WO1994027636A1 - Adjuvants for vaccines against respiratory syncytial virus - Google Patents

Adjuvants for vaccines against respiratory syncytial virus Download PDF

Info

Publication number
WO1994027636A1
WO1994027636A1 PCT/US1994/005833 US9405833W WO9427636A1 WO 1994027636 A1 WO1994027636 A1 WO 1994027636A1 US 9405833 W US9405833 W US 9405833W WO 9427636 A1 WO9427636 A1 WO 9427636A1
Authority
WO
WIPO (PCT)
Prior art keywords
rsv
protein
virus
vaccine
vaccine formulation
Prior art date
Application number
PCT/US1994/005833
Other languages
French (fr)
Inventor
Gerald E. Hancock
Dan J. Speelman
Patrick J. Frenchick
Original Assignee
American Cyanamid Company
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=22078872&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1994027636(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to AT94918109T priority Critical patent/ATE196737T1/en
Priority to DE69426077T priority patent/DE69426077T3/en
Priority to DK94918109T priority patent/DK0705109T4/en
Priority to US08/553,332 priority patent/US5723130A/en
Priority to AU69571/94A priority patent/AU676340B2/en
Application filed by American Cyanamid Company filed Critical American Cyanamid Company
Priority to JP50089995A priority patent/JP3734263B2/en
Priority to CA002163550A priority patent/CA2163550A1/en
Priority to EP94918109A priority patent/EP0705109B2/en
Publication of WO1994027636A1 publication Critical patent/WO1994027636A1/en
Priority to FI955667A priority patent/FI955667A/en
Priority to NO954786A priority patent/NO954786L/en
Priority to GR20000402474T priority patent/GR3034785T3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55572Lipopolysaccharides; Lipid A; Monophosphoryl lipid A
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55577Saponins; Quil A; QS21; ISCOMS

