US20040009177A1 - Compositions and methods for modulating RSV infection and immunity - Google Patents

Compositions and methods for modulating RSV infection and immunity Download PDF

Info

Publication number
US20040009177A1
US20040009177A1 US10/420,387 US42038703A US2004009177A1 US 20040009177 A1 US20040009177 A1 US 20040009177A1 US 42038703 A US42038703 A US 42038703A US 2004009177 A1 US2004009177 A1 US 2004009177A1
Authority
US
United States
Prior art keywords
glycoprotein
rsv
peptide
composition
respiratory syncytial
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
US10/420,387
Other languages
English (en)
Inventor
Ralph Tripp
Les Jones
Larry Anderson
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.)
Individual
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22911027&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20040009177(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US10/420,387 priority Critical patent/US20040009177A1/en
Publication of US20040009177A1 publication Critical patent/US20040009177A1/en
Priority to US11/139,372 priority patent/US8173131B2/en
Priority to US13/358,320 priority patent/US8778354B2/en
Priority to US14/252,836 priority patent/US20140288267A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K9/00Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1027Paramyxoviridae, e.g. respiratory syncytial virus
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • RSV epidemics are seasonal, although the virus likely persists within communities. Peak infection rates occur during cold seasons in temperate climates. The virus affects about 90% of infants and young children by the age two. Most often, infection occurs in infants between the ages of six weeks and six months, with the highest incidence in children under three months of age. Previous infection does not prevent repeated infections that are common in all age groups. For example, in a study in Houston, Tex., infection rates were 68.8 per 100 child-years in infancy, and 82.6 per 100 child-years in the second year of life. In a study in Sweden, antibodies to RSV were produced in 87% of children by age 18 months, and in virtually all children by age three.
  • RSV may also be the cause of up to 5% of community-acquired lower respiratory tract infections in adults. In the elderly, RSV infection can be especially serious, with up to 10% of hospitalized cases leading to death.
  • the RSV genome comprises a single strand of negative sense RNA that is 15,222 nucleotides in length and yields eleven major proteins.
  • F fusion
  • G attachment
  • the SH small hydrophobic protein, the M (matrix) protein, and the M2 (22 kDa) protein are associated with the viral envelope but do not induce a protective immune response.
  • mice have shown that the G glycoprotein primes for a Th2 CD4 + T cell response, characterized by production of IL-4, IL-5, IL-13 and pulmonary eosinophilia. Eosinophil recruitment and activation are promoted by several factors, such as IL-4 and IL-5. Pulmonary eosinophilia is associated with significant to severe lung pathology and is presumably, in part, mediated by RSV G glycoprotein-induced Th2 CD4 + cells.
  • Th1 cytokine expression e.g., IL-2 and gamma interferon
  • chemokine mRNA expression e.g., MIP-1 alpha, MIP-1 beta, MIP-2, IP-10, MCP-1
  • decreased NK cell trafficking to the infected lung e.g., MIP-1 alpha, MIP-1 beta, MIP-2, IP-10, MCP-1
  • live RSV vaccines are produced by modifying the RSV G glycoprotein CX3C motif to render it nonfunctional for viral attachment and infection of host cells.
  • Preferred RSV vaccines are engineered by making deletion or insertion mutations in the CX3C motif in a live RSV virus.
  • the light histograms represent cells stained with biotinylated anti-G glycoprotein cocktail (130-2G, 131-2G) or anti-Fkn antibody (51637.11).
  • the percent positive staining of Fkn or G glycoprotein for a representative experiment is shown: a) 88% Fkn staining of CX3CR1-293 cells; B) 8% Fkn staining of 293 cells; C) 84% G glycoprotein staining of CX3CR1-293 cells; D) 16% G glycoprotein staining of 293 cells; E) 72% staining of G glycoprotein in the presence of peptide RT32 and 74% staining of in the presence of peptide RT34 of CX3CR 1-293 cells; F) 48% staining of G glycoprotein in the presence of peptide RT33 of CX3CR1-293 cells; G) 65% staining of G glycoprotein in the presence of peptide ⁇ +1; H) 73% staining of G glycoprotein in the presence of peptide
  • FIGS. 2 A-B show 293-CX3CR1 and 293 cells were incubated with 1 nM 125 I-fractalkine comprising the 76 amino acid CX3C chemokine domain, in the presence of increasing amounts of unlabeled Fkn or G glycoprotein for 2 h at 37° C. in the presence of azide.
