WO2010044921A2 - Intranasal administration of receptor-binding ligands or genes encoding such ligands as a therapeutic regimen for mitigating infections caused by respiratory pathogens - Google Patents

Intranasal administration of receptor-binding ligands or genes encoding such ligands as a therapeutic regimen for mitigating infections caused by respiratory pathogens Download PDF

Info

Publication number
WO2010044921A2
WO2010044921A2 PCT/US2009/046132 US2009046132W WO2010044921A2 WO 2010044921 A2 WO2010044921 A2 WO 2010044921A2 US 2009046132 W US2009046132 W US 2009046132W WO 2010044921 A2 WO2010044921 A2 WO 2010044921A2
Authority
WO
WIPO (PCT)
Prior art keywords
influenza virus
see
pathogen
therapeutic
adenovirus
Prior art date
Application number
PCT/US2009/046132
Other languages
French (fr)
Other versions
WO2010044921A3 (en
Inventor
De-Chu C. Tang
Kent R. Van Kampen
Original Assignee
Vaxin Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Vaxin Inc. filed Critical Vaxin Inc.
Priority to EP09820933A priority Critical patent/EP2296700A2/en
Publication of WO2010044921A2 publication Critical patent/WO2010044921A2/en
Publication of WO2010044921A3 publication Critical patent/WO2010044921A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, e.g. influenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis virus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16211Influenzavirus B, i.e. influenza B virus
    • C12N2760/16234Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention relates generally to the fields of therapeutic drug, vaccinology, and vector technology.
  • the present invention also relates to methods of non-invasive immunization in an animal, products therefrom and uses for the methods and products therefrom.
  • anti-influenza drugs An under-developed line of defense against influenza is the use of anti-influenza drugs.
  • the licensed anti- influenza drugs (the M2 ion channel blockers, amantadine and rimantadine, and the neuraminidase inhibitors, oseltamivir (Tamiflu) and zanamivir (Relenza)) are beneficial for seasonal influenza, but appropriate dosing regimens for avian influenza have not been established (Beigel and Bray, 2008).
  • these anti-influenza drugs may generate drug-resistant influenza virus strains over time (Aoki et al., 2007; Beigel and Bray, 2008; Moscona, 2008; Reece, 2007; Yen et al., 2007).
  • adverse side effects associated with intranasal inoculation of live attenuated influenza vaccine exceed mild runny nose, sore throat, or low-grade fever (Marwick, 2000), and can include destruction of epithelial cells in the upper respiratory tract which can lead to secondary infections with pulmonary complications (Hilleman, 2002; Marwick, 2000).
  • Another biohazard is shedding of the replicating attenuated influenza virus to bystanders.
  • the present invention relates to a prophylactic-therapeutic multipurpose agent that will provide an efficacious, safe, and rapid-response countermeasure against respiratory pathogens.
  • the present invention relates to a therapeutic as well as immunological composition comprising a pathogen-derived ligand that binds to its receptor on target cells and (1) confers in a subject a rapid, therapeutic effect and (2) may also elicit in the subject long-term protective immunity against the pathogen.
  • the present invention also relates to a method of nasal administration of the immunological composition.
  • the pathogen-derived ligand may be expressed by a viral vector.
  • Viral vector gene delivery systems are well-known in the field and are commonly used as a tool for use in gene transfer and gene therapy applications (Stone, 2000).
  • the viral vector may include but is not limited to a retrovirus, adenovirus, adeno-associated virus, alphavirus, or herpes simplex virus, hi another embodiment, the viral vector is a recombinant adenoviral vector.
  • the recombinant adenoviral vector may be derived from adenovirus serotype 5 (Ad5).
  • the recombinant adenoviral vector may be selected from the group consisting of replication-defective adenovirus, non-replicating adenovirus, replication- competent adenovirus, or wild-type adenovirus.
  • the pathogen-derived ligand may also be delivered as purified proteins, or nucleic acids, or virus-like particles.
  • the viral vector may further comprise a promoter sequence selected from the group consisting of viral promoters, avian promoters, CMV promoter, SV40 promoter, ⁇ -actin promoter, albumin promoter, Elongation Factor 1- ⁇ (EF 1- ⁇ ) promoter, P ⁇ K promoter, MFG promoter, pIX, or Rous sarcoma virus promoter.
  • the pathogen-derived ligand may be a therapeutic ligand, antigen, or immunogen.
  • the pathogen-derived ligand may bind to a receptor that is necessary for infection by a respiratory pathogen.
  • the therapeutic ligand binds to a receptor that is necessary for infection by a respiratory pathogen such as influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS-CoV), human rhinovirus (HRV), or respiratory syncytial virus (RSV).
  • a respiratory pathogen such as influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS-CoV), human rhinovirus (HRV), or respiratory syncytial virus (RSV).
  • the therapeutic ligand may bind to a sialic acid-containing receptor, which thereby prevents the influenza virus from binding to the receptor.
  • the therapeutic ligand may be a hemagglutinin protein, such as hemagglutinin subtype 1, 2, 3, 5, or B.
  • the therapeutic ligand is hemagglutinin HAl domain or an HA subfragment containing a receptor-binding ligand, for example, HAl derived from A/New Caledonia/20/99, A/Panama/2007/99, B/Hong Kong/330/01, A/Vietnam/ 1203/04, A/Puerto Rico/8/34, A/Hong Kong/8/68, or 2009H1N1 swine flu virus.
  • the therapeutic ligand may be a nucleoprotein, matrix, or neuraminidase.
  • the therapeutic ligand may bind to an angiotensin-converting enzyme 2 receptor.
  • the therapeutic ligand may be a SARS-CoV spike (S) protein.
  • the therapeutic ligand may be a SARS-CoV polyproteinl A and IB, envelope protein E, matrix protein M, or nucleocapsid protein N.
  • the therapeutic ligand may bind to inter-cellular adhesion molecule 1. In other embodiments wherein the respiratory pathogen may be RSV, the therapeutic ligand may bind to annexin II.
  • the present invention also relates to a composition.
  • the composition may comprise a viral vector as described above.
  • the composition may be delivered intranasally.
  • the present invention relates to a composition such as a vaccine, pharmaceutical, or immunological composition, for in vivo delivery to a subject, comprising a pharmaceutically acceptable carrier or excipient and a pathogen-derived ligand that binds to its receptor on target cells, confers in the subject a rapid, therapeutic effect, and elicits in the animal long- term protective immunity against the pathogen.
  • the present invention also relates to a viral vector that contains and expresses a nucleic acid molecule having a sequence encoding a pathogen-derived ligand.
  • the present invention further relates to immunological compositions containing viral vectors, hi certain embodiments, the composition is administered intranasally.
  • Figure 1 shows protection of mice against influenza virus by intranasal instillation of an Ad vector encoding HAl shortly before challenge.
  • Figure 2 shows protection of mice against influenza virus by intranasal instillation of an Ad vector encoding HAl shortly after challenge.
  • Figure 3 shows protection of mice against HPAI virus by Ad- vectored nasal vaccines.
  • Figure 4 shows protection of ferrets against HPAI virus by Ad- vectored nasal vaccines.
  • the present invention relates to (1) a therapeutic as well as immunological composition comprising a pathogen-derived ligand that binds to its receptor on target cells, confers in a subject a rapid, therapeutic effect, and may also elicit in the subject long-term protective immunity against the pathogen, and (2) a viral vector, preferably a recombinant adenoviral vector, that contains and expresses in vivo a nucleic acid molecule having a sequence encoding a pathogen-derived ligand, and (3) a method of conferring in a subject a rapid therapeutic effect against a pathogen and may also elicit in the subject long-term protective immunity against the pathogen, comprising intranasally administering to the subject a therapeutically effective amount of the therapeutic-immunological composition.
  • nucleic acid refers to a deoxyribonucleic or ribonucleic oligonucleotide in either single- or double-stranded form.
  • the term encompasses nucleic acids, i.e., oligonucleotides, containing known analogues of natural nucleotides.
  • the term also encompasses nucleic-acid-like structures with synthetic backbones, see e.g., Eckstein, 1991; Baserga et al., 1992; Milligan, 1993; WO 97/03211; WO 96/39154; Mata, 1997; Strauss-Soukup, 1997; and Straus, 1996.
  • recombinant refers to a polynucleotide synthesized or otherwise manipulated in vitro (e.g., “recombinant polynucleotide”), to methods of using recombinant polynucleotides to produce gene products in cells or other biological systems, or to a polypeptide ("recombinant protein") encoded by a recombinant polynucleotide.
  • “Recombinant means” encompasses the ligation of nucleic acids having various coding regions or domains or promoter sequences from different sources into an expression cassette or vector for expression of, e.g., inducible or constitutive expression of polypeptide coding sequences in the vectors of invention.
  • heterologous when used with reference to a nucleic acid, indicates that the nucleic acid is in a cell or a virus where it is not normally found in nature; or, comprises two or more subsequences that are not found in the same relationship to each other as normally found in nature, or is recombinantly engineered so that its level of expression, or physical relationship to other nucleic acids or other molecules in a cell, or structure, is not normally found in nature.
  • a similar term used in this context is "exogenous".
  • a heterologous nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged in a manner not found in nature; e.g., a human gene operably linked to a promoter sequence inserted into an adenovirus-based vector of the invention.
  • a heterologous nucleic acid of interest can encode an immunogenic gene product, wherein the adenovirus is administered therapeutically or prophylactically as a carrier or drug- vaccine composition.
  • Heterologous sequences can comprise various combinations of promoters and sequences, examples of which are described in detail herein.
  • a “therapeutic ligand” may be a substance which can bind to a receptor of a target cell with therapeutic effects.
  • a "therapeutic effect" may be a consequence of a medical treatment of any kind, the results of which are judged by one of skill in the filed to be desirable and beneficial.
  • therapeutic effect may be a behavioral or physiologic change which occurs as a response to the medical treatment. The result may be expected, unexpected, or even an unintended consequence of the medical treatment.
  • a “therapeutic effect” may include, for example, a reduction of symptoms in a subject suffering from infection by a pathogen.
  • a “target cell” may be a cell in which an alteration in its activity can induce a desired result or response.
  • an “antigen” may be a substance that is recognized by the immune system and induces an immune response.
  • An “irnmunogen” may be a substance that elicits an immune response from the immune system.
  • a “ligand” may be any substance that binds to and forms a complex with a biomolecule to serve a biological purpose.
  • ligand may also refer to an "antigen” or “immunogen”.
  • antigen and “immunogen” are used interchangeably.
  • a "pathogen” may refer to a viral pathogen (e.g. virus) or a bacterial pathogen.
  • Pathogen also encompasses “respiratory pathogens”.
  • "Expression” of a gene or nucleic acid encompasses not only cellular gene expression, but also the transcription and translation of nucleic acid(s) in cloning systems and in any other context.
  • a "vector” is a tool that allows or facilitates the transfer of an entity from one environment to another.
  • some vectors used in recombinant DNA techniques allow entities, such as a segment of DNA (such as a heterologous DNA segment, such as a heterologous cDNA segment), to be transferred into a target cell.
  • the present invention comprehends recombinant vectors that can include viral vectors, bacterial vectors, protozoan vectors, DNA vectors, or recombinants thereof.
  • exogenous DNA for expression in a vector e.g., encoding an epitope of interest and/or an antigen and/or a therapeutic
  • documents providing such exogenous DNA as well as with respect to the expression of transcription and/or translation factors for enhancing expression of nucleic acid molecules, and as to terms such as "epitope of interest", “therapeutic”, “immune response”, “immunological response”, “protective immune response”, “immunological composition”, “immunogenic composition”, and “vaccine composition”, inter alia, reference is made to U.S . Patent No.
  • drug composition and “drug”, “vaccinal composition” and “vaccine” and “vaccine composition” and “drug-vaccine composition” and “drug-vaccine dual agent” and “therapeutic composition” and “therapeutic-immunologic composition” cover any composition that induces protection against a pathogen.
  • the protection may be due to an inhibition or prevention of infection by a pathogen.
  • the protection may be induced by an immune response against the antigen(s) of interest, or which efficaciously protects against the antigen; for instance, after administration or injection into the subject, elicits a protective immune response against the targeted antigen or immunogen or provides efficacious protection against the antigen or immunogen expressed from the inventive adenovirus vectors of the invention.
  • pharmaceutical composition means any composition that is delivered to a subject. In some embodiments, the composition may be delivered to inhibit or prevent infection by a pathogen.
  • immunogenic composition and "immunological composition” and
  • immunological composition cover any composition that confers in a subject a therapeutic effect and/or elicits in a subject an immune response against the antigen, immunogen, or pathogen of interest; for instance, after administration into a subject, elicits an immune response against the targeted immunogen or antigen of interest.
  • An "immunological response" to a composition, vaccine, antigen, immunogen, pathogen or ligand is the development in the host of a cellular and/or antibody-mediated immune response to the composition, vaccine, antigen, immunogen, pathogen or ligand interest.
  • an "immunological response" includes but is not limited to one or more of the following effects: the production of antibodies, B cells, helper T cells, and/or cytotoxic T cells, directed specifically to an antigen or antigens included in the composition or vaccine of interest.
  • the host will display both a rapid (e.g. within ⁇ 24 hrs.) therapeutic effect and a long-term protective immunological response such that resistance to new infection will be enhanced and/or the clinical severity of the disease reduced.
  • Such protection will be demonstrated by either a reduction or lack of symptoms normally displayed by an infected host, a quicker recovery time and/or a lowered viral titer in the infected host.
  • a “therapeutically effective amount” or an “immunologically effective amount” is an amount or concentration of the recombinant vector encoding the gene of interest, that, when administered to a subject, produces a therapeutic response or an immune response to the gene product of interest.
  • a “circulating recombinant form” refers to recombinant viruses that have undergone genetic reassortment among two or more subtypes or strains. Other terms used in the context of the present invention is “hybrid form", “recombined form”, and “reassortant form”.
  • Chronic isolates refer to viruses or microbes isolated from infected subjects in a clinical setting.
  • Field isolates refer to viruses or microbes that are isolated from infected subjects or from the environment.
  • the methods of the invention can be appropriately applied to prevent diseases as prophylactic vaccination or provide relief against symptoms of disease as therapeutic treatments.
  • the recombinant vectors of the present invention can be administered to a subject either alone or as part of pharmaceutical or immunological or immunogenic composition.
  • the recombinant vectors of the invention can also be used to deliver or administer one or more proteins to a subject of interest by in vivo expression of the protein(s). It is noted that expressed and/or immunological products and/or antibodies obtained in accordance with this invention can be expressed in vitro and used in a manner in which such expressed and/or immunological products and/or antibodies are typically used.
  • cells that express such immunological products and/or antibodies and/or drugs can be employed in in vitro and/or ex vivo applications, e.g., such uses and applications can include diagnostics, assays, ex vivo therapy (e.g., wherein cells that express the gene product and/or immunological response are expanded in vitro and reintroduced into the host or animal), etc., see U.S. Patent No. 5,990,091, WO 99/60164 and WO 98/00166 and documents cited therein.
  • expressed gene products or antibodies that are isolated from herein methods, or that are isolated from cells expanded in vitro following herein administration methods can be administered in compositions, akin to the administration of therapeutics or subunit epitopes or antigens or antibodies to induce protective immunity and/or confer therapeutic effects, stimulate a therapeutic response and/or stimulate adaptive immunity.
  • viral vector includes but is not limited to retroviruses, adenoviruses, adeno-associated viruses, alphavirus, and herpes simplex virus.
  • human adenovirus as used herein is intended to encompass all human adenoviruses of the Adenoviridae family, which include members of the Mastadenovirus genera.
  • the adenovirus can be of serogroup A, B, C, D, E, or F.
  • the human adenovirus can be a serotype 1 (AdI), serotype 2 (Ad2), serotype 3 (Ad3), serotype 4 (Ad4), serotype 6 (Ad6), serotype 7 (Ad7), serotype 8 (Ad8), serotype 9 (Ad9), serotype 10 (AdIO), serotype 11 (AdI 1), serotype 12 (Adl2), serotype 13 (AdB), serotype 14 (Adl4), serotype 15 (Adl5), serotype 16 (Adl6), serotype 17 (Adl7), serotype 18 (Adl8), serotype 19 (Adl9), serotype 19a (Adl9a), serotype 19 ⁇ (AdI 9p), serotype 20 (Ad20), serotype 21 (Ad21), serotype 22 (Ad22), serotype 23 (Ad23), serotype 24 (Ad24), serotype 25
  • receptor-binding ligands recombinant vectors
  • drug- vaccine compositions recombinant adenoviruses
  • adenovirus vectors can display an altered tropism for specific tissues or cell types (Havenga, M.J.E. et al., 2002), and therefore, mixing and matching of different adenoviral capsids, i.e., fiber, or penton proteins from various adenoviral serotypes may be advantageous.
  • adenoviral capsids including fiber and penton
  • Modification of the adenoviral capsids, including fiber and penton can result in an adenoviral vector with a tropism that is different from the unmodified adenovirus.
  • Adenovirus vectors that are modified and optimized in their ability to infect target cells can allow for a significant reduction in the therapeutic or prophylactic dose, resulting in reduced local and disseminated toxicity.
  • Viral vector gene delivery systems are commonly used in gene transfer and gene therapy applications. Different viral vector systems have their own unique advantages and disadvantages.
  • Viral vectors that may be used to express the pathogen-derived ligand of the present invention include but are not limited to adenoviral vectors, adeno-associated viral vectors, alphavirus vector, herpes simplex viral vector, and retroviral vectors, described in more detail below.
  • Adenovirus vectors have many characteristics which are ideal for gene delivery, especially delivery into the respiratory tract. Examples of these characteristics include:
  • potency of an intranasally-administered adenovirus vector may not be interfered by preexisting immunity to adenovirus (Hoelscher et al., 2006; Shi et al., 2001; Van Kampen et al., 2005); while not wishing to be bound by theory, this may be attributed to the high efficiency of gene delivery, high level of transgene expression, and high degree of antigen presentation along the mucosal barrier in the respiratory tract;
  • Adenovirus is a non-enveloped DNA virus.
  • the genome of adenovirus is a linear double-stranded DNA molecule of approximately 36,000 base pairs ("bp") with a 55-kDa terminal protein covalently bound to the 5 '-terminus of each strand.
  • the adenovirus DNA contains identical inverted terminal repeats ("ITRs") of about 100 bp, with the exact length depending on the serotype.
  • ITRs inverted terminal repeats
  • the viral origins of replication are located within the ITRs exactly at the genome ends.
  • DNA synthesis occurs in two stages. First, replication proceeds by strand displacement, generating a daughter duplex molecule and a parental displaced strand. The displaced strand is single stranded and can form a "panhandle" intermediate, which allows replication initiation and generation of a daughter duplex molecule. Alternatively, replication may proceed from both ends of the genome simultaneously, obviating the requirement to form the panhandle structure.
  • the viral genes are expressed in two phases: the early phase, which is the period up to viral DNA replication, and the late phase, which coincides with the initiation of viral DNA replication.
  • the early phase only the early gene products, encoded by regions El, E2, E3 and E4, are expressed, which carry out a number of functions that prepare the cell for synthesis of viral structural proteins (Berk, A. J., 1986).
  • the late phase the late viral gene products are expressed in addition to the early gene products and host cell DNA and protein synthesis are shut off. Consequently, the cell becomes dedicated to the production of viral DNA and of viral structural proteins (Tooze, J., 1981).
  • the El region of adenovirus is the first region of adenovirus expressed after infection of the target cell. This region consists of two transcriptional units, the ElA and ElB genes, both of which are required for oncogenic transformation of primary (embryonal) rodent cultures.
  • the main functions of the ElA gene products are to induce quiescent cells to enter the cell cycle and resume cellular DNA synthesis, and to transcriptionally activate the ElB gene and the other early regions (E2, E3 and E4) of the viral genome. Transfection of primary cells with the ElA gene alone can induce unlimited proliferation (immortalization), but does not result in complete transformation.
  • the ElB encoded proteins assist El A in redirecting the cellular functions to allow viral replication.
  • the ElB 55 kD and E4 33 kD proteins which form a complex that is essentially localized in the nucleus, function in inhibiting the synthesis of host proteins and in facilitating the expression of viral genes. Their main influence is to establish selective transport of viral mRNAs from the nucleus to the cytoplasm, concomitantly with the onset of the late phase of infection.
  • the ElB 21 kD protein is important for correct temporal control of the productive infection cycle, thereby preventing premature death of the host cell before the virus life cycle has been completed.
  • Mutant viruses incapable of expressing the ElB 21 kD gene product exhibit a shortened infection cycle that is accompanied by excessive degradation of host cell chromosomal DNA (deg-phenotype) and in an enhanced cytopathic effect (cyt-phenotype; Telling et al., 1994).
  • the deg and cyt phenotypes are suppressed when in addition the ElA gene is mutated, indicating that these phenotypes are a function of ElA (White, E. et al., 1988).
  • the ElB 21 kDa protein slows down the rate by which ElA switches on the other viral genes. It is not yet known by which mechanisms ElB 21 kD quenches these ElA dependent functions.
  • adenoviruses do not efficiently integrate into the host cell's genome, are able to infect non-dividing cells, and are able to efficiently transfer recombinant genes in vivo (Brady et al., 1994). These features make adenoviruses attractive candidates for in vivo gene transfer of, for example, an antigen or immunogen of interest into cells, tissues or subjects in need thereof.
  • Adenovirus vectors containing multiple deletions are preferred to both increase the carrying capacity of the vector and reduce the likelihood of recombination to generate replication competent adenovirus (RCA).
  • RCA replication competent adenovirus
  • the adenovirus contains multiple deletions, it is not necessary that each of the deletions, if present alone, would result in a replication defective and/or non-replicating adenovirus.
  • the additional deletions may be included for other purposes, e.g., to increase the carrying capacity of the adenovirus genome for heterologous nucleotide sequences.
  • more than one of the deletions prevents the expression of a functional protein and renders the adenovirus replication defective and/or non-replicating and/or attenuated. More preferably, all of the deletions are deletions that would render the adenovirus replication-defective and/or non-replicating and/or attenuated.
  • the invention also encompasses adenovirus and adenovirus vectors that are replication competent and/or wild-type, i.e. comprises all of the adenoviral genes necessary for infection and replication in a subject.
  • Embodiments of the invention employing adenovirus recombinants may include El- defective or deleted, or E3 -defective or deleted, or E4-defective or deleted or adenovirus vectors comprising deletions of El and E3, or El and E4, or E3 and E4, or El, E3, and E4 deleted, or the "gutless" adenovirus vector in which all viral genes are deleted.
  • the adenovirus vectors can comprise mutations in El, E3, or E4 genes, or deletions in these or all adenoviral genes.
  • the El mutation raises the safety margin of the vector because El- defective adenovirus mutants are said to be replication-defective and/or non-replicating in non-permissive cells, and are, at the very least, highly attenuated.
  • the E3 mutation enhances the immunogenicity of the antigen by disrupting the mechanism whereby adenovirus down- regulates MHC class I molecules.
  • the E4 mutation reduces the immunogenicity of the adenovirus vector by suppressing the late gene expression, thus may allow repeated re- vaccination utilizing the same vector.
  • the present invention comprehends adenovirus vectors of any serotype or serogroup that are deleted or mutated in El, or E3, or E4, or El and E3, or El and E4.
  • the "gutless" adenovirus vector is another type of vector in the adenovirus vector family. Its replication requires a helper virus and a special human 293 cell line expressing both EIa and Cre, a condition that does not exist in natural environment; the vector is deprived of all viral genes, thus the vector as a vaccine carrier is non-immunogenic and may be inoculated multiple times for re-vaccination.
  • the "gutless" adenovirus vector also contains 36 kb space for accommodating antigen or immunogen(s) of interest, thus allowing co-delivery of a large number of antigen or immunogens into cells.
  • AdEasy system He et al., 1998) and the subsequently modified AdEasier system (Zeng et al., 2001), which were developed to generate recombinant Ad vectors in 293 cells rapidly by allowing homologous recombination between donor vectors and Ad helper vectors to occur in Escherichia coli cells, such as BJ5183 cells, overnight.
  • pAdEasy comprises adenoviral structural sequences that, when supplied in trans with a donor vector such as pShuttle-CMV expressing an antigen or immunogen of interest, results in packaging of the antigen or immunogen (e.g., immunogens and/or antigens) in an adenoviral capsid.
  • a donor vector such as pShuttle-CMV expressing an antigen or immunogen of interest
  • the sequence of p AdEasy is well known in the art and is publicly and commercially available through Stratagene.
  • the present invention can be generated using the AdHigh system (U.S. Patent Application Serial No. 11/943,901, incorporated herein by reference) (Tang et al., 2009).
  • AdHigh is a safe, rapid, and efficient method of generating high titers of recombinant adenovirus without the risk of generating RCA, which may be detrimental or fatal to subjects.
  • the AdHigh system uses modified shuttle plasmids, such as pAdHigh, to promote the production of RCA-free adenoviruses in permissive cells, such as PER.C6 cells after generating recombinants with an adenovirus backbone plasmid in E. coli cells.