Definitions

  • Respiratory Syncytial Virus is a major cause of lower respiratory disease in infancy and early childhood (Mclntosh and Chanock, 1985, in
  • RS virus is an enveloped RNA virus of the family ParamyxoViridae and of the genus pneumovirus.
  • the two major envelope proteins are the G protein, which is responsible for attachment of the virus to the host cell membrane, and the fusion protein (F protein), which is responsible for fusing the virus and cell membranes.
  • G protein which is responsible for attachment of the virus to the host cell membrane
  • F protein fusion protein
  • Virus-cell fusion is a necessary step for infection. Fusion protein is also required for cell-cell fusion which is another way to spread the infection from an infected cell to an uninfected cell.
  • Antibodies directed against the fusion protein or against the G protein can neutralize the virus. However, only antibodies to the fusion protein will block the spread of the virus between cells, i.e., have anti-fusion activity. Thus, antibodies to the fusion protein will protect against circulating virus as well as inhibit the spread, between cells, of an established infection. Antibodies to the fusion protein (both polyclonal antisera against purified fusion protein and monoclonal antibodies which contain both neutralizing and anti-fusion activity) have been found to be protective in animal models against infection (Walsh et al., 1984, Infect. Immun.
  • RS virus vaccine A practical means for protection of infants and young children against upper and lower respiratory disease would be protective vaccination against RS virus. Vaccination of expectant mothers (active immunization) would protect young children by passive transfer of immunity, either transplacentally, or through the mother's milk. Several approaches to an RS virus vaccine are possible, but some of them have proven unsuccessful in the past.
  • a useful vaccine to this virus must be effective in inducing production of antibodies to the fusion protein and/or the G protein.
  • protective immunity is the production of antibodies that inhibit fusion and therefore, can stop the spread of virus between cells in the respiratory tract. Additionally, it is helpful to induce a cell mediated immune response, including the stimulation of cytotoxic T cells
  • CTL's which are useful against RS virus infected cells.
  • the various vaccine formulations of the present invention are directed to meeting both these objectives.
  • This invention pertains to the discovery of certain adjuvants that are capable of increasing the immunological response to envelope proteins of
  • 3D-monophosphoryl lipid A (MPL) plus alum significantly increase the ability of antibodies raised against RSV glycoproteins F and/or G to
  • vaccine formulations can be made comprising envelope protein (s) of RSV and an adjuvant selected from QS-21, MPL, 3D-MPL and combinations.
  • the formulation may optionally contain alum.
  • the addition of alum can further boost the immunological response to the RSV antigen (s) when administered with these adjuvants.
  • the presence of these adjuvants provides enhanced immunogenicity to the antigen by augmentation of the immune response, in particular, complement mediated plaque reduction neutralization when compared to alum. Additionally, the presence of adjuvant allows a vaccine to be made with a reduced amount of antigen (s).
  • Figure 1 depicts the cell-mediated cytoxicity results from the experiments of Example 5, which is discussed herein.
  • the present invention pertains to novel vaccine formulations and therapeutic uses therefore for prevention of RSV infection.
  • the vaccine
  • formulation of the present invention comprises an RSV protein or an immunological fragment thereof and an adjuvant that has been shown to boost the
  • the adjuvant is selected from QS-21 and monophosphoryl lipid A and combinations thereof, and optionally alum.
  • the presence of alum in the vaccine acts
  • QS-21 is formulated with RSV envelope protein G and/or F.
  • QS-21 is a saponin which is purified from a crude
  • Antibodies raised against formulations comprising QS-21 and RSV protein F or RSV protein G and F can neutralize RS virus.
  • the immunogenicity of RSV F and G proteins is greatly increased using QS-21 as the adjuvant compared to formulations that are not adjuvanted or that contain other known adjuvants, such as alum when used solely as the adjuvant.
  • the adjuvants can be employed in a vaccine with RSV G protein or F protein to elicit an immune response, such as antibody response, which neutralizes both subgroup A and subgroup B of the RSV virus.
  • an immune response such as antibody response
  • MPL and specifically 3D-MPL can be used in combination with alum to produce a vaccine formulation that can enhance stimulation of complement dependent neutralizing antibodies to RSV.
  • the immunogencity of RSV subunit components is greatly increased with this adjuvant compared to formulations that are not adjuvanted or that contain alum as the sole adjuvant.
  • Proteins and polypeptides related to a neutralizing and/or fusion epitope(s) of the fusion protein and/or G protein of RS virus are useful as immunogens in a subunit vaccine to protect against lower respiratory disease and other disease symptoms of RS virus infection and can be formulated in the vaccines of the present invention.
  • Subunit vaccines comprise the relevant immunogenic material necessary to immunize a host and the adjuvants, identified herein as potent immunomodulators.
  • Vaccines prepared from genetically engineered immunogens, chemically synthesized immunogens and/or immunogens comprising authentic substantially pure RS virus fusion protein or fragments thereof alone or in combination with similarly prepared RS virus G protein or fragments thereof, which are capable of eliciting a protective immune response are particularly advantageous because there is no risk of infection of the recipients.
  • immunogenic fragment from both RSV glycoproteins F and G can also be used in vaccine formulations of this invention.
  • Such chimeric RSV polypeptides have been described by Wathen, U.S. Patent 5,194,595, the teachings of which are incorporated herein by
  • the RS virus fusion protein and/or G protein and polypeptides can be purified from recombinants that express the neutralizing and/or fusion epitopes.
  • recombinants include any bacterial transformants, yeast transformants, cultured insect cells infected with recombinant baculoviruses or cultured mammalian cells as known in the art, for example, such as
  • Chinese hamster ovary cells that express the RS virus fusion protein epitopes.
  • the recombinant protein or polypeptides can comprise multiple copies of the epitope of interest.
  • the RS virus fusion protein and/or G protein related protein or polypeptide can be chemically synthesized.
  • the RS virus fusion protein related protein or polypeptide or G related protein can be isolated in substantially pure form from RS virus or cultures of cells infected with RS virus and formulated with the novel adjuvants as a vaccine against RSV.
  • the RS virus fusion protein or G protein, related protein or polypeptide is adjusted to an appropriate
  • MPL and its derivative 3-deacylated MPL (3D-MPL) can be
  • 3D-MPL may be obtained according to the methods described in British Patent No. 2220211 (Ribi Immunochem.).
  • the amount of protein in each vaccine dose is selected as an amount which induces an
  • each dose will comprise from about 0.1 to about 100 ⁇ g of protein, with from about 5 to about 50 ⁇ g being preferred and from about 5 to about 25 ⁇ g/dose being alternatively preferred.
  • the amount of adjuvant will be an amount that will induce an immunomodulating response without
  • An optional amount for a particular vaccine can be ascertained by
  • the amount of adjuvant will be from about 1 to about 100 ⁇ g/dose, with from about 5 to about 50 ⁇ g/dose being preferred, and from about 20 to about 50 ⁇ g/dose being alternatively preferred.
  • Immunopotency of vaccines containing the RS virus fusion or G protein or immunologic fragments thereof and genetic or physical mixtures thereof can be determined by monitoring the immune response of test animals following immunization with the purified protein, synthetic peptide or recombinant protein.
  • Test animals may include but are not limited to mice, rats, rabbits, primates, and eventually human subjects.
  • Methods of introduction of the immunogen may include intradermal, intramuscular,
  • the immune response of the test subjects can be analyzed by multiple approaches: (a) the reactivity of the resultant immune serum to authentic RS viral antigens, as assayed by known techniques, e.g., enzyme linked immunosorbant assay (ELISA), immunoglots, radio-immunoprecipitations, etc, (b) the ability of the immune serum to neutralize RS virus infectivity in vitro, (c) the ability of the immune serum to inhibit virus fusion in vitro, the ability of immunized animals to generate antigen dependent cytotoxic T lymphocyte (CTL) activity and (e) protection from RS virus infection.
  • ELISA enzyme linked immunosorbant assay
  • CTL antigen dependent cytotoxic T lymphocyte
  • the secretory IgA antibodies produced by the mucosal associated lymphoid tissue may play a major role in protection against RS virus infection by preventing the initial interaction of the pathogens with the mucosal surface, or by neutralizing the important epitopes of the pathogens that are involved in infection/or spreading of the disease. Stimulation of mucosal immune responses, including production of secretory IgA antibodies may be of major importance in conferring protection against lower and upper respiratory tract infection.
  • polypeptides and proteins may generally be formulated at concentrations in the range of from about 0.1 ⁇ g to about 100 ⁇ g per dose.
  • Physiologically acceptable media may be used as carriers. These include, but are not limited to:
  • Suitable adjuvants may be added to the novel vaccine formulations of this invention and include, mineral gels, e.g., aluminum hydroxide, aluminum phosphate, etc.
  • the immunogen may also be incorporated into liposomes or conjugated to
  • polysaccharides and/or other polymers for use in a vaccine formulation.
  • polypeptides and proteins that can be incorporated into vaccine formulations of the present invention may be linked to a soluble macromolecular carrier.
  • the carrier and the polypeptides and proteins are in excess of five thousand daltons after linking, and more preferably, the carrier is in excess of five kilodaltons.
  • the carrier is a polyamino acid, either natural or synthetic, which is immunogenic in animals, including humans.
  • the manner of linking is conventional. Many linking techniques are disclosed in U.S. Pat. No. 4,629,783 which is incorporated herein by reference. Many cross-linking agents are disclosed in 1986-87 Handbook and General Catalog, Pierce Chemical Company,
  • Recombinant viruses are prepared that express RS virus fusion protein and/or G protein related epitopes. These viruses can be used to prepare inactivated recombinant viral vaccines to protect against lower respiratory infections and other disease symptoms of RS virus.
  • Inactivated vaccines are "dead” in the sense that their infectivity has been destroyed, usually by chemical treatment (e.g., formaldehyde). Ideally, the infectivity of the virus is destroyed without affecting the proteins which are related to
  • polypeptides must be grown in culture to provide the necessary quantity of relevant antigens.
  • a mixture of inactivated viruses which express different epitopes may be used for the formulation of "multivalent” vaccines.
  • these "multivalent" inactivated vaccines may be preferable to live vaccine formulation because of potential difficulties which mutual interference of live viruses administered together.
  • the inactivated recombinant virus or mixture of viruses can be formulated with the adjuvant of this invention in order to enhance the immunological response to the antigens.
  • the vaccines of this invention can be administered to an individual to prevent an infection or disease symptoms associated with RSV. Such administration can be accomplished by a single dose or by multiple doses for eliciting a primary immune response in the individual. Typically multiple vaccination will be given three times at essentially two month intervals for humans. Booster doses may be given to stimulate an existingimmune response from previous vaccination or natural infection.
  • PFP-1 is prepared by the procedure of Walsh et al. J. Gen. Virol. 66:409-415 (1985) with the following modifications.
  • the immunoaffinity eluted material is passed over a DEAE column and the flow through is collected, dialised against PBS/0.1% Triton X-100 and sterile filtered through a 0.2 ⁇ m filter.
  • Ion exchange fusion protein-2 (IF)
  • IF is prepared by passing clarified RSV- infected cell lkysate over a DEAE column. The flow through is collected and passed over a hydroxy-apatite (HA) column. Following HA elution, the eluted F protein is dialysed against PBS/0.1% Triton X-100, and sterile filtered through a 0.2 ⁇ m filter.
  • HA hydroxy-apatite
  • G. protein is prepared by the procedure of Walsh et al. J. Gen. Virol. 66: 761-767 (1984) with the following modifications. Following elution, the G protein is passed over an immunoaffinity column specific for RSV F protein. The flow through is collected, dialysed against PSB/0.1% Triton X-100, and sterile filtered through a 0.2 ⁇ m filter.
  • F/G protein chimeric is prepared by U.S. Patent No. 5,194,595 and provided by Upjohn
  • EIA Enzyme Immunoassay
  • RS virus fusion protein is diluted to 200 ng/ml in carbonate-bicarbonate buffer, pH 9.6.
  • One hundred ⁇ l of the diluted antigen is added to each well of rows B-G of a flat-bottomed, 96 well NUNCTM assay plate.
  • rows A and H 100 ⁇ l of carbonatebicarbonate buffer alone is added to each well. The plate is covered and incubated for 2 hours at 37°C with shaking and stored overnight at 4°C to immobilize the antigen.
  • Each sample is first diluted to a primary dilution in 0.2% Tween, 0.01 M EDTA/PBS pH 7.5 (0.2% TWN) .
  • the primary dilutions are further serially diluted as follows in a 96 well U-bottomed FALCONTM plate :
  • Sample 1 is inoculated in triplicate, e.g., in wells A2, B2 and C2 ; Sample 2 in duplicate e.g., in wells D2 , E2 ; Sample 3 in triplicate e.g., in wells F2, G2 an H2.
  • One hundred ⁇ l of the primary dilutions are transferred from each well of the FALCONTM plate to the corresponding well in the NUNCTM plate, e.g., A2
  • NUNCTM FALCONTM to A2 (NUNCTM) .
  • the NUNCTM assay plate is covered and incubated for one hour at 37°C with shaking The supernatants are removed from the assay plate, and the plate is washed with 0.1% Tween/PBS and pat dried.
  • Goat anti-Mouse IgG alkaline phosphatase conjugate (TAGOTM) is diluted with 0.3% Tween/PBS pH 7.0 (0.3%TWN) to a working dilution, e.g., 1:1500.
  • the diluted conjugate (100 ⁇ l) is added to each well in rows 2-12.
  • the plate is covered and incubated for 1 hour at 37°C with shaking. The inocula is then removed, and the plate is washed with 0.1% Tween/PBS pH 7.4 and pat dried.
  • Rows A and H serve as negative controls because no antigen is present; row 1 also serves as a negative control because no antibodies are present.
  • Example 3 Virus Neutralization Assay (Plaque Reduction Neutralization test, PRNT)
  • Test serum samples which are serially diluted and the positive control serum are heat inactivated at 56°C for 30 min. All sera are then diluted with an equal volume containing about 30 plaque forming units (PFU) of RS virus, and incubated at 37°C for one hour, with (C plus PRNT) or without (PRNT) the addition of 5% rabbit complement.
  • PFU plaque forming units
  • Each incubated serum-virus mixture is inoculated to HEp-2 cells (ATCC No. CCL23) in separate wells of 96 well plates and virus absorption is allowed to take place for 2 hours at 37°C.
  • the inocula are removed.
  • the cell monolayers are washed and overlayed with modified Eagle's medium plus 5% fetal bovine serum and 1% SEPHADEX ® , and incubated at 37°C for 3 days.
  • the overlay medium is removed and the cells are washed with phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the BLOTTO is removed. 50 ⁇ l per well of monoclonal antibodies against RS virus (previously titered and diluted with BLOTTO to a working
  • BLOTTO BLOTTO is added and the plate is incubated for 1 hour at 37 ⁇ C.
  • the goat antibodies are removed, and the fixed cells are again washed twice with BLOTTO, 30 minutes each time.
  • Seronegative Swiss Webster mice were immunized (100 ⁇ l) with F and G proteins in various adjuvants on week ) and week 3.
  • mice were bled for serology on weeks 0, 3 and 6.
  • EIA-Ga Ga protein specific enzyme immunoassay
  • PRNT plaque reduction neutralization test
  • a subgroup A strain of RSSV i.e. A2
  • a subgroup B strain of RSV i.e. 18537
  • C'-PRNT plaque reduction neutralization test with 5% complement
  • EIA assays were performed on individual sera and the geometric neon titers (GMT) were calculated and are reported.
  • PRNT and C'-PRNT assays were performed on pooled sera (one pool per group, n-5).
  • Immunogens IF- ion exchange purified RSV F protein; G-affinity purified RSV G protein; PFP-1- affinity purified RSV F protein; F/G chimeric-F/G chimeric protein purified from baculovirus- infected Sf9 culture.
  • Immunogens were administered with the following adjuvants: Alum - 1 ⁇ g/ml aluminum hydroxide, OS-21 - 200 ⁇ g/ml OS-21, 3D-MPL- 250 ⁇ g/ml 3D-MPL, 3D-MPL + alum- a combination of 250 ⁇ /ml 3D-MPL plus 1 ⁇ g/ml aluminum hydroxide.
  • Seronegative Balb/C mice were immunized (100 ⁇ l) on weeks 0 and 4 with 0.5 ⁇ g doses of the various immunogens.
  • Alum Aluminum hydroxide, 1mg/ml.
  • 3D-MPL 3D-Monophospholipid A, 250 ⁇ g/ml (25 ⁇ g/dose).
  • OS21 200 ⁇ g/ ⁇ l (20 ⁇ g/dose).
  • Seronegative Swiss Webster mice were immunized (100 ⁇ l) with ion exchange-purified RSV F protein (IF) or PBS in various adjuvants on week 0 and week 3.
  • IF ion exchange-purified RSV F protein
  • mice were bled for serology on weeks 0, 3 and 6.
  • EIA-Ga Ga protein specific enzyme immunoassay
  • PRNT plaque reduction neutralization test
  • a subgroup A strain of BSV i.e. A2
  • a subgroup B strain of RSV i.e. 18537
  • C'-PRNT plaque reduction neutralization test with 5% complement
  • EIA assays were performed on individual sera and the geometric mean titers (GMT) were calculated and are reported.
  • PRNT and C'PRNT assays were performed on pooled sera (one pool per group, n-5).
  • Seronegative mice were immunized (100 ⁇ L, IM) with 1 ⁇ g protein adjuvanted with either alum (AL (OB) 3 , lmg/ml> or OS-21
  • mice were bled for serology at weeks 0, 3, 6.
  • the A2 and 18537 strains of RSV are used and virus stocks are grown in either Vero
  • DMEM Modified Eagle's medium
  • Anti-F protein antibody subclass determination The titer of anti-F protein antibody subclass of mice primed with 5 ⁇ g of F protein mixed with QS-21, ALOH or natural infection are determined by ELISA.
  • 96 well plates are prepared with the 20 ng F protein or 5 ug RSV A2 as follows. Purifed F protein
  • carbonate/bicarbonate buffer pH 9.6 is coated onto 96-well plates (Nunc, Roskilde, Denmark) for 2h at 37°C and stored overnight at 4°C. Thereafter, the plates are washed 5 times with PBS/0.05% Tween 20 (Sigma) followed by 2 additional rinses with PBS alone.
  • IgGl (1:3000, Zymed), or IgG2a (1:5000, Zymed) is added and the plates are incubated lh at room temperature.
  • strepavidin conjugated to horseradish peroxidase (1:10,000 dilution in PBS/0.3% Tween 20, Zymed) is added to the wells and incubated at room temperature for an additional 30 minutes.
  • Peroxidase substrate (2,2'-azino-di[3-ethyl-benzthiazoline sulfonate (6)], Kirkegaard and Perry Laboratories) was added to the wells after washing and incubated at room temperature for 20 minutes at which time the reaction is stopped with 100 ul of 1% sodium dodecal sulphate (Pierce, Rockford, IL). End point titers are determined at 410nM.
  • Virus Neutralization Assay PRNT is performed as in Example 3.
  • neutralizing antibody is performed in duplicate on HEp-2 cell monolayers in 96-well tissue culture plates as described in Example 3.
  • an adjuvant can enable RSV protein to elicit a complement dependent IgG antibody response which neutralizes both subgroup A and subgroup B viruses (these subgroups being indentified as A2 and 18537, respectively in the table below).
  • This cross neutralizing immune response of heterologous subtype RS virus has not been acheived before using purified G protein alone.
  • a vaccine formulated with QS-21 adjuvant and RS virus G protein generates a desirable heterotypic neutralizing antibody response which is substantially greater than that which is elicted by alum alone or a natural infection.
  • Seronegative BALB/c mice are immunized (0.1ml, I.M.) at weeks 0 and 3 with RSV G protein at the doses indicated above. Animals are challenged (0.1ml, I.N.) with 6 log 10 PFU of RSV strain A2 at week 6 and bled for serology at 4 days post-challenge.
  • Adjuvants QS-21 (200 ⁇ g/ml) or aluminum hydroxide (alum, 1mg/mlm are mixed with RSV G protein or PBS 24 hours prior to use.
  • Example 7 Comparison of QS-21 vs. ALOH for ability to elicit local F protein-dependent killer cell activity
  • PMC Pulmonary Mononuclear Cells
  • the PMC are isolated from the lungs following
  • the fragments are passed through a 100 mesh stainless steel tissue culture seive (Sigma).
  • Antigen-dependent cellular cytotoxicity is determined in a 4h 51 Cr (Amersham Corp., Arlington Heights, IL) release assay. Briefly, 50 ul (5000 cells) of syngeneic 51 Cr-labeled control or RSV-infected (BCH4) target cell lines are incubated (37°C, 5% CO 2 ) in triplicate V-bottom micro-wells (Costar, Cambridge, MA) with 100 ul of spleen or pulmonary mononuclear cells (serially 2-fold diluted in RPMI 1640 containing 10% heat-inactivated FBS, V/V). The final volume is 150 ul per well. After incubation, the supernatants are
  • H2K d clone SFl-1.1, IgG 2a
  • H-2D d clone AF4-62.4, IgG 2b
  • H-2K b clone AF6-88.5, IgG 2a
  • a monoclonal antibody (E37-10, IgG 2b) directed against diphtheria toxoid antigen serves as subclass control.
  • the monoclonal antibody directed against murine CD8 surface molecules (53-6.72, ATCC No. TIB 105) is purified from hybridoma culture supernatants over a recombinant protein G column
  • mice are vaccinated at weeks 0 and 3 with 5 ⁇ g of F protein mixed with either 20 ⁇ g QS-21 ( ) or 100 ⁇ g ALOH ( ⁇ ) and compared to mice immunized by experimental infection ( ⁇ ). Two weeks after secondary immunization, the mice are challenged with virus.