  • Non-specific 125 I-fractalkine binding to 293 cells is defined as the total amount of cell-associated radioactivity in the presence of 1000-fold excess of unlabeled Fkn, and was ⁇ 5% of total binding at the concentrations examined.
  • FIG. 6 is a computer-generated representation of an electrophoretic gel showing RNA Protection Analysis (RPA) of Vero cell mRNA from RSV/A2-infected (lane A) and CX3CR1-293 cells (lane B). mRNA was probed for the CXC-chemokine receptor-2 (CXCR2), CXC chemokine receptor-4 (CXCR4), CX3CR1 and housekeeping genes L32 and GAPDH.
  • RPA RNA Protection Analysis
  • FIG. 7 shows RSV plaque reduction by small molecule peptide inhibitors.
  • Vero cells were treated with PBS, RT33, RT34, ⁇ +1, ⁇ 1 and EYp1 peptides (100 ⁇ m) in the presence (B) or absence of heparin (A) (5 ⁇ g/ml). Percentage inhibition was determined by dividing the mean plaque forming units (PFU) of treated Vero cells by the mean PFU of PBS-treated Vero cells.
  • PFU plaque forming units
  • FIG. 8 shows RSV lung titers following in vivo peptide treatment.
  • the lungs of peptide-treated and control mice were harvested at days 3, 5, 7, and 14 post-RSV infection. The results are expressed as PFU/g ⁇ SEM.
  • FIG. 10 shows BAL cell types in RSV-infected mice following in vivo peptide treatment.
  • BAL cells were stained with antibodies against B220 + (A, D, G), CD3 + (B, E, H) and CD11b + (C, F, I) cells. Data is expressed as the total BAL cells/lung ⁇ SEM at days 3, 5, 7 and 14 post-infection. A representative experiment from 3 independent experiments is shown.
  • FIG. 11 shows BAL cell types in RSV-infected mice following in vivo peptide treatment.
  • BAL cells were stained with antibodies against NK (DX5 + ) (A, C, E) and PMN (RB6-8C5 + ) cells (B, D, F). Data is expressed as the total BAL cells/lung ⁇ SEM at days 3, 5, 7 and 14 post-infection. A representative experiment from 3 independent experiments is shown.
  • the chemokine motif of RSV G glycoprotein located at amino acid positions 182-186 is a C-X-X-X-C (or CX3C) motif.
  • the motif is biologically active (i.e., it acts as a CX3C chemokine) and it participates in virus binding to, and infection of, susceptible cells.
  • C is a cysteine residue and X is any amino acid residue.
  • Each of the three X residues can be a different amino acid or the same amino acid, but not cysteine.
  • the CX3C motif binds to the CX3C receptor (CX3CR1) on the surface of human and animal cells, thereby facilitating RSV infection of the cells.
  • live RSV virus vaccines are produced by altering the CX3C motif in a wild type virus in such a way that the virus cannot use it to attach to and infect cells or modulate the host response to viral infection.
  • the virus containing the altered motif is administered as a live virus vaccine to induce an immune response that confers subsequent protection against RSV infection.
  • live RSV vaccines that are produced, or existing live RSV vaccines that are improved, by modifying the RSV G glycoprotein CX3C motif or proximal amino acid residues or other parts of the G glycoprotein so that, when administered to a human or animal, higher titers of antibodies are produced that block the biological function of the CX3C motif on the G glycoproteins of subsequently-infecting RSV viruses.
  • “Higher titer” means an antibody titer which is greater than an antibody titer previously detected upon immunization with existing live RSV vaccines.
  • live and non-live RSV vaccines are provided that, when administered to a human or animal, induce the production of antibodies that block the biological function of the CX3C motif on the G glycoprotein of subsequently-infecting RSV viruses.
  • the vaccine can comprise one or more G glycoprotein fragments, G glycoprotein peptides or polypeptides from different RSV strains, G glycoproteins from non-live virus vaccines, or G glycoproteins from a live virus vaccine, such as an RSV infectious clone, that, when administered to a human or animal, induce the production of antibodies that inhibit CX3C biological function, such as binding to the CX3C receptor or RSV plaque formation in monolayers of susceptible cells or G glycoprotein-induced leukocyte migration.
  • the RSV infectious clone is available from several sources such as the National Institutes of Health (Dr. Brian Murphy) or from Aviron Corporation (Mountain View, Calif.).
  • the vaccine can comprise one or more G glycoprotein peptides or polypeptides from different RSV strains having the foregoing ability.