  • shuttle plasmids contain polylinkers or multiple cloning sites for easy insertion of antigens such as, for example, influenza immunogens or antigens.
  • Recombination of the adenoviral shuttle plasmids in conjunction with an adenoviral helper plasmid such as pAdEasy in bacterial cells (i.e., BJ5183) can be easily implemented to produce the recombinant human adenoviruses expressing antigens or immunogens of the invention.
  • Methods of producing recombinant vectors by cloning and restriction analysis are well known to those skilled in the art.
  • RGD motif may be inserted into the H-I loop of an adenovirus vector to enhance its infectivity. This sequence has been shown to be essential for the interaction of certain extracellular matrix and adhesion proteins with a superfamily of cell-surface receptors called integrins. Insertion of the RGD motif may be advantageously useful in immunocompromised subjects.
  • An adenovirus recombinant is constructed by cloning specific antigen or immunogen or fragments thereof into any of the adenovirus vectors such as those described above. The adenovirus recombinant is used to transduce cells of a vertebrate use as an immunizing agent. (See, for example, U.S. Patent Application Ser. No.
  • Adeno-associated virus is a single-stranded DNA parvovirus which is endogenous to the human population. Although capable of productive infection in cells from a variety of species, AAV is a dependovirus, requiring helper functions from either adenovirus or herpes virus for its own replication. In the absence of helper functions from either of these helper viruses, AAV will infect cells, uncoat in the nucleus, and integrate its genome into the host chromosome, but will not replicate or produce new viral particles.
  • the genome of AAV has been cloned into bacterial plasmids and is well characterized.
  • the viral genome consists of 4682 bases which include two terminal repeats of 145 bases each. These terminal repeats serve as origins of DNA replication for the virus. Some investigators have also proposed that they have enhancer functions.
  • the rest of the genome is divided into two functional domains. The left portion of the genome codes for the rep functions which regulate viral DNA replication and vital gene expression.
  • the right side of the vital genome contains the cap genes that encode the structural capsid proteins VPl, VP2 and VP3.
  • the proteins encoded by both the rep and cap genes function in trans during productive AAV replication.
  • AAV is considered an ideal candidate for use as a transducing vector , and it has been used in this manner.
  • Such AAV transducing vectors comprise sufficient cis-acting functions to replicate in the presence of adenovirus or herpes virus helper functions provided in trans.
  • Recombinant AAV rAAV
  • rAAV Recombinant AAV
  • these vectors the AAV cap and/or rep genes are deleted from the viral genome and replaced with a DNA segment of choice.
  • Current vectors can accommodate up to 4300 bases of inserted DNA.
  • plasmids containing the desired vital construct are transfected into adenovirus-infected cells.
  • a second helper plasmid is cotransfected into these cells to provide the AAV rep and cap genes which are obligatory for replication and packaging of the recombinant viral construct.
  • the rep and cap proteins of AAV act in trans to stimulate replication and packaging of the rAAV construct.
  • rAAV is harvested from the cells along with adenovirus. The contaminating adenovirus is then inactivated by heat treatment.
  • Herpes Simplex Virus 1 (HSV-I) is an enveloped, double-stranded DNA virus with a genome of 153 kb encoding more than 80 genes. Its wide host range is due to the binding of viral envelope glycoproteins to the extracellular heparin sulphate molecules found in cell membranes (WuDunn & Spear, 1989). Internalization of the virus then requires envelope glycoprotein gD and fibroblast growth factor receptor (Kaner, 1990). HSV is able to infect cells lyrically or can establish latency. HSV vectors have been used to infect a wide variety of cell types (Lowenstein, 1994; Huard, 1995; Miyanohara, 1992; Liu, 1996; Goya, 1998).
  • HSV vectors There are two types of HSV vectors, called the recombinant HSV vectors and the amplicon vectors.
  • Recombinant HSV vectors are generated by the insertion of transcription units directly intot he HSV genome, through homologous recombination events.
  • the amplicon vectors are based on plasmids bearing the transcription unit of choice, an origin of replication, and a packaging signal.
  • HSV vectors have the obvious advantages of a large capacity for insertion of foreign genes, the capacity to establish latency in neurons, a wide host range, and the ability to confer transgene expression to the CNS for up to 18 months (Carpenter & Stevens, 1996).
  • Retroviruses are enveloped single-stranded RNA viruses, which have been widely used in gene transfer protocols. Retroviruses have a diploid genome of about 7-10 kb, composed of four gene regions termed gag, pro, pol and env. These gene regions encode for structural capsid proteins, viral protease, integrase and viral reverse transcriptase, and envelope glycoproteins, respectively. The genome also has a packaging signal and czs-acting sequences, termed long-terminal repeats (LTRs), at each end, which have a role in transcriptional control and integration.
  • LTRs long-terminal repeats
  • the most commonly used retroviral vectors are based on the Moloney murine leukaemia virus (Mo-MLV) and have varying cellular tropisms, depending on the receptor binding surface domain of the envelope glycoprotein.
  • Mo-MLV Moloney murine leukaemia virus
  • Recombinant retroviral vectors are deleted of all retroviral genes, which are replaced with marker or therapeutic genes, or both. To propagate recombinant retroviruses, it is necessary to provide the viral genes, gag, pol and env in trans.
  • Lentiviruses are complex retroviruses that have the ability to infect and express their genes in both mitotic and post-mitotic cells.
  • the most commonly known lentivirus is the human immunodeficiency virus (HIV), which uses the envelope glycoproteins of other viruses to target a broad range of cell types.
  • HIV human immunodeficiency virus
  • Alphaviruses including the prototype Sindbis virus (SIN), Semliki Forest virus (SFV), and Venezuelan equine encephalitis virus (VEE), constitute a group of enveloped viruses containing plus-stranded RNA genomes within icosahedral capsids.
  • the viral vectors of the present invention are useful for the delivery of nucleic acids expressing antigens or immunogens to cells both in vitro and in vivo.
  • the inventive vectors can be advantageously employed to deliver or transfer nucleic acids to animal, more preferably avian and mammalian cells.
  • Nucleic acids of interest include nucleic acids encoding peptides and proteins, preferably therapeutic (e.g., for medical or veterinary uses) or immunogenic (e.g., for vaccines) peptides or proteins.
  • the codons encoding the antigen or immunogen of interest are "optimized" codons, i.e., the codons are those that appear frequently in, i.e., highly expressed genes in the subject's species, instead of those codons that are frequently used by, for example, influenza virus.
  • Such codon usage provides for efficient expression of the antigen or immunogen in animal cells.
  • the codon usage pattern is altered to represent the codon bias for highly expressed genes in the organism in which the antigen or immunogen is being expressed.
  • Codon usage patterns are known in the literature for highly expressed genes of many species (e.g., Nakamura et al., 1996; Wang et al, 1998; McEwan et al. 1998).
  • the viral vectors can be used to infect a cell in culture to express a desired gene product, e.g., to produce a protein or peptide of interest.
  • the protein or peptide is secreted into the medium and can be purified therefrom using routine techniques known in the art.
  • nucleotide sequences encoding the same can be operably linked to the nucleotide sequence encoding the peptide or protein of interest by routine techniques known in the art.
  • the cells can be lysed and the expressed recombinant protein can be purified from the cell lysate.
  • the cell is an animal cell, more preferably a mammalian cell.
  • cells that are competent for transduction by particular viral vectors of interest include PER.C6 cells, 911 cells, and HEK293 cells.
  • a culture medium for culturing host cells includes a medium commonly used for tissue culture, such as M199-earle base, Eagle MEM (E-MEM), Dulbecco MEM (DMEM), SC-UCMl 02, UP-SFM (GIBCO BRL), EX-CELL302 (Nichirei), EX-CELL293-S (Nichirei), TFBM-Ol (Nichirei), ASF104, among others.
  • Suitable culture media for specific cell types can be found at the American Type Culture Collection (ATCC) or the European Collection of Cell Cultures (ECACC).
  • Culture media can be supplemented with amino acids such as L- glutamine, salts, anti-fungal or anti-bacterial agents such as Fungizone®, penicillin- streptomycin, animal serum, and the like.
  • the cell culture medium can optionally be serum-free.
  • the present invention also provides vectors useful as drug and/or vaccine carriers.
  • the immunogen or antigen can be presented in the viral capsid, alternatively, the antigen can be expressed from a transgene introduced into a recombinant viral genome and carried by the inventive virus.
  • the viral vector can provide any antigen or immunogen of interest. Examples of immunogens are detailed herein.
  • Expression vectors include expression control sequences, such as an origin of replication (which can be eukaryotic origins, e.g., autonomously replicating sequences (ARS)), a promoter, an enhancer, and necessary information processing sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, packaging signals, and transcriptional terminator sequences.
  • an origin of replication which can be eukaryotic origins, e.g., autonomously replicating sequences (ARS)
  • ARS autonomously replicating sequences
  • necessary information processing sites such as ribosome binding sites, RNA splice sites, polyadenylation sites, packaging signals, and transcriptional terminator sequences.
  • the viral vectors of the invention can contain appropriate transcription/translation control signals and polyadenylation signals (e.g., polyadenylation signals derived from bovine growth hormone, SV40 polyadenylation signal) operably associated with the antigen or immunogen sequence(s) to be delivered to the target cell.
  • polyadenylation signals e.g., polyadenylation signals derived from bovine growth hormone, SV40 polyadenylation signal
  • a variety of promoter/enhancer elements may be used depending on the level and tissue- specific expression desired.
  • the promoter can be constitutive or inducible (e.g., the metallothionein promoter), depending on the pattern of expression desired.
  • the promoter may be native or foreign and can be a natural or a synthetic sequence. By foreign, it is intended that the transcriptional initiation region is not found in the wild-type host into which the transcriptional initiation region is introduced.
  • the promoter is chosen so that it will function in the target cell(s) or tissue(s) of interest. Brain-specific, hepatic-specific, and muscle-specific (including skeletal, cardiac, smooth, and/or diaphragm-specific) promoters are contemplated by the present invention. Mammalian promoters are also preferred.
  • the promoter can advantageously be an "early" promoter.
  • An "early” promoter is known in the art and is defined as a promoter that drives expression of a gene that is rapidly and transiently expressed in the absence of de novo protein synthesis.
  • the promoter can also be an inducible promoter that can over-drive transgene expression upon induction by a specific substance.
  • the antigens or immunogens are operatively associated with, for example, a human cytomegalovirus (CMV) major immediate-early promoter, a simian virus 40 (SV40) promoter, a ⁇ -actin promoter, an albumin promoter, an Elongation Factor 1- ⁇ (EF 1- ⁇ ) promoter, a P ⁇ K promoter, a MFG promoter, a pIX promoter, or a Rous sarcoma virus promoter.
  • CMV human cytomegalovirus
  • SV40 simian virus 40
  • a ⁇ -actin promoter an albumin promoter
  • Elongation Factor 1- ⁇ (EF 1- ⁇ ) promoter a P ⁇ K promoter
  • MFG promoter a MFG promoter
  • pIX promoter a Rous sarcoma virus promoter
  • Rous sarcoma virus promoter Rous sarcoma virus promoter.
  • any mammalian viral promoter can also be used in the practice of the invention, in addition to any avian viral promoter.
  • the promoters of immediate early (i.e., ICP4, ICP27) genes of the infectious laryngotracheitis virus (ILTV) virus early (i.e., thymidine kinase, DNA helicase, ribonucleotide reductase) or late (i.e., gB, gD, gC, gK), of the Marek's disease virus, (i.e., gB, gC, pp38, ppl4, ICP4, Meq) or of the herpes virus of turkeys (i.e., gB, gC, ICP4) can be used in the methods and vectors of the present invention.
  • the sequences may be operatively associated with a single upstream promoter and one or more downstream internal ribosome entry site (IRES) sequences (e.g., the picornavirus EMC IRES sequence).
  • IRES internal ribosome entry site
  • specific initiation signals are generally required for efficient translation of inserted protein coding sequences.
  • exogenous translational control sequences which may include the ATG initiation codon and adjacent sequences, can be of a variety of origins, both natural and synthetic.
  • the recombinant vectors and methods of the present invention can be used in the treatment or prevention of various respiratory pathogens.
  • pathogens include, but are not limited to, influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS- CoV), human rhinovirus (HRV), and respiratory syncytial virus (RSV).
  • Influenza viruses which belong to the Orthomyxoviridae family, are classified as A, B, and C based on antigenic differences in their nucleoprotein (NP) and matrix (Ml) protein. Influenza viruses B and C infect humans, while influenza virus A infects human, avian species, equine species, swine, and even sea mammals. Further subtyping is based on the antigenicity of two surface glycoproteins, hemagglutinin (HA) and neuraminidase (NA). The HA protein enables the virion to attach to cell surface sialyloligosaccharides (Paulson, J.C., 1985) and is responsible for its hemagglutinating activity (Hirst, G.K., 1941).
  • NP nucleoprotein
  • Ml matrix
  • Influenza viruses B and C infect humans, while influenza virus A infects human, avian species, equine species, swine, and even sea mammals. Further subtyping is based on the anti
  • the HA protein comprises two disulfide-linked chains, HA 1 and HA 2 .
  • HA 1 forms the sialic acid-binding sites and mediates HA attachment to the cell
  • HA 2 forms the membrane- spanning anchor and mediates fusion of viral and cellular membranes.
  • the amino acid sequences of the HA 1 region which is responsible for HA antigenicity, differ from subtype to subtype by 30% or more (Rohm, C. et al, 1996b).
  • There are currently 16 known HA antigenic subtypes Hl to H 16
  • sustained epidemics in humans have been limited to the Hl, H2, and H3 subtypes.
  • NA is a sialidase that cleaves sialic acid residues from the cell surface and is important for release of virus from infected cells. Blocking NA activity, such as through neuraminidase inhibitors, prevents release of new virions from infected cells (Moscona, 2005).
  • Nl to N9 Nine NA subtypes have been identified (Nl to N9), of which the Nl and N2 subtypes have been found in human influenza viruses.
  • Avian species, in particular shorebirds and waterfowl are the natural source of influenza type A viruses. Influenza A viruses can also be transmitted from the aquatic bird reservoir to mammalian species, including humans, seals, whales, horses, pigs, and domestic poultry (Gillim-Ross et al., 2006).
  • Avian influenza A viruses are defined by their virulence; highly virulent types can cause fowl plagues, while avirulent types generally cause only mild disease or asymptomatic infection. In rare instances, however, viruses with low pathogenicity in the laboratory cause outbreaks of severe disease in the field. Nonetheless, the morbidity and mortality associated with these viruses tend to be much lower than those caused by lethal viruses.
  • H4 A/Chicken/Alabama/7395/75 (H4N8)] (Johnson, D.C. et al, 1976) and HlO [A/Cbicken/Germany/N/49 (H10N7)] have been isolated from chickens during severe fowl plague-like outbreaks.
  • Ml proteins are integrated into the virions (Zebedee, S.L. et al, 1988). They form tetramers, have Hl ion channel activity, and, when activated by the low pH in endosomes, acidify the inside of the virion, facilitating its uncoating (Pinto, L.H. et al, 1992). Ml protein that lies within the envelope is thought to function in assembly and budding.
  • RNA molecules (negative sense, or complementary to mRNA) are contained within the viral envelope, in association with NP and three subunits of viral polymerase (PBl, PB2, and PA), which together form a ribonucleoprotein (RNP) complex that participates in RNA replication and transcription.
  • RNP ribonucleoprotein
  • NS2 protein now known to exist in virions (Richardson, J.C. et al, 1991; Yasuda, J. et al, 1993), is thought to play a role in the export of RNP from the nucleus (O'Neill, R.E. et al, 1998) through interaction with Ml protein (Ward, A.C. et al, 1995).
  • NSl protein the only nonstructural protein of influenza A viruses, has multiple functions, including regulation of splicing and nuclear export of cellular niRNAs as well as stimulation of translation (Lamb, R. A. et al, 1996). Its major function is believed to counteract the interferon activity of the host, since an NSl knockout virus was viable although it grew less efficiently than the parent virus in interferon-non-defective cells (Garcia-Sastre, A. et al, 1988).
  • influenza therapeutic ligands, immunogens or antigens useful in the present invention include, but are not limited to, HA, NA, as well as Ml, NS2, and NSl.
  • subtypes that are encompassed by the present invention include, but are not limited to, H10N4, H10N5, H10N7, H10N8, H10N9, HIlNl, H11N13, H11N2, H11N4, H11N6, H11N8, H11N9, H12N1, H12N4, H12N5, H12N8, H13N2, H13N3, H13N6, H13N7,
  • influenza therapeutic ligands, immunogens or antigens can be derived from any known strain of influenza, including all influenza A and B strains, clinical isolates, field isolates, and reassortments thereof.
  • the invention also relates to the use of mutated or otherwise altered influenza genes that reflect, among other things, antigenic drift and antigenic shift.
  • influenza strains include, but are not limited to, turkey influenza virus strain A/Turkey/Ireland/1378/83 (H5N8) (see, e.g., Taylor et al., 1988b), turkey influenza virus strain A/Turkey/England/63 (H7N3) (see, e.g., Alexander et al., 1979; Rott et al., 1979; Horimoto et al., 2001), turkey influenza virus strain A/Turkey/England/66 (H6N2) (see, e.g., Alexander et al., 1979), A/Turkey/England/69 (H7N2) (see, e.g., Alexander et al., 1979; Horimoto et al., 2001), A/Turkey/Scotland/70 (H6N2) (see, e.g., Banks et al., 2000; Alexander et al., 1979), turkey influenza virus strain A/Turkey/England
  • A/Chicken/Vietnam/C58/04 (H5N1), (see, e.g., Anwar et al., 2006;), chicken influenza virus strain A/Chicken/Vietnam/38/2004(H5Nl).
  • H9N1 A/Duck/New York/17542-4/86 (H9N1) (see, e.g., Banks et al., 2000), duck influenza virus strain A/Duck/Alberta/28/76 (H4N6) (see, e.g., Blok et al., 1982), duck influenza virus strain A/Duck/Nanchang/4- 165/2000 (H4N6) (see, e.g., Liu et al., 2003), duck influenza virus strain A/Duck/Germany/49 (H10N7) (see, e.g., Blok et al., 1982), duck influenza virus strain A/Black Duck/Australia/702/78 (H3N8) (see, e.g., Blok et al, 1982), duck influenza virus strain A/Duck/Vietnam/I 1/2004 (H5N1), (see, e.g., Anwar et al., 2006), duck influenza virus strain A/Duck/Alber
  • A/Duck/Alberta/35/76 HlNl (see, e.g., Austin et al., 1990), avian influenza virus strain A/Mallard duck/Gurjev/263/82 (H14N5), (see, e.g., Kawaoka et al., 1990), avian influenza virus strain A/Mallard duck/PA/ 10218/84 (H5N2) (see, e.g., Smirnov et al., 2000), avian influenza virus strain A/Mallard duck/Astrakhan/244/82 (H14N6) (see, e.g., Karasin et al., 2002), goose influenza virus strain A/Goose/Guangdong/1/96 (see, e.g., Xu et al., 1999), goose influenza virus strain A/Goose/Leipzig/I 37-8/79 (H7N7) (see, e.g., Horimoto et al
  • avian influenza virus strain A/Gull/Maryland/704/77 H13N6
  • H13N6 avian influenza virus strain A/Gull/Maryland/704/77
  • H16N3 avian influenza virus strain A/Black-headed gull/Sweden/5/99
  • H2N8 avian influenza virus strain A/Herring gull/DE/677/88
  • H5N1 see, e.g., Terregino et al., 2006
  • avian influenza virus strain A/Hong Kong/156/97 A/HK/156/97
  • A/Quail/HK/Gl/97 (H9N2) (see, e.g., Leneva et al., 2001), avian influenza virus strain A/Quail/Hong Kong/AF157/93 (H9N2) (see, e.g., Karasin et al., 2002), avian influenza virus strain A/Teal/HK/W312/97 (H6N1) (see, e.g., Leneva et al., 2001), avian influenza virus strain A/Shearwater/West Australia/2576/79 (H15N9) (see, e.g., Rohm et al., 1996), avian influenza virus strain A/Shearwater/Australia/72 (H6N5) (see, e.g., Harley et al., 1990), avian influenza virus strain A/Hong Kong/212/03 (see, e.g., Shinya et al., 2005), avian influenza virus strain A/England/
  • influenza virus C see, e.g., Chare et al., 2003
  • influenza virus strain A/Equi/2/Kildare/89 see, e.g., Quinlivan et al., 2004
  • influenza virus type B/Panama 45/90 see, e.g., Zakay-Rones et al., 1995
  • live, cold-adapted, temperature-sensitive (ca/ts) Russian influenza A vaccines see, e.g., Palker et al., 2004
  • swine Hl and H3 influenza viruses see, e.g., Gambaryan et al., 2005
  • swine influenza A viruses see, e.g., Landolt et al., 2005
  • swine influenza virus (SIV) see, e.g., Clavijo et al., 2002
  • H3N2 A/Shanghai/6/90 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Akita/1/94 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Akita/1/95 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Memphis/6/90 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Udorn/307/72 (H3N2) (see, e.g., Karasin et al., 2002; Iuferov et al., 1984), influenza virus strain A/Singapore/1/57 (H2N2) (see, e.g., Karasin et al., 2002; Zhukova et al., 1975), influenza virus strain A/Ohi
  • antigenicity of influenza viruses changes gradually by point mutation (antigenic drift) or drastically by genetic reassortment (antigenic shift) (Murphy, B. R. et al, 1996). Immunological pressure on HA and NA is thought to drive antigenic drift. Antigenic shift can be caused by either direct transmission of nonhuman influenza viruses to humans or the reassortment of genes from two different influenza viruses that have infected a single cell (Webster, R.G. et al, 1982). Theoretically, 256 different combinations of RNA can be produced from the shuffling of the eight different genomic segments of the virus. Genetic reassortment is well documented both in vitro and in vivo under laboratory conditions (Webster, R. G. et al, 1975).
  • influenza therapeutic ligands immunogens or antigens that have undergone antigenic drift or antigenic shift, including clinical isolates of influenza, field or environmental isolates of influenza, hybrid forms, and reassortant forms of influenza.
  • Severe acute respiratory syndrome-associated coronavirus is the virus which causes SARS.
  • Coronaviruses are enveloped, positive-stranded RNA viruses that can cause enteric or respiratory tract infectious in a variety of species including human, livestock, and pets.
  • the SARS-CoV genome contains 11 significant ORFs, which include the following: IA and IB, which encode polyproteins of the replicase complex; envelope spike protein S, which mediates attachment to cellular receptors and entry by fusion with cell membranes; small envelope protein E, which serves as a scaffold protein to trigger assembly; matrix protein M, which is an integral membrane protein involved in budding and which interacts with the nucleocapsid and the S proteins; and nucleocapsid protein N.
  • IA and IB which encode polyproteins of the replicase complex
  • envelope spike protein S which mediates attachment to cellular receptors and entry by fusion with cell membranes
  • small envelope protein E which serves as a scaffold protein to trigger assembly
  • matrix protein M which is an integral membrane protein involved in budding and which interacts with the nucleocapsid and the S proteins
  • nucleocapsid protein N nucleocapsid protein
  • the S protein extends from the surface of the virion and serves as the major viral attachment protein.
  • the interaction between receptor and S protein is an important determinant of species specificity and tissue tropism.
  • Such receptors include angiotensin- converting enzyme 2 and CD209L, although angiotensin-converting enzyme 2 serves as a more efficient receptor (Li et al., 2007).
  • NAbs neutralizing antibodies
  • SARS-CoV neutralizing antibodies
  • Subbarao, et al., 2005 Analysis of the epitopes recognized by these NAbs have revealed that the receptor binding domain of the S protein is a neutralization determinant (Greenough, et al., 2005).
  • monoclonal antibodies capable of neutralizing SARS-CoV by targeting the S protein have been identified and protected mice against subsequent virus challenge (Greenough, et al., 2005).
  • the SARS-CoV therapeutic ligands, immunogens or antigens include, but are not limited to, IA, IB, spike protein S, envelope protein E, matrix protein M, and nucleocapsid protein N.
  • HRV Human rhinovirus
  • IAM-I human intercellular adhesion molecule- 1
  • the second group binds to low- density lipoprotein and related proteins (Rossmann et al., 2007).
  • HRV are non-enveloped viruses with an icosahedral capsid that encloses a single- stranded, positive-sense RNA genome.
  • the viral polyprotein is divided into a Pl region, P2 region, and P3 region.
  • the Pl region encodes capsid proteins VPl, VP2, VP3, and VP4, while the P2 and P3 regions include proteins 2APro, 2B, 2C, 3A, 3B (VPg), 3CPro, and 3DPoI.
  • HRV typically infects through docking to epithelial cells via specific cellular receptors.
  • the binding of HRV to the receptors induces conformational changes of the capsid and leads to the release of viral RNA (Nurani, et al., 2003).
  • a majority of HRV uses the cell surface receptor ICAM-I to bind to and infect epithelial cells.
  • the HRV therapeutic ligands, immunogens or antigens that may be used include, but are not limited to, proteins or ligands which bind to ICAM-I .
  • Such ligands may be acquired through methods known in the art, such high throughput screening or high throughout virtual screening (see Taylor et al., 2007, which is incorporated herein by reference), fragment-based ligand discovery (see Erlanson 2007, which is incorporated herein by reference), and tethering technology (see U.S. Patent Nos. 5,925,529 and 6,998,233, which is incorporated herein by reference).
  • Human respiratory syncytial virus causes respiratory tract infections. It is a large negative-sense, single-stranded RNA virus, member of the family of Paramyxoviridae, subfamily pneumovirinae. Its name comes from the fact that F proteins on the surface of the virus cause the cell membranes on nearby cells to merge, forming syncytia.. Annexin II that binds to RSV G glycoprotein may be RSVs receptor on human cells (Malhotra et al., 2003). Human RSV is responsible for a spectrum of respiratory tract diseases in people of all ages throughout the world. It is the major cause of lower respiratory tract illness during infancy and childhood.
  • Ribavirin a nucleoside analogue
  • the other two drugs, RespiGam.RTM. and palivizumab, polyclonal and monoclonal antibody immunostimulants, are intended to be used in a preventive way.
  • the present invention comprehends the use of more than therapeutic ligand, immunogen or antigen in the vectors and methods disclosed herein, delivered either in separate recombinant vectors, or together in one recombinant vector so as to provide a multivalent vaccine or immunogenic composition that stimulates or modulates immunogenic response to one or more influenza strains and/or hybrids.
  • the present invention encompasses the use of a therapeutic ligand, immunogen or antigen from more than one pathogen in the vectors and methods disclosed herein, delivered either in separate recombinant vectors, or together in one recombinant vector.
  • the recombinant vectors and methods of the invention comprehend the use of adjuvant molecules that can modulate immune responses upon delivery of recombinant vectors or pharmaceutical or immunogenic/immunological compositions.