Abstract

Vaccine formulations and therapeutic methods therefor containing respiratory syncytial viral proteins or immunological fragment and an adjuvant selected from the group consisting of QS-21, 3-deacylated monophosphoryl lipid A, monophosphoryl lipid A and combination are described herein. Presence of the adjuvant(s) significantly increases the humoral and cell-mediated immunogenicity of the RSV proteins.

Description

ADJUVANTS FOR VACCINES AGAINST
RESPIRATORY SYNCYTIAL VIRUS
Background of the Invention
Respiratory Syncytial Virus (RSV) is a major cause of lower respiratory disease in infancy and early childhood (Mclntosh and Chanock, 1985, in
Virology, Fields, B. (ed), Raven, NY, pp. 1285-1304). In all geographical areas, it is the major cause of bronchiolitis and pneumonia in infants and young children. The agent reinfects frequently during childhood, but illness produced by reinfection is generally milder than that associated with the initial infection and rarely causes major problems.
RS virus is an enveloped RNA virus of the family ParamyxoViridae and of the genus pneumovirus. The two major envelope proteins are the G protein, which is responsible for attachment of the virus to the host cell membrane, and the fusion protein (F protein), which is responsible for fusing the virus and cell membranes. Virus-cell fusion is a necessary step for infection. Fusion protein is also required for cell-cell fusion which is another way to spread the infection from an infected cell to an uninfected cell.
Antibodies directed against the fusion protein or against the G protein can neutralize the virus. However, only antibodies to the fusion protein will block the spread of the virus between cells, i.e., have anti-fusion activity. Thus, antibodies to the fusion protein will protect against circulating virus as well as inhibit the spread, between cells, of an established infection. Antibodies to the fusion protein (both polyclonal antisera against purified fusion protein and monoclonal antibodies which contain both neutralizing and anti-fusion activity) have been found to be protective in animal models against infection (Walsh et al., 1984, Infect. Immun.
43:756-758).
A practical means for protection of infants and young children against upper and lower respiratory disease would be protective vaccination against RS virus. Vaccination of expectant mothers (active immunization) would protect young children by passive transfer of immunity, either transplacentally, or through the mother's milk. Several approaches to an RS virus vaccine are possible, but some of them have proven unsuccessful in the past.
Vaccination with killed RS virus vaccine has been tried and found to be ineffective (Kim et al., 1969, Am. J. Epid. 89:422). Not only were children not protected, but in some cases, subsequent
infections with RS virus resulted in atypical and more severe disease than in the unimmunized controls. This phenomenon is not unique to RS virus and has been seen also in killed paramyxovirus vaccines such as measles. It has been suggested that the reason for the failure of the past inactivated RS virus vaccine was due to inactivation of the biologically functional epitopes on either or both of the viral envelope qlycoproteins. That is to say, the neutralizing and fusion epitopes on the killed virus vaccine were "denatured". As a result, the vaccinated subject did not experience the biologically functional neutralizing and fusion epitopes. Therefore, when the vaccinated subject encountered a live virus, the resultant antibody response did not yield protective immunity. Instead, there was an antibody mediated inflammatory response which often resulted in a more severe disease (Choppin and Scheid, 1980, Rev. Inf. Dis. 2:40-61).
The second approach to an RS virus vaccine has been to attenuate live virus. Temperature sensitive mutants (Wright et al., 1982, Infect. Immun. 37:397-400) and passage attenuated virus
(Belshe et al., 1982, J. Inf. Dis. 145:311-319) have proven to be poorly infectious and not efficacious in the prevention of disease when used as immunogens in RS virus vaccines. However, in these cases, there was no atypical disease as a result of vaccination.
Based on our current knowledge of the structure of RS virus and the immune response to infection, it is clear that a useful vaccine to this virus must be effective in inducing production of antibodies to the fusion protein and/or the G protein. Of particular importance to protective immunity is the production of antibodies that inhibit fusion and therefore, can stop the spread of virus between cells in the respiratory tract. Additionally, it is helpful to induce a cell mediated immune response, including the stimulation of cytotoxic T cells
(CTL's) which are useful against RS virus infected cells. The various vaccine formulations of the present invention are directed to meeting both these objectives.
Summary of the Invention
This invention pertains to the discovery of certain adjuvants that are capable of increasing the immunological response to envelope proteins of
respiratory syncytial virus, specifically to RSV glycoprotein F and RSV glycoprotein G. In particular, it is shown herein that the adjuvant QS-21, or
alternatively, 3D-monophosphoryl lipid A (MPL) plus alum, significantly increase the ability of antibodies raised against RSV glycoproteins F and/or G to
neutralize the virus as well as provide immunological protection via cell-mediated response against the virus. Additionally, these adjuvants have been shown to prevent syncytia formation in virally infected cells. Based on these findings, vaccine formulations can be made comprising envelope protein (s) of RSV and an adjuvant selected from QS-21, MPL, 3D-MPL and combinations. The formulation may optionally contain alum. The addition of alum can further boost the immunological response to the RSV antigen (s) when administered with these adjuvants. The presence of these adjuvants provides enhanced immunogenicity to the antigen by augmentation of the immune response, in particular, complement mediated plaque reduction neutralization when compared to alum. Additionally, the presence of adjuvant allows a vaccine to be made with a reduced amount of antigen (s).
Brief Description of the Drawings
Figure 1. depicts the cell-mediated cytoxicity results from the experiments of Example 5, which is discussed herein.
Detailed Description of the Invention
The present invention pertains to novel vaccine formulations and therapeutic uses therefore for prevention of RSV infection. The vaccine
formulation of the present invention comprises an RSV protein or an immunological fragment thereof and an adjuvant that has been shown to boost the
immunological response to the RSV protein. The adjuvant is selected from QS-21 and monophosphoryl lipid A and combinations thereof, and optionally alum. The presence of alum in the vaccine acts
synergistically with MPL to elicit a neutralization response to RSV.
In one embodiment of the invention, QS-21 is formulated with RSV envelope protein G and/or F.
QS-21 is a saponin which is purified from a crude
Quillaja saponaria extract and has been described by Kensil and Marciani, U.S. Patent 5,057,540.
Antibodies raised against formulations comprising QS-21 and RSV protein F or RSV protein G and F can neutralize RS virus. The immunogenicity of RSV F and G proteins is greatly increased using QS-21 as the adjuvant compared to formulations that are not adjuvanted or that contain other known adjuvants, such as alum when used solely as the adjuvant.
Another aspect of the present invention is that the adjuvants can be employed in a vaccine with RSV G protein or F protein to elicit an immune response, such as antibody response, which neutralizes both subgroup A and subgroup B of the RSV virus. This is a significant discovery since other adjuvants, specifically alum, with G protein have been found to neutralize only the subgroup from which the proteinO is purified.
In another embodiment, MPL and specifically 3D-MPL can be used in combination with alum to produce a vaccine formulation that can enhance stimulation of complement dependent neutralizing antibodies to RSV. The immunogencity of RSV subunit components is greatly increased with this adjuvant compared to formulations that are not adjuvanted or that contain alum as the sole adjuvant.
Proteins and polypeptides related to a neutralizing and/or fusion epitope(s) of the fusion protein and/or G protein of RS virus are useful as immunogens in a subunit vaccine to protect against lower respiratory disease and other disease symptoms of RS virus infection and can be formulated in the vaccines of the present invention. Subunit vaccines comprise the relevant immunogenic material necessary to immunize a host and the adjuvants, identified herein as potent immunomodulators. Vaccines prepared from genetically engineered immunogens, chemically synthesized immunogens and/or immunogens comprising authentic substantially pure RS virus fusion protein or fragments thereof alone or in combination with similarly prepared RS virus G protein or fragments thereof, which are capable of eliciting a protective immune response are particularly advantageous because there is no risk of infection of the recipients.
Chimeric polypeptides comprising at least one
immunogenic fragment from both RSV glycoproteins F and G can also be used in vaccine formulations of this invention. Such chimeric RSV polypeptides have been described by Wathen, U.S. Patent 5,194,595, the teachings of which are incorporated herein by
reference.
The RS virus fusion protein and/or G protein and polypeptides can be purified from recombinants that express the neutralizing and/or fusion epitopes. Such recombinants include any bacterial transformants, yeast transformants, cultured insect cells infected with recombinant baculoviruses or cultured mammalian cells as known in the art, for example, such as
Chinese hamster ovary cells that express the RS virus fusion protein epitopes. The recombinant protein or polypeptides can comprise multiple copies of the epitope of interest.
The RS virus fusion protein and/or G protein related protein or polypeptide can be chemically synthesized. Alternatively, the RS virus fusion protein related protein or polypeptide or G related protein can be isolated in substantially pure form from RS virus or cultures of cells infected with RS virus and formulated with the novel adjuvants as a vaccine against RSV.
Regardless of the method of production, the RS virus fusion protein or G protein, related protein or polypeptide is adjusted to an appropriate
concentration and can be formulated with an adjuvant selected from QS-21 or MPL plus alum. MPL and its derivative 3-deacylated MPL (3D-MPL) can be
co-formulated with TDM and squalene and used in vaccine formulations of the present invention. 3D-MPL may be obtained according to the methods described in British Patent No. 2220211 (Ribi Immunochem.).
The amount of protein in each vaccine dose is selected as an amount which induces an
immunoprotective response without significant adverse side effects. Such amount will vary depending upon the immunogen used. Generally each dose will comprise from about 0.1 to about 100 μg of protein, with from about 5 to about 50 μg being preferred and from about 5 to about 25 μg/dose being alternatively preferred. The amount of adjuvant will be an amount that will induce an immunomodulating response without
significant adverse side effect. An optional amount for a particular vaccine can be ascertained by
standard studies involving observation of a vaccine's antibody titers and their virus neutralization
capabilities. The amount of adjuvant will be from about 1 to about 100 μg/dose, with from about 5 to about 50 μg/dose being preferred, and from about 20 to about 50 μg/dose being alternatively preferred.
Immunopotency of vaccines containing the RS virus fusion or G protein or immunologic fragments thereof and genetic or physical mixtures thereof can be determined by monitoring the immune response of test animals following immunization with the purified protein, synthetic peptide or recombinant protein. Test animals may include but are not limited to mice, rats, rabbits, primates, and eventually human subjects. Methods of introduction of the immunogen may include intradermal, intramuscular,
