EP0817854A2 - Antigen presentation system based on retrovirus-like particles - Google Patents

Antigen presentation system based on retrovirus-like particles

Info

Publication number
EP0817854A2
EP0817854A2 EP96911989A EP96911989A EP0817854A2 EP 0817854 A2 EP0817854 A2 EP 0817854A2 EP 96911989 A EP96911989 A EP 96911989A EP 96911989 A EP96911989 A EP 96911989A EP 0817854 A2 EP0817854 A2 EP 0817854A2
Authority
EP
European Patent Office
Prior art keywords
presentation system
antigen presentation
hiv
virus
antigen
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.)
Withdrawn
Application number
EP96911989A
Other languages
German (de)
English (en)
French (fr)
Inventor
Hans Wolf
Ralf Wagner
Ludwig Deml
Klaus Osterrieder
Frank Notka
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.)
Wolf Hans Prof Dr
Original Assignee
Wolf Hans Prof Dr
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 Wolf Hans Prof Dr filed Critical Wolf Hans Prof Dr
Priority to EP96911989A priority Critical patent/EP0817854A2/en
Publication of EP0817854A2 publication Critical patent/EP0817854A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C12N2710/14011Baculoviridae
    • C12N2710/14111Nucleopolyhedrovirus, e.g. autographa californica nucleopolyhedrovirus
    • C12N2710/14141Use of virus, viral particle or viral elements as a vector
    • C12N2710/14143Use 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
    • C12N2710/16011Herpesviridae
    • C12N2710/16711Varicellovirus, e.g. human herpesvirus 3, Varicella Zoster, pseudorabies
    • C12N2710/16722New 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16711Varicellovirus, e.g. human herpesvirus 3, Varicella Zoster, pseudorabies
    • C12N2710/16734Use 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
    • C12N2740/16023Virus like particles [VLP]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the technical problem underlying the present invention is to provide an antigen presentation system on the basis of retroviral group specific antigens ⁇ gag) that can be used for the preventive and therapeutic immunization of mammals against infectious diseases or neoplasias.
  • This invention particularly relates to a newly designed antigen delivery system produced in appropriate expression systems e.g. recombinant baculoviruses, semiiki forest viruses or stably transfected insect or mammalian cells, respectively.
  • the invention is based on retroviral group specific antigens such as the human immunodeficiency virus type 1 (HIV-1) Pr559 a 9 precursor protein constituing immature forms of retroviral particles when expressed in eucaryotic cells.
  • HIV-1 human immunodeficiency virus type 1
  • VLPs immature virus-like particles
  • the immunogenicity of the immature virus-like particles can be extended by anchoring complete proteins on the surface of the VLPs. These proteins can either constitute autologous membrane proteins derived from the same retrovirus as the particulate gag carrier component or from any other virus, infectious agent or neopiastic cell. Stable anchoring of the autologous or "foreign" proteins on the surface of the VLPs requires the co-expression of the retroviral gag-precursor together with the antigen to be presented.
  • the antigen to be presented must include an aminoterminal signal sequence as well as a transmembrane domain in order to allow transport of the antigen via endoplasmic reticulum to the cytoplasmic membrane and stable anchoring on the surface of the budding VLP.
  • the mode of processing and presentation by antigen presenting cells determines which T-cell effector functions are specifically activated in an immune response to a protein antigen.
  • APC antigen presenting cells
  • proteins in the extracellular fluid or in the cell membrane enter the APC through the endocytic pathway to be denatured and proteolyticatly degraded to peptides 12-15 residues long in an acid milieu at a late endosomal stage.
  • Peptides generated in this pathway bind to MHC class II molecules, transit to the APC surface, and selectively stimulate CD4+ T cells. Immunization with soluble protein antigens thus stimulate preferentially CD4+ T cells (Germain, 1991; Germain and Hendrix, 1991).
  • CD8 + cytotoxic T lymphocytes are selectively stimulated in the alternative endogenous processing pathway.
  • Intraceliuiar proteins are degraded to peptides of an optimal 8-15 residue size in the cytosol. These peptides are transported into the endoplasmic reticulum (ER) where they bind to nascent MHC class I heavy chain B2m microgiobulin dimers. This generates transport competent trimeric complexes that move rapidly by the default secretory route to the surface membrane of the APC.
  • Peptides presented in the context of class I molecules stimulate selectively CD8+ T cells (Yewdell and Bennink, 1992; Townsend and Bodmer, 1989).
  • Particulate carrier systems which were also demonstrated to induce CD8+, MHC class l-restricted cytotoxic T cells in vivo are mainly based on viral antigens or on the yeast TY-particles (Layton et al. 1993; Martin et al. 1993). These antigen presentation systems appear to evoke strong immune responses without need of additional adjuvants, but suffer from the presentation of a limited number of relevant "foreign" epitopes.
  • VLPs noninfectious, morphologically immature HIV-1 retrovirus-like particles
