Influenza B viruses having alterations in the hemagglutinin polypeptide

Abstract
The present invention encompasses methods of producing influenza B viruses in cell culture. The influenza B viruses may have desirable characteristics, such as enhanced replication in eggs and may be used, for example, in vaccines and in methods of treatment to protect against influenza B virus infection.
Description
BACKGROUND OF THE INVENTION

Influenza viruses are made up of an internal ribonucleoprotein core containing a segmented single-stranded RNA genome and an outer lipoprotein envelope lined by a matrix protein. Influenza A and B viruses each contain eight segments of single stranded RNA with negative polarity. The eight genome segments of influenza B encode 11 proteins. The three largest genes code for components of the RNA polymerase, PB 1, PB2 and PA. Segment 4 encodes the HA protein. Segment 5 encodes NP. Segment 6 encodes the NA protein and the NB protein. Both proteins, NB and NA, are translated from overlapping reading frames of a biscistronic mRNA. Segment 7 of influenza B also encodes two proteins: M1 and BM2. The smallest segment encodes two products: NS1 is translated from the full length RNA, while NS2 is translated from a spliced mRNA variant.


Vaccines capable of producing a protective immune response specific for influenza viruses have been produced for over 50 years. Vaccines can be characterized as whole virus vaccines, split virus vaccines, surface antigen vaccines and live attenuated virus vaccines. While appropriate formulations of any of these vaccine types is able to produce a systemic immune response, live attenuated virus vaccines are also able to stimulate local mucosal immunity in the respiratory tract.


FluMist™ is a live, attenuated vaccine that protects children and adults from influenza illness (Belshe et al. (1998) The efficacy of live attenuated, cold-adapted, trivalent, intranasal influenza virus vaccine in children N Engl J Med 338:1405-12; Nichol et al. (1999) Effectiveness of live, attenuated intranasal influenza virus vaccine in healthy, working adults: a randomized controlled trial JAMA 282:137-44). FluMist™ vaccine strains contain HA and NA gene segments derived from the currently circulating wild-type strains along with six internal gene segments from a common master donor virus (MDV).


To date, commercially available influenza vaccines in the United States are propagated in embryonated hen's eggs. Many strains of influenza B viruses do not grow well in eggs and must become “egg-adapted.” Unfortunately, egg adaptation of influenza B viruses results in loss of an N-linked glycosylation site at amino acid residue 196 or 197 of the HA polypeptide. Loss of the N-linked glycosylation site affects virus antigenicity and corresponding vaccine efficacy. Stabilization of the N-linked glycosylation site in influenza B viruses grown in eggs could be of significance in, inter alia, influenza B vaccine manufacture.


SUMMARY OF THE INVENTION

One embodiment of the invention encompasses a method of preparing an influenza B virus. A mutation resulting in an amino acid substitution at HA position 141 to arginine is introduced into an influenza B virus genome. The mutated influenza B virus genome is replicated under conditions whereby influenza B virus is produced.


Another embodiment of the invention encompasses a method of preparing an influenza B virus. A plurality of vectors is introduced into a population of host cells. The vectors comprise nucleotide sequences corresponding to: (a) at least 6 internal genome segments of a first influenza B strain, and (b) one or more genome segments encoding HA and NA polypeptides of at least a second influenza B strain. The HA polypeptide comprises an arginine at amino acid residue 141. The population of host cells is cultured at a temperature that does not exceed 35 degrees. The influenza virus is recovered.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1: Illustration of pAD3000 plasmid.



FIG. 2: Illustration of eight plasmid system for the production of influenza B virus.



FIG. 3A-D: FIG. 3A and FIG. 3B, Characterization of recombinant MDV-B virus by RT-PCR, PB1 (SEQ ID NO: 40), HA (SEQ ID NO: 41), NS (SEQ ID NO: 42); FIG. 3C and FIG. 3D; Characterization of recombinant B/Yamanashi/166/98 by RT-PCR, wt-B/Yamanashi/166/98 (SEQ ID NO: 43), rec-B/Yamanashi/166/98 (SEQ ID NO: 44).



FIGS. 4A and B: Sequence of pAD3000 in GeneBank format (SEQ ID NO:3).


FIG. 5A-5AE: Sequence alignment with MDV-B and eight plasmids, FIGS. 5A-E, PB1 segment, MDV-B-PB1 (SEQ ID NO:4) and pAB121-PB1 (SEQ ID NO: 45); FIGS. 5F-J, PB2 segment, MDV-B-PB2 and pAB122-PB2 (SEQ ID NO:5); FIGS. 5K-0,PA segment, MDV-B-PA and pAB123-PA (SEQ ID NO:6); FIGS. 5P-5S, HA segment, MDV-B-HA (SEQ ID NO: 46) and pAB124-HA (SEQ ID NO:7); FIGS. 5T-5W, NP segment, MDV-B-NP and pAB125-NP (SEQ ID NO:8); FIGS. 5X-5Z, NA segment, MDV-B-NA and pAB126-NA (SEQ ID NO:9); FIGS. 5AA-5AC, M segment, MDV-B-M and pAB127-M (SEQ ID NO:10); FIGS. 5AD-5AE, NS segment, MDV-B-NS (SEQ ID NO:11) and pAB128-NS (SEQ ID NO: 47).



FIG. 6: RT-PCR products derived from simultaneous amplification of HA and NA segments of influenza B strains.



FIG. 7: Bar graph illustrating relative titers of recombinant and reassortant virus.



FIG. 8: Schematic illustration of triple-gene recombinants with wild type residues in PA, NP, and M1 proteins.



FIG. 9: Tabulation of growth of single-gene and double-gene recombinant viruses.



FIG. 10: Tabulation of amino acid residue of the nucleoprotein corresponding to non-ts phenotype.



FIG. 11: Bar graphs illustrating differential replication of reassortant viruses. Gray boxes represent wild type amino acid residues. The dotted line represents the shut-off temperature (ts) of 2.0 log10.



FIG. 12: Alignment of the HA sequences near the 196/197 glycosylation site of several egg amplified influenza B strains. The Victoria lineage viruses are aligned with reference strain B/Victoria/2/87 (SEQ ID NO:12). The Yamagata lineage viruses are aligned with B/Yamagata/16/88 (SEQ ID NO:13). Only the residues differing from the reference strain are shown in the alignment. The potential N-glycosylation site (N—X-T/S) at position of 196/197 is indicated as underlined and in arrow. “.” indicates amino acid deletion in the B/Yamagata lineages. “x” indicates mixed amino acid.



FIG. 13: Confirmation of HA glycosylation by Western Blot. 6:2 B/Shanghai/361/02 (B/SH), 6:2 B/Jilin/20/03 (B/JL) and 6:2 B/Jiangsu/10/03 (B/JS) with the indicated 196-199 sequence (SEQ ID NOS: 48, 49, 49, 48, 50, 48, 50, 51, 48 and 49, respectively, in order of appearance) were electrophoresed on 10% SDS-PAGE. The HA 1 and HA2 proteins were detected by Western Blotting using polyclonal anti HA antibody. Underlining indicates the original sequence present in the virus egg isolate.



FIG. 14: Egg-grown viruses with arginine at HA residue 141 retain glycosylation at residue 196-197. 6:2 B/Shanghai/361/02 (B/SH), 6:2 B/Ohio/1/05 (B/Ohio) and 6:2 B/Jiangsu/10/03 (B/JS) with indicated residues at 141 and 196/197 were grown in eggs and viruses were electrophoresed on 10% SDS-PAGE. The HA1 and HA2 proteins were detected by Western Blotting using polyclonal anti HA antibody. The slower migrating HA1 indicates the 196/197 site was glycosylated as indicated by *. Underlining indicates the original sequence present in the virus egg isolate.





DETAILED DESCRIPTION

The present invention encompasses a system for producing influenza B viruses by introducing vectors into cultured cells. The influenza B viruses produced by the method may have amino acid residues at particular positions which influence the viruses ability to replicate in eggs, or may influence the characteristics of the viruses once replicated in eggs.


Unless defined otherwise, all scientific and technical terms are understood to have the same meaning as commonly used in the art to which they pertain. For the purpose of the present invention the following terms are defined below.


A “nucleic acid,” “polynucleotide,” “polynucleotide sequence” and “nucleic acid sequence” may be a single-stranded or double-stranded deoxyribonucleotide or ribonucleotide polymer, or a chimera or analogue thereof. These terms may also include polymers of analogs of naturally occurring nucleotides having the essential nature of natural nucleotides in that they hybridize to single-stranded nucleic acids in a manner similar to naturally occurring nucleotides (e.g., peptide nucleic acids).


A “gene” may refer to any nucleic acid associated with a biological function. Genes include coding sequences and/or the regulatory sequences required for their expression. A “gene” may refer to a specific genomic sequence, as well as to a cDNA or an mRNA encoded by that genomic sequence.


Genes may further include non-expressed nucleic acid segments that, for example, form recognition sequences for other proteins. Non-expressed regulatory sequences include “promoters” and “enhancers,” to which regulatory proteins such as transcription factors bind, resulting in transcription of adjacent or nearby sequences. A “tissue specific” promoter or enhancer is one which regulates transcription in a specific tissue type or cell type, or types.


A “vector” may be a means by which a nucleic can be propagated and/or transferred between organisms, cells, or cellular components. Vectors include plasmids, viruses, bacteriophage, pro-viruses, phagemids, transposons, and artificial chromosomes, and the like, that replicate autonomously or can integrate into a chromosome of a host cell. A vector can also be a naked RNA polynucleotide, a naked DNA polynucleotide, a polynucleotide composed of both DNA and RNA within the same strand, a poly-lysine-conjugated DNA or RNA, a peptide-conjugated DNA or RNA, a liposome-conjugated DNA, or the like, that are not autonomously replicating.


An “expression vector” may be a vector, such as a plasmid, which is capable of promoting expression, as well as replication of a nucleic acid incorporated therein. A nucleic acid to be expressed may be “operably linked” to a promoter and/or enhancer, and subject to transcription regulatory control by the promoter and/or enhancer.


A “bi-directional expression vector” is typically characterized by two alternative promoters oriented in opposite directions relative to a nucleic acid situated between the two promoters, such that expression can be initiated in both orientations resulting in, e.g., transcription of both plus (+) or sense strand, and negative (−) or antisense strand RNAs. Alternatively, the bi-directional expression vector can be an ambisense vector, in which the viral mRNA and viral genomic RNA (as a cRNA) are expressed from the same strand.


“Isolated,” when referring to a biological material, such as a nucleic acid or a protein, may be a biological material which is substantially free from components that normally accompany or interact with it in its naturally occurring environment. The isolated material may optionally comprise materials not found with the material in its natural environment, e.g., a cell.


“Recombinant” may indicate a material (e.g., a nucleic acid or protein) that has been artificially or synthetically (non-naturally) altered by human intervention. The alteration can be performed on the material within, or removed from, its natural environment or state.


Reassortant viruses include viruses that include genetic and/or polypeptide components derived from more than one parental viral strain or source. For example, a 7:1 reassortant includes 7 viral genomic segments (or gene segments) derived from a first parental virus, and 1 viral genomic segment, e.g., encoding hemagglutinin or neuraminidase, from a second parental virus. A 6:2 reassortant includes 6 genomic segments, most commonly the 6 internal genes from a first parental virus, and two genomic segments, e.g., hemagglutinin and neuraminidase, from a second parental virus. A 6:1:1 reassortant may include 6 genomic segments, most commonly the 6 internal genes from a first parental virus, 1 genomic segment from a second parental virus encoding hemagglutinin, and 1 genomic segment from a third parental virus encoding neuraminidase. The 6 internal genes may be those of more than one parental virus as well.


Introduction of vectors or nucleic acids may refer to the incorporation of the nucleic acids into a eukaryotic or prokaryotic cell. The vectors or nucleic acids may be incorporated into the cell by incorporation in its genome (e.g., chromosome, plasmid, plastid or mitochondrial DNA), may be converted into an autonomous replicon, or may be transiently expressed (e.g., transfected mRNA). Introduction includes such methods as “infection,” “transfection,” “transformation” and “transduction.” Introduction may be performed by electroporation, calcium phosphate precipitation, or lipid mediated transfection (lipofection).


A host cell may be a cell which contains a heterologous nucleic acid, such as a vector, and which supports the replication and/or expression of the nucleic acid. Host cells can be prokaryotic cells such as E. coli, or eukaryotic cells such as yeast, insect, amphibian, avian or mammalian cells, including human cells. Host cells include Vero (African green monkey kidney) cells, Per.C6 cells (human embryonic retinal cells), BHK (baby hamster kidney) cells, primary chick kidney (PCK) cells, Madin-Darby Canine Kidney (MDCK) cells, Madin-Darby Bovine Kidney (MDBK) cells, 293 cells (e.g., 293T cells), and COS cells (e.g., COS1, COST cells). Host cell also encompasses combinations or mixtures of cells including, e.g., mixed cultures of different cell types or cell lines (e.g., Vero and CEK cells). Co-cultivation of electroporated Vero cells is described, for example, in PCT/US04/42669 filed Dec. 22, 2004, which is incorporated by reference in their entirety.


A temperature sensitive (ts) virus typically exhibits a 100-fold or greater reduction in titer at 37° C. relative to 33° C. for influenza B strains. A cold adapted (ca) virus typically exhibits growth at 25° C. within 100-fold of its growth at 33° C. An attenuated (att) virus typically replicates in the upper airways of ferrets but is not detectable in lung tissues, and does not cause influenza-like illness in the animals. Growth indicates viral quantity as indicated by titer, plaque size or morphology, particle density or other measures known to those of skill in the art.


An artificially engineered virus, viral nucleic acid, or virally encoded product, e.g., a polypeptide, a vaccine, is a virus, nucleic acid or product, which includes at least one mutation introduced by recombinant methods, e.g., site directed mutagenesis, PCR mutagenesis, etc. An artificially engineered virus (or viral component or product) comprising one or more nucleotide mutations and/or amino acid substitutions indicates that the viral genome or genome segment encoding the virus (or viral component or product) is not derived from naturally occurring sources, such as a naturally occurring or previously existing laboratory strain of virus produced by non-recombinant methods (such as progressive passage at 25° C.), e.g., a wild type or cold adapted A/Ann Arbor/6/60 or B/Ann Arbor/1/66 strain.


Vectors


In some methods encompassed by the invention, viral genomic segments corresponding to each of the eight segments of the influenza B virus may be inserted into a plurality of vectors for manipulation and production of influenza viruses. Eight vectors may be included in the plurality of vectors; eight vectors comprising nucleic acid sequences corresponding to the eight genomic segments of one or more influenza B viruses. The plurality of vectors may include more or fewer vectors. For instance, 11 vectors may be included in the plurality of vectors; 11 vectors comprising nucleic acid sequences corresponding to the coding sequences of the 11 influenza B virus proteins. Alternatively, one vector may be included in the plurality of vectors; one vector comprising each of the eight genomic segments of the one or more influenza B viruses. Two, three, four, five, six, seven, nine, or ten vectors may also be included in the plurality of vectors.


The vectors may be viral vectors, plasmids, cosmids, phage, or artificial chromosomes. If the vectors are plasmids, the plasmids may provide one or more origins of replication functional in bacterial and eukaryotic cells, and, optionally, a marker convenient for screening or selecting cells incorporating the plasmid sequence. An example vector, plasmid pAD3000 is illustrated in FIG. 1.


If the vectors are plasmids the plasmids may be bi-directional expression vectors capable of initiating transcription of the viral genomic segments in either direction, that is, giving rise to both (+) strand and (−) strand viral RNA molecules. To effect bi-directional transcription, each of the viral genomic segments is inserted into a vector having at least two independent promoters, such that copies of viral genomic RNA are transcribed by a first RNA polymerase promoter (e.g., Pol I), from one strand, and viral mRNAs are synthesized from a second RNA polymerase promoter (e.g., Pol II). Accordingly, the two promoters are arranged in opposite orientations flanking at least one cloning site (i.e., a restriction enzyme recognition sequence) preferably a unique cloning site, suitable for insertion of viral genomic RNA segments. Alternatively, an “ambisense” vector can be employed in which the (+) strand mRNA and the (−) strand viral RNA (as a cRNA) are transcribed from the same strand of the vector.


Expression Vectors


The influenza virus genome segment to be expressed is operably linked to an appropriate transcription control sequence (promoter) to direct mRNA synthesis. A variety of promoters are suitable for use in expression vectors for regulating transcription of influenza virus genome segments. In certain embodiments, e.g., wherein the vector is the plasmid pAD3000, the cytomegalovirus (CMV) DNA dependent RNA Polymerase II (Pol II) promoter is utilized. If desired, e.g., for regulating conditional expression, other promoters can be substituted which induce RNA transcription under the specified conditions, or in the specified tissues or cells. Numerous viral and mammalian, e.g., human promoters are available, or can be isolated according to the specific application contemplated. For example, alternative promoters obtained from the genomes of animal and human viruses include such promoters as the adenovirus (such as Adenovirus 2), papilloma virus, hepatitis-B virus, polyoma virus, and Simian Virus 40 (SV40), and various retroviral promoters. Mammalian promoters include, among many others, the actin promoter, immunoglobulin promoters, heat-shock promoters, and the like. In addition, bacteriophage promoters can be employed in conjunction with the cognate RNA polymerase, e.g., the T7 promoter.


Transcription is optionally increased by including an enhancer sequence. Enhancers are typically short, e.g., 10-500 bp, cis-acting DNA elements that act in concert with a promoter to increase transcription. Many enhancer sequences have been isolated from mammalian genes (hemoglobin, elastase, albumin, alpha.-fetoprotein, and insulin), and eukaryotic cell viruses. The enhancer can be spliced into the vector at a position 5′ or 3′ to the heterologous coding sequence, but is typically inserted at a site 5′ to the promoter. Typically, the promoter, and if desired, additional transcription enhancing sequences are chosen to optimize expression in the host cell type into which the heterologous DNA is to be introduced (Scharf et al. (1994) Heat stress promoters and transcription factors Results Probl Cell Differ 20:125-62; Kriegler et al. (1990) Assembly of enhancers, promoters, and splice signals to control expression of transferred genes Methods in Enzymol 185: 512-27). Optionally, the amplicon can also contain a ribosome binding site or an internal ribosome entry site (IRES) for translation initiation.


The vectors of the invention may also include sequences necessary for the termination of transcription and for stabilizing the mRNA, such as a polyadenylation site or a terminator sequence. Such sequences are commonly available from the 5′ and, occasionally 3′, untranslated regions of eukaryotic or viral DNAs or cDNAs. In one embodiment, e.g., involving the plasmid pAD3000, the SV40 polyadenylation sequences provide a polyadenylation signal.


In addition, as described above, the expression vectors optionally include one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells, in addition to genes previously listed, markers such as dihydrofolate reductase or neomycin resistance are suitable for selection in eukaryotic cell culture.


The vector containing the appropriate DNA sequence as described above, as well as an appropriate promoter or control sequence, can be employed to transform a host cell permitting expression of the protein.


Additional Expression Elements


A genome segment encoding an influenza virus protein may include any additional sequences necessary for expression of the segment. For example, specific initiation signals which aid in the efficient translation of the heterologous coding sequence may be included. These signals can include, e.g., the ATG initiation codon and adjacent sequences. To insure translation of the entire protein encoded by the genome segment, the initiation codon is inserted in the correct reading frame relative to the viral protein. Exogenous transcriptional elements and initiation codons can be of various origins, both natural and synthetic. The efficiency of expression can be enhanced by the inclusion of enhancers appropriate to the cell system in use.


Additional polynucleotide sequences such as signal sequences, secretion or localization sequences, and the like can be incorporated into the vector, usually, in-frame with the polynucleotide sequence of interest, e.g., to target polypeptide expression to a desired cellular compartment, membrane, or organelle, or into the cell culture media. Such sequences are known to those of skill, and include secretion leader peptides, organelle targeting sequences (e.g., nuclear localization sequences, ER retention signals, mitochondrial transit sequences), membrane localization/anchor sequences (e.g., stop transfer sequences, GPI anchor sequences), and the like.


Internal Genome Segments


Internal genomic segments of an influenza B virus strain may be the internal genomic segments of one or more master influenza B virus. The one or more master influenza B virus may be selected on the basis of desirable properties relevant to vaccine administration. For example, a master donor influenza B virus strain may be selected for an attenuated phenotype, cold adaptation and/or temperature sensitivity. In this context, ca B/Ann Arbor/1/66, or an artificially engineered influenza B strain incorporating one or more of the amino acid substitutions specified in Table 17 may be the master donor influenza B strain. These amino acid substitutions may include substitutions at one or more of PB2630; PA431; PA497; NP55; NP114; NP410; NP509; M1159 and M1183. The amino acid substitutions may include one or more of the following: PB2630 (S630R); PA431 (V431M); PA497 (Y497H); NP55 (T55A); NP114 (V114A); NP410 (P410H); NP509 (A509T); M1159 (H159Q) and M1183 (M183V). The amino acid substitutions may include substitutions at all of PB2630; PA431; PA497; NP55; NP114; NP410; NP509; M1159 and M1183. The substitutions may be all of PB2630 (S630R); PA431 (V431M); PA497 (Y497H); NP55 (T55A); NP114 (V114A); NP410 (P410H); NP509 (A509T); M1159 (H159Q) and M1183 (M183V).


