WO2004087062A2 - Compositions and methods involving respiratory syncytial virus subgroup b strain 9320 - Google Patents

Compositions and methods involving respiratory syncytial virus subgroup b strain 9320 Download PDF

Info

Publication number
WO2004087062A2
WO2004087062A2 PCT/US2004/009438 US2004009438W WO2004087062A2 WO 2004087062 A2 WO2004087062 A2 WO 2004087062A2 US 2004009438 W US2004009438 W US 2004009438W WO 2004087062 A2 WO2004087062 A2 WO 2004087062A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
leu
amino acid
nucleic acid
polypeptide
Prior art date
Application number
PCT/US2004/009438
Other languages
French (fr)
Other versions
WO2004087062A3 (en
Inventor
Xing Cheng
Hyun Jung Park
Hong Jin
Original Assignee
Medimmune Vaccines, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medimmune Vaccines, Inc. filed Critical Medimmune Vaccines, Inc.
Priority to EP04749465A priority Critical patent/EP1613345B1/en
Priority to AT04749465T priority patent/ATE469170T1/en
Priority to DE602004027362T priority patent/DE602004027362D1/en
Priority to CA2520671A priority patent/CA2520671C/en
Priority to JP2006509391A priority patent/JP4783726B2/en
Priority to AU2004226369A priority patent/AU2004226369A1/en
Publication of WO2004087062A2 publication Critical patent/WO2004087062A2/en
Publication of WO2004087062A3 publication Critical patent/WO2004087062A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1027Paramyxoviridae, e.g. respiratory syncytial virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5254Virus avirulent or attenuated
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18522New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • the present invention is in the field of virology. More specifically, the invention relates to human respiratory syncytial virus, including the diagnosis, treatment, and prevention of human RSV infections.
  • RSV Human Respiratory Syncytial Virus
  • RSV is an enveloped virus that has a single-stranded negative sense non- segmented RNA genome, and it is classified in the Pneumovirus genus of the Paramyxoviridae family (Collins et al. (2001) Respiratory syncytial virus, pp. 1443-1485. In; Knipe and Howley (eds.) Fields Virology vol. 1. Lippincott, Williams and Wilkins, Philadelphia; Lamb and olakofsky (2001) Paramyxoviridae: the viruses and their replication pp. 1305-1340. In; Knipe and Howley (eds.) Fields Virology vol. 1. Lippincott, Williams and Wilkins, Philadelphia). Human RSV is classified into two subgroups, subgroups A and B, based on antigenic diversity and nucleotide sequence divergence. For example, the attachment protein G is most divergent and the fusion protein F is relatively conserved between the two subgroups.
  • RSV strain A2 has been sequenced.
  • the genome of the A2 strain RSV is 15,222 nt in length and contains 10 transcriptional units that encode 11 proteins (NS1, NS2, N, P, M, SH, G, F, M2-1, M2-2, and L).
  • the genome is tightly bound by the N protein to form the nucleocapsid, which is the template for the viral RNA polymerase comprising the N, P and L proteins (Grosfeld et al. (1995) J. Virol. 69:5677-5686; Yu et al.
  • Each transcription unit is flanked by a highly conserved 10-nt gene-start (GS) signal, at which mRNA synthesis begins, and ends with a semiconserved 12- to 13-nt gene-end (GE) signal that directs polyadenylation and release of mRNAs (Harmon et al. (2001) J. Virol. 75:36-44; Kuo et al. (1996) J. Virol. 70:6892-6901). Transcription of RSV genes is sequential, and there is a gradient of decreasing mRNA synthesis due to transcription attenuation (Barik (1992) J. Virol. 66:6813-6818; Pickens et al. (1984) J. Virol. 52:364-369).
  • the viral RNA polymerase must first terminate synthesis of the upstream message in order to initiate synthesis of the downstream mRNA.
  • the nucleocapsid protein (N), phosphoprotein (P), and large polymerase protein (L) constitute the minimal components for viral RNA replication and transcription in vitro (Grosfield et al. (1995) J. Virol. 69:5677-5686; Yu et al. (1995) J. Virol. 69:2412- 2419).
  • the N protein associates with the genomic RNA to form the nucleocapsid, which serves as the template for RNA synthesis.
  • the L protein is a multifunctional protein that contains RNA-dependent RNA polymerase catalytic motifs and is also probably responsible for capping and polyadenylation of viral mRNAs.
  • M2-1 is a transcription antitermination (or elongation) factor required for processive RNA synthesis and transcription read-through at gene junctions, essential for RNA transcription and virus replication (Collins et al.
  • M2-2 though not essential for virus replication in tissue culture, is involved in the switch between viral RNA transcription and replication (Bermingham and Collins (1999) Proc. Natl. Acad. Sci. USA 96:11259-11264; Jin et al. (2000) J. Virol. 74:74-82).
