US20060024670A1 - Influenza virus vaccine composition and methods of use - Google Patents

Influenza virus vaccine composition and methods of use Download PDF

Info

Publication number
US20060024670A1
US20060024670A1 US11/131,479 US13147905A US2006024670A1 US 20060024670 A1 US20060024670 A1 US 20060024670A1 US 13147905 A US13147905 A US 13147905A US 2006024670 A1 US2006024670 A1 US 2006024670A1
Authority
US
United States
Prior art keywords
codons
composition
seq
nucleic acid
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/131,479
Inventor
Catherine Luke
Adrian Vilalta
Mary Wloch
Thomas Evans
Andrew Geall
Gretchen Jimenez
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fresh Tracks Therapeutics Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US11/131,479 priority Critical patent/US20060024670A1/en
Publication of US20060024670A1 publication Critical patent/US20060024670A1/en
Assigned to VICAL INCORPORATED reassignment VICAL INCORPORATED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WLOCH, MARY K., GEALL, ANDREW J., JIMENEZ, GRETCHEN S., VILALTA, ADRIAN, EVANS, THOMAS G., LUKE, CATHERINE J.
Priority to US11/704,251 priority patent/US7537768B2/en
Priority to US11/715,973 priority patent/US7785603B2/en
Priority to US11/892,016 priority patent/US8128938B1/en
Priority to US12/688,614 priority patent/US20100197771A1/en
Priority to US13/250,508 priority patent/US8821890B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1018Orthomyxoviridae, 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
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • 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
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16151Methods of production or purification of viral material