  • adjuvant molecules can include, but are not limited to, immunomodulatory molecules such as interleukins, interferon, and co-stimulatory molecules.
  • the immunomodulatory molecules can be co-administered with the inventive pharmaceutical or immunogenic compositions, or alternatively, the nucleic acid of the immunomodulatory molecule(s) can be co-expressed along with the therapeutic ligands or immunogens or antigens in the recombinant vectors of the invention.
  • expression in the subject of the heterologous sequence can result in expression products which can bind to or block receptors on target cells. Consequently, respiratory pathogens are prevented from binding to these receptors themselves, and thereby cannot initiate infection.
  • expression products such as influenza HAl molecules may bind to sialic acid-containing receptors on target cells and prevent initiation of influenza virus infection. Partial reduction in the number of receptors in the upper respiratory tract is sufficient in alleviating transmission of influenza virus (Tumpey et al., 2007).
  • expression products such as SARS-CoV S protein molecules may bind to the angiotensin- converting enzyme 2 receptor on target cells and prevent SARS-COV infection (Li et al., 2003).
  • expression products comprising ligands of ICAM-I may bind to the ICAM-I receptor on target cells and prevent infection of HRV (Rossmann et al., 2000).
  • expression products such as RSV G glycoprotein may bind to annexin II on target cells (Malhotra et al., 2003) and prevent infection of RSV.
  • the recombinant vectors of the present invention may be used in a vaccine or pharmaceutical to provide a means to inhibit or prevent respiratory pathogen infection.
  • Target cells as used herein may be any cell involved in the pathogenicity of respiratory viruses, including, but not limited to, airway epithelial or mucosal cells.
  • the recombinant vectors of the present invention may be used in an immunological and/or therapeutic compositions to provide a means to confer protection against a pathogen.
  • the molecular biology techniques used in the context of the invention are described by Sambrook et al. (2001).
  • the nucleotide sequence encoding the therapeutic ligands or antigens can have deleted therefrom a portion encoding a transmembrane domain.
  • the vector or pharmaceutical or immunogenic composition can further contain and express in a host cell a nucleotide sequence encoding a heterologous tPA signal sequence such as human or avian tPA and/or a stabilizing intron, such as intron II of the rabbit ⁇ -globin gene.
  • a vector can be administered to a subject in an amount to achieve the amounts stated for gene product (e.g., therapeutic, epitope, antigen, and/or antibody) compositions.
  • the invention envisages dosages below and above those exemplified herein, and for any composition to be administered to a subject, including the components thereof, and for any particular method of administration, it is preferred to determine therefor: toxicity, such as by determining the lethal dose (LD) and LD 50 in a suitable model; and the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response, such as by titrations of sera and analysis thereof, e.g., by ELISA and/or seroneutralization analysis.
  • toxicity such as by determining the lethal dose (LD) and LD 50 in a suitable model
  • the dosage of the composition(s), concentration of components therein and timing of administering the composition(s) which elicit a suitable response, such as by titrations of sera and analysis thereof, e.g., by ELISA and/or seroneutralization analysis.
  • a suitable response such as by titrations of sera and analysis
  • Subjects to which the recombinant vectors and/or pharmaceutical or immunological compositions can be administered include all animal species such as human, primate, feline, canine, avian, murine, bovine, equine, porcine, etc.
  • compositions of the invention include liquid preparations for orifice, or mucosal, e.g., intranasal, oral, anal, vaginal, peroral, intragastric, etc., administration such as suspensions, solutions, sprays, syrups or elixirs; and, preparations for parenteral, epicutaneous, subcutaneous (i.e., through lower neck), intradermal, intraperitoneal, intramuscular, intranasal, or intravenous administration (e.g., injectable administration) such as sterile suspensions or emulsions.
  • parenteral epicutaneous, subcutaneous (i.e., through lower neck), intradermal, intraperitoneal, intramuscular, intranasal, or intravenous administration (e.g., injectable administration) such as sterile suspensions or emulsions.
  • parenteral epicutaneous, subcutaneous (i.e., through lower neck), intradermal, intraperitoneal, intramuscular, intranasal, or intrave
  • inventive compositions or sequential performance of herein methods, e.g., periodic administration of inventive compositions such as in the course of therapy or treatment for a condition and/or booster administration of pharmaceutical or immunological compositions and/or in prime-boost regimens; and, the time and manner for sequential administrations can be ascertained without undue experimentation.
  • compositions and methods for making and using vectors including methods for producing gene products and/or immunological products and/or antibodies in vivo and/or in vitro and/or ex vivo (e.g., the latter two being, for instance, after isolation therefrom from cells from a host that has had an administration according to the invention, e.g., after optional expansion of such cells), and uses for such gene and/or pharmaceutical or immunological products and/or antibodies, including in diagnostics, assays, therapies, treatments, and the like.
  • Vector compositions are formulated by admixing the vector with a suitable carrier or diluent; and, gene product and/or pharmaceutical or immunological product and/or antibody compositions are likewise formulated by admixing the gene and/or immunological product and/or antibody with a suitable carrier or diluent; see, e.g., U.S. Patent No. 5,990,091, WO 99/60164, WO 98/00166, documents cited therein, and other documents cited herein, and other teachings herein (for instance, with respect to carriers, diluents and the like).
  • the recombinant vectors may be in admixture with a suitable veterinarily or pharmaceutically acceptable carrier, diluent, or excipient such as sterile water, physiological saline, glucose or the like.
  • a suitable veterinarily or pharmaceutically acceptable carrier such as sterile water, physiological saline, glucose or the like.
  • the compositions can also be lyophilized.
  • the compositions can contain auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, adjuvants, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
  • DMSO has been known to enhance the potency of vaccine and immunogenic compositions, particularly in regard to in ovo delivery of vectors or immunogenic compositions comprising vectors.
  • DMSO is thought to enhance the potency of vaccines by increasing the permeability of cellular membranes (Oshop et al, 2003).
  • Other agents or additives that are capable of permeabilizing cells, reducing the viscosity of amniotic fluid, and exhibiting a higher compliance rate as compared to DMSO can be used in the formulation of vaccines or immunogenic compositions, especially when administered by in ovo delivery.
  • TDM tetradecyl maltoside
  • Formulations containing 0.125% TDM can cause moderate alterations in cell morphology, while higher concentrations of TDM (i.e., 0.5%) can transiently induce more extensive morphological changes.
  • the quantity of vector to be administered will vary for the subject and condition being treated and will vary from one or a few to a few hundred or thousand micrograms of body weight per day and preferably the dose of vaccine or pharmaceutical or immunological composition being chosen preferably between 10 — 10 plaque forming units (PFU), preferably 10 2 -10 10 PFU per subject.
  • vaccines containing the above titer should be diluted with a pharmaceutically or veterinarily acceptable liquid such as physiological saline to a final volume of approximately 0.5 ml or 0.01 ml.
  • a vector can be non-invasively administered to a subject in an amount to achieve the amounts stated for gene product (e.g., epitope, antigen, therapeutic, and/or antibody) compositions.
  • gene product e.g., epitope, antigen, therapeutic, and/or antibody
  • the invention envisages dosages below and above those exemplified herein, and for any composition to be administered to a subject, including the components thereof, and for any particular method of administration, it is preferred to determine: toxicity, such as by determining the lethal dose (LD) and LD 50 in a suitable model; and, the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response, such as by titrations of sera and analysis thereof, e.g., by ELISA and/or seroneutralization analysis.
  • LD lethal dose
  • Recombinant vectors can be administered in a suitable amount to obtain in vivo expression corresponding to the dosages described herein and/or in herein cited documents. For instance, suitable ranges for viral suspensions can be determined empirically. If more than one gene product is expressed by more than one recombinant, each recombinant can be administered in these amounts; or, each recombinant can be administered such that there is, in combination, a sum of recombinants comprising these amounts.
  • the therapeutic effects can also be achieved by administration of receptor-binding ligands (purified receptor-binding proteins or killed viruses that display receptor-binding ligands or receptor-binding ligands embedded in virus- like particles).
  • receptor-binding ligands purified receptor-binding proteins or killed viruses that display receptor-binding ligands or receptor-binding ligands embedded in virus- like particles.
  • dosages can be as described in documents cited herein or as described herein or as in documents referenced or cited in herein cited documents.
  • the dosage should be a sufficient amount of composition to confer an effect analogous to compositions wherein the ligand(s) are directly present; or to have expression analogous to dosages in such compositions; or to have expression analogous to expression obtained in vivo by recombinant compositions.
  • the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable immunological response can be determined by methods such as by antibody titrations of sera, e.g., by ELISA and/or seroneutralization assay analysis. Such determinations do not require undue experimentation from the knowledge of the skilled artisan, this disclosure and the documents cited herein. And, the time for sequential administrations can be likewise ascertained with methods ascertainable from this disclosure, and the knowledge in the art, without undue experimentation.
  • the pharmaceutical or immunological compositions contemplated by the invention can also contain an adjuvant.
  • Suitable adjuvants include fMLP (N-formyl-methionyl-leucyl- phenylalanine; U.S. Patent No. 6,017,537) and/or acrylic acid or methacrylic acid polymer and/or a copolymer of maleic anhydride and of alkenyl derivative.
  • the acrylic acid or methacrylic acid polymers can be cross-linked, e.g., with polyalkenyl ethers of sugars or of polyalcohols. These compounds are known under the term "carbomer” (Pharmeuropa, Vol. 8, No. 2, June 1996). A person skilled in the art may also refer to U.S.
  • Patent No. 2,909,462 (incorporated by reference), which discusses such acrylic polymers cross-linked with a polyhydroxylated compound containing at least 3 hydroxyl groups: in one embodiment, a polyhydroxylated compound contains not more than 8 hydroxyl groups; in another embodiment, the hydrogen atoms of at least 3 hydroxyls are replaced with unsaturated aliphatic radicals containing at least 2 carbon atoms; in other embodiments, radicals contain from about 2 to about 4 carbon atoms, e.g., vinyls, allyls and other ethylenically unsaturated groups. The unsaturated radicals can themselves contain other substituents, such as methyl.
  • Carbopol® Noveon Inc., Ohio, USA
  • Carbopol® 974P, 934P, and 971P are particularly suitable for use as an adjuvant. They are cross-linked with an allyl sucrose or with allylpentaerythritol, as to which, mention is made of the products Carbopol® 974P, 934P, and 971P.
  • copolymers of maleic anhydride and of alkenyl derivative mention is made of the EMA® products (Monsanto), which are copolymers of maleic anhydride and of ethylene, which may be linear or cross-linked, for example cross-linked with divinyl ether. Also, reference may be made to U.S. Patent No. 6,713,068 and crizson, W. et al., 1960; incorporated by reference).
  • Cationic lipids containing a quaternary ammonium salt are described in U.S. Patent No. 6,713,068, the contents of which are incorporated by reference, can also be used in the methods and compositions of the present invention.
  • cationic lipids preference is given to DMRIE (N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-l-propane ammonium; WO96/34109), advantageously associated with a neutral lipid, advantageously DOPE (dioleoyl-phosphatidyl-ethanol amine; Behr J. P., 1994), to form DMRIE-DOPE.
  • a recombinant vaccine or pharmaceutical or immunogenic or immunological composition can also be formulated in the form of an oil-in-water emulsion.
  • the oil-in-water emulsion can be based, for example, on light liquid paraffin oil (European Pharmacopea type); isoprenoid oil such as squalane, squalene, EICOSANE TM or tetratetracontane; oil resulting from the oligomerization of alkene(s), e.g., isobutene or decene; esters of acids or of alcohols containing a linear alkyl group, such as plant oils, ethyl oleate, propylene glycol di(caprylate/caprate), glyceryl tri(caprylate/caprate) or propylene glycol dioleate; esters of branched fatty acids or alcohols, e.g., isostearic acid esters.
  • the oil advantageously is used in combination with emulsifiers to form the emulsion.
  • the emulsifiers can be nonionic surfactants, such as esters of sorbitan, mannide (e.g., anhydromannitol oleate), glycerol, polyglycerol, propylene glycol, and oleic, isostearic, ricinoleic, or hydroxystearic acid, which are optionally ethoxylated, and polyoxypropylene-polyoxyethylene copolymer blocks, such as the Pluronic® products, e.g., L121.
  • the adjuvant can be a mixture of emulsifier(s), micelle-forming agent, and oil such as that which is available under the name Provax® (IDEC Pharmaceuticals, San Diego, CA).
  • the viral vector expressing one or more antigen or ligand or immunogen of interest can be preserved and/or conserved and stored either in liquid form, at about 5 0 C, or in lyophilized or freeze-dried form, in the presence of a stabilizer. Freeze-drying can be according to well-known standard freeze-drying procedures.
  • the pharmaceutically acceptable stabilizers may be SPGA (sucrose phosphate glutamate albumin; Bovarnick, et al., 1950), carbohydrates (e.g., sorbitol, mannitol, lactose, sucrose, glucose, dextran, trehalose), sodium glutamate (Tsvetkov, T.
  • proteins such as peptone, albumin or casein, protein containing agents such as skimmed milk (Mills, CK. et al., 1988; Wolff, E. et al., 1990), and buffers (e.g., phosphate buffer, alkaline metal phosphate buffer).
  • buffers e.g., phosphate buffer, alkaline metal phosphate buffer.
  • Example 1 Development of the AdHi gh system for rapid generation of RCA-free Ad vectors in PER.C6 cells. Production of RCA-free Ad vectors through recombination in PER.C6 cells is a slow process. Generation of Ad vectors by AdEasy in 293 cells is fast; however, AdEasy is not compatible with the PER.C6 cell line for propagation of high-titer RCA-free Ad vectors. Hence, a new system named AdHigh was constructed by repairing the backbone of the AdEasy' s pShuttleCMV vector. pAdHigh retains the essential components for recombination with pAdEasyl in E.
  • the low-titer production of AdEasy vectors in PER.C6 cells may be attributed to the lack of adenovirus nucleotides 3511-3533, as this segment is present in pAdApt (sequence information provided by Crucell) but missing in pShuttleCMV.
  • the pIX promoter (Babiss and Vales, 1991) is intact in pAdApt but defective in pShuttleCMV.
  • pIX participates in the stability of adenovirus particles as a capsid cement (Rosa-Calatrava et al., 2001). The approach that was used in generating pAdHigh by repairing pShuttleCMV is as follows.
  • pShuttleCMV' s CMV promoter, the adjacent multiple cloning site, and flanking Ad sequences were replaced as one unit with their counterpart from pAdApt through homologous recombination. This was possible because these two shuttle vectors share extensive overlapping sequences.
  • the full-length tetracycline (Tc) resistance gene (Backman and Boyer, 1983; Peden, 1983) was amplified from the plasmid pBR322 by polymerase chain reaction (PCR) using primers 5'- GAGCTCGGTACCTTCTCATGTTTGACAGCTTATCAT-3' and 5'- TCTAGAGGTACCAACGCTGCCCGAGATGCGCCGCGT-3' with built-in Kpnl sites.
  • the amplified Tc gene was inserted into the Kpnl site of the ampicillin (Amp)-resistant plasmid pAdApt to generate a new plasmid ⁇ AdA ⁇ t-Tc, which was selected by applying both Amp and Tc to the growth medium.
  • pShuttleCMV was transformed into E. coli BJ5183 cells, and transformants were selected by kanamycin (Kan) resistance. Kan-resistant cells were immediately transformed with pAdApt-Tc, and recombinants were selected by applying both Kan and Tc. The recombinant can confer both Kan and Tc resistance to E.
  • coli BJ5183 cells only when the defective Ad sequence in pShuttleCMV has been replaced through homologous recombination with its counterpart in pAdApt.
  • the resultant pAdHigh-Tc was purified from E. coli BJ5183 cells and transformed into E. coli DHlOB cells.
  • the plasmid was validated by DNA sequencing. Transgenes were subsequently inserted into convenient restriction sites of pAdHigh-Tc to replace the Tc gene.
  • the resultant plasmid was allowed to recombine with the adenovirus backbone plasmid pAdEasyl in E. coli BJ5183 cells as described (Zeng et al., 2001).
  • AdHigh-derived adenovirus vectors were RCA-free (Apptec reports on RCA analysis of Vaxin's AdHigh-derived adenovirus vectors).
  • Example 2 Construction of RCA-free Ad vectors encoding HAl
  • the Centers for Disease Control and Prevention provided the PI with the influenza virus strains A/New Caledonia/20/99 (HlNl), A/Panama/2007/99 (H3N2), and
  • Caledonia/20/99 HAl (AdNCHl.1) was generated in human PER.C6 cells as described above.
  • AdPNM.H3.1 An RCA-free adenovirus vector (AdPNM.H3.1) encoding a humanized HAl fragment (349 amino acids) of the A/Panama/2007/99 (H3N2) influenza virus was generated in PER.C6 cells as described above.
  • the full-length humanized HA template was synthesized at GENEART, and the HAl fragment was PCR amplified using primers 5'-
  • the amplified HAl fragment was inserted into the Hindlll-Xbal site of pAdHigh.
  • An RCA-free Ad vector (AdHK.B.1) encoding a humanized HAl fragment (370 amino acids) of the A/Hong Kong/330/01 influenza virus was generated in PER.C6 cells as described above.
  • the full-length humanized HA template was synthesized at GENEART, and the HAl fragment was PCR amplified using primers 5'-
  • AdVN.H5.1 An RCA-free Ad vector (AdVN.H5.1) encoding a humanized HAl fragment (340 amino acids) of the A/Vietnam/ 1203/04 (H5N1) avian influenza virus was generated in
  • PER.C6 cells as described above.
  • the full-length humanized HA template was synthesized at GENEART, and the HAl fragment was PCR amplified using primers 5'- TGCATTGGAAGCTTGCCACCATGGAGAAGA-3' and 5'-
  • Example 3 Intranasal administration of AdNCHl.1 as a novel anti-influenza drug
  • HAl expressed from adenovirus vectors following intranasal instillation and subsequently secreted from transduced cells, may bind to sialic acid-containing receptors in the upper respiratory tract and may block a productive infection by influenza virus during a challenge, as a partial reduction of receptors in the upper respiratory tract may alleviate transmission of influenza virus (Tumpey et al., 2007).
  • This hypothesis was tested by instilling an escalating dose of AdNCHl.1 into the nostril of mice at different time points prior to challenge with a lethal dose of A/Puerto Rico/8/34 (HlNl) influenza virus.
  • mice 100% of mice were protected by intranasal administration of 1 X 10 5 or 1 X 10 7 ifu of AdNCH 1.1 vectors one day prior to challenge, or by intranasal administration of 1 X 10 7 ifu of AdNC .Hl.1 vectors three days prior to challenge whereas control mice all died within 13 days post-challenge. Since one and three days are too short for eliciting an adaptive immune response against HAl , the protection may have been mediated by interference with influenza virus infection through HAl binding to sialic acid-containing receptors in the upper respiratory tract.
  • Example 4 Intranasal administration of Ad vectors encoding HAl as a vaccination modality Intranasal administration of adenovirus- vectored vaccines is an effective approach for immunizing animals (Hoelscher et al., 2006; Shi et al., 2001; Xiang et al., 1996) and humans (Van Kampen et al., 2005). Intranasal instillation of an adenovirus vector encoding HAl was as effective as its counterpart containing both HAl and HA2 in eliciting protective immunity against a virulent HPAI virus in mice ( Figure 3) and ferrets ( Figure 4), respectively. Results suggest that this regimen may be utilized as an anti-influenza drug- vaccine multipurpose treatment. In addition to influenza, this regimen ought to be effective in protecting animals and humans against other pathogens that require receptor binding to initiate an infection in a wide variety of disease settings.
  • Example 5 Determination of the minimal dose of AdNCH 1.1 vectors in pre-challenge administration for preventing influenza in mice.
  • Balb/c mice are treated with an escalating dose of AdNCH 1.1 vectors at different time points prior to influenza virus challenge in order to determine the minimal dose required for preventing influenza. Since preexisting immunity to Ad is commonly found in humans and the potency of adenovirus- vectored nasal vaccine may not be appreciably interfered by preexisting immunity to adenovirus (Hoelscher et al., 2006; Shi et al., 2001 ; Van Kampen et al., 2005), all mice are primed by intranasal instillation of 1 X 10 7 ifu of wild-type adenovirus serotype 5 one month prior to adenovirus treatment.
  • mice Control and treated mice are challenged by intranasal instillation of lOXLDso of A/Puerto Rico/8/34 (HlNl) influenza virus. Challenged animals are monitored for survival for 30 days. There are 10 mice per group, and each experiment is repeated once (Table 1).
  • Example 6 Determining the minimal dose of AdNCHl.1 vectors in post-challenge administration for alleviating influenza in mice.
  • mice are treated with an escalating dose of AdNCH 1.1 at different time points post- challenge to determine the minimal dose required for alleviating lethal symptoms of influenza. All mice in these studies are primed and challenged as described above. There are 10 mice per group, and each experiment is repeated once (Table 2).
  • Example 7 Determining the potential of the HAl drug in providing a broad protection against different influenza virus strains.
  • Balb/c mice are treated with AdNCHl.1 vectors encoding an Hl HAl followed by challenge with A/Puerto Rico/8/34 (HlNl) and A/Hong Kong/8/68 (H3N2) influenza virus strains as described (Ulmer et al., 1993) one day later.
  • AdNCHl.1 vectors encoding an Hl HAl followed by challenge with A/Puerto Rico/8/34 (HlNl) and A/Hong Kong/8/68 (H3N2) influenza virus strains as described (Ulmer et al., 1993) one day later.
  • mice are also treated with AdPNM.H3.1 encoding an H3 HAl (Example T), AdHK.B.l encoding an influenza virus B HAl (Example T), and AdVN.H5.1 encoding an H5 HAl (Example T), followed by challenge with A/Puerto Rico/8/34 (HlNl) and A/Hong Kong/8/68 (H3N2) influenza viruses, respectively, in an attempt to determine the cross-reactivity between a specific HAl and receptors utilized by a variety of influenza virus strains. All mice are primed and challenged as described above. There are 10 mice per group, and each experiment is repeated once (Table 3).
  • Example 8 Construction of an Ad vector encoding the HAl domain of the A/Puerto Rico/8/34 influenza virus and evaluation of HAl as a drug- vaccine multipurpose agent.
  • AdPR8.Hl.l An RCA-free Ad vector (AdPR8.Hl.l) encoding a humanized HAl domain of the A/Puerto Rico/8/34 influenza virus is generated in PER.C6 cells as described in Example 2. Mice are immunized by intranasal instillation of an escalating dose of AdPR ⁇ .Hl.l vectors encoding the HAl domain of the A/Puerto Rico/8/34 (HlNl) influenza virus, or AdNCHl.1 vectors encoding the HAl domain of the A/New Caledonia/20/99 (HlNl) influenza virus, in a single-dose regimen.
  • mice After one month, control and immunized mice will be challenged by A/Puerto Rico/8/34 (HlNl) and A/Hong Kong/8/68 (H3N2) influenza virus strains, respectively. All mice are primed and challenged as described above. There are 10 mice per group, and each experiment is repeated once (Table 4).
  • a therapeutic-immunological composition comprising a pathogen-derived ligand, wherein (i) the ligand binds to its receptor on target cells;
  • the therapeutic-immunological composition of claim 1 wherein the virus- derived ligand is expressed by a viral vector.
  • the viral vector is selected from the group consisting of an adenovirus vector, an adeno-associated virus vector, a herpes simplex virus vector, an alphavirus vector, and a retrovirus vector.
  • adenovirus vector is derived from adenovirus serotype 5 (Ad5).
  • Ad5 adenovirus serotype 5
  • the adenovirus vector is selected from the group consisting of replication-defective adenovirus, non-replicating adenovirus, replication-competent adenovirus, or wild-type adenovirus.
  • SARS-CoV severe acute respiratory syndrome-associated coronavirus
  • HRV human rhinovirus
  • RSV respiratory syncytial virus
  • hemagglutinin protein is selected from the group consisting of hemagglutinin subtype 1, 2, 3, 5, or B.
  • hemagglutinin protein is hemagglutinin HAl domain.
  • HAl is derived from a strain selected from the group consisting of A/New Caledonia/20/99, A/Panama/2007/99, B/Hong Kong/330/01, A/Vietnam/I 203/04, A/Puerto Rico/8/34, A/Hong Kong/8/68, and 2009H1N1 swine flu.
  • the pathogen-derived ligand binds to an angiotensin-converting enzyme 2 receptor.
  • a method of conferring in a subject rapid protection against a virus and eliciting in the subject long-term protective immunity against the virus comprising: intranasally administering to the subject a therapeutically effective amount of the immunological composition of claim 1 or 2.
  • the viral vector is selected from the group consisting of an adenovirus vector, an adeno-associated virus vector, a herpes simplex virus vector an alphavirus vector, and a retrovirus vector.
  • recombinant viral vector is a recombinant adenovirus vector.
  • adenovirus vector is derived from adenovirus serotype 5 (Ad5).
  • adenovirus vector is selected from the group consisting of replication-defective adenovirus, non-replicating adenovirus, replication- competent adenovirus, or wild-type adenovirus.
  • pathogen from which the ligand is derived is selected from the group consisting of influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS-CoV), human rhinovirus (HRV), and respiratory syncytial virus (RSV).
  • SARS-CoV severe acute respiratory syndrome-associated coronavirus
  • HRV human rhinovirus
  • RSV respiratory syncytial virus
  • pathogen is influenza virus.
  • pathogen-derived ligand binds to a sialic acid-containing receptor.
  • the pathogen-derived ligand is hemagglutinin protein.
  • the hemagglutinin protein is selected from the group consisting of hemagglutinin subtype 1, 2, 3, 5, or B.
  • hemagglutinin protein is hemagglutinin HAl domain.
  • HAl is derived from a strain selected from the group consisting of A/New Caledonia/20/99, A/Panama/2007/99, B/Hong Kong/330/01 ,
  • Tetracycline resistance determined by pBR322 is mediated by one polypeptide. Gene 26, 197-203. Beigel, J., and Bray, M. (2008). Current and future antiviral therapy of severe seasonal and avian influenza. Antiviral Res 78, 91-102.
  • Point mutations define a sequence flanking the AUG initiator codon that modulates translation by eukaryotic ribosomes.
  • Cell 44, 283-292. Malhotra, R., Ward, M., Bright, H., Priest, R., Foster, M. R., Hurle, M., Blair, E., and Bird,
  • a replication-defective human adenovirus recombinant serves as a highly efficacious vaccine carrier.
  • Neuraminidase inhibitor-resistant recombinant A/Vietnam/ 1203/04 (H5N1) influenza viruses retain their replication efficiency and pathogenicity in vitro and in vivo. J
  • AdEasy system made easier by selecting the viral backbone plasmid preceding homologous recombination. Biotechniques 31, 260-262.