intraperitoneal, intravenous, subcutaneous, intranasal or any other standard routes of immunizations. The immune response of the test subjects can be analyzed by multiple approaches: (a) the reactivity of the resultant immune serum to authentic RS viral antigens, as assayed by known techniques, e.g., enzyme linked immunosorbant assay (ELISA), immunoglots, radio-immunoprecipitations, etc, (b) the ability of the immune serum to neutralize RS virus infectivity in vitro, (c) the ability of the immune serum to inhibit virus fusion in vitro, the ability of immunized animals to generate antigen dependent cytotoxic T lymphocyte (CTL) activity and (e) protection from RS virus infection.
Many methods may be used to administer the vaccine formulations described herein to humans for prophylatic purposes. These include, but are not limited to: intradermal, intramuscular,
intraperitoneal, intravenous, subcutaneous and
intranasal routes. The secretory IgA antibodies produced by the mucosal associated lymphoid tissue may play a major role in protection against RS virus infection by preventing the initial interaction of the pathogens with the mucosal surface, or by neutralizing the important epitopes of the pathogens that are involved in infection/or spreading of the disease. Stimulation of mucosal immune responses, including production of secretory IgA antibodies may be of major importance in conferring protection against lower and upper respiratory tract infection.
The polypeptides and proteins may generally be formulated at concentrations in the range of from about 0.1 μg to about 100 μg per dose. Physiologically acceptable media may be used as carriers. These include, but are not limited to:
sterile water, saline, phosphate buffered saline and the like. Other suitable adjuvants may be added to the novel vaccine formulations of this invention and include, mineral gels, e.g., aluminum hydroxide, aluminum phosphate, etc. The immunogen may also be incorporated into liposomes or conjugated to
polysaccharides and/or other polymers for use in a vaccine formulation.
The polypeptides and proteins that can be incorporated into vaccine formulations of the present invention may be linked to a soluble macromolecular carrier. Preferably, the carrier and the polypeptides and proteins are in excess of five thousand daltons after linking, and more preferably, the carrier is in excess of five kilodaltons. Preferably, the carrier is a polyamino acid, either natural or synthetic, which is immunogenic in animals, including humans. The manner of linking is conventional. Many linking techniques are disclosed in U.S. Pat. No. 4,629,783 which is incorporated herein by reference. Many cross-linking agents are disclosed in 1986-87 Handbook and General Catalog, Pierce Chemical Company,
(Rockford, Illinois) pages 311-340.
Recombinant viruses are prepared that express RS virus fusion protein and/or G protein related epitopes. These viruses can be used to prepare inactivated recombinant viral vaccines to protect against lower respiratory infections and other disease symptoms of RS virus.
Inactivated vaccines are "dead" in the sense that their infectivity has been destroyed, usually by chemical treatment (e.g., formaldehyde). Ideally, the infectivity of the virus is destroyed without affecting the proteins which are related to
immunogenicity of the virus. In order to prepare inactivated vaccines, large quantities of the
recombinant virus expressing the RS virus fusion protein and/or G protein, related proteins or
polypeptides must be grown in culture to provide the necessary quantity of relevant antigens. A mixture of inactivated viruses which express different epitopes may be used for the formulation of "multivalent" vaccines. In certain instances, these "multivalent" inactivated vaccines may be preferable to live vaccine formulation because of potential difficulties which mutual interference of live viruses administered together. In either case, the inactivated recombinant virus or mixture of viruses can be formulated with the adjuvant of this invention in order to enhance the immunological response to the antigens.
The vaccines of this invention can be administered to an individual to prevent an infection or disease symptoms associated with RSV. Such administration can be accomplished by a single dose or by multiple doses for eliciting a primary immune response in the individual. Typically multiple vaccination will be given three times at essentially two month intervals for humans. Booster doses may be given to stimulate an existingimmune response from previous vaccination or natural infection.
The following Examples are offered for the purpose of illustrating the present invention and are not to be construed to limit the scope of the present invention.
Exemplification
Example 1: RSV Protein Preparation
A. Immunoaffinity fusion protein-1 (PFP-1)
PFP-1 is prepared by the procedure of Walsh et al. J. Gen. Virol. 66:409-415 (1985) with the following modifications. The immunoaffinity eluted material is passed over a DEAE column and the flow through is collected, dialised against PBS/0.1% Triton X-100 and sterile filtered through a 0.2 μm filter.
B. Ion exchange fusion protein-2 (IF)
IF is prepared by passing clarified RSV- infected cell lkysate over a DEAE column. The flow through is collected and passed over a hydroxy-apatite (HA) column. Following HA elution, the eluted F protein is dialysed against PBS/0.1% Triton X-100, and sterile filtered through a 0.2 μm filter.
C. Immunoaffinity G protein (G)
G. protein is prepared by the procedure of Walsh et al. J. Gen. Virol. 66: 761-767 (1984) with the following modifications. Following elution, the G protein is passed over an immunoaffinity column specific for RSV F protein. The flow through is collected, dialysed against PSB/0.1% Triton X-100, and sterile filtered through a 0.2 μm filter.
D. F/G Protein Chimeric
F/G protein chimeric is prepared by U.S. Patent No. 5,194,595 and provided by Upjohn
Corporation.
Example 2: Enzyme Immunoasay (EIA)
Antibody titer in serum samples is determined using an Enzyme Immunoassay (EIA) performed as follows:
RS virus fusion protein is diluted to 200 ng/ml in carbonate-bicarbonate buffer, pH 9.6. One hundred μl of the diluted antigen is added to each well of rows B-G of a flat-bottomed, 96 well NUNC™ assay plate. In rows A and H, 100 μl of carbonatebicarbonate buffer alone is added to each well. The plate is covered and incubated for 2 hours at 37°C with shaking and stored overnight at 4°C to immobilize the antigen.
The supernatants are removed from the NUNC™ assay plate and the plate is washed with 0.1%
Tween/PBS pH 7.4 and pat dried.
Three antibody samples are assayed on each plate. Each sample is first diluted to a primary dilution in 0.2% Tween, 0.01 M EDTA/PBS pH 7.5 (0.2% TWN) . The primary dilutions are further serially diluted as follows in a 96 well U-bottomed FALCON™ plate :
(a) The primary ilutions of the samples are inoculated into row 2 at 200 μl/well. Sample 1 is inoculated in triplicate, e.g., in wells A2, B2 and C2 ; Sample 2 in duplicate e.g., in wells D2 , E2 ; Sample 3 in triplicate e.g., in wells F2, G2 an H2.
(b) 100 μl of 0.2% TWN were inoculated into each well of rows 3-12.
(c) Serial dilutions were created by
transferring sequentially 100 μl from a well in row2 to the corresponding well in row 3 (e.g., B2 to B3 ; C2 to C3), a well in row 3 to the corresponding well in row 4, until row 12 was reached.
(d) To row 1, 100 μl of 0.2% TWN was added to each well as control.
One hundred μl of the primary dilutions are transferred from each well of the FALCON™ plate to the corresponding well in the NUNC™ plate, e.g., A2
(FALCON™) to A2 (NUNC™) . The NUNC™ assay plate is covered and incubated for one hour at 37°C with shaking The supernatants are removed from the assay plate, and the plate is washed with 0.1% Tween/PBS and pat dried.
Goat anti-Mouse IgG alkaline phosphatase conjugate (TAGO™) is diluted with 0.3% Tween/PBS pH 7.0 (0.3%TWN) to a working dilution, e.g., 1:1500. The diluted conjugate (100 μl) is added to each well in rows 2-12. The row 1, 100 μl of 0.3% TWN are added to each well as control. The plate is covered and incubated for 1 hour at 37°C with shaking. The inocula is then removed, and the plate is washed with 0.1% Tween/PBS pH 7.4 and pat dried.
To each and every well, 100 μl substrate solution, 1 mg/ml in diethanolamine buffer pH 9.8 (SIGMA-104™) are added. The enzymatic reaction is allowed to take place at room temperature for 1 hours. The reaction is stopped by adding 100 μl of 3N NaOH to each well. The extent of enzymatic reaction is determined by reading the optical density of 410 nm.
Rows A and H serve as negative controls because no antigen is present; row 1 also serves as a negative control because no antibodies are present. Example 3: Virus Neutralization Assay (Plaque Reduction Neutralization test, PRNT)
Test serum samples which are serially diluted and the positive control serum are heat inactivated at 56°C for 30 min. All sera are then diluted with an equal volume containing about 30 plaque forming units (PFU) of RS virus, and incubated at 37°C for one hour, with (C plus PRNT) or without (PRNT) the addition of 5% rabbit complement. A pool of human adult sera which had previously been
characterized by enzyme immunoassay, neutralization and antifusion assays is used for positive control. Sera which had previously been characterized and was known to be non-immune is used as negative control.
Each incubated serum-virus mixture is inoculated to HEp-2 cells (ATCC No. CCL23) in separate wells of 96 well plates and virus absorption is allowed to take place for 2 hours at 37°C. The inocula are removed. The cell monolayers are washed and overlayed with modified Eagle's medium plus 5% fetal bovine serum and 1% SEPHADEX®, and incubated at 37°C for 3 days. The overlay medium is removed and the cells are washed with phosphate buffered saline (PBS).
200 μl of chilled PBS-methanol (1:5)
solution is added to each well, and the cells are fixed for 30 min. at room temperature. The PBS-methanol fixative is removed, an 200 μl per well of 5% CARNATION® instant milk in PBS, pH 6.8 (BLOTTO) is added. The plate is incubated for 30 minutes at 37°C.
The BLOTTO is removed. 50 μl per well of monoclonal antibodies against RS virus (previously titered and diluted with BLOTTO to a working
concentration) is added, and the plate is incubated at 37°C for 1 hour. The antibodies ae removed, and the fixed cells are washed twice with BLOTTO, 30 minutes each time.
50 μl/well of horseradish peroxidase
conjugated goat anti-mouse IgG (diluted 1:250 in
BLOTTO) is added and the plate is incubated for 1 hour at 37μC. The goat antibodies are removed, and the fixed cells are again washed twice with BLOTTO, 30 minutes each time.
50 μl/well of a peroxidase substrate
solution (0.05% 4-chloro-1-naphthol, 0.09% H2O2 in PBS pH 6.8) is added, and color is allowed to develop for 15-30 minutes at room temperature. The substrate solution is removed, and the wells are washed with water and air dried. The number of plaques in each well is determined. The neutralization ability of a test serum sample is expressed as the dilution which results in a 60% reduction in plaque formation when compared to non-immune control serum. The results are tabulated in Tables 1-4.
Data in Tables 1, 2, 3 and 4 representing the EIA and Plaque Reduction Neutralization Test results show the improvement in the biological immune response with the use of these novel adjuvants when compared to alum alone. The vaccine formulations of RS virus fusion protein, G protein, mixtures thereof ad F/G Chimeric protein with the novel adjuvants (with or without additional alum) were ignificantly enhanced when compared to formulations containing alum alone.
Figure imgf000018_0001
Figure imgf000019_0001
1 Seronegative Swiss Webster mice were immunized (100μl) with F and G proteins in various adjuvants on week ) and week 3.
Mice were bled for serology on weeks 0, 3 and 6.
2 Serological assays: EIA-F (F protein specific enzyme
immunoassay), EIA-Ga (Ga protein specific enzyme immunoassay), PRNT (plaque reduction neutralization test) against a subgroup A strain of RSSV (i.e. A2) and a subgroup B strain of RSV (i.e. 18537). C'-PRNT (plaque reduction neutralization test with 5% complement) were also performed against RSV strains A2 and 18537. EIA assays were performed on individual sera and the geometric neon titers (GMT) were calculated and are reported. PRNT and C'-PRNT assays were performed on pooled sera (one pool per group, n-5).
3 Immunogens: IF- ion exchange purified RSV F protein; G-affinity purified RSV G protein; PFP-1- affinity purified RSV F protein; F/G chimeric-F/G chimeric protein purified from baculovirus- infected Sf9 culture.