Definitions

  • This application includes a “Sequence Listing,” which is provided as an electronic document on a compact disk (CD-R).
  • This compact disk contains the file “Sequence Listing.txt” (340,000 bytes, created on May 18, 2005), which is hereby incorporated by reference in its entirety.
  • influenza virus vaccine compositions and methods of treating or preventing influenza infection and disease in mammals.
  • Influenza is an acute febrile illness caused by infection of the respiratory tract.
  • influenza viruses There are three types of influenza viruses: A, B, and C “IAV,” “IBV” or “IAC,” respectively, or generally “IV”.
  • Type A which includes several subtypes, causes widespread epidemics and global pandemics such as those that occurred in 1918, 1957 and 1968.
  • Type B causes regional epidemics.
  • Type C causes sporadic cases and minor, local outbreaks.
  • These virus types are distinguished in part on the basis of differences in two structural proteins, the nucleoprotein, found in the center of the virus, and the matrix protein, which forms the viral shell.
  • the disease can cause significant systemic symptoms, severe illness requiring hospitalization (such as viral pneumonia), and complications such as secondary bacterial pneumonia. More than 20 million people died during the pandemic flu season of 1918/1919, the largest pandemic of the 20 th century. Recent epidemics in the United States are believed to have resulted in greater than 10,000 (up to 40,000) excess deaths per year and 5,000-10,000 deaths per year in non-epidemic years.
  • Vaccination is especially recommended for people in high-risk groups, such as residents of nursing or residential homes, as well as for diabetes, chronic renal failure, or chronic respiratory conditions.
  • the influenza vaccines currently in use are designated whole virus (WV) vaccine or subvirion (SV) (also called “split” or “purified surface antigen”).
  • WV vaccine contains intact, inactivated virus
  • SV vaccine contains purified virus disrupted with detergents that solubilize the lipid-containing viral envelope, followed by chemical inactivation of residual virus.
  • Attenuated viral vaccines against influenza are also in development.
  • a discussion of methods of preparing conventional vaccine may be found in Wright, P. F. & Webster, R. G., FIELDS VIROLOGY, 4d Ed. (Knipe, D. M. et al. Ed.), 1464-65 (2001), for example.
  • An IV is roughly spherical, but it can also be elongated or irregularly shaped.
  • eight segments of single-stranded RNA contain the genetic instructions for making the virus.
  • the most striking feature of the virus is a layer of spikes projecting outward over its surface.
  • spikes There are two different types of spikes: one is composed of the molecule hemagglutinin (HA), the other of neuraminidase (NA).
  • HA hemagglutinin
  • NA neuraminidase
  • the HA molecule allows the virus to “stick” to a cell, initiating infection.
  • the NA molecule allows newly formed viruses to exit their host cell without sticking to the cell surface or to each other.
  • the viral capsid is comprised of viral ribonucleic acid and several so called “internal” proteins (polymerases (PB1, PB2, and PA, matrix protein (M1) and nucleoprotein (NP)). Because antibodies against HA and NA have traditionally proved the most effective in fighting infection, much research has focused on the structure, function, and genetic variation of those molecules. researchers are also interested in a two non-structural proteins M2 and NS1; both molecules play important roles in viral infection.
  • PB1, PB2, and PA matrix protein
  • NP nucleoprotein
  • Type A subtypes are described by a nomenclature system that includes the geographic site of discovery, a lab identification number, the year of discovery, and in parentheses the type of HA and NA it possesses, for example, A/Hong Kong/156/97 (H5N1). If the virus infects non-humans, the host species is included before the geographical site, as in A/Chicken/Hong Kong/G9/97 (H9N2).
  • Virions contain 7 segments (influenza C virus) to 8 segments (influenza A and B virus) of linear negative-sense single stranded RNA. Most of the segments of the virus genome code for a single protein. For many influenza viruses, the whole genome is now known. Genetic reassortment of the virus results from intermixing of the parental gene segments in the progeny of the viruses when a cell is co-infected by two different viruses of a given type. This phenomenon is facilitated by the segmental nature of the genome of influenza virus. Genetic reassortment is manifested as sudden changes in the viral surface antigens.
  • Antigenic changes in HA and NA allow the influenza virus to have tremendous variability. Antigenic drift is the term used to indicate minor antigenic variations in HA and NA of the influenza virus from the original parent virus, while major changes in HA and NA which make the new virions significantly different, are called Antigenic shift. The difference between the two phenomena is a matter of degree.
  • Antigenic drift occurs due to accumulation of point mutations in the gene which results in changes in the amino acids in the proteins. Changes which are extreme, and drastic (too drastic to be explained by mutation alone) result in antigenic shift of the virus.
  • the segmented genomes of the influenza viruses reassort readily in double infected cells. Genetic reassortment between human and non-human influenza virus has been suggested as a mechanism for antigenic shift.
  • Influenza is a zoonotic disease, and an important pathogen in a number of animal species, including swine, horses, and birds, both wild and domestic. Influenza viruses are transferred to humans from other species.
  • DNA-based vaccines for a number of infectious diseases where the need for a vaccine, or an improved vaccine, exists.
  • Several well-recognized advantages of DNA-based vaccines include the speed, ease and cost of manufacture, the versatility of developing and testing multivalent vaccines, the finding that DNA vaccines can produce a robust cellular response in a wide variety of animal models as well as in humans, and the proven safety of using plasmid DNA as a delivery vector (Donnelly, J. J., et al., Annu. Rev. Immunol. 15:617-648 (1997); Manickan, E., et al., Crit. Rev. Immunol. 17(2):139-154 (1997); U.S. Pat.
  • DNA vaccines represent the next generation in the development of vaccines (Nossal, G., Nat. Med. 4(5 Supple):475-476 (1998)) and numerous DNA vaccines are in clinical trials.
  • the above references are herein incorporated by reference in their entireties.
  • mice could be protected by an IV nucleoprotein DNA vaccine alone against severe disease and death resulting from either a homologous or a heterologous IV challenge. Further studies have substantiated this model, and comparative studies of live influenza vaccines versus DNA influenza vaccines show them to be relatively equivalent in immune induction and protection in the murine model.
  • WO 94/21797 discloses IV vaccine compositions comprising DNA constructs encoding NP, HA, M1, PB1 and NS1.
  • WO 94/21797 also discloses methods of protecting against IV infection comprising immunization with a prophylactically effective amount of these DNA vaccine compositions.
  • the IV nucleoprotein is relatively conserved (see Shu, L. L. et al., J. Virol. 67:2723-9 (1993)), but just as conserved are the M1 matrix protein (which is a major T-cell target), and the M2 protein, which are encoded by separate reading frames of RNA segment 7.
  • M1 matrix protein which is a major T-cell target
  • M2 protein which are encoded by separate reading frames of RNA segment 7.
  • Neirynck S. et al., Nat. Med. 5:1157-63 (1999); Lamb, R. A. & Lai, C. J., Virology 112:746-51 (1981); Ito, T. et al., J. Virol. 65:5491-8 (1991).
  • Animal DNA vaccine trials have been performed with DNA constructs encoding these genes alone or in combination, usually with success.
  • M2 protein is involved as part of an ion channel, is critical in resistance to the antiviral agents amantadine and rimantidine, and approximately 24 amino acids are extracellular (eM2).
  • eM2 extracellular, highly conserved protein
  • DNA vaccine priming followed by boosting with protein in adjuvant or by viral vector delivery of DNA encoding antigen appears to be the most effective way of improving antigen specific antibody and CD4+ T-cell responses or CD8+ T-cell responses respectively.
  • US 2002/0165172 A1 describes simultaneous administration of a vector construct encoding an immunogenic portion of an antigen and a protein comprising the immunogenic portion of an antigen such that an immune response is generated.
  • the document is limited to hepatitis B antigens and HIV antigens.
  • U.S. Pat. No. 6,500,432 is directed to methods of enhancing an immune response of nucleic acid vaccination by simultaneous administration of a polynucleotide and polypeptide of interest.
  • simultaneous administration means administration of the polynucleotide and the polypeptide during the same immune response, preferably within 0-10 or 3-7 days of each other.
  • the antigens contemplated by the patent include, among others, those of Hepatitis (all forms), HSV, HIV, CMV, EBV, RSV, VZV, HPV, polio, influenza, parasites (e.g., from the genus Plasmodium ), and pathogenic bacteria (including but not limited to M. tuberculosis, M. leprae, Chlamydia, Shigella, B. burgdorferi, enterotoxigenic E. coli, S. typhosa, H. pylori, V. cholerae, B. pertussis, etc.). All of the above references are herein incorporated by reference in their entireties.
  • the present invention is directed to enhancing the immune response of a vertebrate in need of protection against IV infection by administering in vivo, into a tissue of the vertebrate, at least one polynucleotide, wherein the polynucleotide comprises one or more nucleic acid fragments, where the one or more nucleic acid fragments are optionally fragments of codon-optimized coding regions operably encoding one or more IV polypeptides, or fragments, variants, or derivatives thereof.
  • the present invention is further directed to enhancing the immune response of a vertebrate in need of protection against IV infection by administering, in vivo, into a tissue of the vertebrate, a polynucleotide described above plus at least one isolated IV polypeptide or a fragment, a variant, or derivative thereof.
  • the isolated IV polypeptide can be, for example, a purified subunit, a recombinant protein, a viral vector expressing an isolated IV polypeptide, or can be an inactivated or attentuated IV, such as those present in conventional IV vaccines.
  • the polynucleotide is incorporated into the cells of the vertebrate in vivo, and an immunologically effective amount of an immunogenic epitope of the encoded IV polypeptide, or a fragment, variant, or derivative thereof, is produced in vivo.
  • an isolated IV polypeptide or a fragment, variant, or derivative thereof is also administered in an immunologically effective amount.
  • the polynucleotide can be administered either prior to, at the same time (simultaneously), or subsequent to the administration of the isolated IV polypeptide.
  • the IV polypeptide or fragment, variant, or derivative thereof encoded by the polynucleotide comprises at least one immunogenic epitope capable of eliciting an immune response to influenza virus in a vertebrate.
  • an isolated IV polypeptide or fragment, variant, or derivative thereof when used, comprises at least one immunogenic epitope capable of eliciting an immune response in a vertebrate.
  • the IV polypeptide or fragment, variant, or derivative thereof encoded by the polynucleotide can, but need not, be the same protein or fragment, variant, or derivative thereof as the isolated IV polypeptide which can be administered according to the method.
  • the polynucleotide of the invention can comprise a nucleic acid fragment, where the nucleic acid fragment is a fragment of a codon-optimized coding region operably encoding any IV polypeptide or fragment, variant, or derivative thereof, including, but not limited to, HA, NA, NP, M1 or M2 proteins or fragments (e.g., eM2), variants or derivatives thereof.
  • a polynucleotide of the invention can also encode a derivative fusion protein, wherein two or more nucleic acid fragments, at least one of which encodes an IV polypeptide or fragment, variant, or derivative thereof, are joined in frame to encode a single polypeptide, e.g., NP fused to eM2.
  • a polynucleotide of the invention can further comprise a heterologous nucleic acid or nucleic acid fragment.
  • Such heterologous nucleic acid or nucleic acid fragment may encode a heterologous polypeptide fused in frame with the polynucleotide encoding the IV polypeptide, e.g., a hepatitis B core protein or a secretory signal peptide.
  • the polynucleotide encodes an IV polypeptide or fragment, variant, or derivative thereof comprising at least one immunogenic epitope of IV, wherein the epitope elicits a B-cell (antibody) response, a T-cell (e.g., CTL) response, or both.
  • the isolated IV polypeptide or fragment, variant, or derivative thereof to be delivered can be any isolated IV polypeptide or fragment, variant, or derivative thereof, including but not limited to the HA, NA, NP, M1 or M2 proteins or fragments (e.g., eM2), variants or derivatives thereof.
  • a derivative protein can be a fusion protein, e.g., NP-eM2.
  • the isolated IV polypeptide or fragment, variant, or derivative thereof can be fused to a heterologous protein, e.g., a secretory signal peptide or the hepatitis B virus core protein.
  • a heterologous protein e.g., a secretory signal peptide or the hepatitis B virus core protein.
  • the isolated IV polypeptide or fragment, variant, or derivative thereof comprises at least one immunogenic epitope of IV, wherein the antigen elicits a B-cell antibody response, a T-cell antibody response, or both.
  • Nucleic acids and fragments thereof of the present invention can be altered from their native state in one or more of the following ways.
  • a nucleic acid or fragment thereof which encodes an IV polypeptide or fragment, variant, or derivative thereof can be part or all of a codon-optimized coding region, optimized according to codon usage in the animal in which the vaccine is to be delivered.
  • a nucleic acid or fragment thereof which encodes an IV polypeptide can be a fragment which encodes only a portion of a full-length polypeptide, and/or can be mutated so as to, for example, remove from the encoded polypeptide non-desired protein motifs present in the encoded polypeptide or virulence factors associated with the encoded polypeptide.
  • the nucleic acid sequence could be mutated so as not to encode a membrane anchoring region that would prevent release of the polypeptide from the cell as with, e.g., eM2.
  • the polynucleotide of the invention is incorporated into the cells of the vertebrate in vivo, and a prophylactically or therapeutically effective amount of an immunologic epitope of an IV is produced in vivo.
  • the proteins of the invention can be a fragment of a full-length IV polypeptide and/or can be altered so as to, for example, remove from the polypeptide non-desired protein motifs present in the polypeptide or virulence factors associated with the polypeptide.
  • the polypeptide could be altered so as not to encode a membrane anchoring region that would prevent release of the the polypeptide from the cell.
  • the invention further provides immunogenic compositions comprising at least one polynucleotide, wherein the polynucleotide comprises one or more nucleic acid fragments, where each nucleic acid fragment is a fragment of a codon-optimized coding region encoding an IV polypeptide or a fragment, a variant, or a derivative thereof; and immunogenic compositions comprising a polynucleotide as described above and at least one isolated IV polypeptide or a fragment, a variant, or derivative thereof.
  • Such compositions can further comprise, for example, carriers, excipients, transfection facilitating agents, and/or adjuvants as described herein.
  • immunogenic compositions comprising a polynucleotide and an isolated IV polypeptide or fragment, variant, or derivative thereof as described above can be provided so that the polynucleotide and protein formulation are administered separately, for example, when the polynucleotide portion of the composition is administered prior (or subsequent) to the isolated IV polypeptide portion of the composition.
  • immunogenic compositions comprising the polynucleotide and the isolated IV polypeptide or fragment, variant, or derivative thereof can be provided as a single formulation, comprising both the polynucleotide and the protein, for example, when the polynucleotide and the protein are administered simultaneously.
  • the polynucleotide portion of the composition and the isolated IV polypeptide portion of the composition can be provided simultaneously, but in separate formulations.
  • compositions comprising at least one polynucleotide comprising one or more nucleic acid fragments, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide or fragment, variant, or derivative thereof together with and one or more isolated IV polypeptides or fragments, variants or derivatives thereof (as either a recombinant protein, a purified subunit, a viral vector expressing the protein, or in the form of an inactivated or attenuated IV vaccine) will be referred to herein as “combinatorial polynucleotide (e.g., DNA) vaccine compositions” or “single formulation heterologous prime-boost vaccine compositions.”
  • compositions of the invention can be univalent, bivalent, trivalent or mulitvalent.
  • a univalent composition will comprise only one polynucleotide comprising a nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide or a fragment, variant, or derivative thereof, and optionally the same IV polypeptide or a fragment, variant, or derivative thereof in isolated form.
  • a univalent composition can include a polynucleotide comprising a nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide or a fragment, variant, or derivative thereof and an isolated polypeptide having the same antigenic region as the polynucleotide.
  • a bivalent composition will comprise, either in polynucleotide or protein form, two different IV polypeptides or fragments, variants, or derivatives thereof, each capable of eliciting an immune response.
  • the polynucleotide(s) of the composition can encode two IV polypeptides or alternatively, the polynucleotide can encode only one IV polypeptide and the second IV polypeptide would be provided by an isolated IV polypeptide of the invention as in, for example, a single formulation heterologous prime-boost vaccine composition.
  • the nucleic acid fragments operably encoding those IV polypeptides need not be on the same polynucleotide, but can be on two different polynucleotides.
  • a trivalent or further multivalent composition will comprise three IV polypeptides or fragments, variants or derivatives thereof, either in isolated form or encoded by one or more polynucleotides of the invention.
  • the present invention further provides plasmids and other polynucleotide constructs for delivery of nucleic acid fragments of the invention to a vertebrate, e.g., a human, which provide expression of IV polypeptides, or fragments, variants, or derivatives thereof.
  • the present invention further provides carriers, excipients, transfection-facilitating agents, immunogenicity-enhancing agents, e.g., adjuvants, or other agent or agents to enhance the transfection, expression or efficacy of the administered gene and its gene product.
  • a mulitvalent composition comprises a single polynucleotide, e.g., plasmid, comprising one or more nucleic acid regions operably encoding IV polypeptides or fragments, variants, or derivatives thereof. Reducing the number of polynucleotides, e.g., plasmids in the compositions of the invention can have significant impacts on the manufacture and release of product, thereby reducing the costs associated with manufacturing the compositions. There are a number of approaches to include more than one expressed antigen coding sequence on a single plasmid. These include, for example, the use of Internal Ribosome Entry Site (IRES) sequences, dual promoters/expression cassettes, and fusion proteins.
  • IRS Internal Ribosome Entry Site
  • the invention also provides methods for enhancing the immune response of a vertebrate to IV infection by administering to the tissues of a vertebrate one or more polynucleotides each comprising one or more nucleic acid fragments, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide or fragment, variant, or derivative thereof; and optionally administering to the tissues of the vertebrate one or more isolated IV polypeptides, or fragments, variants, or derivatives thereof.
  • the isolated IV polypeptide can be administered prior to, at the same time (simultaneously), or subsequent to administration of the polynucleotides encoding IV polypeptides.
  • the invention provides consensus amino acid sequences for IV polypeptides, or fragments, variants or derivatives thereof, including, but not limited to the HA, NA, NP, M1 or M2 proteins or fragments (e.g. eM2), variants or derivatives thereof.
  • Polynucleotides which encode the consensus polypeptides or fragments, variants or derivatives thereof, are also embodied in this invention. Such polynucleotides can be obtained by known methods, for example by backtranslation of the amino acid sequence and PCR synthesis of the corresponding polynucleotide as described below.
  • FIG. 1 shows an alignment of nucleotides 46-1542 of SEQ ID NO:1 (native NP coding region) with a coding region fully codon-optimized for human usage (SEQ ID NO:23).
  • FIG. 2 shows the protocol for the preparation of a formulation comprising 0.3 mM BAK, 7.5 mg/ml CRL 1005 and 5 mg/ml of DNA in a final volume of 3.6 ml, through the use of thermal cycling.
  • FIG. 3 shows the protocol for the preparation of a formulation comprising 0.3 mM BAK, 34 mg/ml or 50 mg/ml CRL 1005 and 2.5 mg/ml DNA in a final volumne of 4.0 ml, through the use of thermal cycling.
  • FIG. 4 shows the protocol for the simplified preparation (without thermal cycling) of a formulation comprising 0.3 mM BAK, 7.5 mg/ml CRL 1005 and 5 mg/ml DNA.
  • FIG. 5 shows the anti-NP antibody response three weeks after a single administration of a combinatorial prime-boost vaccine formulation against the influenza virus NP protein.
  • FIG. 6 shows the anti-NP antibody response twelve days after a second administration of a combinatorial prime-boost vaccine formulation against the influenza virus NP protein.
  • FIG. 7 shows the CD8+ T Cell response to a combinatorial prime-boost vaccine formulation against the influenza virus NP protein.
  • FIG. 8 shows the CD4+ T Cell response to a combinatorial prime-boost vaccine formulation against the influenza virus NP protein.
  • FIGS. 9A and 9B show the results of a two dose mouse immunization regimen study with plasmid DNA encoding IAV HA (H3).
  • FIGS. 10A and 10B show the in vitro expression of M1 and M2 from segment 7 and an M1M2 fusion.
  • FIGS. 11A and 11B show the in vitro expression of eM2-NP and codon-optimized influenza virus NP protein.
  • FIG. 12 shows the influenza A NP protein consensus amino acid sequence aligned with 22 full length NP sequences.
  • FIG. 13 is a schematic diagram of various vectors encoding influenza proteins described herein.
  • FIG. 14 are the results of western blot experiments as described in Example 13, Experiment 3.
  • the blots show lysates of VM92 cells transfected with plasmids which express M2 or NP to compare expression of the influenza protein from different expression vectors.
  • FIG. 15 are the results of western blot experiments as described in Example 13, Experiment 3.
  • the blots show lysates of VM92 cells transfected with plasmids which express M1, M2 or NP to compare expression of the influenza protein from expression vectors.
  • the present invention is directed to compositions and methods for enhancing the immune response of a vertebrate in need of protection against IV infection by administering in vivo, into a tissue of a vertebrate, at least one polynucleotide comprising one or more nucleic acid fragments, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide, or a fragment, variant, or derivative thereof in cells of the vertebrate in need of protection.
  • the present invention is also directed to administering in vivo, into a tissue of the vertebrate the above described polynucleotide and at least one isolated IV polypeptide, or a fragment, variant, or derivative thereof.
  • the isolated IV polypeptide or fragment, variant, or derivative thereof can be, for example, a recombinant protein, a purified subunit protein, a protein expressed and carried by a heterologous live or inactivated or attentuated viral vector expressing the protein, or can be an inactivated IV, such as those present in conventional, commercially available, inactivated IV vaccines.
  • the polynucleotide is incorporated into the cells of the vertebrate in vivo, and an immunologically effective amount of the influenza protein, or fragment or variant encoded by the polynucleotide is produced in vivo.
  • the isolated protein or fragment, variant, or derivative thereof is also administered in an immunologically effective amount.
  • the polynucleotide can be administered to the vertebrate in need thereof either prior to, at the same time (simultaneously), or subsequent to the administration of the isolated IV polypeptide or fragment, variant, or derivative thereof.
  • IV polypeptides within the scope of the invention include, but are not limited to, NP, HA, NA, M1 and M2 polypeptides, and fragments, e.g., eM2, derivatives, e.g., an NP-eM2 fusion, and variants thereof.
  • Nucleotide and amino acid sequences of IV polypeptides from a wide variety of IV types and subtypes are known in the art. The nucleotide sequences set out below are the wild-type sequences.
  • nucleotide sequence of the NP protein of Influenza A/PR/8/34 is available as GenBank Accession Number M38279.1, and has the following sequence, referred to herein as SEQ ID NO:1: AGCAAAAGCAGGGTAGATAATCACTCACTGAGTGACATCAAAATCATGGC GTCTCAAGGCACCAAACGATCTTACGAACAGATGGAGACTGATGGAGAAC GCCAGAATGCCACTGAAATCAGAGCATCCGTCGGAAAAATGATTGGTGGA ATTGGACGATTCTACATCCAAATGTGCACCGAACTCAAACTCAGTGATTA TGAGGGACGGTTGATCCAAAACAGCTTAACAATAGAGAGAATGGTGCTCT CTGCTTTTGACGAAAGGAGAAATAAATACCTTGAAGAACATCCCAGTGCG GGGAAAGATCCTAAGAAAACTGGAGGACCTATATACAGGAGAGTAAACGG AAAGTGGATGAGAGAACTCATCCTTTATGACAAAAAATAAAATAAGAAATAAGCCAGTGCG GG
  • SEQ ID NO:2 The amino acid sequence of the NP protein of Influenza A/PR/8/34 (H1N1), encoded by nucleotides 46-1494 of SEQ ID NO:1 is as follows, referred to herein as SEQ ID NO:2: MASQGTKRSYEQMETDGERQNATEIRASVGKMIGGIGRFYIQMCTELKLS DYEGRLIQNSLTIERMVLSAFDERRNKYLEEHPSAGKDPKKTGGPIYRRV NGKWMRELILYDKEEIRRIWRQANNGDDATAGLTHMMIWHSNLNDATYQR TRALVRTGMDPRMCSLMQGSTLPRRSGAAGAAVKGVGTMVMELVRMIKRG INDRNFWRGENGRKTRIAYERMCNILKGKFQTAAQKAMMDQVRESRNPGN AEFEDLTFLARSALILRGSVAHKSCLPACVYGPAVASGYDFEREGYSLVG IDPFRLLQNSQVYSLIRPNENPAHKSQLVWMACHSAAF
  • Segment 7 of the IAV genome encodes both M1 and M2.
  • Seqment 7 of Influenza A virus (A/Puerto Rico/8/34/Mount Sinai (H1N1)), is available as GenBank Accession No. AF389121.1, and has the following sequence, referred to herein as SEQ ID NO:3: AGCGAAAGCAGGTAGATATTGAAAGATGAGTCTTCTAACCGAGGTCGAAA CGTACGTACTCTCTATCATCCCGTCAGGCCCCCTCAAAGCCGAGATCGCA CAGAGACTTGAAGATGTCTTTGCAGGGAAGAACACTGATCTTGAGGTTCT CATGGAATGGCTAAAGACAAGACCAATCCTGTCACCTCTGACTAAGGGGA TTTTAGGATTTGTGTTCACGCTCACCGTGCCCAGTGAGCGAGGACTGCAG CGTAGACGCTTTGTCCAAAATGCCCTTAATGGGAACGGATCCAAATAA CATGGACAAAGCAGTTAAACTGTATAGGAAGCTCAAGGGAGATT
  • SEQ ID NO:4 The amino acid sequence of the M1 protein of Influenza A/Puerto Rico/8/34/Mount Sinai(H1N1), encoded by nucleotides 26 to 784 of SEQ ID NO:3 is as follows, referred to herein as SEQ ID NO:4: MSLLTEVETYVLSIIPSGPLKAEIAQRLEDVFAGKNTDLEVLMEWLKTRP ILSPLTKGILGFVFTLTVPSERGLQRRRFVQNALNGNGDPNNMDKAVKLY RKLKREITFHGAKEISLSYSAGALASGMGLIYNRMGAVTTEVAFGLVCAT CEQIADSQHRSHRQMVTTNPLIRHENRMVLASTTAKAMEQMAGSSEQAAE AMEVASQARQMVQAMRTIGTHPSSSAGLKNDLLENLQAYQKRMGVQMQ RFK
  • SEQ ID NO:5 The amino acid sequence of the M2 protein of Influenza A/Puerto Rico/8/34/Mount Sinai (H1N1), encoded (in spliced form) by nucleotides 26 to 51 and 740 to 1007 of SEQ ID NO:3 is as follows, referred to herein as SEQ ID NO:5: MSLLTEVETPIRNEWGCRCNGSSD PLAIAANIIGILHLTLWILDRLFFKC IYRRFKYGLKGGPSTEGVPKSMREEYRKEQQSAVDADDGHFVSIELE
  • Extracellular region of the M2 protein corresponds to the first 24 amino acids of the N-terminal end of the protein, and is underlined above. See Fischer, W. B. et al., Biochim. Biophys. Acta. 1561:27-45 (2002); Zhong, Q. et al., FEBS Lett. 434:265-71 (1998).
  • a derivative of NP and eM2 described herein is encoded by a construct which encodes the first 24 amino acids of M2 and all or a portion of NP.
  • the fusion constructs may be constructed with the eM2 sequences followed by the NP sequences, or with the NP sequences followed by the eM2 sequences.
  • Exemplary fusion constructs using the NP and M2 sequences from Influenza A/PR/8/34 (H1N1) are set out below.
  • a sequence, using the original influenza virus nucleotide sequences, which encodes the first 24 amino acids of M2 fused at its 3′ end to a sequence which encodes NP in its entirety eM2-NP is referred to herein as SEQ ID NO:6: 1 ATGAGTCTTC TAACCGAGGT CGAAACGCCT ATCAGAAACG AATGGGGGTG CAGATGCAAC 61 GGTTCAAGTG ATATGGCGTC TCAAGGCACC AAACGATCTT ACGAACAGAT GGAGACTGAT 121 GGAGAACGCC AGAATGCCAC TGAAATCAGA GCATCCGTCG GAAAAATGAT TGGTGGAATT 181 GGACGATTCT ACATCCAAAT GTGCACCGAA CTCAAACTCA GTGATTATGA GGGACGGTTG 241 ATCCAAAACA GCTTAACAAT AGAGAGAATG GTGCTCTCTG CTTTTGACGA AAGGAGAAAT 301 AAATACCTTG AAGAACATCC CAGTGCGGGG AAAGATCC
  • SEQ ID NO:7 The amino acid sequence of the eM2-NP fusion protein of Influenza A/PR/8/341 (H1N1), encoded by nucleotides 1 to 1566 SEQ ID NO:6 is as follows, referred to herein as SEQ ID NO:7 (eM2 amino acid sequence underlined): MSLLTEVETPIRNEWGCRCNGSSDMASQGTKRSYEQMETDGERQNATEIR ASVGKMIGGIGRFYIQMCTELKLSDYEGRLIQNSLTIERMVLSAFDERRN KYLEEHPSAGKDPKKTGGPIYRRVNGKWMRELILYDKEEIRRIWRQANNG DDATAGLTHMMIWHSNLNDATYQRTRALVRTGMDPRMCSLMQGSTLPRRS GAAGAAVKGVGTMVMELVRMIKRGINDRNFWRGENGRKTRIAYERMCNIL KGKFQTAAQKAMMDQVRESRNPGNAEFEDLTFLARSALILRGSVAHKSCL PACVYGPAVASGYDFEREG
  • SEQ ID NO:8 ATGGCGTCTCAAGGCACCAAACGATCTTACGAACAGATGGAGACTGATGG AGAACGCCAGAATGCCACTGAAATCAGAGCATCCGTCGGAAAAATGATTG GTGGAATTGGACGATTCTACATCCAAATGTGCACCGAACTCAAACTCAGT GATTATGAGGGACGGTTGATCCAAAACAGCTTAACAATAGAGAGAATGGT GCTCTCTGCTTTTGACGAAAGGAGAAATAAATACCTTGAAGAACATCCCA GTGCGGGGAAAGATCCTAAGAAAACTGGAGGACCTATATACAGGAGAGTA AACGGAAAGTGGATGAGAACTCATCCTTTATGACAGGAGTA AACGGAAAGTGGATGAGAACTCATCCTTTATGACAAAAAATAAGAAATAAG GCGAATCTGGCGCCAAGCTAATAATGGTG
  • SEQ ID NO:9 amino acid sequence underlined: MASQGTKRSYEQMETDGERQNATEIRASVGKMIGGIGRFYIQMCTELKLS DYEGRLIQNSLTIERMVLSAFDERRNKYLEEHPSAGKDPKKTGGPIYRRV NGKWMRELILYDKEEIRRIWRQANNGDDATAGLTHMMIWHSNLNDATYQR TRALVRTGMDPRMCSLMQGSTLPRRSGAAGAAVKGVGTMVMELVRMIKRG INDRNFWRGENGRKTRIAYERMCNILKGKFQTAAQKAMMDQVRESRNPGN AEFEDLTFLARSALILRGSVAHKSCLPACVYGPAVASGYDFEREGYSLVG IDPFRLLQNSQVYSL
  • the nucleotide sequence of the NP protein of Influenza B/LEE/40 is available as GenBank Accession Number K01395, and has the following sequence, referred to herein as SEQ ID NO:15: 1 ATGTCCAACA TGGATATTGA CAGTATAAAT ACCGGAACAA TCGATAAAAC ACCAGAAGAA 61 CTGACTCCCG GAACCAGTGG GGCAACCAGA CCAATCATCA AGCCAGCAAC CCTTGCTCCG 121 CCAAGCAACA AACGAACCCG AAATCCATCT CCAGAAAGGA CAACCACAAG CAGTGAAACC 181 GATATCGGAA GGAAAATCCA AAAGAAACAA ACCCCAACAG AGATAAAGAA GAGCGTCTAC 241 AAAATGGTGG TAAAACTGGG TGAATTCTAC AACCAGATGA TGGTCAAAGC TGGACTTAAT 301 GATGACATGG AAAGGAATCT AATTCAAAAT GCACAAGCTG TGGAGAAT CCTATTGGCT 361 G
  • SEQ ID NO:16 MSNMDIDSINTGTIDKTPEELTPGTSGATRPIIKPATLAPPSNKRTRNPS PERTTTSSETDIGRKIQKKQTPTEIKKSVYKMVVKLGEFYNQMMVKAGLN DDMERNLIQNAQAVERILLAATDDKKTEYQKKRNARDVKEGKEEIDHNKT GGTFYKMVRDDKTIYFSPIKITFLKEEVKTMYKTTMGSDGFSGLNHIMIG HSQMNDVCFQRSKGLKRVGLDPSLISTFAGSTLPRRSGTTGVAIKGGGTL VDEAIRFIGRAMADRGLLRDIKAKTAYEKILLNLKNKCSAPQQKALVDQV IGSRNPGIADIEDLTLLARSMVVVRPSVASKVVLPISIYAKIPQLGFNTE
  • Non limiting examples of nucleotide sequences encoding the IAV hemagglutinin (HA) are as follows. It should be noted that HA sequences vary significantly between IV subtypes. Virtually any nucleotide sequence encoding an IV HA is suitable for the present invention. In fact, HA sequences included in vaccines and therapeutic formulations of the present invention (discussed in more detail below) might change from year to year depending on the prevalent strain or strains of IV.
  • the partial nucleotide sequence of the HA protein of IAV A/New_York/1/18(H1N1) is available as GenBank Accession Number AF116576, and has the following sequence, referred to herein as SEQ ID NO: 17: 1 atggaggcaa gactactggt cttgttatgt gcatttgcag ctacaaatgc agacacaata 61 tgtataggct accatgcgaa taactcaacc gacactgttg acacagtact cgaaaagaat 121 gtgaccgtga cacactctgt taacctgctc gaagacagcc acaacggaaact actatgtaaa 181 ttaaaggaa tagccccatt acaattgggg aaatgtaata tcgcggatg gctcggagga
  • amino acid sequence of the partial HA protein of IAV A/New_York/1/18(H1N1), encoded by nucleotides 1 to 1218 of SEQ ID NO:17 is as follows, referred to herein as SEQ ID NO:18: MEARLLVLLCAFAATNADTICIGYHANNSTDTVDTVLEKNVTVTHSVNLL EDSHNGKLCKLKGIAPLQLGKCNIAGWLLGNPECDLLLTASSWSYIVETS NSENGTGYPGDFLDYEELREQLSSVSSFEKFEIFPKTSSWPNHETTKGVT AACSYAGASSFYRNLLWLTKGSSYPKLSKSYVNNKGKEVLVLWGVHHPPT GTDQQSLYQNADAYVSVGSSKYNRRFTPEIAARPKVRGQAGRMNYYWTLL EPGDTITFEATGNLIAPWYAFALNRGSGSGIITSDAPVHDCNTKCQTPHG AINSSLPFQNIHPVTIGECPKYVRSTKLRMATGLRNIPS
  • the nucleotide sequence of the IAV A/Hong Kong/482/97 hemagglutinin (H5) is available as GenBank Accession Number AF046098, and has the following sequence, referred to herein as SEQ ID NO:19: 1 ctgtcaaaat qgagaaaata gtgcttcttc ttgcaacagt cagtcttgtt aaaagtgatc 61 agatttgcat tggttaccat gcaaacaact cgacagagca ggttgacaca ataatggaaa 121 agaatgttac tgttacacat gcccaagaca tactggaaag gacacacaac gggaagctct 181 gcgatctaaa tggagtgaaa cctctcattttgagggattg tagtgtagc
  • SEQ ID NO:20 The amino acid sequence of the HA protein of IAV A/Hong Kong/482/97 (H5), encoded by nucleotides 9 to 1715 of SEQ ID NO:19 is as follows, referred to herein as SEQ ID NO:20: MEKIVLLLATVSLVKSDQICIGYHANNSTEQVDTIMEKNVTVTHAQDILE RTHNGKLGDLNGVKPLILRDCSVAGWLLGNPMCDEFINVPEWSYIVEKAS PANDLCYPGNFNDYEELKHLLSRINHFEKIQIIPKSSWSNHDASSGVSSA GPYLGRSSFFRNVVWLIKKNSAYPTIKRSYNNTNQEDLLVLWGIHHPNDA AEQTKLYQNPTTYISVGTSTLNQRLVPEIATRPKVNGQSGRMEFFWTILK PNDAINFESNGNFIAPEYAYKIVKKGDSTIMKSELEYGNCNTKCQTPMGA INSSMPFHNIHPLTIGECPKYVKSNR
  • the nucleotide sequence of the IAV A/Hong Kong/1073/99(H9N2) is available as GenBank Accession Number INA404626, and has the following sequence, referred to herein as SEQ ID NO:21: 1 gcaaaagcag gggaattact taactagcaa aatggaaaca atatcactaa taactatact 61 actagtagta acagcaagca atgcagataa aatctgcatc ggccaccagt caacaaactc 121 cacagaaact gtgqacacgc taacagaaac caatgttcct gtgacacatg ccaaagaatt 181 gctccacaca gagcataatg gaatgctgtg tgcaacaagc ctgggacatc cctcattct 241 agacacatgc actattgaag
  • SEQ ID NO:22 The amino acid sequence of the HA protein of IAV A/Hong Kong/1073/99 (H9N2), encoded by nucleotides 32 to 1711 of SEQ ID NO:21 is as follows, referred to herein as SEQ ID NO:22: METISLITILLVVTASNADKICIGHQSTNSTETVDTLTETNVPVTHAKEL LHTEHNGMLCATSLGHPLILDTCTIEGLVYGNPSGDLLLGGREWSYIVER SSAVNGTCYPGNVENLEELRTLFSSASSYQRIQIFPDTTWNVTYTGTSRA CSGSFYRSMRWLTQKSGFYPVQDAQYTNNRGKSILFVWGIHHPPTYTEQT NLYIRNDTTTSVTTEDLNRTFKPVIGPRPLVNGLQGRIDYYWSVLKPGQT LRVRSNGNLIAPWYGHVLSGGSHGRILKTDLKGGNCVVQCQTEKGGLNST LPFHNISKYAFGTCPKY
  • the present invention also provides vaccine compositions and methods for delivery of IV coding sequences to a vertebrate with optimal expression and safety conferred through codon optimization and/or other manipulations.
  • These vaccine compositions are prepared and administered in such a manner that the encoded gene products are optimally expressed in the vertebrate of interest.
  • these compositions and methods are useful in stimulating an immune response against IV infection.
  • expression systems, delivery systems, and codon-optimized IV coding regions are also included in the invention.
  • the invention provides combinatorial polynucleotide (e.g., DNA) vaccines which combine both a polynucleotide vaccine and polypeptide (e.g., either a recombinant protein, a purified subunit protein, a viral vector expressing an isolated IV polypeptide, or in the form of an inactivated or attenuated IV vaccine) vaccine in a single formulation.
  • the single formulation comprises an IV polypeptide-encoding polynucleotide vaccine as described herein, and optionally, an effective amount of a desired isolated IV polypeptide or fragment, variant, or derivative thereof.
  • the polypeptide may exist in any form, for example, a recombinant protein, a purified subunit protein, a viral vector expressing an isolated IV polypeptide, or in the form of an inactivated or attenuated IV vaccine.
  • the IV polypeptide or fragment, variant, or derivative thereof encoded by the polynucleotide vaccine may be identical to the isolated IV polypeptide or fragment, variant, or derivative thereof.
  • the IV polypeptide or fragment, variant, or derivative thereof encoded by the polynucleotide may be different from the isolated IV polypeptide or fragment, variant, or derivative thereof.
  • a or “an” entity refers to one or more of that entity; for example, “a polynucleotide,” is understood to represent one or more polynucleotides.
  • the terms “a” (or “an”), “one or more,” and “at least one” can be used interchangeably herein.
  • polynucleotide is intended to encompass a singular nucleic acid or nucleic acid fragment as well as plural nucleic acids or nucleic acid fragments, and refers to an isolated molecule or construct, e.g., a virus genome (e.g., a non-infectious viral genome), messenger RNA (mRNA), plasmid DNA (pDNA), or derivatives of pDNA (e.g., minicircles as described in (Darquet, A-M et al., Gene Therapy 4:1341-1349 (1997)) comprising a polynucleotide.
  • a polynucleotide may comprise a conventional phosphodiester bond or a non-conventional bond (e.g., an amide bond, such as found in peptide nucleic acids (PNA)).
  • nucleic acid or “nucleic acid fragment” refer to any one or more nucleic acid segments, e.g., DNA or RNA fragments, present in a polynucleotide or construct.
  • a nucleic acid or fragment thereof may be provided in linear (e.g., mRNA) or circular (e.g., plasmid) form as well as double-stranded or single-stranded forms.
  • isolated nucleic acid or polynucleotide is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment. For example, a recombinant polynucleotide contained in a vector is considered isolated for the purposes of the present invention.
  • an isolated polynucleotide examples include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution.
  • Isolated RNA molecules include in vivo or in vitro RNA transcripts of the polynucleotides of the present invention.
  • Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically.
  • a “coding region” is a portion of nucleic acid which consists of codons translated into amino acids. Although a “stop codon” (TAG, TGA, or TAA) is not translated into an amino acid, it may be considered to be part of a coding region, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, and the like, are not part of a coding region.
  • Two or more nucleic acids or nucleic acid fragments of the present invention can be present in a single polynucleotide construct, e.g., on a single plasmid, or in separate polynucleotide constructs, e.g., on separate (different) plasmids.
  • any nucleic acid or nucleic acid fragment may encode a single IV polypeptide or fragment, derivative, or variant thereof, e.g., or may encode more than one polypeptide, e.g., a nucleic acid may encode two or more polypeptides.
  • a nucleic acid may include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator, or may encode heterologous coding regions fused to the IV coding region, e.g., specialized elements or motifs, such as a secretory signal peptide or a heterologous functional domain.
  • fragment when referring to IV polypeptides of the present invention include any polypeptides which retain at least some of the immunogenicity or antigenicity of the corresponding native polypeptide. Fragments of IV polypeptides of the present invention include proteolytic fragments, deletion fragments and in particular, fragments of IV polypeptides which exhibit increased secretion from the cell or higher immunogenicity or reduced pathogenicity when delivered to an animal. Polypeptide fragments further include any portion of the polypeptide which comprises an antigenic or immunogenic epitope of the native polypeptide, including linear as well as three-dimensional epitopes.
  • Variants of IV polypeptides of the present invention include fragments as described above, and also polypeptides with altered amino acid sequences due to amino acid substitutions, deletions, or insertions. Variants may occur naturally, such as an allelic variant.
  • allelic variant is intended alternate forms of a gene occupying a given locus on a chromosome or genome of an organism or virus. Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985), which is incorporated herein by reference. For example, as used herein, variations in a given gene product.
  • each such protein is a “variant,” in that native IV strains are distinguished by the type of NA and HA proteins encoded by the virus. However, within a single HA or NA variant type, further naturally or non-naturally occurring variations such as amino acid deletions, insertions or substitutions may occur. Non-naturally occurring variants may be produced using art-known mutagenesis techniques. Variant polypeptides may comprise conservative or non-conservative amino acid substitutions, deletions or additions. Derivatives of IV polypeptides of the present invention, are polypeptides which have been altered so as to exhibit additional features not found on the native polypeptide. Examples include fusion proteins. An analog is another form of an IV polypeptide of the present invention. An example is a proprotein which can be activated by cleavage of the proprotein to produce an active mature polypeptide.
  • infectious polynucleotide or “infectious nucleic acid” are intended to encompass isolated viral polynucleotides and/or nucleic acids which are solely sufficient to mediate the synthesis of complete infectious virus particles upon uptake by permissive cells.
  • infectious nucleic acids do not require pre-synthesized copies of any of the polypeptides it encodes, e.g., viral replicases, in order to initiate its replication cycle in a permissive host cell.
  • non-infectious polynucleotide or “non-infectious nucleic acid” as defined herein are polynucleotides or nucleic acids which cannot, without additional added materials, e.g, polypeptides, mediate the synthesis of complete infectious virus particles upon uptake by permissive cells.
  • An infectious polynucleotide or nucleic acid is not made “non-infectious” simply because it is taken up by a non-permissive cell.
  • an infectious viral polynucleotide from a virus with limited host range is infectious if it is capable of mediating the synthesis of complete infectious virus particles when taken up by cells derived from a permissive host (i.e., a host permissive for the virus itself).
  • a permissive host i.e., a host permissive for the virus itself.
  • the fact that uptake by cells derived from a non-permissive host does not result in the synthesis of complete infectious virus particles does not make the nucleic acid “non-infectious.”
  • the term is not qualified by the nature of the host cell, the tissue type, or the species taking up the polynucleotide or nucleic acid fragment.
  • an isolated infectious polynucleotide or nucleic acid may produce fully-infectious virus particles in a host cell population which lacks receptors for the virus particles, i.e., is non-permissive for virus entry. Thus viruses produced will not infect surrounding cells. However, if the supernatant containing the virus particles is transferred to cells which are permissive for the virus, infection will take place.
  • replicating polynucleotide or “replicating nucleic acid” are meant to encompass those polynucleotides and/or nucleic acids which, upon being taken up by a permissive host cell, are capable of producing multiple, e.g., one or more copies of the same polynucleotide or nucleic acid.
  • Infectious polynucleotides and nucleic acids are a subset of replicating polynucleotides and nucleic acids; the terms are not synonymous.
  • a defective virus genome lacking the genes for virus coat proteins may replicate, e.g., produce multiple copies of itself, but is NOT infectious because it is incapable of mediating the synthesis of complete infectious virus particles unless the coat proteins, or another nucleic acid encoding the coat proteins, are exogenously provided.
  • the polynucleotide, nucleic acid, or nucleic acid fragment is DNA.
  • a polynucleotide comprising a nucleic acid which encodes a polypeptide normally also comprises a promoter and/or other transcription or translation control elments operably associated with the polypeptide-encoding nucleic acid fragment.
  • An operable association is when a nucleic acid fragment encoding a gene product, e.g., a polypeptide, is associated with one or more regulatory sequences in such a way as to place expression of the gene product under the influence or control of the regulatory sequence(s).
  • Two DNA fragments are “operably associated” if induction of promoter function results in the transcription of mRNA encoding the desired gene product and if the nature of the linkage between the two DNA fragments does not (1) result in the introduction of a frame-shift mutation, (2) interfere with the ability of the expression regulatory sequences to direct the expression of the gene product, or (3) interfere with the ability of the DNA template to be transcribed.
  • a promoter region would be operably associated with a nucleic acid fragment encoding a polypeptide if the promoter was capable of effecting transcription of that nucleic acid fragment.
  • the promoter may be a cell-specific promoter that directs substantial transcription of the DNA only in predetermined cells.
  • Other transcription control elements besides a promoter, for example enhancers, operators, repressors, and transcription termination signals, can be operably associated with the polynucleotide to direct cell-specific transcription. Suitable promoters and other transcription control regions are disclosed herein.
  • transcription control regions are known to those skilled in the art. These include, without limitation, transcription control regions which function in vertebrate cells, such as, but not limited to, promoter and enhancer segments from cytomegaloviruses (the immediate early promoter, in conjunction with intron-A), simian virus 40 (the early promoter), and retroviruses (such as Rous sarcoma virus).
  • Other transcription control regions include those derived from vertebrate genes such as actin, heat shock protein, bovine growth hormone and rabbit ⁇ -globin, as well as other sequences capable of controlling gene expression in eukaryotic cells. Additional suitable transcription control regions include tissue-specific promoters and enhancers as well as lymphokine-inducible promoters (e.g., promoters inducible by interferons or interleukins).
  • translation control elements include, but are not limited to ribosome binding sites, translation initiation and termination codons, elements from picornaviruses (particularly an internal ribosome entry site, or IRES, also referred to as a CITE sequence).
  • a DNA polynucleotide of the present invention may be a circular or linearized plasmid or vector, or other linear DNA which may also be non-infectious and nonintegrating (i.e., does not integrate into the genome of vertebrate cells).
  • a linearized plasmid is a plasmid that was previously circular but has been linearized, for example, by digestion with a restriction endonuclease. Linear DNA may be advantageous in certain situations as discussed, e.g., in Cherng, J. Y., et al., J. Control. Release 60:343-53 (1999), and Chen, Z. Y., et al. Mol. Ther. 3:403-10 (2001), both of which are incorporated herein by reference.
  • the terms plasmid and vector can be used interchangeably
  • DNA virus genomes may be used to administer DNA polynucleotides into vertebrate cells.
  • a DNA virus genome of the present invention is nonreplicative, noninfectious, and/or nonintegrating.
  • Suitable DNA virus genomes include without limitation, herpesvirus genomes, adenovirus genomes, adeno-associated virus genomes, and poxvirus genomes. References citing methods for the in vivo introduction of non-infectious virus genomes to vertebrate tissues are well known to those of ordinary skill in the art, and are cited supra.
  • a polynucleotide of the present invention is RNA, for example, in the form of messenger RNA (mRNA).
  • mRNA messenger RNA
  • Polynucleotides, nucleic acids, and nucleic acid fragments of the present invention may be associated with additional nucleic acids which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a nucleic acid fragment or polynucleotide of the present invention.
  • proteins secreted by mammalian cells have a signal peptide or secretory leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated.
  • polypeptides secreted by vertebrate cells generally have a signal peptide fused to the N-terminus of the polypeptide, which is cleaved from the complete or “full length” polypeptide to produce a secreted or “mature” form of the polypeptide.
  • the native leader sequence is used, or a functional derivative of that sequence that retains the ability to direct the secretion of the polypeptide that is operably associated with it.
  • a heterologous mammalian leader sequence, or a functional derivative thereof may be used.
  • the wild-type leader sequence may be substituted with the leader sequence of human tissue plasminogen activator (TPA) or mouse ⁇ -glucuronidase.
  • a polynucleotide construct for example, a plasmid, comprising a nucleic acid fragment, where the nucleic acid fragment is a fragment of a codon-optimized coding region operably encoding an IV-derived polypeptide, where the coding region is optimized for expression in vertebrate cells, of a desired vertebrate species, e.g., humans, to be delivered to a vertebrate to be treated or immunized.
  • Suitable IV polypeptides, or fragments, variants, or derivatives thereof may be derived from, but are not limited to, the IV HA, NA, NP, M1, or M2 proteins.
  • Additional IV-derived coding sequences may also be included on the plasmid, or on a separate plasmid, and expressed, either using native IV codons or codons optimized for expression in the vertebrate to be treated or immunized.
  • a plasmid encoding one or more optimized influenza sequences is delivered, in vivo to a tissue of the vertebrate to be treated or immunized, one or more of the encoded gene products will be expressed, i.e., transcribed and translated.
  • the level of expression of the gene product(s) will depend to a significant extent on the strength of the associated promoter and the presence and activation of an associated enhancer element, as well as the degree of optimization of the coding region.
  • plasmid refers to a construct made up of genetic material (i.e., nucleic acids). Typically a plasmid contains an origin of replication which is functional in bacterial host cells, e.g., Escherichia coli, and selectable markers for detecting bacterial host cells comprising the plasmid. Plasmids of the present invention may include genetic elements as described herein arranged such that an inserted coding sequence can be transcribed and translated in eukaryotic cells. Also, the plasmid may include a sequence from a viral nucleic acid.
  • a plasmid is a closed circular DNA molecule.
  • RNA product refers to the biological production of a product encoded by a coding sequence.
  • a DNA sequence including the coding sequence, is transcribed to form a messenger-RNA (mRNA).
  • mRNA messenger-RNA
  • the messenger-RNA is then translated to form a polypeptide product which has a relevant biological activity.
  • the process of expression may involve further processing steps to the RNA product of transcription, such as splicing to remove introns, and/or post-translational processing of a polypeptide product.
  • polypeptide is intended to encompass a singular “polypeptide” as well as plural “polypeptides,” and comprises any chain or chains of two or more amino acids.
  • terms including, but not limited to “peptide,” “dipeptide,” “tripeptide,” “protein,” “amino acid chain,” or any other term used to refer to a chain or chains of two or more amino acids are included in the definition of a “polypeptide,” and the term “polypeptide” can be used instead of, or interchangeably with any of these terms.
  • polypeptides which have undergone post-translational modifications, for example, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
  • polypeptides of the present invention are fragments, derivatives, analogs, or variants of the foregoing polypeptides, and any combination thereof.
  • Polypeptides, and fragments, derivatives, analogs, or variants thereof of the present invention can be antigenic and immunogenic polypeptides related to IV polypeptides, which are used to prevent or treat, i.e., cure, ameliorate, lessen the severity of, or prevent or reduce contagion of infectious disease caused by the IV.
  • an “antigenic polypeptide” or an “immunogenic polypeptide” is a polypeptide which, when introduced into a vertebrate, reacts with the vertebrate's immune system molecules, i.e., is antigenic, and/or induces an immune response in the vertebrate, i.e., is immunogenic. It is quite likely that an immunogenic polypeptide will also be antigenic, but an antigenic polypeptide, because of its size or conformation, may not necessarily be immunogenic. Examples of antigenic and immunogenic polypeptides of the present invention include, but are not limited to, e.g., HA or fragments or variants thereof, e.g.
  • NP or fragments thereof, e.g., PB1, or fragments or variants thereof, e.g., NS1 or fragments or variants thereof, e.g., M1 or fragments or variants thereof, and e.g. M2 or fragments or variants thereof including the extracellular fragment of M2 (eM2), or e.g., any of the foregoing polypeptides or fragments fused to a heterologous polypeptide, for example, a hepatitis B core antigen.
  • M2 extracellular fragment of M2
  • Isolated antigenic and immunogenic polypeptides of the present invention in addition to those encoded by polynucleotides of the invention, may be provided as a recombinant protein, a purified subunit, a viral vector expressing the protein, or may be provided in the form of an inactivated IV vaccine, e.g., a live-attenuated virus vaccine, a heat-killed virus vaccine, etc.
  • an inactivated IV vaccine e.g., a live-attenuated virus vaccine, a heat-killed virus vaccine, etc.
  • an “isolated” IV polypeptide or a fragment, variant, or derivative thereof is intended an IV polypeptide or protein that is not in its natural form. No particular level of purification is required.
  • an isolated IV polypeptide can be removed from its native or natural environment.
  • Recombinantly produced IV polypeptides and proteins expressed in host cells are considered isolated for purposed of the invention, as are native or recombinant IV polypeptides which have been separated, fractionated, or partially or substantially purified by any suitable technique, including the separation of IV virions from eggs or culture cells in which they have been propagated.
  • an isolated IV polypeptide or protein can be provided as a live or inactivated viral vector expressing an isolated IV polypeptide and can include those found in inactivated IV vaccine compositions.
  • isolated IV polypeptides and proteins can be provided as, for example, recombinant IV polypeptides, a purified subunit of IV, a viral vector expressing an isolated IV polypeptide, or in the form of an inactivated or attenuated IV vaccine.
  • epitopes refers to portions of a polypeptide having antigenic or immunogenic activity in a vertebrate, for example a human.
  • An “immunogenic epitope,” as used herein, is defined as a portion of a protein that elicits an immune response in an animal, as determined by any method known in the art.
  • antigenic epitope as used herein, is defined as a portion of a protein to which an antibody or T-cell receptor can immunospecifically bind as determined by any method well known in the art. Immunospecific binding excludes non-specific binding but does not exclude cross-reactivity with other antigens. Where all immunogenic epitopes are antigenic, antigenic epitopes need not be immunogenic.
  • immunogenic carrier refers to a first polypeptide or fragment, variant, or derivative thereof which enhances the immunogenicity of a second polypeptide or fragment, variant, or derivative thereof.
  • an “immunogenic carrier” is fused to or conjugated to the desired polypeptide or fragment thereof.
  • An example of an “immunogenic carrier” is a recombinant hepatitis B core antigen expressing, as a surface epitope, an immunogenic epitope of interest. See, e.g., European Patent No. EP 0385610 B 1, which is incorporated herein by reference in its entirety.
  • antigenic epitopes preferably contain a sequence of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, or between about 8 to about 30 amino acids contained within the amino acid sequence of an IV polypeptide of the invention, e.g., an NP polypeptide, an M1 polypeptide or an M2 polypeptide.
  • Certain polypeptides comprising immunogenic or antigenic epitopes are at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length.
  • Antigenic as well as immunogenic epitopes may be linear, i.e., be comprised of contiguous amino acids in a polypeptide, or may be three dimensional, i.e., where an epitope is comprised of non-contiguous amino acids which come together due to the secondary or tertiary structure of the polypeptide, thereby forming an epitope.
  • peptides or polypeptides bearing an antigenic epitope e.g., that contain a region of a protein molecule to which an antibody or T cell receptor can bind
  • relatively short synthetic peptides that mimic part of a protein sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked protein. See, e.g., Sutcliffe, J. G., et al., Science 219:660-666 (1983), which is herein incorporated by reference.
  • Peptides capable of eliciting an immunogenic response are frequently represented in the primary sequence of a protein, can be characterized by a set of simple chemical rules, and are confined neither to immunodominant regions of intact proteins nor to the amino or carboxyl terminals. Peptides that are extremely hydrophobic and those of six or fewer residues generally are ineffective at inducing antibodies that bind to the mimicked protein; longer peptides, especially those containing proline residues, usually are effective. Sutcliffe et al., supra, at 661.
  • Codon optimization is defined as modifying a nucleic acid sequence for enhanced expression in the cells of the vertebrate of interest, e.g. human, by replacing at least one, more than one, or a significant number, of codons of the native sequence with codons that are more frequently or most frequently used in the genes of that vertebrate.
  • Various species exhibit particular bias for certain codons of a particular amino acid.
  • the present invention relates to polynucleotides comprising nucleic acid fragments of codon-optimized coding regions which encode IV polypeptides, or fragments, variants, or derivatives thereof, with the codon usage adapted for optimized expression in the cells of a given vertebrate, e.g., humans.
  • These polynucleotides are prepared by incorporating codons preferred for use in the genes of the vertebrate of interest into the DNA sequence.
  • polynucleotide expression constructs comprising nucleic acid fragments of codon-optimized coding regions which encode IV polypeptides, and fragments, variants, or derivatives thereof, and various methods of using the polynucleotide expression constructs, vectors, host cells to treat or prevent influenza disease in a vertebrate.
  • codon-optimized coding region means a nucleic acid coding region that has been adapted for expression in the cells of a given vertebrate by replacing at least one, or more than one, or a significant number, of codons with one or more codons that are more frequently used in the genes of that vertebrate.
  • Deviations in the nucleotide sequence that comprise the codons encoding the amino acids of any polypeptide chain allow for variations in the sequence coding for the gene. Since each codon consists of three nucleotides, and the nucleotides comprising DNA are restricted to four specific bases, there are 64 possible combinations of nucleotides, 61 of which encode amino acids (the remaining three codons encode signals ending translation).
  • the “genetic code” which shows which codons encode which amino acids is reproduced herein as Table 1. As a result, many amino acids are designated by more than one codon.
  • amino acids alanine and proline are coded for by four triplets, serine and arginine by six, whereas tryptophan and methionine are coded by just one triplet.
  • This degeneracy allows for DNA base composition to vary over a wide range without altering the amino acid sequence of the proteins encoded by the DNA.
  • Codon preference or codon bias differences in codon usage between organisms, is afforded by degeneracy of the genetic code, and is well documented among many organisms. Codon bias often correlates with the efficiency of translation of messenger RNA (mRNA), which is in turn believed to be dependent on, inter alia, the properties of the codons being translated and the availability of particular transfer RNA (tRNA) molecules.
  • mRNA messenger RNA
  • tRNA transfer RNA
  • the predominance of selected tRNAs in a cell is generally a reflection of the codons used most frequently in peptide synthesis. Accordingly, genes can be tailored for optimal gene expression in a given organism based on codon optimization.
  • Codon usage tables are readily available, for example, at the “Codon Usage Database” available at http://www.kazusa.or.jp/codon/ (visited Jul. 9, 2002), and these tables can be adapted in a number of ways. See Nakamura, Y., et al. “Codon usage tabulated from the international DNA sequence databases: status for the year 2000” Nucl. Acids Res. 28:292 (2000), which is incorporated by reference. As examples, the codon usage tables for human, mouse, domestic cat, and cow, calculated from GenBank Release 128.0 (15 Feb.
  • Tables 2-5 are reproduced below as Tables 2-5. These Tables use mRNA nomenclature, and so instead of thymine (T) which is found in DNA, the Tables use uracil (U) which is found in RNA. The Tables have been adapted so that frequencies are calculated for each amino acid, rather than for all 64 codons.
  • Codon-optimized coding regions can be designed by various different methods.
  • codon usage table is used to find the single most frequent codon used for any given amino acid, and that codon is used each time that particular amino acid appears in the polypeptide sequence. For example, referring to Table 2 above, for leucine, the most frequent codon in humans is CUG, which is used 41% of the time. Thus all the leucine residues in a given amino acid sequence would be assigned the codon CUG.
  • SEQ ID NO 24 A coding region for IAV NP (SEQ ID NO:2) optimized by the “uniform optimization” method is presented herein as SEQ ID NO 24: 1 ATGGCCAGCC AGGGCACCAA GCGGAGCTAC GAGCAGATGG AGACCGACGG CGAGCGGCAG 61 AACGCCACCG AGATCCGGGC CAGCGTGGGC AAGATGATCG GCGGCATCGG CCGGTTCTAC 121 ATCCAGATGT GCACCGAGCT GAAGCTGAGC GACTACGAGG GCCGGCTGAT CCAGAACAGC 181 CTGACCATCG AGCGGATGGT GCTGAGCGCC TTCGACGAGC GGCGGAACAA GTACCTGGAG 241 GAGCACCCCA GCGCCGGCAA GGACCCCAAG AAGACCGGCG GCCCCATCTA CCGGCGGGTG 301 AACGGCAAGT GGATGCGGGA GCTGATCCTG TACGACAAGG AGGAGATCCG GCGGATCTGG 3
  • full-optimization the actual frequencies of the codons are distributed randomly throughout the coding region.
  • a hypothetical polypeptide sequence had 100 leucine residues, referring to Table 2 for frequency of usage in humans, about 7, or 7% of the leucine codons would be UUA, about 13, or 13% of the leucine codons would be WUG, about 13, or 13% of the leucine codons would be CUU, about 20, or 20% of the leucine codons would be CUC, about 7, or 7% of the leucine codons would be CUA, and about 41, or 41% of the leucine codons would be CUG.
  • nucleotide sequence for NP (SEQ ID NO:2) fully optimized for human codon usage, is shown as SEQ ID NO:23.
  • an entire polypeptide sequence may be codon-optimized as described above.
  • the fragment variant, or derivative may first be designed, and is then codon-optimized individually.
  • a full-length polypeptide sequence is codon-optimized for a given species resulting in a codon-optimized coding region encoding the entire polypeptide, and then nucleic acid fragments of the codon-optimized coding region, which encode fragments, variants, and derivatives of the polypeptide are made from the original codon-optimized coding region.
  • nucleic acid fragments encoding fragments, variants, and derivatives would not necessarily be fully codon-optimized for the given species. However, such sequences are still much closer to the codon usage of the desired species than the native codon usage.
  • the advantage of this approach is that synthesizing codon-optimized nucleic acid fragments encoding each fragment, variant, and derivative of a given polypeptide, although routine, would be time consuming and would result in significant expense.
  • the term “about” is used precisely to account for fractional percentages of codon frequencies for a given amino acid.
  • “about” is defined as one amino acid more or one amino acid less than the value given. The whole number value of amino acids is rounded up if the fractional frequency of usage is 0.50 or greater, and is rounded down if the fractional frequency of use is 0.49 or less.
  • the fractional frequency of codon usage would be calculated by multiplying 62 by the frequencies for the various codons.
  • 7.28 percent of 62 equals 4.51 UUA codons, or “about 5,” i.e., 4, 5, or 6 UUA codons, 12.66 percent of 62 equals 7.85 UUG codons or “about 8,” i.e., 7, 8, or 9 TUG codons, 12.87 percent of 62 equals 7.98 CUU codons, or “about 8,” i.e., 7, 8, or 9 CTU codons, 19.56 percent of 62 equals 12.13 CUC codons or “about 12,” i.e., 11, 12, or 13 CUC codons, 7.00 percent of 62 equals 4.34 CUA codons or “about 4,” i.e., 3, 4, or 5 CUA codons, and 40.62 percent of 62 equals 25.19 CUG codons, or “about 25,” i.e., 24, 25, or 26 CUG codons.
  • coding regions are only partially optimized.
  • the invention includes a nucleic acid fragment of a codon-optimized coding region encoding a polypeptide in which at least about 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% of the codon positions have been codon-optimized for a given species.
  • a codon that is preferentially used in the genes of a desired species e.g., a vertebrate species, e.g., humans
  • a codon that is normally used in the native nucleic acid sequence Codons that are rarely found in the genes of the vertebrate of interest are changed to codons more commonly utilized in the coding regions of the vertebrate of interest.
  • those codons which are used more frequently in the IV gene of interest than in genes of the vertebrate of interest are substituted with more frequently-used codons.
  • the difference in frequency at which the IV codons are substituted may vary based on a number factors as discussed below. For example, codons used at least twice more per thousand in IV genes as compared to genes of the vertebrate of interest are substituted with the most frequently used codon for that amino acid in the vertebrate of interest. This ratio may be adjusted higher or lower depending on various factors such as those discussed below.
  • a codon in an IV native coding region would be substituted with a codon used more frequently for that amino acid in coding regions of the vertebrate of interest if the codon is used 1.1 times, 1.2 times, 1.3 times, 1.4 times, 1.5 times, 1.6 times, 1.7 times, 1.8 times, 1.9 times, 2.0 times, 2.1 times, 2.2 times, 2.3 times, 2.4 times, 2.5 times, 2.6 times, 2.7 times, 2.8 times, 2.9 times, 3.0 times, 3.1 times, 3.2 times, 3.3. times, 3.4 times, 3.5 times, 3.6 times.
  • This minimal human codon optimization for highly variant codons has several advantages, which include but are not limited to the following examples. Since fewer changes are made to the nucleotide sequence of the gene of interest, fewer manipulations are required, which leads to reduced risk of introducing unwanted mutations and lower cost, as well as allowing the use of commercially available site-directed mutagenesis kits, and reducing the need for expensive oligonucleotide synthesis. Further, decreasing the number of changes in the nucleotide sequence decreases the potential of altering the secondary structure of the sequence, which can have a significant impact on gene expression in certain host cells. The introduction of undesirable restriction sites is also reduced, facilitating the subcloning of the genes of interest into the plasmid expression vector.
  • the present invention also provides isolated polynucleotides comprising coding regions of IV polypeptides, e.g., NP, M1, M2, HA, NA, PB1, PB2, PA, NS1 or NS2, or fragments, variants, or derivatives thereof.
  • isolated polynucleotides can also be codon-optimized.
  • a codon-optimized coding region encoding SEQ ID NO:2 is optimized according to codon usage in humans (Homo sapiens).
  • a codon-optimized coding region encoding SEQ ID NO:2 may be optimized according to codon usage in any plant, animal, or microbial species.
  • Codon-optimized coding regions encoding SEQ ID NO:2, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:2 is shown in Table 6.
  • a human codon-optimized coding region which encodes SEQ ID NO:2 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid.
  • codons are assigned to the coding region encoding SEQ ID NO:2 as follows: the 18 phenylalanine codons are TTC, the 33 leucine codons are CTG, the 26 isoleucine codons are ATC, the 25 methionine codons are ATG, the 23 valine codons are GTG, the 40 serine codons are AGC, the 17 proline codons are CCC, the 28 threonine codons are ACC, the 39 alanine codons are GCC, the 15 tyrosine codons are TAC, the 6 histidine codons are CAC, the 21 glutamine codons are CAG, the 26 asparagine codons are AAC, the 21 lysine codons are AAG, the 22 aspartic acid codons are GAC, the 36 glutamic acid codons are GAG, the 6 tryptophan codons are TGG, the 49 arginine codons are CGG
  • a human codon-optimized coding region which encodes SEQ ID NO:2 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 6 above.
  • codons are assigned to the coding region encoding SEQ ID NO:2 as follows: about 8 of the 18 phenylalanine codons are TTT, and about 10 of the phenylalanine codons are TTC; about 2 of the 33 leucine codons are TTA, about 4 of the leucine codons are TTG, about 4 of the leucine codons are CTT, about 6 of the leucine codons are CTC, about 2 of the leucine codons are CTA, and about 13 of the leucine codons are CTG; about 9 of the 26 isoleucine codons are ATT, about 13 of the isoleucine codons are ATC, and about 4 of the isoleucine codons are ATA; the 25 methionine codons are ATG; about 4 of the 23 valine codons are GTT, about 5 of the valine codons are GTG, about 3 of the valine codons are GTA, and about
  • the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • SEQ ID NO:23 A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:2, optimized according to codon usage in humans is presented herein as SEQ ID NO:23.
  • a minimally codon-optimized nucleotide sequence encoding SEQ ID NO:2 can be designed by changing only certain codons found more frequently in IV genes than in human genes, as shown in Table 7. For example, if it is desired to substitute more frequently used codons in humans for those codons that occur at least 2 times more frequently in IV genes (designated with an asterisk in Table 7), Arg AGA, which occurs 2.3 times more frequently in IV genes than in human genes, is changed to, e.g., CGG; Asn AAT, which occurs 2.0 times more frequently in IV genes than in human genes, is changed to, e.g., AAC; Ile ATA, which occurs 3.6 times more frequently in IV genes than in human genes, is changed to, e.g., ATC; and Leu CTA, which occurs 2.0 times more frequently in IV genes than is human, is changed to, e.g., CTG.
  • a Codon Usage Table for the specific IV sequence in question is generated and compared to CUT for human genomic DNA (see Table 7, supra). Amino acids are identified for which there is a difference of at least 10 percentage points in codon usage between human and IV DNA (either more or less). Then the wild type IV codon is modified to conform to predominant human codon for each such amino acid. Furthermore, the remainder of codons for that amino acid are also modified such that they conform to the predominant human codon for each such amino acid.
  • SEQ ID NO:25 A representative “minimally optimized” codon-optimized coding region encoding SEQ ID NO:2, minimally optimized according to codon usage in humans by this latter method, is presented herein as SEQ ID NO:25: 1 ATGGCCTCAC AGGGCACCAA GCGGAGTTAT GAGCAGATGG AGACCGATGG CGAGAGACAG 61 AACGCCACAG AGATCAGAGC CTCAGTTGGC AAGATGATCG GCGGCATCGG CCGGTTCTAT 121 ATCCAGATGT GCACGGAGCT GAAGCTGAGC GACTACGAGG GCAGACTGAT TCAGAACTCT 181 CTGACCATCG AGAGAATGGT CCTGAGTGCC TTCGATGAGA GACGAAACAA GTATCTGGAG 241 GAGCATCCCT CCGCCGGCAA GGACCCCAAG AAGACGGGCG GCCCCATATA TAGAAGAGTT 301 AACGGCAAGT GGATGAGAGA GCTGATCCTG TA
  • a codon-optimized coding region encoding SEQ ID NO:4 is optimized according to codon usage in humans ( Homo sapiens ).
  • a codon-optimized coding region encoding SEQ ID NO:4 may be optimized according to codon usage in any plant, animal, or microbial species.
  • Codon-optimized coding regions encoding SEQ ID NO:4, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:4 is shown in Table 8.
  • a human codon-optimized coding region which encodes SEQ ID NO:4 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid.
  • codons are assigned to the coding region encoding SEQ ID NO:4 as follows: the 7 phenylalanine codons are TTC, the 26 leucine codons are CTG, the 11 isoleucine codons are ATC, the 14 methionine codons are ATG, the 16 valine codons are GTG, the 18 serine codons are AGC, the 8 proline codons are CCC, the 18 threonine codons are ACC, the 25 alanine codons are GCC, the 5 tyrosine codons are TAC, the 5 histidine codons are CAC, the 15 glutamine codons are CAG, the 11 asparagine codons are AAC, the 13 lysine codons are AAG, the 6 aspartic acid codons are GAC, the 17 glutamic acid codons are GAG, the 1 tryptophan codon is TGG, the 17 arginine codons are CGG,
  • a human codon-optimized coding region which encodes SEQ ID NO:4 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 8 above.
  • codons are assigned to the coding region encoding SEQ ID NO:4 as follows: about 3 of the 7 phenylalanine codons are TTT, and about 4 of the phenylalanine codons are TTC; about 2 of the 26 leucine codons are TTA, about 3 of the leucine codons are TTG, about 3 of the leucine codons are CTT, about 5 of the leucine codons are CTC, about 2 of the leucine codons are CTA, and about 11 of the leucine codons are CTG; about 4 of the 11 isoleucine codons are ATT, about 5 of the isoleucine codons are ATC, and about 2 of the isoleucine codons are ATA; the 14 methionine codons are ATG; about 3 of the 16 valine codons are GTT, about 4 of the valine codons are GTG, about 2 of the valine codons are GTA, and about
  • the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • SEQ ID NO:26 ATGAGCTTGCTAACAGAAGTGGAAACCTATGTCCTCAGTATCATTCCTAG CGGCCCCTTAAAAGCCGAAATCGCTCAGCGGCTCGAGGATGTTTTTGCCG GCAAGAACACCGACCTGGAGGTATTGATGGAGTGGCTGAAAACGCGACCT ATTCTGAGCCCCCTGACTAAGGGAATACTCGGCTTCGTTTTTACATTGAC CGTGCCCTCAGAGAGGTCTCCAAAGGAGGCGCTTCGTGCAGAACGCCT TAAACGGGAACGGGGACCCAAATAATATGGATAAGGCAGTGAAACTGTAT CGCAAATTAAAGCGGGAGATAACCTTCCATGGAGCCAAGGAGATCTCCCT GTCTTACTCTGCAGGTGCTCTCGCGTCGTGTATGGGACTTATCTACAACC GAATGGGCCGTCACAACAGAAGTGGC
  • a minimally codon-optimized nucleotide sequence encoding SEQ ID NO:4 can be designed by changing only certain codons found more frequently in IV genes than in human genes, as shown in Table 7. For example, if it is desired to substitute more frequently used codons in humans for those codons that occur at least 2 times more frequently in IV genes (designated with an asterisk in Table 7), Arg AGA, which occurs 2.3 times more frequently in IV genes than in human genes, is changed to, e.g., CGG; Asn AAT, which occurs 2.0 times more frequently in IV genes than in human genes, is changed to, e.g., AAC; Ile ATA, which occurs 3.6 times more frequently in IV genes than in human genes, is changed to, e.g., ATC; and Leu CTA, which occurs 2.0 times more frequently in IV genes than is human, is changed to, e.g., CTG.
  • a Codon Usage Table for the specific IV sequence in question is generated and compared to CUT for human genomic DNA (see Table 7, supra). Amino acids are identified for which there is a difference of at least 10 percentage points in codon usage between human and IV DNA (either more or less). Then the wild type IV codon is modified to conform to predominant human codon for each such amino acid. Furthermore, the remainder of codons for that amino acid are also modified such that they conform to the predominant human codon for each such amino acid.
  • SEQ ID NO:28 ATGAGTCTGCTGACAGAGGTTGAGACGTACGTGCTGTCCATCATTCCCTC AGGCCCCCTGAAGGCCGAGATTGCCCAGAGACTGGAGGACGTCTTCGCCG GCAAGAACACCGATCTGGAGGTGCTGATGGAGTGGCTGAAGACTCGCCCC ATCCTGTCTCCCCTGACAAAGGGCATCCTGGGCTTCGTATTTACACTGAC CGTCCCCTCCGAGAGAGGCCTGCAGCGGAGGAGGTTCGTTCAGAACGCCC TGAACGGCAACGGCGATCCCAACAACATGGATAAGGCCGTGAAGCTGTAT AGAAAGCTGAAGCGAGAGATCACATTTCATGGCGCCAAGGAGATATCGCT GAGCTACAGTGCCGGCCCTGGCCTCTCTTGCATGGGCCTGATATACAACA GAATGGGC
  • a codon-optimized coding region encoding SEQ ID NO:5 is optimized according to codon usage in humans ( Homo sapiens ).
  • a codon-optimized coding region encoding SEQ ID NO:5 may be optimized according to codon usage in any plant, animal, or microbial species.
  • Codon-optimized coding regions encoding SEQ ID NO:5, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:5 is shown in Table 9.
  • a human codon-optimized coding region which encodes SEQ ID NO:5 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid.
  • codons are assigned to the coding region encoding SEQ ID NO:5 as follows: the 4 phenylalanine codons are TTC, the 10 leucine codons are CTG, the 8 isoleucine codons are ATC, the 2 methionine codons are ATG, the 4 valine codons are GTG, the 7 serine codons are AGC, the 4 proline codons are CCC, the 4 threonine codons are ACC, the 5 alanine codons are GCC, the 3 tyrosine codons are TAC, the 2 histidine codons are CAC, the 2 glutamine codons are CAG, the 3 asparagine codons are AAC, the 5 lysine codons are AAG, the 5 aspartic acid codons are GAC, the 9 glutamic acid codons are GAG, the 2 tryptophan codons are TGG, the 7 arginine codons are CGG
  • the codon-optimized PA coding region designed by this method is presented herein as SEQ ID NO:30: 1 ATGAGCCTGC TGACCGAGGT GGAGACCCCC ATCCGGAACG AGTGGGGCTG CCGGTGCAAC 61 GGCAGCAGCG ACCCCCTGGC CATCGCCGCC AACATCATCG GCATCCTGCA CCTGACCCTG 121 TGGATCCTGG ACCGGCTGTT CTTCAAGTGC ATCTACCGGC GGTTCAAGTA CGGCCTGAAG 181 GGCGGCCCCA GCACCGAGGG CGTGCCCAAG AGCATGCGGG AGGAGTACCG GAAGGAGCAG 241 CAGAGCGCCG TGGACGCCGA CGACGGCCAC TTCGTGAGCA TCGAGCTGGA GTGA
  • a human codon-optimized coding region which encodes SEQ ID NO:5 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 9 above.
  • codons are assigned to the coding region encoding SEQ ID NO:5 as follows: about 2 of the 4 phenylalanine codons are TTT, and about 2 of the phenylalanine codons are TTC; about 1 of the 10 leucine codons are TTA, about 1 of the leucine codons are TTG, about 1 of the leucine codons are CTT, about 2 of the leucine codons are CTC, about 1 of the leucine codons are CTA, and about 4 of the leucine codons are CTG; about 3 of the 8 isoleucine codons are ATT, about 4 of the isoleucine codons are ATC, and about 1 of the isoleucine codons are ATA; the 2 methionine codons are ATG; about 1 of the 4 valine codons are GTT, about 1 of the valine codons are GTG, about 0 of the valine codons are GTA, and
  • the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • SEQ ID NO:29 A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:5, optimized according to codon usage in humans is presented herein as SEQ ID NO:29: 1 ATGAGTCTTC TAACCGAGGT CGAAACGCCT ATCAGAAACG AATGGGGGTG CAGATGCAAC 61 GGTTCAAGTG ATCCTCTCGC TATTGCCGCA AATATCATTG GGATCTTGCA CTTGACATTG 121 TGGATTCTTG ATCGTCTTTT TTTCAAATGC ATTTACCGTC GCTTTAAATA CGGACTGAAA 181 GGAGGGCCTT CTACGGAAGG AGTGCCAAAG TCTATGAGGG AAGAATATCG AAAGGAACAG 241 CAGAGTGCTG TGGATGCTGA CGATGGTCAT TTTGTCAGCA TAGAGCTGGA GTAA
  • a minimally codon-optimized nucleotide sequence encoding SEQ ID NO:5 can be designed by changing only certain codons found more frequently in IV genes than in human genes, as shown in Table 7. For example, if it is desired to substitute more frequently used codons in humans for those codons that occur at least 2 times more frequently in IV genes (designated with an asterisk in Table 7), Arg AGA, which occurs 2.3 times more frequently in IV genes than in human genes, is changed to, e.g., CGG; Asn AAT, which occurs 2.0 times more frequently in IV genes than in human genes, is changed to, e.g., AAC; Ile ATA, which occurs 3.6 times more frequently in IV genes than in human genes, is changed to, e.g., ATC; and Leu CTA, which occurs 2.0 times more frequently in IV genes than is human, is changed to, e.g., CTG.
  • a Codon Usage Table for the specific IV sequence in question is generated and compared to CUT for human genomic DNA (see Table 7, supra). Amino acids are identified for which there is a difference of at least 10 percentage points in codon usage between human and IV DNA (either more or less). Then the wild type IV codon is modified to conform to predominant human codon for each such amino acid. Furthermore, the remainder of codons for that amino acid are also modified such that they conform to the predominant human codon for each such amino acid.
  • SEQ ID NO:31 A representative “minimally optimized” codon-optimized coding region encoding SEQ ID NO:5, minimally optimized according to codon usage in humans by this latter method, is presented herein as SEQ ID NO:31: 1 ATGTCTCTGC TGACAGAGGT GGAGACACCC ATAAGGAACG AGTGGGGCTG CAGGTGCAAC 61 GGCTCTAGTG ATCCCCTGGC CATCGCCGCC AACATCATTG GCATACTGCA TCTGACCCTG 121 TGGATCCTGG ATAGACTGTT CTTTAAGTGC ATTTACAGAC GATTTAAGTA TGGCCTGAAG 181 GGCGGCCCCT CAACTGAGGG CGTGCCCAAG AGTATGAGAG AGGAGTACCG GAAGGAGCAG 241 CAGAGCGCCG TTGACGCCGA TGACGGCCAC TTCGTCTCCA TCGAGCTGGA GTGA
  • a codon-optimized coding region encoding SEQ ID NO:7 is optimized according to codon usage in humans ( Homo sapiens ).
  • a codon-optimized coding region encoding SEQ ID NO:7 may be optimized according to codon usage in any plant, animal, or microbial species.
  • Codon-optimized coding regions encoding SEQ ID NO:7, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:7 is shown in Table 10.
  • a human codon-optimized coding region which encodes SEQ ID NO:7 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid.
  • codons are assigned to the coding region encoding SEQ ID NO:7 as follows: the 18 phenylalanine codons are TTC, the 35 leucine codons are CTG, the 27 isoleucine codons are ATC, the 26 methionine codons are ATG, the 24 valine codons are GTG, the 43 serine codons are AGC, the 18 proline codons are CCC, the 30 threonine codons are ACC, the 39 alanine codons are GCC, the 15 tyrosine codons are TAC, the 6 histidine codons are CAC, the 21 glutamine codons are CAG, the 28 asparagine codons are AAC, the 21 lysine codons are AAG, the 23 aspartic acid codons are GAC, the 39 glutamic acid codons are GAG, the 7 tryptophan codons are TGG, the 51 arginine codons are CGG
  • a human codon-optimized coding region which encodes SEQ ID NO:7 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 10 above.
  • codons are assigned to the coding region encoding SEQ ID NO:7 as follows: about 8 of the 18 phenylalanine codons are TTT, and about 10 of the phenylalanine codons are TTC; about 3 of the 35 leucine codons are TTA, about 4 of the leucine codons are TTG, about 5 of the leucine codons are CTT, about 7 of the leucine codons are CTC, about 2 of the leucine codons are CTA, and about 14 of the leucine codons are CTG; about 10 of the 27 isoleucine codons are ATT, about 13 of the isoleucine codons are ATC, and about 4 of the isoleucine codons are ATA; the 26 methionine codons are ATG; about 4 of the 24 valine codons are GTT, about 6 of the valine codons are GTG, about 3 of the valine codons are GTA, and about
  • the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • SEQ ID NO:32 ATGAGCCTTCTCACAGAAGTGGAAACACCTATCAGAAATGAATGGGGATG CAGATGCAATGGGTCGAGTGATATGGCCTCTCAAGGTACGAAAAGAAGCT ACGAGCAAATGGAAACGGATGGAGAAAGACAAAACGCGACCGAAATCAGA GCATCCGTCGGGAAGATGATTGGAGGAATCGGACGATTCTACATCCAGAT GTGCACAGAGCTAAAGCTATCGGATTATGAAGGGAGACTAATACAAAATA GCCTAACTATCGAGAGAATGGTGCTGTCTGCATTTGACGAAAGGAGAAAC AAATACCTGGAAGAACACCCCTCTGCAGGGAAAGACCCAAAAAAAAAACTGG AGGTCCGATATACCGGAGAGTCAACGGTAAATGGATGAAAATAAAAAAAAAAAAAAAAACTGG AGGTCCGATATACCGGAGAGTCAACGGTAAATGGATGAAAATAAAAAAAAAAAAAAAAACTGG AGGTCCGATATACCGGAGAGTCAACGGT
  • a codon-optimized coding region encoding SEQ ID NO:9 is optimized according to codon usage in humans ( Homo sapiens ).
  • a codon-optimized coding region encoding SEQ ID NO:9 may be optimized according to codon usage in any plant, animal, or microbial species.
  • Codon-optimized coding regions encoding SEQ ID NO:9, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:9 is shown in Table 11.
  • a human codon-optimized coding region which encodes SEQ ID NO:9 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid.
  • codons are assigned to the coding region encoding SEQ ID NO:9 as follows: the 18 phenylalanine codons are TTC, the 35 leucine codons are CTG, the 27 isoleucine codons are ATC, the 26 methionine codons are ATG, the 24 valine codons are GTG, the 43 serine codons are AGC, the 18 proline codons are CCC, the 30 threonine codons are ACC, the 39 alanine codons are GCC, the 15 tyrosine codons are TAC, the 6 histidine codons are CAC, the 21 glutamine codons are CAG, the 28 asparagine codons are AAC, the 21 lysine codons are AAG, the 23 aspartic acid codons are GAC, the 39 glutamic acid codons are GAG, the 7 tryptophan codons are TGG, the 51 arginine codons are CGG
  • a human codon-optimized coding region which encodes SEQ ID NO:9 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 11 above.
  • codons are assigned to the coding region encoding SEQ ID NO:9 as follows: about 8 of the 18 phenylalanine codons are TTT, and about 10 of the phenylalanine codons are TTC; about 3 of the 35 leucine codons are TTA, about 4 of the leucine codons are TTG, about 5 of the leucine codons are CTT, about 7 of the leucine codons are CTC, about 2 of the leucine codons are CTA, and about 14 of the leucine codons are CTG; about 10 of the 27 isoleucine codons are ATT, about 13 of the isoleucine codons are ATC, and about 4 of the isoleucine codons are ATA; the 26 methionine codons are ATG; about 4 of the 24 valine codons are GTT, about 6 of the valine codons are GTG, about 3 of the valine codons are GTA, and about
  • the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • SEQ ID NO:34 ATGGCAAGCCAGGGCACAAAACGCAGTTACGAGCAGATGGAGACTGATGG TGAGAGGCAGAACGCCACCGAAATCCGGGCCTCCGTCGGCAAGATGATTG GTGGCATCGGAAGATTCTATATCCAGATGTGCACGGAGCTTAAGCTGTCC GATTACGAGGGGCGCTTAATACAGAACTCTCTGACTATCGAGCGAATGGT CTTGAGCGCCTTTGATGAGCGGCGTAATAAGTATCTCGAAGAGCACCCTT CTGCTGGAAAAGACCCCAAAAAGACCGGGGGACCTATCTACCGACGTGTG AACGGAAAATGGATGCGCGAACTGATACTGTACGACAAGGAGGATCCG TAGGATCTGGAGACAGGCTAATAACGGAGATGATGCCACAGCTGGGCTGA CCCATATGATGATATGGCATAG
  • a codon-optimized coding region encoding SEQ ID NO:16 is optimized according to codon usage in humans ( Homo sapiens ).
  • a codon-optimized coding region encoding SEQ ID NO:16 may be optimized according to codon usage in any plant, animal, or microbial species.
  • Codon-optimized coding regions encoding SEQ ID NO:16, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:16 is shown in Table 12.
  • a human codon-optimized coding region which encodes SEQ ID NO: 16 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid.
  • codons are assigned to the coding region encoding SEQ ID NO:16 as follows: the 21 phenylalanine codons are TTC, the 39 leucine codons are CTG, the 38 isoleucine codons are ATC, the 27 methionine codons are ATG, the 32 valine codons are GTG, the 40 serine codons are AGC, the 26 proline codons are CCC, the 38 threonine codons are ACC, the 41 alanine codons are GCC, the 14 tyrosine codons are TAC, the 4 histidine codons are CAC, the 19 glutamine codons are CAG, the 25 asparagine codons are AAC, the 52 lysine codons are AAG, the 34 aspartic acid codons are GAC, the 30 glutamic acid codons are GAG, the 1 tryptophan codon is TGG, the 30 arginine codons are CGG,
  • a human codon-optimized coding region which encodes SEQ ID NO:16 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 12 above.
  • codons are assigned to the coding region encoding SEQ ID NO:16 as follows: about 10 of the 21 phenylalanine codons are TTT, and about 12 of the phenylalanine codons are TTC; about 3 of the 39 leucine codons are TTA, about 5 of the leucine codons are TTG, about 5 of the leucine codons are CTT, about 8 of the leucine codons are CTC, about 3 of the leucine codons are CTA, and about 16 of the leucine codons are CTG; about 14 of the 38 isoleucine codons are ATT, about 18 of the isoleucine codons are ATC, and about 6 of the isoleucine codons are ATA; the 27 methionine codons are ATG; about 6 of the 32 valine codons are GTT, about 8 of the valine codons are GTG, about 4 of the valine codons are GTA, and about
  • the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • SEQ ID NO:36 A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:16, optimized according to codon usage in humans is presented herein as SEQ ID NO:36: ATGTCGAACATGGACATCGACAGCATTAACACAGGTACTATTGACAAAAC CCCCGAAGAACTAACCCCTGGAACCTCAGGAGCAACACGCCCAATAATCA AACCGGCCACCCTCGCGCCCCCTAGCAATAAGAGGACCCGCAATCCAAGT CCTGAGAGAACCACTACTTCATCTGAAACGGATATCGGTCGGAAAATTCA AAAAAAGCAGACGCCCACAGAGATAAAGAAGTCTGTTTACAAAATGGTGG TAAAGCTCGGTGAGTTTTATAACCAGATGATGGTCAAGGCGGGGCTTAAC GACGATATGGAACGAAATCTTATACAGAATGCACAGGCAGTAGAGAAAAT ACTGCTGGCCGCTACTGATGACAAGAAAACGGAGTACCAAAAAACGGAATGCACAGAAATTAAACCAAAAAA
  • a minimally codon-optimized nucleotide sequence encoding SEQ ID NO:16 can be designed by changing only certain codons found more frequently in IV genes than in human genes, as shown in Table 7. For example, if it is desired to substitute more frequently used codons in humans for those codons that occur at least 2 times more frequently in IV genes (designated with an asterisk in Table 7), Arg AGA, which occurs 2.3 times more frequently in IV genes than in human genes, is changed to, e.g., CGG; Asn AAT, which occurs 2.0 times more frequently in IV genes than in human genes, is changed to, e.g., AAC; Ile ATA, which occurs 3.6 times more frequently in IV genes than in human genes, is changed to, e.g., ATC; and Leu CTA, which occurs 2.0 times more frequently in IV genes than is human, is changed to, e.g., CTG.
  • a Codon Usage Table for the specific IV sequence in question is generated and compared to CUT for human genomic DNA (see Table 7, supra). Amino acids are identified for which there is a difference of at least 10 percentage points in codon usage between human and IV DNA (either more or less). Then the wild type IV codon is modified to conform to predominant human codon for each such amino acid. Furthermore, the remainder of codons for that amino acid are also modified such that they conform to the predominant human codon for each such amino acid.
  • SEQ ID NO:38 ATGTCTAACATGGACATCGACTCTATAAACACAGGCACGATCGATAAGAC CCCCGAGGAGCTGACACCCGGGACTTCAGGCGCCACCAGACCCATAATAA AGGCCGCGCGACTCTGGCCCCCCTCTAACAAGAGGAGGAGGAACCCCTCT CCCGAGCGCACCACAACGAGTAGCGAGACGGACATCGGCAGGAAGATACA GAAGAAGCAGACTCCCACTGAGATTAAGAAGTGCGTGTATAAGATGGTGG TTAAGCTGGGCGAGTTTTACAACCAGATGATGGTGAAGGCCGGCCTGAAC GATGACATGGAGAGGAACCTGATACAGAACGCCCAGGCCGTGGAGAGGAT TCTGCTGGCCGCCACCGATGACAAGAAGACTGAGTATCAGAAGAAGAA ACGCCCGGGACGT
  • Randomly assigning codons at an optimized frequency to encode a given polypeptide sequence using the “full-optimization” or “minimal optimization” methods can be done manually by calculating codon frequencies for each amino acid, and then assigning the codons to the polypeptide sequence randomly.
  • various algorithms and computer software programs are readily available to those of ordinary skill in the art. For example, the “EditSeq” function in the Lasergene Package, available from DNAstar, Inc., Madison, Wis., the backtranslation function in the VectorNTI Suite, available from InforMax, Inc., Bethesda, Md., and the “backtranslate” function in the GCG—Wisconsin Package, available from Accelrys, Inc., San Diego, Calif.
  • a number of options are available for synthesizing codon-optimized coding regions designed by any of the methods described above, using standard and routine molecular biological manipulations well known to those of ordinary skill in the art.
  • a series of complementary oligonucleotide pairs of 80-90 nucleotides each in length and spanning the length of the desired sequence are synthesized by standard methods.
  • oligonucleotide pairs are synthesized such that upon annealing, they form double stranded fragments of 80-90 base pairs, containing cohesive ends, e.g., each oligonucleotide in the pair is synthesized to extend 3, 4, 5, 6, 7, 8, 9, 10, or more bases beyond the region that is complementary to the other oligonucleotide in the pair.
  • the single-stranded ends of each pair of oligonucleotides is designed to anneal with the single-stranded end of another pair of oligonucleotides.
  • the oligonucleotide pairs are allowed to anneal, and approximately five to six of these double-stranded fragments are then allowed to anneal together via the cohesive single stranded ends, and then they ligated together and cloned into a standard bacterial cloning vector, for example, a TOPO® vector available from Invitrogen Corporation, Carlsbad, Calif.
  • the construct is then sequenced by standard methods. Several of these constructs consisting of 5 to 6 fragments of 80 to 90 base pair fragments ligated together, i.e., fragments of about 500 base pairs, are prepared, such that the entire desired sequence is represented in a series of plasmid constructs.
  • the inserts of these plasmids are then cut with appropriate restriction enzymes and ligated together to form the final construct.
  • the final construct is then cloned into a standard bacterial cloning vector, and sequenced. Additional methods would be immediately apparent to the skilled artisan. In addition, gene synthesis is readily available commercially.
  • the codon-optimized coding regions can be versions encoding any gene products from any strain, derivative, or variant of IV, or fragments, variants, or derivatives of such gene products.
  • Codon-optimized coding regions encoding other IV polypeptides or fragments, variants, or derivatives thereof e.g. HA, NA, PB1, PB2, PA, NS1 or NS2
  • Additional, non-codon-optimized polynucleotides encoding IV polypeptides or other polypeptides are included as well.
  • the present invention is further directed to specific consensus sequences of influenza virus proteins, and fragments, derivatives and variants thereof.
  • a “consensus sequence” is, e.g., an idealized sequence that represents the amino acids most often present at each position of two or more sequences which have been compared to each other.
  • a consensus sequence is a theoretical representative amino acid sequence in which each amino acid is the one which occurs most frequently at that site in the different sequences which occur in nature. The term also refers to an actual sequence which approximates the theoretical consensus.
  • a consensus sequence can be derived from sequences which have, e.g., shared functional or structural purposes. It can be defined by aligning as many known examples of a particular structural or functional domain as possible to maximize the homology.
  • a sequence is generally accepted as a consensus when each particular amino acid is reasonably predominant at its position, and most of the sequences which form the basis of the comparison are related to the consensus by rather few substitutions, e.g., from 0 to about 100 substitutions.
  • the wild-type comparison sequences are at least about 50%, 75%, 80%, 90%, 95%, 96%, 97%, 98% or 99% identical to the consensus sequence.
  • polypeptides of the invention are about 50%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the consensus sequence.
  • Consensus amino acid sequences can be prepared for any of the influenza antigens.
  • a “consensus amino acid” is an amino acid chosen to occupy a given position in the consensus protein.
  • a system which is organized to select consensus amino acids can be a computer program, or a combination of one or more computer programs with “by hand” analysis and calculation. When a consensus amino acid is obtained for each position of the aligned amino acid sequences, then these consensus amino acids are “lined up” to obtain the amino acid sequence of the consensus protein.
  • Another embodiment of this invention is directed to a process for the preparation of a consensus protein comprising a process to calculate an amino acid residue for nearly all positions of a so-called consensus protein and to synthesize a complete gene from this sequence that could be expressed in a prokaryotic or eukaryotic expression system.
  • Polynucleotides which encode the consensus influenza polypeptides, or fragments, variants or derivatives thereof, are also part of this invention.
  • Such polynucleotides can be obtained by known methods, for example by backtranslation of the amino acid sequence and PCR synthesis of the corresponding polynucleotide.
  • the present invention is directed to compositions and methods of enhancing the immune response of a vertebrate in need of protection against IV infection by administering in vivo, into a tissue of a vertebrate, one or more polynucleotides comprising at least one codon-optimized coding region encoding an IV polypeptide, or a fragment, variant, or derivative thereof.
  • the present invention is directed to compositions and methods of enhancing the immune response of a vertebrate in need of protection against IV infection by administering to the vertebrate a composition comprising one or more polynucleotides as described herein, and at least one isolated IV polypeptide, or a fragment, variant, or derivative thereof.
  • the polynucleotide may be administered either prior to, at the same time (simultaneously), or subsequent to the administration of the isolated polypeptide.
  • the coding regions encoding IV polypeptides or fragments, variants, or derivatives thereof may be codon optimized for a particular vertebrate. Codon optimization is carried out by the methods described herein, for example, in certain embodiments codon-optimized coding regions encoding polypeptides of IV, or nucleic acid fragments of such coding regions encoding fragments, variants, or derivatives thereof are optimized according to the codon usage of the particular vertebrate.
  • the polynucleotides of the invention are incorporated into the cells of the vertebrate in vivo, and an immunologically effective amount of an IV polypeptide or a fragment, variant, or derivative thereof is produced in vivo.
  • the coding regions encoding an IV polypeptide or a fragment, variant, or derivative thereof may be codon optimized for mammals, e.g., humans, apes, monkeys (e.g., owl, squirrel, cebus, rhesus, African green, patas, cynomolgus, and cercopithecus), orangutans, baboons, gibbons, and chimpanzees, dogs, wolves, cats, lions, and tigers, horses, donkeys, zebras, cows, pigs, sheep, deer, giraffes, bears, rabbits, mice, ferrets, seals, whales; birds, e.g., ducks, geese, terns, shearwaters, gulls, turkeys, chickens, quail, pheasants, geese, starlings and budgerigars, or other vertebrates.
  • mammals e.g.,
  • the present invention relates to codon-optimized coding regions encoding polypeptides of IV, or nucleic acid fragments of such coding regions fragments, variants, or derivatives thereof which have been optimized according to human codon usage.
  • human codon-optimized coding regions encoding polypeptides of IV, or fragments, variants, or derivatives thereof are prepared by substituting one or more codons preferred for use in human genes for the codons naturally used in the the DNA sequence encoding the IV polypeptide or a fragment, variant, or derivative thereof.
  • Coding regions encoding IV polypeptides can be uniformly optimized, fully optimized, minimally optimized, codon-optimized by region and/or not codon-optimized, as described herein.
  • the present invention is further directed towards polynucleotides comprising codon-optimized coding regions encoding polypeptides of IV antigens, for example, HA, NA, NP, M1 and M2, optionally in conjunction with other antigens.
  • the invention is also directed to polynucleotides comprising codon-optimized nucleic acid fragments encoding fragments, variants and derivatives of these polypeptides, e.g., an eM2 or a fusion of NP and eM2.
  • the present invention provides an isolated polynucleotide comprising a nucleic acid fragment, where the nucleic acid fragment is a fragment of a codon-optimized coding region encoding a polypeptide at least 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to an IV polypeptide, e.g., HA, NA, NP, M1 or M2, and where the nucleic acid fragment is a variant of a codon-optimized coding region encoding an IV polypeptide, e.g., HA, NA, NP, M1 or M2.
  • the human codon-optimized coding region can be optimized for any vertebrate species and by any of the methods described herein.
  • compositions which include at least one polynucleotide comprising one or more nucleic acid fragments, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide or fragment, variant, or derivative thereof; together with one or more isolated IV component or isolated polypeptide.
  • the IV component may be inactivated virus, attenuated virus, a viral vector expressing an isolated influenza virus polypeptide, or an influenza virus protein, fragment, variant or derivative thereof.
  • polypeptides or fragments, variants or derivatives thereof, in combination with the codon-optimized nucleic acid compositions may be referred to as “combinatorial polynucleotide vaccine compositions” or “single formulation heterologous prime-boost vaccine compositions.”
  • isolated IV polypeptides of the invention may be in any form, and are generated using techniques well known in the art. Examples include isolated IV proteins produced recombinantly, isolated IV proteins directly purified from their natural milieu, recombinant (non-IV) virus vectors expressing an isolated IV protein, or proteins delivered in the form of an inactivated IV vaccine, such as conventional vaccines
  • an isolated IV polypeptide or fragment, variant or derivative thereof is administered in an immunologically effective amount.
  • Conventional IV vaccines have been standardized to micrograms of viral antigens HA and NA. See Subbarao, K., Advances in Viral Research 54:349-373 (1999), incorporated herein by reference in its entirety. The recommended dose for these vaccines is 15 ⁇ g of each HA per 0.5 ml. Id.
  • the effective amount of conventional IV vaccines is determinable by one of ordinary skill in the art based upon several factors, including the antigen being expressed, the age and weight of the subject, and the precise condition requiring treatment and its severity, and route of administration.
  • the combination of conventional antigen vaccine compositions with the codon-optimized nucleic acid compositions provides for therapeutically beneficial effects at dose sparing concentrations.
  • immunological responses sufficient for a therapeutically beneficial effect in patients predetermined for an approved commercial product, such as for the conventional product described above, can be attained by using less of the approved commercial product when supplemented or enhanced with the appropriate amount of codon-optimized nucleic acid.
  • dose sparing is contemplated by administration of conventional IV vaccines administered in combination with the codon-optimized nucleic acids of the invention
  • the dose of conventional vaccine may be reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% or at least 70% when administered in combination with the codon-optimized nucleic acid compositions of the invention.
  • a desirable level of an immunological response afforded by a DNA based pharmaceutical alone may be attained with less DNA by including an aliquot of a conventional vaccine.
  • using a combination of conventional and DNA based pharmaceuticals may allow both materials to be used in lesser amounts while still affording the desired level of immune response arising from administration of either component alone in higher amounts (e.g. one may use less of either immunological product when they are used in combination). This may be manifest not only by using lower amounts of materials being delivered at any time, but also to reducing the number of administrations points in a vaccination regime (e.g. 2 versus 3 or 4 injections), and/or to reducing the kinetics of the immunological response (e.g. desired response levels are attained in 3 weeks in stead of 6 after immunization).
  • the dose of DNA based pharmaceuticals may be reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% or at least 70% when administered in combination with conventional IV vaccines.
  • Determining the precise amounts of DNA based pharmaceutical and conventional antigen is based on a number of factors as described above, and is readily determined by one of ordinary skill in the art.
  • the claimed combinatorial compositions provide for a broadening of the immune response and/or enhanced beneficial immune responses.
  • Such broadened or enhanced immune responses are achieved by: adding DNA to enhance cellular responses to a conventional vaccine; adding a conventional vaccine to a DNA pharmaceutical to enhance humoral response; using a combination that induces additional epitopes (both humoral and/or cellular) to be recognized and/or more desirably responded to (epitope broadening); employing a DNA-conventional vaccine combination designed for a particular desired spectrum of immunological responses; obtaining a desirable spectrum by using higher amounts of either component.
  • the broadened immune response is measurable by one of ordinary skill in the art by standard immunological assay specific for the desirable response spectrum.
  • the isolated IV polypeptide or fragment, variant, or derivative thereof to be delivered can be any isolated IV polypeptide or fragment, variant, or derivative thereof, including but not limited to the HA, NA, NP, M1, or M2 proteins or fragments, variants or derivatives thereof. Fragments include, but are not limited to, the eM2 protein.
  • a derivative protein can be a fusion protein, e.g., NP-eM2.
  • any isolated IV polypeptide or fragment, variant, or derivative thereof described herein can be combined in a composition with any polynucleotide comprising a nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide or fragment, variant, or derivative thereof.
  • the proteins can be different, the same, or can be combined in any combination of one or more isolated IV proteins and one or more polynucleotides.
  • the isolated IV polypeptides, or fragments, derivatives or variants thereof can be fused to or conjugated to a second isolated IV polypeptide, or fragment, derivative or variant thereof, or can be fused to other heterologous proteins, including for example, hepatitis B proteins including, but not limited to the hepatitis B core antigen (HBcAg), or those derived from diphtheria or tetanus.
  • the second isolated IV polypeptide or other heterologous protein can act as a “carrier” that potentiates the immunogenicity of the IV polypeptide or a fragment, variant, or derivative thereof to which it is attached.
  • Hepatitis B virus proteins and fragments and variants thereof useful as carriers within the scope of the invention are disclosed in U.S. Pat. Nos. 6,231,864 and 5,143,726, which are incorporated by reference in their entireties. Polynucleotides comprising coding regions encoding said fused or conjugated proteins are also within the scope of the invention.
  • HBcAg hepatitis B core antigen
  • heterologous protein sequences as potent immunogenic moieties.
  • addition of heterologous sequences to the amino terminus of a recombinant HBcAg results in the spontaneous assembly of particulate structures which express the heterologous epitope on their surface, and which are highly immunogenic when inoculated into experimental animals.
  • Heterologous epitopes can also be inserted into HBcAg particles by replacing approximately 40 amino acids of the carboxy terminus of the protein with the heterologous sequences.
  • HBcAg particles may be constructed where the heterologous epitope is inserted in or replaces all or part of the sequence of amino acid residues in a more central region of the HBcAg protein, in an immunodominant loop, thereby allowing the heterologous epitope to be displayed on the surface of the resulting particles. See EP Patent No. 0421635 B1.
  • HBc human hepatitis B core protein
  • SEQ ID NOs 39 and 40 subtype ayw
  • All of the above references are incorporated herein by reference in their entireties.
  • Chimaeric HBcAg particles comprising isolated IV proteins or variants, fragments or derivatives thereof are prepared by recombinant techniques well known to those of ordinary skill in the art.
  • a polynucleotide e.g., a plasmid, which carries the coding region for the HBcAg operably associated with a promoter is constructed.
  • Convenient restrictions sites are engineered into the coding region encoding the N-terminal, central, and/or C-terminal portions of the HBcAg, such that heterologous sequences may be inserted.
  • a construct which expresses a HBcAg/IV fusion protein is prepared by inserting a DNA sequence encoding an IV protein or variant, fragment or derivative thereof, in frame, into a desired restriction site in the coding region of the HBcAg.
  • the resulting construct is then inserted into a suitable host cell, e.g., E. coli, under conditions where the chimeric HBcAg will be expressed.
  • the chimaeric HBcAg self-assembles into particles when expressed, and can then be isolated, e.g., by ultracentrifugation.
  • the particles formed resemble the natural 27 nm HBcAg particles isolated from a hepatitis B virus, except that an isolated IV protein or fragment, variant, or derivative thereof is contained in the particle, preferably exposed on the outer particle surface.
  • the IV protein or fragment, variant, or derivative thereof expressed in a chimaeric HBcAg particle may be of any size which allows suitable particles of the chimeric HBcAg to self-assemble.
  • suitable particles of the chimeric HBcAg may be immunogenic when expressed in the context of an immunogenic carrier, e.g., a HBcAg.
  • HBcAg particles of the invention may comprise at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, or between about 15 to about 30 amino acids of an IV protein fragment of interest inserted therein.
  • HBcAg particles of the invention may further comprise immunogenic or antigenic epitopes of at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues of an IV protein fragment of interest inserted therein.
  • the immunodominant loop region of HBcAg was mapped to about amino acid residues 75 to 83, to about amino acids 75 to 85 or to about amino acids 130 to 140. See Colucci et al., J. Immunol. 141:4376-4380 (1988), and Salfeld et al. J. Virol. 63:798 (1989), which are incorporated by reference.
  • a chimeric HBcAg is still often able to form core particles when foreign epitopes are cloned into the immunodominant loop.
  • amino acids of the IV protein fragment may be inserted into the sequence of HBcAg amino acids at various positions, for example, at the N-terminus, from about amino acid 75 to about amino acid 85, from about amino acid 75 to about amino acid 83, from about amino acid 130 to about amino acid 140, or at the C-terminus.
  • amino acids of the IV protein fragment replace all or part of the native core protein sequence
  • the inserted IV sequence is generally not shorter, but may be longer, than the HBcAg sequence it replaces.
  • full-length IV coding sequences can be fused to the coding region for the HBcAg.
  • the HBcAg sequences can be fused either at the N- or C-terminus of any of the Influenza antigens described herein, including the eM2-NP constructs. Fusions could include flexible protein linkers as described for NP-eM2 fusions above.
  • IV coding sequences fused to the HBcAg coding sequence of SEQ ID NO:41 include an IAV NP-HBcAg fusion (SEQ ID NO:43), ATGGCGTCTCAAGGCACCAAACGATCTTACGAACAGATGGAGACTGATG GAGAACGCCAGAATGCCACTGAAATCAGAGCATCCGTCGGAAAAATGAT TGGTGGAATTGGACGATTCTACATCCAAATGTGCACCGAACTCAAACTCA GTGATTATGAGGGACGGTTGATCCAAAACAGCTTAACAATAGAGAGAAAAT GGTGCTCTCTGCTTTTGACGAAAGGAGAAATAAATACCTTGAAGAACATC CCAGTGCGGGGAAAGATCCTAAGAAAACTGGAGGACCTATATACAGGAG AGTAAACGGAAAGTGGATGAGAGAACTCATCCTTTATGACAAAGAAGAA ATAAGAAATAAGAAATAAGAAATAAGAAATAAGAAATAAGAAATAAGAAATAAGAAATAAGAAATAAGAAATAAGAAATAAGAAATAAG
  • the chimeric HBcAg can be used in the present invention in conjunction with a polynucleotide comprising a nucleic acid fragment, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide, or a fragment, variant, or derivative thereof, as an influenza vaccine for a vertebrate.
  • the present invention also provides methods for delivering an IV polypeptide or a fragment, variant, or derivative thereof to a human, which comprise administering to a human one or more of the compositions described herein; such that upon administration of compositions such as those described herein, an IV polypeptide or a fragment, variant, or derivative thereof is expressed in human cells, in an amount sufficient to generate an immune response to the IV or administering the IV polypeptide or a fragment, variant, or derivative thereof itself to the human in an amount sufficient to generate an immune response.
  • the present invention further provides methods for delivering an IV polypeptide or a fragment, variant, or derivative thereof to a human, which comprise administering to a vertebrate one or more of the compositions described herein; such that upon administration of compositions such as those described herein, an immune response is generated in the vertebrate.
  • vertebrate is intended to encompass a singular “vertebrate” as well as plural “vertebrates” and comprises mammals and birds, as well as fish, reptiles, and amphibians.
  • mammal is intended to encompass a singular “mammal” and plural “mammals,” and includes, but is not limited to humans; primates such as apes, monkeys (e.g., owl, squirrel, cebus, rhesus, African green, patas, cynomolgus, and cercopithecus), orangutans, baboons, gibbons, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equines such as horses, donkeys, and zebras, food animals such as cows, pigs, and sheep; ungulates such as deer and giraffes; ursids such as bears; and others such as rabbits, mice, ferrets, seals, whales.
  • the mammal can be a human subject, a food animal or a companion animal.
  • bird is intended to encompass a singular “bird” and plural “birds,” and includes, but is not limited to feral water birds such as ducks, geese, terns, shearwaters, and gulls; as well as domestic avian species such as turkeys, chickens, quail, pheasants, geese, and ducks.
  • the term “bird” also encompasses passerine birds such as starlings and budgerigars.
  • the present invention further provides a method for generating, enhancing or modulating an immune response to an W comprising administering to a vertebrate one or more of the compositions described herein.
  • the compositions may include one or more isolated polynucleotides comprising at least one nucleic acid fragment where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide, or a fragment, variant, or derivative thereof.
  • compositions may include both a polynucleotide as described above, and also an isolated IV polypeptide, or a fragment, variant, or derivative thereof, wherein the protein is provided as a recombinant protein, in particular, a fusion protein, a purified subunit, viral vector expressing the protein, or in the form of an inactivated IV vaccine.
  • the latter compositions include both a polynucleotide encoding an IV polypeptide or a fragment, variant, or derivative thereof and an isolated IV polypeptide or a fragment, variant, or derivative thereof.
  • compositions to be used according to this method may be univalent, bivalent, trivalent or multivalent.
  • the polynucleotides of the compositions may comprise a fragment of a human (or other vertebrate) codon-optimized coding region encoding a protein of the IV, or a fragment, variant, or derivative thereof.
  • the polynucleotides are incorporated into the cells of the vertebrate in vivo, and an antigenic amount of the IV polypeptide, or fragment, variant, or derivative thereof, is produced in vivo.
  • the IV polypeptide or a fragment, variant, or derivative thereof is expressed in the vertebrate in an amount sufficient to elicit an immune response.
  • an immune response might be used, for example, to generate antibodies to the IV for use in diagnostic assays or as laboratory reagents, or as therapeutic or preventative vaccines as described herein.
  • the present invention further provides a method for generating, enhancing, or modulating a protective and/or therapeutic immune response to IV in a vertebrate, comprising administering to a vertebrate in need of therapeutic and/or preventative immunity one or more of the compositions described herein.
  • the compositions include one or more polynucleotides comprising at least one nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide, or a fragment, variant, or derivative thereof.
  • the composition used in this method includes both an isolated polynucleotide comprising at least one nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide, or a fragment, variant, or derivative thereof; and at least one isolated IV polypeptide, or a fragment, variant, or derivative thereof.
  • the latter composition includes both an isolated polynucleotide encoding an IV polypeptide or a fragment, variant, or derivative thereof and an isolated IV polypeptide or a fragment, variant, or derivative thereof, for example, a recombinant protein, a purified subunit, viral vector expressing the protein, or an inactivated virus vaccine.
  • the IV polypeptide or a fragment, variant, or derivative thereof is expressed in the human in a therapeutically or prophylactically effective amount.
  • an “immune response” refers to the ability of a vertebrate to elicit an immune reaction to a composition delivered to that vertebrate.
  • immune responses include an antibody response or a cellular, e.g., cytotoxic T-cell, response.
  • One or more compositions of the present invention may be used to prevent influenza infection in vertebrates, e.g., as a prophylactic vaccine, to establish or enhance immunity to IV in a healthy individual prior to exposure to influenza or contraction of influenza disease, thus preventing the disease or reducing the severity of disease symptoms.
  • compositions of the present invention can be used both to prevent IV infection, and also to therapeutically treat IV infection.
  • the present invention is used to further stimulate the immune system of the vertebrate, thus reducing or eliminating the symptoms associated with that disease or disorder.
  • treatment refers to the use of one or more compositions of the present invention to prevent, cure, retard, or reduce the severity of influenza disease symptoms in a vertebrate, and/or result in no worsening of influenza disease over a specified period of time in a vertebrate which has already been exposed to IV and is thus in need of therapy.
  • prevention refers to the use of one or more compositions of the present invention to generate immunity in a vertebrate which has not yet been exposed to a particular strain of IV, thereby preventing or reducing disease symptoms if the vertebrate is later exposed to the particular strain of IV.
  • the methods of the present invention therefore may be referred to as therapeutic vaccination or preventative or prophylactic vaccination. It is not required that any composition of the present invention provide total immunity to influenza or totally cure or eliminate all influenza disease symptoms.
  • a “vertebrate in need of therapeutic and/or preventative immunity” refers to an individual for whom it is desirable to treat, i.e., to prevent, cure, retard, or reduce the severity of influenza disease symptoms, and/or result in no worsening of influenza disease over a specified period of time.
  • Vertebrates to treat and/or vaccinate include humans, apes, monkeys (e.g., owl, squirrel, cebus, rhesus, African green, patas, cynomolgus, and cercopithecus), orangutans, baboons, gibbons, and chimpanzees, dogs, wolves, cats, lions, and tigers, horses, donkeys, zebras, cows, pigs, sheep, deer, giraffes, bears, rabbits, mice, ferrets, seals, whales, ducks, geese, terns, shearwaters, gulls, turkeys, chickens, quail, pheasants, geese, starlings and budgerigars.
  • monkeys e.g., owl, squirrel, cebus, rhesus, African green, patas, cynomolgus, and cercopi
  • compositions of the present invention are utilized in a “prime boost” regimen.
  • An example of a “prime boost” regimen may be found in Yang, Z. et al. J. Virol. 77:799-803 (2002), which is incorporated herein by reference in its entirety.
  • one or more polynucleotide vaccine compositions of the present invention are delivered to a vertebrate, thereby priming the immune response of the vertebrate to an IV, and then a second immunogenic composition is utilized as a boost vaccination.
  • compositions of the present invention are used to prime immunity, and then a second immunogenic composition, e.g., a recombinant viral vaccine or vaccines, a different polynucleotide vaccine, or one or more purified subunit isolated IV polypeptides or fragments, variants or derivatives thereof is used to boost the anti-IV immune response.
  • a second immunogenic composition e.g., a recombinant viral vaccine or vaccines, a different polynucleotide vaccine, or one or more purified subunit isolated IV polypeptides or fragments, variants or derivatives thereof is used to boost the anti-IV immune response.
  • a priming composition and a boosting composition are combined in a single composition or single formulation.
  • a single composition may comprise an isolated IV polypeptide or a fragment, variant, or derivative thereof as the priming component and a polynucleotide encoding an influenza protein as the boosting component.
  • the compositions may be contained in a single vial where the priming component and boosting component are mixed together.
  • the polynucleotide component may provide a boost to the isolated protein component.
  • compositions comprising both a priming component and a boosting component are referred to herein as “combinatorial vaccine compositions” or “single formulation heterologous prime-boost vaccine compositions.”
  • the priming composition may be administered before the boosting composition, or even after the boosting composition, if the boosting composition is expected to take longer to act.
  • the priming composition may be administered simultaneously with the boosting composition, but in separate formulations where the priming component and the boosting component are separated.
  • priming or “primary” and “boost” or “boosting” as used herein may refer to the initial and subsequent immunizations, respectively, i.e., in accordance with the definitions these terms normally have in immunology. However, in certain embodiments, e.g., where the priming component and boosting component are in a single formulation, initial and subsequent immunizations may not be necessary as both the “prime” and the “boost” compositions are administered simultaneously.
  • compositions of the present invention are delivered to a vertebrate by methods described herein, thereby achieving an effective therapeutic and/or an effective preventative immune response. More specifically, the compositions of the present invention may be administered to any tissue of a vertebrate, including, but not limited to, muscle, skin, brain tissue, lung tissue, liver tissue, spleen tissue, bone marrow tissue, thymus tissue, heart tissue, e.g., myocardium, endocardium, and pericardium, lymph tissue, blood tissue, bone tissue, pancreas tissue, kidney tissue, gall bladder tissue, stomach tissue, intestinal tissue, testicular tissue, ovarian tissue, uterine tissue, vaginal tissue, rectal tissue, nervous system tissue, eye tissue, glandular tissue, tongue tissue, and connective tissue, e.g., cartilage.
  • tissue of a vertebrate including, but not limited to, muscle, skin, brain tissue, lung tissue, liver tissue, spleen tissue, bone marrow tissue, thymus tissue, heart tissue,
  • compositions of the present invention may be administered to any internal cavity of a vertebrate, including, but not limited to, the lungs, the mouth, the nasal cavity, the stomach, the peritoneal cavity, the intestine, any heart chamber, veins, arteries, capillaries, lymphatic cavities, the uterine cavity, the vaginal cavity, the rectal cavity, joint cavities, ventricles in brain, spinal canal in spinal cord, the ocular cavities, the lumen of a duct of a salivary gland or a liver.
  • a vertebrate including, but not limited to, the lungs, the mouth, the nasal cavity, the stomach, the peritoneal cavity, the intestine, any heart chamber, veins, arteries, capillaries, lymphatic cavities, the uterine cavity, the vaginal cavity, the rectal cavity, joint cavities, ventricles in brain, spinal canal in spinal cord, the ocular cavities, the lumen of a duct of a salivary gland or a liver.
  • compositions of the present invention When the compositions of the present invention is administered to the lumen of a duct of a salivary gland or liver, the desired polypeptide is expressed in the salivary gland and the liver such that the polypeptide is delivered into the blood stream of the vertebrate from each of the salivary gland or the liver.
  • Certain modes for administration to secretory organs of a gastrointestinal system using the salivary gland, liver and pancreas to release a desired polypeptide into the bloodstream is disclosed in U.S. Pat. Nos. 5,837,693 and 6,004,944, both of which are incorporated herein by reference in their entireties.
  • compositions are administered into embryonated chicken eggs or by intra-muscular injection into the defeathered breast area of chicks as described in Kodihalli S. et al., Vaccine 18:2592-9 (2000), which is incorporated herein by reference in its entirety.
  • compositions are administered to muscle, either skeletal muscle or cardiac muscle, or to lung tissue.
  • lung tissue Specific, but non-limiting modes for administration to lung tissue are disclosed in Wheeler, C. J., et al., Proc. Natl. Acad. Sci. USA 93:11454-11459 (1996), which is incorporated herein by reference in its entirety.
  • compositions of the present invention can be administered by intramuscular (i.m.), subcutaneous (s.c.), or intrapulmonary routes.
  • suitable routes of administration include, but are not limited to intratracheal, transdermal, intraocular, intranasal, inhalation, intracavity, intravenous (i.v.), intraductal (e.g., into the pancreas) and intraparenchymal (i.e., into any tissue) administration.
  • Transdermal delivery includes, but not limited to intradermal (e.g., into the dermis or epidermis), transdermal (e.g., percutaneous) and transmucosal administration (i.e., into or through skin or mucosal tissue).
  • Intracavity administration includes, but not limited to administration into oral, vaginal, rectal, nasal, peritoneal, or intestinal cavities as well as, intrathecal (i.e., into spinal canal), intraventricular (i.e., into the brain ventricles or the heart ventricles), inraatrial (i.e., into the heart atrium) and sub arachnoid (i.e., into the sub arachnoid spaces of the brain) administration.
  • intrathecal i.e., into spinal canal
  • intraventricular i.e., into the brain ventricles or the heart ventricles
  • inraatrial i.e., into the heart atrium
  • sub arachnoid i.e., into the sub arachnoid spaces of the brain
  • Administration means of the present invention include needle injection, catheter infusion, biolistic injectors, particle accelerators (e.g., “gene guns” or pneumatic “needleless” injectors) Med-E-Jet (Vahlsing, H., et al., J. Immunol.
  • gelfoam sponge depots other commercially available depot materials (e.g., hydrogels), osmotic pumps (e.g., Alza minipumps), oral or suppositorial solid (tablet or pill) pharmaceutical formulations, topical skin creams, and decanting, use of polynucleotide coated suture (Qin, Y., et al., Life Sciences 65: 2193-2203 (1999)) or topical applications during surgery.
  • Certain modes of administration are intramuscular needle-based injection and pulmonary application via catheter infusion.
  • Energy-assisted plasmid delivery (EAPD) methods may also be employed to administer the compositions of the invention.
  • Determining an effective amount of one or more compositions of the present invention depends upon a number of factors including, for example, the antigen being expressed or administered directly, e.g., HA, NA, NP, M1 or M2, or fragments, e.g., eM2, variants, or derivatives thereof, the age and weight of the subject, the precise condition requiring treatment and its severity, and the route of administration. Based on the above factors, determining the precise amount, number of doses, and timing of doses are within the ordinary skill in the art and will be readily determined by the attending physician or veterinarian.
  • compositions of the present invention may include various salts, excipients, delivery vehicles and/or auxiliary agents as are disclosed, e.g., in U.S. patent application Publication No. 2002/0019358, published Feb. 14, 2002, which is incorporated herein by reference in its entirety.
  • compositions of the present invention may include one or more transfection facilitating compounds that facilitate delivery of polynucleotides to the interior of a cell, and/or to a desired location within a cell.
  • transfection facilitating compound As used herein, the terms “transfection facilitating compound,” “transfection facilitating agent,” and “transfection facilitating material” are synonymous, and may be used interchangeably. It should be noted that certain transfection facilitating compounds may also be “adjuvants” as described infra, i.e., in addition to facilitating delivery of polynucleotides to the interior of a cell, the compound acts to alter or increase the immune response to the antigen encoded by that polynucleotide.
  • transfection facilitating compounds include, but are not limited to inorganic materials such as calcium phosphate, alum (aluminum sulfate), and gold particles (e.g., “powder” type delivery vehicles); peptides that are, for example, cationic, intercell targeting (for selective delivery to certain cell types), intracell targeting (for nuclear localization or endosomal escape), and ampipathic (helix forming or pore forming); proteins that are, for example, basic (e.g., positively charged) such as histones, targeting (e.g., asialoprotein), viral (e.g., Sendai virus coat protein), and pore-forming; lipids that are, for example, cationic (e.g., DMRIE, DOSPA, DC-Chol), basic (e.g., steryl amine), neutral (e.g., cholesterol), anionic (e.g., phosphatidyl serine), and zwitterionic (e.g., DOPE,
  • a transfection facilitating material can be used alone or in combination with one or more other transfection facilitating materials.
  • Two or more transfection facilitating materials can be combined by chemical bonding (e.g., covalent and ionic such as in lipidated polylysine, PEGylated polylysine) (Toncheva, et al., Biochim. Biophys. Acta 1380(3):354-368 (1988)), mechanical mixing (e.g., free moving materials in liquid or solid phase such as “polylysine+cationic lipids”) (Gao and Huang, Biochemistry 35:1027-1036 (1996); Trubetskoy, et al., Biochem. Biophys.
  • cationic lipids are 5-carboxyspermylglycine dioctadecylamide (DOGS) and dipalmitoyl-phophatidylethanolamine-5-carboxyspermylamide (DPPES).
  • DOGS 5-carboxyspermylglycine dioctadecylamide
  • DPES dipalmitoyl-phophatidylethanolamine-5-carboxyspermylamide
  • Cationic cholesterol derivatives are also useful, including ⁇ 3 ⁇ -[N-N′,N′-dimethylamino)ethane]-carbomoyl ⁇ -cholesterol (DC-Chol).
  • Dimethyldioctdecyl-ammonium bromide (DDAB), N-(3-aminopropyl)-N,N-(bis-(2-tetradecyloxyethyl))-N-methyl-ammonium bromide (PA-DEMO), N-(3-aminopropyl)-N,N-(bis-(2-dodecyloxyethyl))-N-methyl-ammonium bromide (PA-DELO), N,N,N-tris-(2-dodecyloxy)ethyl-N-(3-amino)propyl-ammonium bromide (PA-TELO), and N1-(3-aminopropyl)((2-dodecyloxy)ethyl)-N2-(2-dodecyloxy)ethyl-1-piperazinaminium bromide (GA-LOE-BP) can also be employed in the present invention.
  • Non-diether cationic lipids such as DL-1,2-dioleoyl-3-dimethylaminopropyl- ⁇ -hydroxyethylammonium (DORI diester), 1-O-oleyl-2-oleoyl-3-dimethylaminopropyl-p-hydroxyethylammonium (DORI ester/ether), and their salts promote in vivo gene delivery.
  • cationic lipids comprise groups attached via a heteroatom attached to the quaternary ammonium moiety in the head group.
  • a glycyl spacer can connect the linker to the hydroxyl group.
  • DMRIE (( ⁇ )-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide)
  • GAP-DMORIE (( ⁇ )-N
  • the cationic surfactant is Pr-DOctRIE-OAc.
  • cationic lipids include ( ⁇ )-N,N-dimethyl-N-[2-(sperminecarboxamido)ethyl]-2,3-bis(dioleyloxy)-1-propaniminium pentahydrochloride (DOSPA), ( ⁇ )-N-(2-aminoethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propaniminium bromide ( ⁇ -aminoethyl-DMRIE or ⁇ AE-DMRIE) (Wheeler, et al., Biochim. Biophys.
  • DOSPA dioleyloxy-1-propaniminium pentahydrochloride
  • ⁇ -N-(2-aminoethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propaniminium bromide ⁇ -aminoethyl-DMRIE or ⁇ AE-DMRIE
  • DMRIE-derived cationic lipids that are useful for the present invention are ( ⁇ )-N-(3-aminopropyl)-N,N-dimethyl-2,3-(bis-decyloxy)-1-propanaminium bromide (GAP-DDRIE), ( ⁇ )-N-(3-aminopropyl)-N,N-dimethyl-2,3-(bis-tetradecyloxy)-1-propanaminium bromide (GAP-DMRIE), ( ⁇ )-N-((N′′-methyl)-N′-ureyl)propyl-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (GMU-DMRIE), ( ⁇ )-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(dodecyloxy)-1-propanaminium bromide (DLRIE), and ( ⁇ )-N-(2-hydroxy)-
  • the cationic lipid may be mixed with one or more co-lipids.
  • co-lipid refers to any hydrophobic material which may be combined with the cationic lipid component and includes amphipathic lipids, such as phospholipids, and neutral lipids, such as cholesterol.
  • amphipathic lipids such as phospholipids
  • neutral lipids such as cholesterol.
  • Cationic lipids and co-lipids may be mixed or combined in a number of ways to produce a variety of non-covalently bonded macroscopic structures, including, for example, liposomes, multilamellar vesicles, unilamellar vesicles, micelles, and simple films.
  • co-lipids are the zwitterionic phospholipids, which include the phosphatidylethanolamines and the phosphatidylcholines.
  • phosphatidylethanolamines include DOPE, DMPE and DPyPE.
  • the co-lipid is DPyPE, which comprises two phytanoyl substituents incorporated into the diacylphosphatidylethanolamine skeleton.
  • the co-lipid is DOPE, CAS name 1,2-diolyeoyl-sn-glycero-3-phosphoethanolamine.
  • the cationic lipid:co-lipid molar ratio may be from about 9:1 to about 1:9, from about 4:1 to about 1:4, from about 2:1 to about 1:2, or about 1:1.
  • the cationic lipid and co-lipid components may be dissolved in a solvent such as chloroform, followed by evaporation of the cationic lipid/co-lipid solution under vacuum to dryness as a film on the inner surface of a glass vessel (e.g., a Rotovap round-bottomed flask).
  • a glass vessel e.g., a Rotovap round-bottomed flask
  • the amphipathic lipid component molecules self-assemble into homogenous lipid vesicles.
  • These lipid vesicles may subsequently be processed to have a selected mean diameter of uniform size prior to complexing with, for example, a codon-optimized polynucleotide of the present invention, according to methods known to those skilled in the art.
  • compositions include a cationic lipid
  • polynucleotides of the present invention are complexed with lipids by mixing, for example, a plasmid in aqueous solution and a solution of cationic lipid:co-lipid as prepared herein are mixed.
  • concentration of each of the constituent solutions can be adjusted prior to mixing such that the desired final plasmid/cationic lipid:co-lipid ratio and the desired plasmid final concentration will be obtained upon mixing the two solutions.
  • the cationic lipid:co-lipid mixtures are suitably prepared by hydrating a thin film of the mixed lipid materials in an appropriate volume of aqueous solvent by vortex mixing at ambient temperatures for about 1 minute.
  • the thin films are prepared by admixing chloroform solutions of the individual components to afford a desired molar solute ratio followed by aliquoting the desired volume of the solutions into a suitable container.
  • the solvent is removed by evaporation, first with a stream of dry, inert gas (e.g. argon) followed by high vacuum treatment.
  • inert gas e.g. argon
  • hydrophobic and amphiphilic additives such as, for example, sterols, fatty acids, gangliosides, glycolipids, lipopeptides, liposaccharides, neobees, niosomes, prostaglandins and sphingolipids, may also be included in compositions of the present invention. In such compositions, these additives may be included in an amount between about 0.1 mol % and about 99.9 mol % (relative to total lipid), about 1-50 mol %, or about 2-25 mol %.
  • compositions comprising an auxiliary agent which is administered before, after, or concurrently with the polynucleotide.
  • an “auxiliary agent” is a substance included in a composition for its ability to enhance, relative to a composition which is identical except for the inclusion of the auxiliary agent, the entry of polynucleotides into vertebrate cells in vivo, and/or the in vivo expression of polypeptides encoded by such polynucleotides.
  • Certain auxiliary agents may, in addition to enhancing entry of polynucleotides into cells, enhance an immune response to an immunogen encoded by the polynucleotide.
  • Auxiliary agents of the present invention include nonionic, anionic, cationic, or zwitterionic surfactants or detergents, with nonionic surfactants or detergents being preferred, chelators, DNase inhibitors, poloxamers, agents that aggregate or condense nucleic acids, emulsifying or solubilizing agents, wetting agents, gel-forming agents, and buffers.
  • Auxiliary agents for use in compositions of the present invention include, but are not limited to non-ionic detergents and surfactants IGEPAL CA 6300, NONIDET NP-40, Nonidet® P40, Tween-20TM, Tween-80TM, Pluronic® F68 (ave. MW: 8400; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 80%), Pluronic F77® (ave. MW: 6600; approx. MW of hydrophobe, 2100; approx. wt. % of hydrophile, 70%), Pluronic P65®(ave. MW: 3400; approx. MW of hydrophobe, 1800; approx. wt.
  • the auxiliary agent is DMSO, Nonidet P40, Pluronic F68® (ave. MW: 8400; approx. MW of hydrophobe, 1800; approx. wt.
  • Pluronic F77® (ave. MW: 6600; approx. MW of hydrophobe, 2100; approx. wt. % of hydrophile, 70%), Pluronic P65® (ave. MW: 3400; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 50%), Pluronic L64® (ave. MW: 2900; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 40%), and Pluronic F108® (ave. MW: 14600; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 80%). See, e.g., U.S. patent application Publication No. 2002/0019358, published Feb. 14, 2002, which is incorporated herein by reference in its entirety.
  • compositions of the present invention can further include one or more adjuvants before, after, or concurrently with the polynucleotide.
  • adjuvant refers to any material having the ability to (1) alter or increase the immune response to a particular antigen or (2) increase or aid an effect of a pharmacological agent. It should be noted, with respect to polynucleotide vaccines, that an “adjuvant,” can be a transfection facilitating material. Similarly, certain “transfection facilitating materials” described supra, may also be an “adjuvant.” An adjuvant may be used with a composition comprising a polynucleotide of the present invention.
  • an adjuvant may be used with either the priming immunization, the booster immunization, or both.
  • Suitable adjuvants include, but are not limited to, cytokines and growth factors; bacterial components (e.g., endotoxins, in particular superantigens, exotoxins and cell wall components); aluminum-based salts; calcium-based salts; silica; polynucleotides; toxoids; serum proteins, viruses and virally-derived materials, poisons, venoms, imidazoquiniline compounds, poloxamers, and cationic lipids.
  • cytokines and growth factors include, but are not limited to, cytokines and growth factors; bacterial components (e.g., endotoxins, in particular superantigens, exotoxins and cell wall components); aluminum-based salts; calcium-based salts; silica; polynucleotides; toxoids; serum proteins, viruses and virally-derived materials, poisons, venoms, imidazoquini
  • Any compound which may increase the expression, antigenicity or immunogenicity of the polypeptide is a potential adjuvant.
  • the present invention provides an assay to screen for improved immune responses to potential adjuvants.
  • adjuvants which may be screened for their ability to enhance the immune response according to the present invention include, but are not limited to: inert carriers, such as alum, bentonite, latex, and acrylic particles; pluronic block polymers, such as TiterMax® (block copolymer CRL-8941, squalene (a metabolizable oil) and a microparticulate silica stabilizer); depot formers, such as Freunds adjuvant, surface active materials, such as saponin, lysolecithin, retinal, Quil A, liposomes, and pluronic polymer formulations; macrophage stimulators, such as bacterial lipopolysaccharide; alternate pathway complement activators, such as insulin, zymosan, endotoxin, and levamisole; and non-ionic surfactants, such as poloxamers, poly(oxyethylene)-poly(oxypropylene) tri-block copolymers. Also included as adjuvants are transfection-facilitating materials, such
  • Poloxamers which may be screened for their ability to enhance the immune response according to the present invention include, but are not limited to, commercially available poloxamers such as Pluronic® surfactants, which are block copolymers of propylene oxide and ethylene oxide in which the propylene oxide block is sandwiched between two ethylene oxide blocks.
  • Pluronic® surfactants include Pluronic® L121 (ave. MW: 4400; approx. MW of hydrophobe, 3600; approx. wt. % of hydrophile, 10%), Pluronic® L101 (ave. MW: 3800; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 10%), Pluronic® L81 (ave.
  • Pluronic® L43 (ave. MW: 1850; approx. MW of hydrophobe, 1200; approx. wt. % of hydrophile, 30%), Pluronic® L64 (ave. MW: 2900; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 40%), Pluronic® L44 (ave. MW: 2200; approx. MW of hydrophobe, 1200; approx. wt. % of hydrophile, 40%), Pluronic® L35 (ave. MW: 1900; approx. MW of hydrophobe, 900; approx. wt.
  • Pluronic® P123 (ave. MW: 5750; approx. MW of hydrophobe, 3600; approx. wt. % of hydrophile, 30%)
  • Pluronic® P103 (ave. MW: 4950; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 30%)
  • Pluronic® P104 (ave. MW: 5900; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 40%)
  • Pluronic® P84 (ave. MW: 4200; approx. MW of hydrophobe, 2400; approx. wt.
  • Pluronic® P105 (ave. MW: 6500; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 50%)
  • Pluronic® P85 (ave. MW: 4600; approx. MW of hydrophobe, 2400; approx. wt. % of hydrophile, 50%)
  • Pluronic® P75 (ave. MW: 4150; approx. MW of hydrophobe, 2100; approx. wt. % of hydrophile, 50%)
  • Pluronic® P65 (ave. MW: 3400; approx. MW of hydrophobe, 1800; approx. wt.
  • Pluronic® F127 (ave. MW: 12600; approx. MW of hydrophobe, 3600; approx. wt. % of hydrophile, 70%)
  • Pluronic® F98 (ave. MW: 13000; approx. MW of hydrophobe, 2700; approx. wt. % of hydrophile, 80%)
  • Pluronic® F87 (ave. MW: 7700; approx. MW of hydrophobe, 2400; approx. wt. % of hydrophile, 70%)
  • Pluronic® F77 (ave. MW: 6600; approx. MW of hydrophobe, 2100; approx. wt.
  • Reverse poloxamers which may be screened for their ability to enhance the immune response according to the present invention include, but are not limited to Pluronic® R 31R1 (ave. MW: 3250; approx. MW of hydrophobe, 3100; approx. wt. % of hydrophile, 10%), Pluronic® R 25R1 (ave. MW: 2700; approx. MW of hydrophobe, 2500; approx. wt. % of hydrophile, 10%), Pluronic® R 17R1 (ave. MW: 1900; approx. MW of hydrophobe, 1700; approx. wt. % of hydrophile, 10%), Pluronic® R 31R2 (ave. MW: 3300; approx.
  • MW: 4600 in which L indicates that the surfactants are liquids, P that they are pastes, the first digit is a measure of the molecular weight of the polypropylene portion of the surfactant and the last digit of the number, multiplied by 10, gives the percent ethylene oxide content of the surfactant; and compounds that are nonylphenyl polyethylene glycol such as Synperonic® NP10 (nonylphenol ethoxylated surfactant—10% solution), Synperonic® NP30 (condensate of 1 mole of nonylphenol with 30 moles of ethylene oxide) and Synperonic® NP5 (condensate of 1 mole of nonylphenol with 5.5 moles of naphthalene oxide).
  • Synperonic® NP10 nonylphenol ethoxylated surfactant—10% solution
  • Synperonic® NP30 condensate of 1 mole of nonylphenol with 30 moles of ethylene oxide
  • Synperonic® NP5 conden
  • poloxamers of interest include CRL1005 (12 kDa, 5% POE), CRL8300 (11 kDa, 5% POE), CRL2690 (12 kDa, 10% POE), CRL4505 (15 kDa, 5% POE) and CRL1415 (9 kDa, 10% POE).
  • the adjuvant is a cytokine.
  • a composition of the present invention can comprise one or more cytokines, chemokines, or compounds that induce the production of cytokines and chemokines, or a polynucleotide encoding one or more cytokines, chemokines, or compounds that induce the production of cytokines and chemokines.
  • Examples include, but are not limited to granulocyte macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), macrophage colony stimulating factor (M-CSF), colony stimulating factor (CSF), erythropoietin (EPO), interleukin 2 (IL-2), interleukin-3 (IL-3), interleukin 4 (IL-4), interleukin 5 (IL-5), interleukin 6 (IL-6), interleukin 7 (IL-7), interleukin 8 (IL-8), interleukin 10 (IL-10), interleukin 12 (IL-12), interleukin 15 (IL-15), interleukin 18 (IL-18), interferon alpha (IFN ⁇ ), interferon beta (IFN ⁇ ), interferon gamma (IFN ⁇ ), interferon omega (IFN ⁇ ), interferon tau (IFN ⁇ ), interferon gamma inducing factor I (IGIF), transforming growth factor beta (TGF- ⁇ ), RANTES (regulated upon
  • the polynucleotide construct may be complexed with an adjuvant composition comprising ( ⁇ )-N-(3-aminopropyl)-N,N-dimethyl-2,3-bis(syn-9-tetradeceneyloxy)-1-propanaminium bromide (GAP-DMORIE).
  • the composition may also comprise one or more co-lipids, e.g., 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE), and/or 1,2-dimyristoyl-glycer-3-phosphoethanolamine (DMPE).
  • DOPE 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine
  • DPyPE 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine
  • DMPE 1,2-dimyristoyl-glycer
  • VaxfectinTM An adjuvant composition comprising GAP-DMORIE and DPyPE at a 1:1 molar ratio is referred to herein as VaxfectinTM. See, e.g., PCT Publication No. WO 00/57917, which is incorporated herein by reference in its entirety.
  • the polynucleotide itself may function as an adjuvant as is the case when the polynucleotides of the invention are derived, in whole or in part, from bacterial DNA.
  • Bacterial DNA containing motifs of unmethylated CpG-dinucleotides (CpG-DNA) triggers innate immune cells in vertebrates through a pattern recognition receptor (including toll receptors such as TLR 9) and thus possesses potent immunostimulatory effects on macrophages, dendritic cells and B-lymphocytes. See, e.g., Wagner, H., Curr. Opin. Microbiol. 5:62-69 (2002); Jung, J. et al., J. Immunol.
  • an adjuvant to increase the immune response to an antigen is typically manifested by a significant increase in immune-mediated protection.
  • an increase in humoral immunity is typically manifested by a significant increase in the titer of antibodies raised to the antigen
  • an increase in T-cell activity is typically manifested in increased cell proliferation, or cellular cytotoxicity, or cytokine secretion.
  • An adjuvant may also alter an immune response, for example, by changing a primarily humoral or Th 2 response into a primarily cellular, or Th 1 response.
  • Nucleic acid molecules and/or polynucleotides of the present invention may be solubilized in any of various buffers.
  • Suitable buffers include, for example, phosphate buffered saline (PBS), normal saline, Tris buffer, and sodium phosphate (e.g., 150 mM sodium phosphate).
  • PBS phosphate buffered saline
  • Tris buffer Tris buffer
  • sodium phosphate e.g. 150 mM sodium phosphate
  • Insoluble polynucleotides may be solubilized in a weak acid or weak base, and then diluted to the desired volume with a buffer. The pH of the buffer may be adjusted as appropriate.
  • a pharmaceutically acceptable additive can be used to provide an appropriate osmolarity.
  • Such additives are within the purview of one skilled in the art.
  • aqueous compositions used in vivo sterile pyrogen-free water can be used.
  • Such formulations will contain an effective amount of a polynucleotide together with a suitable amount of an aqueous solution in order to prepare pharmaceutically acceptable compositions suitable for administration to a human.
  • compositions of the present invention can be formulated according to known methods. Suitable preparation methods are described, for example, in Remington's Pharmaceutical Sciences, 16th Edition, A. Osol, ed., Mack Publishing Co., Easton, Pa. (1980), and Remington's Pharmaceutical Sciences, 19th Edition, A. R. Gennaro, ed., Mack Publishing Co., Easton, Pa. (1995), both of which are incorporated herein by reference in their entireties.
  • the composition may be administered as an aqueous solution, it can also be formulated as an emulsion, gel, solution, suspension, lyophilized form, or any other form known in the art.
  • the composition may contain pharmaceutically acceptable additives including, for example, diluents, binders, stabilizers, and preservatives.
  • Constructs of the present invention are constructed based on the sequence information provided herein or in the art utilizing standard molecular biology techniques, including, but not limited to the following.
  • the single-stranded ends of each pair of oligonucleotides are designed to anneal with a single-stranded end of an adjacent oligonucleotide duplex.
  • oligonucleotide pairs prepared in this manner are allowed to anneal, and approximately five to six adjacent oligonucleotide duplex fragments are then allowed to anneal together via the cohesive single stranded ends.
  • This series of annealed oligonucleotide duplex fragments is then ligated together and cloned into a suitable plasmid, such as the TOPO® vector available from Invitrogen Corporation, Carlsbad, Calif. The construct is then sequenced by standard methods.
  • Constructs prepared in this manner comprising 5 to 6 adjacent 80 to 90 base pair fragments ligated together, i.e., fragments of about 500 base pairs, are prepared, such that the entire desired sequence of the construct is represented in a series of plasmid constructs.
  • the inserts of these plasmids are then cut with appropriate restriction enzymes and ligated together to form the final construct.
  • the final construct is then cloned into a standard bacterial cloning vector, and sequenced.
  • oligonucleotides and primers referred to herein can easily be designed by a person of skill in the art based on the sequence information provided herein and in the art, and such can be synthesized by any of a number of commercial nucleotide providers, for example Retrogen, San Diego, Calif., and GENEART, Regensburg, Germany.
  • Constructs of the present invention can be inserted, for example, into eukaryotic expression vectors VR1012 or VR10551. These vectors are built on a modified pUC18 background (see Yanisch-Perron, C., et al. Gene 33:103-119 (1985)), and contain a kanamycin resistance gene, the human cytomegalovirus immediate early promoter/enhancer and intron A, and the bovine growth hormone transcription termination signal, and a polylinker for inserting foreign genes. See Hartikka, J., et al., Hum. Gene Ther. 7:1205-1217 (1996).
  • eukaryotic expression vectors may be used in the present invention, including, but not limited to: plasmids pcDNA3, pHCMV/Zeo, pCR3.1, pEF1/His, pIND/GS, pRc/HCMV2, pSV40/Zeo2, pTRACER-HCMV, pUB6/V5-His, pVAX1, and pZeoSV2 (available from Invitrogen, San Diego, Calif.), and plasmid pCI (available from Promega, Madison, Wis.).
  • VR10551 An optimized backbone plasmid, termed VR10551, has minor changes from the VR1012 backbone described above.
  • the VR10551 vector is derived from and similar to VR1012 in that it uses the human cytomegalovirus immediate early (hCMV-IE) gene enhancer/promoter and 5′ untranslated region (UTR), including the hCMV-IE Intron A.
  • the changes from the VR1012 to the VR10551 include some modifications to the multiple cloning site, and a modified rabbit ⁇ globin 3′ untranslated region/polyadenylation signal sequence/transcriptional terminator has been substituted for the same functional domain derived from the bovine growth hormone gene.
  • constructs of the present invention can be inserted into other eukaryotic expression vector backbones such as VR10682 or VR10686.
  • the VR10682 expression vector backbone contains a modified rous sarcoma virus (RSV) promoter from expression plasmid VCL1005, the bovine growth hormone (BGH) poly-adenylation site and a polylinker for inserting foreign genes and a kanamycin resistance gene.
  • the RSV promoter in VCL1005 and VR10682 contains a XbaI endonuclease restriction site near the transcription start site in the sequence TAC TCT AGA CG (SEQ ID NO:82).
  • the modified RSV promoter contained in VR10682.
  • Expression plasmid VCL1005 is described in U.S. Pat. No. 5,561,064 and is incorporated herein by reference.
  • the VR10686 expression vector backbone (SEQ ID NO:112) was created by replacing the West Nile Virus (WNV) antigen insert in VR6430 (SEQ ID NO:89) with the multiple cloning site from the VR1012 vector.
  • the VR10686 and VR6430 expression vector backbones contain the RSV promoter, derived from VCL1005, which has been modified back to the wild-type RSV sequence (TAC AAT AAA CG (SEQ ID NO:83)).
  • the wild-type RSV promoter is fused to the “R” region plus the first 39 nucleotides of the U5 region from Human T-Cell Leukemia Virus I (HTLV-I), hereinafter refered to as the RU5 element.
  • the R and U5 regions are portions of the long terminal repeat region (LTR) of HTLV-I which control expression of the HTLV-I transcript and is duplicated at either end of the integrated viral genome as a result of the retroviral integration mechanism.
  • LTR long terminal repeat region
  • the LTR of HTLV-1 and most retroviruses are divided into three regions, U3, R and U5. Transcription from the intigrated viral genome commences at the U3-R boundary of the 5′ LTR and the transcript is polyadenylated at the R-U5 boundary of the 3′ LTR.
  • This RU5 HTLV-I element has been shown to be a potent stimulator of translation when fused to the SV40 early gene promoter. See Takebe et al., Mol. Cell Biol. 8:466-472 (1988). It has been proposed that the stimulation of translation by the HTLV-I RU5 element is due to its function, in part, as a translational enhancing internal ribosome entry site (IRES). See Attal et al. FEBS Letters 392:220-224 (1996). Additionally the HTLV-I RU5 element provides the 5′-splice donor site. Immediately downstream of the RU5 element is the 3′-end of the HCMV intron A sequence containing the splice acceptor sequence.
  • the VR10686 and VR6430 expression vectors contain a hybrid intron composed of the 5′-HTLV I intron sequence fused to the 3′-end of the HCMV intron A, a bovine growth hormone poly-adenylation site, a polylinker for insertion of forign genes and a kanamycin resistance gene.
  • the VR6430 vector expresses the prM and E West Nile Virus antigens (Genebank Accession No. AF202541).
  • the vector backbones described above may by used to create expression vectors which express multiple influenza proteins, fragments, variants or derivatives thereof.
  • An expression vector as desribed herein may contain an additional promoter.
  • construct VR4774 (described in Example 13), contains a CMV promoter and an RSV promoter.
  • the vector backbones described herein may contain multiple expression cassettes which comprise a promoter and an influenza coding sequence including, inter alia, polynucleotides as described herein.
  • the expression cassettes may encode the same or different influenza polypeptides.
  • the expression cassettes may be in the same or opposite orientation relative to each other. As such transcription from each cassette may be in the same or opposition direction (i.e. 5′ to 3′ in both expression cassettes or, alternatively, 5′ to 3′ in one expression cassette and 3′ to 5′ in the other expression cassette).
  • Plasmid DNA may be transformed into competent cells of an appropriate Escherichia coli strain (including but not limited to the DH5 ⁇ strain) and highly purified covalently closed circular plasmid DNA was isolated by a modified lysis procedure (Horn, N. A., et al., Hum. Gene Ther. 6:565-573 (1995)) followed by standard double CsCl-ethidium bromide gradient ultracentrifugation (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y. (1989)).
  • plasmid DNAs are purified using Giga columns from Qiagen (Valencia, Calif.) according to the kit instructions. All plasmid preparations were free of detectable chromosomal DNA, RNA and protein impurities based on gel analysis and the bicinchoninic protein assay (Pierce Chem. Co., Rockford Ill.). Endotoxin levels were measured using Limulus Amebocyte Lysate assay (LAL, Associates of Cape Cod, Falmouth, Mass.) and were less than 0.6 Endotoxin Units/mg of plasmid DNA. The spectrophotometric A 260 /A 280 ratios of the DNA solutions were typically above 1.8.
  • Plasmids were ethanol precipitated and resuspended in an appropriate solution, e.g., 150 mM sodium phosphate (for other appropriate excipients and auxiliary agents, see U.S. patent application Publication 2002/0019358, published Feb. 14, 2002). DNA was stored at ⁇ 20EC until use. DNA was diluted by mixing it with 300 mM salt solutions and by adding appropriate amount of USP water to obtain 1 mg/ml plasmid DNA in the desired salt at the desired molar concentration.
  • an appropriate solution e.g. 150 mM sodium phosphate (for other appropriate excipients and auxiliary agents, see U.S. patent application Publication 2002/0019358, published Feb. 14, 2002).
  • DNA was stored at ⁇ 20EC until use. DNA was diluted by mixing it with 300 mM salt solutions and by adding appropriate amount of USP water to obtain 1 mg/ml plasmid DNA in the desired salt at the desired molar concentration.
  • VM-92 mouse melanoma cell line
  • VM-92 also known as UM-449.
  • MRC-5 cells ATCC Accession No. CCL-171 or human rhabdomyosarcoma cell line RD (ATCC CCL-136).
  • the transfection was performed using cationic lipid-based transfection procedures well known to those of skill in the art.
  • Other transfection procedures are well known in the art and may be used, for example electroporation and calcium chloride-mediated transfection (Graham F. L. and A. J. van der Eb Virology 52:456-67 (1973)).
  • cell lysates and culture supernatants of transfected cells were evaluated to compare relative levels of expression of IV antigen proteins.
  • the samples were assayed by western blots and ELISAs, using commercially available polyclonal and/or monoclonal antibodies (available, e.g., from Research Diagnostics Inc., Flanders N.J.), so as to compare both the quality and the quantity of expressed antigen.
  • the quadriceps muscles of restrained awake mice are injected bilaterally with 1-50 ⁇ g of DNA in 50 ⁇ l solution (100 ⁇ g in 100 ⁇ l total per mouse) using a disposable plastic insulin syringe and 28G 1 ⁇ 2 needle (Becton-Dickinson, Franklin Lakes, N.J., Cat. No. 329430) fitted with a plastic collar cut from a micropipette tip, as previously described (Hartikka, J., et al., Hum. Gene Ther. 7:1205-1217 (1996).
  • Plasmid constructs comprising the native coding regions encoding NP, M1, M2, HA, and eM2, IV proteins or fragments, variants or derivatives are constructed as follows.
  • the NP, M1, and M2 genes from IV are isolated from viral RNA by RT PCR, or prepared by direct synthesis if the wildtype sequence is known, by standard methods and are inserted into the vector VR10551 via standard restriction sites, by standard methods.
  • Plasmid constructs comprising human codon-optimized coding regions encoding NP, M1, M2, HA, eM2, and/or an eM2-NP fusion; or other codon-optimized coding regions encoding other IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, are prepared as follows.
  • the codon-optimized coding regions are generated using the full, minimal, or uniform codon optimization methods described herein.
  • the codon optimized coding regions are constructed using standard PCR methods described herein, or are ordered commercially.
  • Oligonucleotides representing about the first 23-24 aa extracellular region of M2 are constructed, and are used in an overlap PCR reaction with the NP coding regions described above, to create a coding region coding for an eM2/NP fusion protein, for example as shown in SEQ ID NOs 6 and 7.
  • the codon-optimized coding regions are inserted into the vector VR10551 via standard restriction sites, by standard methods.
  • Plasmids constructed as above are propagated in Escherichia coli and purified by the alkaline lysis method (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., ed. 2 (1989)). CsCl-banded DNA are ethanol precipitated and resuspended in 0.9% saline or PBS to a final concentration of 2 mg/ml for injection. Alternately, plasmids are purified using any of a variety of commercial kits, or by other known procedures involving differential precipitation and/or chromatographic purification.
  • Expression is tested by formulating each of the plasmids in DMRIE/DOPE and transfecting VM92 cells. The supernatants are collected and the protein production tested by Western blot or ELISA. The relative expression of the wild type and codon optimized constructs are compared.
  • the pDNA expression vector VR4700 which encodes the influenza NP protein has been described in the art. See, e.g. Sankar, V., Baccaglilni, L., Sawddey, M., Wheeler, C. J., Pillemer, S. R., Baum, B. J. and Atkinson, J. C., “Salivary Gland Delivery of pDNA-Cationic Lipolplexes Elicits Systemic Immune Responses,” Oral Diseases 8:275-281 (2002).
  • SEQ ID NO:46 1 atggatgcaa tgaagagagg gctctgctgt gtgctgctgc tgtgtggagc agtcttcgtt 61 tcgcccagcg ctagaggatc gggaatggcg tcccaaggca ccaaacggtc ttacgaacag 121 atggagactg atggagaacg ccagaatgcc actgaaatca gagcatccgt cggaaaatg 181 attggtggaa ttggacgatt ctacatccaa atgtgcaccg aactcaaact cagtgattat 241 gagggacggt tgatccaaaa cagcttaaca atagagagaa
  • Plasmid VR4707 expresses a secreted form of M2, i.e., TPA-M2.
  • the sequence was assembled using synthetic oligonucleotides in which the oligos were annealed amongst themselves, and then ligated and gel purified. The purified product was then ligated (cloned) into Eco RI/Sal I of VR10551.
  • the M2 sequence lacks the transmembrane domain; the cloned sequence contains amino acids [TPA(1-23)]ARGSG[M2(1-25)]GGG[M2(44-97)]. Amino acid residues between TPA and M2 and between M2 domains were added as flexible linkers.
  • TPA-M2ATM from VR4707), referred to herein as SEQ ID NO:47: 1 atggatgcaa tgaagagagg gctctgctgt gtgctgctgc tgtgtggagc agtcttcgtt 61 tcgcccagcg ctagaggatc gggaatgagt cttctgaccg aggtcgaaac ccctatcaga 121 aacgaatggg ggtgcagatg caacgattca agtgatcctg gcggcggcga tcggcttttt 181 ttcaaatgca tttatcggcg cttaaata
  • VR4710 was created by fusing the TPA leader and the first 24 amino acids of M2 from VR4707 to the full-length NP gene from VR4700.
  • Primers 5′-GCCGAATCCATGGATGCAATGAAG-3′ (SEQ ID NO:48) and 5′-GGTGCCTTGGGACGCCATATCACTTGAATCGTTGCA-3′ (SEQ ID NO:49) were used to amplify the TPA-M2 fragment from VR4707.
  • Primers 5′-TGCAACGATTCAAGTGATATGGCGTCCCAAGGCACC-3′ (SEQ ID NO:50) and 5′-GCCGTCGACTTAATTGTCGTACTC-3′ (SEQ ID NO:51) were used to amplify the NP gene from VR4700.
  • VR4750 was created by first reverse transcribing RNA from the mouse-adapted A/Hong Kong/1/68 virus stock using random hexamer to create a cDNA library. Then primers 5′ GGGCTAGCGCCGCCACCATGAAGACCATCATTGCT 3′ (SEQ ID NO:53) and 5′ CCGTCGACTCAAATGCAAATGTTGCA 3′ (SEQ ID NO:54) were employed to PCR the HA gene. The gene was inserted into the Invitrogen TOPO-TA vector first, and then sub-cloned into VR10551 using restriction enzymes NheI and SalI.
  • SEQ ID NO:55 1 atgaagacca tcattgcttt gagctacatt ttctgtctgg ctctcggcca agaccttcca 61 ggaaatgaca acaacacagc aacgctgtgc ctgggacatc atgcggtgcc aaacggaaca 121 ctagtgaaaa caatcacaga tgatcagatt gaagtgacta atgctactga gctagttcag 181 agctcctcaa cggggaaat atgcaacaat cctcatcgaa tccttgatgg aatagactgc 241 acactgatag atgctctat
  • VR4750 expression was not clearly detected in vitro by Western blot Assay, two 100 ⁇ g vaccinations of VR4750 have been shown to protect mice from intranasal challenge with mouse-adapted A/Hong Kong/68 virus.
  • VR4752 was created by first reverse transcribing RNA from the mouse-adapted A/Puerto Rico/8/34 virus stock using random hexamer to create a cDNA library. Then primers 5′ GGGCTAGCGCCGCCACCATGAAGGCAAACCTACTG 3′ (SEQ ID NO:56) and 5′ CCGTCGACTCAGATGCATATTCTGCA 3′ (SEQ ID NO:57) were employed to PCR the HA gene. The gene was then cloned into the TOPO-TA vector first, and then sub-cloned into VR10551 using restriction enzymes NheI and SalI.
  • SEQ ID NO:58 1 atgaaggcaa acctactggt cctgttatgt gcacttgcag ctgcagatgc agacacaata 61 tgtataggct accatgcgaa caattcaacc gacactgttg acacagtgct cgagaagaat 121 gtgacagtga cacactctgt taacctgctc gaagacagcc acaacggaa actatgtaga 181 ttaaaggaa tagccccact acaattgggg aaatgtaaca tcgcggatg gcttggga 241 aacccagaat gcgacc
  • Purified VR4752 DNA was used to transfect the murine cell line VM92 to determine expression of the HA protein. Expression of HA was confirmed with a Western Blot assay. Expression was visualized with a commercially available goat anti-influenza A (H1N1) antibody.
  • a direct fusion of the M2 gene to the M1 gene was synthesized based on a codon-optimized sequence derived from methods described in Example 4 using the “universal” optimization strategy.
  • the synthesized gene was received in the pUC119 vector and then sub-cloned into VR10551 as an EcoRI-SalI fragment.
  • SEQ ID NO:59 1 atgagcctgc tgaccgaggt ggagaccccc atcagaaacg agtggggctg cagatgcaac 61 gacagcagcg accccctggt ggtggccgcc agcatcatcg gcatcctgca cctgatcctg 121 tggatcctgg acagactgtt cttcaagtgc atctacagac tgttcaagca cggcctgaag 181 agaggcccca gcaccgaggg cgtgcccgag agcatgagag aggagtacag aaggagcag 241 cagaacgccg tggacg
  • Purified VR4755 DNA was used to transfect the murine cell line VM92 to determine expression of the M2M1 fusion protein. Expression of M2M1 was confirmed with a Western Blot assay. Expression of the M2M1 fusion was visualized with commercially available anti-M1 and anti-M2 monoclonal antibodies.
  • the segment 7 RNA of influenza A encodes both the M1 and M2 genes.
  • a consensus amino acid sequence for M1 and M2 was derived according to methods described herein.
  • the consensus sequences for both proteins are identical to the M1 and M2 amino acid sequences derived from the IV strain A/Niigata/137/96, represented herein as SEQ ID NO:77 and SEQ ID NO:78, respectively.
  • the native sequence for segment 7, A/Niigata/137/96 was synthesized and received as an insert in pUC119.
  • the segment 7 insert was sub-cloned into VR10551 as an EcoRI-SalI fragment.
  • SEQ ID NO:60 1 atgagccttc taaccgaggt cgaaacgtat gttctcta tcgttccatc aggccccctc 61 aaagccgaaa tcgcgcagag acttgaagat gtctttgctg ggaaaacac agatcttgag 121 gctctcatgg aatggctaaa gacaagacca atcctgtcac ctctgactaa ggggattttg 181 gggtttgtgt tcacgctcac cgtgcccagt gagcgaggac tgcagcgtag acgctttgtc 241 caaaatg
  • SEQ ID NO:77 (“consensus” (A/Niigata/137/96) M1): MSLLTEVETYVLSIVPSGPLKAEIAQRLEDVFAGKNTDLEALMEWLKTRP ILSPLTKGILGFVFTLTVPSERGLQRRRFVQNALNGNGDPNNMDRAVKLY RKLKREITFHGAKEIALSYSAGALASCMGLIYNRMGAVTTEVAFGLVCAT CEQIADSQHRSHRQMVATTNPLIRHENRMVLASTTAKAMEQMAGSSEQAA EAMEIASQARQMVQAMRAIGTHPSSSAGLKDDLLENLQTYQKRMGVQM QRFK
  • SEQ ID NO:78 (“consensus” (A/Niigata/137/96) M2): MSLLTEVETPIRNEWGCRCNDSSDPLVVAASIIGILHLILWILDRLFFKC IYRLFKHGLKRGPSTEGVPESMREEYRKEQQNAVDADDSHFVSIELE
  • segment 7 contains selectively codon-optimized regions of segment 7. Optimization of the coding regions in segment 7 is selective, because segment 7 contains two overlapping coding regions (i.e., encoding M1 and M2,) and these coding regions are partially in different reading frames.
  • M1 is encoded by bp 1 through 759 of the segment 7 RNA
  • M2 is encoded by a spliced messenger RNA which includes nucleotides 1 to 26 of segment 7 spliced to nucleotides 715 to 982 of segment 7.
  • optimization of the region from 715 to 759 is avoided because the M1 and M2 coding sequences (in different reading frames) overlap in that region. Due to the splicing that occurs to join bp 26 to an alternate frame at bp 715 of the segment 7 sequence, optimization in these splicing regions is also avoided; adjacent regions that arguably could also participate in splicing are likewise avoided. Optimization is done in a manner to insure that no new splicing sites are inadvertently introduced. The areas that are optimized are done so using “universal” strategy, e.g. inserting the most frequently used codon for each amino acid.
  • SEQ ID NO:61 1 atgagcctgc tgaccgaggt cgaaacgtat gttctctcta tcgtgcccag cggcccctg 61 aaggccgaga tcgcccagag actggaggac gtgttcgccg gcaagaacac cgacctggag 121 gccctgatgg agtggctgaa gaccagaccc atcctgagcc cctgaccaa gggcatcctg 181 ggcttcgtgt tcaccctgac cgtgcccagc gagagaggcc tgcagagaag aaagattcgtg 241 caga
  • the codon optimized coding region for M1 extends from nucleotide 1 to nucleotide 759 of SEQ ID NO:61 including the stop codon, and is represented herein as SEQ ID NO:79.
  • the codon-optimized coding region for M2 extends from nucleotide 1 to nucleotide 26 of SEQ ID NO:61 spliced to nucleotide 715 through nucleotide 959 of SEQ ID NO:61, including the stop codon, and is represented herein as SEQ ID NO:80.
  • Optimized M1 Coding Region (SEQ ID NO:79): ATGAGCCTGCTGACCGAGGTCGAAACGTATGTTCTCTCTATCGTGCCCAG CGGCCCCCTGAAGGCCGAGATCGCCCAGAGACTGGAGGACGTGTTCGCCG GCAAGAACACCGACCTGGAGGCCCTGATGGAGTGGCTGAAGACCAGACCC ATCCTGAGCCCCCTGACCAAGGGCATCCTGGGCTTCGTGTTCACCCTGAC CGTGCCCAGCGAGAGAGGCCTGCAGAGAAGAAGATTCGTGCAGAACGCCC TGAACGGCAACGGCGACCCCAACAACATGGACAGAGCCGTGAAGCTGTAC AGAAAGCTGAAGAGAGATCACCTTCCACGGCGCCAAGGAGATCGCCCT GAGCTACAGCCGGCCGGCCCTGGCCAGCTGCATGGGCCTGATCTACAACA GAATGGGCCGTGACCACCGAGGTGGCCTTCGGCCTGGTGTGCGCCACC TGCGAGCAGATCGCCGACAGCCAGCACAACA
  • Optimized M2 Coding Region (SEQ ID NO:80): ATGAGCCTGCTGACCGAGGTCGAAACACCTATCAGAAACGAATGGGGGTG CAGATGCAACGATTCAAGTGACCCCCTGGTGGTGGCCGCCAGCATCATCG GCATCCTGCACCTGATCCTGTGGATCCTGGACAGACTGTTCTTCAAGTGC ATCTACAGACTGTTCAAGCACGGCCTGAAGAGAGGCCCCAGCACCGAGGG CGTGCCCGAGAGCATGAGAGAGGAGTACAGAAAGGAGCAGCAGAACGCCG TGGACGCCGACGACAGCCACTTCGTGAGCATCGAGCTGGAGTGA
  • the eM2-NP fusion was codon-optimized, inserted in pUC119 and sub-cloned into VR10551 as an EcoRI-SalI fragment.
  • the following is the open reading frame for eM2-NP: codon-optimized by Contract (from VR4757), referred to herein as SEQ ID NO:62: 1 atgagcttgc tcactgaagt cgagacacca atcagaaacg aatggggatg tagatgcaac 61 gatagctcag acatggcctc ccagggaacc aaaagaagct atgaacagat ggagactgac 121 ggagagagac agaacgccac agagatcaga gctagtgtag gaaagatgat agacggtatc 181 gggcgatttt acattcaaat gtgtacggaa ttgaactca gcg
  • the eM2-NP fusion gene in VR4758 was codon-optimized and synthesized. The gene was inserted into pUC119 and sub-cloned into VR10551 as an EcoRI-SalI fragment.
  • the following is the open reading frame for eM2-NP: codon-optimized by Applicants (from VR4758), referred to herein as SEQ ID NO:63: 1 atgagcctgc tgaccgaggt ggagaccccc atcagaaacg agtggggctg cagatgcaac 61 gacagcagcg acatggccag ccagggcacc aagagaagct acgagcagat ggagaccgac 121 ggcgagagac agaacgccac cgagatcaga gccagcgtgg gacggcatc 181 ggcagattct acatccagat gt
  • the M2 gene was PCR-amplified from VR4755 using the primers 5′-GCCGAATTCGCCACCATGAGCCTGCTGACC-3′ (SEQ ID NO:64) and 5′-GCCGTCGACTGATCACTCCAGCTCGATGCTCAC-3′ (SEQ ID NO:65) and sub-cloned into VR10551 as an EcoRI-SalI fragment.
  • the M1 gene was PCR-amplified from VR4755 using the primers 5′-GCCGAATTCGCCACCATGTCCCTGCTGACAGAAGTG-3′ (SEQ ID NO:67) and 5′-GCCGTCGACTGATCACTTGAATCTCTGCATC-3′ (SEQ ID NO:68) and sub-cloned into VR10551 as an EcoRI-SalI fragment.
  • the NP gene was PCR-amplified from VR4757 using primers 5′-GCCGAATTCGCCACCATGGCCTCCCAGGGAACCAAAAG-3′ (SEQ ID NO:70) and 5′-GCCGTCGACTGATCAATTGTCGTACTCTTC-3′ (SEQ ID NO:71) and sub-cloned into VR10551 as an EcoRI-SalI fragment.
  • NP codon-optimized by Contract (from VR4761), referred to herein as SEQ ID NO:72: 1 atg gcc tcc cag gga acc aaa aga agc tat gaa cag atg gag act gac 49 gga gag aga cag aac gcc aca gag atc aga gct agt gta gga aag atg 97 ata gac ggt atc ggg cga ttt tac att caa atg tgt acg gaa ttg aaaa 145 ctc agc gac tat gaa ggc aga ctt atc cag aac tca ctc aca att gag 193 cgc atg gta ctc agt gca ttt gat gaa aga agg
  • VR4761 DNA was used to transfect the murine cell line VM92 to determine expression of the NP protein. Expression of NP was confirmed with a Western Blot assay. Expression was visualized with a mouse polyclonal anti-NP antibody. In vitro expression of VR4761 was significantly higher than VR4700 and comparable to VR4762.
  • the NP gene was PCR-amplified from VR4758 using primers 5′-GCCGAATTCGCCACCATGGCCAGCCAGGGCACCAAG-3′ (SEQ ID NO:73) and 5′-GCCGTCGACTGATCAGTTGTCGTACTCC-3′ (SEQ ID NO:74) and sub-cloned into VR10551 as an EcoRI-SalI fragment.
  • NP codon-optimized by Applicants (from VR4762), referred to herein as SEQ ID NO:75: 1 atg gcc agc cag ggc acc aag aga agc tac gag cag atg gag acc gac 49 ggc gag aga cag aac gcc acc gag atc aga gcc agc gtg ggc aag atg 97 atc gac ggc atc ggc aga ccc tac atc cag atg tgc acc gag ctg aag 145 ctg agc gac tac gag ggc aga ctg atc cag aac agc ctg acc atc gag 193 aga atg gtg ctg agc gcccc gac gag aga aga aac aga tac ctg gag 24
  • VR4762 DNA was used to transfect the murine cell line VM92 to determine expression of the NP protein. Expression of NP was confirmed with a Western Blot assay. Expression was visualized with a mouse polyclonal anti-NP antibody. In vitro expression of VR4762 was significantly higher than VR4700 and comparable to VR4761.
  • single plasmids which contain two or more IV coding regions are constructed according to standard methods.
  • a polycistronic construct where two or more IV coding regions are transcribed as a single transcript in eukaryotic cells may be constructed by separating the various coding regions with IRES sequences.
  • two or more coding regions may be inserted into a single plasmid, each with their own promoter sequence.
  • Recombinant NP DNA and protein may be prepared using the following procedure.
  • Eukaryotic cells may be used to express the NP protein from a transfected expression plasmid.
  • a baculovirus system can be used wherein insect cells such as, but not limited to, Sf9, Sf21, or D.Mel-2 cells are infected with a recombinant baculovirus which can expresses the NP protein.
  • Cells which have been infected with recombinant baculoviruses, or contain expression plasmids, encoding recombinant NP are collected by knocking and scraping cells off the bottom of the flask in which they are grown.
  • Cells infected for 24 or 48 hours are less easy to detach from flask and may lyse, thus care must be taken with their removal.
  • the flask containing the cells is then rinsed with PBS and the cells are transfered to 250 ml conical tubes.
  • the tubes are spun at 1000 rpm in J-6 centrifuge (300 ⁇ g) for about 5-10 minutes.
  • the cell pellets are washed two times with PBS and then resuspended in about 10-20 ml of PBS in order to count.
  • Approximately 10 6 cells are used per lane of a standard SDS-PAGE mini-protein gel which is equivalent to the whole cell fraction for gel analysis purposes.
  • 10% NP40 is added to the cells for a final concentration of 0.5%.
  • the cell-NP40 mixture is vortexed and placed on ice for 10 minutes, vortexing occasionally. After ice incubation, the cells are spun at 1500 rpm in a J-6 centrifuge (600 ⁇ 1) for 10 minutes. The supernantant is removed which is the cytoplasmic fration.
  • the nuclei are resuspended in buffer C to 5 ⁇ 10 7 nuclei/ml.
  • the nuclei are vortexed vigorously to break up particles and an aliquot is removed for the mini-protein gel which is the nuclei fraction.
  • a quarter of the volume of 5M NaCl is added and the mixture is sonicated for 5 minutes at a maximum output in a bath-type sonicator at 4° C., in 1-2 minute bursts, resting 30 seconds between bursts.
  • the sonicated mixture is stirred at 4° C., then spun at 12000 ⁇ g for 10 minutes.
  • a sample is removed for the protein mini-gel equivalent to approximately 10 6 nuclei.
  • the sample for the gel is centrifuged and the supernatant is the nuclear extract and the pellet is the nuclear pellet for gel analysis.
  • NP nuclear equivalents
  • a small amount (about 10 6 nuclear equivalents) of the nuclear pellet is resuspended directly in gel sample buffer and run with equivalent amounts of whole cells, cytoplasm, nuclei, nuclear extract and nuclear pellet.
  • the above method gives relatively crude NP.
  • 2.1 M NaCl can be added to the nuclear pellet instead of 5M NaCl. This will bring the salt content to 0.42M NaCl.
  • the supernatant will then contain about 60-70% of the total NP plus nuclear proteins.
  • the resulting pellet is then extracted with 1M NaCl and centrifuged as above. The supernatant will contain NP at more than 95% purity.
  • influenza NP strain A
  • the method by which amino acid sequences for influenza NP was chosen is as follows. The http://www.flu.lanl.gov database containing influenza sequences for each segment was searched for influenza A strains, human, NP, amino acids. Results gave about 400 sequences, the majority of which were only partial sequences. The sequences were subsequently narrowed down to 85 approximately full length sequences. If different passages of the same strain were found, the earliest passage was chosen. The sequences were further narrowed down to 28 full length NP sequences isolated from 1990 to 2000 (no full-length sequences from 2001-2003).
  • Balb/c epitope TYQRTRALV The dominant Balb/c epitope TYQRTRALV is still maintained in the new consensus; changes to other theoretical human epitopes have not been determined as yet.
  • SEQ ID NO:76 1 masqgtkrsy eqmetdgerq nateirasvg kmidgigrfy iqmctelkls dyegrliqns 61 ltiermvlsa fderrnryle ehpsagkdpk ktggpiyrrv dgkwmrelvl ydkeeirriw 121 rqanngedat aglthmmiwh snlndttyqr tralvrtgmd prmcslmqgs tlprrsgaag 181 aavkgigtmv melirmikrg indrnfwrge ngrktrsaye rmcnilkgkf qtaaqrammd 241 qvresrnpgn aeiedlif
  • Consensus sequences for M1 and M2 were determined in a similar fashion, as follows.
  • the search parameters on the http://www.flu.lanl.gov/website were: influenza A strains, human, segment 7, nucleotide (both M1 and M2 are derived from segment 7).
  • Full-length sequences from 1990-1999 (no 2000+ sequences were available) were chosen. For sequences with the same year and city, only the earliest passage was used. For entries for the same year, sequences were eliminated that were identical to another sequence isolated from the same year (even if different city). Twenty one sequences, full-length for both M1 and M2 from 1993-1999, were compared. At each position, the amino acid with the simple majority was used.
  • the M1 amino acid consensus sequence is referred to herein as SEQ ID NO:77: 1 mslltevety vlsivpsgpl kaeiaqrled vfagkntdle almewlktrp ilspltkgil 61 gfvftltvps erglqrrrfv qnalngngdp nnmdravkly rklkreitfh gakeialsys 121 agalascmgl iynrmgavtt evafglvcat ceqiadsqhr shrqmvattn plirhenrmv 181 lasttakame qmagsseqaa eameiasqar qmvqamraig thpsssaglk ddllenlqty 241 qkrmgvqmqr fk
  • the M2 amino acid consensus sequence is referred to herein as SEQ ID NO:78: 1 mslltevetp irnewgcrcn dssdplvvaa siigilhlil wildrlffkc iyrlfkhglk 61 rgpstegvpe smreeyrkeq qnavdaddsh fvsiele
  • the program did not always use the most frequent codon for amino acids such as cysteine proline, and arginine. To change this, go back to the Edit CUT TAB and manually drag the rainbow colored bar to 100% for the desired codon. Then re-do start under the Output TAB.
  • Default minimum score is 0.4, where: % splice % false sites recognized positives Human 5′ Splice sites 93.2% 5.2% Human 3′ Splice sites 83.8% 3.1%
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, HA, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are formulated with the poloxamer CRL 1005 and BAK (Benzalkonium chloride 50% solution, available from Ruger Chemical Co. Inc.) by the following methods.
  • a carrier protein e.g., HBcAg
  • various controls e.g., empty vector
  • the concentration of CRL 1005 is adjusted depending on, for example, transfection efficiency, expression efficiency, or immunogenicity, to achieve a final concentration of between about 1 mg/ml to about 75 mg/ml, for example, about 1 mg/ml, about 2 mg/ml, about 3 mg/ml, about 4 mg/ml, about 5 mg/ml, about 6.5 mg/ml, about 7 mg/ml, about 7.5 mg/ml, about 8 mg/ml, about 9 mg/ml, about 10 mg/ml, about 15 mg/ml, about 20 mg/ml, about 25 mg/ml, about 30 mg/ml, about 35 mg/ml, about 40 mg/ml, about 45 mg/ml, about 50 mg/ml, about 55 mg/ml, about 60 mg/ml, about 65 mg/ml, about 70 mg/ml, or about 75 mg/ml of CRL 1005.
  • concentration of DNA is adjusted depending on many factors, including the amount of a formulation to be delivered, the age and weight of the subject, the delivery method and route and the immunogenicity of the antigen being delivered.
  • formulations of the present invention are adjusted to have a final concentration from about 1 ng/ml to about 30 mg/ml of plasmid (or other polynucleotide).
  • a formulation of the present invention may have a final concentration of about 1 ng/ml, about 5 ng/ml, about 10 ng/ml, about 50 ng/ml, about 100 ng/ml, about 500 ng/ml, about 1 ⁇ g/ml, about 5 ⁇ g/ml, about 10 ⁇ g/ml, about 50 ⁇ g/ml, about 200 ⁇ g/ml, about 400 ⁇ g/ml, about 600 ⁇ g/ml, about 800 ⁇ g/ml, about 1 mg/ml, about 2 mg/ml, about 2.5, about 3 mg/ml, about 3.5, about 4 mg/ml, about 4.5, about 5 mg/ml, about 5.5 mg/ml, about 6 mg/ml, about 7 mg/ml, about 8 mg/ml, about 9 mg/ml, about 10 mg/ml, about 20 mg/ml, or about 30 mg mg/ml of a plasmid.
  • Certain formulations of the present invention include a cocktail of plasmids (see, e,g., Example 2 supra) of the present invention, e.g., comprising coding regions encoding IV proteins NP, M1 and/or M2 and optionally, plasmids encoding immunity enhancing proteins, e.g., cytokines.
  • plasmids desired in a cocktail are combined together in PBS or other diluent prior to the addition to the other ingredients.
  • plasmids may be present in a cocktail at equal proportions, or the ratios may be adjusted based on, for example, relative expression levels of the antigens or the relative immunogenicity of the encoded antigens.
  • various plasmids in the cocktail may be present in equal proportion, or up to twice or three times as much of one plasmid may be included relative to other plasmids in the cocktail.
  • concentration of BAK may be adjusted depending on, for example, a desired particle size and improved stability.
  • formulations of the present invention include CRL 1005 and DNA, but are free of BAK.
  • BAK-containing formulations of the present invention are adjusted to have a final concentration of BAK from about 0.05 mM to about 0.5 mM.
  • a formulation of the present invention may have a final BAK concentration of about 0.05 mM, 0.1 mM, 0.2 mM, 0.3 mM, 0.4 mM or 0.5 mM.
  • the total volume of the formulations produced by the methods below may be scaled up or down, by choosing apparatus of proportional size.
  • the three components of the formulation, BAK, CRL 1005, and plasmid DNA may be added in any order.
  • the term “cloud point” refers to the point in a temperature shift, or other titration, at which a clear solution becomes cloudy, i.e., when a component dissolved in a solution begins to precipitate out of solution.
  • This example describes the preparation of a formulation comprising 0.3 mM BAK, 7.5 mg/ml CRL 1005, and 5 mg/ml of DNA in a total volume of 3.6 ml.
  • the ingredients are combined together at a temperature below the cloud point and then the formulation is thermally cycled to room temperature (above the cloud point) several times, according to the protocol outlined in FIG. 2 .
  • a 1.28 mM solution of BAK is prepared in PBS, 846 ⁇ l of the solution is placed into a 15 ml round bottom flask fitted with a magnetic stirring bar, and the solution is stirred with moderate speed, in an ice bath on top of a stirrer/hotplate (hotplate off) for 10 minutes.
  • CRL 1005 (27 ⁇ l) is then added using a 100 ⁇ l positive displacement pipette and the solution is stirred for a further 60 minutes on ice.
  • Plasmids comprising codon-optimized coding regions encoding, for example, NP, M1, and M2 as described herein, and optionally, additional plasmids comprising codon-optimized or non-codin-optimized coding regions encoding, e.g., additional IV proteins, and or other proteins, e.g., cytokines, are mixed together at desired proportions in PBS to achieve 6.4 mg/ml total DNA.
  • This plasmid cocktail is added drop wise, slowly, to the stirring solution over 1 min using a 5 ml pipette. The solution at this point (on ice) is clear since it is below the cloud point of the poloxamer and is further stirred on ice for 15 min. The ice bath is then removed, and the solution is stirred at ambient temperature for 15 minutes to produce a cloudy solution as the poloxamer passes through the cloud point.
  • the flask is then placed back into the ice bath and stirred for a further 15 minutes to produce a clear solution as the mixture is cooled below the poloxamer cloud point.
  • the ice bath is again removed and the solution stirred at ambient temperature for a further 15 minutes. Stirring for 15 minutes above and below the cloud point (total of 30 minutes), is defined as one thermal cycle.
  • the mixture is cycled six more times.
  • the resulting formulation may be used immediately, or may be placed in a glass vial, cooled below the cloud point, and frozen at ⁇ 80 ° C. for use at a later time.
  • This example describes the preparation of a formulation comprising 0.3 mM BAK, 34 mg/ml or 50 mg/ml CRL 1005, and 5.0 mg/ml of DNA in a final volume of 4.0 ml.
  • the ingredients are combined together at a temperature below the cloud point, then the formulation is thermally cycled to room temperature (above the cloud point) several times, diluted, and filtered according to the protocol outlined in FIG. 3 .
  • Plasmids comprising codon-optimized coding regions encoding, for example, NP, M1, and M2 as described herein, and optionally, additional plasmids comprising codon-optimized or non-codin-optimized coding regions encoding, e.g., additional IV proteins, and or other proteins, e.g., cytokines, are mixed together at desired proportions in PBS to achieve 6.4 mg/ml total DNA.
  • This plasmid cocktail is placed into the 15 ml round bottom flask fitted with a magnetic stirring bar, and for the formulation containing 50 mg/ml CRL 1005, 3.13 ml of a solution containing about 3.2 mg/ml of NP encoding plasmid and about 3.2 mg/ml M2 encoding plasmid (about 6.4 mg/ml total DNA) is placed into the 15 ml round bottom flask fitted with a magnetic stirring bar, and the solutions are stirred with moderate speed, in an ice bath on top of a stirrer/hotplate (hotplate off) for 10 minutes.
  • a stirrer/hotplate hotplate off
  • CRL 1005 (136 ⁇ l for 34 mg/ml final concentration, and 200 ⁇ l for 50 mg/ml final concentration) is then added using a 200 ⁇ l positive displacement pipette and the solution is stirred for a further 30 minutes on ice.
  • Solutions of 1.6 mM and 1.8 mM BAK are prepared in PBS, and 734 ⁇ l of 1.6 mM and 670 ⁇ l of 1.8 mM are then added drop wise, slowly, to the stirring poloxamer solutions with concentrations of 34 mg/ml or 50 mg/ml mixtures, respectively, over 1 min using a 1 ml pipette.
  • the solutions at this point are clear since they are below the cloud point of the poloxamer and are stirred on ice for 30 min.
  • the ice baths are then removed; the solutions stirred at ambient temperature for 15 minutes to produce cloudy solutions as the poloxamer passes through the cloud point.
  • the flasks are then placed back into the ice baths and stirred for a further 15 minutes to produce clear solutions as the mixtures cooled below the poloxamer cloud point.
  • the ice baths are again removed and the solutions stirred for a further 15 minutes. Stirring for 15 minutes above and below the cloud point (total of 30 minutes), is defined as one thermal cycle. The mixtures are cycled two more times.
  • the resulting formulations may be used immediately, or may be transferred to glass vials, cooled below the cloud point, and frozen at ⁇ 80 ° C. for use at a later time.
  • This example describes a simplified preparation of a formulation comprising 0.3 mM BAK, 7.5 mg/ml CRL 1005, and 5 mg/ml of DNA in a total volume of 2.0 ml.
  • the ingredients are combined together at a temperature below the cloud point and then the formulation is simply filtered and then used or stored, according to the protocol outlined in FIG. 4 .
  • a 0.77 mM solution of BAK is prepared in PBS, and 780 ⁇ l of the solution is placed into a 15 ml round bottom flask fitted with a magnetic stirring bar, and the solution is stirred with moderate speed, in an ice bath on top of a stirrer/hotplate (hotplate off) for 15 minutes.
  • CRL 1005 (15 ⁇ l) is then added using a 100 ⁇ l positive displacement pipette and the solution is stirred for a further 60 minutes on ice.
  • Plasmids comprising codon-optimized coding regions encoding, for example, NP, M1, and M2 as described herein, and optionally, additional plasmids comprising codon-optimized or non-codin-optimized coding regions encoding, e.g., additional IV proteins, and or other proteins, e.g., cytokines, are mixed together at desired proportions in PBS to achieve a final concentration of about 8.3 mg/ml total DNA.
  • This plasmid cocktail is added drop wise, slowly, to the stirring solution over 1 min using a 5 ml pipette. The solution at this point (on ice) is clear since it is below the cloud point of the poloxamer and is further stirred on ice for 15 min.
  • one Steriflip® 50 ml disposable vacuum filtration devices with a 0.22 ⁇ m Millipore Express® membrane (available from Millipore, cat # SCGP00525) is placed in an ice bucket, with a vacuum line attached and left for 1 hour to allow the device to equilibrate to the temperature of the ice.
  • the poloxamer formulation is then filtered under vacuum, below the cloud point and then allowed to warm above the cloud point.
  • the resulting formulations may be used immediately, or may be transferred to glass vials, cooled below the cloud point and then frozen at ⁇ 80° C. for use at a later time.
  • the immunogenicity of the various IV expression products encoded by the codon-optimized polynucleotides described herein are initially evaluated based on each plasmid's ability to mount an immune response in vivo. Plasmids are tested individually and in combinations by injecting single constructs as well as multiple constructs. Immunizations are initially carried out in animals, such as mice, rabbits, goats, sheep, non-human primates, or other suitable animal, by intramuscular (IM) injections. Serum is collected from immunized animals, and the antigen specific antibody response is quantified by ELISA assay using purified immobilized antigen proteins in a protein—immunized subject antibody—anti-species antibody type assay, according to standard protocols.
  • IM intramuscular
  • the tests of immunogenicity further include measuring antibody titer, neutralizing antibody titer, T-cell proliferation, T-cell secretion of cytokines, cytolytic T cell responses, and by direct enumeration of antigen specific CD4+ and CD8+ T-cells. Correlation to protective levels of the immune responses in humans are made according to methods well known by those of ordinary skill in the art. See above.
  • Plasmid DNA is formulated with a poloxamer by any of the methods described in Example 3.
  • plasmid DNA is prepared as described above and dissolved at a concentration of about 0.1 mg/ml to about 10 mg/ml, preferably about 1 mg/ml, in PBS with or without transfection-facilitating cationic lipids, e.g., DMRIE/DOPE at a 4:1 DNA:lipid mass ratio.
  • Alternative DNA formulations include 150 mM sodium phosphate instead of PBS, adjuvants, e.g., VaxfectinTM at a 4:1 DNA: VaxfectinTM mass ratio, mono-phosphoryl lipid A (detoxified endotoxin) from S.
  • MPL minnesota
  • TDM trehalosedicorynomycolateAF
  • MPL+TDM solubilized mono-phosphoryl lipid A formulation
  • AF available from Corixa
  • compound # VC1240 see Shriver, J. W. et al., Nature 415:331-335 (2002), and P.C.T. Publication No. WO 02/00844 A2, each of which is incorporated herein by reference in its entirety).
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are injected into BALB/c mice as single plasmids or as cocktails of two or more plasmids, as either DNA in PBS or formulated with the poloxamer-based delivery system: 2 mg/ml DNA, 3 mg/ml CRL 1005, and 0.1 mM BAK.
  • a carrier protein e.g., HBcAg
  • various controls e.g., empty vector
  • mice Groups of 10 mice are immunized three times, at biweekly intervals, and serum is obtained to determine antibody titers to each of the antigens. Groups are also included in which mice are immunized with a trivalent preparation, containing each of the three plasmid constructs in equal mass.
  • the immunization schedule is as follows: Day ⁇ 3 Pre-bleed Day 0 Plasmid injections, intramuscular, bilateral in rectus femoris, 5-50 ⁇ g/leg Day 21 Plasmid injections, intramuscular, bilateral in rectus femoris, 5-50 ⁇ g/leg Day 49 Plasmid injections, intramuscular, bilateral in rectus femoris, 5-50 ⁇ g/leg Day 59 Serum collection
  • Serum antibody titers are determined by ELISA with recombinant proteins, peptides or transfection supernatants and lysates from transfected VM-92 cells live, inactivated, or lysed virus.
  • VaxfectinTM (a 1:1 molar ratio of the cationic lipid VC1052 and the neutral co-lipid DPyPE) is a synthetic cationic lipid formulation which has shown promise for its ability to enhance antibody titers against when administered with DNA intramuscularly to mice.
  • VaxfectinTM shows promise as a delivery system and as an adjuvant in a DNA vaccine.
  • VaxfectinTM mixtures are prepared by mixing chloroform solutions of VC1052 cationic lipid with chloroform solutions of DpyPE neutral co-lipid.
  • Dried films are prepared in 2 ml sterile glass vials by evaporating the chloroform under a stream of nitrogen, and placing the vials under vacuum overnight to remove solvent traces. Each vial contains 1.5 ⁇ mole each of VC1052 and DPyPE.
  • Liposomes are prepared by adding sterile water followed by vortexing. The resulting liposome solution is mixed with DNA at a phosphate mole:cationic lipid mole ratio of 4:1.
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are mixed together at desired proportions in PBS to achieve a final concentration of 1.0 mg/ml.
  • the plasmid cocktail, as well as the controls, are formulated with VaxfectinTM.
  • mice Groups of 5 BALB/c female mice are injected bilaterally in the rectus femoris muscle with 50 ⁇ l of DNA solution (100 ⁇ l total/mouse), on days 1 and 21 and 49 with each formulation. Mice are bled for serum on days 0 (prebleed), 20 (bleed 1), and 41 (bleed 2), and 62 (bleed 3), and up to 40 weeks post-injection. Antibody titers to the various IV proteins encoded by the plasmid DNAs are measured by ELISA as described elsewhere herein.
  • Cytolytic T-cell responses are measured as described in Hartikka et al. “Vaxfectin Enhances the Humoral Response to Plasmid DNA-encoded Antigens,” Vaccine 19:1911-1923 (2001) and is incorporated herein in its entirety by reference. Standard ELISPOT technology is used for the CD4+ and CD8+ T-cell assays as described in Example 6, part A.
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are prepared according to the immunization scheme described above and injected into a suitable animal for generating polyclonal antibodies. Serum is collected and the antibody titered as above.
  • Monoclonal antibodies are also produced using hybridoma technology (Kohler, et al., Nature 256:495 (1975); Kohler, et al., Eur. J. Immunol. 6:511 (1976); Kohler, et al., Eur. J. Immunol. 6:292 (1976); Hammerling, et al., in Monoclonal Antibodies and T - Cell Hybridomas, Elsevier, N.Y., (1981), pp. 563-681, each of which is incorporated herein by reference in its entirety).
  • such procedures involve immunizing an animal (preferably a mouse) as described above.
  • the splenocytes of such mice are extracted and fused with a suitable myeloma cell line.
  • a suitable myeloma cell line may be employed in accordance with the present invention; however, it is preferable to employ the parent myeloma cell line (SP2O), available from the American Type Culture Collection, Rockville, Md.
  • SP2O parent myeloma cell line
  • the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands et al., Gastroenterology 80:225-232 (1981), incorporated herein by reference in its entirety.
  • the hybridoma cells obtained through such a selection are then assayed to identify clones which secrete antibodies capable of binding the various IV proteins.
  • additional antibodies capable of binding to IV proteins described herein may be produced in a two-step procedure through the use of anti-idiotypic antibodies.
  • Such a method makes use of the fact that antibodies are themselves antigens, and that, therefore, it is possible to obtain an antibody which binds to a second antibody.
  • various IV-specific antibodies are used to immunize an animal, preferably a mouse.
  • the splenocytes of such an animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones which produce an antibody whose ability to bind to the IV protein-specific antibody can be blocked by the cognate IV protein.
  • Such antibodies comprise anti-idiotypic antibodies to the IV protein-specific antibody and can be used to immunize an animal to induce formation of further IV-specific antibodies.
  • Fab and F(ab′) 2 and other fragments of the antibodies of the present invention may be used according to the methods disclosed herein.
  • Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab′) 2 fragments).
  • enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab′) 2 fragments).
  • NP, M1, M2, HA and eM2 binding fragments can be produced through the application of recombinant DNA technology or through synthetic chemistry.
  • “humanized” chimeric monoclonal antibodies Such antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above. Methods for producing chimeric antibodies are known in the art. See, for review, Morrison, Science 229:1202 (1985); Oi, et al., BioTechniques 4:214 (1986); Cabilly, et al., U.S. Pat. No.
  • anti-IV antibodies are used, for example, in diagnostic assays, as a research reagent, or to further immunize animals to generate IV-specific anti-idiotypic antibodies.
  • uses for anti-IV antibodies include use in Western blots, ELISA (competitive, sandwich, and direct), immunofluorescence, immunoelectron microscopy, radioimmunoassay, immunoprecipitation, agglutination assays, immunodiffusion, immunoelectrophoresis, and epitope mapping (Weir, D. Ed. Handbook of Experimental Immunology, 4 th ed. Vols. I and II, Blackwell Scientific Publications (1986)).
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, HA, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, (100 ⁇ g/50 ⁇ l total DNA) are delivered to BALB/c mice at 0, 2 and 4 weeks via i.m., intranasal (i.n.), intravenous (i.v.), intravaginal (i.vag.), intrarectal (i.r.) or oral routes.
  • a carrier protein e.g., HBcAg
  • various controls e.g., empty vector, (100 ⁇ g/50 ⁇ l total DNA)
  • the DNA is delivered unformulated or formulated with the cationic lipids DMRIE/DOPE (DD) or GAP-DLRIE/DOPE (GD).
  • DD cationic lipid
  • GD GAP-DLRIE/DOPE
  • serum IgG titers against the various IV antigens are measured by ELISA and splenic T-cell responses are measured by antigen-specific production of IFN-gamma and IL-4 in ELISPOT assays.
  • Standard chromium release assays are used to measure specific cytotoxic T lymphocyte (CTL) activity against the various IV antigens.
  • Tetramer assays are used to detect and quantify antigen specific T-cells, with quantification being confirmed and phenotypic characterization accomplished by intracellular cytokine staining.
  • IgG and IgA responses against the various IV antigens are analyzed by ELISA of vaginal washes.
  • In vivo gene delivery may be enhanced through the application of brief electrical pulses to injected tissues, a procedure referred to herein as electrically-assisted plasmid delivery.
  • electrically-assisted plasmid delivery See, e.g., Aihara, H. & Miyazaki, J. Nat. Biotechnol. 16:867-70 (1998); Mir, L. M. et al., Proc. Natl Acad. Sci. USA 96:4262-67 (1999); Hartikka, J. et al., Mol. Ther. 4:407-15 (2001); and Mir, L. M. et al.; Rizzuto, G. et al., Hum Gene Ther 11:1891-900 (2000); Widera, G.
  • the electroporation procedure can be performed with various electroporation devices. These devices include external plate type electrodes or invasive needle/rod electrodes and can possess two electrodes or multiple electrodes placed in an array. Distances between the plate or needle electrodes can vary depending upon the number of electrodes, size of target area and treatment subject.
  • the TriGrid needle array used in examples described herein, is a three electrode array comprising three elongate electrodes in the approximate shape of a geometric triangle. Needle arrays may include single, double, three, four, five, six or more needles arranged in various array formations. The electrodes are connected through conductive cables to a high voltage switching device that is connected to a power supply.
  • the electrode array is placed into the muscle tissue, around the site of nucleic acid injection, to a depth of approximately 3 mm to 3 cm.
  • the depth of insertion varys depending upon the target tissue and size of patient receiving electroporation.
  • square wave electrical pulses are applied to the tissue.
  • the amplitude of each pulse ranges from about 100 volts to about 1500 volts, e.g., about 100 volts, about 200 volts, about 300 volts, about 400 volts, about 500 volts, about 600 volts, about 700 volts, about 800 volts, about 900 volts, about 1000 volts, about 1100 volts, about 1200 volts, about 1300 volts, about 1400 volts, or about 1500 volts or about 1-1.5 kV/cm, based on the spacing between electrodes.
  • Each pulse has a duration of about 1 ⁇ s to about 1000 ⁇ s, e.g., about 1 ⁇ s, about 10 ⁇ s, about 50 ⁇ s, about 100 ⁇ s, about 200 ⁇ s, about 300 ⁇ s, about 400 ⁇ s, about 500 ⁇ s, about 600 ⁇ s, about 700 ⁇ s, about 800 ⁇ s, about 900 ⁇ s, or about 1000 ⁇ s, and a pulse frequency on the order of about 1-10 Hz.
  • the polarity of the pulses may be reversed during the electroporation procedure by switching the connectors to the pulse generator. Pulses are repeated multiple times.
  • the electroporation parameters e.g. voltage amplitude, duration of pulse, number of pulses, depth of electrode insertion and frequency
  • membrane stabilizing agents include, but are not limited to, steroids (e.g. dexamethasone, methylprednisone and progesterone), angiotensin II and vitamin E.
  • steroids e.g. dexamethasone, methylprednisone and progesterone
  • angiotensin II e.g. angiotensin II
  • vitamin E e.g. vitamin E
  • EAPD techniques such as electroporation can also be used for plasmids contained in liposome formulations.
  • the liposome—plasmid suspension is administered to the animal or patient and the site of injection is treated with a safe but effective electrical field generated, for example, by a TriGrid needle array.
  • the electroporation may aid in plasmid delivery to the cell by destabilizing the liposome bilayer so that membrane fusion between the liposome and the target cellular structure occurs.
  • Electroporation may also aid in plasmid delivery to the cell by triggering the release of the plasmid, in high concentrations, from the liposome at the surface of the target cell so that the plasmid is driven across the cell membrane by a concentration gradient via the pores created in the cell membrane as a result of the electroporation.
  • a carrier protein e.g., HBcAg
  • Electrodes are applied. Modified caliper electrodes are used to apply the electrical pulse. See Hartikka J. et al. Mol Ther 188:407-415 (2001). The caliper electrode plates are coated with conductivity gel and applied to the sides of the injected muscle before closing to a gap of 3 mm for administration of pulses. EAPD is applied using a square pulse type at 1-10 Hz with a field strength of 100-500 V/cm, 1-10 pulses, of 10-100 ms each.
  • mice are vaccinated ⁇ EAPD at 0, 2 and 4 weeks.
  • serum IgG titers against the various IV antigens are measured by ELISA and splenic T-cell responses are measured by antigen-specific production of IFN-gamma and IL-4 in ELISPOT assays.
  • Standard chromium release assays are used to measure specific cytotoxic T lymphocyte (CTL) activity against the various IV antigens.
  • Rabbits are given bilateral injections in the quadriceps muscle with plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, HA, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector.
  • the implantation area is shaved and the TriGrid electrode array is implanted into the target region of the muscle.
  • Electroporation begins approximately one minute after injection of the plasmid DNA is complete. Electroporation is administered with a TriGrid needle array, with eletrodes evenly spaced 7 mm apart, using an Ichor TGP-2 pulse generator. The array is inserted into the target muscle to a depth of about I to 2 cm. 4-8 pulses are administered. Each pulse has a duration of about 50-100 ⁇ s, an amplitude of about 1-1.2 kV/cm and a pulse frequency of 1 Hz. The injection and electroporation may be repeated.
  • Sera are collected from vaccinated rabbits at various time point. As endpoints, serum IgG titers against the various IV antigens are measured by ELISA and PBMC T-cell proliferative responses.
  • plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, (0.1 to 10 mg DNA total per animal).
  • a carrier protein e.g., HBcAg
  • various controls e.g., empty vector, (0.1 to 10 mg DNA total per animal).
  • Target muscle groups include, but are not limited to, bilateral rectus fermoris, cranial tibialis, biceps, gastrocenemius or deltoid muscles.
  • the target area is shaved and a needle array, comprising between 4 and 10 electrodes, spaced between 0.5-1.5 cm apart, is implanted into the target muscle.
  • a sequence of brief electrical pulses are applied to the electrodes implanted in the target muscle using an Ichor TGP-2 pulse generator.
  • the pulses have an amplitude of approximately 120 - 200V.
  • the pulse sequence is completed within one second. During this time, the target muscle may make brief contractions or twitches. The injection and electroporation may be repeated.
  • Sera are collected from vaccinated monkeys at various time points. As endpoints, serum IgG titers against the various IV antigens are measured by ELISA and PBMC T-cell proliferative responses are measured by antigen-specific production of IFN-gamma and IL-4 in ELISPOT assays or by tetramer assays to detect and quantify antigen specific T-cells, with quantification being confirmed and phenotypic characterization accomplished by intracellular cytokine staining. Standard chromium release assays are used to measure specific cytotoxic T lymphocyte (CTL) activity against the various TV antigens.
  • CTL cytotoxic T lymphocyte
  • This Example describes vaccination with a combinatorial formulation including one or more polynucleotides comprising one codon-optimized coding regions encoding an IV protein or fragment, variant, or derivative thereof prepared with an adjuvant and/or transfection facilitating agent; and also an isolated IV protein or fragment, variant, or derivative thereof.
  • antigen is provided in two forms.
  • the exogenous isolated protein stimulates antigen specific antibody and CD4+ T-cell responses, while the polynucleotide-encoded protein, produced as a result of cellular uptake and expression of the coding region, stimulates a CD8+ T-cell response.
  • this approach provides different forms of antigen in the same formulation. Because antigen expression from the DNA vaccine doesn't peak until 7-10 days after injection, the DNA vaccine provides a boost for the protein component.
  • the formulation takes advantage of the immunostimulatory properties of the bacterial plasmid DNA.
  • NP DNA formulated with cationic lipid
  • the DNA was diluted in 2 ⁇ PBS to 0.2 mg/ml ⁇ purified recombinant NP protein (produced in baculovirus as described in Example 2) at 0.08 mg/ml.
  • Each cationic lipid was reconstituted from a dried film by adding 1 ml of sterile water for injection (SWFI) to each vial and vortexing continuously for 2 min., then diluted with SWFI to a final concentration of 0.15 mM.
  • Equal volumes of NP DNA ( ⁇ NP protein) and cationic lipid were mixed to obtain a DNA to cationic lipid molar ratio of 4:1.
  • Ribi I was reconstituted with saline to twice the final concentration.
  • Ribi I (2 ⁇ ) was mixed with an equal volume of NP DNA at 0.2 mg/ml in saline ⁇ NP protein at 0.08 mg/ml.
  • NP DNA was prepared in 150 mM sodium phosphate buffer, pH 7.2.
  • groups of 9 BALB/c female mice at 7-9 weeks of age were injected with 50 ⁇ l of NP DNA ⁇ NP protein, cationic lipid or Ribi I. Injections were given bilaterally in each rectus femoris at day 0 and day 21. The mice were bled by OSP on day 20 and day 33 and serum titers of individual mice were measured.
  • NP specific serum antibody titers were determined by indirect binding ELISA using 96 well ELISA plates coated overnight at 4° C. with purified recombinant NP protein at 0.5 ⁇ g per well in BBS buffer pH 8.3. NP coated wells were blocked with 1% bovine serum albumin in BBS for 1 h at room temperature. Two-fold serial dilutions of sera in blocking buffer were incubated for 2 h at room temperature and detected by incubating with alkaline phosphatase conjugated (AP) goat anti-mouse IgG-Fc (Jackson Immunoresearch, West Grove, Pa.) at 1:5000 for 2 h at room temperature.
  • AP alkaline phosphatase conjugated
  • Color was developed with 1 mg/ml para-nitrophenyl phosphate (Calbiochem, La Jolla, Calif.) in 50 mM sodium bicarbonate buffer, pH 9.8 and 1 mM MgCl 2 and the absorbance read at 405 nm.
  • the titer is the reciprocal of the last dilution exhibiting an absorbance value 2 times that of pre-bleed samples.
  • Standard ELISPOT technology used to identify the number of interferon gamma (IFN-y) secreting cells after stimulation with specific antigen (spot forming cells per million splenocytes, expressed as SFU/million), was used for the CD4+ and CD8+ T-cell assays.
  • IFN-y interferon gamma
  • specific antigen spot forming cells per million splenocytes, expressed as SFU/million
  • CD4+ and CD8+ assays cells were serially diluted 3-fold, starting at 10 6 cells per well and transferred to 96 well ELISPOT plates pre-coated with anti-murine IFN- ⁇ monoclonal antibody.
  • Spleen cells were stimulated with the H-2K d binding peptide, TYQRTRALV (SEQ ID NO:81), at 1 ⁇ g/ml and recombinant murine IL-2 at 1 U/ml for the CD8+ assay and with purified recombinant NP protein at 20 ⁇ g/ml for the CD4+ assay. Cells were stimulated for 20-24 hours at 37° C.
  • mice receiving vaccine formulations containing purified protein were 6 to 8-fold higher than for mice receiving NP DNA only ( FIG. 5 , Table 15).
  • the titers for mice receiving DNA and protein formulated with a cationic lipid were similar to those for mice receiving protein in Ribi adjuvant or DNA and protein in Ribi adjuvant.
  • mice receiving vaccine formulations containing purified protein were 9 to 129-fold higher than for mice receiving NP DNA only ( FIG. 6 , Table 15).
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are used in the prime-boost compositions described herein.
  • a carrier protein e.g., HBcAg
  • various controls e.g., empty vector
  • the same protein may be used for the boost, e.g., DNA encoding NP with NP protein, or a heterologous boost may be used, e.g., DNA encoding NP with an M1 protein boost.
  • a heterologous boost may be used, e.g., DNA encoding NP with an M1 protein boost.
  • Each formulation, the plasmid comprising a coding region for the IV protein alone, or the plasmid comprising a coding region for the IV protein plus the isolated protein are formulated with Ribi I or the cationic lipids, DMRIE:DOPE or VaxfectinTM.
  • the formulations are prepared in the recommended buffer for that vaccine modality. Exemplary formulations, using NP as an example, are described herein.
  • plasmid/protein formulations can be easily prepared by one of ordinary skill in the art by following this example.
  • the DNA is diluted in 2 ⁇ PBS to 0.2 mg/ml ⁇ purified recombinant NP protein at 0.08 mg/ml.
  • Each cationic lipid is reconstituted from a dried film by adding 1 ml of sterile water for injection (SWFI) to each vial and vortexing continuously for 2 min., then diluted with SWFI to a final concentration of 0.15 mM.
  • Equal volumes of NP DNA ( ⁇ NP protein) and cationic lipid are mixed to obtain a DNA to cationic lipid molar ratio of 4:1.
  • Ribi I is reconstituted with saline to twice the final concentration.
  • Ribi I (2 ⁇ ) is mixed with an equal volume of NP DNA at 0.2 mg/ml in saline ⁇ NP protein at 0.08 mg/ml.
  • NP DNA is prepared in 150 mM sodium phosphate buffer, pH 7.2.
  • groups of 9 BALB/c female mice at 7-9 weeks of age are injected with 50 ⁇ l of NP DNA ⁇ NP protein, cationic lipid or Ribi I.
  • mice are bled on day 20 and day 33 and serum titers of individual mice to the various IV antigens are measured. Serum antibody titers specific for the various IV antigens are determined by ELISA. Standard ELISPOT technology, used to identify the number of interferon gamma (IFN- ⁇ ) secreting cells after stimulation with specific antigen (spot forming cells per million splenocytes, expressed as SFU/million), is used for the CD4+ and CD8+ T-cell assays using 3 mice from each group vaccinated above, sacrificed on day 34, 35 and 36, post vaccination.
  • IFN- ⁇ interferon gamma
  • a murine challenge model with influenza A virus is used to test the efficacy of the immunotherapies.
  • the model used is based on that described in Ulmer, J. B., et al., Science 259:1745-49 (1993) and Ulmer, J. B. et al., J Virol. 72:5648-53 (1998), both of which are incorporated herein by reference in their entireties.
  • This model utilizes a mouse-adapted strain of influenza A/HK/8/68 which replicates in mouse lungs and is titered in tissue culture in Madin Darby Canine Kidney cells.
  • the LD 90 of this mouse-adapted influenza virus is determined in female BALB/c mice age 13-15 weeks.
  • lethal challenge where the virus is administered intranasally to heavily sedated mice under ketamine anesthesia
  • sub-lethal challenge mice are not anesthetized when the viral inoculum is administered (also intranasally).
  • the endpoint for lethal challenge is survival, but loss in body mass and body temperature can also be monitored.
  • the read-outs for the sublethal challenge include lung virus titer and loss in body mass and body temperature.
  • mice are subjected to lethal challenge.
  • Mice that are previously vaccinated with DNA encoding IV antigens are anesthetized and challenged intranasally with 0.02 mL of mouse-adapted influenza A/HK/8/68 (mouse passage #6), diluted 1 to 10,000 (500 PFU) in PBS containing 0.2% wt/vol BSA.
  • mice The route of administration is intramuscular in rectus femoris (quadriceps), using 0.1 ⁇ g up to 1 mg total plasmid DNA. Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are tested singly and in multivalent cocktails for the ability to protect against challenge.
  • a carrier protein e.g., HBcAg
  • various controls e.g., empty vector
  • the plasmids are formulated with an adjuvant and/or a transfection facilitating agent, e.g., VaxfectinTM by methods described elsewhere herein.
  • Mice are vaccinated on days 0 and 21 using amounts of plasmids as described in Example 6. Subsequent injections can be administered. Nasal challenge of mice takes place 3 weeks after the final immunization, and animals are monitored daily for body mass, hypothermia, general appearance and then death.
  • VR10551 is an expression vector without any transgene insert.
  • VR4750 contains the coding sequence for hemagglutinin (HA) (H3N2) from mouse adapted A/Hong Kong/68.
  • the DNA was prepared using Qiagen plasmid purification kits.
  • HA hemagglutinin
  • mice were vaccinated twice at 3 week intervals with either 100 ⁇ g pDNA VR4750 encoding the HA gene cloned directly from the mouse-adapted influenza A/HK/8/68 strain, or with 100 ⁇ g blank vector pDNA (VR10551).
  • An additional control group was immunized intranasally with live A/HK/8/68 virus (500 PFU).
  • mice were challenged intranasally with mouse-adapted influenza A/HK/8168 with one of 3 doses (50, 500 and 5,000 PFU). Following viral challenge, mice were monitored daily for symptoms of disease, loss in body mass and survival.
  • FIG. 9 shows that homologous HA-pDNA vaccinated mice are completely protected over a range of viral challenge doses ( FIG. 9A ) and did not suffer significant weight loss ( FIG. 9B ) during the 3 week period following challenge.
  • future mouse flu challenge studies can include VR4750 (HA) pDNA as a positive control for protection and utilize 500 PFU, which is the LD90 for this mouse-adapted virus, as the challenge dose.
  • Rhesus or cynomologus monkeys (6/group) are vaccinated with plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, HA, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, intramuscularly 0.1 to 2 mg DNA combined with cationic lipid, and/or poloxamer and/or aluminum phosphate based or other adjuvants at 0, 1 and 4 months.
  • a carrier protein e.g., HBcAg
  • Plasma is drawn twice at baseline and then again at the time of and two weeks following each vaccination, and then again 4 months following the last vaccination.
  • Plasma is analyzed for humoral response and PBMCs are monitored for cellular responses, by standard methods described herein. Animals are monitored for 4 months following the final vaccination to determine the durability of the immune response.
  • Animals are challenged within 2-4 weeks following the final vaccination. Animals are challenged intratracheally with the suitable dose of virus based on preliminary challege studies. Nasal swabs, pharyngeal swabs and lung lavages are collected at days 0, 2, 4, 6, 8 and 11 post-challenge and will be assayed for cell-free virus titers on monkey kidney cells. After challenge, animals are monitored for clinical symptoms, e.g., rectal temperature, body weight, leukocyte counts, and in addition, hematocrit and respiratory rate. Oropharyngeal swab samples are taken to allow determination of the length of viral shedding. Illness is scored using the system developed by Berendt & Hall ( Infect Immun 16:476-479 (1977)), and will be analyzed by analysis of variance and the method of least significant difference.
  • various vaccine formulations of the present invention are tested in the chicken influenza model.
  • an IV H5N1 virus known to infect birds, is used.
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are formulated with cationic lipid, and/or poloxamer and/or aluminum phosphate based or other adjuvants.
  • the vaccine formulations are delivered at a dose of about 1-10 ⁇ g, delivered IM into the defeathered breast area, at 0 and 1 month.
  • the animals are bled for antibody results 3 weeks following the second vaccine.
  • Antibody titers against the various IV antigens are determined using techniques described in the literature. See, e.g., Kodihalli S. et al., Vaccine 18:2592-9 (2000).
  • the birds are challenged intranasally with 0.1 mL containing 100 LD 50 3 weeks post second vaccination.
  • the birds are monitored daily for 10 days for disease symptoms, which include loss of appetite, diarrhea, swollen faces, cyanosis, paralysis and death. Tracheal and cloacal swabs are taken 4 days following challenge for virus titration.
  • mice Groups of nine, six- to eight-week old BALB/c mice (Harlan-Sprague-Dawley) received bilateral (50 ⁇ L/leg) intramuscular (rectus femoris) injections of plasmid DNA. Control mice received DNA in PBS alone. Mice received injections on days 0, 20 and 49. Mice were bled by OSP on day 62, and NP-specific antibodies analyzed by ELISA. Splenocytes were harvested from 3 mice/group/day for three sequential days beginning day 63, and NP-specific specific T cells were analyzed by IFN ⁇ ELISPOT using overlapping peptide stimulation.
  • Splenocyte cultures were grown in RPMI-1640 medium containing 25 mM HEPES buffer and L-glutamine and supplemented with 10% (v/v) FBS, 55 ⁇ M ⁇ -mercaptoethanol, 100 U/mL of penicillin G sodium salt, and 100 ⁇ g/mL of streptomycin sulfate.
  • NP specific serum antibody titers were determined by indirect binding ELISA using 96 well ELISA plates coated overnight at 4° C. with purified recombinant NP protein at 0.5 ⁇ g per well in BBS buffer, pH 8.3. NP coated wells were blocked with 1% bovine serum albumin in BBS for 1 hour at room temperature. Two-fold serial dilutions of sera in blocking buffer were incubated for 2 hours at room temperature and detected by incubating with alkaline phosphatase conjugated (AP) goat anti-mouse IgG-Fc (Jackson Immunoresearch, West Grove, Pa.) at 1:5000 for 2 hours at room temperature.
  • AP alkaline phosphatase conjugated
  • Color was developed with 1 mg/ml para-nitrophenyl phosphate (Calbiochem, La Jolla, Calif.) in 50 mM sodium bicarbonate buffer, pH 9.8 and 1 mM MgCl 2 and the absorbance read at 405 nm.
  • the titer is the reciprocal of the last dilution exhibiting an absorbance value 2 times that of pre-bleed samples.
  • Standard ELISPOT technology used to identify the number of interferon gamma (IFN- ⁇ ) secreting cells after stimulation with specific antigen (spot forming cells per million splenocytes, expressed as SFU/million), was used for the CD4+ and CD8+ T-cell assays.
  • IFN- ⁇ interferon gamma
  • Three mice from each group were sacrificed on each of three consecutive days. At the time of collection, spleens from each group were pooled, and single cell suspensions were made in cell culture media using a dounce homogenizer. Red blood cells were lysed, and cells were washed and counted.
  • CD4+ and CD8+ assays cells were serially diluted 3- fold, starting at 10 6 cells per well and transferred to 96 well ELISPOT plates pre-coated with anti-murine IFN- ⁇ monoclonal antibody.
  • Spleen cells were stimulated with the H-2K d binding peptide, TYQRTRALV, at 1 ⁇ g/ml and recombinant murine IL-2 at 1 U/ml for the CD8+ assay and with purified recombinant NP protein at 20 ⁇ g/ml for the CD4+ assay. Cells were stimulated for 20-24 hours at 37° C.
  • VR4700 is a plasmid encoding influenza A/PR/8/34 nucleoprotein (NP) in a VR10551 backbone.
  • VR10551 is an expression vector without any transgene insert.
  • VF-P1205-02A is a formulation containing a poloxamer with a POP molecular weight of 12 KDa and POE of 5% (CRL1005) at a DNA:poloxamer:BAK ratio of 5 mg/ml:7.5 mg/ml:0.3 mM.
  • the purpose of this experiment was first to compare immune responses of DMRIE:DOPE (1:1, mol:mol) at pDNA/cationic lipid molar ratios of 4:1 as an MLV (multi lamellar vesicle formulation—multi-vial) or SUV (small unilamellar vesicles—single-vial) formulation. Second, it was to compare sucrose (lyophilized and frozen) and PBS based formulations.
  • Polynucleotides of the present invention were inserted into eukaryotic expression vector backbones VR10551, VR10682 and VR6430 all of which are described previously.
  • the VR10551 vector is built on a modified pUC18 background (see Yanisch-Perron, C., et al. Gene 33:103-119 (1985)), and contains a kanamycin resistance gene, the human cytomegalovirus immediate early 1 promoter/enhancer and intron A, and the bovine growth hormone transcription termination signal, and a polylinker for inserting foreign genes. See Hartikka, J., et al., Hum. Gene Ther. 7:1205-1217 (1996).
  • eukaryotic expression vectors may be used in the present invention, including, but not limited to: plasmids pcDNA3, pHCMV/Zeo, pCR3.1, pEF1/His, pIND/GS, pRc/HCMV2, pSV40/Zeo2, pTRACER-HCMV, pUB6/V5-His, pVAX1, and pZeoSV2 (available from Invitrogen, San Diego, Calif.), and plasmid pCI (available from Promega, Madison, Wis.).
  • segment 7 encodes both M1 and M2 proteins via differential splicing
  • M2 and NP into expression constructions as described below and pictured in FIG. 13 .
  • Plasmids VR4756 (SEQ ID NO:91), VR4759 (SEQ ID NO:92) and VR4762 (SEQ ID NO:93) were created by cloning the nucleotide sequence encoding the consensus sequence for the following influenza A antigens respectively: segment 7 (encoding both the M1 and M2 proteins by differential splicing), M2 and NP into the VR10551 backbone.
  • segment 7 encoding both the M1 and M2 proteins by differential splicing
  • M2 and NP into the VR10551 backbone.
  • the VR4756, VR4759 and VR4762 plasmids are also described in Table 13.
  • the VR4764 (SEQ ID NO:95) and VR4765 (SEQ ID NO:96) plasmids were constructed by ligating the segment 7 and NP coding regions from VR4756 and VR4762 respectively into the VR10682 vector.
  • the VR4756 vector was digested with EcoRV and SalI restriction endonucleases and the blunted fragment was ligated into the VR10682 backbone, which had been digested with the EcoRV restriction endonuclease.
  • the VR4765 vector was constructed by digesting the VR4762 vector with EcoRV and NotI and ligating the NP coding region into the VR10682 backbone digested with the same restriction endonucleases.
  • VR4766 (SEQ ID NO:97) and VR4767 (SEQ ID NO:98) contain a CMV promoter/intron A-NP expression cassette and a RSV promoter (from VCL1005)-segment 7 expression cassette in the same orientation (VR4766) or opposite orientation (VR4767).
  • These plasmids were generated by digesting VR4762 with the DraIII restriction endonuclease and cutting the RSV-segment 7-mRBG cassette from VR4764 with EcoRV and BamHI restriction endonucleases. After exonuclease digestion with the Klenow fragment of DNA polymerase I, the the EcoRV/BamHI fragment was cloned into the DraIII digested VR4762 vector. Both insert orientations were obtained by this blunt end cloning method.
  • VR4768 (SEQ ID NO:99) and VR4769 (SEQ ID NO:100), containing a CMV promoter/intron A-segment 7 expression cassette and a RSV promoter-NP expression cassette, were similarly derived.
  • VR4756 was digested with the DraIII restriction endonuclease and blunted by treatment with the Klenow fragment of DNA Polymerase I.
  • the cassette containing the RSV promoter, NP coding region and mRBG terminator was removed from VR4765 by digesting with KpnI and NdeI restriction endonucleases. The fragment was also blunted with the Klenow fragment of DNA polymerase I and ligated into the DraIII-digested VR4756 vector in both gene orientations.
  • VR4770 (SEQ ID NO:101), VR4771 (SEQ ID NO:102) and VR4772 (SEQ ID NO:103) were constructed by cloning the coding regions from VR4756, VR4762 and VR4759 respectively into the VR6430 vector backbone.
  • the segment 7 gene from VR4756 was removed using SalI and EcoRV restriction endonucleases and blunted with the Klenow fragment of DNA polymerase I.
  • the VR6430 plasmid was digested with EcoRV and BamHI and the vector backbone fragment was blunted with the Klenow fragment of DNA polymerase I.
  • the segment 7 gene fragment was then ligated into the VR6430 vector backbone.
  • VR4771 was derived by removing the NP insert from VR4762 following EcoRV and BglII restriction endonuclease digestion and the fragment was ligated into the VR6430 vector backbone which had been digested the same restriction endonucleases.
  • VR4772 was derived by subcloning the M2 coding region from VR4759 as a blunted SalI-EcoRV fragment and ligating into the VR6430 vector backbone from a blunted EcoRV-BamHI digest.
  • VR4773 (SEQ ID NO:104) and VR4774 (SEQ ID NO:105) contain a CMV promoter/intron A-segment 7 expression cassette and a RSV/R-NP expression cassette with the genes in the same or opposite orientation. These plasmids were generated by digesting VR4756 with the DraIII restriction endonuclease, blunting, and ligating to the RSV/R-NP-BGH fragment from VR4771 (VR4771 digested with NdeI and SfiI and then blunted).
  • VR4775 (SEQ ID NO:106) and VR4776 (SEQ ID NO:107) contain a CMV promoter/intron A-NP expression cassette and a RSV/R-segment 7 expression cassette with the genes in the same or opposite orientation. These plasmids were generated by digesting VR4762 with the DraIII restriction enzyme and blunting with the Klenow fragment of DNA polymerase. The RSV/R-segment 7-BGH fragment was generated by digesting VR4770 with NdeI and SfiI restriction endonucleases and ligating the blunted fragment with the DraIII restriction endonuclease digested VR4762.
  • VR4777 (SEQ ID NO:108) and VR4778 (SEQ ID NO:109) contain a CMV promoter/intron A-NP expression cassette and a RSV/R-M2 expression cassette in the same or opposite orientation.
  • These plasmids were generated by digesting VR4762 with the MscI restriction endonuclease, digesting VR4772 with NdeI and SfiI restriction endonucleases and treating the RSV/R-M2-BGH with the Klenow fragment of DNA polymerase, followed by ligation of these two gel purified fragments.
  • VR4779 and VR4780 contain a CMV promoter/intron A-M2 expression cassette and a RSV/R-NP expression cassette in the same or opposite orientation. These plasmids were generated by digesting VR4759 with the MscI restriction endonuclease, digesting VR4771 with NdeI and SfiI restriction endonucleases and treating the RSV/R-NP-BGH segment with the Klenow fragment of DNA polymerase, followed by ligation of these two gel purified fragments.
  • Plasmid DNA was transformed into Escherichia coli DH5 ⁇ competent cells, and highly purified covalently closed circular plasmid DNA was isolated by a modified lysis procedure (Horn, N. A., et al., Hum. Gene Ther. 6:565-573 (1995)) followed by standard double CsCl-ethidium bromide gradient ultracentrifugation (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y. (1989)).
  • plasmid preparations were free of detectable chromosomal DNA, RNA and protein impurities based on gel analysis and the bicinchoninic protein assay (Pierce Chem. Co., Rockford Ill.). Endotoxin levels were measured using Limulus Amebocyte Lysate assay (LAL, Associates of Cape Cod, Falmouth, Mass.) and were less than 0.6 Endotoxin Units/mg of plasmid DNA. The spectrophotometric A 260 /A 280 ratios of the DNA solutions were typically above 1.8.
  • Plasmids were ethanol precipitated and resuspended in an appropriate solution, e.g., 150 mM sodium phosphate (for other appropriate excipients and auxiliary agents, see U.S. patent application Publication 2002/0019358, published Feb. 14, 2002). DNA was stored at ⁇ 20° C. until use. DNA was diluted by mixing it with 300 mM salt solutions and by adding appropriate amount of USP water to obtain 1 mg/ml plasmid DNA in the desired salt at the desired molar concentration.
  • an appropriate solution e.g. 150 mM sodium phosphate (for other appropriate excipients and auxiliary agents, see U.S. patent application Publication 2002/0019358, published Feb. 14, 2002).
  • DNA was stored at ⁇ 20° C. until use. DNA was diluted by mixing it with 300 mM salt solutions and by adding appropriate amount of USP water to obtain 1 mg/ml plasmid DNA in the desired salt at the desired molar concentration.
  • the expression plasmids were analyzed in vitro by transfecting the plasmids into a well characterized mouse melanoma cell line (VM-92, also known as UM-449) and the human rhabdomyosarcoma cell line RD (ATCC CCL-136) both available from the American Type Culture Collection, Manassas, Va.
  • VM-92 mouse melanoma cell line
  • RD human rhabdomyosarcoma cell line
  • Other well-characterized human cell lines may also be used, e.g. MRC-5 cells, ATCC Accession No. CCL-171.
  • the transfection was performed using cationic lipid-based transfection procedures well known to those of skill in the art. Other transfection procedures are well known in the art and may be used, for example electroporation and calcium chloride-mediated transfection (Graham F. L. and A. J.
  • eM2-NP full-length NP
  • One nucleotide sequence was derived from the above codon optimization algorithm, while the other was done by an outside vendor. Comparison of expression levels from the two eM2-NP pDNAs was measured in vitro, and comparison of immunogenicity in vivo is on-going. Additionally, the full-length NP genes for both codon-optimized versions were sub-cloned from the eM2-NP pDNAs and analyzed for expression in vitro.
  • NP consensus protein expression in vitro was also detected using a goat polyclonal antibody (Fitzgerald) generated against whole H1N1 or H3N2 virus (data not shown). Expression levels of both of these NP constructs were much higher than a pDNA containing A/PR/34 NP (VR4700).
  • Influenza antigen-encoding plasmids were transfected into VM92 cells using methods described above. Cell lysates and media were collected 48 hours after transfection. Cells were lysed in 200 ⁇ l of Laemmli buffer, cell debris removed by microcentrifuge spin, and 20 ⁇ l was heated and loaded on a 4-12% Bis-Tris gel. To determine expression of those vectors encoding secreted NP protein, 15 ⁇ l of media was mixed with 5 ⁇ l of loading buffer, heated, and loaded on a gel. Western blots were processed as described above.
  • Primary antibodies were as follows: monoclonal antibody MA1-082 (ABR) to detect M2 protein, monoclonal antibody MCA401 (Serotec) to detect M1 protein, and a polyclonal antibody against VR4762-injected rabbits generated in-house. All primary antibodies were used at a 1:500 dilution.
  • FIG. 14 shows Western blot results wherein M2 protein expression from segment 7-enocoding plasmids are higher in CMV promoter/intron A-segment 7 (VR4756) and RSV/R-segment 7 (VR4770) than VR4764 (RSV promoter).
  • NP expression appeared highest from the RSV/R-NP plasmid (VR4771), followed by CMV/intron A-NP (VR4762) and then RSV-NP (VR4765). Similar results were seen in Western blots from human RD-transfected cells.
  • M2 expression from segment 7 is very low, independent of orientation.
  • the CMV/intron A-NP expression in these dual promoter plasmids does not differ significantly compared to VR4762.
  • RSV-NP expression in dual promoter plasmids (VR4768 and VR4769), where segment 7 is expressed from CMV/intron A, NP expression decreases somewhat, but not as drastically as M2 expression in the dual promoter VR4766 and VR4767.
  • FIG. 15 shows expression of the M1 and M2 proteins from segment 7, as well as NP, from CMV promoter/intron A, RSV promoter, and RSV/R-containing plasmids.
  • dual promoter plasmids contain the CMV promoter/intron A and RSV/R driving either NP or segment 7. Similar results were seen in Western blots from human RD-transfected cells.
  • a model influenza A challenge model has been established utilizing a mouse-adapted A/BK/8/68 strain.
  • Positive and negative control Hemagluttinin (HA)-containing plasmids were generated by PCR of the HA genes directly from mouse-adapted A/Hong Kong/68 (H3N2) and A/Puerto Rico/34 (H1N1) viruses, respectively.
  • plasmid DNA vaccinations are given as bilateral, rectus femoris injections at 0 and 3 weeks, followed by orbital sinus puncture (OSP) bleed at 5 weeks and intranasal viral challenge at 6 weeks with 500 pfu (1 LD 90 ) of virus. Mice are monitored for morbidity and weight loss for about 3 weeks following viral challenge. Endpoint antibody titers for NP and M2 were determined by ELISA. For study GSJ08, 5 additional mice per test group were vaccinated and interferon- ⁇ ELISPOT assays were performed at week number 5.
  • OSP orbital sinus puncture
  • mice received 100 ⁇ g of each plasmid formulated in poloxamer CRL1005, 02A formulation.
  • the performance criteria for this study was survival of >90% for the positive controls, ⁇ 10% for the negative controls, and >75% for the experimental groups.
  • Table 21 shows that all of the control groups, as well as two experimental groups met the performance criteria.
  • the M2+NP and S7+NP plamsid DNA combinations resulted in 100% and 75% survival, respectively. There was no statistically significant difference (p ⁇ 0.05) between the two lead plasmid combinations, but there was statistical significance in the S7, S7+NP, and M2+NP groups vs. the negative controls.
  • Weight loss data showed that the positive control groups did not exhibit any weight loss following viral challenge, as opposed to the weight loss seen in all of the experimental groups. Mice that survived the viral challenge recovered to their starting weight by the end of the study. Tables 22 and 23 show endpoint antibody titers for test groups containing M2, Segment 7, and NP antigens. Shaded boxes represent mice that died following viral challenge.
  • NP Antibody Titers Group A Group E Group F Group G mouse (NP) (NP + M2) (NP + M1) (NP + seg7) 1 204800 51200 25600 2 51200 204800 51200 3 204800 51200 51200 4 25600 51200 5 102400 6 51200 102400 7 204800 102400 8 102400 102400 9 102400 51200 10 102400 102400 11 51200 25600 12 51200 25600 Study GSJ05:
  • mice were injected with 100 ⁇ g, 30 ⁇ g, or 10 ⁇ g per plasmid in the 02A poloxamer formulation at 0 and 3 weeks, followed by bleed at 5 weeks and viral challenge at 6 weeks. Sixteen mice per group were vaccinated for test groups A-H, while 12 mice per group were vaccinated for the controls. Poloxamer 02A-formulated HA plasmids, VR4750 (HA H3) and VR4752 (HA H1), were included as positive and negative controls, respectively.
  • the plasmid combination VR4759 (M2) and VR4762 (NP) was utilized in further mouse influenza challenge studies to examine additional formulations.
  • Poloxamer 02A and VaxfectinTM-formulated plasmid DNA led to 92% and 100% survival at the 20 ⁇ g pDNA dose, and 58% and 75% at the 2 ⁇ g dose, respectively (Table 27).
  • mice per test group were challenged with influenza virus at week number 6. Five mice per test group were sacrificed at days 36-38 for T cell assays (IFN- ⁇ ELISPOT).
  • the test groups and 21 day survival post-challenge are shown in Table 31. Groups A-D, and F-G were vaccinated with 20 ⁇ g total plasmid DNA per injection to further explore the weight loss/recovery phenomena seen in study GSJ06 with the VaxfectinTM-formulated pDNA.
  • the DMRIE:DOPE and VaxfectinTM formulated groups resulted in 92-100% survival at a 20 ⁇ g pDNA dose.
  • Group A Polyxaamer 02A
  • Group G Group G
  • the VaxfectinTM and DMIRE:DOPE Groups were shown to be statistically superior (p ⁇ 0.05) as compared to the negative control. Therefore, the plasmid DNA formulated with lipids appear to provide superior protection in the mouse influenza model challenge.
  • T cell responses as measured by IFN- ⁇ ELISPOT assay, were conducted on the last 5 mice per group using an M2 peptide encompassing the first 24 amino acids of M2 (TABLE 33), an NP protein expressed in baculovirus (TABLE 34), and an NP CD8+ Balb/c immunodominant peptide (TABLE 35).
  • Antibody titers Tables 36 and 37, were determined for M2 and NP proteins. The first 12 mice listed for each group were challenge at day 42 and the last 5 mice per group were sacrificed for IFN- ⁇ ELISPOT. The shaded boxes represent mice that died following viral challenge.