The six internal genomic segments of the one or more influenza master influenza B virus strain, (i.e., PB1, PB2, PA, NP, NB, M1, BM2, NS1 and NS2) may transfected into suitable host cells in combination with hemagglutinin and neuraminidase segments from an antigenically desirable strain, e.g., a strain predicted to cause significant local or global influenza infection. Following replication of the reassortant virus in cell culture at appropriate temperatures for efficient recovery, e.g., equal to or less than 35° C., such as between about 30° C. and 35° C., such as between about 32° C. and 35° C., such as between about 32° C. and 34° C., or at about 30° C., or at about 31° C., or at about 32° C., or at about 33° C., or at about 34° C. or at about 35° C., reassortant viruses is recovered. The recovered virus may be replicated in embryonated eggs. The recovered virus may be replicated in cultured cells. The recovered virus, which may have been replicated in embryonated eggs or cultured cells, may be inactivated using a denaturing agent such as formaldehyde or β-propiolactone.


Influenza B Viruses with Altered Attributes


The methods of the present invention also encompass introducing a mutation resulting in an amino acid substitution at HA position 141. The mutation may increase the ability of the influenza B viruses to replicate in embryonated chicken eggs when compared to HA unsubstituted influenza viruses. The substitution at HA position 141 may further allow the influenza virus to retain glycosylation at HA amino acid residue 196/197. The substitution at HA position 141 may further not significantly alter antigenicity of the HA. The substitution at HA position 141 may be for an arginine, a histine, or a cysteine.


The introduction of the amino acid substitution into HA may enhance the ability of the influenza B virus to replicate in eggs by at least 10%, or by at least 20%, or by at least 30%, or by at least 40%, or by at least 50%, or by at least 60%, or by at least 70%, or by at least 80%, or by at least 90%, or by at least 100%, or by at least 200%, or by at least 300%, or by at least 400%, or by at least 500% when compared to the unmodified influenza virus. The titer of the virus with the enhanced ability to replicate in eggs may be at least 5.0 log10 PFU/ml, at least 6.0 log10 PFU/ml, at least 6.5 log10 PFU/ml, at least 7.0 log10 PFU/ml, at least 7.25 log10 PFU/ml, at least 7.5 log10 PFU/ml, at least 7.75 log10 PFU/ml, at least 8.0 log10 PFU/ml, at least 8.25 log10 PFU/ml, at least 8.5 log10 PFU/ml, at least 8.75 log10 PFU/ml, at least 9.0 log10 PFU/ml, or at least 9.5 log10 PFU/ml. The influenza B virus with the enhanced ability to replicate in eggs when compared to the unmodified influenza virus will also retain HA glycosylation at amino acid residue position 196/197.


The introduction of the amino acid substitution may further not significantly alter the antigenicity of the substituted influenza virus when compared to the unsubstituted virus. The antigenicity of the substituted influenza virus when compared to the unsubstituted virus differs by less then 5%, 10%, 20%, 25%, 30%, 40%, or 50%. Methods to determine viral antigenicity are well known in the art.


Introduction of a mutation which results in the amino acid substitution in the HA at residue position 141 may modulate receptor binding activity of the HA. Receptor binding activity of the HA includes but is not limited to the binding of HA to sialic acid residues (e.g., 2,6-linked sialyl-galactosyl moieties [Siaα(2,6)Gal] and 2,3-linked sialyl-galactosyl moieties [Siaα(2,3)Gal]) present on the cell surface glycoproteins or glycolipids. Methods to assay HA binding are well known in the art. Introduction of the mutation that results in an amino acid substitution at HA residue 141 may enhance the binding of HA to [Siaα(2,3)Gal] moieties. Enhanced binding to [Siaα(2,3)Gal] moieties may be by at least 10%, or by at least 20%, or by at least 30%, or by at least 40%, or by at least 50%, or by at least 60%, or by at least 70%, or by at least 80%, or by at least 90%, or by at least 100%, or by at least 200% in an, e.g., hemaagglutination, assay well known to those of skill in the art.


The influenza B variant virus may further have one or more attributes including attenuation, a cold adaptation, temperature sensitivity, or any combination thereof. The influenza B variant virus may have one or more of these attributes owing to incorporation of internal genome segments of a master influenza B donor virus, such as influenza B/Ann Arbor/1/66.


The influenza B variant virus may be any influenza B virus that comprises an HA polypeptide with a glycine residue at position 141. The influenza B virus HA polypeptide may be that of influenza strain B/Victoria/2/87, B/Hong Kong/330/01, B/Brisbane/32/02, B/Malaysia/2506/04, B/Hawaii/13/04, B/Ohio/1/05, B/Yamagata/16/88, B/Yamanashi/166/98, B/Johannesburg/5/99, B/Vicotria/504/00, B/Shanghai/361/02, B/Jilin/20/03, or B/Florida/7/04.


Cell Culture


In some methods encompassed by the invention, a plurality of vectors is introduced into host cells. These host cells include, e.g., Vero cells, Per.C6 cells, BHK cells, MDCK cells, 293 cells and COS cells, including 293T cells, COST cells. Alternatively, co-cultures including two of the above cell lines, e.g., MDCK cells and either 293T or COS cells may employed at a ratio, e.g., of 1:1. The cells may be maintained in suitable commercial culture medium, such as Dulbecco's modified Eagle's medium supplemented with serum (e.g., 10% fetal bovine serum), or in serum free medium, under controlled humidity and CO2 concentration suitable for maintaining neutral buffered pH (e.g., at pH between 7.0 and 7.2). Optionally, the medium contains antibiotics to prevent bacterial growth, e.g., penicillin, streptomycin, etc., and/or additional nutrients, such as L-glutamine, sodium pyruvate, non-essential amino acids, additional supplements to promote favorable growth characteristics, e.g., trypsin, β-mercaptoethanol, and the like.


Procedures for maintaining mammalian cells in culture have been extensively reported, and are known to those of skill in the art. General protocols are provided, e.g., in Freshney (1983) Culture of Animal Cells: Manual of Basic Technique, Alan R. Liss, New York; Paul (1975) Cell and Tissue Culture, 5th ed., Livingston, Edinburgh; Adams (1980) Laboratory Techniques in Biochemistry and Molecular Biology-Cell Culture for Biochemists, Work and Burdon (eds.) Elsevier, Amsterdam. Additional details regarding tissue culture procedures of particular interest in the production of influenza virus in vitro include, e.g., Merten et al. (1996) Production of influenza virus in cell cultures for vaccine preparation. In Cohen and Shafferman (eds) Novel Strategies in Design and Production of Vaccines, which is incorporated herein in its entirety. Additionally, variations in such procedures adapted to the present invention are readily determined through routine experimentation.


Cells for production of influenza B virus may be cultured in serum-containing or serum free medium. In some case, e.g., for the preparation of purified viruses, it may be desirable to grow the host cells in serum free conditions.


Cells may be cultured on any scale. Cells may be cultured on small scale, e.g., less than 25 ml medium, in culture tubes or flasks or in large flasks with agitation, in rotator bottles, or on microcarrier beads (e.g., DEAE-Dextran microcarrier beads, such as Dormacell, Pfeifer & Langen; Superbead, Flow Laboratories; styrene copolymer-tri-methylamine beads, such as Hillex, SoloHill, Ann Arbor) in flasks, bottles or reactor cultures. Microcarrier beads are small spheres (in the range of 100-200 microns in diameter) that provide a large surface area for adherent cell growth per volume of cell culture. For example a single liter of medium can include more than 20 million microcarrier beads providing greater than 8000 square centimeters of growth surface. For commercial production of viruses, e.g., for vaccine production, it may be desirable to culture the cells in a bioreactor or fermenter. Bioreactors are available in volumes from under 1 liter to in excess of 100 liters, e.g., Cyto3 Bioreactor (Osmonics, Minnetonka, Minn.); NBS bioreactors (New Brunswick Scientific, Edison, N.J.); laboratory and commercial scale bioreactors from B. Braun Biotech International (B. Braun Biotech, Melsungen, Germany).


Regardless of the culture volume, the cultures may be maintained at a temperature less than or equal to 35° C., less than or equal to 34° C., less than or equal to 33° C., less than or equal to 32° C., less than or equal to 31° C., or less than or equal to 30° C. The cells may be cultured at a temperature between about 30° C. and 35° C., between about 32° C. and 35° C., between about 32° C. and about 34° C., or between about 32° C. and 33° C.


Introduction of Vectors into Host Cells


Vectors comprising nucleotide sequences corresponding to influenza genome segments may be introduced (e.g., transfected) into host cells according to methods well known in the art including, e.g., calcium phosphate co-precipitation, electroporation, microinjection, lipofection, and transfection employing polyamine transfection reagents. By way of example, vectors, e.g., plasmids, can be transfected into host cells, such as COS cells, 293T cells or combinations of COS or 293T cells and MDCK cells, using the polyamine transfection reagent TransIT-LT1 (Minis) according to the manufacturer's instructions. Approximately 1 μg of each vector can be introduced into the population of host cells with approximately 2 μl of TransIT-LT1 diluted in 160 μl medium in a total volume of 200 μl. The DNA:transfection reagent mixtures are incubated at room temperature for 45 min followed by addition of 800 μl of medium. The transfection mixture is added to the host cells, and the cells are cultured as described above.


Alternatively, electroporation can be employed to introduce vectors comprising nucleotide sequences corresponding to influenza genome segments into host cells. By way of example, plasmid vectors comprising nucleotide sequences corresponding to influenza B genome segments may be introduced into Vero cells using electroporation according to the following procedure. 5×106 Vero cells, e.g., grown in Modified Eagle's Medium (MEM) supplemented with 10% Fetal Bovine Serum (FBS) are resuspended in 0.4 ml OptiMEM and placed in an electroporation cuvette. Twenty micrograms of DNA in a volume of up to 25 μl is added to the cells in the cuvette, which is then mixed gently by tapping. Electroporation is performed according to the manufacturer's instructions (e.g., BioRad Gene Pulser II with Capacitance Extender Plus connected) at 300 volts, 950 microFarads with a time constant of between 28-33 msec. The cells are remixed by gently tapping and approximately 1-2 minutes following electroporation 0.7 ml MEM with 10% FBS is added directly to the cuvette. The cells are then transferred to two wells of a standard 6 well tissue culture dish containing 2 ml MEM, 10% FBS or OPTI-MEM without serum. The cuvette is washed to recover any remaining cells and the wash suspension is divided between the two wells. Final volume is approximately 3.5 mls. The cells are then incubated under conditions permissive for viral growth.


Recovery of Viruses


Viruses may be recovered from the culture medium of cells into which a plurality of vectors had been introduced. Crude medium may be obtained and clarified, and influenza viruses in the clarified medium may then be concentrated. Common methods of concentration include filtration, ultrafiltration, adsorption on barium sulfate and elution, and centrifugation. By way of example, crude medium from infected cultures may first be clarified by centrifugation at, e.g., 1000-2000×g for a time sufficient to remove cell debris and other large particulate matter, e.g., between 10 and 30 minutes. Alternatively, the medium may be filtered through a 0.8 μm cellulose acetate filter to remove intact cells and other large particulate matter. Optionally, the clarified medium supernatant may then be centrifuged to pellet the influenza viruses, e.g., at 15,000×g, for approximately 3-5 hours. Following resuspension of the virus pellet in an appropriate buffer, such as STE (0.01 M Tris-HCl; 0.15 M NaCl; 0.0001 M EDTA) or phosphate buffered saline (PBS) at pH 7.4, the virus may be concentrated by density gradient centrifugation on sucrose (60%-12%) or potassium tartrate (50%-10%). Either continuous or step gradients, e.g., a sucrose gradient between 12% and 60% in four 12% steps, are suitable. The gradients may be centrifuged at a speed, and for a time, sufficient for the viruses to concentrate into a visible band for recovery. Alternatively, and for large scale commercial applications, virus may be elutriated from density gradients using a zonal-centrifuge rotor operating in continuous mode. Additional details sufficient to guide one of skill through the preparation of influenza viruses from tissue culture are provided, e.g., in Furminger. Vaccine Production, in Nicholson et al. (eds) Textbook of Influenza pp. 324-332; Merten et al. (1996) Production of influenza virus in cell cultures for vaccine preparation, in Cohen & Shafferman (eds) Novel Strategies in Design and Production of Vaccines pp. 141-151, and U.S. Pat. No. 5,690,937. If desired, the recovered viruses can be stored at −80° C. in the presence of sucrose-phosphate-glutamate (SPG) as a stabilizer.


Methods and Compositions for Prophylactic Administration of Vaccines


Recombinant and reassortant viruses of the invention can be administered prophylactically in an appropriate carrier or excipient to stimulate an immune response specific for one or more strains of influenza virus. The carrier or excipient may be a pharmaceutically acceptable carrier or excipient, such as sterile water, aqueous saline solution, aqueous buffered saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, ethanol, allantoic fluid from uninfected Hens' eggs (i.e., normal allantoic fluid “NAF”) or combinations thereof. The preparation of such solutions insuring sterility, pH, isotonicity, and stability is effected according to protocols established in the art. Generally, a carrier or excipient is selected to minimize allergic and other undesirable effects, and to suit the particular route of administration, e.g., subcutaneous, intramuscular, intranasal, etc.


Generally, the influenza viruses of the invention are administered in a quantity sufficient to stimulate an immune response specific for one or more strains of influenza virus. Dosages and methods for eliciting a protective immune response against one or more influenza strains are known to those of skill in the art. By way of example, inactivated influenza viruses may be provided in the range of about 1-1000 HID50 (human infectious dose), i.e., about 105-108 pfu (plaque forming units) per dose administered. Alternatively, about 10-50 μg, e.g., about 15 μg HA is administered without an adjuvant, with smaller doses being administered with an adjuvant. Typically, the dose will be adjusted within this range based on, e.g., age, physical condition, body weight, sex, diet, time of administration, and other clinical factors. The prophylactic vaccine formulation may be systemically administered, e.g., by subcutaneous or intramuscular injection using a needle and syringe, or a needleless injection device. Alternatively, the vaccine formulation may be administered intranasally, either by drops, large particle aerosol (greater than about 10 microns), or spray into the upper respiratory tract. For intranasal administration, attenuated live virus vaccines may be used, e.g., an attenuated, cold adapted and/or temperature sensitive recombinant or reassortant influenza virus. While stimulation of a protective immune response with a single dose is preferred, additional dosages may be administered, by the same or different route, to achieve the desired prophylactic effect.


Alternatively, an immune response can be stimulated by ex vivo or in vivo targeting of dendritic cells with influenza viruses. For example, proliferating dendritic cells can be exposed to viruses in a sufficient amount and for a sufficient period of time to permit capture of the influenza antigens by the dendritic cells. The cells are then transferred into a subject to be vaccinated by standard intravenous transplantation methods.


One or more influenza B viruses may be present in a formulation for prophylactic or therapeutic treatment of influenza. A formulation may comprise one influenza B virus. A formulation may comprise one influenza B virus and one influenza A virus. A formulation may comprise one influenza B virus and two influenza A viruses. A formulation may comprise two influenza B viruses and two influenza A viruses. A formulation may comprise two influenza B viruses. At least one influenza B virus in the formulation may comprise an arginine at amino acid residue 141.


A formulation for prophylactic administration of the influenza viruses, or subunits thereof, may also contain one or more adjuvants for enhancing the immune response to the influenza antigens. Suitable adjuvants include: saponin, mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil or hydrocarbon emulsions, bacille Calmette-Guerin (BCG), Corynebacterium parvum, and the synthetic adjuvants QS-21 and MF59.


The formulation for prophylactic administration of influenza viruses may be performed in conjunction with administration of one or more immunostimulatory molecules. Immunostimulatory molecules include various cytokines, lymphokines and chemokines with immunostimulatory, immunopotentiating, and pro-inflammatory activities, such as interleukins (e.g., IL-1, IL-2, IL-3, IL-4, IL-12, IL-13); growth factors (e.g., granulocyte-macrophage (GM)-colony stimulating factor (CSF)); and other immunostimulatory molecules, such as macrophage inflammatory factor, Flt3 ligand, B7.1; B7.2, etc. The immunostimulatory molecules can be administered in the same formulation as the influenza viruses, or can be administered separately. Either the protein or an expression vector encoding the protein can be administered to produce an immunostimulatory effect.


In another embodiment, the vectors of the invention comprising nucleotide sequences corresponding to influenza genome segments may be employed to introduce heterologous nucleic acids into a host organism or host cell, such as a mammalian cell, e.g., cells derived from a human subject, in combination with a suitable pharmaceutical carrier or excipient as described above. A heterologous nucleic acid may be inserted into a non-essential region of a gene or genome segment. The heterologous polynucleotide sequence can encode a polypeptide or peptide, or an RNA such as an antisense RNA or ribozyme. The heterologous nucleic acid is then introduced into a host or host cells by producing recombinant viruses incorporating the heterologous nucleic, and the viruses are administered as described above.


Alternatively, a vector of the invention including a heterologous nucleic acid can be introduced and expressed in a host cells by co-transfecting the vector into a cell infected with an influenza virus. Optionally, the cells are then returned or delivered to the subject, typically to the site from which they were obtained. In some applications, the cells are grafted onto a tissue, organ, or system site (as described above) of interest, using established cell transfer or grafting procedures. For example, stem cells of the hematopoietic lineage, such as bone marrow, cord blood, or peripheral blood derived hematopoietic stem cells can be delivered to a subject using standard delivery or transfusion techniques.


Alternatively, the viruses comprising a heterologous nucleic acid can be delivered to the cells of a subject in vivo. Such methods may involve the administration of vector particles to a target cell population (e.g., blood cells, skin cells, liver cells, neural (including brain) cells, kidney cells, uterine cells, muscle cells, intestinal cells, cervical cells, vaginal cells, prostate cells, etc., as well as tumor cells derived from a variety of cells, tissues and/or organs. Administration can be either systemic, e.g., by intravenous administration of viral particles, or by delivering the viral particles directly to a site or sites of interest by a variety of methods, including injection (e.g., using a needle or syringe), needleless vaccine delivery, topical administration, or pushing into a tissue, organ or skin site. For example, the viral vector particles can be delivered by inhalation, orally, intravenously, subcutaneously, subdermally, intradermally, intramuscularly, intraperitoneally, intrathecally, by vaginal or rectal administration, or by placing the viral particles within a cavity or other site of the body, e.g., during surgery.


The methods and viruses encompassed by the present invention can be used to therapeutically or prophylactically treat a wide variety of disorders, including genetic and acquired disorders, e.g., as vaccines for infectious diseases, due to viruses, bacteria, and the like.


Kits


To facilitate use of the vectors and influenza viruses encompassed by the invention any of these, and additional components, such as, buffer, cells, culture medium, useful for packaging and infection of influenza viruses for experimental or therapeutic purposes, can be packaged in the form of a kit. The kit may contain, in addition to the above components, additional materials, e.g., instructions for performing the methods of the invention, packaging material, and a container.


Manipulation of Viral Nucleic Acids and Proteins


In the context of the invention, influenza virus nucleic acids and/or proteins are manipulated according to well known molecular biology techniques. Detailed protocols for numerous such procedures, including amplification, cloning, mutagenesis, transformation, and the like, are described in, e.g., in Ausubel et al. Current Protocols in Molecular Biology (supplemented through 2000) John Wiley & Sons, New York (“Ausubel”); Sambrook et al. Molecular Cloning—A Laboratory Manual (2nd Ed.), Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989 (“Sambrook”), and Berger and Kimmel Guide to Molecular Cloning Techniques, Methods in Enzymology volume 152 Academic Press, Inc., San Diego, Calif. (“Berger”).


In addition to the above references, protocols for in vitro amplification techniques, such as the polymerase chain reaction (PCR), the ligase chain reaction (LCR), Qβ-replicase amplification, and other RNA polymerase mediated techniques (e.g., NASBA), useful e.g., for amplifying cDNA probes of the invention, are found in Mullis et al. (1987) U.S. Pat. No. 4,683,202; PCR Protocols A Guide to Methods and Applications (Innis et al. eds) Academic Press Inc. San Diego, Calif. (1990) (“Innis”); Arnheim and Levinson (1990) C&EN 36; The Journal Of NIH Research (1991) 3:81; Kwoh et al. (1989) Proc Natl Acad Sci USA 86, 1173; Guatelli et al. (1990) Proc Natl Acad Sci USA 87:1874; Lomeli et al. (1989) J Clin Chem 35:1826; Landegren et al. (1988) Science 241:1077; Van Brunt (1990) Biotechnology 8:291; Wu and Wallace (1989) Gene 4: 560; Barringer et al. (1990) Gene 89:117, and Sooknanan and Malek (1995) Biotechnology 13:563. Additional methods, useful for cloning nucleic acids in the context of the present invention, include Wallace et al. U.S. Pat. No. 5,426,039. Improved methods of amplifying large nucleic acids by PCR are summarized in Cheng et al. (1994) Nature 369:684 and the references therein.


Certain polynucleotides of the invention, e.g., oligonucleotides can be synthesized utilizing various solid-phase strategies including mononucleotide- and/or trinucleotide-based phosphoramidite coupling chemistry. For example, nucleic acid sequences can be synthesized by the sequential addition of activated monomers and/or trimers to an elongating polynucleotide chain. See e.g., Caruthers, M. H. et al. (1992) Meth Enzymol 211:3.