  • Isolated, recombinant or synthetic proteins or peptides containing the CX3C motif of the RSV G glycoprotein, or active fragments thereof or fusion proteins thereof can be utilized as blocking molecules as described above, but are also useful as scientific research tools to identify and produce other blocking molecules, thereby promoting an understanding of the mechanisms of RSV viral pathology and the development of antiviral therapies.
  • the isolated, recombinant or synthetic proteins, or antigenic portions thereof (including epitope-bearing fragments), or fusion proteins thereof can be administered to animals as immunogens or antigens, alone or in combination with an adjuvant, for the production of antisera reactive with the CX3C motif.
  • the proteins can be used to screen antisera from hyperimmune patients from whom antibodies having a very high affinity for the proteins can be derived.
  • the proteins, peptides or polypeptides of this invention contain the CX3C motif and all or a biologically active or immunogenic portion of the amino acid sequence VPCSICSNNPTC (referred to herein as RT32) (SEQ ID NO: 2), TCWAICKRIPNK ((referred to herein as RT33) (SEQ ID NO: 3), NKKPGKKTTTKP (referred to herein as RT34) (SEQ ID NO: 4), or combinations thereof.
  • isolated nucleic acid means a nucleic acid separated or substantially free from at least some of the other components of the naturally occurring organism, for example, the cell structural components commonly found associated with nucleic acids in a cellular environment and/or other nucleic acids.
  • the isolation of nucleic acids can therefore be accomplished by techniques such as cell lysis followed by phenol plus chloroform extraction, followed by ethanol precipitation of the nucleic acids.
  • the nucleic acids of this invention can be isolated from cells according to methods well known in the art for isolating nucleic acids.
  • the nucleic acids of the present invention can be synthesized according to standard protocols well described in the literature for synthesizing nucleic acids. Modifications to the nucleic acids of the invention are also contemplated, provided that the essential structure and function of the peptide or polypeptide encoded by the nucleic acid are maintained.
  • E. coli Escherichia coli
  • E. coli Escherichia coli
  • Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis, and other enterobacteria, such as Salmonella, Serratia, as well as various Pseudomonas species.
  • These prokaryotic hosts can support expression vectors which will typically contain expression control sequences compatible with the host cell (e.g., an origin of replication).
  • the nucleic acid sequences can be expressed in hosts after the sequences have been positioned to ensure the functioning of an expression control sequence.
  • These expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA.
  • expression vectors can contain selection markers, e.g., tetracycline resistance or hygromycin resistance, to permit detection and/or selection of those cells transformed with the desired nucleic acid sequences (50).
  • yeast expression system For eukaryotic system expression, a yeast expression system can be used. There are several advantages to yeast expression systems. First, evidence exists that polypeptides produced in a yeast expression system exhibit correct disulfide pairing. Second, post-translational glycosylation is efficiently carried out by yeast expression systems.
  • the Saccharomyces cerevisiae pre-pro-alpha-factor leader region (encoded by the MF ⁇ -1 gene) is routinely used to direct protein secretion from yeast (51).
  • the leader region of pre-pro-alpha-factor contains a signal peptide and a pro-segment which includes a recognition sequence for a yeast protease encoded by the KEX2 gene.
  • Hybridomas are subsequently screened for the ability to produce monoclonal antibodies that block RSV G glycoprotein binding to CX3CR1 and/or modulate biological activity associated with RSV G glycoprotein binding to CX3CR1.
  • Hybridomas producing antibodies are cloned, expanded and stored frozen for future production.
  • Polyclonal antibodies are prepared by screening human blood donors and selecting those that have high titer antibodies that block RSV G glycoprotein binding to CX3CR1 or block the activities induced by RSV G glycoprotein binding to CX3CR1 as well as antibodies that have other important biologic functions, e.g., neutralizing antibodies. Polyclonal antibodies can also be obtained by immunizing donors with vaccines that induce antibodies that block RSV G glycoprotein binding to CX3CR1 or block the activities induced by RSV G glycoprotein binding to CX3CR1, and/or have other important biologic functions, e.g., neutralizing antibodies. Serum from the selected donors is then pooled and made into immunoglobulin preparations.
