lymph node histomorphology at 12 weeks after challenge.
  • A Typical lymph node from a vaccinated macaque showing evidence of follicular hyperplasia characterized by the presence of numerous secondary follicles with expanded germinal centers and discrete dark and light zones.
  • B Typical lymph node from an infected control animal showing follicular depletion and paracortical lymphocellular atrophy.
  • C A representative lymph node from an age-matched, uninfected macaque displaying nonreactive germinal centers.
  • D The percentage of the total lymph node area occupied by germinal centers was measured to give a non-specific indicator of follicular hyperplasia. Data for uninfected controls are for four age-matched rhesus macaques.
  • FIG. 11 Temporal antibody responses. Micrograms of total Gag (A) or Env (B) antibody were determined with ELISAs. The titers of neutralizing antibody for SHIV-89.6 (C) and SHIV-89.6P (D) were determined with MT-2 cell killing and neutral red staining (D. C. Montefiori et al. 1988 J Clin Microbiol 26:231). Titers are the reciprocal of the serum dilution giving 50% neutralization of the indicated viruses grown in human PBMC. Symbols for animals are the same as in FIG. 8.
  • FIG. 12 Gag-CM9-Mamu-A*01 tetramer-specific T cells in Mamu-A*01 vaccinated and control macaques at various times before challenge and at two weeks after challenge.
  • the number at the upper right corner of each plot represents the frequency of tetramer-specific CD8 T cells as a % of total CD8 T cells.
  • the numbers above each column of FACS data designate individual animals.
  • FIG. 13 Map of plasmid transfer vector pLW-48.
  • FIG. 14 Sequence of plasmid transfer vector pLW-48.
  • FIG. 15 Sequences of plasmid transfer vector pLW-48, Psy II promoter (which controls ADA envelope expression), ADA envelope truncated, PmH5 promoter (which controls HXB2 gag pol expression), and HXB2 gag pol (with safety mutations, ⁇ integrase).
  • FIG. 16 Plasmid transfer vector pLW-48 and making MVA recombinant virus MVA/HIV 48.
  • FIG. 17 A clade B gag pol.
  • FIG. 18 Sequence of new Psyn II promoter.
  • Vaccinia virus a member of the genus Orthopoxvirus in the family of Poxviridae, was used as live vaccine to immunize against the human smallpox disease.
  • Successful worldwide vaccination with vaccinia virus culminated in the eradication of variola virus, the causative agent of the smallpox (The global eradication of smallpox. Final report of the global commission for the certification of smallpox eradication. History of Public Health, No. 4, Geneva: World Health Organization, 1980). Since that WHO declaration, vaccination has been universally discontinued except for people at high risk of poxvirus infections (e.g. laboratory workers).
  • vaccinia viruses have also been used to engineer viral vectors for recombinant gene expression and for the potential use as recombinant live vaccines (Mackett, M. et al. 1982 PNAS USA 79:7415-7419; Smith, G. L. et al. 1984 Biotech Genet Engin Rev 2:383-407). This entails DNA sequences (genes) which code for foreign antigens being introduced, with the aid of DNA recombination techniques, into the genome of the vaccinia viruses.
  • the gene is integrated at a site in the viral DNA which is non-essential for the life cycle of the virus, it is possible for the newly produced recombinant vaccinia virus to be infectious, that is to say able to infect foreign cells and thus to express the integrated DNA sequence (EP Patent Applications No. 83,286 and No. 110,385).
  • the recombinant vaccinia viruses prepared in this way can be used, on the one hand, as live vaccines for the prophylaxis of infectious diseases, on the other hand, for the preparation of heterologous proteins in eukaryotic cells.
  • MVA modified vaccinia Ankara
  • CVA Ankara strain of vaccinia virus
  • MVA is distinguished by its great attenuation, that is to say by diminished virulence and ability to replicate in primate cells while maintaining good immunogenicity.
  • the MVA virus has been analyzed to determine alterations in the genome relative to the parental CVA strain. Six major deletions of genomic DNA (deletion I, II, III, IV, V, and VI) totaling 31,000 base pairs have been identified (Meyer, H. et al. 1991 J Gen Virol 72:1031-1038). The resulting MVA virus became severely host cell restricted to avian cells.
  • MVA is characterized by its extreme attenuation. When tested in a variety of animal models, MVA was proven to be avirulent even in immunosuppressed animals. More importantly, the excellent properties of the MVA strain have been demonstrated in extensive clinical trials (Mayr A. et al. 1978 Monilial Bakteriol [B] 167:375-390; Stickl et al. 1974 Dtsch Med Wschr 99:2386-2392). During these studies in over 120,000 humans, including high-risk patients, no side effects were associated with the use of MVA vaccine.
  • MVA replication in human cells was found to be blocked late in infection preventing the assembly to mature infectious virions. Nevertheless, MVA was able to express viral and recombinant genes at high levels even in non-permissive cells and was proposed to serve as an efficient and exceptionally safe gene expression vector (Sutter, G. and Moss, B. 1992 PNAS USA 89:10847-10851). Additionally, novel vaccinia vector vaccines were established on the basis of MVA having foreign DNA sequences inserted at the site of deletion III within the MVA genome (Sutter, G. et al. 1994 Vaccine 12:1032-1040).
  • the recombinant MVA vaccinia viruses can be prepared as set out hereinafter.
  • a DNA-construct which contains a DNA-sequence which codes for a foreign polypeptide flanked by MVA DNA sequences adjacent to a naturally occurring deletion, e.g. deletion III, or other non-essential sites, within the MVA genome is introduced into cells infected with MVA, to allow homologous recombination.
  • Once the DNA-construct has been introduced into the eukaryotic cell and the foreign DNA has recombined with the viral DNA, it is possible to isolate the desired recombinant vaccinia virus in a manner known per se, preferably with the aid of a marker.
  • the DNA-construct to be inserted can be linear or circular.
  • a plasmid or polymerase chain reaction product is preferred.
  • the DNA-construct contains sequences flanking the left and the right side of a naturally occurring deletion, e.g. deletion III, within the MVA genome.
  • the foreign DNA sequence is inserted between the sequences flanking the naturally occurring deletion.
  • regulatory sequences which are required for the transcription of the gene, to be present on the DNA.
  • promoters are known to those skilled in the art, and include for example those of the vaccinia 11 kDa gene as are described in EP-A-198,328, and those of the 7.5 kDa gene (EP-A-110,385).
  • the DNA-construct can be introduced into the MVA infected cells by transfection, for example by means of calcium phosphate precipitation (Graham et al. 1973 Virol 52:456-467; Wigler et al. 1979 Cell 16:777-785), by means of electroporation (Neumann et al. 1982 EMBO J 1:841-845), by microinjection (Graessmann et al. 1983 Meth Enzymol 101:482-492), by means of liposomes (Straubinger et al. 1983 Meth Enzymol 101:512-527), by means of spheroplasts (Schaffner 1980 PNAS USA 77:2163-2167) or by other methods known to those skilled in the art.
  • transfection for example by means of calcium phosphate precipitation (Graham et al. 1973 Virol 52:456-467; Wigler et al. 1979 Cell 16:777-785), by means of electroporation (Neumann et al. 1982 EMBO J 1:841
  • HIV-2 is more closely related to SIV smm , a virus isolated from sooty mangabey monkeys in the wild, than to HIV-1. It is currently believed that HIV-2 represents a zoonotic transmission of SIV smm to man.
  • SIV cpz A series of lentiviral isolates from captive chimpanzees, designated SIV cpz , are close genetic relatives of HIV-1.
  • the M group of HIV-1 which includes over 95% of the global virus isolates, consists of at least eight discrete clades (A, B, C, D, F, G, H, and J), based on the sequence of complete viral genomes.
  • HIV-1 group O has been recovered from individuals living in Cameroon, Gabon, and Equatorial Guinea; their genomes share less than 50% identity in nucleotide sequence with group M viruses.
  • group N HIV-1strains have been identified in infected Cameroonians, fail to react serologically in standard whole-virus enzyme-linked immunosorbent assay (ELISA), yet are readily detectable by conventional Western blot analysis.
  • ELISA enzyme-linked immunosorbent assay
  • HIV-1 genetic variation comes from studies of group M viruses of diverse geographic origin. Data collected during the past decade indicate that the HIV-1 population present within an infected individual can vary from 6% to 10% in nucleotide sequence. HIV-1 isolates within a clade may exhibit nucleotide distances of 15% in gag and up to 30% in gp120 coding sequences. Interclade genetic variation may range between 30% and 40% depending on the gene analyzed.
  • Clade A viruses are genetically the most divergent and were the most common HIV-1 subtype in Africa early in the epidemic. With the rapid spread of HIV-1 to southern Africa during the mid to late 1990s, clade C viruses have become the dominant subtype and now account for 48% of HIV-1 infections worldwide. Clade B viruses, the most intensively studied HIV-1 subtype, remain the most prevalent isolates in Europe and North America.
  • HIV-1 recombinants have arisen in Africa and a majority contain segments originally derived from clade A viruses.
  • the composition of the predominant circulating strain consists of a clade A gag plus pol gene segment and a clade E env gene. Because the clade E env gene in Thai HIV-1 strains is closely related to the lade E env present in virus isolates from the Central African Republic, it is believed that the original recombination event occurred in Africa, with the subsequent introduction of a descendent virus into Thailand. Interestingly, no full-length HIV-1 subtype E isolate (i.e., with subtype E gag, pol, and env genes) has been reported to date.
  • HIV-1 isolates using the CXCR4 receptor are usually T cell line (TCL)-tropic syncytium inducing (SI) strains, whereas those exclusively utilizing the CCR5 receptor (R5 viruses) are predominantly macrophage (M)-tropic and non-syncytium inducing (NSI).
  • TCL T cell line
  • SI macrophage
  • NBI non-syncytium inducing
  • the three primary HIV-1 translation products are initially synthesized as polyprotein precursors, which are subsequently processed by viral or cellular proteases into mature particle-associated proteins (FIG. 4).
  • the 55-kd Gag precursor Pr55 Gag is cleaved into the matrix (MA), capsid (CA), nucleocapsid (NC), and p6 proteins.
  • Retroviral Gag proteins are generally synthesized as polyprotein precursors; the HIV-1 Gag precursor has been named, based on its apparent molecular mass, Pr55 Gag .
  • the mRNA for Pr55 Gag is the unspliced 9.2-kb transcript (FIG. 4) that requires Rev for its expression in the cytoplasm.
  • the viral protease (PR) cleaves Pr55 Gag during or shortly after budding from the cell to generate the mature Gag proteins p17 (MA), p24 (CA), p7 (NC), and p6 (see FIG. 4).
  • MA is localized immediately inside the lipid bilayer of the viral envelope
  • CA forms the outer portion of the cone-shaped core structure in the center of the particle
  • NC is present in the core in a ribonucleoprotein complex with the viral RNA genome (FIG. 5).
  • the HIV Pr55 Gag precursor oligomerizes following its translation and is targeted to the plasma membrane, where particles of sufficient size and density to be visible by EM are assembled.
  • Formation of virus-like particles by Pr55 Gag is a self-assembly process, with critical Gag-Gag interactions taking place between multiple domains along the Gag precursor.
  • the assembly of virus-like particles does not require the participation of genomic RNA (although the presence of nucleic acid appears to be essential), pol-encoded enzymes, or Env glycoproteins, but the production of infectious virions requires the encapsidation of the viral RNA genome and the incorporation of the Env glycoproteins and the Gag-Pol polyprotein precursor Pr160 Gag-Pol .
  • telomeres Downstream of gag lies the most highly conserved region of the HIV genome, the pol gene, which encodes three enzymes: PR, RT, and IN (see FIG. 4).
  • RT and IN are required, respectively, for reverse transcription of the viral RNA genome to a double-stranded DNA copy, and for the integration of the viral DNA into the host cell chromosome.
  • PR plays a critical role late in the life cycle by mediating the production of mature, infectious virions.
  • the pol gene products are derived by enzymatic cleavage of a 160-kd Gag-Pol fusion protein, referred to as Pr160 Gag-Pol . This fusion protein is produced by ribosomal frameshifting during translation of Pr55 Gag (see FIG. 4).