In lieu of synthesizing the desired sequences, essentially any nucleic acid can be custom ordered from any of a variety of commercial sources, such as The Midland Certified Reagent Company (mcrc@oligos.com), The Great American Gene Company (www.genco.com), ExpressGen, Inc. (www.expressgen.com), Operon Technologies, Inc. (www.operon.com), and many others.


In addition, substitutions of selected amino acid residues in viral polypeptides can be accomplished by, e.g., site directed mutagenesis. For example, viral polypeptides with amino acid substitutions functionally correlated with desirable phenotypic characteristic, e.g., an attenuated phenotype, cold adaptation, temperature sensitivity, can be produced by introducing specific mutations into a viral nucleic acid segment encoding the polypeptide. Methods for site directed mutagenesis are well known in the art, and described, e.g., in Ausubel, Sambrook, and Berger, supra. Numerous kits for performing site directed mutagenesis are commercially available, e.g., the Chameleon Site Directed Mutagenesis Kit (Stratagene, La Jolla), and can be used according to the manufacturers instructions to introduce, e.g., one or more amino acid substitutions described in Table 6 or Table 17, into a genome segment encoding a influenza A or B polypeptide, respectively.


SPECIFIC EMBODIMENTS

1. A method of preparing an HA glycosylated influenza B virus having increased replication in eggs comprising:

    • (a) introducing a mutation resulting in an amino acid substitution at HA position 141 to arginine in an influenza B virus genome; and
    • (b) replicating the mutated influenza virus genome under conditions whereby influenza B virus is produced.


2. The method of embodiment 2 wherein the step of introducing is performed by site-directed mutagenesis.


3. A method of preparing an HA glycosylated influenza B virus having increased replication in eggs comprising:

    • (a) introducing into a population of host cells a plurality of vectors, said vectors comprising nucleotide sequences corresponding to:
      • (i) at least 6 internal genome segments of a first influenza B strain; and
      • (ii) one or more genome segments encoding HA and NA polypeptides of at least a second influenza B strain, wherein the HA polypeptide comprises an arginine at amino acid residue 141;
    • (b) culturing the population of host cells at a temperature that does not exceed 35 degrees; and
    • (c) recovering the influenza virus.


4. The method of embodiment 3 further comprising, prior to step (i):

    • introducing a mutation in one vector of the plurality of vectors,
  • wherein the one vector comprises nucleotide sequences corresponding to the genome segment encoding HA, and
  • wherein the mutation results in the arginine at amino acid residue 141.


5. The method of embodiment 3 or 4 wherein the first influenza B virus has one of the following attributes: temperature sensitivity, attenuation, or cold-adaptation.


6. The method of any one of embodiments 3-5 wherein the first influenza B virus comprises amino acid residues: PB2630 (630R); PA431 (431M); PA497 (497H); NP55 (55A); NP114 (114A); NP410 (410H); NP510 (510T); M1159 (159Q) and M1183 (183V).


7. The method of embodiment 6 further comprising a step of:

    • introducing mutations in vectors of the plurality of vectors,
    • wherein the vectors comprise nucleotide sequences corresponding to the 6 internal genome segments of the first influenza B strains,
    • wherein the mutations result in presence of the amino acid residues PB2630 (630R); PA431 (431M); PA497 (497H); NP55 (55A); NP114 (114A); NP410 (410H); NP510 (510T); M1159 (159Q) and M1183 (183V).


8. The method of any one of embodiments 3-7 wherein the first influenza B virus is strain B/Ann Arbor/1/66.


9. The method of any one of embodiments 3-8 wherein the cells are one of Vero cells, Per.C6 cells, BHK cells, PCK cells, MDCK cells, MDBK cells, 293 cells, or COS cells.


10. The method of any one of embodiments 3-9 wherein the vectors are plasmids.


11. The method of any one of embodiments 3-10 wherein the plurality comprises sets of eight plasmids, wherein each of the eight plasmids comprises a nucleotide sequence corresponding to a different genome segment of the first or the second influenza B strain.


12. The method of any one of embodiments 3-11 wherein each plasmid of the plurality comprises all the nucleotide sequences.


13. The method of any one of embodiments 3-12, wherein the method does not comprise employing a helper virus.


14. The method of any one of embodiments 3-13 wherein the step of introducing is performed by lipid-mediated transfection or electroporation.


15. The method of any one of embodiments 3-14 where the temperature is between 30 and 35 degrees.


16. The method of any one of embodiments 3-15 wherein the temperature is between 32 and 35 degrees.


17. The method of any one of embodiments 3-16 further comprising replicating the recovered influenza virus on eggs;


wherein the influenza virus replicated on eggs retains the HA amino acid residue position 196/197 glycosylation site; and


wherein the influenza virus replicates to at least a peak titer of 7.0 log 10 PFU/ml on the eggs.


18. An influenza B virus prepared by the method of any one of embodiments 1-17.


19. An immunogenic composition comprising the influenza B virus of embodiment 18.


20. A vaccine comprising the influenza B virus of embodiment 19.


21. The vaccine of embodiment 20 which is suitable for intranasal administration.


22. The method of any one of embodiments 3-17 further comprising:


killing the recovered virus.


23. The method of embodiment 1 or 2 further comprising:

    • a) recovering the influenza virus; and
    • b) killing the recovered virus.


24. A live attenuated influenza B virus vaccine comprising the virus produced by the method of any one of embodiments 1-17.


24. A method of treatment of viral infection in a subject comprising:

    • administering to the subject the virus produced by the method of any one of embodiments 1-17 in an amount effective to produce an immunogenic response against the viral infection.


EXAMPLES
Example 1
Construction of pAD3000

The plasmid pHW2000 (Hoffmann et al. (2000) A DNA transfection system for generation of influenza A virus from eight plasmids Proc Natl Acad Sci USA 97:6108-6113) was modified to replace the bovine growth hormone (BGH) polyadenylation signals with a polyadenylation signal sequences derived from Simian virus 40 (SV40).


Sequences derived from SV40 were amplified with Taq MasterMix (Qiagen) using the following oligonucleotides, designated in the 5′ to 3′ direction:









(SEQ ID NO: 1)








polyA.1:
AACAATTGAGATCTCGGTCACCTCAGACATGATAAGATAC



ATTGATGAGT










(SEQ ID NO: 2)








polyA.2:
TATAACTGCAGACTAGTGATATCCTTGTTTATTGCAGCTT



ATAATGGTTA






The plasmid pSV2H is was used as a template. A fragment consistent with the predicted 175 bp product was obtained and cloned into pcDNA3.1, using a Topo TA cloning vector (Invitrogen) according to the manufacturer's directions. The desired 138 bp fragment containing the SV40 polyadenylation signals was excised from the resulting plasmid with EcoRV and BstEII, isolated from an agarose gel, and ligated between the unique PvuII and BstEII sites in pHW2000 using conventional techniques (see, e.g., Ausubel, Berger, Sambrook). The resulting plasmid, pAD3000 (FIG. 1), was sequenced and found to contain the SV40 polyadenylation site in the correct orientation. Nucleotides 295-423 in pAD3000 correspond to nucleotides 2466-2594, respectively, in SV40 strain 777 (AF332562).


Example 2
Eight Plasmid System for Production of MDV-B

Viral RNA from a cold adapted variant of influenza B/Ann Arbor/1/66 (ca/Master Ann Arbor/1/66 P1 Aviron 10/2/97), an exemplary influenza B master donor strain (MDV-B) was extracted from 100 μl of allantoic fluid from infected embryonated eggs using the RNeasy Kit (Qiagen, Valencia, Calif.), and the RNA was eluted into 40 μl H20. RT-PCR of genomic segments was performed using the One Step RT-PCR kit (Qiagen, Valencia, Calif.) according to the protocol provided, using 1 μl of extracted RNA for each reaction. The RT-reaction was performed 50 min at 50° C., followed by 15 min at 94° C. The PCR was performed for 25 cycles at 94° C. for 1 min, 54° C. for 1 min, and 72° C. for 3 min. The P-genes were amplified using segment specific primers with BsmBI-sites that resulted in the generation of two fragments (Table 1).









TABLE 1







RT-PCR primers for amplification of the eight vRNAs of influenza ca B/Ann


Arbor/1/66.










Forward primer
Reverse primer





PB1
Bm-PB1b-1: (SEQ ID NO: 14)
Bm-PB1b-1200R: (SEQ ID NO:15)


[1A]
TATTCGTCTCAGGGAGCAGAAGCGGAGCCTTTAAGATG
TATTCGTCTCGATGCCGTTCCTTCTTCATTGAAGAATGG





PB1
Bm-PB1b-1220: (SEQ ID NO: 16)
Bm-PB1b-2369R: (SEQ ID NO:17)


[1B]
TATTCGTCTCGGCATCTTTGTCGCCTGGGATGATGATG
ATATCGTCTCGTATTAGTAGAAACACGAGCCTT





PB2
Bm-PB2b-1: (SEQ ID NO: 18)
Bm-PB2b-1145R: (SEQ ID NO:19)


[2A]
TATTCGTCTCAGGGAGCAGAAGCGGAGCGTTTTCAAGATG
TATTCGTCTCTCTCATTTTGCTCTTTTTTAATATTCCCC





PB2
Bm-PB2b-1142: (SEQ ID NO: 20)
Bm-PB2b-2396R: (SEQ ID NO:21)


[2B]
TATTCGTCTCATGAGAATGGAAAAACTACTAATAAATTCAGC
ATATCGTCTCGTATTAGTAGAAACACGAGCATT





PA
Bm-Pab-1: (SEQ ID NO: 22)
Bm-PAb-1261R: (SEQ ID NO:23)


[3A]
TATTCGTCTCAGGGAGCAGAAGCGGTGCGTTTGA
TATTCGTCTCCCAGGGCCCTTTTACTTGTCAGAGTGC





PA
Bm-Pab-1283: (SEQ ID NO: 24)
Bm-PAb-2308R: (SEQ ID NO:25)


[3B]
TATTCGTCTCTCCTGGATCTACCAGAAATAGGGCCAGAC
ATATCGTCTCGTATTAGTAGAAACACGTGCATT





HA
MDV-B 5′BsmBI-HA: (SEQ ID NO: 26)
MDV-B 3′BsmBI-HA: (SEQ ID NO:27)



TATTCGTCTCAGGGAGCAGAAGCAGAGCATTTTCTAATATC
ATATCGTCTCGTATTAGTAGTAACAAGAGCATTTTTC





NP
Ba-NPb-1: (SEQ ID NO: 28)
Ba-NPb-1842R: (SEQ ID NO:29)



TATTGGTCTCAGGGAGCAGAAGCACAGCATTTTCTTGT
ATATGGTCTCGTATTAGTAGAAACAACAGCATTTTT





NA
MDV-B 5′BsmBI-NA: (SEQ ID NO: 30)
MDV-B 3′BsmBI-NA: (SEQ ID NO:31)



TATTCGTCTCAGGGAGCAGAAGCAGAGCATCTTCTCAAAAC
ATATCGTCTCGTATTAGTAGTAACAAGAGCATTTTTCAG





M
MDV-B 5′BsmBI-M: (SEQ ID NO: 32)
MDV-B 3′BsmBI-M: (SEQ ID NO:33)



TATTCGTCTCAGGGAGCAGAAGCACGCACTTTCTTAAAATG
ATATCGTCTCGTATTAGTAGAAACAACGCACTTTTTCCAG





NS
MDV-B 5′BsmBI-NS: (SEQ ID NO: 34)
MDV-B 3′BsmBI-NS: (SEQ ID NO:35)



TATTCGTCTCAGGGAGCAGAAGCAGAGGATTTGTTTAGTC
ATATCGTCTCGTATTAGTAGTAACAAGAGGATTTTTAT





The sequences complementary to the influenza sequences are shown in bold. The 5′-ends have recognition sequences for the restriction endonucleases BsmBI (Bm) or BsaI (Ba).







Cloning of Plasmids


PCR fragments were isolated, digested with BsmBI (or BsaI for NP) and inserted into pAD3000 (a derivative of pHW2000 which allows the transcription of negative sense vRNA and positive mRNA) at the BsmBI site as described above. Two to four each of the resultant plasmids were sequenced and compared to the consensus sequence of MDV-B based on sequencing the RT-PCR fragments directly. Plasmids which had nucleotide substitutions resulting in amino acid changes different from the consensus sequence were “repaired” either by cloning of plasmids or by utilizing the Quikchange kit (Stratagene, La Jolla, Calif.). The resultant B/Ann Arbor/1/66 plasmids were designated pAB 121-PB 1, pAB 122-PB2, pAB 123-PA, pAB 124-HA, pAB 125-NP, pAB126-NA, pAB127-M, and pAB128-NS. Using this bi-directional transcription system all viral RNAs and proteins are produced intracellularly, resulting in the generation of infectious influenza B viruses (FIG. 2).


It is noteworthy that pAB121-PB1 and pAB124-HA had 2 and pAB128-NS had 1 silent nucleotide substitution compared to the consensus sequence (Table 2). These nucleotide changes do not result in amino acid alterations, and are not anticipated to affect viral growth and rescue. These silent substitutions have been retained to facilitate genotyping of the recombinant viruses.









TABLE 2







Plasmid Set Representing The Eight Segments


of B/Ann Arbor/1/66 (MDV-B)










Seg.
plasmids
nucleotides
protein





PB1
PAB121-PB1
A924 > G924; C1701 > T1701
silent


PB2
PAB122-PB2
consensus



PA
PAB123-PA
consensus



HA
PAB124-HA
T150 > C150; T153 > C153
silent


NP
PAB125-NP
consensus



NA
PAB126-NA
consensus



M
PAB127-M
consensus



NS
PAB128-NS
A416 > G416
NS1: silent









For construction of the plasmids with nucleotide substitution in PA, NP, and M1 genes the plasmids pAB123-PA, pAB125-NP, pAB127-M were used as templates. Nucleotides were changed by Quikchange kit (Stratagene, La Jolla, Calif.). Alternatively, two fragments were amplified by PCR using primers which contained the desired mutations, digested with BsmBI and inserted into pAD3000-BsmBI in a three fragment ligation reaction. The generated plasmids were sequenced to ensure that the cDNA did not contain unwanted mutations.


The sequence of template DNA was determined by using Rhodamine or dRhodamine dye-terminator cycle sequencing ready reaction kits with AmpliTaq® DNA polymerase FS (Perkin-Elmer Applied Biosystems, Inc, Foster City, Calif.). Samples were separated by electrophoresis and analyzed on PE/ABI model 373, model 373 Stretch, or model 377 DNA sequencers.


In a separate experiment, viral RNA from influenza B/Yamanshi/166/98 was amplified and cloned into pAD3000 as described above with respect to the MDV-B strain, with the exception that amplification was performed for 25 cycles at 94° C. for 30 seconds, 54° C. for 30 seconds and 72° C. for 3 minutes. Identical primers were used for amplification of the B/Yamanashi/166/98 strain segments, with the substitution of the following primers for amplification of the NP and NA segments: MDV-B 5′BsmBI-NP: TATTCGTCTCAGGGAGCAGAAGCACAGCATTTTCTTGTG (SEQ ID NO:36) and MDV-B 3′BsmBI-NP:ATATCGTCTCGTATTAGTAGAAACAACAGCATTTTTTAC (SEQ ID NO:37) and Bm-NAb-1: TATTCGTCTCAGGGAGCAGAAGCAGAGCA (SEQ ID NO:38) and Bm-NAb-1557R:ATATCGTCTCGTATTAGTAGTAACAAGAGCA TTTT (SEQ ID NO:39), respectively. The B/Yamanashi/166/98 plasmids were designated pAB251-PB1, pAB252-PB2, pAB253-PA, pAB254-HA, pAB255-NP, pAB256-NA, pAB257-M, and pAB258-NS. Three silent nucleotide differences were identified in PA facilitating genotyping of recombinant and reassortant B/Yamanashi/166/98 virus.


Example 3
Generation of Infectious Recombinant Influenza B and Reassorted Influenza Virus

Infectious recombinant influenza B viruses were produced by co-culturing 293T or COS-7 cells (primate cells with high transfection efficiency and poll activity) with MDCK cells (permissive for influenza virus). 293T cells were maintained in OptiMEM I-AB medium containing 5% FBS cells, COS-7 cells were maintained in DMEM I-AB medium containing 10% FBS. MDCK cells were maintained in 1×MEM, 10% FBS with the addition of antibiotic and antimycotic agents. Prior to transfection with the viral genome vectors, the cells were washed once with 5 ml PBS or medium without FBS. Ten ml trypsin-EDTA was added to confluent cells in a 75 cm2 flask (MDCK cells were incubated for 20-45 min, 293T cells were incubated for 1 min) The cells were centrifuged, and resuspended in 10 ml OptiMEM I-AB. One ml of each suspended cell line was then diluted into 18 ml OptiMEM I-AB, and mixed. The cells were then aliquoted into a 6 well plate at 3 ml/well. After 6-24 hours, 1 ng of each plasmid was mixed in an 1.5 ml Eppendorf tube with OptiMEM I-AB to the plasmids (x μl plasmids+x μl OptiMEM I-AB+x μl TransIT-LT1=200 μ1); 2 μ1 TransIT-LT1 per μg of plasmid DNA. The mixture was incubated at room temperature for 45 min. Then 800 μl of OptiMEM I-AB was added. The medium was removed from the cells, and the transfection mixture was added to the cells (t=0) at 33° C. for 6-15 hours. The transfection mixture was slowly removed from the cells, and 1 ml of OptiMEM I-AB was added, and the cells were incubated at 33° C. for 24 hours. Forty-eight hours following transfection, 1 ml of OptiMEM I-AB containing 1 μg/ml TPCK-trypsin was added to the cells. At 96 hours post-transfection, 1 ml of OptiMEM I-AB containing 1 μg/ml TPCK-trypsin was added to the cells.


Between 4 days and 7 days following transfection 1 ml of the cell culture supernatant was withdrawn and monitored by HA or plaque assay. Briefly, 1 ml of supernatant was aliquoted into an Eppendorf tube and centrifuge at 5000 rpm for 5 min. Nine hundred μl of supernatant was transferred to a new tube, and serial dilutions were performed at 500 μl/well to MDCK cells (e.g., in 12 well plates). The supernatant was incubated with the cells for 1 hour then removed, and replaced with infection medium (1×MEM) containing 1 μg/ml of TPCK-trypsin. HA assay or plaque assays were then performed. For example, for the plaque assays supernatants were titrated on MDCK cells which were incubated with an 0.8% agarose overlay for three days at 33° C. For infection of eggs the supernatant of transfected cells were harvested six or seven days after transfection, 100 μl of the virus dilutions in Opti-MEM I were injected into 11 days old embryonated chicken eggs at 33° C. The titer was determined three days after inoculation by TCID50 assay in MDCK cells.


To generate MDV-B, either co-cultured 293T-MDCK or COS-7-MDCK cells were transfected with 1 μg of each plasmid. When examined at 5 to 7 days post-transfection the co-cultured MDCK cells showed cytopathic effects (CPE), indicating the generation of infectious MDV-B virus from cloned cDNA. No CPE was observed in cells transfected with seven plasmids (Table 3). To determine the efficiency of the DNA transfection system for virus generation, supernatants of cells were titrated seven days after transfection on MDCK cells and the virus titer was determined by plaque assay. The virus titer of the supernatant of co-cultured 293T-MDCK was 5.0×106 pfu/ml and 7.6×106 pfu/ml in COST-MDCK cells.









TABLE 3







Generation of infectious Influenza-B virus from eight plasmids









segment












1
2
3
4















PB1
pAB121-PB1

PAB121-PB1



PB2
pAB122-PB2
pAB122-PB2
PAB122-PB2
pAB122-PB2


PA
pAB123-PA
pAB123-PA
pAB123-PA
pAB123-PA


HA
pAB124-HA
pAB124-HA
pAB124-HA
pAB124-HA


NP
pAB125-NP
pAB125-NP
pAB125-NP
pAB125-NP


NA
pAB126-NA
pAB126-NA
pAB126-NA
pAB126-NA


M
pAB127-M
pAB127-M
pAB127-M
pAB127-M


NS
pAB128-NS
pAB128-NS
pAB128-NS
pAB128-NS













co-cultured 293T-MDCK cells
co-cultured COS-7-MDCK cells











CPE
+

+






pfu/
5.0 × 106
0
7.6 × 106
0


ml









Transiently co-cultured 293T-MDCK (1, 2) or co-cultured COST-MDCK cells (3, 4) were transfected with seven or eight plasmids. Cytopathic effect (CPE) was monitored seven days after transfection in the co-cultured MDCK cells. Seven days after transfection the supernatants of transfected cells were titrated on MDCK cells. The data of pfu/ml represent the average of multiple, (e.g., three or four) transfection experiments.


Comparable results were obtained in transfection experiments utilizing the B/Yamanashi/166/98 plasmid vectors. These results show that the transfection system allows the reproducible de novo generation of influenza B virus from eight plasmids.


Genotyping of Recombinant Influenza B


After a subsequent passage on MDCK cells, RT-PCR of the supernatant of infected cells was used to confirm the authenticity of the generated virus. RT-PCR was performed with segment specific primers for all eight segments (Table 1). As shown in FIG. 3A, PCR products were generated for all segments. Direct sequencing of the PCR products of the PB1, HA, and NS segments revealed that the four nucleotides analyzed were the same as found in the plasmid pAB121-PB1, pAB124-HA, and pAB128-NS. These results confirmed that the generated virus was generated from the designed plasmids and exclude (in addition to the negative controls) any possible laboratory contamination with the parent virus (FIG. 3B).


