RECOMBINANT SWINE INFLUENZA VIRUS AND USES THEREOF

Recombinant, chimeric porcine influenza viruses are disclosed that include hemagglutinin segments from more than one influenza virus subtype. Also described are methods of producing the recombinant influenza viruses, immunogenic compositions comprising the recombinant influenza viruses, methods of stimulating an immune response against influenza virus, and methods of treating and preventing influenza virus infection.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATION

This application claims the benefit under 35 U.S.C. §119(e)(1) to U.S. Provisional Application No. 61/514,156, filed Aug. 2, 2011, which application is incorporated herein by reference in its entirety.

TECHNICAL FIELD

The present invention pertains generally to influenza virus and immunogenic compositions and methods for treating and preventing influenza infection. In particular, the invention relates to recombinant, chimeric swine influenza viruses expressing more than one hemagglutinin (HA) subtype.

BACKGROUND

Swine influenza (SI) is an acute respiratory disease of swine caused by type A and type C influenza viruses. Influenza A viruses are segmented negative-strand RNA viruses and can be isolated from a number of other animal host species, including birds, humans, horses, whales, and mink. Although whole influenza viruses rarely cross the species barrier, gene segments can cross this barrier through the process of genetic reassortment, or genetic shift. Pigs support the replication of both human and avian influenza A viruses and have been postulated to play an important role in interspecies transmission by acting as a “mixing vessel” for reassortment between viruses specific to different host species (Scholtissek, Eur. J. Epidemiol. (1994) 10:455-458). This may lead to the generation of influenza viruses capable of crossing the species barrier to humans.

Influenza virions include an internal ribonucleoprotein core (a helical nucleocapsid) containing the single-stranded RNA genome, and an outer lipoprotein envelope lined inside by a matrix protein (M1). The genome of influenza A virus consists of eight segmented negative sense single-stranded RNA molecules. Each segment possesses segment-specific RNA packaging signals which are composed of both the noncoding regions and short coding regions at both 5′ and 3′ ends. The eight segmented RNAs encode 11 viral proteins, including RNA-dependent RNA polymerase proteins (PB2, PB1 and PA) and nucleoprotein (NP) which form the nucleocapsid; the matrix membrane proteins (M1, M2); hemagglutinin (HA) and neuraminidase (NA), both surface glycoproteins which project from the lipid-containing envelope; the nonstructural protein (NS1), nuclear export protein (NEP, also termed NS2), the proapoptotic factor PB1-F2. HA is critical for virus binding and entry to the cells, and is the major neutralizing antibody target, whereas NA plays a role in progeny virus release and is essential for virus propagation. Transcription and replication of the genome take place in the nucleus and assembly occurs via budding on the plasma membrane. The viruses can reassort genes during mixed infections.

Multiple swine influenza virus (SIV) subtypes continue to circulate in swine populations despite available vaccines. Currently, H1N1, H3N2, and H1N2 are the dominant subtypes that cause disease in the North American swine population. SIVs of the subtype H3N2 were generated by reassortment between human, avian and classical swine viruses, are undergoing rapid evolution and in general cause more severe disease than classical H1N1 SIV. Current SIV vaccines do not provide cross-protection against multiple antigenic SIV variants.

Thus, there remains a need for the development of effective strategies for the treatment and prevention of swine influenza infection.

SUMMARY OF THE INVENTION

The present invention relates to recombinant, chimeric influenza viruses that possess HAs from two or more subtypes of SIVs and methods of producing and using the same. In preferred embodiments, all or a portion of the NA segment is absent from the recombinant virus such that virus propagation is hindered. Because NA is essential for virus propagation, the function of NA can be provided in culture by growing the virus in the presence of sialidase. The recombinant virus that expresses more than one HA type can be used in immunogenic compositions to stimulate an immune response against influenza virus, and for treating and preventing influenza virus infection. Because HAs from different subtypes of SIVs are present, compositions including the chimeric influenza viruses can be used to provide broad coverage against a number of influenza strains.

In particular, the inventors herein have found that a chimeric virus including both H1 and H3, and retaining NA 3′ and 5′ viral RNA-specific packaging signals but lacking the remainder of the NA segment, grows efficiently in culture and is attenuated in pigs as no sialidase is present in swine. The NA packaging signals are largely retained for efficient packaging. Such chimeric constructs can be used as effective and safe live, attenuated vaccines.

Accordingly, in one embodiment, the invention is directed to a recombinant, chimeric porcine influenza virus comprising more than one hemagglutinin (HA) segment (segment 5) from more than one influenza subtype. In particular, the virus comprises segments 1-5, 7 and 8 from a first influenza subtype and a second segment 5 from a second influenza subtype. Further, all or a portion of the neuraminidase (NA) segment (segment 6) of the first influenza subtype is missing to render an attenuated virus.

In certain embodiments, the second segment 5 comprises NA packaging sequences from said first influenza subtype located 3′ and optionally 5′ to said second segment 5. In additional embodiments, the NA packaging sequences comprise 3′ NA packaging sequences from the 3′ NA UTR and the 3′ NA coding sequence and, optionally 5′ NA packaging sequences from the 5′ NA UTR and the 5′ NA coding sequence.

In further embodiments, the influenza virus described above is from an influenza A virus. In certain embodiments, the influenza virus comprises an HA segment from an H1N1 subtype and an HA segment from an H3N2 subtype. In certain embodiments, the first influenza subtype is H1N1, such as A/swine/Saskatchewan/18789/02. In other embodiments, the second influenza subtype is H3N2, such as A/Swine/Texas/4199-2/98.

In yet additional embodiments, the invention is directed to an attenuated, recombinant, porcine influenza virus comprising segments 1-5, 7 and 8 from an H1N1 influenza subtype, and segment 5 from an H3N2 influenza subtype. Further, all or a portion of segment 6 from the H1N1 influenza subtype is missing and the H3N2 segment 5 is flanked by NA packaging sequences from the H1N1 subtype. The packaging sequences comprise 3′ NA packaging sequences from the 3′ NA UTR and the 3′ NA coding sequence and 5′ NA packaging sequences from the 5′ NA UTR and the 5′ NA coding sequence. In certain embodiments, the H1N1 subtype is A/swine/Saskatchewan/18789/02 and the H3N2 subtype is A/Swine/Texas/4199-2/98.

In further embodiments, the invention is directed to a composition comprising any one of the recombinant viruses described above, and a pharmaceutically acceptable excipient. In certain embodiments, the composition further comprises an adjuvant. In yet additional embodiments, the invention is directed to a method of eliciting an immunological response in a vertebrate subject, comprising administering the composition to the subject. In other embodiments, the invention is directed to a method of treating or preventing an influenza infection in a vertebrate subject, comprising administering to the subject a therapeutically effective amount of the composition. In other embodiments, the invention is directed to a method of vaccinating a subject against an influenza virus, comprising administering an effective amount of the composition to the subject. In certain embodiments, the subject is a porcine subject.

In additional embodiments, the invention is directed to a recombinant construct comprising: (a) a porcine influenza H3N2 subtype HA segment; and (b) porcine influenza H1N1 subtype NA packaging sequences located 3′ and optionally 5′ to said H3N2 HA segment. In certain embodiments, the H3N2 HA segment is flanked by H1N1 NA packaging sequences that comprise 3′ NA packaging sequences from the 3′ NA UTR and the 3′ NA coding sequence and 5′ NA packaging sequences from the 5′ NA UTR and the 5′ NA coding sequence. In additional embodiments the H1N1 subtype is A/swine/Saskatchewan/18789/02 and the H3N2 subtype is A/Swine/Texas/4199-2/98.

In further embodiments, the invention is directed to a method of producing a recombinant, chimeric influenza virus, comprising transfecting a host cell with (a) individual plasmids comprising segments 1-5, 7 and 8 from an H1N1 influenza subtype; and (b) a recombinant construct described above, and culturing the host cell under conditions that result in the production of the recombinant, chimeric influenza virus.

In other embodiments, the invention is directed to a cell transformed with (a) individual plasmids comprising segments 1-5, 7 and 8 from an H1N1 influenza subtype; and (b) a recombinant construct as described above.

In further embodiments, the invention is directed to a method of producing a composition comprising combining any of the recombinant, chimeric porcine influenza viruses described above with a pharmaceutically acceptable excipient.

In other embodiments, the invention is directed to a method of producing an influenza vaccine comprising: (a) propagating any one of the recombinant, chimeric porcine influenza viruses described above; (b) purifying the virus; and (c) combining the purified virus with a pharmaceutically acceptable excipient.

In yet additional embodiments, the invention is directed to a kit comprising one or more containers of any one of the recombinant viruses described above, or the compositions described above.

These and other embodiments of the subject invention will readily occur to those of skill in the art in view of the disclosure herein.

BRIEF DESCRIPTION OF THE FIGURES

FIGS. 1A and 1B depict various influenza segments for use in the present invention. FIG. 1A depicts the 8 segments of a wild-type Influenza A H1N1 SIV virus, A/swine/Saskatchewan/18789/02 (termed “SK02” herein) and the 8 segments of the recombinant, chimeric attenuated virus produced as described in the examples (termed “SIV-606” herein). FIG. 1B is a schematic representation of the segment termed “H3-HA” in FIG. 1A. The HA segment depicted in FIG. 1B was derived from an H3N2 Influenza A virus, A/Swine/Texas/4199-2/98 (termed “Tx98” herein) and included the 3′ and 5′ NA packaging signals from SK02.

FIG. 2 shows the growth curves of SIV-606 and SIV/SK02.

FIGS. 3A-3C show the body temperature of pigs infected with high and low doses of SK02 (FIG. 3A), Tx98 (FIG. 3B) and SIV-606 (FIG. 3C).

FIG. 4 shows the lung virus titers of pigs infected with high and low doses of SIV/SK02, SIV Tx98 and SIV-606.

FIGS. 5A and 5B (SEQ ID NOS:1 and 2) show the nucleotide sequence and amino acid sequence, respectively, of HA from SIV SK02 (GenBank: AY619961.1).

FIGS. 6A and 6B (SEQ ID NOS:3 and 4) show the nucleotide sequence and amino acid sequence, respectively, of NA from SIV SK02 (GenBank: AY619960.1).

FIGS. 7A-7C (SEQ ID NOS:5, 6 and 7) show the matrix nucleotide sequence (FIG. 7A) and the amino acid sequences of M2 (FIG. 7B) and M1 (FIG. 7C) from SIV SK02 (GenBank: AY619959.1).

FIGS. 8A and 8B (SEQ ID NOS:8 and 9) show the nucleotide sequence and amino acid sequence, respectively, of NP from SIV SK02 (GenBank: AY619958.1).

FIGS. 9A-9C (SEQ ID NOS:10, 11 and 12) show the nonstructural protein nucleotide sequence (FIG. 9A) and the amino acid sequences of NEP (FIG. 9B) and NS1 (FIG. 9C) from SIV SK02 (GenBank: AY619957.1).

FIGS. 10A and 10B (SEQ ID NOS:13 and 14) show the nucleotide sequence and amino acid sequence, respectively, of PA from SIV SK02 (GenBank: AY619956).

FIGS. 11A and 11B (SEQ ID NOS:15 and 16) show the nucleotide sequence and amino acid sequence, respectively, of PB1 from SIV SK02 (GenBank: AY619955.1).

FIGS. 12A and 12B (SEQ ID NOS:17 and 18) show the nucleotide sequence and amino acid sequence, respectively, of PB2 from SIV SK02 (GenBank: AY619954.1).

FIGS. 13A and 13B (SEQ ID NOS:19 and 20) show the nucleotide sequence and amino acid sequence, respectively, of HA from SIV Tx98.

FIGS. 14A-14C show SIV/SK02-specific serum IgG titers (FIG. 14A); SIV/Tx98-specific serum IgG titers (FIG. 14B); and H1N1 Halifax-specific serum IgG titers (FIG. 14C) in pigs vaccinated with SIV-606.

FIGS. 15A-15C show SIV/SK02-specific nasal IgA titers (FIG. 15A); SIV/Tx98-specific nasal IgA titers (FIG. 15B); and H1N1 Halifax-specific nasal IgA titers (FIG. 15C) in pigs vaccinated with SIV-606.

FIGS. 16A-16C show SIV/SK02-specific BALF IgA titers (FIG. 16A); SIV/Tx98-specific BALF IgA titers (FIG. 16B); and H1N1 Halifax-specific BALF IgA titers (FIG. 16C) in pigs vaccinated with SIV-606.

FIGS. 17A and 17B show rectal temperature in unvaccinated control pigs and SIV-606 vaccinated pigs challenged with SIV/SK02 (FIG. 17A) and challenged with SIV/Tx98 (FIG. 17B).

FIGS. 18A and 18B show the percentage of lung lesions (FIG. 18A) and lung viral load (FIG. 18B) in unvaccinated SIV/SK02 and SIV/Tx98 pigs, as well as in pigs vaccinated with SIV-606.

FIGS. 19A-19E show histopathological lesions in unvaccinated, unchallenged pigs (FIG. 19A); MEM vaccinated and challenged pigs (FIG. 19B); SIV-606 vaccinated and SIV/SK02 challenged pigs (FIG. 19C); MEM vaccinated and SIV/Tx98 challenged pigs (FIG. 19D); and SIV-606 vaccinated and SIV/Tx98 challenged pigs (FIG. 19E).