  • Microencapsulation of the vaccine will also give a controlled release.
  • a number of factors contribute to the selection of a particular polymer for microencapsulation.
  • the reproducibility of polymer synthesis and the microencapsulation process, the cost of the microencapsulation materials and process, the toxicological profile, the requirements for variable release kinetics and the physicochemical compatibility of the polymer and the antigens are all factors that must be considered.
  • useful polymers are polycarbonates, polyesters, polyurethanes, polyorthoesters polyamides, poly (d,1-lactide-co-glycolide) (PLGA) and other biodegradable polymers.
  • a composition of this invention can be administered and can be in an adjuvant, at one to three week intervals for approximately 12 weeks or until an evaluation of the subject's clinical parameters (e.g., symptoms and RSV RNA levels indicate that the subject is not infected by RSV).
  • the treatment can be continued or resumed if the subject's clinical parameters indicate that HCV infection is present and can be maintained until the infection is no longer detected by these parameters.
  • Leukocyte chemotaxis toward Fkn, G glycoprotein or media was measured according to Boyden 46 using 24 well plates (Costar, Cambridge, Mass.) with 3 ⁇ m filter inserts (Nalge Nunc, Rochester, N.Y.) coated with an extracellular matrix consisting primarily of laminin, collagen IV and proteoglycan (ECM, Sigma, St. Louis, Mo.).
  • ECM proteoglycan
  • 5 ⁇ 10 5 na ⁇ ve spleen cells was placed in the upper chamber and 10 nM mouse Fkn or 10 nM G glycoprotein, anti-CX3CR1 antibody, anti-G or anti-F glycoprotein monoclonal antibody, normal rabbit sera, rabbit anti-CCR5 sera or media was placed in the upper or lower chamber as appropriate.
  • CX3CR1 The receptor for Fkn, CX3CR1
  • CX3CR1 was previously identified and characterized using anti-CX3CR1 rabbit antiserum and CX3CR1-transfected human embryonic kidney cells (293-CX3CR1 cells) 33 .
  • Flow cytometry using anti-CX3CR1 rabbit antiserum 33 indicated that the percent of detectable CX3CR1 on 293-CX3CR1 cells increased from 4-10% on untransfected 293 cells to 80-92% for 293-CX3CR1 cells.
  • G glycoprotein binding to 293-CX3CR1 cells in the presence of heparin was also further inhibited by peptide RT33 that contained the CX3C motif (from 78-84% to 85-94%) and by the addition of the two anti-CX3CR1 antibodies (from 78-84% to 88-94%).
  • peptide RT33 that contained the CX3C motif (from 78-84% to 85-94%) and by the addition of the two anti-CX3CR1 antibodies (from 78-84% to 88-94%).
  • a similar increase in inhibition of Fkn binding to 293-CX3CR1 cells in the presence of heparin was noted with the addition of G glycoprotein (from 57-65% to 80-85%) or peptide RT33 (from 57-65% to 70-80%) or the two anti-CX3CR1 antibodies (78-84% to 88-97%).
  • Radioligand inhibition studies 36 demonstrated that Fkn and G glycoprotein inhibited >90% of 125 -I Fkn polypeptide binding to 293-CX3CR1 cells at concentrations from 10 pM to 10 nM (FIG. 2).
  • naive BALB/c mice show that G and Fkn also recruit leukocytes in vivo (FIG. 5B and Table 6).
  • CX3CR1 is expressed on mouse and rat leukocytes, and by immunoprecipitation CX3CR1 was detected in na ⁇ ve mouse splenocytes, a mouse monocytic cell line (LADMAC), as well as in a human epithelial cell line (HUT-292, data not shown).
  • LADMAC mouse monocytic cell line
  • HUT-292 human epithelial cell line
  • the inhibition studies with reagents known to bind to CX3CR1 support the specific role of CX3CR1 in this interaction.
  • the inhibition studies with G glycoprotein peptides with or without the CX3C motif indicate that G glycoprotein interaction with CX3CR1 occurs, as expected, through the CX3C motif in the G glycoprotein.
  • the data presented herein also indicate that much of the G glycoprotein and RSV binding to cells occurs via HBD-GAG interaction, and that the remaining binding to cells occurs mostly via G glycoprotein binding to CX3CR1.
  • Virus and Infection The A2 strain of RSV (RSV/A2) was used in all experiments and propagated in Vero cells as previously described (54, 55). Mice were anesthetized by i.p. administration of Avertin (2,2,2-tribromoethanol, 0.2 ml/g body weight, Sigma-Aldrich, St. Louis, Mo.), and i.n. challenged with 106 PFU of RSV in Dulbecco's PBS (GIBCO Laboratories, Grand Island, N.Y.). No fewer than three mice per treatment were examined per time point in three separate experiments.