Abstract

The present invention relates to a receptor-binding ligand either administered intranasally or expressed following intranasal administration from a recombinant vector that expresses at least one therapeutic ligand which binds to a receptor necessary for infection by a respiratory pathogen. The receptor-binding ligand in the respiratory tract not only confers rapid protection as a therapeutic drug but also elicits long-term protective immunity as a vaccine against a respiratory pathogen. This regimen is not compromised by the emergence of drug resistance because the receptor for any pathogen can be blocked by the pathogen's own receptor-binding ligand.

Description

TITLE OF THE INVENTION
INTRANASAL ADMINISTRATION OF RECEPTOR-BINDING LIGANDS OR GENES
ENCODING SUCH LIGANDS AS A THERAPEUTIC REGIMEN FOR MITIGATING
INFECTIONS CAUSED BY RESPIRATORY PATHOGENS
INCORPORATION BY REFERENCE
This application claims benefit of U.S. Provisional Application No. 61/058,413, filed June 3, 2008.
The foregoing application, and all documents cited therein or during their prosecution ("appln cited documents") and all documents cited or referenced in the appln cited documents, and all documents cited or referenced herein ("herein cited documents"), and all documents cited or referenced in herein cited documents, together with any manufacturer's instructions, descriptions, product specifications, and product sheets for any products mentioned herein or in any document incorporated by reference herein, are hereby incorporated herein by reference, and may be employed in the practice of the invention. FIELD OF THE INVENTION
The present invention relates generally to the fields of therapeutic drug, vaccinology, and vector technology. The present invention also relates to methods of non-invasive immunization in an animal, products therefrom and uses for the methods and products therefrom.
BACKGROUND OF THE INVENTION
The increased susceptibility to influenza due to human population growth, the emergence of lethal avian influenza strains (Subbarao et al., 1998), and the potential use of designer influenza virus as a bioweapon (Hoffmann et al., 2002; Neumann et al., 1999) collectively highlight a need for development of rapid-response countermeasures against influenza. Although annual vaccination is effective in protecting people against seasonal influenza (Nichol et al., 1995), it is difficult to produce sufficient amounts of an effective vaccine using the egg-based system to fully respond to a surge in demand, particularly during the first wave of an avian influenza pandemic. In addition, even seasonal influenza can be life-threatening to infants, the elderly, and immunodeficient subjects. An under-developed line of defense against influenza is the use of anti-influenza drugs. The licensed anti- influenza drugs (the M2 ion channel blockers, amantadine and rimantadine, and the neuraminidase inhibitors, oseltamivir (Tamiflu) and zanamivir (Relenza)) are beneficial for seasonal influenza, but appropriate dosing regimens for avian influenza have not been established (Beigel and Bray, 2008). Moreover, these anti-influenza drugs may generate drug-resistant influenza virus strains over time (Aoki et al., 2007; Beigel and Bray, 2008; Moscona, 2008; Reece, 2007; Yen et al., 2007).
Even when anti-influenza drugs are available, vaccination is still the most cost- effective regimen against influenza. The contemporary clinically licensed influenza vaccines for all age groups consist of trivalent inactivated viruses (TFV) that have been administered intramuscularly (Pfleiderer et al., 2001). The annual fall inoculations using TIV are effective in protecting people against influenza (Nichol et al., 1995). However, the necessity of intramuscular injection by licensed medical personnel is associated with a series of problems, including limited supply of medical personnel qualified to perform hypodermic injections, fear of pain from needle injections amongst target population, transmission of bloodborne pathogens by contaminated and reused needles, and necessity to dispose of contaminated needles as medical waste. It is conceivable that a great number of people will not have access to adequate medical services if a pandemic influenza outbreak should occur.
Recently, a live attenuated influenza virus vaccine (FluMist), which is administered directly to the respiratory tract by nasal spray, has been developed as a needle-free alternative for influenza vaccination (Nolan et al., 2003). Both inactivated and live attenuated influenza vaccines are currently produced in embryonated chicken eggs (Hilleman, 2002). However, the presence of chicken pathogens in eggs in FluMist is a biohazard (Hilleman, 2002). Potentially harmful reassortments generated by recombination between live attenuated and wild influenza viruses present additional biohazardous concerns (Hilleman, 2002). Further, adverse side effects associated with intranasal inoculation of live attenuated influenza vaccine exceed mild runny nose, sore throat, or low-grade fever (Marwick, 2000), and can include destruction of epithelial cells in the upper respiratory tract which can lead to secondary infections with pulmonary complications (Hilleman, 2002; Marwick, 2000). Another biohazard is shedding of the replicating attenuated influenza virus to bystanders. Some of these unwanted side effects could be minimized or even eliminated if an egg- and syringe needle-independent non-replicating influenza vaccine is developed.
Citation or identification of any document in this application is not an admission that such document is available as prior art to the present invention. SUMMARY OF THE INVENTION AND DETAILED DESCRIPTION
The present invention relates to a prophylactic-therapeutic multipurpose agent that will provide an efficacious, safe, and rapid-response countermeasure against respiratory pathogens. In particular, the present invention relates to a therapeutic as well as immunological composition comprising a pathogen-derived ligand that binds to its receptor on target cells and (1) confers in a subject a rapid, therapeutic effect and (2) may also elicit in the subject long-term protective immunity against the pathogen. The present invention also relates to a method of nasal administration of the immunological composition.
In embodiments of the present invention, the pathogen-derived ligand may be expressed by a viral vector. Viral vector gene delivery systems are well-known in the field and are commonly used as a tool for use in gene transfer and gene therapy applications (Stone, 2000). hi one embodiment the viral vector may include but is not limited to a retrovirus, adenovirus, adeno-associated virus, alphavirus, or herpes simplex virus, hi another embodiment, the viral vector is a recombinant adenoviral vector. In a preferred embodiment the recombinant adenoviral vector may be derived from adenovirus serotype 5 (Ad5). In other embodiments, the recombinant adenoviral vector may be selected from the group consisting of replication-defective adenovirus, non-replicating adenovirus, replication- competent adenovirus, or wild-type adenovirus.
In addition to virus-derived vectors, the pathogen-derived ligand may also be delivered as purified proteins, or nucleic acids, or virus-like particles.
The viral vector may further comprise a promoter sequence selected from the group consisting of viral promoters, avian promoters, CMV promoter, SV40 promoter, β-actin promoter, albumin promoter, Elongation Factor 1-α (EF 1-α) promoter, PγK promoter, MFG promoter, pIX, or Rous sarcoma virus promoter. In certain embodiments, the pathogen-derived ligand may be a therapeutic ligand, antigen, or immunogen. In some embodiments, the pathogen-derived ligand may bind to a receptor that is necessary for infection by a respiratory pathogen.
In some embodiments, the therapeutic ligand binds to a receptor that is necessary for infection by a respiratory pathogen such as influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS-CoV), human rhinovirus (HRV), or respiratory syncytial virus (RSV).
In embodiments in which the respiratory pathogen may be influenza virus, the therapeutic ligand may bind to a sialic acid-containing receptor, which thereby prevents the influenza virus from binding to the receptor. In some embodiments, the therapeutic ligand may be a hemagglutinin protein, such as hemagglutinin subtype 1, 2, 3, 5, or B. In further embodiments, the therapeutic ligand is hemagglutinin HAl domain or an HA subfragment containing a receptor-binding ligand, for example, HAl derived from A/New Caledonia/20/99, A/Panama/2007/99, B/Hong Kong/330/01, A/Vietnam/ 1203/04, A/Puerto Rico/8/34, A/Hong Kong/8/68, or 2009H1N1 swine flu virus. In other embodiments, the therapeutic ligand may be a nucleoprotein, matrix, or neuraminidase.
In embodiments in which the respiratory pathogen may be SARS-CoV, the therapeutic ligand may bind to an angiotensin-converting enzyme 2 receptor. In certain embodiments, the therapeutic ligand may be a SARS-CoV spike (S) protein. In other embodiments, the therapeutic ligand may be a SARS-CoV polyproteinl A and IB, envelope protein E, matrix protein M, or nucleocapsid protein N.
In another embodiment wherein the respiratory pathogen may be HRV, the therapeutic ligand may bind to inter-cellular adhesion molecule 1. In other embodiments wherein the respiratory pathogen may be RSV, the therapeutic ligand may bind to annexin II.
The present invention also relates to a composition. The composition may comprise a viral vector as described above. Moreover, the composition may be delivered intranasally. The present invention relates to a composition such as a vaccine, pharmaceutical, or immunological composition, for in vivo delivery to a subject, comprising a pharmaceutically acceptable carrier or excipient and a pathogen-derived ligand that binds to its receptor on target cells, confers in the subject a rapid, therapeutic effect, and elicits in the animal long- term protective immunity against the pathogen. The present invention also relates to a viral vector that contains and expresses a nucleic acid molecule having a sequence encoding a pathogen-derived ligand. The present invention further relates to immunological compositions containing viral vectors, hi certain embodiments, the composition is administered intranasally.
Accordingly, it is an object of the invention to not encompass within the invention any previously known product, process of making the product, or method of using the product such that Applicants reserve the right and hereby disclose a disclaimer of any previously known product, process, or method. It is further noted that the invention does not intend to encompass within the scope of the invention any product, process, or making of the product or method of using the product, which does not meet the written description and enablement requirements of the USPTO (35 U.S.C. §112, first paragraph) or the EPO (Article 83 of the EPC), such that Applicants reserve the right and hereby disclose a disclaimer of any previously described product, process of making the product, or method of using the product.
It is noted that in this disclosure and particularly in the claims and/or paragraphs, terms such as "comprises", "comprised", "comprising" and the like can have the meaning attributed to it in U.S. Patent law; e.g., they can mean "includes", "included", "including", and the like; and that terms such as "consisting essentially of and "consists essentially of have the meaning ascribed to them in U.S. Patent law, e.g., they allow for elements not explicitly recited, but exclude elements that are found in the prior art or that affect a basic or novel characteristic of the invention. These and other embodiments are disclosed or are obvious from and encompassed by, the following Detailed Description. BRIEF DESCRIPTION OF FIGURES
The following Detailed Description, given by way of example, but not intended to limit the invention to specific embodiments described, may be understood in conjunction with the accompanying Figures, incorporated herein by reference, in which:
Figure 1 shows protection of mice against influenza virus by intranasal instillation of an Ad vector encoding HAl shortly before challenge.
Figure 2 shows protection of mice against influenza virus by intranasal instillation of an Ad vector encoding HAl shortly after challenge. Figure 3 shows protection of mice against HPAI virus by Ad- vectored nasal vaccines.
Figure 4 shows protection of ferrets against HPAI virus by Ad- vectored nasal vaccines.
DETAILED DESCRIPTION
The present invention relates to (1) a therapeutic as well as immunological composition comprising a pathogen-derived ligand that binds to its receptor on target cells, confers in a subject a rapid, therapeutic effect, and may also elicit in the subject long-term protective immunity against the pathogen, and (2) a viral vector, preferably a recombinant adenoviral vector, that contains and expresses in vivo a nucleic acid molecule having a sequence encoding a pathogen-derived ligand, and (3) a method of conferring in a subject a rapid therapeutic effect against a pathogen and may also elicit in the subject long-term protective immunity against the pathogen, comprising intranasally administering to the subject a therapeutically effective amount of the therapeutic-immunological composition..
The term "nucleic acid" or "nucleic acid sequence" refers to a deoxyribonucleic or ribonucleic oligonucleotide in either single- or double-stranded form. The term encompasses nucleic acids, i.e., oligonucleotides, containing known analogues of natural nucleotides. The term also encompasses nucleic-acid-like structures with synthetic backbones, see e.g., Eckstein, 1991; Baserga et al., 1992; Milligan, 1993; WO 97/03211; WO 96/39154; Mata, 1997; Strauss-Soukup, 1997; and Samstag, 1996. As used herein, "recombinant" refers to a polynucleotide synthesized or otherwise manipulated in vitro (e.g., "recombinant polynucleotide"), to methods of using recombinant polynucleotides to produce gene products in cells or other biological systems, or to a polypeptide ("recombinant protein") encoded by a recombinant polynucleotide. "Recombinant means" encompasses the ligation of nucleic acids having various coding regions or domains or promoter sequences from different sources into an expression cassette or vector for expression of, e.g., inducible or constitutive expression of polypeptide coding sequences in the vectors of invention.
The term "heterologous" when used with reference to a nucleic acid, indicates that the nucleic acid is in a cell or a virus where it is not normally found in nature; or, comprises two or more subsequences that are not found in the same relationship to each other as normally found in nature, or is recombinantly engineered so that its level of expression, or physical relationship to other nucleic acids or other molecules in a cell, or structure, is not normally found in nature. A similar term used in this context is "exogenous". For instance, a heterologous nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged in a manner not found in nature; e.g., a human gene operably linked to a promoter sequence inserted into an adenovirus-based vector of the invention. As an example, a heterologous nucleic acid of interest can encode an immunogenic gene product, wherein the adenovirus is administered therapeutically or prophylactically as a carrier or drug- vaccine composition. Heterologous sequences can comprise various combinations of promoters and sequences, examples of which are described in detail herein.
A "therapeutic ligand" may be a substance which can bind to a receptor of a target cell with therapeutic effects.
A "therapeutic effect" may be a consequence of a medical treatment of any kind, the results of which are judged by one of skill in the filed to be desirable and beneficial. The
"therapeutic effect" may be a behavioral or physiologic change which occurs as a response to the medical treatment. The result may be expected, unexpected, or even an unintended consequence of the medical treatment. A "therapeutic effect" may include, for example, a reduction of symptoms in a subject suffering from infection by a pathogen. A "target cell" may be a cell in which an alteration in its activity can induce a desired result or response.
An "antigen" may be a substance that is recognized by the immune system and induces an immune response. An "irnmunogen" may be a substance that elicits an immune response from the immune system.
A "ligand" may be any substance that binds to and forms a complex with a biomolecule to serve a biological purpose. As used herein, "ligand" may also refer to an "antigen" or "immunogen". As used herein "antigen" and "immunogen" are used interchangeably.
As used herein, a "pathogen" may refer to a viral pathogen (e.g. virus) or a bacterial pathogen. "Pathogen" also encompasses "respiratory pathogens". "Expression" of a gene or nucleic acid encompasses not only cellular gene expression, but also the transcription and translation of nucleic acid(s) in cloning systems and in any other context.
As used herein, a "vector" is a tool that allows or facilitates the transfer of an entity from one environment to another. By way of example, some vectors used in recombinant DNA techniques allow entities, such as a segment of DNA (such as a heterologous DNA segment, such as a heterologous cDNA segment), to be transferred into a target cell. The present invention comprehends recombinant vectors that can include viral vectors, bacterial vectors, protozoan vectors, DNA vectors, or recombinants thereof.
With respect to exogenous DNA for expression in a vector (e.g., encoding an epitope of interest and/or an antigen and/or a therapeutic) and documents providing such exogenous DNA, as well as with respect to the expression of transcription and/or translation factors for enhancing expression of nucleic acid molecules, and as to terms such as "epitope of interest", "therapeutic", "immune response", "immunological response", "protective immune response", "immunological composition", "immunogenic composition", and "vaccine composition", inter alia, reference is made to U.S . Patent No. 5,990,091 issued November 23, 1999, and WO 98/00166 and WO 99/60164, and the documents cited therein and the documents of record in the prosecution of that patent and those PCT applications; all of which are incorporated herein by reference. Thus, U.S. Patent No. 5,990,091 and WO 98/00166 and WO 99/60164 and documents cited therein and documents of record in the prosecution of that patent and those PCT applications, and other documents cited herein or otherwise incorporated herein by reference, can be consulted in the practice of this invention; and, all exogenous nucleic acid molecules, promoters, and vectors cited therein can be used in the practice of this invention. In this regard, mention is also made of U.S. Patents Nos. 6,706,693; 6,716,823; 6,348,450; U.S. Patent Application Serial Nos. 10/424,409; 10/052,323; 10/116,963; 10/346,021; and WO 99/08713, published February 25, 1999, from PCT/US98/16739.
As used herein, the terms "drug composition" and "drug", "vaccinal composition" and "vaccine" and "vaccine composition" and "drug-vaccine composition" and "drug-vaccine dual agent" and "therapeutic composition" and "therapeutic-immunologic composition" cover any composition that induces protection against a pathogen. In some embodiments, the protection may be due to an inhibition or prevention of infection by a pathogen. In other embodiments, the protection may be induced by an immune response against the antigen(s) of interest, or which efficaciously protects against the antigen; for instance, after administration or injection into the subject, elicits a protective immune response against the targeted antigen or immunogen or provides efficacious protection against the antigen or immunogen expressed from the inventive adenovirus vectors of the invention. The term "pharmaceutical composition" means any composition that is delivered to a subject. In some embodiments, the composition may be delivered to inhibit or prevent infection by a pathogen. The terms "immunogenic composition" and "immunological composition" and
"immunogenic or immunological composition" cover any composition that confers in a subject a therapeutic effect and/or elicits in a subject an immune response against the antigen, immunogen, or pathogen of interest; for instance, after administration into a subject, elicits an immune response against the targeted immunogen or antigen of interest. An "immunological response" to a composition, vaccine, antigen, immunogen, pathogen or ligand is the development in the host of a cellular and/or antibody-mediated immune response to the composition, vaccine, antigen, immunogen, pathogen or ligand interest. Usually, an "immunological response" includes but is not limited to one or more of the following effects: the production of antibodies, B cells, helper T cells, and/or cytotoxic T cells, directed specifically to an antigen or antigens included in the composition or vaccine of interest. Preferably, the host will display both a rapid (e.g. within <24 hrs.) therapeutic effect and a long-term protective immunological response such that resistance to new infection will be enhanced and/or the clinical severity of the disease reduced. Such protection will be demonstrated by either a reduction or lack of symptoms normally displayed by an infected host, a quicker recovery time and/or a lowered viral titer in the infected host.
A "therapeutically effective amount" or an "immunologically effective amount" is an amount or concentration of the recombinant vector encoding the gene of interest, that, when administered to a subject, produces a therapeutic response or an immune response to the gene product of interest. A "circulating recombinant form" refers to recombinant viruses that have undergone genetic reassortment among two or more subtypes or strains. Other terms used in the context of the present invention is "hybrid form", "recombined form", and "reassortant form".
"Clinical isolates" refer to viruses or microbes isolated from infected subjects in a clinical setting.
"Field isolates" refer to viruses or microbes that are isolated from infected subjects or from the environment.
The methods of the invention can be appropriately applied to prevent diseases as prophylactic vaccination or provide relief against symptoms of disease as therapeutic treatments.
The recombinant vectors of the present invention can be administered to a subject either alone or as part of pharmaceutical or immunological or immunogenic composition. The recombinant vectors of the invention can also be used to deliver or administer one or more proteins to a subject of interest by in vivo expression of the protein(s). It is noted that expressed and/or immunological products and/or antibodies obtained in accordance with this invention can be expressed in vitro and used in a manner in which such expressed and/or immunological products and/or antibodies are typically used. It is also noted that cells that express such immunological products and/or antibodies and/or drugs can be employed in in vitro and/or ex vivo applications, e.g., such uses and applications can include diagnostics, assays, ex vivo therapy (e.g., wherein cells that express the gene product and/or immunological response are expanded in vitro and reintroduced into the host or animal), etc., see U.S. Patent No. 5,990,091, WO 99/60164 and WO 98/00166 and documents cited therein. Further, expressed gene products or antibodies that are isolated from herein methods, or that are isolated from cells expanded in vitro following herein administration methods, can be administered in compositions, akin to the administration of therapeutics or subunit epitopes or antigens or antibodies to induce protective immunity and/or confer therapeutic effects, stimulate a therapeutic response and/or stimulate adaptive immunity. The term "viral vector" as used herein includes but is not limited to retroviruses, adenoviruses, adeno-associated viruses, alphavirus, and herpes simplex virus. The term "human adenovirus" as used herein is intended to encompass all human adenoviruses of the Adenoviridae family, which include members of the Mastadenovirus genera. To date, over fifty-one human serotypes of adenoviruses have been identified (see, e.g., Fields et al., Virology 2, Ch. 67 (3d ed., Lippincott-Raven Publishers)). The adenovirus can be of serogroup A, B, C, D, E, or F. The human adenovirus can be a serotype 1 (AdI), serotype 2 (Ad2), serotype 3 (Ad3), serotype 4 (Ad4), serotype 6 (Ad6), serotype 7 (Ad7), serotype 8 (Ad8), serotype 9 (Ad9), serotype 10 (AdIO), serotype 11 (AdI 1), serotype 12 (Adl2), serotype 13 (AdB), serotype 14 (Adl4), serotype 15 (Adl5), serotype 16 (Adl6), serotype 17 (Adl7), serotype 18 (Adl8), serotype 19 (Adl9), serotype 19a (Adl9a), serotype 19ρ (AdI 9p), serotype 20 (Ad20), serotype 21 (Ad21), serotype 22 (Ad22), serotype 23 (Ad23), serotype 24 (Ad24), serotype 25 (Ad25), serotype 26 (Ad26), serotype 27 (Ad27), serotype 28 (Ad28), serotype 29 (Ad29), serotype 30 (Ad30), serotype 31 (Ad31), serotype 32 (Ad32), serotype 33 (Ad33), serotype 34 (Ad34), serotype 35 (Ad35), serotype 36 (Ad36), serotype 37 (Ad37), serotype 38 (Ad38), serotype 39 (Ad39), serotype 40 (Ad40), serotype 41 (Ad41), serotype 42 (Ad42), serotype 43 (Ad43), serotype 44 (Ad44), serotype 45 (Ad45), serotype 46 (Ad46), serotype 47 (Ad47), serotype 48 (Ad48), serotype 49 (Ad49), serotype 50 (Ad50), serotype 51 (Ad51), or preferably, serotype 5 (Ad5), but are not limited to these examples.
Also contemplated by the present invention are receptor-binding ligands, recombinant vectors, drug- vaccine compositions, and recombinant adenoviruses that can comprise subviral particles from more than one adenovirus serotype. For example, it is known that adenovirus vectors can display an altered tropism for specific tissues or cell types (Havenga, M.J.E. et al., 2002), and therefore, mixing and matching of different adenoviral capsids, i.e., fiber, or penton proteins from various adenoviral serotypes may be advantageous. Modification of the adenoviral capsids, including fiber and penton can result in an adenoviral vector with a tropism that is different from the unmodified adenovirus. Adenovirus vectors that are modified and optimized in their ability to infect target cells can allow for a significant reduction in the therapeutic or prophylactic dose, resulting in reduced local and disseminated toxicity. Viral vector gene delivery systems are commonly used in gene transfer and gene therapy applications. Different viral vector systems have their own unique advantages and disadvantages. Viral vectors that may be used to express the pathogen-derived ligand of the present invention include but are not limited to adenoviral vectors, adeno-associated viral vectors, alphavirus vector, herpes simplex viral vector, and retroviral vectors, described in more detail below.
Adenovirus vectors have many characteristics which are ideal for gene delivery, especially delivery into the respiratory tract. Examples of these characteristics include:
1) ability of adenovirus vectors to transduce both mitotic and postmitotic cells in situ; 2) existing technology to prepare stocks containing high titers of virus [greater than 1012 ifu (infectious units) per ml] to transduce cells in situ at high multiplicity of infection (MOI);
3) inhalation of adenovirus is in compliance with evolutionary medicine (Tang and Van Kampen, 2008);
4) potency of an intranasally-administered adenovirus vector may not be interfered by preexisting immunity to adenovirus (Hoelscher et al., 2006; Shi et al., 2001; Van Kampen et al., 2005); while not wishing to be bound by theory, this may be attributed to the high efficiency of gene delivery, high level of transgene expression, and high degree of antigen presentation along the mucosal barrier in the respiratory tract;
5) capability of adenovirus to induce high levels of transgene expression (at least as an initial burst); and
6) ease with which replication-defective adenovirus vectors can be bioengineered. Additional general features of adenoviruses are that the biology of the adenovirus is characterized in detail; the adenovirus is not associated with severe human pathology; the adenovirus is extremely efficient in introducing its DNA into the host cell; the adenovirus can infect a wide variety of cells and has a broad host range; the adenovirus can be produced in large quantities with relative ease; and the adenovirus can be rendered replication defective and/or non-replicating by deletions in the early region 1 ("El ") of the viral genome. Adenovirus is a non-enveloped DNA virus. The genome of adenovirus is a linear double-stranded DNA molecule of approximately 36,000 base pairs ("bp") with a 55-kDa terminal protein covalently bound to the 5 '-terminus of each strand. The adenovirus DNA contains identical inverted terminal repeats ("ITRs") of about 100 bp, with the exact length depending on the serotype. The viral origins of replication are located within the ITRs exactly at the genome ends. DNA synthesis occurs in two stages. First, replication proceeds by strand displacement, generating a daughter duplex molecule and a parental displaced strand. The displaced strand is single stranded and can form a "panhandle" intermediate, which allows replication initiation and generation of a daughter duplex molecule. Alternatively, replication may proceed from both ends of the genome simultaneously, obviating the requirement to form the panhandle structure.
During the productive infection cycle, the viral genes are expressed in two phases: the early phase, which is the period up to viral DNA replication, and the late phase, which coincides with the initiation of viral DNA replication. During the early phase, only the early gene products, encoded by regions El, E2, E3 and E4, are expressed, which carry out a number of functions that prepare the cell for synthesis of viral structural proteins (Berk, A. J., 1986). During the late phase, the late viral gene products are expressed in addition to the early gene products and host cell DNA and protein synthesis are shut off. Consequently, the cell becomes dedicated to the production of viral DNA and of viral structural proteins (Tooze, J., 1981).
The El region of adenovirus is the first region of adenovirus expressed after infection of the target cell. This region consists of two transcriptional units, the ElA and ElB genes, both of which are required for oncogenic transformation of primary (embryonal) rodent cultures. The main functions of the ElA gene products are to induce quiescent cells to enter the cell cycle and resume cellular DNA synthesis, and to transcriptionally activate the ElB gene and the other early regions (E2, E3 and E4) of the viral genome. Transfection of primary cells with the ElA gene alone can induce unlimited proliferation (immortalization), but does not result in complete transformation. However, expression of ElA, in most cases, results in induction of programmed cell death (apoptosis), and only occasionally is immortalization obtained (Jochemsen et al., 1987). Co-expression of the ElB gene is required to prevent induction of apoptosis and for complete morphological transformation to occur. In established immortal cell lines, high-level expression of ElA can cause complete transformation in the absence of ElB (Roberts, B. E. et al., 1985).
The ElB encoded proteins assist El A in redirecting the cellular functions to allow viral replication. The ElB 55 kD and E4 33 kD proteins, which form a complex that is essentially localized in the nucleus, function in inhibiting the synthesis of host proteins and in facilitating the expression of viral genes. Their main influence is to establish selective transport of viral mRNAs from the nucleus to the cytoplasm, concomitantly with the onset of the late phase of infection. The ElB 21 kD protein is important for correct temporal control of the productive infection cycle, thereby preventing premature death of the host cell before the virus life cycle has been completed. Mutant viruses incapable of expressing the ElB 21 kD gene product exhibit a shortened infection cycle that is accompanied by excessive degradation of host cell chromosomal DNA (deg-phenotype) and in an enhanced cytopathic effect (cyt-phenotype; Telling et al., 1994). The deg and cyt phenotypes are suppressed when in addition the ElA gene is mutated, indicating that these phenotypes are a function of ElA (White, E. et al., 1988). Furthermore, the ElB 21 kDa protein slows down the rate by which ElA switches on the other viral genes. It is not yet known by which mechanisms ElB 21 kD quenches these ElA dependent functions. In contrast to, for example, retroviruses, adenoviruses do not efficiently integrate into the host cell's genome, are able to infect non-dividing cells, and are able to efficiently transfer recombinant genes in vivo (Brady et al., 1994). These features make adenoviruses attractive candidates for in vivo gene transfer of, for example, an antigen or immunogen of interest into cells, tissues or subjects in need thereof.