DETAILED DESCRIPTION OF THE INVENTION

The practice of the present invention will employ, unless otherwise indicated, conventional methods of virology, chemistry, biochemistry, recombinant DNA techniques and immunology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Fundamental Virology, Current Edition, vol. I & II (B. N. Fields and D. M. Knipe, eds.); Handbook of Experimental Immunology, Vols. I-IV (D. M. Weir and C. C. Blackwell eds., Blackwell Scientific Publications); T. E. Creighton, Proteins: Structures and Molecular Properties (W.H. Freeman and Company); A. L. Lehninger, Biochemistry (Worth Publishers, Inc., current edition); Sambrook, et al., Molecular Cloning: A Laboratory Manual (current edition); Methods In Enzymology (S. Colowick and N. Kaplan eds., Academic Press, Inc.).

All publications, patents and patent applications cited herein, whether supra or infra, are hereby incorporated by reference in their entireties.

The following amino acid abbreviations are used throughout the text:

    • Alanine: Ala (A) Arginine: Arg (R)
    • Asparagine: Asn (N) Aspartic acid: Asp (D)
    • Cysteine: Cys (C) Glutamine: Gln (O)
    • Glutamic acid: Glu (E) Glycine: Gly (G)
    • Histidine: H is (H) Isoleucine: Ile (I)
    • Leucine: Leu (L) Lysine: Lys (K)
    • Methionine: Met (M) Phenylalanine: Phe (F)
    • Proline: Pro (P) Serine: Ser (S)
    • Threonine: Thr (T) Tryptophan: Trp (W)
    • Tyrosine: Tyr (Y) Valine: Val (V)

1. DEFINITIONS

In describing the present invention, the following terms will be employed, and are intended to be defined as indicated below.

It must be noted that, as used in this specification and the appended claims, the singular forms “a”, “an” and “the” include plural referents unless the content clearly dictates otherwise. Thus, for example, reference to “an influenza A virus” includes a mixture of two or more such viruses, and the like.

As used herein, the term “influenza virus” refers to members of the orthomyxoviridae family of enveloped viruses with a segmented antisense RNA genome (Knipe and Howley (eds.) Fields Virology, 4th edition, Lippincott Williams and Wilkins, Philadelphia, Pa., 2001). The term influenza virus may include any strain of influenza virus, such as influenza A, B, or C, which is capable of causing disease in an animal or human subject. In particular, the term encompasses any subtype of influenza A virus selected from H1-H15 and N1-N9, such as but not limited to H1N1, H1N2, H3N2, H3N1, H9N2 and H5N1, or any combination of H's and N's. A large number of influenza isolates have been partially or completely sequenced. See, e.g., the Influenza Sequence Database (ISD) (website at flu.lanl.gov; described by Macken et al., “The value of a database in surveillance and vaccine selection.” in Options for the Control of Influenza IV. A. D. M. E. Osterhaus, N. Cox & A. W. Hampson (Eds.) Amsterdam: Elsevier Science, 2001, 103-106) and the GenBank database, particularly the Influenza Virus Resource (website at ncbi.nlm.nih.gov/genomes/FLU/FLU.html). The ISD and GenBank databases contain complete sequences for influenza A, B and C genome segments.

The term “derived from” is used herein to identify the original source of a molecule but is not meant to limit the method by which the molecule is made which can be, for example, by chemical synthesis or recombinant means.

An influenza virus molecule is a molecule derived from an influenza virus, including, without limitation, polypeptide, protein, polynucleotide, oligonucleotide, and nucleic acid molecules, as defined herein, from any of the various isolates of influenza subtypes A, B, or C. The molecule need not be physically derived from the particular isolate in question, but may be synthetically or recombinantly produced.

Nucleic acid and polypeptide sequences for a number of influenza virus isolates are known. Representative influenza sequences are presented in FIGS. 5-13 herein. Additional representative sequences, including additional sequences for the 8 influenza segments, including those segments coding for hemagglutinin (HA), neuraminidase (NA), polymerase acidic protein (PA), polymerase basic proteins 1 and 2 (PB1 and PB2), matrix membrane proteins 1 and 2 (M1 and M2), nucleoprotein (NP), and nonstructural proteins 1 and 2 (NS1 and NEP, also termed NS2) from influenza isolates found in various species are listed in the National Center for Biotechnology Information (NCBI) database and the Influenza Research Database found at fludb.org. See also Ferguson et al. (2003) Nature 422: 428-433; Lin et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97, 9654-9658; Nguyen et al. (2005) J. Virol. 79:4201-4212; Ha et al. (2002) EMBO J. 21:865-875; and Chan et al. (2004) J. Microbiol. Immunol. Infect. 37:135-144; for sequence comparisons and a discussion of genetic diversity and phylogenetic analysis of influenza virus.

As used herein, the term “swine influenza virus” refers to a type A or type C influenza virus from the family orthomyxovirus that causes swine influenza. While orthomyxovirus has three groups: type A, type B and type C, only type A and type C influenza viruses infect pigs. Subtypes of swine influenza virus include H1N1, H1N2, H3N2, H3N1, H9N2 and H5N1. In certain embodiments, a swine influenza virus is an influenza virus that has been isolated from swine. For purposes of the present invention, a swine influenza virus is either a wild-type swine influenza virus or a recombinant, chimeric influenza virus derived from a wild-type swine influenza virus.

As used herein, the phrase “wild-type swine influenza virus” refers to the types of a swine virus that are prevalent, circulating naturally and producing typical outbreaks of disease. Examples of wild-type swine influenza viruses include, but are not limited to, A/swine/Saskatchewan/18789/02, A/Swine/Colorado/1/77, A/Swine/Colorado/23619/99, A/Swine/Cote d'Armor/3633/84, A/Swine/Cote d'Armor/3633/84, A/Swine/England/195852/92, A/Swine/Finistere/2899/82, A/Swine/Hong Kong/10/98, A/Swine/Hong Kong/9/98, A/Swine/Hong Kong/81/78, A/Swine/Illinois/100084/01, A/Swine/Illinois/100085A/01, A/Swine/Illinois/21587/99, A/Swine/Indiana/1726/88, A/Swine/Indiana/9K035/99, A/Swine/Indiana/P 12439/00, A/Swine/Iowa/30, A/Swine/Iowa/15/30, A/Swine/Iowa/533/99, A/Swine/Iowa/569/99, A/Swine/Iowa/3421/90, A/Swine/Iowa/8548-1/98, A/Swine/Iowa/930/01, A/Swine/Iowa/17672/88, A/Swine/Italy/1513-1/98, A/Swine/Italy/1523/98, A/Swine/Korea/CY02/02, A/Swine/Minnesota/55551/00, A/Swine/Minnesota/593/99, A/Swine/Minnesota/9088-2/98, A/Swine/Nebraska/1/92, A/Swine/Nebraska/209/98, A/Swine/Netherlands/12/85, A/Swine/North Carolina/16497/99, A/Swine/North Carolina/35922/98, A/Swine/North Carolina/93523/01, A/Swine/North Carolina/98225/01, A/Swine/Oedenrode/7C/96, A/Swine/Ohio/891/01, A/Swine/Oklahoma/18717/99, A/Swine/Oklahoma/18089/99, A/Swine/Ontario/01911-1/99, A/Swine/Ontario/01911-2/99, A/Swine/Ontario/41848/97, A/Swine/Ontario/97, A/Swine/Quebec/192/81, A/Swine/Quebec/192/91, A/Swine/Quebec/5393/91, A/Swine/Taiwan/7310/70, A/Swine/Tennessee/24/77, A/Swine/Texas/4199-2/98, A/Swine/Wisconsin/125/97, A/Swine/Wisconsin/136/97, A/Swine/Wisconsin/163/97, A/Swine/Wisconsin/164/97, A/Swine/Wisconsin/166/97, A/Swine/Wisconsin/168/97, A/Swine/Wisconsin/235/97, A/Swine/Wisconsin/238/97, A/Swine/Wisconsin/457/98, A/Swine/Wisconsin/458/98, A/Swine/Wisconsin/464/98 and A/Swine/Wisconsin/14094/99.

The term “HA gene” refers to the gene which encodes the hemagglutinin (HA) surface glycoprotein which projects from the lipid-containing envelope in influenza. HA is one of the molecules encoded by the segmented genome of influenza A and other viruses. A “swine influenza virus HA gene” is an HA gene isolated from a swine influenza virus, such as from any of the strains described above. The polynucleotide and amino acid sequences of representative swine HA genes can be found in public sequence databases such as GenBank. For example, HA genes from H1N1 and include, but are not limited to, GenBank Accession Nos. AY619961.1 (see FIGS. 5A and 5B); GQ457549.1; GQ457548.1; GQ457547.1; CY091769.1; CY091745.1; CY091737.1; CY091729.1; GU721143.3; JF820285.1; JF820277.1; JF707784.1; CY087136.1; CY087104.1; CY087096.1; CY087080.1; CY087072.1; CY087064.1; CY087056.1; CY087048.1; CY086863.1; CY086839.1; CY086353.1; CY086006.1; CY085990.1; CY085982.1; CY085974.1; CY085966.1; CY085958.1; CY085950.1; CY085942.1; CY085934.1; CY085926.1; CY085918.1; CY085910.1; CY085902.1; CY085894.1; CY085886.1; CY085878.1; CY085870.1; CY085854.1; CY085846.1; CY085838.1; CY085830.1; CY085822.1; CY085814.1; CY085806.1; CY085798.1; CY085790.1; CY085782.1; CY085774.1; CY085766.1; CY085758.1; CY085742.1; CY085726.1; CY085718.1; CY085710.1; CY085702.1; CY085694.1; CY085686.1; CY085670.1; JF833344.1; JF833343.1; JF833341.1; JF833339.1; JF833338.1; JF833337.1; JF833335.1; JF916682.1; JF812292.1; JF812291.1; JF812290.1; JF812287.1; JF812284.1; JF812281.1; JF812280.1; JF812279.1; JF812278.1; JF812273.1; JF812272.1; JF812271.1; AF091317.1; AF091315.1; AF091314.1.

HA genes from H3N2 and include, but are not limited to, the sequence shown in FIGS. 13A and 13B; as well as GenBank Accession Nos. AY377927.2; CY092324.1; AF153233.1; JN105973.1; HQ315643.1; FJ519977.1; FJ519976.1; FJ519975.1; FJ519974.1; FJ519973.1; FJ519972.1; FJ519971.1; GU937743.1; JF833345.1; JF833340.1; JF833336.1; JF833334.1; JF812293.1; JF812289.1; JF812277.1; JF812276.1; JF812275.1; JF812274.1; CY045575.1; CY045567.1; CY045559.1; CY045551.1; HQ825243.1; HQ825235.1; HQ825229.1; HQ825226.1; HQ825223.1; HQ825218.1; HQ825210.1; HQ825210.1; HQ825198.1; HQ825190.1; HQ825182.1; HQ825174.1; HQ825166.1; JF312065.1; JF312064.1; CY086920.1; JF312073.1; JF312072.1; JF312071.1; JF316643.1; JF263536.1; JF263535.1; HQ734204.1; HQ734201.1; HQ734198.1; HQ734195.1; HQ734192.1; HQ734189.1; HQ734186.1; CY077942.1; CY077934.1.

The term “NA gene” refers to the gene which encodes the neuraminidase (NA) surface glycoprotein which projects from the lipid-containing envelope in influenza. NA is one of the molecules encoded by the segmented genome of influenza A and other viruses. A “swine influenza virus NA gene” is an NA gene isolated from a swine influenza virus, such as from any of the strains described above. The polynucleotide and amino acid sequences of representative swine NA genes can be found in public sequence databases such as GenBank. For example, NA genes from H1N1 and include, but are not limited to, AY619960.1 (see FIGS. 6A and 6B); JF833356.1.; JF833355.1; JF833353.1; JF833351.1; JF833350.1; JF833349.1; JF833355.1; JF833347.1; JF812315.1; JF812314.1; JF812313.1; JF812310.1; JF812307.1; JF812304.1; JF812303.1; JF812302.1; JF812301.1; JF812294.1; FJ791299.1; FJ791298.1; FJ791297.1; FJ791296.1; FJ791295.1; FJ791294.1; FJ791293.1; FJ791292.1; FJ791291.1; FJ791290.1; FJ791289.1; FJ791288.1; FJ791287.1.

The term “NA packaging signal” refers to the 3′ and 5′ viral RNA-specific packaging signals for NA that provide for efficient incorporation of viral RNA into viral particles. The packaging signals are present in the 5′ and 3′ untranslated regions (UTRs) and extend into the coding region of the NA segment. Preferably, the NA packaging signals used in the production of the recombinant, chimeric viruses will include only so much of the NA region sufficient for packaging and will not include the entire NA coding sequence. NA packaging signals are discussed in greater detail below.

As used herein, the phrase “multiplicity of infection” or “MOI” is the average number of virus per infected cell. The MOI is determined by dividing the number of virus added (ml added x PFU) by the number of cells added (ml added x cells/ml).

As used herein, the term “attenuated” means that an influenza virus variant, such as a recombinant, chimeric virus described herein, exhibits a measurable reduction in replication efficiency relative to wild-type influenza virus. The replication efficiency of an influenza virus may be determined, for example, by measuring plaque size in MDCK cells, by measuring virus titers over multiple growth cycles, or by isolating virus from infected lung tissue and measuring titers.