  • the frame-shifting mechanism for Gag-Pol expression also utilized by many other retroviruses, ensures that the pol-derived proteins are expressed at a low level, approximately 5% to 10% that of Gag.
  • the N-terminus of Pr160 Gag-Pol is myristylated and targeted to the plasma membrane.
  • retroviral Gag proteins are initially synthesized as polyprotein precursors that are cleaved to generate smaller products. Subsequent studies demonstrated that the processing function is provided by a viral rather than a cellular enzyme, and that proteolytic digestion of the Gag and Gag-Pol precursors is essential for virus infectivity. Sequence analysis of retroviral PRs indicated that they are related to cellular “aspartic” proteases such as pepsin and renin. Like these cellular enzymes, retroviral PRs use two apposed Asp residues at the active site to coordinate a water molecule that catalyzes the hydrolysis of a peptide bond in the target protein.
  • retroviral PRs function as true dimers.
  • X-ray crystallographic data from HIV-1 PR indicate that the two monomers are held together in part by a four-stranded antiparallel ⁇ -sheet derived from both N- and C-terminal ends of each monomer.
  • the substrate-binding site is located within a cleft formed between the two monomers.
  • the HIV PR dimer contains flexible “flaps” that overhang the binding site and may stabilize the substrate within the cleft; the active-site Asp residues lie in the center of the dimer.
  • the primary sequences of retroviral PRs are highly divergent, yet their structures are remarkably similar.
  • Retroviral RTs have three enzymatic activities: (a) RNA-directed DNA polymerization (for minus-strand DNA synthesis), (b) RNaseH activity (for the degradation of the tRNA primer and genomic RNA present in DNA-RNA hybrid intermediates), and (c) DNA-directed DNA polymerization (for second- or plus-strand DNA synthesis).
  • the mature HIV-1 RT holoenzyme is a heterodimer of 66 and 51 kd subunits.
  • the 51-kd subunit (p51) is derived from the 66-kd (p66) subunit by proteolytic removal of the C-terminal 15-kd RNaseH domain of p66 by PR (see FIG. 4).
  • the crystal structure of HIV-1 RT reveals a highly asymmetric folding in which the orientations of the p66 and p51 subunits differ substantially.
  • the p66 subunit can be visualized as a right hand, with the polymerase active site within the palm, and a deep template-binding cleft formed by the palm, fingers, and thumb subdomains.
  • the polymerase domain is linked to RNaseH by the connection subdomain.
  • the active site located in the palm, contains three critical Asp residues (110, 185, and 186) in close proximity, and two coordinated Mg 2+ ions. Mutation of these Asp residues abolishes RT polymerizing activity.
  • the orientation of the three active-site Asp residues is similar to that observed in other DNA polymerases (e.g., the Klenow fragment of E. coli DNA poll).
  • the p51 subunit appears to be rigid and does not form a polymerizing cleft; Asp 110, 185, and 186 of this subunit are buried within the molecule.
  • Approximately 18 base pairs of the primer-template duplex lie in the nucleic acid binding cleft, stretching from the polymerase active site to the RNaseH domain.
  • the presence of a dideoxynucleotide at the 3′ end of the primer allows visualization of the catalytic complex trapped just prior to attack on the incoming dNTP.
  • Comparison with previously obtained structures suggests a model whereby the fingers close in to trap the template and dNTP prior to nucleophilic attack of the 3′-OH of the primer on the incoming dNTP.
  • the fingers adopt a more open configuration, thereby releasing the pyrophosphate and enabling RT to bind the next dNTP.
  • the structure of the HIV-1 RNaseH has also been determined by x-ray crystallography; this domain displays a global folding similar to that of E. coli RNaseH.
  • a distinguishing feature of retrovirus replication is the insertion of a DNA copy of the viral genome into the host cell chromosome following reverse transcription.
  • the integrated viral DNA (the provirus) serves as the template for the synthesis of viral RNAs and is maintained as part of the host cell genome for the lifetime of the infected cell.
  • Retroviral mutants deficient in the ability to integrate generally fail to establish a productive infection.
  • Retroviral IN proteins are composed of three structurally and functionally distinct domains: an N-terminal, zinc-finger-containing domain, a core domain, and a relatively nonconserved C-terminal domain. Because of its low solubility, it has not yet been possible to crystallize the entire 288-amino-acid HIV-1 IN protein. However, the structure of all three domains has been solved independently by x-ray crystallography or NMR methods. The crystal structure of the core domain of the avian sarcoma virus IN has also been determined.
  • the N-terminal domain (residues 1 to 55), whose structure was solved by NMR spectroscopy, is composed of four helices with a zinc coordinated by amino acids His-12, His-16, Cys-40, and Cys-43.
  • the structure of the N-terminal domain is reminiscent of helical DNA binding proteins that contain a so-called helix-turn-helix motif; however, in the HIV-1 structure this motif contributes to dimer formation.
  • helix-turn-helix motif a so-called helix-turn-helix motif
  • this motif contributes to dimer formation.
  • poor solubility hampered efforts to solve the structure of the core domain.
  • attempts at crystallography were successful when it was observed that a Phe-to-Lys change at IN residue 185 greatly increased solubility without disrupting in vitro catalytic activity.
  • Each monomer of the HIV-1 IN core domain (IN residues 50 to 212) is composed of a five-stranded ⁇ -sheet flanked by helices; this structure bears striking resemblance to other polynucleotidyl transferases including RNaseH and the bacteriophage MuA transposase.
  • Three highly conserved residues are found in analogous positions in other polynucleotidyl transferases; in HIV-1 IN these are Asp-64, Asp-116 and Glu-152, the so-called D,D-35-E motif. Mutations at these positions block HIV IN function both in vivo and in vitro.
  • the HIV Env glycoproteins play a major role in the virus life cycle. They contain the determinants that interact with the CD4 receptor and coreceptor, and they catalyze the fusion reaction between the lipid bilayer of the viral envelope and the host cell plasma membrane. In addition, the HIV Env glycoproteins contain epitopes that elicit immune responses that are important from both diagnostic and vaccine development perspectives.
  • the HIV Env glycoprotein is synthesized from the singly spliced 4.3-kb Vpu/Env bicistronic mRNA (see FIG. 4); translation occurs on ribosomes associated with the rough endoplasmic reticulum (ER).
  • the 160-kd polyprotein precursor (gp160) is an integral membrane protein that is anchored to cell membranes by a hydrophobic stop-transfer signal in the domain destined to be the mature TM Env glycoprotein, gp41 (FIG. 6).
  • the gp160 is cotranslationally glycosylated, forms disulfide bonds, and undergoes oligomerization in the ER.
  • the predominant oligomeric form appears to be a trimer, although dimers and tetramers are also observed.
  • the gp160 is transported to the Golgi, where, like other retroviral envelope precursor proteins, it is proteolytically cleaved by cellular enzymes to the mature SU glycoprotein gp120 and TM glycoprotein gp41 (see FIG. 6).
  • the cellular enzyme responsible for cleavage of retroviral Env precursors following a highly conserved Lys/Arg-X-Lys/Arg-Arg motif is furin or a furin-like protease, although other enzymes may also catalyze gp160 processing. Cleavage of gp160 is required for Env-induced fusion activity and virus infectivity.
  • gp120 and gp41 form a noncovalent association that is critical for transport of the Env complex from the Golgi to the cell surface.
  • the gp120-gp41 interaction is fairly weak, and a substantial amount of gp120 is shed from the surface of Env-expressing cells.
  • the HIV Env glycoprotein complex in particular the SU (gp120) domain, is very heavily glycosylated; approximately half the molecular mass of gp160 is composed of oligosaccharide side chains.
  • SU gp120
  • oligosaccharide side chains During transport of Env from its site of synthesis in the ER to the plasma membrane, many of the side chains are modified by the addition of complex sugars.
  • the numerous oligosaccharide side chains form what could be imagined as a sugar cloud obscuring much of gp120 from host immune recognition.
  • gp120 contains interspersed conserved (C 1 to C 5 ) and variable (V 1 to V 5 ) domains.
  • the Cys residues present in the gp120s of different isolates are highly conserved and form disulfide bonds that link the first four variable regions in large loops.
  • a primary function of viral Env glycoproteins is to promote a membrane fusion reaction between the lipid bilayers of the viral envelope and host cell membranes. This membrane fusion event enables the viral core to gain entry into the host cell cytoplasm.
  • a number of regions in both gp120 and gp41 have been implicated, directly or indirectly, in Env-mediated membrane fusion.
  • C-terminal to the gp41 fusion peptide are two amphipathic helical domains (see FIG. 6) which play a central role in membrane fusion. Mutations in the N-terminal helix (referred to as the N-helix), which contains a Leu zipper-like heptad repeat motif, impair infectivity and membrane fusion activity, and peptides derived from these sequences exhibit potent antiviral activity in culture.
  • the structure of the ectodomain of HIV-1 and SIV gp41, the two helical motifs in particular, has been the focus of structural analyses in recent years.
  • gp41 adopts a hypothetical conformation that allows the fusion peptide to insert into the target lipid bilayer.
  • the formation of the gp41 six-helix bundle brings the viral and cellular membranes together and membrane fusion takes place.
  • the present invention relates to generation of a CD8 + T cell immune response against an antigen and also eliciting an antibody response. More particularly, the present invention relates to “prime and boost” immunization regimes in which the immune response induced by administration of a priming composition is boosted by administration of a boosting composition.
  • the present invention is based on inventors' experimental demonstration that effective boosting can be achieved using modified vaccinia Ankara (MVA) vectors, following priming with any of a variety of different types of priming compositions including recombinant MVA itself.
  • MVA modified vaccinia Ankara
  • a major protective component of the immune response against a number of pathogens is mediated by T lymphocytes of the CD8 + type, also known as cytotoxic T lymphocytes (CTL).
  • T lymphocytes of the CD8 + type also known as cytotoxic T lymphocytes (CTL).
  • An important function of CD8 + cells is secretion of gamma interferon (IFN ⁇ ), and this provides a measure of CD8 + T cell immune response.
  • IFN ⁇ gamma interferon
  • a second component of the immune response is antibody directed to the proteins of the pathogen.
  • the present invention employs MVA which, as the experiments described below show, has been found to be an effective means for providing a boost to a CD8 + T cell immune response primed to antigen using any of a variety of different priming compositions and also eliciting an antibody response.
  • the experimental work described below demonstrates that use of embodiments of the present invention allows for recombinant MVA virus expressing an HIV antigen to boost a CD8 + T cell immune response primed by a DNA vaccine and also eliciting an antibody response.
  • the MVA was found to induce a CD8 + T cell response after intradermal, intramuscular or mucosal immunization.
  • Recombinant MVA has also been shown to prime an immune response that is boosted by one or more inoculations of recombinant MVA.
  • Non-human primates immunized with plasmid DNA and boosted with the MVA were effectively protected against intramucosal challenge with live virus.
  • a vaccination regime used intradermal, intramuscular or mucosal immunization for both prime and boost can be employed, constituting a general immunization regime suitable for inducing CD8 + T cells and also eliciting an antibody response, e.g. in humans.
  • the present invention in various aspects and embodiments employs an MVA vector encoding an HIV antigen for boosting a CD8 + T cell immune response to the antigen primed by previous administration of nucleic acid encoding the antigen and also eliciting an antibody response.