  • This concept follows the construction of Pr55-? a .? expression cassetts allowing the insertion of carefully selected epitopes from HIV reading frames other than gag. Resulting chimeric proteins should assemble into premature VLP when expressed in eucaryotic cells and allow the presentation of additional imunologically relevant epitopes. This concept also allowes to exclude epitopes suggested to be associated with adverse side effects such as induction of graft versus host-like diseases, antibodies enhancing the infection of CD4+ cells by HIV or gp120 mediated apoptosis.
  • V3 specific CTL were not only found in splenocytes, but in lymphnodes when tested. Immunization of BALB/c mice with naked Pr55-?
  • a /V3 VLP efficiently primed the CTL response in absence of adjuvant or replicating vector (69% specific lysis): In contrast VLP adsorbed to alum or emulsified in IFA only weakly stimulated CTL response (24%, 37% specific lysis).
  • Pr559 a 9N3 recombinant vaccinia viruses the position of the V3-domain within different variants of chimeric VLP ⁇ Pr559 a 9/V3-3, Pr559 a 9/V3-4, Pr559 a 9N3-5) did not influence the induction of a V3-specific CTL response.
  • the technical problem underlying the invention is to provide DNA sequences encoding authentic or modified polypeptides derived from HIV, or from any other virus, infectious agent or neoplastic cell which allow the presentation of the polypeptides on the surface of noninfectious retroviral virus-like particles (VLPs).
  • VLPs noninfectious retroviral virus-like particles
  • the present invention relates to the presentation of immunologicaily important epitopes, authentic or chimeric polypeptides via noninfectious retrovirus- like particles to the immune system.
  • the retrovirus-like particulate carrier is encoded by the group specific antigen (gag) of a retrovirus being pathogenic to humans, subhuman primates or other mammals.
  • the DNA sequence encoding the retrovirus- like particles is derived from any of the retroviruses HTLV-1 , HTLV-2, HIV-1 , HIV-2, SIV or FIV.
  • gag polypeptid is pr555 a 5 of HIV-1.
  • gag polypeptides spontaneously form said retrovirus-like particles.
  • retrovirus-like particles which are composed by retroviral gag polypeptides are spiked by additional immunologicaily relevant peptides or proteins which are presented to the immune system.
  • immunologicaily relevant peptides or proteins can be derived from any infectious agent or neoplastic cell.
  • the proteins to be presented by the retrovirus-like particles represent authentic (occurring in nature) or chimeric (not occurring in nature) membrane proteins.
  • these membrane antigens are derived from different viruses such as retroviruses or herpesviruses.
  • the envelope proteins being anchored on the surface of a retrovirus-like particle are derived from any of the retroviruses HTLV-1 , HTLV-2, HIV-1. HIV-2, SIV or FIV or the Epstein-Barr (EBV) virus or the equine herpesvirus EHV. More specifically the antigen anchored on the surface of said retrovirus-like particles is the complete envelope protein gp160 of HIV-1 or the major membrane antigen of EBV gp220/350 or the herpes simplex virus gB homologue of EHV.
  • the transmembrane and cytoplasmic domain of a given membrane protein may be replaced by a heterologous membrane anchor sequence.
  • This heterologous transmembrane domain may be encoded by any viral envelope protein or cellular membrane protein.
  • transmembrane domain including a short cytoplasmic tail is derived from the Epstein-Barr virus major membrane antigen gp220/350.
  • the DNA sequence encoding the EBV gp220/350 transmembrane domain including a short cytoplasmic tail is fused by a short linker sequence encoding a flexible glycin/serin stretch to the DNA sequence encoding different derivatives of the HIV-1 external glycoprotein gp120.
  • a 5 ' DNA sequence derived from the lnterieukin-3 (IL-3) gene and encoding the IL-3 signal peptide is connected via a short multiple cloning site with the DNA sequence encoding the glycin/serin linker and the EBV gp220/350 transmembrane anchor sequence.
  • IL-3 lnterieukin-3
  • the multiple cloning site allows the insertion of any other DNA sequence encoding an immunologicaily relevant protein.
  • the NH 2 terminal fused IL-3 signal sequence induces the transport of the chimeric proteins via endoplasmic reticulum to the cytoplasmic membran upon expression of the construct in eucaryotic cells.
  • Anchoring of said chimeric polypeptides in the cell membrane is achieved by the EBV gp220/350 transmembrane anchor sequence fused to the COOH-terminus of the chimeric polypeptides.
  • retroviral gag polypeptides with authentic (occurring in nature) or chimeric (not occurring in nature) membrane proteins spontaneously leads to the formation retrovirus-like particles, which are spiked with the authentic or chimeric membrane proteins.
  • said spiked retrovirus- like particies are secreted into the cell culture supematantallowing the recovery of the expression product from the medium.
  • spiked retrovirus- like particles in a baculovirus dependent expression system in insect cells, (ii) in stably transfected Drosophila Schneider cells, (iii) in a Semliki-Forest virus driven expression system or (iv) in any other mammalian cell line such as CHO cells.
  • Polyvalent antigens which contain at least one antigenic domain are suitable for diagnosis of a variety of infectious agents and neoplasias based on antibodies binding to the antigens presentet on the surface of retrovirus-like particles.
  • Said recombinant VLPs represent a pharmaceutical composition delivering at least one antigenic domain suitable for prevention and therapy of a variety of infectious agents and neoplasias to the immune system by means of inducing a humoral and cell mediated immune response.