The terms “polypeptide” and “protein” refer to a polymer of amino acid residues and are not limited to a minimum length of the product. Thus, peptides, oligopeptides, dimers, multimers, and the like, are included within the definition. Both full-length proteins and fragments thereof are encompassed by the definition. The terms also include postexpression modifications of the polypeptide, for example, glycosylation, acetylation, phosphorylation and the like. Furthermore, for purposes of the present invention, a “polypeptide” refers to a protein which includes modifications, such as deletions, additions and substitutions, to the native sequence, so long as the protein maintains the desired activity. These modifications may be deliberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the proteins or errors due to PCR amplification.

“Substantially purified” generally refers to isolation of a substance (recombinant virus, compound, polynucleotide, protein, polypeptide, polypeptide composition) such that the substance comprises the majority percent of the sample in which it resides. Typically in a sample, a substantially purified component comprises 50%, preferably 80%-85%, more preferably 90-95% of the sample. Techniques for purifying molecules of interest are well-known in the art and include, for example, ion-exchange chromatography, affinity chromatography and sedimentation according to density.

By “isolated” is meant, when referring to a polypeptide, that the indicated molecule is separate and discrete from the whole organism with which the molecule is found in nature or is present in the substantial absence of other biological macro-molecules of the same type. The term “isolated” with respect to a polynucleotide is a nucleic acid molecule devoid, in whole or part, of sequences normally associated with it in nature; or a sequence, as it exists in nature, but having heterologous sequences in association therewith; or a molecule disassociated from the chromosome.

“Homology” refers to the percent identity between two polynucleotide or two polypeptide moieties. Two nucleic acid, or two polypeptide sequences are “substantially homologous” to each other when the sequences exhibit at least about 50% sequence identity, preferably at least about 75% sequence identity, more preferably at least about 80%-85% sequence identity, more preferably at least about 90% sequence identity, and most preferably at least about 95%-98% sequence identity over a defined length of the molecules. As used herein, substantially homologous also refers to sequences showing complete identity to the specified sequence.

In general, “identity” refers to an exact nucleotide-to-nucleotide or amino acid-to-amino acid correspondence of two polynucleotides or polypeptide sequences, respectively. Percent identity can be determined by a direct comparison of the sequence information between two molecules by aligning the sequences, counting the exact number of matches between the two aligned sequences, dividing by the length of the shorter sequence, and multiplying the result by 100. Readily available computer programs can be used to aid in the analysis, such as ALIGN, Dayhoff, M. O. in Atlas of Protein Sequence and Structure M. O. Dayhoff ed., 5 Suppl. 3:353-358, National biomedical Research Foundation, Washington, D.C., which adapts the local homology algorithm of Smith and Waterman Advances in Appl. Math. 2:482-489, 1981 for peptide analysis. Programs for determining nucleotide sequence identity are available in the Wisconsin Sequence Analysis Package, Version 8 (available from Genetics Computer Group, Madison, Wis.) for example, the BESTFIT, FASTA and GAP programs, which also rely on the Smith and Waterman algorithm. These programs are readily utilized with the default parameters recommended by the manufacturer and described in the Wisconsin Sequence Analysis Package referred to above. For example, percent identity of a particular nucleotide sequence to a reference sequence can be determined using the homology algorithm of Smith and Waterman with a default scoring table and a gap penalty of six nucleotide positions.

Another method of establishing percent identity in the context of the present invention is to use the MPSRCH package of programs copyrighted by the University of Edinburgh, developed by John F. Collins and Shane S. Sturrok, and distributed by IntelliGenetics, Inc. (Mountain View, Calif.). From this suite of packages the Smith-Waterman algorithm can be employed where default parameters are used for the scoring table (for example, gap open penalty of 12, gap extension penalty of one, and a gap of six). From the data generated the “Match” value reflects “sequence identity.” Other suitable programs for calculating the percent identity or similarity between sequences are generally known in the art, for example, another alignment program is BLAST, used with default parameters. For example, BLASTN and BLASTP can be used using the following default parameters: genetic code=standard; filter=none; strand=both; cutoff=60; expect=10; Matrix=BLOSUM62; Descriptions=50 sequences; sort by ═HIGH SCORE; Databases=non-redundant, GenBank+EMBL+DDBJ+PDB+GenBank CDS translations+Swiss protein+Spupdate+PIR. Details of these programs are readily available.

Alternatively, homology can be determined by hybridization of polynucleotides under conditions which form stable duplexes between homologous regions, followed by digestion with single-stranded-specific nuclease(s), and size determination of the digested fragments. DNA sequences that are substantially homologous can be identified in a Southern hybridization experiment under, for example, stringent conditions, as defined for that particular system. Defining appropriate hybridization conditions is within the skill of the art. See, e.g., Sambrook et al., supra; DNA Cloning, supra; Nucleic Acid Hybridization, supra.

The teens “polynucleotide,” “oligonucleotide,” “nucleic acid” and “nucleic acid molecule” are used herein to include a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. This term refers only to the primary structure of the molecule. Thus, the term includes triple-, double- and single-stranded DNA, as well as triple-, double- and single-stranded RNA. It also includes modifications, such as by methylation and/or by capping, and unmodified forms of the polynucleotide. More particularly, the terms “polynucleotide,” “oligonucleotide,” “nucleic acid” and “nucleic acid molecule” include polydeoxyribonucleotides (containing 2-deoxy-D-ribose), polyribonucleotides (containing D-ribose), any other type of polynucleotide which is an N- or C-glycoside of a purine or pyrimidine base, and other polymers containing normucleotidic backbones, for example, polyamide (e.g., peptide nucleic acids (PNAs)) and polymorpholino (commercially available from the Anti-Virals, Inc., Corvallis, Oreg., as Neugene) polymers, and other synthetic sequence-specific nucleic acid polymers providing that the polymers contain nucleobases in a configuration which allows for base pairing and base stacking, such as is found in DNA and RNA. There is no intended distinction in length between the terms “polynucleotide,” “oligonucleotide,” “nucleic acid” and “nucleic acid molecule,” and these terms will be used interchangeably. Thus, these terms include, for example, 3′-deoxy-2′,5′-DNA, oligodeoxyribonucleotide N3′ P5′ phosphoramidates, 2′-O-alkyl-substituted RNA, double- and single-stranded DNA, as well as double- and single-stranded RNA, DNA:RNA hybrids, and hybrids between PNAs and DNA or RNA, and also include known types of modifications, for example, labels which are known in the art, methylation, “caps,” substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as, for example, those with uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), with negatively charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), and with positively charged linkages (e.g., aminoalklyphosphoramidates, aminoalkylphosphotriesters), those containing pendant moieties, such as, for example, proteins (including nucleases, toxins, antibodies, signal peptides, poly-L-lysine, etc.), those with intercalators (e.g., acridine, psoralen, etc.), those containing chelators (e.g., metals, radioactive metals, boron, oxidative metals, etc.), those containing alkylators, those with modified linkages (e.g., alpha anomeric nucleic acids, etc.), as well as unmodified forms of the polynucleotide or oligonucleotide. In particular, DNA is deoxyribonucleic acid.

A polynucleotide “derived from” a designated sequence refers to a polynucleotide sequence which comprises a contiguous sequence of approximately at least about 6 nucleotides, preferably at least about 8 nucleotides, more preferably at least about 10-12 nucleotides, and even more preferably at least about 15-20 nucleotides corresponding, i.e., identical or complementary to, a region of the designated nucleotide sequence. The derived polynucleotide will not necessarily be derived physically from the nucleotide sequence of interest, but may be generated in any manner, including, but not limited to, chemical synthesis, replication, reverse transcription or transcription, which is based on the information provided by the sequence of bases in the region(s) from which the polynucleotide is derived. As such, it may represent either a sense or an antisense orientation of the original polynucleotide.

“Recombinant” as used herein to describe a nucleic acid molecule means a polynucleotide of genomic, RNA, cDNA, viral, semisynthetic, or synthetic origin which, by virtue of its origin or manipulation is not associated with all or a portion of the polynucleotide with which it is associated in nature. The term “recombinant” as used with respect to a virus, means a virus produced by manipulation of the viral genome.

“Recombinant host cells,” “host cells,” “cells,” “cell lines,” “cell cultures,” and other such terms denoting microorganisms or higher eukaryotic cell lines cultured as unicellular entities refer to cells which can be, or have been, used as recipients for recombinant viruses and vectors or other transferred nucleic acid, and include the original progeny of the original cell which has been transfected.

A “coding sequence” or a sequence which “encodes” a selected polypeptide, is a nucleic acid molecule which is transcribed and translated into a polypeptide in vivo when placed under the control of appropriate regulatory sequences (or “control elements”). The boundaries of the coding sequence can be determined by a start codon at the 5′ (amino) terminus and a translation stop codon at the 3′ (carboxy) terminus. A coding sequence can include, but is not limited to, RNA or cDNA from viral, procaryotic or eucaryotic mRNA, genomic DNA sequences from viral or procaryotic DNA, and even synthetic DNA sequences. A transcription termination sequence may be located 3′ to the coding sequence.

Typical “control elements” include, but are not limited to, transcription promoters, transcription enhancer elements, transcription termination signals, polyadenylation sequences (located 3′ to the translation stop codon), sequences for optimization of initiation of translation (located 5′ to the coding sequence), and translation termination sequences.

“Operably linked” refers to an arrangement of elements wherein the components so described are configured so as to perform their usual function. Thus, a given promoter operably linked to a coding sequence is capable of effecting the expression of the coding sequence when the proper enzymes are present. The promoter need not be contiguous with the coding sequence, so long as it functions to direct the expression thereof. Thus, for example, intervening untranslated yet transcribed sequences can be present between the promoter sequence and the coding sequence and the promoter sequence can still be considered “operably linked” to the coding sequence.

“Encoded by” refers to a nucleic acid sequence which codes for a polypeptide sequence, wherein the polypeptide sequence or a portion thereof contains an amino acid sequence of at least 3 to 5 amino acids, more preferably at least 8 to 10 amino acids, and even more preferably at least 15 to 20 amino acids from a polypeptide encoded by the nucleic acid sequence.

“Expression cassette” or “expression construct” refers to an assembly which is capable of directing the expression of the sequence(s) or gene(s) of interest. An expression cassette generally includes control elements, as described above, such as a promoter which is operably linked to (so as to direct transcription of) the sequence(s) or gene(s) of interest, and often includes a polyadenylation sequence as well. An expression cassette may be contained within a plasmid construct. In addition to the components of the expression cassette, the plasmid construct may also include, one or more selectable markers, a signal which allows the plasmid construct to exist as single-stranded DNA (e.g., a M13 origin of replication), at least one multiple cloning site, and a “mammalian” origin of replication (e.g., a SV40 or adenovirus origin of replication).

The term “transfection” is used to refer to the uptake of foreign nucleic acid by a cell. A cell has been “transfected” when exogenous nucleic acid has been introduced inside the cell membrane. A number of transfection techniques are generally known in the art. See, e.g., Graham et al. (1973) Virology, 52:456, Sambrook et al. (1989) Molecular Cloning, a laboratory manual, Cold Spring Harbor Laboratories, New York, Davis et al. (1986) Basic Methods in Molecular Biology, Elsevier, and Chu et al. (1981) Gene 13:197. Such techniques can be used to introduce one or more exogenous DNA moieties into suitable host cells. The term refers to both stable and transient uptake of the genetic material, and includes uptake of peptide- or antibody-linked nucleic acids.

A “vector” is capable of transferring nucleic acid sequences to target cells (e.g., viral vectors, non-viral vectors, particulate carriers, and liposomes). Typically, “vector construct,” “expression vector,” and “gene transfer vector,” mean any nucleic acid construct capable of directing the expression of a nucleic acid of interest and which can transfer nucleic acid sequences to target cells. Thus, the term includes cloning and expression vehicles, as well as viral vectors.

An “immunological response” to an antigen or composition is the development in a subject of a humoral and/or a cellular immune response to an antigen present in the composition of interest. For purposes of the present invention, a “humoral immune response” refers to an immune response mediated by antibody molecules, while a “cellular immune response” is one mediated by T-lymphocytes and/or other white blood cells. One important aspect of cellular immunity involves an antigen-specific response by cytolytic T-cells (“CTL”s). CTLs have specificity for peptide antigens that are presented in association with proteins encoded by the major histocompatibility complex (MHC) and expressed on the surfaces of cells. CTLs help induce and promote the destruction of intracellular microbes, or the lysis of cells infected with such microbes. Another aspect of cellular immunity involves an antigen-specific response by helper T-cells. Helper T-cells act to help stimulate the function, and focus the activity of nonspecific effector cells against cells displaying peptide antigens in association with MHC molecules on their surface. A “cellular immune response” also refers to the production of cytokines, chemokines and other such molecules produced by activated T-cells and/or other white blood cells, including those derived from CD4+ and CD8+ T-cells.

A composition or vaccine that elicits a cellular immune response may serve to sensitize a vertebrate subject by the presentation of antigen in association with MHC molecules at the cell surface. The cell-mediated immune response is directed at, or near, cells presenting antigen at their surface. In addition, antigen-specific T-lymphocytes can be generated to allow for the future protection of an immunized host.

The ability of a particular antigen to stimulate a cell-mediated immunological response may be determined by a number of assays, such as by lymphoproliferation (lymphocyte activation) assays, CTL cytotoxic cell assays, or by assaying for T-lymphocytes specific for the antigen in a sensitized subject. Such assays are well known in the art. See, e.g., Erickson et al., J. Immunol. (1993) 151:4189-4199; Doe et al., Eur. J. Immunol. (1994) 24:2369-2376. Recent methods of measuring cell-mediated immune response include measurement of intracellular cytokines or cytokine secretion by T-cell populations, or by measurement of epitope specific T-cells (e.g., by the tetramer technique) (reviewed by McMichael, A. J., and O'Callaghan, C. A., J. Exp. Med. (1998) 187:1367-1371; Mcheyzer-Williams, M. G., et al, Immunol. Rev. (1996) 150:5-21; Lalvani, A., et al, J. Exp. Med. (1997) 186:859-865).