  • a general aspect of the present invention provides for the use of an MVA vector for boosting a CD8 + T cell immune response to an HIV antigen and also eliciting an antibody response.
  • One aspect of the present invention provides a method of boosting a CD8 + T cell immune response to an HIV antigen in an individual, and also eliciting an antibody response, the method including provision in the individual of an MVA vector including nucleic acid encoding the antigen operably linked to regulatory sequences for production of antigen in the individual by expression from the nucleic acid, whereby a CD8 + T cell immune response to the antigen previously primed in the individual is boosted.
  • An immune response to an HIV antigen may be primed by immunization with plasmid DNA or by infection with an infectious agent.
  • a further aspect of the invention provides a method of inducing a CD8 + T cell immune response to an HIV antigen in an individual, and also eliciting an antibody response, the method comprising administering to the individual a priming composition comprising nucleic acid encoding the antigen and then administering a boosting composition which comprises an MVA vector including nucleic acid encoding the antigen operably linked to regulatory sequences for production of antigen in the individual by expression from the nucleic acid.
  • a further aspect provides for use of an MVA vector, as disclosed, in the manufacture of a medicament for administration to a mammal to boost a CD8 + T cell immune response to an HIV antigen, and also eliciting an antibody response.
  • a medicament is generally for administration following prior administration of a priming composition comprising nucleic acid encoding the antigen.
  • the priming composition may comprise any viral vector, such as a vaccinia virus vector such as a replication-deficient strain such as modified vaccinia Ankara (MVA) or NYVAC (Tartaglia et al. 1992 Virology 118:217-232), an avipox vector such as fowlpox or canarypox, e.g. the strain known as ALVAC (Paoletti et al. 1994 Dev Biol Stand 82:65-69), or an adenovirus vector or a vesicular stomatitis virus vector or an alphavirus vector.
  • a viral vector such as a vaccinia virus vector such as a replication-deficient strain such as modified vaccinia Ankara (MVA) or NYVAC (Tartaglia et al. 1992 Virology 118:217-232), an avipox vector such as fowlpox or canarypox, e.g. the strain
  • the priming composition may comprise DNA encoding the antigen, such DNA preferably being in the form of a circular plasmid that is not capable of replicating in mammalian cells.
  • Any selectable marker should not be resistance to an antibiotic used clinically, so for example Kanamycin resistance is preferred to Ampicillin resistance.
  • Antigen expression should be driven by a promoter which is active in mammalian cells, for instance the cytomegalovirus immediate early (CMV IE) promoter.
  • CMV IE cytomegalovirus immediate early
  • a priming composition is followed by boosting with a boosting composition, or first and second boosting compositions, the first and second boosting compositions being the same or different from one another.
  • Still further boosting compositions may be employed without departing from the present invention.
  • a triple immunization regime employs DNA, then adenovirus as a first boosting composition, then MVA as a second boosting composition, optionally followed by a further (third) boosting composition or subsequent boosting administration of one or other or both of the same or different vectors.
  • DNA then MVA then adenovirus, optionally followed by subsequent boosting administration of one or other or both of the same or different vectors.
  • the antigen to be encoded in respective priming and boosting compositions need not be identical, but should share at least one CD8 + T cell epitope.
  • the antigen may correspond to a complete antigen, or a fragment thereof.
  • Peptide epitopes or artificial strings of epitopes may be employed, more efficiently cutting out unnecessary protein sequence in the antigen and encoding sequence in the vector or vectors.
  • One or more additional epitopes may be included, for instance epitopes which are recognized by T helper cells, especially epitopes recognized in individuals of different HLA types.
  • An HIV antigen of the invention to be encoded by a recombinant MVA virus includes polypeptides having immunogenic activity elicited by an amino acid sequence of an HIV Env, Gag, Pol, Vif, Vpr, Tat, Rev, Vpu, or Nef amino acid sequence as at least one CD8 + T cell epitope.
  • This amino acid sequence substantially corresponds to at least one 10-900 amino acid fragment and/or consensus sequence of a known HIV Env or Pol; or at least one 10-450 amino acid fragment and/or consensus sequence of a known HIV Gag; or at least one 10-100 amino acid fragment and/or consensus sequence of a known HIV Vif, Vpr, Tat, Rev, Vpu, or Nef.
  • Env is optionally deleted of subsequences. For example, regions of the gp120 surface and gp41 transmembrane cleavage products can be deleted.
  • Gag is optionally deleted of subsequences.
  • regions of the matrix protein (p17), regions of the capsid protein (p24), regions of the nucleocapsid protein (p7), and regions of p6 (the C-terminal peptide of the Gag polyprotein) can be deleted.
  • Pol is optionally deleted of subsequences.
  • regions of the protease protein (p10), regions of the reverse transcriptase protein (p66/p51), and regions of the integrase protein (p32) can be deleted.
  • Such an HIV Env, Gag, or Pol can have overall identity of at least 50% to a known Env, Gag, or Pol protein amino acid sequence, such as 50-99% identity, or any range or value therein, while eliciting an immunogenic response against at least one strain of an HIV.
  • Percent identify can be determined, for example, by comparing sequence information using the GAP computer program, version 6.0, available from the University of Wisconsin Genetics Computer Group (UWGCG).
  • the GAP program utilizes the alignment method of Needleman and Wunsch ( J Mol Biol 1970 48:443), as revised by Smith and Waterman ( Adv Appl Math 1981 2:482). Briefly, the GAP program defines identity as the number of aligned symbols (i.e., nucleotides or amino acids) which are identical, divided by the total number of symbols in the shorter of the two sequences.
  • the preferred default parameters for the GAP program include: (1) a unitary comparison matrix (containing a value of 1 for identities and 0 for non-identities) and the weighted comparison matrix of Gribskov and Burgess ( Nucl Acids Res 1986 14:6745), as described by Schwartz and Dayhoff (eds., Atlas of Protein Sequence and Structure , National Biomedical Research Foundation, Washington, D.C. 1979, pp. 353-358); (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps.
  • an Env of the present invention is a variant form of at least one HIV envelope protein.
  • the Env is composed of gp120 and the membrane-spanning and ectodomain of gp41 but lacks part or all of the cytoplasmic domain of gp41.
  • HIV sequences are readily available from commercial and institutional HIV sequence databases, such as GENBANK, or as published compilations, such as Myers et al. eds., Human Retroviruses and AIDS, A Compilation and Analysis of Nucleic Acid and Amino Acid Sequences , Vol. I and II, Theoretical Biology and Biophysics, Los Alamos, N. Mex. (1993), or http://hiv-web.lanl.gov/.
  • Substitutions or insertions of an HIV Env, Gag, or Pol to obtain an additional HIV Env, Gag, or Pol, encoded by a nucleic acid for use in a recombinant MVA virus of the present invention can include substitutions or insertions of at least one amino acid residue (e.g., 1-25 amino acids).
  • at least one amino acid e.g., 1-25 amino acids
  • substitutions, insertions or deletions are identified based on safety features, expression levels, immunogenicity and compatibility with high replication rates of MVA.
  • Amino acid sequence variations in an HIV Env, Gag, or Pol of the present invention can be prepared e.g., by mutations in the DNA.
  • HIV Env, Gag, or Pol include, for example, deletions, insertions or substitutions of nucleotides coding for different amino acid residues within the amino acid sequence.
  • mutations that will be made in nucleic acid encoding an HIV Env, Gag, or Pol must not place the sequence out of reading frame and preferably will not create complementary domains that could produce secondary mRNA structures.
  • HIV Env, Gag, or Pol-encoding nucleic acid of the present invention can also be prepared by amplification or site-directed mutagenesis of nucleotides in DNA or RNA encoding an HIV Env, Gag, or Pol and thereafter synthesizing or reverse transcribing the encoding DNA to produce DNA or RNA encoding an HIV Env, Gag, or Pol, based on the teaching and guidance presented herein.
  • Recombinant MVA viruses expressing HIV Env, Gag, or Pol of the present invention include a finite set of HIV Env, Gag, or Pol-encoding sequences as substitution nucleotides that can be routinely obtained by one of ordinary skill in the art, without undue experimentation, based on the teachings and guidance presented herein.
  • substitution nucleotides can be routinely obtained by one of ordinary skill in the art, without undue experimentation, based on the teachings and guidance presented herein.
  • For a detailed description of protein chemistry and structure see Schulz, G. E. et al., 1978 Principles of Protein Structure , Springer-Verlag, New York, N.Y., and Creighton, T. E., 1983 Proteins: Structure and Molecular Properties , W. H. Freeman & Co., San Francisco, Calif.
  • regulatory sequences for expression of the encoded antigen will include a natural, modified or synthetic poxvirus promoter.
  • promoter is meant a sequence of nucleotides from which transcription may be initiated of DNA operably linked downstream (i.e. in the 3′ direction on the sense strand of double-stranded DNA).
  • operably linked means joined as part of the same nucleic acid molecule, suitably positioned and oriented for transcription to be initiated from the promoter.
  • DNA operably linked to a promoter is “under transcriptional initiation regulation” of the promoter.
  • Terminator fragments include terminator fragments, polyadenylation sequences, marker genes and other sequences.
  • Other regulatory sequences including terminator fragments, polyadenylation sequences, marker genes and other sequences may be included as appropriate, in accordance with the knowledge and practice of the ordinary person skilled in the art: see, for example, Moss, B. (2001). Poxviridae: the viruses and their replication. In Fields Virology, D. M. Knipe, and P. M. Howley, eds. (Philadelphia, Lippincott Williams & Wilkins), pp. 2849-2883.
  • Promoters for use in aspects and embodiments of the present invention must be compatible with poxvirus expression systems and include natural, modified and synthetic sequences.
  • Either or both of the priming and boosting compositions may include an adjuvant, such as granulocyte macrophage-colony stimulating factor (GM-CSF) or encoding nucleic acid therefor.
  • an adjuvant such as granulocyte macrophage-colony stimulating factor (GM-CSF) or encoding nucleic acid therefor.
  • GM-CSF granulocyte macrophage-colony stimulating factor
  • Administration of the boosting composition is generally about 1 to 6 months after administration of the priming composition, preferably about 1 to 3 months.
  • priming composition is intradermal, intramuscular or mucosal immunization.
  • boosting composition is intradermal, intramuscular or mucosal immunization.
  • Administration of MVA vaccines may be achieved by using a needle to inject a suspension of the virus.
  • a needleless injection device to administer a virus suspension (using, e.g., BiojectorTM needleless injector) or a resuspended freeze-dried powder containing the vaccine, providing for manufacturing individually prepared doses that do not need cold storage. This would be a great advantage for a vaccine that is needed in rural areas of Africa.
  • MVA is a virus with an excellent safety record in human immunizations.
  • the generation of recombinant viruses can be accomplished simply, and they can be manufactured reproducibly in large quantities.
  • Intradermal, intramuscular or mucosal administration of recombinant MVA virus is therefore highly suitable for prophylactic or therapeutic vaccination of humans against AIDS which can be controlled by a CD8 + T cell response.
  • the individual may have AIDS such that delivery of the antigen and generation of a CD8 + T cell immune response to the antigen is of benefit or has a therapeutically beneficial effect.
  • administration will have prophylactic aim to generate an immune response against HIV or AIDS before infection or development of symptoms.