  • VLPs represents a general method of preventing or treating HlV-infection. EBV-infection or EBV-related diseases and EHV-infection after administration to humans or horses in amounts sufficient to modulate or induce an immune response.
  • Figure 1 Scematic drawing illustrating the construction of the plasmids encoding the chimeric gp160 and gp120 genes gp160, gp120/TM, gp120 5 "/ M and gp120 °-/TM.
  • the numbers below the hatched boxes refer to the 1 st nucleotide of the coding region (A of ATG start codon); . refers to synthetic oligonucleotides (OI)
  • OI 3a and OI 4a were synthetic oligonucleotides used to generate a PCR fragment encoding the IL-3 signal peptide from annealed and filled up oligonucleotides OI 3 and OI 4.
  • the redigested 75 nucleotide (nc) PCR product was inserted into the EcoRI/Kspl site of plin20 to generate plin20-S.
  • OI 7 and OI 8 were used to generate a PCR fragment encoding a 6 aa Gly/Ser hinge strech, fused to the EBV gp220/350 transmembrane domaine NH2-terminus.
  • the Mrol/Pstl redigested PCR fragment (153 bp in length) was inserted into the Mrol/Pstl site of plin20-S to generate plin20-ST.
  • Figure 2 Expression of the rgp160 and chimeric rgp120 derivatives in insect cells.
  • Spodoptera frugiperda cells were infected with recombinant baculoviruses rAc160 (lane 4), rAc120/TM (lane 5), rAc120 5 - TM (lane 6) and rAc120 20 YTM (lane 7) at a MOI of 10.
  • Sf 9 cells were either not infected (lane 1), or infected with wildtype baculovirus (lane 2) or a recombinant baculovirus expressing the H1V-1 Pr559 ⁇ g gene product (rAcgag; lane 3). Correct expression of the different HIV-1 gp160/120 derivatives was proven by analyzing extracts of 10 4 infected cells harvested 3 days p.i.
  • Figure 3 Co-expression of the chimeric HIV-1 envelope proteins in insect cells.
  • HighFive insect cells were co-infected with a Pr55Sra9 recombinant baculovirus (rAcgag) and a recombinant baculovirus expressing one of the HIV-1 envelope constructs rAc160 (lane 4) or rAc120/TM (lane 5) or rAc120 5 7TM (lane 6) or rAc120 20 7TM (lane 7) at a MOI of 10 for each virus, respectively.
  • rAcgag Pr55Sra9 recombinant baculovirus
  • rAcgag a recombinant baculovirus expressing one of the HIV-1 envelope constructs rAc160 (lane 4) or rAc120/TM (lane 5) or rAc120 5 7TM (lane 6) or rAc120 20 7TM (lane 7) at a MOI of 10 for each virus, respectively.
  • FIG. 4 Expression of gp160 or derivatives thereoff on the surface of recombinant retrovirus-like particles (VLP).
  • Sf 9 cells were either not infected (lane 1), or infected with wildtype baculovirus (lane 2) or coinfected with wildtype baculovirus and a recombinant baculovirus expressing the HIV-1 Pr559- i 9 gene product (rAcgag; lane 3).
  • Immunoprecipitates were separated by SDS-Page and analyzed after conventional western blotting. Recombinant antigens were detected by using using monoclonal antibodies to ⁇ 24 (16/4/2) (C) and to the V3-domain within gp120 (D). Positions of the molecular weight standart are given from the left, positions of specifically detected recombinant proteins are indicated at the right side of the figure.
  • VLPs Virus-like particles spiked with gp14 were generated in insect cells by co- infection of two different recombinant baculoviruses. 4 days p.i. supernatants were harvested and the particles were collected by isopycnic centrifugation in a sucrose gradient and checked for purity by electron microscopy. Five microliters of these preparations were run in a 15% SDS-PAGE, transferred to nitrocellulose and probed with anti-HIV gag mab 16/4/2 or with anti-EHV-1 serum 528/84. HIV-VLPs produced by infection with rAcgag alone and harvested at 72 h p.i. were used as a control (lane Co). The MWs of the reactive proteins are indicated in kD.
  • FIG. 7 Immunoeiectron microscopy of VLPs.
  • VLP-gp14 preparations were adsorbed to grids and incubated with anti-gp14 mab 3F6.
  • Bound mab 3F6 was detected with an anti-mouse IgG gold conjugate and and analyzed by electron microscopy. A representative immunogold labeled particle is shown.
  • Panel A shows the mean increases in ear thickness of two individual mice after i.m. immunization at 0, 24 and 48 h post inoculation of inactivated RacL11 and uninfected cell culture supernatants (see Materials and Methods).
  • Panel B shows the DTH response of mice immunized i.nas. with the same antigens. Standard deviations ranged from 0 to 4% and are not shown.
  • Figure 9 shows the mean increases in ear thickness of two individual mice after i.m. immunization at 0, 24 and 48 h post inoculation of inactivated RacL11 and uninfected cell culture supernatants (see Materials and Methods).
  • Panel B shows the DTH response of mice immunized i.nas. with the same antigens. Standard deviations ranged from 0 to 4% and are not shown.
  • Figure 9 shows the mean increases in ear thickness of two individual mice after i.m. immunization at 0, 24 and 48 h post inoculation of in
  • Example 1 Development of vector modules allowing the construction of authentic or chimeric membrane proteins (fig. 1).
  • PCR polymerase chain reaction
  • plin20-S The 5 ' 75 nucleotides of the murine interleukin 3 (IL-3) gene encoding a eucaryotic signal sequence were cloned into the above described plin20 vector.
  • Two overlapping synthetic oligonucleotides (OI 3 and OI 4) served after annealing and filling up the protruding single stranded- DNA sequences as template in a PCR (reaction 1).
  • Two amplification primers OI 3a and OI 4a were used to amplify the IL-3 leader template and to introduce terminal restriction sites.