  • Avertin 2,2,2-tribromoethanol, 0.2 ml/g body weight, Sigma-Aldrich, St. Louis, Mo.
  • RT33 a 12-mer G glycoprotein peptide encompassing the G glycoprotein CX3C motif (underlined, T CWAIC KRIPNK (SEQ ID NO: 3)) in the amino acid sequence of the A2 strain of RSV (GENBANK, attachment protein locus 1912305, accession number M111486) (54), 2) RT34, a 12-mer G glycoprotein peptide C-terminal to the CX3C motif (NKKPGKKTTTKP) (SEQ ID NO: 4) (54), 3) Eyp1, a 13-mer G glycoprotein peptide variant of RT33 containing a ALA substitution for ILE in the CX3C motif (T CAAAC KRIPNKK) (SEQ ID NO: 5), 4) ⁇ 1, a 11-mer G glycoprotein peptide variant of RT33 with a deletion of ILE in the CX3C motif (T CWAC KRIPNK), (54), and
  • the peptides were synthesized using a simultaneous, multiple solid-phase peptide synthesis method on a peptide synthesizer (Perkin-Elmer Applied Biosystems, Berkeley, Calif.), and tested for homogeneity by reverse-phase liquid chromatography and capillary electrophoresis as previously described (56).
  • mice were i.p. treated (0.1 ml/mouse) with 100 ⁇ M of G glycoprotein peptides RT33 (CWAIC), RT34 (lacks the CX3C motif) or peptides with changes in the CX3C motif ( ⁇ +1, ⁇ 1, EYp1).
  • CWAIC G glycoprotein peptides RT33
  • RT34 lats the CX3C motif
  • peptides with changes in the CX3C motif ⁇ +1, ⁇ 1, EYp1
  • Control mice were treated with PBS containing a similar dilution of DMSO used to solubilize the peptides.
  • BAL cells were washed in Dulbecco's PBS (GIBCO) containing 1% bovine serum albumin (Sigma), blocked with 10% normal mouse sera diluted in PBS containing 1% bovine serum albumin, and then stained (4° C., 30 min) with an appropriate dilution of FITC-conjugated or PE-conjugated anti-CD3 ⁇ (145-2C11), anti-CD45R/B220 (RA3-6B2), anti-CD8 (Ly-2), anti-neutrophil (PMN) (RB6-8C5), anti-CD11b (M1/70) and isotype antibody controls (all from Pharmingen, San Diego, Calif.).
  • the cells were washed in Cytofix/Cytoperm buffer and stained (4° C., 30 min) with appropriate dilutions of anti-IL-2 (JES6-5H4), anti-IL-4 (BVD4-1D11), anti-IL-5 (TRFK5), IL-10 (JES3-16E3), anti-IFN ⁇ (XMG1.2), or anti-TNF ⁇ (MP6-XT22) antibodies (Pharmingen) diluted in Cytofix/Cytoperm. Extra- and intracellular staining was analyzed by flow cytometry using a FACScan and Cell Quest software (Becton Dickinson, San Diego, Calif.).
  • Virus titers in the lungs of RSV-infected mice were determined as previously described (55, 58). Briefly, lungs were aseptically removed from 3-5 mice per group at days 3, 5, 7 and 9 pi, and stored at ⁇ 70° C. until assay. Identical weights (0.1 g) of individual lung samples were homogenized in 1 ml of Dulbecco's PBS (GIBCO), and ten-fold serial dilutions of the lung homogenates were added to 80-90% confluent Vero cell monolayers.
  • RT33 or EYp1 had dramatically lower virus titers (0.1-0.4 ⁇ 10 4 pfu/g lung tissue).
  • day 14 pi no virus was detected in lungs of any treated mice.
  • peptide treatment was associated with lower virus titers at day 3 pi, compared to PBS-treated mice, suggesting that treatment may augment aspects of innate immunity.
  • B220 + , CD3 + , and CD11b + cells (FIG. 10) or NK or PMN cells (FIG. 11) was determined at days 3, 5, 7 and 14 pi.
  • B220 + (FIG. 10A) and CD11b + (FIG. 10C) cells were the predominate cell types in peptide-or PBS-treated mice, compared to CD3 + (T cell), DX5 + (NK cell) and RB6-8C5 + (PMN) cells.
  • Peptide EYp1 has a neutral CX3C motif (CAAAC) compared to peptide RT33 (CWAIC).
  • CAAAC neutral CX3C motif
  • CWAIC peptide RT33
  • Replacement of TRP-ALA-ILE in peptide RT33, with ALA-ALA-ALA in peptide EYp1 may enhance the avidity of CX3CR1 interaction, and trigger the induction of chemokines or other factors that induce pulmonary leukocyte chemotaxis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pulmonology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Steroid Compounds (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US10/420,387 2000-10-18 2003-04-18 Compositions and methods for modulating RSV infection and immunity Abandoned US20040009177A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US10/420,387 US20040009177A1 (en) 2000-10-18 2003-04-18 Compositions and methods for modulating RSV infection and immunity
US11/139,372 US8173131B2 (en) 2000-10-18 2005-05-26 Compositions and methods for modulating RSV infection and immunity
US13/358,320 US8778354B2 (en) 2000-10-18 2012-01-25 Compositions and methods for modulating RSV infection and immunity
US14/252,836 US20140288267A1 (en) 2000-10-18 2014-04-15 Compositions and methods for modulating rsv infection and immunity