4 Immunogens were administered with the following adjuvants: Alum - 1μg/ml aluminum hydroxide, OS-21 - 200μg/ml OS-21, 3D-MPL- 250μg/ml 3D-MPL, 3D-MPL + alum- a combination of 250μ/ml 3D-MPL plus 1μg/ml aluminum hydroxide.
5 nd- not done.
Figure imgf000021_0001
1 Seronegative Balb/C mice were immunized (100μl) on weeks 0 and 4 with 0.5 μg doses of the various immunogens.
Mice were bled for serology at weeks 0, 4 and 8. 2 Serological assays: EIA-F (F protein specific enzyme
immunoassay), EIA-GA (GA protein specific enzyme immunoassay), PRNT (Plaque reduction neutralization test) against a subgroup A strain of RSV (i.e. A2) and a subgroup B strain of RSV (i.e. 18537). All assays were performed on pooled sera (1 pool/group, n-5. Additionally, the week 8 pools were tested by Complement enhanced PRNT by the addition of 5% rabbit complement. 3 Alum: Aluminum hydroxide, 1mg/ml. 4 3D-MPL: 3D-Monophospholipid A, 250μg/ml (25μg/dose). 5 OS21: 200μg/μl (20μg/dose). 6 Alum + 30-MPL: mixture of lmg/ml alumiδnum hydroxide and
250μg/ml 3D-MPL.
Figure imgf000023_0001
1 Seronegative Swiss Webster mice were immunized (100μl) with ion exchange-purified RSV F protein (IF) or PBS in various adjuvants on week 0 and week 3.
Mice were bled for serology on weeks 0, 3 and 6.
2 Serological assays: EIA-F (F protein specif8c enzyme
immunoassay), EIA-Ga (Ga protein specific enzyme immunoassay), PRNT (plaque reduction neutralization test) against a subgroup A strain of BSV (i.e. A2) and a subgroup B strain of RSV (i.e. 18537). C'-PRNT (plaque reduction neutralization test with 5% complement) were also performed against RSV strains A2 and 18537. EIA assays were performed on individual sera and the geometric mean titers (GMT) were calculated and are reported. PRNT and C'PRNT assays were performed on pooled sera (one pool per group, n-5).
3 Ion exchange-purified F protein (IF) was administered with the following adjuvants: alum=1mg/ml aluminum hydroxide, OS-21 = 200μg/ml OS-21, 3D-MPL = 250μg/ml 3D-MPL, 3D-MPL + alum = a mixture of 250μg/ml 3D-MPL plus lmg/ml aluminum hydroxide.
4. ND = Not done.
Figure imgf000025_0001
1 Seronegative mice were immunized (100μL, IM) with 1μg protein adjuvanted with either alum (AL (OB)3, lmg/ml> or OS-21
(250μg/ml) at weeks 0 and 3.
Mice were bled for serology at weeks 0, 3, 6.
2 Serological assays: EIA-F (F protein specific enzyme
immunoassay), PRNT (plaque reduction neutralization test) for subgroup A strain of RSV (i.e. A2) and a subgroup B strain of RSV (i.e 18537) with the addition of 5% rabbit complement. EIA assays were performed on individual sera and the geometric mean titers (GMT) were calculated and are reported (#mice/group =5).
3 Ion-exchange purified F protein (triton X-100 lysis) combined with an equal quantity of affinity purified G protein (triton X- 100/deoxycholate lysis).
4 Ion-exchange purified F protein.
Example 4
Virus and Cell Lines. The A2 and 18537 strains of RSV are used and virus stocks are grown in either Vero
[American Type Culture Collection (ATCC) No. CCL 81] or HEp-2 (ATCC No. CCL 23) cells following standard procedures, purified over sorbitol density gradients and stored at -70°C until use. A BCH4 cell line persistently infected with the Long strain of RSV and the uninfected BALB/c cell line (for both cell lines see Fernie et al., Proc. Soc. Exp. Biol. Med., 1981, 167:83-86) are a gift of Dr. Bruce F. Fernie. The latter cell lines are maintained in Dulbecco's
Modified Eagle's medium (DMEM, Gibco BRL,
Gaithersburg, MD) with 10% (V/V) heat-inactivated FBS
(Hyclone Laboratories Inc., Logan, UT).
Example 5
Anti-F protein antibody subclass determination. The titer of anti-F protein antibody subclass of mice primed with 5 μg of F protein mixed with QS-21, ALOH or natural infection are determined by ELISA.
Briefly, 96 well plates are prepared with the 20 ng F protein or 5 ug RSV A2 as follows. Purifed F protein
(200 ng/ml) or RSV A2 (50μg/ml) in
carbonate/bicarbonate buffer (pH 9.6) is coated onto 96-well plates (Nunc, Roskilde, Denmark) for 2h at 37°C and stored overnight at 4°C. Thereafter, the plates are washed 5 times with PBS/0.05% Tween 20 (Sigma) followed by 2 additional rinses with PBS alone.
Serial 3 -fold dilutions of serum prepared in PBS/0.3% Tween 20/0.01M EDTA buffer (pH 7.0) are then added to the wells and incubated for lh at room temperature. After washing 5 times with PBS/0.1% Tween 20, 100 ul of biotinylated goat anti-mouse IgG (1:4000,
Kirkegaard and Perry Laboratories), IgGl (1:3000, Zymed), or IgG2a (1:5000, Zymed) is added and the plates are incubated lh at room temperature.
Following another series of washes, 100ul of
strepavidin conjugated to horseradish peroxidase (1:10,000 dilution in PBS/0.3% Tween 20, Zymed) is added to the wells and incubated at room temperature for an additional 30 minutes. Peroxidase substrate (2,2'-azino-di[3-ethyl-benzthiazoline sulfonate (6)], Kirkegaard and Perry Laboratories) was added to the wells after washing and incubated at room temperature for 20 minutes at which time the reaction is stopped with 100 ul of 1% sodium dodecal sulphate (Pierce, Rockford, IL). End point titers are determined at 410nM.
Virus Neutralization Assay (PRNT) is performed as in Example 3.
The heightened complement-assisted serum neutralizing antibody titers elicited by F/QS-21 correlated with the induction of anti-F protein antibodies of the IgG2a subclass (Table 5). Three weeks after primary immunization, there is a QS-21 dose-related increase in protein-specific IgG2a as well as IgGl antibodies. In comparison, a single injection of F protein mixed in saline alone or F/ALOH elicits primarily protein-specific antibodies of the IgGl subclass (Table 5). The data indicates that F/QS-21 induces humoral immune responses that are similar to those generated by experimental infection and consist of both complement-fixing IgG2a as well as IgGl antibodies.
Example 6
Determination of Cross Neutralizing Antibody Titers and RSV Infectivity. The titration of serum
neutralizing antibody is performed in duplicate on HEp-2 cell monolayers in 96-well tissue culture plates as described in Example 3.
In this example, as shown in the table below, it is observed that an adjuvant can enable RSV protein to elicit a complement dependent IgG antibody response which neutralizes both subgroup A and subgroup B viruses (these subgroups being indentified as A2 and 18537, respectively in the table below). This cross neutralizing immune response of heterologous subtype RS virus has not been acheived before using purified G protein alone. A vaccine formulated with QS-21 adjuvant and RS virus G protein generates a desirable heterotypic neutralizing antibody response which is substantially greater than that which is elicted by alum alone or a natural infection.
Table 6: Heterotypic Neutralizing Antibody Response Elicited by RSV G Protein When Adjuvanted with QS-211
PRNT2
Immunogen (μg)3 Adiuvant4 A2 18537
G protein (2.5) QS-21 7940 6039
G protein (1.2) QS-21 >10,240 5154
G protein (0.6) QS-21 2092 719
G protein (0.3) QS-21 308 1906
Controls:
G protein (2.5) alum 212 <10
G protein (2.5) none <10 <10
PBS QS-21 <10 <10
- - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -
1 Seronegative BALB/c mice are immunized (0.1ml, I.M.) at weeks 0 and 3 with RSV G protein at the doses indicated above. Animals are challenged (0.1ml, I.N.) with 6 log10 PFU of RSV strain A2 at week 6 and bled for serology at 4 days post-challenge.
2 PRNT = plaque reduction neutralization test
performed against a subgroup A (A2) and a subgroup B
(18537) strain of RSV. Assays are performed on pooled sera (n=5) in the presence of 5% rabbit complement.
3 Immunogen: RSV G protein is immunoaffinity
purified from Vero cell lysates infected with RSV strain A2. This purified protein is further processed using immunoaffinity chromatography to reduce the level of residual F protein. No detectable F proteinspecific antibodies (as measured by EIA) are elicited by these immunogens.
4 Adjuvants: QS-21 (200μg/ml) or aluminum hydroxide (alum, 1mg/mlm are mixed with RSV G protein or PBS 24 hours prior to use. Example 7: Comparison of QS-21 vs. ALOH for ability to elicit local F protein-dependent killer cell activity
The ability of QS-21 to elicit local F protein-dependent killer cell activity is also
examined and compared to the cell-mediated
cytotoxicity generated by immunization with F/ALOH or experimental infection.
The Isolation of Pulmonary Mononuclear Cells (PMC).
The PMC are isolated from the lungs following
collagenase digestion (see Hancock et. al., Vaccine, 12:267-274, 1994 and Anderson et. al., J. Gen. Virol., 71:1561-1570, 1990). Briefly, excised lungs are placed in cold DMEM and rinsed free of peripheral blood. The lungs are then minced in fresh DMEM, transferred to a 50 ml centrifuge tube and nutated at 37°C in the presence of collagenase (collagenase type IV, Sigma Chemical Co., St. Louis, MO) at a final concentration of 2 mg/ml, 10 mM HEPES buffer, and 1% (V/V) heat-inactivated FBS. After 90 minutes
incubation, the fragments are passed through a 100 mesh stainless steel tissue culture seive (Sigma).
The resulting suspension is pelleted (400g),
resuspendend in metrizamide (16%, W/V, Accurate
Chemical & Scientific Corp., Westbury, NY), overlayed with RPMI 1640 (Gibco BRL) containing 10% heat-inactivated FBS, and spun (150g) for 20 minutes at 5°C. The PMC layers are then collected, washed free of gradient, and tested ex vivo for their cytolytic capacity.
Determination of Percent Cytotoxicity. Antigen-dependent cellular cytotoxicity is determined in a 4h 51Cr (Amersham Corp., Arlington Heights, IL) release assay. Briefly, 50 ul (5000 cells) of syngeneic51Cr-labeled control or RSV-infected (BCH4) target cell lines are incubated (37°C, 5% CO2) in triplicate V-bottom micro-wells (Costar, Cambridge, MA) with 100 ul of spleen or pulmonary mononuclear cells (serially 2-fold diluted in RPMI 1640 containing 10% heat-inactivated FBS, V/V). The final volume is 150 ul per well. After incubation, the supernatants are
collected (Skatron Harvester, Skatron Inc., Sterling, VA), measured for 51Cr release in a ClinGamma counter (Pharmacia LKB), and compared with spontanteous release (targets incubated with medium alone, 20-25%) and total release (targets incubated in culture medium with 1.0% Triton X-100, V/V in PBS). Percent specific release is calculated by: 100 X [(mean cpm
experimental) - (mean cpm spontaneous release)] /
[(mean cpm total release) - (mean cpm spontaneous release)].
Antibody Blocking Studies. Purified monoclonal antibodies directed against major histocompatibility complex (MHC) antigens H2Kd (clone SFl-1.1, IgG 2a), H-2Dd (clone AF4-62.4, IgG 2b), and H-2Kb (clone AF6-88.5, IgG 2a) are purchased from PharMingen, San
Diego, CA. A monoclonal antibody (E37-10, IgG 2b) directed against diphtheria toxoid antigen serves as subclass control. The monoclonal antibody directed against murine CD8 surface molecules (53-6.72, ATCC No. TIB 105) is purified from hybridoma culture supernatants over a recombinant protein G column
(Pharmacia). Purified rat IgG is purchased from
Calbiochem (San Diego, CA). To block cell-mediated cytolysis, 50 ul antibody is added to 50 ul effector cells prior to the addition of 50 ul of target cells. The final effector to target ratio was 60:1.
Balb/c mice are vaccinated at weeks 0 and 3 with 5μg of F protein mixed with either 20μg QS-21 ( ) or 100μg ALOH (Δ) and compared to mice immunized by experimental infection (●). Two weeks after secondary immunization, the mice are challenged with virus.
Four days after challenge, the PMC from BALB/c mice vaccinated with F/QS-21 are able to kill RSV-infected targets (solid lines in Figure) in an antigen-dependent manner (see Figure 1A). Most noteworthy, this cytotoxic activity is as potent as that of the PMC from mice previously infected with RSV and nearly 3-fold greater than the activity induced in the PMC of mice vaccinated with F/ALOH. Control syngeneic targets (dashed lines) not infected with RSV are not killed (Figure 1A). The activity is local because the spleen cells from the same mice are not cytolytic.
The results further suggested that the local killer cell activity induced by the F/QS-21 vaccine is mediated by T cells of the CD8 phenotype. Cytolysis was inhibited when increasing doses of monoclonal antibody directed against cells bearing CD8 surface determinants (filled symbol) are added to the assay mixture (Figure 1B). Likewise, increasing
concentrations of anti-H2Dd and H2Kd monoclonal antibodies (filled symbol) block cytolysis (Figure 1C). Control immunoglobulin (open symbols) is not inhibitory (Figure 1 B&C).