  • the reaction yielded a double-stranded synthetic oligonucleotide containing a EcoRI- restriction site at the 5 ' - and a Kspl -restriction site at the 3 ' -end.
  • this double-stranded oligonucleotide was cleaved with EcoRI/Kspl and inserted into the EcoRI/Kspl linearized plin20.
  • the introduction of the 5 ' Ksp restriction site into the gp 160 open reading frame resulted in a conversion of the residues 32 (E) and 33 (K) of HIV-1 HX-10 isolate to A, E, N.
  • the subcloned HIV-1 gp160 fragment reasembles the gp160 sequence from amino acid (aa) position 31 to 856 (nucleotide position 6314-8791).
  • (D) Construction of pfin20-ST To generate the plin20-ST plasmid the coding region of the EBV gp 220/350 transmembrane (TM) domaine was amplified in a PCR reaction using the plasmid pBRBamHI-L as a template and the oligonucleotides 7 and 8 as primer. Furthermore the oligonucleotides 7 and 8 introduced a Mrol restriction site at the 5 ' - and a Pstl restriction site at the 3 ' -end respectively, flanking the EBV gp220/350 TM coding sequence.
  • TM transmembrane
  • the 5 ' primer OI 7 additionally accomplished the fusion of the Gly/Ser hinge region coding to the 5 ' end of the EBV gp220/350 TM domaine coding nucleotide-sequence.
  • the PCR product was digested with Mrol/Pstl and ligated into the plin20-S vector.
  • the position of the cloned EBV fragment in the virus genome is nucleotide 89433-89576 on the complementary strand corresponding to aa 860 - aa 907 of the EBV gp220/350 (EBV B95-8, Baer et al. 1984, Nature 310: 207-211 ; Genebank, accession V01555).
  • All subgenomic fragments included a Kspl restriction site at the 5 ' - and a Mrol restriction site at the 3 ' -end. Using these restriction sites all three gp120 derivatives were subcloned into the plin20-ST vector.
  • the cloned HIV-1 gp120 variants reasemble the aa sequences from residue 31 to residue 506, 502 and 487 (the corresponding nucleotide positions are 6314 - 7741 , 7729 and 7684) for 120, 120 5 "/TM and 120 20 7TM respectively.
  • above described gp160/120 derivatives are referred to as gp160, gp120 TM, gp120 5 7TM and gp120 20 7TM.
  • the HIV-1 gp160/120TM derivative constructs exhibiting the autologous HIV-1 signal sequence were obtained.
  • the PCR amplifications of above mentioned gene segments were performed using the pNL4-3 template, one 5 ' primer (OI 10) for all four constructs (introducing a 5 ' EcoRI restriction site) and the above described 3 ' primer OI 6, OI 9a, OI 9b or OI 9c.
  • the resulting DNA fragments were subcloned into plin20 derivatives.
  • the gp160 original coding region (aa 1-856) was inserted into plin20 after EcoRI/Pstl digestion and ligation.
  • gp120 coding sequences (gp120: aa 1-506, gp120 5 ': aa 1-502, gp120 20 " aa 1-487) were subcloned into plin20-ST using the EcoRI/Mrol restriction sites.
  • Example 2 Subcloning of the chimeric HIV-1 envelope genes into the baculovirus transfervector pVL1393 and construction of recombinant baculoviruses.
  • the EcoRI/Pstl DNA fragments encoding the gp160 and gp120 derivatives have been subcloned into the EcoRI/Pstl site of the transvervector pVL1393. Plasmid DNAs have been purified by using a Quiagen tip 100 kit (Diagen).
  • Spodoptera frugiperda cells were infected by recombinant baculoviruses (rAc160, rAc120/TM, rAc120/ 5 M, rAc120/ 2 °-TM) at a MOI of 10. Correct expression of the different HIV-1 gp160/120 derivatives was proven by analyzing extracts of 10 4 infected cells by conventional Western blot analysis. Briefly, the cell lysates were diluted in sample buffer (Sambrook et al., 1989), separated by electrophoresis on 10- 12,5% SDS-polyacrylamide gels and transferred to nitrocellulose (Schleicher and Schuell) by electroblotting.
  • Sheets were incubated for 1 h at RT with 10% nonfat dry milk in Tris-buffered saline (TBS) containing 0.05% Tween 20 (Sigma) and washed in TBS-Tween 20. Filters were then incubated overnight at 4°C with mabs to the third variable domain V3 of gp120 (DuPont 9303). After removing the antibodies, blots were washed twice with TBS-Tween20 and incubated for 1 h at RT with anti-mouse IgG-POD conjugate. Blots were washed again and substrate (4-chloro-1-naphtole) was added (fig. 2). Exposition of the envelope derivatives on the cell surface was proven by immunoflourescence analysis and confirmed by FACSscan analysis (tab.1).
  • infected insect cells were fixed with paraformaldehyde (1% in PBS). Cells were incubated with a mab directed to the V3 domain of gp120 (1/100 dilution in PBS), washed twice and incubated for 10 min at RT with an anti-mouse IgG fluoroisothio-cyanate (FITC) conjugate. After two washes in PBS, DNA was stained with propidium iodide and samples were analyzed with a fluorocytometer (FACSscan, Becton- Dickinson) or by UV-microscopy.
  • FACSscan Fluorocytometer
  • Example 4 Co-expression of the chimeric H1V-1 envelope proteins in insect cells.
  • Example 5 Analysis of particle formation. Ultrathin sections of insect cells coexpressing Pr559 a 9 and the gp120 derivatives (fig. 1 : gp160, gp120/TM, gp120/ 5" TM, gpl20 2°-TM) after co-infection with the respective recombinant baculoviruses revealed efficient budding of recombinant VLP (not shown).
  • Example 6 Expression of gp160 or derivatives thereoff on the surface of recombinant retrovirus-like particles (VLP).