Similarly, following transfection with the B/Yamanashi/166/98 plasmid vectors, virus was recovered and the region encompassing nucleotides 1280-1290 of the PA segment were amplified. Sequencing confirmed that the recovered virus corresponded to the plasmid-derived recombinant B/Yamanashi/166/98 (FIGS. 3C and D).


Phenotyping of rMDV-B


The MDV-B virus shows two characteristic phenotypes: temperature sensitivity (ts) and cold adaptation (ca). By definition a 2 log(or higher) difference in virus titer at 37° C. compared to 33° C. defines ts, ca is defined by less than 2 log difference in virus growth at 25° C. compared to 33° C. Primary chicken kidney (PCK) cells were infected with the parent virus MDV-B and with the transfected virus derived from plasmids to determine the viral growth at three temperatures.


For plaque assay confluent MDCK cells (ECACC) in six well plates were used. Virus dilutions were incubated for 30-60 min. at 33° C. The cells were overlayed with an 0.8% agarose overlay. Infected cells were incubated at 33° C. or 37° C. Three days after infection the cells were stained with 0.1% crystal violet solution and the number of plaques determined.


The ca-ts phenotype assay was performed by TCID50 titration of the virus samples at 25, 33, and 37° C. This assay format measures the TCID50 titer by examining the cytopathic effect (CPE) of influenza virus on primary chick kidney cell monolayers in 96-well cell culture plates at different temperatures (25° C., 33° C., 37° C.). This assay is not dependent on the plaque morphology, which varies with temperature and virus strains; instead it is dependent solely on the ability of influenza virus to replicate and cause CPE. Primary chicken kidney (PCK) cell suspension, prepared by trypsinization of the primary tissue, were suspended in MEM (Earl's) medium containing 5% FCS. PCK cells were seeded in 96 well cell culture plates for 48 hours in order to prepare monolayer with >90% confluency. After 48 hrs, the PCK cell monolayer were washed for one hour with serum free MEM medium containing 5 mM L-Glutamine, antibiotics, non-essential amino acid, referred as Phenotype Assay Medium (PAM). Serial ten-fold dilution of the virus samples were prepared in 96 well blocks containing PAM. The diluted virus samples were then plated onto the washed PCK monolayer in the 96 well plates. At each dilution of the virus sample, replicates of six wells were used for infection with the diluted virus. Un-infected cells as cell control were included as replicate of 6 wells for each sample. Each virus sample was titered in 2-4 replicates. Phenotype control virus with pre-determined titers at 25° C., 33° C., and 37° C. is included in each assay. In order to determine the ts phenotype of the virus samples, the plates were incubated for 6 days at 33° C. and 37° C. in 5% CO2 cell culture incubators. For ca-phenotype characterization the plates were incubated at 2° C. for 10 days. The virus titer was calculated by the Karber Method and reported as Log10 Mean (n=4) TCID50 Titer/ml±Standard Deviation. The standard deviations of the virus titers presented in FIG. 1-3 ranged from 0.1 to 0.3. The difference in virus titer at 33° C. and 37° C. were used to determine the ts phenotype and difference in titer at 25° C. and 33° C. of the virus were used to determine the ca phenotype.


The plasmid derived recombinant MDV-B (recMDV-B) virus expressed the two characteristic phenotypes in cell culture, ca and ts, as expected. The ca phenotype, efficient replication at 25° C., is functionally measured as a differential in titer between 25° C. and 33° C. of less than or equal to 2 log 10 when assayed on PCK cells. Both the parental MDV-B and recMDV-B expressed ca; the difference between 25° C. and 33° C. was 0.3 and 0.4 log 10, respectively (Table 4). The ts phenotype is also measured by observing the titers at two different temperatures on PCK cells; for this phenotype, however, the titer at 37° C. should be less than the titer at 33° C. by 2 log 10 or more. The difference between 33° C. and 37° C. for the parental MDV-B and recMDV-B was 3.4 and 3.7 log 10, respectively (Table 4). Thus, the recombinant plasmid-derived MDV-B virus expressed both the ca and ts phenotypes.


The recombinant virus had a titer of 7.0 log10 TCID50/ml at 33° C. and 3.3 TCID50/ml at 37° C. and 8.8 log10 TCID50/ml at 25° C. (Table 4). Thus, the recombinant virus derived from transfection with the eight influenza MDV-B genome segment plasmids has both the ca and ts phenotype.









TABLE 4







Phenotype assay for MDV-B and


rMDV-B generated from plasmids










Temperature (0 C.)












25
33
37









Virus
Log10 TCID50/ml (Mean + SD)
Phenotype














ca B/Ann Arbor/
8.8 + 0.3
8.5 + 0.05
5.1 + 0.1
ca, ts


01/66 (MDV-B)


RecMDV-B
7.4 + 0.3
7.0 + 0.13
3.3 + 0.12
ca, ts


Rec53-MDV-B
5.9 + 0.1
5.7 + 0.0
5.3 + 0.1
ca, non-ts





Primary chicken kidney cells were infected with the parent virus MDV-B and the plasmid-derived recombinant virus (recMDV-B). The virus titer was determined at three different temperatures.






Example 7
Production of Reassortant B/Yamanashi/166/98 Virus

The HA and NA segments of several different strains representing the major lineages of influenza B were amplified and cloned into pAD3000, essentially as described above. The primers were optimized for simultaneous RT-PCR amplification of the HA and NA segments. Comparison of the terminal regions of the vRNA representing the non coding region of segment 4 (HA) and segment 6 (NB/NA) revealed that the 20 terminal nucleotides at the 5′ end and 15 nucleotides at the 3′ end were identical between the HA and NA genes of influenza B viruses. A primer pair for RT-PCR (italicized sequences are influenza B virus specific) Bm-NAb-1: TAT TCG TCT CAG GGA GCA GAA GCA GAG CA (SEQ ID NO:38); Bm-NAb-1557R: ATA TCG TCT CGT ATT AGT AGT AAC AAG AGC ATT TT (SEQ ID NO:39) was synthesized and used to simultaneously amplify the HA and NA genes from various influenza B strains (FIG. 6). The HA and NA PCR-fragments of B/Victoria/504/2000, B/Hawaii/10/2001, and B/Hong Kong/330/2001 were isolated, digested with BsmBI and inserted into pAD3000. These results demonstrated the applicability of these primers for the efficient generation of plasmids containing the influenza B HA and NA genes from several different wild type viruses representing the major lineages of influenza B. The RT-PCR products can be used for sequencing and/or cloning into the expression plasmids.


In order to demonstrate the utility of B/Yamanashi/166/98 (a B/Yamagata/16/88-like virus) to efficiently express antigens from various influenza B lineages, reassortants containing PB1, PB2, PA, NP, M, NS from B/Yamanashi/166/98 and the HA and NA from strains representing both the Victoria and Yamagata lineages (6+2 reassortants) were generated. Transiently cocultured COS7-MDCK cells were cotransfected with six plasmids representing B/Yamanashi/166/98 and two plasmids containing the cDNA of the HA and NA segments of two strains from the B/Victoria/2/87 lineage, B/Hong Kong/330/2001 and B/Hawaii/10/2001, and one strain from the B/Yamagata/16/88 lineage, B/Victoria/504/2000, according to the methods described above. Six to seven days after transfection the supernatants were titrated on fresh MDCK cells. All three 6+2 reassortant viruses had titers between 4-9×106 pfu/ml (Table 5). These data demonstrated that the six internal genes of B/Yamanashi/166/98 could efficiently form infectious virus with HA and NA gene segments from both influenza B lineages.


Supernatants of cocultured COST-MDCK cells were titrated six or seven days after transfection and the viral titer determined by plaque assays on MDCK cells.









TABLE 5







Plasmid set used for the generation of B/Yamanashi/166/98 and 6 + 2 reassortants.







segment















1

pAB251-PB1
pAB251-PB1
pAB251-PB1
pAB251-PB1


2
pAB252-PB2
pAB252-PB2
pAB252-PB2
pAB252-PB2
pAB252-PB2


3
pAB253-PA
pAB253-PA
pAB253-PA
pAB253-PA
pAB253-PA


4
pAB254-HA
pAB254-HA
pAB281-HA
pAB285-HA
pAB287-HA


5
pAB255-NP
pAB255-NP
pAB255-NP
pAB255-NP
pAB255-NP


6
pAB256-NA
pAB256-NA
pAB291-NA
pAB295-NA
pAB297-NA


7
pAB257-M
pAB257-M
pAB257-M
pAB257-M
pAB257-M


8
pAB258-NA
pAB258-NA
pAB258-NA
pAB258-NA
pAB258-NA












Recombinant virus












8
6 + 2
6 + 2
6 + 2














B/Yamanashi/
B/Victoria/
B/Hawaii/
B/Hong Kong/




166/98
504/2000
10/2001
330/2001





pfu/mla
0
4 × 106
9 × 106
6 × 106
7 × 106









Relatively high titers are obtained by replication of wild type B/Yamanashi/166/98 in eggs. Experiments were performed to determine whether this property was an inherent phenotype of the six “internal” genes of this virus. To evaluate this property, the yield of wild type B/Victoria/504/2000, which replicated only moderately in eggs, was compared to the yield of the 6+2 reassortant expressing the B/Victoria/504/2000 HA and NA. These viruses in addition to wild type and recombinant B/Yamanashi/166/98 were each inoculated into 3 or 4 embryonated chicken eggs, at either 100 or 1000 pfu. Three days following infection, the allantoic fluids were harvested from the eggs and the TCID50 titers determined on MDCK cells. The 6+2 reassortants produced similar quantities of virus in the allantoic fluid to the wt and recombinant B/Yamanashi/166/98 strain (FIG. 7). The difference in titer between B/Victoria/504/2000 and the 6+2 recombinant was approximately 1.6 log10 TCID50 (0.7-2.5 log10 TCID50/mL, 95% CI). The difference between B/Victoria/504/2000 and the 6+2 recombinant were confirmed on three separate experiments (P<0.001). These results demonstrated that the egg growth properties of B/Yamanashi/166/98 could be conferred to HA and NA antigens that are normally expressed from strains that replicated poorly in eggs.


Example 8
Molecular Basis for Attenuation of Ca B/Ann Arbor/1/66

The MDV-B virus (ca B/Ann Arbor/1/66) is attenuated in humans, shows an attenuated phenotype in ferrets and shows a cold adapted and temperature sensitive phenotype in cell culture. The deduced amino acid sequences of the internal genes of MDV-B were compared with sequences in the Los Alamos influenza database (on the world wide web at: flu.lanl.gov) using the BLAST search algorithm. Eight amino acids unique to MDV-B, and not present in any other strain were identified (Table 6). Genome segments encoding PB1, BM2, NS1, and NS2 show no unique substituted residues. The PA and M1 proteins each have two, and the NP protein has four unique substituted amino acids (Table 6). One substituted amino acid is found in PB2 at position 630 (an additional strain B/Harbin/7/94 (AF170572) also has an arginine residue at position 630).


These results suggested that the gene segments PB2, PA, NP and M1 may be involved in the attenuated phenotype of MDV-B. In a manner analogous to that described above for MDV-A, the eight plasmid system can be utilized to generate recombinant and reassortant (single and/or double, i.e., 7:1; 6:2 reassortants) in a helper independent manner simply by co-transfection of the relevant plasmids into cultured cells as described above with respect to MDV-A. For example, the 6 internal genes from B/Lee/40 can be used in conjunction with HA and NA segments derived from MDV-B to generate 6+2 reassortants.









TABLE 6







Unique substituted amino acids of B/Ann


Arbor/1/66















ca B/Ann
Aligned
Number





Arbor/
sequences (wild
of





1/66
type viruses)
align-

















amino

amino

ed seq-



Nr.
pos.
acid
codon
acid
codon
uences





PB1
0





23





PB2
1
630
Arg630
AGA
Ser630
AGC
23





PA
2
431
Met431
ATG
Va1431
GTG
23







497
His497
CAT
Tyr497
TAT






NP
4
55
Ala55
GCC
Thr55
ACC
26







114
A1a114
GCG
Va1114
GTG








410
His410
CAT
Pro410
CCT,









CCC








509
Thr509
GAC
A1a509
GGC






M1
2
159
G1n159
CAA
His159
CAT
24







183
Va1183
GTG
M183
ATG






BM2
0





24





NS1
0





80





NS2
0





80





The deduced amino acid sequence of eight proteins of ca B/Ann Arbor was used in a BLAST search Amino acid position which were different between MDV-B and the aligned sequences are shown. The nucleotides in the codons that are underlined represent the substituted positions.






In order to determine whether the 8 unique amino acid differences had any impact on the characteristic MDV-B phenotypes, a recombinant virus was constructed in which all eight nucleotide positions encoded the amino acid reflecting the wt influenza genetic complement. A set of plasmids was constructed in which the eight residues of the PA, NP, and M1 genes were changed by site directed mutagenesis to reflect the wild type amino acids (as indicated in Table 6). A recombinant with all eight changes, designated rec53-MDV-B, was generated by cotransfection of the constructed plasmids onto cocultured COST-MDCK cells. The coculturing of MDCK cells and growth at 33° C. ensured that the supernatant contained high virus titers six to seven days after transfection. The supernatants of the transfected cells were titrated and the titer determined on MDCK cells by plaque assay and PCK cells at 33° C. and 37° C.


As shown in FIG. 8, in two different independent experiments, recMDV-B expressed the ts-phenotype in both MDCK cells and PCK cells. The triple reassortant virus rec53-MDV-B designed harboring all eight amino acid changes expressed the non-ts-phenotype, the difference in titer between 33° C. and 37° C. was only 0.7 log10 in PCK cells. This titer was less than the required 2 log10 difference characteristic of the ts definition and significantly lower than the ˜3 log10 difference observed with recMDV-B. These results show that the alteration of the eight amino acids within PA, NP, and M1 proteins was sufficient to generate a non-ts, wild type-like virus with both homologous and heterologous glycoproteins.


The contribution of each gene segment to the ts phenotype was then determined Plasmid derived recombinants harboring either the PA, NP, or M gene segment with the wild-type amino acid complement were generated by the DNA cotransfection technique. All single gene recombinants exhibited growth restriction at 37° C. in MDCK cells and in PCK cells (FIG. 9), indicating that changes in no one gene segment were capable of reverting the ts phenotype. In addition, recombinant viruses that carried both the NP and M or PA and M gene segments together also retained the ts-phenotype. In contrast, recombinant viruses that harbored both the PA and NP gene segments had a difference in titer between 37° C. and 33° C. of 2.0 log10 or less, similar to the rec53-MDV-B. These results show that the NP and PA genes have a major contribution to the ts-phenotype.


To determine whether all of the four amino acids in the NP protein and two in the PA protein contribute to non-ts, triple gene and double-gene recombinants with altered NP and PA genes were generated (FIG. 10). The substitution of two amino acids in the NP protein, A114→V114 and H410→P410 resulted in non-ts phenotype. Viruses with single substitution H410→P410 in the nucleoprotein showed non-ts phenotype in MDCK and PCK. On the other hand, the single substitution A55→T55 showed a ts-phenotype, as did the single substitution at position 509. These results indicate that amino acid residues V114 and P410 in NP are involved in efficient growth at 37° C. (FIG. 11A). A similar strategy was employed to dissect the contribution of the two amino acids in the PA gene. A set of recombinants was constructed, each harboring an NP gene segment with four wild-type consensus amino acids and a PA gene with only one of the two consensus wild type amino acids. Substitution of H497→Y497 remained ts (FIG. 11B), demonstrating that this locus had little impact on expression of the phenotype. In contrast, substitution of M431 with V431 resulted in reversion of the ts phenotype. These results show that amino acids A114 and H410 in NP and M431 in PA are the major determinants for temperature sensitivity of MDV-B.


Based on prior evidence, a ts-phenotype and an attenuated phenotype are highly correlated. It is well established that ca B/Ann Arbor/1/66 virus is not detectable in lung tissue of infected ferrets, whereas non attenuated influenza B viruses are detectable in lungs after intranasal infection. To determine whether identical mutation underlie the ts and att phenotypes, the following studies were performed.


Recombinant viruses obtained after transfection were passaged in embryonated chicken eggs to produce a virus stock. Nine week old ferrets were inoculated intranasally with 0.5 ml per nostril of viruses with titers of 5.5, 6.0 or 7.0 login pfu/ml. Three days after infection ferrets were sacrificed and their lungs and turbinates were examined as described previously.


Ferrets (four animals in each group) were infected intranasally with recMDV-B or rec53-MDV-B. Three days after infection virus nasal turbinates and lung tissue were harvested and the existence of virus was tested. No virus was detected in lung tissues of ferrets infected with 7.0 log10 pfu recMDV-B. From the four animals infected with rec53-MDV-B virus with 7.0 log10 pfu in three animals virus was detected in lung tissue (one animal in this group for unknown reasons). In two out of four lung tissues of ferrets infected with rec53-MDV-B at a lower dose (5.5 log pfu/ml) virus could be isolated from lung tissue. Thus, the change of the eight unique amino acids in PA, NP, and M1 protein into wild type residues were sufficient to convert a att phenotype into a non-att phenotype.


Since the data in cell culture showed that PA and NP are main contributors to the ts-phenotype, in a second experiment, ferrets were infected with rec53-MDV-B (PA,NP,M), rec62-MDV-B (PA), NP rec71-MDV-B (NP) with 6 log pfu. Two out of four animals infected with rec53-MDV-B had virus in the lung. None of the lung tissues of ferrets infected with single and double reassortant viruses had detectable levels of virus. Thus, in addition to the amino acids in the PA and NP proteins, the M1 protein is important for the att phenotype. Virus with wt PA and NP did not replicate in ferret lung, indicating that a subset of the mutations involved in attenuation are involved in the ts phenotype.


Thus, the ts and att phenotypes of B/Ann Arbor/1/66 are determined by at most three genes. The conversion of eight amino acids in the PA, NP, and M1 protein into wild type residues resulted in a recombinant virus that replicated efficiently at 37° C.


Similarly, a 6+2 recombinant virus representing the six internal genes of MDV-B with the HA and NA segments from B/HongKong/330/01 showed a ts-phenotype and the triple recombinant was non-ts.


Our results using the MDV-B backbone indicated that six amino acids were sufficient to convert a ts/att phenotype into a non-ts/non-att phenotype. Therefore, we were interested in determining whether the introduction of those six ‘attenuation’ residues would transfer these biological properties to a heterologous wildtype, non attenuated influenza B virus, such as B/Yamanashi/166/98.


Recombinant wildtype B/Yamanashi/166/98 (recYam) (7) and a recombinant virus (recti-Yam): with six amino acid changes PA (V431→M431, H497→Y497), NP (V114→A114, P410→H410), and M1 (H159→Q159, M183→V183) were produced. RecYam showed a 0.17 log 10 titer reduction in titer at 37° C. compared to 33° C., whereas rec6Yam was clearly ts, the difference in viral titer between 37° C. and 33° C. was 4.6 log 10. Virus was efficiently recovered from ferrets infected with recYam, as expected for a typical wildtype influenza B virus. When rec6Yam was inoculated into ferrets, no virus was detected in the lung tissues (Table 7). Thus, the transfer of the ts/att loci from MDV-B are sufficient to transfer the ts- and att-phenotypes to a divergent virus.









TABLE 7







Attenuation studies in ferrets


















Nasal



Recombinant

Ts-

Dose
turbinatesb
Lung tissue


virus
wt componentsa
phenotype
ferrets
[log10 pfu]
[log10 pfu/g]
[log10 EID50/g]c
















rMDV-B
none
ts
4
6.0
4.01
<1.5


rec53-B
NP, PA, M
Non-ts
4
6.0
4.65
3.81


rec62-B
NP, PA
Non-ts
4
6.0
4.69
<1.5


rec71NP-B
NP
ts
4
6.0
4.13
<1.5


rec71M-B
M
ts
4
6.0
4.17
<1.5


RecYam

Non-ts
4
6.0
4.92
3.31


rec6Yam

ts
4
6.0
4.02
<1.5






aRecombinant viruses with MDV-B backbone that differed in wildtype amino acids were used to infected ferrets intranassally. RecYam is recombinant B/Yamanashi/166/98 and Rec6Yam represents a virus that has six ‘MDV-B-attenuation’ amino acid changes in NP, PA, and M1 with a B/Yamanashi backbone.




bThree days after infection the virus titer of the nasal turbinates and lung tissue was determined, the average titer of four infected ferrets is shown.




c<1.5 indicates that no virus was detected.







Accordingly, artificially engineered variants of influenza B strain virus having one or more of these amino acid substitutions exhibit the ts and att phenotypes and are suitable for use, e.g., as master donor strain viruses, in the production of attenuated live influenza virus vaccines.


Example 9
Determination of the Loci Controlling the Cold-Adapted Phenotype of B/Ann Arbor/1/66 Influenza Virus

The cold adapted (ca) B/Ann Arbor/1/66 is the master donor virus (MDV-B) for the live attenuated influenza B Flumist® vaccines. The 6:2 influenza B vaccines carrying the six internal genes derived from ca B/Ann Arbor/1/66 and the HA and NA surface glycoproteins from the circulating wild-type strains are characterized by the cold-adapted (c a), temperature-sensitive (ts) and attenuated (au) phenotypes. Sequence analysis revealed that MDV-B contains nine amino acids in the PB2, PA, NP and M1 proteins that are not found in wild-type influenza B strains. We have determined that three amino acids in the PA(M431V) and NP(A114V, H410P) determined the ts phenotype and, in addition to these three is loci, two amino acids in the M1 (Q159H, V183M) conferred the att phenotype.