Thus, an immunological response as used herein may be one that stimulates the production of antibodies (e.g., neutralizing antibodies that block pathogens such as viruses entering cells and replicating by binding to toxins and pathogens, typically protecting cells from infection and destruction). The antigen of interest may also elicit production of CTLs. Hence, an immunological response may include one or more of the following effects: the production of antibodies by B-cells; and/or the activation of suppressor T-cells and/or memory/effector T-cells directed specifically to an antigen or antigens present in the composition or vaccine of interest. These responses may serve to neutralize infectivity, and/or mediate antibody-complement, or antibody dependent cell cytotoxicity (ADCC) to provide protection to an immunized host. Such responses can be determined using standard immunoassays and neutralization assays, well known in the art. (See, e.g., Montefiori et al. J. Clin Microbiol. (1988) 26:231-235; Dreyer et al., AIDS Res Hum Retroviruses (1999) 15:1563-1571). The innate immune system of mammals also recognizes and responds to molecular features of pathogenic organisms via activation of Toll-like receptors and similar receptor molecules on immune cells. Upon activation of the innate immune system, various non-adaptive immune response cells. are activated to, e.g., produce various cytokines, lymphokines and chemokines. Cells activated by an innate immune response include immature and mature Dendritic cells of the monocyte and plamsacytoid lineage (MDC, PDC), as well as gamma, delta, alpha and beta T cells and B cells and the like. Thus, the present invention also contemplates an immune response wherein the immune response involves both an innate and adaptive response.

An “immunogenic composition” is a composition that comprises an antigenic molecule where administration of the composition to a subject results in the development in the subject of an immunological response as defined above.

An “antigen” refers to a molecule, such as a protein, polypeptide, or fragment thereof, or an attenuated virus, containing one or more epitopes (either linear, conformational or both) that will stimulate a host's immune-system to make an immunological response, as defined above. The term is used interchangeably with the term “immunogen.” The term “antigen” denotes both subunit antigens, (i.e., antigens which are separate and discrete from a whole organism with which the antigen is associated in nature), as well as, killed, attenuated or inactivated bacteria, viruses, fungi, parasites or other microbes. Similarly, an oligonucleotide or polynucleotide which expresses an antigen or antigenic determinant in vivo, such as in gene therapy and nucleic acid immunization applications, is also included in the definition of antigen herein.

By “vertebrate subject” is meant any member of the subphylum chordata, including, without limitation, humans and other primates, including non-human primates such as chimpanzees and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs; birds, including domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like. The term does not denote a particular age. Thus, both adult and newborn individuals are intended to be covered.

By “therapeutically effective amount” in the context of the immunogenic compositions is meant an amount of an immunogen, e.g., a recombinant, chimeric influenza virus, which will induce an immunological response, either for antibody production or for treatment or prevention of influenza virus infection. Such a response will generally result in the development in the subject of an antibody-mediated and/or a secretory or cellular immune response to the composition. Usually, such a response includes but is not limited to one or more of the following effects; the production of antibodies from any of the immunological classes, such as immunoglobulins A, D, E, G or M; the proliferation of B and T lymphocytes; the provision of activation, growth and differentiation signals to immunological cells; expansion of helper T cell, suppressor T cell, and/or cytotoxic T cell and/or γδT cell populations.

“Parenteral administration” refers to introduction into the body outside the digestive tract, such as by subcutaneous, intramuscular, intradermal or intravenous administration. This is to be contrasted with delivery to a mucosal surface, such as oral, nasal, vaginal or rectal. “Mucosal administration” refers to introduction into the body via any mucosal surface, such as intragastrically, pulmonarily, transdermally, intestinally, ocularly, intranasally, orally, vaginally, rectally, intratracheally or the like.

As used herein, “treatment” refers to any of (i) the prevention of infection or reinfection, as in a traditional vaccine, (ii) the reduction or elimination of symptoms, and (iii) the substantial or complete elimination of influenza virus from an infected individual. Treatment may be effected prophylactically (prior to infection) or therapeutically (following infection).

2. MODES OF CARRYING OUT THE INVENTION

Before describing the present invention in detail, it is to be understood that this invention is not limited to particular formulations or process parameters as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments of the invention only, and is not intended to be limiting.

Although a number of methods and materials similar or equivalent to those described herein can be used in the practice of the present invention, the preferred materials and methods are described herein.

The present invention provides recombinant, chimeric swine influenza viruses which are attenuated with an impaired ability to replicate in vivo, methods for producing such attenuated swine influenza viruses, and the use of such viruses in vaccine and pharmaceutical formulations. Such viruses are capable of generating an immune response and creating immunity but either do not cause illness or cause fewer and/or less severe symptoms, i.e., the viruses have decreased virulence. Therefore, they are ideal candidates for live virus vaccines. Moreover, because HAs from different subtypes of SIVs are present, compositions including the chimeric influenza viruses can be used to provide broad coverage against a number of influenza strains.

In particular, the invention pertains to recombinant, chimera influenza viruses that include HA segments from more than one influenza subtype and include a deletion of all or part of the NA segment, immunogenic compositions comprising the viruses, as well as methods of stimulating an immune response against influenza virus, and methods of interfering with influenza virus replication.

In order to further an understanding of the invention, a more detailed discussion is provided below regarding the production of recombinant, chimeric influenza viruses and methods of using the same in compositions in the treatment and/or prevention of influenza virus infection.

A. Recombinant, Chimeric Influenza Viruses

Wild-type swine influenza viruses typically include an 8 segmented genome with the segments designated as follows:

SEGMENT GENE PRODUCT NAME 1 PB2 (Polymerase (basic) protein 2) 2 PB1 (Polymerase (basic) protein 1) 3 PA (Polymerase (acidic) protein) 4 HA (Hemagglutinin) 5 NP (Nucleoprotein) 6 NA (Neuraminidase) 7 M1 (Matrix protein 1); M2 (Matrix protein 2) 8 NS1 (Non-structural protein 1); NEP, also termed NS2 (Non-structural protein 2)

The recombinant, chimeric influenza viruses described herein include two or more HA segments (segment 4) from two or more subtypes of influenza viruses. The recombinant influenza viruses can include HAs from any subtype of influenza virus and preferably from influenza A virus, selected from H1-H15 and N1-N9, such as but not limited to H1N2, H1N1, H3N2, H3N1, H9N2 and H5N1 or any combination of H's and N's. Particularly preferred are HA segments from viruses that infect pigs. The polynucleotide and amino acid sequences of representative swine HA genes can be found in public sequence databases such as GenBank. For example, HA genes from H1N1 and include, but are not limited to, GenBank Accession Nos. AY619961.1 (see FIGS. 5A and 5B); GQ457549.1; GQ457548.1; GQ457547.1; CY091769.1; CY091745.1; CY091737.1; CY091729.1; GU721143.3; JF820285.1; JF820277.1; JF707784.1; CY087136.1; CY087104.1; CY087096.1; CY087080.1; CY087072.1; CY087064.1; CY087056.1; CY087048.1; CY086863.1; CY086839.1; CY086353.1; CY086006.1; CY085990.1; CY085982.1; CY085974.1; CY085966.1; CY085958.1; CY085950.1; CY085942.1; CY085934.1; CY085926.1; CY085918.1; CY085910.1; CY085902.1; CY085894.1; CY085886.1; CY085878.1; CY085870.1; CY085854.1; CY085846.1; CY085838.1; CY085830.1; CY085822.1; CY085814.1; CY085806.1; CY085798.1; CY085790.1; CY085782.1; CY085774.1; CY085766.1; CY085758.1; CY085742.1; CY085726.1; CY085718.1; CY085710.1; CY085702.1; CY085694.1; CY085686.1; CY085670.1; JF833344.1; JF833343.1; JF833341.1; JF833339.1; JF833338.1; JF833337.1; JF833335.1; JF916682.1; JF812292.1; JF812291.1; JF812290.1; JF812287.1; JF812284.1; JF812281.1; JF812280.1; JF812279.1; JF812278.1; JF812273.1; JF812272.1; JF812271.1; AF091317.1; AF091315.1; AF091314.1.

HA genes from H3N2 and include, but are not limited to, GenBank Accession Nos. AF153233.1 (see FIGS. 13A and 13B); AY377927.2; CY092324.1; JN105973.1; HQ315643.1; FJ519977.1; FJ519976.1; FJ519975.1; FJ519974.1; FJ519973.1; FJ519972.1; FJ519971.1; GU937743.1; JF833345.1; JF833340.1; JF833336.1; JF833334.1; JF812293.1; JF812289.1; JF812277.1; JF812276.1; JF812275.1; JF812274.1; CY045575.1; CY045567.1; CY045559.1; CY045551.1; HQ825243.1; HQ825235.1; HQ825229.1; HQ825226.1; HQ825223.1; HQ825218.1; HQ825210.1; HQ825210.1; HQ825198.1; HQ825190.1; HQ825182.1; HQ825174.1; HQ825166.1; JF312065.1; JF312064.1; CY086920.1; JF312073.1; JF312072.1; JF312071.1; JF316643.1; JF263536.1; JF263535.1; HQ734204.1; HQ734201.1; HQ734198.1; HQ734195.1; HQ734192.1; HQ734189.1; HQ734186.1; CY077942.1; CY077934.1.

Any of the above HAs or other readily available HA sequences can be used with the subject invention.

Additionally, the recombinant, chimeric influenza viruses typically include a mutation in the NA genomic segment (segment 6) coding for neuraminidase such that replication of the virus is impaired. Mutations can include deletions, inversions, insertions or substitutions that impair replication of the virus. In certain embodiments, the virus variant comprises a deletion of all or part of the NA segment such that virus propagation is hindered. Because NA is essential for virus propagation, the function of NA can be provided in culture by growing the virus in the presence of sialidase. Preferably, NA packaging sequences at the 3′ and optionally the 5′ untranslated regions (UTRs) flanking the NA sequence and extending into the coding sequence are retained in the recombinant viruses.

In particular, specific cis-acting packaging signals exist in 3′ and 5′ (UTRs) that extend into the coding regions of most if not all segments, including the NA segment, which is responsible for viral release from infected cells by removing sialic acids from cellular glycoconjugates and viral glycoproteins. Each viral RNA consists predominantly of coding sequences (in antisense orientation), flanked at both ends by UTRs that range from 19 to 58 bases long. Within these UTRs, the distal 12 and 13 noncoding bases that form the extreme 3′ and 5′ termini, respectively, of every segment are highly conserved among viral strains and among the eight segments themselves. These distal conserved sequences are partially complementary to each other and can anneal to form a bulged duplex structure that is essential for transcription and replication of the segment. The UTRs harbor cis-acting signals that contribute to RNA packaging, since the attachment of authentic UTRs onto a heterologous RNA can enable it to be packaged into, and transduced by, influenza virus particles. Optimal packaging of at least some segments, such as NA, HA and NS requires not only both UTRs but also short portions of the coding region.

Deletion analysis of reporter constructs indicates that the minimal sequences needed for efficient packaging extend beyond each UTR to include 9 to 80 bases of adjacent coding sequence at either end of the segment (Fujii et al., J. Virol. (2005) 79:3766-3774; Fujii et al., Proc. Natl. Acad. Sci. USA (2003) 100:2002-2007; Watanabe et al., J. Virol. (2003) 77:10575-10583). Sequences at the 3′ end of the coding region appear to exert a greater quantitative effect than those at the 5′ end. These regions are therefore useful for packaging and maintaining wild-type NA RNA as well as mutant NA RNAs, e.g., RNAs with internal deletions and/or insertions. Accordingly, the recombinant, chimeric viruses of the invention will include at least packaging signals from the 3′ UTR and a portion of the 3′ NA coding region, and preferably will include packaging signals from both the 3′ and 5′ UTRs and 3′ and 5′ portions of the NA sequence.

Methods for locating packaging signals are known. In particular, Gog et al., Nucl. Acids Res. (2007) 35:1897-1907 found highly statistically significant clusters of codons with lower than expected synonymous variation within the influenza virus genome, located at the terminal regions of segments, where the presence of specific packaging signals are known. Synonymous mutational analysis of these regions confirmed the ability of their method to identify functionally significant cis-acting elements (i.e., packaging signals) in the virus genome at the single nucleotide level. Using these methods, then, packaging signals for the NA segment of various virus strains and subtypes can be readily identified. Determination of packaging efficiency of recombinant viral RNA segments can be carried out using techniques known in the art. See, e.g., Dos et al., Virology (2005) 341:34-46.