  • gp160 or derivatives thereoff on the surface of recombinant retrovirus- like particles was assessed by a co-immunoprecipitation analysis from the antigenic peak fractions of the sucrose gradients. Immunoprecipitations were performed according to standart procedures (Sambrook et al., 1989) with 10 ⁇ l of a gp120 V3- loop specific murine monoclonal antibody (DuPont 9303) in absence of detergent. Immunoprecipitates were diluted in sample buffer (Sambrook et al., 1989), separated by electrophoresis on 10-12,5% SDS-polyacrylamide gels and transferred to nitrocellulose (Schleicher and Schuell) by electroblotting.
  • the Pr559 a 9 precorsor has been co-immunoprecipitated only from peak fractions of the sucrose sedimentation analysis which were derived from supernatants of co-infected cells. This clearly indicates the gp160 or derivatives thereoff are exposed on the surface of the infected cells.
  • co-expression of P ⁇ 559 a 9 with the wild type gp160 construct significantly reduces the exposition of the envelope protein if compared to the chimeric derivatives of gp120 (fig. 4 C, D).
  • VLPs were generated by co-infection of HighFive cells with a multiplicity of infection (MOI) of 1 per cell with the recombinant baculoviruses expressing gp160 (rAc160), gp120 or dervatives thereoff (rAc120TM, rAc120/ 5 M, rAc120/ 20 "TM) and rAcgag, the latter encoding the HIV 55 kD gag protein Pr550 a S.
  • Supernatants of infected HighFive cells were collected 4 days p.i., purified by isopycnic sucrose gradient centrifugation as described above, and checked for purity and absence of baculoviruses by electron microscopy. After dilution of the baculovirus-free fractions VLP were pelletted by centrifugation in a TFT 41.14 rotor in Kontron centrifuge and resuspended PBS.
  • Example 8 Induction of a humoral immune response by recombinant VLPs.
  • Antigen ELISA Micro-ELISA plates (Greiner, Frickenhausen, Germany) were coated with 500 ng HIV-1 HX10 lysate, 500 ng recombinant p24, 80 ng rgp120 or 300 ng V3-peptide (36mer)/well in 50 ⁇ l 0.05 M sodium carbonate puffer pH 9.5 at 4°C overnight in a wetchamber. Sera diluted 1 :10 to 1 :1000 in PBS with 3% FCS and 2% Tween-20 were added to the coated wells. After incubation for 2 hours at 37°C the plates were washed 5 times.
  • Bound antibody was detected with horseradish peroxidase-conjugated anti-rabbit antibody (Dakopatts, Copenhagen, Denmark) at a dilution of 1 :1000, followed by incubation with o-phenylendiamine-0.01% hydrogen peroxide in phosphate buffered saline (pH 6.0). The reaction was stopped by adding 1 M H2SO4 and read at 492 nm. Values above the mean optical density +3 SD of negative controls were considered positive. All rabbits immunized with recombinant VLPs developed high titers of antibodies ranging from 1/64000 to the Pr55-? a -3 carrier component to 1/64000-1/32000 towards purified gp120. Only low titers (1/256) to a 36 amino acid peptide representing the homologous V3-peptide hav been detected.
  • HIV-1 neutralisation assays Serial twofold dilutions of heat inactivated serum were incubated for 1.5 h at 37°C with 50 TCID50 of H ⁇ v " 1 HX10-strain produced on MT4 cells. The virus-serum mixture was incubated for 1 h at 37°C with 5x10 4 MT4 suspension cells. After virus adsorbtion, the unbound virus was removed and 100 ⁇ l of medium (RPMI-1640 supplemented with 10% FCS) was added to each well. 7 days p.i., 100 ⁇ l of supernatant medium was removed in order to quantify the amount of virions released from the cells using a commercial p24 sandwich assay (Abbott Laboratories, Chicago III.).
  • Neutralisation titres were calculated by comparison with control wells of virus only and are expressed as the reciprocal of the highest serum dilution that inhibited p24 production and release by more than 90 %.
  • Sera from rabbits obtained after 3 immunisations were used for the neutralisation assay. As shown in table 2 ail of the sera showed neutralization titers in a range of 1 :128 to 1 :256, depending to the Pr559 a 9/er,v preperation used for immunisation.
  • Example 9 Induction of a cell mediated immune response by recombinant VLPs
  • BALB/cJ mice H-2 d were immunized with 10 ⁇ g of the different gag/env hybrid VLP in complete absence of adjuvants or replicating vector.
  • BALB/c mice were injected with 50 ⁇ g of a 16mer V3-peptide (RIQRGPGRAFVTIGKI) or 10 ⁇ g of Pr559 9 VLP only.
  • Lymphoid cells were prepared from immuniced mice 6 days post immunisation and cocultivated with syngenic V3-16mer peptide labelled syngenic P815 cells, irradiated with 20000rad.
  • a control group included unprimed BALB/c cells stimulated in vitro with V3 peptide labelled P815 cells. Cytotoxic effector cell populations were harvested after 5 days of in vitro culture. The cytotoxic response was determined against the syngenic target cell line A20 pulsed for 1 h with 10" 8 M V3-16mer peptide. Negative controls were not pulsed target A20 cells in a standard 51 Cr release test. Neither the synthetic V3- peptide nor Pr559 a 9 VLP were sufficient to prime an adequate V3-specific CTL response. A comparably weak CTL response could be demonstrated after administration of purified gp160 (fig.5).
  • Example 10 Presentation of membrane proteins derived from other viruses than HIV-1 by recombinant VLPs: The equine herpesvirus gp14 (gB)
  • VLPs were generated by. co-infection of HighFive cells with a multiplicity of infection (MOI) of 1 per cell with the recombinant baculoviruses rAd 7-11 expressing the EHV gp14 membrane protein (Osterrieder et al. 1994) and rAcgag, the latter encoding the HIV 55 kD gag protein Pr559 a 9.
  • MOI multiplicity of infection
  • Example 11 Humoral immune response to recombinant gp14 induced by chimeric VLPs in comparison to other antigen formulations