To understand the molecular basis of the ca phenotype, the plasmid-based reverse genetics system was used to evaluate the contribution of these nine MDV-B specific amino acids to the ca phenotype. Recombinant MDV-B replicated efficiently at 25° C. and 33° C. in the chicken embryonic kidney (CEK) cells. In contrast, recombinant wild type B/Ann Arbor/1/66, containing the nine wild type amino acids, replicated inefficiently at 25° C. It was determined that a total of five wild type amino acids, one in PB2 (R630S), one in PA(M431V) and three in NP(A114V, H410P, T509A), were required for to completely revert the MDV-B ca phenotype. In addition, replacing two amino acids in the M1 protein (Q159H, V183M) of MDV-B or 6:2 vaccine strains with the wild-type amino acids significantly increased virus replication at 33° C. but not at 25° C. in CEK cells; the V183M change had a larger impact on the change.


Example 10
Rescue of Influenza from Eight Plasmids by Electroporation of Vero Cells

Recombinant influenza viruses may also be rescued from Vero cells using electroporation. These methods are suitable for the production of both influenza A and influenza B strain viruses, and permit the recovery of, e.g., cold adapted, temperature sensitive, attenuated virus from Vero cells grown under serum free conditions facilitating the preparation of live attenuated vaccine suitable for administration in, e.g., intranasal vaccine formulations. In addition to its broad applicability across virus strains, electroporation requires no additional reagents other than growth medium for the cell substrate and thus has less potential for undesired contaminants. In particular, this method is effective for generating recombinant and reassortant virus using Vero cells adapted to growth under serum free condition, such as Vero cell isolates qualified as pathogen free and suitable for vaccine production. This characteristic supports the choice of electroporation as an appropriate method for commercial introduction of DNA into cell substrates.


Electroporation was compared to a variety of methods for introduction of DNA into Vero cells, including transfection using numerous lipid based reagents, calcium phosphate precipitation and cell microinjection. Although some success was obtained using lipid based reagents for the rescue of influenza A, only electroporation was demonstrated to rescue influenza B as well as influenza A from Vero cells.


One day prior to electroporation, 90-100% confluent Vero cells were split, and seeded at a density of 9×106 cells per T225 flask in MEM supplemented with pen/strep, L-glutamine, nonessential amino acids and 10% FBS (MEM, 10% FBS). The following day, the cells were trypsinized and resuspended in 50 ml phosphate buffered saline (PBS) per T225 flask. The cells are then pelleted and resuspended in 0.5 ml OptiMEM I per T225 flask. Optionally, customized OptiMEM medium containing no human or animal-derived components can be employed. Following determination of cell density, e.g., by counting a 1:40 dilution in a hemocytometer, 5×106 cells were added to a 0.4 cm electroporation cuvette in a final volume of 400 μl OptiMEM I. Twenty μg DNA consisting of an equimolar mixture of eight plasmids incorporating either the MDV-A or MDV-B genome in a volume of no more than 25 μl was then added to the cells in the cuvette. The cells were mixed gently by tapping and electroporated at 300 volts, 950 microFarads in a BioRad Gene Pulser II with Capacitance Extender Plus connected (BioRad, Hercules, Calif.). The time constant should be in the range of 28-33 msec.


The contents of the cuvette were mixed gently by tapping and 1-2 min after electroporation, 0.7 ml MEM, 10% FBS was added with a 1 ml pipet. The cells were again mixed gently by pipetting up and down a few times and then split between two wells of a 6 well dish containing 2 ml per well MEM, 10% FBS. The cuvette was then washed with 1 ml MEM, 10% FBS and split between the two wells for a final volume of about 3.5 ml per well.


In alternative experiments, Vero cells adapted to serum free growth conditions, e.g., in OptiPro (SFM) (Invitrogen, Carlsbad, Calif.) were electroporated as described above except that following electroporation in OptiMEM I, the cells were diluted in OptiPro (SFM) in which they were subsequently cultured for rescue of virus.


The electroporated cells were then grown under conditions appropriate for replication and recovery of the introduced virus, i.e., at 33° C. for the cold adapted Master Donor Strains. The following day (e.g., approximately 19 hours after electroporation), the medium was removed, and the cells were washed with 3 ml per well OptiMEM I or OptiPro (SFM). One ml per well OptiMEM I or OptiPro (SFM) containing pen/strep was added to each well, and the supernatants were collected daily by replacing the media. Supernatants were stored at −80° C. in SPG. Peak virus production was typically observed between 2 and 3 days following electroporation.









TABLE 8







Results of 8 Plasmid Rescue of MDV strains on Different


Cell Types and by Different Transfection Methods













Result





(Infectious


Substrate
Method
No of Test
Virus Recovered)










MDV-B










COS-7/MDCK
Lipo
3
positive


COS-7/MDCK
CaPO4
2
positive


MRC-5
Lipo
5
negative


MRC-5
CaPO4
3
negative


MRC-5
Electroporation
2
negative


WI-38
Lipo
2
negative


WI-38
Electroporation
4
negative


WI-38
Microinjection
1
negative


LF1043
Lipo
1
negative


LF1043
CaPO4
2
negative


Vero
Lipo
7
negative


Vero
CaPO4
2
negative


Vero/MDCK
Lipo
1
negative


Vero (serum)
Electroporation
5
positive (5/5)


Vero (serum free)
Electroporation
4
positive (4/4)







MDV-A










Vero (serum)
Electroporation
3
positive (3/3)


Vero (serum Free)
Electroporation
3
positive (3/3)









Example 11
Influenza B Virus Growth in Eggs Results in Loss of HA 196/197 Glycosylation Site

Most influenza B virus clinical isolates contain a potential HA N-linked glycosylation site. This HA N-linked glycosylation site is present around amino acid residues 196-199 for B/Yamagata strains and amino acid residues 197-199 for B/Victoria strains. Recently circulating B/Victoria strains, such as B/Malaysia/2506/04 and B/Ohio/1/05, and recently circulating B/Yamagata strains, such as B/Florida/7/04, contain this potential HA N-linked glycosylation site.


To determine whether the HA glycosylation site of these strains is retained following egg passage, each strain was grown on eggs and nucleotide sequencing was performed to determine the amino acid sequence of the encoded HA polypeptide. The described virus strains used in this study were obtained from the Centers for Disease Control and Prevention (CDC, Atlanta, Ga.). The virus was used to inoculate embryonated chicken eggs obtained from Charles River SPAFAS (Franklin, Conn., North) that had been fertilized 10-11 days prior to virus inoculation. The inoculated eggs were incubated at 33° C. HA viral RNAs from viruses in the inoculated eggs were amplified by RT-PCR, and then sequenced.


The amino acid sequence of the HA polypeptide of influenza B strains B/Ohio/1/05, B/Malaysia/2506/04, and B/Florida/7/04 all changed at the N-linked glycosylation site following egg passage. The sequence at the glycosylation site of B/Ohio/1/05 changed from NET to SET. The sequence at the glycosylation site of B/Malaysia/2506/04 changed from NET to NEA or SET. The sequence at the glycosylation site of B/Florida/7/04 changed from NKT to NKP, DKT, or IKT. See Table 9, below.









TABLE 9







Influenza B HA 196/197 glycosylation site


sequences before and after passage in eggs










Amino acid 196-198 (197-199)














Clinical
Egg
cDNA



Virus
isolate*
isolate
clones







B/Ohio/1/05
NET
SET
SET



B/Malaysia/2506/04
NET
XaEX
NEA






SET



B/Florida/7/04
NKT
XKX
NKP






DKT






IKT







*HA sequences of clinical isolated provided by Dr. M. Shaw of the CDC.




aX indicates mixed sequences







The amino acid sequence at the HA glycosylation site of various other strains of influenza B viruses was examined See FIG. 12, which provides a portion of the HA amino acid sequence for six B/Victoria and eight B/Yamagata following passage on eggs. The potential N-linked glycosylation site (N—X-T/S) is underlined in the figure. It was noted that none of the fourteen influenza B virus strains examined retained their potential N—X-T/S N-linked glycosylation site following egg passage.


Example 12
Loss of the HA 196/197 Glycosylation Site Reduces Influenza B Virus Antigenicity

The effect of the HA 196-197 glycosylation site on antigenicity of the influenza B strains B/Ohio/1/05, B/Malaysia/2506/04, and B/Florida/7/04 was next examined To compare antigenicity of the glycosylated versus nonglycosylated viruses, a pair of viruses corresponding to each of the influenza B strains B/Ohio/1/05, B/Malaysia/2506/04, and B/Florida/7/04 was produced using reverse genetics (see Example 3). The two members of each pair were identical except the first member contained an HA polypeptide with a wild-type amino acid sequence, i.e., an HA amino acid sequence containing the N-linked glycosylation site present in the strain obtained from the CDC, and the second member contained an HA polypeptide lacking the N-linked glycosylation site, i.e., an HA amino acid sequence obtained from the virus following egg passage.


Six of the plasmids used in the reverse genetics technique provided nucleotide sequences corresponding to the internal genome segments of ca B/Ann Arbor/1/66 (MDV-B). A seventh plasmid provided a nucleotide sequence corresponding to the genome segment encoding the wild-type NA polypeptide from each wild-type virus, e.g., each member of the pair of B/Ohio/1/05 viruses was produced using the wild-type NA polynucleotide sequence of the B/Ohio/1/05 strain. An eighth plasmid provided a nucleotide sequence corresponding to a genome segment encoding an HA polypeptide. The HA polypeptide was either the wild-type or egg-passaged HA, depending on whether the influenza virus was the first or second member of the pair of viruses.


The NA and HA polynucleotide sequences of the wild-type viruses were obtained by RT-PCR amplification of the NA or HA vRNA of the wild-type viruses, and cloning of the amplified cDNAs between the two BsmBI sites of pAD3000. Plasmids containing nucleotide sequences corresponding the to genome segments encoding the egg passaged HA polypeptides were prepared by subjecting the plasmids containing the wild-type HA segments to site-directed mutagenesis using a QuikChange® site-directed mutagenesis kit (Stratagene, La Jolla, Calif.).


The plasmids were transfected into co-cultured MDCK and 293 cells. All rescued viruses replicated efficiently in MDCK cells with titers of 6-7 log10 PFU/mL. Seven days after transfection, supernatants from the transfected cells were collected and titrated by plaque assay. Sequence analysis of the recovered viruses confirmed that the wild-type or egg-passaged HA amino acid sequence was retained, in accordance with the HA plasmid used to produce the virus during the transfection.


Antigenicity of each pair of viruses was examined by HAI assay using post-infection ferret sera. Sera were collected from ferrets 21 days following intranasal inoculated with 6-7 log10 PFU virus. Antibody levels in ferret serum against the various viruses were assessed by the hemagglutination-inhibition (HAI) assay. The HAI assay was performed by adding 25 μL serial diluted serum samples with 4 HA units of influenza virus (in a 25 μL volume) in V-bottom 96-well microplates. Following 30 min incubation, 50 μl of 0.5% turkey erythrocytes was added to measure hemagglutination. HAI titer was expressed as the highest serum dilution which inhibits virus hemagglutination. Table 10 provides the antigenicity of the paired wt (HA glycosylation+)/egg-passaged (HA glycosylation) viruses.









TABLE 10







Antigenicity of HA 196/197 glycosylation site variants in ferrets











Geometric mean HAI titer of post



Amino Acid
infection ferret serum against











196-198
Glycosylated
Non-glycosylated


Virus
(197-199)
(G−)
(G+)





B/Ohio/1/05
 SET (G−)

101.6

16.0




NET (G+)

64.0

64.0



B/Malaysia/2506/04
NEA (G−)

64.0

32.0




NET (G+)

25.4

50.8



B/Florida/7/04
DKT (G−)

161.3

28.5



NKT (G+)
35.9

80.6










Sera generated against HA glycosylated viruses had higher HAI titers against HA glycosylated viruses than paired HA nonglycosylated viruses, and sera generated against HA nonglycosylated viruses had higher HAI titers against paired HA nonglycosylated viruses. The antigenic differences between each paired HA glycosylated/HA non-glycosylated virus in the HAI assay varied from 1.5-4.5-fold. This variance indicated that the 196/197 glycosylation site affected virus antigenicity.


Example 13
Influenza B Viruses Having the HA 196/197 GLYCOSYLATION SITE WERE UNABLE TO REPLICATE IN EGGS

To determine whether each member of the paired influenza strains of Example 12 could replicate in eggs, embryonated eggs were inoculated with 102 PFU/egg or 104-105 PFU/egg virus and incubated at 33° C. for three days. Virus peak titers were then determined by plaque assay in MDCK cells. Replication of the paired viruses on eggs (virus titer) and sequence at HA amino acid residues 196-199 for each of the viruses is shown in Table 11.









TABLE 11







Replication of paired HA 196/197 


glycosylation variants in eggs











Amino Acid
Virus




196-198
titer
Amino Acid




(log10
196-199 (197-200)


Virus
(197-199)
PFU/ml)
after growth in eggs





B/Ohio/1/05
SET (G−)
8.7a
SETQ (SEQ ID NO: 52)






NET (G+)
2.1a

ND
d








8.8b
SETQ (SEQ ID NO: 52)





B/Malaysia/
NEA (G−)
8.7a
NEAQ (SEQ ID NO: 53)





2506/04
NET (G+)
1.7a

ND








7.3b
SETQ (SEQ ID NO: 52)








NENQ (SEQ ID NO: 54)





B/Florida/
DKT (G−)
8.2a
DKTQ (SEQ ID NO: 49)





7/04
NKT (G+)
3.0a
NKTQ (SEQ ID NO: 48)







6.7b
NKIQ (SEQ ID NO: 55)








NKTP (SEQ ID NO: 50)






a,bEggs were inoculated with 102 PFU/egg (a) or 104-105PFU/egg (b) of the indicated 6:2 reassortant viruses.




cThe HA sequence of the virus recovered from eggs were determined and amino acid sequence changes are indicated as underlined.




dND: Not determined.







For each virus pair, the member virus lacking the glycosylation site grew well in eggs, to titers greater than 8.0 log10 PFU/mL. However, the member virus containing the glycosylation site (NXT) did not replicate well in eggs inoculated with 102 PFU virus. See Table 11, which indicates that HA glycosylated viruses B/Ohio/1/05, B/Malaysia/2506/04, and B/Florida/7/04, grew to virus titers of only 2.1 log10 PFU/mL, 1.7 log10 PFU/mL, and 3.0 log10 PFU/mL, respectively. Replication of the HA glycosylated member viruses became detectable when the eggs were inoculated with higher amounts of virus, 104-105 PFU/egg. Sequence analysis of these replicating viruses revealed that an amino acid substitution had been introduced at the 196/197 glycosylation site. See Table 11, which indicates that wt glycosylation sequence of B/Ohio/1/05 changed from NET to SET, the wt glycosylation sequence of B/Malaysia/2506/04 changed from NET to SET or NEN, and that the wt glycosylation sequence of B/Florida/7/04 changed from NKT to NKI or a proline was substituted for glutamine immediately C-terminal to the NXT glycosylation sequence. Prior studies (Bause, Biochem J. 209 (1983):331-336; Gavel and Von Heijne, Protein Eng. 3 (1990):433-442) have shown that proline C-terminally adjacent to the HA NXT glycosylation site prevents N-linked glycosylation. Thus, it appeared that lack of glycosylation at HA 196/197 was needed for the influenza B viruses to replicate well on eggs.


Example 14
Identification of an HA Glycosylation+ Influenza B Strain Able to Replicate on Eggs

To determine whether any influenza B strains containing the 196/197 glycosylation site were able to replicate in eggs, eggs were inoculated with various wildtype influenza B virus strains. The HA sequence of the replicating viruses was then determined Most of the influenza B viruses that were able to replicate on eggs did not contain the NXT glycosylation site at residues 197-199 (or 196-198). If the egg-passaged viruses did contain the NXT glycosylation site they were in the process of losing it; the NXT sequence was one of a population of sequences at residues 197-199/196-198 of the HA protein.


Two virus strains, B/Jilin/20/03 (B/JL) and B/Jiangsu/10/03 (B/JS), were identified as having the NXT glycosylation sequence, NKT, following egg passage. B/JL had a proline at position 199, immediately C-terminal to the 196-198 glycosylation site. As discussed above, proline immediately C-terminal to the glycosylation site residues likely interferes with and prevents 196/197 glycosylation. To more closely examine replication of B/JL and B/JS on eggs, paired influenza B virus strains, lacking and containing the NXT glycosylation site sequence were prepared for each of B/JL, B/JS, and related influenza B strain B/Shanghai/361/02 (B/SH) by reverse genetics as described in Example 12. Replication of these paired viruses on MDCK cells and eggs was then determined See Table 12.









TABLE 12







B/Jiangsu/10/03 maintained the 196-197 glycosylation site in eggs











Amino Acid
Virus Titer (log10PFU/ml)
Amino Acid 196-199











Virusa
196-199
NDCK
Egg
after growth in eggsc





B/JS/10/03
DKTQ (G−)
6.5
7.3a
DKTQ (SEQ ID



(SEQ ID



NO: 49)




NO: 49)









NKTQ (G+)
7.4
8.4a
NKTQ (SEQ ID



(SEQ ID



NO: 48)




NO: 48)








B/SH/361/02
DKTQ (G−)
7.3
8.7a
DKTQ (SEQ ID



(SEQ ID



NO: 49)




NO: 49)









NKTQ (G+)
6.9
3.9a
NKTQ (SEQ ID



(SEQ ID



NO: 48)




NO: 48)











6.2b

SKTQ (SEQ ID








NO: 56)











DKTQ (SEQ ID








NO: 49)






B/JL/20/03
NKTP (G−)
6.4
7.6a
NKTP (SEQ ID



(SEQ ID



NO: 50)




NO: 50)









NKTQ (G+)
7.5
3.0a
NKTQ (SEQ ID



(SEQ ID



NO: 48)




NO: 48)











6.8b
NKSQ (SEQ ID







NO: 57)







a,bMDCK cells were infected with the indicated virus at moi of 0.004 and eggs were inoculated with 102 PFU/egg (a) or 104-105PFU/egg (b) of the indicated 6:2 reassortant viruses amplified in MDCK cells that either had (G+) or did not have (G−) the 196/197 HA glycosylation site and incubated at 33° C. for three days. Virus peak titers were determined by plaque assay in MDCK cells.




CThe HA sequence of the virus recovered from eggs were determined and amino acid sequence changes are indicated as underlined.







All three paired virus sets replicated well in MDCK cells, with titers ranging from 6.4 to 7.5 log10 PFU/mL. However, not all viruses replicated well in eggs. Eggs inoculated with 102 log10 PFU of either of the HA 196/197 glycosylated (glycosylation sequence NKTQ (SEQ ID NO: 48)) B/SH or B/JL viruses did not replicate well. Raising the inoculating dose of the B/SH or B/JL HA glycosylated viruses to 104-105 log10 PFU resulted in detectable virus replication. Sequencing these replicating viruses revealed loss of the glycosylation site (from NKT to SKT or DKT in B/SH and from NKT to NKS in B/JL). Unlike the B/SH and B/JL viruses, the B/JS virus was able to replicate well in eggs in the presence or absence of the glycosylation site, titers of 7.3 and 8.4 log10 PFU, respectively.


Western blotting with an HA specific antibody confirmed the glycosylation status of each of the viruses grown in MDCK cells and in eggs. Western blotting was performed by mixing virus from MDCK cell culture supernatants or allantoic fluid with 2× protein lysis buffer (Invitrogen) and electrophoresing on a 10% SDS-PAGE gel. The electrophoresed proteins on the gel were transferred to a nitrocellulose membrane and subjected to Western blot using chicken anti-influenza B antiserum. The protein-antibody complex was detected by a chemiluminescent detection kit (GE Healthcare Bio-Sciences) following incubation with HRP conjugated anti-chicken antibodies.


Western blot analysis showed that when replicated on MDCK cells, HA glycosylation viruses retained their glycosylation site and therefore migrated more slowly on the gel than did their paired counterpart HA glycosylation viruses. See, for example, lanes 1 and 2 of FIG. 13a, which show a band the cross-reacts with the HA antiserum of the glycosylation+ HA (lane 1) virus migrating more slowly than the band in the lane with the virus having the glycosylation+ HA (lane 2). Similar results were obtained for both the B/SH (FIG. 13a, lanes 3 and 4) and B/JL (FIG. 13a, lines 5 and 6) viruses.


When replicated on eggs, only one virus, the B/JS virus, retained the migration pattern in which the band for the glycosylation+ HA virus (FIG. 13b, lane 3) migrated more slowly than the band for the glycosylation HA virus (FIG. 13b, lane 4). This pattern suggested that the B/JS virus was the only virus tested which could replicate on eggs and retain the HA glycosylation site.