Abstract

The present invention is directed to enhancing the immune response of a human in need of protection against IV infection by administering in vivo, into a tissue of the human, at least one polynucleotide comprising one or more regions of nucleic acid encoding an IV protein or a fragment, a variant, or a derivative thereof. The present invention is further directed to enhancing the immune response of a human in need of protection against IV infection by administering, in vivo, into a tissue of the human, at least one IV protein or a fragment, a variant, or derivative thereof. The IV protein can be, for example, in purified form or can be an inactivated IV, such as those present in inactivated IV vaccines. The polynucleotide is incorporated into the cells of the human in vivo, and an immunologically effective amount of an immunogenic epitope of an IV, or a fragment, variant, or derivative thereof is produced in vivo. The IV protein (in purified form or in the form of an inactivated IV vaccine) is also administered in an immunologically effective amount.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application claims the benefit of the filing date of U.S. Provisional Application No. 60/571,854 filed May 18, 2004, which is incorporated herein by reference in its entirety.
  • REFERENCE TO A SEQUENCE LISTING SUBMITTED ON A COMPACT DISC
  • This application includes a “Sequence Listing,” which is provided as an electronic document on a compact disk (CD-R). This compact disk contains the file “Sequence Listing.txt” (340,000 bytes, created on May 18, 2005), which is hereby incorporated by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • The present invention relates to influenza virus vaccine compositions and methods of treating or preventing influenza infection and disease in mammals. Influenza is an acute febrile illness caused by infection of the respiratory tract. There are three types of influenza viruses: A, B, and C “IAV,” “IBV” or “IAC,” respectively, or generally “IV”. Type A, which includes several subtypes, causes widespread epidemics and global pandemics such as those that occurred in 1918, 1957 and 1968. Type B causes regional epidemics. Type C causes sporadic cases and minor, local outbreaks. These virus types are distinguished in part on the basis of differences in two structural proteins, the nucleoprotein, found in the center of the virus, and the matrix protein, which forms the viral shell.
  • The disease can cause significant systemic symptoms, severe illness requiring hospitalization (such as viral pneumonia), and complications such as secondary bacterial pneumonia. More than 20 million people died during the pandemic flu season of 1918/1919, the largest pandemic of the 20th century. Recent epidemics in the United States are believed to have resulted in greater than 10,000 (up to 40,000) excess deaths per year and 5,000-10,000 deaths per year in non-epidemic years.
  • The best strategy for prevention of morbidity and mortality associated with influenza is vaccination. Vaccination is especially recommended for people in high-risk groups, such as residents of nursing or residential homes, as well as for diabetes, chronic renal failure, or chronic respiratory conditions.
  • Traditional methods of producing influenza vaccines involve growth of an isolated strain in embryonated hens' eggs. Initially, the virus is recovered from a throat swab or similar source and isolated in eggs. The initial isolation in egg is difficult, but the virus adapts to its egg host and subsequent propagation in eggs takes place relatively easily. It is widely recognized, however, that the egg-derived production of IV for vaccine purposes has several disadvantages. One disadvantage is that such production process is rather vulnerable due to the varying (micro)biological quality of the eggs. Another disadvantage is that the process completely lacks flexibility if demand suddenly increases, i.e., in case of a serious epidemic or pandemic, because of the logistical problems due to the non-availability of large quantities of suitable eggs. Also, vaccines thus produced are contra-indicated for persons with a known hypersensitivity to chicken and/or egg proteins.
  • The influenza vaccines currently in use are designated whole virus (WV) vaccine or subvirion (SV) (also called “split” or “purified surface antigen”). The WV vaccine contains intact, inactivated virus, whereas the SV vaccine contains purified virus disrupted with detergents that solubilize the lipid-containing viral envelope, followed by chemical inactivation of residual virus. Attenuated viral vaccines against influenza are also in development. A discussion of methods of preparing conventional vaccine may be found in Wright, P. F. & Webster, R. G., FIELDS VIROLOGY, 4d Ed. (Knipe, D. M. et al. Ed.), 1464-65 (2001), for example.
  • Virus Structures
  • An IV is roughly spherical, but it can also be elongated or irregularly shaped. Inside the virus, eight segments of single-stranded RNA contain the genetic instructions for making the virus. The most striking feature of the virus is a layer of spikes projecting outward over its surface. There are two different types of spikes: one is composed of the molecule hemagglutinin (HA), the other of neuraminidase (NA). The HA molecule allows the virus to “stick” to a cell, initiating infection. The NA molecule allows newly formed viruses to exit their host cell without sticking to the cell surface or to each other. The viral capsid is comprised of viral ribonucleic acid and several so called “internal” proteins (polymerases (PB1, PB2, and PA, matrix protein (M1) and nucleoprotein (NP)). Because antibodies against HA and NA have traditionally proved the most effective in fighting infection, much research has focused on the structure, function, and genetic variation of those molecules. Researchers are also interested in a two non-structural proteins M2 and NS1; both molecules play important roles in viral infection.
  • Type A subtypes are described by a nomenclature system that includes the geographic site of discovery, a lab identification number, the year of discovery, and in parentheses the type of HA and NA it possesses, for example, A/Hong Kong/156/97 (H5N1). If the virus infects non-humans, the host species is included before the geographical site, as in A/Chicken/Hong Kong/G9/97 (H9N2).
  • Virions contain 7 segments (influenza C virus) to 8 segments (influenza A and B virus) of linear negative-sense single stranded RNA. Most of the segments of the virus genome code for a single protein. For many influenza viruses, the whole genome is now known. Genetic reassortment of the virus results from intermixing of the parental gene segments in the progeny of the viruses when a cell is co-infected by two different viruses of a given type. This phenomenon is facilitated by the segmental nature of the genome of influenza virus. Genetic reassortment is manifested as sudden changes in the viral surface antigens.
  • Antigenic changes in HA and NA allow the influenza virus to have tremendous variability. Antigenic drift is the term used to indicate minor antigenic variations in HA and NA of the influenza virus from the original parent virus, while major changes in HA and NA which make the new virions significantly different, are called Antigenic shift. The difference between the two phenomena is a matter of degree.
  • Antigenic drift (minor changes) occurs due to accumulation of point mutations in the gene which results in changes in the amino acids in the proteins. Changes which are extreme, and drastic (too drastic to be explained by mutation alone) result in antigenic shift of the virus. The segmented genomes of the influenza viruses reassort readily in double infected cells. Genetic reassortment between human and non-human influenza virus has been suggested as a mechanism for antigenic shift. Influenza is a zoonotic disease, and an important pathogen in a number of animal species, including swine, horses, and birds, both wild and domestic. Influenza viruses are transferred to humans from other species.
  • Because of antigenic shift and antigenic drift, immunity to an IV carrying a particular HA and/or NA protein does not necessarily confer protective immunity against IV strains carrying variant, or different HA and/or NA proteins. Because antibodies against HA and NA have traditionally proved the most effective in fighting IV infection, much research has focused on the structure, function and genetic variation of those molecules.
  • Recent IV Vaccine Candidates
  • During the past few years, there has been substantial interest in testing DNA-based vaccines for a number of infectious diseases where the need for a vaccine, or an improved vaccine, exists. Several well-recognized advantages of DNA-based vaccines include the speed, ease and cost of manufacture, the versatility of developing and testing multivalent vaccines, the finding that DNA vaccines can produce a robust cellular response in a wide variety of animal models as well as in humans, and the proven safety of using plasmid DNA as a delivery vector (Donnelly, J. J., et al., Annu. Rev. Immunol. 15:617-648 (1997); Manickan, E., et al., Crit. Rev. Immunol. 17(2):139-154 (1997); U.S. Pat. No. 6,214,804). DNA vaccines represent the next generation in the development of vaccines (Nossal, G., Nat. Med. 4(5 Supple):475-476 (1998)) and numerous DNA vaccines are in clinical trials. The above references are herein incorporated by reference in their entireties.
  • Studies have already been performed using DNA-based vaccines in animals. Ulmer, J. B. et al., Science 259:1745-9 (1993) revealed that mice could be protected by an IV nucleoprotein DNA vaccine alone against severe disease and death resulting from either a homologous or a heterologous IV challenge. Further studies have substantiated this model, and comparative studies of live influenza vaccines versus DNA influenza vaccines show them to be relatively equivalent in immune induction and protection in the murine model.
  • WO 94/21797, incorporated herein by reference in its entirety, discloses IV vaccine compositions comprising DNA constructs encoding NP, HA, M1, PB1 and NS1. WO 94/21797 also discloses methods of protecting against IV infection comprising immunization with a prophylactically effective amount of these DNA vaccine compositions.
  • The IV nucleoprotein is relatively conserved (see Shu, L. L. et al., J. Virol. 67:2723-9 (1993)), but just as conserved are the M1 matrix protein (which is a major T-cell target), and the M2 protein, which are encoded by separate reading frames of RNA segment 7. See Neirynck, S. et al., Nat. Med. 5:1157-63 (1999); Lamb, R. A. & Lai, C. J., Virology 112:746-51 (1981); Ito, T. et al., J. Virol. 65:5491-8 (1991). Animal DNA vaccine trials have been performed with DNA constructs encoding these genes alone or in combination, usually with success. See Okuda, K., et al., Vaccine 19:3681-91 (2001); Watabe, S. et al., Vaccine 19:4434-44 (2001). Of interest, the M2 protein is involved as part of an ion channel, is critical in resistance to the antiviral agents amantadine and rimantidine, and approximately 24 amino acids are extracellular (eM2). See Fischer, W. B., Biochim Biophys Acta 1561:27-45 (2002); Zhong, Q., FEBS Lett 434:265-71 (1998). Antibodies to this extracellular, highly conserved protein (eM2), which is highly expressed in infected cells (Lamb, R. A., et al., Cell 40:627-33 (1985)), have been shown to be involved in animal models. Treanor, J. J., J. Virol. 64:1375-7 (1990); Slepushkin, V. A. et al., Vaccine 13:1399-402 (1995). An approach using a conjugate hepatitis B core-eM2 protein has been evaluated in an animal model and proposed as a pandemic influenza vaccine. Neirynck, S. et al., Nat. Med. 5:1157-63 (1999). However, in one study vaccination of pigs with a DNA construct expressing eM2-NP fusion protein exacerbated disease after challenge with influenza A virus. Heinen, P. P., J. Gen. Virol. 83:1851-59 (2002). All of the above references are herein incorporated by reference in their entireties.
  • Heterologous “prime boost” strategies have been effective for enhancing immune responses and protection against numerous pathogens. Schneider et al., Immunol. Rev. 170:29-38 (1999); Robinson, H. L., Nat. Rev. Immunol. 2:239-50 (2002); Gonzalo, R. M. et al., Vaccine 20:1226-31 (2002); Tanghe, A., Infect. Immun. 69:3041-7 (2001). Providing antigen in different forms in the prime and the boost injections appears to maximize the immune response to the antigen. DNA vaccine priming followed by boosting with protein in adjuvant or by viral vector delivery of DNA encoding antigen appears to be the most effective way of improving antigen specific antibody and CD4+ T-cell responses or CD8+ T-cell responses respectively. Shiver J. W. et al., Nature 415: 331-5 (2002); Gilbert, S. C. et al., Vaccine 20:1039-45 (2002); Billaut-Mulot, O. et al., Vaccine 19:95-102 (2000); Sin, J. I. et al., DNA Cell Biol. 18:771-9 (1999). Recent data from monkey vaccination studies suggests that adding CRL1005 poloxamer (12 kDa, 5% POE), to DNA encoding the HIV gag antigen enhances T-cell responses when monkeys are vaccinated with an HIV gag DNA prime followed by a boost with an adenoviral vector expressing HIV gag (Ad5-gag). The cellular immune responses for a DNA/poloxamer prime followed by an Ad5-gag boost were greater than the responses induced with a DNA (without poloxamer) prime followed by Ad5-gag boost or for Ad5-gag only. Shiver, J. W. et al. Nature 415:331-5 (2002). U.S. patent application Publication No. US 2002/0165172 A1 describes simultaneous administration of a vector construct encoding an immunogenic portion of an antigen and a protein comprising the immunogenic portion of an antigen such that an immune response is generated. The document is limited to hepatitis B antigens and HIV antigens. Moreover, U.S. Pat. No. 6,500,432 is directed to methods of enhancing an immune response of nucleic acid vaccination by simultaneous administration of a polynucleotide and polypeptide of interest. According to the patent, simultaneous administration means administration of the polynucleotide and the polypeptide during the same immune response, preferably within 0-10 or 3-7 days of each other. The antigens contemplated by the patent include, among others, those of Hepatitis (all forms), HSV, HIV, CMV, EBV, RSV, VZV, HPV, polio, influenza, parasites (e.g., from the genus Plasmodium), and pathogenic bacteria (including but not limited to M. tuberculosis, M. leprae, Chlamydia, Shigella, B. burgdorferi, enterotoxigenic E. coli, S. typhosa, H. pylori, V. cholerae, B. pertussis, etc.). All of the above references are herein incorporated by reference in their entireties.
  • SUMMARY OF THE INVENTION
  • The present invention is directed to enhancing the immune response of a vertebrate in need of protection against IV infection by administering in vivo, into a tissue of the vertebrate, at least one polynucleotide, wherein the polynucleotide comprises one or more nucleic acid fragments, where the one or more nucleic acid fragments are optionally fragments of codon-optimized coding regions operably encoding one or more IV polypeptides, or fragments, variants, or derivatives thereof. The present invention is further directed to enhancing the immune response of a vertebrate in need of protection against IV infection by administering, in vivo, into a tissue of the vertebrate, a polynucleotide described above plus at least one isolated IV polypeptide or a fragment, a variant, or derivative thereof. The isolated IV polypeptide can be, for example, a purified subunit, a recombinant protein, a viral vector expressing an isolated IV polypeptide, or can be an inactivated or attentuated IV, such as those present in conventional IV vaccines. According to either method, the polynucleotide is incorporated into the cells of the vertebrate in vivo, and an immunologically effective amount of an immunogenic epitope of the encoded IV polypeptide, or a fragment, variant, or derivative thereof, is produced in vivo. When utilized, an isolated IV polypeptide or a fragment, variant, or derivative thereof is also administered in an immunologically effective amount.
  • According to the present invention, the polynucleotide can be administered either prior to, at the same time (simultaneously), or subsequent to the administration of the isolated IV polypeptide. The IV polypeptide or fragment, variant, or derivative thereof encoded by the polynucleotide comprises at least one immunogenic epitope capable of eliciting an immune response to influenza virus in a vertebrate. In addition, an isolated IV polypeptide or fragment, variant, or derivative thereof, when used, comprises at least one immunogenic epitope capable of eliciting an immune response in a vertebrate. The IV polypeptide or fragment, variant, or derivative thereof encoded by the polynucleotide can, but need not, be the same protein or fragment, variant, or derivative thereof as the isolated IV polypeptide which can be administered according to the method.
  • The polynucleotide of the invention can comprise a nucleic acid fragment, where the nucleic acid fragment is a fragment of a codon-optimized coding region operably encoding any IV polypeptide or fragment, variant, or derivative thereof, including, but not limited to, HA, NA, NP, M1 or M2 proteins or fragments (e.g., eM2), variants or derivatives thereof. A polynucleotide of the invention can also encode a derivative fusion protein, wherein two or more nucleic acid fragments, at least one of which encodes an IV polypeptide or fragment, variant, or derivative thereof, are joined in frame to encode a single polypeptide, e.g., NP fused to eM2. Additionally, a polynucleotide of the invention can further comprise a heterologous nucleic acid or nucleic acid fragment. Such heterologous nucleic acid or nucleic acid fragment may encode a heterologous polypeptide fused in frame with the polynucleotide encoding the IV polypeptide, e.g., a hepatitis B core protein or a secretory signal peptide. Preferably, the polynucleotide encodes an IV polypeptide or fragment, variant, or derivative thereof comprising at least one immunogenic epitope of IV, wherein the epitope elicits a B-cell (antibody) response, a T-cell (e.g., CTL) response, or both.
  • Similarly, the isolated IV polypeptide or fragment, variant, or derivative thereof to be delivered (either a recombinant protein, a purified subunit, or viral vector expressing an isolated IV polypeptide, or in the form of an inactivated IV vaccine) can be any isolated IV polypeptide or fragment, variant, or derivative thereof, including but not limited to the HA, NA, NP, M1 or M2 proteins or fragments (e.g., eM2), variants or derivatives thereof. In certain embodiments, a derivative protein can be a fusion protein, e.g., NP-eM2. In other embodiments, the isolated IV polypeptide or fragment, variant, or derivative thereof can be fused to a heterologous protein, e.g., a secretory signal peptide or the hepatitis B virus core protein. Preferably, the isolated IV polypeptide or fragment, variant, or derivative thereof comprises at least one immunogenic epitope of IV, wherein the antigen elicits a B-cell antibody response, a T-cell antibody response, or both.
  • Nucleic acids and fragments thereof of the present invention can be altered from their native state in one or more of the following ways. First, a nucleic acid or fragment thereof which encodes an IV polypeptide or fragment, variant, or derivative thereof can be part or all of a codon-optimized coding region, optimized according to codon usage in the animal in which the vaccine is to be delivered. In addition, a nucleic acid or fragment thereof which encodes an IV polypeptide can be a fragment which encodes only a portion of a full-length polypeptide, and/or can be mutated so as to, for example, remove from the encoded polypeptide non-desired protein motifs present in the encoded polypeptide or virulence factors associated with the encoded polypeptide. For example, the nucleic acid sequence could be mutated so as not to encode a membrane anchoring region that would prevent release of the polypeptide from the cell as with, e.g., eM2. Upon delivery, the polynucleotide of the invention is incorporated into the cells of the vertebrate in vivo, and a prophylactically or therapeutically effective amount of an immunologic epitope of an IV is produced in vivo.
  • Similarly, the proteins of the invention can be a fragment of a full-length IV polypeptide and/or can be altered so as to, for example, remove from the polypeptide non-desired protein motifs present in the polypeptide or virulence factors associated with the polypeptide. For example, the polypeptide could be altered so as not to encode a membrane anchoring region that would prevent release of the the polypeptide from the cell.
  • The invention further provides immunogenic compositions comprising at least one polynucleotide, wherein the polynucleotide comprises one or more nucleic acid fragments, where each nucleic acid fragment is a fragment of a codon-optimized coding region encoding an IV polypeptide or a fragment, a variant, or a derivative thereof; and immunogenic compositions comprising a polynucleotide as described above and at least one isolated IV polypeptide or a fragment, a variant, or derivative thereof. Such compositions can further comprise, for example, carriers, excipients, transfection facilitating agents, and/or adjuvants as described herein.
  • The immunogenic compositions comprising a polynucleotide and an isolated IV polypeptide or fragment, variant, or derivative thereof as described above can be provided so that the polynucleotide and protein formulation are administered separately, for example, when the polynucleotide portion of the composition is administered prior (or subsequent) to the isolated IV polypeptide portion of the composition. Alternatively, immunogenic compositions comprising the polynucleotide and the isolated IV polypeptide or fragment, variant, or derivative thereof can be provided as a single formulation, comprising both the polynucleotide and the protein, for example, when the polynucleotide and the protein are administered simultaneously. In another alternative, the polynucleotide portion of the composition and the isolated IV polypeptide portion of the composition can be provided simultaneously, but in separate formulations.
  • Compositions comprising at least one polynucleotide comprising one or more nucleic acid fragments, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide or fragment, variant, or derivative thereof together with and one or more isolated IV polypeptides or fragments, variants or derivatives thereof (as either a recombinant protein, a purified subunit, a viral vector expressing the protein, or in the form of an inactivated or attenuated IV vaccine) will be referred to herein as “combinatorial polynucleotide (e.g., DNA) vaccine compositions” or “single formulation heterologous prime-boost vaccine compositions.”
  • The compositions of the invention can be univalent, bivalent, trivalent or mulitvalent. A univalent composition will comprise only one polynucleotide comprising a nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide or a fragment, variant, or derivative thereof, and optionally the same IV polypeptide or a fragment, variant, or derivative thereof in isolated form. In a single formulation heterologous prime-boost vaccine composition, a univalent composition can include a polynucleotide comprising a nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide or a fragment, variant, or derivative thereof and an isolated polypeptide having the same antigenic region as the polynucleotide. A bivalent composition will comprise, either in polynucleotide or protein form, two different IV polypeptides or fragments, variants, or derivatives thereof, each capable of eliciting an immune response. The polynucleotide(s) of the composition can encode two IV polypeptides or alternatively, the polynucleotide can encode only one IV polypeptide and the second IV polypeptide would be provided by an isolated IV polypeptide of the invention as in, for example, a single formulation heterologous prime-boost vaccine composition. In the case where both IV polypeptides of a bivalent composition are delivered in polynucleotide form, the nucleic acid fragments operably encoding those IV polypeptides need not be on the same polynucleotide, but can be on two different polynucleotides. A trivalent or further multivalent composition will comprise three IV polypeptides or fragments, variants or derivatives thereof, either in isolated form or encoded by one or more polynucleotides of the invention.
  • The present invention further provides plasmids and other polynucleotide constructs for delivery of nucleic acid fragments of the invention to a vertebrate, e.g., a human, which provide expression of IV polypeptides, or fragments, variants, or derivatives thereof. The present invention further provides carriers, excipients, transfection-facilitating agents, immunogenicity-enhancing agents, e.g., adjuvants, or other agent or agents to enhance the transfection, expression or efficacy of the administered gene and its gene product.
  • In one embodiment, a mulitvalent composition comprises a single polynucleotide, e.g., plasmid, comprising one or more nucleic acid regions operably encoding IV polypeptides or fragments, variants, or derivatives thereof. Reducing the number of polynucleotides, e.g., plasmids in the compositions of the invention can have significant impacts on the manufacture and release of product, thereby reducing the costs associated with manufacturing the compositions. There are a number of approaches to include more than one expressed antigen coding sequence on a single plasmid. These include, for example, the use of Internal Ribosome Entry Site (IRES) sequences, dual promoters/expression cassettes, and fusion proteins.
  • The invention also provides methods for enhancing the immune response of a vertebrate to IV infection by administering to the tissues of a vertebrate one or more polynucleotides each comprising one or more nucleic acid fragments, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide or fragment, variant, or derivative thereof; and optionally administering to the tissues of the vertebrate one or more isolated IV polypeptides, or fragments, variants, or derivatives thereof. The isolated IV polypeptide can be administered prior to, at the same time (simultaneously), or subsequent to administration of the polynucleotides encoding IV polypeptides.
  • In addition, the invention provides consensus amino acid sequences for IV polypeptides, or fragments, variants or derivatives thereof, including, but not limited to the HA, NA, NP, M1 or M2 proteins or fragments (e.g. eM2), variants or derivatives thereof. Polynucleotides which encode the consensus polypeptides or fragments, variants or derivatives thereof, are also embodied in this invention. Such polynucleotides can be obtained by known methods, for example by backtranslation of the amino acid sequence and PCR synthesis of the corresponding polynucleotide as described below.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 shows an alignment of nucleotides 46-1542 of SEQ ID NO:1 (native NP coding region) with a coding region fully codon-optimized for human usage (SEQ ID NO:23).
  • FIG. 2 shows the protocol for the preparation of a formulation comprising 0.3 mM BAK, 7.5 mg/ml CRL 1005 and 5 mg/ml of DNA in a final volume of 3.6 ml, through the use of thermal cycling.
  • FIG. 3 shows the protocol for the preparation of a formulation comprising 0.3 mM BAK, 34 mg/ml or 50 mg/ml CRL 1005 and 2.5 mg/ml DNA in a final volumne of 4.0 ml, through the use of thermal cycling.
  • FIG. 4 shows the protocol for the simplified preparation (without thermal cycling) of a formulation comprising 0.3 mM BAK, 7.5 mg/ml CRL 1005 and 5 mg/ml DNA.
  • FIG. 5 shows the anti-NP antibody response three weeks after a single administration of a combinatorial prime-boost vaccine formulation against the influenza virus NP protein.
  • FIG. 6 shows the anti-NP antibody response twelve days after a second administration of a combinatorial prime-boost vaccine formulation against the influenza virus NP protein.
  • FIG. 7 shows the CD8+ T Cell response to a combinatorial prime-boost vaccine formulation against the influenza virus NP protein.
  • FIG. 8 shows the CD4+ T Cell response to a combinatorial prime-boost vaccine formulation against the influenza virus NP protein.
  • FIGS. 9A and 9B show the results of a two dose mouse immunization regimen study with plasmid DNA encoding IAV HA (H3).
  • FIGS. 10A and 10B show the in vitro expression of M1 and M2 from segment 7 and an M1M2 fusion.
  • FIGS. 11A and 11B show the in vitro expression of eM2-NP and codon-optimized influenza virus NP protein.
  • FIG. 12 shows the influenza A NP protein consensus amino acid sequence aligned with 22 full length NP sequences.
  • FIG. 13 is a schematic diagram of various vectors encoding influenza proteins described herein.
  • FIG. 14 are the results of western blot experiments as described in Example 13, Experiment 3. The blots show lysates of VM92 cells transfected with plasmids which express M2 or NP to compare expression of the influenza protein from different expression vectors.
  • FIG. 15 are the results of western blot experiments as described in Example 13, Experiment 3. The blots show lysates of VM92 cells transfected with plasmids which express M1, M2 or NP to compare expression of the influenza protein from expression vectors.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to compositions and methods for enhancing the immune response of a vertebrate in need of protection against IV infection by administering in vivo, into a tissue of a vertebrate, at least one polynucleotide comprising one or more nucleic acid fragments, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide, or a fragment, variant, or derivative thereof in cells of the vertebrate in need of protection. The present invention is also directed to administering in vivo, into a tissue of the vertebrate the above described polynucleotide and at least one isolated IV polypeptide, or a fragment, variant, or derivative thereof. The isolated IV polypeptide or fragment, variant, or derivative thereof can be, for example, a recombinant protein, a purified subunit protein, a protein expressed and carried by a heterologous live or inactivated or attentuated viral vector expressing the protein, or can be an inactivated IV, such as those present in conventional, commercially available, inactivated IV vaccines. According to either method, the polynucleotide is incorporated into the cells of the vertebrate in vivo, and an immunologically effective amount of the influenza protein, or fragment or variant encoded by the polynucleotide is produced in vivo. The isolated protein or fragment, variant, or derivative thereof is also administered in an immunologically effective amount. The polynucleotide can be administered to the vertebrate in need thereof either prior to, at the same time (simultaneously), or subsequent to the administration of the isolated IV polypeptide or fragment, variant, or derivative thereof.
  • Non-limiting examples of IV polypeptides within the scope of the invention include, but are not limited to, NP, HA, NA, M1 and M2 polypeptides, and fragments, e.g., eM2, derivatives, e.g., an NP-eM2 fusion, and variants thereof. Nucleotide and amino acid sequences of IV polypeptides from a wide variety of IV types and subtypes are known in the art. The nucleotide sequences set out below are the wild-type sequences. For example, the nucleotide sequence of the NP protein of Influenza A/PR/8/34 (H1N1) is available as GenBank Accession Number M38279.1, and has the following sequence, referred to herein as SEQ ID NO:1:
    AGCAAAAGCAGGGTAGATAATCACTCACTGAGTGACATCAAAATCATGGC
    GTCTCAAGGCACCAAACGATCTTACGAACAGATGGAGACTGATGGAGAAC
    GCCAGAATGCCACTGAAATCAGAGCATCCGTCGGAAAAATGATTGGTGGA
    ATTGGACGATTCTACATCCAAATGTGCACCGAACTCAAACTCAGTGATTA
    TGAGGGACGGTTGATCCAAAACAGCTTAACAATAGAGAGAATGGTGCTCT
    CTGCTTTTGACGAAAGGAGAAATAAATACCTTGAAGAACATCCCAGTGCG
    GGGAAAGATCCTAAGAAAACTGGAGGACCTATATACAGGAGAGTAAACGG
    AAAGTGGATGAGAGAACTCATCCTTTATGACAAAGAAGAAATAAGGCGAA
    TCTGGCGCCAAGCTAATAATGGTGACGATGCAACGGCTGGTCTGACTCAC
    ATGATGATCTGGCATTCCAATTTGAATGATGCAACTTATCAGAGGACAAG
    AGCTCTTGTTCGCACCGGAATGGATCCCAGGATGTGCTCTCTGATGCAAG
    GTTCAACTCTCCCTAGGAGGTCTGGAGCCGCAGGTGCTGCAGTCAAAGGA
    GTTGGAACAATGGTGATGGAATTGGTCAGAATGATCAAACGTGGGATCAA
    TGATCGGAACTTCTGGAGGGGTGAGAATGGACGAAAAACAAGAATTGCTT
    ATGAAAGAATGTGCAACATTCTCAAAGGGAAATTTCAAACTGCTGCACAA
    AAAGCAATGATGGATCAAGTGAGAGAGAGCCGGAACCCAGGGAATGCTGA
    GTTCGAAGATCTCACTTTTCTAGCACGGTCTGCACTCATATTGAGAGGGT
    CGGTTGCTCACAAGTCCTGCCTGCCTGCCTGTGTGTATGGACCTGCCGTA
    GCCAGTGGGTACGACTTTGAAAGGGAGGGATACTCTCTAGTCGGAATAGA
    CCCTTTCAGACTGCTTCAAAACAGCCAAGTGTACAGCCTAATCAGACCAA
    ATGAGAATCCAGCACACAAGAGTCAACTGGTGTGGATGGCATGCCATTCT
    GCCGCATTTGAAGATCTAAGAGTATTAAGCTTCATCAAAGGGACGAAGGT
    GCTCCCAAGAGGGAAGCTTTCCACTAGAGGAGTTCAAATTGCTTCCAATG
    AAAATATGGAGACTATGGAATCAAGTACACTTGAACTGAGAAGCAGGTAC
    TGGGCCATAAGGACCAGAAGTGGAGGAAACACCAATCAACAGAGGGCATC
    TGCGGGCCAAATCAGCATACAACCTACGTTCTCAGTACAGAGAAATCTCC
    CTTTTGACAGAACAACCGTTATGGCAGCATTCAGTGGGAATACAGAGGGG
    AGAACATCTGACATGAGGACCGAAATCATAAGGATGATGGAAAGTGCAAG
    ACCAGAAGATGTGTCTTTCCAGGGGCGGGGAGTCTTCGAGCTCTCGGACG
    AAAAGGCAGCGAGCCCGATCGTGCCTTCCTTTGACATGAGTAATGAAGGA
    TCTTATTTCTTCGGAGACAATGCAGAGGAATACGATAATTAAAGAAAAAT
    ACCCTTGTTTCTACT
  • The amino acid sequence of the NP protein of Influenza A/PR/8/34 (H1N1), encoded by nucleotides 46-1494 of SEQ ID NO:1 is as follows, referred to herein as SEQ ID NO:2:
    MASQGTKRSYEQMETDGERQNATEIRASVGKMIGGIGRFYIQMCTELKLS
    DYEGRLIQNSLTIERMVLSAFDERRNKYLEEHPSAGKDPKKTGGPIYRRV
    NGKWMRELILYDKEEIRRIWRQANNGDDATAGLTHMMIWHSNLNDATYQR
    TRALVRTGMDPRMCSLMQGSTLPRRSGAAGAAVKGVGTMVMELVRMIKRG
    INDRNFWRGENGRKTRIAYERMCNILKGKFQTAAQKAMMDQVRESRNPGN
    AEFEDLTFLARSALILRGSVAHKSCLPACVYGPAVASGYDFEREGYSLVG
    IDPFRLLQNSQVYSLIRPNENPAHKSQLVWMACHSAAFEDLRVLSFIKGT
    KVLPRGKLSTRGVQIASNENMETMESSTLELRSRYWAIRTRSGGNTNQQR
    ASAGQISIQPTFSVQRNLPFDRTTVMAAFSGNTEGRTSDMRTEIIRMMES
    ARPEDVSFQGRGVFELSDEKAASPIVPSFDMSNEGSYFFGDNAEEYDN
  • Segment 7 of the IAV genome encodes both M1 and M2. Seqment 7 of Influenza A virus (A/Puerto Rico/8/34/Mount Sinai (H1N1)), is available as GenBank Accession No. AF389121.1, and has the following sequence, referred to herein as SEQ ID NO:3:
    AGCGAAAGCAGGTAGATATTGAAAGATGAGTCTTCTAACCGAGGTCGAAA
    CGTACGTACTCTCTATCATCCCGTCAGGCCCCCTCAAAGCCGAGATCGCA
    CAGAGACTTGAAGATGTCTTTGCAGGGAAGAACACTGATCTTGAGGTTCT
    CATGGAATGGCTAAAGACAAGACCAATCCTGTCACCTCTGACTAAGGGGA
    TTTTAGGATTTGTGTTCACGCTCACCGTGCCCAGTGAGCGAGGACTGCAG
    CGTAGACGCTTTGTCCAAAATGCCCTTAATGGGAACGGGGATCCAAATAA
    CATGGACAAAGCAGTTAAACTGTATAGGAAGCTCAAGAGGGAGATTAACA
    TTCCATGGGGCCAAAGAAATCTCACTCAGTTATTCTGCTGGTGCACTTGC
    CAGTTGTATGGGCCTCATATACAACAGGATGGGGGCTGTGACCACTGAAG
    TGGCATTTGGCCTGGTATGTGCAACCTGTGAACAGATTGCTGACTCCCAG
    CATCGGTCTCATAGGCAAATGGTGACAACAACCAATCCACTAATCAGACA
    TGAGAACAGAATGGTTTTAGCCAGCACTACAGCTAAGGCTATGGAGCAAA
    TGGCTGGATCGAGTGAGCAAGCAGCAGAGGCCATGGAGGTTGCTAGTCAG
    GCTAGACAAATGGTGCAAGCGATGAGAACCATTGGGACTCATCCTAGCTC
    CAGTGCTGGTCTGAAAAATGATCTTCTTGAAAATTTGCAGGCCTATCAGA
    AACGAATGGGGGTGCAGATGCAACGGTTCAAGTGATCCTCTCGCTATTGC
    CGCAAATATCATTGGGATCTTGCACTTGACATTGTGGATTCTTGATCGTC
    TTTTTTTCAAATGCATTTACCGTCGCTTTAAATACGGACTGAAAGGAGGG
    CCTTCTACGGAAGGAGTGCCAAAGTCTATGAGGGAAGAATATCGAAAGGA
    ACAGCAGAGTGCTGTGGATGCTGACGATGGTCATTTTGTCAGCATAGAGC
    TGGAGTAAAAAACTACCTTGTTTCTACT
  • The amino acid sequence of the M1 protein of Influenza A/Puerto Rico/8/34/Mount Sinai(H1N1), encoded by nucleotides 26 to 784 of SEQ ID NO:3 is as follows, referred to herein as SEQ ID NO:4:
    MSLLTEVETYVLSIIPSGPLKAEIAQRLEDVFAGKNTDLEVLMEWLKTRP
    ILSPLTKGILGFVFTLTVPSERGLQRRRFVQNALNGNGDPNNMDKAVKLY
    RKLKREITFHGAKEISLSYSAGALASGMGLIYNRMGAVTTEVAFGLVCAT
    CEQIADSQHRSHRQMVTTNPLIRHENRMVLASTTAKAMEQMAGSSEQAAE
    AMEVASQARQMVQAMRTIGTHPSSSAGLKNDLLENLQAYQKRMGVQMQ
    RFK
  • The amino acid sequence of the M2 protein of Influenza A/Puerto Rico/8/34/Mount Sinai (H1N1), encoded (in spliced form) by nucleotides 26 to 51 and 740 to 1007 of SEQ ID NO:3 is as follows, referred to herein as SEQ ID NO:5:
    MSLLTEVETPIRNEWGCRCNGSSDPLAIAANIIGILHLTLWILDRLFFKC
    IYRRFKYGLKGGPSTEGVPKSMREEYRKEQQSAVDADDGHFVSIELE
  • The Extracellular region of the M2 protein (eM2) corresponds to the first 24 amino acids of the N-terminal end of the protein, and is underlined above. See Fischer, W. B. et al., Biochim. Biophys. Acta. 1561:27-45 (2002); Zhong, Q. et al., FEBS Lett. 434:265-71 (1998).
  • A derivative of NP and eM2 described herein is encoded by a construct which encodes the first 24 amino acids of M2 and all or a portion of NP. The fusion constructs may be constructed with the eM2 sequences followed by the NP sequences, or with the NP sequences followed by the eM2 sequences. Exemplary fusion constructs using the NP and M2 sequences from Influenza A/PR/8/34 (H1N1) are set out below. A sequence, using the original influenza virus nucleotide sequences, which encodes the first 24 amino acids of M2 fused at its 3′ end to a sequence which encodes NP in its entirety eM2-NP is referred to herein as SEQ ID NO:6:
    1 ATGAGTCTTC TAACCGAGGT CGAAACGCCT ATCAGAAACG AATGGGGGTG CAGATGCAAC
    61 GGTTCAAGTG ATATGGCGTC TCAAGGCACC AAACGATCTT ACGAACAGAT GGAGACTGAT
    121 GGAGAACGCC AGAATGCCAC TGAAATCAGA GCATCCGTCG GAAAAATGAT TGGTGGAATT
    181 GGACGATTCT ACATCCAAAT GTGCACCGAA CTCAAACTCA GTGATTATGA GGGACGGTTG
    241 ATCCAAAACA GCTTAACAAT AGAGAGAATG GTGCTCTCTG CTTTTGACGA AAGGAGAAAT
    301 AAATACCTTG AAGAACATCC CAGTGCGGGG AAAGATCCTA AGAAAACTGG AGGACCTATA
    361 TACAGGAGAG TAAACGGAAA GTGGATGAGA GAACTCATCC TTTATGACAA AGAAGAAATA
    421 AGGCGAATCT GGCGCCAAGC TAATAATGGT GACGATGCAA CGGCTGGTCT GACTCACATG
    481 ATGATCTGGC ATTCCAATTT GAATGATGCA ACTTATCAGA GGACAAGAGC TCTTGTTCGC
    541 ACCGGAATGG ATCCCAGGAT GTGCTCTCTG ATGCAAGGTT CAACTCTCCC TAGGAGGTCT
    601 GGAGCCGCAG GTGCTGCAGT CAAAGGAGTT GGAACAATGG TGATGGAATT GGTCAGAATG
    661 ATCAAACGTG GGATCAATGA TCGGAACTTC TGGAGGGGTG AGAATGGACG AAAAACAAGA
    721 ATTGCTTATG AAAGAATGTG CAACATTCTC AAAGGGAAAT TTCAAACTGC TGCACAAAAA
    781 GCAATGATGG ATCAAGTGAG AGAGAGCCGG AACCCAGGGA ATGCTGAGTT CGAAGATCTC
    841 ACTTTTCTAG CACGGTCTGC ACTCATATTG AGAGGGTCGG TTGCTCACAA GTCCTGCCTG
    901 CCTGCCTGTG TGTATGGACC TGCCGTAGCC AGTGGGTACG ACTTTGAAAG GGAGGGATAC
    961 TCTCTAGTCG GAATAGACCC TTTCAGACTG CTTCAAAACA GCCAAGTGTA CAGCCTAATC
    1021 AGACCAAATG AGAATCCAGC ACACAAGAGT CAACTGGTGT GGATGGCATG CCATTCTGCC
    1081 GCATTTGAAG ATCTAAGAGT ATTAAGCTTC ATCAAAGGGA CGAAGGTGCT CCCAAGAGGG
    1141 AAGCTTTCCA CTAGAGGAGT TCAAATTGCT TCCAATGAAA ATATGGAGAC TATGGAATCA
    1201 AGTACACTTG AACTGAGAAG CAGGTACTGG GCCATAAGGA CCAGAAGTGG AGGAAACACC
    1261 AATCAACAGA GGGCATCTGC GGGCCAAATC AGCATACAAC CTACGTTCTC AGTACAGAGA
    1321 AATCTCCCTT TTGACAGAAC AACCGTTATG GCAGCATTCA GTGGGAATAC AGAGGGGAGA
    1381 ACATCTGACA TGAGGACCGA AATCATAAGG ATGATGGAAA GTGCAAGACC AGAAGATGTG
    1441 TCTTTCCAGG GGCGGGGAGT CTTCGAGCTC TCGGACGAAA AGGCAGCGAG CCCGATCGTG
    1501 CCTTCCTTTG ACATGAGTAA TGAAGGATCT TATTTCTTCG GAGACAATGC AGAGGAATAC
    1561 GATAAT
  • The amino acid sequence of the eM2-NP fusion protein of Influenza A/PR/8/341 (H1N1), encoded by nucleotides 1 to 1566 SEQ ID NO:6 is as follows, referred to herein as SEQ ID NO:7 (eM2 amino acid sequence underlined):
    MSLLTEVETPIRNEWGCRCNGSSDMASQGTKRSYEQMETDGERQNATEIR
    ASVGKMIGGIGRFYIQMCTELKLSDYEGRLIQNSLTIERMVLSAFDERRN
    KYLEEHPSAGKDPKKTGGPIYRRVNGKWMRELILYDKEEIRRIWRQANNG
    DDATAGLTHMMIWHSNLNDATYQRTRALVRTGMDPRMCSLMQGSTLPRRS
    GAAGAAVKGVGTMVMELVRMIKRGINDRNFWRGENGRKTRIAYERMCNIL
    KGKFQTAAQKAMMDQVRESRNPGNAEFEDLTFLARSALILRGSVAHKSCL
    PACVYGPAVASGYDFEREGYSLVGIDPFRLLQNSQVYSLIRPNENPAHKS
    QLVWMACHSAAFEDLRVLSFIKGTKVLPRGKLSTRGVQIASNENMETMES
    STLELRSRYWAIRTRSGGNTNQQRASAGQISIQPTFSVQRNLPFDRTTVM
    AAFSGNTEGRTSDMRTEIIRMMESARPEDVSFQGRGVFELSDEKAASPIV
    PSFDMSNEGSYFFGDNAEEYDN
  • A sequence, using the original influenza virus nucleotide sequences, which encodes NP in its entirety fused at its 3′ end to the first 24 amino acids of M2 fused to a sequence which encodes NP in its entirety is referred to herein as SEQ ID NO:8:
    ATGGCGTCTCAAGGCACCAAACGATCTTACGAACAGATGGAGACTGATGG
    AGAACGCCAGAATGCCACTGAAATCAGAGCATCCGTCGGAAAAATGATTG
    GTGGAATTGGACGATTCTACATCCAAATGTGCACCGAACTCAAACTCAGT
    GATTATGAGGGACGGTTGATCCAAAACAGCTTAACAATAGAGAGAATGGT
    GCTCTCTGCTTTTGACGAAAGGAGAAATAAATACCTTGAAGAACATCCCA
    GTGCGGGGAAAGATCCTAAGAAAACTGGAGGACCTATATACAGGAGAGTA
    AACGGAAAGTGGATGAGAGAACTCATCCTTTATGACAAAGAAGAAATAAG
    GCGAATCTGGCGCCAAGCTAATAATGGTGACGATGCAACGGCTGGTCTGA
    CTCACATGATGATCTGGCATTCCAATTTGAATGATGCAACTTATCAGAGG
    ACAAGAGCTCTTGTTCGCACCGGAATGGATCCCAGGATGTGCTCTCTGAT
    GCAAGGTTCAACTCTCCCTAGGAGGTCTGGAGCCGCAGGTGCTGCAGTCA
    AAGGAGTTGGAACAATGGTGATGGAATTGGTCAGAATGATCAAACGTGGG
    ATCAATGATCGGAACTTCTGGAGGGGTGAGAATGGACGAAAAACAAGAAT
    TGCTTATGAAAGAATGTGCAACATTCTCAAAGGGAAATTTCAAACTGCTG
    CACAAAAAGCAATGATGGATCAAGTGAGAGAGAGCCGGAACCCAGCGAAT
    GCTGAGTTCGAAGATCTCACTTTTCTAGCACGGTCTGCACTCATATTGAG
    AGCGTCGGTTGCTCACAAGTCCTGCCTGCCTGCCTGTGTGTATGGACCTG
    CCGTAGCCAGTGGGTACGACTTTGAAAGGGAGGGATACTCTCTAGTCGGA
    ATAGACCCTTTCAGACTGCTTCAAAACAGCCAAGTGTACAGCCTAATCAG
    ACCAAATGAGAATCCAGCACACAAGAGTCAACTGGTGTGGATGGCATGCC
    ATTCTGCCGCATTTGAAGATCTAAGAGTATTAAGCTTCATCAAAGGGACG
    AAGGTGCTCCCAAGAGGGAAGCTTTCCACTAGAGGAGTTCAAATTGCTTC
    CAATGAAAATATGGAGACTATGGAATCAAGTACACTTGAACTGAGAAGCA
    GGTACTGGGCCATAAGGACCAGAAGTGGAGGAAACACCAATCAACAGAGG
    GCATCTGCGGGCCAAATCAGCATACAACCTACGTTCTCAGTACAGAGAAA
    TCTCCCTTTTGACAGAACAACCGTTATGGCAGCATTCAGTGGGAATACAG
    AGGGGAGAACATCTGACATGAGGACCGAAATCATAAGGATGATGGAAAGT
    GCAAGACCAGAAGATGTGTCTTTCCAGGGGCGGGGAGTCTTCGAGCTCTC
    GGACGAAAAGGCAGCGAGCCCGATCGTGCCTTCCTTTGACATGAGTAATG
    AAGGATCTTATTTCTTCGGAGACAATGCAGAGGAATACGATAATATGAGT
    CTTCTAACCGAGGTCGAAACGCCTATCAGAAACGAATGGGGGTGCAGATG
    CAACGGTTCAAGTGAT
  • The amino acid sequence of the NP-eM2 fusion protein of Influenza A/PR/8/34/ (H1N1), encoded by nucleotides 1 to 1566 of SEQ ID NO:8 is as follows, referred to herein as SEQ ID NO:9 (eM2 amino acid sequence underlined):
    MASQGTKRSYEQMETDGERQNATEIRASVGKMIGGIGRFYIQMCTELKLS
    DYEGRLIQNSLTIERMVLSAFDERRNKYLEEHPSAGKDPKKTGGPIYRRV
    NGKWMRELILYDKEEIRRIWRQANNGDDATAGLTHMMIWHSNLNDATYQR
    TRALVRTGMDPRMCSLMQGSTLPRRSGAAGAAVKGVGTMVMELVRMIKRG
    INDRNFWRGENGRKTRIAYERMCNILKGKFQTAAQKAMMDQVRESRNPGN
    AEFEDLTFLARSALILRGSVAHKSCLPACVYGPAVASGYDFEREGYSLVG
    IDPFRLLQNSQVYSLIRPNENPAHKSQLVWMACHSAAFEDLRVLSFIKGT
    KVLPRGKLSTRGVQIASNENMETMESSTLELRSRYWAIRTRSGGNTNQQR
    ASAGQISIQPTFSVQRNLPFDRTTVMAAFSGNTEGRTSDMRTEIIRMMES
    ARPEDVSFQGRGVFELSDEKAASPIVPSFDMSNEGSYFFGDNAEEYDN
    MSLLTEVETPIRNEWGCRCNGSSD
  • The construction of functional fusion proteins often requires a linker sequence between the two fused fragments, in order to adopt an extended conformation to allow maximal flexibility. We used program LINKER (Chiquita J. Crasto C. J. and Feng, J. Protein Engineering 13:309-312 (2000), program publicly available at http://chutney.med.yale.edu/linker/linker.html (visited Apr. 16, 2003)), that can automatically generate a set of linker sequences, which are known to adopt extended conformations as determined by X-ray crystallography and NMR. Examples of suitable linkers to use in various eM2-NP or NP-eM2 fusion proteins are as follows:
    1. GYNTRA (SEQ ID NO:10)
    2. FQMGET (SEQ ID NO:11)
    3. FDRVKHLK (SEQ ID NO:12)
    4. GRNTNGVIT (SEQ ID NO:13)
    5. VNEKTIPDHD (SEQ ID NO:14)
  • The nucleotide sequence of the NP protein of Influenza B/LEE/40 is available as GenBank Accession Number K01395, and has the following sequence, referred to herein as SEQ ID NO:15:
    1 ATGTCCAACA TGGATATTGA CAGTATAAAT ACCGGAACAA TCGATAAAAC ACCAGAAGAA
    61 CTGACTCCCG GAACCAGTGG GGCAACCAGA CCAATCATCA AGCCAGCAAC CCTTGCTCCG
    121 CCAAGCAACA AACGAACCCG AAATCCATCT CCAGAAAGGA CAACCACAAG CAGTGAAACC
    181 GATATCGGAA GGAAAATCCA AAAGAAACAA ACCCCAACAG AGATAAAGAA GAGCGTCTAC
    241 AAAATGGTGG TAAAACTGGG TGAATTCTAC AACCAGATGA TGGTCAAAGC TGGACTTAAT
    301 GATGACATGG AAAGGAATCT AATTCAAAAT GCACAAGCTG TGGAGAGAAT CCTATTGGCT
    361 GCAACTGATG ACAAGAAAAC TGAATACCAA AAGAAAAGGA ATGCCAGAGA TGTCAAAGAA
    421 GGGAAGGAAG AAATAGACCA CAACAAGACA GGAGGCACCT TTTATAAGAT GGTAAGAGAT
    481 GATAAAACCA TCTACTTCAG CCCTATAAAA ATTACCTTTT TAAAAGAAGA GGTGAAAACA
    541 ATGTACAAGA CCACCATGGG GAGTGATGGT TTCAGTGGAC TAAATCACAT TATGATTGGA
    601 CATTCACAGA TGAACGATGT CTGTTTCCAA AGATCAAAGG GACTGAAAAG GGTTGGACTT
    661 GACCCTTCAT TAATCAGTAC TTTTGCCGGA AGCACACTAC CCAGAAGATC AGGTACAACT
    721 GGTGTTGCAA TCAAAGGAGG TGGAACTTTA GTGGATGAAG CCATCCGATT TATAGGAAGA
    781 GCAATGGCAG ACAGAGGGCT ACTGAGAGAC ATCAAGGCCA AGACGGCCTA TGAAAAGATT
    841 CTTCTGAATC TGAAAAACAA GTGCTCTGCG CCGCAACAAA AGGCTCTAGT TGATCAAGTG
    901 ATCGGAAGTA GGAACCCAGG GATTGCAGAC ATAGAAGACC TAACTCTGCT TGCCAGAAGC
    961 ATGGTAGTTG TCAGACCCTC TGTAGCGAGC AAAGTGGTGC TTCCCATAAG CATTTATGCT
    1021 AAAATACCTC AACTAGGATT CAATACCGAA GAATACTCTA TGGTTGGGTA TGAAGCCATG
    1081 GCTCTTTATA ATATGGCAAC ACCTGTTTCC ATATTAAGAA TGGGAGATGA CGCAAAAGAT
    1141 AAATCTCAAC TATTCTTCAT GTCGTGCTTC GGAGCTGCCT ATGAAGATCT AAGAGTGTTA
    1201 TCTGCACTAA CGGGCACCGA ATTTAAGCCT AGATCAGCAC TAAAATGCAA GGGTTTCCAT
    1261 GTCCCGGCTA AGGAGCAAGT AGAAGGAATG GGGGCAGCTC TGATGTCCAT CAAGCTTCAG
    1321 TTCTGGGCCC CAATGACCAG ATCTGGAGGG AATGAAGTAA GTGGAGAAGG AGGGTCTGGT
    1381 CAAATAAGTT GCAGCCCTGT GTTTGCAGTA GAAAGACCTA TTGCTCTAAG CAAGCAAGCT
    1441 GTAAGAAGAA TGCTGTCAAT GAACGTTGAA GGACGTGATG CAGATGTCAA AGGAAATCTA
    1501 CTCAAAATGA TGAATGATTC AATGGCAAAG AAAACCAGTG GAAATGCTTT CATTGGGAAG
    1561 AAAATGTTTC AAATATCAGA CAAAAACAAA GTCAATCCCA TTGAGATTCC AATTAAGCAG
    1621 ACCATCCCCA ATTTCTTCTT TGGGAGGGAC ACAGCAGAGG ATTATGATGA CCTCGATTAT
    1681 TAA
  • The amino acid sequence of the NP protein of IBV B/LEE/40, encoded by nucleotides 1-1680 of SEQ ID NO:1 is as follows, referred to herein as SEQ ID NO:16:
    MSNMDIDSINTGTIDKTPEELTPGTSGATRPIIKPATLAPPSNKRTRNPS
    PERTTTSSETDIGRKIQKKQTPTEIKKSVYKMVVKLGEFYNQMMVKAGLN
    DDMERNLIQNAQAVERILLAATDDKKTEYQKKRNARDVKEGKEEIDHNKT
    GGTFYKMVRDDKTIYFSPIKITFLKEEVKTMYKTTMGSDGFSGLNHIMIG
    HSQMNDVCFQRSKGLKRVGLDPSLISTFAGSTLPRRSGTTGVAIKGGGTL
    VDEAIRFIGRAMADRGLLRDIKAKTAYEKILLNLKNKCSAPQQKALVDQV
    IGSRNPGIADIEDLTLLARSMVVVRPSVASKVVLPISIYAKIPQLGFNTE
    EYSMVGYEAMALYNMATPVSILRMGDDAKDKSQLFFMSCFGAAYEDLRVL
    SALTGTEFKPRSALKCKGFHVPAKEQVEGMGAALMSIKLQFWAPMTRSGG
    NEVSGEGGSGQISCSPVFAVERPIALSKQAVRRMLSMNVEGRDADVKGNL
    LKMMNDSMAKKTSGNAFIGKKMFQISDKNKVNPIEIPIKQTIPNFFFGRD
    TAEDYDDLDY
  • Non limiting examples of nucleotide sequences encoding the IAV hemagglutinin (HA) are as follows. It should be noted that HA sequences vary significantly between IV subtypes. Virtually any nucleotide sequence encoding an IV HA is suitable for the present invention. In fact, HA sequences included in vaccines and therapeutic formulations of the present invention (discussed in more detail below) might change from year to year depending on the prevalent strain or strains of IV.
  • The partial nucleotide sequence of the HA protein of IAV A/New_York/1/18(H1N1) is available as GenBank Accession Number AF116576, and has the following sequence, referred to herein as SEQ ID NO: 17:
    1 atggaggcaa gactactggt cttgttatgt gcatttgcag ctacaaatgc agacacaata
    61 tgtataggct accatgcgaa taactcaacc gacactgttg acacagtact cgaaaagaat
    121 gtgaccgtga cacactctgt taacctgctc gaagacagcc acaacggaaa actatgtaaa
    181 ttaaaaggaa tagccccatt acaattgggg aaatgtaata tcgccggatg gctcttggga
    241 aacccggaat gcgatttact gctcacagcg agctcatggt cctatattgt agaaacatcg
    301 aactcagaga atggaacatg ttacccagga gatttcatcg actatgaaga actgagggag
    361 caattgagct cagtgtcatc gtttgaaaaa ttcgaaatat ttcccaagac aagctcgtgg
    421 cccaatcatg aaacaaccaa aggtgtaacg gcagcatgct cctatgcggg agcaagcagt
    481 ttttacagaa atttgctgtg gctgacaaag aagggaagct catacccaaa gcttagcaag
    541 tcctatgtga acaataaagg gaaagaagtc cttgtactat ggggtgttca tcatccgcct
    601 accggtactg atcaacagag tctctatcag aatgcagatg cttatgtctc tgtagggtca
    661 tcaaaatata acaggagatt caccccggaa atagcagcga gacccaaagt aagaggtcaa
    721 gctgggagga tgaactatta ctggacatta ctagaacccg gagacacaat aacatttgag
    781 gcaactggaa atctaatagc accatggtat gctttcgcac tgaatagagg ttctggatcc
    841 ggtatcatca cttcagacgc accagtgcat gattgtaaca cgaagtgtca aacaccccat
    901 ggtgctataa acagcagtct ccctttccag aatatacatc cagtcacaat aggagagtgc
    961 ccaaaatacg tcaggagtac caaattgagg atggctacag gactaagaaa cattccatct
    1021 attcaatcca ggggtctatt tggagccatt gccggtttta ttgagggggg atggactgga
    1081 atgatagatg gatggtatgg ttatcatcat cagaatgaac agggatcagg ctatgcagcg
    1141 gatcaaaaaa gcacacaaaa tgccattgac gggattacaa acaaggtgaa ttctgttatc
    1201 gagaaaatga acacccaatt
  • The amino acid sequence of the partial HA protein of IAV A/New_York/1/18(H1N1), encoded by nucleotides 1 to 1218 of SEQ ID NO:17 is as follows, referred to herein as SEQ ID NO:18:
    MEARLLVLLCAFAATNADTICIGYHANNSTDTVDTVLEKNVTVTHSVNLL
    EDSHNGKLCKLKGIAPLQLGKCNIAGWLLGNPECDLLLTASSWSYIVETS
    NSENGTGYPGDFLDYEELREQLSSVSSFEKFEIFPKTSSWPNHETTKGVT
    AACSYAGASSFYRNLLWLTKGSSYPKLSKSYVNNKGKEVLVLWGVHHPPT
    GTDQQSLYQNADAYVSVGSSKYNRRFTPEIAARPKVRGQAGRMNYYWTLL
    EPGDTITFEATGNLIAPWYAFALNRGSGSGIITSDAPVHDCNTKCQTPHG
    AINSSLPFQNIHPVTIGECPKYVRSTKLRMATGLRNIPSIQSRGLFGAIA
    GFIEGGWTGMIDGWYGYHHQNEQGSGYAADQKSTQNAIDGITNKVNSVIE
    KMNTQ
  • The nucleotide sequence of the IAV A/Hong Kong/482/97 hemagglutinin (H5) is available as GenBank Accession Number AF046098, and has the following sequence, referred to herein as SEQ ID NO:19:
    1 ctgtcaaaat qgagaaaata gtgcttcttc ttgcaacagt cagtcttgtt aaaagtgatc
    61 agatttgcat tggttaccat gcaaacaact cgacagagca ggttgacaca ataatggaaa
    121 agaatgttac tgttacacat gcccaagaca tactggaaag gacacacaac gggaagctct
    181 gcgatctaaa tggagtgaaa cctctcattt tgagggattg tagtgtagct ggatggctcc
    241 tcggaaaccc tatgtgtgac gaattcatca atgtgccgga atggtcttac atagtggaga
    301 aggccagtcc agccaatgac ctctgttatc cagggaattt caacgactat gaagaactga
    361 aacacctatt gagcagaata aaccattttg agaaaattca gatcatcccc aaaagttctt
    421 gqtccaatca tgatgcctca tcaggggtga gctcagcatg tccatacctt gggaggtcct
    481 cctttttcag aaatgtggta tggcttatca aaaagaacag tgcataccca acaataaaga
    541 ggagctacaa taataccaac caagaagatc ttttggtact gtgggggatt caccatccta
    601 atgatgcggc agaycagaca aagctctatc aaaatccaac cacctacatt tccgttggaa
    661 catcaacact gaaccagaga ttggttccag aaatagctac tagacccaaa gtaaacgggc
    721 aaagtggaag aatggagttc ttctggacaa ttttaaagcc gaatgatgcc atcaatttcg
    781 agagtaatgg aaatttcatt gccccagaat atgcatacaa aattgtcaag aaaggggact
    841 caacaattat gaaaagtgaa ttggaatatg gtaactgcaa caccaagtgt caaactccaa
    901 tgggggcgat aaactctagt atgccattcc acaacataca ccccctcacc atcggggaat
    961 gccccaaata tgtgaaatca aacagattag ttcttgcgac tggactcaga aatacccctc
    1021 aaagggagag aagaagaaaa aagagaggac tatttggagc tatagcaggt tttatagagg
    1081 gaggatggca gggcatggta gatggttggt atgggtacca ccatagcaat gagcagggga
    1141 gtggatacgc tgcagacaaa gaatccactc aaaaggcaat agatggagtc accaataagg
    1201 tcaactcgat cattaacaaa atgaacactc agtttgaggc cgttggaagg gaatttaata
    1261 acttagaaag gagaatagag aatttaaaca agaaaatgga agacggattc ctagatgtct
    1321 ggacttacaa tgctgaactt ctggttctca tggaaaatga gagaactctc gactttcatg
    1381 actcaaatgt caagaacctt tacgacaagg tccgactaca gcttagggat aatgcaaagg
    1441 aactgggtaa tggttgtttc gaattctatc acaaatgtga taatgaatgt atggaaagtg
    1501 taaaaaacgg aacgtatgac tacccgcagt attcagaaga agcaagacta aacagagagg
    1561 aaataagtgg agtaaaattg gaatcaatgg gaacttacca aatactgtca atttattcaa
    1621 cagtggcgag ttccctagca ctggcaatca tggtagctgg tctatcttta tggatgtgct
    1681 ccaatggatc gttacaatgc agaatttgca tttaaatttg tgagttcaga ttgtagttaa
    1741 a
  • The amino acid sequence of the HA protein of IAV A/Hong Kong/482/97 (H5), encoded by nucleotides 9 to 1715 of SEQ ID NO:19 is as follows, referred to herein as SEQ ID NO:20:
    MEKIVLLLATVSLVKSDQICIGYHANNSTEQVDTIMEKNVTVTHAQDILE
    RTHNGKLGDLNGVKPLILRDCSVAGWLLGNPMCDEFINVPEWSYIVEKAS
    PANDLCYPGNFNDYEELKHLLSRINHFEKIQIIPKSSWSNHDASSGVSSA
    GPYLGRSSFFRNVVWLIKKNSAYPTIKRSYNNTNQEDLLVLWGIHHPNDA
    AEQTKLYQNPTTYISVGTSTLNQRLVPEIATRPKVNGQSGRMEFFWTILK
    PNDAINFESNGNFIAPEYAYKIVKKGDSTIMKSELEYGNCNTKCQTPMGA
    INSSMPFHNIHPLTIGECPKYVKSNRLVLATGLRNTPQRERRRKKRGLFG
    AIAGFIEGGWQGMVDGWYGYHHSNEQGSGYAADKESTQKAIDGVTNKVNS
    IINKMNTQFEAVGREFNNLERRIENLNKKMEDGFLDVWTYNAELLVLMEN
    ERTLDFHDSNVKNLYDKVRLQLRDNAKELGNGCFEFYHKCDNECMESVKN
    GTYDYPQYSEEARLNREEISGVKLESMGTYQILSIYSTVASSLALAIMVA
    GLSLWMCSNGSLQCRICI
  • The nucleotide sequence of the IAV A/Hong Kong/1073/99(H9N2) is available as GenBank Accession Number INA404626, and has the following sequence, referred to herein as SEQ ID NO:21:
    1 gcaaaagcag gggaattact taactagcaa aatggaaaca atatcactaa taactatact
    61 actagtagta acagcaagca atgcagataa aatctgcatc ggccaccagt caacaaactc
    121 cacagaaact gtgqacacgc taacagaaac caatgttcct gtgacacatg ccaaagaatt
    181 gctccacaca gagcataatg gaatgctgtg tgcaacaagc ctgggacatc ccctcattct
    241 agacacatgc actattgaag gactagtcta tggcaaccct tcttgtgacc tgctgttggg
    301 aggaagagaa tggtcctaca tcgtcgaaag atcatcagct gtaaatggaa cgtgttaccc
    361 tgggaatgta gaaaacctaq aggaactcag gacacttttt agttccgcta gttcctacca
    421 aagaatccaa atcttcccag acacaacctg gaatgtgact tacactggaa caagcagagc
    481 atgttcaggt tcattctaca ggagtatgag atggctgact caaaagagcg gtttttaccc
    541 tgttcaagac gcccaataca caaataacag gggaaagagc attcttttcg tgtggggcat
    601 acatcaccca cccacctata ccgagcaaac aaatttgtac ataagaaacg acacaacaac
    661 aagcgtgaca acagaagatt tgaataggac cttcaaacca gtgatagggc caaggcccct
    721 tgtcaatggt ctgcagggaa gaattgatta ttattggtcg gtactaaaac caggccaaac
    781 attgcgagta cgatccaatg ggaatctaat tgctccatgg tatggacacg ttctttcagg
    841 agggagccat ggaagaatcc tgaagactga tttaaaaggt ggtaattqtg tagtgcaatg
    901 tcagactgaa aaaggtggct taaacagtac attgccattc cacaatatca gtaaatatgc
    961 atttggaacc tgccccaaat atgtaagagt taatagtctc aaactggcag tcggtctgag
    1021 gaacgtgcct gctagatcaa gtagaggact atttggagcc atagctggat tcatagaagg
    1081 aggttggcca ggactagtcg ctggctggta tggtttccag cattcaaatg atcaaggggt
    1141 tggtatggct gcagataggg attcaactca aaaggcaatt gataaaataa catccaaggt
    1201 gaataatata gtcgacaaga tgaacaagca atatgaaata attgatcatg aattcagtga
    1261 ggttgaaact agactcaata tgatcaataa taagattgat gaccaaatac aaqacgtatg
    1321 ggcatataat gcagaattgc tagtactact tgaaaatcaa aaaacactcg atgagcatga
    1381 tgcgaacgtg aacaatctat ataacaaggt gaagagggca ctgggctcca atgctatgga
    1441 agatgggaaa ggctgtttcg agctatacca taaatgtgat gatcagtgca tggaaacaat
    1501 tcggaacggg acctataata ggagaaagta tagagaggaa tcaagactag aaaggcagaa
    1561 aatagagggg gttaagctgg aatctgaggg aacttacaaa atcctcacca tttattcgac
    1621 tgtcgcctca tctcttgtgc ttgcaatggg gtttgctgcc ttcctgttct gggccatgtc
    1681 caatggatct tgcagatgca acatttgtat ataa
  • The amino acid sequence of the HA protein of IAV A/Hong Kong/1073/99 (H9N2), encoded by nucleotides 32 to 1711 of SEQ ID NO:21 is as follows, referred to herein as SEQ ID NO:22:
    METISLITILLVVTASNADKICIGHQSTNSTETVDTLTETNVPVTHAKEL
    LHTEHNGMLCATSLGHPLILDTCTIEGLVYGNPSGDLLLGGREWSYIVER
    SSAVNGTCYPGNVENLEELRTLFSSASSYQRIQIFPDTTWNVTYTGTSRA
    CSGSFYRSMRWLTQKSGFYPVQDAQYTNNRGKSILFVWGIHHPPTYTEQT
    NLYIRNDTTTSVTTEDLNRTFKPVIGPRPLVNGLQGRIDYYWSVLKPGQT
    LRVRSNGNLIAPWYGHVLSGGSHGRILKTDLKGGNCVVQCQTEKGGLNST
    LPFHNISKYAFGTCPKYVRVNSLKLAVGLRNVPARSSRGLFGAIAGFIEG
    GWPGLVAGWYGFQHSNDQGVGMAADRDSTQKAIDKITSKVNNIVDKMNKQ
    YEIIDHEFSEVETRLNMINNKIDDQIQDVWAYNAELLVLLENQKTLDEHD
    ANVNNLYNKVKRALGSNAMEDGKGCFELYHKCDDQCMETIRNGTYNRRKY
    REESRLERQKIEGVKLESEGTYKILTIYSTVASSLVLAMGFAAFLFWAMS
    NGSGRGNICI
  • The present invention also provides vaccine compositions and methods for delivery of IV coding sequences to a vertebrate with optimal expression and safety conferred through codon optimization and/or other manipulations. These vaccine compositions are prepared and administered in such a manner that the encoded gene products are optimally expressed in the vertebrate of interest. As a result, these compositions and methods are useful in stimulating an immune response against IV infection. Also included in the invention are expression systems, delivery systems, and codon-optimized IV coding regions.
  • In a specific embodiment, the invention provides combinatorial polynucleotide (e.g., DNA) vaccines which combine both a polynucleotide vaccine and polypeptide (e.g., either a recombinant protein, a purified subunit protein, a viral vector expressing an isolated IV polypeptide, or in the form of an inactivated or attenuated IV vaccine) vaccine in a single formulation. The single formulation comprises an IV polypeptide-encoding polynucleotide vaccine as described herein, and optionally, an effective amount of a desired isolated IV polypeptide or fragment, variant, or derivative thereof. The polypeptide may exist in any form, for example, a recombinant protein, a purified subunit protein, a viral vector expressing an isolated IV polypeptide, or in the form of an inactivated or attenuated IV vaccine. The IV polypeptide or fragment, variant, or derivative thereof encoded by the polynucleotide vaccine may be identical to the isolated IV polypeptide or fragment, variant, or derivative thereof. Alternatively, the IV polypeptide or fragment, variant, or derivative thereof encoded by the polynucleotide may be different from the isolated IV polypeptide or fragment, variant, or derivative thereof.
  • It is to be noted that the term “a” or “an” entity refers to one or more of that entity; for example, “a polynucleotide,” is understood to represent one or more polynucleotides. As such, the terms “a” (or “an”), “one or more,” and “at least one” can be used interchangeably herein.
  • The term “polynucleotide” is intended to encompass a singular nucleic acid or nucleic acid fragment as well as plural nucleic acids or nucleic acid fragments, and refers to an isolated molecule or construct, e.g., a virus genome (e.g., a non-infectious viral genome), messenger RNA (mRNA), plasmid DNA (pDNA), or derivatives of pDNA (e.g., minicircles as described in (Darquet, A-M et al., Gene Therapy 4:1341-1349 (1997)) comprising a polynucleotide. A polynucleotide may comprise a conventional phosphodiester bond or a non-conventional bond (e.g., an amide bond, such as found in peptide nucleic acids (PNA)).
  • The terms “nucleic acid” or “nucleic acid fragment” refer to any one or more nucleic acid segments, e.g., DNA or RNA fragments, present in a polynucleotide or construct. A nucleic acid or fragment thereof may be provided in linear (e.g., mRNA) or circular (e.g., plasmid) form as well as double-stranded or single-stranded forms. By “isolated” nucleic acid or polynucleotide is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment. For example, a recombinant polynucleotide contained in a vector is considered isolated for the purposes of the present invention. Further examples of an isolated polynucleotide include recombinant polynucleotides maintained in heterologous host cells or purified (partially or substantially) polynucleotides in solution. Isolated RNA molecules include in vivo or in vitro RNA transcripts of the polynucleotides of the present invention. Isolated polynucleotides or nucleic acids according to the present invention further include such molecules produced synthetically.
  • As used herein, a “coding region” is a portion of nucleic acid which consists of codons translated into amino acids. Although a “stop codon” (TAG, TGA, or TAA) is not translated into an amino acid, it may be considered to be part of a coding region, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, and the like, are not part of a coding region. Two or more nucleic acids or nucleic acid fragments of the present invention can be present in a single polynucleotide construct, e.g., on a single plasmid, or in separate polynucleotide constructs, e.g., on separate (different) plasmids. Furthermore, any nucleic acid or nucleic acid fragment may encode a single IV polypeptide or fragment, derivative, or variant thereof, e.g., or may encode more than one polypeptide, e.g., a nucleic acid may encode two or more polypeptides. In addition, a nucleic acid may include a regulatory element such as a promoter, ribosome binding site, or a transcription terminator, or may encode heterologous coding regions fused to the IV coding region, e.g., specialized elements or motifs, such as a secretory signal peptide or a heterologous functional domain.
  • The terms “fragment,” “variant,” “derivative” and “analog” when referring to IV polypeptides of the present invention include any polypeptides which retain at least some of the immunogenicity or antigenicity of the corresponding native polypeptide. Fragments of IV polypeptides of the present invention include proteolytic fragments, deletion fragments and in particular, fragments of IV polypeptides which exhibit increased secretion from the cell or higher immunogenicity or reduced pathogenicity when delivered to an animal. Polypeptide fragments further include any portion of the polypeptide which comprises an antigenic or immunogenic epitope of the native polypeptide, including linear as well as three-dimensional epitopes. Variants of IV polypeptides of the present invention include fragments as described above, and also polypeptides with altered amino acid sequences due to amino acid substitutions, deletions, or insertions. Variants may occur naturally, such as an allelic variant. By an “allelic variant” is intended alternate forms of a gene occupying a given locus on a chromosome or genome of an organism or virus. Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985), which is incorporated herein by reference. For example, as used herein, variations in a given gene product. When referring to IV NA or HA proteins, each such protein is a “variant,” in that native IV strains are distinguished by the type of NA and HA proteins encoded by the virus. However, within a single HA or NA variant type, further naturally or non-naturally occurring variations such as amino acid deletions, insertions or substitutions may occur. Non-naturally occurring variants may be produced using art-known mutagenesis techniques. Variant polypeptides may comprise conservative or non-conservative amino acid substitutions, deletions or additions. Derivatives of IV polypeptides of the present invention, are polypeptides which have been altered so as to exhibit additional features not found on the native polypeptide. Examples include fusion proteins. An analog is another form of an IV polypeptide of the present invention. An example is a proprotein which can be activated by cleavage of the proprotein to produce an active mature polypeptide.
  • The terms “infectious polynucleotide” or “infectious nucleic acid” are intended to encompass isolated viral polynucleotides and/or nucleic acids which are solely sufficient to mediate the synthesis of complete infectious virus particles upon uptake by permissive cells. Thus, “infectious nucleic acids” do not require pre-synthesized copies of any of the polypeptides it encodes, e.g., viral replicases, in order to initiate its replication cycle in a permissive host cell.
  • The terms “non-infectious polynucleotide” or “non-infectious nucleic acid” as defined herein are polynucleotides or nucleic acids which cannot, without additional added materials, e.g, polypeptides, mediate the synthesis of complete infectious virus particles upon uptake by permissive cells. An infectious polynucleotide or nucleic acid is not made “non-infectious” simply because it is taken up by a non-permissive cell. For example, an infectious viral polynucleotide from a virus with limited host range is infectious if it is capable of mediating the synthesis of complete infectious virus particles when taken up by cells derived from a permissive host (i.e., a host permissive for the virus itself). The fact that uptake by cells derived from a non-permissive host does not result in the synthesis of complete infectious virus particles does not make the nucleic acid “non-infectious.” In other words, the term is not qualified by the nature of the host cell, the tissue type, or the species taking up the polynucleotide or nucleic acid fragment.
  • In some cases, an isolated infectious polynucleotide or nucleic acid may produce fully-infectious virus particles in a host cell population which lacks receptors for the virus particles, i.e., is non-permissive for virus entry. Thus viruses produced will not infect surrounding cells. However, if the supernatant containing the virus particles is transferred to cells which are permissive for the virus, infection will take place.
  • The terms “replicating polynucleotide” or “replicating nucleic acid” are meant to encompass those polynucleotides and/or nucleic acids which, upon being taken up by a permissive host cell, are capable of producing multiple, e.g., one or more copies of the same polynucleotide or nucleic acid. Infectious polynucleotides and nucleic acids are a subset of replicating polynucleotides and nucleic acids; the terms are not synonymous. For example, a defective virus genome lacking the genes for virus coat proteins may replicate, e.g., produce multiple copies of itself, but is NOT infectious because it is incapable of mediating the synthesis of complete infectious virus particles unless the coat proteins, or another nucleic acid encoding the coat proteins, are exogenously provided.
  • In certain embodiments, the polynucleotide, nucleic acid, or nucleic acid fragment is DNA. In the case of DNA, a polynucleotide comprising a nucleic acid which encodes a polypeptide normally also comprises a promoter and/or other transcription or translation control elments operably associated with the polypeptide-encoding nucleic acid fragment. An operable association is when a nucleic acid fragment encoding a gene product, e.g., a polypeptide, is associated with one or more regulatory sequences in such a way as to place expression of the gene product under the influence or control of the regulatory sequence(s). Two DNA fragments (such as a polypeptide-encoding nucleic acid fragment and a promoter associated with the 5′ end of the nucleic acid fragment) are “operably associated” if induction of promoter function results in the transcription of mRNA encoding the desired gene product and if the nature of the linkage between the two DNA fragments does not (1) result in the introduction of a frame-shift mutation, (2) interfere with the ability of the expression regulatory sequences to direct the expression of the gene product, or (3) interfere with the ability of the DNA template to be transcribed. Thus, a promoter region would be operably associated with a nucleic acid fragment encoding a polypeptide if the promoter was capable of effecting transcription of that nucleic acid fragment. The promoter may be a cell-specific promoter that directs substantial transcription of the DNA only in predetermined cells. Other transcription control elements, besides a promoter, for example enhancers, operators, repressors, and transcription termination signals, can be operably associated with the polynucleotide to direct cell-specific transcription. Suitable promoters and other transcription control regions are disclosed herein.
  • A variety of transcription control regions are known to those skilled in the art. These include, without limitation, transcription control regions which function in vertebrate cells, such as, but not limited to, promoter and enhancer segments from cytomegaloviruses (the immediate early promoter, in conjunction with intron-A), simian virus 40 (the early promoter), and retroviruses (such as Rous sarcoma virus). Other transcription control regions include those derived from vertebrate genes such as actin, heat shock protein, bovine growth hormone and rabbit β-globin, as well as other sequences capable of controlling gene expression in eukaryotic cells. Additional suitable transcription control regions include tissue-specific promoters and enhancers as well as lymphokine-inducible promoters (e.g., promoters inducible by interferons or interleukins).
  • Similarly, a variety of translation control elements are known to those of ordinary skill in the art. These include, but are not limited to ribosome binding sites, translation initiation and termination codons, elements from picornaviruses (particularly an internal ribosome entry site, or IRES, also referred to as a CITE sequence).
  • A DNA polynucleotide of the present invention may be a circular or linearized plasmid or vector, or other linear DNA which may also be non-infectious and nonintegrating (i.e., does not integrate into the genome of vertebrate cells). A linearized plasmid is a plasmid that was previously circular but has been linearized, for example, by digestion with a restriction endonuclease. Linear DNA may be advantageous in certain situations as discussed, e.g., in Cherng, J. Y., et al., J. Control. Release 60:343-53 (1999), and Chen, Z. Y., et al. Mol. Ther. 3:403-10 (2001), both of which are incorporated herein by reference. As used herein, the terms plasmid and vector can be used interchangeably
  • Alternatively, DNA virus genomes may be used to administer DNA polynucleotides into vertebrate cells. In certain embodiments, a DNA virus genome of the present invention is nonreplicative, noninfectious, and/or nonintegrating. Suitable DNA virus genomes include without limitation, herpesvirus genomes, adenovirus genomes, adeno-associated virus genomes, and poxvirus genomes. References citing methods for the in vivo introduction of non-infectious virus genomes to vertebrate tissues are well known to those of ordinary skill in the art, and are cited supra.
  • In other embodiments, a polynucleotide of the present invention is RNA, for example, in the form of messenger RNA (mRNA). Methods for introducing RNA sequences into vertebrate cells are described in U.S. Pat. No. 5,580,859, the disclosure of which is incorporated herein by reference in its entirety.
  • Polynucleotides, nucleic acids, and nucleic acid fragments of the present invention may be associated with additional nucleic acids which encode secretory or signal peptides, which direct the secretion of a polypeptide encoded by a nucleic acid fragment or polynucleotide of the present invention. According to the signal hypothesis, proteins secreted by mammalian cells have a signal peptide or secretory leader sequence which is cleaved from the mature protein once export of the growing protein chain across the rough endoplasmic reticulum has been initiated. Those of ordinary skill in the art are aware that polypeptides secreted by vertebrate cells generally have a signal peptide fused to the N-terminus of the polypeptide, which is cleaved from the complete or “full length” polypeptide to produce a secreted or “mature” form of the polypeptide. In certain embodiments, the native leader sequence is used, or a functional derivative of that sequence that retains the ability to direct the secretion of the polypeptide that is operably associated with it. Alternatively, a heterologous mammalian leader sequence, or a functional derivative thereof, may be used. For example, the wild-type leader sequence may be substituted with the leader sequence of human tissue plasminogen activator (TPA) or mouse β-glucuronidase.
  • In accordance with one aspect of the present invention, there is provided a polynucleotide construct, for example, a plasmid, comprising a nucleic acid fragment, where the nucleic acid fragment is a fragment of a codon-optimized coding region operably encoding an IV-derived polypeptide, where the coding region is optimized for expression in vertebrate cells, of a desired vertebrate species, e.g., humans, to be delivered to a vertebrate to be treated or immunized. Suitable IV polypeptides, or fragments, variants, or derivatives thereof may be derived from, but are not limited to, the IV HA, NA, NP, M1, or M2 proteins. Additional IV-derived coding sequences, e.g., coding for HA, NA, NP, M1, M2 or eM2, may also be included on the plasmid, or on a separate plasmid, and expressed, either using native IV codons or codons optimized for expression in the vertebrate to be treated or immunized. When such a plasmid encoding one or more optimized influenza sequences is delivered, in vivo to a tissue of the vertebrate to be treated or immunized, one or more of the encoded gene products will be expressed, i.e., transcribed and translated. The level of expression of the gene product(s) will depend to a significant extent on the strength of the associated promoter and the presence and activation of an associated enhancer element, as well as the degree of optimization of the coding region.
  • As used herein, the term “plasmid” refers to a construct made up of genetic material (i.e., nucleic acids). Typically a plasmid contains an origin of replication which is functional in bacterial host cells, e.g., Escherichia coli, and selectable markers for detecting bacterial host cells comprising the plasmid. Plasmids of the present invention may include genetic elements as described herein arranged such that an inserted coding sequence can be transcribed and translated in eukaryotic cells. Also, the plasmid may include a sequence from a viral nucleic acid. However, such viral sequences normally are not sufficient to direct or allow the incorporation of the plasmid into a viral particle, and the plasmid is therefore a non-viral vector. In certain embodiments described herein, a plasmid is a closed circular DNA molecule.
  • The term “expression” refers to the biological production of a product encoded by a coding sequence. In most cases a DNA sequence, including the coding sequence, is transcribed to form a messenger-RNA (mRNA). The messenger-RNA is then translated to form a polypeptide product which has a relevant biological activity. Also, the process of expression may involve further processing steps to the RNA product of transcription, such as splicing to remove introns, and/or post-translational processing of a polypeptide product.
  • As used herein, the term “polypeptide” is intended to encompass a singular “polypeptide” as well as plural “polypeptides,” and comprises any chain or chains of two or more amino acids. Thus, as used herein, terms including, but not limited to “peptide,” “dipeptide,” “tripeptide,” “protein,” “amino acid chain,” or any other term used to refer to a chain or chains of two or more amino acids, are included in the definition of a “polypeptide,” and the term “polypeptide” can be used instead of, or interchangeably with any of these terms. The term further includes polypeptides which have undergone post-translational modifications, for example, glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-naturally occurring amino acids.
  • Also included as polypeptides of the present invention are fragments, derivatives, analogs, or variants of the foregoing polypeptides, and any combination thereof. Polypeptides, and fragments, derivatives, analogs, or variants thereof of the present invention can be antigenic and immunogenic polypeptides related to IV polypeptides, which are used to prevent or treat, i.e., cure, ameliorate, lessen the severity of, or prevent or reduce contagion of infectious disease caused by the IV.
  • As used herein, an “antigenic polypeptide” or an “immunogenic polypeptide” is a polypeptide which, when introduced into a vertebrate, reacts with the vertebrate's immune system molecules, i.e., is antigenic, and/or induces an immune response in the vertebrate, i.e., is immunogenic. It is quite likely that an immunogenic polypeptide will also be antigenic, but an antigenic polypeptide, because of its size or conformation, may not necessarily be immunogenic. Examples of antigenic and immunogenic polypeptides of the present invention include, but are not limited to, e.g., HA or fragments or variants thereof, e.g. NP, or fragments thereof, e.g., PB1, or fragments or variants thereof, e.g., NS1 or fragments or variants thereof, e.g., M1 or fragments or variants thereof, and e.g. M2 or fragments or variants thereof including the extracellular fragment of M2 (eM2), or e.g., any of the foregoing polypeptides or fragments fused to a heterologous polypeptide, for example, a hepatitis B core antigen. Isolated antigenic and immunogenic polypeptides of the present invention in addition to those encoded by polynucleotides of the invention, may be provided as a recombinant protein, a purified subunit, a viral vector expressing the protein, or may be provided in the form of an inactivated IV vaccine, e.g., a live-attenuated virus vaccine, a heat-killed virus vaccine, etc.
  • By an “isolated” IV polypeptide or a fragment, variant, or derivative thereof is intended an IV polypeptide or protein that is not in its natural form. No particular level of purification is required. For example, an isolated IV polypeptide can be removed from its native or natural environment. Recombinantly produced IV polypeptides and proteins expressed in host cells are considered isolated for purposed of the invention, as are native or recombinant IV polypeptides which have been separated, fractionated, or partially or substantially purified by any suitable technique, including the separation of IV virions from eggs or culture cells in which they have been propagated. In addition, an isolated IV polypeptide or protein can be provided as a live or inactivated viral vector expressing an isolated IV polypeptide and can include those found in inactivated IV vaccine compositions. Thus, isolated IV polypeptides and proteins can be provided as, for example, recombinant IV polypeptides, a purified subunit of IV, a viral vector expressing an isolated IV polypeptide, or in the form of an inactivated or attenuated IV vaccine.
  • The term “epitopes,” as used herein, refers to portions of a polypeptide having antigenic or immunogenic activity in a vertebrate, for example a human. An “immunogenic epitope,” as used herein, is defined as a portion of a protein that elicits an immune response in an animal, as determined by any method known in the art. The term “antigenic epitope,” as used herein, is defined as a portion of a protein to which an antibody or T-cell receptor can immunospecifically bind as determined by any method well known in the art. Immunospecific binding excludes non-specific binding but does not exclude cross-reactivity with other antigens. Where all immunogenic epitopes are antigenic, antigenic epitopes need not be immunogenic.
  • The term “immunogenic carrier” as used herein refers to a first polypeptide or fragment, variant, or derivative thereof which enhances the immunogenicity of a second polypeptide or fragment, variant, or derivative thereof. Typically, an “immunogenic carrier” is fused to or conjugated to the desired polypeptide or fragment thereof. An example of an “immunogenic carrier” is a recombinant hepatitis B core antigen expressing, as a surface epitope, an immunogenic epitope of interest. See, e.g., European Patent No. EP 0385610 B 1, which is incorporated herein by reference in its entirety.
  • In the present invention, antigenic epitopes preferably contain a sequence of at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, or between about 8 to about 30 amino acids contained within the amino acid sequence of an IV polypeptide of the invention, e.g., an NP polypeptide, an M1 polypeptide or an M2 polypeptide. Certain polypeptides comprising immunogenic or antigenic epitopes are at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length. Antigenic as well as immunogenic epitopes may be linear, i.e., be comprised of contiguous amino acids in a polypeptide, or may be three dimensional, i.e., where an epitope is comprised of non-contiguous amino acids which come together due to the secondary or tertiary structure of the polypeptide, thereby forming an epitope.
  • As to the selection of peptides or polypeptides bearing an antigenic epitope (e.g., that contain a region of a protein molecule to which an antibody or T cell receptor can bind), it is well known in that art that relatively short synthetic peptides that mimic part of a protein sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked protein. See, e.g., Sutcliffe, J. G., et al., Science 219:660-666 (1983), which is herein incorporated by reference.
  • Peptides capable of eliciting an immunogenic response are frequently represented in the primary sequence of a protein, can be characterized by a set of simple chemical rules, and are confined neither to immunodominant regions of intact proteins nor to the amino or carboxyl terminals. Peptides that are extremely hydrophobic and those of six or fewer residues generally are ineffective at inducing antibodies that bind to the mimicked protein; longer peptides, especially those containing proline residues, usually are effective. Sutcliffe et al., supra, at 661. For instance, 18 of 20 peptides designed according to these guidelines, containing 8-39 residues covering 75% of the sequence of the IV hemagglutinin HA1 polypeptide chain, induced antibodies that reacted with the HA1 protein or intact virus; and 12/12 peptides from the MuLV polymerase and 18/18 from the rabies glycoprotein induced antibodies that precipitated the respective proteins.
  • Codon Optimization
  • “Codon optimization” is defined as modifying a nucleic acid sequence for enhanced expression in the cells of the vertebrate of interest, e.g. human, by replacing at least one, more than one, or a significant number, of codons of the native sequence with codons that are more frequently or most frequently used in the genes of that vertebrate. Various species exhibit particular bias for certain codons of a particular amino acid.
  • In one aspect, the present invention relates to polynucleotides comprising nucleic acid fragments of codon-optimized coding regions which encode IV polypeptides, or fragments, variants, or derivatives thereof, with the codon usage adapted for optimized expression in the cells of a given vertebrate, e.g., humans. These polynucleotides are prepared by incorporating codons preferred for use in the genes of the vertebrate of interest into the DNA sequence. Also provided are polynucleotide expression constructs, vectors, and host cells comprising nucleic acid fragments of codon-optimized coding regions which encode IV polypeptides, and fragments, variants, or derivatives thereof, and various methods of using the polynucleotide expression constructs, vectors, host cells to treat or prevent influenza disease in a vertebrate.
  • As used herein the term “codon-optimized coding region” means a nucleic acid coding region that has been adapted for expression in the cells of a given vertebrate by replacing at least one, or more than one, or a significant number, of codons with one or more codons that are more frequently used in the genes of that vertebrate.
  • Deviations in the nucleotide sequence that comprise the codons encoding the amino acids of any polypeptide chain allow for variations in the sequence coding for the gene. Since each codon consists of three nucleotides, and the nucleotides comprising DNA are restricted to four specific bases, there are 64 possible combinations of nucleotides, 61 of which encode amino acids (the remaining three codons encode signals ending translation). The “genetic code” which shows which codons encode which amino acids is reproduced herein as Table 1. As a result, many amino acids are designated by more than one codon. For example, the amino acids alanine and proline are coded for by four triplets, serine and arginine by six, whereas tryptophan and methionine are coded by just one triplet. This degeneracy allows for DNA base composition to vary over a wide range without altering the amino acid sequence of the proteins encoded by the DNA.
    TABLE 1
    The Standard Genetic Code
    T C A G
    T TTT Phe (F) TCT Ser (S) TAT Tyr (Y) TGT Cys (C)
    TTC Phe (F) TCC Ser (S) TAC Tyr (Y) TGC
    TTA Leu (L) TCA Ser (S) TAA Ter TGA Ter
    TTG Leu (L) TCG Ser (S) TAG Ter TGG Trp (W)
    C CTT Leu (L) CCT Pro (P) CAT His (H) CGT Arg (R)
    CTC Leu (L) CCC Pro (P) CAC His (H) CGC Arg (R)
    CTA Leu (L) CCA Pro (P) CAA Gln (Q) CGA Arg (R)
    CTG Leu (L) CCG Pro (P) CAG Gln (Q) CGG Arg (R)
    A ATT Ile (I) ACT Thr (T) AAT Asn (N) AGT Ser (S)
    ATC Ile (I) ACC Thr (T) AAC Asn (N) AGC Ser (S)
    ATA Ile (I) ACA Thr (T) AAA Lys (K) AGA Arg (R)
    ATG Met (M) ACG Thr (T) AAG Lys (K) AGG Arg (R)
    G GTT Val (V) GCT Ala (A) GAT Asp (D) GGT Gly (G)
    GTC Val (V) GCC Ala (A) GAC Asp (D) GGC Gly (G)
    GTA Val (V) GCA Ala (A) GAA Glu (E) GGA Gly (G)
    GTG Val (V) GCG Ala (A) GAG Glu (E) GGG Gly (G)
  • Many organisms display a bias for use of particular codons to code for insertion of a particular amino acid in a growing peptide chain. Codon preference or codon bias, differences in codon usage between organisms, is afforded by degeneracy of the genetic code, and is well documented among many organisms. Codon bias often correlates with the efficiency of translation of messenger RNA (mRNA), which is in turn believed to be dependent on, inter alia, the properties of the codons being translated and the availability of particular transfer RNA (tRNA) molecules. The predominance of selected tRNAs in a cell is generally a reflection of the codons used most frequently in peptide synthesis. Accordingly, genes can be tailored for optimal gene expression in a given organism based on codon optimization.
  • Given the large number of gene sequences available for a wide variety of animal, plant and microbial species, it is possible to calculate the relative frequencies of codon usage. Codon usage tables are readily available, for example, at the “Codon Usage Database” available at http://www.kazusa.or.jp/codon/ (visited Jul. 9, 2002), and these tables can be adapted in a number of ways. See Nakamura, Y., et al. “Codon usage tabulated from the international DNA sequence databases: status for the year 2000” Nucl. Acids Res. 28:292 (2000), which is incorporated by reference. As examples, the codon usage tables for human, mouse, domestic cat, and cow, calculated from GenBank Release 128.0 (15 Feb. 2002), are reproduced below as Tables 2-5. These Tables use mRNA nomenclature, and so instead of thymine (T) which is found in DNA, the Tables use uracil (U) which is found in RNA. The Tables have been adapted so that frequencies are calculated for each amino acid, rather than for all 64 codons.
    TABLE 2
    Codon Usage Table for Human Genes (Homo sapiens)
    Amino Acid Codon Number Frequency
    Phe UUU 326146 0.4525
    Phe UUC 394680 0.5475
    Total 720826
    Leu UUA 139249 0.0728
    Leu UUG 242151 0.1266
    Leu CUU 246206 0.1287
    Leu CUC 374262 0.1956
    Leu CUA 133980 0.0700
    Leu CUG 777077 0.4062
    Total 1912925
    Ile AUU 303721 0.3554
    Ile AUC 414483 0.4850
    Ile AUA 136399 0.1596
    Total 854603
    Met AUG 430946 1.0000
    Total 430946
    Val GUU 210423 0.1773
    Val GUC 282445 0.2380
    Val GUA 134991 0.1137
    Val GUG 559044 0.4710
    Total 1186903
    Ser UCU 282407 0.1840
    Ser UCC 336349 0.2191
    Ser UCA 225963 0.1472
    Ser UCG 86761 0.0565
    Ser AGU 230047 0.1499
    Ser AGC 373362 0.2433
    Total 1534889
    Pro CCU 333705 0.2834
    Pro CCC 386462 0.3281
    Pro CCA 322220 0.2736
    Pro CCG 135317 0.1149
    Total 1177704
    Thr ACU 247913 0.2419
    Thr ACC 371420 0.3624
    Thr ACA 285655 0.2787
    Thr ACG 120022 0.1171
    Total 1025010
    Ala GCU 360146 0.2637
    Ala GCC 551452 0.4037
    Ala GCA 308034 0.2255
    Ala GCG 146233 0.1071
    Total 1365865
    Tyr UAU 232240 0.4347
    Tyr UAC 301978 0.5653
    Total 534218
    His CAU 201389 0.4113
    His CAC 288200 0.5887
    Total 489589
    Gln CAA 227742 0.2541
    Gln CAG 668391 0.7459
    Total 896133
    Asn AAU 322271 0.4614
    Asn AAC 376210 0.5386
    Total 698481
    Lys AAA 462660 0.4212
    Lys AAG 635755 0.5788
    Total 1098415
    Asp GAU 430744 0.4613
    Asp GAC 502940 0.5387
    Total 933684
    Glu GAA 561277 0.4161
    Glu GAG 787712 0.5839
    Total 1348989
    Cys UGU 190962 0.4468
    Cys UGC 236400 0.5532
    Total 427362
    Trp UGG 248083 1.0000
    Total 248083
    Arg CGU 90899 0.0830
    Arg CGC 210931 0.1927
    Arg CGA 122555 0.1120
    Arg CGG 228970 0.2092
    Arg AGA 221221 0.2021
    Arg AGG 220119 0.2011
    Total 1094695
    Gly GGU 209450 0.1632
    Gly GGC 441320 0.3438
    Gly GGA 315726 0.2459
    Gly GGG 317263 0.2471
    Total 1283759
    Stop UAA 13963
    Stop UAG 10631
    Stop UGA 24607
  • TABLE 3
    Codon Usage Table for Mouse Genes (Mus musculus)
    Amino Acid Codon Number Frequency
    Phe UUU 150467 0.4321
    Phe UUC 197795 0.5679
    Total 348262
    Leu UUA 55635 0.0625
    Leu UUG 116210 0.1306
    Leu CUU 114699 0.1289
    Leu CUC 179248 0.2015
    Leu CUA 69237 0.0778
    Leu CUG 354743 0.3987
    Total 889772
    Ile AUU 137513 0.3367
    Ile AUC 208533 0.5106
    Ile AUA 62349 0.1527
    Total 408395
    Met AUG 204546 1.0000
    Total 204546
    Val GUU 93754 0.1673
    Val GUC 140762 0.2513
    Val GUA 64417 0.1150
    Val GUG 261308 0.4664
    Total 560241
    Ser UCU 139576 0.1936
    Ser UCC 160313 0.2224
    Ser UCA 100524 0.1394
    Ser UCG 38632 0.0536
    Ser AGU 108413 0.1504
    Ser AGC 173518 0.2407
    Total 720976
    Pro CCU 162613 0.3036
    Pro CCC 164796 0.3077
    Pro CCA 151091 0.2821
    Pro CCG 57032 0.1065
    Total 535532
    Thr ACU 119832 0.2472
    Thr ACC 172415 0.3556
    Thr ACA 140420 0.2896
    Thr ACG 52142 0.1076
    Total 484809
    Ala GCU 178593 0.2905
    Ala GCC 236018 0.3839
    Ala GCA 139697 0.2272
    Ala GCG 60444 0.0983
    Total 614752
    Tyr UAU 108556 0.4219
    Tyr UAC 148772 0.5781
    Total 257328
    His CAU 88786 0.3973
    His CAC 134705 0.6027
    Total 223491
    Gln CAA 101783 0.2520
    Gln CAG 302064 0.7480
    Total 403847
    Asn AAU 138868 0.4254
    Asn AAC 187541 0.5746
    Total 326409
    Lys AAA 188707 0.3839
    Lys AAG 302799 0.6161
    Total 491506
    Asp GAU 189372 0.4414
    Asp GAC 239670 0.5586
    Total 429042
    Glu GAA 235842 0.4015
    Glu GAG 351582 0.5985
    Total 587424
    Cys UGU 97385 0.4716
    Cys UGC 109130 0.5284
    Total 206515
    Trp UGG 112588 1.0000
    Total 112588
    Arg CGU 41703 0.0863
    Arg CGC 86351 0.1787
    Arg CGA 58928 0.1220
    Arg CGG 92277 0.1910
    Arg AGA 101029 0.2091
    Arg AGG 102859 0.2129
    Total 483147
    Gly GGU 103673 0.1750
    Gly GGC 198604 0.3352
    Gly GGA 151497 0.2557
    Gly GGG 138700 0.2341
    Total 592474
    Stop UAA 5499
    Stop UAG 4661
    Stop UGA 10356
  • TABLE 4
    Codon Usage Table for Domestic Cat Genes (Felis cattus)
    Amino Acid Codon Number Frequency of usage
    Phe UUU 1204.00 0.4039
    Phe UUC 1777.00 0.5961
    Total 2981
    Leu UUA 404.00 0.0570
    Leu UUG 857.00 0.1209
    Leu CUU 791.00 0.1116
    Leu CUC 1513.00 0.2135
    Leu CUA 488.00 0.0688
    Leu CUG 3035.00 0.4282
    Total 7088
    Ile AUU 1018.00 0.2984
    Ile AUC 1835.00 0.5380
    Ile AUA 558.00 0.1636
    Total 3411
    Met AUG 1553.00 0.0036
    Total 1553
    Val GUU 696.00 0.1512
    Val GUC 1279.00 0.2779
    Val GUA 463.00 0.1006
    Val GUG 2164.00 0.4702
    Total 4602
    Ser UCU 940.00 0.1875
    Ser UCC 1260.00 0.2513
    Ser UCA 608.00 0.1213
    Ser UCG 332.00 0.0662
    Ser AGU 672.00 0.1340
    Ser AGC 1202.00 0.2397
    Total 5014
    Pro CCU 958.00 0.2626
    Pro CCC 1375.00 0.3769
    Pro CCA 850.00 0.2330
    Pro CCG 465.00 0.1275
    Total 3648
    Thr ACU 822.00 0.2127
    Thr ACC 1574.00 0.4072
    Thr ACA 903.00 0.2336
    Thr ACG 566.00 0.1464
    Total 3865
    Ala GCU 1129.00 0.2496
    Ala GCC 1951.00 0.4313
    Ala GCA 883.00 0.1952
    Ala GCG 561.00 0.1240
    Total 4524
    Tyr UAU 837.00 0.3779
    Tyr UAC 1378.00 0.6221
    Total 2215
    His CAU 594.00 0.3738
    His CAC 995.00 0.6262
    Total 1589
    Gln CAA 747.00 0.2783
    Gln CAG 1937.00 0.7217
    Total 2684
    Asn AAU 1109.00 0.3949
    Asn AAC 1699.00 0.6051
    Total 2808
    Lys AAA 1445.00 0.4088
    Lys AAG 2090.00 0.5912
    Total 3535
    Asp GAU 1255.00 0.4055
    Asp GAC 1840.00 0.5945
    Total 3095
    Glu GAA 1637.00 0.4164
    Glu GAG 2294.00 0.5836
    Total 3931
    Cys UGU 719.00 0.4425
    Cys UGC 906.00 0.5575
    Total 1625
    Trp UGG 1073.00 1.0000
    Total 1073
    Arg CGU 236.00 0.0700
    Arg CGC 629.00 0.1865
    Arg CGA 354.00 0.1050
    Arg CGG 662.00 0.1963
    Arg AGA 712.00 0.2112
    Arg AGG 779.00 0.2310
    Total 3372
    Gly GGU 648.00 0.1498
    Gly GGC 1536.00 0.3551
    Gly GGA 1065.00 0.2462
    Gly GGG 1077.00 0.2490
    Total 4326
    Stop UAA 55
    Stop UAG 36
    Stop UGA 110
  • TABLE 5
    Codon Usage Table for Cow Genes (Bos taurus)
    Amino Acid Codon Number Frequency of usage
    Phe UUU 13002 0.4112
    Phe UUC 18614 0.5888
    Total 31616
    Leu UUA 4467 0.0590
    Leu UUG 9024 0.1192
    Leu CUU 9069 0.1198
    Leu CUC 16003 0.2114
    Leu CUA 4608 0.0609
    Leu CUG 32536 0.4298
    Total 75707
    Ile AUU 12474 0.3313
    Ile AUC 19800 0.5258
    Ile AUA 5381 0.1429
    Total 37655
    Met AUG 17770 1.0000
    Total 17770
    Val GUU 8212 0.1635
    Val GUC 12846 0.2558
    Val GUA 4932 0.0982
    Val GUG 24222 0.4824
    Total 50212
    Ser UCU 10287 0.1804
    Ser UCC 13258 0.2325
    Ser UCA 7678 0.1347
    Ser UCG 3470 0.0609
    Ser AGU 8040 0.1410
    Ser AGC 14279 0.2505
    Total 57012
    Pro CCU 11695 0.2684
    Pro CCC 15221 0.3493
    Pro CCA 11039 0.2533
    Pro CCG 5621 0.1290
    Total 43576
    Thr ACU 9372 0.2203
    Thr ACC 16574 0.3895
    Thr ACA 10892 0.2560
    Thr ACG 5712 0.1342
    Total 42550
    Ala GCU 13923 0.2592
    Ala GCC 23073 0.4295
    Ala GCA 10704 0.1992
    Ala GCG 6025 0.1121
    Total 53725
    Tyr UAU 9441 0.3882
    Tyr UAC 14882 0.6118
    Total 24323
    His CAU 6528 0.3649
    His CAC 11363 0.6351
    Total 17891
    Gln CAA 8060 0.2430
    Gln CAG 25108 0.7570
    Total 33168
    Asn AAU 12491 0.4088
    Asn AAC 18063 0.5912
    Total 30554
    Lys AAA 17244 0.3897
    Lys AAG 27000 0.6103
    Total 44244
    Asp GAU 16615 0.4239
    Asp GAC 22580 0.5761
    Total 39195
    Glu GAA 21102 0.4007
    Glu GAG 31555 0.5993
    Total 52657
    Cys UGU 7556 0.4200
    Cys UGC 10436 0.5800
    Total 17992
    Trp UGG 10706 1.0000
    Total 10706
    Arg CGU 3391 0.0824
    Arg CGC 7998 0.1943
    Arg CGA 4558 0.1108
    Arg CGG 8300 0.2017
    Arg AGA 8237 0.2001
    Arg AGG 8671 0.2107
    Total 41155
    Gly GGU 8508 0.1616
    Gly GGC 18517 0.3518
    Gly GGA 12838 0.2439
    Gly GGG 12772 0.2427
    Total 52635
    Stop UAA 555
    Stop UAG 394
    Stop UGA 392
  • By utilizing these or similar tables, one of ordinary skill in the art can apply the frequencies to any given polypeptide sequence, and produce a nucleic acid fragment of a codon-optimized coding region which encodes the polypeptide, but which uses codons more optimal for a given species. Codon-optimized coding regions can be designed by various different methods.
  • In one method, termed “uniform optimization,” a codon usage table is used to find the single most frequent codon used for any given amino acid, and that codon is used each time that particular amino acid appears in the polypeptide sequence. For example, referring to Table 2 above, for leucine, the most frequent codon in humans is CUG, which is used 41% of the time. Thus all the leucine residues in a given amino acid sequence would be assigned the codon CUG. A coding region for IAV NP (SEQ ID NO:2) optimized by the “uniform optimization” method is presented herein as SEQ ID NO 24:
    1 ATGGCCAGCC AGGGCACCAA GCGGAGCTAC GAGCAGATGG AGACCGACGG CGAGCGGCAG
    61 AACGCCACCG AGATCCGGGC CAGCGTGGGC AAGATGATCG GCGGCATCGG CCGGTTCTAC
    121 ATCCAGATGT GCACCGAGCT GAAGCTGAGC GACTACGAGG GCCGGCTGAT CCAGAACAGC
    181 CTGACCATCG AGCGGATGGT GCTGAGCGCC TTCGACGAGC GGCGGAACAA GTACCTGGAG
    241 GAGCACCCCA GCGCCGGCAA GGACCCCAAG AAGACCGGCG GCCCCATCTA CCGGCGGGTG
    301 AACGGCAAGT GGATGCGGGA GCTGATCCTG TACGACAAGG AGGAGATCCG GCGGATCTGG
    361 CGGCAGGCCA ACAACGGCGA CGACGCCACC GCCGGCCTGA CCCACATGAT GATCTGGCAC
    421 AGCAACCTGA ACGACGCCAC CTACCAGCGG ACCCGGGCCC TGGTGCGGAC CGGCATGGAC
    481 CCCCGGATGT GCAGCCTGAT GCAGGGCAGC ACCCTGCCCC GGCGGAGCGG CGCCGCCGGC
    541 GCCGCCGTGA AGGGCGTGGG CACCATGGTG ATGGAGCTGG TGCGGATGAT CAAGCGGGGC
    601 ATCAACGACC GGAACTTCTG GCGGGGCGAG AACGGCCGGA AGACCCGGAT CGCCTACGAG
    661 CGGATGTGCA ACATCCTGAA GGGCAAGTTC CAGACCGCCG CCCAGAAGGC CATGATGGAC
    721 CAGGTGCGGG AGAGCCGGAA CCCCGGCAAC GCCGAGTTCG AGGACCTGAC CTTCCTGGCC
    781 CGGAGCGCCC TGATCCTGCG GGGCAGCGTG GCCCACAAGA GCTGCCTGCC CGCCTGCGTG
    841 TACGGCCCCG CCGTGGCCAG CGGCTACGAC TTCGAGCGGG AGGOCTACAG CCTGGTGGGC
    901 ATCGACCCCT TCCGGCTGCT GCAGAACAGC CAGGTGTACA GCCTGATCCG GCCCAACGAG
    961 AACCCCGCCC ACAAGAGCCA GCTGGTGTGG ATGGCCTGCC ACAGCGCCGC CTTCGAGGAC
    1021 CTGCGGGTGC TGAGCTTCAT CAAGGGCACC AAGGTGCTGC CCCGGGGCAA GCTGAGCACC
    1081 CGGGGCGTGC AGATCGCCAG CAACGAGAAC ATGGAGACCA TGGAGAGCAG CACCCTGGAG
    1141 CTGCGGAGCC GGTACTGGGC CATCCGGACC CGGAGCGGCG GCAACACCAA CCAGCAGCGG
    1201 GCCAGCGCCG GCCAGATCAG CATCCAGCCC ACCTTCAGCG TGCAGCGGAA CCTGCCCTTC
    1261 GACCGGACCA CCGTGATGGC CGCCTTCAGC GGCAACACCG AGGGCCGGAC CAGCGACATG
    1321 CGGACCGAGA TCATCCGGAT GATGGAGAGC GCCCGGCCCG AGGACGTGAG CTTCCAGGGC
    1381 CGGGGCGTGT TCGAGCTGAG CGACGAGAAG GCCGCCAGCC CCATCGTGCC CAGCTTCGAC
    1441 ATGAGCAACG AGGGCAGCTA CTTCTTCGGC GACAACGCCG AGGAGTACGA CAACTGA
  • In another method, termed “full-optimization,” the actual frequencies of the codons are distributed randomly throughout the coding region. Thus, using this method for optimization, if a hypothetical polypeptide sequence had 100 leucine residues, referring to Table 2 for frequency of usage in humans, about 7, or 7% of the leucine codons would be UUA, about 13, or 13% of the leucine codons would be WUG, about 13, or 13% of the leucine codons would be CUU, about 20, or 20% of the leucine codons would be CUC, about 7, or 7% of the leucine codons would be CUA, and about 41, or 41% of the leucine codons would be CUG. These frequencies would be distributed randomly throughout the leucine codons in the coding region encoding the hypothetical polypeptide. As will be understood by those of ordinary skill in the art, the distribution of codons in the sequence can vary significantly using this method; however, the sequence always encodes the same polypeptide.
  • As an example, a nucleotide sequence for NP (SEQ ID NO:2) fully optimized for human codon usage, is shown as SEQ ID NO:23. An alignment of nucleotides 46-1542 of SEQ ID NO:1 (native NP coding region) with the codon-optimized coding region (SEQ ID NO:23) is presented in FIG. 1.
  • In using the “full-optimization” method, an entire polypeptide sequence may be codon-optimized as described above. With respect to various desired fragments, variants or derivatives of the complete polypeptide, the fragment variant, or derivative may first be designed, and is then codon-optimized individually. Alternatively, a full-length polypeptide sequence is codon-optimized for a given species resulting in a codon-optimized coding region encoding the entire polypeptide, and then nucleic acid fragments of the codon-optimized coding region, which encode fragments, variants, and derivatives of the polypeptide are made from the original codon-optimized coding region. As would be well understood by those of ordinary skill in the art, if codons have been randomly assigned to the full-length coding region based on their frequency of use in a given species, nucleic acid fragments encoding fragments, variants, and derivatives would not necessarily be fully codon-optimized for the given species. However, such sequences are still much closer to the codon usage of the desired species than the native codon usage. The advantage of this approach is that synthesizing codon-optimized nucleic acid fragments encoding each fragment, variant, and derivative of a given polypeptide, although routine, would be time consuming and would result in significant expense.