  • rAc17-11-gp14 rgp14 exressed in and purified from insect cells after infection with a gp14 recombinant baculovirus; (Osterrieder er al. 1994)) of VLP-gp14 (recombinant HIV-1 Pr559 a 9 VLPs presenting gp14 on their surface)
  • pDES-gp14 DNA vaccine; pcDNA/Amp (Invitrogen) derived expression plasmid containing the EHV-1 gp14 without its transmembrane and cytoplasmic domain under the control of the CMV immediate early promotor; Osterrieder et al., in press
  • the ELISA and NT antibody titres obtained for the specific gp14-formulations in immunized mice are summarized in table 3.
  • the highest ELISA and NT antibody titres were observed after im. and inas. immunization with gp14-spiked VLPs.
  • the ELISA antibody titres were even higher than those obtained after i.m. and i.nas. application of live EHV-1 virus (RacL11 ; RacH).
  • Example 12 DTH response to recombinant gp14 induced by chimeric VLPs in comparison to other antigen formulations
  • the DTH response after immunization with the various recombinants was assayed by determining the increase in ear thickness of two individual mice at different time points post inoculation (p. inoc.) of inactivated antigen into the ear pinna.
  • p. inoc. time points post inoculation
  • a readily detectable increase in ear thickness was observed in mice immunized with VLPs and the E.Coli derived gp14 (pCEP-8-gp14) via the i.m. and i.nas route. This reaction was decreasing at 48 h p.inoc.
  • Example 13 Clinical observations in mice immunized with gp14 chimeric VLPs in comparison to other antigen formulations after challenge with wt- EHV-1
  • mice were challenged inas with 106.5 PFU of EHV-1 wt strain RacL11. All mice immunized with BSA developed signs of illness such as ruffled fur, respiratory symptoms, and hunched posture associated with a dramatic loss of body weight of up to 23% by day 2 after challenge infection. Similar observations have been reported for EHV-1 strain Ab4 (Awan et al. 1990).
  • mice immunized im with pDES1 DNA vaccine In contrast to others (Awan et al. 1990; Inazu et al. 1993), no deaths in BSA- immunized mice were observed after challenge infection and animals recovered from the weight losses by day 4/5 p.chall. but did not reach the preinfection weight until day 8 p.chall. In contrast, no or only a mild (up to 6%) decrease in the mean body weights was observed after challenge infection of mice immunized im with all recombinant gp 14 formulations and subsequent challenge infection. In mice immunized im with pDES1 DNA vaccine, however, one individual mouse exhibited ruffled fur, dyspnoea, and a body weight loss of 15% on Day 4 p.chall..
  • pCEP-8- gp14- and pDES-1 immunized mice one and two individual animals, respectively, showed signs of illness and body weight losses of up to 20% after challenge infection was seen in mice previously infected inas with the live virus strains RacL11 or RacH (fig. 9).
  • Example 14 Virus reisolation from mice immunized with gp14 chimeric VLPs in comparison to other antigen formulations after challenge with wt- EHV-1
  • virus was recovered from lungs of two individual mice killed on days 1 , 3, 5 and 8 p. chall., respectively.
  • virus titers recovered from lungs were higher compared to those after immunization with RacL, RacH or the VLPs, but we were not able to demonstrate any virus on day 5 p.chall..
  • pCEP-8-gp14 E.Coli derived gp14
  • protection against challenge infection appeared to be efficient after both im. and inas. immunization.
  • the virus titres in lungs were in general comparable to those seen in insect cell derived gp14 (rAc17-11gp14) immunized mice and were also reduced significantly after i.m. and i.nas. immunization from day 3 p.chall. (p ⁇ 0.05).
  • mice immunized with gp14-spiked VLPs both i.m. and i.nas. were found to be best protected against subsequent EHV-1 challenge.
  • ° Serum antibody leveis were tested by ELISA three weeks after the second booster immunization and expressed u . reciprocal o the dilution that gave rise to one half the maximal density at 492 nm (midpoint tire). The antibody levels were determined against various antigens such as HIV-1 lysate. rp24, rgpl20 and a synthetic V3 peptide (36-mer). Titers below 1/16 were considered to be unspecific and reactions counted as negative. c Reciprocal of the highest serum dilution that inhibited p24 production by more than 90% 7 days p.i. were classified as neutralizine.
  • Endpoint ELISA titers were determined with purified EHV-1 virions ( 1 ⁇ g/ml) by twofold log 2 dilutions of sera starting with a
  • SN titers were determined on Rk-u cells with 50 TCID50 RacH and triplicate log 2 dilutions of sera inactivated at 56° for 30 min. No complement was added. Titers reflect the serum dilution with complete protection in all wells.
  • Electroporation and commercial liposomes efficiently deliver soluble protein into the MHC class I presentation pathway. Priming in vitro and in vivo for class l-restricted recognition of soluble antigen. J. Immunol. Methods 160, 49-57.
  • Liposome-mediated delivery stimulates a class l-restricted cytotoxic T cell response to soluble antigen. Eur. J. Immunol. 22, 2 ⁇ 7 - 290.
  • HIV-1 gag-specific cytotoxic T lymphocytes defined with recombinant vaccinia virus and synthetic peptides. Nature 336, 484-487.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Virology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
EP96911989A 1995-03-31 1996-04-01 Antigen presentation system based on retrovirus-like particles Withdrawn EP0817854A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP96911989A EP0817854A2 (en) 1995-03-31 1996-04-01 Antigen presentation system based on retrovirus-like particles