Adenovirus vectors containing multiple deletions are preferred to both increase the carrying capacity of the vector and reduce the likelihood of recombination to generate replication competent adenovirus (RCA). Where the adenovirus contains multiple deletions, it is not necessary that each of the deletions, if present alone, would result in a replication defective and/or non-replicating adenovirus. As long as one of the deletions renders the adenovirus replication defective or non-replicating, the additional deletions may be included for other purposes, e.g., to increase the carrying capacity of the adenovirus genome for heterologous nucleotide sequences. Preferably, more than one of the deletions prevents the expression of a functional protein and renders the adenovirus replication defective and/or non-replicating and/or attenuated. More preferably, all of the deletions are deletions that would render the adenovirus replication-defective and/or non-replicating and/or attenuated. However, the invention also encompasses adenovirus and adenovirus vectors that are replication competent and/or wild-type, i.e. comprises all of the adenoviral genes necessary for infection and replication in a subject. Embodiments of the invention employing adenovirus recombinants may include El- defective or deleted, or E3 -defective or deleted, or E4-defective or deleted or adenovirus vectors comprising deletions of El and E3, or El and E4, or E3 and E4, or El, E3, and E4 deleted, or the "gutless" adenovirus vector in which all viral genes are deleted. The adenovirus vectors can comprise mutations in El, E3, or E4 genes, or deletions in these or all adenoviral genes. The El mutation raises the safety margin of the vector because El- defective adenovirus mutants are said to be replication-defective and/or non-replicating in non-permissive cells, and are, at the very least, highly attenuated. The E3 mutation enhances the immunogenicity of the antigen by disrupting the mechanism whereby adenovirus down- regulates MHC class I molecules. The E4 mutation reduces the immunogenicity of the adenovirus vector by suppressing the late gene expression, thus may allow repeated re- vaccination utilizing the same vector. The present invention comprehends adenovirus vectors of any serotype or serogroup that are deleted or mutated in El, or E3, or E4, or El and E3, or El and E4. Deletion or mutation of these adenoviral genes result in impaired or substantially complete loss of activity of these proteins. The "gutless" adenovirus vector is another type of vector in the adenovirus vector family. Its replication requires a helper virus and a special human 293 cell line expressing both EIa and Cre, a condition that does not exist in natural environment; the vector is deprived of all viral genes, thus the vector as a vaccine carrier is non-immunogenic and may be inoculated multiple times for re-vaccination. The "gutless" adenovirus vector also contains 36 kb space for accommodating antigen or immunogen(s) of interest, thus allowing co-delivery of a large number of antigen or immunogens into cells.
Other adenovirus vector systems known in the art include the AdEasy system (He et al., 1998) and the subsequently modified AdEasier system (Zeng et al., 2001), which were developed to generate recombinant Ad vectors in 293 cells rapidly by allowing homologous recombination between donor vectors and Ad helper vectors to occur in Escherichia coli cells, such as BJ5183 cells, overnight. pAdEasy comprises adenoviral structural sequences that, when supplied in trans with a donor vector such as pShuttle-CMV expressing an antigen or immunogen of interest, results in packaging of the antigen or immunogen (e.g., immunogens and/or antigens) in an adenoviral capsid. The sequence of p AdEasy is well known in the art and is publicly and commercially available through Stratagene.
The present invention can be generated using the AdHigh system (U.S. Patent Application Serial No. 11/943,901, incorporated herein by reference) (Tang et al., 2009). AdHigh is a safe, rapid, and efficient method of generating high titers of recombinant adenovirus without the risk of generating RCA, which may be detrimental or fatal to subjects. The AdHigh system uses modified shuttle plasmids, such as pAdHigh, to promote the production of RCA-free adenoviruses in permissive cells, such as PER.C6 cells after generating recombinants with an adenovirus backbone plasmid in E. coli cells. These shuttle plasmids contain polylinkers or multiple cloning sites for easy insertion of antigens such as, for example, influenza immunogens or antigens. Recombination of the adenoviral shuttle plasmids in conjunction with an adenoviral helper plasmid such as pAdEasy in bacterial cells (i.e., BJ5183) can be easily implemented to produce the recombinant human adenoviruses expressing antigens or immunogens of the invention. Methods of producing recombinant vectors by cloning and restriction analysis are well known to those skilled in the art. Specific sequence motifs such as the RGD motif may be inserted into the H-I loop of an adenovirus vector to enhance its infectivity. This sequence has been shown to be essential for the interaction of certain extracellular matrix and adhesion proteins with a superfamily of cell-surface receptors called integrins. Insertion of the RGD motif may be advantageously useful in immunocompromised subjects. An adenovirus recombinant is constructed by cloning specific antigen or immunogen or fragments thereof into any of the adenovirus vectors such as those described above. The adenovirus recombinant is used to transduce cells of a vertebrate use as an immunizing agent. (See, for example, U.S. Patent Application Ser. No. 10/424,409, incorporated by reference). Adeno-associated virus (AAV) is a single-stranded DNA parvovirus which is endogenous to the human population. Although capable of productive infection in cells from a variety of species, AAV is a dependovirus, requiring helper functions from either adenovirus or herpes virus for its own replication. In the absence of helper functions from either of these helper viruses, AAV will infect cells, uncoat in the nucleus, and integrate its genome into the host chromosome, but will not replicate or produce new viral particles.
The genome of AAV has been cloned into bacterial plasmids and is well characterized. The viral genome consists of 4682 bases which include two terminal repeats of 145 bases each. These terminal repeats serve as origins of DNA replication for the virus. Some investigators have also proposed that they have enhancer functions. The rest of the genome is divided into two functional domains. The left portion of the genome codes for the rep functions which regulate viral DNA replication and vital gene expression. The right side of the vital genome contains the cap genes that encode the structural capsid proteins VPl, VP2 and VP3. The proteins encoded by both the rep and cap genes function in trans during productive AAV replication. AAV is considered an ideal candidate for use as a transducing vector , and it has been used in this manner. Such AAV transducing vectors comprise sufficient cis-acting functions to replicate in the presence of adenovirus or herpes virus helper functions provided in trans. Recombinant AAV (rAAV) have been constructed in a number of laboratories and have been used to carry exogenous genes into cells of a variety of lineages. In these vectors , the AAV cap and/or rep genes are deleted from the viral genome and replaced with a DNA segment of choice. Current vectors can accommodate up to 4300 bases of inserted DNA.
To produce rAAV, plasmids containing the desired vital construct are transfected into adenovirus-infected cells. In addition, a second helper plasmid is cotransfected into these cells to provide the AAV rep and cap genes which are obligatory for replication and packaging of the recombinant viral construct. Under these conditions, the rep and cap proteins of AAV act in trans to stimulate replication and packaging of the rAAV construct. Three days after transfection, rAAV is harvested from the cells along with adenovirus. The contaminating adenovirus is then inactivated by heat treatment. Herpes Simplex Virus 1 (HSV-I) is an enveloped, double-stranded DNA virus with a genome of 153 kb encoding more than 80 genes. Its wide host range is due to the binding of viral envelope glycoproteins to the extracellular heparin sulphate molecules found in cell membranes (WuDunn & Spear, 1989). Internalization of the virus then requires envelope glycoprotein gD and fibroblast growth factor receptor (Kaner, 1990). HSV is able to infect cells lyrically or can establish latency. HSV vectors have been used to infect a wide variety of cell types (Lowenstein, 1994; Huard, 1995; Miyanohara, 1992; Liu, 1996; Goya, 1998).
There are two types of HSV vectors, called the recombinant HSV vectors and the amplicon vectors. Recombinant HSV vectors are generated by the insertion of transcription units directly intot he HSV genome, through homologous recombination events. The amplicon vectors are based on plasmids bearing the transcription unit of choice, an origin of replication, and a packaging signal.
HSV vectors have the obvious advantages of a large capacity for insertion of foreign genes, the capacity to establish latency in neurons, a wide host range, and the ability to confer transgene expression to the CNS for up to 18 months (Carpenter & Stevens, 1996).
Retroviruses are enveloped single-stranded RNA viruses, which have been widely used in gene transfer protocols. Retroviruses have a diploid genome of about 7-10 kb, composed of four gene regions termed gag, pro, pol and env. These gene regions encode for structural capsid proteins, viral protease, integrase and viral reverse transcriptase, and envelope glycoproteins, respectively. The genome also has a packaging signal and czs-acting sequences, termed long-terminal repeats (LTRs), at each end, which have a role in transcriptional control and integration.
The most commonly used retroviral vectors are based on the Moloney murine leukaemia virus (Mo-MLV) and have varying cellular tropisms, depending on the receptor binding surface domain of the envelope glycoprotein.
Recombinant retroviral vectors are deleted of all retroviral genes, which are replaced with marker or therapeutic genes, or both. To propagate recombinant retroviruses, it is necessary to provide the viral genes, gag, pol and env in trans.
Lentiviruses are complex retroviruses that have the ability to infect and express their genes in both mitotic and post-mitotic cells. The most commonly known lentivirus is the human immunodeficiency virus (HIV), which uses the envelope glycoproteins of other viruses to target a broad range of cell types. Alphaviruses, including the prototype Sindbis virus (SIN), Semliki Forest virus (SFV), and Venezuelan equine encephalitis virus (VEE), constitute a group of enveloped viruses containing plus-stranded RNA genomes within icosahedral capsids.
The viral vectors of the present invention are useful for the delivery of nucleic acids expressing antigens or immunogens to cells both in vitro and in vivo. In particular, the inventive vectors can be advantageously employed to deliver or transfer nucleic acids to animal, more preferably avian and mammalian cells. Nucleic acids of interest include nucleic acids encoding peptides and proteins, preferably therapeutic (e.g., for medical or veterinary uses) or immunogenic (e.g., for vaccines) peptides or proteins. Preferably, the codons encoding the antigen or immunogen of interest are "optimized" codons, i.e., the codons are those that appear frequently in, i.e., highly expressed genes in the subject's species, instead of those codons that are frequently used by, for example, influenza virus. Such codon usage provides for efficient expression of the antigen or immunogen in animal cells. In other embodiments, for example, when the antigen or immunogen of interest is expressed in bacteria, yeast or other expression system, the codon usage pattern is altered to represent the codon bias for highly expressed genes in the organism in which the antigen or immunogen is being expressed. Codon usage patterns are known in the literature for highly expressed genes of many species (e.g., Nakamura et al., 1996; Wang et al, 1998; McEwan et al. 1998). As a further alternative, the viral vectors can be used to infect a cell in culture to express a desired gene product, e.g., to produce a protein or peptide of interest. Preferably, the protein or peptide is secreted into the medium and can be purified therefrom using routine techniques known in the art. Signal peptide sequences that direct extracellular secretion of proteins are known in the art and nucleotide sequences encoding the same can be operably linked to the nucleotide sequence encoding the peptide or protein of interest by routine techniques known in the art. Alternatively, the cells can be lysed and the expressed recombinant protein can be purified from the cell lysate. Preferably, the cell is an animal cell, more preferably a mammalian cell. Also preferred are cells that are competent for transduction by particular viral vectors of interest. Such cells include PER.C6 cells, 911 cells, and HEK293 cells.
A culture medium for culturing host cells includes a medium commonly used for tissue culture, such as M199-earle base, Eagle MEM (E-MEM), Dulbecco MEM (DMEM), SC-UCMl 02, UP-SFM (GIBCO BRL), EX-CELL302 (Nichirei), EX-CELL293-S (Nichirei), TFBM-Ol (Nichirei), ASF104, among others. Suitable culture media for specific cell types can be found at the American Type Culture Collection (ATCC) or the European Collection of Cell Cultures (ECACC). Culture media can be supplemented with amino acids such as L- glutamine, salts, anti-fungal or anti-bacterial agents such as Fungizone®, penicillin- streptomycin, animal serum, and the like. The cell culture medium can optionally be serum- free.
The present invention also provides vectors useful as drug and/or vaccine carriers. The immunogen or antigen can be presented in the viral capsid, alternatively, the antigen can be expressed from a transgene introduced into a recombinant viral genome and carried by the inventive virus. The viral vector can provide any antigen or immunogen of interest. Examples of immunogens are detailed herein.
The antigens or immunogens are preferably operably associated with the appropriate expression control sequences. Expression vectors include expression control sequences, such as an origin of replication (which can be eukaryotic origins, e.g., autonomously replicating sequences (ARS)), a promoter, an enhancer, and necessary information processing sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, packaging signals, and transcriptional terminator sequences.
For example, the viral vectors of the invention can contain appropriate transcription/translation control signals and polyadenylation signals (e.g., polyadenylation signals derived from bovine growth hormone, SV40 polyadenylation signal) operably associated with the antigen or immunogen sequence(s) to be delivered to the target cell. A variety of promoter/enhancer elements may be used depending on the level and tissue- specific expression desired. The promoter can be constitutive or inducible (e.g., the metallothionein promoter), depending on the pattern of expression desired. The promoter may be native or foreign and can be a natural or a synthetic sequence. By foreign, it is intended that the transcriptional initiation region is not found in the wild-type host into which the transcriptional initiation region is introduced. The promoter is chosen so that it will function in the target cell(s) or tissue(s) of interest. Brain-specific, hepatic-specific, and muscle-specific (including skeletal, cardiac, smooth, and/or diaphragm-specific) promoters are contemplated by the present invention. Mammalian promoters are also preferred. The promoter can advantageously be an "early" promoter. An "early" promoter is known in the art and is defined as a promoter that drives expression of a gene that is rapidly and transiently expressed in the absence of de novo protein synthesis. The promoter can also be an inducible promoter that can over-drive transgene expression upon induction by a specific substance. More preferably, the antigens or immunogens are operatively associated with, for example, a human cytomegalovirus (CMV) major immediate-early promoter, a simian virus 40 (SV40) promoter, a β-actin promoter, an albumin promoter, an Elongation Factor 1-α (EF 1-α) promoter, a PγK promoter, a MFG promoter, a pIX promoter, or a Rous sarcoma virus promoter. Other expression control sequences include promoters derived from immunoglobin genes, adenovirus, bovine papilloma virus, herpes virus, and so forth. Any mammalian viral promoter can also be used in the practice of the invention, in addition to any avian viral promoter. Among avian promoters of viral origin, the promoters of immediate early (i.e., ICP4, ICP27) genes of the infectious laryngotracheitis virus (ILTV) virus, early (i.e., thymidine kinase, DNA helicase, ribonucleotide reductase) or late (i.e., gB, gD, gC, gK), of the Marek's disease virus, (i.e., gB, gC, pp38, ppl4, ICP4, Meq) or of the herpes virus of turkeys (i.e., gB, gC, ICP4) can be used in the methods and vectors of the present invention. Moreover, it is well within the purview of the skilled artisan to select a suitable promoter that expresses the drug and/or antigen or immunogen of interest at sufficiently high levels so as to confer protection against a pathogen without undue experimentation.
In embodiments wherein there is more than one therapeutic ligand or antigen, the sequences may be operatively associated with a single upstream promoter and one or more downstream internal ribosome entry site (IRES) sequences (e.g., the picornavirus EMC IRES sequence). In embodiments of the invention in which the drug or antigen or immunogen sequence(s) will be transcribed and then translated in the target cells, specific initiation signals are generally required for efficient translation of inserted protein coding sequences. These exogenous translational control sequences, which may include the ATG initiation codon and adjacent sequences, can be of a variety of origins, both natural and synthetic. The recombinant vectors and methods of the present invention can be used in the treatment or prevention of various respiratory pathogens. Such pathogens include, but are not limited to, influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS- CoV), human rhinovirus (HRV), and respiratory syncytial virus (RSV).
Influenza viruses, which belong to the Orthomyxoviridae family, are classified as A, B, and C based on antigenic differences in their nucleoprotein (NP) and matrix (Ml) protein. Influenza viruses B and C infect humans, while influenza virus A infects human, avian species, equine species, swine, and even sea mammals. Further subtyping is based on the antigenicity of two surface glycoproteins, hemagglutinin (HA) and neuraminidase (NA). The HA protein enables the virion to attach to cell surface sialyloligosaccharides (Paulson, J.C., 1985) and is responsible for its hemagglutinating activity (Hirst, G.K., 1941). The HA protein comprises two disulfide-linked chains, HA1 and HA2. HA1 forms the sialic acid-binding sites and mediates HA attachment to the cell, while HA2 forms the membrane- spanning anchor and mediates fusion of viral and cellular membranes. The amino acid sequences of the HA1 region, which is responsible for HA antigenicity, differ from subtype to subtype by 30% or more (Rohm, C. et al, 1996b). There are currently 16 known HA antigenic subtypes (Hl to H 16), although sustained epidemics in humans have been limited to the Hl, H2, and H3 subtypes. NA is a sialidase that cleaves sialic acid residues from the cell surface and is important for release of virus from infected cells. Blocking NA activity, such as through neuraminidase inhibitors, prevents release of new virions from infected cells (Moscona, 2005). Nine NA subtypes have been identified (Nl to N9), of which the Nl and N2 subtypes have been found in human influenza viruses. Avian species, in particular shorebirds and waterfowl, are the natural source of influenza type A viruses. Influenza A viruses can also be transmitted from the aquatic bird reservoir to mammalian species, including humans, seals, whales, horses, pigs, and domestic poultry (Gillim-Ross et al., 2006). Avian influenza A viruses are defined by their virulence; highly virulent types can cause fowl plagues, while avirulent types generally cause only mild disease or asymptomatic infection. In rare instances, however, viruses with low pathogenicity in the laboratory cause outbreaks of severe disease in the field. Nonetheless, the morbidity and mortality associated with these viruses tend to be much lower than those caused by lethal viruses.
Highly virulent avian influenza viruses have caused outbreaks in poultry in Australia (1976 [H7] (Bashiruddin, J.B. et al, 1992); 1985 [H7] (Cross, G.M., 1987; Nestorowicz, A. et al, 1987), 1992 [H7] (Perdue, M.L. et al, 1997), 1995 [H7], and 1997 [H7]), England (1979 [H7] (Wood, G. W., et al, 1993) and 1991 [H5] (Alexander, DJ. et al, 1993), the United States (1983 to 1984 [H5] (Eckroade, RJ. et al, 1987), Ireland (1983 to 1984 [H5]) (Kawaoka, Y. et al, 1987), Germany (1979 [H7] (Rohm, C. et al, 1996a), Mexico (1994 to 1995 [H5] (Garcia, M. et al, 1996; Horimoto, T. et al, 1995), Pakistan (1995 [H7] (Perdue, M.L. et al, 1997), Italy (1997 [H5]), and Hong Kong (1997 [H5] (Claas, EJ. et al, 1988). Without wishing to be bound by any one theory, it is believed that all of the pathogenic avian influenza A viruses are of the H5 or H7 subtype, although the reason for this subtype specificity remains unknown. There appears to be no association of NA subtypes with virulent viruses. Two additional subtypes, H4 [A/Chicken/Alabama/7395/75 (H4N8)] (Johnson, D.C. et al, 1976) and HlO [A/Cbicken/Germany/N/49 (H10N7)], have been isolated from chickens during severe fowl plague-like outbreaks.
In addition to HA and NA, a limited number of Ml proteins are integrated into the virions (Zebedee, S.L. et al, 1988). They form tetramers, have Hl ion channel activity, and, when activated by the low pH in endosomes, acidify the inside of the virion, facilitating its uncoating (Pinto, L.H. et al, 1992). Ml protein that lies within the envelope is thought to function in assembly and budding. Eight segments of single-stranded RNA molecules (negative sense, or complementary to mRNA) are contained within the viral envelope, in association with NP and three subunits of viral polymerase (PBl, PB2, and PA), which together form a ribonucleoprotein (RNP) complex that participates in RNA replication and transcription. NS2 protein, now known to exist in virions (Richardson, J.C. et al, 1991; Yasuda, J. et al, 1993), is thought to play a role in the export of RNP from the nucleus (O'Neill, R.E. et al, 1998) through interaction with Ml protein (Ward, A.C. et al, 1995). NSl protein, the only nonstructural protein of influenza A viruses, has multiple functions, including regulation of splicing and nuclear export of cellular niRNAs as well as stimulation of translation (Lamb, R. A. et al, 1996). Its major function is believed to counteract the interferon activity of the host, since an NSl knockout virus was viable although it grew less efficiently than the parent virus in interferon-non-defective cells (Garcia-Sastre, A. et al, 1988).
The influenza therapeutic ligands, immunogens or antigens useful in the present invention include, but are not limited to, HA, NA, as well as Ml, NS2, and NSl. Examples of subtypes that are encompassed by the present invention include, but are not limited to, H10N4, H10N5, H10N7, H10N8, H10N9, HIlNl, H11N13, H11N2, H11N4, H11N6, H11N8, H11N9, H12N1, H12N4, H12N5, H12N8, H13N2, H13N3, H13N6, H13N7,
H14N5, H14N6, H15N8, H15N9, H16N3, HlNl, H1N2, H1N3, H1N6, H2N1, H2N2, H2N3, H2N5, H2N7, H2N8, H2N9, H3N1, H3N2, H3N3, H3N4, H3N5, H3N6, H3N8, H4N1, H4N2, H4N3, H4N4, H4N5, H4N6, H4N8, H4N9, H5N1, H5N2, H5N3, H5N7, H5N8, H5N9, H6N1, H6N2, H6N4, H6N5, H6N6, H6N7, H6N8, H6N9, H7N1, H7N2, H7N3, H7N5, H7N7, H7N8, H8N4, H8N5, H9N1, H9N2, H9N3, H9N5, H9N6, H9N7, H9N8, and H9N9.
The influenza therapeutic ligands, immunogens or antigens can be derived from any known strain of influenza, including all influenza A and B strains, clinical isolates, field isolates, and reassortments thereof. The invention also relates to the use of mutated or otherwise altered influenza genes that reflect, among other things, antigenic drift and antigenic shift.
Examples of influenza strains include, but are not limited to, turkey influenza virus strain A/Turkey/Ireland/1378/83 (H5N8) (see, e.g., Taylor et al., 1988b), turkey influenza virus strain A/Turkey/England/63 (H7N3) (see, e.g., Alexander et al., 1979; Rott et al., 1979; Horimoto et al., 2001), turkey influenza virus strain A/Turkey/England/66 (H6N2) (see, e.g., Alexander et al., 1979), A/Turkey/England/69 (H7N2) (see, e.g., Alexander et al., 1979; Horimoto et al., 2001), A/Turkey/Scotland/70 (H6N2) (see, e.g., Banks et al., 2000; Alexander et al., 1979), turkey influenza virus strain A/Turkey/England/N28/73 (H5N2) (see, e.g., Alexander et al., 1979), turkey influenza virus strain A/Turkey/England/110/77 (H6N2) (see, e.g., Alexander et al., 1979), turkey influenza virus strain A/Turkey/England/647/77 (HlNl) (see, e.g., Alexander et al., 1979; Karasin et al., 2002)), turkey influenza virus strain A/turkey/Ontario/7732/66 (H5N9) (see, e.g., Slemons et al., 1972; Philpott et al., 1989), turkey influenza virus strain A/Turkey/England/199/79 (H7N7) (see, e.g., Horimoto et al., 2001 ), turkey influenza virus strain A/Turkey/Ontario/7732/66 (H5N9) (see, e.g., Horimoto et al., 2001; Panigrahy et al., 1996), turkey influenza virus strain A/Turkey/Ireland/1378/85 (H5N8) (see, e.g., Horimoto et al., 2001 ; Walker et al., 1993), turkey influenza virus strain A/Turkey/England/50-92/91 (H5N1) (see, e.g., Horimoto et al., 2001; Howard et al., 2006), turkey influenza virus strain A/Turkey/Wisconsin/68 (H5N9), turkey influenza virus strain A/Turkey/Masschusetts/65 (H6N2), turkey influenza virus strain A/Turkey/Oregon/71 (H7N3), (see, e.g., Orlich et al., 1990), turkey influenza virus strain A/Turkey/Ontario/6228/67 (H8N4), turkey influenza virus strain A/Turkey/Wisconsin/66 (H9N2), (see, e.g., ZakstePskaia et al., 1977), turkey influenza virus strain A/Turkey/England/647/77 (HlNl) (see, e.g., Karasin et al., 2002; Alexander et al., 1979), turkey influenza virus strain A/Turkey/Ontario/6118/68 (H8N4) (see, e.g., Blok et al., 1982), turkey influenza virus strain A/Tur/Ger 3/91 (see, e.g., Zakay-Rones et al., 1995), turkey influenza virus strain A/Turkey/Minnesota/833/80 (H4N2) (see, e.g., Gubareva et al., 1997) chicken influenza virus strain A/Chicken/Indonesia/03 (H5N1), chicken influenza virus strain A/Chicken/FPV/Rostock/1934 (see, e.g., Ohuchi et al., 1994), chicken influenza virus strain A/Chicken/Texas/298313/04 (see, e.g., Lee et al., 2005), chicken influenza virus strain A/Chicken/Texas/I 67280-4-/02 (see, e.g., Lee et al., 2005), chicken influenza virus strain A/Chicken/Hong Kong/220/97 (see, e.g., Perkins et al., 2001), chicken influenza virus strain A/Chicken/Italy/8/98 (see, e.g., Capua et al., 1999), chicken influenza virus strain A/Chicken/Victoria/76 (H7N7) (see, e.g., Zambon, 2001; Nestorowicz et al., 1987), chicken influenza virus strain A/Chicken/Germany/79 (H7N7) (see, e.g., Rohm et al., 1996), chicken influenza virus strain A/Chicken/Scotland/59 (H5N1) (see, e.g., Horimoto et al., 2001; De et al., 1988; Wood et al., 1993), chicken influenza virus strain A/Chicken/Pennsylvania/1370/83 (H5N2) (see, e.g., Bean et al., 1985; van der Goot et al., 2002), chicken influenza virus strain A/Chicken/Queretaro- 19/95 (H5N2) (see, e.g., Horimoto et al., 2001 ; Garcia et al., 1998), chicken influenza virus strain A/Chicken/Queretaro-20/95 (H5N2) (see, e.g., Horimoto et al., 2001), chicken influenza virus strain A/Chicken/Hong Kong/258/97 (H5N1) (see, e.g., Horimoto et al., 2001; Webster, 1998), chicken influenza virus strain A/Chicken/Italy/1487/97 (H5N2) (see, e.g., Horimoto et al., 2001), chicken influenza virus strain A/Chicken/Leipzig/79 (H7N7) (see, e.g., Horimoto et al., 2001; Rohm et al., 1996), chicken influenza virus strain A/Chicken/Victoria/85 (H7N7) (see, e.g., Horimoto et al., 2001), chicken influenza virus strain A/Chicken/Victoria/92 (H7N3) (see, e.g., Horimoto et al., 2001), chicken influenza virus strain A/Chicken/Queensland/95 (H7N3) (see, e.g., Horimoto et al., 2001), chicken influenza virus strain A/Chicken/Pakistan/1369/95 (H7N2) (see, e.g., Horimoto et al., 2001), chicken influenza virus strain A/Chicken/Pakistan/447-4/95 (H7N3) (see, e.g., Horimoto et al., 2001), chicken influenza virus strain A/Chicken/HK/G9/97 (H9N2) (see, e.g., Leneva et al., 2001), chicken influenza virus strain A/Chicken/Nakorn-Patom/Thailand/CU-K2/2004(H5Nl) (see, e.g., Anwar et al., 2006; Viseshakul et al., 2004), chicken influenza virus strain A/Chicken/Hong Kong/31.2/2002 (H5N1), (see, e.g., Anwar et al., 2006), chicken influenza virus strain
A/Chicken/Vietnam/C58/04 (H5N1), (see, e.g., Anwar et al., 2006;), chicken influenza virus strain A/Chicken/Vietnam/38/2004(H5Nl). (see, e.g., Anwar et al., 2006), chicken influenza virus strain A/Chicken/Alabama/7395/75 (H4N8), (see, e.g., Swayne et al., 1994), chicken influenza virus strain A/Chicken/Germany/N/49 (H10N7), (see, e.g., Yamane et al., 1981), chicken influenza virus strain A/Chicken/Beijing/1/94 (H9N2) (see, e.g., Karasin et al., 2002), chicken influenza virus strain A/Chicken/Hong Kong/G23/97 (H9N2) (see, e.g., Karasin et al., 2002), chicken influenza virus strain A/Chicken/Pennsylvania/8125/83 (H5N2) (see, e.g., Karasin et al., 2002; Shortridge et al., 1998), chicken influenza virus strain A/Chicken/Hong Kong/97 (H5N1) (see, e.g., Chen et al., 2003), duck influenza virus strain A/Duck/Anyang/AVL-1/01 (see, e.g., Tumpey et al., 2002), duck influenza virus strain
A/Duck/New York/17542-4/86 (H9N1) (see, e.g., Banks et al., 2000), duck influenza virus strain A/Duck/Alberta/28/76 (H4N6) (see, e.g., Blok et al., 1982), duck influenza virus strain A/Duck/Nanchang/4- 165/2000 (H4N6) (see, e.g., Liu et al., 2003), duck influenza virus strain A/Duck/Germany/49 (H10N7) (see, e.g., Blok et al., 1982), duck influenza virus strain A/Black Duck/Australia/702/78 (H3N8) (see, e.g., Blok et al, 1982), duck influenza virus strain A/Duck/Vietnam/I 1/2004 (H5N1), (see, e.g., Anwar et al., 2006), duck influenza virus strain A/Duck/Alberta/60/76 (H12N5), (see, e.g., Baez et al., 1981), duck influenza virus strain A/Duck/Hong Kong/196/77 (Hl) (see, e.g., Karasin et al., 2002; Kanegae et al., 1994), duck influenza virus strain A/Duck/Wisconsin/I 938/80 (HlNl) (see, e.g., Karasin et al., 2002), duck influenza virus strain A/Duck/Bavaria/2/77 (HlNl) (see, e.g., Karasin et al., 2002; Ottis et al., 1980), duck influenza virus strain A/Duck/Bavaria/1/77 (HlNl) (see, e.g., Ottis et al., 1980), duck influenza virus strain A/Duck/Australia/749/80 (HlNl) (see, e.g., Karasin et al., 2002), duck influenza virus strain A/Duck/Hong Kong/Y280/97 (H9N2) (see, e.g., Karasin et al., 2002; Guan et al., 2000), duck influenza virus strain
A/Duck/Alberta/35/76 HlNl) (see, e.g., Austin et al., 1990), avian influenza virus strain A/Mallard duck/Gurjev/263/82 (H14N5), (see, e.g., Kawaoka et al., 1990), avian influenza virus strain A/Mallard duck/PA/ 10218/84 (H5N2) (see, e.g., Smirnov et al., 2000), avian influenza virus strain A/Mallard duck/Astrakhan/244/82 (H14N6) (see, e.g., Karasin et al., 2002), goose influenza virus strain A/Goose/Guangdong/1/96 (see, e.g., Xu et al., 1999), goose influenza virus strain A/Goose/Leipzig/I 37-8/79 (H7N7) (see, e.g., Horimoto et al., 2001), goose influenza virus strain A/Goose/Hong Kong/W222/97 (H6N7) (see, e.g., Chin et al., 2002), goose influenza virus strain A/Goose/Leipzig/ 187-7/79 (H7N7) (see, e.g., Horimoto et al., 2001), goose influenza virus strain A/Goose/Leipzig/I 92-7/79 (H7N7) (see, e.g., Horimoto et al., 2001), avian influenza virus strain A/Env/HK/437-4/99 (see, e.g.,
Cauthen et al., 2000), avian influenza virus strain A/Env/HK/437-6/99 (see, e.g., Cauthen et al., 2000), avian influenza virus strain A/Env/HK/437-8/99 (see, e.g., Cauthen et al., 2000), avian influenza virus strain A/Env/HK/437-10/99, (see, e.g., Cauthen et al., 2000), avian influenza virus strain A/Fowl plague virus strain/Dutch/27 (H7N7) (see, e.g., Horimoto et al., 2001 ; Carter et al., 1982), avian influenza virus strain A/Fowl plague virus strain/Dobson/27 (H7N7) (see, e.g., Horimoto et al., 2001), avian influenza virus strain A/Fowl plague virus strain/Rostock/34 (H7N1) (see, e.g., Horimoto et al., 2001; Takeuchi et al., 1994), avian influenza virus strain A/Fowl plague virus strain/Egypt/45 (H7N1) (see, e.g., Horimoto et al., 2001), avian influenza virus strain A/Fowl plague virus strain/Weybridge (H7N7) (see, e.g., Tonew et al., 1982), avian influenza virus strain A/Tern/South Afiϊca/61 (H5N3) (see, e.g., Horimoto et al., 2001; Perkins et al., 2002; Walker et al., 1992), avian influenza virus strain A/Tern/Australia/G70C/75 (Hl 1N9) (see, e.g., Pruett et al., 1998), avian influenza virus strain A/Quail/Vietnam/36/04(H5Nl). (see, e.g., Anwar et al., 2006), avian influenza virus strain A/Gull/Maryland/704/77 (H13N6), (see, e.g., Iamnikova et al., 1989), avian influenza virus strain A/Black-headed gull/Sweden/5/99 (H16N3) (see, e.g., Fouchier et al., 2005), avian influenza virus strain A/Herring gull/DE/677/88 (H2N8) (see, e.g., Saito et al., 1993), avian influenza virus strain A/Swan/Italy/179/06 (H5N1) (see, e.g., Terregino et al., 2006), avian influenza virus strain A/Hong Kong/156/97 (A/HK/156/97) (see, e.g., Leneva et al., 2001 ; Claas et al., 1998; Cauthen et al., 2000), avian influenza virus strain