Claims

WE CLAIM :
1. A vaccine formulation comprising a respiratory syncytial viral (RSV) protein or an immunological fragment thereof and an adjuvant selected from the group consisting of QS-21, monophosphoryl lipid A, 3-deacylated monophosphoryl lipid A and combination thereof , in a physiologically acceptable vehicle.
2. A vaccine formulation comprising a respiratory syncytial viral (RSV) protein or an immunological fragment thereof and QS-21, in a physiologically acceptable vehicle.
3. The vaccine formulation of Claim 1 further comprising alum.
4. The vaccine formulation of Claim 2 further comprising alum.
5. A vaccine formulation comprising a respiratory syncytial viral (RSV) protein or an immunological fragment thereof, alum and 3-deacylated monophosphoryl A, in a physiologically acceptable vehicle.
6. A vaccine formulation of Claim 1 wherein the RSV protein is selected from the group consisting of RSV glycoprotein G, RSV glycoprotein F, a chimeric polypeptide comprising at least one immunogenic fragment from both RSV glycoproteins F and G, and combinations thereof.
7. The vaccine formulation of Claim 2 wherein the RSV protein is selected from the group consisting of RSV glycoprotein G, RSV glycoprotein F, a chimeric polypeptide comprising at least one immunogenic fragment from both RSV glycoproteins F and G, and combinations thereof_.
8. The vaccine formulation of Claim 3 wherein the RSV protein is selected from the group consisting of RSV glycoprotein G, RSV glycoprotein F, a chimeric
polypeptide comprising at least one immunogenic fragment from both RSV glycoprotein F and G,and combinations thereof.
9. The vaccine formulation of Claim 4 wherein the RSV protein is selected from the group consisting of RSV glycoprotein G, RSV glycoprotein F, a chimeric
polypeptide comprising at least one immunogenic fragment from both RSV glycoproteins F and G, and combinations thereof.
10. The vaccine formulation of Claim 5 wherein the RSV protein is selected from the group consisting of RSV glycoprotein G, RSV glycoprotein F, a chimeric polypeptide comprising at least one immunogenic fragment from both RSV glycoproteins F and G, and combinations thereof .
11. A method of preventing an infection or disease symptoms associated with respiratory syncytial virus in an individual comprising administering an effective amount of the vaccine of Claim 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
PCT/US1994/005833 1993-05-25 1994-05-24 Adjuvants for vaccines against respiratory syncytial virus WO1994027636A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
EP94918109A EP0705109B2 (en) 1993-05-25 1994-05-24 Adjuvants for vaccines against respiratory syncytial virus
DE69426077T DE69426077T3 (en) 1993-05-25 1994-05-24 ADJUVANTS FOR VACCINE AGAINST THE RESPIRATORY SYNCITIAL VIRUS
DK94918109T DK0705109T4 (en) 1993-05-25 1994-05-24 Adjuvants for vaccines against respiratory syncytial virus
US08/553,332 US5723130A (en) 1993-05-25 1994-05-24 Adjuvants for vaccines against respiratory syncytial virus
AU69571/94A AU676340B2 (en) 1993-05-25 1994-05-24 Adjuvants for vaccines against respiratory syncytial virus
AT94918109T ATE196737T1 (en) 1993-05-25 1994-05-24 ADJUVANTS FOR VACCINES AGAINST RESPIRATORY SYNCITIAL VIRUS
JP50089995A JP3734263B2 (en) 1993-05-25 1994-05-24 Adjuvants for vaccines against respiratory syncytial virus
CA002163550A CA2163550A1 (en) 1993-05-25 1994-05-24 Adjuvants for vaccines against respiratory syncytial virus
FI955667A FI955667A (en) 1993-05-25 1995-11-24 Adjuvants for vaccine against RS virus
NO954786A NO954786L (en) 1993-05-25 1995-11-24 Excipients for respiratory syncytal virus vaccines
GR20000402474T GR3034785T3 (en) 1993-05-25 2000-11-08 Adjuvants for vaccines against respiratory syncytial virus

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US6785593A 1993-05-25 1993-05-25
US08/067,855 1993-05-25

Publications (1)

Publication Number Publication Date
WO1994027636A1 true WO1994027636A1 (en) 1994-12-08

Family

ID=22078872

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1994/005833 WO1994027636A1 (en) 1993-05-25 1994-05-24 Adjuvants for vaccines against respiratory syncytial virus

Country Status (15)