  • compositions may comprise a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • the precise nature of the carrier or other material may depend on the route of administration, e.g. intravenous, cutaneous or subcutaneous, nasal, intramuscular, intraperitoneal routes.
  • administration is preferably intradermal, intramuscular or mucosal.
  • Physiological saline solution dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
  • the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • a parenterally acceptable aqueous solution which is pyrogen-free and has suitable pH, isotonicity and stability.
  • isotonic vehicles such as Sodium Chloride Injection, Ringer's Injection, Lactated Ringer's Injection.
  • Preservatives, stabilizers, buffers, antioxidants and/or other additives may be included as required.
  • a slow-release formulation may be employed.
  • the particles may be administered to an individual, particularly human or other primate. Administration may be to another mammal, e.g. rodent such as mouse, rat or hamster, guinea pig, rabbit, sheep, goat, pig, horse, cow, donkey, dog or cat.
  • rodent such as mouse, rat or hamster, guinea pig, rabbit, sheep, goat, pig, horse, cow, donkey, dog or cat.
  • Administration is preferably in a “prophylactically effective amount” or a “therapeutically effective amount” (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual.
  • the actual amount administered, and rate and time-course of administration, will depend on the nature and severity of what is being treated. Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors, or in a veterinary context a veterinarian, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of the techniques and protocols mentioned above can be found in Remington's Pharmaceutical Sciences , 16th edition, 1980, Osol, A. (ed.).
  • DNA is administered at a dose of 250 ⁇ g to 2.5 mg/injection, followed by MVA at a dose of 10 6 to 10 9 infectious virus particles/injection.
  • a composition may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.
  • Delivery to a non-human mammal need not be for a therapeutic purpose, but may be for use in an experimental context, for instance in investigation of mechanisms of immune responses to an antigen of interest, e.g. protection against HIV or AIDS.
  • SHIV-89.6 The 89.6 chimera of simian and human immunodeficiency viruses (SHIV-89.6) was used for the construction of immunogens and its highly pathogenic derivative, SHIV-89.6P, for challenge (G. B. Karlsson et al. 1997 J Virol 71:4218). SHIV-89.6 and SHIV-89.6P do not generate cross-neutralizing antibody (D. C . . . Montefiori et al.
  • MVA Modified vaccinia Ankara
  • rMVA DNA and recombinant MVA
  • the DNA prime (DNA/89.6) expressed simian immunodeficiency virus (SIV) Gag, Pol, Vif, Vpx, and Vpr and human immunodeficiency virus-1 (HIV-1) Env, Tat, and Rev from a single transcript (R. J. Gorelick et al. 1999 Virology 253:259; M. M. Sauter et al. 1996 J Cell Biol 132:795).
  • SIV simian immunodeficiency virus
  • HIV-1 human immunodeficiency virus-1
  • SHIV-89.6 sequences were cloned into the vector pGA2 using ClaI and RsrII sites. This cloning deleted both long terminal repeats (LTRs) and nef.
  • the SHIV-89.6 sequences also were internally mutated for a 12-base pair region encoding the first four amino acids of the second zinc finger in nucleocapsid. This mutation renders SHIV viruses noninfectious (R. J. Gorelick et al. 1999 Virology 253:259).
  • a mutation in gp41 converted the tyrosine at position 710 to cysteine to achieve better expression of Env on the plasma membrane of DNA-expressing cells (M. M.
  • pGA2 uses the CMV immediate early promoter without intron A and the bovine growth hormone polyadenylation sequence to express vaccine inserts.
  • Vaccine DNA was produced by Althea (San Diego, Calif.). In transient transfections of 293T cells, DNA/89.6 produced about 300 ng of Gag and 85 ng of Env per 1 ⁇ 10 6 cells.
  • the rMVA booster (MVA/89.6) expressed SIV Gag, Pol, and HIV-1 Env under the control of vaccinia virus early/late promoters.
  • the MVA double recombinant virus expressed both the HIV 89.6 Env and the SIV 239 Gag-Pol, which were inserted into deletion II and deletion III of MVA, respectively.
  • the 89.6 Env protein was truncated for the COOH-terminal 115 amino acids of gp41.
  • the modified H5 promoter controlled the expression of both foreign genes.
  • Vaccination was accomplished by priming with DNA at 0 and 8 weeks and boosting with rMVA at 24 weeks (FIG. 7A).
  • DNA immunizations were delivered in phosphate-buffered saline (PBS) with a needleless jet injector (Bioject, Portland, Oreg.) to deliver five i.d. 100- ⁇ l injections to each outer thigh for the 2.5-mg dose of DNA or one i.d. 100- ⁇ l injection to the right outer thigh for the 250- ⁇ g dose of plasmid.
  • PBS phosphate-buffered saline
  • Bioject Bioject, Portland, Oreg.
  • I.m. deliveries of DNA were done with one 0.5-ml injection of DNA in PBS to each outer thigh for the 2.5-mg dose and one 100- ⁇ l injection to the right outer thigh for the 250- ⁇ g dose.
  • 1 ⁇ 10 8 pfu of MVA/89.6 was administered both i.d. and i.m. with a needle.
  • One 100- ⁇ l dose was delivered to each outer thigh for the i.d. dose and one 500- ⁇ l dose to each outer thigh for the i.m dose.
  • Control animals received 2.5 mg of the pGA2 vector without vaccine insert with the Bioject device to deliver five 100- ⁇ l doses i.d. to each outer thigh.
  • the control MVA booster immunization consisted of 2 ⁇ 10 8 pfu of MVA without an insert delivered i.d. and i.m. as described for MVA/89.6.
  • Animal numbers are as follows: 1, RBr-5*; 2, RIm-5*; 3, RQf-5*; 4, RZe-5; 5, ROm-5; 6, RDm-5; 7, RAj-5*; 8, RJi-5*; 9, RAl-5*; 10, RDe-5*; 11, RAi-5; 12, RPr-5; 13, RKw-4*; 14, RWz-5*; 15, RGo-5; 16, RLp-4; 17, RWd-6; 18, RAt-5; 19, RPb-5*; 20, RIi-5*; 21, RIq-5; 22, RSp-4; 23, RSn-5; 24, RGd-6; 25, RMb-5*; 26, RGy-5*; 27, RUs-4; and 28, RPm-5. Animals with the A*01 allele are indicated with asterisks.
  • DNA priming followed by rMVA boosting generated high frequencies of virus-specific T cells that peaked at one week following the rMVA booster (FIG. 7).
  • the frequencies of T cells recognizing the Gag-CM9 epitope were assessed by means of Mamu-A*01 tetramers, and the frequencies of T cells recognizing epitopes throughout Gag were assessed with pools of overlapping peptides and an enzyme-linked immunospot (ELISPOT) assay (C. A. Power et al. 1999 J Immunol Methods 227:99).
  • ELISPOT enzyme-linked immunospot
  • PBMC peripheral blood mononuclear cells
  • FITC fluorescein isothiocyanate
  • PerCP peridinin chlorophyl protein
  • SEQ ID NO: 6 Gag-CM9-Mamu-A*01 tetramer
  • Cells were washed twice with cold PBS containing 2% fetal bovine serum (FBS), fixed with 1% paraformaldehyde in PBS, and analyzed within 24 hrs on a FACScaliber (Becton Dickinson, San Jose, Calif.). Cells were initially gated on lymphocyte populations with forward scatter and side scatter and then on CD3 cells. The CD3 cells were then analyzed for CD8 and tetramer-binding cells. About 150,000 lymphocytes were acquired for each sample. Data were analyzed using FloJo software (Tree Star, San Carlos, Calif.).
  • IFN- ⁇ ELISPOTs For interferon- ⁇ (IFN- ⁇ ) ELISPOTs, MULTISCREEN 96 well filtration plates (Millipore Inc. Bedford, Mass.) were coated overnight with antibody to human IFN- ⁇ (Clone B27, Pharmingen, San Diego, Calif.) at a concentration of 2 ⁇ g/ml in sodium bicarbonate buffer (pH 9.6) at 8° to 10° C. Plates were washed two times with RPMI medium and then blocked for 1 hour with complete medium (RPMI containing 10% FBS) at 37° C. Plates were washed five more times with plain RPMI medium, and cells were seeded in duplicate in 100 ⁇ l complete medium at numbers ranging from 2 ⁇ 10 4 to 5 ⁇ 10 5 cells per well.
  • IFN- ⁇ interferon- ⁇
  • Peptide pools were added to each well to a final concentration of 2 ⁇ g/ml of each peptide in a volume of 100 ⁇ l in complete medium.
  • Cells were cultured at 37° C. for about 36 hrs under 5% CO 2 . Plates were washed six times with wash buffer (PBS with 0.05% Tween-20) and then incubated with 1 ⁇ g of biotinylated antibody to human IFN- ⁇ per milliliter (clone 7-86-1; Diapharma Group, West Chester, Ohio) diluted in wash buffer containing 2% FBS. Plates were incubated for 2 hrs at 37° C. and washed six times with wash buffer.
  • wash buffer PBS with 0.05% Tween-20
  • ELISPOTs The frequencies of ELISPOTs are approximate because different dilutions of cells have different efficiencies of spot formation in the absence of feeder cells (C. A. Power et al. 1999 J Immunol Methods 227: 99). The same dilution of cells was used for all animals at a given time point, but different dilutions were used to detect memory and acute responses.
  • Gag-CM9 tetramer analyses were restricted to macaques that expressed the Mamu-A*01 histocompatibility type, whereas ELISPOT responses did not depend on a specific histocompatibility type.
  • the DNA immunizations raised low levels of memory cells that expanded to high frequencies within 1 week of the rMVA booster (FIGS. 7 and 12).
  • CD8 cells specific to the Gag-CM9 epitope expanded to frequencies as high as 19% of total CD8 T cells (FIG. 12). This peak of specific cells underwent a 10- to 100-fold contraction into the DNA/MVA memory pool (FIGS. 7A and 12).
  • ELISPOTs for three pools of Gag peptides also underwent a major expansion (frequencies up to 4000 spots for 1 ⁇ 10 6 PBMC) before contracting from 5- to 20-fold into the DNA/MVA memory response (FIG. 7B).
  • the frequencies of ELISPOTs were the same in macaques with and without the A*01 histocompatibility type (P>0.2).
  • the rank order for the height of the ELISPOTs in the vaccine groups was 2.5 mg i.d.>2.5 mg i.m.>250 ⁇ g i.d.>250 ⁇ g i.m. (FIG. 7B).
  • the IFN- ⁇ ELISPOTs included both CD4 and CD8 cells. Gag-CM9-specific CD8 cells had good lytic activity after restimulation with peptide.
  • the challenge stock (5.7 ⁇ 10 9 copies of viral RNA per milliliter) was produced by one intravenous followed by one intrarectal passage in rhesus macaques of the original SHIV-89.6P stock (G. B. Karlsson et al. 1997 J Virol 71:4218). Lymphoid cells were harvested from the intrarectally infected animal at peak viremia, CD8-depleted, and mitogen-stimulated for stock production. Before intrarectal challenge, fasted animals were anesthetized (ketamine, 10 mg/kg) and placed on their stomach with the pelvic region slightly elevated. A feeding tube (8Fr (2.7 mm) ⁇ 16 inches (41 cm); Sherwood Medical, St.
  • a syringe containing 20 intrarectal infectious doses in 2 ml of RPMI-1640 plus 10% FBS was attached to the tube and the inoculum was slowly injected into the rectum.
  • the feeding tube was flushed with 3.0 ml of RPMI without FBS and then slowly withdrawn. Animals were left in place, with pelvic regions slightly elevated, for a period of ten minutes after the challenge.
  • the reaction mix was adjusted to a final volume of 50 ⁇ l containing 50 mM KCl, 10 mM Tris-HCl (pH 8.3), 4 mM MgCl 2 , 0.4 mM each dNTP, 0.2 ⁇ M forward primer, 0.2 ⁇ M reverse primer, 0.1 ⁇ M probe, and 5 units AmpliTaq Gold DNA polymerase (all reagents from PerkinElmer Applied Biosystems, Foster City, Calif.).
  • the primer sequences within a conserved portion of the SIV gag gene are the same as those described previously (S. Staprans et al. in: Viral Genome Methods K. Adolph, ed. CRC Press, Boca Raton, Fla., 1996 pp.
  • PCR product accumulation was monitored with the 7700 sequence detector and a probe to an internal conserved gag gene sequence: 6FAM-CTGTCTGCGTCATTTGGTGC-Tamra (SEQ ID NO: 8), where FAM and Tamra denote the reporter and quencher dyes.
  • SHIV RNA copy number was determined by comparison with an external standard curve consisting of virion-derived SIVmac239 RNA quantified by the SIV bDNA method (Bayer Diagnostics, Emeryville, Calif.).
  • the intraassay coefficient of variation was ⁇ 20% for samples containing >10 4 SHIV RNA copies per milliliter, and ⁇ 25% for samples containing 10 3 to 10 4 SHIV RNA copies per milliliter.
  • both high-dose DNA-primed groups and the low-dose i.d. DNA-primed group had reduced their geometric mean loads to about 1000 copies of viral RNA per milliliter.
  • the low-dose i.m. DNA-primed group had a geometric mean of 6 ⁇ 10 3 copies of viral RNA and the nonvaccinated controls had a geometric mean of 2 ⁇ 10 6 .
  • the low-dose i.m. group had reduced its geometric mean copies of viral RNA to 1000.
  • the 24 vaccinated animals only one animal, animal number 22 in the low-dose i.m. group, had intermittent viral loads above 1 ⁇ 10 4 copies per milliliter (FIG. 8D).
  • PBMC peripheral blood mononuclear cells
  • CTPYDINQM Gag-CM9 peptide
  • SEQ ID NO: 6 Gag-CM9 peptide
  • RPMI RPMI containing 10% FBS and monensin (10 ⁇ g/ml) was added, and the cells were cultured for an additional 5 hrs at 37° C. at an angle of 5° under 5% CO 2 .