Example 15
Arginine at HA Amino Acid Residue Position 141 Stabilizes the 196-197 Glycosylation Site

Review of Table 12 revealed that although both B/JS and B/JL influenza strains had the amino acid sequence NKTQ (SEQ ID NO: 48) at HA amino acid residues 196-199, only B/JS was able to replicate well on eggs and retain the NKTQ (SEQ ID NO: 48) glycosylation site. Comparison of the HA amino acid sequence of the B/JS and B/JL viruses identified three differing amino acid residues. Among these three residues, 141R and 237E were unique to B/JS (relative to other influenza B viruses). At amino acid residue positions 141 and 237, most influenza B strains contain glycine. To test whether one or both of the 141R and/or 237E amino acid residues contributed to stabilization of the B/JS HA 196 glycosylation site, B/JS HA was mutagenized to change 141R and/or 237E to glycine. Replication of the various B/JS viruses on eggs was then determined.


As shown in Table 13, when B/JS HA residue 141 was changed from R to G, the virus was unable to replicate on eggs inoculated at a dose of 102 PFU. Increasing the inoculating dose to 104-105 PFU permitted the virus to replicate on eggs. The replicating B/JS virus having the HA 141G residue was sequenced to determine whether the 196/197 glycosylation site was retained. Sequencing revealed that the NKT glycosylation site had been lost and replaced with either DKT or NKTP (SEQ ID NO: 50). This finding indicated that the HA 141 arginine residue of B/JS may be stabilizing the 196/197 HA glycosylation site. Substituting a glycine for glutamate at B/JS HA amino acid residue 237 did not affect growth on eggs. Data not shown.









TABLE 13







HA 141R stabilizes the 196/197 glycosylation 


site during egg passage











Amino acid at
Virus|




the indicated
Titers|
Amino acid 196-



position
(log10|
199 (197-200)













196-198
PFU/ml)|
after growth












Virus
141
(197-199)
NDCK
Egg
in eggsc





B/JS/
R
NKT
7.4
8.4a
NKTQ (SEQ ID NO: 48)





10/03
G
NKT
7.0
2.4a
NKTQ (SEQ ID NO: 48)









8.5b

DKTQ (SEQ ID NO: 49)











NKTP (SEQ ID NO: 50)





B/SH/
R
NKT
7.6
8.0a
NKTQ (SEQ ID NO: 48)


361/02










B/Ohio/
R
NET
7.6
7.9a
NETQ (SEQ ID NO: 58)


1/05






a,bMDCK cells were infected with the indicated virus at moi of 0.004 and eggs were inoculated with 102 PFU/egg (a) or 104-105PFU/egg (b) of the indicated 6:2 reassortant viruses amplified in MDCK cells that either had (G+) or did not have (G−) the 196/197 HA glycosylation site and incubated at 33° C. for three days. Virus peak titers were determined by plaque assay in MDCK cells.




cThe HA sequence of the virus recovered from eggs were determined and amino acid sequence changes are indicated as underlined.







To further confirm that HA residue 141R was sufficient to stabilize the influenza B HA 196/197 glycosylation site during egg replication, an amino acid substitution of arginine for glycine at HA 141 of B/SH and B/Ohio/1/05 was introduced. As shown in Table 13, both B/SH and B/Ohio/1/05 viruses having the glycine to arginine substitution at HA position 141 were able to replicate efficiently in eggs, titers of approximately 8.0 log10 PFU/mL. The B/SH and B/Ohio/1/05 viruses with the HA 141R substitution also retained HA glycosylation during egg replication. See FIG. 14, which provides a Western blot confirming HA glycosylation of egg passaged B/SH (lane 2), B/Ohio (lane 4), and B/JS (lane 6) viruses having the HA 141R residue. These data indicated that HA residue 141 plays a role in influencing the use of the HA 196/197 glycosylation site of influenza B viruses grown on eggs.


Example 16
Arginine at HA Residue 1410F Influenza B does not Effect Virus Antigenicity

The effect of substituting an arginine residue at HA amino acid position 141 on antigenicity of the influenza B strains was tested. To determine whether the 141R residue affects virus antigenicity, ferret sera was generated against different glycosylated and nonglycosylated viruses. The ferret sera was tested for reactivity against viruses that contained different modifications in the 141 and 196/197 residues.


Ferret sera was prepared by intranasally inoculating ferrets with 7.0 log10 PFU egg-derived viruses with genetic signatures of GD (nonglycosylated) or RN (glycosylated) at the 141 and 196/197 sites, respectively. Post-infection serum was collected from the ferrets twenty-one days later for antigenicity testing in the HAI assay.


B/SH/361/02, B/Ohio/1/05, and B/JS/10/03 viruses having each of the genetic signatures of GD, RN or GN at HA amino acid positions 141 and 196/197, respectively, were prepared to test for antigenicity against the ferret sera. These viruses were prepared from infected MDCK cells; influenza viruses with the G141 and 196/197N residues were unable to grow in eggs.


In the HAI assay, ferret serum generated against nonglycosylated (GD) B/SH/361/02 reacted well with the nonglycosylated B/SH/361/02 virus, but not the glycosylated B/SH/361/02 virus; the HAI titer of the post infection ferret serum was four-fold greater for the nonglycosylated relative to the glycosylated virus. Similarly, ferret serum generated against glycosylated (RN) B/SH/361/02 virus reacted well with glycosylated B/SH/361/02 virus, but not nonglycosylated B/SH/361/02 virus. Again, the difference in HAI titer of the post infection ferret serum was four-fold. These four-fold differences are indicative of an antigenic difference between nonglycosylated and glycosylated viruses, also discussed in Example 12, Table 10.


Ferret serum generated against glycosylated (RN) B/SH/361/02, reacted similarly against the RN and GN glycosylated viruses in the HAI assay; 2-fold greater against the RN glycosylated virus relative to the GN glycosylated virus. This slight difference in reactivity indicated that the amino acid residue change at position 141 from glycine to arginine did not have a significant impact on B/SH/361/02 antigenicity. Similar results were obtained when the same set of HAI assays were performed using influenza B virus strains B/Ohio/1/05 and B/JS/10/03. See Table 14.









TABLE 14







Lack of Effect of Amino Acid 141 on


Antigenicity of Influenza B Strains











Geometric mean HAI titer of post



Amino acid at
infection ferret serum











Virus
141
196/197
GD
RN





B/SH/361/02
G
D (G−)

203.2

40.3



R
N (G+)
40.3

161.3




G
N (G+)
40.3
80.6


B/Ohio/1/05
G

S (G−)


101.6

32.0



R
N (G+)
32.0

161.3




G
N (G+)
25.4
80.6


B/JS/10/03
G
D (G−)

256.0

16.0



R
N (G+)
32.0

90.5




G
N (G+)
128.0 
128.0 





The ferret serum was tested for HAI titers against MDCK-derived viruses using chicken red blood cells.


Geometric mean HAI titers were calculated from three ferret post infection sera.


Homologous HAI titers underlined.






Example 17
Glycosylation at HA 196/197 Affects Binding to α-2,3 Linked Sialic Acids

Because influenza B viruses in which the HA 196/197 site is glycosylated grow well in MDCK cells but not in eggs, glycosylation at HA 196/197 may affect virus receptor binding specificity. Sia (α-2,3) Gal and Sia (α-2,6) Gal are the two major receptor moieties differentially distributed in different host cells. MDCK cells express both Sia (α-2,3) Gal and Sia (α-2,6) Gal moieties. Chicken embryo chorio-allantoic membrane cells express only Sia (α-2,3) Gal moieties. Virus receptor binding specificity can be examined by the hemaagglutination assay using erythrocytes (RBC) from different animal species that differentially express Sia (α-2,3) and Sia (α-2,6) Gal moieties. Horse RBC mainly express Sia (α-2,3) Gal receptors while guinea pig RBC mainly express Sia (α-2,6) Gal receptors. Turkey and chicken RBC are enriched in expression of both Sia (α-2,3) and Sia (α-2,6) Gal moieties (Ito et al., Virol. 156 (1997):493-499).


Egg derived B/Ohio/1/05 and B/Jiangsu/10/03 viruses that were glycosylation+ (RN) or glycosylation (GS, RS, GD, or RD) were tested for their HA titers using horse RBCs (hRBCs), guinea pig RBCs (gpRBCs) and turkey RBCs (tRBCs). Regardless of glycosylation status of influenza B viruses, they all bound similarly well to gpRBCs and tRBCs, both of which express Sia (α-2,6) Gal moieties. In contrast, glycosylation+ (RN) viruses bound poorly or at undetectable levels to hRBC, which only express Sia (α-2,3) moieties, suggesting that glycosylation at HA 196/197 inhibited virus binding to Sia (α-2,3) Gal receptors. See Table 15.









TABLE 15







HA 196/197 glycosylation inhibits HA binding


to receptors having α-2,3 linked sialic acid













Hemagglutination (HA)





titer with the



Amino acid at
Virus Titer
indicated red blood cells













Virus
141
196/197
(log10 PFU/ml)
hRBC
gpRBC
tRBC
















B/Ohio/1/05
G
S (G−)
8.9
128
128
128



R
S (G−)
9.3
512
64
128



R
N (G+)
8.1
<2
64
64


B/JS/10/03
G
D (G−)
8.9
1024
128
256



R
D (G−)
7.6
64
32
32



R
N (G+)
8.6
4
128
256









The inability of the glycosylated viruses to bind to cells expressing Sia (α-2,3) moieties, such as allantoic cells of embryonated chicken eggs, makes it difficult to grow influenza B vaccine strains in eggs. Loss of the glycosylation site, which permits growth of influenza B strains in eggs, alters the antigenicity of the strains. The ability to retain the HA 196/197 glycosylation site of influenza B viruses, while maintaining growth on eggs and virus antigenicity would aid vaccine manufacture. The introduction of an arginine at HA amino acid position 141 of influenza B strains is a means of accomplishing this.


While the foregoing invention has been described in some detail for purposes of clarity and understanding, it will be clear to one skilled in the art from a reading of this disclosure that various changes in form and detail can be made without departing from the true scope of the invention. For example, all the techniques and apparatus described above may be used in various combinations. All publications, patents, patent applications, or other documents cited in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, patent, patent application, or other document were individually indicated to be incorporated by reference for all purposes.


In particular, the following patent application is incorporated by reference in its entirety: U.S. Provisional Application Nos. 60/944,600, filed Jun. 18, 2007.

Claims
  • 1. A method of preparing influenza B virus comprising: (a) introducing a mutation in a hemagglutinin (HA) genome segment resulting in an amino acid substitution at position 141 to arginine, thereby generating a modified HA polypeptide; and(b) replicating an influenza B virus comprising the modified HA polypeptide under conditions whereby influenza B virus is produced, wherein the influenza B virus comprising the modified HA polypeptide replicates to a higher titer than an influenza B virus comprising an HA polypeptide that is unmodified at position 141.
  • 2. The method of claim 1, wherein the introducing a mutation in (a) is performed by site-directed mutagenesis.
  • 3. The method of claim 1, further comprising: (c) determining viral antigenicity for the influenza B virus produced in (b).
  • 4. The method of claim 3, wherein the viral antigenicity of the influenza B virus produced in (b) is not significantly altered by the amino acid substitution in (a).
  • 5. The method of claim 3, wherein the viral antigenicity of the influenza B virus produced in (b) and the viral antigenicity of an influenza B virus comprising an HA polypeptide that is unmodified at position 141 differ by less than 20%.
  • 6. The method of claim 3, wherein the viral antigenicity of the influenza B virus produced in (b) and the viral antigenicity of an influenza B virus comprising an HA polypeptide that is unmodified at position 141 differ by less than 10%.
  • 7. The method of claim 3, wherein the viral antigenicity of the influenza B virus produced in (b) and the viral antigenicity of an influenza B virus comprising an HA polypeptide that is unmodified at position 141 differ by less than 5%.
  • 8. The method of claim 1, wherein the influenza B virus produced in (b) is a reassortant influenza B virus.
  • 9. The method of claim 8, wherein the reassortant influenza B virus comprises at least six internal genome segments from a donor influenza B virus.
  • 10. The method of claim 9, wherein the donor influenza B virus has one or more phenotypes chosen from temperature-sensitive, cold-adapted, and attenuated.
  • 11. The method of claim 10, wherein the donor influenza B virus is B/Ann Arbor/1/66.
RELATED PATENT APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 12/599,761, filed Nov. 11, 2009, now U.S. Pat. No. 8,673,613, which was filed under 35 U.S.C. § 371 as the U.S. national phase of International Application PCT/US2008/067301, filed Jun. 18, 2008, entitled INFLUENZA B VIRUSES HAVING ALTERATIONS IN THE HEMAGLUTININ POLYPEPTIDE, naming as inventors Hong Jin and Zhongying Chen, which designated the U.S. and claims priority to U.S. application Ser. No. 60/944,600, filed Jun. 18, 2007. Each of the foregoing patent applications is incorporated herein by reference in its entirety, including all text, tables and drawings. The instant application contains a Sequence Listing which has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Feb. 19, 2013, is named MDI-0150-US_SL.txt and is 41,021 bytes in size.