  • When using the “full-optimization” method, the term “about” is used precisely to account for fractional percentages of codon frequencies for a given amino acid. As used herein, “about” is defined as one amino acid more or one amino acid less than the value given. The whole number value of amino acids is rounded up if the fractional frequency of usage is 0.50 or greater, and is rounded down if the fractional frequency of use is 0.49 or less. Using again the example of the frequency of usage of leucine in human genes for a hypothetical polypeptide having 62 leucine residues, the fractional frequency of codon usage would be calculated by multiplying 62 by the frequencies for the various codons. Thus, 7.28 percent of 62 equals 4.51 UUA codons, or “about 5,” i.e., 4, 5, or 6 UUA codons, 12.66 percent of 62 equals 7.85 UUG codons or “about 8,” i.e., 7, 8, or 9 TUG codons, 12.87 percent of 62 equals 7.98 CUU codons, or “about 8,” i.e., 7, 8, or 9 CTU codons, 19.56 percent of 62 equals 12.13 CUC codons or “about 12,” i.e., 11, 12, or 13 CUC codons, 7.00 percent of 62 equals 4.34 CUA codons or “about 4,” i.e., 3, 4, or 5 CUA codons, and 40.62 percent of 62 equals 25.19 CUG codons, or “about 25,” i.e., 24, 25, or 26 CUG codons.
  • In a third method termed “minimal optimization,” coding regions are only partially optimized. For example, the invention includes a nucleic acid fragment of a codon-optimized coding region encoding a polypeptide in which at least about 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% of the codon positions have been codon-optimized for a given species. That is, they contain a codon that is preferentially used in the genes of a desired species, e.g., a vertebrate species, e.g., humans, in place of a codon that is normally used in the native nucleic acid sequence. Codons that are rarely found in the genes of the vertebrate of interest are changed to codons more commonly utilized in the coding regions of the vertebrate of interest.
  • Thus, those codons which are used more frequently in the IV gene of interest than in genes of the vertebrate of interest are substituted with more frequently-used codons. The difference in frequency at which the IV codons are substituted may vary based on a number factors as discussed below. For example, codons used at least twice more per thousand in IV genes as compared to genes of the vertebrate of interest are substituted with the most frequently used codon for that amino acid in the vertebrate of interest. This ratio may be adjusted higher or lower depending on various factors such as those discussed below. Accordingly, a codon in an IV native coding region would be substituted with a codon used more frequently for that amino acid in coding regions of the vertebrate of interest if the codon is used 1.1 times, 1.2 times, 1.3 times, 1.4 times, 1.5 times, 1.6 times, 1.7 times, 1.8 times, 1.9 times, 2.0 times, 2.1 times, 2.2 times, 2.3 times, 2.4 times, 2.5 times, 2.6 times, 2.7 times, 2.8 times, 2.9 times, 3.0 times, 3.1 times, 3.2 times, 3.3. times, 3.4 times, 3.5 times, 3.6 times. 3.7 times, 3.8 times, 3.9 times, 4.0 times, 4.1 times, 4.2 times, 4.3 times, 4.4 times, 4.5 times, 4.6 times, 4.7 times, 4.8 times, 4.9 times, 5.0 times, 5.5 times, 6.0 times, 6.5 times, 7.0 times, 7.5 times, 8.0 times, 8.5 times, 9.0 times, 9.5 times, 10.0 times, 10.5 times, 11.0 times, 11.5 times, 12.0 times, 12.5 times, 13.0 times, 13.5 times, 14.0 times, 14.5 times, 15.0 times, 15.5 times, 16.0 times, 16.5 times, 17.0 times, 17.5 times, 18.0 times, 18.5 times, 19.0 times, 19.5 times, 20 times, 21 times, 22 times, 23 times, 24 times, 25 times, or greater more frequently in IV coding regions than in coding regions of the vertebrate of interest.
  • This minimal human codon optimization for highly variant codons has several advantages, which include but are not limited to the following examples. Since fewer changes are made to the nucleotide sequence of the gene of interest, fewer manipulations are required, which leads to reduced risk of introducing unwanted mutations and lower cost, as well as allowing the use of commercially available site-directed mutagenesis kits, and reducing the need for expensive oligonucleotide synthesis. Further, decreasing the number of changes in the nucleotide sequence decreases the potential of altering the secondary structure of the sequence, which can have a significant impact on gene expression in certain host cells. The introduction of undesirable restriction sites is also reduced, facilitating the subcloning of the genes of interest into the plasmid expression vector.
  • The present invention also provides isolated polynucleotides comprising coding regions of IV polypeptides, e.g., NP, M1, M2, HA, NA, PB1, PB2, PA, NS1 or NS2, or fragments, variants, or derivatives thereof. The isolated polynucleotides can also be codon-optimized.
  • In certain embodiments described herein, a codon-optimized coding region encoding SEQ ID NO:2 is optimized according to codon usage in humans (Homo sapiens). Alternatively, a codon-optimized coding region encoding SEQ ID NO:2 may be optimized according to codon usage in any plant, animal, or microbial species. Codon-optimized coding regions encoding SEQ ID NO:2, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:2 is shown in Table 6.
    TABLE 6
    Number in
    AMINO ACID SEQ ID NO: 2
    A Ala 39
    R Arg 49
    C Cys 6
    G Gly 41
    H His 6
    I Ile 26
    L Leu 33
    K Lys 21
    M Met 25
    F Phe 18
    P Pro 17
    S Ser 40
    T Thr 28
    W Trp 6
    Y Tyr 15
    V Val 23
    N Asn 26
    D Asp 22
    Q Gln 21
    E Glu 36
  • Using the amino acid composition shown in Table 6, a human codon-optimized coding region which encodes SEQ ID NO:2 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid. According to this method, codons are assigned to the coding region encoding SEQ ID NO:2 as follows: the 18 phenylalanine codons are TTC, the 33 leucine codons are CTG, the 26 isoleucine codons are ATC, the 25 methionine codons are ATG, the 23 valine codons are GTG, the 40 serine codons are AGC, the 17 proline codons are CCC, the 28 threonine codons are ACC, the 39 alanine codons are GCC, the 15 tyrosine codons are TAC, the 6 histidine codons are CAC, the 21 glutamine codons are CAG, the 26 asparagine codons are AAC, the 21 lysine codons are AAG, the 22 aspartic acid codons are GAC, the 36 glutamic acid codons are GAG, the 6 tryptophan codons are TGG, the 49 arginine codons are CGG, AGA, or AGG (the frequencies of usage of these three codons in the human genome are not significantly different), and the 41 glycine codons are GGC.
  • Alternatively, a human codon-optimized coding region which encodes SEQ ID NO:2 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 6 above. Using this latter method, codons are assigned to the coding region encoding SEQ ID NO:2 as follows: about 8 of the 18 phenylalanine codons are TTT, and about 10 of the phenylalanine codons are TTC; about 2 of the 33 leucine codons are TTA, about 4 of the leucine codons are TTG, about 4 of the leucine codons are CTT, about 6 of the leucine codons are CTC, about 2 of the leucine codons are CTA, and about 13 of the leucine codons are CTG; about 9 of the 26 isoleucine codons are ATT, about 13 of the isoleucine codons are ATC, and about 4 of the isoleucine codons are ATA; the 25 methionine codons are ATG; about 4 of the 23 valine codons are GTT, about 5 of the valine codons are GTG, about 3 of the valine codons are GTA, and about 11 of the valine codons are GTG; about 7 of the 40 serine codons are TCT, about 9 of the serine codons are TCC, about 6 of the serine codons are TCA, about 2 of the serine codons are TCG, about 6 of the serine codons are AGT, and about 10 of the serine codons are AGC; about 5 of the 17 proline codons are CCT, about 6 of the proline codons are CCC, about 5 of the proline codons are CCA, and about 2 of the proline codons are CCG; about 7 of the 28 threonine codons are ACT, about 10 of the threonine codons are ACC, about 8 of the threonine codons are ACA, and about 3 of the threonine codons are ACG; about 10 of the 39 alanine codons are GCT, about 16 of the alanine codons are GCC, about 9 of the alanine codons are GCA, and about 4 of the alanine codons are GCG; about 7 of the 15 tyrosine codons are TAT and about 8 of the tyrosine codons are TAC; about 2 of the 6 histidine codons are CAT and about 4 of the histidine codons are CAC; about 5 of the 21 glutamine codons are CAA and about 16 of the glutamine codons are CAG; about 12 of the 26 asparagine codons are AAT and about 14 of the asparagine codons are AAC; about 9 of the 21 lysine codons are AAA and about 12 of the lysine codons are AAG; about 10 of the 22 aspartic acid codons are GAT and about 12 of the aspartic acid codons are GAC; about 11 of the 26 glutamic acid codons are GAA and about 15 of the glutanic acid codons are GAG; about 3 of the 6 cysteine codons are TGT and about 3 of the cysteine codons are TGC; the 6 tryptophan codons are TGG; about 4 of the 49 arginine codons are CGT, about 9 of the arginine codons are CGC, about 5 of the arginine codons are CGA, about 10 of the arginine codons are CGG, about 10 of the arginine codons are AGA, and about 10 of the arginine codons are AGG; and about 7 of the 41 glycine codons are GGT, about 14 of the glycine codons are GGC, about 10 of the glycine codons are GGA, and about 10 of the glycine codons are GGG.
  • As described above, the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:2, optimized according to codon usage in humans is presented herein as SEQ ID NO:23.
  • Additionally, a minimally codon-optimized nucleotide sequence encoding SEQ ID NO:2 can be designed by changing only certain codons found more frequently in IV genes than in human genes, as shown in Table 7. For example, if it is desired to substitute more frequently used codons in humans for those codons that occur at least 2 times more frequently in IV genes (designated with an asterisk in Table 7), Arg AGA, which occurs 2.3 times more frequently in IV genes than in human genes, is changed to, e.g., CGG; Asn AAT, which occurs 2.0 times more frequently in IV genes than in human genes, is changed to, e.g., AAC; Ile ATA, which occurs 3.6 times more frequently in IV genes than in human genes, is changed to, e.g., ATC; and Leu CTA, which occurs 2.0 times more frequently in IV genes than is human, is changed to, e.g., CTG.
    TABLE 7
    Codon Usage Table for Human Genes and IV Genes
    Amino Acid Codon Human IV
    Ala A GCA 16 25
    GCG 8  5
    GCC 19 11
    GCT 19 15
    Arg R AGA 12  28*
    AGG 11 14
    CGA 6  7
    CGG 12  4
    CGC 11  3
    CGT 5  3
    Asn N AAC 20 27
    AAT 17  34*
    Asp D GAC 26 20
    GAT 22 25
    Cys C TGC 12 13
    TGT 10 12
    Gln Q CAA 12 18
    CAG 35 20
    Glu E GAA 30 39
    GAG 40 28
    Gly G GGA 16 30
    GGG 16 19
    GGC 23  9
    GGT 11 13
    His H CAC 15 13
    CAT 11  7
    Ile I ATA 7  25*
    ATC 22 18
    ATT 16 23
    Leu L CTA 7  14*
    CTG 40 17
    CTC 20 14
    CTT 13 14
    TTA 7  8
    TTG 13 14
    Lys K AAA 24 35
    AAG 33 20
    Met M ATG 22 30
    Phe F TTC 21 17
    TTT 17 19
    Pro P CCA 17 12
    CCG 7  4
    CCC 20  8
    CCT 17 13
    Ser S AGC 19 14
    AGT 12 16
    TCA 12 23
    TCG 5  4
    TCC 18 12
    TCT 15 15
    Thr T ACA 15 24
    ACG 6  4
    ACC 19 13
    ACT 13 19
    Trp W TGG 13 18
    Tyr Y TAC 16 12
    TAT 12 19
    Val V GTA 7 13
    GTG 29 20
    GTC 15 12
    GTT 11 15
    Term TAA 1  2
    TAG 0.5   0.4
    TGA 1  1
  • In another form of minimal optimization, a Codon Usage Table (CUT) for the specific IV sequence in question is generated and compared to CUT for human genomic DNA (see Table 7, supra). Amino acids are identified for which there is a difference of at least 10 percentage points in codon usage between human and IV DNA (either more or less). Then the wild type IV codon is modified to conform to predominant human codon for each such amino acid. Furthermore, the remainder of codons for that amino acid are also modified such that they conform to the predominant human codon for each such amino acid.
  • A representative “minimally optimized” codon-optimized coding region encoding SEQ ID NO:2, minimally optimized according to codon usage in humans by this latter method, is presented herein as SEQ ID NO:25:
    1 ATGGCCTCAC AGGGCACCAA GCGGAGTTAT GAGCAGATGG AGACCGATGG CGAGAGACAG
    61 AACGCCACAG AGATCAGAGC CTCAGTTGGC AAGATGATCG GCGGCATCGG CCGGTTCTAT
    121 ATCCAGATGT GCACGGAGCT GAAGCTGAGC GACTACGAGG GCAGACTGAT TCAGAACTCT
    181 CTGACCATCG AGAGAATGGT CCTGAGTGCC TTCGATGAGA GACGAAACAA GTATCTGGAG
    241 GAGCATCCCT CCGCCGGCAA GGACCCCAAG AAGACGGGCG GCCCCATATA TAGAAGAGTT
    301 AACGGCAAGT GGATGAGAGA GCTGATCCTG TACGATAAGG AGGAGATCCG CAGAATATGG
    361 AGGCAGGCCA ACAACGGCGA CGATGCCACT GCCGGCCTGA CACATATGAT GATATGGCAC
    421 AGTAACCTGA ACGACGCCAC CTACCAGAGA ACAAGGGCCC TGGTTCGCAC GGGCATGGAT
    481 CCCAGAATGT GTTCACTGAT GCAGGGCTCT ACACTGCCCA GAAGGTCTGG CGCCGCCGGC
    541 GCCGCCGTCA AGGGCGTTGG CACAATGGTG ATGGAGCTGG TGCGGATGAT CAAGAGAGGC
    601 ATTAACGATC GGAACTTTTG GAGGGGCGAG AACGGCAGAA AGACCAGGAT AGCCTACGAG
    661 CGAATGTGCA ACATTCTGAA GGGCAAGTTC CAGACTGCCG CCCAGAAGGC CATGATGGAT
    721 CAGGTGCGGG AGAGCAGAAA CCCCGGCAAC GCCGAGTTCG AGGACCTGAC TTTCCTGGCC
    781 AGATCTGCCC TGATACTGAG GGGCTCTGTA GCCCACAAGT CCTGCCTGCC CGCCTGCGTG
    841 TACGGCCCCG CCGTGGCCTC CGGCTATGAC TTCGAGCGAG AGGGCTACTC CCTGGTAGGC
    901 ATCGATCCCT TTAGACTGCT GCAGAACTCT CAGGTCTACA GTCTGATTAG ACCCAACGAG
    961 AACCCCGCCC ATAAGAGCCA GCTGGTGTGG ATGGCCTGCC ACAGTGCCGC CTTCGAGGAC
    1021 CTGAGGGTGC TGTCTTTTAT AAAGGGCACA AAGGTGCTGC CCCGCGGCAA GCTGTCTACT
    1081 AGGGGCGTCC AGATAGCCTC CAACGAGAAC ATGGAGACAA TGGAGTCTAG TACTCTGGAG
    1141 CTGAGGTCTA GGTACTGGGC CATCAGGACT AGGAGCGGCG GCAACACCAA CCAGCAGAGG
    1201 GCCAGCGCCG GCCAGATCAG CATTCAGCCC ACCTTCAGTG TACAGAGAAA CCTGCCCTTT
    1261 GATAGAACTA CTGTTATGGC CGCCTTCTCT GGCAACACTG AGGGCAGAAC TAGTGACATG
    1321 CGAACAGAGA TCATAAGAAT GATGGAGTCG GCCCGTCCCG AGGATGTGTC CTTTCAGGGC
    1381 AGGGGCGTCT TCGAGCTGAG CGACGAGAAG GCCGCCAGCC CCATCGTACC CTCTTTCGAT
    1441 ATGAGTAACG AGGGCTCGTA CTTTTTTGGC GACAACGCCG AGGAGTATGA TAACTGA
  • In certain embodiments described herein, a codon-optimized coding region encoding SEQ ID NO:4 is optimized according to codon usage in humans (Homo sapiens). Alternatively, a codon-optimized coding region encoding SEQ ID NO:4 may be optimized according to codon usage in any plant, animal, or microbial species. Codon-optimized coding regions encoding SEQ ID NO:4, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:4 is shown in Table 8.
    TABLE 8
    Number in
    AMINO ACID SEQ ID NO: 4
    A Ala 25
    R Arg 17
    C Cys 3
    G Gly 16
    H His 5
    I Ile 11
    L Leu 26
    K Lys 13
    M Met 14
    F Phe 7
    P Pro 8
    S Ser 18
    T Thr 18
    W Trp 1
    Y Tyr 5
    V Val 16
    N Asn 11
    D Asp 6
    Q Gln 15
    E Glu 17
  • Using the amino acid composition shown in Table 8, a human codon-optimized coding region which encodes SEQ ID NO:4 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid. According to this method, codons are assigned to the coding region encoding SEQ ID NO:4 as follows: the 7 phenylalanine codons are TTC, the 26 leucine codons are CTG, the 11 isoleucine codons are ATC, the 14 methionine codons are ATG, the 16 valine codons are GTG, the 18 serine codons are AGC, the 8 proline codons are CCC, the 18 threonine codons are ACC, the 25 alanine codons are GCC, the 5 tyrosine codons are TAC, the 5 histidine codons are CAC, the 15 glutamine codons are CAG, the 11 asparagine codons are AAC, the 13 lysine codons are AAG, the 6 aspartic acid codons are GAC, the 17 glutamic acid codons are GAG, the 1 tryptophan codon is TGG, the 17 arginine codons are CGG, AGA, or AGG (the frequencies of usage of these three codons in the human genome are not significantly different), and the 16 glycine codons are GGC. The codon-optimized coding region designed by this method is presented herein as SEQ ID NO:27:
    ATGAGCCTGCTGACCGAGGTGGAGACCTACGTGCTGAGCATCATCCCCAG
    CGGCCCCCTGAAGGCCGAGATCGCCCAGAGGCTGGAGGACGTGTTCGCCG
    GCAAGAACACCGACCTGGAGGTGCTGATGGAGTGGCTGAAGACCAGGCCC
    ATCCTGAGCCCCCTGACCAAGGGCATCCTGGGCTTCGTGTTCACCCTGAC
    CGTGCCCAGCGAGAGGGGCCTGCAGAGGAGGAGGTTCGTGCAGAACGCCC
    TGAACGGCAACGGCGACCCCAACAACATGGACAAGGCCGTGAAGCTGTAC
    AGGAAGCTGAAGAGGGAGATCACCTTCCACGGCGCCAAGGAGATCAGCCT
    GAGCTACAGCGCCGGCGCCCTGGCCAGCTGCATGGGCCTGATCTACAACA
    GGATGGGCGCCGTGACCACCGAGGTGGCCTTCGGCCTGGTGTGCGCCACC
    TGCGAGCAGATCGCCGACAGCCAGCACAGGAGCCACAGGCAGATGGTGAC
    CACCACCAACCCCCTGATCAGGCACGAGAACAGGATGGTGCTGGCCAGCA
    CCACCGCCAAGGCCATGGAGCAGATGGCCGGCAGCAGCGAGCAGGCCGCC
    GAGGCCATGGAGGTGGCCAGCCAGGCCAGGCAGATGGTGCAGGCCATGAG
    GACCATCGGCACCCACCCCAGCAGCAGCGCCGGCCTGAAGAACGACCTGC
    TGGAGAACCTGCAGGCCTACCAGAAGAGGATGGGCGTGCAGATGCAGAGG
    TTCAAG
  • Alternatively, a human codon-optimized coding region which encodes SEQ ID NO:4 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 8 above. Using this latter method, codons are assigned to the coding region encoding SEQ ID NO:4 as follows: about 3 of the 7 phenylalanine codons are TTT, and about 4 of the phenylalanine codons are TTC; about 2 of the 26 leucine codons are TTA, about 3 of the leucine codons are TTG, about 3 of the leucine codons are CTT, about 5 of the leucine codons are CTC, about 2 of the leucine codons are CTA, and about 11 of the leucine codons are CTG; about 4 of the 11 isoleucine codons are ATT, about 5 of the isoleucine codons are ATC, and about 2 of the isoleucine codons are ATA; the 14 methionine codons are ATG; about 3 of the 16 valine codons are GTT, about 4 of the valine codons are GTG, about 2 of the valine codons are GTA, and about 8 of the valine codons are GTG; about 3 of the 18 serine codons are TCT, about 4 of the serine codons are TCC, about 3 of the serine codons are TCA, about 1 of the serine codons is TCG, about 3 of the serine codons are AGT, and about 4 of the serine codons are AGC; about 2 of the 8 proline codons are CCT, about 3 of the proline codons are CCC, about 2 of the proline codons are CCA, and about 1 of the proline codons is CCG; about 4 of the 18 threonine codons are ACT, about 7 of the threonine codons are ACC, about 5 of the threonine codons are ACA, and about 2 of the threonine codons are ACG; about 7 of the 25 alanine codons are GCT, about 10 of the alanine codons are GCC, about 6 of the alanine codons are GCA, and about 3 of the alanine codons are GCG; about 2 of the 5 tyrosine codons are TAT and about 3 of the tyrosine codons are TAC; about 2 of the 5 histidine codons are CAT and about 3 of the histidine codons are CAC; about 4 of the 15 glutamine codons are CAA and about 11 of the glutamine codons are CAG; about 5 of the 11 asparagine codons are AAT and about 6 of the asparagine codons are AAC; about 5 of the 13 lysine codons are AAA and about 8 of the lysine codons are AAG; about 3 of the 6 aspartic acid codons are GAT and about 3 of the aspartic acid codons are GAC; about 7 of the 17 glutamic acid codons are GAA and about 10 of the glutamic acid codons are GAG; about 1 of the 3 cysteine codons is TGT and about 2 of the cysteine codons are TGC; the 1 tryptophan codons is TGG; about 1 of the 17 arginine codons are CGT, about 3 of the arginine codons are CGC, about 2 of the arginine codons are CGA, about 4 of the arginine codons are CGG, about 3 of the arginine codons are AGA, and about 3 of the arginine codons are AGG; and about 3 of the 16 glycine codons are GGT, about 6 of the glycine codons are GGC, about 4 of the glycine codons are GGA, and about 4 of the glycine codons are GGG.
  • As described above, the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:4, optimized according to codon usage in humans is presented herein as SEQ ID NO:26:
    ATGAGCTTGCTAACAGAAGTGGAAACCTATGTCCTCAGTATCATTCCTAG
    CGGCCCCTTAAAAGCCGAAATCGCTCAGCGGCTCGAGGATGTTTTTGCCG
    GCAAGAACACCGACCTGGAGGTATTGATGGAGTGGCTGAAAACGCGACCT
    ATTCTGAGCCCCCTGACTAAGGGAATACTCGGCTTCGTTTTTACATTGAC
    CGTGCCCTCAGAGAGGGGTCTCCAAAGGAGGCGCTTCGTGCAGAACGCCT
    TAAACGGGAACGGGGACCCAAATAATATGGATAAGGCAGTGAAACTGTAT
    CGCAAATTAAAGCGGGAGATAACCTTCCATGGAGCCAAGGAGATCTCCCT
    GTCTTACTCTGCAGGTGCTCTCGCGTCGTGTATGGGACTTATCTACAACC
    GAATGGGCGCCGTCACAACAGAAGTGGCTTTCGGGCTGGTGTGCGCAACT
    TGCGAACAGATTGCTGACAGTCAGCACCGGTCCCACCGTCAAATGGTCAC
    CACCACCAATCCGCTGATTAGACATGAAAATCGCATGGTTCTAGCATCAA
    CTACAGCCAAAGCAATGGAACAAATGGCCGGAAGCTCCGAGCAGGCTGCC
    GAGGCGATGGAGGTGGCGTCCCAGGCCAGACAGATGGTACAGGCTATGAG
    AACTATCGGTACGCACCCAAGTTCTTCAGCTGGGCTGAAGAATGATCTTC
    TTGAGAACCTGCAGGCCTACCAAAAGCGGATGGGCGTCCAGATGCAGAGA
    TTTAAA
  • Additionally, a minimally codon-optimized nucleotide sequence encoding SEQ ID NO:4 can be designed by changing only certain codons found more frequently in IV genes than in human genes, as shown in Table 7. For example, if it is desired to substitute more frequently used codons in humans for those codons that occur at least 2 times more frequently in IV genes (designated with an asterisk in Table 7), Arg AGA, which occurs 2.3 times more frequently in IV genes than in human genes, is changed to, e.g., CGG; Asn AAT, which occurs 2.0 times more frequently in IV genes than in human genes, is changed to, e.g., AAC; Ile ATA, which occurs 3.6 times more frequently in IV genes than in human genes, is changed to, e.g., ATC; and Leu CTA, which occurs 2.0 times more frequently in IV genes than is human, is changed to, e.g., CTG.
  • In another form of minimal optimization, a Codon Usage Table (CUT) for the specific IV sequence in question is generated and compared to CUT for human genomic DNA (see Table 7, supra). Amino acids are identified for which there is a difference of at least 10 percentage points in codon usage between human and IV DNA (either more or less). Then the wild type IV codon is modified to conform to predominant human codon for each such amino acid. Furthermore, the remainder of codons for that amino acid are also modified such that they conform to the predominant human codon for each such amino acid.
  • A representative “minimally optimized” codon-optimized coding region encoding SEQ ID NO:4, minimally optimized according to codon usage in humans by this latter method, is presented herein as SEQ ID NO:28:
    ATGAGTCTGCTGACAGAGGTTGAGACGTACGTGCTGTCCATCATTCCCTC
    AGGCCCCCTGAAGGCCGAGATTGCCCAGAGACTGGAGGACGTCTTCGCCG
    GCAAGAACACCGATCTGGAGGTGCTGATGGAGTGGCTGAAGACTCGCCCC
    ATCCTGTCTCCCCTGACAAAGGGCATCCTGGGCTTCGTATTTACACTGAC
    CGTCCCCTCCGAGAGAGGCCTGCAGCGGAGGAGGTTCGTTCAGAACGCCC
    TGAACGGCAACGGCGATCCCAACAACATGGATAAGGCCGTGAAGCTGTAT
    AGAAAGCTGAAGCGAGAGATCACATTTCATGGCGCCAAGGAGATATCGCT
    GAGCTACAGTGCCGGCGCCCTGGCCTCTTGCATGGGCCTGATATACAACA
    GAATGGGCGCCGTTACTACAGAGGTAGCCTTTGGCCTGGTCTGCGCCACT
    TGCGAGCAGATCGCCGACTCTCAGCATAGATCTCACAGACAGATGGTGAC
    GACTACAAACCCCCTGATACGGCACGAGAACAGGATGGTGCTGGCCTCTA
    CTACCGCCAAGGCCATGGAGCAGATGGCCGGCAGCAGTGAGCAGGCCGCC
    GAGGCCATGGAGGTAGCCTCACAGGCCAGGCAGATGGTGCAGGCCATGCG
    AACCATCGGCACTCACCCCTCCAGCTCTGCCGGCCTGAAGAACGACCTGC
    TGGAGAACCTGCAGGCCTATCAGAAGAGAATGGGCGTACAGATGCAGAGG
    TTCAAG
  • In certain embodiments described herein, a codon-optimized coding region encoding SEQ ID NO:5 is optimized according to codon usage in humans (Homo sapiens). Alternatively, a codon-optimized coding region encoding SEQ ID NO:5 may be optimized according to codon usage in any plant, animal, or microbial species. Codon-optimized coding regions encoding SEQ ID NO:5, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:5 is shown in Table 9.
    TABLE 9
    Number in
    AMINO ACID SEQ ID NO: 5
    A Ala 5
    R Arg 7
    C Cys 3
    G Gly 8
    H His 2
    I Ile 8
    L Leu 10
    K Lys 5
    M Met 2
    F Phe 4
    P Pro 4
    S Ser 7
    T Thr 4
    W Trp 2
    Y Tyr 3
    V Val 4
    N Asn 3
    D Asp 5
    Q Gln 2
    E Glu 9
  • Using the amino acid composition shown in Table 9, a human codon-optimized coding region which encodes SEQ ID NO:5 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid. According to this method, codons are assigned to the coding region encoding SEQ ID NO:5 as follows: the 4 phenylalanine codons are TTC, the 10 leucine codons are CTG, the 8 isoleucine codons are ATC, the 2 methionine codons are ATG, the 4 valine codons are GTG, the 7 serine codons are AGC, the 4 proline codons are CCC, the 4 threonine codons are ACC, the 5 alanine codons are GCC, the 3 tyrosine codons are TAC, the 2 histidine codons are CAC, the 2 glutamine codons are CAG, the 3 asparagine codons are AAC, the 5 lysine codons are AAG, the 5 aspartic acid codons are GAC, the 9 glutamic acid codons are GAG, the 2 tryptophan codons are TGG, the 7 arginine codons are CGG, AGA, or AGG (the frequencies of usage of these three codons in the human genome are not significantly different), and the 8 glycine codons are GGC. The codon-optimized PA coding region designed by this method is presented herein as SEQ ID NO:30:
    1 ATGAGCCTGC TGACCGAGGT GGAGACCCCC ATCCGGAACG AGTGGGGCTG CCGGTGCAAC
    61 GGCAGCAGCG ACCCCCTGGC CATCGCCGCC AACATCATCG GCATCCTGCA CCTGACCCTG
    121 TGGATCCTGG ACCGGCTGTT CTTCAAGTGC ATCTACCGGC GGTTCAAGTA CGGCCTGAAG
    181 GGCGGCCCCA GCACCGAGGG CGTGCCCAAG AGCATGCGGG AGGAGTACCG GAAGGAGCAG
    241 CAGAGCGCCG TGGACGCCGA CGACGGCCAC TTCGTGAGCA TCGAGCTGGA GTGA
  • Alternatively, a human codon-optimized coding region which encodes SEQ ID NO:5 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 9 above. Using this latter method, codons are assigned to the coding region encoding SEQ ID NO:5 as follows: about 2 of the 4 phenylalanine codons are TTT, and about 2 of the phenylalanine codons are TTC; about 1 of the 10 leucine codons are TTA, about 1 of the leucine codons are TTG, about 1 of the leucine codons are CTT, about 2 of the leucine codons are CTC, about 1 of the leucine codons are CTA, and about 4 of the leucine codons are CTG; about 3 of the 8 isoleucine codons are ATT, about 4 of the isoleucine codons are ATC, and about 1 of the isoleucine codons are ATA; the 2 methionine codons are ATG; about 1 of the 4 valine codons are GTT, about 1 of the valine codons are GTG, about 0 of the valine codons are GTA, and about 2 of the valine codons are GTG; about 1 of the 7 serine codons are TCT, about 2 of the serine codons are TCC, about 1 of the serine codons are TCA, about 0 of the serine codons are TCG, about 1 of the serine codons are AGT, and about 2 of the serine codons are AGC; about 1 of the 4 proline codons are CCT, about 1 of the proline codons are CCC, about 2 of the proline codons are CCA, and about 0 of the proline codons are CCG; about 1 of the 4 threonine codons are ACT, about 1 of the threonine codons are ACC, about 1 of the threonine codons are ACA, and about 0 of the threonine codons are ACG; about 1 of the 5 alanine codons are GGT, about 2 of the alanine codons are GCC, about 1 of the alanine codons are GCA, and about 1 of the alanine codons are GCG; about 1 of the 3 tyrosine codons are TAT and about 2 of the tyrosine codons are TAC; about 1 of the 2 histidine codons are CAT and about 1 of the histidine codons are CAC; about 1 of the 2 glutamine codons are CAA and about 1 of the glutamine codons are CAG; about 1 of the 3 asparagine codons are AAT and about 2 of the asparagine codons are AAC; about 2 of the 5 lysine codons are AAA and about 3 of the lysine codons are AAG; about 2 of the 5 aspartic acid codons are GAT and about 3 of the aspartic acid codons are GAC; about 4 of the 9 glutamic acid codons are GAA and about 5 of the glutamic acid codons are GAG; about 1 of the 3 cysteine codons are TGT and about 2 of the cysteine codons are TGC; the 2 tryptophan codons are TGG; about 1 of the 7 arginine codons are CGT, about 1 of the arginine codons are CGC, about 1 of the arginine codons are CGA, about 1 of the arginine codons are CGG, about 1 of the arginine codons are AGA, and about 1 of the arginine codons are AGG; and about 1 of the 8 glycine codons are GGT, about 3 of the glycine codons are GGC, about 2 of the glycine codons are GGA, and about 2 of the glycine codons are GGG.
  • As described above, the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:5, optimized according to codon usage in humans is presented herein as SEQ ID NO:29:
    1 ATGAGTCTTC TAACCGAGGT CGAAACGCCT ATCAGAAACG AATGGGGGTG CAGATGCAAC
    61 GGTTCAAGTG ATCCTCTCGC TATTGCCGCA AATATCATTG GGATCTTGCA CTTGACATTG
    121 TGGATTCTTG ATCGTCTTTT TTTCAAATGC ATTTACCGTC GCTTTAAATA CGGACTGAAA
    181 GGAGGGCCTT CTACGGAAGG AGTGCCAAAG TCTATGAGGG AAGAATATCG AAAGGAACAG
    241 CAGAGTGCTG TGGATGCTGA CGATGGTCAT TTTGTCAGCA TAGAGCTGGA GTAA
  • Additionally, a minimally codon-optimized nucleotide sequence encoding SEQ ID NO:5 can be designed by changing only certain codons found more frequently in IV genes than in human genes, as shown in Table 7. For example, if it is desired to substitute more frequently used codons in humans for those codons that occur at least 2 times more frequently in IV genes (designated with an asterisk in Table 7), Arg AGA, which occurs 2.3 times more frequently in IV genes than in human genes, is changed to, e.g., CGG; Asn AAT, which occurs 2.0 times more frequently in IV genes than in human genes, is changed to, e.g., AAC; Ile ATA, which occurs 3.6 times more frequently in IV genes than in human genes, is changed to, e.g., ATC; and Leu CTA, which occurs 2.0 times more frequently in IV genes than is human, is changed to, e.g., CTG.
  • In another form of minimal optimization, a Codon Usage Table (CUT) for the specific IV sequence in question is generated and compared to CUT for human genomic DNA (see Table 7, supra). Amino acids are identified for which there is a difference of at least 10 percentage points in codon usage between human and IV DNA (either more or less). Then the wild type IV codon is modified to conform to predominant human codon for each such amino acid. Furthermore, the remainder of codons for that amino acid are also modified such that they conform to the predominant human codon for each such amino acid.
  • A representative “minimally optimized” codon-optimized coding region encoding SEQ ID NO:5, minimally optimized according to codon usage in humans by this latter method, is presented herein as SEQ ID NO:31:
    1 ATGTCTCTGC TGACAGAGGT GGAGACACCC ATAAGGAACG AGTGGGGCTG CAGGTGCAAC
    61 GGCTCTAGTG ATCCCCTGGC CATCGCCGCC AACATCATTG GCATACTGCA TCTGACCCTG
    121 TGGATCCTGG ATAGACTGTT CTTTAAGTGC ATTTACAGAC GATTTAAGTA TGGCCTGAAG
    181 GGCGGCCCCT CAACTGAGGG CGTGCCCAAG AGTATGAGAG AGGAGTACCG GAAGGAGCAG
    241 CAGAGCGCCG TTGACGCCGA TGACGGCCAC TTCGTCTCCA TCGAGCTGGA GTGA
  • In certain embodiments described herein, a codon-optimized coding region encoding SEQ ID NO:7 is optimized according to codon usage in humans (Homo sapiens). Alternatively, a codon-optimized coding region encoding SEQ ID NO:7 may be optimized according to codon usage in any plant, animal, or microbial species. Codon-optimized coding regions encoding SEQ ID NO:7, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:7 is shown in Table 10.
    TABLE 10
    Number in
    AMINO ACID SEQ ID NO: 7
    A Ala 39
    R Arg 51
    C Cys 8
    G Gly 43
    H His 6
    I Ile 27
    L Leu 35
    K Lys 21
    M Met 26
    F Phe 18
    P Pro 18
    S Ser 43
    T Thr 30
    W Trp 7
    Y Tyr 15
    V Val 24
    N Asn 28
    D Asp 23
    Q Gln 21
    E Glu 39
  • Using the amino acid composition shown in Table 10, a human codon-optimized coding region which encodes SEQ ID NO:7 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid. According to this method, codons are assigned to the coding region encoding SEQ ID NO:7 as follows: the 18 phenylalanine codons are TTC, the 35 leucine codons are CTG, the 27 isoleucine codons are ATC, the 26 methionine codons are ATG, the 24 valine codons are GTG, the 43 serine codons are AGC, the 18 proline codons are CCC, the 30 threonine codons are ACC, the 39 alanine codons are GCC, the 15 tyrosine codons are TAC, the 6 histidine codons are CAC, the 21 glutamine codons are CAG, the 28 asparagine codons are AAC, the 21 lysine codons are AAG, the 23 aspartic acid codons are GAC, the 39 glutamic acid codons are GAG, the 7 tryptophan codons are TGG, the 51 arginine codons are CGG, AGA, or AGG (the frequencies of usage of these three codons in the human genome are not significantly different), and the 43 glycine codons are GGC. The codon-optimized PA coding region designed by this method is presented herein as SEQ ID NO:33:
    ATGAGCCTGCTGACCGAGGTGGAGACCCCCATCAGGAACGAGTGGGGCT
    GCAGGTGCAACGGCAGCAGCGACATGGCCAGCCAGGGCACCAAGAGGAGC
    TACGAGCAGATGGAGACCGACGGCGAGAGGCAGAACGCCACCGAGATCAG
    GGCCAGCGTGGGCAAGATGATCGGCGGCATCGGCAGGTTCTACATCCAGA
    TGTGCACCGAGCTGAAGCTGAGCGACTACGAGGGCAGGCTGATCCAGAAC
    AGCCTGACCATCGAGAGGATGGTGCTGAGCGCCTTCGACGAGAGGAGGAA
    CAAGTACCTGGAGGAGCACCCCAGCGCCGGCAAGGACCCCAAGAAGACCG
    GCGGCCCCATCTACAGGAGGGTGAACGGCAAGTGGATGAGGGAGCTGATC
    CTGTACGACAAGGAGGAGATCAGGAGGATCTGGAGGCAGGCCAACAACGG
    CGACGACGCCACCGCCGGCCTGACCCACATGATGATCTGGCACAGCAACC
    TGAACGACGCCACCTACCAGAGGACCAGGGCCCTGGTGAGGACCGGCATG
    GACCCCAGGATGTGCAGCCTGATGCAGGGCAGCACCCTGCCCAGGAGGAG
    CGGCGCCGCCGGCGCCGCCGTGAAGGGCGTGGGCACCATGGTGATGGAGC
    TGGTGAGGATGATCAAGAGGGGCATCAACGACAGGAACTTCTGGAGGGGC
    GAGAACGGCAGGAAGACCAGGATCGCCTACGAGAGGATGTGCAACATCCT
    GAAGGGCAAGTTCCAGACCGCCGCCCAGAAGGCCATGATGGACCAGGTGA
    GGGAGAGCAGGAACCCCGGCAACGCCGAGTTCGAGGACCTGACCTTCCTG
    GCCAGGAGCGCCCTGATCCTGAGGGGCAGCGTGGCCCACAAGAGCTGCCT
    GCCCGCCTGCGTGTACGGCCCCGCCGTGGCCAGCGGCTACGACTTCGAGA
    GGGAGGGCTACAGCCTGGTGGGCATCGACCCCTTCAGGCTGCTGCAGAAC
    AGCCAGGTGTACAGCCTGATCAGGCCCAACGAGAACCCCGCCCACAAGAG
    CCAGCTGGTGTGGATGGCCTGCCACAGCGCCGCCTTCGAGGACCTGAGGG
    TGCTGAGCTTCATCAAGGGCACCAAGGTGCTGCCCAGGGGCAAGCTGAGC
    ACCAGGGGCGTGCAGATCGCCAGCAACGAGAACATGGAGACCATGGAGAG
    CAGCACCCTGGAGCTGAGGAGCAGGTACTGGGCCATCAGGACCAGGAGCG
    GCGGCAACACCAACCAGCAGAGGGCCAGCGCCGGCCAGATCAGCATCCAG
    CCCACCTTCAGCGTGCAGAGGAACCTGCCCTTCGACAGGACCACCGTGAT
    GGCCGCCTTCAGCGGCAACACCGAGGGCAGGACCAGCGACATGAGGACCG
    AGATCATCAGGATGATGGAGAGCGCCAGGCCCGAGGACGTGAGCTTCCAG
    GGCAGGGGCGTGTTCGAGCTGAGCGACGAGAAGGCCGCCAGCCCCATCGT
    GCCCAGCTTCGACATGAGCAACGAGGGCAGCTACTTCTTCGGCGACAACG
    CCGAGGAGTACGACAAC
  • Alternatively, a human codon-optimized coding region which encodes SEQ ID NO:7 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 10 above. Using this latter method, codons are assigned to the coding region encoding SEQ ID NO:7 as follows: about 8 of the 18 phenylalanine codons are TTT, and about 10 of the phenylalanine codons are TTC; about 3 of the 35 leucine codons are TTA, about 4 of the leucine codons are TTG, about 5 of the leucine codons are CTT, about 7 of the leucine codons are CTC, about 2 of the leucine codons are CTA, and about 14 of the leucine codons are CTG; about 10 of the 27 isoleucine codons are ATT, about 13 of the isoleucine codons are ATC, and about 4 of the isoleucine codons are ATA; the 26 methionine codons are ATG; about 4 of the 24 valine codons are GTT, about 6 of the valine codons are GTG, about 3 of the valine codons are GTA, and about 11 of the valine codons are GTG; about 8 of the 43 serine codons are TCT, about 9 of the serine codons are TCC, about 6 of the serine codons are TCA, about 2 of the serine codons are TCG, about 6 of the serine codons are AGT, and about 10 of the serine codons are AGC; about 5 of the 18 proline codons are CCT, about 6 of the proline codons are CCC, about 5 of the proline codons are CCA, and about 2 of the proline codons are CCG; about 7 of the 30 threonine codons are ACT, about 11 of the threonine codons are ACC, about 8 of the threonine codons are ACA, and about 4 of the threonine codons are ACG; about 10 of the 39 alanine codons are GGT, about 16 of the alanine codons are GCC, about 9 of the alanine codons are GCA, and about 4 of the alanine codons are GCG; about 7 of the 15 tyrosine codons are TAT and about 8 of the tyrosine codons are TAC; about 2 of the 6 histidine codons are CAT and about 4 of the histidine codons are CAC; about 5 of the 21 glutamine codons are CAA and about 16 of the glutamine codons are CAG; about 13 of the 28 asparagine codons are AAT and about 15 of the asparagine codons are AAC; about 9 of the 21 lysine codons are AAA and about 12 of the lysine codons are AAG; about 11 of the 23 aspartic acid codons are GAT and about 12 of the aspartic acid codons are GAC; about 16 of the 39 glutamic acid codons are GAA and about 23 of the glutamic acid codons are GAG; about 4 of the 8 cysteine codons are TGT and about 4 of the cysteine codons are TGC; the 7 tryptophan codons are TGG; about 4 of the 51 arginine codons are CGT, about 10 of the arginine codons are CGC, about 6 of the arginine codons are CGA, about 11 of the arginine codons are CGG, about 10 of the arginine codons are AGA, and about 10 of the arginine codons are AGG; and about 7 of the 43 glycine codons are GGT, about 15 of the glycine codons are GGC, about 11 of the glycine codons are GGA, and about 11 of the glycine codons are GGG.
  • As described above, the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:7, optimized according to codon usage in humans is presented herein as SEQ ID NO:32:
    ATGAGCCTTCTCACAGAAGTGGAAACACCTATCAGAAATGAATGGGGATG
    CAGATGCAATGGGTCGAGTGATATGGCCTCTCAAGGTACGAAAAGAAGCT
    ACGAGCAAATGGAAACGGATGGAGAAAGACAAAACGCGACCGAAATCAGA
    GCATCCGTCGGGAAGATGATTGGAGGAATCGGACGATTCTACATCCAGAT
    GTGCACAGAGCTAAAGCTATCGGATTATGAAGGGAGACTAATACAAAATA
    GCCTAACTATCGAGAGAATGGTGCTGTCTGCATTTGACGAAAGGAGAAAC
    AAATACCTGGAAGAACACCCCTCTGCAGGGAAAGACCCAAAAAAAACTGG
    AGGTCCGATATACCGGAGAGTCAACGGTAAATGGATGAGAGAGCTGATCT
    TGTATGATAAGGAAGAAATAAGACGCATCTGGCGGCAAGCTAATAATGGA
    GACGACGCTACTGCAGGGCTCACGCATATGATGATCTGGCACTCTAATTT
    GAATGATGCAACGTACCAAAGAACCCGCGCACTTGTGCGGACCGGAATGG
    ACCCTCGTATGTGCAGCCTTATGCAGGGGTCCACACTGCCCAGAAGGTCC
    GGAGCAGCTGGAGCAGCAGTAAAGGGGGTTGGAACCATGGTGATGGAGCT
    GGTGAGAATGATTAAGAGGGGGATCAATGACAGGAACTTCTGGCGAGGAG
    AAAACGGGAGAAAAACTAGGATAGCATATGAGAGGATGTGTAACATCCTC
    AAAGGAAAATTCCAAACCGCTGCTCAGAAAGCAATGATGGATCAAGTACG
    CGAAAGTAGAAATCCTGGAAATGCAGAGTTTGAAGATCTCACTTTCCTCG
    CGCGAAGCGCTCTCATCCTCAGAGGGAGTGTCGCTCATAAAAGTTGCCTG
    CCTGCCTGCGTATATGGTCCTGCCGTGGCAAGTGGATACGACTTTGAGAG
    AGAGGGGTACTCTCTTGTTGGAATAGATCCATTCAGATTACTTCAGAATT
    CCCAGGTGTACAGTTTAATAAGGCCAAACGAAAATCCTGCACACAAATCA
    CAACTTGTTTGGATGGCATGCCATAGTGCCGCATTCGAAGATCTAAGAGT
    TCTCTCTTTCATCAAAGGTACAAAGGTCCTTCCAAGGGGAAAACTCTCTA
    CCAGAGGGGTACAAATAGCTTCAAATGAGAACATGGAGACAATGGAATCT
    AGCACATTGGAATTGAGAAGTAGGTATTGGGCCATTAGAACCAGGAGTGG
    AGGCAATACTAATCAACAGCGGGCTTCTGCCGGTCAAATTAGCATACAAC
    CTACTTTTTCAGTGCAACGGAATCTCCCTTTTGATAGGACAACTGTCATG
    GCGGCATTCTCTGGAAATACCGAAGGAAGGACTTCCGATATGAGGACTGA
    GATCATTAGGATGATGGAAAGTGCCCGACCTGAAGACGTCAGTTTTCAAG
    GAAGAGGTGTGTTCGAACTCTCTGACGAAAAGGCAGCTAGCCCAATCGTT
    CCTTCTTTTGATATGTCAAATGAAGGATCCTACTTCTTCGGCGATAATGC
    GGAGGAATATGACAAC
  • In certain embodiments described herein, a codon-optimized coding region encoding SEQ ID NO:9 is optimized according to codon usage in humans (Homo sapiens). Alternatively, a codon-optimized coding region encoding SEQ ID NO:9 may be optimized according to codon usage in any plant, animal, or microbial species. Codon-optimized coding regions encoding SEQ ID NO:9, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:9 is shown in Table 11.
    TABLE 11
    Number in
    AMINO ACID SEQ ID NO: 9
    A Ala 39
    R Arg 51
    C Cys 8
    G Gly 43
    H His 6
    I Ile 27
    L Leu 35
    K Lys 21
    M Met 26
    F Phe 18
    P Pro 18
    S Ser 43
    T Thr 30
    W Trp 7
    Y Tyr 15
    V Val 24
    N Asn 28
    D Asp 23
    Q Gln 21
    E Glu 39
  • Using the amino acid composition shown in Table 11, a human codon-optimized coding region which encodes SEQ ID NO:9 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid. According to this method, codons are assigned to the coding region encoding SEQ ID NO:9 as follows: the 18 phenylalanine codons are TTC, the 35 leucine codons are CTG, the 27 isoleucine codons are ATC, the 26 methionine codons are ATG, the 24 valine codons are GTG, the 43 serine codons are AGC, the 18 proline codons are CCC, the 30 threonine codons are ACC, the 39 alanine codons are GCC, the 15 tyrosine codons are TAC, the 6 histidine codons are CAC, the 21 glutamine codons are CAG, the 28 asparagine codons are AAC, the 21 lysine codons are AAG, the 23 aspartic acid codons are GAC, the 39 glutamic acid codons are GAG, the 7 tryptophan codons are TGG, the 51 arginine codons are CGG, AGA, or AGG (the frequencies of usage of these three codons in the human genome are not significantly different), and the 43 glycine codons are GGC. The codon-optimized PA coding region designed by this method is presented herein as SEQ ID NO:35:
    ATGGCCAGCCAGGGCACCAAGAGGAGCTACGAGCAGATGGAGACCGACGG
    CGAGAGGCAGAACGCCACCGAGATCAGGGGCAGCGTGGGCAAGATGATCG
    GCGGCATCGGCAGGTTCTACATCCAGATGTGCACCGAGCTGAAGCTGAGC
    GACTACGAGGGCAGGCTGATCCAGAACAGCCTGACCATCGAGAGGATGGT
    GCTGAGCGCCTTCGACGAGAGGAGGAACAAGTACCTGGAGGAGCACCCCA
    GCGCCGGCAAGGACCCCAAGAAGACCGGCGGCCCCATCTACAGGAGGGT
    GAACGGCAAGTGGATGAGGGAGCTGATCCTGTACGACAAGGAGGAGATCA
    GGAGGATCTGGAGGCAGGCCAACAACGGCGACGACGCCACCGCCGGCCTG
    ACCCACATGATGATCTGGCACAGCAACCTGAACGACGCCACCTACCAGAG
    GACCAGGGCCCTGGTGAGGACCGGCATGGACCCCAGGATGTGCAGCCTGA
    TGCAGGGCAGCACCCTGCCCAGGAGGAGCGGCGCCGCCGGCGCCGCCGTG
    AAGGGCGTGGGCACCATGGTGATGGAGCTGGTGAGGATGATCAAGAGGGG
    CATCAACGACAGGAACTTCTGGAGGGGCGAGAACGGCAGGAAGACCAGGA
    TCGCCTACGAGAGGATGTGCAACATCCTGAAGGGCAAGTTCCAGACCGCC
    GCCCAGAAGGCCATGATGGACCAGGTGAGGGAGAGCAGGAACCCCGGCAA
    CGCCGAGTTCGAGGACCTGACCTTCCTGGCCAGGAGCGCCCTGATCCTGA
    GGGGCAGCGTGGCCCACAAGAGCTGCCTGCCCGCCTGCGTGTACGGCCCC
    GCCGTGGCCAGCGGCTACGACTTCGAGAGGGAGGGCTACAGCCTGGTGGG
    CATCGACCCCTTCAGGCTGCTGCAGAACAGCCAGGTGTACAGCCTGATCA
    GGCCCAACGAGAACCCCGCCCACAAGAGCCAGCTGGTGTGGATGGCCTGC
    CACAGCGCCGCCTTCGAGGACCTGAGGGTGCTGAGCTTCATCAAGGGCAC
    CAAGGTGCTGCCCAGGGGCAAGCTGAGCACCAGGGGCGTGCAGATGGGCA
    GCAAGGAGAACATGGAGACCATGGAGAGCAGCACCCTGGAGCTGAGGAGC
    AGGTACTGGGCCATCAGGACCAGGAGCGGCGGCAACACCAACCAGCAGAG
    GGCCAGCGCCGGCCAGATCAGCATCCAGCCCACCTTCAGCGTGCAGAGGA
    ACCTGCCCTTCGACAGGACCACCGTGATGGCCGCCTTCAGCGGCAACACC
    GAGGGCAGGACCAGCGACATGAGGACCGAGATCATCAGGATGATGGAGAG
    CGCCAGGCCCGAGGACGTGAGCTTCCAGGGCAGGGGCGTGTTCGAGCTGA
    GCGACGAGAAGGCCGCCAGCCCCATCGTGCCCAGCTTCGACATGAGCAAC
    GAGGGCAGCTACTTCTTCGGCGACAACGCCGAGGAGTACGACAACATGAG
    CCTGCTGACCGAGGTGGAGACCCCCATCAGGAACGAGTGGGGCTGCAGGT
    GCAACGGCAGCAGCGAC
  • Alternatively, a human codon-optimized coding region which encodes SEQ ID NO:9 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 11 above. Using this latter method, codons are assigned to the coding region encoding SEQ ID NO:9 as follows: about 8 of the 18 phenylalanine codons are TTT, and about 10 of the phenylalanine codons are TTC; about 3 of the 35 leucine codons are TTA, about 4 of the leucine codons are TTG, about 5 of the leucine codons are CTT, about 7 of the leucine codons are CTC, about 2 of the leucine codons are CTA, and about 14 of the leucine codons are CTG; about 10 of the 27 isoleucine codons are ATT, about 13 of the isoleucine codons are ATC, and about 4 of the isoleucine codons are ATA; the 26 methionine codons are ATG; about 4 of the 24 valine codons are GTT, about 6 of the valine codons are GTG, about 3 of the valine codons are GTA, and about 11 of the valine codons are GTG; about 8 of the 43 serine codons are TCT, about 9 of the serine codons are TCC, about 6 of the serine codons are TCA, about 2 of the serine codons are TCG, about 6 of the serine codons are AGT, and about 10 of the serine codons are AGC; about 5 of the 18 proline codons are CCT, about 6 of the proline codons are CCC, about 5 of the proline codons are CCA, and about 2 of the proline codons are CCG; about 7 of the 30 threonine codons are ACT, about 11 of the threonine codons are ACC, about 8 of the threonine codons are ACA, and about 4 of the threonine codons are ACG; about 10 of the 39 alanine codons are GGT, about 16 of the alanine codons are GCC, about 9 of the alanine codons are GCA, and about 4 of the alanine codons are GCG; about 7 of the 15 tyrosine codons are TAT and about 8 of the tyrosine codons are TAC; about 2 of the 6 histidine codons are CAT and about 4 of the histidine codons are CAC; about 5 of the 21 glutamine codons are CAA and about 16 of the glutamine codons are CAG; about 13 of the 28 asparagine codons are AAT and about 15 of the asparagine codons are AAC; about 9 of the 21 lysine codons are AAA and about 12 of the lysine codons are AAG; about 11 of the 23 aspartic acid codons are GAT and about 12 of the aspartic acid codons are GAC; about 16 of the 39 glutamic acid codons are GAA and about 23 of the glutamic acid codons are GAG; about 4 of the 8 cysteine codons are TGT and about 4 of the cysteine codons are TGC; the 7 tryptophan codons are TGG; about 4 of the 51 arginine codons are CGT, about 10 of the arginine codons are CGC, about 6 of the arginine codons are CGA, about 11 of the arginine codons are CGG, about 10 of the arginine codons are AGA, and about 10 of the arginine codons are AGG; and about 7 of the 43 glycine codons are GGT, about 15 of the glycine codons are GGC, about 11 of the glycine codons are GGA, and about 11 of the glycine codons are GGG.
  • As described above, the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:9, optimized according to codon usage in humans is presented herein as SEQ ID NO:34:
    ATGGCAAGCCAGGGCACAAAACGCAGTTACGAGCAGATGGAGACTGATGG
    TGAGAGGCAGAACGCCACCGAAATCCGGGCCTCCGTCGGCAAGATGATTG
    GTGGCATCGGAAGATTCTATATCCAGATGTGCACGGAGCTTAAGCTGTCC
    GATTACGAGGGGCGCTTAATACAGAACTCTCTGACTATCGAGCGAATGGT
    CTTGAGCGCCTTTGATGAGCGGCGTAATAAGTATCTCGAAGAGCACCCTT
    CTGCTGGAAAAGACCCCAAAAAGACCGGGGGACCTATCTACCGACGTGTG
    AACGGAAAATGGATGCGCGAACTGATACTGTACGACAAGGAGGAGATCCG
    TAGGATCTGGAGACAGGCTAATAACGGAGATGATGCCACAGCTGGGCTGA
    CCCATATGATGATATGGCATAGCAACCTGAACGACGCAACCTATCAACGC
    ACTAGAGCACTCGTGAGGACCGGTATGGACCCACGCATGTGCTCATTGAT
    GCAAGGTAGCACATTGCCTCGGAGGTCAGGCGCCGCCGGTGCCGCCGTAA
    AGGGGGTGGGCACAATGGTGATGGAACTGGTCCGAATGATCAAAAGAGGC
    ATCAATGACAGGAACTTTTGGCGCGGAGAAAACGGGCGCAAGACCCGCAT
    TGCCTACGAGCGCATGTGTAACATTTTAAAAGGCAAATTCCAGACTGCAG
    CCCAGAAAGCAATGATGGACCAAGTTAGAGAAAGTAGAAATCCCGGGAAT
    GCCGAGTTTGAAGACCTGACTTTCCTGGCTAGAAGCGCCTTGATCCTGCG
    GGGCTCTGTCGCCCACAAGAGCTGCCTCCCCGCTTGCGTTTACGGCCCCG
    CGGTCGCAAGTGGCTACGATTTCGAGAGGGAGGGGTATTCCCTAGTTGGG
    ATCGATCCCTTCCGGCTCCTACAGAATTCTCAGGTGTATAGTCTGATTAG
    ACCCAACGAAAACCCGGCTCACAAGAGTCAGCTTGTTTGGATGGCATGTC
    ACTCAGCAGCTTTCGAAGACCTGCGGGTACTCAGCTTTATTAAAGGCACC
    AAGGTCCTGCCAAGAGGAAAGCTCTCCACGAGGGGAGTACAGATCGCCTC
    AAACGAGAACATGGAGACAATGGAAAGCTCCACCCTTGAGCTTAGGTCGC
    GGTATTGGGCTATTAGAACACGATCTGGGGGGAATACCAATCAGCAACGA
    GCGAGTGCTGGTCAGATTTCCATTCAGCCTACTTTCTCTGTGCAACGGAA
    TCTACCATTTGACAGGACAACTGTGATGGCAGCGTTCTCCGGCAATACAG
    AAGGACGAACATCAGACATGAGGACCGAAATTATCCGGATGATGGAGAGC
    GCTCGGCCAGAAGATGTGTCGTTCCAGGGCCGGGGCGTGTTTGAGCTCAG
    CGACGAGAAGGCCGCGTCTCCAATTGTGCCTTCCTTTGATATGAGCAATG
    AGGGGTCATACTTTTTCGGAGACAATGCCGAAGAGTATGATAATATGTCT
    CTGCTTACCGAGGTGGAAACGCCGATACGCAACGAATGGGGTTGTCGTTG
    TAACGGCTCCAGTGAT
  • In certain embodiments described herein, a codon-optimized coding region encoding SEQ ID NO:16 is optimized according to codon usage in humans (Homo sapiens). Alternatively, a codon-optimized coding region encoding SEQ ID NO:16 may be optimized according to codon usage in any plant, animal, or microbial species. Codon-optimized coding regions encoding SEQ ID NO:16, optimized according to codon usage in humans are designed as follows. The amino acid composition of SEQ ID NO:16 is shown in Table 12.
    TABLE 12
    AMINO Number in
    ACID SEQ ID NO: 16
    A Ala 41
    R Arg 30
    C Cys 5
    G Gly 44
    H His 4
    I Ile 38
    L Leu 39
    K Lys 52
    M Met 27
    F Phe 21
    P Pro 26
    S Ser 40
    T Thr 38
    W Trp 1
    Y Tyr 14
    V Val 32
    N Asn 25
    D Asp 34
    Q Gln 19
    E Glu 30
  • Using the amino acid composition shown in Table 12, a human codon-optimized coding region which encodes SEQ ID NO: 16 can be designed by any of the methods discussed herein. For “uniform” optimization, each amino acid is assigned the most frequent codon used in the human genome for that amino acid. According to this method, codons are assigned to the coding region encoding SEQ ID NO:16 as follows: the 21 phenylalanine codons are TTC, the 39 leucine codons are CTG, the 38 isoleucine codons are ATC, the 27 methionine codons are ATG, the 32 valine codons are GTG, the 40 serine codons are AGC, the 26 proline codons are CCC, the 38 threonine codons are ACC, the 41 alanine codons are GCC, the 14 tyrosine codons are TAC, the 4 histidine codons are CAC, the 19 glutamine codons are CAG, the 25 asparagine codons are AAC, the 52 lysine codons are AAG, the 34 aspartic acid codons are GAC, the 30 glutamic acid codons are GAG, the 1 tryptophan codon is TGG, the 30 arginine codons are CGG, AGA, or AGG (the frequencies of usage of these three codons in the human genome are not significantly different), and the 44 glycine codons are GGC. The codon-optimized PA coding region designed by this method is presented herein as SEQ ID NO:37:
    ATGAGCAACATGGACATCGACAGGATCAACACCGGCACCATCGACAAGAC
    CGGCGAGGAGCTGAGGGCCGGCACCAGCGGGGCCAGCCGGCCGATCATCA
    AGCCGGGCAGCGTGGCCCCCCCGAGCAACAAGCGGACCCGGAACCCCAGC
    CCCGAGCGGACCAGGAGCAGGAGCGAGACCGAGATCGGCCGGAAGATCCA
    GAAGAAGGAGAGGCCCACCGAGATGAAGAAGAGCGTGTACAAGATGGTGG
    TGAAGCTGGGCGAGTTGTACAACCAGATGATGGTGAAGGCCGGGCTGAAC
    GACGACATGGAGCGGAAGCTGATGGAGAACGCCGAGGCGGTGGAGGGGAT
    GCTGCTGGCCGGCAGCGACGACAAGAAGAGCGAGTACCAGAAGAAGGGGA
    ACGCCGGGGACGTGAAGGAGGGCAAGGAGGAGATCGACGACAACAAGAGC
    GGCGGCACCTTCTACAAGATGGTGCGGGACGACAAGACCATGTAGTTGAG
    CCCCATGAAGATCACCTTCGTGAAGGAGGAGGTGAAGACCATGTACAAGA
    CCACGATGGGCAGCGACGGCTTCAGCGGCCTGAACCACATCATGATCGGC
    CACAGCCAGATGAACGAGGTGTGCTTCGAGCGGAGCAAGGGGGTGAAGCG
    GGTGGGCCTGGACCCCAGCCTGATCAGCAGCTTCGCCGGCAGCACCGTGC
    CCCGGCGGAGCGGCACCACCGGCGTGGCCATCAAGGGCGGCGGCACGCTG
    GTGGACGAGGGCATCCGGTTCATCGGCCGGGCCATGGCCGACGGGGGCCT
    GCTGGGGGACATCAAGGCCAAGACCGCCTACGAGAAGATCCTGCTGAACC
    TGAAGAACAAGTGCAGCGCCCCCCAGCAGAAGGCCCTGGTGGACCAGGTG
    ATCGGCAGCCGGAAGCCCGGCATCGCCGACATCGAGGACCTGACCCTGCT
    GGCCCGGAGCATGGTGGTGGTGCGGCCCAGCGTGGCCAGCAAGGTGGTGC
    TGCCCATCAGCATCTACGCCAAGATGCCCCAGCTGGGCTTCAACACCGAG
    GAGTACAGCATGGTGGGCTACGAGGCCATGGCCCTGTACAACATGGCCAC
    CCCCGTGAGCATCCTGCGGATGGGCGACGACGCCAAGGACAAGAGCCAGC
    TGTTCTTCATGAGCTGCTTCGGCGCCGCCTACGAGGACCTGCGGGTGCTG
    AGCGCCCTGACCGGCACCGAGTTCAAGCCCCGGAGCGCCCTGAAGTGCAA
    GGGCTTCCACGTGCCCGCCAAGGAGCAGGTGGAGGGCATGGGCGCCGCCC
    TGATGAGCATCAAGCTGCAGTTCTGGGCCCCCATGACCCGGAGCGGCGGG
    AAGGAGGTGAGCGGCGAGGGCGGGAGCGGCCAGATCAGCTGCAGCCCCGT
    GTTGGCCGTGGAGCGGCCCATCGCCCTGAGCAAGCAGGCCGTGCGGCGGA
    TGCTGAGCATGAACGTGGAGGGCCGGGACGCCGAGGTGAAGGGCAACCTG
    CTGAAGATGATGAACGACAGCATGGCCAAGAAGACCAGCGGCAACGCCTT
    CATCGGCAAGAAGATGTTCCAGATCAGCGACAAGAACAAGGTGAACCCCA
    TCGAGATCCCCATCAAGCAGACCATCCCCAACTTCTTCTTCGGCCGGGAC
    ACCGCCGAGGACTACGACGACCTGGACTACTGA
  • Alternatively, a human codon-optimized coding region which encodes SEQ ID NO:16 can be designed by the “full optimization” method, where each amino acid is assigned codons based on the frequency of usage in the human genome. These frequencies are shown in Table 12 above. Using this latter method, codons are assigned to the coding region encoding SEQ ID NO:16 as follows: about 10 of the 21 phenylalanine codons are TTT, and about 12 of the phenylalanine codons are TTC; about 3 of the 39 leucine codons are TTA, about 5 of the leucine codons are TTG, about 5 of the leucine codons are CTT, about 8 of the leucine codons are CTC, about 3 of the leucine codons are CTA, and about 16 of the leucine codons are CTG; about 14 of the 38 isoleucine codons are ATT, about 18 of the isoleucine codons are ATC, and about 6 of the isoleucine codons are ATA; the 27 methionine codons are ATG; about 6 of the 32 valine codons are GTT, about 8 of the valine codons are GTG, about 4 of the valine codons are GTA, and about 15 of the valine codons are GTG; about 7 of the 40 serine codons are TCT, about 9 of the serine codons are TCC, about 6 of the serine codons are TCA, about 2 of the serine codons are TCG, about 6 of the serine codons are AGT, and about 10 of the serine codons are AGC; about 7 of the 26 proline codons are CCT, about 9 of the proline codons are CCC, about 7 of the proline codons are CCA, and about 3 of the proline codons are CCG; about 9 of the 38 threonine codons are ACT, about 14 of the threonine codons are ACC, about 11 of the threonine codons are ACA, and about 4 of the threonine codons are ACG; about 11 of the 41 alanine codons are GGT, about 17 of the alanine codons are GCC, about 9 of the alanine codons are GCA, and about 4 of the alanine codons are GCG; about 6 of the 14 tyrosine codons are TAT and about 8 of the tyrosine codons are TAC; about 2 of the 4 histidine codons are CAT and about 2 of the histidine codons are CAC; about 5 of the 19 glutamine codons are CAA and about 14 of the glutamine codons are CAG; about 12 of the 25 asparagine codons are AAT and about 13 of the asparagine codons are AAC; about 22 of the 52 lysine codons are AAA and about 30 of the lysine codons are AAG; about 16 of the 34 aspartic acid codons are GAT and about 18 of the aspartic acid codons are GAC; about 12 of the 30 glutamic acid codons are GAA and about 18 of the glutamic acid codons are GAG; about 2 of the 5 cysteine codons are TGT and about 3 of the cysteine codons are TGC; the single tryptophan codon is TGG; about 2 of the 30 arginine codons are CGT, about 6 of the arginine codons are CGC, about 3 of the arginine codons are CGA, about 6 of the arginine codons are CGG, about 6 of the arginine codons are AGA, and about 6 of the arginine codons are AGG; and about 7 of the 44 glycine codons are GGT, about 15 of the glycine codons are GGC, about 11 of the glycine codons are GGA, and about 11 of the glycine codons are GGG.
  • As described above, the term “about” means that the number of amino acids encoded by a certain codon may be one more or one less than the number given. It would be understood by those of ordinary skill in the art that the total number of any amino acid in the polypeptide sequence must remain constant, therefore, if there is one “more” of one codon encoding a give amino acid, there would have to be one “less” of another codon encoding that same amino acid.
  • A representative “fully optimized” codon-optimized coding region encoding SEQ ID NO:16, optimized according to codon usage in humans is presented herein as SEQ ID NO:36:
    ATGTCGAACATGGACATCGACAGCATTAACACAGGTACTATTGACAAAAC
    CCCCGAAGAACTAACCCCTGGAACCTCAGGAGCAACACGCCCAATAATCA
    AACCGGCCACCCTCGCGCCCCCTAGCAATAAGAGGACCCGCAATCCAAGT
    CCTGAGAGAACCACTACTTCATCTGAAACGGATATCGGTCGGAAAATTCA
    AAAAAAGCAGACGCCCACAGAGATAAAGAAGTCTGTTTACAAAATGGTGG
    TAAAGCTCGGTGAGTTTTATAACCAGATGATGGTCAAGGCGGGGCTTAAC
    GACGATATGGAACGAAATCTTATACAGAATGCACAGGCAGTAGAGAGAAT
    ACTGCTGGCCGCTACTGATGACAAGAAAACGGAGTACCAAAAAAAACGGA
    ATGCTCGAGATGTGAAAGAAGGAAAAGAAGAAATTGACCATAACAAAACT
    GGGGGGACATTCTATAAGATGGTGCGGGACGATAAGACAATCTATTTTAG
    CCCGATAAAGATTACCTTCCTGAAGGAGGAGGTTAAAACAATGTACAAGA
    CGACGATGGGCAGCGATGGTTTTCCGGACTTAATCATATAATGATTGGTC
    ACTCGCAGATGAACGATGTATGTTTCCAGCGCTCCAAGGGCTTAAAGAGG
    GTAGGTCTTGACCCGTCTCTAATATCAACTTTCGCAGGATCCACTTTGCC
    GAGGCGTTCTGGCACGACAGGCGTGGCTATCAAGGGCGGGGGGACGCTGG
    TCGATGAGGCCATTCGCTTTATTGGTAGGGCCATGGCCGATAGAGGGCTT
    CTACGAGACATCAAAGCAAAAACAGCATATGAGAAGATATTATTAAACTT
    AAAGAACAAATGCTCCGCTCCTCAGCAAAAAGCGCTCGTTGACCAAGTAA
    TCGGTTCGAGAAATCCAGGCATTGCCGATATCGAAGATCTTACACTCTTG
    GCGCGAAGCATGGTCGTTGTCCGTCCCAGTGTCGCTAGTAAGGTGGTACT
    ACCAATCTCGATTTACGCAAAAATTCCACAACTCGGCTTTAATACAGAGG
    AATATTCTATGGTAGGTTATGAAGCCATGGCGTTGTATAATATGGCTACA
    CCAGTCTCCATATTGCGTATGGGAGATGACGCAAAAGATAAGAGTCAACT
    CTTTTTCATGTCATGTTTCGGCGCAGCGTACGAAGATCTGAGAGTACTAT
    CCGCCTTGACTGGAACGGAATTTAAACCACGGTCAGCCTTAAAGTGTAAG
    GGTTTTCACGTCCCTGCTAAGGAGCAAGTTGAGGGAATGGGCGCGGCACT
    GATGAGTATAAAATTACAATTTTGGGCTCCAATGACGCGTTCGGGAGGGA
    ATGAAGTTTCTGGTGAGGGAGGGAGTGGACAGATATCATGCTCGCCCGTG
    TTCGCGGTTGAACGTCCGATTGCTTTGAGTAAGCAGGCGGTTAGGCGGAT
    GTTAAGTATGAATGTGGAGGGCCGCGATGCCGACGTCAAAGGCAACTTAT
    TAAAAATGATGAACGACAGCATGGCAAAGAAGACTAGTGGGAATGCTTTT
    ATAGGGAAAAAAATGTTCCAAATAAGTGACAAAAACAAAGTGAACCCCAT
    CGAAATACCTATCAAGCAAACCATCCCGAATTTCTTTTTCGGTCGAGACA
    CCGCGGAGGACTACGATGACCTAGATTACTAA
  • Additionally, a minimally codon-optimized nucleotide sequence encoding SEQ ID NO:16 can be designed by changing only certain codons found more frequently in IV genes than in human genes, as shown in Table 7. For example, if it is desired to substitute more frequently used codons in humans for those codons that occur at least 2 times more frequently in IV genes (designated with an asterisk in Table 7), Arg AGA, which occurs 2.3 times more frequently in IV genes than in human genes, is changed to, e.g., CGG; Asn AAT, which occurs 2.0 times more frequently in IV genes than in human genes, is changed to, e.g., AAC; Ile ATA, which occurs 3.6 times more frequently in IV genes than in human genes, is changed to, e.g., ATC; and Leu CTA, which occurs 2.0 times more frequently in IV genes than is human, is changed to, e.g., CTG.
  • In another form of minimal optimization, a Codon Usage Table (CUT) for the specific IV sequence in question is generated and compared to CUT for human genomic DNA (see Table 7, supra). Amino acids are identified for which there is a difference of at least 10 percentage points in codon usage between human and IV DNA (either more or less). Then the wild type IV codon is modified to conform to predominant human codon for each such amino acid. Furthermore, the remainder of codons for that amino acid are also modified such that they conform to the predominant human codon for each such amino acid.
  • A representative “minimally optimized” codon-optimized coding region encoding SEQ ID NO:16, minimally optimized according to codon usage in humans by this latter method, is presented herein as SEQ ID NO:38:
    ATGTCTAACATGGACATCGACTCTATAAACACAGGCACGATCGATAAGAC
    CCCCGAGGAGCTGACACCCGGGACTTCAGGCGCCACCAGACCCATAATAA
    AGGCCGCGACTCTGGCCCCCCCCTCTAACAAGAGGAGGAGGAACCCCTCT
    CCCGAGCGCACCACAACGAGTAGCGAGACGGACATCGGCAGGAAGATACA
    GAAGAAGCAGACTCCCACTGAGATTAAGAAGTGCGTGTATAAGATGGTGG
    TTAAGCTGGGCGAGTTTTACAACCAGATGATGGTGAAGGCCGGCCTGAAC
    GATGACATGGAGAGGAACCTGATACAGAACGCCCAGGCCGTGGAGAGGAT
    TCTGCTGGCCGCCACCGATGACAAGAAGACTGAGTATCAGAAGAAGAGAA
    ACGCCCGGGACGTTAAGGAGGGCAAGGAGGAGATCGATCACAACAAGACA
    GGCGGCACTTTCTATAAGATGGTCCGTGATGACAAGACAATCTACTTTTC
    TCCCATCAAGATCACATTCCTGAAGGAGGAGGTAAAGACTATGTACAAGA
    CAACTATGGGCTCCGATGGCTTCAGTGGCCTGAACCACATAATGATAGGC
    CATAGTCAGATGAACGATGTGTGCTTCCAGAGAAGCAAGGGCCTGAAGAG
    GGTCGGCCTGGATCCCTCGCTGATTAGTACCTTCGCCGGCAGCACTCTGC
    CCAGAAGATCTGGCACTACTGGCGTAGCCATAAAGGGCGGCGGCACACTG
    GTAGACGAGGCCATAAGGTTTATTGGCAGAGCCATGGCCGACCGGGGGGT
    GCTGAGAGATATGAAGGCCAAGACCGCCTACGAGAAGATACTGCTGAACC
    TGAAGAACAAGTGCTCAGCCCCCCAGCAGAAGGCCCTGGTGGATCAGGTG
    ATCGGCAGTAGAAACCCCGGCATCGCCGACATCGAGGATCTGACTCTGCT
    GGCCAGAAGCATGGTAGTCGTAAGACCCTCTGTGGCCTCTAAGGTTGTGC
    TGCCCATCTCCATCTACGCCAAGATTCCCCAGCTGGGCTTTAACACTGAG
    GAGTACTCCATGGTGGGCTATGAGGCCATGGCCCTGTATAACATGGCCAC
    ACCCGTCTCTATCCTGCGGATGGGCGACGATGCCAAGGACAAGTCTCAGC
    TGTTTTTTATGAGTTGTTTCGGCGCCGCCTATGAGGATCTGAGAGTGCTG
    TCAGCCCTGACAGGCACTGAGTTCAAGCGCAGGTCGGCGCTGAAGTGCAA
    GGGCTTTCATGTGGCGGCCAAGGAGCAGGTGGAGGGCATGGGCGCCGGCC
    TGATGAGCATCAAGCTGCAGTTCTGGGCCCGCATGACCCGGTCTGGCGGC
    AAGGAGGTCTCGGGCGAGGGCGGCAGTGGCCAGATAAGTTGGAGCCCCGT
    TTTTGCCGTTGAGAGACCCATCGCCCTGTCTAAGCAGGCCGTTAGACGAA
    TGCTGAGTATGAACGTCGAGGGCGGAGAGGCCGATGTGAAGGGCAACCTG
    CTGAAGATGATGAACGATTCCATGGCCAAGAAGACAAGCGGCAACGGCTT
    CATTGGCAAGAAGATGTTCCAGATAAGCGATAAGAACAAGGTTAACCCCA
    TCGAGATTCCCATCAAGCAGACCATCCCCAACTTCTTCTTCGGCAGGGAT
    ACCGCCGAGGATTAGGATGACCTGGACTACTGA
  • Randomly assigning codons at an optimized frequency to encode a given polypeptide sequence using the “full-optimization” or “minimal optimization” methods, can be done manually by calculating codon frequencies for each amino acid, and then assigning the codons to the polypeptide sequence randomly. Additionally, various algorithms and computer software programs are readily available to those of ordinary skill in the art. For example, the “EditSeq” function in the Lasergene Package, available from DNAstar, Inc., Madison, Wis., the backtranslation function in the VectorNTI Suite, available from InforMax, Inc., Bethesda, Md., and the “backtranslate” function in the GCG—Wisconsin Package, available from Accelrys, Inc., San Diego, Calif. In addition, various resources are publicly available to codon-optimize coding region sequences. For example, the “backtranslation” function found at http://www.entelechon.com/eng/backtranslation.html (visited Jul. 9, 2002), and the “backtranseq” function available at http:/bioinfo.pbi.nrc.ca:8090/EMBOSS/index.html (visited Oct. 15, 2002). Constructing a rudimentary algorithm to assign codons based on a given frequency can also easily be accomplished with basic mathematical functions by one of ordinary skill in the art.
  • A number of options are available for synthesizing codon-optimized coding regions designed by any of the methods described above, using standard and routine molecular biological manipulations well known to those of ordinary skill in the art. In one approach, a series of complementary oligonucleotide pairs of 80-90 nucleotides each in length and spanning the length of the desired sequence are synthesized by standard methods. These oligonucleotide pairs are synthesized such that upon annealing, they form double stranded fragments of 80-90 base pairs, containing cohesive ends, e.g., each oligonucleotide in the pair is synthesized to extend 3, 4, 5, 6, 7, 8, 9, 10, or more bases beyond the region that is complementary to the other oligonucleotide in the pair. The single-stranded ends of each pair of oligonucleotides is designed to anneal with the single-stranded end of another pair of oligonucleotides. The oligonucleotide pairs are allowed to anneal, and approximately five to six of these double-stranded fragments are then allowed to anneal together via the cohesive single stranded ends, and then they ligated together and cloned into a standard bacterial cloning vector, for example, a TOPO® vector available from Invitrogen Corporation, Carlsbad, Calif. The construct is then sequenced by standard methods. Several of these constructs consisting of 5 to 6 fragments of 80 to 90 base pair fragments ligated together, i.e., fragments of about 500 base pairs, are prepared, such that the entire desired sequence is represented in a series of plasmid constructs. The inserts of these plasmids are then cut with appropriate restriction enzymes and ligated together to form the final construct. The final construct is then cloned into a standard bacterial cloning vector, and sequenced. Additional methods would be immediately apparent to the skilled artisan. In addition, gene synthesis is readily available commercially.
  • The codon-optimized coding regions can be versions encoding any gene products from any strain, derivative, or variant of IV, or fragments, variants, or derivatives of such gene products. For example, nucleic acid fragments of codon-optimized coding regions encoding the NP, M1 and M2 polypeptides, or fragments, variants or derivatives thereof. Codon-optimized coding regions encoding other IV polypeptides or fragments, variants, or derivatives thereof (e.g. HA, NA, PB1, PB2, PA, NS1 or NS2), are included within the present invention. Additional, non-codon-optimized polynucleotides encoding IV polypeptides or other polypeptides are included as well.
  • Consensus Sequences
  • The present invention is further directed to specific consensus sequences of influenza virus proteins, and fragments, derivatives and variants thereof. A “consensus sequence” is, e.g., an idealized sequence that represents the amino acids most often present at each position of two or more sequences which have been compared to each other. A consensus sequence is a theoretical representative amino acid sequence in which each amino acid is the one which occurs most frequently at that site in the different sequences which occur in nature. The term also refers to an actual sequence which approximates the theoretical consensus. A consensus sequence can be derived from sequences which have, e.g., shared functional or structural purposes. It can be defined by aligning as many known examples of a particular structural or functional domain as possible to maximize the homology. A sequence is generally accepted as a consensus when each particular amino acid is reasonably predominant at its position, and most of the sequences which form the basis of the comparison are related to the consensus by rather few substitutions, e.g., from 0 to about 100 substitutions. In general, the wild-type comparison sequences are at least about 50%, 75%, 80%, 90%, 95%, 96%, 97%, 98% or 99% identical to the consensus sequence. Accordingly, polypeptides of the invention are about 50%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the consensus sequence. Consensus amino acid sequences can be prepared for any of the influenza antigens. By analyzing amino acid sequences from influenza A strains sequenced since 1990, consensus amino acid sequences were derived for the influenza A NP (SEQ ID NO: 76), M1 (SEQ ID NO:77) and M2 (SEQ ID NO:78) proteins (Example 3).
  • A “consensus amino acid” is an amino acid chosen to occupy a given position in the consensus protein. A system which is organized to select consensus amino acids can be a computer program, or a combination of one or more computer programs with “by hand” analysis and calculation. When a consensus amino acid is obtained for each position of the aligned amino acid sequences, then these consensus amino acids are “lined up” to obtain the amino acid sequence of the consensus protein.
  • Another embodiment of this invention is directed to a process for the preparation of a consensus protein comprising a process to calculate an amino acid residue for nearly all positions of a so-called consensus protein and to synthesize a complete gene from this sequence that could be expressed in a prokaryotic or eukaryotic expression system.
  • Polynucleotides which encode the consensus influenza polypeptides, or fragments, variants or derivatives thereof, are also part of this invention. Such polynucleotides can be obtained by known methods, for example by backtranslation of the amino acid sequence and PCR synthesis of the corresponding polynucleotide.
  • Compositions and Methods
  • In certain embodiments, the present invention is directed to compositions and methods of enhancing the immune response of a vertebrate in need of protection against IV infection by administering in vivo, into a tissue of a vertebrate, one or more polynucleotides comprising at least one codon-optimized coding region encoding an IV polypeptide, or a fragment, variant, or derivative thereof. In addition, the present invention is directed to compositions and methods of enhancing the immune response of a vertebrate in need of protection against IV infection by administering to the vertebrate a composition comprising one or more polynucleotides as described herein, and at least one isolated IV polypeptide, or a fragment, variant, or derivative thereof. The polynucleotide may be administered either prior to, at the same time (simultaneously), or subsequent to the administration of the isolated polypeptide.
  • The coding regions encoding IV polypeptides or fragments, variants, or derivatives thereof may be codon optimized for a particular vertebrate. Codon optimization is carried out by the methods described herein, for example, in certain embodiments codon-optimized coding regions encoding polypeptides of IV, or nucleic acid fragments of such coding regions encoding fragments, variants, or derivatives thereof are optimized according to the codon usage of the particular vertebrate. The polynucleotides of the invention are incorporated into the cells of the vertebrate in vivo, and an immunologically effective amount of an IV polypeptide or a fragment, variant, or derivative thereof is produced in vivo. The coding regions encoding an IV polypeptide or a fragment, variant, or derivative thereof may be codon optimized for mammals, e.g., humans, apes, monkeys (e.g., owl, squirrel, cebus, rhesus, African green, patas, cynomolgus, and cercopithecus), orangutans, baboons, gibbons, and chimpanzees, dogs, wolves, cats, lions, and tigers, horses, donkeys, zebras, cows, pigs, sheep, deer, giraffes, bears, rabbits, mice, ferrets, seals, whales; birds, e.g., ducks, geese, terns, shearwaters, gulls, turkeys, chickens, quail, pheasants, geese, starlings and budgerigars, or other vertebrates.
  • In one embodiment, the present invention relates to codon-optimized coding regions encoding polypeptides of IV, or nucleic acid fragments of such coding regions fragments, variants, or derivatives thereof which have been optimized according to human codon usage. For example, human codon-optimized coding regions encoding polypeptides of IV, or fragments, variants, or derivatives thereof are prepared by substituting one or more codons preferred for use in human genes for the codons naturally used in the the DNA sequence encoding the IV polypeptide or a fragment, variant, or derivative thereof. Also provided are polynucleotides, vectors, and other expression constructs comprising codon-optimized coding regions encoding polypeptides of IV, or nucleic acid fragments of such coding regions encoding fragments, variants, or derivatives thereof, pharmaceutical compositions comprising polynucleotides, vectors, and other expression constructs comprising codon-optimized coding regions -encoding polypeptides of IV, or nucleic acid fragments of such coding regions encoding fragments, variants, or derivatives thereof, and various methods of using such polynucleotides, vectors and other expression constructs. Coding regions encoding IV polypeptides can be uniformly optimized, fully optimized, minimally optimized, codon-optimized by region and/or not codon-optimized, as described herein.
  • The present invention is further directed towards polynucleotides comprising codon-optimized coding regions encoding polypeptides of IV antigens, for example, HA, NA, NP, M1 and M2, optionally in conjunction with other antigens. The invention is also directed to polynucleotides comprising codon-optimized nucleic acid fragments encoding fragments, variants and derivatives of these polypeptides, e.g., an eM2 or a fusion of NP and eM2.
  • In certain embodiments, the present invention provides an isolated polynucleotide comprising a nucleic acid fragment, where the nucleic acid fragment is a fragment of a codon-optimized coding region encoding a polypeptide at least 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to an IV polypeptide, e.g., HA, NA, NP, M1 or M2, and where the nucleic acid fragment is a variant of a codon-optimized coding region encoding an IV polypeptide, e.g., HA, NA, NP, M1 or M2. The human codon-optimized coding region can be optimized for any vertebrate species and by any of the methods described herein.
  • Isolated IV Polypeptides
  • The present invention is further drawn to compositions which include at least one polynucleotide comprising one or more nucleic acid fragments, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide or fragment, variant, or derivative thereof; together with one or more isolated IV component or isolated polypeptide. The IV component may be inactivated virus, attenuated virus, a viral vector expressing an isolated influenza virus polypeptide, or an influenza virus protein, fragment, variant or derivative thereof.
  • The polypeptides or fragments, variants or derivatives thereof, in combination with the codon-optimized nucleic acid compositions may be referred to as “combinatorial polynucleotide vaccine compositions” or “single formulation heterologous prime-boost vaccine compositions.”
  • The isolated IV polypeptides of the invention may be in any form, and are generated using techniques well known in the art. Examples include isolated IV proteins produced recombinantly, isolated IV proteins directly purified from their natural milieu, recombinant (non-IV) virus vectors expressing an isolated IV protein, or proteins delivered in the form of an inactivated IV vaccine, such as conventional vaccines
  • When utilized, an isolated IV polypeptide or fragment, variant or derivative thereof is administered in an immunologically effective amount. Conventional IV vaccines have been standardized to micrograms of viral antigens HA and NA. See Subbarao, K., Advances in Viral Research 54:349-373 (1999), incorporated herein by reference in its entirety. The recommended dose for these vaccines is 15 μg of each HA per 0.5 ml. Id. The effective amount of conventional IV vaccines is determinable by one of ordinary skill in the art based upon several factors, including the antigen being expressed, the age and weight of the subject, and the precise condition requiring treatment and its severity, and route of administration.
  • In the instant invention, the combination of conventional antigen vaccine compositions with the codon-optimized nucleic acid compositions provides for therapeutically beneficial effects at dose sparing concentrations. For example, immunological responses sufficient for a therapeutically beneficial effect in patients predetermined for an approved commercial product, such as for the conventional product described above, can be attained by using less of the approved commercial product when supplemented or enhanced with the appropriate amount of codon-optimized nucleic acid. Thus, dose sparing is contemplated by administration of conventional IV vaccines administered in combination with the codon-optimized nucleic acids of the invention
  • In particular, the dose of conventional vaccine may be reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% or at least 70% when administered in combination with the codon-optimized nucleic acid compositions of the invention.
  • Similarly, a desirable level of an immunological response afforded by a DNA based pharmaceutical alone may be attained with less DNA by including an aliquot of a conventional vaccine. Further, using a combination of conventional and DNA based pharmaceuticals may allow both materials to be used in lesser amounts while still affording the desired level of immune response arising from administration of either component alone in higher amounts (e.g. one may use less of either immunological product when they are used in combination). This may be manifest not only by using lower amounts of materials being delivered at any time, but also to reducing the number of administrations points in a vaccination regime (e.g. 2 versus 3 or 4 injections), and/or to reducing the kinetics of the immunological response (e.g. desired response levels are attained in 3 weeks in stead of 6 after immunization).
  • In particular, the dose of DNA based pharmaceuticals, may be reduced by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% or at least 70% when administered in combination with conventional IV vaccines.
  • Determining the precise amounts of DNA based pharmaceutical and conventional antigen is based on a number of factors as described above, and is readily determined by one of ordinary skill in the art.
  • In addition to dose sparing, the claimed combinatorial compositions provide for a broadening of the immune response and/or enhanced beneficial immune responses. Such broadened or enhanced immune responses are achieved by: adding DNA to enhance cellular responses to a conventional vaccine; adding a conventional vaccine to a DNA pharmaceutical to enhance humoral response; using a combination that induces additional epitopes (both humoral and/or cellular) to be recognized and/or more desirably responded to (epitope broadening); employing a DNA-conventional vaccine combination designed for a particular desired spectrum of immunological responses; obtaining a desirable spectrum by using higher amounts of either component. The broadened immune response is measurable by one of ordinary skill in the art by standard immunological assay specific for the desirable response spectrum.
  • Both broadening and dose sparing can be obtained simultaneously.
  • The isolated IV polypeptide or fragment, variant, or derivative thereof to be delivered (either a recombinant protein, a purified subunit, or viral vector expressing an isolated IV polypeptide, or in the form of an inactivated IV vaccine) can be any isolated IV polypeptide or fragment, variant, or derivative thereof, including but not limited to the HA, NA, NP, M1, or M2 proteins or fragments, variants or derivatives thereof. Fragments include, but are not limited to, the eM2 protein. In certain embodiments, a derivative protein can be a fusion protein, e.g., NP-eM2. It should be noted that any isolated IV polypeptide or fragment, variant, or derivative thereof described herein can be combined in a composition with any polynucleotide comprising a nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide or fragment, variant, or derivative thereof. The proteins can be different, the same, or can be combined in any combination of one or more isolated IV proteins and one or more polynucleotides.
  • In certain embodiments, the isolated IV polypeptides, or fragments, derivatives or variants thereof can be fused to or conjugated to a second isolated IV polypeptide, or fragment, derivative or variant thereof, or can be fused to other heterologous proteins, including for example, hepatitis B proteins including, but not limited to the hepatitis B core antigen (HBcAg), or those derived from diphtheria or tetanus. The second isolated IV polypeptide or other heterologous protein can act as a “carrier” that potentiates the immunogenicity of the IV polypeptide or a fragment, variant, or derivative thereof to which it is attached. Hepatitis B virus proteins and fragments and variants thereof useful as carriers within the scope of the invention are disclosed in U.S. Pat. Nos. 6,231,864 and 5,143,726, which are incorporated by reference in their entireties. Polynucleotides comprising coding regions encoding said fused or conjugated proteins are also within the scope of the invention.
  • The use of recombinant particles comprising hepatitis B core antigen (“HBcAg”) and heterologous protein sequences as potent immunogenic moieties is well documented. For example, addition of heterologous sequences to the amino terminus of a recombinant HBcAg results in the spontaneous assembly of particulate structures which express the heterologous epitope on their surface, and which are highly immunogenic when inoculated into experimental animals. See Clarke et al., Nature 330:381-384 (1987). Heterologous epitopes can also be inserted into HBcAg particles by replacing approximately 40 amino acids of the carboxy terminus of the protein with the heterologous sequences. These recombinant HBcAg proteins also spontaneously form immunogenic particles. See Stahl and Murray, Proc. Natl. Acad. Sci. USA, 86:6283-6287 (1989). Additionally, chimeric HBcAg particles may be constructed where the heterologous epitope is inserted in or replaces all or part of the sequence of amino acid residues in a more central region of the HBcAg protein, in an immunodominant loop, thereby allowing the heterologous epitope to be displayed on the surface of the resulting particles. See EP Patent No. 0421635 B1. Shown below are the DNA and amino acid sequences of the human hepatitis B core protein (HBc), subtype ayw (SEQ ID NOs 39 and 40), as described in Galibert, F., et al., Nature 281:646-650 (1979); see also U.S. Pat. Nos. 4,818,527, 4,882,145 and 5,143,726. All of the above references are incorporated herein by reference in their entireties. The nucleotide and amino acid sequences are presented herein as SEQ ID NO 39:
    ATGGACATCGACCCTTATAAAGAATTTGGAGCTACTGTGGAGTTACTCTC
    GTTTTTGCCTTCTGACTTCTTTCCTTCAGTACGAGATCTTCTAGATACCG
    CCTCAGCTCTGTATCGGGAAGCCTTAGAGTCTCCTGAGCATTGTTCACCT
    CACCATACTGCACTCAGGCAAGCAATTCTTTGCTGGGGGGAACTAATGAC
    TCTAGCTACCTGGGTGGGTGTTAATTTGGAAGATCCAGCGTCTAGAGACC
    TAGTAGTCAGTTATGTCAACACTAATATGGGCCTAAAGTTCAGGCAACTC
    TTGTGGTTTCACATTTCTTGTCTCACTTTTGGAAGAGAAACAGTTATAGA
    GTATTTGGTGTCTTTCGGAGTGTGGATTCGCACTCCTCCAGCTTATAGAC
    CACCAAATGCCCCTATCCTATCAACACTTCCGGAGACTACTGTTGTTAGA
    CGACGAGGCAGGTCCCCTAGAAGAAGAACTCCCTCGCCTCGCAGACGAAG
    GTCTCAATCGCCGCGTCGCAGAAGATCTCAATCTCGGGAATCTCAATG
    TTAG
  • and SEQ ID NO:40:
    MDIDPYKEFGATVELLSFLPSDFFPSVRDLLDTASALYREALESPEHCSP
    HHTALRQAILCWGELMTLATWVGVNLEDPASRDLVVSYVNTNMGLKFRQL
    LWFHISCLTFGRETVIEYLVSFGVWIRTPPAYRPPNAPILSTLPETTVVR
    RRGRSPRRRTPSPRRRRSQSPRRRRSQSRESQC
  • A completely synthetic HBcAg has been synthesized as well. See Nassal, M. Gene 66:279-294 (1988). The nucleotide and amino acid sequences are presented herein as SEQ ID NO 41:
    ATGGATATCGATCCTTATAAAGAATTCGGAGCTACTGGGAGTTACTCTCG
    TTTCTCCCGAGTGACTTCTTTCCTTCAGTACGAGATCTTCTGGATACCGC
    CAGCGCGCTGTATCGGGAAGCCTTGGAGTCTCCTGAGCACTGCAGCCCTC
    ACCATACTGCCCTCAGGCAAGCAATTCTTTGCTGGGGGGAGCTCATGACT
    CTGGCCACGTGGGTGGGTGTTAACTTGGAAGATCCAGCTAGCAGGGACCT
    GGTAGTCAGTTATGTCAACACTAATATGGGTTTAAAGTTCAGGCAACTCT
    TGTGGTTTCACATTAGCTGCCTCACTTTCGGCCGAGAAACAGTTCTAGAA
    TATTTGGTGTCTTTCGGAGTGTGGATCCGCACTCCTCCAGCTTATAGGCC
    TCCGAATGCCCCTATCCTGTCGACACTCCCGGAGACTACTGTTGTTAGAC
    GTCGAGGCAGGTCACCTAGAAGAAGAACTCCTTCGCCTCGCAGGCGAAGG
    TCTCAATCGCCGCGGCGCCGAAGATCTCAATCTCGGGAATCTCAATGTTA
    GTGA
  • and SEQ ID NO:42:
    MDIDPYKEFGATVELLSFLPSDFFPSVRDLLDTASALYREALESPEHCSP
    HHTALRQAILCWGELMTLATWVGVNLEDPASRDLVVSYVNTNMGLKFRQL
    LWFHISCLTFGRETVLEYLVSFGVWIRTPPAYRPPNAPILSTLPETTVVR
    RRGRSPRRRTPSPRRRRSQSPRRRRSQSRESQC
  • Chimaeric HBcAg particles comprising isolated IV proteins or variants, fragments or derivatives thereof are prepared by recombinant techniques well known to those of ordinary skill in the art. A polynucleotide, e.g., a plasmid, which carries the coding region for the HBcAg operably associated with a promoter is constructed. Convenient restrictions sites are engineered into the coding region encoding the N-terminal, central, and/or C-terminal portions of the HBcAg, such that heterologous sequences may be inserted. A construct which expresses a HBcAg/IV fusion protein is prepared by inserting a DNA sequence encoding an IV protein or variant, fragment or derivative thereof, in frame, into a desired restriction site in the coding region of the HBcAg. The resulting construct is then inserted into a suitable host cell, e.g., E. coli, under conditions where the chimeric HBcAg will be expressed. The chimaeric HBcAg self-assembles into particles when expressed, and can then be isolated, e.g., by ultracentrifugation. The particles formed resemble the natural 27 nm HBcAg particles isolated from a hepatitis B virus, except that an isolated IV protein or fragment, variant, or derivative thereof is contained in the particle, preferably exposed on the outer particle surface.
  • The IV protein or fragment, variant, or derivative thereof expressed in a chimaeric HBcAg particle may be of any size which allows suitable particles of the chimeric HBcAg to self-assemble. As discussed above, even small antigenic epitopes may be immunogenic when expressed in the context of an immunogenic carrier, e.g., a HBcAg. Thus, HBcAg particles of the invention may comprise at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, or between about 15 to about 30 amino acids of an IV protein fragment of interest inserted therein. HBcAg particles of the invention may further comprise immunogenic or antigenic epitopes of at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues of an IV protein fragment of interest inserted therein.
  • The immunodominant loop region of HBcAg was mapped to about amino acid residues 75 to 83, to about amino acids 75 to 85 or to about amino acids 130 to 140. See Colucci et al., J. Immunol. 141:4376-4380 (1988), and Salfeld et al. J. Virol. 63:798 (1989), which are incorporated by reference. A chimeric HBcAg is still often able to form core particles when foreign epitopes are cloned into the immunodominant loop. Thus, for example, amino acids of the IV protein fragment may be inserted into the sequence of HBcAg amino acids at various positions, for example, at the N-terminus, from about amino acid 75 to about amino acid 85, from about amino acid 75 to about amino acid 83, from about amino acid 130 to about amino acid 140, or at the C-terminus. Where amino acids of the IV protein fragment replace all or part of the native core protein sequence, the inserted IV sequence is generally not shorter, but may be longer, than the HBcAg sequence it replaces.
  • Alternatively, if particle formation is not desired, full-length IV coding sequences can be fused to the coding region for the HBcAg. The HBcAg sequences can be fused either at the N- or C-terminus of any of the Influenza antigens described herein, including the eM2-NP constructs. Fusions could include flexible protein linkers as described for NP-eM2 fusions above. Examples of IV coding sequences fused to the HBcAg coding sequence of SEQ ID NO:41 include an IAV NP-HBcAg fusion (SEQ ID NO:43),
    ATGGCGTCTCAAGGCACCAAACGATCTTACGAACAGATGGAGACTGATG
    GAGAACGCCAGAATGCCACTGAAATCAGAGCATCCGTCGGAAAAATGAT
    TGGTGGAATTGGACGATTCTACATCCAAATGTGCACCGAACTCAAACTCA
    GTGATTATGAGGGACGGTTGATCCAAAACAGCTTAACAATAGAGAGAAT
    GGTGCTCTCTGCTTTTGACGAAAGGAGAAATAAATACCTTGAAGAACATC
    CCAGTGCGGGGAAAGATCCTAAGAAAACTGGAGGACCTATATACAGGAG
    AGTAAACGGAAAGTGGATGAGAGAACTCATCCTTTATGACAAAGAAGAA
    ATAAGGCGAATCTGGCGCCAAGCTAATAATGGTGACGATGCAACGGCTG
    GTCTGACTCACATGATGATCTGGGATTCCAATTTGAATGATGCAACTTAT
    CAGAGGACAAGAGCTCTTGTTCGCACCGGAATGGATCCCAGGATGTGCTC
    TCTGATGCAAGGTTCAACTCTCCCTAGGAGGTCTGGAGCCGCAGGTGCTG
    CAGTCAAAGGAGTTTGGAACAATGGTGATGGAATTGGTCAGAATGATCAA
    ACGTGGGATCAATGATCGGAACTTCTGGAGGGGTGAGAATGGACGAAAAA
    CAAGAATTGCTTATGAAAGAATGTGCAACATTCTCAAAGGGAAATTTCAA
    ACTGCTGCACAAAAAGCAATGATGGATCAAGTGAGAGAGAGCCGGAACC
    CAGGGAATGCTGAGTTTCGAAGATGTCAGTTTCTAGCACGGTCTGCACTC
    ATATTGAGAGGGTCGGTTGCTCACAAGTCCTGCCTGCCTGCCTGTGTGTA
    TGGACCTGGCGTAGCCAGTGGGTACGACTTTGAAAGGGAGGGATACTCTC
    TAGTCGGAATAGACCGTTTCAGACTGCTTCAAAACAGCCAAGTGTACAGC
    CTAATCAGACCAAATGAGAATCCAGGACACAAGAGTCAACTGGTGTGGA
    TGGCATGCCATTCTGCCGCATTTGAAGATCTAAGAGTATTAAGCTTCATC
    AAAGGGACGAAGGTGCTCCCAAGAGGGAAGCTTTCCACTAGAGGAGTTC
    AAATTGCTTCCAATGAAAATATGGAGACTATGGAATCAAGTACACTTGAA
    CTGAGAAGCAGGTACTGGGCCATAAGGACCAGAAGTGGAGGAAACACCA
    ATCAACAGAGGGCATCTGCGGGCCAAATCAGCATACAAGGTACGTTCTCA
    GTACAGAGAAATCTCCCTTTTGACAGAACAACCGTTATGGCAGCATTCAG
    TGGGAATACAGAGGGGAGATGGCGTCTCAAGGCACCAAACGATCTACG
    AACAGATGGAGACTGATGGAGAACGCCAGAATGCCACTGAAATCAGAGG
    ATCCGTCGGAAAAATGATTGGTGGAATGGACGATTCTACATCCAAATGT
    GCACCGAACTCAAACTCAGTGATTATGAGGGACGGTTGATCCAAAACAG
    CTTAACAATAGAGAGAATGGTGCTCTCTGCTTTTGACGAAAGGAGAAATA
    AATACCTTGAAGAACATCCCAGTGCGGGGAAAGATCCTAAGAAAACTGG
    AGGACCTATATACAGGAGAGTAAACGGAAAGTGGATGAGAGAACTCATC
    CTTTATGACAAAGAAGAAATAAGGCGAATCTGGCGCCAAGCTAATAATG
    GTGACGATGCAACGGCTGGTCTGACTCACATGATGATCTGGCATTCCAAT
    TTGAATGATGCAACTTATCAGAGGACAAGAGCTCTTGTTCGCACCGGAAT
    GGATCCCAGGATGTGCTCTCTGATGCAAGGTTCAACTCTCCCTAGGAGGT
    CTGGAGCCGCAGGTGCTGCAGTCAAAGGAGTTGGAACAATGGTGATGGA
    ATTGGTCAGAATGATCAAACGTGGGATCAATGATCGGAACTTCTGGAGG
    GGTGAGAATGGACGAAAAACAAGAATTGCTTATGAAAGAATGTGCAACA
    TTCTCAAAGGGAAATTTCAAACTGGTGCAGAAAAAGGAATGATGGATCA
    AGTGAGAGAGAGGCGGAAGCCAGGGAATGCTGAGTTCGAAGATCTCACT
    TTTCTAGCACGGTCTGCACTCATATTGAGAGGGTCGGTTGCTCACAAGTC
    CTGCCTGCCTGCCTGTGTGTATGGACCTGCCGTAGCCAGTGGGTACGACT
    TTGAAAGGGAGGGATACTCTCTAGTCGGAATAGACCCTTTCAGACTGCTT
    CAAAACAGCCAAGTGTACAGCCTAATCAGACCAAATGAGAATCCAGCAC
    ACAAGAGTCAACTGGTGTGGATGGCATGCGATTCTGCCGCATTTGAAGAT
    CTAAGAGTATTAAGCTTCATCAAAGGGACGAAGGTGGTCCCAAGAGGGA
    AGCTTTCCACTAGAGGAGTTCAAATTGCTTCCAATGAAAATATGGAGACT
    ATGGAATCAAGTACACTTGAACTGAGAAGCAGGTACTGGGCGATAAGGA
    CCAGAAGTGGAGGAAACACCAATCAACAGAGGGCATCTGCGGGCCAAAT
    CAGCATACAACCTACGTTCTCAGTACAGAGAAATCTCCCTTTTGACAGAA
    CAACCGTTATGGCAGCATTCAGTGGGAATACAGAGGGGAGAACATCTGA
    CATGAGGACCGAAATGATAAGGATGATGGAAAGTGGAAGACCAGAAGAT
    GTGTCTTCCAGGGGCGGGGAGTCTTCGAGCTCTCGGACGAAAAGGCAGC
    GAGCCCGATCGTGCCTTCCTTTGACATGAGTAATGAAGGATCTTATTTC
    TTCGGAGACAATGCAGAGGAATACGATAATATGGATATCGATCCTTATA
    AAGAATTCGGAGCTACTGTGGAGTTACTCTCGTTTCTCCCGAGTGACTT
    CTTTCCTTCAGTACGAGATCTTCTGGATACCGGCAGCGCGCTGTATCGG
    GAAGCCTTGGAGTCTCCTGAGCACTGCAGCCCTGACCATACTGGCCTCA
    GGGAAGCAATTCTTTGCTGGGGGGAGCTCATGACTCTGGCCACGTGGGT
    GGGTGTTAACTTGGAAGATGGAGCTAGCAGGGACCTGGTAGTCAGTTAT
    GTCAACACTAATATGGGTTTAAAGTTCAGGCAACTCTTGTGGTTTCACA
    TTAGCTGCCTCACTTTCGGCCGAGAAACAGTTCTAGAATATTTGGTGTC
    TTTCGGAGTGTGGATCCGCACTCCTCCAGCTTATAGGCCTCCGAATGCC
    CCTATCCTGTCGACAGTCCCGGAGACTACTGTTTGTTAGACGTCGAGGC
    AGGTCACCTAGAAGAAGAACTCCTTCGCCTCGCAGGCGAAGGTCTCAAT
    CGCCGCGGCGCCGAAGATCTCAATCTCGGGAATCTCAATGT
  • an IBV NP-HBcAg fusion (SEQ ID NO:44),
    ATGTCCAACATGGATATTGACAGTATAAATACCGGAACAATGGATAAAA
    GACCAGAAGAACTGACTGCCGGAACCAGTGGGGCAACCAGACCAATCAT
    CAAGCCAGCAACCCTTGCTCCGCCAAGCAACAAACGAACCCGAAATCCA
    TCTCCAGAAAGGACAACCACAAGCAGTGAAACCGATATCGGAAGGAAAA
    TCCAAAAGAAACAAACCCCAACAGAGATAAAGAAGAGCGTCTACAAAAT
    GGTGGTAAAACTGGGTGAATTTGTACAACCAGATGATGGTCAAAGGTGGA
    CTTAATGATGACATGGAAAGGAATCTAATTCAAAATGCACAAGCTGTGG
    AGAGAATCCTATTGGCTGCAACTGATGACAAGAAAACTGAATACCAAAA
    GAAAAGGAATGCGAGAGATGTCAAAGAAGGGAAGGAAGAAATAGACCA
    CAACAAGACAGGAGGCACCTTTTATAAGATGGTAAGAGATGATAAAACC
    ATCTACTTCAGCCCTATAAAAATTACCTTTTTAAAAGAAGAGGTGAAAAC
    AATGTACAAGACCACCATGGGGAGTGATGGTTTCAGTGGACTAAATCAC
    ATTATGATTGGACATTCACAGATGAACGATGTCTGTTTCCAAAGATCAAA
    GGGACTGAAAAGGGTTGGACTTGACCCTTCATTAATCAGTACTTTTGCCG
    GAAGGACAGTACCCAGAAGATCAGGTACAACTGGTGTTGCAATCAAAGG
    AGGTGGAACTTAGTGGATGAAGCCATCCGATTTATAGGAAGAGCAATG
    GCAGACAGAGGGGTACTGAGAGACATGAAGGCCAAGACGGCCTATGAAA
    AGATTCTTCTGAATGTGAAAAACAAGTGCTCTGCGGCGCAACAAAAGGCT
    CTAGTTTGATCAAGTGATCGGAAGTAGGAACCCAGGGATTGCAGACATAG
    AAGACCTAACTCTGCTTGCCAGAAGCATGGTAGTTGTCAGACCCTCTGTA
    GCGAGCAAAGTGGTGCTTCCCATAAGGATTTATGCTAAAATACCTCAACT
    AGGATTCAATACCGAAGAATACTCTATGGTTGGGTATGAAGCCATGGCTC
    TTTATAATATGGCAACACCTGTTCCATATTAAGAATGGGAGATGACGCA
    AAAGATAAATCTCAACTATTCTTCATGTCGTGCTTCGGAGCTGCCTATGA
    AGATCTAAGAGTGTTATCTGCACTAACGGGCACCGAATTTAAGCCTAGAT
    CAGCACTAAAATGCAAGGGTTTCCATGTCCCGGCTAAGGAGCAAGTAGA
    AGGAATGGGGGCAGCTCTGATGTCCATCAAGCTTCAGTTCTGGGCCCCAA
    TGACCAGATCTGGAGGGAATGAAGTAAGTGGAGAAGGAGGGTCTGGTGA
    AATAAGTTGCAGCCCTGTGTTTGCAGTAGAAAGACCTATTGCTCTAAGGA
    AGCAAGCTGTAAGAAGAATGCTGTCAATGAACGTTGAAGGACGTGATGC
    AGATGTCAAAGGAAATCTACTCAAAATGATGAATGATTCAATGGCAAAG
    AAAACCAGTGGAAATGCTTTCATTGGGAAGAAAATGTTTCAAATATCAGA
    CAAAAACAAAGTCAATCCCATTGAGATTCCAATTAAGCAGACCATCCCCA
    ATTTCTTCTTTGGGAGGGACACAGCAGAGGATTATGATGACCTCGATTAT
    ATGGATATCGATCCTTATAAAGAATTCGGAGCTACTGTGGAGTTACTCTC
    GTTTCTCCCGAGTGACTTCTTCCTTCAGTACGAGATCTTCTGGATACCGC
    CAGCGCGCTGTATCGGGAAGCCTTGGAGTCTCCTGAGCACTGCAGCCCTC
    ACCATACTGCCCTCAGGCAAGCAATTCTTTGCTGGGGGGAGCTCATGACT
    CTGGCCACGTGGGTGGGTGTTAACTTGGAAGATCCAGCTAGCAGGGACCT
    GGTAGTCAGTTATGTCAACACTAATATGGGTTTAAAGTTCAGGCAACTCT
    TGTGGTTTGACATTAGCTGCCTCACTTTCGGCCGAGAAACAGTTCTAGAA
    TATTTGGTGTCTTTCGGAGTGTGGATCCGCACTCCTCCAGCTTATAGGCC
    TCCGAATGCCCCTATCCTGTCGACACTCCCGGAGACTACTGTTGTTAGAC
    GTCGAGGCAGGTCACCTAGAAGAAGAACTCCTTCGCCTCGCAGGCGAAGG
    TCTCAATCGCCGCGGCGCCGAAGATCTCAATCTCGGGAATCTCAATGTT
  • or an IAV M1-HBcAg fusion (SEQ ID NO:45),
    ATGAGTCTTCTAACCGAGGTCGAAACGTACGTACTCTCTATCATCCCGTC
    AGGCCCCCTCAAAGCCGAGATCGCACAGAGACTTGAAGATGTCTTTGCAG
    GGAAGAACACTGATCTTGAGGTTCTCATGGAATGGCTAAAGACAAGACC
    AATCCTGTCACCTCTGACTAAGGGGATTTTAGGATTTGTGTTCACGCTCA
    CCGTGCCCAGTGAGCGAGGACTGCAGCGTAGACGCTTTGTCCAAAATGCC
    CTTAATGGGAACGGGGATCCAAATAACATGGACAAAGCAGTTAAACTGTA
    TAGGAAGCTCAAGAGGGAGATAACATTCCATGGGGCCAAAGAAATCTCA
    CTCAGTTATTCTGCTGGTGCACTTGCCAGTTGTATGGGCCTCATATACAA
    CAGGATGGGGGCTGTGACCACTGAAGTGGCATTTGGCCTGGTATGTGCAA
    CCTGTGAACAGATTGCTGACTCCCAGCATCGGTCTCATAGGCAAATGGTG
    ACAACAACCAATCCACTAATCAGACATGAGAACAGAATGGTTTTAGCCAG
    CACTACAGCTAAGGCTATGGAGCAAATGGCTGGATCGAGTGAGCAAGCA
    GCAGAGGCCATGGAGGTTGCTAGTCAGGCTAGACAAATGGTGCAAGCGA
    TGAGAACCATTGGGACTCATCCTAGCTCCAGTGCTGGTCTGAAAAATGAT
    CTTCTTGAAAATTTGCAGGCCTATCAGAAACGAATGGGGGTGCAGATGCA
    ACGGTTCAAGATGGATATCGATCCTTATAAAGAATTCGGAGCTACTGTGG
    AGTTACTCTCGTTTCTCCCGAGTGACTTCTTTCCTTCAGTACGAGATCTT
    CTGGATACCGCCAGCGCGCTGTATCGGGAAGCCTTGGAGTCTCCTGAGCA
    CTGCAGCCCTCACCATACTGCCCTCAGGCAAGCAATTCTTTGCTGGGGGG
    AGCTCATGACTCTGGCCACGTGGGTGGGTGTTAACTTGGAAGATCCAGCT
    AGCAGGGACCTGGTAGTCAGTTATGTCAACACTAATATGGGTTTAAAGTT
    CAGGCAACTCTTGTGGTTTCACATTAGCTGCCTCACTTTCGGCCGAGAAA
    CAGTTCTAGAATATTTGGTGTCTTTCGGAGTGTGGATCCGCACTCCTCCA
    GCTTATAGGCCTCCGAATGCCCCTATCCTGTCGACACTCCCGGAGACTAC
    TGTTGTTAGACGTCGAGGCAGGTCACCTAGAAGAAGAACTCCTTCGCCTC
    GCAGGCGAAGGTCTCAATCGCCGCGGCGCCGAAGATCTCAATCTCGGGAA
    TCTCAATGT
  • These fusion constructs could be codon optimized by any of the methods described.
  • The chimeric HBcAg can be used in the present invention in conjunction with a polynucleotide comprising a nucleic acid fragment, where each nucleic acid fragment is optionally a fragment of a codon-optimized coding region operably encoding an IV polypeptide, or a fragment, variant, or derivative thereof, as an influenza vaccine for a vertebrate.
  • Methods and Administration
  • The present invention also provides methods for delivering an IV polypeptide or a fragment, variant, or derivative thereof to a human, which comprise administering to a human one or more of the compositions described herein; such that upon administration of compositions such as those described herein, an IV polypeptide or a fragment, variant, or derivative thereof is expressed in human cells, in an amount sufficient to generate an immune response to the IV or administering the IV polypeptide or a fragment, variant, or derivative thereof itself to the human in an amount sufficient to generate an immune response.
  • The present invention further provides methods for delivering an IV polypeptide or a fragment, variant, or derivative thereof to a human, which comprise administering to a vertebrate one or more of the compositions described herein; such that upon administration of compositions such as those described herein, an immune response is generated in the vertebrate.
  • The term “vertebrate” is intended to encompass a singular “vertebrate” as well as plural “vertebrates” and comprises mammals and birds, as well as fish, reptiles, and amphibians.
  • The term “mammal” is intended to encompass a singular “mammal” and plural “mammals,” and includes, but is not limited to humans; primates such as apes, monkeys (e.g., owl, squirrel, cebus, rhesus, African green, patas, cynomolgus, and cercopithecus), orangutans, baboons, gibbons, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equines such as horses, donkeys, and zebras, food animals such as cows, pigs, and sheep; ungulates such as deer and giraffes; ursids such as bears; and others such as rabbits, mice, ferrets, seals, whales. In particular, the mammal can be a human subject, a food animal or a companion animal.
  • The term “bird” is intended to encompass a singular “bird” and plural “birds,” and includes, but is not limited to feral water birds such as ducks, geese, terns, shearwaters, and gulls; as well as domestic avian species such as turkeys, chickens, quail, pheasants, geese, and ducks. The term “bird” also encompasses passerine birds such as starlings and budgerigars.
  • The present invention further provides a method for generating, enhancing or modulating an immune response to an W comprising administering to a vertebrate one or more of the compositions described herein. In this method, the compositions may include one or more isolated polynucleotides comprising at least one nucleic acid fragment where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide, or a fragment, variant, or derivative thereof. In another embodiment, the compositions may include both a polynucleotide as described above, and also an isolated IV polypeptide, or a fragment, variant, or derivative thereof, wherein the protein is provided as a recombinant protein, in particular, a fusion protein, a purified subunit, viral vector expressing the protein, or in the form of an inactivated IV vaccine. Thus, the latter compositions include both a polynucleotide encoding an IV polypeptide or a fragment, variant, or derivative thereof and an isolated IV polypeptide or a fragment, variant, or derivative thereof. The IV polypeptide or a fragment, variant, or derivative thereof encoded by the polynucleotide of the compositions need not be the same as the isolated IV polypeptide or a fragment, variant, or derivative thereof of the compositions. Compositions to be used according to this method may be univalent, bivalent, trivalent or multivalent.
  • The polynucleotides of the compositions may comprise a fragment of a human (or other vertebrate) codon-optimized coding region encoding a protein of the IV, or a fragment, variant, or derivative thereof. The polynucleotides are incorporated into the cells of the vertebrate in vivo, and an antigenic amount of the IV polypeptide, or fragment, variant, or derivative thereof, is produced in vivo. Upon administration of the composition according to this method, the IV polypeptide or a fragment, variant, or derivative thereof is expressed in the vertebrate in an amount sufficient to elicit an immune response. Such an immune response might be used, for example, to generate antibodies to the IV for use in diagnostic assays or as laboratory reagents, or as therapeutic or preventative vaccines as described herein.
  • The present invention further provides a method for generating, enhancing, or modulating a protective and/or therapeutic immune response to IV in a vertebrate, comprising administering to a vertebrate in need of therapeutic and/or preventative immunity one or more of the compositions described herein. In this method, the compositions include one or more polynucleotides comprising at least one nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide, or a fragment, variant, or derivative thereof. In a further embodiment, the composition used in this method includes both an isolated polynucleotide comprising at least one nucleic acid fragment, where the nucleic acid fragment is optionally a fragment of a codon-optimized coding region encoding an IV polypeptide, or a fragment, variant, or derivative thereof; and at least one isolated IV polypeptide, or a fragment, variant, or derivative thereof. Thus, the latter composition includes both an isolated polynucleotide encoding an IV polypeptide or a fragment, variant, or derivative thereof and an isolated IV polypeptide or a fragment, variant, or derivative thereof, for example, a recombinant protein, a purified subunit, viral vector expressing the protein, or an inactivated virus vaccine. Upon administration of the composition according to this method, the IV polypeptide or a fragment, variant, or derivative thereof is expressed in the human in a therapeutically or prophylactically effective amount.
  • As used herein, an “immune response” refers to the ability of a vertebrate to elicit an immune reaction to a composition delivered to that vertebrate. Examples of immune responses include an antibody response or a cellular, e.g., cytotoxic T-cell, response. One or more compositions of the present invention may be used to prevent influenza infection in vertebrates, e.g., as a prophylactic vaccine, to establish or enhance immunity to IV in a healthy individual prior to exposure to influenza or contraction of influenza disease, thus preventing the disease or reducing the severity of disease symptoms.