Generally, NA packaging sequences for use in the present invention will include at least 19 nucleotides from the 3′ UTR adjacent to the NA coding sequence, preferably 19-30 nucleotides, such as 19, 20 . . . 25 . . . 30 . . . 35 nucleotides and at least 28 nucleotides from the 5′ UTR adjacent to the NA coding sequence, preferably 28-50 nucleotides, such as 28, 29, 30 . . . 35 . . . 40 . . . 45 . . . 50 nucleotides. The NA packaging sequences also will include about 145 to 250, preferably 150-200 nucleotides from at least the 3′ end of the coding sequence and optionally from each end of the coding region for the NA segment. Thus, for example, influenza virus packaging sequences can comprise sequences corresponding to the 3′ end of NA viral RNA including sequences corresponding to the N-terminus of the NA coding region, e.g., at least 150 nucleotides of the 3′ end of a type A NA viral RNA such as 15O, 151, 152, 153, 154, 155 . . . 160 . . . 165 . . . 170 . . . 175 . . . 180 . . . 185 . . . 190, and so on, and, optionally, packaging sequences corresponding to the 5′ end of NA viral RNA including sequences corresponding to the C-terminus of the NA coding region, e.g., 150, 151, 152, 153, 154, 155 . . . 160 . . . 165 . . . 170 . . . 175 . . . 180 . . . 185 . . . 190, and so on.

In one particular embodiment, a construct can be provided that includes an HA segment from one porcine influenza subtype and NA packaging sequences from another porcine influenza subtype located 3′ and optionally 5′ to the HA segment. As described in the examples, a construct was prepared that included an H3N2 HA flanked by H1N1 packaging sequences. This particular construct comprises an H3N2 HA sequence, flanked by 19 nucleotides from the 3′ UTR adjacent to an H1N1 NA sequence and 183 nucleotides from the 3′ NA coding region and 28 nucleotides from the 5′ UTR adjacent to the NA sequence and 157 nucleotides from the 5′ NA coding region. However, the remainder of the NA coding region is absent. Typically, the packaging sequences used are homologous to the backbone virus. Thus, if an H1N1 subtype is used as the backbone (i.e., all H1N1 segments are present in the recombinant virus except for all or a portion of the NA segment), NA packaging sequences from H1N1 will be retained and the remainder of the H1N1 NA sequence conveniently replaced with an HA sequence from a different subtype, such as an H3N2 HA sequence.

If desired, rather than a deletion, the NA coding region can be mutated such that virus propagation is hindered. The NA region can be mutagenized in vitro by the replacement of the appropriate nucleotides to result in the desired amino acid changes. Such a change can include as little as one nucleotide, effecting a change in a single amino acid, or can encompass several nucleotide changes. Mutants can be produced by standard methods of site-directed mutagenesis. The mutations can be effected using a mismatched primer which hybridizes to the parent nucleotide sequence (generally cDNA corresponding to the RNA sequence), at a temperature below the melting temperature of the mismatched duplex. The primer can be made specific by keeping primer length and base composition within relatively narrow limits and by keeping the mutant base centrally located. See, e.g., Innis et al, (1990) PCR Applications: Protocols for Functional Genomics; Zoller and Smith, Methods Enzymol. (1983) 100:468; Wu (Ed.), Meth. In Enzymol. Vol. 217, San Diego: Academic Press (1993); Kunkel (1985) Proc. Natl. Acad. Sci. USA, 82:488-492; all of which are incorporated herein by reference.

The NA mutation (e.g., deletion of all or a portion of the NA coding sequence except the sequences harboring packaging signals) is preferably one that hinders virus propagation. The replication efficiency of an attenuated influenza virus may be determined, for example, by measuring plaque size in Madin-Darby canine kidney (MDCK) cells, by measuring virus titers over multiple growth cycles, or by isolating virus from infected lung tissue and measuring titers. For example, an attenuated swine influenza virus of the invention permits the attenuated virus, at a multiplicity of infection (MOI) of between 0.0005 and 0.001, 0.001 and 0.01, 0.01 and 0.1, or 0.1 and 1, or a MOI of 0.0005, 0.0007, 0.001, 0.005, 0.01, 0.05, 0.1, 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 5.5, or 6.0, to grow to titers between approximately 1 to approximately 100 fold, approximately 2 to approximately 90 fold, approximately 5 to approximately 80 fold, approximately 20 to approximately 80 fold, or approximately 40 to approximately 80 fold, approximately 1 to approximately 10 fold, approximately 1 to approximately 5 fold, approximately 1 to approximately 4 fold, approximately 1 to approximately 3 fold, approximately 1 to approximately 2 fold, approximately 3 to approximately 15 fold, or approximately 1, 2, 3, 4, 5, 6, 7, 8, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100 fold lower than wild-type swine influenza virus in cell culture, e.g. MDCK cells; cells of a human (e.g., PerC6, a producer cell line derived from human embryonic retinoblasts transformed with the E1 region of Adenovirus 5); mouse; chicken (e.g., chicken embryo fibroblasts); rat, birds; or pig (e.g., PK(D1) cells, PK(15) cells, PK13 cells, NSK cells, LLC-PK1 cells, LLC-PK1A cells, ESK-4 cells, ST cells, PT-K75 cells, PK-2a/CL 13 or SJPL cells). Replication efficiency can be determined by a hemagglutination assay of BALF from pigs or supernatants of pig cells approximately 2 to 10 days, 3 to 7 days, 3 to 5 days, or 2, 3, 5, 6, 7, 8, 9, 10 days post-infection or when the viruses are plagued on MDCK cells. In one embodiment, the growth of an attenuated swine influenza virus of the invention is compared to a particular standard or reference, e.g., wild-type swine influenza virus A/Swine/Texas/4199-2/98. Another measure of attenuation is to grow the virus in the absence of sialidase and measure titers as compared to a reference wild-type strain as above.

In addition to the HA sequences, and the packaging sequences described above, the recombinant, chimeric influenza virus will also include the remaining viral segments, segments 1-3, 5, 7 and 8, that is, segments encoding PB2 (segment 1), PB1 (segment 2), PA (segment 3), NP (segment 5), M1 and M2 (segment 7), NS1 and NEP (segment 8). Nucleic acid and polypeptide sequences for these segments, as well as segments 4 (encoding HA) and 6 (encoding NA) from a number of influenza virus isolates are known. Representative influenza sequences are presented in FIGS. 5-13 herein. Additional representative sequences for the 8 influenza segments from influenza isolates found in various species are listed in the National Center for Biotechnology Information (NCBI) database and the Influenza Research Database found at fludb.org. See also Ferguson et al. (2003) Nature 422: 428-433; Lin et al. (2000) Proc. Natl. Acad. Sci. U.S.A. 97, 9654-9658; Nguyen et al. (2005) J. Virol. 79:4201-4212; Ha et al. (2002) EMBO J. 21:865-875; and Chan et al. (2004) J. Microbiol. Immunol. Infect. 37:135-144; for sequence comparisons and a discussion of genetic diversity and phylogenetic analysis of influenza virus.

Any of these sequences, as well as variants thereof can be used to produce the recombinant, chimeric influenza viruses. Thus, the invention includes variants of the above sequences displaying at least about 80-100% sequence identity thereto, including any percent identity within these ranges, such as 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100% sequence identity thereto.

The above-described segments can be derived from any of various swine influenza viruses, including, without limitation, A/swine/Saskatchewan/18789/02, A/Swine/Colorado/1/77, A/Swine/Colorado/23619/99, A/Swine/Cote d'Armor/3633/84, A/Swine/Cote d'Armor/3633/84, A/Swine/England/195852/92, A/Swine/Finistere/2899/82, A/Swine/Hong Kong/10/98, A/Swine/Hong Kong/9/98, A/Swine/Hong Kong/81/78, A/Swine/Illinois/100084/01, A/Swine/Illinois/100085A/01, A/Swine/Illinois/21587/99, A/Swine/Indiana/1726/88, A/Swine/Indiana/9K035/99, A/Swine/Indiana/P 12439/00, A/Swine/Iowa/30, A/Swine/Iowa/15/30, A/Swine/Iowa/533/99, A/Swine/Iowa/569/99, A/Swine/Iowa/3421/90, A/Swine/Iowa/8548-1/98, A/Swine/Iowa/930/01, A/Swine/Iowa/17672/88, A/Swine/Italy/1513-1/98, A/Swine/Italy/1523/98, A/Swine/Korea/CY02/02, A/Swine/Minnesota/55551/00, A/Swine/Minnesota/593/99, A/Swine/Minnesota/9088-2/98, A/Swine/Nebraska/1/92, A/Swine/Nebraska/209/98, A/Swine/Netherlands/12/85, A/Swine/North Carolina/16497/99, A/Swine/North Carolina/35922/98, A/Swine/North Carolina/93523/01, A/Swine/North Carolina/98225/01, A/Swine/Oedenrode/7C/96, A/Swine/Ohio/891/01, A/Swine/Oklahoma/18717/99, A/Swine/Oklahoma/18089/99, A/Swine/Ontario/01911-1/99, A/Swine/Ontario/01911-2/99, A/Swine/Ontario/41848/97, A/Swine/Ontario/97, A/Swine/Quebec/192/81, A/Swine/Quebec/192/91, A/Swine/Quebec/5393/91, A/Swine/Taiwan/7310/70, A/Swine/Tennessee/24/77, A/Swine/Texas/4199-2/98, A/Swine/Wisconsin/125/97, A/Swine/Wisconsin/136/97, A/Swine/Wisconsin/163/97, A/Swine/Wisconsin/164/97, A/Swine/Wisconsin/166/97, A/Swine/Wisconsin/168/97, A/Swine/Wisconsin/235/97, A/Swine/Wisconsin/238/97, A/Swine/Wisconsin/457/98, A/Swine/Wisconsin/458/98, A/Swine/Wisconsin/464/98 and A/Swine/Wisconsin/14094/99.

In one particular embodiment, an H3N2 HA is used in place of all or part of an H1N1 NA sequence in an H1N1 backbone. Thus, the resulting recombinant virus includes two, HAs and the remainder of the viral segments. See, FIG. 1.

Each of the above described segments can be isolated from viral RNA using known methods. For example, nucleic acids can be obtained by screening cDNA and/or genomic libraries from cells infected with virus, or by deriving the gene from a vector known to include the same. For example, polynucleotides of interest can be isolated from a genomic library derived from viral RNA from an infected subject. Alternatively, influenza virus can be isolated from infected mammals or from biological samples (e.g., nasal, nasopharyngeal, throat, or conjunctival secretions, blood, or anal swabs) collected from infected subjects. Once obtained, the virus can be propagated using known techniques, such as described in Mochalova et al., Virology (2003) 313:473-480; Lin et al., Virology (1997) 233:402-410; Hardy et al., Virology (1995) 211:302-306; Hinshaw et al., J. Gen. Virol. (1978) 41:115-127. Nucleic acid can also be obtained directly from the influenza virus in question.

Thus, particular nucleotide sequences can be obtained from vectors harboring the desired sequences or synthesized completely or in part using various oligonucleotide synthesis techniques known in the art, such as site-directed mutagenesis and polymerase chain reaction (PCR) techniques where appropriate. See, e.g., Sambrook, supra. One method of obtaining nucleotide sequences encoding the desired sequences is by annealing complementary sets of overlapping synthetic oligonucleotides produced in a conventional, automated polynucleotide synthesizer, followed by ligation with an appropriate DNA ligase and amplification of the ligated nucleotide sequence via PCR. See, e.g., Jayaraman et al. (1991) Proc. Natl. Acad. Sci. USA 88:4084-4088. Additionally, oligonucleotide directed synthesis (Jones et al. (1986) Nature 54:75-82), oligonucleotide directed mutagenesis of pre-existing nucleotide regions (Riechmann et al. (1988) Nature 332:323-327 and Verhoeyen et al. (1988) Science 239:1534-1536), and enzymatic filling-in of gapped oligonucleotides using T4 DNA polymerase (Queen et al. (1989) Proc. Natl. Acad. Sci. USA 86:10029-10033) can be used to produce modified molecules.

An amplification method such as PCR can be used to amplify polynucleotides including the various segments. In one embodiment, these segments are reverse-transcribed into cDNA and amplified using RT-PCR. See, e.g., Hoffmann et al., Proc. Natl. Acad. Sci. USA (2000) 97:6108-6113. The cDNA from each segment is cloned to provide separate plasmids for use in preparing the recombinant, chimeric influenza virus. In some embodiments, cloning vector pHW2000 (Hoffmann et al., Proc. Natl. Acad. Sci. USA (2000) 97:6108-6113) can be used. However, the segments can be cloned into any suitable vector. Numerous cloning vectors are known to those of skill in the art, and the selection of an appropriate cloning vector is a matter of choice. Examples of recombinant DNA vectors for cloning and host cells which they can transform include the bacteriophage λ (E. coli), pBR322 (E. coli), pACYC177 (E. coli), pKT230 (gram-negative bacteria), pGV1106 (gram-negative bacteria), pLAFR1 (gram-negative bacteria), pME290 (non-E. coli gram-negative bacteria), pHV14 (E. coli and Bacillus subtilis), pBD9 (Bacillus), pIJ61 (Streptomyces), pUC6 (Streptomyces), YIp5 (Saccharomyces), YCp19 (Saccharomyces) and bovine papilloma virus (mammalian cells). See, generally, DNA Cloning: Vols. I & II, supra; Sambrook et al., supra; B. Perbal, supra.

The recombinant, chimeric, attenuated influenza virus comprising more than one HA sequence from more than one influenza subtype can then be produced by any method well know in the art. Preferably reverse genetics is used to produce the recombinant viruses. Reverse genetics uses RNA polymerase complexes isolated from influenza virus-infected cells to transcribe artificial influenza virus genome segments containing the mutation(s). The synthesized RNA segment(s) are incorporated into virus particles using a helper virus, and viruses containing the desired changes are then selected. Reverse genetics methods for influenza viruses are described, for example, in Enami et al., Proc. Natl. Acad. Sci. (1990) 87:3802 3805; Enami and Palese, J. Virol. (1991) 65:2711-13; Luytjes, Cell (1989) 59:1107-13; Fodor et al., J. Virol. (1999) 73:9679-9682; Gao et al., J. Virol. (2008) 82:6419-6426; Quinlivan et al., J. Virol. (2005) 79:8431-8439; and U.S. Pat. Nos. 5,578,473, 6,974,686 and 7,037,707, all of which are incorporated herein by reference in their entireties.