US Referenced Citations (64)
Number Name Date Kind
3874999 Zaremba et al. Apr 1975 A
3992522 Chanock et al. Nov 1976 A
4000257 Cano Dec 1976 A
4057626 Metzgar et al. Nov 1977 A
4071618 Konobe et al. Jan 1978 A
4337242 Markus et al. Jun 1982 A
4338296 Lobmann Jul 1982 A
4500512 Barme Feb 1985 A
4512285 McGehee Apr 1985 A
4512972 Schmidt-Ruppin Apr 1985 A
4634666 Engleman et al. Jan 1987 A
4659569 Mitsuhashi et al. Apr 1987 A
5166057 Palese et al. Nov 1992 A
5690937 Parkin Nov 1997 A
5716821 Wertz Feb 1998 A
5789229 Wertz et al. Aug 1998 A
5820871 Palese et al. Oct 1998 A
5840520 Clark et al. Nov 1998 A
5854037 Palese et al. Dec 1998 A
5922326 Murphy Jul 1999 A
6033886 Conzelmann Mar 2000 A
6039958 Koyama Mar 2000 A
6090391 Parkin Jul 2000 A
6146642 Garcia-Sastre et al. Nov 2000 A
6146873 Kistner et al. Nov 2000 A
6168943 Rose Jan 2001 B1
6177082 Dowling et al. Jan 2001 B1
6344354 Webster Feb 2002 B1
6649372 Palese et al. Nov 2003 B1
6656720 Groner et al. Dec 2003 B2
6887699 Palese et al. May 2005 B1
6951754 Hoffmann Oct 2005 B2
7037707 Webster et al. May 2006 B2
7262045 Schwartz et al. Aug 2007 B2
7465456 Hoffmann Dec 2008 B2
8012736 Jin et al. Sep 2011 B2
8093033 Kemble Jan 2012 B2
8409843 Kemble Apr 2013 B2
8574591 Hoffmann et al. Nov 2013 B2
8673613 Jin et al. Mar 2014 B2
8722059 Hoffman et al. May 2014 B2
20020119445 Parkin Aug 2002 A1
20020164770 Hoffmann Nov 2002 A1
20030035814 Kawaoka et al. Feb 2003 A1
20030108859 Kistner et al. Jun 2003 A1
20030147916 Ferko Aug 2003 A1
20040029251 Hoffman et al. Feb 2004 A1
20040137013 Katinger Jul 2004 A1
20050042229 Yang Feb 2005 A1
20050054846 Webster et al. Mar 2005 A1
20050158342 Kemble Jul 2005 A1
20050186563 Hoffmann Aug 2005 A1
20050266026 Hoffmann Dec 2005 A1
20060110406 Kemble May 2006 A1
20070161085 Trager et al. Jul 2007 A1
20090175907 Hoffman et al. Jul 2009 A1
20090208527 Kemble Aug 2009 A1
20100322969 Jin et al. Dec 2010 A1
20120020997 Hoffman et al. Jan 2012 A1
20120196371 Kemble et al. Aug 2012 A1
20120288521 Hoffmann et al. Nov 2012 A1
20130189762 Kemble et al. Jul 2013 A1
20140134208 Hoffmann et al. May 2014 A1
20140220075 Hoffman et al. Aug 2014 A1
Foreign Referenced Citations (34)
Number Date Country
2118234 Apr 1993 CA
1221795 Jul 1999 CN
0480949 Apr 1992 EP
0702085 Mar 1996 EP
0780475 Jun 1997 EP
0863202 Sep 1998 EP
0864645 Sep 1998 EP
1597400 Feb 2005 EP
1826269 Aug 2007 EP
660109 Oct 1951 GB
WO 9103552 Mar 1991 WO
WO 9321306 Oct 1993 WO
WO 9610632 Apr 1996 WO
WO 9610633 Apr 1996 WO
WO 9634625 Nov 1996 WO
WO 9706270 Feb 1997 WO
WO 9712032 Apr 1997 WO
WO 9714434 Apr 1997 WO
WO 9802530 Jan 1998 WO
WO 9813501 Apr 1998 WO
WO 9853078 Nov 1998 WO
WO 9902657 Jan 1999 WO
WO 9915672 Apr 1999 WO
WO 0003019 Jan 2000 WO
WO 0053786 Sep 2000 WO
WO 0060050 Oct 2000 WO
WO 0122992 Apr 2001 WO
WO 0183794 Nov 2001 WO
WO 03091401 Jun 2003 WO
WO 2005014862 Feb 2005 WO
WO 2005062820 Jul 2005 WO
WO 2005115448 Dec 2005 WO
WO 2006041819 Apr 2006 WO
WO 2008157583 Dec 2008 WO
Non-Patent Literature Citations (253)
Entry
“Influenza Strain Details for \B/Jiangsu/10/03”, Apr. 5, 2011, XP002633783, Retrieved from the Internet: URL: http://www.fludb.org/brc/fluStrainDetails.do?strainName=B/Jiangsu/10/03&decorator=influenza [retrieved on Apr. 20, 2011].
“Influenza B virus (B/Jiangsu/10/2003 (recomb)) segment 4 hemagglutinin (HA) gene, partial cds.,” [online], 2007. 05, [searched on Jun. 20, 2013], Accession No. EF473637.
Anderson, et al, “:Evaluation of a Cold-Adapted Influenza B/Texas/84 Reassortant Virus (CRB-87) Vaccine in Young Children,” Journal of Clinical Microbiologu, Sep. 1993, p. 2230-2234.
Banerjee and Barik, 1992, “Gene expression of vesicular stomatitis virus genome RNA”, Virology. 188(2):417-28.
Baron and Barrett, 1997, “Rescue of Rinderpest Virus from Cloned cDNA”, J. Virol. 71:1265-1271.
Baron et al., Electroporation of antibodies, DNA, and other macromolecules into cells: a highly efficient method, Journal of Immunological Methods, 2000, vol. 242, pp. 115-126.
Basler et al., Mutation of Neuraminidase Cysteine Residues Yields Temperature-Sensitive Influenza Viruses, Journal of Virology, Oct. 1999, vol. 73, No. 10, p. 8095-8103.
Beare et al., 1975, “Trials in Man with Live Recombinants Made from A/NPR/8/34 (HO N1) and Wild H3 N2 Influenza Viruses”, Lancet 2(7938):729-732.
Belshe, 1995 “A Review of Attenuation of Influenza Viruses by Genetic manipulationn,” American Journal of Respiratory and Critical Care Medicine 152[4 Pt 2], 572-575. 1995.
Belshe, et al., “The Efficacy of live attenuated, cold-adapted, trivalent intranasal influenza virus vaccine in children,” N. Eng J Med 338:1405-1412.
Bergmann, el al., “The relative amount of an influenza A virus segment present in the viral particle is not affected by a reduction in replication of that segment,”. Journal of General Virology, 1995,76:3211-3215.
Boyce et al., 2001, “Safety and immunogonicity of adjuvanted and unadjuvanled subunit influenza vaccines administered Intranasally to healthy adults”, Vaccine 19:217-226.
Boyer et al., 1994, “Infectious transcripts and cDNA clones of RNA viruses”, Virology. 198(2):415-26.
Brandt et al., 2001, “Molecular Determinants of Virulence, Cell Tropism. and Pathogenic Phenotype of Infectious Bursal Disease Virus”. Journal of Virology 75(24):11974-11982.
Brigden and Elliott. 1996, “Rescue of a Segmented Negative-Strand RNA Virus Entirely from Cloned Complementary DNAS”, Proc. Natl. Acad. Sci. USA 93:15400-15404.
Buchholz et al., 1999 “Generation of Bovine Respiratory Syncytial Virus (BRSV) from cDNA: BRSV NS2 Is Not Essential for Virus Replication in Tissue Culture. and the Human RSV Leader Region Acts as a Functional BRSV Genome Promoter”. J. Virol. 73:251-259.
Bukreyev et al., 1996, “Recovery of infectious respiratory syncytial virus expressing an additional, foreign gene”, J Virol. 70(10):6634-6641.
Burmeister, “Sequence and crystallization of influenza virus b/Beijing/1/87 neuraminidase” Virology, 1991, vol. 180, No. 1, pp. 266-272.
Castrucci et al., 1995, “Reverse genetics system for generation of an influenza A virus mutant containing a deletion of the carboxyl-terminal residue of M2 protein”, J Virol. 69(5):2725-2728.
Chen et al., 1999, “Influenza A virus NS1 protein targets poly (A)-binding protein II of the cellular 3′-end processing machinery”, EMBO 18: 2273-2283.
Chen et al., “Genetic mapping of the cold-adapted phenotype of B/Ann Arbor/1/66, the master donor virus for live attenuated influenza vaccines (FluMist)” Virology vol. 345, No. 2, 2006, pp. 416-423.
Chen et al., “Molecular studies of temperature-sensitive replication of the cold-adapted B/Ann Arbor/1/66, the master donor virus for live attenuated influenza FluMist vaccines.”, Virology Oct. 25, 2008 LNKDPUBMED: 18804834, vol. 380, No. 2, Oct. 25, 2008, pp. 354-362.
Chen et al., “Stabilizing the glycosylation pattern of influenza B hemagglutinin following adaptation to growth in eggs”, Vaccine, Elsevier Ltd, GB, vol. 26, No. 3, Nov. 26, 2007, pp. 361-371.
Clarke et al., 2000, “Rescue of mumps virus from cDNAJ”, J Virol. 74(10):4831-8.
Collins et al., 1991, “Rescue of Synthetic Analogs of Respiratory Syncytial Virus Genomic RNA and Effect of Truncations and Mutations on the Expression of a Foreign Reporter Gene”, Proc. Natl. Acad. Sci. USA 88:9663 9657.
Collins et al., 1995, “Production of infectious human respiratory syncytial virus from cloned cDNA confirms an essential role . . . ” PNAS 92: 11563-7.
Collins et al., 1996, “Parainfluenza Viruses”, Fields Virology, Lippincott-Raven Publishers, Phila., Chapter 41, pp. 1205-1241.
Conzelmann et al., 1994, “Rescue of synthetic genomic RNA analogs of rabies virus by plasmid-encoded proteins”, J Virol. 68(2):713-9.
Conzelmann et al., 1996, “Genetic engineering of animal RNA viruses”, Trends Microbiol. 4(10):386-93.
Conzelmann et al., 1996, “Genetic manipulation of non-segmented negative-strand RNA viruses”, J Gen Virol. 77 (Pt 3):381-389.
Conzelmann et al., 1998, “Nonsegmented negative-strand RNA viruses: genetics and manipulation of viral genomes”, Annu Rev Genet. 32:123-62.
Cox. NJ et al., “Identification of sequence changes in the cold-adapted, live attenuated influenza vaccine strain . . . ”. Virology. Dec. 1998; 167(2)554-567.
De and Banerjee, 1985, “Requirements and Functions of Vesicular Stomatitis Virus Land NS Proteins in the Transcription Process in vitro”, Biochem. Biophys. Res. Commun. 126:40-49.
De and Banerjee, 1993, “Rescue of synthetic analogs of genome RNA of human parainfluenza virus type 3”, Virology, 96(1 ):344-8.
De and Banerjee, 1994, “Reverse genetics of negative strand RNA viruses”, Indian J Biochem Biophys. 31(5):367-76.
De la Luna et al., 1993. “Influenza virus naked RNA can be expressed upon transfection into cells co-expressing the three subunits of the polymerase and the nucleoprotein from simian virus 40 recombinant viruses”, J Gen Virol. 74 (Pt 3):535-9.
De La Luna et al., 1995, “Influenza virus NS1 Protein Enhances the Rate of Translation Initiation of Viral mRNAs”, J. of Virol. 69: 2427-2433.
DeBorde et al., 1988, Sequence comparison of wild-type and cold-adapted B/Ann Arbor/1/66 influenza virus genes Virology 163(2):429-443.
Dimock et al., 1993, Rescue of synthetic analogs of genomic RNA and replicative-intermediate RNA of human parainfluenza virus type 3 . . . J Virol. 67(5):2772-8.
Donabedian et al., “A Mutation in the PA Protein Gene of Cold-Adapted B/Ann Arbor/1/66 Influenza Virus Associated with Reversion of Temperature Sensitivity and Attenuated Virulence,” Virology, 163, p. 444-451, (1988).
Dreher and Hall, 1988, “Mutational Analysis of the Sequence and Structural Requirements in Brome Mosaic Virus RNA for Minus Strand Promoter Activity”, J. Mol. Biol. 201:31-40.
Dreher et al., 1984, “Mutant Viral RNAs Synthesized in vitro Show Altered Aminoacylation and Replicase Template Activities”, Nature 311:171-175.
Dunn et al., 1995, “Transcription of a recombinant bunyavirus RNA template by transiently expressed bunyavirus proteins”, Virology, 211 (1): 133-43.
Durbin et al., 1997, “Recovery of infectious Human Parainfluenza Virus Type 3 from cDNA”, Virol. 235:323-332.
Edwards et al.. 1994. “A randomized controlled trial of cold adapted and inactivated vaccines for the prevention of influenza A disease”, J Infect Dis 169:68-76.
Egorov et al., Transfectant Influenza A Viruses with Long Deletions in the NS1 Protein Grow Efficiently in Vero Cells, Journal of Virology, Aug. 1998, vol. 72, No. 8, p. 6437-6441.
Elliot et al., 1997, Abstract # 96 10.sup.th International conference on Negative Strand Viruses.
Elliott et al., 1991, “Some highlights of virus research in 1990”, J Gen Virol.72 (Pt. 8):1761-79. Review. No abstract available.
Emerson and Yu, 1975, “Both NS and L Proteins are Required for in vitro RNA SynthesiS by Vosicular Stomatitis Virus”, J. Virol. 15:1348-1356.
Enami and Palese, 1991, “High-Efficiency Formation of Influenza Virus Transfectants”, J. Virol. 65:2711-2713.
Enami et al., 1991, “An influenza virus containing nine different RNA segments”, Virology. 185(1):291-8.
Enami et aL, 1990, “Introduction of Site SpeCific Mutations into the Genome of Influenza Virus”, Proc Natl Acad Sci USA 87: 3802-3805.
Enami et al., “Characterization of Influenza Virus NS1 Protein by Using a Novel Helper-Virus-Free Reverse Genetic System” Journal of Virology, 2000, 74(12):5556-5561.
European Search Report mailed on: May 4, 2011 in European Application No. 08771329 filed on: Jun. 18, 2008.
Extended European Search Report dated: Aug. 9, 2012 in European Application No. EP12168901 filed: Apr. 25, 2003.
Fahey and Schooley, 1992, “Status of Immune-Based Therapies in HIV Infection and AIDS”, Clin. Exp. Immunol. 88:1-5.
Flandorfer et al., 2003, •Chimeric Influenza A Viruses with a Functional Influenza B Virus Neuraminidase or Hemagglutinin, J. of Virology—77(17):9116-9123.
Flick. et al., “Promoter elements in the influenza vRNA terminal structure,” RNA, 1996: 2(10):1046-1057.
Fodor et al., “Rescue of Influenza A Virus from Recombinant DNA”. J. of Virology, Am. Society for Microbiology. Nov. 1999, vol. 73, No. 11, pp. 9679-9682.
Fortes et al., 1994, “Influenza virus NS 1 protein inhibits pre-mRNA splicing and blocks mRNA nucleocytoplasmic transport”, EMBO 13: 704-712.
Furminger, “Vaccine Production,” Textbook of Influenza, pp. 324-332 (1996).
Garcia-Sastre A, Palese p, 1993. “Genetic manipulation of negative-strand RNA virus genomes”, Annu Rev Microbiol. :47:765-90.
Garcin et al., 1995, A highly recombinogenic system for the recovery of infectious sendal paramyxovirus from cDNA: generation of a novel copy-back nondefeclive interfering virus•, EMBO J. 14: 6087-6094.
Ghendon, “Cold-Adapted, Live Influenza Vaccines Developed in Russia,” Textbook of Influenza, Chapter 29, pp. 391-399 (1998).
Giudice et al., An MF59-adjuvanted inactivated influenza vaccine containing A/Panama/1999 (H3N2) induced broader serological protein against hetervariant influenza vaccine strain A/Fujian/2002 than a subunit and split influenza vaccine, 2006, Vaccine, vol. 24, pp. 3063-3065.
Goto et al., 1997, “Mutations Affecting the Sensitivity of the Influenza Virus Neuraminidase to 4-Guanidino-2,4-Dideoxy-2,3 Dehydro-N-Acetyineuraminic Acid”, Virol. 238:265-272.
Govorkova, et al., “African Green Monkey Kidney (Vero) Cells Provide an Alternative Host Cell System for Influenza A and B Viruses”. Journal of Virology. American Society for Microbiology. Aug. 1996. vol. 70. No. 8, pp. 5519-5524.
Grosfeld et al., 1995, RNA replication by respiratory syncytial virus (RSV) is directed by the N. P. and L proteins: transcription also occurs under lhese conditions but requires RSV superinfection for efficient synthesis of full-length mRNA. J Virol. 69(9):5677-86.
Guan, Vi, et al., “Molecular Characterization of H9N2 Influenza Viruses: Were They the Donors of the “Internal” Genes of H5N1 Viruses in Hong Kong?”Proc. Nail. Acad. Sci., U.S.A., Aug. 1999, vol. 96, pp. 9363-9367.
Ha et al., “X-ray structures of H5 avian and H9 swine influenza virus hem agglutinins bound to avian and human receptor analogs”, PNAS, USA, vol. 98, No. 20, Sep. 25, 2001, pp. 11181-11186.
Halperin et al., “Saftey and immunogenicity of a new influenza vaccine grown in a mammailian cell culture,” Vaccine 1998, vol. 16, No. 13, p. 1331-1335.
Hardy et al., Egg Fluids and Cells of the Chorioallantoic Membrane of Embryonated Chicken Eggs Can Select Different Variants of Influenza A (H3N2) Viruses, 1995. Virology, vol. 211, pp. 302-306.
Hatada and Fukudo, 1992, “Binding of influenza A virus NS1 protein to dsRNA in vitro”, J. of Gen. Virol. 73: 3325-3329.
He et al., 1997, “Recovery of Infectious SV5 from Cloned DNA and Expression of a Foreign Gene”, Virol. 237:249-260.
Herlocher et al., “Sequence Comparisons of AIAAJ6/60 Influenza Viruses: Mutations Which May Contribute to Attenuation”, Virus Research, 42:11-25; (1996).
Hillman Maurice R., 2000, “Vaccines in historic evolution and perspective: a narrative of vaccine discoveries”, Vaccine 18:1436-1447.
Hoffman and Banerjee, 1997. “An Infectious Clone of a Human Parainfluenza Virus Type 3”, J. Virol. 71:4272-4277.
Hoffman et al., “A DNA transfection system for generation of influenza A virus from eight plasm ids”, PNAS, May 23, 2000, vol. 97, No. 11, pp. 6108-6113.
Hoffman et al., 2002, “Rescue of influenza B virus from eight plasmids”, PNAS 99: 11411-11416.
Hoffman et al., “Multiple gene 1-15 segments control the temperature sensitivity and attenuation phenotypes of ca B/Ann Arbor/1/66.”, Journal of Virology Sep. 2005 LNKDPUBMED: 16103152, vol. 79, No. 17, Sep. 2005, pp. 11014-11021.
Hoffman et al., “Unidirectional RNA polymerase I-polymerase II transcription system for generation of influenza A virus from eight plasmids”, J. of Gen Vir, 2000, 61, 2843-2847.
Hoffman et al.. “Eight-Plasmid Resue System for Influenza A Virus”. International Congress Series. 1219:1007-1013; (2001).
Hoffman et al.. “Eight-Plasmid Resue System for Rapid Generation of Influenza Virus Vaccines”, Vaccine, 20:3165-3170; (2002).
Hoffman et al.. 2000. “Ambisense approach for the generation of influenza A virus: vRNA and mRNA synthesis from one template”, Virology 267:310-7.
Hoffmann et al., “Characterization of the Influenza A Virus Gene Pool in Avian Species in Southern China: Was H6N1 a Derivative or a Precursor of H5N1?” J. Virology. 2000. vol. 74. No. 14. pp. 6309-6315.
Hoffmann et al., “Universal primer set for the full-length amplification of all Influenza A viruses.” Arch Virol. Dec. 2001; 146(12):2275-89.
Hoffmann, Erich, Aufbau eines RNA-Polymerase I-Vektorsystems zur gezlelten Mutagenese von Influenza A Vlren, Glessen 1997 (Doctoral Dissertation).With translation (Generation of an RNA-Polymerase Vector System for the Selective Mutagenesis of Influenza A).
Huang et al.. 1990, “Determination of Influenza virus proteins required for genome replication”. J Virol. 64( 11 ):5669-5673.
International Search Report and Written Opinion maild on: Feb. 10, 2006 in International Application No. PCT/US2004/42669 filed on: Dec. 22, 2004 and published as WO 2005/062820 on Jul. 14, 2005.
International Search Report and Written Opinion maild on: Feb. 9, 2004 in International Application No. PCT/US2003/12728 filed on: Apr. 23, 2003 and published as WO 2003/091401 on Nov. 6, 2003.
International Search Report and Written Opinion maild on: Oct. 11, 2006 in International Application No. PCT/US2005/017734 filed on: May 20, 2005 and published as WO 2005/115448 on Dec. 8, 2005.
International Search Report and Written Opinion maild on: Sep. 2, 2008 in International Application No. PCT/US2008/067301 filed on: Jun. 18, 2008 and published as WO 2008/0157583 on Dec. 24, 2008.
International Search Report and Written Opinion mailed on: Feb. 10, 2006 in International application No. PCT/US45/42669 filed on Dec. 22, 2004.
Jackson et al. 2002, “A reverse genetics approach for recovery of recombinant influenza B Viruses . . . ” J. of Virology 76(22): 11744-11747.
Jin et al., “Imparting Temperature Sensitivity and Attenuation in Ferrets to AlPuerto Rico/6/34 Influenza Virus by . . . ”. J. of Virology. Am. Society for Microbiology, pp. 995-998, Jan 2004.
Jin et al., Multiple Amino acid residues confer temperature sensitivity to human influenza vaccine strains (FluMist) derived from cold-adapted A/Ann Arbor/6/60, 2003, Virology, vol. 302, pp. 18-24.
Jin-Hua Liu et al: “Genetic Conservation of Hemagglutinin Gene of H9 Influenza Virus in Chicken Population in Mainland China” Virus Genes, Kluwer Academic Publishers, Bo, vol. 29, No. 3, Dec. 1, 2004, pp. 329-334.
Kaplan et al.. 1985. “In vitro Synthesis of Infectious Poliovirus RNA”. Proc. Natl. Acad. Sci. USA 82:8424-8428.
Katinger et al., “Attenuated Influenza Virus as a Vector for Mucosal Immunization against HIV-1”, Vaccines, pp. 315-319, (1997).
Kato et al., 1996, “Initiation of Sendai Virus Multiplication from Transfected cDNA or RNA with Negative or Positive Sense”, Genes Cells 1 :569-579.
Keitel. et al., “Live Cold-Adapted, Reassortant Influenza Vaccines (USA),” Textbook of Influenza, Chapter 28, pp. 373-390 (1998).
Kimura et al., 1993, “An in vivo study of the replication origin in the influenza virus complementary RNA”. J Biochem (Tokyo) 113(1):88-92.
Kimura et al., 1992, Transcription of a recombinant influenza virus RNA in cells that can express the influenza virus RNA polymerase and nucleoprotein genes•, J Gen Virol. 73 (Pt 6):1321-1328.
Kistner et al., Development of a Mammalian Cell (Vero) Derived Candidate Infleunza Virus Vaccine, Vaccine, 1998, vol. 16, No. 9-10, pp. 960-968.
Kobayashi, 1992, Reconstitution of influenza virus RNA polymerase from three subunits expressed using recombinant baculovirus system. Virus Res. 22(3):235-245.
Konarska et al., 1990, “Structure of RNAs replicated by the DNA-dependent T7 RNA polymerase”, Cell. 63(3):609-18.
Krystal et al., 1986, Expression of the Three Influenza Virus Polymerase Proteins in a Single Cell Allows Growth Complementation of Viral Mutants•, Proc. Nail. Acad. Sci. USA 83:2709-2713.
Kunkel, 1985. “Rapid and Efficient Site-Specific MutagenesiS without Phenotypic Selection”, Proc. Natl. Acad. Sci. USA 82:488•492.
Lamb et al., 1996, Fundamental Virology 3.sup.rd ed. Chapters 20 and 21.
Lawson et al., 1995, “Recombinant vesicular stomatitis viruses from DNA”, Proc Natl Acad Sci U S A.92(1 0):4477-81.
Levis et al., 1986, “Deletion Mapping of Sindbis Virus 01 RNAs Derived from cDNAs Defines the Sequences Essential for Replication and Packaging”, Cell 44:137-145.
Li et al., Virus Research, 1995, 37:153-161.
Li et al.. 1999, “Recombinant influenza A virus vaccines for the pathogenic human A/Hong Kong/97 (H5N1) viruses,” J. of Infectious Diseases. 179:1132-8.
Lu Bin et al: “Improvement of influenza A/Fujian/411/02 (H3N2) virus growth in embryonated chicken eggs by balancing the hemagglutinin and neuraminidase activities, using reverse genetics” Journal of Virology, vol. 79, No. 11, Jun. 2005, pp. 6763-6771.
Lugovtsev et al., “Changes of the receptor-binding properties of influenza B virus B/Victoria/504/2000 during adaptation in chicken eggs”, Virology, Academic Press,Orlando, US, vol. 394, No. 2, Nov. 25, 2009, pp. 218-226.
Lugovtsev V.Y. et al.: ‘Generation of the influenza B viruses with improved growth phenotype by substitution of specific amino acids of hemagglutinin’ Virology vol. 365, pp. 315-323.
Lugovtsev V.Y. et al.: ‘Mutational pattern of influenza B viruses adapted to high growth replication in embryonated eggs’ Virus Research vol. 109, No. 2, 2005, pp. 149-157.