  • As mentioned above, compositions of the present invention can be used both to prevent IV infection, and also to therapeutically treat IV infection. In individuals already exposed to influenza, or already suffering from influenza disease, the present invention is used to further stimulate the immune system of the vertebrate, thus reducing or eliminating the symptoms associated with that disease or disorder. As defined herein, “treatment ” refers to the use of one or more compositions of the present invention to prevent, cure, retard, or reduce the severity of influenza disease symptoms in a vertebrate, and/or result in no worsening of influenza disease over a specified period of time in a vertebrate which has already been exposed to IV and is thus in need of therapy. The term “prevention” refers to the use of one or more compositions of the present invention to generate immunity in a vertebrate which has not yet been exposed to a particular strain of IV, thereby preventing or reducing disease symptoms if the vertebrate is later exposed to the particular strain of IV. The methods of the present invention therefore may be referred to as therapeutic vaccination or preventative or prophylactic vaccination. It is not required that any composition of the present invention provide total immunity to influenza or totally cure or eliminate all influenza disease symptoms. As used herein, a “vertebrate in need of therapeutic and/or preventative immunity” refers to an individual for whom it is desirable to treat, i.e., to prevent, cure, retard, or reduce the severity of influenza disease symptoms, and/or result in no worsening of influenza disease over a specified period of time. Vertebrates to treat and/or vaccinate include humans, apes, monkeys (e.g., owl, squirrel, cebus, rhesus, African green, patas, cynomolgus, and cercopithecus), orangutans, baboons, gibbons, and chimpanzees, dogs, wolves, cats, lions, and tigers, horses, donkeys, zebras, cows, pigs, sheep, deer, giraffes, bears, rabbits, mice, ferrets, seals, whales, ducks, geese, terns, shearwaters, gulls, turkeys, chickens, quail, pheasants, geese, starlings and budgerigars.
  • One or more compositions of the present invention are utilized in a “prime boost” regimen. An example of a “prime boost” regimen may be found in Yang, Z. et al. J. Virol. 77:799-803 (2002), which is incorporated herein by reference in its entirety. In these embodiments, one or more polynucleotide vaccine compositions of the present invention are delivered to a vertebrate, thereby priming the immune response of the vertebrate to an IV, and then a second immunogenic composition is utilized as a boost vaccination. One or more compositions of the present invention are used to prime immunity, and then a second immunogenic composition, e.g., a recombinant viral vaccine or vaccines, a different polynucleotide vaccine, or one or more purified subunit isolated IV polypeptides or fragments, variants or derivatives thereof is used to boost the anti-IV immune response.
  • In one embodiment, a priming composition and a boosting composition are combined in a single composition or single formulation. For example, a single composition may comprise an isolated IV polypeptide or a fragment, variant, or derivative thereof as the priming component and a polynucleotide encoding an influenza protein as the boosting component. In this embodiment, the compositions may be contained in a single vial where the priming component and boosting component are mixed together. In general, because the peak levels of expression of protein from the polynucleotide does not occur until later (e.g. 7-10 days) after administration, the polynucleotide component may provide a boost to the isolated protein component. Compositions comprising both a priming component and a boosting component are referred to herein as “combinatorial vaccine compositions” or “single formulation heterologous prime-boost vaccine compositions.” In addition, the priming composition may be administered before the boosting composition, or even after the boosting composition, if the boosting composition is expected to take longer to act.
  • In another embodiment, the priming composition may be administered simultaneously with the boosting composition, but in separate formulations where the priming component and the boosting component are separated.
  • The terms “priming” or “primary” and “boost” or “boosting” as used herein may refer to the initial and subsequent immunizations, respectively, i.e., in accordance with the definitions these terms normally have in immunology. However, in certain embodiments, e.g., where the priming component and boosting component are in a single formulation, initial and subsequent immunizations may not be necessary as both the “prime” and the “boost” compositions are administered simultaneously.
  • In certain embodiments, one or more compositions of the present invention are delivered to a vertebrate by methods described herein, thereby achieving an effective therapeutic and/or an effective preventative immune response. More specifically, the compositions of the present invention may be administered to any tissue of a vertebrate, including, but not limited to, muscle, skin, brain tissue, lung tissue, liver tissue, spleen tissue, bone marrow tissue, thymus tissue, heart tissue, e.g., myocardium, endocardium, and pericardium, lymph tissue, blood tissue, bone tissue, pancreas tissue, kidney tissue, gall bladder tissue, stomach tissue, intestinal tissue, testicular tissue, ovarian tissue, uterine tissue, vaginal tissue, rectal tissue, nervous system tissue, eye tissue, glandular tissue, tongue tissue, and connective tissue, e.g., cartilage.
  • Furthermore, the compositions of the present invention may be administered to any internal cavity of a vertebrate, including, but not limited to, the lungs, the mouth, the nasal cavity, the stomach, the peritoneal cavity, the intestine, any heart chamber, veins, arteries, capillaries, lymphatic cavities, the uterine cavity, the vaginal cavity, the rectal cavity, joint cavities, ventricles in brain, spinal canal in spinal cord, the ocular cavities, the lumen of a duct of a salivary gland or a liver. When the compositions of the present invention is administered to the lumen of a duct of a salivary gland or liver, the desired polypeptide is expressed in the salivary gland and the liver such that the polypeptide is delivered into the blood stream of the vertebrate from each of the salivary gland or the liver. Certain modes for administration to secretory organs of a gastrointestinal system using the salivary gland, liver and pancreas to release a desired polypeptide into the bloodstream is disclosed in U.S. Pat. Nos. 5,837,693 and 6,004,944, both of which are incorporated herein by reference in their entireties.
  • In certain embodiments, the compositions are administered into embryonated chicken eggs or by intra-muscular injection into the defeathered breast area of chicks as described in Kodihalli S. et al., Vaccine 18:2592-9 (2000), which is incorporated herein by reference in its entirety.
  • In certain embodiments, the compositions are administered to muscle, either skeletal muscle or cardiac muscle, or to lung tissue. Specific, but non-limiting modes for administration to lung tissue are disclosed in Wheeler, C. J., et al., Proc. Natl. Acad. Sci. USA 93:11454-11459 (1996), which is incorporated herein by reference in its entirety.
  • According to the disclosed methods, compositions of the present invention can be administered by intramuscular (i.m.), subcutaneous (s.c.), or intrapulmonary routes. Other suitable routes of administration include, but are not limited to intratracheal, transdermal, intraocular, intranasal, inhalation, intracavity, intravenous (i.v.), intraductal (e.g., into the pancreas) and intraparenchymal (i.e., into any tissue) administration. Transdermal delivery includes, but not limited to intradermal (e.g., into the dermis or epidermis), transdermal (e.g., percutaneous) and transmucosal administration (i.e., into or through skin or mucosal tissue). Intracavity administration includes, but not limited to administration into oral, vaginal, rectal, nasal, peritoneal, or intestinal cavities as well as, intrathecal (i.e., into spinal canal), intraventricular (i.e., into the brain ventricles or the heart ventricles), inraatrial (i.e., into the heart atrium) and sub arachnoid (i.e., into the sub arachnoid spaces of the brain) administration.
  • Any mode of administration can be used so long as the mode results in the expression of the desired peptide or protein, in the desired tissue, in an amount sufficient to generate an immune response to IV and/or to generate a prophylactically or therapeutically effective immune response to IV in a human in need of such response. Administration means of the present invention include needle injection, catheter infusion, biolistic injectors, particle accelerators (e.g., “gene guns” or pneumatic “needleless” injectors) Med-E-Jet (Vahlsing, H., et al., J. Immunol. Methods 171:11-22 (1994)), Pigjet (Schrijver, R., et al., Vaccine 15: 1908-1916 (1997)), Biojector (Davis, H., et al., Vaccine 12: 1503-1509 (1994); Gramzinski, R., et al., Mol. Med. 4: 109-118 (1998)), AdvantaJet (Linmayer, I., et al., Diabetes Care 9:294-297 (1986)), Medi-jector (Martins, J., and Roedl, E. J. Occup. Med. 21:821-824 (1979)), gelfoam sponge depots, other commercially available depot materials (e.g., hydrogels), osmotic pumps (e.g., Alza minipumps), oral or suppositorial solid (tablet or pill) pharmaceutical formulations, topical skin creams, and decanting, use of polynucleotide coated suture (Qin, Y., et al., Life Sciences 65: 2193-2203 (1999)) or topical applications during surgery. Certain modes of administration are intramuscular needle-based injection and pulmonary application via catheter infusion. Energy-assisted plasmid delivery (EAPD) methods may also be employed to administer the compositions of the invention. One such method involves the application of brief electrical pulses to injected tissues, a procedure commonly known as electroporation. See generally Mir, L. M. et al., Proc. Natl. Acad. Sci USA 96:4262-7 (1999); Hartikka, J. et al., Mol. Ther. 4:407-15 (2001); Mathiesen, I., Gene Ther. 6:508-14(1999); Rizzuto G. et al., Hum. Gen. Ther. 11:1891-900 (2000). Each of the references cited in this paragraph is incorporated herein by reference in its entirety.
  • Determining an effective amount of one or more compositions of the present invention depends upon a number of factors including, for example, the antigen being expressed or administered directly, e.g., HA, NA, NP, M1 or M2, or fragments, e.g., eM2, variants, or derivatives thereof, the age and weight of the subject, the precise condition requiring treatment and its severity, and the route of administration. Based on the above factors, determining the precise amount, number of doses, and timing of doses are within the ordinary skill in the art and will be readily determined by the attending physician or veterinarian.
  • Compositions of the present invention may include various salts, excipients, delivery vehicles and/or auxiliary agents as are disclosed, e.g., in U.S. patent application Publication No. 2002/0019358, published Feb. 14, 2002, which is incorporated herein by reference in its entirety.
  • Furthermore, compositions of the present invention may include one or more transfection facilitating compounds that facilitate delivery of polynucleotides to the interior of a cell, and/or to a desired location within a cell. As used herein, the terms “transfection facilitating compound,” “transfection facilitating agent,” and “transfection facilitating material” are synonymous, and may be used interchangeably. It should be noted that certain transfection facilitating compounds may also be “adjuvants” as described infra, i.e., in addition to facilitating delivery of polynucleotides to the interior of a cell, the compound acts to alter or increase the immune response to the antigen encoded by that polynucleotide. Examples of the transfection facilitating compounds include, but are not limited to inorganic materials such as calcium phosphate, alum (aluminum sulfate), and gold particles (e.g., “powder” type delivery vehicles); peptides that are, for example, cationic, intercell targeting (for selective delivery to certain cell types), intracell targeting (for nuclear localization or endosomal escape), and ampipathic (helix forming or pore forming); proteins that are, for example, basic (e.g., positively charged) such as histones, targeting (e.g., asialoprotein), viral (e.g., Sendai virus coat protein), and pore-forming; lipids that are, for example, cationic (e.g., DMRIE, DOSPA, DC-Chol), basic (e.g., steryl amine), neutral (e.g., cholesterol), anionic (e.g., phosphatidyl serine), and zwitterionic (e.g., DOPE, DOPC); and polymers such as dendrimers, star-polymers, “homogenous” poly-amino acids (e.g., poly-lysine, poly-arginine), “heterogeneous” poly-amino acids (e.g., mixtures of lysine & glycine), co-polymers, polyvinylpyrrolidinone (PVP), poloxamers (e.g. CRL 1005) and polyethylene glycol (PEG). A transfection facilitating material can be used alone or in combination with one or more other transfection facilitating materials. Two or more transfection facilitating materials can be combined by chemical bonding (e.g., covalent and ionic such as in lipidated polylysine, PEGylated polylysine) (Toncheva, et al., Biochim. Biophys. Acta 1380(3):354-368 (1988)), mechanical mixing (e.g., free moving materials in liquid or solid phase such as “polylysine+cationic lipids”) (Gao and Huang, Biochemistry 35:1027-1036 (1996); Trubetskoy, et al., Biochem. Biophys. Acta 1131:311-313 (1992)), and aggregation (e.g., co-precipitation, gel forming such as in cationic lipids+poly-lactide, and polylysine+gelatin). Each of the references cited in this paragraph is incorporated herein by reference in its entirety.
  • One category of transfection facilitating materials is cationic lipids. Examples of cationic lipids are 5-carboxyspermylglycine dioctadecylamide (DOGS) and dipalmitoyl-phophatidylethanolamine-5-carboxyspermylamide (DPPES). Cationic cholesterol derivatives are also useful, including {3β-[N-N′,N′-dimethylamino)ethane]-carbomoyl}-cholesterol (DC-Chol). Dimethyldioctdecyl-ammonium bromide (DDAB), N-(3-aminopropyl)-N,N-(bis-(2-tetradecyloxyethyl))-N-methyl-ammonium bromide (PA-DEMO), N-(3-aminopropyl)-N,N-(bis-(2-dodecyloxyethyl))-N-methyl-ammonium bromide (PA-DELO), N,N,N-tris-(2-dodecyloxy)ethyl-N-(3-amino)propyl-ammonium bromide (PA-TELO), and N1-(3-aminopropyl)((2-dodecyloxy)ethyl)-N2-(2-dodecyloxy)ethyl-1-piperazinaminium bromide (GA-LOE-BP) can also be employed in the present invention.
  • Non-diether cationic lipids, such as DL-1,2-dioleoyl-3-dimethylaminopropyl-β-hydroxyethylammonium (DORI diester), 1-O-oleyl-2-oleoyl-3-dimethylaminopropyl-p-hydroxyethylammonium (DORI ester/ether), and their salts promote in vivo gene delivery. In some embodiments, cationic lipids comprise groups attached via a heteroatom attached to the quaternary ammonium moiety in the head group. A glycyl spacer can connect the linker to the hydroxyl group.
  • Specific, but non-limiting cationic lipids for use in certain embodiments of the present invention include DMRIE ((±)-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide), GAP-DMORIE ((±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-bis(syn-9-tetradeceneyloxy)-1-propanaminium bromide), and GAP-DMRIE ((±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-(bis-dodecyloxy)-1-propanaminium bromide).
  • Other specific but non-limiting cationic surfactants for use in certain embodiments of the present invention include Bn-DHRIE, DhxRIE, DhxRIE-OAc, DhxRIE-OBz and Pr-DOctRIE-OAc. These lipids are disclosed in copending U.S. patent application Ser. No. 10/725,015. In another aspect of the present invention, the cationic surfactant is Pr-DOctRIE-OAc.
  • Other cationic lipids include (±)-N,N-dimethyl-N-[2-(sperminecarboxamido)ethyl]-2,3-bis(dioleyloxy)-1-propaniminium pentahydrochloride (DOSPA), (±)-N-(2-aminoethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propaniminium bromide (β-aminoethyl-DMRIE or βAE-DMRIE) (Wheeler, et al., Biochim. Biophys. Acta 1280:1-11 (1996), and (±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-bis(dodecyloxy)-1-propaniminium bromide (GAP-DLRIE) (Wheeler, et al., Proc. Natl. Acad. Sci. USA 93:11454-11459 (1996)), which have been developed from DMRIE. Both of the references cited in this paragraph are incorporated herein by reference in their entirety.
  • Other examples of DMRIE-derived cationic lipids that are useful for the present invention are (±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-(bis-decyloxy)-1-propanaminium bromide (GAP-DDRIE), (±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-(bis-tetradecyloxy)-1-propanaminium bromide (GAP-DMRIE), (±)-N-((N″-methyl)-N′-ureyl)propyl-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (GMU-DMRIE), (±)-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(dodecyloxy)-1-propanaminium bromide (DLRIE), and (±)-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis-([Z]-9-octadecenyloxy)propyl-1-propaniminium bromide (HP-DORIE).
  • In the embodiments where the immunogenic composition comprises a cationic lipid, the cationic lipid may be mixed with one or more co-lipids. For purposes of definition, the term “co-lipid” refers to any hydrophobic material which may be combined with the cationic lipid component and includes amphipathic lipids, such as phospholipids, and neutral lipids, such as cholesterol. Cationic lipids and co-lipids may be mixed or combined in a number of ways to produce a variety of non-covalently bonded macroscopic structures, including, for example, liposomes, multilamellar vesicles, unilamellar vesicles, micelles, and simple films. One non-limiting class of co-lipids are the zwitterionic phospholipids, which include the phosphatidylethanolamines and the phosphatidylcholines. Examples of phosphatidylethanolamines, include DOPE, DMPE and DPyPE. In certain embodiments, the co-lipid is DPyPE, which comprises two phytanoyl substituents incorporated into the diacylphosphatidylethanolamine skeleton. In other embodiments, the co-lipid is DOPE, CAS name 1,2-diolyeoyl-sn-glycero-3-phosphoethanolamine.
  • When a composition of the present invention comprises a cationic lipid and co-lipid, the cationic lipid:co-lipid molar ratio may be from about 9:1 to about 1:9, from about 4:1 to about 1:4, from about 2:1 to about 1:2, or about 1:1.
  • In order to maximize homogeneity, the cationic lipid and co-lipid components may be dissolved in a solvent such as chloroform, followed by evaporation of the cationic lipid/co-lipid solution under vacuum to dryness as a film on the inner surface of a glass vessel (e.g., a Rotovap round-bottomed flask). Upon suspension in an aqueous solvent, the amphipathic lipid component molecules self-assemble into homogenous lipid vesicles. These lipid vesicles may subsequently be processed to have a selected mean diameter of uniform size prior to complexing with, for example, a codon-optimized polynucleotide of the present invention, according to methods known to those skilled in the art. For example, the sonication of a lipid solution is described in Felgner et al., Proc. Natl. Acad. Sci. USA 8:,7413-7417 (1987) and in U.S. Pat. No. 5,264,618, the disclosures of which are incorporated herein by reference.
  • In those embodiments where the composition includes a cationic lipid, polynucleotides of the present invention are complexed with lipids by mixing, for example, a plasmid in aqueous solution and a solution of cationic lipid:co-lipid as prepared herein are mixed. The concentration of each of the constituent solutions can be adjusted prior to mixing such that the desired final plasmid/cationic lipid:co-lipid ratio and the desired plasmid final concentration will be obtained upon mixing the two solutions. The cationic lipid:co-lipid mixtures are suitably prepared by hydrating a thin film of the mixed lipid materials in an appropriate volume of aqueous solvent by vortex mixing at ambient temperatures for about 1 minute. The thin films are prepared by admixing chloroform solutions of the individual components to afford a desired molar solute ratio followed by aliquoting the desired volume of the solutions into a suitable container. The solvent is removed by evaporation, first with a stream of dry, inert gas (e.g. argon) followed by high vacuum treatment.
  • Other hydrophobic and amphiphilic additives, such as, for example, sterols, fatty acids, gangliosides, glycolipids, lipopeptides, liposaccharides, neobees, niosomes, prostaglandins and sphingolipids, may also be included in compositions of the present invention. In such compositions, these additives may be included in an amount between about 0.1 mol % and about 99.9 mol % (relative to total lipid), about 1-50 mol %, or about 2-25 mol %.
  • Additional embodiments of the present invention are drawn to compositions comprising an auxiliary agent which is administered before, after, or concurrently with the polynucleotide. As used herein, an “auxiliary agent” is a substance included in a composition for its ability to enhance, relative to a composition which is identical except for the inclusion of the auxiliary agent, the entry of polynucleotides into vertebrate cells in vivo, and/or the in vivo expression of polypeptides encoded by such polynucleotides. Certain auxiliary agents may, in addition to enhancing entry of polynucleotides into cells, enhance an immune response to an immunogen encoded by the polynucleotide. Auxiliary agents of the present invention include nonionic, anionic, cationic, or zwitterionic surfactants or detergents, with nonionic surfactants or detergents being preferred, chelators, DNase inhibitors, poloxamers, agents that aggregate or condense nucleic acids, emulsifying or solubilizing agents, wetting agents, gel-forming agents, and buffers.
  • Auxiliary agents for use in compositions of the present invention include, but are not limited to non-ionic detergents and surfactants IGEPAL CA 6300, NONIDET NP-40, Nonidet® P40, Tween-20™, Tween-80™, Pluronic® F68 (ave. MW: 8400; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 80%), Pluronic F77® (ave. MW: 6600; approx. MW of hydrophobe, 2100; approx. wt. % of hydrophile, 70%), Pluronic P65®(ave. MW: 3400; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 50%), Triton X-100™, and Triton X-114™; the anionic detergent sodium dodecyl sulfate (SDS); the sugar stachyose; the condensing agent DMSO; and the chelator/DNAse inhibitor EDTA, CRL 1005 (12 kDa, 5% POE), and BAK (Benzalkonium chloride 50% solution, available from Ruger Chemical Co. Inc.). In certain specific embodiments, the auxiliary agent is DMSO, Nonidet P40, Pluronic F68® (ave. MW: 8400; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 80%), Pluronic F77® (ave. MW: 6600; approx. MW of hydrophobe, 2100; approx. wt. % of hydrophile, 70%), Pluronic P65® (ave. MW: 3400; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 50%), Pluronic L64® (ave. MW: 2900; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 40%), and Pluronic F108® (ave. MW: 14600; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 80%). See, e.g., U.S. patent application Publication No. 2002/0019358, published Feb. 14, 2002, which is incorporated herein by reference in its entirety.
  • Certain compositions of the present invention can further include one or more adjuvants before, after, or concurrently with the polynucleotide. The term “adjuvant” refers to any material having the ability to (1) alter or increase the immune response to a particular antigen or (2) increase or aid an effect of a pharmacological agent. It should be noted, with respect to polynucleotide vaccines, that an “adjuvant,” can be a transfection facilitating material. Similarly, certain “transfection facilitating materials” described supra, may also be an “adjuvant.” An adjuvant may be used with a composition comprising a polynucleotide of the present invention. In a prime-boost regimen, as described herein, an adjuvant may be used with either the priming immunization, the booster immunization, or both. Suitable adjuvants include, but are not limited to, cytokines and growth factors; bacterial components (e.g., endotoxins, in particular superantigens, exotoxins and cell wall components); aluminum-based salts; calcium-based salts; silica; polynucleotides; toxoids; serum proteins, viruses and virally-derived materials, poisons, venoms, imidazoquiniline compounds, poloxamers, and cationic lipids.
  • A great variety of materials have been shown to have adjuvant activity through a variety of mechanisms. Any compound which may increase the expression, antigenicity or immunogenicity of the polypeptide is a potential adjuvant. The present invention provides an assay to screen for improved immune responses to potential adjuvants. Potential adjuvants which may be screened for their ability to enhance the immune response according to the present invention include, but are not limited to: inert carriers, such as alum, bentonite, latex, and acrylic particles; pluronic block polymers, such as TiterMax® (block copolymer CRL-8941, squalene (a metabolizable oil) and a microparticulate silica stabilizer); depot formers, such as Freunds adjuvant, surface active materials, such as saponin, lysolecithin, retinal, Quil A, liposomes, and pluronic polymer formulations; macrophage stimulators, such as bacterial lipopolysaccharide; alternate pathway complement activators, such as insulin, zymosan, endotoxin, and levamisole; and non-ionic surfactants, such as poloxamers, poly(oxyethylene)-poly(oxypropylene) tri-block copolymers. Also included as adjuvants are transfection-facilitating materials, such as those described above.
  • Poloxamers which may be screened for their ability to enhance the immune response according to the present invention include, but are not limited to, commercially available poloxamers such as Pluronic® surfactants, which are block copolymers of propylene oxide and ethylene oxide in which the propylene oxide block is sandwiched between two ethylene oxide blocks. Examples of Pluronic® surfactants include Pluronic® L121 (ave. MW: 4400; approx. MW of hydrophobe, 3600; approx. wt. % of hydrophile, 10%), Pluronic® L101 (ave. MW: 3800; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 10%), Pluronic® L81 (ave. MW: 2750; approx. MW of hydrophobe, 2400; approx. wt. % of hydrophile, 10%), Pluronic® L61 (ave. MW: 2000; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 10%), Pluronic® L31 (ave. MW: 1100; approx. MW of hydrophobe, 900; approx. wt. % of hydrophile, 10%), Pluronic® L122 (ave. MW: 5000; approx. MW of hydrophobe, 3600; approx. wt. % of hydrophile, 20%), Pluronic® L92 (ave. MW: 3650; approx. MW of hydrophobe, 2700; approx. wt. % of hydrophile, 20%), Pluronic® L72 (ave. MW: 2750; approx. MW of hydrophobe, 2100; approx. wt. % of hydrophile, 20%), Pluronic® L62 (ave. MW: 2500; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 20%), Pluronic® L42 (ave. MW: 1630; approx. MW of hydrophobe, 1200; approx. wt. % of hydrophile, 20%), Pluronic® L63 (ave. MW: 2650; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 30%), Pluronic® L43 (ave. MW: 1850; approx. MW of hydrophobe, 1200; approx. wt. % of hydrophile, 30%), Pluronic® L64 (ave. MW: 2900; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 40%), Pluronic® L44 (ave. MW: 2200; approx. MW of hydrophobe, 1200; approx. wt. % of hydrophile, 40%), Pluronic® L35 (ave. MW: 1900; approx. MW of hydrophobe, 900; approx. wt. % of hydrophile, 50%), Pluronic® P123 (ave. MW: 5750; approx. MW of hydrophobe, 3600; approx. wt. % of hydrophile, 30%), Pluronic® P103 (ave. MW: 4950; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 30%), Pluronic® P104 (ave. MW: 5900; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 40%), Pluronic® P84 (ave. MW: 4200; approx. MW of hydrophobe, 2400; approx. wt. % of hydrophile, 40%), Pluronic® P105 (ave. MW: 6500; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 50%), Pluronic® P85 (ave. MW: 4600; approx. MW of hydrophobe, 2400; approx. wt. % of hydrophile, 50%), Pluronic® P75 (ave. MW: 4150; approx. MW of hydrophobe, 2100; approx. wt. % of hydrophile, 50%), Pluronic® P65 (ave. MW: 3400; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 50%), Pluronic® F127 (ave. MW: 12600; approx. MW of hydrophobe, 3600; approx. wt. % of hydrophile, 70%), Pluronic® F98 (ave. MW: 13000; approx. MW of hydrophobe, 2700; approx. wt. % of hydrophile, 80%), Pluronic® F87 (ave. MW: 7700; approx. MW of hydrophobe, 2400; approx. wt. % of hydrophile, 70%), Pluronic® F77 (ave. MW: 6600; approx. MW of hydrophobe, 2100; approx. wt. % of hydrophile, 70%), Pluronic® F108 (ave. MW: 14600; approx. MW of hydrophobe, 3000; approx. wt. % of hydrophile, 80%), Pluronic® F98 (ave. MW: 13000; approx. MW of hydrophobe, 2700; approx. wt. % of hydrophile, 80%), Pluronic® F88 (ave. MW: 11400; approx. MW of hydrophobe, 2400; approx. wt. % of hydrophile, 80%), Pluronic® F68 (ave. MW: 8400; approx. MW of hydrophobe, 1800; approx. wt. % of hydrophile, 80%), Pluronic® F38 (ave. MW: 4700; approx. MW of hydrophobe, 900; approx. wt. % of hydrophile, 80%).
  • Reverse poloxamers which may be screened for their ability to enhance the immune response according to the present invention include, but are not limited to Pluronic® R 31R1 (ave. MW: 3250; approx. MW of hydrophobe, 3100; approx. wt. % of hydrophile, 10%), Pluronic® R 25R1 (ave. MW: 2700; approx. MW of hydrophobe, 2500; approx. wt. % of hydrophile, 10%), Pluronic® R 17R1 (ave. MW: 1900; approx. MW of hydrophobe, 1700; approx. wt. % of hydrophile, 10%), Pluronic® R 31R2 (ave. MW: 3300; approx. MW of hydrophobe, 3100; approx. wt. % of hydrophile, 20%), Pluronic® R 25R2 (ave. MW: 3100; approx. MW of hydrophobe, 2500; approx. wt. % of hydrophile, 20%), Pluronic® R 17R2 (ave. MW: 2150; approx. MW of hydrophobe, 1700; approx. wt. % of hydrophile, 20%), Pluronic® R 12R3 (ave. MW: 1800; approx. MW of hydrophobe, 1200; approx. wt. % of hydrophile, 30%), Pluronic® R 31R4 (ave. MW: 4150; approx. MW of hydrophobe, 3100; approx. wt. % of hydrophile, 40%), Pluronic® R 25R4 (ave. MW: 3600; approx. MW of hydrophobe, 2500; approx. wt. % of hydrophile, 40%), Pluronic® R 22R4 (ave. MW: 3350; approx. MW of hydrophobe, 2200; approx. wt. % of hydrophile, 40%), Pluronic® R 17R4 (ave. MW: 3650; approx. MW of hydrophobe, 1700; approx. wt. % of hydrophile, 40%), Pluronic® R 25R5 (ave. MW: 4320; approx. MW of hydrophobe, 2500; approx. wt. % of hydrophile, 50%), Pluronic® R 10R5 (ave. MW: 1950; approx. MW of hydrophobe, 1000; approx. wt. % of hydrophile, 50%), Pluronic® R 25R8 (ave. MW: 8550; approx. MW of hydrophobe, 2500; approx. wt. % of hydrophile, 80%), Pluronic® R 17R8 (ave. MW: 7000; approx. MW of hydrophobe, 1700; approx. wt. % of hydrophile, 80%), and Pluronic® R 10R8 (ave. MW: 4550; approx. MW of hydrophobe, 1000; approx. wt. % of hydrophile, 80%).
  • Other commercially available poloxamers which may be screened for their ability to enhance the immune response according to the present invention include compounds that are block copolymer of polyethylene and polypropylene glycol such as Synperonic® L121 (ave. MW: 4400), Synperonic® L122 (ave. MW: 5000), Synperonic® P104 (ave. MW: 5850), Synperonic® P105 (ave. MW: 6500), Synperonic® P123 (ave. MW: 5750), Synperonic® P85 (ave. MW: 4600) and Synperonic® P94 (ave. MW: 4600), in which L indicates that the surfactants are liquids, P that they are pastes, the first digit is a measure of the molecular weight of the polypropylene portion of the surfactant and the last digit of the number, multiplied by 10, gives the percent ethylene oxide content of the surfactant; and compounds that are nonylphenyl polyethylene glycol such as Synperonic® NP10 (nonylphenol ethoxylated surfactant—10% solution), Synperonic® NP30 (condensate of 1 mole of nonylphenol with 30 moles of ethylene oxide) and Synperonic® NP5 (condensate of 1 mole of nonylphenol with 5.5 moles of naphthalene oxide).
  • Other poloxamers which may be screened for their ability to enhance the immune response according to the present invention include: (a) a polyether block copolymer comprising an A-type segment and a B-type segment, wherein the A-type segment comprises a linear polymeric segment of relatively hydrophilic character, the repeating units of which contribute an average Hansch-Leo fragmental constant of about −0.4 or less and have molecular weight contributions between about 30 and about 500, wherein the B-type segment comprises a linear polymeric segment of relatively hydrophobic character, the repeating units of which contribute an average Hansch-Leo fragmental constant of about −0.4 or more and have molecular weight contributions between about 30 and about 500, wherein at least about 80% of the linkages joining the repeating units for each of the polymeric segments comprise an ether linkage; (b) a block copolymer having a polyether segment and a polycation segment, wherein the polyether segment comprises at least an A-type block, and the polycation segment comprises a plurality of cationic repeating units; and (c) a polyether-polycation copolymer comprising a polymer, a polyether segment and a polycationic segment comprising a plurality of cationic repeating units of formula —NH—R0, wherein R0 is a straight chain aliphatic group of 2 to 6 carbon atoms, which may be substituted, wherein said polyether segments comprise at least one of an A-type of B-type segment. See U.S. Pat. No. 5,656,611, by Kabonov, et al., which is incorporated herein by reference in its entirety. Other poloxamers of interest include CRL1005 (12 kDa, 5% POE), CRL8300 (11 kDa, 5% POE), CRL2690 (12 kDa, 10% POE), CRL4505 (15 kDa, 5% POE) and CRL1415 (9 kDa, 10% POE).
  • Other auxiliary agents which may be screened for their ability to enhance the immune response according to the present invention include, but are not limited to Acacia (gum arabic); the poloxyethylene ether R—O—(C2H4O)x—H (BRIJ®), e.g., polyethylene glycol dodecyl ether (BRIJ® 35, x=23), polyethylene glycol dodecyl ether (BRIJ® 30, x=4), polyethylene glycol hexadecyl ether (BRIJ® 52 x=2), polyethylene glycol hexadecyl ether (BRIJ® 56, x=10), polyethylene glycol hexadecyl ether (BRIJ® 58P, x=20), polyethylene glycol octadecyl ether (BRIJ® 72, x=2), polyethylene glycol octadecyl ether (BRIJ® 76, x=10), polyethylene glycol octadecyl ether (BRIJ® 78P, x=20), polyethylene glycol oleyl ether (BRIJ® 92V, x=2), and polyoxyl 10 oleyl ether (BRIJ® 97, x=10); poly-D-glucosamine (chitosan); chlorbutanol; cholesterol; diethanolamine; digitonin; dimethylsulfoxide (DMSO), ethylenediamine tetraacetic acid (EDTA); glyceryl monosterate; lanolin alcohols; mono- and di-glycerides; monoethanolamine; nonylphenol polyoxyethylene ether (NP-40®); octylphenoxypolyethoxyethanol (NONIDET NP-40 from Amresco); ethyl phenol poly (ethylene glycol ether)n, n=11 (Nonidet® P40 from Roche); octyl phenol ethylene oxide condensate with about 9 ethylene oxide units (nonidet P40); IGEPAL CA 630® ((octyl phenoxy) polyethoxyethanol; structurally same as NONIDET NP-40); oleic acid; oleyl alcohol; polyethylene glycol 8000; polyoxyl 20 cetostearyl ether; polyoxyl 35 castor oil; polyoxyl 40 hydrogenated castor oil; polyoxyl 40 stearate; polyoxyethylene sorbitan monolaurate (polysorbate 20, or TWEEN-20®; polyoxyethylene sorbitan monooleate (polysorbate 80, or TWEEN-80®); propylene glycol diacetate; propylene glycol monstearate; protamine sulfate; proteolytic enzymes; sodium dodecyl sulfate (SDS); sodium monolaurate; sodium stearate; sorbitan derivatives (SPAN®), e.g., sorbitan monopalmitate (SPAN® 40), sorbitan monostearate (SPAN® 60), sorbitan tristearate (SPAN® 65), sorbitan monooleate (SPAN® 80), and sorbitan trioleate (SPAN® 85); 2,6,10,15,19,23-hexamethyl-2,6,10,14,18,22-tetracosa-hexaene (squalene); stachyose; stearic acid; sucrose; surfactin (lipopeptide antibiotic from Bacillus subtilis); dodecylpoly(ethyleneglycolether)9 (Thesit®) MW 582.9; octyl phenol ethylene oxide condensate with about 9-10 ethylene oxide units (Triton X-100™); octyl phenol ethylene oxide condensate with about 7-8 ethylene oxide units (Triton X-114™); tris(2-hydroxyethyl)amine (trolamine); and emulsifying wax.
  • In certain adjuvant compostions, the adjuvant is a cytokine. A composition of the present invention can comprise one or more cytokines, chemokines, or compounds that induce the production of cytokines and chemokines, or a polynucleotide encoding one or more cytokines, chemokines, or compounds that induce the production of cytokines and chemokines. Examples include, but are not limited to granulocyte macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), macrophage colony stimulating factor (M-CSF), colony stimulating factor (CSF), erythropoietin (EPO), interleukin 2 (IL-2), interleukin-3 (IL-3), interleukin 4 (IL-4), interleukin 5 (IL-5), interleukin 6 (IL-6), interleukin 7 (IL-7), interleukin 8 (IL-8), interleukin 10 (IL-10), interleukin 12 (IL-12), interleukin 15 (IL-15), interleukin 18 (IL-18), interferon alpha (IFNα), interferon beta (IFNβ), interferon gamma (IFNγ), interferon omega (IFNω), interferon tau (IFNτ), interferon gamma inducing factor I (IGIF), transforming growth factor beta (TGF-β), RANTES (regulated upon activation, normal T-cell expressed and presumably secreted), macrophage inflammatory proteins (e.g., MIP-1 alpha and MIP-1 beta), Leishmania elongation initiating factor (LEIF), and Flt-3 ligand.
  • In certain compositions of the present invention, the polynucleotide construct may be complexed with an adjuvant composition comprising (±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-bis(syn-9-tetradeceneyloxy)-1-propanaminium bromide (GAP-DMORIE). The composition may also comprise one or more co-lipids, e.g., 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE), and/or 1,2-dimyristoyl-glycer-3-phosphoethanolamine (DMPE). An adjuvant composition comprising GAP-DMORIE and DPyPE at a 1:1 molar ratio is referred to herein as Vaxfectin™. See, e.g., PCT Publication No. WO 00/57917, which is incorporated herein by reference in its entirety.
  • In other embodiments, the polynucleotide itself may function as an adjuvant as is the case when the polynucleotides of the invention are derived, in whole or in part, from bacterial DNA. Bacterial DNA containing motifs of unmethylated CpG-dinucleotides (CpG-DNA) triggers innate immune cells in vertebrates through a pattern recognition receptor (including toll receptors such as TLR 9) and thus possesses potent immunostimulatory effects on macrophages, dendritic cells and B-lymphocytes. See, e.g., Wagner, H., Curr. Opin. Microbiol. 5:62-69 (2002); Jung, J. et al., J. Immunol. 169: 2368-73 (2002); see also Klinman, D. M. et al., Proc. Natl Acad. Sci. U.S.A. 93:2879-83 (1996). Methods of using unmethylated CpG-dinucleotides as adjuvants are described in, for example, U.S. Pat. Nos. 6,207,646, 6,406,705 and 6,429,199, the disclosures of which are herein incorporated by reference.
  • The ability of an adjuvant to increase the immune response to an antigen is typically manifested by a significant increase in immune-mediated protection. For example, an increase in humoral immunity is typically manifested by a significant increase in the titer of antibodies raised to the antigen, and an increase in T-cell activity is typically manifested in increased cell proliferation, or cellular cytotoxicity, or cytokine secretion. An adjuvant may also alter an immune response, for example, by changing a primarily humoral or Th2 response into a primarily cellular, or Th1 response.
  • Nucleic acid molecules and/or polynucleotides of the present invention, e.g., plasmid DNA, mRNA, linear DNA or oligonucleotides, may be solubilized in any of various buffers. Suitable buffers include, for example, phosphate buffered saline (PBS), normal saline, Tris buffer, and sodium phosphate (e.g., 150 mM sodium phosphate). Insoluble polynucleotides may be solubilized in a weak acid or weak base, and then diluted to the desired volume with a buffer. The pH of the buffer may be adjusted as appropriate. In addition, a pharmaceutically acceptable additive can be used to provide an appropriate osmolarity. Such additives are within the purview of one skilled in the art. For aqueous compositions used in vivo, sterile pyrogen-free water can be used. Such formulations will contain an effective amount of a polynucleotide together with a suitable amount of an aqueous solution in order to prepare pharmaceutically acceptable compositions suitable for administration to a human.
  • Compositions of the present invention can be formulated according to known methods. Suitable preparation methods are described, for example, in Remington's Pharmaceutical Sciences, 16th Edition, A. Osol, ed., Mack Publishing Co., Easton, Pa. (1980), and Remington's Pharmaceutical Sciences, 19th Edition, A. R. Gennaro, ed., Mack Publishing Co., Easton, Pa. (1995), both of which are incorporated herein by reference in their entireties. Although the composition may be administered as an aqueous solution, it can also be formulated as an emulsion, gel, solution, suspension, lyophilized form, or any other form known in the art. In addition, the composition may contain pharmaceutically acceptable additives including, for example, diluents, binders, stabilizers, and preservatives.
  • The following examples are included for purposes of illustration only and are not intended to limit the scope of the present invention, which is defined by the appended claims. All references cited in the Examples are incorporated herein by reference in their entireties.
  • EXAMPLES
  • Materials and Methods
  • The following materials and methods apply generally to all the examples disclosed herein. Specific materials and methods are disclosed in each example, as necessary.
  • The practice of the present invention will employ, unless otherwise indicated, conventional techniques of cell biology, cell culture, molecular biology (including PCR), vaccinology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, for example, Molecular Cloning A Laboratory Manual, 2nd Ed., Sambrook et al., ed., Cold Spring Harbor Laboratory Press: (1989); DNA Cloning, Volumes I and II (D. N. Glover ed., 1985); Oligonucleotide Synthesis (M. J. Gait ed., 1984); Mullis et al. U.S. Pat. No: 4,683,195; Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. 1984); Transcription And Translation (B. D. Hames & S. J. Higgins eds. 1984); Culture Of Animal Cells (R. I. Freshney, Alan R. Liss, Inc., 1987); Immobilized Cells And Enzymes (IRL Press, 1986); B. Perbal, A Practical Guide To Molecular Cloning (1984); the treatise, Methods In Enzymology (Academic Press, Inc., N.Y.); Gene Transfer Vectors For Mammalian Cells (J. H. Miller and M. P. Calos eds., 1987, Cold Spring Harbor Laboratory); Methods In Enzymology, Vols. 154 and 155 (Wu et al. eds.), Immunochemical Methods In Cell And Molecular Biology (Mayer and Walker, eds., Academic Press, London, 1987); and in Ausubel et al., Current Protocols in Molecular Biology, John Wiley and Sons, Baltimore, Md. (1989). Each of the references cited in this paragraph is incorporated herein by reference in its entirety.
  • Gene Construction
  • Constructs of the present invention are constructed based on the sequence information provided herein or in the art utilizing standard molecular biology techniques, including, but not limited to the following. First, a series complementary oligonucleotide pairs of 80-90 nucleotides each in length and spanning the length of the construct are synthesized by standard methods. These oligonucleotide pairs are synthesized such that upon annealing, they form double stranded fragments of 80-90 base pairs, containing cohesive ends. The single-stranded ends of each pair of oligonucleotides are designed to anneal with a single-stranded end of an adjacent oligonucleotide duplex. Several adjacent oligonucleotide pairs prepared in this manner are allowed to anneal, and approximately five to six adjacent oligonucleotide duplex fragments are then allowed to anneal together via the cohesive single stranded ends. This series of annealed oligonucleotide duplex fragments is then ligated together and cloned into a suitable plasmid, such as the TOPO® vector available from Invitrogen Corporation, Carlsbad, Calif. The construct is then sequenced by standard methods. Constructs prepared in this manner, comprising 5 to 6 adjacent 80 to 90 base pair fragments ligated together, i.e., fragments of about 500 base pairs, are prepared, such that the entire desired sequence of the construct is represented in a series of plasmid constructs. The inserts of these plasmids are then cut with appropriate restriction enzymes and ligated together to form the final construct. The final construct is then cloned into a standard bacterial cloning vector, and sequenced. The oligonucleotides and primers referred to herein can easily be designed by a person of skill in the art based on the sequence information provided herein and in the art, and such can be synthesized by any of a number of commercial nucleotide providers, for example Retrogen, San Diego, Calif., and GENEART, Regensburg, Germany.
  • Plasmid Vectors
  • Constructs of the present invention can be inserted, for example, into eukaryotic expression vectors VR1012 or VR10551. These vectors are built on a modified pUC18 background (see Yanisch-Perron, C., et al. Gene 33:103-119 (1985)), and contain a kanamycin resistance gene, the human cytomegalovirus immediate early promoter/enhancer and intron A, and the bovine growth hormone transcription termination signal, and a polylinker for inserting foreign genes. See Hartikka, J., et al., Hum. Gene Ther. 7:1205-1217 (1996). However, other standard commercially available eukaryotic expression vectors may be used in the present invention, including, but not limited to: plasmids pcDNA3, pHCMV/Zeo, pCR3.1, pEF1/His, pIND/GS, pRc/HCMV2, pSV40/Zeo2, pTRACER-HCMV, pUB6/V5-His, pVAX1, and pZeoSV2 (available from Invitrogen, San Diego, Calif.), and plasmid pCI (available from Promega, Madison, Wis.).
  • An optimized backbone plasmid, termed VR10551, has minor changes from the VR1012 backbone described above. The VR10551 vector is derived from and similar to VR1012 in that it uses the human cytomegalovirus immediate early (hCMV-IE) gene enhancer/promoter and 5′ untranslated region (UTR), including the hCMV-IE Intron A. The changes from the VR1012 to the VR10551 include some modifications to the multiple cloning site, and a modified rabbit β globin 3′ untranslated region/polyadenylation signal sequence/transcriptional terminator has been substituted for the same functional domain derived from the bovine growth hormone gene.
  • Additionally, constructs of the present invention can be inserted into other eukaryotic expression vector backbones such as VR10682 or VR10686. The VR10682 expression vector backbone (SEQ ID NO:94) contains a modified rous sarcoma virus (RSV) promoter from expression plasmid VCL1005, the bovine growth hormone (BGH) poly-adenylation site and a polylinker for inserting foreign genes and a kanamycin resistance gene. The RSV promoter in VCL1005 and VR10682 contains a XbaI endonuclease restriction site near the transcription start site in the sequence TAC TCT AGA CG (SEQ ID NO:82). The modified RSV promoter contained in VR10682. Expression plasmid VCL1005 is described in U.S. Pat. No. 5,561,064 and is incorporated herein by reference.
  • The VR10686 expression vector backbone (SEQ ID NO:112) was created by replacing the West Nile Virus (WNV) antigen insert in VR6430 (SEQ ID NO:89) with the multiple cloning site from the VR1012 vector. The VR10686 and VR6430 expression vector backbones contain the RSV promoter, derived from VCL1005, which has been modified back to the wild-type RSV sequence (TAC AAT AAA CG (SEQ ID NO:83)). The wild-type RSV promoter is fused to the “R” region plus the first 39 nucleotides of the U5 region from Human T-Cell Leukemia Virus I (HTLV-I), hereinafter refered to as the RU5 element. The R and U5 regions are portions of the long terminal repeat region (LTR) of HTLV-I which control expression of the HTLV-I transcript and is duplicated at either end of the integrated viral genome as a result of the retroviral integration mechanism. The LTR of HTLV-1 and most retroviruses are divided into three regions, U3, R and U5. Transcription from the intigrated viral genome commences at the U3-R boundary of the 5′ LTR and the transcript is polyadenylated at the R-U5 boundary of the 3′ LTR. (See Goff, S. P. Retroviridae, Field's Virology 4th ed. 2:1871-1939 (2001). This RU5 HTLV-I element has been shown to be a potent stimulator of translation when fused to the SV40 early gene promoter. See Takebe et al., Mol. Cell Biol. 8:466-472 (1988). It has been proposed that the stimulation of translation by the HTLV-I RU5 element is due to its function, in part, as a translational enhancing internal ribosome entry site (IRES). See Attal et al. FEBS Letters 392:220-224 (1996). Additionally the HTLV-I RU5 element provides the 5′-splice donor site. Immediately downstream of the RU5 element is the 3′-end of the HCMV intron A sequence containing the splice acceptor sequence. The VR10686 and VR6430 expression vectors contain a hybrid intron composed of the 5′-HTLV I intron sequence fused to the 3′-end of the HCMV intron A, a bovine growth hormone poly-adenylation site, a polylinker for insertion of forign genes and a kanamycin resistance gene. The VR6430 vector expresses the prM and E West Nile Virus antigens (Genebank Accession No. AF202541).
  • The vector backbones described above may by used to create expression vectors which express multiple influenza proteins, fragments, variants or derivatives thereof. An expression vector as desribed herein may contain an additional promoter. For example, construct VR4774 (described in Example 13), contains a CMV promoter and an RSV promoter. Thus, the vector backbones described herein may contain multiple expression cassettes which comprise a promoter and an influenza coding sequence including, inter alia, polynucleotides as described herein. The expression cassettes may encode the same or different influenza polypeptides. Additionally, the expression cassettes may be in the same or opposite orientation relative to each other. As such transcription from each cassette may be in the same or opposition direction (i.e. 5′ to 3′ in both expression cassettes or, alternatively, 5′ to 3′ in one expression cassette and 3′ to 5′ in the other expression cassette).
  • Plasmid DNA Purification
  • Plasmid DNA may be transformed into competent cells of an appropriate Escherichia coli strain (including but not limited to the DH5α strain) and highly purified covalently closed circular plasmid DNA was isolated by a modified lysis procedure (Horn, N. A., et al., Hum. Gene Ther. 6:565-573 (1995)) followed by standard double CsCl-ethidium bromide gradient ultracentrifugation (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y. (1989)). Alternatively, plasmid DNAs are purified using Giga columns from Qiagen (Valencia, Calif.) according to the kit instructions. All plasmid preparations were free of detectable chromosomal DNA, RNA and protein impurities based on gel analysis and the bicinchoninic protein assay (Pierce Chem. Co., Rockford Ill.). Endotoxin levels were measured using Limulus Amebocyte Lysate assay (LAL, Associates of Cape Cod, Falmouth, Mass.) and were less than 0.6 Endotoxin Units/mg of plasmid DNA. The spectrophotometric A260/A280 ratios of the DNA solutions were typically above 1.8. Plasmids were ethanol precipitated and resuspended in an appropriate solution, e.g., 150 mM sodium phosphate (for other appropriate excipients and auxiliary agents, see U.S. patent application Publication 2002/0019358, published Feb. 14, 2002). DNA was stored at −20EC until use. DNA was diluted by mixing it with 300 mM salt solutions and by adding appropriate amount of USP water to obtain 1 mg/ml plasmid DNA in the desired salt at the desired molar concentration.
  • Plasmid Expression in Mammalian Cell Lines
  • The expression plasmids were analyzed in vitro by transfecting the plasmids into a well characterized mouse melanoma cell line (VM-92, also known as UM-449). See, e.g., Wheeler, C. J., Sukhu, L., Yang, G., Tsai, Y., Bustamente, C., Felgner, P. Norman, J & Manthorpe, M. “Converting an Alcohol to an Amine in a Cationic Lipid Dramatically Alters the Co-lipid Requirement, Cellular Transfection Activity and the Ultrastructure of DNA-Cytofectin Complexes,” Biochim. Biophys. Acta. 1280:1-11 (1996). Other well-characterized human cell lines can also be used, e.g. MRC-5 cells, ATCC Accession No. CCL-171 or human rhabdomyosarcoma cell line RD (ATCC CCL-136). The transfection was performed using cationic lipid-based transfection procedures well known to those of skill in the art. Other transfection procedures are well known in the art and may be used, for example electroporation and calcium chloride-mediated transfection (Graham F. L. and A. J. van der Eb Virology 52:456-67 (1973)). Following transfection, cell lysates and culture supernatants of transfected cells were evaluated to compare relative levels of expression of IV antigen proteins. The samples were assayed by western blots and ELISAs, using commercially available polyclonal and/or monoclonal antibodies (available, e.g., from Research Diagnostics Inc., Flanders N.J.), so as to compare both the quality and the quantity of expressed antigen.
  • Injections of Plasmid DNA
  • The quadriceps muscles of restrained awake mice (e.g., female 6-12 week old BALB/c mice from Harlan Sprague Dawley, Indianapolis, Ind.) are injected bilaterally with 1-50 μg of DNA in 50 μl solution (100 μg in 100 μl total per mouse) using a disposable plastic insulin syringe and 28G ½ needle (Becton-Dickinson, Franklin Lakes, N.J., Cat. No. 329430) fitted with a plastic collar cut from a micropipette tip, as previously described (Hartikka, J., et al., Hum. Gene Ther. 7:1205-1217 (1996).
  • Animal care throughout the study was in compliance with the “Guide for the Use and Care of Laboratory Animals”, Institute of Laboratory Animal Resources, Commission on Life Sciences, National Research Council, National Academy Press, Washington, D.C., 1996 as well as with Vical's Institutional Animal Care and Use Committee.
  • Example 1 Construction of Expression Vectors
  • Plasmid constructs comprising the native coding regions encoding NP, M1, M2, HA, and eM2, IV proteins or fragments, variants or derivatives are constructed as follows. The NP, M1, and M2 genes from IV (A/PR/8/34) are isolated from viral RNA by RT PCR, or prepared by direct synthesis if the wildtype sequence is known, by standard methods and are inserted into the vector VR10551 via standard restriction sites, by standard methods.
  • Plasmid constructs comprising human codon-optimized coding regions encoding NP, M1, M2, HA, eM2, and/or an eM2-NP fusion; or other codon-optimized coding regions encoding other IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, are prepared as follows. The codon-optimized coding regions are generated using the full, minimal, or uniform codon optimization methods described herein. The codon optimized coding regions are constructed using standard PCR methods described herein, or are ordered commercially. Oligonucleotides representing about the first 23-24 aa extracellular region of M2 are constructed, and are used in an overlap PCR reaction with the NP coding regions described above, to create a coding region coding for an eM2/NP fusion protein, for example as shown in SEQ ID NOs 6 and 7. The codon-optimized coding regions are inserted into the vector VR10551 via standard restriction sites, by standard methods.
  • Plasmids constructed as above are propagated in Escherichia coli and purified by the alkaline lysis method (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., ed. 2 (1989)). CsCl-banded DNA are ethanol precipitated and resuspended in 0.9% saline or PBS to a final concentration of 2 mg/ml for injection. Alternately, plasmids are purified using any of a variety of commercial kits, or by other known procedures involving differential precipitation and/or chromatographic purification.
  • Expression is tested by formulating each of the plasmids in DMRIE/DOPE and transfecting VM92 cells. The supernatants are collected and the protein production tested by Western blot or ELISA. The relative expression of the wild type and codon optimized constructs are compared.
  • Examples of constructs made according to the above methods are listed in Table 13. The experimental procedure for generating the listed constructs is as described above, with particular parameters and materials employed as described herein.
    TABLE 13
    Plasmid # Description
    VR4700 TPA leader - NP (A/PR/34) in VR 1255
    VR4707 TPA leader-M2 with transmembrane deletion,
    glycine linker inserted
    VR4710 TPA leader - 1st 24 amino acids of M2 from
    VR4707 fused to NP from VR4700
    VR4750 full length HA from mouse adapted virus (H3, Hong Kong 68)
    VR4752 full length HA from mouse adapted virus
    (H1, Puerto Rico 34)
    VR4755 algorithm to codon optimize consensus amino acid
    sequence, direct fusion M2 to ATG of M1
    VR4756 native sequence from A/Niigata/137/96 influenza strain
    (matches amino acid consensus sequence)
    VR4757 Contracted codon optimized - 1st 24 amino acids
    of M2 from consensus fused to full-
    length NP consensus
    VR4758 Applicants' codon optimized - 1st 24 amino
    acids of M2 from consensus fused to full-
    length NP consensus
    VR4759 Full-length M2 derived from VR4755
    VR4760 Full-length M1 derived from VR4755
    VR4761 Full-length NP derived from VR4757
    VR4762 Full-length NP derived from VR4758
    VR4763 Selectively codon-optimized regions of segment 7
  • The pDNA expression vector VR4700 which encodes the influenza NP protein has been described in the art. See, e.g. Sankar, V., Baccaglilni, L., Sawddey, M., Wheeler, C. J., Pillemer, S. R., Baum, B. J. and Atkinson, J. C., “Salivary Gland Delivery of pDNA-Cationic Lipolplexes Elicits Systemic Immune Responses,” Oral Diseases 8:275-281 (2002). The following is the open reading frame for TPA-NP (from VR4700), referred to herein as SEQ ID NO:46:
    1 atggatgcaa tgaagagagg gctctgctgt gtgctgctgc tgtgtggagc agtcttcgtt
    61 tcgcccagcg ctagaggatc gggaatggcg tcccaaggca ccaaacggtc ttacgaacag
    121 atggagactg atggagaacg ccagaatgcc actgaaatca gagcatccgt cggaaaaatg
    181 attggtggaa ttggacgatt ctacatccaa atgtgcaccg aactcaaact cagtgattat
    241 gagggacggt tgatccaaaa cagcttaaca atagagagaa tggtgctctc tgcttttgac
    301 gaaaggagaa ataaatacct ggaagaacat cccagtgcgg ggaaagatcc taagaaaact
    361 ggaggaccta tatacaggag agtaaacgga aagtggatga gagaactcat cctttatgac
    421 aaagaagaaa taaggcgaat ctggcgccaa gctaataatg gtgacgatgc aacggctggt
    481 ctgactcaca tgatgatctg gcattccaat ttgaatgatg caacttatca gaggacaaga
    541 gctcttgttc gcaccggaat ggatcccagg atgtgctctc tgatgcaagg ttcaactctc
    601 cctaggaggt ctggagccgc aggtgctgca gtcaaaggag ttggaacaat ggtgatggaa
    661 ttggtcagga tgatcaaacg tgggatcaat gatcggaact tctggagggg tgagaatgga
    721 cgaaaaacaa gaattgctta tgaaagaatg tgcaacattc tcaaagggaa atttcaaact
    781 gctgcacaaa aagcaatgat ggatcaagtg agagagagcc ggaacccagg gaatgctgag
    841 ttcgaagatc tcacttttct agcacggtct gcactcatat tgagagggtc ggttgctcac
    901 aagtcctgcc tgcctgcctg tgtgtatgga cctgccgtag ccagtgggta cgactttgaa
    961 agagagggat actctctagt cggaatagac cctttcagac tgcttcaaaa cagccaagtg
    1021 tacagcctaa tcagaccaaa tgagaatcca gcacacaaga gtcaactggt gtggatggca
    1081 tgccattctg ccgcatttga agatctaaga gtattaagct tcatcaaagg gacgaaggtg
    1141 ctcccaagag ggaagctttc cactagagga gttcaaattg cttccaatga aaatatggag
    1201 actatggaat caagtacact tgaactgaga agcaggtact gggccataag gaccagaagt
    1261 ggaggaaaca ccaatcaaca gagggcatct gcgggccaaa tcagcataca acctacgttc
    1321 tcagtacaga gaaatctccc ttttgacaga acaaccatta tggcagcatt caatgggaat
    1381 acagagggaa gaacatctga catgaggacc gaaatcataa ggatgatgga aagtgcaaga
    1441 ccagaagatg tgtctttcca ggggcgggga gtcttcgagc tctcggacga aaaggcagcg
    1501 agcccgatcg tgccttcctt tgacatgagt aatgaaggat cttatttctt cggagacaat
    1561 gcagatgagt acgacaatta a
  • Purified VR4700 DNA was used to transfect the murine cell line VM92 to determine expression of the NP protein. Expression of NP was confirmed with a Western Blot assay. Western blot analysis showed very low level expression of VR4700 in vitro as detected with mouse polyclonal anti-NP antibody. In vivo antibody response was detected by ELISA with an average titer of 62,578.
  • Plasmid VR4707 expresses a secreted form of M2, i.e., TPA-M2. The sequence was assembled using synthetic oligonucleotides in which the oligos were annealed amongst themselves, and then ligated and gel purified. The purified product was then ligated (cloned) into Eco RI/Sal I of VR10551. The M2 sequence lacks the transmembrane domain; the cloned sequence contains amino acids [TPA(1-23)]ARGSG[M2(1-25)]GGG[M2(44-97)]. Amino acid residues between TPA and M2 and between M2 domains were added as flexible linkers. The following mutations were introduced to generate appropriate T-cell epitopes: 74S→G and 78S→N. The following is the open reading frame for TPA-M2ATM (from VR4707), referred to herein as SEQ ID NO:47:
    1 atggatgcaa tgaagagagg gctctgctgt gtgctgctgc tgtgtggagc agtcttcgtt
    61 tcgcccagcg ctagaggatc gggaatgagt cttctgaccg aggtcgaaac ccctatcaga
    121 aacgaatggg ggtgcagatg caacgattca agtgatcctg gcggcggcga tcggcttttt
    181 ttcaaatgca tttatcggcg ctttaaatac ggcttgaaaa gagggccttc taccgaagga
    241 gtgccagagt ctatgaggga agaatatcgg aaggaacagc agaatgctgt ggatgttgac
    301 gatagccatt ttgtcagcat cgagctggag taa
  • Purified VR4707 DNA was used to transfect the murine cell line VM92 to determine expression of the M2 protein. Expression of M2 was confirmed with a Western Blot assay. Expression was visualized with a commercially available anti-M2 monoclonal antibody. In vivo M2 antibody response to VR4707, as assayed by ELISA, resulted in an average titer of 110, which is lower than the average titer of 9,240 for VR4756, encoding full-length M2 from segment 7. An IFNγ ELISPOT assay for M2-specific T cells resulted in an average of 61 SFU/106 cells versus an average of 121 SFU/106 cells for the segment 7 construct.
  • VR4710 was created by fusing the TPA leader and the first 24 amino acids of M2 from VR4707 to the full-length NP gene from VR4700. Primers 5′-GCCGAATCCATGGATGCAATGAAG-3′ (SEQ ID NO:48) and 5′-GGTGCCTTGGGACGCCATATCACTTGAATCGTTGCA-3′ (SEQ ID NO:49) were used to amplify the TPA-M2 fragment from VR4707. Primers 5′-TGCAACGATTCAAGTGATATGGCGTCCCAAGGCACC-3′ (SEQ ID NO:50) and 5′-GCCGTCGACTTAATTGTCGTACTC-3′ (SEQ ID NO:51) were used to amplify the NP gene from VR4700. Then the N-terminal and C-terminal primers were used to assemble the fusion, and the eM2NP fusion was cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for TPA-M2-NP (from VR4710), referred to herein as SEQ ID NO:52:
    1 atggatgcaa tgaagagagg gctctgctgt gtgctgctgc tgtgtggagc agtcttcgtt
    61 tcgcccagcg ctagaggatc gggaatgagt cttctgaccg aggtcgaaac ccctatcaga
    121 aacgaatggg ggtgcagatg caacgattca agtgatatgg cgtcccaagg caccaaacgg
    181 tcttacgaac agatggagac tgatggagaa cgccagaatg ccactgaaat cagagcatcc
    241 gtcggaaaaa tgattggtgg aattggacga ttctacatcc aaatgtgcac cgaactcaaa
    301 ctcagtgatt atgagggacg gttgatccaa aacagcttaa caatagagag aatggtgctc
    361 tctgcttttg acgaaaggag aaataaatac ctggaagaac atcccagtgc ggggaaagat
    421 cctaagaaaa ctggaggacc tatatacagg agagtaaacg gaaagtggat gagagaactc
    481 atcctttatg acaaagaaga aataaggcga atctggcgcc aagctaataa tggtgacgat
    541 gcaacggctg gtctgactca catgatgatc tggcattcca atttgaatga tgcaacttat
    601 cagaggacaa gagctcttgt tcgcaccgga atggatccca ggatgtgctc tctgatgcaa
    661 ggttcaactc tccctaggag gtctggagcc gcaggtgctg cagtcaaagg agttggaaca
    721 atggtgatgg aattggtcag gatgatcaaa cgtgggatca atgatcggaa cttctggagg
    781 ggtgagaatg gacgaaaaac aagaattgct tatgaaagaa tgtgcaacat tctcaaaggg
    841 aaatttcaaa ctgctgcaca aaaagcaatg atggatcaag tgagagagag ccggaaccca
    901 gggaatgctg agttcgaaga tctcactttt ctagcacggt ctgcactcat attgagaggg
    961 tcggttgctc acaagtcctg cctgcctgcc tgtgtgtatg gacctgccgt agccagtggg
    1021 tacgactttg aaagagaggg atactctcta gtcggaatag accctttcag actgcttcaa
    1081 aacagccaag tgtacagcct aatcagacca aatgagaatc cagcacacaa gagtcaactg
    1141 gtgtggatgg catgccattc tgccgcattt gaagatctaa gagtattaag cttcatcaaa
    1201 gggacgaagg tgctcccaag agggaagctt tccactagag gagttcaaat tgcttccaat
    1261 gaaaatatgg agactatgga atcaagtaca cttgaactga gaagcaggta ctgggccata
    1321 aggaccagaa gtggaggaaa caccaatcaa cagagggcat ctgcgggcca aatcagcata
    1381 caacctacgt tctcagtaca gagaaatctc ccttttgaca gaacaaccat tatggcagca
    1441 ttcaatggga atacagagyg aagaacatct gacatgagga ccgaaatcat aaggatgatg
    1501 gaaagtgcaa gaccagaaga tgtgtctttc caggggcggg gagtcttcga gctctcggac
    1561 gaaaaggcag cgagcccgat cgtgccttcc tttgacatga gtaatgaagg atcttatttc
    1621 ttcggagaca atgcagatga gtacgacaat taa
  • Purified VR4710 DNA was used to transfect the murine cell line VM92 to determine expression of the eM2-NP fusion protein. Expression of EM2-NP was confirmed with a Western Blot assay. Expression was visualized with a commercially available monoclonal antibody to M2 and with mouse polyclonal antibody to NP. ELISA assay results following 2 injections of pDNA into mice revealed little antibody response to M2, but an average titer of 66,560 for anti-NP antibody.
  • VR4750 was created by first reverse transcribing RNA from the mouse-adapted A/Hong Kong/1/68 virus stock using random hexamer to create a cDNA library. Then primers 5′ GGGCTAGCGCCGCCACCATGAAGACCATCATTGCT 3′ (SEQ ID NO:53) and 5′ CCGTCGACTCAAATGCAAATGTTGCA 3′ (SEQ ID NO:54) were employed to PCR the HA gene. The gene was inserted into the Invitrogen TOPO-TA vector first, and then sub-cloned into VR10551 using restriction enzymes NheI and SalI. The following is the open reading frame for HA (H3N2) from mouse-adapted A/Hong Kong/68 (from VR4750), referred to herein as SEQ ID NO:55:
    1 atgaagacca tcattgcttt gagctacatt ttctgtctgg ctctcggcca agaccttcca
    61 ggaaatgaca acaacacagc aacgctgtgc ctgggacatc atgcggtgcc aaacggaaca
    121 ctagtgaaaa caatcacaga tgatcagatt gaagtgacta atgctactga gctagttcag
    181 agctcctcaa cggggaaaat atgcaacaat cctcatcgaa tccttgatgg aatagactgc
    241 acactgatag atgctctatt gggggaccct cattgtgatg tttttcaaaa tgagacatgg
    301 gaccttttcg ttgaacgcag caaagctttc agcaactgtt acccttatga tgtgccagat
    361 tatgcccccc ttaggtcact agttgcctcg tcaggcactc tggagtttat cactgagggt
    421 ttcacttgga ctggggtcac tcagaatggg ggaagcagtg cttgcaaaag gggacctggt
    481 agcggttttt tcagtagact gaactggttg accaaatcag gaagcacata tccagtgctg
    541 aacgtgacta tgccaaacaa tgacaatttt gacaaactat acatttgggg ggttcaccac
    601 ccgagcacga accaagaaca aaccagcctg tatgttcaag catcagggag agtcacagtc
    661 tctaccagga gaagccagca aactataatc ccgaatatcg agtccagacc ctgggtaagg
    721 ggtctgtcta gtagaataag catctattgg acaatagtta agccgggaga cgtactggta
    781 attaatagta atgggaacct aatcgctcct cggggttatt tcaagatgcg cactgggaaa
    841 agctcaataa tgaggtcaga tgcacctatt gatacctgta tttctgaatg catcactcca
    901 aatggaagca ttcccaatga caagcccttt caaaacgtaa acaaaatcac gtatggagca
    961 tgccccaagt atgttaagca aaacaccctg aagttggcaa cagggatgcg gaatgtacca
    1021 gagaaacaaa ctagaggcct attcggcgca atagcaggtt tcatagaaaa tggttgggag
    1081 ggaatgatag acggttggta cggtttcagg catcaaaatt ctgagggcac aggacaagca
    1141 gcagatctta aaagcactca agcagccatc gaccaaatca atgggaaatt gaacaggata
    1201 atcaagaaga cgaacgagaa attccatcaa atcgaaaagg aattctcaga agtagaaggg
    1261 agaattcagg acctcgagaa atacgttgaa gacactaaaa tagatctctg gtcttacaat
    1321 gcggagcttc ttgtcgctct ggagaatcaa catacaattg acctgactga ctcggaaatg
    1381 aacaagctgt ttgaaaaaac aaggaggcaa ctgagggaaa atgctgaaga catgggcaat
    1441 ggttgcttca aaatatacca caaatgtgac aacgcttgca tagagtcaat cagaactggg
    1501 acttatgacc atgatgtata cagagacgaa gcattaaaca accggtttca gatcaaaggt
    1561 gttgaactga agtctggata caaagactgg atcctgtgga tttcctttgc catatcatgc
    1621 tttttgcttt gtgttgtttt gctggggttc atcatgtggg cctgccagaa aggcaacatt
    1681 aggtgcaaca tttgcatttg a
  • While VR4750 expression was not clearly detected in vitro by Western blot Assay, two 100 μg vaccinations of VR4750 have been shown to protect mice from intranasal challenge with mouse-adapted A/Hong Kong/68 virus.
  • VR4752 was created by first reverse transcribing RNA from the mouse-adapted A/Puerto Rico/8/34 virus stock using random hexamer to create a cDNA library. Then primers 5′ GGGCTAGCGCCGCCACCATGAAGGCAAACCTACTG 3′ (SEQ ID NO:56) and 5′ CCGTCGACTCAGATGCATATTCTGCA 3′ (SEQ ID NO:57) were employed to PCR the HA gene. The gene was then cloned into the TOPO-TA vector first, and then sub-cloned into VR10551 using restriction enzymes NheI and SalI. The following is the open reading frame for HA (H1N1) cloned from mouse-adapted A/Puerto Rico/34 (from VR4752), referred to herein as SEQ ID NO:58:
    1 atgaaggcaa acctactggt cctgttatgt gcacttgcag ctgcagatgc agacacaata
    61 tgtataggct accatgcgaa caattcaacc gacactgttg acacagtgct cgagaagaat
    121 gtgacagtga cacactctgt taacctgctc gaagacagcc acaacggaaa actatgtaga
    181 ttaaaaggaa tagccccact acaattgggg aaatgtaaca tcgccggatg gctcttggga
    241 aacccagaat gcgacccact gcttccagtg agatcatggt cctacattgt agaaacacca
    301 aactctgaga atggaatatg ttatccagga gatttcatcg actatgagga gctgagggag
    361 caattgagct cagtgtcatc attcgaaaga ttcgaaatat ttcccaaaga aagctcatgg
    421 cccaaccaca acacaaccaa aggagtaacg gcagcatgct cccatgcggg gaaaagcagt
    481 ttttacagaa atttgctatg gctgacggag aaggagggct catacccaaa gctgaaaaat
    541 tcttatgtga acaagaaagg gaaagaagtc cttgtactgt ggggtattca tcacccgtct
    601 aacagtaagg atcaacagaa tatctatcag aatgaaaatg cttatgtctc tgtagtgact
    661 tcaaattata acaggagatt taccccggaa atagcagaaa gacccaaagt aagagatcaa
    721 gctgggagga tgaactatta ctggaccttg ctaaaacccg gagacacaat aatatttgag
    781 gcaaatggaa atctaatagc accaaggtat gctttcgcac tgagtagagg ctttgggtcc
    841 ggcatcatca cctcaaacgc atcaatgcat gagtgtaaca cgaagtgtca aacacccctg
    901 ggagctataa acagcagtct ccctttccag aatatacacc cagtcacaat aggagagtgc
    961 ccaaaatacg tcaggagtgc caaattgagg atggttacag gactaaggaa cattccgtcc
    1021 attcaatcca gaggtctatt tggagccatt gccggtttta ttgaaggggg atggactgga
    1081 atgatagatg gatggtacgg ttatcatcat cagaatgaac agggatcagg ctatgcagcg
    1141 gatcaaaaaa gcacacaaaa tgccattaac gggattacaa acaaggtgaa ctctgttatc
    1201 gagaaaatga acattcaatt cacagctgtg ggtaaagaat tcaacaaatt agaaaaaagg
    1261 atggaaaatt taaataaaaa agttgatgat ggatttctgg acatttggac atataatgca
    1321 gaattgttag ttctactgga aaatgaaagg actctggatt tccatgactc aaatgtgaag
    1381 aatctgtatg agaaagtaaa aagccaatta aagaataatg ccaaagaaat cggaaatgga
    1441 tgttttgagt tctaccacaa gtgtgacaat gaatgcatgg aaagtgtaag aaatgggact
    1501 tatgattatc ccaaatattc agaagagtca aagttgaaca gggaaaaggt agatggagtg
    1561 aaattggaat caatggggat ctatcagatt ctggcgatct actcaactgt cgccagttca
    1621 ctggtgcttt tggtctccct gggggcaatc agtttctgga tgtgttctaa tggatctttg
    1681 cagtgcagaa tatgcatctg a
  • Purified VR4752 DNA was used to transfect the murine cell line VM92 to determine expression of the HA protein. Expression of HA was confirmed with a Western Blot assay. Expression was visualized with a commercially available goat anti-influenza A (H1N1) antibody.
  • A direct fusion of the M2 gene to the M1 gene was synthesized based on a codon-optimized sequence derived from methods described in Example 4 using the “universal” optimization strategy. The synthesized gene was received in the pUC119 vector and then sub-cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for the M2M1 fusion (from VR4755), referred to herein as SEQ ID NO:59:
    1 atgagcctgc tgaccgaggt ggagaccccc atcagaaacg agtggggctg cagatgcaac
    61 gacagcagcg accccctggt ggtggccgcc agcatcatcg gcatcctgca cctgatcctg
    121 tggatcctgg acagactgtt cttcaagtgc atctacagac tgttcaagca cggcctgaag
    181 agaggcccca gcaccgaggg cgtgcccgag agcatgagag aggagtacag aaaggagcag
    241 cagaacgccg tggacgccga cgacagccac ttcgtgagca tcgagctgga gatgtccctg
    301 ctgacagaag tggaaacata cgtgctgagc atcgtgccca gcggccccct gaaggccgag
    361 atcgcccaga gactggagga cgtgttcgcc ggcaagaaca ccgacctgga ggccctgatg
    421 gagtggctga agaccagacc catcctgagc cccctgacca agggcatcct gggcttcgtg
    481 ttcaccctga ccgtgcccag cgagagaggc ctgcagagaa gaagattcgt gcagaacgcc
    541 ctgaacggca acggcgaccc caacaacatg gaccgggccg tgaagctgta ccggaagctg
    601 aagagagaga tcaccttcca cggcgccaag gagatcgccc tgagctacag cgccggcgcc
    661 ctggccagct gcatgggcct gatctacaac agaatgggcg ccgtgaccac cgaggtggcc
    721 ttcggcctgg tgtgcgccac ctgcgagcag atcgccgaca gccagcacag aagccacaga
    781 cagatggtgg ccaccaccaa ccccctgatc agacacgaga acagaatggt gctggccagc
    841 accaccgcca aggccatgga gcagatggcc ggcagcagcg agcaggccgc cgaggccatg
    901 gagatcgcca gccaggccag acagatggtg caggccatga gagccatcgg cacccacccc
    961 agcagcagcg ccggcctgaa ggacgacctg ctggagaacc tgcagaccta ccagaagaga
    1021 atgggcgtgc agatgcagag attcaagtga
  • Purified VR4755 DNA was used to transfect the murine cell line VM92 to determine expression of the M2M1 fusion protein. Expression of M2M1 was confirmed with a Western Blot assay. Expression of the M2M1 fusion was visualized with commercially available anti-M1 and anti-M2 monoclonal antibodies.
  • The segment 7 RNA of influenza A encodes both the M1 and M2 genes. A consensus amino acid sequence for M1 and M2 was derived according to methods described herein. The consensus sequences for both proteins, however, are identical to the M1 and M2 amino acid sequences derived from the IV strain A/Niigata/137/96, represented herein as SEQ ID NO:77 and SEQ ID NO:78, respectively. Accordingly, the native sequence for segment 7, A/Niigata/137/96, was synthesized and received as an insert in pUC119. The segment 7 insert was sub-cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for segment 7 (from VR4756), referred to herein as SEQ ID NO:60:
    1 atgagccttc taaccgaggt cgaaacgtat gttctctcta tcgttccatc aggccccctc
    61 aaagccgaaa tcgcgcagag acttgaagat gtctttgctg ggaaaaacac agatcttgag
    121 gctctcatgg aatggctaaa gacaagacca atcctgtcac ctctgactaa ggggattttg
    181 gggtttgtgt tcacgctcac cgtgcccagt gagcgaggac tgcagcgtag acgctttgtc
    241 caaaatgccc tcaatgggaa tggggatcca aataacatgg acagagcagt taaactatat
    301 agaaaactta agagggagat tacattccat ggggccaaag aaatagcact cagttattct
    361 gctggtgcac ttgccagttg catgggcctc atatacaaca gaatgggggc tgtaaccact
    421 gaagtggcct ttggcctggt atgtgcaaca tgtgaacaga ttgctgactc ccagcacagg
    481 tctcataggc aaatggtggc aacaaccaat ccattaataa ggcatgagaa cagaatggtt
    541 ttggccagca ctacagctaa ggctatggag caaatggctg gatcaagtga gcaggcagcg
    601 gaggccatgg aaattgctag tcaggccagg caaatggtgc aggcaatgag agccattggg
    661 actcatccta gctccagtgc tggtctaaaa gatgatcttc ttgaaaattt gcagacctat
    721 cagaaacgaa tgggggtgca gatgcaacga ttcaagtgac ccgcttgttg ttgctgcgag
    781 tatcattggg atcttgcact tgatattgtg gattcttgat cgtctttttt tcaaatgcat
    841 ctatcgactc ttcaaacacg gtctgaaaag agggccttct acggaaggag tacctgagtc
    901 tatgagggaa gaatatcgaa aggaacagca gaatgctgtg gatgctgacg acagtcattt
    961 tgtcagcata gagctggagt aa
  • SEQ ID NO:77 (“consensus” (A/Niigata/137/96) M1):
    MSLLTEVETYVLSIVPSGPLKAEIAQRLEDVFAGKNTDLEALMEWLKTRP
    ILSPLTKGILGFVFTLTVPSERGLQRRRFVQNALNGNGDPNNMDRAVKLY
    RKLKREITFHGAKEIALSYSAGALASCMGLIYNRMGAVTTEVAFGLVCAT
    CEQIADSQHRSHRQMVATTNPLIRHENRMVLASTTAKAMEQMAGSSEQAA
    EAMEIASQARQMVQAMRAIGTHPSSSAGLKDDLLENLQTYQKRMGVQM
    QRFK
  • SEQ ID NO:78 (“consensus” (A/Niigata/137/96) M2):
    MSLLTEVETPIRNEWGCRCNDSSDPLVVAASIIGILHLILWILDRLFFKC
    IYRLFKHGLKRGPSTEGVPESMREEYRKEQQNAVDADDSHFVSIELE
  • Purified VR4756 DNA was used to transfect the murine cell line VM92 to determine expression of the the proteins encoded by segment 7. Expression of both M1 and M2 was confirmed with a Western blot assay using commercially available anti-M1 and anti-M2 monoclonal antibodies. ELISA assay results following 2 injections of pDNA into mice revealed an average anti-M2 antibody titer of 9,240 versus a 110 average titer for VR4707. An IFNγ ELISPOT assay for M2-specific T cells resulted in an average of 121 SFU/106 cells for VR4756 injected mice versus an average of 61 SFU/106 cells for the VR4707 construct.
  • An additional segment 7 sequence is created, VR4763, which contains selectively codon-optimized regions of segment 7. Optimization of the coding regions in segment 7 is selective, because segment 7 contains two overlapping coding regions (i.e., encoding M1 and M2,) and these coding regions are partially in different reading frames. From the AUG encoded by nucleotides 1 to 3 of segment 7, M1 is encoded by bp 1 through 759 of the segment 7 RNA, while M2 is encoded by a spliced messenger RNA which includes nucleotides 1 to 26 of segment 7 spliced to nucleotides 715 to 982 of segment 7. Optimization of the region from 715 to 759 is avoided because the M1 and M2 coding sequences (in different reading frames) overlap in that region. Due to the splicing that occurs to join bp 26 to an alternate frame at bp 715 of the segment 7 sequence, optimization in these splicing regions is also avoided; adjacent regions that arguably could also participate in splicing are likewise avoided. Optimization is done in a manner to insure that no new splicing sites are inadvertently introduced. The areas that are optimized are done so using “universal” strategy, e.g. inserting the most frequently used codon for each amino acid. The following is the nucleotide sequence for codon-optimized segment 7 (from VR4763), referred to herein as SEQ ID NO:61:
    1 atgagcctgc tgaccgaggt cgaaacgtat gttctctcta tcgtgcccag cggccccctg
    61 aaggccgaga tcgcccagag actggaggac gtgttcgccg gcaagaacac cgacctggag
    121 gccctgatgg agtggctgaa gaccagaccc atcctgagcc ccctgaccaa gggcatcctg
    181 ggcttcgtgt tcaccctgac cgtgcccagc gagagaggcc tgcagagaag aagattcgtg
    241 cagaacgccc tgaacggcaa cggcgacccc aacaacatgg acagagccgt gaagctgtac
    301 agaaagctga agagagagat caccttccac ggcgccaagg agatcgccct gagctacagc
    361 gccggcgccc tggccagctg catgggcctg atctacaaca gaatgggcgc cgtgaccacc
    421 gaggtggcct tcggcctggt gtgcgccacc tgcgagcaga tcgccgacag ccagcacaga
    481 agccacagac agatggtggc caccaccaac cccctgatca gacacgagaa cagaatggtg
    541 ctggccagca ccaccgccaa ggccatggag cagatggccg gcagcagcga gcaggccgcc
    601 gaggccatgg agatcgccag ccaggccaga cagatggtgc aggccatgag agccatcggc
    661 acccacccca gcagcagcgc cggcctgaaa gatgatcttc ttgaaaattt gcagacctat
    721 cagaaacgaa tgggggtgca gatgcaacga ttcaagtgac cccctggtgg tggccgccag
    781 catcatcggc atcctgcacc tgatcctgtg gatcctggac agactgttct tcaagtgcat
    841 ctacagactg ttcaagcacg gcctgaagag aggccccagc accgagggcg tgcccgagag
    901 catgagagag gagtacagaa aggagcagca gaacgccgtg gacgccgacg acagccactt
    961 cgtgagcatc gagctggagt ga
  • The codon optimized coding region for M1 extends from nucleotide 1 to nucleotide 759 of SEQ ID NO:61 including the stop codon, and is represented herein as SEQ ID NO:79. The codon-optimized coding region for M2 extends from nucleotide 1 to nucleotide 26 of SEQ ID NO:61 spliced to nucleotide 715 through nucleotide 959 of SEQ ID NO:61, including the stop codon, and is represented herein as SEQ ID NO:80.
  • Optimized M1 Coding Region (SEQ ID NO:79):
    ATGAGCCTGCTGACCGAGGTCGAAACGTATGTTCTCTCTATCGTGCCCAG
    CGGCCCCCTGAAGGCCGAGATCGCCCAGAGACTGGAGGACGTGTTCGCCG
    GCAAGAACACCGACCTGGAGGCCCTGATGGAGTGGCTGAAGACCAGACCC
    ATCCTGAGCCCCCTGACCAAGGGCATCCTGGGCTTCGTGTTCACCCTGAC
    CGTGCCCAGCGAGAGAGGCCTGCAGAGAAGAAGATTCGTGCAGAACGCCC
    TGAACGGCAACGGCGACCCCAACAACATGGACAGAGCCGTGAAGCTGTAC
    AGAAAGCTGAAGAGAGAGATCACCTTCCACGGCGCCAAGGAGATCGCCCT
    GAGCTACAGCGCCGGCGCCCTGGCCAGCTGCATGGGCCTGATCTACAACA
    GAATGGGCGCCGTGACCACCGAGGTGGCCTTCGGCCTGGTGTGCGCCACC
    TGCGAGCAGATCGCCGACAGCCAGCACAGAAGCCACAGACAGATGGTGGC
    CACCACCAACCCCCTGATCAGACACGAGAACAGAATGGTGCTGGCCAGCA
    CCACCGCCAAGGCCATGGAGCAGATGGCCGGCAGCAGCGAGCAGGCCGCC
    GAGGCCATGGAGATCGCCAGCCAGGCCAGACAGATGGTGCAGGCCATGAG
    AGCCATCGGCACCCACCCCAGCAGCAGCGCCGGCCTGAAAGATGATCTTC
    TTGAAAATTTGCAGACCTATCAGAAACGAATGGGGGTGCAGATGCAACGA
    TTCAAGTGA
  • Optimized M2 Coding Region (SEQ ID NO:80):
    ATGAGCCTGCTGACCGAGGTCGAAACACCTATCAGAAACGAATGGGGGTG
    CAGATGCAACGATTCAAGTGACCCCCTGGTGGTGGCCGCCAGCATCATCG
    GCATCCTGCACCTGATCCTGTGGATCCTGGACAGACTGTTCTTCAAGTGC
    ATCTACAGACTGTTCAAGCACGGCCTGAAGAGAGGCCCCAGCACCGAGGG
    CGTGCCCGAGAGCATGAGAGAGGAGTACAGAAAGGAGCAGCAGAACGCCG
    TGGACGCCGACGACAGCCACTTCGTGAGCATCGAGCTGGAGTGA
  • The eM2-NP fusion was codon-optimized, inserted in pUC119 and sub-cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for eM2-NP: codon-optimized by Contract (from VR4757), referred to herein as SEQ ID NO:62:
    1 atgagcttgc tcactgaagt cgagacacca atcagaaacg aatggggatg tagatgcaac
    61 gatagctcag acatggcctc ccagggaacc aaaagaagct atgaacagat ggagactgac
    121 ggagagagac agaacgccac agagatcaga gctagtgtag gaaagatgat agacggtatc
    181 gggcgatttt acattcaaat gtgtacggaa ttgaaactca gcgactatga aggcagactt
    241 atccagaact cactcacaat tgagcgcatg gtactcagtg catttgatga aagaaggaat
    301 aggtacctcg aagaacaccc cagcgccggc aaagatccca agaagactgg cggcccaatt
    361 tacagaagag tggacggtaa gtggatgaga gagctggtat tgtacgataa agaagaaatt
    421 agaagaatct ggaggcaagc aaacaatgga gaggatgcta cagctggcct gacccacatg
    481 atgatttggc atagtaacct gaatgatacc acctaccagc ggacaagggc tctcgttcga
    541 accgggatgg atccccgcat gtgctcattg atgcagggta gtacactccc gaggaggtca
    601 ggcgcggccg gtgcagccgt gaaaggaatc ggcactatgg taatggaatt gataagaatg
    661 attaaaaggg ggattaatga caggaacttt tggagaggag aaaatggacg caaaacaagg
    721 agtgcgtatg aacggatgtg caatattttg aaaggaaaat tccaaactgc agcacagcgc
    781 gccatgatgg atcaggtacg agaaagtcgc aacccaggta atgctgaaat agaggacctt
    841 atatttctcg cccggagtgc tctcatactt agaggaagcg tggcccataa aagttgtctc
    901 cccgcatgcg tatacggtcc cgctgtgtct tccggatacg attttgaaaa agagggatat
    961 tcattggtgg gaatcgaccc ttttaagctg cttcagaact cacaggttta cagtttgatt
    1021 agaccaaacg agaacccagc ccacaaatca caactcgtgt ggatggcatg ccactctgcc
    1081 gctttcgaag atctgagact gctctcattt attagaggca ctaaagtgag cccgagggga
    1141 aaactgagca cacgaggagt acagatagca tctaacgaaa atatggataa tatgggatct
    1201 agcacactcg aattgaggtc acgatactgg gctattagaa cacggagcgg agggaacacc
    1261 aaccagcaga gagcatccgc cggtcagata agcgttcagc ctacattttc agtacaacga
    1321 aacctgccat ttgaaaagag tacagtgatg gccgcattta ctggcaacac cgagggacga
    1381 acaagcgaca tgagagcaga gattattaga atgatggaag gagctaaacc agaggaggtt
    1441 tcatttagag gaaggggagt cttcgaattg tccgatgaga aagccacaaa tcccatagta
    1501 cctagcttcg acatgtccaa cgaaggctct tacttttttg gtgacaatgc cgaagagtac
    1561 gacaattga
  • Purified VR4757 DNA was used to transfect the murine cell line VM92 to determine expression of the eM2-NP fusion protein. Expression of eM2-NP was confirmed with a Western Blot assay. Expression was visualized with a commercially available monoclonal antibody to M2 and with mouse polyclonal antibody to NP. In vivo antibody response to NP was detected by ELISA with an average titer of 51,200.
  • The eM2-NP fusion gene in VR4758 was codon-optimized and synthesized. The gene was inserted into pUC119 and sub-cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for eM2-NP: codon-optimized by Applicants (from VR4758), referred to herein as SEQ ID NO:63:
    1 atgagcctgc tgaccgaggt ggagaccccc atcagaaacg agtggggctg cagatgcaac
    61 gacagcagcg acatggccag ccagggcacc aagagaagct acgagcagat ggagaccgac
    121 ggcgagagac agaacgccac cgagatcaga gccagcgtgg gcaagatgat cgacggcatc
    181 ggcagattct acatccagat gtgcaccgag ctgaagctga gcgactacga gggcagactg
    241 atccagaaca gcctgaccat cgagagaatg gtgctgagcg ccttcgacga gagaagaaac
    301 agatacctgg aggagcaccc cagcgccggc aaggacccca agaagaccgg cggccccatc
    361 tacagaagag tggacggcaa gtggatgaga gagctggtgc tgtacgacaa ggaggagatc
    421 agaagaatct ggagacaggc caacaacggc gaggacgcca ccgccggcct gacccacatg
    481 atgatctggc acagcaacct gaacgacacc acctaccaga gaaccagagc cctggtgcgg
    541 accggcatgg accccagaat gtgcagcctg atgcagggca gcaccctgcc cagaagaagc
    601 ggcgccgccg gcgccgccgt gaagggcatc ggcaccatgg tgatggagct gatcagaatg
    661 atcaagagag gcatcaacga cagaaacttc tggagaggcg agaacggcag aaagaccaga
    721 agcgcctacg agagaatgtg caacatcctg aagggcaagt tccagaccgc cgcccagaga
    781 gccatgatgg accaggtccg ggagagcaga aaccccggca acgccgagat cgaggacctg
    841 atcttcctgg ccagaagcgc cctgatcctg agaggcagcg tggcccacaa gagctgcctg
    901 cccgcctgcg tgtacggccc cgccgtgagc agcggctacg acttcgagaa ggagggctac
    961 agcctggtgg gcatcgaccc cttcaagctg ctgcagaaca gccaggtgta cagcctgatc
    1021 agacccaacg agaaccccgc ccacaagagc cagctggtgt ggatggcctg ccacagcgcc
    1081 gccttcgagg acctgagact gctgagcttc atcagaggca ccaaggtgtc ccccagaggc
    1141 aagctgagca ccagaggcgt gcagatcgcc agcaacgaga acatggacaa catgggcagc
    1201 agcaccctgg agctgagaag cagatactgg gccatcagaa ccagaagcgg cggcaacacc
    1261 aaccagcaga gagccagcgc cggccagatc agcgtgcagc ccaccttcag cgtgcagaga
    1321 aacctgccct tcgagaagag caccgtgatg gccgccttca ccggcaacac cgagggcaga
    1381 accagcgaca tgagagccga gatcatcaga atgatggagg gcgccaagcc cgaggaggtg
    1441 tccttcagag gcagaggcgt gttcgagctg agcgacgaga aggccaccaa ccccatcgtg
    1501 cctagcttcg acatgagcaa cgagggcagc tacttcttcg gcgacaacgc cgaggagtac
    1561 gacaactga
  • Purified VR4758 DNA was used to transfect the murine cell line VM92 to determine expression of the eM2-NP protein. Expression of eM2-NP was confirmed with a Western Blot assay. Expression was visualized with a commercially available monoclonal antibody to M2 and with mouse polyclonal antibody to NP. In vivo antibody response to NP was detected by ELISA with an average titer of 48,640.
  • The M2 gene was PCR-amplified from VR4755 using the primers 5′-GCCGAATTCGCCACCATGAGCCTGCTGACC-3′ (SEQ ID NO:64) and 5′-GCCGTCGACTGATCACTCCAGCTCGATGCTCAC-3′ (SEQ ID NO:65) and sub-cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for M2 (from VR4759), referred to herein as SEQ ID NO:66:
    1 atgagcctgc tgaccgaggt ggagaccccc atcagaaacg agtggggctg cagatgcaac
    61 gacagcagcg accccctggt ggtggccgcc agcatcatcg gcatcctgca cctgatcctg
    121 tggatcctgg acagactgtt cttcaagtgc atctacagac tgttcaagca cggcctgaag
    181 agaggcccca gcaccgaggg cgtgcccgag agcatgagag aggagtacag aaaggagcag
    241 cagaacgccg tggacgccga cgacagccac ttcgtgagca tcgagctgga gtga
  • Purified VR4759 DNA was used to transfect the murine cell line VM92 to determine expression of the M2 protein. Expression of M2 was confirmed with a Western Blot assay. Expression was visualized with a commercially available anti-M2 monoclonal antibody.
  • The M1 gene was PCR-amplified from VR4755 using the primers 5′-GCCGAATTCGCCACCATGTCCCTGCTGACAGAAGTG-3′ (SEQ ID NO:67) and 5′-GCCGTCGACTGATCACTTGAATCTCTGCATC-3′ (SEQ ID NO:68) and sub-cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for M1 (from VR4760), referred to herein as SEQ ID NO:69:
    1 atgtccctgc tgacagaagt ggaaacatac gtgctgagca tcgtgcccag cggccccctg
    61 aaggccgaga tcgcccagag actggaggac gtgttcgccg gcaagaacac cgacctggag
    121 gccctgatgg agtggctgaa gaccagaccc atcctgagcc ccctgaccaa gggcatcctg
    181 ggcttcgtgt tcaccctgac cgtgcccagc gagagaggcc tgcagagaag aagattcgtg
    241 cagaacgccc tgaacggcaa cggcgacccc aacaacatgg accgggccgt gaagctgtac
    301 cggaagctga agagagagat caccttccac ggcgccaagg agatcgccct gagctacagc
    361 gccggcgccc tggccagctg catgggcctg atctacaaca gaatgggcgc cgtgaccacc
    421 gaggtggcct tcggcctggt gtgcgccacc tgcgagcaga tcgccgacag ccagcacaga
    481 agccacagac agatggtggc caccaccaac cccctgatca gacacgagaa cagaatggtg
    541 ctggccagca ccaccgccaa ggccatggag cagatggccg gcagcagcga gcaggccgcc
    601 gaggccatgg agatcgccag ccaggccaga cagatggtgc aggccatgag agccatcggc
    661 acccacccca gcagcagcgc cggcctgaag gacgacctgc tggagaacct gcagacctac
    721 cagaagagaa tgggcgtgca gatgcagaga ttcaagtga
  • Purified VR4760 DNA was used to transfect the murine cell line VM92 to determine expression of the M1 protein. Expression of M1 was confirmed with a Western Blot assay. Expression was visualized with a commercially available anti-M1 monoclonal antibody.
  • The NP gene was PCR-amplified from VR4757 using primers 5′-GCCGAATTCGCCACCATGGCCTCCCAGGGAACCAAAAG-3′ (SEQ ID NO:70) and 5′-GCCGTCGACTGATCAATTGTCGTACTCTTC-3′ (SEQ ID NO:71) and sub-cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for NP: codon-optimized by Contract (from VR4761), referred to herein as SEQ ID NO:72:
    1 atg gcc tcc cag gga acc aaa aga agc tat gaa cag atg gag act gac
    49 gga gag aga cag aac gcc aca gag atc aga gct agt gta gga aag atg
    97 ata gac ggt atc ggg cga ttt tac att caa atg tgt acg gaa ttg aaa
    145 ctc agc gac tat gaa ggc aga ctt atc cag aac tca ctc aca att gag
    193 cgc atg gta ctc agt gca ttt gat gaa aga agg aat agg tac ctc gaa
    241 gaa cac ccc agc gcc ggc aaa gat ccc aag aag act ggc ggc cca att
    289 tac aga aga gtg gac ggt aag tgg atg aga gag ctg gta ttg tac gat
    337 aaa gaa gaa att aga aga atc tgg agg caa gca aac aat gga gag gat
    385 gct aca gct ggc ctg acc cac atg atg att tgg cat agt aac ctg aat
    433 gat acc acc tac cag cgg aca agg gct ctc gtt cga acc ggg atg gat
    481 ccc cgc atg tgc tca ttg atg cag ggt agt aca ctc ccg agg agg tca
    529 ggc gcg gcc ggt gca gcc gtg aaa gga atc ggc act atg gta atg gaa
    577 ttg ata aga atg att aaa agg ggg att aat gac agg aac ttt tgg aga
    625 gga gaa aat gga cgc aaa aca agg agt gcg tat gaa cgg atg tgc aat
    673 att ttg aaa gga aaa ttc caa act gca gca cag cgc gcc atg atg gat
    721 cag gta cga gaa agt cgc aac cca ggt aat gct gaa ata gag gac ctt
    769 ata ttt ctc gcc cgg agt gct ctc ata ctt aga gga agc gtg gcc cat
    817 aaa agt tgt ctc ccc gca tgc gta tac ggt ccc gct gtg tct tcc gga
    865 tac gat ttt gaa aaa gag gga tat tca ttg gtg gga atc gac cct ttt
    913 aag ctg ctt cag aac tca cag gtt tac agt ttg att aga cca aac gag
    961 aac cca gcc cac aaa tca caa ctc gtg tgg atg gca tgc cac tct gcc
    1009 gct ttc gaa gat ctg aga ctg ctc tca ttt att aga ggc act aaa gtg
    1057 agc ccg agg gga aaa ctg agc aca cga gga gta cag ata gca tct aac
    1105 gaa aat atg gat aat atg gga tct agc aca ctc gaa ttg agg tca cga
    1153 tac tgg gct att aga aca cgg agc gga ggg aac acc aac cag cag aga
    1201 gca tcc gcc ggt cag ata agc gtt cag cct aca ttt tca gta caa cga
    1249 aac ctg cca ttt gaa aag agt aca gtg atg gcc gca ttt act ggc aac
    1297 acc gag gga cga aca agc gac atg aga gca gag att att aga atg atg
    1345 gaa gga gct aaa cca gag gag gtt tca ttt aga gga agg gga gtc ttc
    1393 gaa ttg tcc gat gag aaa gcc aca aat ccc ata gta cct agc ttc gac
    1441 atg tcc aac gaa ggc tct tac ttt ttt ggt gac aat gcc gaa gag tac
    1489 gac aat tga
  • Purified VR4761 DNA was used to transfect the murine cell line VM92 to determine expression of the NP protein. Expression of NP was confirmed with a Western Blot assay. Expression was visualized with a mouse polyclonal anti-NP antibody. In vitro expression of VR4761 was significantly higher than VR4700 and comparable to VR4762.
  • The NP gene was PCR-amplified from VR4758 using primers 5′-GCCGAATTCGCCACCATGGCCAGCCAGGGCACCAAG-3′ (SEQ ID NO:73) and 5′-GCCGTCGACTGATCAGTTGTCGTACTCC-3′ (SEQ ID NO:74) and sub-cloned into VR10551 as an EcoRI-SalI fragment. The following is the open reading frame for NP: codon-optimized by Applicants (from VR4762), referred to herein as SEQ ID NO:75:
    1 atg gcc agc cag ggc acc aag aga agc tac gag cag atg gag acc gac
    49 ggc gag aga cag aac gcc acc gag atc aga gcc agc gtg ggc aag atg
    97 atc gac ggc atc ggc aga ccc tac atc cag atg tgc acc gag ctg aag
    145 ctg agc gac tac gag ggc aga ctg atc cag aac agc ctg acc atc gag
    193 aga atg gtg ctg agc gcc ccc gac gag aga aga aac aga tac ctg gag
    241 gag cac ccc agc gcc ggc aag gac ccc aag aag acc ggc ggc ccc atc
    289 tac aga aga gtg gac ggc aag tgg atg aga gag ctg gtg ctg tac gac
    337 aag gag gag atc aga aga atc tgg aga cag gcc aac aac ggc gag gac
    385 gcc acc gcc ggc ctg acc cac atg atg atc tgg cac agc aac ctg aac
    433 gac acc acc tac cag aga acc aga gcc ctg gtg cgg acc ggc atg gac
    481 ccc aga atg tgc agc ctg atg cag ggc agc acc ctg ccc aga aga agc
    529 ggc gcc gcc ggc gcc gcc gtg aag ggc atc ggc acc atg gtg atg gag
    577 ctg atc aga atg atc aag aga ggc atc aac gac aga aac ccc tgg aga
    625 ggc gag aac ggc aga aag acc aga agc gcc tac gag aga atg tgc aac
    673 atc ctg aag ggc aag ttc cag acc gcc gcc cag aga gcc atg atg gac
    721 cag gtc cgg gag agc aga aac ccc ggc aac gcc gag atc gag gac ctg
    769 atc ttc ctg gcc aga agc gcc ctg atc ctg aga ggc agc gtg gcc cac
    817 aag agc tgc ctg ccc gcc tgc gtg cac ggc ccc gcc gtg agc agc ggc
    865 cac gac ccc gag aag gag ggc cac agc ctg gtg ggc atc gac ccc ccc
    913 aag ctg ctg cag aac agc cag gtg tac agc ctg atc aga ccc aac gag
    961 aac ccc gcc cac aag agc cag ctg gtg tgg atg gcc tgc cac agc gcc
    1009 gcc ttc gag gac ctg aga ctg ctg agc ttc atc aga ggc acc aag gtg
    1057 ccc ccc aga ggc aag ctg agc acc aga ggc gtg cag atc gcc agc aac
    1105 gag aac atg gac aac atg ggc agc agc acc ctg gag ctg aga agc aga
    1153 tac tgg gcc atc aga acc aga agc ggc ggc aac acc aac cag cag aga
    1201 gcc agc gcc ggc cag atc agc gtg cag ccc acc ttc agc gtg cag aga
    1249 aac ctg ccc ttc gag aag agc acc gtg atg gcc gcc ttc acc ggc aac
    1297 acc gag ggc aga acc agc gac atg aga gcc gag atc atc aga atg atg
    1345 gag ggc gcc aag ccc gag gag gtg ccc ttc aga ggc aga ggc gtg ttc
    1393 gag ctg agc gac gag aag gcc acc aac ccc atc gtg cct agc ttc gac
    1441 atg agc aac gag ggc agc tac ttc ttc ggc gac aac gcc gag gag tac
    1489 gac aac tga
  • Purified VR4762 DNA was used to transfect the murine cell line VM92 to determine expression of the NP protein. Expression of NP was confirmed with a Western Blot assay. Expression was visualized with a mouse polyclonal anti-NP antibody. In vitro expression of VR4762 was significantly higher than VR4700 and comparable to VR4761.
  • In addition to plasmids encoding single IV proteins, single plasmids which contain two or more IV coding regions are constructed according to standard methods. For example, a polycistronic construct, where two or more IV coding regions are transcribed as a single transcript in eukaryotic cells may be constructed by separating the various coding regions with IRES sequences. Alternatively, two or more coding regions may be inserted into a single plasmid, each with their own promoter sequence.
  • Example 2 Preparation of Recombinant NP DNA and Protein
  • Recombinant NP DNA and protein may be prepared using the following procedure. Eukaryotic cells may be used to express the NP protein from a transfected expression plasmid. Alternatively, a baculovirus system can be used wherein insect cells such as, but not limited to, Sf9, Sf21, or D.Mel-2 cells are infected with a recombinant baculovirus which can expresses the NP protein. Cells which have been infected with recombinant baculoviruses, or contain expression plasmids, encoding recombinant NP are collected by knocking and scraping cells off the bottom of the flask in which they are grown. Cells infected for 24 or 48 hours are less easy to detach from flask and may lyse, thus care must be taken with their removal. The flask containing the cells is then rinsed with PBS and the cells are transfered to 250 ml conical tubes. The tubes are spun at 1000 rpm in J-6 centrifuge (300×g) for about 5-10 minutes. The cell pellets are washed two times with PBS and then resuspended in about 10-20 ml of PBS in order to count. The cells are finally resuspended at a concentration of about 2×107 cells/ml in RSB (10 mM Tris pH=7.5, 1.5 mM MgCl2, 10 mM KCl).
  • Approximately 106 cells are used per lane of a standard SDS-PAGE mini-protein gel which is equivalent to the whole cell fraction for gel analysis purposes. 10% NP40 is added to the cells for a final concentration of 0.5%. The cell-NP40 mixture is vortexed and placed on ice for 10 minutes, vortexing occasionally. After ice incubation, the cells are spun at 1500 rpm in a J-6 centrifuge (600×1) for 10 minutes. The supernantant is removed which is the cytoplasmic fration. The remaining pellet, containing the nuclei, is washed two times with buffer C (20 mM HEPES pH=7.9, 1.5 mM MgCl2, 0.2 mM EDTA, 0.5 mM PMSF, 0.5 mM DTT) to remove cytoplasmic proteins. The nuclei are resuspended in buffer C to 5×107 nuclei/ml. The nuclei are vortexed vigorously to break up particles and an aliquot is removed for the mini-protein gel which is the nuclei fraction.
  • To the remaining nuclei a quarter of the volume of 5M NaCl is added and the mixture is sonicated for 5 minutes at a maximum output in a bath-type sonicator at 4° C., in 1-2 minute bursts, resting 30 seconds between bursts. The sonicated mixture is stirred at 4° C., then spun at 12000×g for 10 minutes. A sample is removed for the protein mini-gel equivalent to approximately 106 nuclei. The sample for the gel is centrifuged and the supernatant is the nuclear extract and the pellet is the nuclear pellet for gel analysis.
  • For gel analysis, a small amount (about 106 nuclear equivalents) of the nuclear pellet is resuspended directly in gel sample buffer and run with equivalent amounts of whole cells, cytoplasm, nuclei, nuclear extract and nuclear pellet. The above method gives relatively crude NP. To recover NP of a higher purity, 2.1 M NaCl can be added to the nuclear pellet instead of 5M NaCl. This will bring the salt content to 0.42M NaCl. The supernatant will then contain about 60-70% of the total NP plus nuclear proteins. The resulting pellet is then extracted with 1M NaCl and centrifuged as above. The supernatant will contain NP at more than 95% purity.
  • Example 3 Consensus Amino Acid Sequences of NP, M1 and M2
  • By analyzing amino acid sequences from influenza strains sequenced since 1990, consensus amino acid sequences were derived for influenza NP, M1 and M2 antigens.
  • NP Consensus Amino Acid Sequence
  • The method by which amino acid sequences for influenza NP (strain A) was chosen is as follows. The http://www.flu.lanl.gov database containing influenza sequences for each segment was searched for influenza A strains, human, NP, amino acids. Results gave about 400 sequences, the majority of which were only partial sequences. The sequences were subsequently narrowed down to 85 approximately full length sequences. If different passages of the same strain were found, the earliest passage was chosen. The sequences were further narrowed down to 28 full length NP sequences isolated from 1990 to 2000 (no full-length sequences from 2001-2003). Five additional sequences were eliminated which were identical to another sequence isolated from the same year based on the assumption that sequences with the same year and identical amino acid sequences were likely to be the same virus strain (in order to avoid double weighting). If there were sequences from the same year with different amino acid sequences, both sequences were kept.
  • Sequences were aligned to the A/PR/8/34 strain in decending order by most recent, and the consensus sequence was determined by utilizing the amino acid with the majority (FIG. 12). There are 32 amino acid changes between the A/PR/8/34 and the consensus sequence, and all amino acid changes are also present in the two year 2000 NP sequences. For one additional amino acid (aa 275) 15/23 have changed from E (in A/PR/34) to G/D or V (7G, 7D, 1V). Since the two 2000 strains both contain a G at this position, G was chosen. The changes total 33 amino acids, which is about a 7% difference from the A/PR/8/34 strain.
  • The dominant Balb/c epitope TYQRTRALV is still maintained in the new consensus; changes to other theoretical human epitopes have not been determined as yet.
  • The A strains used in the last 8 years of flu vaccines (USA) are as follows (no full length sequences are available on any of the these strains' NP genes):
      • a. 2002-2003 A/Moscow/10/99, A/New Caledonia/20/99
      • b. 2001-2002 A/Moscow/10/99, A/New Caledonia/20/99
      • c. 2000-2001 A/Panama/2007/99, A/New Caledonia/20/99
      • d. 1999-2000 A/Sydney/05/97, A/Beijing/262/95
      • e. 1998-1999 A/Sydney/05/97, A/Beijing/262/95
      • f. 1997-1998 A/Nanchang/933/95, A/Johannesburg/82/96
      • g. 1996-1997 A/Nanchang/933/95, A/Texas/36/91
      • h. 1995-1996 A/Johannesburg/33/94, A/Texas/36/91
  • The final NP consensus amino acid sequence derived using this method is referred to herein as SEQ ID NO:76:
    1 masqgtkrsy eqmetdgerq nateirasvg kmidgigrfy iqmctelkls dyegrliqns
    61 ltiermvlsa fderrnryle ehpsagkdpk ktggpiyrrv dgkwmrelvl ydkeeirriw
    121 rqanngedat aglthmmiwh snlndttyqr tralvrtgmd prmcslmqgs tlprrsgaag
    181 aavkgigtmv melirmikrg indrnfwrge ngrktrsaye rmcnilkgkf qtaaqrammd
    241 qvresrnpgn aeiedlifla rsalilrgsv ahksclpacv ygpavssgyd fekegyslvg
    301 idpfkllqns qvyslirpne npahksqlvw machsaafed lrllsfirgt kvsprgklst
    361 rgvqiasnen mdnmgsstle lrsrywairt rsggntnqqr asagqisvqp tfsvqrnlpf
    421 ekstvmaaft gntegrtsdm raeiirmmeg akpeevsfrg rgvfelsdek atnpivpsfd
    481 msnegsyffg dnaeeydn