Country Link
US (1) US5723130A (en)
EP (1) EP0705109B2 (en)
JP (1) JP3734263B2 (en)
AT (1) ATE196737T1 (en)
AU (1) AU676340B2 (en)
CA (1) CA2163550A1 (en)
DE (1) DE69426077T3 (en)
DK (1) DK0705109T4 (en)
ES (1) ES2150493T5 (en)
FI (1) FI955667A (en)
GR (1) GR3034785T3 (en)
NO (1) NO954786L (en)
PT (1) PT705109E (en)
RU (1) RU2160119C2 (en)
WO (1) WO1994027636A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996011019A1 (en) * 1994-10-05 1996-04-18 Vanderbilt University Interleukin-12 as an adjuvant for paramyxoviridae vaccines
WO1996033739A1 (en) * 1995-04-25 1996-10-31 Smithkline Beecham Biologicals S.A. Vaccines containing a saponin and a sterol
WO1998002179A1 (en) * 1996-07-12 1998-01-22 Connaught Laboratories Limited Two-step immunization procedure against the pyramyxoviridae family of viruses using recombinant virus and subunit protein preparation
WO1998002457A1 (en) * 1996-07-12 1998-01-22 Connaught Laboratories Limited Subunit respiratory syncytial virus vaccine preparation
WO1998002180A1 (en) * 1996-07-12 1998-01-22 Connaught Laboratories Limited Two-step immunization procedure against the pyramyxoviridae family of viruses using attenuated viral strains and subunit protein preparation
US6146632A (en) * 1993-12-23 2000-11-14 Smithkline Beecham Biologicals S.A. Vaccines
WO2001051082A1 (en) * 2000-01-14 2001-07-19 Allergy Therapeutics Limited Composition of antigen and glycolipid adjuvant sublingual administration
WO2003022878A2 (en) * 2001-09-13 2003-03-20 Aventis Pasteur Limited Subunit respiratory syncytial virus vaccine preparation
US7718178B2 (en) 1997-04-05 2010-05-18 Allergy Therapeutics Limited Allergen formulation
US7815920B2 (en) 1998-09-21 2010-10-19 Allergy Therapeutics (UK) Ltd Method of preparing an antigen-containing formulation
US8628784B2 (en) * 1998-10-16 2014-01-14 Glaxosmithkline Biologicals S.A. Adjuvant systems and vaccines
US10059757B2 (en) 2007-06-01 2018-08-28 Medimmune Limited RSV-specific binding molecules and means for producing them

Families Citing this family (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9105992D0 (en) * 1991-03-21 1991-05-08 Smithkline Beecham Biolog Vaccine
US6727230B1 (en) * 1994-03-25 2004-04-27 Coley Pharmaceutical Group, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US6488934B1 (en) * 1995-02-25 2002-12-03 Smithkline Beecham Biologicals S.A. Hepatitis B vaccine
US6975708B1 (en) * 1996-04-17 2005-12-13 Convergys Cmg Utah, Inc. Call processing system with call screening
US5867562A (en) * 1996-04-17 1999-02-02 Scherer; Gordon F. Call processing system with call screening
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6699478B1 (en) * 1997-09-19 2004-03-02 Wyeth Holdings Corporation Enhanced immune response to attachment (G) protein of Respiratory Syncytial Virus
US7790856B2 (en) * 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US20080050367A1 (en) * 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
TWI239847B (en) * 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US6787523B1 (en) * 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US20050059802A1 (en) * 1998-04-07 2005-03-17 Neuralab Ltd Prevention and treatment of amyloidogenic disease
JP2002513773A (en) * 1998-05-07 2002-05-14 コリクサ コーポレイション Adjuvant composition and use thereof
US7198920B1 (en) * 1999-01-29 2007-04-03 Corika Corporation HER-2/neu fusion proteins
GB9909077D0 (en) * 1999-04-20 1999-06-16 Smithkline Beecham Biolog Novel compositions
ATE419006T1 (en) * 1999-03-26 2009-01-15 Us Army MULTIVALENT DENGUE VIRUS VACCINE
CA2368673A1 (en) * 1999-03-26 2000-10-05 Walter Reed Army Institute Of Research Attenuated dengue-4 virus vaccine
US6511667B1 (en) * 1999-03-26 2003-01-28 The United States Of America As Represented By The Secretary Of The Army Attenuated dengue-2 virus vaccine
KR100863367B1 (en) * 1999-05-13 2008-10-13 와이어쓰 홀딩스 코포레이션 Adjuvant combination formulations
CN1437481A (en) * 2000-06-22 2003-08-20 美国氰胺公司 QS-21 and IL-12 as an adjuvant combination
US7229623B1 (en) * 2000-08-03 2007-06-12 Corixa Corporation Her-2/neu fusion proteins
PE20020530A1 (en) * 2000-11-10 2002-06-18 Wyeth Corp ADJUVANT COMBINATION FORMULATIONS
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
DE10205373B4 (en) * 2002-02-09 2007-07-19 Aloys Wobben Fire protection
MY139983A (en) * 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
ZA200505782B (en) * 2003-02-01 2006-09-27 Neuralab Ltd Active immunization to generate antibodies to soluble A-beta
US20060095001A1 (en) * 2004-10-29 2006-05-04 Transcutaneous Technologies Inc. Electrode and iontophoresis device
PE20061329A1 (en) * 2004-12-15 2006-12-08 Neuralab Ltd HUMANIZED AB ANTIBODIES TO IMPROVE COGNITION
JP2006346368A (en) * 2005-06-20 2006-12-28 Transcutaneous Technologies Inc Iontophoresis apparatus and manufacturing method
JP2007000342A (en) * 2005-06-23 2007-01-11 Transcutaneous Technologies Inc Iontophoresis device for controlling quantity and time of dosing a plurality of medicaments
US8295922B2 (en) 2005-08-08 2012-10-23 Tti Ellebeau, Inc. Iontophoresis device
US8386030B2 (en) * 2005-08-08 2013-02-26 Tti Ellebeau, Inc. Iontophoresis device
US20070088332A1 (en) * 2005-08-22 2007-04-19 Transcutaneous Technologies Inc. Iontophoresis device
JPWO2007023907A1 (en) * 2005-08-24 2009-02-26 Tti・エルビュー株式会社 Electrode structure for frozen iontophoresis
US20070048362A1 (en) * 2005-08-29 2007-03-01 Transcutaneous Technologies Inc. General purpose electrolyte solution composition for iontophoresis
JPWO2007029611A1 (en) * 2005-09-06 2009-03-19 Tti・エルビュー株式会社 Iontophoresis device
US20070112294A1 (en) * 2005-09-14 2007-05-17 Transcutaneous Technologies Inc. Iontophoresis device
BRPI0616165A2 (en) * 2005-09-15 2011-06-07 Tti Ellebeau Inc rod type iontophoresis device
WO2007032423A1 (en) * 2005-09-16 2007-03-22 Tti Ellebeau, Inc. Catheter type iontophoresis apparatus
US20090299264A1 (en) * 2005-09-28 2009-12-03 Tti Ellebeau, Inc. Electrode Assembly for Dry Type Iontophoresis
US20090187134A1 (en) * 2005-09-30 2009-07-23 Hidero Akiyama Iontophoresis Device Controlling Amounts of a Sleep-Inducing Agent and a Stimulant to be Administered and Time at Which the Drugs are Administered
JP2009509677A (en) * 2005-09-30 2009-03-12 Tti・エルビュー株式会社 Iontophoretic delivery of vesicle encapsulated active substances
US20070093787A1 (en) * 2005-09-30 2007-04-26 Transcutaneous Technologies Inc. Iontophoresis device to deliver multiple active agents to biological interfaces
EP1928539A1 (en) * 2005-09-30 2008-06-11 Tti Ellebeau, Inc. Functionalized microneedles transdermal drug delivery systems, devices, and methods
WO2007041115A1 (en) * 2005-09-30 2007-04-12 Tti Ellebeau Inc. Method and system to detect malfunctions in an iontophoresis device that delivers active agents to biological interfaces
EP1931417A2 (en) * 2005-09-30 2008-06-18 Transcutaneous Technologies Inc. Transdermal drug delivery systems, devices, and methods employing novel pharmaceutical vehicles
US20070135754A1 (en) * 2005-09-30 2007-06-14 Hidero Akiyama Electrode assembly for iontophoresis for administering active agent enclosed in nanoparticle and iontophoresis device using the same
US20090299265A1 (en) * 2005-09-30 2009-12-03 Tti Ellebeau, Inc. Electrode Assembly for Iontophoresis Having Shape-Memory Separator and Iontophoresis Device Using the Same
KR20080066712A (en) * 2005-09-30 2008-07-16 티티아이 엘뷰 가부시키가이샤 Functionalized microneedles transdermal drug delivery systems, devices, and methods
US20070197955A1 (en) * 2005-10-12 2007-08-23 Transcutaneous Technologies Inc. Mucous membrane adhesion-type iontophoresis device
US20080033338A1 (en) * 2005-12-28 2008-02-07 Smith Gregory A Electroosmotic pump apparatus and method to deliver active agents to biological interfaces
WO2007079190A2 (en) * 2005-12-29 2007-07-12 Tti Ellebeau, Inc. Device and method for enhancing immune response by electrical stimulation
US8784810B2 (en) * 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
CN101528300A (en) * 2006-09-05 2009-09-09 Tti优而美株式会社 Transdermal drug delivery systems, devices, and methods using inductive power supplies
CA2671069A1 (en) 2006-12-01 2008-06-12 Tti Ellebeau, Inc. Systems, devices, and methods for powering and/or controlling devices, for instance transdermal delivery devices
MX2009011127A (en) * 2007-04-18 2010-03-10 Janssen Alzheimer Immunotherap Prevention and treatment of cerebral amyloid angiopathy.
US8003097B2 (en) * 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
SI2182983T1 (en) * 2007-07-27 2014-09-30 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases with humanised anti-abeta antibodies
JO3076B1 (en) * 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
BRPI0819210A2 (en) * 2007-10-25 2015-06-23 Trellis Bioscience Inc Anti-rsv G Protein Antibodies
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
EP2440694A4 (en) * 2009-06-09 2012-12-19 Tti Ellebeau Inc Long life high capacity electrode, device, and method of manufacture
US9433671B2 (en) 2012-03-30 2016-09-06 Artificial Cell Technologies, Inc. Anti-malaria compositions and methods
US8883717B2 (en) 2012-03-30 2014-11-11 Artificial Cell Technologies, Inc. Antigenic compositions and methods
US10272160B2 (en) * 2015-04-16 2019-04-30 The Research Foundation For The State University Of New York Nanostructures comprising cobalt porphyrin-phospholipid conjugates and polyhistidine-tags
EP3349788A4 (en) 2015-09-16 2019-06-05 Artificial Cell Technologies, Inc. Anti-malaria compositions and methods
US11043823B2 (en) * 2017-04-06 2021-06-22 Tesla, Inc. System and method for facilitating conditioning and testing of rechargeable battery cells
US20210290754A1 (en) * 2018-08-07 2021-09-23 Wisconsin Alumni Research Foundation (Warf) Recombinant biologically contained filovirus vaccine
JP2022522112A (en) 2019-02-08 2022-04-14 ウィスコンシン アルムニ リサーチ ファンデイション Humanized cell line
US11807872B2 (en) 2019-08-27 2023-11-07 Wisconsin Alumni Research Foundation (Warf) Recombinant influenza viruses with stabilized HA for replication in eggs
US20220226465A1 (en) 2021-01-18 2022-07-21 ConserV Bioscience Coronavirus Immunogenic Compositions, Methods and Uses Thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057540A (en) * 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
WO1992016231A1 (en) * 1991-03-21 1992-10-01 Smithkline Beecham Biologicals S.A. HERPES SIMPLEX VACCINE COMPRISING HSV GLYCOPROTEIN gD AND 3 dEACYLATED MONOPHOSPHORYL LIPID A
US5194595A (en) * 1987-12-23 1993-03-16 The Upjohn Company Chimeric glycoproteins containing immunogenic segments of the glycoproteins of human respiratory syncytial virus