Luytjes et al., “Amplification, expression, and packaging of foreign gene by influenza virus,” 1989, Cell, 59:1107-1113.
Maassab et al., Evaluation of a Cold-Recombinant Influenza Virus Vaccine in Ferrets, J. of Infectious Diseases, 146:780-900; (1982).
Maassab et al., The Development of Live Attenuated Cold-Adapted Influenza Virus Vaccine for Humans,Reviews in Medical Virology, 1999, vol. 9, pp. 237-244.
Maassab et al., “Development and characterization of cold-adapted viruses for use as live virus vaccines,” Vaccine, vol. 3, Dec. 1985, pp. 355-369.
Maassab et al.. “Evaluation of a Cold-Recombinant Influenza Virus Vaccine in Ferrets”, J. of Infectious Diseases. 146:780-900; (1982).
Maassab, Adaptation and growth characteristics of influenza virus at 25 degrees C Nature. 213:612-614 (1967).
Marten et al., “Production of influenza virus in Cell Cultures for Vaccine Preparation”, Novel Slrategies in Design and Production of Vaccines, pp. 141-151; (1996).
Martin at al., 1998, “Studies of the Binding Properties of Influenza Hemagglutinin Receptor-Site Mutants”, Virology 241:101-111.
Medeiros Rita et al: “Hemagglutinin residues of recent human A (H3N2) influenza viruses that contribute to the inability to agglutinate chicken erythrocytes”, Virology, vol. 289, No. 1, Oct. 10, 2001, pp. 74-85.
Melkonyan et al., Electroporation efficiency in mammalian cells is increased by dimethyl sulfoxide (DMSO). Nucleic Acids Research, 1996, vol. 24, No. 21, pp. 4356-4357.
Mena et al., 1994, “Synthesis of biologically active influenza virus core proteins using a vaccinia virus-T7 RNA polymerase expression system”, J Gen Virol. 75 (Pt 8):2109-14.
Mena et al., 1996, “Rescue of a Synthetic Chloramphenicol Acetyltransferase RNA into Influenza Virus-Like Particles Obtained from Recombinant Plasmids”, J. Virol. 70: 5015-S024.
Merten at at. “Production of influenza virus in Cell Cultures for Vaccine Preparation”, Novel Strategies in Design and Production of Vaccines, pp. 141-151; (1996).
Mochalova L et al.: “Receptor-binding properties of modern human influenza viruses primarily isolated in Vero and MDCK ceils and chicken embryonated eggs”, Virology, Academic Press,Orlando, US, vol. 313, No. 2, Sep. 1, 2003, pp. 473-480.
Moyer et al., 1991, “Assembly and transcription of synthetic vesicular stomatitis virus nucleocapsids”, J Virol. 65(5):2170-8.
Murphy & Coelingh, “Principles Underlying the Development and Use of Live Attenuated Cold-Adapted Influenza A and B Virus Vaccines”, ViralImmunol. 15:295-323; (2002).
Muster et al., 1991, “An influenza A virus containing influenza B virus S′ and 3′ noncoding regions on the neuraminidase gene is attenuated in mice:”. Proc Natl Acad Sci U S A.88(12):5177-81.
Naito and Ishihama, 1976, “Function and Structure of RNA Polymerase from Vesicular Stomatitis Virus”, J. Biol. Chern. 251 :4307-4314.
Nakagawa et al., “Neutralizing epitopes specific for influenza B virus Yamagata group strains are in the loop”, Journal of General Virology vol. 84, No. 4, Apr. 2003, pp. 769-773.
Nakajima et al., 2003. “Restriction of Amino Acid Change in Influenza A Virus H3HA: Comparison of Amino Acid Changes Observed . . . ”; J, of Virology 77(18):10088-10098.
Nara et al., 1987. “Simple, Rapid, Quantitative, Syncytlum-Fonmlng MIcorassay for the Detection of Human Immunodeficiency Virus Neutralizing Antibody”, AIDS Res. Hum. Retroviruses 3:283-302.
Nemeroff et al., 1998, “Influenza Virus NS1 Protein Interacts with the Cellular 30 kDa Subunit of CPSF and Inhibits 3′ End Formation of Cellular Pre-mRNAs”, Mol. Cell1 :991•1000.
Neumann et al., 1994, “RNA Polymerase I-Mediated Expression of Influenza Viral RNA Molecules”, Virol, 202:477-479.
Neumann et al. Generation of influenza A viruses entirely from cloned cDNAsn, Proc. Natl. Acad. Sci.. Microbiology, Aug. 1999, vol. 96, pp. 9345-9350.
Neumann G., et al., “Generation of Influenza A Virus from Clones cDNAs—Historical Perspective and Outlook for the New Millenium,” Rev.Med. Virol, (2002)12; 13-30.
Neumann, et al., “Genetic Engineering of Influenza and Other Negative-Strand RNA Viruses Containing Segmented Genomes,” Advances in Virus Research, 1999; 53: 265-300.
Nichol et al., “Effectiveness of live, attenuated Intranasal influenza vlrus vaccine in healthy, working adults: a randomized controlled trial”, JAMA 281:137-44.
Oxford et al., “A host-cell-selected variant of influenza B virus with a single nucleotide substitution in HA affecting a potential glycosylation site was attenuated in virulence for volunteers,” Arch Virol., vol. 110, pp. 37-46.
Oxford et al., “Direct isolation in eggs of influenza A (H1N1) and B Virus with haemagglutinins of different antigenic and amino acid compositions,” J. Gen Virol 1991, vol. 72, No. 1, pp. 185-189.
Palese et al., 1996, “Negative-Strand RNA Viruses: Genetic Engineering and Applications”, Proc. Natl. Acad. Sci. USA 93,11354-11358.
Paltnaik et al., 1991, •Cells that express all flVe proteins of vesicular stomatitis virus from cloned cDNAs support replication, assembly, and budding of defective Interfering particles, Proc Nail Acad Sci USA. 88(4):1379-83.
Paragas et al., “Influenza B and C Virus NEP (NS2) Proteins Possess Nuclear Export Activities,” Journal of Virology, Aug. 2001, p. 7375-7383.
Park et al., 1991, “Rescue of a Foreign Gene by Sendai Virus”, Proc. Natl. Acad. Sci. USA 88:5537-5541.
Parkin et al.. “Temperature Sensitive Mutants of Influenza A Virus Generated by Reverse Genetics . . . ”. Vir. Res .• 46:31-44; (1996).
Parkin N. et al., “Genetically Engineered Live Atenuated Influenza A Virus Vaccine Candidates”, J. Virol., pp. 2772-2778; (1997).
Peeters et al., 1999, “Rescue of Newcastle Disease Virus from Cloned cDNA: Evidence that Cleavability of the Fusion Protein is a Major Determinant for Virulence”, J. Virol. 73:5001-5009.
Pekosz et al., 1999, “Reverse genetics of negative-strand RNA viruses: closing the circle”, Proc Natl Acad Sci USA. 96(16):8804-6.
Percy et al., 1994, “Expression of a foreign protein by influenza A virus”, J Virol 68(7):4486-92.
Perez, Daniel R. et al., “The Matrix 1 Protein of Influenza A Virus Inhibits the TransciptaseActivity of a Model Influenza Reporter Genome in Vivo”, Article No. VY989318, Virology, 1998. vol. 249. pp. 52-61.
Perkin N. et al., “Genetically Engineered live Atenuated Influenza A Virus Vaccine Candidates”, J. ViraL, pp. 2772-2778; (1997).
Pleschka et al., 1996, “A Plasmid-Based Reverse Genetics System for Influenza A Virus”, J. Virol. 70:4188-4192.
Qiu et. al.. 1994, “The influenza virus NS1 protein is a poly(A)-binding protein that inhibits nuclear export of mRNAs containing poly(A)”, J Virol. 68(4):2425-32.
Qiu et.al., 1995. the influenza virus NS1 protein binds to a specific region in human U6 snRNA and inhibits U6-U2 and U6-U4 snRNA . . . , RNA 1:304-16.
Racaniello et aL. 1981. “Cloned Poliovirus Complementary DNA is Infectious in Mammalian Cells”, Science 214:916-919.
Radecke et al. 1995, “Rescue of measles viruses from cloned DNA”. EMBO J. 14(23):5773-84.
Radecke et al.. “Reverse Genetics Meets the Nonsegmented Negative-Strand RNA Viruses”, Medical Virology. vol. 7: 49-63 (1997).
Roberts and Rose. 1998. “Recovery of Negative-Strand RNA Viruses from Plasmid DNAs: a Positive Approach Revitalizes a Negative Field”, Virol. 247:1-6.
Rocha et al., Comparison of 10 influenza A (H1 N1 and H3N2) haemagglutinin sequences obtained directly from clinical specimens to those of MOCK cell- and egg-grown viruses, 1993, Joumal of General Virology, vol. 74, pp. 2513-2518.
Rogers G N et al: “Single Amino-Acid Substitutions in Influenza Hem Agglutinin Change Receptor Binding Specificity”, Nature (London), vol. 304, No. 5921, 1983, pp. 76-78.
Rose et al., 1996, “Positive Strands to the Rescue Again: . . . ” PNAS USA 94:14998-15000.
Schickli et al., “Plasmid-only rescue of influenza A virus vaccine candidates,” Philosophical Transactions of the Royal Society of London. Series B. Biological Sciences (London), 2001, 356:1965-1973.
Schlesinger et al., 1995. “RNA viruses as vectors for the expression of heterologous proteins”, Mol Biotechnol. 3(2):155-165.
Schlicki et al., Plasmid-only rescue of influenza A virus vaccine candidates, Philosophical Transactions of the Royal Society of London Series S, 2001, vol. 356, p. 1965-1973.
Schnell et al.. 1994. “Infectious Rabies Viruses from Cloned eDNA”, EMBO J. 13:4195-4203.
Scholtissek, et al., “The Nucleoprotein as a Possible Major Factor in Determining Host Specificity of Influenza H3N2 Viruses,” Virology, 1985; 147:287-294.
Seong et al.. 1992. A new method for reconstituting influenza polymerase and RNA in vitro: a study of the promoter elements for cRNA and vRNA synthesis in vitro and viral rescue in vivo. Virology. 166(1):247-260.
Sidhu et al., 1995, “Rescue of synthetic measles virus minireplicons: measles genomic termini direct efficient expression and propagation of a reporter gene”. Virology, 208(2):600-607.
Snyder et al., Four Viral Genes Independently Contribute to Attenuation of Live Influenza AIAnn Arbor/6/60 (H2N2) Cold-Adapted . . . J, Virol.. 62:488-95; (1988).
Stoeckle, “Segment-specific and common nucleotide sequences in the noncoding regions of influenza B virus genome RNAs,” PNAS USA, 1987, vol. 84, No. 9, pp. 2703-2707.
Subbarao et al., The Attenuation Phenotype Conferred by the M Gene of the Influenza AIAnn Arbor/6/60 Cold-Adapted Virus (H2N2) on the . . . Virus. Res ., 25:37-50; (1992).
Subbarao et al., “Sequential Addition of Temperature-Sensitive Missense Mutations into the PB2 Gene of Influenza A Transfectanl . . . ”. J. of Vir., Am. Society for Microbiology. Oct. 1995. pp. 5969-5977.
Subbarao, et al., “Rescue of a Influenza A Virus Wild-Type PB2 Gene and a Mutant Derivative Bearing a Site-Specific . . . ” J. of Virology, 1993, pp. 7223-7228.
Subbarao, K., et al., “Evaluation of Genetically Modified Reassortant H5N1 Influenza A Virus Vaccine Candidate Generated by Plasmid-Based Reverse Genetics.” Virology (2003) 305: 192-200.
Subrehmanyan et al., The Development of Double-Seeded and Mixed Cell Culture Systems for the Use in Diagnostic Virology, Archiv fur die desamte Virusforschung, 1974, vol. 44. pp. 291-297.
Supplementary European Search Report mailed on: Dec. 11, 2007 in European Patent Application No. EP03724208.8 filed on Apr. 25, 2003.
Supplementary European Search Report mailed on: Dec. 29, 2006 in European Patent Application No. EP0481407.6 filed on Dec. 22, 2004.
Supplementary Partial European Search Report mailed on: Apr. 1, 2009 in European Patent Application No. EP05750661.0 filed on May 20, 2005.
Supplementary Partial European Search Report mailed on: Sep. 24, 2007 in European Patent Application No. EP03724208.8 filed on Apr. 25, 2003.
Supplemetary Eurpoean Search Report mailed Dec. 29, 2006 in Eurpoean Application No. 04814807.6 filed on Dec. 22, 2004.
Szewczyk et al., 1988, •Purification, Thioredoxin Renaturation, and Reconstituted Activity of the Three Subunits of the Influenza A Virus RNA Polymerase. Proc. Nat. Acad. Sci. USA 85:7907-7911.
Taylor et al., 1990, “Newcastle Disease Virus Fusion Protein Expressed in a Fowlpox Virus Recombinant Confers Protection in ChiCkens”, J. Viral. 64:1441-1450.
Treanor et al., “Evaluation of the Genetic Stability of the Temperature-Sensitive PB2 Gene Mutation of the Influenza A/Ann Arbor/6/60 Cold-Adapted Vaccine Virus,” Journal of Virology Dec. 1994, p. 7684-7688.
Verhoeyen, “Complete nucleotide sequence of the influenza B/Singapore/222/79 virus hemagglutinin gene and comparison with the B/Lee/40 hemagglutinin” Nucleic Acids Res., 1983, vol. 11, No. 14, pp. 4703-4712.
Wang et al. Extensive Hetergeneity in the Hemagglutinin of Egg-Grown Influenza Viruses from different Patients, 1989, Virology, vol. 171, p. 275-279.
Ward et al., 1988, “Direct Measurement of the Poliovirus RNA Polymerase Error Frequency in Vitro”, J. Virol. 62:558-562.
Wareing at al., 2001. Immunogenic and Isotype-Specific Responses to Russian and US Cold-Adapted Influenza A Vaccine Donor Strains . . . , J of Medical Virology 65:171-177.
Wareing, M. D., et al. “Preparation and Characterisation of Attenuated Cold-Adapted Influenza A Reassortants Derived from the AlLeningradl1341l7/57 Donor Strain.” Vaccine (2002) 20: 2082-90.
Webby et al., 2004, “Responsiveness to a pandemic alert: use of reverse genetics for rapid development of influenza vaccines”, Lancet 363:1099-1103.
Whelan et al., 1995, “Effiecient recovery of infectious vesicular stomatitis virus entirely from cDNA clones”, Proc.Natl.Acad.Sci. USA 92: 8388-8392.
Xu et al., 1995 #AAB06964 (abstract only).
Xu et al., 1996, “Genetic Variation in Neuraminidase Genes of Influenza A (H3N2) Viruses”, Virology 224:175-183.
Xu, Xiyan et al., “Genetic Characterization of the Pathogenic Influenza A/Goose/Guandong/1/96 (H5N1) Virus: Similarly of its Hemagglutinin Gene to Those of H5N1 Viruses form the 1997 Outbreaks in Hong Kong”, Article 10 viro. 1999.9820, Virology, 1999, vol. 261, pp. 15-19.
Yamanaka et al.. “In vivo analysis of the promoter structure of the influenza virus RNA genome using a transfection system with an engineered RNA.” Proc Nail Aced Sci USA 88: 5369-5373. 1991.
Yu et al., 1995, “Functional coNA clones of the human respiratory syncytial (RS) virus N, P, and L proteins support replication RS virus genomic RNA analogs and define minimal trans-acting requirements for RNA replication”, J Virol. 69(4):2412-9.
Yusoff et al.. 1987, “Nucleotide Sequence Analysis of the L Gene of Newcastle Disease Virus: Homologies with Sendi and Vesicular Stomatitis Viruses” Nucleic Acids Res. 15: 3961-3976.
Zaghouani el al., 1991, “Induction of antibodies to the envelope protein of the human immunodeficiency virus by Immunization with monoclonal anti-idlotypes”, Proc. Natl. Acad. Sci. USA 88:5645-5649.
Zaghouani et al., 1992. “Cells Expressing an H Chain to Gene Carrying a Viral T Cell Epitope Are Lysed by Specific Cytolytic T Cells”, J. Immunol. 148:3604-3609.
Zambon et al., The Pathogenesis of Influenza in Humans, Reviews in Medical Virology, Jul.-Aug. 2001, vol. 11, No. 4, pp. 227-241.
Zhang and Air, 1994, “Expression of Functional Influenza Virus A Polymerase Proteins and Template from Cloned cDNAs in Recombinant Vaccinia Virus Infected Cells”, Biochem. Biophys. Res. Commun. 200:95-101.
Zhang et al.. Persistence of four related human munodeficiency virus subtypes during the course of zidovudine therapy . . . J. Virol. 1994 66: 425-432.
Zhou, Yan, et al., “Membrane-Anchored Incorporation of a Foreign Protein in Recombinant Influenza Virions”, Article No. VY989169, Virology, 1998, vol. 246, pp. 83-94.
Zobel et aL, 1993, “RNA polymerase I catalysed transcription of insert viral cDNA”, Nucleic Acids Res. 21 (16):3607-14.
Extended European Search Report mailed on Sep. 2, 2013 in European Patent Application No. 13159978.9, filed on May 20, 2005.
Office Action mailed on: Jun. 20, 2008 in U.S. Appl. No. 11/018,624, filed Dec. 22, 2004 and published as: 2005-0158342 on: Jul. 21, 2005, now abandoned.
Office Action mailed on: Sep. 24, 2007 in U.S. Appl. No. 11/018,624, filed Dec. 22, 2004 and published as: 2005-0158342 on: Jul. 21, 2005, now abandoned.
Office Action mailed on: Feb. 2, 2007 in U.S. Appl. No. 11/018,624, filed Dec. 22, 2004 and published as: 2005-0158342 on: Jul. 21, 2005, now abandoned.
Office Action mailed on: Jun. 13, 2006 in U.S. Appl. No. 11/018,624, filed Dec. 22, 2004 and published as: 2005-0158342 on: Jul. 21, 2005, now abandoned.
Office Action mailed on: Apr. 28, 2006 in U.S. Appl. No. 11/018,624, filed Dec. 22, 2004 and published as: 2005-0158342 on: Jul. 21, 2005, now abandoned.
Office Action mailed on: May 18, 2010 in U.S. Appl. No. 12/336,158, filed Dec. 16, 2008 and published as: 2009-0208527 on: Aug. 20, 2009.
Office Action mailed on: Jun. 1, 2010 in U.S. Appl. No. 12/336,158, filed Dec. 16, 2008 and published as: 2009-0208527 on: Aug. 20, 2009.
Office Action mailed on:Nov. 8, 2010 in U.S. Appl. No. 12/336,158, filed Dec. 16, 2008 and published as: 2009-0208527 on: Aug. 20, 2009.
Office Action mailed on: May 18, 2011 in U.S. Appl. No. 12/336,158, filed Dec. 16, 2008 and published as: 2009-0208527 on: Aug. 20, 2009.
Office Action mailed on: Sep. 8, 2011 in U.S. Appl. No. 12/336,158, filed Dec. 16, 2008 and published as: 2009-0208527 on: Aug. 20, 2009.
Office Action mailed on: Nov. 30, 2012 in U.S. Appl. No. 13/309,498, filed Dec. 1, 2011 and published as: on:.
Office Action mailed on: Aug. 6, 2012 in U.S. Appl. No. 13/309,498, filed Dec. 1, 2011 and published as: on:.
Office Action mailed on: Jul. 22, 2008 in U.S. Appl. No. 11/133,345, filed May 20, 2005 and published as: 2005-0266026 on: Dec. 1, 2005, and issued as 7,465,456 on Dec. 16, 2008.
Office Action mailed on: Aug. 20, 2007 in U.S. Appl. No. 11/133,345, filed May 20, 2005 and published as: 2005-0266026 on: Dec. 1, 2005, and issued as 7,465,456 on Dec. 16, 2008.
Office Action mailed on: Nov. 27, 2006 in U.S. Appl. No. 11/133,345, filed May 20, 2005 and published as: 2005-0266026 on: Dec. 1, 2005, and issued as 7,465,456 on Dec. 16, 2008.
Office Action mailed on: Aug. 8, 2006 in U.S. Appl. No. 11/133,345, filed May 20, 2005 and published as: 2005-0266026 on: Dec. 1, 2005, and issued as 7,465,456 on Dec. 16, 2008.
Office Action mailed on: Jun. 28, 2013 in U.S. Appl. No. 13/296,933, filed Nov. 15, 2011 and published as: 2012-0288521 on: Nov. 15, 2012.
Office Action mailed on: Feb. 7, 2013 in U.S. Appl. No. 13/296,933, filed Nov. 15, 2011 and published as: 2012-0288521 on: Nov. 15, 2012.
Office Action mailed on: Oct. 24, 2012 in U.S. Appl. No. 13/296,933, filed Nov. 15, 2011 and published as: 2012-0288521 on: Nov. 15, 2012.
Office Action mailed on: Aug. 25, 2011 in U.S. Appl. No. 12/254,131, filed Oct. 20, 2008 and published as: 2009-0175907 on: Jul. 9, 2009 and issued as: 8,114,415 on Feb. 14, 2012.
Office Action mailed on: Apr. 21, 2011 in U.S. Appl. No. 12/254,131, filed Oct. 20, 2008 and published as: 2009-0175907 on: Jul. 9, 2009 and issued as: 8,114,415 on Feb. 14, 2012.
Office Action mailed on:Aug. 19, 2010 in U.S. Appl. No. 12/254,131, filed Oct. 20, 2008 and published as: 2009-0175907 on: Jul. 9, 2009 and issued as: 8,114,415 on Feb. 14, 2012.
Office Action mailed on: Mar. 23, 2010 in U.S. Appl. No. 12/254,131, filed Oct. 20, 2008 and published as: 2009-0175907 on: Jul. 9, 2009 and issued as: 8,114,415 on Feb. 14, 2012.
Office Action mailed on: Nov. 28, 2012 in U.S. Appl. No. 13/214,110, filed Aug. 19, 2011 and published as: 2012-0020997 on: Jan. 26, 2012.
Office Action mailed on: Jun. 11, 2012 in U.S. Appl. No. 13/214,110, filed Aug. 19, 2011 and published as: 2012-0020997 on: Jan. 26, 2012.
Office Action mailed on: Jul. 15, 2011 in U.S. Appl. No. 10/423,828, filed Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Office Action mailed on: Jul. 5, 2011 in U.S. Appl. No. 10/423,828 filed, Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Office Action mailed on: Oct. 13, 2010 in U.S. Appl. No. 10/423,828, filed Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Office Action mailed on:Feb. 5, 2010 in U.S. Appl. No. 10/423,828, filed Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Office Action mailed on:Dec. 8, 2008 in U.S. Appl. No. 10/423,828, filed Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Office Action mailed on: Mar. 26, 2008 in U.S. Appl. No. 10/423,828, filed Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Office Action mailed on: Jun. 11, 2007 in U.S. Appl. No. 10/423,828, filed Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Office Action mailed on: Sep. 22, 2006 in U.S. Appl. No. 10/423,828, filed Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Office Action mailed on: Feb. 7, 2006 in U.S. Appl. No. 10/423,828, filed Apr. 25, 2003 and published as: 2004-0029251 on: Feb. 12, 2004 and issued as: 8,012,736 on Sep. 6, 2011.
Extended European Search Report mailed on Nov. 15, 2013 in European Patent Application No. 13170051.0, filed on Jun. 18, 2008 and published as EP 2 674 486 on Dec. 18, 2013.
Office Action dated Oct. 25, 2013 in U.S. Appl. No. 12/599,761, filed Sep. 10, 2010 and published as US 2010-0322969 on Dec. 23, 2010.
Office Action mailed on: Jun. 10, 2013 in U.S. Appl. No. 12/599,761, filed Sep. 10, 2010 and published as: 2010-0322969 on: Dec. 23, 2010.
Office Action mailed on: Nov. 21, 2012 in U.S. Appl. No. 12/599,761, filed Sep. 10, 2010 and published as: 2010-0322969 on: Dec. 23, 2010.
Office Action dated Dec. 23, 2013 in U.S. Appl. No. 13/214,110, filed Aug. 19, 2011 and published as US 2012-0020997 on Jan. 26, 2012.
Office Action dated Sep. 8, 2014 in U.S. Appl. No. 13/779,549, filed Feb. 27, 2013 and published as US 2013-0189762 on Jul. 25, 2013.
Donabedian et al., “Genetics of cold-adapted B/Ann Arbor/1/66 influenza virus reassortants: the acidic polymerase (PA) protein gene confers temperature sensitivity and attenuated virulence” Microb. Pathog. (1987) 3(2):97-108.
Office Action dated Dec. 24, 2014 in U.S. Appl. No. 13/779,549, filed Feb. 27, 2013 and published as US 2013-0189762 on Jul. 25, 2013.
Office Action dated Jan. 16, 2015 in U.S. Appl. No. 14/222,461, filed Mar. 21, 2014 and published as US 2014-0220075 on Aug. 7, 2014.
Related Publications (1)
Number Date Country
20140199683 A1 Jul 2014 US
Provisional Applications (1)
Number Date Country
60944600 Jun 2007 US
Continuations (1)
Number Date Country
Parent 12599761 US
Child 14161430 US