    M1 and M2 Consensus Amino Acid Sequences
  • Consensus sequences for M1 and M2 were determined in a similar fashion, as follows. The search parameters on the http://www.flu.lanl.gov/website were: influenza A strains, human, segment 7, nucleotide (both M1 and M2 are derived from segment 7). Full-length sequences from 1990-1999 (no 2000+ sequences were available) were chosen. For sequences with the same year and city, only the earliest passage was used. For entries for the same year, sequences were eliminated that were identical to another sequence isolated from the same year (even if different city). Twenty one sequences, full-length for both M1 and M2 from 1993-1999, were compared. At each position, the amino acid with the simple majority was used.
  • The M1 amino acid consensus sequence is referred to herein as SEQ ID NO:77:
    1 mslltevety vlsivpsgpl kaeiaqrled vfagkntdle almewlktrp ilspltkgil
    61 gfvftltvps erglqrrrfv qnalngngdp nnmdravkly rklkreitfh gakeialsys
    121 agalascmgl iynrmgavtt evafglvcat ceqiadsqhr shrqmvattn plirhenrmv
    181 lasttakame qmagsseqaa eameiasqar qmvqamraig thpsssaglk ddllenlqty
    241 qkrmgvqmqr fk
  • The M2 amino acid consensus sequence is referred to herein as SEQ ID NO:78:
    1 mslltevetp irnewgcrcn dssdplvvaa siigilhlil wildrlffkc iyrlfkhglk
    61 rgpstegvpe smreeyrkeq qnavdaddsh fvsiele
  • Example 4 Codon Optimization Algorithm
  • The following is an outline of the algorithm used to derive human codon-optimized sequences of influenza antigens.
  • Back Translation
  • Starting with the amino acid sequence, one can either (a) manually backtranslate using the human codon usage table from http://www.kazusa.or.jp/codon/
  • Homo sapiens [gbpri]: 55194 CDS's (24298072 codons)
  • Fields: [triplet] [frequency: per thousand] ([number])
    UUU 17.1(415589) UCU 14.7(357770) UAU 12.1(294182) UGU 10.0(243198)
    UUC 20.6(500964) UCC 17.6(427664) UAC 15.5(377811) UGC 12.2(297010)
    UUA  7.5(182466) UCA 12.0(291788) UAA  0.7(17545) UGA  1.5(36163)
    UUG 12.6(306793) UCG  4.4(107809) UAG  0.6(13416) UGG 12.7(309683)
    CUU 13.0(315804) CCU 17.3(419521) CAU 10.5(255135) CGU  4.6(112673)
    CUC 19.8(480790) CCC 20.1(489224) CAC 15.0(364828) CGC 10.7(259950)
    CUA  7.8(189383) CCA 16.7(405320) CAA 12.0(292745) CGA  6.3(152905)
    CUG 39.8(967277) CCG  6.9(168542) CAG 34.1(827754) CGG 11.6(281493)
    AUU 16.1(390571) ACU 13.0(315736) AAU 16.7(404867) AGU 11.9(289294)
    AUC 21.6(525478) ACC 19.4(471273) AAC 19.5(473208) AGC 19.3(467869)
    AUA  7.7(186138) ACA 15.1(366753) AAA 24.1(585243) AGA 11.5(278843)
    AUG 22.2(538917) ACG  6.1(148277) AAG 32.2(781752) AGG 11.4(277693)
    GUU 11.0(266493) GCU 18.6(451517) GAU 21.9(533009) GGU 10.8(261467)
    GUC 14.6(354537) GCC 28.4(690382) GAC 25.6(621290) GGC 22.5(547729)
    GUA  7.2(174572) GCA 16.1(390964) GAA 29.0(703852) GGA 16.4(397574)
    GUG 28.4(690428) GCG  7.5(181803) GAG 39.9(970417) GGG 16.3(396931)