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US24152100P 2000-10-18 2000-10-18
PCT/US2001/032459 WO2002032942A2 (en) 2000-10-18 2001-10-18 Compositions and methods for modulating rsv infection and immunity
US10/420,387 US20040009177A1 (en) 2000-10-18 2003-04-18 Compositions and methods for modulating RSV infection and immunity

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/032459 Continuation WO2002032942A2 (en) 2000-10-18 2001-10-18 Compositions and methods for modulating rsv infection and immunity

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/139,372 Division US8173131B2 (en) 2000-10-18 2005-05-26 Compositions and methods for modulating RSV infection and immunity

Publications (1)

Publication Number Publication Date
US20040009177A1 true US20040009177A1 (en) 2004-01-15

Family

ID=22911027

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/420,387 Abandoned US20040009177A1 (en) 2000-10-18 2003-04-18 Compositions and methods for modulating RSV infection and immunity
US11/139,372 Expired - Fee Related US8173131B2 (en) 2000-10-18 2005-05-26 Compositions and methods for modulating RSV infection and immunity
US13/358,320 Expired - Fee Related US8778354B2 (en) 2000-10-18 2012-01-25 Compositions and methods for modulating RSV infection and immunity
US14/252,836 Abandoned US20140288267A1 (en) 2000-10-18 2014-04-15 Compositions and methods for modulating rsv infection and immunity

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/139,372 Expired - Fee Related US8173131B2 (en) 2000-10-18 2005-05-26 Compositions and methods for modulating RSV infection and immunity
US13/358,320 Expired - Fee Related US8778354B2 (en) 2000-10-18 2012-01-25 Compositions and methods for modulating RSV infection and immunity
US14/252,836 Abandoned US20140288267A1 (en) 2000-10-18 2014-04-15 Compositions and methods for modulating rsv infection and immunity

Country Status (9)