  • Cells were surface stained with antibodies to CD8 conjugated to PerCP (clone SK1, Becton Dickinson) at 8° to 10° C. for 30 min, washed twice with cold PBS containing 2% FBS, and fixed and permeabilized with Cytofix/Cytoperm solution (Pharmingen).
  • the tetramer + cells in the control group like those in the vaccine group, produced IFN- ⁇ after peptide stimulation (FIG. 9B).
  • IFN- ⁇ after peptide stimulation
  • three of the four controls had undetectable levels of IFN- ⁇ -producing ELISPOTs. This rapid loss of antiviral T cells in the presence of high viral loads may reflect the lack of CD4 help.
  • T cell proliferative responses demonstrated that virus-specific CD4 cells had survived the challenge and were available to support the antiviral immune response (FIG. 9C).
  • Stimulation indices are the counts of tritiated thymidine incorporated in PBMC stimulated with 89.6 antigens divided by the counts of tritiated thymidine incorporated by the same PBMC stimulated with mock antigen.
  • lymph nodes from the vaccinated animals were morphologically intact and responding to the infection, whereas those from the infected controls had been functionally destroyed (FIG. 10).
  • Nodes from vaccinated animals contained large numbers of reactive secondary follicles with expanded germinal centers and discrete dark and light zones (FIG. 10A).
  • lymph nodes from the non-vaccinated control animals showed follicular and paracortical depletion (FIG. 10B), while those from unvaccinated and unchallenged animals displayed normal numbers of minimally reactive germinal centers (FIG. 10C).
  • Germinal centers occupied ⁇ 0.05% of total lymph node area in the infected controls, 2% of the lymph node area in the uninfected controls, and up to 18% of the lymph node area in the vaccinated groups (FIG. 10 D). More vigorous immune reactivity in the low-dose than the high-dose DNA-primed animals was suggested by more extensive germinal centers in the low dose group (FIG. 10D).
  • in situ hybridization for viral RNA revealed rare virus-expressing cells in lymph nodes from 3 of the 24 vaccinated macaques, whereas virus-expressing cells were readily detected in lymph nodes from each of the infected control animals.
  • the cytomorphology of infected lymph node cells was consistent with a macrophage phenotype.
  • the prime/boost strategy raised low levels of antibody to Gag and undetectable levels of antibody to Env (FIG. 11).
  • Postchallenge antibodies to both Env and Gag underwent anamnestic responses with total Gag antibody reaching heights approaching 1 mg/ml and total Env antibody reaching heights of up to 100 ⁇ g/ml.
  • Enzyme-linked immunosorbent assays for total antibody to Gag used bacterially produced SIV gag p27 to coat wells (2 ⁇ g per milliliter in bicarbonate buffer).
  • ELISAs for antibody to Env antibody used 89.6 Env produced in transiently transfected 293T cells and captured with sheep antibody against Env (catalog number 6205; International Enzymes, Fairbrook Calif.).
  • Standard curves for Gag and Env ELISAs were produced with serum from a SHIV-89.6-infected macaque with known amounts of immunoglobulin G (IgG) specific for Gag or Env.
  • IgG immunoglobulin G
  • Bound antibody was detected with peroxidase-conjugated goat antibody to macaque IgG (catalog # YNGMOIGGFCP; Accurate Chemical, Westbury, N.Y.) and TMB substrate (Catalog # T3405; Sigma, St. Louis, Mo.). Sera were assayed at threefold dilutions in duplicate wells. Dilutions of test sera were performed in whey buffer (4% whey and 0.1% tween 20 in 1 ⁇ PBS). Blocking buffer consisted of whey buffer plus 0.5% nonfat dry milk. Reactions were stopped with 2M H 2 SO 4 and the optical density read at 450 nm. Standard curves were fitted and sample concentrations were interpolated as ⁇ g of antibody per ml of serum using SOFTmax 2.3 software (Molecular Devices, Sunnyvale, Calif.).
  • the high-dose DNA-primed animals had slightly lower geometric mean levels of viral RNA (7 ⁇ 10 2 and 5 ⁇ 10 2 ) than the low-dose DNA-primed animals (9 ⁇ 10 2 and 1 ⁇ 10 3 ).
  • the DNA/MVA vaccine controlled the infection, rapidly reducing viral loads to near or below 1000 copies of viral RNA per milliliter of blood. Containment, rather than prevention of infection, affords the opportunity to establish a chronic infection (H. L. Robinson et al. 1999 Nat Med 5:526). By rapidly reducing viral loads, a multiprotein DNA/MVA vaccine will extend the prospect for long-term non-progression and limit HIV transmission. (J. W. Mellors et al. 1996 Science 272:1167; T. C. Quinn et al. 2000 N Engl J Med 342:921).
  • This disclosure describes the construction of a modified vaccinia Ankara (MVA) recombinant virus, MVA/HIV clade B recombinant virus expressing the HIV strain ADA env and the HXB2 gag pol (MVA/HIV ADA env+HXB2 gag pol).
  • MVA/HIV 48 the lab name of MVA/HIV 48 will be used, which denotes the plasmid from which the construct comes.
  • the HIV gag-pol genes were derived from the Clade B infectious HXB2 virus.
  • the gag-pol gene was truncated so that most of the integrase coding sequences were removed and amino acids 185, 266, and 478 were mutated to inactivate reverse transcriptase, inhibit strand transfer activity, and inhibit the RNaseH activity, respectively.
  • the Clade B CCR5 tropic envelope gene was derived from the primary ADA isolate; TTTTTNT sequences were mutated without changing coding capacity to prevent premature transcription termination and the cytoplasmic tail was truncated in order to improve surface expression, immunogenicity, and stability of the MVA vector.
  • the HIV genes were inserted into a plasmid transfer vector so that gag-pol gene was regulated by the modified H5 early/late vaccinia virus promoter and the env gene was regulated by the newly designed early/late Psyn II promoter to provide similar high levels of expression.
  • a self-deleting GUS reporter gene was included to allow detection and isolation of the recombinant virus.
  • the HIV genes were flanked by MVA sequences to allow homologous recombination into the deletion 3 site so that the recombinant MVA would remain TK positive for stability and high expression in resting cells. The recombinant MVA was isolated and shown to express abundant amounts of gag-pol-env and to process gag.
  • the plasmid transfer vector used to make the MVA recombinant virus, pLW-48, (FIG. 16) by homologous recombination was constructed as follows:
  • flanking areas on either side of deletion III designated flank 1 and flank 2, containing 926 and 520 base pairs respectively, were amplified by PCR from the MVA stains of vaccinia virus.
  • a promoter, the mH5 which had been modified from the originally published sequence by changing two bases that had been shown by previously published work to increase the expression of the cloned gene, was added.
  • a clade B gag pol (FIG. 17) was truncated so that the integrase was removed and was cloned into the plasmid so that it was controlled by the mH5 promoter.
  • This gene contained the complete HXB2 sequence of the gag.
  • the pol gene has reverse transcriptase safety mutations in amino acid 185 within the active site of RT, in amino acid 266 which inhibits strand transfer activity, and at amino acid 478 which inhibits the RNaseH activity.
  • the integrase gene was deleted past EcoRI site.
  • MVA virus which may be obtained from ATCC Number VR-1508, was plaque purified three times by terminal dilutions in chicken embryo fibroblasts (CEF), which were made from 9 day old SPF Premium SPAFAS fertile chicken eggs, distributed by B and E Eggs, Stevens, Pa.
  • CEF chicken embryo fibroblasts
  • the recombinant virus was shown to produce gag particles in the supernatant of infected cells by pelleting the 35 S-labeled particles on a 20% sucrose cushion.
  • Gag particles were also visualized both outside and budding from cells as well as within vacuoles of cells in the electron microscope in thin sections. These gag particles had envelope protein on their surface.
  • nucleotide sequences determined by sequencing a DNA molecule herein were determined using an automated DNA sequencer, and all amino acid sequences of polypeptides encoded by DNA molecules determined herein were predicted by translation of a DNA sequence determined as above. Therefore, as is known in the art for any DNA sequence determined by this automated approach, any nucleotide sequence determined herein may contain some errors. Nucleotide sequences determined by automation are typically at least about 90% identical, more typically at least about 95% to at least about 99.9% identical to the actual nucleotide sequence of the sequenced DNA molecule. The actual sequence can be more precisely determined by other approaches including manual DNA sequencing methods well known in the art.
  • a single insertion or deletion in a determined nucleotide sequence compared to the actual sequence will cause a frame shift in translation of the nucleotide sequence such that the predicted amino acid sequence encoded by a determined nucleotide sequence will be completely different from the amino acid sequence actually encoded by the sequenced DNA molecule, beginning at the point of such an insertion or deletion.
  • MVA/HIV 48 which has high expression of the ADA truncated envelope and the HXB2 gag pol.
  • the MVA recombinant virus is made using a transiently expressed GUS marker that is deleted in the final virus.
  • High expression of the ADA envelope is possible because of a new hybrid early/late promoter, Psyn II.
  • the envelope has been truncated because we have shown truncation of the envelope enhances the amount of protein on the surface of the infected cells, and hence enhances immunogenicity; stability of the recombinant is also enhanced.
  • the MVA recombinant makes gag particles which has been shown by pelleting the particles through sucrose and analyzing by PAGE. Gag particles with envelope protein on the surface have also been visualized in the electron microscope.
  • Additional modified or synthetic promoters were designed for gene expression in MVA or other poxviruses. Promoters were modified to allow expression at early and late times after infection and to reduce possibility of homologous recombination between identical sequences when multiple promoters are used in same MVA vector. Promoters are placed upstream of protein coding sequence.
  • m7.5 promoter CGCTTTTTATAGTAAGTTTTTCACCCATAAATAATAAATACAATAATTAATTTC (SEQ ID NO:10) TCGTAAAAATTGAAAAACTATTCTAATTTATTGCACGGT Psyn II promoter: TAAAAAATGAAAAAATATTCTAATTTATAGGACGGTTTTGATTTTCTTTTTTTC (SEQ ID NO:2) TATGCTATAAATAATAAATA Psyn III promoter: TAAAAATTGAAAAAATATTCTAATTTATAGGACGGTTTTGATTTTCTTTTTTTC (SEQ ID NO:11) TATACTATAAATAATAAATA Psyn IV promoter: TAAAAATTGAAAAACTATTCTAATTTATAGGACGGTTTTGATTTTCTTTTTTTC (SEQ ID NO:12) TATACTATAAATAATAAATA Psyn V promoter: AAAAAATGATAAAGTAGGTTCAGTTTTATTGCTGGTTTAAAATCACGCTTTCG (SEQ ID NO:
  • Table A MVA/48 immunization—guinea pigs.
  • Table B Frequencies of HIV-1 gag-specific T cells following immunization of mice with MVA/48.
  • Table D Antibody response following immunization of macaques with MVA/SHIV KB9.
  • SHIV-89.6P and SHIV-89.6 were neutralized in an MT-2 cell assay. Titers are the reciprocal serum dilution at which 50% of the cells were protected from virus-induced killing.
  • SHIV-89.6P and SHIV-89.6 are heterologous, i.e. sera from animals infected with one of the viruses does not neutralize the other virus.
  • two immunizations with MVA/SHIV-KB9 elicited good ELISA binding antibodies in all animals and neutralizing antibodies to the homologous virus (SHIV-89.6P) in some animals.
  • heterologous neutralizing antibodies were observed in a subset of animals.
  • Nab titer Nab titer animals animals 598 tonsil 25,600 31,086 20,383 ⁇ 20 ⁇ 20 3 2 601 ′′ 51,200 ⁇ 20 ⁇ 20 606 ′′ 25,600 ⁇ 20 ⁇ 20 642 ′′ 51,200 75 31 646 ′′ 51,200 61 48 653 ′′ 6,400 ⁇ 20 ⁇ 20 654 ′′ 6,400 22 ⁇ 20 602 i.d.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Plant Pathology (AREA)
  • Hematology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Physics & Mathematics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Oncology (AREA)
  • AIDS & HIV (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
US10/646,628 2001-03-08 2003-08-22 MVA expressing modified HIV envelope, gag, and pol genes Abandoned US20040146528A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US10/646,628 US20040146528A1 (en) 2001-03-08 2003-08-22 MVA expressing modified HIV envelope, gag, and pol genes
US12/018,150 US7867982B2 (en) 2001-03-08 2008-01-22 MVA expressing modified HIV envelope, gag, and pol genes
US12/987,791 US8916172B2 (en) 2001-03-08 2011-01-10 MVA expressing modified HIV envelope, gag, and pol genes
US14/566,066 US20150238593A1 (en) 2001-03-08 2014-12-10 Mva expressing modified hiv envelope, gag and pol genes