Recently developed reverse-genetics systems, based entirely on cDNA, have allowed the manipulation of the influenza viral genome. See, e.g, Palese et., Proc. Natl. Acad. Sci. USA (1996) 93:11354; Neumann and Kawaoka, Adv. Virus Res. (1999) 53:265; Neumann et al., Proc. Natl. Acad. Sci. USA (1999) 96:9345; Fodor et al., J. Virol. (1999) 73:9679, incorporated by reference in their entireties. In one technique, modified viral RNA transcripts are transcribed in vitro from cDNA constructs in the presence of purified NP, PB1, PB2, and PA proteins. The resulting synthetic RNP is then transfected into cells previously infected with an influenza helper virus. This helper virus usually has a conditional growth defect, such as host range restriction or temperature sensitivity, which allows the subsequent selection of transfectant viruses. For example, host-range helper viruses have been successfully used to rescue synthetic NA and PB2 genes. See Enami, supra, and Subbarao, J Virol (1993) 67:7223-28.

In preferred embodiments, an eight plasmid system is used to generate attenuated influenza viruses. See, e.g., Hoffmann et al., Vaccine (2002) 20:3165-3170; Hoffmann et al., Proc. Natl. Acad. Sci. USA (2000) 97:6108-6113; and U.S. Patent Publication No. 20040029251, incorporated herein by reference in their entireties. In this embodiment, the plasmids harboring the eight segments of the desired influenza virus, such as the two HA segments, as well as segments encoding polymerase acidic protein (PA), polymerase basic proteins 1 and 2 (PB1 and PB2), the matrix (M) segment encoding matrix proteins 1 and 2 (M1 and M2), the nucleoprotein (NP), and the nonstructural (NS) segment encoding nonstructural proteins 1 and 2 (NS1 and NEP), are cotransfected into an appropriate cell resulting in the recombinant, chimeric virus described herein. See also, U.S. Pat. No. 6,951,754 that describes eight plasmid dual promoter reverse genetic systems for the production of attenuated influenza viruses using a pol I-pol II system, incorporated herein by reference in its entirety.

Production of live attenuated virus vaccine formulations is accomplished using conventional methods involving propagation of the recombinant, chimeric virus in any substrate that allows the virus to grow to titers sufficient for further use. Typically, the viruses are propagated in cells, embryonated eggs, and/or animals followed by purification. Generally, influenza viruses are grown in embryonated chicken eggs or mammalian cells, such as Madin-Darby canine kidney (MDCK) cells, Madin Darby bovine kidney (MDBK) cells, pig cells, or African green monkey kidney (Vero) cells, using known techniques. See, e.g., Mochalova et al., Virology (2003) 313:473-480; Lin et al., Virology (1997) 233:402-410; Hardy et al., Virology (1995) 211:302-306; Hinshaw et al., J. Gen. Virol. (1978) 41:115-127. Representative pig cells include porcine kidney cell lines, porcine testis cell lines and porcine lung cell lines, such as but not limited to, PK(D1) cells, PK(15) cells, PK13 cells, SJPL cells, NSK cells, LLC-PK1 cells, LLC-PK1A cells, ESK-4 cells, ST cells, PT-K75 cells, and PK-2a/CL 13 cells.

In another embodiment, the recombinant, chimeric swine influenza viruses are propagated in chicken cells, e.g., chicken embryo fibroblasts derived from, e.g., 6-14 day-old embryonated eggs. In other embodiments, young or immature embryonated eggs can be used to propagate the viruses of the invention. Immature embryonated eggs encompass eggs which are less than ten-day-old eggs. Immature embryonated eggs may also be eggs which artificially mimic immature eggs as a result of alterations to the growth conditions, e.g., changes in incubation temperatures; treating with drugs; or any other alteration which results in an egg with a retarded development. The swine influenza viruses can be propagated in different locations of the embryonated egg, e.g., the allantoic cavity.

In a specific embodiment, the attenuated swine influenza viruses of the present invention are propagated in any substrate that allows the virus to grow to titers comparable to those determined for wild type swine influenza virus strains. Preferably, the virions are cultured in the presence of sialidase since the NA segment in the recombinant, chimeric virus is deficient.

It is preferred that the virus is highly purified prior to vaccine formulation. Generally, the purification procedures will result in the extensive removal of cellular DNA, other cellular components, and adventitious agents. Procedures that extensively degrade or denature DNA can also be used. See, e.g., Mizrahi, ed., Viral Vaccines, Wiley-Liss, New York (1990). Methods of purification are known in the art and may include one or more of, for instance, gradient centrifugation, ultracentrifugation, zonal ultracentrifugation, continuous-flow ultracentrifugation and chromatography, such as ion exchange chromatography, size exclusion chromatography, and liquid affinity chromatography, polyethylene glycol or ammonium sulfate precipitation.

B. Anti-Viral Compositions

The recombinant, chimeric influenza viruses, as well as recombinant, chimeric influenza viruses that have been subsequently inactivated, can be formulated into compositions for delivery to subjects for either inhibiting infection, or for enhancing an immune response to influenza virus. Thus, either a live recombinant swine influenza virus vaccine or immunogenic formulation or an inactivated recombinant swine influenza virus vaccine or immunogenic formulation can be formulated. A live vaccine or immunogenic formulation may be preferred because multiplication in the host leads to a prolonged stimulus of similar kind and magnitude to that occurring in natural infections, and therefore, confers substantial, long-lasting immunity. Production of such live recombinant swine influenza virus vaccine formulations and immunogenic formulations may be accomplished using conventional methods involving propagation as described above. When formulated as a live virus vaccine, a range of about 102 to 108 can be used, preferably from about 103 to 107, more preferably 104 pfu to about 5×106, and most preferably from 104 to 107 pfu per dose should be used.

Inactivated vaccine formulations may be prepared using conventional techniques to “kill” the attenuated viruses. Inactivated vaccines are “dead” in the sense that their infectivity has been destroyed. Ideally, the infectivity of the virus is destroyed without affecting its immunogenicity. In order to prepare inactivated vaccines, the attenuated virus is grown and purified as described above. The purified virus is then inactivated using one of several methods known in the art. Such methods include both chemical or physical means. Chemical means for inactivating an influenza virus include treatment of the virus with an effective amount of one or more of the following agents: detergents, formaldehyde, formalin, β-propiolactone, or UV light. Other methods of viral inactivation are known in the art, such as for example binary ethylamine, acetyl ethyleneimine, or gamma irradiation. See, e.g., U.S. Pat. Nos. 6,635,246; 5,891,705; 5,106,619; and 4,693,981, incorporated herein by reference in their entireties.

Compositions of the invention may comprise or be coadministered with a non-influenza antigen or combination of antigens, such as with a combination influenza vaccine. Methods of preparing such formulations are described in, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 18 Edition, 1990. The compositions of the present invention can be prepared for mucosal delivery, parenteral delivery, e.g., as injectables, either as liquid solutions or suspensions. Solid forms suitable for solution in or suspension in liquid vehicles prior to injection may also be prepared. The preparation may also be emulsified or the active ingredient encapsulated in liposome vehicles. The active immunogenic ingredient is generally mixed with a compatible pharmaceutical vehicle, such as, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof. In addition, if desired, the vehicle may contain minor amounts of auxiliary substances such as wetting or emulsifying agents and pH buffering agents.

If used to modulate an immune response, additional adjuvants which enhance the effectiveness of the composition may also be added to the formulation. Adjuvants may include for example, muramyl dipeptides, pyridine, aluminum hydroxide, dimethyldioctadecyl ammonium bromide (DDA), oils, oil-in-water emulsions, saponins, cytokines, and other substances known in the art.

Carriers may also be used in order to increase the immunogenicity of the vaccine. Suitable carriers include large, slowly metabolized macromolecules such as proteins, including serum albumins, keyhole limpet hemocyanin, immunoglobulin molecules, thyroglobulin, ovalbumin, and other proteins well known to those skilled in the art; polysaccharides, such as sepharose, agarose, cellulose, cellulose beads and the like; polymeric amino acids such as polyglutamic acid, polylysine, and the like; amino acid copolymers; and inactive virus particles.

Furthermore, influenza molecules may be formulated into compositions in either neutral or salt forms. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the active polypeptides) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed from free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.

Formulations will contain a “therapeutically effective amount” of the active ingredient, that is, an amount capable of achieving the desired response in a subject to which the composition is administered. In the treatment and prevention of influenza infection, for example, a “therapeutically effective amount” would preferably be an amount which prevents, reduces or ameliorates the symptoms of flu. The exact amount necessary will vary depending on the subject being treated; the age and general condition of the subject to be treated; the capacity of the subject's immune system to synthesize antibodies; the degree of protection desired; the severity of the condition being treated; the particular virus preparation selected and its mode of administration, among other factors. An appropriate effective amount can be readily determined by one of skill in the art. Thus, a “therapeutically effective amount” will fall in a relatively broad range that can be determined through routine trials. The recombinant, chimeric influenza virus will typically range from about 1% to about 95% (w/w) of the composition, or even higher or lower if appropriate.

Additional formulations which are suitable for other modes of administration include suppositories and, in some cases, aerosol, intranasal, oral formulations, and sustained release formulations. For suppositories, the vehicle composition will include traditional binders and carriers, such as, polyalkaline glycols, or triglycerides. Such suppositories may be formed from mixtures containing the active ingredient in the range of about 0.5% to about 10% (w/w), preferably about 1% to about 2%. Oral vehicles include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium, stearate, sodium saccharin cellulose, magnesium carbonate, and the like. These oral vaccine compositions may be taken in the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations, or powders, and contain from about 10% to about 95% of the active ingredient, preferably about 25% to about 70%.

Intranasal formulations will usually include vehicles that neither cause irritation to the nasal mucosa nor significantly disturb ciliary function. Diluents such as water, aqueous saline or other known substances can be employed with the subject invention. The nasal formulations may also contain preservatives such as, but not limited to, chlorobutanol and benzalkonium chloride. A surfactant may be present to enhance absorption of the subject proteins by the nasal mucosa.

Controlled or sustained release formulations are made by incorporating the protein into carriers or vehicles such as liposomes, nonresorbable impermeable polymers such as ethylenevinyl acetate copolymers and HYTREL copolymers, swellable polymers such as hydrogels, resorbable polymers such as collagen and certain polyacids or polyesters such as those used to make resorbable sutures, polyphosphazenes, alginate, microparticles, gelatin nanospheres, chitosan nanoparticles, and the like. The influenza virus can also be delivered using implanted mini-pumps, well known in the art.

C. Administration

Compositions of the invention will generally be administered directly to a patient. Direct delivery may be accomplished by parenteral injection (e.g. subcutaneously, intraperitoneally, intravenously, intramuscularly, or to the interstitial space of a tissue), or mucosally, such as by intratracheal, rectal, oral (e.g. tablet, spray), vaginal, topical, transdermal (See e.g. WO99/27961) or transcutaneous (See e.g. WO02/074244 and WO02/064162), intranasal (See e.g. WO03/028760), ocular, aural, pulmonary or other mucosal administration. Immunogenic compositions can also be administered topically by direct transfer to the surface of the skin. Topical administration can be accomplished without utilizing any devices, or by contacting naked skin with the immunogenic composition utilizing a bandage or a bandage-like device (see, e.g., U.S. Pat. No. 6,348,450).

Preferably the mode of administration is parenteral, mucosal or a combination of mucosal and parenteral immunizations. Even more preferably, the mode of administration is parenteral, mucosal or a combination of mucosal and parenteral immunizations in a total of 1-2 vaccinations 1-3 weeks apart. Preferably the route of administration includes but is not limited to oral delivery, intramuscular delivery and a combination of oral and intramuscular delivery.

In one embodiment, the method for treating an infection by an influenza virus, comprises mucosally administering to a subject in need thereof a first immunogenic composition comprising the influenza viruses of the invention followed by parenterally administering a therapeutically effective amount of a second immunogenic composition comprising the influenza viruses of the invention.

The immunogenic composition may be used to elicit systemic and/or mucosal immunity, preferably to elicit an enhanced systemic and/or mucosal immunity. Preferably the immune response is characterized by the induction of a serum IgG and/or an IgA immune response.

In any method involving coadministration, the various compositions can be delivered in any order. Thus, in embodiments including delivery of multiple different compositions or molecules, the influenza virus need not be delivered prior to other immunogenic substances. For example, the priming step may include delivery of one or more polypeptides and the boosting may comprise delivery of one or more attenutated influenza viruses. Multiple administrations of influenza virus can be followed by multiple administrations of other substances. Administrations can be performed in any order. Therefore, any combination of influenza virus and other immunogenic substances can be used to elicit an immune reaction.

D. Dosage Regime

Dosage treatment can be according to a single dose schedule or a multiple dose schedule. Multiple doses may be used in a primary immunization schedule and/or in a booster immunization schedule. In a multiple dose schedule, the various doses may be given by the same or different routes, e.g. a parenteral prime and mucosal boost, a mucosal prime and parenteral boost, etc.