Country Link
US (4) US20040009177A1 (de)
EP (1) EP1334119B2 (de)
AT (1) ATE412666T1 (de)
AU (2) AU2002224416A1 (de)
CA (1) CA2426312C (de)
DE (1) DE60136381D1 (de)
DK (1) DK1334119T4 (de)
ES (1) ES2316487T5 (de)
WO (1) WO2002032942A2 (de)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080226630A1 (en) * 2007-03-06 2008-09-18 Symphogen A/S Recombinant antibodies for treatment of respiratory syncytial virus infections
US20100040606A1 (en) * 2006-03-06 2010-02-18 Symphogen A/S Recombinant polyclonal antibody for treatment of respiratory syncytial virus infections
US20130122362A1 (en) * 2010-07-28 2013-05-16 Nissan Motor Co., Ltd. Bipolar electrode, bipolar secondary battery using the same and method for manufacturing bipolar electrode
WO2019075400A1 (en) * 2017-10-13 2019-04-18 Trellis Bioscience, Llc CONFORMATIONAL EPITOPES IN A CENTRAL PROTECTED REGION OF SYNCYTIAL RESPIRATORY VIRUS PROTEIN G

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006023029A2 (en) * 2004-06-16 2006-03-02 The Johns Hopkins University The cysteine-rich region of respiratory syncytial virus and methods of use therefor
CA2703667C (en) 2007-10-25 2015-12-29 Lawrence M. Kauvar Anti-rsv g protein antibodies
GB0724253D0 (en) * 2007-12-12 2008-01-30 Fermentas Uab Transfection reagent
ES2594102T3 (es) * 2008-04-18 2016-12-15 Vaxinnate Corporation Mutantes por deleción de la flagelina y métodos para su uso
US20110318376A1 (en) * 2008-12-24 2011-12-29 University Of Rochester Recombinant expression of self-folding neutralizing epitope-bearing subdomains of the respiratory syncytial virus attachment and fusion proteins
CA2770304C (en) 2009-08-04 2019-11-26 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services, Centers For Disease Control And Prevention Anti-rsv immunogens and methods of immunization
US9856456B2 (en) 2009-10-12 2018-01-02 Thermo Fisher Scientific Baltics Uab Delivery agent
GB0917792D0 (en) 2009-10-12 2009-11-25 Fermentas Uab Delivery agent
CN103037898B (zh) 2010-07-07 2016-06-29 人工细胞科技公司 呼吸道合胞病毒抗原组合物和方法
EP3656396A1 (de) 2012-08-01 2020-05-27 Bavarian Nordic A/S Rekombinanter modifizierter impfstoffvirus ankara (mva) gegen das respiratorische synzytialvirus (rsv)
EP2993009A1 (de) * 2014-09-08 2016-03-09 HILTI Aktiengesellschaft Verfahren zur Steuerung eines Wandsägesystems beim Erstellen eines Trennschnittes
SE539731C2 (sv) 2016-03-04 2017-11-14 Vertical Wind Ab Rotor för en elektrisk generator och förfarande för dess tillverkning
US11201095B1 (en) 2019-08-23 2021-12-14 Xilinx, Inc. Chip package having a cover with window
AU2021241450A1 (en) * 2020-03-23 2022-09-15 Atossa Therapeutics, Inc. Heparin and N-acetylcysteine for the treatment of a respiratory virus

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2718452B1 (fr) 1994-04-06 1996-06-28 Pf Medicament Elément d'immunogène, agent immunogène, composition pharmaceutique et procédé de préparation.
DE69509103T2 (de) 1994-08-25 1999-09-30 Pepscan Systems Bv Antigene peptide abgeleitet vom g-protein des rsv (respiratorisches synzytialvirus) für die typen- und subtypen spezifische diagnose einer rsv-infektionen
ATE389018T1 (de) 1996-01-24 2008-03-15 Schering Corp Cx3c chemokin-gene von säugetieren
AUPO026596A0 (en) 1996-06-05 1996-06-27 Biomolecular Research Institute Limited Viral peptide
US6020182A (en) * 1996-07-12 2000-02-01 Connaught Laboratories Limited Subunit respiratory syncytial virus vaccine preparation
WO1999001433A1 (fr) 1997-07-04 1999-01-14 Chisso Corporation Composes de type cristaux liquides dotes d'une valeur elevee d'anisotropie de permittivite, composition de cristaux liquides contenant ces composes et element d'affichage a cristaux liquides fabrique a partir d'une telle composition de cristaux liquides
ES2236946T3 (es) 1997-09-19 2005-07-16 Wyeth Holdings Corporation Peptidos derivados de la proteina de union (g) del virus respiratorio sincitial.
FR2827605B1 (fr) 2001-07-20 2004-07-16 Pf Medicament Nouveaux peptides derives de la proteine g du vrs et leur utilisation dans un vaccin