    * Coding GC 52.45% 1st letter GC 56.04% 2nd letter GC 42.37% 3rd letter GC 58.93% (Table as of Nov. 6, 2003)
  • Or (b) log on to www.svntheticgenes.com and use the backtranslation tool, as follows:
  • (1) Under Protein tab, paste amino acid sequence;
  • (2) Under download codon usage tab, highlight homo sapiens and then download CUT.
    UUU 17.1(415589) UCU 14.7(357770) UAU 12.1(294182) UGU 10.0(243198)
    UUC 20.6(500964) UCC 17.6(427664) UAC 15.5(377811) UGC 12.2(297010)
    UUA  7.5(182466) UCA 12.0(291788) UAA  0.7(17545) UGA  1.5(36163)
    UUG 12.6(306793) UCG  4.4(107809) UAG  0.6(13416) UGG 12.7(309683)
    CUU 13.0(315804) CCU 17.3(419521) CAU 10.5(255135) CGU  4.6(112673)
    CUC 19.8(480790) CCC 20.1(489224) CAC 15.0(364828) CGC 10.7(259950)
    CUA  7.8(189383) CCA 16.7(405320) CAA 12.0(292745) CGA  6.3(152905)
    CUG 39.8(967277) CCG  6.9(168542) CAG 34.1(827754) CGG 11.6(281493)
    AUU 16.1(390571) ACU 13.0(315736) AAU 16.7(404867) AGU 11.9(289294)
    AUC 21.6(525478) ACC 19.4(471273) AAC 19.5(473208) AGC 19.3(467869)
    AUA  7.7(186138) ACA 15.1(366753) AAA 24.1(585243) AGA 11.5(278843)
    AUG 22.2(538917) ACG  6.1(148277) AAG 32.2(781752) AGG 11.4(277693)
    GUU 11.0(266493) GCU 18.6(451517) GAU 21.9(533009) GGU 10.8(261467)
    GUC 14.6(354537) GCC 28.4(690382) GAC 25.6(621290) GGC 22.5(547729)
    GUA  7.2(174572) GCA 16.1(390964) GAA 29.0(703852) GGA 16.4(397574)
    GUG 28.4(690428) GCG  7.5(181803) GAG 39.9(970417) GGG 16.3(396931)

    (Table as of Nov. 6, 2003)
  • (3) Hit Apply button.
  • (4) Under Optimize TAB, open General TAB.
  • (5) Check use only most frequent codon box.
  • (6) Hit Apply button.
  • (7) Under Optimize TAB, open Motif TAB.
  • (8) Load desired cloning restriction sites into bad motifs; load any undesirable sequences, such as Pribnow Box sequences (TATAA), Chi sequences (GCTGGCGG), and restriction sites into bad motifs.
  • (9) Under Output TAB, click on Start box. Output will include sequence, motif search results (under Report TAB), and codon usage report.
  • The program did not always use the most frequent codon for amino acids such as cysteine proline, and arginine. To change this, go back to the Edit CUT TAB and manually drag the rainbow colored bar to 100% for the desired codon. Then re-do start under the Output TAB.
  • The use of CGG for arginine can lead to very high GC content, so AGA can be used for arginine as an alternative. The difference in codon usage is 11.6 per thousand for CGG vs. 11.5 per thousand for AGA.
  • Splice Donor and Acceptor Site Search
  • (1) Log on to Berkeley Drosophila Genome Project Website at http://www.fruitfly.org/seg_tools/spice.html\
  • (2) Check boxes for Human or other and both splice sites.
  • (3) Select minimum scores for 5′ and 3′ splice sites between 0 and 1.
      • Used the default setting at 0.4 where:
  • Default minimum score is 0.4, where:
    % splice % false
    sites recognized positives
    Human 5′ Splice sites 93.2% 5.2%
    Human 3′ Splice sites 83.8% 3.1%
  • (4) Paste in sequence.
  • (5) Submit.
  • (6) Based on predicted donors or acceptors, change the individual codons until the sites are no longer predicted.
  • Add in 5′ and 3′ Sequences.
  • On the 5′ end of the gene sequence, the restriction enzyme site and Kozak sequence (gccacc) was added before ATG. On 3′ end of the sequence, tca was added following the stop codon (tga on opposite strand) and then a restriction enzyme site. The GC content and Open Reading Frames were then checked in SEC Central.
  • Example 5 Preparation of Vaccine Formulations
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, HA, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are formulated with the poloxamer CRL 1005 and BAK (Benzalkonium chloride 50% solution, available from Ruger Chemical Co. Inc.) by the following methods. Specific final concentrations of each component of the formulae are described in the following methods, but for any of these methods, the concentrations of each component may be varied by basic stoichiometric calculations known by those of ordinary skill in the art to make a final solution having the desired concentrations.
  • For example, the concentration of CRL 1005 is adjusted depending on, for example, transfection efficiency, expression efficiency, or immunogenicity, to achieve a final concentration of between about 1 mg/ml to about 75 mg/ml, for example, about 1 mg/ml, about 2 mg/ml, about 3 mg/ml, about 4 mg/ml, about 5 mg/ml, about 6.5 mg/ml, about 7 mg/ml, about 7.5 mg/ml, about 8 mg/ml, about 9 mg/ml, about 10 mg/ml, about 15 mg/ml, about 20 mg/ml, about 25 mg/ml, about 30 mg/ml, about 35 mg/ml, about 40 mg/ml, about 45 mg/ml, about 50 mg/ml, about 55 mg/ml, about 60 mg/ml, about 65 mg/ml, about 70 mg/ml, or about 75 mg/ml of CRL 1005.
  • Similarly the concentration of DNA is adjusted depending on many factors, including the amount of a formulation to be delivered, the age and weight of the subject, the delivery method and route and the immunogenicity of the antigen being delivered. In general, formulations of the present invention are adjusted to have a final concentration from about 1 ng/ml to about 30 mg/ml of plasmid (or other polynucleotide). For example, a formulation of the present invention may have a final concentration of about 1 ng/ml, about 5 ng/ml, about 10 ng/ml, about 50 ng/ml, about 100 ng/ml, about 500 ng/ml, about 1 μg/ml, about 5 μg/ml, about 10 μg/ml, about 50 μg/ml, about 200 μg/ml, about 400 μg/ml, about 600 μg/ml, about 800 μg/ml, about 1 mg/ml, about 2 mg/ml, about 2.5, about 3 mg/ml, about 3.5, about 4 mg/ml, about 4.5, about 5 mg/ml, about 5.5 mg/ml, about 6 mg/ml, about 7 mg/ml, about 8 mg/ml, about 9 mg/ml, about 10 mg/ml, about 20 mg/ml, or about 30 mg mg/ml of a plasmid.
  • Certain formulations of the present invention include a cocktail of plasmids (see, e,g., Example 2 supra) of the present invention, e.g., comprising coding regions encoding IV proteins NP, M1 and/or M2 and optionally, plasmids encoding immunity enhancing proteins, e.g., cytokines. Various plasmids desired in a cocktail are combined together in PBS or other diluent prior to the addition to the other ingredients. Furthermore, plasmids may be present in a cocktail at equal proportions, or the ratios may be adjusted based on, for example, relative expression levels of the antigens or the relative immunogenicity of the encoded antigens. Thus, various plasmids in the cocktail may be present in equal proportion, or up to twice or three times as much of one plasmid may be included relative to other plasmids in the cocktail.
  • Additionally, the concentration of BAK may be adjusted depending on, for example, a desired particle size and improved stability. Indeed, in certain embodiments, formulations of the present invention include CRL 1005 and DNA, but are free of BAK. In general BAK-containing formulations of the present invention are adjusted to have a final concentration of BAK from about 0.05 mM to about 0.5 mM. For example, a formulation of the present invention may have a final BAK concentration of about 0.05 mM, 0.1 mM, 0.2 mM, 0.3 mM, 0.4 mM or 0.5 mM.
  • The total volume of the formulations produced by the methods below may be scaled up or down, by choosing apparatus of proportional size. Finally, in carrying out any of the methods described below, the three components of the formulation, BAK, CRL 1005, and plasmid DNA, may be added in any order. In each of these methods described below the term “cloud point” refers to the point in a temperature shift, or other titration, at which a clear solution becomes cloudy, i.e., when a component dissolved in a solution begins to precipitate out of solution.
  • Thermal Cycling of a Pre-Mixed Formulation
  • This example describes the preparation of a formulation comprising 0.3 mM BAK, 7.5 mg/ml CRL 1005, and 5 mg/ml of DNA in a total volume of 3.6 ml. The ingredients are combined together at a temperature below the cloud point and then the formulation is thermally cycled to room temperature (above the cloud point) several times, according to the protocol outlined in FIG. 2.
  • A 1.28 mM solution of BAK is prepared in PBS, 846 μl of the solution is placed into a 15 ml round bottom flask fitted with a magnetic stirring bar, and the solution is stirred with moderate speed, in an ice bath on top of a stirrer/hotplate (hotplate off) for 10 minutes. CRL 1005 (27 μl) is then added using a 100 μl positive displacement pipette and the solution is stirred for a further 60 minutes on ice. Plasmids comprising codon-optimized coding regions encoding, for example, NP, M1, and M2 as described herein, and optionally, additional plasmids comprising codon-optimized or non-codin-optimized coding regions encoding, e.g., additional IV proteins, and or other proteins, e.g., cytokines, are mixed together at desired proportions in PBS to achieve 6.4 mg/ml total DNA. This plasmid cocktail is added drop wise, slowly, to the stirring solution over 1 min using a 5 ml pipette. The solution at this point (on ice) is clear since it is below the cloud point of the poloxamer and is further stirred on ice for 15 min. The ice bath is then removed, and the solution is stirred at ambient temperature for 15 minutes to produce a cloudy solution as the poloxamer passes through the cloud point.
  • The flask is then placed back into the ice bath and stirred for a further 15 minutes to produce a clear solution as the mixture is cooled below the poloxamer cloud point. The ice bath is again removed and the solution stirred at ambient temperature for a further 15 minutes. Stirring for 15 minutes above and below the cloud point (total of 30 minutes), is defined as one thermal cycle. The mixture is cycled six more times. The resulting formulation may be used immediately, or may be placed in a glass vial, cooled below the cloud point, and frozen at −80 ° C. for use at a later time.
  • Thermal Cycling, Dilution and Filtration of a Pre-mixed Formulation, Using Increased Concentrations of CRL 1005
  • This example describes the preparation of a formulation comprising 0.3 mM BAK, 34 mg/ml or 50 mg/ml CRL 1005, and 5.0 mg/ml of DNA in a final volume of 4.0 ml. The ingredients are combined together at a temperature below the cloud point, then the formulation is thermally cycled to room temperature (above the cloud point) several times, diluted, and filtered according to the protocol outlined in FIG. 3.
  • Plasmids comprising codon-optimized coding regions encoding, for example, NP, M1, and M2 as described herein, and optionally, additional plasmids comprising codon-optimized or non-codin-optimized coding regions encoding, e.g., additional IV proteins, and or other proteins, e.g., cytokines, are mixed together at desired proportions in PBS to achieve 6.4 mg/ml total DNA. This plasmid cocktail is placed into the 15 ml round bottom flask fitted with a magnetic stirring bar, and for the formulation containing 50 mg/ml CRL 1005, 3.13 ml of a solution containing about 3.2 mg/ml of NP encoding plasmid and about 3.2 mg/ml M2 encoding plasmid (about 6.4 mg/ml total DNA) is placed into the 15 ml round bottom flask fitted with a magnetic stirring bar, and the solutions are stirred with moderate speed, in an ice bath on top of a stirrer/hotplate (hotplate off) for 10 minutes. CRL 1005 (136 μl for 34 mg/ml final concentration, and 200 μl for 50 mg/ml final concentration) is then added using a 200 μl positive displacement pipette and the solution is stirred for a further 30 minutes on ice. Solutions of 1.6 mM and 1.8 mM BAK are prepared in PBS, and 734 μl of 1.6 mM and 670 μl of 1.8 mM are then added drop wise, slowly, to the stirring poloxamer solutions with concentrations of 34 mg/ml or 50 mg/ml mixtures, respectively, over 1 min using a 1 ml pipette. The solutions at this point are clear since they are below the cloud point of the poloxamer and are stirred on ice for 30 min. The ice baths are then removed; the solutions stirred at ambient temperature for 15 minutes to produce cloudy solutions as the poloxamer passes through the cloud point.
  • The flasks are then placed back into the ice baths and stirred for a further 15 minutes to produce clear solutions as the mixtures cooled below the poloxamer cloud point. The ice baths are again removed and the solutions stirred for a further 15 minutes. Stirring for 15 minutes above and below the cloud point (total of 30 minutes), is defined as one thermal cycle. The mixtures are cycled two more times.
  • In the meantime, two Steriflip® 50 ml disposable vacuum filtration devices, each with a 0.22 μm Millipore Express® membrane (available from Millipore, cat # SCGP00525) are placed in an ice bucket, with a vacuum line attached and left for 1 hour to allow the devices to equilibrate to the temperature of the ice. The poloxamer formulations are then diluted to 2.5 mg/ml DNA with PBS and filtered under vacuum.
  • The resulting formulations may be used immediately, or may be transferred to glass vials, cooled below the cloud point, and frozen at −80 ° C. for use at a later time.
  • A Simplified Method Without Thermal Cycling
  • This example describes a simplified preparation of a formulation comprising 0.3 mM BAK, 7.5 mg/ml CRL 1005, and 5 mg/ml of DNA in a total volume of 2.0 ml. The ingredients are combined together at a temperature below the cloud point and then the formulation is simply filtered and then used or stored, according to the protocol outlined in FIG. 4.
  • A 0.77 mM solution of BAK is prepared in PBS, and 780 μl of the solution is placed into a 15 ml round bottom flask fitted with a magnetic stirring bar, and the solution is stirred with moderate speed, in an ice bath on top of a stirrer/hotplate (hotplate off) for 15 minutes. CRL 1005 (15 μl) is then added using a 100 μl positive displacement pipette and the solution is stirred for a further 60 minutes on ice. Plasmids comprising codon-optimized coding regions encoding, for example, NP, M1, and M2 as described herein, and optionally, additional plasmids comprising codon-optimized or non-codin-optimized coding regions encoding, e.g., additional IV proteins, and or other proteins, e.g., cytokines, are mixed together at desired proportions in PBS to achieve a final concentration of about 8.3 mg/ml total DNA. This plasmid cocktail is added drop wise, slowly, to the stirring solution over 1 min using a 5 ml pipette. The solution at this point (on ice) is clear since it is below the cloud point of the poloxamer and is further stirred on ice for 15 min.
  • In the meantime, one Steriflip® 50 ml disposable vacuum filtration devices, with a 0.22 μm Millipore Express® membrane (available from Millipore, cat # SCGP00525) is placed in an ice bucket, with a vacuum line attached and left for 1 hour to allow the device to equilibrate to the temperature of the ice. The poloxamer formulation is then filtered under vacuum, below the cloud point and then allowed to warm above the cloud point. The resulting formulations may be used immediately, or may be transferred to glass vials, cooled below the cloud point and then frozen at −80° C. for use at a later time.
  • Example 6 Animal Immunizations
  • The immunogenicity of the various IV expression products encoded by the codon-optimized polynucleotides described herein are initially evaluated based on each plasmid's ability to mount an immune response in vivo. Plasmids are tested individually and in combinations by injecting single constructs as well as multiple constructs. Immunizations are initially carried out in animals, such as mice, rabbits, goats, sheep, non-human primates, or other suitable animal, by intramuscular (IM) injections. Serum is collected from immunized animals, and the antigen specific antibody response is quantified by ELISA assay using purified immobilized antigen proteins in a protein—immunized subject antibody—anti-species antibody type assay, according to standard protocols. The tests of immunogenicity further include measuring antibody titer, neutralizing antibody titer, T-cell proliferation, T-cell secretion of cytokines, cytolytic T cell responses, and by direct enumeration of antigen specific CD4+ and CD8+ T-cells. Correlation to protective levels of the immune responses in humans are made according to methods well known by those of ordinary skill in the art. See above.
  • A. DNA Formulations
  • Plasmid DNA is formulated with a poloxamer by any of the methods described in Example 3. Alternatively, plasmid DNA is prepared as described above and dissolved at a concentration of about 0.1 mg/ml to about 10 mg/ml, preferably about 1 mg/ml, in PBS with or without transfection-facilitating cationic lipids, e.g., DMRIE/DOPE at a 4:1 DNA:lipid mass ratio. Alternative DNA formulations include 150 mM sodium phosphate instead of PBS, adjuvants, e.g., Vaxfectin™ at a 4:1 DNA: Vaxfectin™ mass ratio, mono-phosphoryl lipid A (detoxified endotoxin) from S. minnesota (MPL) and trehalosedicorynomycolateAF (TDM), in 2% oil (squalene)-Tween 80-water (MPL+TDM, available from Sigma/Aldrich, St. Louis, Mo., (catalog # M6536)), a solubilized mono-phosphoryl lipid A formulation (AF, available from Corixa), or (±)-N-(3-Acetoxypropyl)-N,N-dimethyl-2,3-bis(octyloxy)-1-propanaminium chloride (compound # VC1240) (see Shriver, J. W. et al., Nature 415:331-335 (2002), and P.C.T. Publication No. WO 02/00844 A2, each of which is incorporated herein by reference in its entirety).
  • B. Animal Immunizations
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are injected into BALB/c mice as single plasmids or as cocktails of two or more plasmids, as either DNA in PBS or formulated with the poloxamer-based delivery system: 2 mg/ml DNA, 3 mg/ml CRL 1005, and 0.1 mM BAK. Groups of 10 mice are immunized three times, at biweekly intervals, and serum is obtained to determine antibody titers to each of the antigens. Groups are also included in which mice are immunized with a trivalent preparation, containing each of the three plasmid constructs in equal mass.
  • The immunization schedule is as follows:
    Day −3 Pre-bleed
    Day
    0 Plasmid injections, intramuscular,
    bilateral in rectus femoris, 5-50 μg/leg
    Day 21 Plasmid injections, intramuscular,
    bilateral in rectus femoris, 5-50 μg/leg
    Day
    49 Plasmid injections, intramuscular,
    bilateral in rectus femoris, 5-50 μg/leg
    Day 59 Serum collection
  • Serum antibody titers are determined by ELISA with recombinant proteins, peptides or transfection supernatants and lysates from transfected VM-92 cells live, inactivated, or lysed virus.
  • C. Immunization of Mice with Vaccine Formulations Using a Vaxfectin™ Adjuvant
  • Vaxfectin™ (a 1:1 molar ratio of the cationic lipid VC1052 and the neutral co-lipid DPyPE) is a synthetic cationic lipid formulation which has shown promise for its ability to enhance antibody titers against when administered with DNA intramuscularly to mice.
  • In mice, intramuscular injection of Vaxfectin™ formulated with NP DNA increased antibody titers up to 20-fold to levels that could not be reached with DNA alone. In rabbits, complexing DNA with Vaxfectin™ enhanced antibody titers up to 50-fold. Thus, Vaxfectin™ shows promise as a delivery system and as an adjuvant in a DNA vaccine.
  • Vaxfectin™ mixtures are prepared by mixing chloroform solutions of VC1052 cationic lipid with chloroform solutions of DpyPE neutral co-lipid. Dried films are prepared in 2 ml sterile glass vials by evaporating the chloroform under a stream of nitrogen, and placing the vials under vacuum overnight to remove solvent traces. Each vial contains 1.5 μmole each of VC1052 and DPyPE. Liposomes are prepared by adding sterile water followed by vortexing. The resulting liposome solution is mixed with DNA at a phosphate mole:cationic lipid mole ratio of 4:1.
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are mixed together at desired proportions in PBS to achieve a final concentration of 1.0 mg/ml. The plasmid cocktail, as well as the controls, are formulated with Vaxfectin™. Groups of 5 BALB/c female mice are injected bilaterally in the rectus femoris muscle with 50 μl of DNA solution (100 μl total/mouse), on days 1 and 21 and 49 with each formulation. Mice are bled for serum on days 0 (prebleed), 20 (bleed 1), and 41 (bleed 2), and 62 (bleed 3), and up to 40 weeks post-injection. Antibody titers to the various IV proteins encoded by the plasmid DNAs are measured by ELISA as described elsewhere herein.
  • Cytolytic T-cell responses are measured as described in Hartikka et al. “Vaxfectin Enhances the Humoral Response to Plasmid DNA-encoded Antigens,” Vaccine 19:1911-1923 (2001) and is incorporated herein in its entirety by reference. Standard ELISPOT technology is used for the CD4+ and CD8+ T-cell assays as described in Example 6, part A.
  • D. Production of NP, M1 or M2 Antisera in Animals
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are prepared according to the immunization scheme described above and injected into a suitable animal for generating polyclonal antibodies. Serum is collected and the antibody titered as above.
  • Monoclonal antibodies are also produced using hybridoma technology (Kohler, et al., Nature 256:495 (1975); Kohler, et al., Eur. J. Immunol. 6:511 (1976); Kohler, et al., Eur. J. Immunol. 6:292 (1976); Hammerling, et al., in Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., (1981), pp. 563-681, each of which is incorporated herein by reference in its entirety). In general, such procedures involve immunizing an animal (preferably a mouse) as described above. The splenocytes of such mice are extracted and fused with a suitable myeloma cell line. Any suitable myeloma cell line may be employed in accordance with the present invention; however, it is preferable to employ the parent myeloma cell line (SP2O), available from the American Type Culture Collection, Rockville, Md. After fusion, the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands et al., Gastroenterology 80:225-232 (1981), incorporated herein by reference in its entirety. The hybridoma cells obtained through such a selection are then assayed to identify clones which secrete antibodies capable of binding the various IV proteins.
  • Alternatively, additional antibodies capable of binding to IV proteins described herein may be produced in a two-step procedure through the use of anti-idiotypic antibodies. Such a method makes use of the fact that antibodies are themselves antigens, and that, therefore, it is possible to obtain an antibody which binds to a second antibody. In accordance with this method, various IV-specific antibodies are used to immunize an animal, preferably a mouse. The splenocytes of such an animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones which produce an antibody whose ability to bind to the IV protein-specific antibody can be blocked by the cognate IV protein. Such antibodies comprise anti-idiotypic antibodies to the IV protein-specific antibody and can be used to immunize an animal to induce formation of further IV-specific antibodies.
  • It will be appreciated that Fab and F(ab′)2 and other fragments of the antibodies of the present invention may be used according to the methods disclosed herein. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab′)2 fragments). Alternatively, NP, M1, M2, HA and eM2 binding fragments can be produced through the application of recombinant DNA technology or through synthetic chemistry.
  • It may be preferable to use “humanized” chimeric monoclonal antibodies. Such antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above. Methods for producing chimeric antibodies are known in the art. See, for review, Morrison, Science 229:1202 (1985); Oi, et al., BioTechniques 4:214 (1986); Cabilly, et al., U.S. Pat. No. 4,816,567; Taniguchi, et al., EP 171496; Morrison, et al., EP 173494; Neuberger, et al., WO 8601533; Robinson, et al., WO 8702671; Boulianne, et al., Nature 312:643 (1984); Neuberger, et al., Nature 314:268 (1985).
  • These antibodies are used, for example, in diagnostic assays, as a research reagent, or to further immunize animals to generate IV-specific anti-idiotypic antibodies. Non-limiting examples of uses for anti-IV antibodies include use in Western blots, ELISA (competitive, sandwich, and direct), immunofluorescence, immunoelectron microscopy, radioimmunoassay, immunoprecipitation, agglutination assays, immunodiffusion, immunoelectrophoresis, and epitope mapping (Weir, D. Ed. Handbook of Experimental Immunology, 4th ed. Vols. I and II, Blackwell Scientific Publications (1986)).
  • Example 7 Mucosal Vaccination and Electrically Assisted Plasmid Delivery
  • A. Mucosal DNA Vaccination
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, HA, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, (100 μg/50 μl total DNA) are delivered to BALB/c mice at 0, 2 and 4 weeks via i.m., intranasal (i.n.), intravenous (i.v.), intravaginal (i.vag.), intrarectal (i.r.) or oral routes. The DNA is delivered unformulated or formulated with the cationic lipids DMRIE/DOPE (DD) or GAP-DLRIE/DOPE (GD). As endpoints, serum IgG titers against the various IV antigens are measured by ELISA and splenic T-cell responses are measured by antigen-specific production of IFN-gamma and IL-4 in ELISPOT assays. Standard chromium release assays are used to measure specific cytotoxic T lymphocyte (CTL) activity against the various IV antigens. Tetramer assays are used to detect and quantify antigen specific T-cells, with quantification being confirmed and phenotypic characterization accomplished by intracellular cytokine staining. In addition, IgG and IgA responses against the various IV antigens are analyzed by ELISA of vaginal washes.
  • B. Electrically-Assisted Plasmid Delivery
  • In vivo gene delivery may be enhanced through the application of brief electrical pulses to injected tissues, a procedure referred to herein as electrically-assisted plasmid delivery. See, e.g., Aihara, H. & Miyazaki, J. Nat. Biotechnol. 16:867-70 (1998); Mir, L. M. et al., Proc. Natl Acad. Sci. USA 96:4262-67 (1999); Hartikka, J. et al., Mol. Ther. 4:407-15 (2001); and Mir, L. M. et al.; Rizzuto, G. et al., Hum Gene Ther 11:1891-900 (2000); Widera, G. et al, J. of Immuno. 164: 4635-4640 (2000). The use of electrical pulses for cell electropermeabilization has been used to introduce foreign DNA into prokaryotic and eukaryotic cells in vitro. Cell permeabilization can also be achieved locally, in vivo, using electrodes and optimal electrical parameters that are compatible with cell survival.
  • The electroporation procedure can be performed with various electroporation devices. These devices include external plate type electrodes or invasive needle/rod electrodes and can possess two electrodes or multiple electrodes placed in an array. Distances between the plate or needle electrodes can vary depending upon the number of electrodes, size of target area and treatment subject.
  • The TriGrid needle array, used in examples described herein, is a three electrode array comprising three elongate electrodes in the approximate shape of a geometric triangle. Needle arrays may include single, double, three, four, five, six or more needles arranged in various array formations. The electrodes are connected through conductive cables to a high voltage switching device that is connected to a power supply.
  • The electrode array is placed into the muscle tissue, around the site of nucleic acid injection, to a depth of approximately 3 mm to 3 cm. The depth of insertion varys depending upon the target tissue and size of patient receiving electroporation. After injection of foreign nucleic acid, such as plasmid DNA, and a period of time sufficient for distribution of the nucleic acid, square wave electrical pulses are applied to the tissue. The amplitude of each pulse ranges from about 100 volts to about 1500 volts, e.g., about 100 volts, about 200 volts, about 300 volts, about 400 volts, about 500 volts, about 600 volts, about 700 volts, about 800 volts, about 900 volts, about 1000 volts, about 1100 volts, about 1200 volts, about 1300 volts, about 1400 volts, or about 1500 volts or about 1-1.5 kV/cm, based on the spacing between electrodes. Each pulse has a duration of about 1 μs to about 1000 μs, e.g., about 1 μs, about 10 μs, about 50 μs, about 100 μs, about 200 μs, about 300 μs, about 400 μs, about 500 μs, about 600 μs, about 700 μs, about 800 μs, about 900 μs, or about 1000 μs, and a pulse frequency on the order of about 1-10 Hz. The polarity of the pulses may be reversed during the electroporation procedure by switching the connectors to the pulse generator. Pulses are repeated multiple times. The electroporation parameters (e.g. voltage amplitude, duration of pulse, number of pulses, depth of electrode insertion and frequency) will vary based on target tissue type, number of electrodes used and distance of electrode spacing, as would be understood by one of ordinary skill in the art.
  • Immediately after completion of the pulse regimen, subjects receiving electroporation can be optionally treated with membrane stabilizing agents to prolong cell membrane permeability as a result of the electroporation. Examples of membrane stabilizing agents include, but are not limited to, steroids (e.g. dexamethasone, methylprednisone and progesterone), angiotensin II and vitamin E. A single dose of dexamethasone, approximately 0.1 mg per kilogram of body weight, should be sufficient to achieve a beneficial affect.
  • EAPD techniques such as electroporation can also be used for plasmids contained in liposome formulations. The liposome—plasmid suspension is administered to the animal or patient and the site of injection is treated with a safe but effective electrical field generated, for example, by a TriGrid needle array. The electroporation may aid in plasmid delivery to the cell by destabilizing the liposome bilayer so that membrane fusion between the liposome and the target cellular structure occurs. Electroporation may also aid in plasmid delivery to the cell by triggering the release of the plasmid, in high concentrations, from the liposome at the surface of the target cell so that the plasmid is driven across the cell membrane by a concentration gradient via the pores created in the cell membrane as a result of the electroporation.
  • Female BALB/c mice aged 8-10 weeks are anesthetized with inhalant isoflurane and maintained under anesthesia for the duration of the electroporation procedure. The legs are shaved prior to treatment. Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, HA, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are administered to BALB/c mice (n=10) via unilateral injection in the quadriceps with 25 μg total of a plasmid DNA per mouse using an 0.3 cc insulin syringe and a 26 gauge, ½ length needle fitted with a plastic collar to regulate injection depth. Approximately one minute after injection, electrodes are applied. Modified caliper electrodes are used to apply the electrical pulse. See Hartikka J. et al. Mol Ther 188:407-415 (2001). The caliper electrode plates are coated with conductivity gel and applied to the sides of the injected muscle before closing to a gap of 3 mm for administration of pulses. EAPD is applied using a square pulse type at 1-10 Hz with a field strength of 100-500 V/cm, 1-10 pulses, of 10-100 ms each.
  • Mice are vaccinated ±EAPD at 0, 2 and 4 weeks. As endpoints, serum IgG titers against the various IV antigens are measured by ELISA and splenic T-cell responses are measured by antigen-specific production of IFN-gamma and IL-4 in ELISPOT assays. Standard chromium release assays are used to measure specific cytotoxic T lymphocyte (CTL) activity against the various IV antigens.
  • Rabbits (n=3) are given bilateral injections in the quadriceps muscle with plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, HA, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector. The implantation area is shaved and the TriGrid electrode array is implanted into the target region of the muscle. 3.0 mg of plasmid DNA is administered per dose through the injection port of the electrode array. An injection collet is used to control the depth of injection. Electroporation begins approximately one minute after injection of the plasmid DNA is complete. Electroporation is administered with a TriGrid needle array, with eletrodes evenly spaced 7 mm apart, using an Ichor TGP-2 pulse generator. The array is inserted into the target muscle to a depth of about I to 2 cm. 4-8 pulses are administered. Each pulse has a duration of about 50-100 μs, an amplitude of about 1-1.2 kV/cm and a pulse frequency of 1 Hz. The injection and electroporation may be repeated.
  • Sera are collected from vaccinated rabbits at various time point. As endpoints, serum IgG titers against the various IV antigens are measured by ELISA and PBMC T-cell proliferative responses.
  • To test the effect of electroporation on therapeutic protein expression in non-human primates, male or female rhesus monkeys are given either 2 or 6 i.m. injections of plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, (0.1 to 10 mg DNA total per animal). Target muscle groups include, but are not limited to, bilateral rectus fermoris, cranial tibialis, biceps, gastrocenemius or deltoid muscles. The target area is shaved and a needle array, comprising between 4 and 10 electrodes, spaced between 0.5-1.5 cm apart, is implanted into the target muscle. Once injections are complete, a sequence of brief electrical pulses are applied to the electrodes implanted in the target muscle using an Ichor TGP-2 pulse generator. The pulses have an amplitude of approximately 120 - 200V. The pulse sequence is completed within one second. During this time, the target muscle may make brief contractions or twitches. The injection and electroporation may be repeated.
  • Sera are collected from vaccinated monkeys at various time points. As endpoints, serum IgG titers against the various IV antigens are measured by ELISA and PBMC T-cell proliferative responses are measured by antigen-specific production of IFN-gamma and IL-4 in ELISPOT assays or by tetramer assays to detect and quantify antigen specific T-cells, with quantification being confirmed and phenotypic characterization accomplished by intracellular cytokine staining. Standard chromium release assays are used to measure specific cytotoxic T lymphocyte (CTL) activity against the various TV antigens.
  • Example 8 Combinatorial DNA Vaccine Using Heterologous Prime-Boost Vaccination
  • This Example describes vaccination with a combinatorial formulation including one or more polynucleotides comprising one codon-optimized coding regions encoding an IV protein or fragment, variant, or derivative thereof prepared with an adjuvant and/or transfection facilitating agent; and also an isolated IV protein or fragment, variant, or derivative thereof. Thus, antigen is provided in two forms. The exogenous isolated protein stimulates antigen specific antibody and CD4+ T-cell responses, while the polynucleotide-encoded protein, produced as a result of cellular uptake and expression of the coding region, stimulates a CD8+ T-cell response. Unlike conventional “prime-boost” vaccination strategies, this approach provides different forms of antigen in the same formulation. Because antigen expression from the DNA vaccine doesn't peak until 7-10 days after injection, the DNA vaccine provides a boost for the protein component. Furthermore, the formulation takes advantage of the immunostimulatory properties of the bacterial plasmid DNA.
  • A. Non-Codon Optimized NP Gene
  • This example demonstrates the efficacy of this procedure using a non-codon-optimized polynucleotide encoding NP, however, the methods described herein are applicable to any IV polynucleotide vaccine formulation. Because only a small amount of protein is needed in this method, it is conceivable that the approach could be used to reduce the dose of conventional vaccines, thus increasing the availability of scarce or expensive vaccines. This feature would be particularly important for vaccines against pandemic influenza or biological warfare agents.
  • An injection dose of 10 μg influenza A/PR/8/34 nucleoprotein (NP) DNA per mouse, prepared essentially as described in Ulmer, J. B., et al., Science 259:1745-49 (1993) and Ulmer, J. B. et al., J. Virol. 72:5648-53 (1998) was pre-determined in dose response studies to induce T cell and antibody responses in the linear range of the dose response and results in a response rate of greater than 95% of mice injected. Each formulation, NP DNA alone, or NP DNA±NP protein formulated with Ribi I or the cationic lipids, DMRIE:DOPE or Vaxfectin™, was prepared in the recommended buffer for that vaccine modality. For injections with NP DNA formulated with cationic lipid, the DNA was diluted in 2× PBS to 0.2 mg/ml±purified recombinant NP protein (produced in baculovirus as described in Example 2) at 0.08 mg/ml. Each cationic lipid was reconstituted from a dried film by adding 1 ml of sterile water for injection (SWFI) to each vial and vortexing continuously for 2 min., then diluted with SWFI to a final concentration of 0.15 mM. Equal volumes of NP DNA (±NP protein) and cationic lipid were mixed to obtain a DNA to cationic lipid molar ratio of 4:1. For injections with DNA containing Ribi I adjuvant (Sigmna), Ribi I was reconstituted with saline to twice the final concentration. Ribi I (2×) was mixed with an equal volume of NP DNA at 0.2 mg/ml in saline±NP protein at 0.08 mg/ml. For immunizations without cationic lipid or Ribi, NP DNA was prepared in 150 mM sodium phosphate buffer, pH 7.2. For each experiment, groups of 9 BALB/c female mice at 7-9 weeks of age were injected with 50 μl of NP DNA±NP protein, cationic lipid or Ribi I. Injections were given bilaterally in each rectus femoris at day 0 and day 21. The mice were bled by OSP on day 20 and day 33 and serum titers of individual mice were measured.
  • NP specific serum antibody titers were determined by indirect binding ELISA using 96 well ELISA plates coated overnight at 4° C. with purified recombinant NP protein at 0.5 μg per well in BBS buffer pH 8.3. NP coated wells were blocked with 1% bovine serum albumin in BBS for 1 h at room temperature. Two-fold serial dilutions of sera in blocking buffer were incubated for 2 h at room temperature and detected by incubating with alkaline phosphatase conjugated (AP) goat anti-mouse IgG-Fc (Jackson Immunoresearch, West Grove, Pa.) at 1:5000 for 2 h at room temperature. Color was developed with 1 mg/ml para-nitrophenyl phosphate (Calbiochem, La Jolla, Calif.) in 50 mM sodium bicarbonate buffer, pH 9.8 and 1 mM MgCl2 and the absorbance read at 405 nm. The titer is the reciprocal of the last dilution exhibiting an absorbance value 2 times that of pre-bleed samples.
  • Standard ELISPOT technology, used to identify the number of interferon gamma (IFN-y) secreting cells after stimulation with specific antigen (spot forming cells per million splenocytes, expressed as SFU/million), was used for the CD4+ and CD8+ T-cell assays. For the screening assays, 3 mice from each group were sacrificed on day 34, 35, and 36. At the time of collection, spleens from each group were pooled, and single cell suspensions made in cell culture media using a dounce homogenizer. Red blood cells were lysed, and cells washed and counted. For the CD4+ and CD8+ assays, cells were serially diluted 3-fold, starting at 106 cells per well and transferred to 96 well ELISPOT plates pre-coated with anti-murine IFN-γ monoclonal antibody. Spleen cells were stimulated with the H-2Kd binding peptide, TYQRTRALV (SEQ ID NO:81), at 1 μg/ml and recombinant murine IL-2 at 1 U/ml for the CD8+ assay and with purified recombinant NP protein at 20 μg/ml for the CD4+ assay. Cells were stimulated for 20-24 hours at 37° C. in 5% CO2, then the cells were washed out and biotin labeled anti-IFN-γ monoclonal antibody added for a 2 hour incubation at room temperature. Plates were washed and horseradish peroxidase-labeled avidin was added. After a 1-hour incubation at room temperature, AEC substrate was added and “spots” developed for 15 min. Spots were counted using the Immunospot automated spot counter (C.T.L. Inc., Cleveland Ohio). Thus, CD4+ and CD8+ responses were measured in three separate assays, using spleens collected on each of three consecutive days.
  • Three weeks after a single injection, antibody responses in mice receiving vaccine formulations containing purified protein were 6 to 8-fold higher than for mice receiving NP DNA only (FIG. 5, Table 15). The titers for mice receiving DNA and protein formulated with a cationic lipid were similar to those for mice receiving protein in Ribi adjuvant or DNA and protein in Ribi adjuvant. These data indicate that the levels of antibody seen when protein is injected with an adjuvant can be obtained with DNA vaccines containing DNA and protein formulated with a cationic lipid, without the addition of conventional adjuvant.
  • Twelve days after a second injection, antibody responses in mice receiving vaccine formulations containing purified protein were 9 to 129-fold higher than for mice receiving NP DNA only (FIG. 6, Table 15). With a mean anti-NP antibody titer of 750,933 at day 33, the titers for mice receiving DNA and protein formulated with Vaxfectin™ were 25-fold higher than for mice receiving DNA alone (mean titer=30,578), and nearly as high as those for mice injected with protein in Ribi adjuvant (mean titer=1,748,133).
    TABLE 15
    Fold increase in antibody response over DNA alone
    20 days after one 12 days after second
    Formulation injection injection
    protein + Ribi 7X (p = 0.0002)  57X (p = 0.002)
    DNA + protein + 6X (p = 0.00005)  9X (p = 0.0002)
    DMRIE:DOPE
    DNA + protein + 8X (p = 0.00003)  25X (p = 0.0004)
    Vaxfectin ™
    DNA + protein + Ribi 7X (p = 0.01) 129X (p = 0.003)

    *protein = purified recombinant NP protein
  • As expected, an NP specific CD8+ T-cell IFN-γ response was not detected in spleens of mice injected with NP protein in Ribi (FIG. 7). All of the other groups had detectable NP specific CD8+ T-cell responses. The CD8+ T-cell responses for all groups receiving vaccine formulations containing NP DNA were not statistically different from each other.
  • Mice from all of the groups had detectable NP specific CD4+ T-cell responses (FIG. 8). The CD4+ T-cell responses of splenocytes from groups receiving vaccine formulations containing NP DNA and NP protein formulated with cationic lipid were 2-6 fold higher than the group injected with DNA alone.
  • B. Codon-Optimized IV Constructs
  • Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are used in the prime-boost compositions described herein. For the prime-boost modalities, the same protein may be used for the boost, e.g., DNA encoding NP with NP protein, or a heterologous boost may be used, e.g., DNA encoding NP with an M1 protein boost. Each formulation, the plasmid comprising a coding region for the IV protein alone, or the plasmid comprising a coding region for the IV protein plus the isolated protein are formulated with Ribi I or the cationic lipids, DMRIE:DOPE or Vaxfectin™. The formulations are prepared in the recommended buffer for that vaccine modality. Exemplary formulations, using NP as an example, are described herein. Other plasmid/protein formulations, including multivalent formulations, can be easily prepared by one of ordinary skill in the art by following this example. For injections with DNA formulated with cationic lipid, the DNA is diluted in 2× PBS to 0.2 mg/ml±purified recombinant NP protein at 0.08 mg/ml. Each cationic lipid is reconstituted from a dried film by adding 1 ml of sterile water for injection (SWFI) to each vial and vortexing continuously for 2 min., then diluted with SWFI to a final concentration of 0.15 mM. Equal volumes of NP DNA (±NP protein) and cationic lipid are mixed to obtain a DNA to cationic lipid molar ratio of 4:1. For injections with DNA containing Ribi I adjuvant (Sigma), Ribi I is reconstituted with saline to twice the final concentration. Ribi I (2×) is mixed with an equal volume of NP DNA at 0.2 mg/ml in saline±NP protein at 0.08 mg/ml. For immunizations without cationic lipid or Ribi, NP DNA is prepared in 150 mM sodium phosphate buffer, pH 7.2. For each experiment, groups of 9 BALB/c female mice at 7-9 weeks of age are injected with 50 μl of NP DNA±NP protein, cationic lipid or Ribi I. The formulations are administered to BALB/c mice (n=10) via bilateral injection in each rectus femoris at day 0 and day 21.
  • The mice are bled on day 20 and day 33 and serum titers of individual mice to the various IV antigens are measured. Serum antibody titers specific for the various IV antigens are determined by ELISA. Standard ELISPOT technology, used to identify the number of interferon gamma (IFN-γ) secreting cells after stimulation with specific antigen (spot forming cells per million splenocytes, expressed as SFU/million), is used for the CD4+ and CD8+ T-cell assays using 3 mice from each group vaccinated above, sacrificed on day 34, 35 and 36, post vaccination.
  • Example 9 Murine Challenge Model of Influenza
  • General Experimental Procedure
  • A murine challenge model with influenza A virus is used to test the efficacy of the immunotherapies. The model used is based on that described in Ulmer, J. B., et al., Science 259:1745-49 (1993) and Ulmer, J. B. et al., J Virol. 72:5648-53 (1998), both of which are incorporated herein by reference in their entireties. This model utilizes a mouse-adapted strain of influenza A/HK/8/68 which replicates in mouse lungs and is titered in tissue culture in Madin Darby Canine Kidney cells. The LD90 of this mouse-adapted influenza virus is determined in female BALB/c mice age 13-15 weeks. In this model, two types of challenge study can be conducted: lethal challenge, where the virus is administered intranasally to heavily sedated mice under ketamine anesthesia; and a sub-lethal challenge, where mice are not anesthetized when the viral inoculum is administered (also intranasally). The endpoint for lethal challenge is survival, but loss in body mass and body temperature can also be monitored. The read-outs for the sublethal challenge include lung virus titer and loss in body mass and body temperature.
  • In the studies described here, mice are subjected to lethal challenge. Mice that are previously vaccinated with DNA encoding IV antigens are anesthetized and challenged intranasally with 0.02 mL of mouse-adapted influenza A/HK/8/68 (mouse passage #6), diluted 1 to 10,000 (500 PFU) in PBS containing 0.2% wt/vol BSA.
  • These challenge studies utilize groups of 10 mice. The route of administration is intramuscular in rectus femoris (quadriceps), using 0.1 μg up to 1 mg total plasmid DNA. Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are tested singly and in multivalent cocktails for the ability to protect against challenge. The plasmids are formulated with an adjuvant and/or a transfection facilitating agent, e.g., Vaxfectin™ by methods described elsewhere herein. Mice are vaccinated on days 0 and 21 using amounts of plasmids as described in Example 6. Subsequent injections can be administered. Nasal challenge of mice takes place 3 weeks after the final immunization, and animals are monitored daily for body mass, hypothermia, general appearance and then death.
  • For each group of mice that are studied, blood is taken at 2 weeks following the second injection, and/or any subsequent injection, and the animals are terminally bled two weeks following the last injection. Antibody titers are determined for M2, M1, and NP using ELISAs as previously described.
  • Plasmids
  • As described above, constructs of the present invention were inserted into the expression vector VR10551. VR10551 is an expression vector without any transgene insert.
  • VR4750 contains the coding sequence for hemagglutinin (HA) (H3N2) from mouse adapted A/Hong Kong/68. The DNA was prepared using Qiagen plasmid purification kits.
  • Experimental Procedure
  • The experimental procedure for the following example is as described above, with particular parameters and materials employed as described herein. In order to provide a pDNA control for protection in the mouse influenza challenge model, the hemagglutinin (HA) gene was cloned from the influenza A/HK/8/68 challenge virus stock, which was passaged 6 times in mice.
  • Mice were vaccinated twice at 3 week intervals with either 100 μg pDNA VR4750 encoding the HA gene cloned directly from the mouse-adapted influenza A/HK/8/68 strain, or with 100 μg blank vector pDNA (VR10551). An additional control group was immunized intranasally with live A/HK/8/68 virus (500 PFU). Three weeks after the last injection, mice were challenged intranasally with mouse-adapted influenza A/HK/8168 with one of 3 doses (50, 500 and 5,000 PFU). Following viral challenge, mice were monitored daily for symptoms of disease, loss in body mass and survival.
  • FIG. 9 shows that homologous HA-pDNA vaccinated mice are completely protected over a range of viral challenge doses (FIG. 9A) and did not suffer significant weight loss (FIG. 9B) during the 3 week period following challenge.
  • Based on these results, future mouse flu challenge studies can include VR4750 (HA) pDNA as a positive control for protection and utilize 500 PFU, which is the LD90 for this mouse-adapted virus, as the challenge dose.
  • Example 10 Challenge in Non-Human Primates
  • The purpose of these studies is to evaluate three or more of the optimal plasmid DNA vaccine formulations for immunogenicity in non-human primates. Rhesus or cynomologus monkeys (6/group) are vaccinated with plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, HA, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, intramuscularly 0.1 to 2 mg DNA combined with cationic lipid, and/or poloxamer and/or aluminum phosphate based or other adjuvants at 0, 1 and 4 months.
  • Blood is drawn twice at baseline and then again at the time of and two weeks following each vaccination, and then again 4 months following the last vaccination. At 2 weeks post-vaccination, plasma is analyzed for humoral response and PBMCs are monitored for cellular responses, by standard methods described herein. Animals are monitored for 4 months following the final vaccination to determine the durability of the immune response.
  • Animals are challenged within 2-4 weeks following the final vaccination. Animals are challenged intratracheally with the suitable dose of virus based on preliminary challege studies. Nasal swabs, pharyngeal swabs and lung lavages are collected at days 0, 2, 4, 6, 8 and 11 post-challenge and will be assayed for cell-free virus titers on monkey kidney cells. After challenge, animals are monitored for clinical symptoms, e.g., rectal temperature, body weight, leukocyte counts, and in addition, hematocrit and respiratory rate. Oropharyngeal swab samples are taken to allow determination of the length of viral shedding. Illness is scored using the system developed by Berendt & Hall (Infect Immun 16:476-479 (1977)), and will be analyzed by analysis of variance and the method of least significant difference.
  • Example 11 Challenge in Birds
  • In this example, various vaccine formulations of the present invention are tested in the chicken influenza model. For these studies an IV H5N1 virus, known to infect birds, is used. Plasmid constructs comprising codon-optimized and non-codon-optimized coding regions encoding NP, M1, M2, eM2, and/or an eM2-NP fusion; or alternatively coding regions (either codon-optimized or non-codon optimized) encoding various IV proteins or fragments, variants or derivatives either alone or as fusions with a carrier protein, e.g., HBcAg, as well as various controls, e.g., empty vector, are formulated with cationic lipid, and/or poloxamer and/or aluminum phosphate based or other adjuvants. The vaccine formulations are delivered at a dose of about 1-10 μg, delivered IM into the defeathered breast area, at 0 and 1 month. The animals are bled for antibody results 3 weeks following the second vaccine. Antibody titers against the various IV antigens are determined using techniques described in the literature. See, e.g., Kodihalli S. et al., Vaccine 18:2592-9 (2000). The birds are challenged intranasally with 0.1 mL containing 100 LD50 3 weeks post second vaccination. The birds are monitored daily for 10 days for disease symptoms, which include loss of appetite, diarrhea, swollen faces, cyanosis, paralysis and death. Tracheal and cloacal swabs are taken 4 days following challenge for virus titration.
  • Example 12 Formulation Selection Studies
  • The potency of different vaccine formulations was evaluated in different experimental studies using the NP protein of Influenza A/PR/8134.
  • Vaccination Regimen
  • Groups of nine, six- to eight-week old BALB/c mice (Harlan-Sprague-Dawley) received bilateral (50 μL/leg) intramuscular (rectus femoris) injections of plasmid DNA. Control mice received DNA in PBS alone. Mice received injections on days 0, 20 and 49. Mice were bled by OSP on day 62, and NP-specific antibodies analyzed by ELISA. Splenocytes were harvested from 3 mice/group/day for three sequential days beginning day 63, and NP-specific specific T cells were analyzed by IFNγ ELISPOT using overlapping peptide stimulation.
  • Cell Culture Media
  • Splenocyte cultures were grown in RPMI-1640 medium containing 25 mM HEPES buffer and L-glutamine and supplemented with 10% (v/v) FBS, 55 μM β-mercaptoethanol, 100 U/mL of penicillin G sodium salt, and 100 μg/mL of streptomycin sulfate.
  • Standard Influenza NP Indirect Binding Assay
  • NP specific serum antibody titers were determined by indirect binding ELISA using 96 well ELISA plates coated overnight at 4° C. with purified recombinant NP protein at 0.5 μg per well in BBS buffer, pH 8.3. NP coated wells were blocked with 1% bovine serum albumin in BBS for 1 hour at room temperature. Two-fold serial dilutions of sera in blocking buffer were incubated for 2 hours at room temperature and detected by incubating with alkaline phosphatase conjugated (AP) goat anti-mouse IgG-Fc (Jackson Immunoresearch, West Grove, Pa.) at 1:5000 for 2 hours at room temperature. Color was developed with 1 mg/ml para-nitrophenyl phosphate (Calbiochem, La Jolla, Calif.) in 50 mM sodium bicarbonate buffer, pH 9.8 and 1 mM MgCl2 and the absorbance read at 405 nm. The titer is the reciprocal of the last dilution exhibiting an absorbance value 2 times that of pre-bleed samples.
  • Standard NP CD8+ and CD4+ T-Cell ELISPOT Assay
  • Standard ELISPOT technology, used to identify the number of interferon gamma (IFN-γ) secreting cells after stimulation with specific antigen (spot forming cells per million splenocytes, expressed as SFU/million), was used for the CD4+ and CD8+ T-cell assays. Three mice from each group were sacrificed on each of three consecutive days. At the time of collection, spleens from each group were pooled, and single cell suspensions were made in cell culture media using a dounce homogenizer. Red blood cells were lysed, and cells were washed and counted. For the CD4+ and CD8+ assays, cells were serially diluted 3- fold, starting at 106 cells per well and transferred to 96 well ELISPOT plates pre-coated with anti-murine IFN-γ monoclonal antibody. Spleen cells were stimulated with the H-2Kd binding peptide, TYQRTRALV, at 1 μg/ml and recombinant murine IL-2 at 1 U/ml for the CD8+ assay and with purified recombinant NP protein at 20 μg/ml for the CD4+ assay. Cells were stimulated for 20-24 hours at 37° C. in 5% CO2, and then the cells were washed out and biotin labeled anti-IFN-γ monoclonal antibody added for a 2 hour incubation at room temperature. Plates were washed and horseradish peroxidase-labeled avidin was added. After a 1-hour incubation at room temperature, AEC substrate was added and “spots” developed for 15 minutes. Spots were counted using the Immunospot automated spot counter (C.T.L. Inc., Cleveland Ohio).
  • Experiment 1
  • The purpose of this experiment was to determine a dose response to naked DNA (VR4700) and for pDNA formulated with VF-P1205-02A. VR4700 is a plasmid encoding influenza A/PR/8/34 nucleoprotein (NP) in a VR10551 backbone. VR10551 is an expression vector without any transgene insert. VF-P1205-02A is a formulation containing a poloxamer with a POP molecular weight of 12 KDa and POE of 5% (CRL1005) at a DNA:poloxamer:BAK ratio of 5 mg/ml:7.5 mg/ml:0.3 mM. The results of this experiment are shown in the following Table:
    TABLE 16
    CRL1005 BAK Serum Ab CD8+T CD4+T
    DNA dose dose conc. titers (total cells cells
    (μg) (μg) (μM) IgG, n = 9) (SFU/106) (SFU/106)
    1 11,206 28 24
    10 31,289 77 99
    100 65,422 243 304
    1 1.5 0.06 9,956 48 57
    10 15 0.6 45,511 174 220
    100 150 6 79,644 397 382
  • The results of this experiment indicate that increasing the dose of DNA increases both the humoral and cell mediated immune responses. When the DNA is formulated with poloxamer and BAK, increasing the dose also increases both the humoral and cell mediated immune responses.
  • Experiment 2
  • The purpose of this experiment was to determine a dose response to CRL1005, with a fixed pDNA (VR4700) dose and no BAK. The results of this experiment are shown in the following Table:
    TABLE 17
    CRL1005
    DNA dose dose Serum Ab titers CD8+T cells CD4+T cells
    (μg) (μg) (total IgG, n = 9) (SFU/106) (SFU/106)
    10 27,733 45 46
    10 15 38,400 69 86
    10 50 46,933 66 73
    10 150 54,044 90 97
    10 450 76,800 90 92
    10 750 119,467 83 60
  • The results of this experiment indicate that increasing the dose of CRL1005 increases both the humoral and cell mediated immune responses.
  • Experiment 3
  • The purpose of this experiment was to compare immune responses of DMRIE:DOPE (1:1, mol:mol) and Vaxfectin™ cationic lipid formulations at different pDNA/cationic lipid molar ratios. The results of this experiement are shown in the following Table:
    TABLE 18
    DMRIE:DOPE Vaxfectin ™ Serum CD8+T CD4+T
    DNA pDNA/cationic pDNA/cationic Ab titers cells cells
    dose lipid molar lipid molar (total (SFU/ (SFU/
    (μg) ratios ratios IgG, n = 9) 106) 106)
    10 17,778 57 54
    10 4:1 48,356 47 112
    10 2:1 49,778 44 133
    10 4:1 88,178 68 464
    10 2:1 150,756 46 363
  • The results of this experiment indicate that formulating the plasmid with DMRIE:DOPE or Vaxfectin™ increases both the humoral and cell mediated immune responses.
  • Experiment 4
  • The purpose of this experiment was first to compare immune responses of DMRIE:DOPE (1:1, mol:mol) at pDNA/cationic lipid molar ratios of 4:1 as an MLV (multi lamellar vesicle formulation—multi-vial) or SUV (small unilamellar vesicles—single-vial) formulation. Second, it was to compare sucrose (lyophilized and frozen) and PBS based formulations. The results of this experiment are shown in the following Table:
    TABLE 19
    DNA Serum Ab CD8+T CD4+T
    dose titers (total cells cells
    (μg) Formulation Buffer IgG, n = 9) (SFU/106) (SFU/106)
    10 PBS, pH 21,333 107 118
    7.2
    10 SUV PBS, pH 15,644 144 169
    7.2
    10 SUV PBS, pH 13,511 114 173
    7.8
    10 SUV Sucrose 15,644 103 119
    Frozen/thawed pH 7.8
    10 SUV Sucrose 10,311 ND 246
    Lyophilized pH 7.8
    10 MLV PBS, 29,867 170 259
    pH 7.2