Preferably the dosage regime enhances the avidity of the antibody response leading to antibodies with a neutralizing characteristic. An in vitro neutralization assay may be used to test for neutralizing antibodies (see for example Asanaka et al., J. of Virol. (2005) 102:10327; Wobus et al., PLOS Biology (2004) 2; e432; and Dubekti et al., J. Med. Virol. (2002) 66:400).

E. Tests to Determine the Efficacy of an Immune Response

One way of assessing efficacy of therapeutic treatment involves monitoring infection after administration of a composition of the invention. One way of assessing efficacy of prophylactic treatment involves monitoring immune responses against the antigens in the compositions of the invention after administration of the composition.

Another way of checking efficacy of therapeutic treatment involves monitoring infection after administration of the compositions of the invention. One way of checking efficacy of prophylactic treatment involves monitoring immune responses both systemically (such as monitoring the level of IgG1 and IgG2a production) and mucosally (such as monitoring the level of IgA production) against the antigens in the compositions of the invention after administration of the composition. Typically, serum specific antibody responses are determined post-immunization but pre-challenge whereas mucosal specific antibody body responses are determined post-immunization and post-challenge.

The immunogenic compositions of the present invention can be evaluated in in vitro and in vivo animal models prior to host administration. The efficacy of immunogenic compositions of the invention can be determined in vivo by challenging animal models of infection, e.g., guinea pigs or mice or pigs, with the immunogenic compositions. The immunogenic compositions may or may not be derived from the same strains as the challenge strains. Preferably the immunogenic compositions are derivable from the same strains as the challenge strains. Particularly useful mouse models include those in which intraperitoneal immunization is followed by either intraperitoneal challenge or intranasal challenge. In vivo efficacy mouse models include but are not limited to a murine infection model using swine strains and a murine disease model which is a murine model using a mouse-adapted strain, such as strains which are particularly virulent in mice.

The immune response may be one or both of a TH1 immune response and a TH2 response. The immune response may be an improved or an enhanced or an altered immune response. The immune response may be one or both of a systemic and a mucosal immune response. Preferably the immune response is an enhanced systemic and/or mucosal response.

An enhanced systemic and/or mucosal immunity is reflected in an enhanced TH1 and/or TH2 immune response. Preferably, the enhanced immune response includes an increase in the production of IgG1 and/or IgG2a and/or IgA. Preferably the mucosal immune response is a TH2 immune response. Preferably, the mucosal immune response includes an increase in the production of IgA.

Activated TH2 cells enhance antibody production and are therefore of value in responding to extracellular infections. Activated TH2 cells may secrete one or more of IL-4, IL-5, IL-6, and IL-10. A TH2 immune response may result in the production of IgG1, IgE, IgA and memory B cells for future protection.

A TH2 immune response may include one or more of an increase in one or more of the cytokines associated with a TH2 immune response (such as IL-4, IL-5, IL-6 and IL-10), or an increase in the production of IgG1, IgE, IgA and memory B cells. Preferably, the enhanced TH2 immune response will include an increase in IgG1 production.

A TH1 immune response may include one or more of an increase in CTLs, an increase in one or more of the cytokines associated with a TH1 immune response (such as IL-2, IFNγ, and TNFβ), an increase in activated macrophages, an increase in NK activity, or an increase in the production of IgG2a. Preferably, the enhanced TH1 immune response will include an increase in IgG2a production.

Immunogenic compositions of the invention, in particular, immunogenic composition comprising one or more antigens of the present invention may be used either alone or in combination with other antigens optionally with an immunoregulatory agent capable of eliciting a Th1 and/or Th2 response.

The immunogenic compositions of the invention will preferably elicit both a cell mediated immune response as well as a humoral immune response in order to effectively address an infection. This immune response will preferably induce long lasting (e.g., neutralizing) antibodies and a cell mediated immunity that can quickly respond upon exposure to one or more infectious antigens. By way of example, evidence of neutralizing antibodies in patient blood samples is considered as a surrogate parameter for protection since their formation is of decisive importance for virus elimination in TBE infections (see Kaiser and Holzmann, Infection 28; 78-84).

F. Kits

The invention also provides kits comprising one or more containers of compositions of the invention. Compositions can be in solid form, liquid form or can be lyophilized. Suitable containers for the compositions include, for example, bottles, vials, syringes, and test tubes. Containers can be formed from a variety of materials, including glass or plastic. A container may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).

The kit can further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution, or dextrose solution. It can also contain other materials useful to the end-user, including other pharmaceutically acceptable formulating solutions such as buffers, diluents, filters, needles, and syringes or other delivery device. The kit may further include a third component comprising an adjuvant.

For mucosal routes, the composition may be packaged for intranasal administration, such as by nasal spray, nasal drops, gel or powder. See, e.g., Almeida & Alpar, J. Drug Targeting (1996) 3:455-467; Agarwal & Mishra, Indian J. Exp. Biol. (1999) 37:6-16 or in inhalation devices well known in the art.

The kit can also comprise a package insert containing written instructions for methods of inducing immunity or for treating infections. The package insert can be an unapproved draft package insert or can be a package insert approved by the Food and Drug Administration (FDA) or other regulatory body.

The invention also provides a delivery device pre-filled with the immunogenic compositions of the invention.

3. EXPERIMENTAL

Below are examples of specific embodiments for carrying out the present invention. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.

Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for.

Example 1 Generation of a Recombinant Virus Including Both H1 and H3 HAs

A recombinant, chimeric influenza A virus possessing, an eight segmented genome, was produced as detailed below. The virus included seven segments of a swine H1N1 virus with the majority of the NA segment replaced by an H3 HA coding region sequence flanked by NA packaging sequences. The virus thus included HAs from two different types of swine influenza, H1 HA and H3 HA. Because NA is essential for virus propagation, the function of NA was provided by growing the virus in the presence of sialidase (neuraminidase).

In particular, in order to generate a recombinant swine influenza virus carrying two different HA molecules, the NA segment in an H1N1 swine influenza virus, A/swine/Saskatchewan/18789/02, termed “SIV SK02” herein (obtained from the Prairie Diagnostic Services, Western College of Veterinary Medicine, University of Saskatchewan, Canada) was replaced with an H3 HA segment from the H3N2 Influenza A virus, A/Swine/Texas/4199-2/98 (termed “SIV Tx98” herein) (FIG. 1A). The H3 HA open reading frame (ORF) derived from SIV-Tx98 was flanked by NA packaging signals that included 202 nt at the 3′ end (19 nt from the 3′ UTR and 183 nt from the 3′ NA coding region) and 185 nt at the 5′ end (28 nt from the 5′ UTR and 157 nt from the 5′ NA coding region) from SIV-SK02 (H1N1) strain (FIG. 1B). Plasmid pHW-SIV-NA-H3HA encoding H3 HA flanked by NA packaging signals was constructed by modifying an original plasmid pHW-SIV/SK-NA. Briefly, the NA segment-specific packaging signals at 3′ and 5′ ends (202 nt and 185 nt respectively), were amplified by polymerase chain reaction (PCR) using pHW-SIV/SK-NA as template and the following sets of primers: for amplifying 3′ NA packaging signal, 5′-TAATACGACTCACTATAGGG-3′ (SEQ ID NO:21) and 5′-GTCATTTCCGGGAAGTTTTTGCACCCAAGTATTGTTTTCGTAG-3′ (SEQ ID NO:22) were used; for amplifying 5′ NA packaging signal, 5′-GCTGAAATCAGGATACAAAGATTGAGGCCTTGCTTCTGGGTTG-3′ (SEQ ID NO:23) and 5′-ACAGGTGTCCGTGTCGCG-3′ (SEQ ID NO:24) were used. H3 HA ectodomain (excluding signal peptide sequence) from SIV/Tx98 was amplified by PCR using pHW-Tx98 HA as a template and 5′-CTACGAAAACAATACTTGGGTGCAAAAACTTCCCGGAAATGAC-3′ (SEQ ID NO:25) and 5′-CAACCCAGAAGCAAGGCCTCAATCTTTGTATCCTGATTTCAGC-3′ (SEQ ID NO:26) as primers. The three pieces of PCR products were joined together by overlapping PCR. Finally, this PCR product was digested by NaeI/NheI and replaced the corresponding segment in pHW-SIV/SK-NA. The constructed plasmid was DNA-sequenced to ensure that additional mutations were not introduced during the overlapping PCRs.

This construct was inserted into cloning vector pHW2000 (Hoffmann et al., Proc. Natl. Acad. Sci. USA (2000) 97:6108-6113) to render plasmid-606. Cloning vector pHW2000 contains 225 bp of the human pol I promoter and 33 bp of the murine terminator sequence separated by two BsmB1 sites. The pol I promoter and terminator elements are flanked by a truncated immediate-early promoter of human cytomegalovirus and by the gene encoding bovine growth hormone.

The pHW2000 vector was cotransfected with plasmid-606 and seven plasmids which included the PB2, PB1, PA, HA, NP, M and NS segments from SIV-SK02 strain as described in Masic et al., J. Gen. Virol. (2009) 90:375-385, in the presence of 10 mU/ml of vibrio cholera sialidase which resulted in successful rescuing of a recombinant, chimeric virus termed “SIV/SK-606 or SIV-606” This SIV/H1H3 SIV mutant virus was rescued using an 8-plasmid reverse genetics system described by Hoffmann et al., Proc. Natl. Acad. Sci. USA (2000) 97:6108-6113. Briefly, 293T and MDCK cells were co-cultured at the same density (2.5×105 cells/well) in a 6-well plate and maintained in DMEM containing 10% FBS at 37° C., 5% CO2 for 24 hrs. One hour prior to transfection, medium containing FBS was replaced with fresh Opti-MEM (Invitrogen). To rescue SIV/SK-606, cells were transfected with eight plasmid constructs (pHW-SIV/SK-PB2, pHW-SIV/SK-PB1, pHW-SIV/SK-PA, pHW-SIV/SK-HA, pHW-SIV/SK-NP, pHW-SIV-NA-H3HA, pHW-SIV/SK-M and pHW-SIV/SK-NS) by Transit-LT1 transfection reagent (Minis, Madison, Wis.). Six hours later, the transfection mixture was replaced with 1 ml of fresh Opti-MEM. Twenty four hours post-transfection, one ml of Opti-MEM containing 0.4% BSA, 2 μg/ml of TPCK-treated trypsin and 10 mU/ml Vibrio cholerae neuraminidase (Sigma, N6514) was added to transfected cells. Supernatant was collected at 96 hours post transfection. Cytopathogenic effect (CPE) was observed after the third consecutive passage on MDCK cells and virus presence was confirmed by a hemagglutination test.

Example 2 Characterization of SIV/SK-606

To confirm that recombinant virus SIV/SK-606 possessed both H1 and H3 HA segments in its genome, viral RNA was isolated from purified virons. Briefly, tissue culture grown viruses were collected by ultracentrifugation and subjected to a sucrose gradient centrifugation (Masic et al., J. Gen. Virol. (2009) 90:375-385). For RNA purification, purified virions were processed following manufacturer's instruction of Trizol (Invitrogen). Reverse transcription was performed using the Uni12 primer (Hoffman et al., Arch. Virol. (2001) 146:2275-2289) which specifically amplifies viral RNAs. PCR was carried out by using primers specific for H1 (Fw 5′ TGGCCAAACCATGAGACAAC 3′ (SEQ ID NO:27) and Bw 5′ GGCGTTATTCCTCAGTTGTG 3′ (SEQ ID NO:28)) and H3 HAs (Fw 5′ CGCAATCGCAGGTTTCATAG 3′ (SEQ ID NO:29) and Bw 5′ CAACCCAGAAGCAAGGCCTCAATCTTTGTATCCTGATTTCAGC 3′ (SEQ ID NO:30)).

While PCR products representing the H1 HA segment were detected only in the SK02 and the SIV-606 genomes, PCR bands representing the H3 HA segment were observed in Tx98 and SIV-606 viral RNA extraction. These data demonstrated that the genome of SIV-606 included both H1 and H3 HA segments.

To examine whether both HAs were expressed, viral infected cell lysates were subjected to Western blotting analysis using antibodies specific for H1HA, H3HA and M1. Briefly, MDCK cells were infected with wild-type SIV/SK02, wild-type SIV/Tx98, or SIV-606 at an MOI of 0.01. At 48 hours post-infection, cell lysates were prepared and were subjected to Western blotting analysis using antibodies specific for H1 HA (Anti-HA (A/California/06/2009((H1N1) monoclonal antibody, eEnzyme (Maryland, USA), H3 HA (Anti-multi-Hemagglutinins (H3N2) Antibody, rabbit IgG, eEnzyme (Maryland, USA) and M1 (Shin et al., J Gen Virol (2007) 88:942-950).

M1 protein was detected in all virus infected cells, however, H1 HA was seen only in SK02 and SIV-606 infected cells and H3 HA was seen in Tx98 and SIV-606 samples. Together, these data demonstrate that the H3 segment was incorporated into the genome of SIV-606 and both HAs were expressed.

To observe the morphology of the recombinant virus, negative staining transmission electron microscopy was performed. The majority of virions exhibited spherical enveloped particles of approximately 100 nm in diameter, which resembled the morphology of the wild type virus.

The replication potential of the SIV-606 was investigated in MDCK cells. In the presence of sialidase, SIV-606 formed plaques similar in size as wild-type virus. In contrast, SIV-606 did not grow in the absence of sialidase, indicating that replication of the recombinant virus was dependent on sialidase. The growth potential and kinetics of SIV-606 were also determined. As shown in FIG. 2, SIV-606 reached a plateau at 24 h.p.i. as did the wild-type virus. SIV-606 grew to a titer of 7×106 PFU/ml, which was approximately 1 log lower than wild-type virus. These results indicated that although SIV-606 had a slightly lower titer, it grew to relatively high titer in cell culture, which enables propagation of the virus.