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100040606A1 (en) * 2006-03-06 2010-02-18 Symphogen A/S Recombinant polyclonal antibody for treatment of respiratory syncytial virus infections
US20080226630A1 (en) * 2007-03-06 2008-09-18 Symphogen A/S Recombinant antibodies for treatment of respiratory syncytial virus infections
US7879329B2 (en) 2007-03-06 2011-02-01 Symphogen A/S Recombinant antibodies for treatment of respiratory syncytial virus infections
US20110189171A1 (en) * 2007-03-06 2011-08-04 Symphogen A/S Recombinant antibodies for treatment of respiratory syncytial virus infections
US20130122362A1 (en) * 2010-07-28 2013-05-16 Nissan Motor Co., Ltd. Bipolar electrode, bipolar secondary battery using the same and method for manufacturing bipolar electrode
WO2019075400A1 (en) * 2017-10-13 2019-04-18 Trellis Bioscience, Llc CONFORMATIONAL EPITOPES IN A CENTRAL PROTECTED REGION OF SYNCYTIAL RESPIRATORY VIRUS PROTEIN G
JP2020536934A (ja) * 2017-10-13 2020-12-17 トレリス バイオサイエンス リミテッド ライアビリティー カンパニー 呼吸器合胞体ウイルスgタンパク質中央保存領域の立体構造エピトープ
US11773143B2 (en) 2017-10-13 2023-10-03 Trellis Bioscience, Llc Conformational epitopes in respiratory syncytial virus G protein central conserved region
JP7445895B2 (ja) 2017-10-13 2024-03-08 トレリス・バイオサイエンス・インコーポレーテッド 呼吸器合胞体ウイルスgタンパク質中央保存領域の立体構造エピトープ

Also Published As

Publication number Publication date
AU2002224416A1 (en) 2002-04-29
US8173131B2 (en) 2012-05-08
DK1334119T3 (da) 2009-02-23
ES2316487T5 (es) 2019-05-13
WO2002032942A3 (en) 2002-12-12
ATE412666T1 (de) 2008-11-15
EP1334119A2 (de) 2003-08-13
EP1334119B1 (de) 2008-10-29
DK1334119T4 (en) 2019-03-18
US20140288267A1 (en) 2014-09-25
US8778354B2 (en) 2014-07-15
EP1334119B2 (de) 2018-11-28
WO2002032942A2 (en) 2002-04-25
AU2009200997A1 (en) 2009-04-02
CA2426312A1 (en) 2002-04-25
US20120258111A1 (en) 2012-10-11
US20060018925A1 (en) 2006-01-26
AU2009200997B2 (en) 2011-12-22
CA2426312C (en) 2014-01-14
ES2316487T3 (es) 2009-04-16
DE60136381D1 (de) 2008-12-11

Similar Documents

Publication Publication Date Title
US8778354B2 (en) Compositions and methods for modulating RSV infection and immunity
Olmsted et al. Expression of the F glycoprotein of respiratory syncytial virus by a recombinant vaccinia virus: comparison of the individual contributions of the F and G glycoproteins to host immunity.
US6677127B1 (en) Nucleic acid respiratory syncytial virus vaccines
US20210106673A1 (en) Respiratory syncytial virus vaccine
CA2363593A1 (en) Nucleic acid respiratory syncytial virus vaccines
AU756222B2 (en) Nucleic acid vaccines encoding G protein of respiratory syncytial virus
US11464850B2 (en) Recombinant RSV antigens, nucleic acid molecules encoding the antigens, and vaccine compositions comprising the same
US6017897A (en) Nucleic acid respiratory syncytial virus vaccines
AU644470B2 (en) Hrsv vaccine
WO2005016247A2 (en) Dna sequences, peptides, antibodies and vaccines for prevention and treatment of sars
US20190290754A1 (en) Vaccine composition comprising hepatitis b virus-like particle as adjuvant
WO2022148374A1 (zh) 抗冠状病毒的全人广谱中和抗体76e1及其应用
EP3833677A1 (de) Verbesserte rsv-passive und -aktive impfstoffe
Stott et al. Immunity to respiratory syncytial virus infections
KR20140036610A (ko) 호흡기세포융합 바이러스에 대한 백신

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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