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ZA814386B (en) 1980-07-01 1982-07-28 Nat Res Dev Production of viral antigens
GB9120221D0 (en) 1991-09-23 1991-11-06 Smithkline Beecham Biolog Novel compounds
JP4028593B2 (en) * 1993-03-23 2007-12-26 グラクソスミスクライン・バイオロジカルス・ソシエテ・アノニム 3-O deacylated monophosphoryl lipid A-containing vaccine composition

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057540A (en) * 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US5194595A (en) * 1987-12-23 1993-03-16 The Upjohn Company Chimeric glycoproteins containing immunogenic segments of the glycoproteins of human respiratory syncytial virus
WO1992016231A1 (en) * 1991-03-21 1992-10-01 Smithkline Beecham Biologicals S.A. HERPES SIMPLEX VACCINE COMPRISING HSV GLYCOPROTEIN gD AND 3 dEACYLATED MONOPHOSPHORYL LIPID A

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JOURNAL OF VIROLOGY, Volume 60, No. 2, issued November 1986, E.J. STOTT et al., "Human Respiratory Syncytial Virus Glycoprotein G Expressed from a Recombinant Vaccinia Virus Vector Protects Mice Against Live-Virus Challenge", pages 607-613. *
JOURNAL OF VIROLOGY, Volume 61, No. 2, issued February 1987, G.W. WERTZ, "Expression of the Fusion Protein of Human Respiratory Syncytial Virus from Recombinant Vaccinia Virus Vectors and Protection of Vaccinated Mice", pages 293-301. *
THE JOURNAL OF IMMUNOLOGY, Volume 147, No. 7, issued October 1991, R. SCHNEERSON et al., "Evaluation of Monophosphoryl Lipid A (MPL) as an Adjuvant: Enhancement of the Serum Antibody Response in Mice to Polysaccharide-Protein Conjugates by Concurrent Injection with MPL", pages 2136-2140. *

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6623739B1 (en) 1993-12-23 2003-09-23 Smithkline Beecham Biologicals S.A. Vaccines
US7510698B2 (en) 1993-12-23 2009-03-31 Glaxosmithkline Biologicals Sa Vaccines
US7169391B2 (en) 1993-12-23 2007-01-30 Smithkline Beecham Biologicals (S.A.) Vaccines
US7029678B2 (en) 1993-12-23 2006-04-18 Smithkline Beecham Biologicals (S.A.) Vaccines
US6146632A (en) * 1993-12-23 2000-11-14 Smithkline Beecham Biologicals S.A. Vaccines
WO1996011019A1 (en) * 1994-10-05 1996-04-18 Vanderbilt University Interleukin-12 as an adjuvant for paramyxoviridae vaccines
US6071893A (en) * 1994-10-05 2000-06-06 Vanderbilt University Interleukin-12 as an adjuvant for paramyxoviridae vaccines
WO1996033739A1 (en) * 1995-04-25 1996-10-31 Smithkline Beecham Biologicals S.A. Vaccines containing a saponin and a sterol
EP0884056A1 (en) * 1995-04-25 1998-12-16 SMITHKLINE BEECHAM BIOLOGICALS s.a. Vaccines containing a saponin and a sterol
EA000839B1 (en) * 1995-04-25 2000-04-24 Смитклайн Бичем Байолоджикалз С.А. Liposomatic vaccine composition, use same and method for treating a mammal
EP0955059A3 (en) * 1995-04-25 2000-07-12 SMITHKLINE BEECHAM BIOLOGICALS s.a. Vaccines containing a saponin and a sterol
US6020182A (en) * 1996-07-12 2000-02-01 Connaught Laboratories Limited Subunit respiratory syncytial virus vaccine preparation
WO1998002179A1 (en) * 1996-07-12 1998-01-22 Connaught Laboratories Limited Two-step immunization procedure against the pyramyxoviridae family of viruses using recombinant virus and subunit protein preparation
WO1998002457A1 (en) * 1996-07-12 1998-01-22 Connaught Laboratories Limited Subunit respiratory syncytial virus vaccine preparation
WO1998002180A1 (en) * 1996-07-12 1998-01-22 Connaught Laboratories Limited Two-step immunization procedure against the pyramyxoviridae family of viruses using attenuated viral strains and subunit protein preparation
EP1477494A1 (en) * 1996-07-12 2004-11-17 Aventis Pasteur Limited Subunit respiratory syncytial virus vaccine preparation
US7718178B2 (en) 1997-04-05 2010-05-18 Allergy Therapeutics Limited Allergen formulation
US8105605B2 (en) 1997-04-05 2012-01-31 Allergy Therapeutics (Uk) Ltd. Allergen formulation
US7815920B2 (en) 1998-09-21 2010-10-19 Allergy Therapeutics (UK) Ltd Method of preparing an antigen-containing formulation
US8628784B2 (en) * 1998-10-16 2014-01-14 Glaxosmithkline Biologicals S.A. Adjuvant systems and vaccines
WO2001051082A1 (en) * 2000-01-14 2001-07-19 Allergy Therapeutics Limited Composition of antigen and glycolipid adjuvant sublingual administration
EP2100616A3 (en) * 2000-01-14 2009-09-23 Allergy Therapeutics (UK) Limited Composition of antigen and glycolipid adjuvant for sublingual administration
US8470331B2 (en) 2000-01-14 2013-06-25 Allergy Therapeutics (Uk) Limited Composition of antigen and glycolipid adjuvant for sublingual administration
WO2003022878A3 (en) * 2001-09-13 2003-09-18 Aventis Pasteur Subunit respiratory syncytial virus vaccine preparation
WO2003022878A2 (en) * 2001-09-13 2003-03-20 Aventis Pasteur Limited Subunit respiratory syncytial virus vaccine preparation
US10059757B2 (en) 2007-06-01 2018-08-28 Medimmune Limited RSV-specific binding molecules and means for producing them
US10730931B2 (en) 2007-06-01 2020-08-04 Medimmune Limited RSV-specific binding molecules and means for producing them

Also Published As

Publication number Publication date
PT705109E (en) 2001-02-28
AU676340B2 (en) 1997-03-06
DE69426077T3 (en) 2004-09-02
JP3734263B2 (en) 2006-01-11
EP0705109A4 (en) 1998-09-02
FI955667A (en) 1996-01-12
GR3034785T3 (en) 2001-02-28
NO954786L (en) 1996-01-23
RU2160119C2 (en) 2000-12-10
NO954786D0 (en) 1995-11-24
ATE196737T1 (en) 2000-10-15
JPH08510749A (en) 1996-11-12
CA2163550A1 (en) 1994-12-08
AU6957194A (en) 1994-12-20
ES2150493T3 (en) 2000-12-01
DK0705109T4 (en) 2004-05-10
EP0705109B1 (en) 2000-10-04
EP0705109A1 (en) 1996-04-10
EP0705109B2 (en) 2004-01-02
DE69426077D1 (en) 2000-11-09
ES2150493T5 (en) 2004-07-01
US5723130A (en) 1998-03-03
DK0705109T3 (en) 2000-11-13
FI955667A0 (en) 1995-11-24
DE69426077T2 (en) 2001-05-10

Similar Documents

Publication Publication Date Title
US5723130A (en) Adjuvants for vaccines against respiratory syncytial virus
EP0390799B1 (en) Respiratory syncytial virus: vaccines
AP298A (en) Herpes simplex vaccine comprising HSV Glycoprotein gD and 3 deacylated mono-phosphoryl lipid A.
Prince et al. Enhancement of respiratory syncytial virus pulmonary pathology in cotton rats by prior intramuscular inoculation of formalin-inactiva ted virus
Murphy et al. Immunization of cotton rats with the fusion (F) and large (G) glycoproteins of respiratory syncytial virus (RSV) protects against RSV challenge without potentiating RSV disease
Perrin et al. Rabies immunosome (subunit vaccine) structure and immunogenicity. Pre-and post-exposure protection studies
JP3602448B2 (en) Nucleic acid respiratory syncytial virus vaccine
KR102096937B1 (en) Parenteral norovirus vaccine formulations
CZ116799A3 (en) Adjuvant preparation and vaccine
US20100233250A1 (en) Vaccine
KR20030055275A (en) Split enveloped virus preparation
Ambrose et al. Evaluation of the immunogenicity and protective efficacy of a candidate parainfluenza virus type 3 subunit vaccine in cotton rats
Ewasyshyn et al. Comparative analysis of the immunostimulatory properties of different adjuvants on the immunogenicity of a prototype parainfluenza virus type 3 subunit vaccine
EP0936921B1 (en) Two-step immunization procedure against the pyramyxoviridae family of viruses using attenuated viral strains and subunit protein preparation
Takamura et al. Protection studies on winter dysentery caused by bovine coronavirus in cattle using antigens prepared from infected cell lysates.
Ghazi et al. Immunogenicity of influenza and HSV-1 mixed antigen ISCOMs in mice
CA1336955C (en) Respiratory syncytial virus: vaccines and diagnostic assays
CA1340661C (en) Respriatory synctial virus: vaccines and diagnostic assays
JP2001253833A (en) Method for inducing cell-mediated immunity of live vaccine even in inactivated vaccine, and combined vaccine obtained by the method

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA FI JP NO RU US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)

Free format text: FI

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2163550

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 955667

Country of ref document: FI

WWE Wipo information: entry into national phase

Ref document number: 1994918109

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1994918109

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 08553332

Country of ref document: US

WWG Wipo information: grant in national office

Ref document number: 1994918109

Country of ref document: EP