A/Quail/HK/Gl/97 (H9N2) (see, e.g., Leneva et al., 2001), avian influenza virus strain A/Quail/Hong Kong/AF157/93 (H9N2) (see, e.g., Karasin et al., 2002), avian influenza virus strain A/Teal/HK/W312/97 (H6N1) (see, e.g., Leneva et al., 2001), avian influenza virus strain A/Shearwater/West Australia/2576/79 (H15N9) (see, e.g., Rohm et al., 1996), avian influenza virus strain A/Shearwater/Australia/72 (H6N5) (see, e.g., Harley et al., 1990), avian influenza virus strain A/Hong Kong/212/03 (see, e.g., Shinya et al., 2005), avian influenza virus strain A/England/321/77 (H3N2) (see, e.g., Hauptmann et al., 1983), avian pandemic influenza A viruses of avian origin (see, e.g., Audsley et al., 2004) avian H5N1 influenza virus, , avian H7N1 influenza strain (see, e.g., Foni et al., 2005), avian H9N2 influenza virus (see, e.g., Leneva et al., 2001), and avian influenza virus, cold-adapted (ca) and temperature sensitive (ts) master donor strain, A/Leningrad/I 34/17/57 (H2N2) (see, e.g., Youil et al., 2004), the disclosures of which are incorporated by reference.
Other influenza strains that may be used in methods of the present invention include, but are not limited to, equine influenza virus (A/Equi 2 (H3N8), Newmarket 1/93) (see, e.g., Mohler et al., 2005; Nayak et al., 2005) , equine-2 influenza virus (EIV; subtype H3N8) (see, e.g., Lin et al., 2001), equine-2 influenza virus, A/Equine/Kentucky/1/91 (H3N8) (see, e.g., Youngner et al., 2001), equine influenza virus strain A/Equine/Berlin/2/91 (H3N8) (see, e.g., Ilobi et al., 1998), equine influenza virus strain A/Equine/Cambridge/1/63 (H7N7) (see, e.g., Gibson et al., 1992), equine influenza virus strain A/Equine/Prague/1/56 (H7N7) (see, e.g., Karasin et al., 2002; Appleton et al., 1989), equine influenza virus strain A/Eq/Kentucky/98 (see, e.g., Crouch et al., 2004), equine influenza virus strain A/Equi 2 (Kentucky 81) (see, e.g., Short et al., 1986; Horner et al., 1988), equine influenza virus strain A/Equine/Kentucky/1/81 (Eq/Ky) (see, e.g., Breathnach et al., 2004), equine influenza virus strain A/Equine/Kentucky/1/81 (H3N8) (see, e.g., Olsen et al., 1997; Morley et al., 1995; Ozaki et al., 2001 ; Sugiura et al., 2001 ; Goto et al., 1993), equine influenza virus strain A/Equine/Kentucky/ 1/91 (H3N8) (see, e.g., Youngner et al., 2001), equine influenza virus strain A/Equine/Kentucky/ 1277/90 (Eq/Kentucky) (see, e.g., Webster et al., 1993), equine influenza virus strain A/Equine/Kentucky/2/91 (H3N8) (see, e.g., Donofrio et al., 1994), equine influenza virus strain A/Equine/Kentucky/79 (H3N8) (see, e.g., Donofrio et al., 1994), equine influenza virus strain A/Equine/Kentucky/81 (see, e.g., Sugiura et al., 2001), equine influenza virus strain A/Equine/Kentucky/91 (H3N8) (see, e.g., Gross et al., 1998), equine influenza virus strain A/Equine-2/Kenrucky/95 (H3N8) (see, e.g., Heldens et al., 2004) and equine influenza virus strain A/Equine-2/Kentucky/98 (see, e.g., Chambers et al., 2001), equine influenza virus strain A/Eq/Newmarket/1/77 (see, e.g., Lindstrom et al., 1998), equine influenza virus strain A/Eq/Newmarket/5/03 (see, e.g., Edlund Toulemonde et al., 2005), equine influenza virus strain A/Equi 2 (H3N8), Newmarket 1/93 (see, e.g., Mohler et al., 2005; Nayak et al., 2005), equine influenza virus strain A/Equi-2/Newmarket-l/93 (see, e.g., Heldens et al., 2002), equine influenza virus strain A/Equine/Newmarket/2/93 (see, e.g., Wattrang et al., 2003), equine influenza virus strain A/Equine/Newmarket/79 (H3N8) (see, e.g., Duhaut et al., 2000 ; Noble et al., 1994; Duhaut et al., 1998; Hannant et al., 1989; Hannant et al., 1989; Hannant et al., 1988; Richards et al., 1992; Heldens et al., 2004), equine influenza virus strain A/Equine/Newmarket/1/77 (H7N7) (see, e.g., Goto et al., 1993; Sugiura et al., 2001) and equine influenza virus strain A/Equine-2/Newmarket-2/93 (see, e.g., Heldens et al., 2004), equine influenza virus strain A/Eq/Miami/63 (H3N8) (see, e.g., van Maanen et al., 2003), A/Equi 1 (Prague strain) (see, e.g., Homer et al., 1988; Short et al., 1986), equine influenza virus strain A/Equi 2 (Miami) (see, e.g., Short et al., 1986), equine influenza virus strain A/Equi- 1 /Prague/56 (Pr/56) (see, e.g., Heldens et al., 2002), equine influenza virus strain A/Equi-2/Suffolk/89 (Suf789) (see, e.g., Heldens et al., 2002), equine influenza virus strain A/Equine 2/Sussex/89 (H3N8) (see, e.g., Mumford et al., 1994), equine influenza virus strain A/Equine/Sussex/89 (see, e.g., Wattrang et al., 2003), equine influenza virus strain A/Equine-2/Saskatoon/90 (see, e.g., Chambers et al., 2001), equine influenza virus strain A/Equine/Prague/1/56 (H7N7) (see, e.g., Donofrio et al., 1994; Morley et al., 1995), equine influenza virus strain A/Equine/Miami/I /63 (H3N8) (see, e.g., Morley et al., 1995; Ozaki et al., 2001; Thomson et al., 1977; Mumford et al., 1988; Donofrio et al., 1994; Mumford et al., 1983), A/Aichi/2/68 (H3N2) (see, e.g., Ozaki et al., 2001), equine influenza virus strain A/Equine/Tokyo/2/71 (H3N8) (see, e.g., Goto et al., 1993), equine influenza virus strain A/Eq/LaPlata/1/88 (see, e.g., Lindstrom et al., 1998), equine influenza virus strain A/Equine/Jilin/1/89 (Eq/Jilin) (see, e.g., Webster et al., 1993), equine influenza virus strain A/Equine/Alaska/ 1/91 (H3N8) (see, e.g., Webster et al., 1993), equine influenza virus strain A/Equine/Saskatoon/1/91 (H3N8) (see, e.g., Morley et al., 1995), equine influenza virus strain A/Equine/Rome/5/91 (H3N8) (see, e.g., Sugiura et al., 2001), equine influenza virus strain A/Equine/La Plata/ 1/93 (H3N8) (see, e.g., Ozaki et al., 2001), equine influenza virus strain A/Equine/La Plata/1/93 (LP/93) (see, e.g., Sugiura et al., 2001), equine influenza virus strain A/Eq/Holland/1/95 (H3N8) (see, e.g., van Maanen et al., 2003) and equine influenza virus strain A/Eq/Holland/2/95 (H3N8) (see, e.g., van Maanen et al., 2003), human influenza virus A(H3N2) isolates (see, e.g., Abed et al., 2002), human influenza virus A/Memphis/ 1/71 (H3N2) (see, e.g., Suzuki et al., 1996), human influenza virus A/Nanchang/933/95 (H3N2) virus (see, e.g., Scholtissek et al., 2002), human influenza virus A/PR/8/34 (HlNl) virus (see, e.g., Scholtissek et al., 2002), human influenza virus A/Singapore/57 (H2N2) virus (see, e.g., Scholtissek et al., 2002), influenza virus A (see, e.g., Chare et al., 2003), influenza virus A/HK/213/03 (see, e.g., Guan et al., 2004; Anwar et al., 2006), influenza virus strain A/HK/483/97 (see, e.g., Cheung et al., 2002), influenza virus strain A/HK/486/97 (see, e.g., Cheung et al., 2002), influenza virus strain A/Thailand/5(KK-494)/2004 (H5N1).( see, e.g., Anwar et al., 2006), influenza virus strain A PR/8/34 (PR8) virus strain (HlNl subtype) (see, e.g., Mantani et al., 2001), influenza virus strain A/Aichi/2/68(H3N2) (see, e.g., Miyamoto et al., 1998), influenza virus strain A/Ann Arbor/6/60 cold-adapted virus strain (see, e.g., Treanor et al., 1994), influenza virus strain A/Beijing 32/92 (H3N2) (see, e.g., Zakay-Rones et al., 1995), influenza virus strain A/Charlottesville/31/95 (HlNl) (see, e.g., Gubareva et al., 2002), influenza virus strain A/Kawasaki/86 (HlNl) virus strain (see, e.g., Staschke et al., 1998), influenza virus strain A/Korea/82 (H3N2) (see, e.g., Treanor et al., 1994), influenza virus strain A/Leningrad/ 134/57 (see, e.g., Egorov et al., 1998), influenza virus strain A/NWS/33 (HlNl) (see, e.g., Sidwell et al., 1998), influenza virus strain A/PR/8/34(HlNl) (see, e.g., Miyamoto et al., 1998), influenza virus strain A/PR8/34 (see, e.g., Nunes-Correia et al., 1999; Tree et al., 2001), influenza virus strain A/Puerto Rico (PR)/8/34 (see, e.g., Egorov et al., 1998), influenza virus strain A/Puerto Rico/8-Mount Sinai (see, e.g., Mazanec et al., 1995), influenza virus strain A/Shangdong 9/93 (H3N2) (see, e.g., Zakay-Rones et al., 1995; Sidwell et al., 1998), influenza virus strain A/Shingapol/1/57(H2N2) (see, e.g., Miyamoto et al., 1998), influenza virus strain A/Singapore 6/86 (HlNl) (see, e.g., Zakay-Rones et al., 1995), influenza virus strain A/Singapore/1/57 (H2N2) (see, e.g., Bantia et al., 1998), influenza virus strain A/Texas 36/91 (HlNl) (see, e.g., Zakay-Rones et al., 1995), influenza virus strain A/Texas/36/91 (HlNl) virus strain (see, e.g., Gubareva et al., 2001; Halperin et al., 1998), influenza virus strain A/Texas/36/91 (HlNl) (see, e.g., Hayden et al., 1994), influenza virus strain A/Udorn/72 virus infection (see, e.g., Shimizu et al., 1999), influenza virus A/Victoria/3/75 (H3N2) (see, e.g., Sidwell et al., 1998), influenza virus A/Virginia/88(H3N2) (see, e.g., Hayden et al., 1994), influenza virus A/WSN/33 (HlNl) (see, e.g., Lu et al., 2002), influenza virus A/WSN/33 (see, e.g., Gujuluva et al., 1994), influenza virus B (see, e.g., Chare et al., 2003), influenza virus B/Ann Arbor 1/86 (see, e.g., Zakay-Rones et al., 1995), influenza virus B/Harbin/7/94 (see, e.g., Halperin et al., 1998), influenza virus B/Hong Kong/5/72 (see, e.g., Sidwell et al., 1998), influenza virus B/Lee/40 (see, e.g., Miyamoto et al., 1998), influenza virus B/Victoria group (see, e.g., Nakagawa et al., 1999), influenza virus B/Yamagata 16/88 (see, e.g., Zakay-Rones et al., 1995), influenza virus B/Yamagata group (see, e.g., Nakagawa et al., 1999), influenza virus
B/Yamanashi/166/98 (see, e.g., Hoffmann et al., 2002), influenza virus C (see, e.g., Chare et al., 2003), influenza virus strain A/Equi/2/Kildare/89 (see, e.g., Quinlivan et al., 2004), influenza virus type B/Panama 45/90 (see, e.g., Zakay-Rones et al., 1995), live, cold-adapted, temperature-sensitive (ca/ts) Russian influenza A vaccines (see, e.g., Palker et al., 2004), swine Hl and H3 influenza viruses (see, e.g., Gambaryan et al., 2005), swine influenza A viruses (see, e.g., Landolt et al., 2005), swine influenza virus (SIV) (see, e.g., Clavijo et al., 2002), swine influenza virus A/Sw/Ger 2/81 (see, e.g., Zakay-Rones et al., 1995), swine influenza virus A/Sw/Ger 8533/91 (see, e.g., Zakay-Rones et al., 1995), swine influenza virus strain A/Swine/Wisconsin/ 125/97 (HlNl) (see, e.g., Karasin et al., 2002; Karasin et al., 2006), swine influenza virus strain A/Swine/Wisconsin/ 136/97 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Wisconsin/I 63/97 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/S wine/Wisconsin/ 164/97 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Wisconsin/I 66/97 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Wisconsin/I 68/97 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Wisconsin/235/97 (HlNl) (see, e.g., Karasin et al., 2002; Olsen et al., 2000), swine influenza virus strain A/Swine/Wisconsin/238/97 (HlNl) (see, e.g., Karasin et al., 2002; Ayora-Talavera et al., 2005), swine influenza virus strain A/Swine/Wisconsin/457/98 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Wisconsin/458/98 (HlNl) (see, e.g., Karasin et al., 2002; Karasin et al., 2006), swine influenza virus strain A/Swine/Wisconsin/464/98 (HlNl) (see, e.g., Karasin et al., 2002; Karasin et al., 2006), swine influenza virus strain A/Swine/Indiana/1726/88 (HlNl) (see, e.g., Karasin et al., 2002; Macklin et al., 1998), swine influenza virus strain A/Swine/Indiana/9K035/99 (H1N2) (see, e.g., Karasin et al., 2002; Karasin et al., 2000), swine influenza virus strain A/Swine/Nebraska/ 1/92 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Quebec/91 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Quebec/81 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/New Jersey/11/76 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Ehime/1/80 (H1N2) (see, e.g., Karasin et al., 2002; Nerome et al., 1985), swine influenza virus strain A/Swine/England/283902/93 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/England/ 195852/92 (HlNl) (see, e.g., Karasin et al., 2002; Brown et al., 1993), swine influenza virus strain A/Swine/Germany/8533/91 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Germany/2/81 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Nebraska/209/98 (H3N2) (see, e.g., Karasin et al., 2002), A/Swine/Iowa/533/99 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Iowa/569/99 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Minnesota/593/99 (H3N2) (see, e.g., Karasin et al., 2002; Ayora- Talavera et al., 2005), swine influenza virus strain A/Swine/Iowa/8548-1/98 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Minnesota/9088-2/98 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Texas/4199-2/98 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Ontario/41848/97 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/North Carolina/35922/98 (H3N2) (see, e.g., Karasin et al., 2002), /Swine/Colorado/ 1/77 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Hong Kong/3/76 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Hong Kong/13/77 (H3N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Nagasaki/ 1/90 (H1N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Nagasaki/1/89 (H1N2) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Wisconsin/1915/88 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Iowa/17672/88 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Tennessee/24/77 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Ontario/2/81 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Wisconsin/1/67 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Italy/ 1521/98 (H1N2) (see, e.g., Marozin et al., 2002), swine influenza virus strain A/Swine/Italy/839/89 (HlNl) (see, e.g., Karasin et al., 2002), swine influenza virus strain A/Swine/Hong Kong/126/82 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Idaho/4/95 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Johannesburg/33/94 (H3N2) (see, e.g., Karasin et al., 2002; Johansson et al., 1998), influenza virus strain A/Bangkok/1/79 (H3N2) (see, e.g., Karasin et al., 2002; Nelson et al., 2001), influenza virus strain A/Udorn/72 (H3N2) (see, e.g., Karasin et al., 2002; Markoff et al., 1982), influenza virus strain A/Hokkaido/2/92 (HlNl) (see, e.g., Karasin et al., 2002), influenza virus strain A/Thailand/KAN-1/04 (see, e.g., Puthavathana et al., 2005; Amonsin et al., 2006), influenza virus strain A/England/1/53 (see, e.g., Govorkova EA, et al.,, 1995), influenza virus strain A/Vietnam/3046/2004 (H5N1), (see, e.g., Anwar et al., 2006), influenza virus strain A/Vietnam/I 203/2004 (H5N1), (see, e.g., Anwar et al., 2006; Gao et al., 2006), influenza virus strain A/tiger/Thailand/SPB-l(H5Nl), (see, e.g., Anwar et al., 2006), influenza virus strain A/Japan/305/57 (H2N2) (see, e.g., Naeve et al., 1990; Brown et al., 1982), influenza virus strain A/Adachi/2/57 (H2N2) (see, e.g., Gething et al., 1980), influenza virus strain A/Camel/Mongolia/82 (HlNl) (see, e.g., Yamnikova et al., 1993), influenza virus strain A/RI/5/57 (H2N2) (see, e.g., Elleman et al., 1982), influenza virus strain A/Whale/Maine/1/84 (H13N9) (see, e.g., Air et al., ,1987), influenza virus strain A/Taiwan/1/86 (HlNl) (see, e.g., Karasin et al., 2002; Brown, 1988), influenza virus strain A/Bayern/7/95 (HlNl) (see, e.g., Karasin et al., 2002), influenza virus strain A/USSR/90/77 (HlNl) (see, e.g., Karasin et al., 2002; Iftimovici et al., 1980), influenza virus strain A/Wuhan/359/95 (H3N2) (see, e.g., Karasin et al., 2002; Hardy et al., 2001), influenza virus strain A/Hong Kong/5/83 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Memphis/8/88 (H3N2) (see, e.g., Karasin et al., 2002; Hatta et al., 2002), influenza virus strain A/Beijing/337/89 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain
A/Shanghai/6/90 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Akita/1/94 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Akita/1/95 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Memphis/6/90 (H3N2) (see, e.g., Karasin et al., 2002), influenza virus strain A/Udorn/307/72 (H3N2) (see, e.g., Karasin et al., 2002; Iuferov et al., 1984), influenza virus strain A/Singapore/1/57 (H2N2) (see, e.g., Karasin et al., 2002; Zhukova et al., 1975), influenza virus strain A/Ohio/4/83 (HlNl) (see, e.g., Karasin et al., 2002), influenza virus strain Madin Darby Canine Kidney (MDCK)-derived cell line (see, e.g., Halperin et al., 2002), mouse-adapted influenza virus strain A/Guizhou/54/89 (H3N2 subtype) (see, e.g., Nagai et al., 1995), mouse-adapted influenza virus A/PR/8/34 (A/PR8) (see, e.g., Nagai et al., 1995), mouse-adapted influenza virus B/Ibaraki/2/85 (see, e.g., Nagai et al., 1995), Russian live attenuated influenza vaccine donor strains A/Leningrad/I 34/17/57, A/Leningrad/I 34/47/57 and B/USSR/60/69 (see, e.g., Audsley et al. 2005), the disclosures of which are incorporated by reference.
The antigenicity of influenza viruses changes gradually by point mutation (antigenic drift) or drastically by genetic reassortment (antigenic shift) (Murphy, B. R. et al, 1996). Immunological pressure on HA and NA is thought to drive antigenic drift. Antigenic shift can be caused by either direct transmission of nonhuman influenza viruses to humans or the reassortment of genes from two different influenza viruses that have infected a single cell (Webster, R.G. et al, 1982). Theoretically, 256 different combinations of RNA can be produced from the shuffling of the eight different genomic segments of the virus. Genetic reassortment is well documented both in vitro and in vivo under laboratory conditions (Webster, R. G. et al, 1975). More importantly, mixed infections occur relatively frequently in nature and can lead to genetic reassortment, resulting in new field isolates, hybrid forms, or reassortant forms (Bean, WJ. et al, 1980; Hinshaw, V.S. et al, 1980; Young, J.F., et al, 1979). Reemergence of a previously circulating virus is another mechanism by which antigenic shift can occur.
Thus, the invention also concerns the use of influenza therapeutic ligands, immunogens or antigens that have undergone antigenic drift or antigenic shift, including clinical isolates of influenza, field or environmental isolates of influenza, hybrid forms, and reassortant forms of influenza.
Severe acute respiratory syndrome-associated coronavirus (SARS-CoV) is the virus which causes SARS. Coronaviruses are enveloped, positive-stranded RNA viruses that can cause enteric or respiratory tract infectious in a variety of species including human, livestock, and pets.
The SARS-CoV genome contains 11 significant ORFs, which include the following: IA and IB, which encode polyproteins of the replicase complex; envelope spike protein S, which mediates attachment to cellular receptors and entry by fusion with cell membranes; small envelope protein E, which serves as a scaffold protein to trigger assembly; matrix protein M, which is an integral membrane protein involved in budding and which interacts with the nucleocapsid and the S proteins; and nucleocapsid protein N.
The S protein extends from the surface of the virion and serves as the major viral attachment protein. The interaction between receptor and S protein is an important determinant of species specificity and tissue tropism. Such receptors include angiotensin- converting enzyme 2 and CD209L, although angiotensin-converting enzyme 2 serves as a more efficient receptor (Li et al., 2007).
In several animal models, infection with SARS-CoV induces the production of neutralizing antibodies (NAbs) which protect animals from subsequent virus challenge and are sufficient to restrict SARS-CoV replication (Subbarao, et al., 2005). Analysis of the epitopes recognized by these NAbs have revealed that the receptor binding domain of the S protein is a neutralization determinant (Greenough, et al., 2005). Also, monoclonal antibodies capable of neutralizing SARS-CoV by targeting the S protein have been identified and protected mice against subsequent virus challenge (Greenough, et al., 2005).
In the present invention, the SARS-CoV therapeutic ligands, immunogens or antigens that may be used include, but are not limited to, IA, IB, spike protein S, envelope protein E, matrix protein M, and nucleocapsid protein N.
Human rhinovirus (HRV) belongs to the Picornaviridae family. There are 102 identified serotypes of HRV, which can be divided into two groups based on receptor utilization. The first group comprises over 90% of the identified HRV serotypes, and they bind to human intercellular adhesion molecule- 1 (ICAM-I). The second group binds to low- density lipoprotein and related proteins (Rossmann et al., 2007).
HRV are non-enveloped viruses with an icosahedral capsid that encloses a single- stranded, positive-sense RNA genome. The viral polyprotein is divided into a Pl region, P2 region, and P3 region. The Pl region encodes capsid proteins VPl, VP2, VP3, and VP4, while the P2 and P3 regions include proteins 2APro, 2B, 2C, 3A, 3B (VPg), 3CPro, and 3DPoI.
HRV typically infects through docking to epithelial cells via specific cellular receptors. The binding of HRV to the receptors induces conformational changes of the capsid and leads to the release of viral RNA (Nurani, et al., 2003). A majority of HRV uses the cell surface receptor ICAM-I to bind to and infect epithelial cells.
In the present invention, the HRV therapeutic ligands, immunogens or antigens that may be used include, but are not limited to, proteins or ligands which bind to ICAM-I . Such ligands may be acquired through methods known in the art, such high throughput screening or high throughout virtual screening (see Taylor et al., 2007, which is incorporated herein by reference), fragment-based ligand discovery (see Erlanson 2007, which is incorporated herein by reference), and tethering technology (see U.S. Patent Nos. 5,925,529 and 6,998,233, which is incorporated herein by reference).
Human respiratory syncytial virus (RSV) causes respiratory tract infections. It is a large negative-sense, single-stranded RNA virus, member of the family of Paramyxoviridae, subfamily pneumovirinae. Its name comes from the fact that F proteins on the surface of the virus cause the cell membranes on nearby cells to merge, forming syncytia.. Annexin II that binds to RSV G glycoprotein may be RSVs receptor on human cells (Malhotra et al., 2003). Human RSV is responsible for a spectrum of respiratory tract diseases in people of all ages throughout the world. It is the major cause of lower respiratory tract illness during infancy and childhood. Over half of all infants encounter RSV in their first year of life, and almost all within their first two years. The infection in young children can cause lung damage that persists for years and may contribute to chronic lung disease in later life (chronic wheezing, asthma). Older children and adults often suffer from a (bad) common cold upon RSV infection. In old age, susceptibility again increases, and RSV has been implicated in a number of outbreaks of pneumonia in the aged resulting in significant mortality.
Infection with a virus from a given subgroup does not protect against a subsequent infection with an RSV isolate from the same subgroup in the following winter season. Re- infection with RSV is thus common, despite the existence of only two subtypes, A and B.
So far, only three drugs have been approved for use against RSV infection. Ribavirin, a nucleoside analogue, provides an aerosol treatment for serious RSV infection in hospitalized children. The aerosol route of administration, the toxicity (risk of teratogenicity), the cost and the highly variable efficacy limit its use. The other two drugs, RespiGam.RTM. and palivizumab, polyclonal and monoclonal antibody immunostimulants, are intended to be used in a preventive way.
In addition, the present invention comprehends the use of more than therapeutic ligand, immunogen or antigen in the vectors and methods disclosed herein, delivered either in separate recombinant vectors, or together in one recombinant vector so as to provide a multivalent vaccine or immunogenic composition that stimulates or modulates immunogenic response to one or more influenza strains and/or hybrids. Further, the present invention encompasses the use of a therapeutic ligand, immunogen or antigen from more than one pathogen in the vectors and methods disclosed herein, delivered either in separate recombinant vectors, or together in one recombinant vector.
The recombinant vectors and methods of the invention comprehend the use of adjuvant molecules that can modulate immune responses upon delivery of recombinant vectors or pharmaceutical or immunogenic/immunological compositions. Such adjuvant molecules can include, but are not limited to, immunomodulatory molecules such as interleukins, interferon, and co-stimulatory molecules. The immunomodulatory molecules can be co-administered with the inventive pharmaceutical or immunogenic compositions, or alternatively, the nucleic acid of the immunomodulatory molecule(s) can be co-expressed along with the therapeutic ligands or immunogens or antigens in the recombinant vectors of the invention. Expression in the subject of the heterologous sequence can result in expression products which can bind to or block receptors on target cells. Consequently, respiratory pathogens are prevented from binding to these receptors themselves, and thereby cannot initiate infection. For example, expression products such as influenza HAl molecules may bind to sialic acid-containing receptors on target cells and prevent initiation of influenza virus infection. Partial reduction in the number of receptors in the upper respiratory tract is sufficient in alleviating transmission of influenza virus (Tumpey et al., 2007). Similarly, expression products such as SARS-CoV S protein molecules may bind to the angiotensin- converting enzyme 2 receptor on target cells and prevent SARS-COV infection (Li et al., 2003). In addition, expression products comprising ligands of ICAM-I may bind to the ICAM-I receptor on target cells and prevent infection of HRV (Rossmann et al., 2000).
Moreover, expression products such as RSV G glycoprotein may bind to annexin II on target cells (Malhotra et al., 2003) and prevent infection of RSV. Thus, the recombinant vectors of the present invention may be used in a vaccine or pharmaceutical to provide a means to inhibit or prevent respiratory pathogen infection. Target cells as used herein may be any cell involved in the pathogenicity of respiratory viruses, including, but not limited to, airway epithelial or mucosal cells.
Alternatively, expression in the subject of the heterologous sequence, i.e. therapeutic ligands or immunogens, can result in a response in the subject to the expression products of the therapeutic ligand or antigen or immunogen. Thus, the recombinant vectors of the present invention may be used in an immunological and/or therapeutic compositions to provide a means to confer protection against a pathogen. The molecular biology techniques used in the context of the invention are described by Sambrook et al. (2001).
Even further alternatively or additionally, in the pharmaceutical or immunological compositions encompassed by the present invention, the nucleotide sequence encoding the therapeutic ligands or antigens can have deleted therefrom a portion encoding a transmembrane domain. Yet even further alternatively or additionally, the vector or pharmaceutical or immunogenic composition can further contain and express in a host cell a nucleotide sequence encoding a heterologous tPA signal sequence such as human or avian tPA and/or a stabilizing intron, such as intron II of the rabbit β-globin gene. A vector can be administered to a subject in an amount to achieve the amounts stated for gene product (e.g., therapeutic, epitope, antigen, and/or antibody) compositions. Of course, the invention envisages dosages below and above those exemplified herein, and for any composition to be administered to a subject, including the components thereof, and for any particular method of administration, it is preferred to determine therefor: toxicity, such as by determining the lethal dose (LD) and LD50 in a suitable model; and the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response, such as by titrations of sera and analysis thereof, e.g., by ELISA and/or seroneutralization analysis. Such determinations do not require undue experimentation from the knowledge of the skilled artisan, this disclosure and the documents cited herein.
Subjects to which the recombinant vectors and/or pharmaceutical or immunological compositions can be administered include all animal species such as human, primate, feline, canine, avian, murine, bovine, equine, porcine, etc.
Examples of compositions of the invention include liquid preparations for orifice, or mucosal, e.g., intranasal, oral, anal, vaginal, peroral, intragastric, etc., administration such as suspensions, solutions, sprays, syrups or elixirs; and, preparations for parenteral, epicutaneous, subcutaneous (i.e., through lower neck), intradermal, intraperitoneal, intramuscular, intranasal, or intravenous administration (e.g., injectable administration) such as sterile suspensions or emulsions. Reference is made to U.S. Patent No. 6,716,823 issued April 6, 2004; U.S. Patent No. 6,706,693 issued March 16, 2004; U.S. Patent No. 6,348,450 issued February 19, 2002; U.S. Application Serial Nos. 10/052,323 and 10,116,963; and 10/346,021, the contents of which are incorporated herein by reference and which disclose immunization and delivery of pharmaceutical or immunogenic or vaccine compositions through a non-invasive mode of delivery, i.e. epicutaneous and intranasal administration.
The invention also comprehends sequential administration of inventive compositions or sequential performance of herein methods, e.g., periodic administration of inventive compositions such as in the course of therapy or treatment for a condition and/or booster administration of pharmaceutical or immunological compositions and/or in prime-boost regimens; and, the time and manner for sequential administrations can be ascertained without undue experimentation.
Further, the invention comprehends compositions and methods for making and using vectors, including methods for producing gene products and/or immunological products and/or antibodies in vivo and/or in vitro and/or ex vivo (e.g., the latter two being, for instance, after isolation therefrom from cells from a host that has had an administration according to the invention, e.g., after optional expansion of such cells), and uses for such gene and/or pharmaceutical or immunological products and/or antibodies, including in diagnostics, assays, therapies, treatments, and the like. Vector compositions are formulated by admixing the vector with a suitable carrier or diluent; and, gene product and/or pharmaceutical or immunological product and/or antibody compositions are likewise formulated by admixing the gene and/or immunological product and/or antibody with a suitable carrier or diluent; see, e.g., U.S. Patent No. 5,990,091, WO 99/60164, WO 98/00166, documents cited therein, and other documents cited herein, and other teachings herein (for instance, with respect to carriers, diluents and the like).
In such compositions, the recombinant vectors may be in admixture with a suitable veterinarily or pharmaceutically acceptable carrier, diluent, or excipient such as sterile water, physiological saline, glucose or the like. The compositions can also be lyophilized. The compositions can contain auxiliary substances, such as wetting or emulsifying agents, pH buffering agents, adjuvants, gelling or viscosity enhancing additives, preservatives, flavoring agents, colors, and the like, depending upon the route of administration and the preparation desired.
DMSO has been known to enhance the potency of vaccine and immunogenic compositions, particularly in regard to in ovo delivery of vectors or immunogenic compositions comprising vectors. DMSO is thought to enhance the potency of vaccines by increasing the permeability of cellular membranes (Oshop et al, 2003). Other agents or additives that are capable of permeabilizing cells, reducing the viscosity of amniotic fluid, and exhibiting a higher compliance rate as compared to DMSO can be used in the formulation of vaccines or immunogenic compositions, especially when administered by in ovo delivery. Absorption of a variety of proteins, such as insulin, leptin, and somatotropin, have been shown to be enhanced by surfactants such as tetradecyl maltoside (TDM) without appreciable side effects, following intranasal administration (Arnold, et al, 2004). The present invention therefore comprehends the use of TDM in the methods and compositions described herein.