Example 3 Pathogenicity of SIV-606 in Pigs

The pathogenicity of SIV-606 was evaluated in pigs. Thirty-five 4-week old SIV-negative pigs were split randomly into seven groups of five pigs. These were infected intratracheally with 4 ml MEM containing 1×105 or 1×106 PFU/ml SK02 WT, SIV-606 or Tx98. The animals in the control group were mock infected with medium only (Table 1). Rectal temperature was monitored daily. On day 5 post infection pigs were euthanized and necropsies were performed. As shown in FIGS. 3A-3C, pigs infected with wild-type viruses had an increased temperature on day 1 post infection, and the temperature decreased gradually on the following days. However, pigs infected by SIV-606 did not show elevated temperatures compared to the control group. At necropsy, the macroscopic lung lesions were documented. The mock, SIV-606 high dose- and low dose-infected pigs did not show any typical macroscopic lung lesions. In contrast, gross lesions characterized as purple- to plum-colored consolidated areas were observed in cardiac lobes of pigs infected by SK02 and Tx98 with high and low doses. In agreement with these results, SK02 wild type virus could be recovered from lung tissue of all animals infected with low and high doses of SK02 with median titers of 2.4×104 PFU/ml and 2.6×104 PFU/ml respectively. Similarly, wild type virus could be isolated from lung tissue of all pigs infected with Tx98 virus with median titers of 1×104 PFU/ml and 3.4×104 PFU/ml in low and high dose groups. However, SIV-606 virus was only detected from one pig in the low dose group and 3 pigs from the high dose group with a very low titer (median titers were 0 and 20 PFU/ml respectively). These results demonstrated that the SIV-606 virus is highly attenuated in pigs and thus can be used as a live, attenuated vaccine for swine influenza.

TABLE 1 Assignment of pigs, dose and route of virus infection Group N = 5 Inoculum Concentration Dose Volume Route 1 MEM 4 ml Intratracheal 2 SK02-WT 105 PFU/ml 4 ml Intratracheal 3 SK02-WT 106 PFU/ml 4 ml Intratracheal 4 SIV-606 105 PFU/ml 4 ml Intratracheal 5 SIV-606 106 PFU/ml 4 ml Intratracheal 6 Tx98 105 PFU/ml 4 ml Intratracheal 7 Tx98 106 PFU/ml 4 ml Intratracheal

Example 4 Protective Effect of SIV-606 in Pigs

To determine whether SIV-606 was immunogenic and could provide protection from SIV infection, the following vaccination and viral challenge studies were performed in pigs. Twenty three H1N1 and H3N2 sero-negative pigs were randomly divided into five groups (n=5, except n=3 in group 5) (Table 2). Two groups of pigs were given MEM and two groups of pigs were vaccinated with 4×106 PFU of SIV-606 virus intratracheally. Pigs received a second vaccination on day 21. Ten days after the second vaccination (on day 31), pigs were challenged intratracheally with either SIV/SK02 or SIV/Tx98 and were euthanized on day 5 post infection. Sera were collected prior to the first vaccination, 21 days after the first vaccination and 10 days after the second vaccination (before viral challenge). Antigen specific serum IgG and nasal IgA were measured on day 0, 21 and 31.

After the first vaccination, SIV/SK02 specific IgG in serum increased significantly and the second dose of SIV-606 boosted IgG response measured on day 31 (FIG. 14A). Serum IgG against SIV/Tx98 or a heterologous H1N1 Halifax210 strain, which was isolated during a 2009 pandemic, increased slightly after one vaccination and increased significantly after the second vaccination (FIGS. 14B and 14C).

To assess the presence of IgA antibodies specific for H1N1 and H3N2 influenza viruses at mucosal surfaces in the upper and lower respiratory tract, nasal swabs and bronchoalveolar lavage fluid (BALF) samples from pigs in all groups were collected and tested by ELISA. The first vaccination of SIV-606 induced moderate IgA levels in nasal secretion and the second vaccination boosted IgA induction specific to SIV/SK02, SIV/Tx98 and Halifax210 (FIGS. 15A, 15B and 15C). Similarly, IgA titers remained low in BALF after the first vaccination and were significantly higher after the second vaccination (FIGS. 16A, 16B and 16C).

After the viral challenge on day 31, rectal temperature was measured daily for 5 days until necropsy. On day 1 post infection, the pigs vaccinated with MEM and challenged with SIV/SK02 had an onset of fever with a mean rectal temperature of 40.9° C. In contrast, pigs vaccinated with SIV-606 and challenged with SIV/SK02 had a normal temperature ranging between 39.1° C. to 39.6° C. during these five days (FIG. 17A). Similarly, the temperature of pigs vaccinated with MEM and challenged with Tx98 rose to 40.1° C. on day 1 post infection then decreased to 39.6° C. the following day and went back to 39.3° C. on day 5 post infection. Fever was not seen in pigs vaccinated with SIV-606 and challenged with Tx98 (FIG. 17B). The temperature in this group fluctuated between 39.2° C. and 39.7° C. during the 5 days post infection.

At necropsy, SIV-induced gross lung lesions were examined and scored by the percentage of surface that lesions took up compared to the total lung area (FIG. 18A). All pigs in the unvaccinated groups and challenged with SIV/SK02 or SIV/Tx98 manifested SIV typical gross lesions seen as clear demarcation of dark purple, consolidated areas mostly found in the apical and cardiac lobes. The mean score for these two groups were 8.6 and 14.6, respectively. In contrast, the lungs of pigs vaccinated with SIV-606 and challenged with either SIV/SK02 or SIV/Tx98 had no gross lung lesions.

To measure the viral load in the lungs (FIG. 18B), tissues from the right apical, cardiac and diaphragmatic lobes were collected at necropsy. Virus was detected in the lung tissues from all pigs in the unvaccinated and SIV/SK02 challenged group (mean viral titer was 1.90×104 PFU per gram). In the unvaccinated and SIV/Tx98 challenged group, virus was only isolated from one pig. No virus was detected in the lung tissues of pigs vaccinated with SIV-606 and challenged with SIV.

Histophathological lesions were examined using lung tissue samples taken from the right apical, cardiac, and diaphragmatic lobes at necropsy. As shown in FIGS. 19B and 19D, pathological lesions were observed in the lung tissues of unvaccinated and virus challenged groups. The histophathological lesions included the loss of bronchial epithelium due to the necrosis of bronchiolar epithelium, hypertrophy and hyperplasia of bronchiolar epithelium to compensate for the necrosis of bronchiolar epithelium, neutrophil infiltration through the mucosa and into the lumen of bronchioles, peribronchiolar and perivascular lymphocyte infiltration, interstitial thickening, and proliferation of the bronchiolar associated lymphoid tissues. In contrast, no histopathological changes were observed in the lung tissues of SIV-606 vaccinated and challenged groups (FIGS. 19C and 19E). Both SIV-606 vaccinated groups maintained healthy bronchiolar epithelium and alveolar structures with mild interstitial thickening, similar to the tissue shown in the unvaccinated and unchallenged group (FIG. 19A).

TABLE 2 Assignment of pigs for virus challenge Vaccination Challenge Group 1 (day 0) 2 (day 21) (day 31) 1 (n = 5) MEM MEM SIV/SK02 2 (n = 5) MEM MEM SIV/Tx98 3 (n = 5) SIV-606 SIV-606 SIV/SK02 4 (n = 5) SIV-606 SIV-606 SIV/Tx98 5 (n = 3)

Thus, recombinant, chimeric influenza viruses are disclosed, as well as compositions and methods for treating and preventing influenza. Although preferred embodiments of the subject invention have been described in some detail, it is understood that obvious variations can be made without departing from the spirit and the scope of the invention as defined by the appended claims.

Claims

1. A recombinant, chimeric porcine influenza virus comprising more than one hemagglutinin (HA) segment (segment 5) from more than one influenza subtype, wherein said virus comprises segments 1-5, 7 and 8 from a first influenza subtype and a second segment 5 from a second influenza subtype, and further wherein all or a portion of the neuraminidase (NA) segment (segment 6) of the first influenza subtype is missing to render an attenuated virus.

2. The recombinant, chimeric porcine influenza virus of claim 1, wherein said second segment 5 comprises NA packaging sequences from said first influenza subtype located 3′ and optionally 5′ to said second segment 5.

3. The recombinant, chimeric porcine influenza virus of claim 2, wherein the NA packaging sequences comprise 3′ NA packaging sequences from the 3′ NA UTR and the 3′ NA coding sequence and, optionally 5′ NA packaging sequences from the 5′ NA UTR and the 5′ NA coding sequence.

4. The recombinant, chimeric porcine influenza virus of claim 1, wherein the influenza virus is derived from an influenza A virus.

5. The recombinant, chimeric porcine influenza virus of claim 4, wherein the influenza virus comprises an HA segment from an H1N1 subtype and an HA segment from an H3N2 subtype.

6. The recombinant, chimeric porcine influenza virus of claim 1, wherein the first influenza subtype is H1N1.

7. The recombinant, chimeric porcine influenza virus of claim 6, wherein the H1N1 subtype is A/swine/Saskatchewan/18789/02.

8. The recombinant, chimeric porcine influenza virus of claim 1, wherein the second influenza subtype is H3N2.

9. The recombinant, chimeric porcine influenza virus of claim 8, wherein the H3N2 subtype is A/Swine/Texas/4199-2/98.

10. An attenuated, recombinant, porcine influenza virus comprising segments 1-5, 7 and 8 from an H1N1 influenza subtype, and segment 5 from an H3N2 influenza subtype, wherein all or a portion of segment 6 from the H1N1 influenza subtype is missing, wherein the H3N2 segment 5 is flanked by NA packaging sequences from said H1N1 subtype, wherein the packaging sequences comprise 3′ NA packaging sequences from the 3′ NA UTR and the 3′ NA coding sequence and 5′ NA packaging sequences from the 5′ NA UTR and the 5′ NA coding sequence.

11. The attenuated, recombinant porcine influenza virus of claim 10, wherein the H1N1 subtype is A/swine/Saskatchewan/18789/02 and the H3N2 subtype is A/Swine/Texas/4199-2/98.

12. A composition comprising the recombinant virus of claim 1 and a pharmaceutically acceptable excipient.

13. The composition of claim 12, further comprising an adjuvant.

14. A composition comprising the recombinant virus of claim 10 and a pharmaceutically acceptable excipient.

15. The composition of claim 14, further comprising an adjuvant.

16. A method of eliciting an immunological response in a vertebrate subject, comprising administering the composition of claim 12 to said subject.

17. A method of eliciting an immunological response in a vertebrate subject, comprising administering the composition of claim 14 to said subject.

18. A method of treating or preventing an influenza infection in a vertebrate subject, comprising administering to said subject a therapeutically effective amount of the composition of claim 14.

19. A method of vaccinating a subject against an influenza virus, comprising administering an effective amount of the composition of claim 14 to said subject.

20. The method of claim 16, wherein the subject is a porcine subject.

21. The method of claim 20, wherein the influenza virus is a swine influenza virus.

22. A recombinant construct comprising:

(a) a porcine influenza H3N2 subtype HA segment; and
(b) porcine influenza H1N1 subtype NA packaging sequences located 3′ and optionally 5′ to said H3N2 HA segment.

23. The recombinant construct of claim 22, wherein the H3N2 HA segment is flanked by H1N1 NA packaging sequences that comprise 3′ NA packaging sequences from the 3′ NA UTR and the 3′ NA coding sequence and 5′ NA packaging sequences from the 5′ NA UTR and the 5′ NA coding sequence.

24. The recombinant construct of claim 23, wherein the H1N1 subtype is A/swine/Saskatchewan/18789/02 and the H3N2 subtype is A/Swine/Texas/4199-2/98.

25. A method of producing a recombinant, chimeric influenza virus, comprising transfecting a host cell with (a) individual plasmids comprising segments 1-5, 7 and 8 from an H1N1 influenza subtype; and (b) a recombinant construct according to claim 22, and culturing said host cell under conditions that result in the production of said recombinant, chimeric influenza virus.

26. A cell transformed with (a) individual plasmids comprising segments 1-5, 7 and 8 from an H1N1 influenza subtype; and (b) a recombinant construct according to claim 22.

27. A method of producing a composition comprising combining the recombinant, chimeric porcine influenza virus of claim 1 with a pharmaceutically acceptable excipient.

28. A method of producing a composition comprising combining the recombinant, chimeric porcine influenza virus of claim 10 with a pharmaceutically acceptable excipient.

29. A method of producing an influenza vaccine comprising:

(a) propagating the recombinant, chimeric porcine influenza virus of claim 1;
(b) purifying the virus; and
(c) combining the purified virus with a pharmaceutically acceptable excipient.

30. A method of producing an influenza vaccine comprising:

(a) propagating the recombinant, chimeric porcine influenza virus of claim 10;
(b) purifying the virus; and
(c) combining the purified virus with a pharmaceutically acceptable excipient.

31. A kit comprising one or more containers wherein the one or more containers comprise the recombinant virus of claim 1.

Patent History
Publication number: 20130189303
Type: Application
Filed: Jul 27, 2012
Publication Date: Jul 25, 2013
Inventors: Yan Zhou (Saskatoon), Aleksandar Masic (Belleville)
Application Number: 13/559,940