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US27443401P 2001-03-08 2001-03-08
PCT/US2002/006713 WO2002072754A2 (en) 2001-03-08 2002-03-01 Mva expressing modified hiv envelope, gag, and pol genes
US10/646,628 US20040146528A1 (en) 2001-03-08 2003-08-22 MVA expressing modified HIV envelope, gag, and pol genes

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/006713 Continuation WO2002072754A2 (en) 2000-03-02 2002-03-01 Mva expressing modified hiv envelope, gag, and pol genes

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/018,150 Continuation US7867982B2 (en) 2001-03-08 2008-01-22 MVA expressing modified HIV envelope, gag, and pol genes

Publications (1)

Publication Number Publication Date
US20040146528A1 true US20040146528A1 (en) 2004-07-29

Family

ID=23048178

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/646,628 Abandoned US20040146528A1 (en) 2001-03-08 2003-08-22 MVA expressing modified HIV envelope, gag, and pol genes
US12/018,150 Expired - Fee Related US7867982B2 (en) 2001-03-08 2008-01-22 MVA expressing modified HIV envelope, gag, and pol genes
US12/987,791 Expired - Fee Related US8916172B2 (en) 2001-03-08 2011-01-10 MVA expressing modified HIV envelope, gag, and pol genes
US14/566,066 Abandoned US20150238593A1 (en) 2001-03-08 2014-12-10 Mva expressing modified hiv envelope, gag and pol genes

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/018,150 Expired - Fee Related US7867982B2 (en) 2001-03-08 2008-01-22 MVA expressing modified HIV envelope, gag, and pol genes
US12/987,791 Expired - Fee Related US8916172B2 (en) 2001-03-08 2011-01-10 MVA expressing modified HIV envelope, gag, and pol genes
US14/566,066 Abandoned US20150238593A1 (en) 2001-03-08 2014-12-10 Mva expressing modified hiv envelope, gag and pol genes

Country Status (13)

Country Link
US (4) US20040146528A1 (ja)
EP (1) EP1372710B1 (ja)
JP (2) JP4554887B2 (ja)
CN (1) CN100446812C (ja)
AT (1) ATE445411T1 (ja)
AU (1) AU2002252199B2 (ja)
CA (1) CA2454959C (ja)
CY (1) CY1109659T1 (ja)
DE (1) DE60234018D1 (ja)
DK (1) DK1372710T3 (ja)
ES (1) ES2334335T3 (ja)
PT (1) PT1372710E (ja)
WO (1) WO2002072754A2 (ja)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060134133A1 (en) * 2003-03-28 2006-06-22 Bernard Moss MVA expressing modified HIV envelope, Gag, and Pol genes
WO2008118936A2 (en) * 2007-03-26 2008-10-02 International Aids Vaccine Initiative Method of elicting immune response with a modified mva viral hiv-1 vector
US20090060947A1 (en) * 2006-05-19 2009-03-05 Sanofi Pasteur, Inc. Immunological compositions
WO2009145853A1 (en) * 2008-04-16 2009-12-03 Virostatics Srl Novel assay for precursor t-cells having high proliferative capacity (phpc-assay)
CN101394868B (zh) * 2006-03-07 2011-08-10 浙江海正药业股份有限公司 重组腺病毒5型/35型和痘病毒mva活病毒载体疫苗的联用诱导强大的免疫应答的方法
WO2012137071A2 (en) 2011-04-06 2012-10-11 Biovaxim Limited Pharmaceutical compositions for preventing and/or treating an hiv disease in humans
WO2017143016A1 (en) * 2016-02-16 2017-08-24 Geovax Inc. Multivalent hiv vaccine boost compositions and methods of use