Formulations containing 0.125% TDM can cause moderate alterations in cell morphology, while higher concentrations of TDM (i.e., 0.5%) can transiently induce more extensive morphological changes.
The quantity of vector to be administered will vary for the subject and condition being treated and will vary from one or a few to a few hundred or thousand micrograms of body weight per day and preferably the dose of vaccine or pharmaceutical or immunological composition being chosen preferably between 10 — 10 plaque forming units (PFU), preferably 102-1010 PFU per subject. For injection, vaccines containing the above titer should be diluted with a pharmaceutically or veterinarily acceptable liquid such as physiological saline to a final volume of approximately 0.5 ml or 0.01 ml.
A vector can be non-invasively administered to a subject in an amount to achieve the amounts stated for gene product (e.g., epitope, antigen, therapeutic, and/or antibody) compositions. Of course, the invention envisages dosages below and above those exemplified herein, and for any composition to be administered to a subject, including the components thereof, and for any particular method of administration, it is preferred to determine: toxicity, such as by determining the lethal dose (LD) and LD50 in a suitable model; and, the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable response, such as by titrations of sera and analysis thereof, e.g., by ELISA and/or seroneutralization analysis.
Such determinations do not require undue experimentation from the knowledge of the skilled artisan, this disclosure and the documents cited herein.
Recombinant vectors can be administered in a suitable amount to obtain in vivo expression corresponding to the dosages described herein and/or in herein cited documents. For instance, suitable ranges for viral suspensions can be determined empirically. If more than one gene product is expressed by more than one recombinant, each recombinant can be administered in these amounts; or, each recombinant can be administered such that there is, in combination, a sum of recombinants comprising these amounts.
In addition to recombinant vectors, the therapeutic effects can also be achieved by administration of receptor-binding ligands (purified receptor-binding proteins or killed viruses that display receptor-binding ligands or receptor-binding ligands embedded in virus- like particles).
In vector or ligand compositions employed in the invention, dosages can be as described in documents cited herein or as described herein or as in documents referenced or cited in herein cited documents. Advantageously, the dosage should be a sufficient amount of composition to confer an effect analogous to compositions wherein the ligand(s) are directly present; or to have expression analogous to dosages in such compositions; or to have expression analogous to expression obtained in vivo by recombinant compositions.
However, the dosage of the composition(s), concentration of components therein and timing of administering the composition(s), which elicit a suitable immunological response, can be determined by methods such as by antibody titrations of sera, e.g., by ELISA and/or seroneutralization assay analysis. Such determinations do not require undue experimentation from the knowledge of the skilled artisan, this disclosure and the documents cited herein. And, the time for sequential administrations can be likewise ascertained with methods ascertainable from this disclosure, and the knowledge in the art, without undue experimentation.
The pharmaceutical or immunological compositions contemplated by the invention can also contain an adjuvant. Suitable adjuvants include fMLP (N-formyl-methionyl-leucyl- phenylalanine; U.S. Patent No. 6,017,537) and/or acrylic acid or methacrylic acid polymer and/or a copolymer of maleic anhydride and of alkenyl derivative. The acrylic acid or methacrylic acid polymers can be cross-linked, e.g., with polyalkenyl ethers of sugars or of polyalcohols. These compounds are known under the term "carbomer" (Pharmeuropa, Vol. 8, No. 2, June 1996). A person skilled in the art may also refer to U.S. Patent No. 2,909,462 (incorporated by reference), which discusses such acrylic polymers cross-linked with a polyhydroxylated compound containing at least 3 hydroxyl groups: in one embodiment, a polyhydroxylated compound contains not more than 8 hydroxyl groups; in another embodiment, the hydrogen atoms of at least 3 hydroxyls are replaced with unsaturated aliphatic radicals containing at least 2 carbon atoms; in other embodiments, radicals contain from about 2 to about 4 carbon atoms, e.g., vinyls, allyls and other ethylenically unsaturated groups. The unsaturated radicals can themselves contain other substituents, such as methyl. The products sold under the name Carbopol® (Noveon Inc., Ohio, USA) are particularly suitable for use as an adjuvant. They are cross-linked with an allyl sucrose or with allylpentaerythritol, as to which, mention is made of the products Carbopol® 974P, 934P, and 971P.
As to the copolymers of maleic anhydride and of alkenyl derivative, mention is made of the EMA® products (Monsanto), which are copolymers of maleic anhydride and of ethylene, which may be linear or cross-linked, for example cross-linked with divinyl ether. Also, reference may be made to U.S. Patent No. 6,713,068 and Regelson, W. et al., 1960; incorporated by reference).
Cationic lipids containing a quaternary ammonium salt are described in U.S. Patent No. 6,713,068, the contents of which are incorporated by reference, can also be used in the methods and compositions of the present invention. Among these cationic lipids, preference is given to DMRIE (N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-l-propane ammonium; WO96/34109), advantageously associated with a neutral lipid, advantageously DOPE (dioleoyl-phosphatidyl-ethanol amine; Behr J. P., 1994), to form DMRIE-DOPE.
A recombinant vaccine or pharmaceutical or immunogenic or immunological composition can also be formulated in the form of an oil-in-water emulsion. The oil-in-water emulsion can be based, for example, on light liquid paraffin oil (European Pharmacopea type); isoprenoid oil such as squalane, squalene, EICOSANE ™ or tetratetracontane; oil resulting from the oligomerization of alkene(s), e.g., isobutene or decene; esters of acids or of alcohols containing a linear alkyl group, such as plant oils, ethyl oleate, propylene glycol di(caprylate/caprate), glyceryl tri(caprylate/caprate) or propylene glycol dioleate; esters of branched fatty acids or alcohols, e.g., isostearic acid esters. The oil advantageously is used in combination with emulsifiers to form the emulsion. The emulsifiers can be nonionic surfactants, such as esters of sorbitan, mannide (e.g., anhydromannitol oleate), glycerol, polyglycerol, propylene glycol, and oleic, isostearic, ricinoleic, or hydroxystearic acid, which are optionally ethoxylated, and polyoxypropylene-polyoxyethylene copolymer blocks, such as the Pluronic® products, e.g., L121. The adjuvant can be a mixture of emulsifier(s), micelle-forming agent, and oil such as that which is available under the name Provax® (IDEC Pharmaceuticals, San Diego, CA).
The viral vector expressing one or more antigen or ligand or immunogen of interest, e.g., vector according to this disclosure, can be preserved and/or conserved and stored either in liquid form, at about 50C, or in lyophilized or freeze-dried form, in the presence of a stabilizer. Freeze-drying can be according to well-known standard freeze-drying procedures. The pharmaceutically acceptable stabilizers may be SPGA (sucrose phosphate glutamate albumin; Bovarnick, et al., 1950), carbohydrates (e.g., sorbitol, mannitol, lactose, sucrose, glucose, dextran, trehalose), sodium glutamate (Tsvetkov, T. et al., 1983; Israeli, E. et al., 1993), proteins such as peptone, albumin or casein, protein containing agents such as skimmed milk (Mills, CK. et al., 1988; Wolff, E. et al., 1990), and buffers (e.g., phosphate buffer, alkaline metal phosphate buffer). An adjuvant and/or a vehicle or excipient may be used to make soluble the freeze-dried preparations.
The following examples are given for the purpose of illustrating various embodiments of the invention and are not meant to limit the present invention in any fashion.
EXAMPLES
Example 1: Development of the AdHi gh system for rapid generation of RCA-free Ad vectors in PER.C6 cells. Production of RCA-free Ad vectors through recombination in PER.C6 cells is a slow process. Generation of Ad vectors by AdEasy in 293 cells is fast; however, AdEasy is not compatible with the PER.C6 cell line for propagation of high-titer RCA-free Ad vectors. Hence, a new system named AdHigh was constructed by repairing the backbone of the AdEasy' s pShuttleCMV vector. pAdHigh retains the essential components for recombination with pAdEasyl in E. coli BJ5183 cells and received the sequence encompassing the pIX promoter region from Crucell's pAdApt. This new shuttle vector allows rapid recombination with pAdEasyl in E. coli BJ5183 cells and that the titer of Ad vectors produced in PER.C6 cells can achieve 109 ifu per ml in crude extracts before purification.
The low-titer production of AdEasy vectors in PER.C6 cells may be attributed to the lack of adenovirus nucleotides 3511-3533, as this segment is present in pAdApt (sequence information provided by Crucell) but missing in pShuttleCMV. The pIX promoter (Babiss and Vales, 1991) is intact in pAdApt but defective in pShuttleCMV. pIX participates in the stability of adenovirus particles as a capsid cement (Rosa-Calatrava et al., 2001). The approach that was used in generating pAdHigh by repairing pShuttleCMV is as follows. To repair the defective sequences, pShuttleCMV' s CMV promoter, the adjacent multiple cloning site, and flanking Ad sequences were replaced as one unit with their counterpart from pAdApt through homologous recombination. This was possible because these two shuttle vectors share extensive overlapping sequences. The full-length tetracycline (Tc) resistance gene (Backman and Boyer, 1983; Peden, 1983) was amplified from the plasmid pBR322 by polymerase chain reaction (PCR) using primers 5'- GAGCTCGGTACCTTCTCATGTTTGACAGCTTATCAT-3' and 5'- TCTAGAGGTACCAACGCTGCCCGAGATGCGCCGCGT-3' with built-in Kpnl sites. The amplified Tc gene was inserted into the Kpnl site of the ampicillin (Amp)-resistant plasmid pAdApt to generate a new plasmid ρAdAρt-Tc, which was selected by applying both Amp and Tc to the growth medium.
Next, the adenovirus sequences and expression cassette in pShuttleCMV were replaced with their counterparts in pAdApt using the high-efficiency AdEasier recombination protocol (Zeng et al., 2001). Briefly, pShuttleCMV was transformed into E. coli BJ5183 cells, and transformants were selected by kanamycin (Kan) resistance. Kan-resistant cells were immediately transformed with pAdApt-Tc, and recombinants were selected by applying both Kan and Tc. The recombinant can confer both Kan and Tc resistance to E. coli BJ5183 cells only when the defective Ad sequence in pShuttleCMV has been replaced through homologous recombination with its counterpart in pAdApt. The resultant pAdHigh-Tc was purified from E. coli BJ5183 cells and transformed into E. coli DHlOB cells. The plasmid was validated by DNA sequencing. Transgenes were subsequently inserted into convenient restriction sites of pAdHigh-Tc to replace the Tc gene. The resultant plasmid was allowed to recombine with the adenovirus backbone plasmid pAdEasyl in E. coli BJ5183 cells as described (Zeng et al., 2001). An adenovirus vector encoding an HAl was generated as rapidly as with the AdEasy system and produced a titer as high as that achieved by the Crucell's AdApt system after transfecting the recombinant plasmid into PER.C6 cells. Notably, the AdHigh-derived adenovirus vectors were RCA-free (Apptec reports on RCA analysis of Vaxin's AdHigh-derived adenovirus vectors). Example 2: Construction of RCA-free Ad vectors encoding HAl
The Centers for Disease Control and Prevention (CDC) provided the PI with the influenza virus strains A/New Caledonia/20/99 (HlNl), A/Panama/2007/99 (H3N2), and
B/Hong Kong/330/01 (strains selected for vaccine production in 2003-2004). Humanized versions of their HA genes were synthesized at GENEART with codons optimized to match the tRNA pool found in human cells. The A/New Caledonia/20/99 HAl fragment containing
347 amino acids was amplified by PCR using primers 5'-
CACAGGTACCGCCACCATGAAGGCCAAGCTG-3' and 5'-
GAGTCTAGATTATCAGCCGAACAGGCCTCTGCTCTGG-3'. These primers contain sequences that anneal to the 5' and 3' ends of the A/Caledonia/20/99 HA gene, an eukaryotic ribosomal binding site (Kozak, 1986) immediately upstream from the HA initiation ATG codon, and specific restriction sites for subsequent cloning. The Kpnl-Xbal fragment containing the HAl fragment was inserted into the Kpnl-Xbal site of pAdHigh (described above) in the correct orientation under transcriptional control of the human cytomegalovirus (CMV) early promoter. An RCA-free adenovirus vector encoding the A/New
Caledonia/20/99 HAl (AdNCHl.1) was generated in human PER.C6 cells as described above.
An RCA-free adenovirus vector (AdPNM.H3.1) encoding a humanized HAl fragment (349 amino acids) of the A/Panama/2007/99 (H3N2) influenza virus was generated in PER.C6 cells as described above. The full-length humanized HA template was synthesized at GENEART, and the HAl fragment was PCR amplified using primers 5'-
TTGGAAGCTTGCCACCATGAAAACCATCATC-3' and 5'-
GAGTCTAGATTATCAGCCGAAGATGCCCCGGGTCTGC-3'. The amplified HAl fragment was inserted into the Hindlll-Xbal site of pAdHigh. An RCA-free Ad vector (AdHK.B.1) encoding a humanized HAl fragment (370 amino acids) of the A/Hong Kong/330/01 influenza virus was generated in PER.C6 cells as described above. The full-length humanized HA template was synthesized at GENEART, and the HAl fragment was PCR amplified using primers 5'-
TACCAAGCTTGCCACCATGAAGGCCATCATC-3' and 5'- GAGTCTAGATTATCAGCCGGCGATGGCGCCGAAG-S'. The amplified HAl fragment was inserted into the Hindlll-Xbal site of pAdHigh.
An RCA-free Ad vector (AdVN.H5.1) encoding a humanized HAl fragment (340 amino acids) of the A/Vietnam/ 1203/04 (H5N1) avian influenza virus was generated in
PER.C6 cells as described above. The full-length humanized HA template was synthesized at GENEART, and the HAl fragment was PCR amplified using primers 5'- TGCATTGGAAGCTTGCCACCATGGAGAAGA-3' and 5'-
GAGTCTAGATTATCACTCCCGCTGGGGGCTGTTC-3'. The amplified HAl fragment was inserted into the Hindlll-Xbal site of pAdHigh. All adenovirus vectors encoding HAl were validated by sequencing both 5' and 3' junctions between the HAl insert and the vector backbone.
Example 3: Intranasal administration of AdNCHl.1 as a novel anti-influenza drug
It is hypothesized that sufficient amounts of HAl, expressed from adenovirus vectors following intranasal instillation and subsequently secreted from transduced cells, may bind to sialic acid-containing receptors in the upper respiratory tract and may block a productive infection by influenza virus during a challenge, as a partial reduction of receptors in the upper respiratory tract may alleviate transmission of influenza virus (Tumpey et al., 2007). This hypothesis was tested by instilling an escalating dose of AdNCHl.1 into the nostril of mice at different time points prior to challenge with a lethal dose of A/Puerto Rico/8/34 (HlNl) influenza virus. As shown in Figure 1, 100% of mice were protected by intranasal administration of 1 X 105 or 1 X 107 ifu of AdNCH 1.1 vectors one day prior to challenge, or by intranasal administration of 1 X 107 ifu of AdNC .Hl.1 vectors three days prior to challenge whereas control mice all died within 13 days post-challenge. Since one and three days are too short for eliciting an adaptive immune response against HAl , the protection may have been mediated by interference with influenza virus infection through HAl binding to sialic acid-containing receptors in the upper respiratory tract.
Partial protection against influenza could be achieved by intranasal instillation of the HAl gene a few days post-challenge (Figure 2). Results corroborate the hypothesis that protection conferred by AdNCHl .1 vectors is not mediated by elicitation of adaptive immunity against HAl .
Example 4: Intranasal administration of Ad vectors encoding HAl as a vaccination modality Intranasal administration of adenovirus- vectored vaccines is an effective approach for immunizing animals (Hoelscher et al., 2006; Shi et al., 2001; Xiang et al., 1996) and humans (Van Kampen et al., 2005). Intranasal instillation of an adenovirus vector encoding HAl was as effective as its counterpart containing both HAl and HA2 in eliciting protective immunity against a virulent HPAI virus in mice (Figure 3) and ferrets (Figure 4), respectively. Results suggest that this regimen may be utilized as an anti-influenza drug- vaccine multipurpose treatment. In addition to influenza, this regimen ought to be effective in protecting animals and humans against other pathogens that require receptor binding to initiate an infection in a wide variety of disease settings.
Example 5: Determination of the minimal dose of AdNCH 1.1 vectors in pre-challenge administration for preventing influenza in mice.
Balb/c mice are treated with an escalating dose of AdNCH 1.1 vectors at different time points prior to influenza virus challenge in order to determine the minimal dose required for preventing influenza. Since preexisting immunity to Ad is commonly found in humans and the potency of adenovirus- vectored nasal vaccine may not be appreciably interfered by preexisting immunity to adenovirus (Hoelscher et al., 2006; Shi et al., 2001 ; Van Kampen et al., 2005), all mice are primed by intranasal instillation of 1 X 107 ifu of wild-type adenovirus serotype 5 one month prior to adenovirus treatment. Control and treated mice are challenged by intranasal instillation of lOXLDso of A/Puerto Rico/8/34 (HlNl) influenza virus. Challenged animals are monitored for survival for 30 days. There are 10 mice per group, and each experiment is repeated once (Table 1).
Table 1. Minimal dose of AdNCHl.1 vectors required in pre-challenge administration for preventing influenza as an anti-influenza drug
Figure imgf000044_0001
Example 6: Determining the minimal dose of AdNCHl.1 vectors in post-challenge administration for alleviating influenza in mice.
Mice are treated with an escalating dose of AdNCH 1.1 at different time points post- challenge to determine the minimal dose required for alleviating lethal symptoms of influenza. All mice in these studies are primed and challenged as described above. There are 10 mice per group, and each experiment is repeated once (Table 2).
Example 7: Determining the potential of the HAl drug in providing a broad protection against different influenza virus strains.
Although expression of the A/New Caledonia/20/99 (HlNl) HAl protected mice against another HlNl strain (Example 4), it is not clear whether this regimen may confer a broad protection against other non-HINl strains, particularly against H3N2 and B strains
Table 2. Minimal dose of AdNCHl.1 vectors required in post-challenge administration for alleviating influenza as an anti-influenza drug
Figure imgf000045_0001
that may enter cells through different receptors. Here, Balb/c mice are treated with AdNCHl.1 vectors encoding an Hl HAl followed by challenge with A/Puerto Rico/8/34 (HlNl) and A/Hong Kong/8/68 (H3N2) influenza virus strains as described (Ulmer et al., 1993) one day later. Mice are also treated with AdPNM.H3.1 encoding an H3 HAl (Example T), AdHK.B.l encoding an influenza virus B HAl (Example T), and AdVN.H5.1 encoding an H5 HAl (Example T), followed by challenge with A/Puerto Rico/8/34 (HlNl) and A/Hong Kong/8/68 (H3N2) influenza viruses, respectively, in an attempt to determine the cross-reactivity between a specific HAl and receptors utilized by a variety of influenza virus strains. All mice are primed and challenged as described above. There are 10 mice per group, and each experiment is repeated once (Table 3).
Table 3. Cross-reactivity between a specific HAl and receptors utilized by a variety of influenza virus strains
Figure imgf000047_0001
Example 8: Construction of an Ad vector encoding the HAl domain of the A/Puerto Rico/8/34 influenza virus and evaluation of HAl as a drug- vaccine multipurpose agent.
An RCA-free Ad vector (AdPR8.Hl.l) encoding a humanized HAl domain of the A/Puerto Rico/8/34 influenza virus is generated in PER.C6 cells as described in Example 2. Mice are immunized by intranasal instillation of an escalating dose of AdPRδ.Hl.l vectors encoding the HAl domain of the A/Puerto Rico/8/34 (HlNl) influenza virus, or AdNCHl.1 vectors encoding the HAl domain of the A/New Caledonia/20/99 (HlNl) influenza virus, in a single-dose regimen. After one month, control and immunized mice will be challenged by A/Puerto Rico/8/34 (HlNl) and A/Hong Kong/8/68 (H3N2) influenza virus strains, respectively. All mice are primed and challenged as described above. There are 10 mice per group, and each experiment is repeated once (Table 4).
Table 4. Evaluation of intranasally-administered HAl as an anti-influenza drug and vaccine
Figure imgf000048_0001
The invention is further described by the following numbered paragraphs:
1. A therapeutic-immunological composition, comprising a pathogen-derived ligand, wherein (i) the ligand binds to its receptor on target cells;
(ii) confers in an animal rapid, therapeutic effect; and (iii) elicits in the animal long-term protective immunity against the pathogen.
2. The therapeutic-immunological composition of claim 1 , wherein the virus- derived ligand is expressed by a viral vector. 3. The therapeutic-immunological composition of claim 2, wherein the viral vector is selected from the group consisting of an adenovirus vector, an adeno-associated virus vector, a herpes simplex virus vector, an alphavirus vector, and a retrovirus vector.
4. The therapeutic-immunological composition of claim 3, wherein the viral vector is a recombinant viral vector.
5. The therapeutic-immunological composition of claim 4, wherein the recombinant viral vector is a recombinant adenovirus vector.
6. The therapeutic-immunological composition of claim 5, wherein the recombinant adenovirus vector is derived from adenovirus serotype 5 (Ad5). 7. The therapeutic-immunological composition of claim 3, wherein the adenovirus vector is selected from the group consisting of replication-defective adenovirus, non-replicating adenovirus, replication-competent adenovirus, or wild-type adenovirus.
8. The therapeutic-immunological composition of claim 1 or 2, wherein the pathogen from which the ligand is derived is selected from the group consisting of influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS-CoV), human rhinovirus (HRV), and respiratory syncytial virus (RSV).
9. The therapeutic-immunological composition of claim 8, wherein the pathogen is influenza virus.
10. The therapeutic-immunological composition of claim 9, wherein the pathogen- derived ligand binds to a sialic acid-containing receptor.
11. The therapeutic-immunological composition of claim 9, wherein the pathogen- derived ligand is hemagglutinin protein.
12. The therapeutic-immunological composition of claim 11 , wherein the hemagglutinin protein is selected from the group consisting of hemagglutinin subtype 1, 2, 3, 5, or B.
13. The therapeutic-immunological composition of claim 12, wherein the hemagglutinin protein is hemagglutinin HAl domain.
14. The therapeutic-immunological composition of claim 13, wherein HAl is derived from a strain selected from the group consisting of A/New Caledonia/20/99, A/Panama/2007/99, B/Hong Kong/330/01, A/Vietnam/I 203/04, A/Puerto Rico/8/34, A/Hong Kong/8/68, and 2009H1N1 swine flu.
15. The therapeutic-immunological composition of claim 8, wherein the pathogen is SARS-CoV. 16. The therapeutic-immunological composition of claim 15, wherein the pathogen-derived ligand binds to an angiotensin-converting enzyme 2 receptor.
17. The therapeutic-immunological composition of claim 15, wherein the pathogen-derived ligand is a SARS-CoV S protein. 18. The therapeutic-immunological composition of claim 8, wherein the pathogen is HRV.
19. The therapeutic-immunological composition of claim 18, wherein the pathogen-derived ligand binds to inter-cellular adhesion molecule 1.
20. The therapeutic-immunological composition of claim 8, wherein the pathogen is RSV.
21. The therapeutic-immunological composition of claim 20, wherein the pathogen-derived ligand binds to annexin II.
22. A method of conferring in a subject rapid protection against a virus and eliciting in the subject long-term protective immunity against the virus, comprising: intranasally administering to the subject a therapeutically effective amount of the immunological composition of claim 1 or 2.
23. The method of claim 22, wherein the viral vector is selected from the group consisting of an adenovirus vector, an adeno-associated virus vector, a herpes simplex virus vector an alphavirus vector, and a retrovirus vector.
24. The method of claim 23, wherein the viral vector is a recombinant viral vector.
25. The method of claim 24, wherein the recombinant viral vector is a recombinant adenovirus vector.
26. The method of claim 25, wherein the recombinant adenovirus vector is derived from adenovirus serotype 5 (Ad5).
27. The method of claim 26, wherein the adenovirus vector is selected from the group consisting of replication-defective adenovirus, non-replicating adenovirus, replication- competent adenovirus, or wild-type adenovirus.
28. The method of claim 22, wherein the pathogen from which the ligand is derived is selected from the group consisting of influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS-CoV), human rhinovirus (HRV), and respiratory syncytial virus (RSV).
29. The method of claim 28, wherein the pathogen is influenza virus. 30. The method of claim 28, wherein the pathogen-derived ligand binds to a sialic acid-containing receptor.
30. The method of claim 28, wherein the pathogen-derived ligand is hemagglutinin protein. 31. The method of claim 30, wherein the hemagglutinin protein is selected from the group consisting of hemagglutinin subtype 1, 2, 3, 5, or B.
32. The method of claim 31 , wherein the hemagglutinin protein is hemagglutinin HAl domain.
33. The method of claim 32, wherein HAl is derived from a strain selected from the group consisting of A/New Caledonia/20/99, A/Panama/2007/99, B/Hong Kong/330/01 ,
A/Vietoam/1203/04, A/Puerto Rico/8/34, A/Hong Kong/8/68, and 2009H1N1.
34. The method of claim 28, wherein the pathogen is SARS-CoV.
35. The method of claim 34, wherein the pathogen-derived ligand binds to an angjotensin-converting enzyme 2 receptor. 36. The method of claim 34, wherein the pathogen-derived ligand is a SARS-
CoV S protein.
37. The method of claim 28, wherein the pathogen is HRV.
38. The method of claim 37, wherein the pathogen-derived ligand binds to intercellular adhesion molecule 1. 39. The method of claim 28, wherein the pathogen is RSV.
40. The method of claim 39, wherein the pathogen-derived ligand binds to annexin II.
* * * Having thus described in detail preferred embodiments of the present invention, it is to be understood that the invention defined by the appended claims is not to be limited by particular details set forth in the above description as many apparent variations thereof are possible without departing from the spirit or scope thereof. References cited:
Aoki, F. Y., Boivin, G., and Roberts, N. (2007). Influenza virus susceptibility and resistance to oseltamivir. Antivir Ther 12, 603-616. Babiss, L. E., and Vales, L. D. (1991). Promoter of the adenovirus polypeptide IX gene: similarity to ElB and inactivation by substitution of the simian virus 40 TATA element. J
Virol 65, 598-605.
Backman, K., and Boyer, H. W. (1983). Tetracycline resistance determined by pBR322 is mediated by one polypeptide. Gene 26, 197-203. Beigel, J., and Bray, M. (2008). Current and future antiviral therapy of severe seasonal and avian influenza. Antiviral Res 78, 91-102.
He, T. C, Zhou, S., da Costa, L. T., Yu, J., Kinzler, K. W., and Vogelstein, B. (1998). A simplified system for generating recombinant adenoviruses. Proc Natl Acad Sci USA 95,
2509-2514. Hilleman, M. R. (2002). Realities and enigmas of human viral influenza: pathogenesis, epidemiology and control. Vaccine 20, 3068-3087.
Hoelscher, M. A., Garg, S., Bangari, D. S., Belser, J. A., Lu, X., Stephenson, L, Bright, R. A.,
Katz, J. M., Mittal, S. K., and Sambhara, S. (2006). Development of adenoviral-vector-based pandemic influenza vaccine against antigenically distinct human H5N1 strains in mice. Lancet 367, 475-481.
Hoffmann, E., Krauss, S., Perez, D., Webby, R., and Webster, R. G. (2002). Eight-plasmid system for rapid generation of influenza virus vaccines. Vaccine 20, 3165-3170.
Kozak, M. (1986). Point mutations define a sequence flanking the AUG initiator codon that modulates translation by eukaryotic ribosomes. Cell 44, 283-292. Malhotra, R., Ward, M., Bright, H., Priest, R., Foster, M. R., Hurle, M., Blair, E., and Bird,
M. (2003). Isolation and characterisation of potential respiratory syncytial virus receptor(s) on epithelial cells. Microbes Infect 5, 123-133.
Marwick, C. (2000). Merits, flaws of live virus flu vaccine debated. JAMA 283, 1814-1815.
Moscona, A. (2008). Medical management of influenza infection. Annu Rev Med 59, 397- 413.
Neumann, G., Watanabe, T., Ito, H., Watanabe, S., Goto, H., Gao, P., Hughes, M., Perez, D.
R., Donis, R., Hoffmann, E., et al. (1999). Generation of influenza A viruses entirely from cloned cDNAs. Proc Natl Acad Sci USA 96, 9345-9350. Nichol, K. L., Lind, A., Margolis, K. L., Murdoch, M., McFadden, R., Hauge, M., Magnan,
S., and Drake, M. (1995). The effectiveness of vaccination against influenza in healthy, working adults. N Engl J Med 333, 889-893.
Nolan, T., Lee, M. S., Cordova, J. M., Cho, L, Walker, R. E., August, M. J., Larson, S., Coelingh, K. L., and Mendelman, P. M. (2003). Safety and immunogenicity of a live- attenuated influenza vaccine blended and filled at two manufacturing facilities. Vaccine 21,
1224-1231.
Peden, K. W. (1983). Revised sequence of the tetracycline-resistance gene of pBR322. Gene
22, 277-280. Pfleiderer, M., Lower, J., and Kurth, R. (2001). Cold-attenuated live influenza vaccines, a risk-benefit assessment. Vaccine 20, 886-894.
Reece, P. A. (2007). Neuraminidase inhibitor resistance in influenza viruses. J Med Virol 79,
1577-1586.
Rosa-Calatrava, M., Grave, L., Puvion-Dutilleul, F., Chatton, B., and Kedinger, C. (2001). Functional analysis of adenovirus protein IX identifies domains involved in capsid stability, transcriptional activity, and nuclear reorganization. J Virol 75, 7131-7141.
Shi, Z., Zeng, M., Yang, G., Siegel, F., Cain, L. J., Van Kampen, K. R., Elmets, C. A., and
Tang, D. C. (2001). Protection against tetanus by needle-free inoculation of adenovirus- vectored nasal and epicutaneous vaccines. J Virol 75, 11474-11482. Subbarao, K., Klimov, A., Katz, J., Regnery, H., Lim, W., Hall, H., Perdue, M., Swayne, D.,
Bender, C, Huang, J., et al. (1998). Characterization of an avian influenza A (H5N1) virus isolated from a child with a fatal respiratory illness. Science 279, 393-396.
Tang, D. C, and Van Kampen, K. R. (2008). Toward the development of vectored vaccines in compliance with evolutionary medicine. Expert Rev Vaccines 7, 399-402. Tang, D. C, Zhang, J., Toro, H., Shi, Z., and Van Kampen, K. R. (2009). Adenovirus as a carrier for the development of influenza virus-free avian influenza vaccines. Expert Rev
Vaccines 8, 469-481.
Tumpey, T. M., Maines, T. R., Van Hoeven, N., Glaser, L., Solόrzano, A., Pappas, C, Cox,
N. J., Swayne, D. E., Palese, P., Katz, J. M., and Garcia-Sastre, A. (2007). A two-amino acid change in the hemagglutinin of the 1918 influenza virus abolishes transmission. Science 315,
655-659.
Ulmer, J. B., Donnelly, J. J., Parker, S. E., Rhodes, G. H., Feigner, P. L., Dwarki, V. J.,
Gromkowski, S. H., Deck, R. R., DeWitt, C. M., Friedman, A., etal. (1993). Heterologous protection against influenza by injection of DNA encoding a viral protein. Science 259, 1745-
1748.
Van Kampen, K. R., Shi, Z., Gao, P., Zhang, J., Foster, K. W., Chen, D. T., Marks, D.,
Elmets, C. A., and Tang, D. C. (2005). Safety and immunogenicity of adenovirus- vectored nasal and epicutaneous influenza vaccines in humans. Vaccine 23, 1029-1036.
Xiang, Z. Q., Yang, Y., Wilson, J. M., and Ertl, H. C. (1996). A replication-defective human adenovirus recombinant serves as a highly efficacious vaccine carrier. Virology 219, 220-
227.
Yen, H. L., Ilyushina, N. A., Salomon, R., Hoffmann, E., Webster, R. G., and Govorkova, E. A. (2007). Neuraminidase inhibitor-resistant recombinant A/Vietnam/ 1203/04 (H5N1) influenza viruses retain their replication efficiency and pathogenicity in vitro and in vivo. J
Virol 57, 12418-12426.
Zeng, M., Smith, S. K., Siegel, F., Shi, Z., Van Kampen, K. R., Elmets, C. A., and Tang, D.
C. (2001). AdEasy system made easier by selecting the viral backbone plasmid preceding homologous recombination. Biotechniques 31, 260-262.