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
EP95104850 1995-03-31
EP95104849 1995-03-31
EP95104848 1995-03-31
EP95104848 1995-03-31
EP95104850 1995-03-31
EP95104849 1995-03-31
EP96911989A EP0817854A2 (en) 1995-03-31 1996-04-01 Antigen presentation system based on retrovirus-like particles
PCT/EP1996/001433 WO1996030523A2 (en) 1995-03-31 1996-04-01 Antigen presentation system based on retrovirus-like particles

Publications (1)

Publication Number Publication Date
EP0817854A2 true EP0817854A2 (en) 1998-01-14

Family

ID=27236505

Family Applications (1)

Application Number Title Priority Date Filing Date
EP96911989A Withdrawn EP0817854A2 (en) 1995-03-31 1996-04-01 Antigen presentation system based on retrovirus-like particles

Country Status (3)

Country Link
EP (1) EP0817854A2 (zh)
CN (1) CN1185811A (zh)
WO (1) WO1996030523A2 (zh)

Families Citing this family (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA01005389A (es) 1998-11-30 2003-03-27 Cytos Biotechnology Ag Presentacion molecular ordenada de antigenos, metodos de preparacion y uso.
US7935805B1 (en) 1998-12-31 2011-05-03 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV Type C polypeptides, polypeptides and uses thereof
JP2002533124A (ja) 1998-12-31 2002-10-08 カイロン コーポレイション Hivポリペプチドの改善された発現およびウイルス様粒子の生成
WO2000039304A2 (en) 1998-12-31 2000-07-06 Chiron Corporation Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof
US20040014033A1 (en) 1999-06-30 2004-01-22 Evotec Biosystems Ag, A Corporation Of Germany Virus like particles, their preparation and their use preferably in pharmaceutical screening and functional genomics
ATE362990T1 (de) * 1999-06-30 2007-06-15 Evotec Ag Virusähnliche partikel, vorbereitung und verwendung in screening und funktionelle genomanwendung
US7476517B2 (en) 1999-06-30 2009-01-13 Evotec Ag Virus like particles, their preparation and their use preferably in pharmaceutical screening and functional genomics
DE19957838C2 (de) 1999-11-25 2002-11-21 Pette Heinrich Inst Gentherapie von HIV-Infizierten durch Expression von Membran-verankerten gp41-Peptiden
US20020106798A1 (en) * 2000-03-02 2002-08-08 Robinson Harriet L. DNA expression vectors and methods of use
WO2001085208A2 (en) 2000-05-05 2001-11-15 Cytos Biotechnology Ag Molecular antigen arrays and vaccines
DE60037263T2 (de) * 2000-12-29 2008-10-09 Evotec Ag Virus-ähnliche Partikel, ihre Herstellung und ihre Verwendung beim pharmazeutischen Screening und funktionelle Genomanwendungen
US7320793B2 (en) 2001-01-19 2008-01-22 Cytos Biotechnology Ag Molecular antigen array
US7128911B2 (en) 2001-01-19 2006-10-31 Cytos Biotechnology Ag Antigen arrays for treatment of bone disease
US7094409B2 (en) 2001-01-19 2006-08-22 Cytos Biotechnology Ag Antigen arrays for treatment of allergic eosinophilic diseases
EP2280074A3 (en) 2001-07-05 2011-06-22 Novartis Vaccines and Diagnostics, Inc. Polynucleotides encoding antigenic HIV type B and/or type C polypeptides, polypeptides and uses thereof
AU2002320314A1 (en) 2001-07-05 2003-01-21 Chiron, Corporation Polynucleotides encoding antigenic hiv type c polypeptides, polypeptides and uses thereof
WO2003012085A1 (en) * 2001-07-30 2003-02-13 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Antigen presenting cells, method for their preparation and their use for cancer vaccines
EP1450856B1 (en) 2001-09-14 2009-11-11 Cytos Biotechnology AG Packaging of immunostimulatory cpg into virus-like particles: method of preparation and use
US7115266B2 (en) 2001-10-05 2006-10-03 Cytos Biotechnology Ag Angiotensin peptide-carrier conjugates and uses thereof
BR0213117A (pt) 2001-10-05 2004-09-21 Cytos Biotechnology Ag Conjugados peptìdeo angiotensina-veìculo e seus usos
CN102061288A (zh) 2002-07-17 2011-05-18 希托斯生物技术股份公司 使用衍生自ap205外壳蛋白的病毒样颗粒的分子抗原阵列
BR0312297A (pt) 2002-07-18 2005-04-12 Cytos Biotechnology Ag Conjugados veìculos de hapteno e seu uso
SI1524994T1 (sl) 2002-07-19 2011-08-31 Cytos Biotechnology Ag Sestavki cepiv, ki vsebujejo amiloidne beta 1-6 antigenske mreĹľe
US7537767B2 (en) 2003-03-26 2009-05-26 Cytis Biotechnology Ag Melan-A- carrier conjugates
AU2004224762B2 (en) 2003-03-26 2009-12-24 Kuros Us Llc Packaging of immunostimulatory oligonucleotides into virus-like particles: method of preparation and use
WO2006050394A2 (en) 2004-11-01 2006-05-11 Novartis Vaccines And Diagnostics Inc. Combination approaches for generating immune responses
EP1868642B1 (en) 2005-03-18 2013-05-08 Cytos Biotechnology AG Cat allergen fusion proteins and uses thereof
EP1736538A1 (en) 2005-06-21 2006-12-27 Cytos Biotechnology AG Process for the preparative purification of virus-like-particles (VLPs)
AU2006298767B2 (en) 2005-09-28 2013-01-10 Cytos Biotechnology Ag Interleukin-1 conjugates and uses thereof
AU2006325225B2 (en) 2005-12-14 2013-07-04 Cytos Biotechnology Ag Immunostimulatory nucleic acid packaged particles for the treatment of hypersensitivity
MX2008015529A (es) 2006-06-12 2009-01-13 Cytos Biotechnology Ag Procesos para empacar oligonucleotidos en particulas similares a virus de bacteriofagos de arn.
US9296790B2 (en) 2008-10-03 2016-03-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods and compositions for protein delivery
MX337723B (es) 2008-12-09 2016-03-15 Pfizer Vaccines Llc Vacuna de peptido ch3 de ige.
IN2012DN00446A (zh) 2009-07-30 2015-05-15 Pfizer Vaccines Llc
CA2846746A1 (en) 2009-09-03 2011-03-10 Pfizer Vaccines Llc Pcsk9 vaccine
EP2575868A1 (en) 2010-06-07 2013-04-10 Pfizer Vaccines LLC Ige ch3 peptide vaccine
EP2680883B1 (en) 2011-03-02 2018-09-05 Pfizer Inc Pcsk9 vaccine
US20140004146A1 (en) * 2011-03-17 2014-01-02 Institut Pasteur Of Shanghai, Chinese Academy Of Sciences Method for producing virus-like particle by using drosophila cell and applications thereof
EP3663395A1 (en) 2012-03-26 2020-06-10 The United States of America, as Represented by The Secretary, Department of Health and Human Services Office of Technology Transfer Delivery of packaged rna to mammalian cells
MX2016007726A (es) 2013-12-16 2016-10-28 Us Health Inmunoterapia de cancer mediante el suministro de antigenos mhc clase ii usando un replicon de vlp.
EP3089755A1 (en) 2014-01-03 2016-11-09 Fundacion Biofisica Bizkaia VLPs, METHODS FOR THEIR OBTENTION AND APPLICATIONS THEREOF
WO2015123291A1 (en) 2014-02-11 2015-08-20 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Pcsk9 vaccine and methods of using the same
CN104450631A (zh) * 2014-11-11 2015-03-25 中国人民解放军第四军医大学 肠道病毒ev71型vp1基因病毒样颗粒及其制备方法和应用
CN105177031B (zh) * 2015-06-12 2018-04-24 北京艺妙神州医疗科技有限公司 嵌合抗原受体修饰的t细胞及其用途
WO2017179025A1 (en) 2016-04-15 2017-10-19 Alk-Abelló A/S Epitope polypeptides of ragweed pollen allergens
WO2019173438A1 (en) 2018-03-06 2019-09-12 Stc. Unm Compositions and methods for reducing serum triglycerides
CN115279905A (zh) 2019-10-23 2022-11-01 里珍纳龙药品有限公司 合成rig-i样受体激动剂
WO2021250628A1 (en) 2020-06-12 2021-12-16 Glaxosmithkline Biologicals Sa Bacterial immunization using nanoparticle vaccine
WO2023111826A1 (en) 2021-12-14 2023-06-22 Glaxosmithkline Biologicals Sa Bacterial immunization using qbeta hairpin nanoparticle constructs