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002211948B2 (en) 2000-03-02 2007-09-13 Emory University DNA expression vectors and methods of use
US8623379B2 (en) * 2000-03-02 2014-01-07 Emory University Compositions and methods for generating an immune response
CA2454959C (en) 2001-03-08 2018-07-10 Bernard Moss Mva expressing modified hiv envelope, gag, and pol genes
AU2003220111B2 (en) * 2002-03-08 2008-07-31 Emory University Compositions and methods for generating an immune response
CN1490056A (zh) * 2002-10-18 2004-04-21 ��¡���ɵ°��̲��о����� 针对hiv-1的免疫方法和组合物
WO2005034992A2 (en) * 2003-09-15 2005-04-21 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Hiv vaccines based on env of multiple clades of hif
DK1845164T3 (da) * 2003-11-24 2010-09-20 Bavarian Nordic As Promoterer til ekspression i modificeret vacciniavirus ankara
CN1304580C (zh) * 2004-07-09 2007-03-14 楼伟 重组表达人获得性免疫缺陷病毒I型的表面糖蛋白gp160
EP2397490B1 (en) 2004-07-16 2013-09-04 THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Vaccine constructs and combinations of vaccines designed to improve the breadth of the immune response to diverse strains and clades of HIV
AU2005279799A1 (en) * 2004-08-27 2006-03-09 Emory University Recombinant MVA viruses expressing Clade A/G, Clade B and Clade C modified HIV ENV, GAG and POL genes
AU2012202786B8 (en) * 2004-08-27 2015-05-21 Emory University Recombinant MVA viruses expressing Clade A/G, Clade B, and Clade C modified HIV ENV, GAG and POL genes
ES2281252B1 (es) 2005-07-27 2009-02-16 Consejo Superior De Investigaciones Cientificas Vectores recombinantes basados en el virus modificado de ankara (mva) como vacunas preventivas y terapeuticas contra el sida.
FR2893254B1 (fr) * 2005-11-14 2007-12-21 Biomerieux Sa Composition comprenant un vecteur synthetique colloidal bioresorbable et un vecteur viral et son utilisation prophylactique, therapeutique et diagnostique.
CN101020052A (zh) * 2006-02-16 2007-08-22 中国疾病预防控制中心性病艾滋病预防控制中心 基于复制型痘苗病毒载体的艾滋病疫苗
EP2042604A1 (en) * 2007-09-26 2009-04-01 Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) VV promoter driven overexpression of recombinant antigens
EP2337581B1 (fr) * 2008-09-10 2017-08-23 Institut National De La Sante Et De La Recherche Medicale Construction d'un gène synthétique codant pour gag de vih1 et son utilisation pour l'obtention de vaccins anti-vih-1
WO2011047031A2 (en) * 2009-10-13 2011-04-21 Geovax, Inc. Eliciting immune responses using recombinant mva viruses expressing hiv env, gag and pol anitgens
ES2741881T3 (es) 2010-05-19 2020-02-12 Nippon Steel Corp Método de conformación por prensado de componentes con forma de L
WO2014018850A2 (en) * 2012-07-27 2014-01-30 The Trustees Of The University Of Pennsylvania Cytoplasmic tail modifications to boost surface expression and immunogenicity of envelope glycoproteins
WO2016068919A1 (en) 2014-10-29 2016-05-06 Geovax, Inc. Combination therapy for treating viral reservoirs
US11701418B2 (en) 2015-01-12 2023-07-18 Geovax, Inc. Replication-deficient modified vaccinia Ankara (MVA) expressing Ebola virus glycoprotein (GP) and matrix protein (VP40)
CA3011014A1 (en) 2016-01-08 2017-07-13 Harriet Robinson Compositions and methods for generating an immune response to a tumor associated antigen
WO2017136419A1 (en) 2016-02-03 2017-08-10 Geovax Inc. Compositions and methods for generating an immune response to a flavivirus
WO2018195447A1 (en) * 2017-04-20 2018-10-25 Geovax Labs, Inc. Recombinant mva-based hiv immunogens and uses thereof
US20200171141A1 (en) * 2017-07-18 2020-06-04 Geovax, Inc. Compositions and Methods for Generating an Immune Response to LASV
US11311612B2 (en) 2017-09-19 2022-04-26 Geovax, Inc. Compositions and methods for generating an immune response to treat or prevent malaria

Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5169763A (en) * 1986-04-08 1992-12-08 Transgene S.A., Institut Pasteur Viral vector coding glycoprotein of HIV-1
US5185146A (en) * 1988-01-12 1993-02-09 Hoffmann-Laroche Inc. Recombinant mva vaccinia virus
US5256767A (en) * 1987-06-10 1993-10-26 The Immune Response Corporation Retroviral antigens
US5445953A (en) * 1991-08-26 1995-08-29 Immuno Aktiengesellschaft Direct molecular cloning of a modified poxvirus genome
US5494807A (en) * 1991-03-07 1996-02-27 Virogenetics Corporation NYVAC vaccinia virus recombinants comprising heterologous inserts
US5589466A (en) * 1989-03-21 1996-12-31 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5593972A (en) * 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
US5614404A (en) * 1988-06-10 1997-03-25 Theriod Biologics, Incorporated Self-assembled, defective, non-self-propagating lentivirus particles
US5676950A (en) * 1994-10-28 1997-10-14 University Of Florida Enterically administered recombinant poxvirus vaccines
US5736368A (en) * 1988-06-10 1998-04-07 Therion Biologics Corporation Self assembled defective non-self propagating lentiviral particles
US5741492A (en) * 1996-01-23 1998-04-21 St. Jude Children's Research Hospital Preparation and use of viral vectors for mixed envelope protein vaccines against human immunodeficiency viruses
US5747338A (en) * 1996-08-15 1998-05-05 Chiron Corporation Method and construct for screening for inhibitors of transcriptional activation
US5846946A (en) * 1996-06-14 1998-12-08 Pasteur Merieux Serums Et Vaccins Compositions and methods for administering Borrelia DNA
US5849304A (en) * 1986-04-08 1998-12-15 The United States Of America As Represented By The Department Of Health And Human Services Recombinant vaccinia virus expressing human retrovirus gene
US5858775A (en) * 1994-06-06 1999-01-12 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5863542A (en) * 1991-03-07 1999-01-26 Virogenetics Corporation Recombinant attenuated ALVAC canaryopox virus containing heterologous HIV or SIV inserts
US5879925A (en) * 1994-08-15 1999-03-09 Connaught Laboratories Limited Genetically engineered human immunodeficiency virus-like particles with modified chimeric envelope glycoproteins containing influenza virus transmembrane spanning domains
US5911989A (en) * 1995-04-19 1999-06-15 Polynum Scientific Immunbiologische Forschung Gmbh HIV-vaccines
US5985641A (en) * 1989-10-13 1999-11-16 Connaught Laboratories Limited Non-infectious, replication-impaired, immunogenic human immunodeficiency virus type 1 retrovirus-like particles with multiple genetic deficiencies
US6077662A (en) * 1996-11-27 2000-06-20 Emory University Virus-like particles, methods and immunogenic compositions
US6080408A (en) * 1994-08-22 2000-06-27 Connaught Laboratories Limited Human immunodeficiency virus type 1 nucleic acids devoid of long terminal repeats capable of encoding for non-infectious, immunogenic, retrovirus-like particles
US6099847A (en) * 1997-05-15 2000-08-08 The United States Of America As Represented By The Department Of Health And Human Services Chimeric Gag pseudovirions
US6103244A (en) * 1991-08-26 2000-08-15 Immuno Ag. Methods for generating immune responses employing modified vaccinia of fowlpox viruses
US6121021A (en) * 1997-12-16 2000-09-19 Connaught Laboratories Limited Constitutive expression of non-infectious HIV-like particles
US6140114A (en) * 1996-04-05 2000-10-31 Universite Pierre Et Marie Curie (Paris Vi) Defective viral vaccine particles obtained in vivo or ex vivo
US6156952A (en) * 1998-04-09 2000-12-05 Constituent Institution Of The University Of Maryland System HIV transgenic animals and uses therefor
US6171596B1 (en) * 1993-12-10 2001-01-09 The United States Of America As Represented By The Department Of Health And Human Services Oligomeric HIV-1 envelope glycoproteins
US6201663B1 (en) * 1996-09-05 2001-03-13 Kabushiki Kaisha Ohara Magnetic head having a slider made of a glass-ceramic
US6204250B1 (en) * 1996-11-22 2001-03-20 The Mount Sinai Medical Center Of The City Of New York Immunization of infants
US6214804B1 (en) * 1989-03-21 2001-04-10 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US6291157B1 (en) * 1998-02-23 2001-09-18 Connaught Laboratories Limited Antigenically-marked non-infectious retrovirus-like particles
US6306625B1 (en) * 1988-12-30 2001-10-23 Smithkline Beecham Biologicals, Sa Method for obtaining expression of mixed polypeptide particles in yeast
US6448083B1 (en) * 1997-08-29 2002-09-10 Selective Genetics, Inc. Receptor-mediated gene delivery using bacteriophage vectors
US6554527B1 (en) * 1998-05-13 2003-04-29 Altronic Distributors Pty. Ltd. Mounting assembly

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1340729C (en) 1988-03-23 1999-09-07 Mohan Bir Singh Ryegrass pollen allergen
IL89567A0 (en) 1988-03-28 1989-09-10 Univ Leland Stanford Junior Mutated hiv envelope protein
WO1989012095A1 (en) * 1988-06-10 1989-12-14 Applied Biotechnology, Inc. A method of evaluating recombinant vaccines against immunodeficiency virus
ES2052478T1 (es) * 1989-11-20 1994-07-16 Oncogen Particulas retrovirales no replicantes producidas de forma recombinante empleadas como agentes antivirales e inmunogenos.
IE71643B1 (en) * 1990-11-20 1997-02-26 Virogenetics Corp A recombinant poxviral vaccine for canine distemper
DE69133333T2 (de) * 1991-08-26 2004-07-29 Baxter Healthcare S.A. Ein intaktes FPV-tk-Gen enthaltender rekombinanter Virus der Vogelpocken
GB9711957D0 (en) 1997-06-09 1997-08-06 Isis Innovation Methods and reagents for vaccination
DE19726167B4 (de) 1997-06-20 2008-01-24 Sanofi-Aventis Deutschland Gmbh Insulin, Verfahren zu seiner Herstellung und es enthaltende pharmazeutische Zubereitung
GB2337755B (en) * 1998-05-29 2003-10-29 Secr Defence Virus vaccine
CA2335508A1 (en) 1998-06-26 2000-01-06 Aventis Pasteur Use of poxviruses as enhancer of specific immunity
WO2000011140A1 (en) 1998-08-20 2000-03-02 The Wistar Institute Of Anatomy And Biology Methods of augmenting mucosal immunity through systemic priming and mucosal boosting
WO2001002607A1 (en) 1999-07-06 2001-01-11 Merck & Co., Inc. Adenovirus carrying gag gene hiv vaccine
US6256183B1 (en) * 1999-09-09 2001-07-03 Ferraz Shawmut Inc. Time delay fuse with mechanical overload device and indicator actuator
CN1213068C (zh) * 1999-12-23 2005-08-03 医疗研究局 对hiv免疫应答的改进或与其相关的改进
AUPQ520800A0 (en) 2000-01-21 2000-02-17 Alfred Hospital Prime-boost vaccination strategy
AU2002211948B2 (en) * 2000-03-02 2007-09-13 Emory University DNA expression vectors and methods of use
WO2001082962A2 (en) 2000-04-27 2001-11-08 Aventis Pasteur Limited Immunizing against hiv infection
CA2454959C (en) 2001-03-08 2018-07-10 Bernard Moss Mva expressing modified hiv envelope, gag, and pol genes
EP1409694A4 (en) 2001-07-05 2006-02-08 Chiron Corp POLYNUCLEOTIDES ENCODING ANTIGENIC POLYPEPTIDES OF HIV SUBTYPE B AND / OR SUBTYPE C, POLYPEPTIDES AND USES THEREOF