Claims

WHAT IS CLAIMED IS:
1. A therapeutic-immunological composition, comprising a pathogen-derived ligand, wherein
(i) the ligand binds to its receptor on target cells; (ii) confers in an animal rapid, therapeutic effect; and
(iii) elicits in the animal long-term protective immunity against the pathogen.
2. The therapeutic-immunological composition of claim 1 , wherein the virus- derived ligand is expressed by a viral vector.
3. The therapeutic-immunological composition of claim 2, wherein the viral vector is selected from the group consisting of an adenovirus vector, an adeno-associated virus vector, a herpes simplex virus vector, an alphavirus vector, and a retrovirus vector.
4. The therapeutic-immunological composition of claim 3, wherein the viral vector is a recombinant viral vector.
5. The therapeutic-immunological composition of claim 4, wherein the recombinant viral vector is a recombinant adenovirus vector.
6. The therapeutic-immunological composition of claim 5, wherein the recombinant adenovirus vector is derived from adenovirus serotype 5 (Ad5).
7. The therapeutic-immunological composition of claim 3, wherein the adenovirus vector is selected from the group consisting of replication-defective adenovirus, non-replicating adenovirus, replication-competent adenovirus, or wild-type adenovirus.
8. The therapeutic-immunological composition of claim 1 or 2, wherein the pathogen from which the ligand is derived is selected from the group consisting of influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS-CoV), human rhinovirus (HRV), and respiratory syncytial virus (RSV).
9. The therapeutic-immunological composition of claim 8, wherein the pathogen is influenza virus.
10. The therapeutic-immunological composition of claim 9, wherein the pathogen- derived ligand binds to a sialic acid-containing receptor.
11. The therapeutic-immunological composition of claim 9, wherein the pathogen- derived ligand is hemagglutinin protein.
12. The therapeutic-immunological composition of claim 11, wherein the hemagglutinin protein is selected from the group consisting of hemagglutinin subtype 1, 2, 3, 5, or B.
13. The therapeutic-immunological composition of claim 12, wherein the hemagglutinin protein is hemagglutinin HAl domain.
14. The therapeutic-immunological composition of claim 13, wherein HAl is derived from a strain selected from the group consisting of A/New Caledonia/20/99, A/Panama/2007/99, B/Hong Kong/330/01, A/Vietnam/I 203/04, A/Puerto Rico/8/34, A/Hong Kong/8/68, and 2009H1N1 swine flu.
15. The therapeutic-immunological composition of claim 8, wherein the pathogen is SARS-CoV.
16. The therapeutic-immunological composition of claim 15, wherein the pathogen-derived ligand binds to an angiotensin-converting enzyme 2 receptor.
17. The therapeutic-immunological composition of claim 15, wherein the pathogen-derived ligand is a SARS-CoV S protein.
18. The therapeutic-immunological composition of claim 8, wherein the pathogen is HRV.
19. The therapeutic-immunological composition of claim 18, wherein the pathogen-derived ligand binds to inter-cellular adhesion molecule 1.
20. The therapeutic-immunological composition of claim 8, wherein the pathogen is RSV.
21. The therapeutic-immunological composition of claim 20, wherein the pathogen-derived ligand binds to annexin II.
22. A method of conferring in a subject rapid protection against a virus and eliciting in the subject long-term protective immunity against the virus, comprising: intranasally administering to the subject a therapeutically effective amount of the immunological composition of claim 1 or 2.
23. The method of claim 22, wherein the viral vector is selected from the group consisting of an adenovirus vector, an adeno-associated virus vector, a herpes simplex virus vector an alphavirus vector, and a retrovirus vector.
24. The method of claim 23, wherein the viral vector is a recombinant viral vector.
25. The method of claim 24, wherein the recombinant viral vector is a recombinant adenovirus vector.
26. The method of claim 25, wherein the recombinant adenovirus vector is derived from adenovirus serotype 5 (Ad5).
27. The method of claim 26, wherein the adenovirus vector is selected from the group consisting of replication-defective adenovirus, non-replicating adenovirus, replication- competent adenovirus, or wild-type adenovirus.
28. The method of claim 22, wherein the pathogen from which the ligand is derived is selected from the group consisting of influenza virus, severe acute respiratory syndrome-associated coronavirus (SARS-CoV), human rhinovirus (HRV), and respiratory syncytial virus (RSV).
29. The method of claim 28, wherein the pathogen is influenza virus.
30. The method of claim 28, wherein the pathogen-derived ligand binds to a sialic acid-containing receptor.
30. The method of claim 28, wherein the pathogen-derived ligand is hemagglutinin protein.
31. The method of claim 30, wherein the hemagglutinin protein is selected from the group consisting of hemagglutinin subtype 1, 2, 3, 5, or B.
32. The method of claim 31 , wherein the hemagglutinin protein is hemagglutinin HAl domain.
33. The method of claim 32, wherein HAl is derived from a strain selected from the group consisting of A/New Caledonia/20/99, A/Panama/2007/99, B/Hong Kong/330/01, A/Vietnam/I 203/04, A/Puerto Rico/8/34, A/Hong Kong/8/68, and 2009H1N1.
34. The method of claim 28, wherein the pathogen is SARS-CoV.
35. The method of claim 34, wherein the pathogen-derived ligand binds to an angiotensin-converting enzyme 2 receptor.
36. The method of claim 34, wherein the pathogen-derived ligand is a SARS- CoV S protein.
37. The method of claim 28, wherein the pathogen is HRV.
38. The method of claim 37, wherein the pathogen-derived ligand binds to intercellular adhesion molecule 1.
39. The method of claim 28, wherein the pathogen is RSV.
40. The method of claim 39, wherein the pathogen-derived ligand binds to annexin II.
PCT/US2009/046132 2008-06-03 2009-06-03 Intranasal administration of receptor-binding ligands or genes encoding such ligands as a therapeutic regimen for mitigating infections caused by respiratory pathogens WO2010044921A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP09820933A EP2296700A2 (en) 2008-06-03 2009-06-03 Intranasal administration of receptor-binding ligands or genes encoding such ligands as a therapeutic regimen for mitigating infections caused by respiratory pathogens

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5841308P 2008-06-03 2008-06-03
US61/058,413 2008-06-03

Publications (2)

Publication Number Publication Date
WO2010044921A2 true WO2010044921A2 (en) 2010-04-22
WO2010044921A3 WO2010044921A3 (en) 2010-07-15

Family

ID=42107118

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/046132 WO2010044921A2 (en) 2008-06-03 2009-06-03 Intranasal administration of receptor-binding ligands or genes encoding such ligands as a therapeutic regimen for mitigating infections caused by respiratory pathogens

Country Status (2)

Country Link
EP (1) EP2296700A2 (en)
WO (1) WO2010044921A2 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012145572A1 (en) * 2011-04-20 2012-10-26 The Trustees Of The University Of Pennsylvania Regimens and compositions for aav-mediated passive immunization of airborne pathogens
US10786568B2 (en) 2017-02-28 2020-09-29 The Trustees Of The University Of Pennsylvania AAV mediated influenza vaccines
WO2021224946A1 (en) * 2020-05-07 2021-11-11 Bharat Biotech International Limited Coronavirus vaccine through nasal immunization
WO2021211629A3 (en) * 2020-04-13 2021-12-16 Massachusetts Eye And Ear Infirmary Methods of making and using a vaccine against coronavirus
WO2021254476A1 (en) * 2020-06-19 2021-12-23 南京金斯瑞生物科技有限公司 Magnetic microparticle chemiluminescence reagent kit for detecting sars-cov-2 virus neutralising antibodies and application therefor
WO2022077593A1 (en) * 2020-10-15 2022-04-21 广州达博生物制品有限公司 Sars-cov-2 coronavirus vaccine and preparation method therefor
WO2022155476A1 (en) * 2021-01-15 2022-07-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Replication-competent adenovirus type 4 sars-cov-2 vaccines and their use
US11535665B2 (en) 2015-05-13 2022-12-27 The Trustees Of The University Of Pennsylvania AAV-mediated expression of anti-influenza antibodies and methods of use thereof
EP3953457A4 (en) * 2019-04-06 2023-01-11 Altimmune Inc Broad and long-lasting influenza vaccine
US11578341B2 (en) 2017-02-28 2023-02-14 The Trustees Of The University Of Pennsylvania Compositions useful in treatment of spinal muscular atrophy
US11827906B2 (en) 2017-02-28 2023-11-28 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) clade f vector and uses therefor

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001029054A2 (en) * 1999-10-21 2001-04-26 Glaxo Group Limited Medicaments for treatment of respiratory syncytial virus infections
US20050222264A1 (en) * 2002-05-08 2005-10-06 Unihart Corporation Anti-rhinovirus active agents
US20060093616A1 (en) * 2004-09-29 2006-05-04 Ralf Altmeyer Process for vaccinating eucaryotic hosts and for protecting against SARS-CoV infection
US20070003576A1 (en) * 2004-12-09 2007-01-04 Andrea Gambotto Vaccines for the rapid response to pandemic avian influenza

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001029054A2 (en) * 1999-10-21 2001-04-26 Glaxo Group Limited Medicaments for treatment of respiratory syncytial virus infections
US20050222264A1 (en) * 2002-05-08 2005-10-06 Unihart Corporation Anti-rhinovirus active agents
US20060093616A1 (en) * 2004-09-29 2006-05-04 Ralf Altmeyer Process for vaccinating eucaryotic hosts and for protecting against SARS-CoV infection
US20070003576A1 (en) * 2004-12-09 2007-01-04 Andrea Gambotto Vaccines for the rapid response to pandemic avian influenza

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
GAO ET AL: 'Protection of mice and poultry from lethal H5N1 avian influenza virus through adenovirus-based immunization' J. VIROL. vol. 80, no. 4, February 2006, pages 1959 - 1964, XP002400987 *
HOELSCHER ET AL: 'A broadly protective vaccine against globally dispersed clade 1 and clade 2 H5N1 influenza viruses' J. INFECT. DIS. vol. 197, no. 8, 15 April 2008, pages 1185 - 1188, XP008138968 *
SEE ET AL: 'Comparative evaluation of two severe acute respiratory syndrome (SARS) vaccine candidates in mice challenged with SARS coronavirus' J. GEN. VIROL. vol. 87, no. 3, March 2006, pages 641 - 650, XP008138952 *
SINGH ET AL: 'Bovine adenoviral vector-based H5N1 influenza vaccine overcomes exceptionally high levels of pre-existing immunity against human adenovirus' MOL. THER. vol. 16, no. 5, May 2008, pages 965 - 971, XP008138961 *
TORO ET AL: 'Protection of chickens against avian influenza with non-replicating adenovirus- vectored vaccine' VACCINE vol. 26, no. 21, 19 May 2008, pages 2640 - 2646, XP022639508 *
YU ET AL: 'Single intranasal immunization with recombinant adenovirus-based vaccine induces protective immunity against respiratory syncytial virus infection' J. VIROL. vol. 82, no. 5, March 2008, pages 2350 - 2357, XP008138949 *
ZAKHARTCHOUK ET AL: 'Severe acute respiratory syndrome coronavirus nucleocapsid protein expressed by an adenovirus vector is phosphorylated and immunogenic in mice' J. GEN. VIROL. vol. 86, no. 1, January 2005, pages 211 - 215, XP008138950 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012145572A1 (en) * 2011-04-20 2012-10-26 The Trustees Of The University Of Pennsylvania Regimens and compositions for aav-mediated passive immunization of airborne pathogens
EP3699286A1 (en) * 2011-04-20 2020-08-26 The Trustees of the University of Pennsylvania Regimens and compositions for aav-mediated passive immunization of airborne pathogens
US11535665B2 (en) 2015-05-13 2022-12-27 The Trustees Of The University Of Pennsylvania AAV-mediated expression of anti-influenza antibodies and methods of use thereof
US10786568B2 (en) 2017-02-28 2020-09-29 The Trustees Of The University Of Pennsylvania AAV mediated influenza vaccines
US11578341B2 (en) 2017-02-28 2023-02-14 The Trustees Of The University Of Pennsylvania Compositions useful in treatment of spinal muscular atrophy
US11827906B2 (en) 2017-02-28 2023-11-28 The Trustees Of The University Of Pennsylvania Adeno-associated virus (AAV) clade f vector and uses therefor
EP3953457A4 (en) * 2019-04-06 2023-01-11 Altimmune Inc Broad and long-lasting influenza vaccine
WO2021211629A3 (en) * 2020-04-13 2021-12-16 Massachusetts Eye And Ear Infirmary Methods of making and using a vaccine against coronavirus
WO2021224946A1 (en) * 2020-05-07 2021-11-11 Bharat Biotech International Limited Coronavirus vaccine through nasal immunization
WO2021254476A1 (en) * 2020-06-19 2021-12-23 南京金斯瑞生物科技有限公司 Magnetic microparticle chemiluminescence reagent kit for detecting sars-cov-2 virus neutralising antibodies and application therefor
WO2022077593A1 (en) * 2020-10-15 2022-04-21 广州达博生物制品有限公司 Sars-cov-2 coronavirus vaccine and preparation method therefor
WO2022155476A1 (en) * 2021-01-15 2022-07-21 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Replication-competent adenovirus type 4 sars-cov-2 vaccines and their use

Also Published As

Publication number Publication date
WO2010044921A3 (en) 2010-07-15
EP2296700A2 (en) 2011-03-23

Similar Documents

Publication Publication Date Title
US20200338188A1 (en) Immunizations of avians by administration of non-replicating vectored vaccines
WO2010044921A2 (en) Intranasal administration of receptor-binding ligands or genes encoding such ligands as a therapeutic regimen for mitigating infections caused by respiratory pathogens
US20090175897A1 (en) System for rapid production of high-titer and replication-competent adenovirus-free recombinant adenovirus vectors
US20110268762A1 (en) Immunization of avians by mucosal administration of non-replicating vectored vaccines
JP2012527232A (en) Genetically modified vaccinia Ankara virus (MVA) based influenza universal vaccine
EP3341018A1 (en) Generation of infectious influenza viruses from virus-like particles
Cheng et al. Chimpanzee adenovirus vector-based avian influenza vaccine completely protects mice against lethal challenge of H5N1
Holman et al. Multi-antigen vaccines based on complex adenovirus vectors induce protective immune responses against H5N1 avian influenza viruses
Park et al. Mucosal immunity induced by adenovirus-based H5N1 HPAI vaccine confers protection against a lethal H5N2 avian influenza virus challenge
US20240091336A1 (en) Universal influenza vaccine
RU2618918C2 (en) Universal anti-infectious vaccine
US9220769B2 (en) Composition
He et al. Adenovirus-based vaccines against avian-origin H5N1 influenza viruses
Sayedahmed Adenovirus Vector-based Vaccine Approach for Emerging Influenza Viruses
WO2023019274A1 (en) Methods and compositions for vaccination against heterosubtypic influenza viruses using an adenoviral vector leading to enhanced t cell response through autophagy
Pushko et al. 25 Traditional and Novel Trends
Patel Comparative evaluation of human and porcine adenovirus vectors for vaccine application agianst avian influenza (H5N1)

Legal Events

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

Ref document number: 09820933

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009820933

Country of ref document: EP