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8923123D0 (en) * 1989-10-13 1989-11-29 Connaught Lab A vaccine for human immunodeficiency virus
HUT60506A (en) * 1989-11-20 1992-09-28 Oncogen Process for producing non-replicable, recombinant retrovirus particles and antiviral and immunogenic preparations
WO1991019803A1 (en) * 1990-06-19 1991-12-26 Applied Biotechnology, Incorporated Self assembled, defective, nonself-propagating viral particles
JPH06500232A (ja) * 1990-08-15 1994-01-13 サリオン・バイオロジクス・コーポレイシヨン 自己集合性複製欠陥型ハイブリッドウイルス粒子
GB9107631D0 (en) * 1991-04-10 1991-05-29 British Bio Technology Proteinaceous particles
CA2098029C (en) * 1993-06-09 2000-10-03 Benjamin Rovinski Retrovirus-like particles containing modified envelope glycoproteins
US5955342A (en) * 1994-08-15 1999-09-21 Connaught Laboratories Limited Non-infectious, replication-defective, self-assembling HIV-1 viral particles containing antigenic markers in the gag coding region

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9630523A2 *

Also Published As

Publication number Publication date
WO1996030523A2 (en) 1996-10-03
WO1996030523A3 (en) 1997-04-17
CN1185811A (zh) 1998-06-24

Similar Documents

Publication Publication Date Title
WO1996030523A2 (en) Antigen presentation system based on retrovirus-like particles
Doan et al. Virus‐like particles as HIV‐1 vaccines
Hanke et al. DNA multi-CTL epitope vaccines for HIV and Plasmodium falciparum: immunogenicity in mice
JP4805511B2 (ja) Hivに対する免疫応答における改善、または免疫応答に関する改善
EP0969862B1 (en) Synthetic hiv gag genes
WAGNER et al. Construction, expression, and immunogenicity of chimeric HIV-1 virus-like particles
Buonaguro et al. High efficient production of Pr55gag Virus-like Particles expressing multiple HIV-1 epitopes, including a gp120 protein derived from an Ugandan HIV-1 isolate of subtype A
Griffiths et al. Hybrid human immunodeficiency virus Gag particles as an antigen carrier system: induction of cytotoxic T-cell and humoral responses by a Gag: V3 fusion
EP0904380B1 (en) Synthetic hiv genes
JP2002533124A (ja) Hivポリペプチドの改善された発現およびウイルス様粒子の生成
WO1996006177A2 (en) Retrovirus-like particles made non-infectious by a plurality of mutations
WO1996005292A1 (en) Antigenically-marked non-infectious retrovirus-like particles
Kang et al. Development of HIV/AIDS vaccine using chimeric gag-env virus-like particles
Katz et al. The cytoplasmic and transmembrane domains of the vaccinia virus B5R protein target a chimeric human immunodeficiency virus type 1 glycoprotein to the outer envelope of nascent vaccinia virions
BRAND et al. Comparative analysis of humoral immune responses to HIV type 1 envelope glycoproteins in mice immunized with a DNA vaccine, recombinant Semliki Forest virus RNA, or recombinant Semliki Forest virus particles
Kameoka et al. Cytotoxic T lymphocyte response in mice induced by a recombinant BCG vaccination which produces an extracellular α antigen that fused with the human immunodeficiency virus type 1 envelope immunodominant domain in the V3 loop
US20050287174A1 (en) Immunizing against HIV infection
CA2539864A1 (en) Hiv pharmaccines
KR101370817B1 (ko) Hiv 백신
JP2003516741A (ja) コドン最適化HIV−1Nef及び修飾HIV−1Nefを発現するポリヌクレオチドワクチン
WO1994020621A2 (en) Proteinaceous particles containing retroviral element
Wang et al. Cellular immune responses to helper-free HSV-1 amplicon particles encoding HIV-1 gp120 are enhanced by DNA priming
Notka et al. Construction and characterization of recombinant VLPs and Semliki-Forest virus live vectors for comparative evaluation in the SHIV monkey model
Mustafa et al. HIV-1 Env glycoproteins from two series of primary isolates: replication phenotype and immunogenicity
JP2003520786A (ja) コドン最適化hiv−1polおよび修飾hiv−1polを発現するポリヌクレオチドワクチン

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19971031

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH DE ES FR GB IE IT LI NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20011101