Patent Citations (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5849304A (en) * 1986-04-08 1998-12-15 The United States Of America As Represented By The Department Of Health And Human Services Recombinant vaccinia virus expressing human retrovirus gene
US5169763A (en) * 1986-04-08 1992-12-08 Transgene S.A., Institut Pasteur Viral vector coding glycoprotein of HIV-1
US5256767A (en) * 1987-06-10 1993-10-26 The Immune Response Corporation Retroviral antigens
US5928930A (en) * 1987-06-10 1999-07-27 Immune Response Corporation Prevention and treatment of retroviral disease
US5853725A (en) * 1987-06-10 1998-12-29 The Immune Response Corporation Prevention and treatment of retroviral disease
US5185146A (en) * 1988-01-12 1993-02-09 Hoffmann-Laroche Inc. Recombinant mva vaccinia virus
US6051410A (en) * 1988-06-10 2000-04-18 Therion Biologics, Corp. Self-assembled, defective, nonself-propagating viral particles
US5747324A (en) * 1988-06-10 1998-05-05 Therion Biologics Corporation Self-assembled, defective, non-self-propagating lentivirus particles
US5614404A (en) * 1988-06-10 1997-03-25 Theriod Biologics, Incorporated Self-assembled, defective, non-self-propagating lentivirus particles
US5736368A (en) * 1988-06-10 1998-04-07 Therion Biologics Corporation Self assembled defective non-self propagating lentiviral particles
US6306625B1 (en) * 1988-12-30 2001-10-23 Smithkline Beecham Biologicals, Sa Method for obtaining expression of mixed polypeptide particles in yeast
US6214804B1 (en) * 1989-03-21 2001-04-10 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5589466A (en) * 1989-03-21 1996-12-31 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5985641A (en) * 1989-10-13 1999-11-16 Connaught Laboratories Limited Non-infectious, replication-impaired, immunogenic human immunodeficiency virus type 1 retrovirus-like particles with multiple genetic deficiencies
US5766599A (en) * 1991-03-07 1998-06-16 Virogenetics Corporation Trova fowl pox virus recombinants comprising heterologous inserts
US5756103A (en) * 1991-03-07 1998-05-26 Virogenetics Corporation Alvac canarypox virus recombinants comprising heterlogous inserts
US5863542A (en) * 1991-03-07 1999-01-26 Virogenetics Corporation Recombinant attenuated ALVAC canaryopox virus containing heterologous HIV or SIV inserts
US5494807A (en) * 1991-03-07 1996-02-27 Virogenetics Corporation NYVAC vaccinia virus recombinants comprising heterologous inserts
US6103244A (en) * 1991-08-26 2000-08-15 Immuno Ag. Methods for generating immune responses employing modified vaccinia of fowlpox viruses
US5445953A (en) * 1991-08-26 1995-08-29 Immuno Aktiengesellschaft Direct molecular cloning of a modified poxvirus genome
US6265183B1 (en) * 1991-08-26 2001-07-24 Baxter Aktiengesellschaft Direct molecular cloning of foreign genes into poxviruses and methods for the preparation of recombinant proteins
US5817637A (en) * 1993-01-26 1998-10-06 The Trustees Of The University Of Pennsylvania Genetic immunization
US5593972A (en) * 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
US6171596B1 (en) * 1993-12-10 2001-01-09 The United States Of America As Represented By The Department Of Health And Human Services Oligomeric HIV-1 envelope glycoproteins
US5858775A (en) * 1994-06-06 1999-01-12 Children's Hospital, Inc. Adeno-associated virus materials and methods
US5879925A (en) * 1994-08-15 1999-03-09 Connaught Laboratories Limited Genetically engineered human immunodeficiency virus-like particles with modified chimeric envelope glycoproteins containing influenza virus transmembrane spanning domains
US6080408A (en) * 1994-08-22 2000-06-27 Connaught Laboratories Limited Human immunodeficiency virus type 1 nucleic acids devoid of long terminal repeats capable of encoding for non-infectious, immunogenic, retrovirus-like particles
US5676950A (en) * 1994-10-28 1997-10-14 University Of Florida Enterically administered recombinant poxvirus vaccines
US5911989A (en) * 1995-04-19 1999-06-15 Polynum Scientific Immunbiologische Forschung Gmbh HIV-vaccines
US5741492A (en) * 1996-01-23 1998-04-21 St. Jude Children's Research Hospital Preparation and use of viral vectors for mixed envelope protein vaccines against human immunodeficiency viruses
US6086891A (en) * 1996-01-23 2000-07-11 St. Jude Children's Research Hospital Bi-functional plasmid that can act as both a DNA vaccine and a recombinant virus vector
US6140114A (en) * 1996-04-05 2000-10-31 Universite Pierre Et Marie Curie (Paris Vi) Defective viral vaccine particles obtained in vivo or ex vivo
US5846946A (en) * 1996-06-14 1998-12-08 Pasteur Merieux Serums Et Vaccins Compositions and methods for administering Borrelia DNA
US5747338A (en) * 1996-08-15 1998-05-05 Chiron Corporation Method and construct for screening for inhibitors of transcriptional activation
US6201663B1 (en) * 1996-09-05 2001-03-13 Kabushiki Kaisha Ohara Magnetic head having a slider made of a glass-ceramic
US6204250B1 (en) * 1996-11-22 2001-03-20 The Mount Sinai Medical Center Of The City Of New York Immunization of infants
US6077662A (en) * 1996-11-27 2000-06-20 Emory University Virus-like particles, methods and immunogenic compositions
US6099847A (en) * 1997-05-15 2000-08-08 The United States Of America As Represented By The Department Of Health And Human Services Chimeric Gag pseudovirions
US6448083B1 (en) * 1997-08-29 2002-09-10 Selective Genetics, Inc. Receptor-mediated gene delivery using bacteriophage vectors
US6121021A (en) * 1997-12-16 2000-09-19 Connaught Laboratories Limited Constitutive expression of non-infectious HIV-like particles
US6291157B1 (en) * 1998-02-23 2001-09-18 Connaught Laboratories Limited Antigenically-marked non-infectious retrovirus-like particles
US6156952A (en) * 1998-04-09 2000-12-05 Constituent Institution Of The University Of Maryland System HIV transgenic animals and uses therefor
US6554527B1 (en) * 1998-05-13 2003-04-29 Altronic Distributors Pty. Ltd. Mounting assembly

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7303754B2 (en) 2003-03-28 2007-12-04 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services MVA expressing modified HIV envelope, gag, and pol genes
US20060134133A1 (en) * 2003-03-28 2006-06-22 Bernard Moss MVA expressing modified HIV envelope, Gag, and Pol genes
US20110159036A1 (en) * 2003-03-28 2011-06-30 Bernard Moss MVA expressing modified HIV envelope, GAG, and POL genes
CN101394868B (zh) * 2006-03-07 2011-08-10 浙江海正药业股份有限公司 重组腺病毒5型/35型和痘病毒mva活病毒载体疫苗的联用诱导强大的免疫应答的方法
US20090060947A1 (en) * 2006-05-19 2009-03-05 Sanofi Pasteur, Inc. Immunological compositions
WO2008118936A2 (en) * 2007-03-26 2008-10-02 International Aids Vaccine Initiative Method of elicting immune response with a modified mva viral hiv-1 vector
US20080274992A1 (en) * 2007-03-26 2008-11-06 Jean-Louis Excler Recombinant modified ankara viral hiv-1 vaccines
WO2008118936A3 (en) * 2007-03-26 2008-12-04 Int Aids Vaccine Initiative Method of elicting immune response with a modified mva viral hiv-1 vector
WO2009145853A1 (en) * 2008-04-16 2009-12-03 Virostatics Srl Novel assay for precursor t-cells having high proliferative capacity (phpc-assay)
WO2012137071A2 (en) 2011-04-06 2012-10-11 Biovaxim Limited Pharmaceutical compositions for preventing and/or treating an hiv disease in humans
EP3000476A1 (en) 2011-04-06 2016-03-30 Biovaxim Limited Pharmaceutical compositions for preventing and/or treating an hiv disease in humans
WO2017143016A1 (en) * 2016-02-16 2017-08-24 Geovax Inc. Multivalent hiv vaccine boost compositions and methods of use
US11098086B2 (en) 2016-02-16 2021-08-24 Geovax Inc. Multivalent HIV vaccine boost compositions and methods of use
US11897919B2 (en) 2016-02-16 2024-02-13 Geovax, Inc. Multivalent HIV vaccine boost compositions and methods of use

Also Published As

Publication number Publication date
AU2002252199B2 (en) 2008-01-03
CA2454959C (en) 2018-07-10
EP1372710B1 (en) 2009-10-14
ATE445411T1 (de) 2009-10-15
US20090074726A1 (en) 2009-03-19
JP4554887B2 (ja) 2010-09-29
CN1638795A (zh) 2005-07-13
CY1109659T1 (el) 2014-08-13
WO2002072754A2 (en) 2002-09-19
WO2002072754A3 (en) 2003-05-22
ES2334335T3 (es) 2010-03-09
DE60234018D1 (de) 2009-11-26
JP2010115208A (ja) 2010-05-27
CA2454959A1 (en) 2002-09-19
CN100446812C (zh) 2008-12-31
US20110104199A1 (en) 2011-05-05
US20150238593A1 (en) 2015-08-27
US8916172B2 (en) 2014-12-23
EP1372710A4 (en) 2005-04-27
PT1372710E (pt) 2009-12-21
EP1372710A2 (en) 2004-01-02
WO2002072754A9 (en) 2003-01-03
JP2004537974A (ja) 2004-12-24
DK1372710T3 (da) 2010-01-18
US7867982B2 (en) 2011-01-11

Similar Documents

Publication Publication Date Title
CA2454959C (en) Mva expressing modified hiv envelope, gag, and pol genes
US7303754B2 (en) MVA expressing modified HIV envelope, gag, and pol genes
CA2578403C (en) Recombinant mva viruses expressing clade a/g, clade b, and clade c modified hiv env, gag and pol genes
AU2002252199A1 (en) MVA expressing modified HIV envelope, GAG, and POL genes
JP2011231112A (ja) 免疫応答を作製するための組成物および方法
AU2012202786B8 (en) Recombinant MVA viruses expressing Clade A/G, Clade B, and Clade C modified HIV ENV, GAG and POL genes

Legal Events

Date Code Title Description
AS Assignment

Owner name: GOVERNMENT OF THE UNITED STATES OF AMERICA, AS REP

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MOSS, BERNARD;WYATT, LINDA;EARL, PATRICIA;REEL/FRAME:014974/0687

Effective date: 20040202

Owner name: EMORY UNIVERSITY, GEORGIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ROBINSON, HARRIET L.;REEL/FRAME:014975/0742

Effective date: 20040116

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:EMORY UNIVERSITY;REEL/FRAME:021262/0050

Effective date: 20040618

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH -DIRECTOR DEITR, MAR

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:EMORY UNIVERSITY;REEL/FRAME:037880/0754

Effective date: 20160303