    * ND - could not be counted due to high background
  • The results of this experiment indicate that formulating the plasmid with DMRIE:DOPE stimulates both the humoral and cell mediated immune responses.
  • Experiment 5
  • The purpose of this experiment was first to determine what effect changing the ratio of DMRIE to DOPE has on immune response at pDNA/cationic lipid molar ratios of 4:1 as an MLV (multi-vial, in PBS) or SUV (single-vial in PBS) formulation. Second, it was to compare the effect of changing the co-lipid from DOPE to cholesterol. The results of this experiment are shown in the following Table:
    TABLE 20
    Serum Ab
    DNA titers CD8+T CD4+T
    dose (total IgG, cells cells
    (μg) Formulation DMRIE:DOPE n = 9) (SFU/106) (SFU/106)
    10 19,342 65 98
    10 MLV, 1:0 38,684 70 126
    DM:DP
    10 MLV, 3:1 75,093 82 162
    DM:DP
    10 MLV, 1:1 53,476 78 186
    DM:DP
    10 SUV, 1:1 36,409 96 106
    DM:DP
    10 MLV, 1:1 52,338 65 154
    DM:Chol
  • The results of this experiment indicate that formulating the plasmid with DMRIE:DOPE stimulates both the humoral and cell mediated immune responses. Changing the co-lipid from DOPE to cholesterol also stimulates both the humoral and cell mediated immune responses.
  • Experiment 6
  • The purpose of this experiment was to obtain a dose response to pDNA formulated with DMRIE:DOPE (1:1, mol:mol) at a 4:1 pDNA/cationic lipid molar ratio. The results of this experiemtn are shown in the following Table:
    TABLE 21
    Serum
    DNA dose Ab titers (total CD8+T cells CD4+T cells
    (μg) Formulation IgG, n = 9) (SFU/106) (SFU/106)
    10 22,044 119 154
    1 MLV 5,600 22 67
    3 MLV 22,756 46 97
    10 MLV 45,511 199 250
    30 MLV 60,444 274 473
    100 MLV 91,022 277 262
  • The results of this experiment indicate that when the plasmid is formulated with DMRIE:DOPE, increasing the dose also increases both the humoral and cell mediated immune responses.
  • Example 13 In vitro Expression of Influenza Antigens
  • Plasmid Vector
  • Polynucleotides of the present invention were inserted into eukaryotic expression vector backbones VR10551, VR10682 and VR6430 all of which are described previously. The VR10551 vector is built on a modified pUC18 background (see Yanisch-Perron, C., et al. Gene 33:103-119 (1985)), and contains a kanamycin resistance gene, the human cytomegalovirus immediate early 1 promoter/enhancer and intron A, and the bovine growth hormone transcription termination signal, and a polylinker for inserting foreign genes. See Hartikka, J., et al., Hum. Gene Ther. 7:1205-1217 (1996). However, other standard commercially available eukaryotic expression vectors may be used in the present invention, including, but not limited to: plasmids pcDNA3, pHCMV/Zeo, pCR3.1, pEF1/His, pIND/GS, pRc/HCMV2, pSV40/Zeo2, pTRACER-HCMV, pUB6/V5-His, pVAX1, and pZeoSV2 (available from Invitrogen, San Diego, Calif.), and plasmid pCI (available from Promega, Madison, Wis.).
  • Various plasmids were generated by cloning the nucleotide sequence for the following influenza A antigens: segment 7 (encodes both M1 and M2 proteins via differential splicing), M2 and NP into expression constructions as described below and pictured in FIG. 13.
  • Plasmids VR4756 (SEQ ID NO:91), VR4759 (SEQ ID NO:92) and VR4762 (SEQ ID NO:93) were created by cloning the nucleotide sequence encoding the consensus sequence for the following influenza A antigens respectively: segment 7 (encoding both the M1 and M2 proteins by differential splicing), M2 and NP into the VR10551 backbone. The VR4756, VR4759 and VR4762 plasmids are also described in Table 13.
  • The VR4764 (SEQ ID NO:95) and VR4765 (SEQ ID NO:96) plasmids were constructed by ligating the segment 7 and NP coding regions from VR4756 and VR4762 respectively into the VR10682 vector. Specifically, the VR4756 vector was digested with EcoRV and SalI restriction endonucleases and the blunted fragment was ligated into the VR10682 backbone, which had been digested with the EcoRV restriction endonuclease. The VR4765 vector was constructed by digesting the VR4762 vector with EcoRV and NotI and ligating the NP coding region into the VR10682 backbone digested with the same restriction endonucleases.
  • VR4766 (SEQ ID NO:97) and VR4767 (SEQ ID NO:98) contain a CMV promoter/intron A-NP expression cassette and a RSV promoter (from VCL1005)-segment 7 expression cassette in the same orientation (VR4766) or opposite orientation (VR4767). These plasmids were generated by digesting VR4762 with the DraIII restriction endonuclease and cutting the RSV-segment 7-mRBG cassette from VR4764 with EcoRV and BamHI restriction endonucleases. After exonuclease digestion with the Klenow fragment of DNA polymerase I, the the EcoRV/BamHI fragment was cloned into the DraIII digested VR4762 vector. Both insert orientations were obtained by this blunt end cloning method.
  • VR4768 (SEQ ID NO:99) and VR4769 (SEQ ID NO:100), containing a CMV promoter/intron A-segment 7 expression cassette and a RSV promoter-NP expression cassette, were similarly derived. VR4756 was digested with the DraIII restriction endonuclease and blunted by treatment with the Klenow fragment of DNA Polymerase I. The cassette containing the RSV promoter, NP coding region and mRBG terminator was removed from VR4765 by digesting with KpnI and NdeI restriction endonucleases. The fragment was also blunted with the Klenow fragment of DNA polymerase I and ligated into the DraIII-digested VR4756 vector in both gene orientations.
  • VR4770 (SEQ ID NO:101), VR4771 (SEQ ID NO:102) and VR4772 (SEQ ID NO:103) were constructed by cloning the coding regions from VR4756, VR4762 and VR4759 respectively into the VR6430 vector backbone. Specifically, the segment 7 gene from VR4756 was removed using SalI and EcoRV restriction endonucleases and blunted with the Klenow fragment of DNA polymerase I. The VR6430 plasmid was digested with EcoRV and BamHI and the vector backbone fragment was blunted with the Klenow fragment of DNA polymerase I. The segment 7 gene fragment was then ligated into the VR6430 vector backbone. VR4771 was derived by removing the NP insert from VR4762 following EcoRV and BglII restriction endonuclease digestion and the fragment was ligated into the VR6430 vector backbone which had been digested the same restriction endonucleases. VR4772 was derived by subcloning the M2 coding region from VR4759 as a blunted SalI-EcoRV fragment and ligating into the VR6430 vector backbone from a blunted EcoRV-BamHI digest.
  • VR4773 (SEQ ID NO:104) and VR4774 (SEQ ID NO:105) contain a CMV promoter/intron A-segment 7 expression cassette and a RSV/R-NP expression cassette with the genes in the same or opposite orientation. These plasmids were generated by digesting VR4756 with the DraIII restriction endonuclease, blunting, and ligating to the RSV/R-NP-BGH fragment from VR4771 (VR4771 digested with NdeI and SfiI and then blunted).
  • VR4775 (SEQ ID NO:106) and VR4776 (SEQ ID NO:107) contain a CMV promoter/intron A-NP expression cassette and a RSV/R-segment 7 expression cassette with the genes in the same or opposite orientation. These plasmids were generated by digesting VR4762 with the DraIII restriction enzyme and blunting with the Klenow fragment of DNA polymerase. The RSV/R-segment 7-BGH fragment was generated by digesting VR4770 with NdeI and SfiI restriction endonucleases and ligating the blunted fragment with the DraIII restriction endonuclease digested VR4762.
  • VR4777 (SEQ ID NO:108) and VR4778 (SEQ ID NO:109) contain a CMV promoter/intron A-NP expression cassette and a RSV/R-M2 expression cassette in the same or opposite orientation. These plasmids were generated by digesting VR4762 with the MscI restriction endonuclease, digesting VR4772 with NdeI and SfiI restriction endonucleases and treating the RSV/R-M2-BGH with the Klenow fragment of DNA polymerase, followed by ligation of these two gel purified fragments.
  • VR4779 and VR4780 contain a CMV promoter/intron A-M2 expression cassette and a RSV/R-NP expression cassette in the same or opposite orientation. These plasmids were generated by digesting VR4759 with the MscI restriction endonuclease, digesting VR4771 with NdeI and SfiI restriction endonucleases and treating the RSV/R-NP-BGH segment with the Klenow fragment of DNA polymerase, followed by ligation of these two gel purified fragments.
  • Plasmid DNA Purification
  • Plasmid DNA was transformed into Escherichia coli DH5α competent cells, and highly purified covalently closed circular plasmid DNA was isolated by a modified lysis procedure (Horn, N. A., et al., Hum. Gene Ther. 6:565-573 (1995)) followed by standard double CsCl-ethidium bromide gradient ultracentrifugation (Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y. (1989)). All plasmid preparations were free of detectable chromosomal DNA, RNA and protein impurities based on gel analysis and the bicinchoninic protein assay (Pierce Chem. Co., Rockford Ill.). Endotoxin levels were measured using Limulus Amebocyte Lysate assay (LAL, Associates of Cape Cod, Falmouth, Mass.) and were less than 0.6 Endotoxin Units/mg of plasmid DNA. The spectrophotometric A260/A280 ratios of the DNA solutions were typically above 1.8. Plasmids were ethanol precipitated and resuspended in an appropriate solution, e.g., 150 mM sodium phosphate (for other appropriate excipients and auxiliary agents, see U.S. patent application Publication 2002/0019358, published Feb. 14, 2002). DNA was stored at −20° C. until use. DNA was diluted by mixing it with 300 mM salt solutions and by adding appropriate amount of USP water to obtain 1 mg/ml plasmid DNA in the desired salt at the desired molar concentration.
  • Plasmid Expression in Mammalian Cell Lines
  • The expression plasmids were analyzed in vitro by transfecting the plasmids into a well characterized mouse melanoma cell line (VM-92, also known as UM-449) and the human rhabdomyosarcoma cell line RD (ATCC CCL-136) both available from the American Type Culture Collection, Manassas, Va. Other well-characterized human cell lines may also be used, e.g. MRC-5 cells, ATCC Accession No. CCL-171. The transfection was performed using cationic lipid-based transfection procedures well known to those of skill in the art. Other transfection procedures are well known in the art and may be used, for example electroporation and calcium chloride-mediated transfection (Graham F. L. and A. J. van der Eb Virology 52:456-67 (1973)). Following transfection, cell lysates and culture supernatants of transfected cells were evaluated to compare relative levels of expression of IV antigen proteins. The samples were assayed by western blots and ELISAs, using commercially available monoclonal antibodies (available, e.g., from Research Diagnostics Inc., Flanders, N.J.), so as to compare both the quality and the quantity of expressed antigen.
  • Genes encoding the consensus amino acid sequences (described above) derived for NP, M1 and M2 antigens were cloned in several configurations into several plasmid vector backbones. The pDNAs were tested for in vitro expression and are being assessed in vivo for immunogenicity, as well as for the ability to protect mice from influenza challenge.
  • Experiment 1
  • Following the derivation of an amino acid consensus for M1 and M2, a native segment 7 isolate was found to encode this consensus, and this nucleotide sequence was synthesized according to methods described above. An M2-M1 fusion gene was also created and the nucleotide sequence was human codon-optimized using the above described codon optimization algorithm of Example 4. The individual full-length M2 and M1 genes were also cloned via PCR from this fusion.
  • In vitro expression of influenza antigens in cell lysates was assessed 48 hours after transfection into a mouse melanoma cell line. M2 expression was detected following transfection of VR4756 (segment 7), VR4755 (M2-M1 fusion) and VR4759 (full-length M2) using the anti-M2 monoclonal antibody (14C2) from Affinity BioReagents. The data are shown in FIG. 10 for VR4756 and VR4755. Expression of M1 was detected from transfected VR4756, VR4755 and VR4760 (full-length M1) pDNAs, as detected by anti-M1 monoclonal (Serotec) in FIG. 10 for VR4756 and VR4755, or by anti-M1 goat polyclonal (Virostat, data not shown). VR10551 is the empty cloning vector.
  • Experiment 2
  • In order to compare alternative human codon-optimization methods, two versions of a fusion of the first 24 amino acids of M2 to full-length NP (“eM2-NP”) were constructed. One nucleotide sequence was derived from the above codon optimization algorithm, while the other was done by an outside vendor. Comparison of expression levels from the two eM2-NP pDNAs was measured in vitro, and comparison of immunogenicity in vivo is on-going. Additionally, the full-length NP genes for both codon-optimized versions were sub-cloned from the eM2-NP pDNAs and analyzed for expression in vitro.
  • In vitro expression was tested to compare eM2-NP and NP pDNAs derived from the above described codon-optimization algorithm and an outside vendor algorithm. The data are shown in FIG. 11. Expression levels were approximately the same for VR4757 (eM2-NP vendor optimization) vs. VR4758 (eM2-NP Applicant optimization), as detected by anti-M2 monoclonal (FIG. 11A) or anti-NP mouse polyclonal (data not shown). Similarly, NP expression was approximately equal for VR4761 (vendor optimization) vs. VR4762 (Applicant optimization), detected by anti-NP mouse polyclonal generated by Applicants (FIG. 11B). NP consensus protein expression in vitro was also detected using a goat polyclonal antibody (Fitzgerald) generated against whole H1N1 or H3N2 virus (data not shown). Expression levels of both of these NP constructs were much higher than a pDNA containing A/PR/34 NP (VR4700).
  • Experiment 3
  • Influenza antigen-encoding plasmids were transfected into VM92 cells using methods described above. Cell lysates and media were collected 48 hours after transfection. Cells were lysed in 200 μl of Laemmli buffer, cell debris removed by microcentrifuge spin, and 20 μl was heated and loaded on a 4-12% Bis-Tris gel. To determine expression of those vectors encoding secreted NP protein, 15 μl of media was mixed with 5 μl of loading buffer, heated, and loaded on a gel. Western blots were processed as described above. Primary antibodies were as follows: monoclonal antibody MA1-082 (ABR) to detect M2 protein, monoclonal antibody MCA401 (Serotec) to detect M1 protein, and a polyclonal antibody against VR4762-injected rabbits generated in-house. All primary antibodies were used at a 1:500 dilution.
  • FIG. 14 shows Western blot results wherein M2 protein expression from segment 7-enocoding plasmids are higher in CMV promoter/intron A-segment 7 (VR4756) and RSV/R-segment 7 (VR4770) than VR4764 (RSV promoter). NP expression appeared highest from the RSV/R-NP plasmid (VR4771), followed by CMV/intron A-NP (VR4762) and then RSV-NP (VR4765). Similar results were seen in Western blots from human RD-transfected cells.
  • For dual promoter plasmids, containing RSV-segment 7 and CMV/intron A-NP (VR4766 and VR4767), M2 expression from segment 7 is very low, independent of orientation. The CMV/intron A-NP expression in these dual promoter plasmids does not differ significantly compared to VR4762. RSV-NP expression in dual promoter plasmids (VR4768 and VR4769), where segment 7 is expressed from CMV/intron A, NP expression decreases somewhat, but not as drastically as M2 expression in the dual promoter VR4766 and VR4767.
  • FIG. 15 shows expression of the M1 and M2 proteins from segment 7, as well as NP, from CMV promoter/intron A, RSV promoter, and RSV/R-containing plasmids. For these Western blots, dual promoter plasmids contain the CMV promoter/intron A and RSV/R driving either NP or segment 7. Similar results were seen in Western blots from human RD-transfected cells.
  • Western blot results confirm that the M1 and M2 protein expression from both CMV promoter/intron A-segment 7 (VR4756) and RSV/R-segment 7 (VR4770) is superior to RSV-segment 7 (VR4764). M1 and M2 expression decrease slightly when RSV/R-segment 7 or CMV/intron A-segment 7 is combined with CMV/intron A-NP or RSV/R-NP in a dual promoter plasmid (VR4773, VR4774, VR4775, and VR4776). Results were similar in Western blots from human RD transfected cells. Human RD cells transfected with M2 antigen encoding plasmids, RSV/R-M2 (VR4772) and CMV/intron A-M2 (VR4759), showed a similar level of M2 expression, which was decreased in dual promoter plasmids (VR4777, VR4778, VR4779, and VR4780). Human RD cells transfected with NP antigen-encoding plasmids, VR4762, VR4771, VR4777, VR4778, VR4779, and VR4780, all showed similar NP expression levels.
  • Example 14 Murine Influenza a Challenge Model
  • A model influenza A challenge model has been established utilizing a mouse-adapted A/BK/8/68 strain. Positive and negative control Hemagluttinin (HA)-containing plasmids were generated by PCR of the HA genes directly from mouse-adapted A/Hong Kong/68 (H3N2) and A/Puerto Rico/34 (H1N1) viruses, respectively.
  • For all experiments, plasmid DNA vaccinations are given as bilateral, rectus femoris injections at 0 and 3 weeks, followed by orbital sinus puncture (OSP) bleed at 5 weeks and intranasal viral challenge at 6 weeks with 500 pfu (1 LD90) of virus. Mice are monitored for morbidity and weight loss for about 3 weeks following viral challenge. Endpoint antibody titers for NP and M2 were determined by ELISA. For study GSJ08, 5 additional mice per test group were vaccinated and interferon-γ ELISPOT assays were performed at week number 5.
  • Study CL88:
  • A mouse influenza challenge study was initiated to test the M1, M2, Segment 7, and NP-encoding plamids alone, or in combination. In addition to HA pDNAs, sub-lethal infection and naïve mice serve as additional positive and negative controls, respectively. Mice received 100 μg of each plasmid formulated in poloxamer CRL1005, 02A formulation. The test groups and 21 day post-challenge survival are shown in Table 21:
    TABLE 21
    Total
    pDNA per # mice/ 21 day
    Group Construct(s) vaccination group Survival (%)
    A VR4762 (NP) 100 μg 12 17
    B VR4759 (M2) 100 μg 12 25
    C VR4760 (M1) 100 μg 12 0
    D VR4756 (S7) 100 μg 12 50
    E VR4762 (NP) + 200 μg 12 100
    VR4759 (M2)
    F VR4762 (NP) + 200 μg 12 17
    VR4760 (M1)
    G VR4762 (NP) + 200 μg 12 75
    VR4756 (S7)
    H VR4750 (HA, 100 μg 12 100
    H3N2, + control)
    I VR4752 (HA, 100 μg 12 8
    H1N1, − control)
    J Naive mice (− control) N/A 12 8
    K Sub-lethal (+ control) N/A 12 100

    CL88 Results:
  • The performance criteria for this study was survival of >90% for the positive controls, ≦10% for the negative controls, and >75% for the experimental groups. Table 21 shows that all of the control groups, as well as two experimental groups met the performance criteria. The M2+NP and S7+NP plamsid DNA combinations resulted in 100% and 75% survival, respectively. There was no statistically significant difference (p<0.05) between the two lead plasmid combinations, but there was statistical significance in the S7, S7+NP, and M2+NP groups vs. the negative controls.
  • Weight loss data showed that the positive control groups did not exhibit any weight loss following viral challenge, as opposed to the weight loss seen in all of the experimental groups. Mice that survived the viral challenge recovered to their starting weight by the end of the study. Tables 22 and 23 show endpoint antibody titers for test groups containing M2, Segment 7, and NP antigens. Shaded boxes represent mice that died following viral challenge.
    TABLE 22
    CL88 M2 Antibody Titers
    Group D Group G Group B Group E
    mouse (seg 7) (NP + seg7) (M2) (NP + M2)
    1 800 1600 25600 1600
    2
    Figure US20060024670A1-20060202-C00001
    1600
    Figure US20060024670A1-20060202-C00002
    6400
    3 3200 6400
    Figure US20060024670A1-20060202-C00003
    200
    4 6400
    Figure US20060024670A1-20060202-C00004
    Figure US20060024670A1-20060202-C00005
    6400
    5 12800
    Figure US20060024670A1-20060202-C00006
    3200 3200
    6 800 12800 12800 3200
    7
    Figure US20060024670A1-20060202-C00007
    0
    Figure US20060024670A1-20060202-C00008
    3200
    8
    Figure US20060024670A1-20060202-C00009
    0
    Figure US20060024670A1-20060202-C00010
    6400
    9 800 3200
    Figure US20060024670A1-20060202-C00011
    1600
    10
    Figure US20060024670A1-20060202-C00012
    3200
    Figure US20060024670A1-20060202-C00013
    800
    11 12800 1600
    Figure US20060024670A1-20060202-C00014
    3200
    12
    Figure US20060024670A1-20060202-C00015
    12800
    Figure US20060024670A1-20060202-C00016
    400

    **An M2 antibody titer of 0 represents a titer of <100.
  • TABLE 23
    CL88 NP Antibody Titers
    Group A Group E Group F Group G
    mouse (NP) (NP + M2) (NP + M1) (NP + seg7)
    1 204800 51200
    Figure US20060024670A1-20060202-C00017
    25600
    2
    Figure US20060024670A1-20060202-C00018
    51200 204800 51200
    3 204800 51200
    Figure US20060024670A1-20060202-C00019
    51200
    4
    Figure US20060024670A1-20060202-C00020
    25600 51200
    Figure US20060024670A1-20060202-C00021
    5
    Figure US20060024670A1-20060202-C00022
    102400
    Figure US20060024670A1-20060202-C00023
    Figure US20060024670A1-20060202-C00024
    6
    Figure US20060024670A1-20060202-C00025
    51200
    Figure US20060024670A1-20060202-C00026
    102400
    7
    Figure US20060024670A1-20060202-C00027
    204800
    Figure US20060024670A1-20060202-C00028
    102400
    8
    Figure US20060024670A1-20060202-C00029
    102400
    Figure US20060024670A1-20060202-C00030
    102400
    9
    Figure US20060024670A1-20060202-C00031
    102400
    Figure US20060024670A1-20060202-C00032
    51200
    10
    Figure US20060024670A1-20060202-C00033
    102400
    Figure US20060024670A1-20060202-C00034
    102400
    11
    Figure US20060024670A1-20060202-C00035
    51200
    Figure US20060024670A1-20060202-C00036
    25600
    12
    Figure US20060024670A1-20060202-C00037
    51200
    Figure US20060024670A1-20060202-C00038
    25600

    Study GSJ05:
  • In order to attempt to distinguish between the two antigen combinations, S7+NP and M2+NP, a dose ranging challenge experiment was undertaken with these two plasmid combinations. Mice were injected with 100 μg, 30 μg, or 10 μg per plasmid in the 02A poloxamer formulation at 0 and 3 weeks, followed by bleed at 5 weeks and viral challenge at 6 weeks. Sixteen mice per group were vaccinated for test groups A-H, while 12 mice per group were vaccinated for the controls. Poloxamer 02A-formulated HA plasmids, VR4750 (HA H3) and VR4752 (HA H1), were included as positive and negative controls, respectively. The test groups and 21 day survival post-challenge are shown in Table 24:
    TABLE 24
    Total pDNA # mice/ 21 day
    Group Construct(s) per vaccination group Survival (%)
    A VR4756 (Seg 7) + VR4762 (NP) 200 μg 16 73
    B VR4756 (Seg 7) + VR4762 (NP)  60 μg 16 81
    C VR4756 (Seg 7) + VR4762 (NP)  20 μg 16 69
    D VR4759 (M2) + VR4762 (NP) 200 μg 16 94
    E VR4759 (M2) + VR4762 (NP)  60 μg 16 81
    F VR4759 (M2) + VR4762 (NP)  20 μg 16 75
    G VR4750 (Positive DNA control) 100 μg 12 100
    H VR4752 (Negative DNA control) 100 μg 12 8

    Results
  • The performance criteria of >90% survival with the HA positive control and ≦10% for the HA negative control plasmid again were met. The performance criteria for the experimental groups, >75% survival at the 30 μg per plasmid dose, was met by both M2+NP and S7+NP (Table 24). In fact, at a dose of 10 μg per plasmid, S7+NP and M2+NP resulted in 69% and 75% survival, respectively. There was no statistical significance (p<0.05) between the three doses of M2+NP or between the 3 doses of S7+NP, nor was there statistical significance when comparing M2+NP to S7+NP at the 200 μg, 60 μg, or 20 μg doses. However, there was a statistical difference for the HA positive control vs. S7+NP at 200 μg and 20 μg. Body mass data shows weight loss and recovery by all surviving experimental plasmid DNA-vaccinated groups, while the HA positive control mice did not experience weight loss. Antibody data for M2 and NP are shown in Tables 25 and 26.
    TABLE 25
    GSJ05 M2 Antibody Titers
    Group Group Group Group Group
    mouse # A B C D E Group F
    1
    Figure US20060024670A1-20060202-C00039
    400 3200 6400 800 3200
    2 200
    Figure US20060024670A1-20060202-C00040
    0 25600 1600 0
    3 0
    Figure US20060024670A1-20060202-C00041
    0 3200 3200 3200
    4 100 0
    Figure US20060024670A1-20060202-C00042
    6400 1600 400
    5
    Figure US20060024670A1-20060202-C00043
    0 0 3200 800 1600
    6 3200 400 0 6400 200 100
    7 25600 800 0
    Figure US20060024670A1-20060202-C00044
    Figure US20060024670A1-20060202-C00045
    Figure US20060024670A1-20060202-C00046
    8 0 100
    Figure US20060024670A1-20060202-C00047
    1600 0 400
    9
    Figure US20060024670A1-20060202-C00048
    Figure US20060024670A1-20060202-C00049
    800 3200 12800 0
    10
    Figure US20060024670A1-20060202-C00050
    800
    Figure US20060024670A1-20060202-C00051
    1600 800
    Figure US20060024670A1-20060202-C00052
    11 100 1600
    Figure US20060024670A1-20060202-C00053
    3200 200 1600
    12 3200 0
    Figure US20060024670A1-20060202-C00054
    6400
    Figure US20060024670A1-20060202-C00055
    1600
    13 800 0 400 3200
    Figure US20060024670A1-20060202-C00056
    800
    14
    Figure US20060024670A1-20060202-C00057
    0 1600 3200 400 100
    15 0 1600 800 1600 3200
    Figure US20060024670A1-20060202-C00058
    16 0 0 800 800 3200
    Figure US20060024670A1-20060202-C00059
  • TABLE 26
    GSJ05 NP Antibody Titers
    Group Group Group Group Group
    mouse # A B C D E Group F
    1
    Figure US20060024670A1-20060202-C00060
    51200 51200 51200 25600 25600
    2 25600
    Figure US20060024670A1-20060202-C00061
    12800 51200 25600 6400
    3 102400
    Figure US20060024670A1-20060202-C00062
    51200 12800 51200 25600
    4 25600 12800
    Figure US20060024670A1-20060202-C00063
    25600 12800 12800
    5
    Figure US20060024670A1-20060202-C00064
    102400 6400 25600 12800 12800
    6 25600 51200 25600 25600 12800 6400
    7 102400 51200 6400
    Figure US20060024670A1-20060202-C00065
    Figure US20060024670A1-20060202-C00066
    Figure US20060024670A1-20060202-C00067
    8 51200 25600
    Figure US20060024670A1-20060202-C00068
    12800 51200 6400
    9
    Figure US20060024670A1-20060202-C00069
    Figure US20060024670A1-20060202-C00070
    25600 102400 12800 12800
    10
    Figure US20060024670A1-20060202-C00071
    25600
    Figure US20060024670A1-20060202-C00072
    25600 12800
    Figure US20060024670A1-20060202-C00073
    11 51200 25600
    Figure US20060024670A1-20060202-C00074
    25600 25600 3200
    12 51200 51200
    Figure US20060024670A1-20060202-C00075
    25600
    Figure US20060024670A1-20060202-C00076
    12800
    13 51200 51200 25600 51200
    Figure US20060024670A1-20060202-C00077
    12800
    14
    Figure US20060024670A1-20060202-C00078
    12800 25600 51200 6400 12800
    15 25600 6400 25600 25600 25600
    Figure US20060024670A1-20060202-C00079
    16 51200 51200 25600 12800 12800
    Figure US20060024670A1-20060202-C00080

    Gray shading represents mice that died post-challenge. Group A, mouse 9 (spotted box) died during the OSP bleed procedure.

    Study GSJ06
  • The plasmid combination VR4759 (M2) and VR4762 (NP) was utilized in further mouse influenza challenge studies to examine additional formulations.
  • Using the experimental protocol described above, 12 mice per group were vaccinated with equal weight VR4759 (M2) and VR4762 (NP) in the following formulations:
      • Poloxamer 02A used in the previous two challenge experiments.
      • DMRIE+Cholesterol (DM:Chol) at a 4:1 molar ratio of DNA to DMRIE, the molar ratio of DM:Chol is 3:1.
      • Vaxfectin™ (VC 1052+DPyPE) at a 4:1 molar ratio of DNA: VC1052, the molar ratio of VC1052: DpyPE is 1:1.
  • GSJ06 study design and 21 day survival post-challenge is found in Table 27.
    TABLE 27
    Total 21 day
    Group pDNA pDNA Survival (%)
    A Poloxamer 02A  20 ug 92
    B Poloxamer 02A  2 ug 58
    C DMRIE: Cholesterol  20 ug 58
    D DMRIE: Cholesterol  2 ug 17
    E Vaxfectin  20 ug 100
    F Vaxfectin  2 ug 75
    G VR4750 (HA, positive) 100 ug 100
    H VR4752 (HA, negative) 100 ug 0

    Results
  • Poloxamer 02A and Vaxfectin™-formulated plasmid DNA led to 92% and 100% survival at the 20 μg pDNA dose, and 58% and 75% at the 2 μg dose, respectively (Table 27).
  • Average weights were tracked for each group of mice starting at the day of challenge. As shown in Table 28, it was noted in this experiment that the weight recovery for group E (Vaxfectin™-formulated pDNA, 20 μg total) began after day 4, as opposed to the other groups' recovery beginning at day 7. Antibody titers, Tables 29 and 30, were determined for M2 and NP and shaded boxes represent mice that died following viral challenge.
    TABLE 28
    GSJ06 Average Body Weights Post-Challenge
    Avg Body Weights (g)-Days post-challenge
    Group pDNA Total pDNA 0 2 4 7 9 11 14 16 18 21
    A Poloxamer 02A 20 ug 20.73 19.98 17.98
    Figure US20060024670A1-20060202-C00081
    17.36 18.74 19.94 20.45 20.60 21.08
    B Poloxamer 02A  2 ug 21.08 19.91 17.96 15.17
    Figure US20060024670A1-20060202-C00082
    16.03 16.77 17.41 18.10 19.52
    C DMRIE-Cholesterol 20 ug 21.43 20.24 18.14
    Figure US20060024670A1-20060202-C00083
    18.68 19.24 20.14 20.50 20.90 21.42
    D DMRIE-Cholesterol  2 ug 21.28 20.24 17.58
    Figure US20060024670A1-20060202-C00084
    16.18 17.45 18.80 19.84 20.13 20.98
    E Vaxfectin 20 ug 21.41 19.97
    Figure US20060024670A1-20060202-C00085
    18.10 19.12 19.82 20.39 20.87 20.93 21.34
    F Vaxfectin  2 ug 20.47 18.97 16.86
    Figure US20060024670A1-20060202-C00086
    16.22 16.84 17.87 18.60 19.08 20.02
    G VR4750 (HA, positive) 100 ug  21.30 20.97 21.60 21.21 21.57 21.79 21.84 22.13 21.94 22.13
    H VR4752 (HA, negative) 100 ug  20.89 20.25 17.57 14.67

    Shading represents the lowest group average post-challenge for each test group. Group H (negative control) weight averages are not recorded once the percentage survival has dropped below 50%.
  • TABLE 29
    GSJ06 M2 Antibody Titers
    Group Group Group Group
    mouse # A B C Group D E Group F
    1
    Figure US20060024670A1-20060202-C00087
    400
    Figure US20060024670A1-20060202-C00088
    Figure US20060024670A1-20060202-C00089
    1600 6400
    2 6400
    Figure US20060024670A1-20060202-C00090
    1600 400 800
    3 6400
    Figure US20060024670A1-20060202-C00091
    Figure US20060024670A1-20060202-C00092
    Figure US20060024670A1-20060202-C00093
    12800 3200
    4 1600 0 400
    Figure US20060024670A1-20060202-C00094
    25600 1600
    5 6400 3200
    Figure US20060024670A1-20060202-C00095
    Figure US20060024670A1-20060202-C00096
    100
    Figure US20060024670A1-20060202-C00097
    6 3200 100 100
    Figure US20060024670A1-20060202-C00098
    12800 1600
    7 800 1600 1600
    Figure US20060024670A1-20060202-C00099
    800 3200
    8 400 100
    Figure US20060024670A1-20060202-C00100
    200 6400
    Figure US20060024670A1-20060202-C00101
    9 1600
    Figure US20060024670A1-20060202-C00102
    100
    Figure US20060024670A1-20060202-C00103
    6400
    Figure US20060024670A1-20060202-C00104
    10 100
    Figure US20060024670A1-20060202-C00105
    1600
    Figure US20060024670A1-20060202-C00106
    3200 400
    11 3200 0 800
    Figure US20060024670A1-20060202-C00107
    1600 1600
    12 6400
    Figure US20060024670A1-20060202-C00108
    Figure US20060024670A1-20060202-C00109
    0 6400 1600
  • TABLE 30
    GSJ06 NP Antibody Titers
    Group Group Group Group
    mouse # A B C Group D E Group F
    1
    Figure US20060024670A1-20060202-C00110
    6400
    Figure US20060024670A1-20060202-C00111
    Figure US20060024670A1-20060202-C00112
    51200 51200
    2 51200
    Figure US20060024670A1-20060202-C00113
    6400
    Figure US20060024670A1-20060202-C00114
    102400 102400
    3 12800
    Figure US20060024670A1-20060202-C00115
    Figure US20060024670A1-20060202-C00116
    Figure US20060024670A1-20060202-C00117
    51200 25600
    4 25600 1600 6400
    Figure US20060024670A1-20060202-C00118
    204800 102400
    5 25600 6400
    Figure US20060024670A1-20060202-C00119
    Figure US20060024670A1-20060202-C00120
    51200
    Figure US20060024670A1-20060202-C00121
    6 51200 12800 25600
    Figure US20060024670A1-20060202-C00122
    102400 51200
    7 25600 25600 12800
    Figure US20060024670A1-20060202-C00123
    51200 51200
    8 25600 3200
    Figure US20060024670A1-20060202-C00124
    6400 25600
    Figure US20060024670A1-20060202-C00125
    9 25600
    Figure US20060024670A1-20060202-C00126
    51200
    Figure US20060024670A1-20060202-C00127
    51200
    Figure US20060024670A1-20060202-C00128
    10 51200
    Figure US20060024670A1-20060202-C00129
    12800
    Figure US20060024670A1-20060202-C00130
    51200 51200
    11 25600 12800 25600
    Figure US20060024670A1-20060202-C00131
    102400 51200
    12 51200
    Figure US20060024670A1-20060202-C00132
    Figure US20060024670A1-20060202-C00133
    400 51200 51200

    Study GSJ08
  • Further formulation comparisons were done with utilizing VR4759 (M2) and VR4762 (NP). Seventeen mice per test group (A-G) were vaccinated with equal weight VR4759 (M2) and VR4762 (NP) vectors in the following formulations:
      • Poloxamer 02A
      • Vaxfectin™ (preparations A and B represent different purifications)
      • DMRIE:DOPE at a 4:1 molar ratio of DNA to DMRIE
      • DMRIE:DOPE at a 2.5:1 molar ratio of DNA to DMRIE
      • PBS (unformulated pDNA)
  • Twelve mice per test group were challenged with influenza virus at week number 6. Five mice per test group were sacrificed at days 36-38 for T cell assays (IFN-γ ELISPOT). The test groups and 21 day survival post-challenge are shown in Table 31. Groups A-D, and F-G were vaccinated with 20 μg total plasmid DNA per injection to further explore the weight loss/recovery phenomena seen in study GSJ06 with the Vaxfectin™-formulated pDNA.
    TABLE 31
    Total pDNA 21 Day
    Group Construct(s) per vaccination Survival (%)
    A Poloxamer 02A 20 μg 50
    B DMRIE:DOPE 4:1 20 μg 92
    C DMRIE:DOPE 2.5:1 20 μg 92
    D Vaxfectin - prep A 20 μg 92
    E Vaxfectin - prep A 2 μg 75
    F Vaxfectin - prep B 20 μg 100
    G PBS 20 μg 42
    H VR4750 (HA, H3N2, +control) 100 μg 100
    I VR4752 (HA, H1N1, −control) 100 μg 17

    Results
  • The DMRIE:DOPE and Vaxfectin™ formulated groups resulted in 92-100% survival at a 20 μg pDNA dose. Group A (Poloxaamer 02A) and Group G (PBS) survival results were not statistically different than the negative control (as measured by Fisher exact p, one-tailed), while the Vaxfectin™ and DMIRE:DOPE Groups (Groups B-F) were shown to be statistically superior (p<0.05) as compared to the negative control. Therefore, the plasmid DNA formulated with lipids appear to provide superior protection in the mouse influenza model challenge.
  • A repeated measures ANOVA mixed model analysis of weight data for groups B, C, and D of the weight loss and recovery data showed that Group B and Group D were not statistically different, while Group C and Group D were statistically different.
  • T cell responses, as measured by IFN-γ ELISPOT assay, were conducted on the last 5 mice per group using an M2 peptide encompassing the first 24 amino acids of M2 (TABLE 33), an NP protein expressed in baculovirus (TABLE 34), and an NP CD8+ Balb/c immunodominant peptide (TABLE 35).
  • Antibody titers, Tables 36 and 37, were determined for M2 and NP proteins. The first 12 mice listed for each group were challenge at day 42 and the last 5 mice per group were sacrificed for IFN-γ ELISPOT. The shaded boxes represent mice that died following viral challenge.
    TABLE 32
    GSJ06 Average Body Weights Post-Challenge
    Total pDNA Avg Body Weights (g)-Days post-challenge
    Group Construct(s) par vaccination 0 2 4 5 6 7 9 11 14 16 18 22
    A Poloxamer 02A 20 μg 20.47 18.97 16.30 15.43 14.75
    Figure US20060024670A1-20060202-C00134
    14.36 14.44 16.63 17.64 18.36 20.53
    B DMRIE-DOPE 4:1 20 μg 21.58 19.94 17.43 16.75 16.17
    Figure US20060024670A1-20060202-C00135
    16.43 17.28 18.45 19.50 20.22 20.89
    C DMRIE-DOPE 2.5:1 20 μg 19.95 18.58 16.44 15.77
    Figure US20060024670A1-20060202-C00136
    15.56 15.75 16.22 16.78 17.16 17.31 18.04
    D Vaxfectin - prep A 20 μg 20.87 19.22 16.81 16.47
    Figure US20060024670A1-20060202-C00137
    16.92 17.94 19.48 20.06 20.19 20.64 21.17
    E Vaxfectin - prep A  2 μg 20.40 19.59 17.97 17.47 17.27
    Figure US20060024670A1-20060202-C00138
    18.96 19.83 20.24 20.49 20.57 21.06
    F Vaxfectin - prep B 20 μg 21.33 20.01 17.88
    Figure US20060024670A1-20060202-C00139
    17.74 18.21 18.85 19.85 20.29 20.77 20.88 21.39
    G PBS 20 μg 20.84 19.46 16.97 16.00 15.38
    Figure US20060024670A1-20060202-C00140
    15.80 16.39 17.35
    H VR4750 100 μg  21.25 21.15 21.27 20.77 20.92 21.24 20.74 21.16 21.33 21.40 21.64 21.64
    (HA, H3N2, +control)
    I VR4752 100 μg  21.67 20.65 17.87 16.77 16.05 15.17 15.09
    (HA, H1N1, −control)

    Shading represents the lowest group average post-challenge for each test group. Group G and I weight averages are not recorded once the percentage survival has dropped below 50%.
  • TABLE 33
    M2 peptide Interferon-γ ELISPOT
    M2 peptide IFN gamma ELISPOT (SFU/10E6 cells)
    Mouse A B C D E F G
    1 66 88 145 189 283 253 31
    2 11 115 150 269 62 282 47
    3 115 247 190 233 99 283 112
    4 20 6 51 67 73 93 45
    5 93 277 397 248 202 399 93
    AVG 61 147 187 201 144 262 66
  • TABLE 34
    NP CD4 peptide Interferon-γ ELISPOT
    ND CD4 peptide IFN gamma ELISPOT (SFU/10E6 cells)
    Mouse A B C D E F G
    1 7 32 3 52 72 108 18
    2 8 83 34 125 8 34 8
    3 22 91 106 293 26 51 73
    4 9 15 80 39 53 10 12
    5 37 150 374 117 40 217 43
    AVG 17 74 119 125 40 84 31
  • TABLE 35
    NP CD8 peptide Interferon-γ ELISPOT
    NP CD8 peptide IFN gamma ELISPOT (SFU/10E6 cells)
    Mouse A B C D E F G
    1 11 37 4 14 20 67 8
    2 0 3 4 6 1 0 2
    3 31 19 15 26 23 51 34
    4 1 0 0 12 1 38 3
    5 46 36 39 21 13 15 18
    AVG 18 19 12 16 12 34 13
  • TABLE 36
    GSJ08 M2 Antibody Titers
    mouse # Group A Group B Group C Group D Group E Group F Group G Group H ELISPOT #
    1 1600 3200 3200 6400 400 12800 800 6400
    2
    Figure US20060024670A1-20060202-C00141
    12800 6400 1600 3200 800
    Figure US20060024670A1-20060202-C00142
    Figure US20060024670A1-20060202-C00143
    3
    Figure US20060024670A1-20060202-C00144
    3200 6400
    Figure US20060024670A1-20060202-C00145
    800 3200
    Figure US20060024670A1-20060202-C00146
    800
    4
    Figure US20060024670A1-20060202-C00147
    Figure US20060024670A1-20060202-C00148
    6400 1600
    Figure US20060024670A1-20060202-C00149
    800
    Figure US20060024670A1-20060202-C00150
    0
    5 1600 0
    Figure US20060024670A1-20060202-C00151
    12800 1600 800
    Figure US20060024670A1-20060202-C00152
    Figure US20060024670A1-20060202-C00153
    6
    Figure US20060024670A1-20060202-C00154
    3200 1600 6400 200 12800 400
    Figure US20060024670A1-20060202-C00155
    7
    Figure US20060024670A1-20060202-C00156
    3200 12800 800 1600 3200 1600
    Figure US20060024670A1-20060202-C00157
    8 12800 6400 3200 12800 12800 12800 12800
    Figure US20060024670A1-20060202-C00158
    9 1600 1600 0 12800 6400 12800
    Figure US20060024670A1-20060202-C00159
    Figure US20060024670A1-20060202-C00160
    10 3200 1600 12800 12800 1600 800
    Figure US20060024670A1-20060202-C00161
    12800
    11 1600 6400 3200 3200
    Figure US20060024670A1-20060202-C00162
    6400
    Figure US20060024670A1-20060202-C00163
    Figure US20060024670A1-20060202-C00164
    12 200 800 6400 25600
    Figure US20060024670A1-20060202-C00165
    800
    Figure US20060024670A1-20060202-C00166
    6400
    13 1600 800 6400 12800 3200 6400 6400 6400 1
    14 3200 6400 1600 1600 800 12800 3200 12800 2
    15 0 1600 3200 3200 12800 12800 6400 12800 3
    16 3200 3200 1600 12800 0 12800 200 6400 4
    17 3200 200 400 6400 800 400 1600 3200 5
  • TABLE 37
    GSJ08 NP Antibody Titers
    mouse # Group A Group B Group C Group D Group E Group F Group G Group H ELISPOT #
    1 51200 25600 6400 51200 12800 51200 51200 25600
    2
    Figure US20060024670A1-20060202-C00167
    25600 51200 51200 25600 102400
    Figure US20060024670A1-20060202-C00168
    Figure US20060024670A1-20060202-C00169
    3
    Figure US20060024670A1-20060202-C00170
    51200 12800
    Figure US20060024670A1-20060202-C00171
    6400 102400
    Figure US20060024670A1-20060202-C00172
    12800
    4
    Figure US20060024670A1-20060202-C00173
    Figure US20060024670A1-20060202-C00174
    51200 102400
    Figure US20060024670A1-20060202-C00175
    25600
    Figure US20060024670A1-20060202-C00176
    25600
    5 25600 12800
    Figure US20060024670A1-20060202-C00177
    51200 51200 102400
    Figure US20060024670A1-20060202-C00178
    Figure US20060024670A1-20060202-C00179
    6
    Figure US20060024670A1-20060202-C00180
    12800 51200 102400 25600 51200 25600
    Figure US20060024670A1-20060202-C00181
    7
    Figure US20060024670A1-20060202-C00182
    51200 51200 51200 25600 204800 102400
    Figure US20060024670A1-20060202-C00183
    8 25600 51200 25600 51200 12800 51200 25600
    Figure US20060024670A1-20060202-C00184
    9 25600 12800 25600 51200 51200 51200
    Figure US20060024670A1-20060202-C00185
    Figure US20060024670A1-20060202-C00186
    10 6400 12800 51200 51200 25600 204800
    Figure US20060024670A1-20060202-C00187
    25600
    11 12800 51200 25600 204800
    Figure US20060024670A1-20060202-C00188
    102400
    Figure US20060024670A1-20060202-C00189
    Figure US20060024670A1-20060202-C00190
    12 102400 102400 51200 102400
    Figure US20060024670A1-20060202-C00191
    204800
    Figure US20060024670A1-20060202-C00192
    51200
    13 25600 25600 12800 51200 51200 102400 25600 25600 1
    14 51200 25600 12800 51200 25600 102400 25600 51200 2
    15 51200 51200 51200 51200 25600 25600 102400 12800 3
    16 25600 6400 25600 51200 25600 102400 25600 51200 4
    17 25600 25600 51200 51200 12800 51200 25600 25600 5
  • The present invention is not to be limited in scope by the specific embodiments described which are intended as single illustrations of individual aspects of the invention, and any compositions or methods which are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the appended claims.
  • All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.

Claims (52)

1-392. (canceled)
393. An isolated polynucleotide comprising a nucleic acid fragment which encodes the consensus amino acid sequence of SEQ ID NO:78, wherein the codons of said nucleic acid fragment are optimized for expression in humans.
394. The polynucleotide of claim 393, wherein the nucleotide sequence of said nucleic acid fragment is SEQ ID NO:66.
395. A vector comprising the polynucleotide of claim 393, wherein said vector, upon uptake by a suitable host cell, expresses said amino acid sequence.
396. The polynucleotide of claim 393, further comprising a heterologous nucleic acid ligated to said nucleic acid fragment.
397. A composition comprising the vector of claim 395 and a carrier.
398. The composition of claim 397, further comprising a component selected from the group consisting of an adjuvant and a transfection facilitating compound.
399. The composition of claim 398, wherein said component is a cationic lipid.
400. The composition of claim 399, wherein said adjuvant comprises(±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-bis(syn-9-tetradeceneyloxy)-1-propanaminium bromide (GAP-DMORIE) and a neutral lipid, wherein said neutral lipid is selected from the group consisting of:
(a) 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE);
(b) 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE); and
(c) 1,2-dimyristoyl-glycer-3-phosphoethanolamine (DMPE).
401. The composition of claim 399, wherein said transfection facilitating compound comprises (±)-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (DMRIE).
402. The composition of claim 401, wherein said transfection facilitating compound further comprises a neutral lipid.
403. The composition of claim 402, wherein said neutral lipid is DOPE.
404. The composition of claim 401 further comprising a 1:1 molar ratio of GAP-DMORIE and DPyPE.
405. A method for treating or preventing influenza infection in a vertebrate comprising administering to a vertebrate in need thereof the composition of claim 397.
406. A method for eliciting an immune response to influenza virus in a vertebrate comprising administering to a vertebrate in need thereof the composition of claim 397.
407. An isolated polynucleotide comprising a nucleic acid fragment which encodes the consensus amino acid sequence of SEQ ID NO:76, wherein the codons of said nucleic acid fragment are optimized for expression in humans.
408. The polynucleotide of claim 407, wherein the nucleotide sequence of said nucleic acid fragment is SEQ ID NO:75.
409. A vector comprising the polynucleotide of claim 405, wherein said vector, upon uptake by a suitable host cell, expresses said amino acid sequence.
410. The polynucleotide of claim 407, further comprising a heterologous nucleic acid ligated to said nucleic acid fragment.
411. A composition comprising the vector of claim 409 and a carrier
412. The composition of claim 411, further comprising a component selected from the group consisting of an adjuvant and a transfection facilitating compound.
413. The composition of claim 412, wherein said component is a cationic lipid.
414. The composition of claim 413, wherein said adjuvant comprises(±)-N-(3 -aminopropyl)-N,N-dimethyl-2,3-bis(syn-9-tetradeceneyloxy)-1-propanaminium bromide (GAP-DMORIE) and a neutral lipid, wherein said neutral lipid is selected from the group consisting of:
(a) 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE);
(b) 1,2-diphytanoyl-sn-glycero-3 -phosphoethanolamine (DPyPE); and
(c) 1,2-dimyristoyl-glycer-3-phosphoethanolamine (DMPE).
415. The composition of claim 413, wherein said transfection facilitating compound comprises (±)-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (DMRIE).
416. The composition of claim 415, wherein said transfection facilitating agent further comprises a neutral lipid.
417. The composition of claim 416, wherein the neutral lipid is DOPE.
418. The composition of claim 415 further comprising a 1:1 molar ratio of GAP-DMORIE and DPyPE.
419. An isolated polypeptide produced by the polynucleotide claim 407.
420. A method for treating or preventing influenza infection in a vertebrate comprising administering to a vertebrate in need thereof the composition of claim 411.
421. A method for eliciting an immune response to influenza virus in a vertebrate by administration of the composition of claim 411.
422. An isolated polynucleotide comprising a first nucleic acid fragment which encodes the consensus amino acid sequence of SEQ ID NO:78 and a second nucleic acid fragment which encodes the consensus amino acid sequence of SEQ ID NO:76, wherein the codons of said first and second nucleic acid fragments are optimized for expression in humans.
423. The polynucleotide of claim 422, wherein the nucleotide sequence of said first nucleic acid fragment is SEQ ID NO:66 and wherein the nucleotide sequence of said second nucleic acid fragment is SEQ ID NO:75.
424. A vector comprising the polynucleotide of claim 422, wherein said vector, upon uptake by a suitable host cell, expresses the consensus amino acid sequences of SEQ ID NO:78 and SEQ ID NO:76.
425. The vector of claim 424, wherein said consensus amino acid sequences of SEQ ID NO:78 and SEQ ID NO:76 are expressed as a fusion protein.
426. The vector of claim 422, wherein said vector is DNA and wherein said vector comprises a first expression cassette and second expression cassette, said first expression cassette comprises a first nucleic acid fragment which encodes the consensus amino acid sequence of SEQ ID NO:78 in operable association with a promoter and said second expression cassette comprises a second nucleic acid fragment which encodes the consensus amino acid sequence of SEQ ID NO:76 in operable association with a promoter.
427. The vector of claim 426, wherein said first expression cassette and said second expression cassette are associated with separate promoters.
428. The vector of claim 427, wherein said separate promoters are non-identical.
429. The vector of claim 426, wherein said first expression cassette and said second expression cassette are associated with a single promoter, and wherein said second expression cassette is in operable association with an internal ribosome entry site (IRES).
430. The vector of claim 426, wherein said first expression cassette and said second expression cassette are associated with a single promoter, and wherein said first expression cassette is in operable association with an internal ribosome entry site (IRES).
431. A composition comprising the vector of claim 424 and a carrier.
432. A composition comprising the vector of claim 426 and a carrier.
433. A composition comprising at least two non-identical vectors, wherein one of said vectors comprises a nucleic acid fragment which encodes the consensus amino acid sequence of SEQ ID NO:78 and wherein another of said vectors comprises a nucleic acid fragment which encodes the consensus amino acid sequence of SEQ ID NO:76, wherein the codons of said nucleic acid fragments encoding SEQ ID NO:78 and SEQ ID NO:76 are optimized for expression in humans, and wherein said vectors, upon uptake by a suitable host cell, express said amino acid sequences.
434. The composition of claim 433, further comprising a carrier.
435. The composition of claim 434, further comprising a component selected from the group consisting of an adjuvant and a transfection facilitating compound.
436. The composition of claim 435, wherein said component is a cationic lipid.
437. The composition of claim 436, wherein said adjuvant comprises(±)-N-(3-aminopropyl)-N,N-dimethyl-2,3-bis(syn-9-tetradeceneyloxy)-1-propanaminium bromide (GAP-DMORIE) and a neutral lipid, wherein said neutral lipid is selected from the group consisting of:
(a) 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE);
(b) 1,2-diphytanoyl-sn-glycero-3-phosphoethanolamine (DPyPE); and
(c) 1,2-dimyristoyl-glycer-3-phosphoethanolamine (DMPE).
438. The composition of claim 436, wherein said transfection facilitating compound comprises (±)-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide (DMRIE).
439. The composition of claim 438, wherein said transfection facilitating compound further comprises a neutral lipid.
440. The composition of claim 439, wherein the neutral lipid is DOPE.
441. The composition of claim 438 further comprising a 1:1 molar ratio of GAP-DMORIE and DPyPE.
442. A method for treating or preventing influenza infection in a vertebrate comprising administering to a vertebrate in need thereof the composition of claim 434.
443. A method for eliciting an immune response to influenza virus in a vertebrate by administration of the composition of claim 434.
US11/131,479 2004-05-18 2005-05-18 Influenza virus vaccine composition and methods of use Abandoned US20060024670A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US11/131,479 US20060024670A1 (en) 2004-05-18 2005-05-18 Influenza virus vaccine composition and methods of use
US11/704,251 US7537768B2 (en) 2004-05-18 2007-02-09 Influenza virus vaccine composition and methods of use
US11/715,973 US7785603B2 (en) 2004-05-18 2007-03-09 Influenza virus vaccine composition and methods of use
US11/892,016 US8128938B1 (en) 2004-05-18 2007-08-17 Influenza virus vaccine composition and methods of use
US12/688,614 US20100197771A1 (en) 2004-05-18 2010-01-15 Influenza virus vaccine composition and methods of use
US13/250,508 US8821890B2 (en) 2004-05-18 2011-09-30 Influenza virus vaccine composition and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57185404P 2004-05-18 2004-05-18
US11/131,479 US20060024670A1 (en) 2004-05-18 2005-05-18 Influenza virus vaccine composition and methods of use

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US11/704,251 Division US7537768B2 (en) 2004-05-18 2007-02-09 Influenza virus vaccine composition and methods of use
US11/715,973 Continuation US7785603B2 (en) 2004-05-18 2007-03-09 Influenza virus vaccine composition and methods of use
US12/688,614 Continuation US20100197771A1 (en) 2004-05-18 2010-01-15 Influenza virus vaccine composition and methods of use

Publications (1)

Publication Number Publication Date
US20060024670A1 true US20060024670A1 (en) 2006-02-02

Family

ID=35451484

Family Applications (6)

Application Number Title Priority Date Filing Date
US11/131,479 Abandoned US20060024670A1 (en) 2004-05-18 2005-05-18 Influenza virus vaccine composition and methods of use
US11/704,251 Expired - Fee Related US7537768B2 (en) 2004-05-18 2007-02-09 Influenza virus vaccine composition and methods of use
US11/715,973 Expired - Fee Related US7785603B2 (en) 2004-05-18 2007-03-09 Influenza virus vaccine composition and methods of use
US11/892,016 Expired - Fee Related US8128938B1 (en) 2004-05-18 2007-08-17 Influenza virus vaccine composition and methods of use
US12/688,614 Abandoned US20100197771A1 (en) 2004-05-18 2010-01-15 Influenza virus vaccine composition and methods of use
US13/250,508 Expired - Fee Related US8821890B2 (en) 2004-05-18 2011-09-30 Influenza virus vaccine composition and methods of use

Family Applications After (5)

Application Number Title Priority Date Filing Date
US11/704,251 Expired - Fee Related US7537768B2 (en) 2004-05-18 2007-02-09 Influenza virus vaccine composition and methods of use
US11/715,973 Expired - Fee Related US7785603B2 (en) 2004-05-18 2007-03-09 Influenza virus vaccine composition and methods of use
US11/892,016 Expired - Fee Related US8128938B1 (en) 2004-05-18 2007-08-17 Influenza virus vaccine composition and methods of use
US12/688,614 Abandoned US20100197771A1 (en) 2004-05-18 2010-01-15 Influenza virus vaccine composition and methods of use
US13/250,508 Expired - Fee Related US8821890B2 (en) 2004-05-18 2011-09-30 Influenza virus vaccine composition and methods of use

Country Status (6)

Country Link
US (6) US20060024670A1 (en)
EP (1) EP1766094A4 (en)
JP (1) JP5331340B2 (en)
AU (1) AU2005248361B2 (en)
CA (1) CA2566355C (en)
WO (1) WO2005116270A2 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040162256A1 (en) * 2002-12-23 2004-08-19 Vical Incorporated. Method for producing sterile polynucleotide based medicaments
US20060134221A1 (en) * 2004-12-03 2006-06-22 Vical Incorporated Methods for producing block copolymer/amphiphilic particles
US20060217338A1 (en) * 2005-02-24 2006-09-28 Shan Lu Influenza nucleic acids, polypeptides, and uses thereof
US20070010015A1 (en) * 2003-03-31 2007-01-11 Francisco Rodriguez Aguirre J Complete empty viral particles of infectious bursal disease virus (ibdv), production method thereof and applications of same
US20070015243A1 (en) * 2005-07-15 2007-01-18 Consejo Superior De Investigaciones Cientificas And Bionostra S.L. Chimeric empty viral-like particles derived from the infectious bursal disease virus (IBDV), process for their production and applications
US20070128692A1 (en) * 2004-01-21 2007-06-07 Aguirre Jose Francisco R Chimeric empty capsids of the infectious bursal disease viruse (ibdv), obtainment process and applications
US20070286869A1 (en) * 2004-05-18 2007-12-13 Vical Incorporated Influenza virus vaccine composition and methods of use
US20080299151A1 (en) * 2007-05-31 2008-12-04 Statens Serum Institut Influenza vaccines
EP1998814A2 (en) * 2006-05-11 2008-12-10 Novavax, Inc. Novel influenza m2 vaccines
WO2009073330A2 (en) * 2007-11-12 2009-06-11 The Trustees Of The University Of Pennsylvania Novel vaccines against multiple subtypes of influenza virus
US20090208528A1 (en) * 2004-01-21 2009-08-20 Jose Francisco Rodriguez Aguirre Empty capsids (vlps(-vp4)) of the infectious bursal disease virus (ibdv), obtainment process and applications
US20100040651A1 (en) * 2006-07-20 2010-02-18 Vical Incorporated Compositions and Methods for Vaccinating Against HSV-2
US20100285050A1 (en) * 2007-10-05 2010-11-11 Isis Innovation Limited Compositions and Methods
US20110229518A1 (en) * 2008-11-28 2011-09-22 Statens Serum Institut Optimized influenza vaccines
WO2012012461A3 (en) * 2010-07-19 2012-04-26 Shire Human Genetic Therapies, Inc. Mannose receptor c type 1 (mrc1) codon optimized cell line and uses thereof
US20130260435A1 (en) * 2010-12-15 2013-10-03 Old Dominion University Research Foundation Electroporation-induced electrosensitization
US20160175431A1 (en) * 2009-11-09 2016-06-23 National Jewish Health Vaccine Composition
AU2014268164B2 (en) * 2007-11-12 2016-09-29 Inovio Pharmaceuticals, Inc. Novel vaccines against multiple subtypes of influenza virus
EP2044947B1 (en) 2007-10-05 2017-03-08 Oxford University Innovation Limited MVA-based compositions and methods for inducing an immune response against influenza
US10279032B2 (en) 2006-02-07 2019-05-07 Peptcell Limited Peptide sequences and compositions
WO2022192524A1 (en) * 2021-03-10 2022-09-15 Elicio Therapeutics, Inc. Uses of amphiphiles in immune cell therapy and compositions therefor

Families Citing this family (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1578193A4 (en) * 2002-12-23 2011-06-15 Vical Inc Method for freeze-drying nucleic acid/block copolymer/cationic surfactant complexes
CN104826102A (en) * 2006-02-02 2015-08-12 全球免疫股份有限公司 Yeast-based vaccine for inducing immune response
WO2007103565A2 (en) * 2006-03-09 2007-09-13 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Wnv dna vaccine incorporating a cmv/r backbone and expressing wnv prm and e proteins
CN101015691B (en) * 2006-11-14 2010-08-25 中国医学科学院医学生物学研究所 Recombinant phages influenza vaccine
EP2274320A1 (en) * 2008-04-25 2011-01-19 ID Biomedical Corporation of Quebec Methods for preparing immunogenic compositions
CN102123724B (en) * 2008-06-19 2018-05-01 变异生技公司 Treat the composition and method of influenza
US20110097418A1 (en) * 2009-05-29 2011-04-28 Variation Biotechnologies, Inc. Compositions and methods for treating influenza
CA2803282C (en) 2009-07-06 2018-05-01 David E. Anderson Methods for preparing vesicles and formulations produced therefrom
WO2011005769A1 (en) * 2009-07-06 2011-01-13 Variation Biotechnologies, Inc. Methods for preparing vesicles and formulations produced therefrom
WO2011005799A2 (en) 2009-07-06 2011-01-13 Novartis Ag Self replicating rna molecules and uses thereof
LT3178490T (en) 2009-07-15 2022-07-25 Glaxosmithkline Biologicals S.A. Rsv f protein compositions and methods for making same
GB0918679D0 (en) * 2009-10-23 2009-12-09 Iqur Ltd Influenza vaccine
WO2011056226A1 (en) * 2009-11-05 2011-05-12 Mercia Pharma Llc Adjuvanted nanoparticulate influenza vaccine
EP2544718A1 (en) 2010-03-11 2013-01-16 Immune Design Corp. Vaccines for pandemic influenza
CN102869379B (en) 2010-04-30 2015-04-15 淡马锡生命科学研究院有限公司 Universal vaccine against H5N1 lineages
HUE047796T2 (en) 2010-07-06 2020-05-28 Glaxosmithkline Biologicals Sa Delivery of rna to trigger multiple immune pathways
US20130171241A1 (en) 2010-07-06 2013-07-04 Novartis Ag Liposomes with lipids having an advantageous pka-value for rna delivery
ES2600892T3 (en) 2010-07-06 2017-02-13 Glaxosmithkline Biologicals Sa Virion-like administration particles for self-replicating RNA molecules
US9192661B2 (en) 2010-07-06 2015-11-24 Novartis Ag Delivery of self-replicating RNA using biodegradable polymer particles
US9770463B2 (en) 2010-07-06 2017-09-26 Glaxosmithkline Biologicals Sa Delivery of RNA to different cell types
US9610248B2 (en) 2010-07-06 2017-04-04 Variation Biotechnologies, Inc. Compositions and methods for treating influenza
AU2011276236B2 (en) 2010-07-06 2016-03-10 Glaxosmithkline Biologicals S.A. Cationic oil-in-water emulsions
US10487332B2 (en) 2010-07-06 2019-11-26 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of RNA
WO2012021558A1 (en) 2010-08-09 2012-02-16 Richard Markham Methods and compositions for preventing a condition
SI3970742T1 (en) 2010-08-31 2022-08-31 Glaxosmithkline Biologicals S.A. Pegylated liposomes for delivery of immunogen-encoding rna
MX363307B (en) 2010-10-11 2019-03-20 Novartis Ag Star Antigen delivery platforms.
US10004797B2 (en) 2010-10-27 2018-06-26 Harrisvaccines, Inc. Method of rapidly producing improved vaccines for animals
US8822427B2 (en) 2010-10-27 2014-09-02 Harrisvaccines Methods and compositions to protect aquatic invertebrates from disease
BR112013010288A2 (en) 2010-10-27 2016-09-20 Harrisvaccines Inc method of producing a vaccine to protect an animal from a microorganism biotype, vaccine to protect an animal from a microorganism biotype, method to protect an animal and method to determine if an animal has been vaccinated
AU2012205315B2 (en) 2011-01-13 2017-05-04 Variation Biotechnologies, Inc. Methods for preparing vesicles and formulations produced therefrom
CA2862864C (en) 2011-01-13 2018-12-11 Variation Biotechnologies Inc. Compositions and methods for treating viral infections
EP4144368A1 (en) 2011-01-26 2023-03-08 GlaxoSmithKline Biologicals S.A. Rsv immunization regimen
PL2672992T3 (en) 2011-02-11 2020-11-02 The Trustees Of The University Of Pennsylvania Nucleic acid molecule encoding hepatitis b virus core protein and vaccine comprising the same
US9238679B2 (en) 2011-02-11 2016-01-19 The Trustees Of The University Of Pennslyvania Nucleic acid molecule encoding hepatitis B virus core protein and surface antigen protein and vaccine comprising the same
HUE048777T2 (en) 2011-05-13 2020-08-28 Glaxosmithkline Biologicals Sa Pre-fusion rsv f antigens
RS59037B1 (en) 2011-06-08 2019-08-30 Translate Bio Inc Lipid nanoparticle compositions and methods for mrna delivery
CA2840989A1 (en) 2011-07-06 2013-01-10 Novartis Ag Immunogenic combination compositions and uses thereof
MX350198B (en) 2011-07-06 2017-08-30 Novartis Ag Oil-in-water emulsions that contain nucleic acids.
WO2013006842A2 (en) 2011-07-06 2013-01-10 Novartis Ag Immunogenic compositions and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
RU2014118727A (en) 2011-10-11 2015-11-20 Новартис Аг RECOMBINANT SELF-REPLICING POLYCISTRON RNA MOLECULES
US20140328876A1 (en) 2011-11-18 2014-11-06 Variation Biotechnologies Inc. Synthetic derivatives of mpl and uses thereof
AU2013208693B2 (en) 2012-01-12 2017-12-07 Variation Biotechnologies Inc. Compositions and methods for treating viral infections
US20150079077A1 (en) 2012-01-27 2015-03-19 Variation Biotechnologies, Inc. Methods and compositions for therapeutic agents
CA2865676C (en) 2012-04-17 2020-03-10 Stefan Klostermann Method for the expression of polypeptides using modified nucleic acids
US9969794B2 (en) 2012-05-10 2018-05-15 Visterra, Inc. HA binding agents
EP2869842A1 (en) 2012-07-06 2015-05-13 Novartis AG Immunogenic compositions and uses thereof
BR112015009300A2 (en) 2012-10-23 2018-05-15 Harrisvaccines Inc method of producing a vaccine to protect an animal from a biotype of a microorganism, method of protecting an animal from a biotype of a microorganism and vaccine to protect an animal from a biotype of a microorganism.
CA2897752A1 (en) 2013-01-10 2014-07-17 Novartis Ag Influenza virus immunogenic compositions and uses thereof
US10080792B2 (en) 2013-09-23 2018-09-25 Engen Bio, Inc. Influenza vaccine and therapy
CN104436157A (en) 2013-09-23 2015-03-25 恩金生物有限公司 Influenza vaccine and therapy
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
EA201690675A1 (en) 2013-10-03 2016-08-31 Модерна Терапьютикс, Инк. POLYNUCLEOTES ENCODING THE RECEPTOR OF LOW DENSITY LIPOPROTEINS
JP2015128397A (en) * 2014-01-08 2015-07-16 国立大学法人広島大学 Phosphorous acid dehydrogenase gene, and selective culture method of budding yeast using gene concerned
GB201402890D0 (en) 2014-02-18 2014-04-02 Iqur Ltd Vaccines based on hepatitis B Core antigens
EP3110401A4 (en) 2014-02-25 2017-10-25 Merck Sharp & Dohme Corp. Lipid nanoparticle vaccine adjuvants and antigen delivery systems
LT3134131T (en) 2014-04-23 2022-02-10 Modernatx, Inc. Nucleic acid vaccines
WO2015195049A1 (en) * 2014-06-18 2015-12-23 Agency For Science, Technology And Research Novel promoters for high level expression
EP3374390A1 (en) 2015-11-13 2018-09-19 Visterra, Inc. Compositions and methods for treating and preventing influenza
EP3528827A4 (en) * 2016-10-21 2020-11-04 Merck Sharp & Dohme Corp. Influenza hemagglutinin protein vaccines
KR20190129032A (en) 2016-12-28 2019-11-19 인벡스, 인크. Influenza vaccine
CA3067224A1 (en) 2017-06-15 2018-12-20 Infectious Disease Research Institute Nanostructured lipid carriers and stable emulsions and uses thereof
JP2021511356A (en) 2018-01-29 2021-05-06 メルク・シャープ・アンド・ドーム・コーポレーションMerck Sharp & Dohme Corp. Stabilized RSVF protein and its use
CN112334481A (en) * 2018-06-11 2021-02-05 葛兰素史克消费保健(美国)控股有限责任公司 Antibody pairs for rapid influenza B diagnostic testing
JP7320601B2 (en) * 2018-09-11 2023-08-03 上▲海▼市公共▲衛▼生▲臨▼床中心 Broad-spectrum anti-influenza vaccine immunogen and its use
AU2020248404A1 (en) 2019-03-25 2021-09-30 Visterra, Inc. Compositions and methods for treating and preventing influenza
AU2020304841A1 (en) 2019-06-28 2021-12-23 F. Hoffmann-La Roche Ag Method for the production of an antibody
PE20230493A1 (en) 2020-02-14 2023-03-23 Merck Sharp And Dohme Llc HPV VACCINE
AU2021336217A1 (en) 2020-09-04 2023-04-20 Access To Advanced Health Institute Live-attenuated rna hybrid vaccine technology
TW202245835A (en) 2021-02-04 2022-12-01 美商默沙東有限責任公司 Nanoemulsion adjuvant composition for pneumococcal conjugate vaccines
WO2022221736A2 (en) 2021-04-16 2022-10-20 Genentech, Inc. Optimized tlr7 ligands and uses thereof
WO2023023152A1 (en) 2021-08-19 2023-02-23 Merck Sharp & Dohme Llc Thermostable lipid nanoparticle and methods of use thereof
WO2023228116A1 (en) 2022-05-24 2023-11-30 Access To Advanced Health Institute Intranasal administration of thermostable rna vaccines
WO2024052882A1 (en) 2022-09-09 2024-03-14 Access To Advanced Health Institute Immunogenic vaccine composition incorporating a saponin

Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4818527A (en) * 1986-12-09 1989-04-04 Scripps Clinic And Research Foundation T cell epitopes of the hepatitis B virus nucleocapsid protein
US4882145A (en) * 1986-12-09 1989-11-21 Scripps Clinic And Research Foundation T cell epitopes of the hepatitis B virus nucleocapsid protein
US5143726A (en) * 1986-12-09 1992-09-01 The Scripps Research Institute T cell epitopes of the hepatitis B virus nucleocapsid protein
US5264618A (en) * 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5561064A (en) * 1994-02-01 1996-10-01 Vical Incorporated Production of pharmaceutical-grade plasmid DNA
US5580859A (en) * 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5656611A (en) * 1994-11-18 1997-08-12 Supratek Pharma Inc. Polynucleotide compositions
US5837693A (en) * 1995-03-24 1998-11-17 The Regents Of The University Of California Intravenous hormone polypeptide delivery by salivary gland expression
US6004944A (en) * 1995-03-24 1999-12-21 The Regents Of The University Of California Protein delivery by secretory gland expression
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6214804B1 (en) * 1989-03-21 2001-04-10 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US6231864B1 (en) * 1998-02-12 2001-05-15 Immune Complex Corporation Strategically modified hepatitis B core proteins and their derivatives
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6429199B1 (en) * 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US20020165172A1 (en) * 1996-09-17 2002-11-07 Matti Sallberg Compositions and methods for treating intracellular diseases
US6500432B1 (en) * 1997-12-16 2002-12-31 Smithkline Beecham Biologicals, S.A. Method to enhance an immune response of nucleic acid vaccination
US20030032615A1 (en) * 1989-03-21 2003-02-13 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US6586409B1 (en) * 1999-03-26 2003-07-01 Vical Incorporated Adjuvant compositions and methods for enhancing immune responses to polynucleotide-based vaccines
US20040157244A1 (en) * 2002-12-23 2004-08-12 Vical Incorporated Process for purification of plasmid DNA
US20040157789A1 (en) * 2002-12-23 2004-08-12 Vical Incorporated. Method for freeze-drying nucleic acid/block copolymer/cationic surfactant complexes
US20040162256A1 (en) * 2002-12-23 2004-08-19 Vical Incorporated. Method for producing sterile polynucleotide based medicaments
US20040171572A1 (en) * 1995-11-30 2004-09-02 Wheeler Carl J. Complex cationic lipids having quaternary nitrogens therein
US6875748B2 (en) * 2000-04-21 2005-04-05 Vical Incorporated Compositions and methods for in vivo delivery of polynucleotide-based therapeutics
US20070286869A1 (en) * 2004-05-18 2007-12-13 Vical Incorporated Influenza virus vaccine composition and methods of use

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
DE3689123T2 (en) 1985-11-01 1994-03-03 Xoma Corp MODULAR UNIT OF ANTIBODY GENES, ANTIBODIES MADE THEREOF AND USE.
GB8903313D0 (en) 1989-02-14 1989-04-05 Wellcome Found Conjugates
NZ235315A (en) 1989-09-19 1991-09-25 Wellcome Found Chimaeric hepadnavirus core antigen proteins and their construction
IT221831Z2 (en) 1991-04-18 1994-10-20 Meschi Ind Grafica DEVICE FOR THE DEVELOPMENT OF A PAPER STRIP FROM A REEL
US7105574B1 (en) * 1999-03-26 2006-09-12 Vical Incorporated Adjuvant compositions and methods for enhancing immune responses to polynucleotide-based vaccines
US5591631A (en) * 1993-02-12 1997-01-07 The United States Of America As Represented By The Department Of Health And Human Services Anthrax toxin fusion proteins, nucleic acid encoding same
EP0620277A1 (en) 1993-03-18 1994-10-19 Merck & Co. Inc. Nucleic acid pharmaceuticals
WO1996010631A1 (en) 1994-09-30 1996-04-11 St. Jude Children's Research Hospital Nucleic acid encoding mutant matrix proteins useful for attenuation or enhancement of influenza a virus
US20020045594A1 (en) * 1997-02-14 2002-04-18 Merck & Co., Inc. Polynucleotide vaccine formulations
AU757175B2 (en) 1997-09-05 2003-02-06 Regents Of The University Of California, The Use of immunostimulatory oligonucleotides for preventing or reducing antigen-stimulated, granulocyte-mediated inflammation
WO2001083528A2 (en) 2000-05-01 2001-11-08 Powderject Vaccines, Inc. Nucleic acid immunization
EP1335953A4 (en) 2000-06-23 2005-03-30 Merck & Co Inc Polynucleotide vaccine adjuvants and formulations containing cationic surfactants, and methods of use
EP1358319B1 (en) 2000-09-25 2009-06-17 Polymun Scientific Immunbiologische Forschung GmbH Live influenza vaccine and method of manufacture
TW523465B (en) 2001-02-06 2003-03-11 Olympus Optical Co Image forming apparatus
US20030202982A1 (en) * 2001-08-15 2003-10-30 Birkett Ashley J. Influenza immunogen and vaccine

Patent Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4683195B1 (en) * 1986-01-30 1990-11-27 Cetus Corp
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4818527A (en) * 1986-12-09 1989-04-04 Scripps Clinic And Research Foundation T cell epitopes of the hepatitis B virus nucleocapsid protein
US4882145A (en) * 1986-12-09 1989-11-21 Scripps Clinic And Research Foundation T cell epitopes of the hepatitis B virus nucleocapsid protein
US5143726A (en) * 1986-12-09 1992-09-01 The Scripps Research Institute T cell epitopes of the hepatitis B virus nucleocapsid protein
US20040023911A1 (en) * 1989-03-21 2004-02-05 Vical Incorporated Expression of exogenous polynucleotide sequences in a vertebrate
US6214804B1 (en) * 1989-03-21 2001-04-10 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5580859A (en) * 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US20030032615A1 (en) * 1989-03-21 2003-02-13 Vical Incorporated Lipid-mediated polynucleotide administration to deliver a biologically active peptide and to induce a cellular immune response
US5264618A (en) * 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5561064A (en) * 1994-02-01 1996-10-01 Vical Incorporated Production of pharmaceutical-grade plasmid DNA
US6207646B1 (en) * 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6429199B1 (en) * 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US5656611A (en) * 1994-11-18 1997-08-12 Supratek Pharma Inc. Polynucleotide compositions
US5837693A (en) * 1995-03-24 1998-11-17 The Regents Of The University Of California Intravenous hormone polypeptide delivery by salivary gland expression
US6004944A (en) * 1995-03-24 1999-12-21 The Regents Of The University Of California Protein delivery by secretory gland expression
US20040171572A1 (en) * 1995-11-30 2004-09-02 Wheeler Carl J. Complex cationic lipids having quaternary nitrogens therein
US20020165172A1 (en) * 1996-09-17 2002-11-07 Matti Sallberg Compositions and methods for treating intracellular diseases
US6406705B1 (en) * 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6500432B1 (en) * 1997-12-16 2002-12-31 Smithkline Beecham Biologicals, S.A. Method to enhance an immune response of nucleic acid vaccination
US6231864B1 (en) * 1998-02-12 2001-05-15 Immune Complex Corporation Strategically modified hepatitis B core proteins and their derivatives
US20030191082A1 (en) * 1999-03-26 2003-10-09 Wheeler Carl J. Adjuvant compositions and methods for enhancing immune responses to polynucleotide-based vaccines
US6586409B1 (en) * 1999-03-26 2003-07-01 Vical Incorporated Adjuvant compositions and methods for enhancing immune responses to polynucleotide-based vaccines
US6875748B2 (en) * 2000-04-21 2005-04-05 Vical Incorporated Compositions and methods for in vivo delivery of polynucleotide-based therapeutics
US20040157244A1 (en) * 2002-12-23 2004-08-12 Vical Incorporated Process for purification of plasmid DNA
US20040157789A1 (en) * 2002-12-23 2004-08-12 Vical Incorporated. Method for freeze-drying nucleic acid/block copolymer/cationic surfactant complexes
US20040162256A1 (en) * 2002-12-23 2004-08-19 Vical Incorporated. Method for producing sterile polynucleotide based medicaments
US20070286869A1 (en) * 2004-05-18 2007-12-13 Vical Incorporated Influenza virus vaccine composition and methods of use

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7381422B2 (en) 2002-12-23 2008-06-03 Vical Incorporated Method for producing sterile polynucleotide based medicaments
US20040162256A1 (en) * 2002-12-23 2004-08-19 Vical Incorporated. Method for producing sterile polynucleotide based medicaments
US8435557B2 (en) 2002-12-23 2013-05-07 Vical Incorporated Method for producing sterile polynucleotide based medicaments
US20080287387A1 (en) * 2002-12-23 2008-11-20 Vical Incorporated Method for Producing Sterile Polynucleotide Based Medicaments
US20070010015A1 (en) * 2003-03-31 2007-01-11 Francisco Rodriguez Aguirre J Complete empty viral particles of infectious bursal disease virus (ibdv), production method thereof and applications of same
US20070128692A1 (en) * 2004-01-21 2007-06-07 Aguirre Jose Francisco R Chimeric empty capsids of the infectious bursal disease viruse (ibdv), obtainment process and applications
US20090208528A1 (en) * 2004-01-21 2009-08-20 Jose Francisco Rodriguez Aguirre Empty capsids (vlps(-vp4)) of the infectious bursal disease virus (ibdv), obtainment process and applications
US20070286869A1 (en) * 2004-05-18 2007-12-13 Vical Incorporated Influenza virus vaccine composition and methods of use
US20080057080A1 (en) * 2004-05-18 2008-03-06 Vical Incorporated Influenza virus vaccine composition and methods of use
US8128938B1 (en) 2004-05-18 2012-03-06 Vical Incorporated Influenza virus vaccine composition and methods of use
US8821890B2 (en) 2004-05-18 2014-09-02 Vical Incorporated Influenza virus vaccine composition and methods of use
US7537768B2 (en) 2004-05-18 2009-05-26 Vical Incorporated Influenza virus vaccine composition and methods of use
US7785603B2 (en) 2004-05-18 2010-08-31 Vical Incorporated Influenza virus vaccine composition and methods of use
US20110064755A1 (en) * 2004-12-03 2011-03-17 Vical Incorporated Methods for producing block copolymer/amphiphilic particles
US20060134221A1 (en) * 2004-12-03 2006-06-22 Vical Incorporated Methods for producing block copolymer/amphiphilic particles
US20090291472A1 (en) * 2005-02-24 2009-11-26 University Of Massachusetts, A Massachusetts Corporation Influenza nucleic acids, polypeptides, and uses thereof
US7566454B2 (en) * 2005-02-24 2009-07-28 University Of Massachusetts Influenza nucleic acids, polypeptides, and uses thereof
US20060217338A1 (en) * 2005-02-24 2006-09-28 Shan Lu Influenza nucleic acids, polypeptides, and uses thereof
US7476387B2 (en) 2005-07-15 2009-01-13 Chimera Pharma S.L.U. Chimeric empty viral-like particles derived from the infectious bursal disease virus (IBDV), process for their production and applications
US20070015243A1 (en) * 2005-07-15 2007-01-18 Consejo Superior De Investigaciones Cientificas And Bionostra S.L. Chimeric empty viral-like particles derived from the infectious bursal disease virus (IBDV), process for their production and applications
US10765734B2 (en) 2006-02-07 2020-09-08 Peptcell Limited Influenza peptides and compositions
US11439702B2 (en) 2006-02-07 2022-09-13 Peptcell Limited Influenza peptides and compositions
US20190365883A1 (en) * 2006-02-07 2019-12-05 Peptcell Limited Influenza Peptides and Compositions
US10279032B2 (en) 2006-02-07 2019-05-07 Peptcell Limited Peptide sequences and compositions
US10335480B2 (en) * 2006-02-07 2019-07-02 Peptcell Limited Peptide sequences and compositions
EP1998814A2 (en) * 2006-05-11 2008-12-10 Novavax, Inc. Novel influenza m2 vaccines
EP1998814A4 (en) * 2006-05-11 2010-06-16 Novavax Inc Novel influenza m2 vaccines
US20100143393A1 (en) * 2006-05-11 2010-06-10 Gale Smith Novel influenza m2 vaccines
US8293248B2 (en) 2006-07-20 2012-10-23 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US8263087B2 (en) 2006-07-20 2012-09-11 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US7935352B2 (en) 2006-07-20 2011-05-03 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US20100160419A1 (en) * 2006-07-20 2010-06-24 Vical Incorporated Compositions and Methods for Vaccinating Against HSV-2
US7879339B2 (en) 2006-07-20 2011-02-01 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US20100160418A1 (en) * 2006-07-20 2010-06-24 Vical Incorporated Compositions and Methods for Vaccinating Against HSV-2
US20100040651A1 (en) * 2006-07-20 2010-02-18 Vical Incorporated Compositions and Methods for Vaccinating Against HSV-2
US8840904B2 (en) 2006-07-20 2014-09-23 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US20100158949A1 (en) * 2006-07-20 2010-06-24 Vical Incorporated Compositions and Methods for Vaccinating Against HSV-2
US8852611B2 (en) 2006-07-20 2014-10-07 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US8852610B2 (en) 2006-07-20 2014-10-07 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US9161973B2 (en) 2006-07-20 2015-10-20 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US8840903B2 (en) 2006-07-20 2014-09-23 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US8828408B2 (en) 2006-07-20 2014-09-09 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US8834894B2 (en) 2006-07-20 2014-09-16 Vical Incorporated Compositions and methods for vaccinating against HSV-2
US20080299151A1 (en) * 2007-05-31 2008-12-04 Statens Serum Institut Influenza vaccines
US20100285050A1 (en) * 2007-10-05 2010-11-11 Isis Innovation Limited Compositions and Methods
EP2044947B1 (en) 2007-10-05 2017-03-08 Oxford University Innovation Limited MVA-based compositions and methods for inducing an immune response against influenza
US8133723B2 (en) 2007-11-12 2012-03-13 The Trustees Of The University Of Pennsylvania Vaccines against multiple subtypes of influenza virus
US20090169505A1 (en) * 2007-11-12 2009-07-02 Ruxandra Draghia-Akli Novel vaccines against multiple subtypes of influenza virus
WO2009073330A2 (en) * 2007-11-12 2009-06-11 The Trustees Of The University Of Pennsylvania Novel vaccines against multiple subtypes of influenza virus
WO2009073330A3 (en) * 2007-11-12 2010-01-07 The Trustees Of The University Of Pennsylvania Novel vaccines against multiple subtypes of influenza virus
AU2014268164B2 (en) * 2007-11-12 2016-09-29 Inovio Pharmaceuticals, Inc. Novel vaccines against multiple subtypes of influenza virus
KR101255419B1 (en) 2007-11-12 2013-04-17 이노비오 파마수티컬즈, 인크. Novel vaccines against multiple subtypes of influenza virus
US9592285B2 (en) 2007-11-12 2017-03-14 The Trustees Of The University Of Pennsylvania Vaccines against multiple subtypes of influenza virus
US10076565B2 (en) 2007-11-12 2018-09-18 The Trustees Of The University Of Pennsylvania Vaccines against multiple subtypes of influenza virus
US9764024B2 (en) 2008-11-28 2017-09-19 Statens Serum Institut Optimized influenza vaccines
US20110229518A1 (en) * 2008-11-28 2011-09-22 Statens Serum Institut Optimized influenza vaccines
US20160175431A1 (en) * 2009-11-09 2016-06-23 National Jewish Health Vaccine Composition
WO2012012461A3 (en) * 2010-07-19 2012-04-26 Shire Human Genetic Therapies, Inc. Mannose receptor c type 1 (mrc1) codon optimized cell line and uses thereof
US9453847B2 (en) 2010-07-19 2016-09-27 Shire Human Genetic Therapies, Inc. Mannose receptor C type 1 (MRC1) codon optimized cell line and uses thereof
US10131900B2 (en) * 2010-12-15 2018-11-20 Old Dominion University Research Foundation Electroporation-induced electrosensitization
US20130260435A1 (en) * 2010-12-15 2013-10-03 Old Dominion University Research Foundation Electroporation-induced electrosensitization
US10982206B2 (en) 2010-12-15 2021-04-20 Old Dominion University Research Foundation Electroporation-induced electrosensitization
US11820976B2 (en) 2010-12-15 2023-11-21 Old Dominion University Research Foundation Electroporation-induced electrosensitization
WO2022192524A1 (en) * 2021-03-10 2022-09-15 Elicio Therapeutics, Inc. Uses of amphiphiles in immune cell therapy and compositions therefor

Also Published As

Publication number Publication date
US7537768B2 (en) 2009-05-26
JP2008505660A (en) 2008-02-28
EP1766094A4 (en) 2009-11-25
CA2566355C (en) 2014-04-15
AU2005248361B2 (en) 2010-03-11
US20120128717A1 (en) 2012-05-24
JP5331340B2 (en) 2013-10-30
US8821890B2 (en) 2014-09-02
US20120045467A1 (en) 2012-02-23
US7785603B2 (en) 2010-08-31
CA2566355A1 (en) 2005-12-08
WO2005116270A2 (en) 2005-12-08
WO2005116270A3 (en) 2007-12-21
US20080057080A1 (en) 2008-03-06
US20070286869A1 (en) 2007-12-13
US20100197771A1 (en) 2010-08-05
EP1766094A2 (en) 2007-03-28
AU2005248361A1 (en) 2005-12-08
US8128938B1 (en) 2012-03-06

Similar Documents

Publication Publication Date Title
US8821890B2 (en) Influenza virus vaccine composition and methods of use
Chen et al. Advances in development and application of influenza vaccines
Lee et al. New vaccines against influenza virus
US8080642B2 (en) Severe acute respiratory syndrome DNA compositions and methods of use
US20180339037A1 (en) Oil-in-water emulsion influenza vaccine
US20070141078A1 (en) Influenza vaccine
US20080181911A1 (en) Use Of An Influenza Virus, An Oil-In-Water Emulsion Adjuvant, To Induce Cd4 T-Cell And/Or Improved B-Memory Cell Response
US9636394B2 (en) Process for producing influenza vaccine
US9060972B2 (en) Recombinant hemagglutinin protein of influenza virus and vaccine containing the same
US20230270843A1 (en) Post-Exposure Vaccination Against Viral Respiratory Infections
WO2007126788A2 (en) Methods and compositions for the treatment and prevention of viral infection
WO2024068545A1 (en) Influenza virus vaccines

Legal Events

Date Code Title Description
AS Assignment

Owner name: VICAL INCORPORATED, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LUKE, CATHERINE J.;VILALTA, ADRIAN;WLOCH, MARY K.;AND OTHERS;REEL/FRAME:017257/0881;SIGNING DATES FROM 20050910 TO 20051004

STCB Information on status: application discontinuation

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