  • NSl and NS2 have been shown to inhibit minigenome synthesis in vitro (Atreya et al. (1998) J. Virol. 72:1452-1461).
  • NSl, NS2, SH, M2-2 and G are accessory proteins that can be deleted from the RSV A2 strain without affecting virus viability (Bermingham and Collins (1999) Proc. Natl. Acad. Sci. USA 96:11259-11264; Jin et al. (2000) J. Virol. 74:74-82; Jin et al. (2000) "Recombinant respiratory syncytial viruses with deletions in the NSl, NS2, SH, and M2-2 genes are attenuated in vitro and in vivo" Virology 273:210-218; Bukreyev et al.
  • the G and F proteins are the two major surface antigens that elicit anti-RSV neutralizing antibodies to provide protective immunity against RSV infection and reinfection. High levels of circulating antibodies correlate with protection against RSV infections or reduction of disease severity (Crowe (1999) Microbiol. Immunol. 236:191- 214).
  • a and B two antigenic RSV subgroups have been recognized based on virus antigenic and sequence divergence (Anderson et al. (1985) J. Infect. Pis. 151:626-633; Mufson et al. (1985) J. Gen. Virol. 66:2111-2124).
  • RSV infection in children have suggested that naturally acquired infection elicits a relatively higher protection against disease caused by the homologous subgroup virus (Mclntosh and Chanock (1990) "Respiratory syncytial virus” In: P.M. Knipe et al. (Ed) Second Edition Virology, pp.1045-1072, Raven Press, Ltd., New York).
  • the immunity induced by RSV infection is transient and subsequent reinfection can occur.
  • RSV reinfection usually does not cause severe disease.
  • An RSV vaccine is therefore typically targeted to provide protection against severe lower respiratory disease caused by RSV subgroup A and B viruses.
  • Efforts to produce a safe and effective RSV vaccine have focused on the administration of purified viral antigen or the development of live attenuated RSV for intranasal administration.
  • a formalin-inactivated virus vaccine not only failed to provide protection against RSV infection, but was shown to exacerbate symptoms during subsequent infection by the wild-type virus in infants (Kapi ian et al. (1969) Am. J. Epidemiol. 89:405-421; Chin et al. (1969) Am. J. Epidemiol. 89:449-63).
  • this invention presents the complete polynucleotide sequence of human RSV subgroup B strain 9320.
  • Polypeptides encoded by the B9320 genome are also provided, as are other benefits which will become apparent upon review of the disclosure.
  • the present invention provides the complete polynucleotide sequence of human respiratory syncytial virus subgroup B strain 9320. Amino acid sequences of proteins encoded by the B9320 genome are also provided.
  • the invention provides isolated or recombinant nucleic acids and polypeptides comprising the novel B9320 sequences. Isolated or recombinant RSV comprising the nucleic acids and polypeptides of the invention (e.g., attenuated recombinant RSV) are also provided, as are immunogenic compositions including such nucleic acids, polypeptides, and RSV that are suitable for use as vaccines. Recovery of infectious recombinant 9320 viruses from cPNAs is described.
  • the present invention provides isolated or recombinant nucleic acids comprising a polynucleotide sequence of the invention.
  • an isolated or recombinant nucleic acid comprising the polynucleotide sequence of SEQ ID NO: 1 or a complementary polynucleotide sequence thereof is a favored embodiment of the invention.
  • An isolated or recombinant nucleic acid comprising at least one unique polynucleotide subsequence of SEQ ID NO:l or a complementary polynucleotide sequence thereof, with the proviso that the unique polynucleotide subsequence includes at least one subsequence not included in SEQ ID NOs: 14-19 or a complementary polynucleotide sequence thereof, is another favored embodiment.
  • the unique polynucleotide subsequence can, for example, comprise at least 10 contiguous nucleotides of SEQ ID NO:l or a complementary polynucleotide sequence thereof (e.g., at least 20 contiguous nucleotides, at least 50 contiguous nucleotides, at least 100 contiguous nucleotides, at least 500 contiguous nucleotides, or even at least 1000 contiguous nucleotides).
  • the polynucleotide subsequence includes at least one compete open reading frame (ORF) of SEQ ED NO-.l.
  • polynucleotide sequences that are highly related structurally and/or functionally are polynucleotides of the invention.
  • a polynucleotide sequence that is greater than 97.8% identical to SEQ ID NO:l or a complementary polynucleotide sequence thereof, as determined by BLASTN using default parameters is a polynucleotide of the invention.
  • polynucleotide sequences of the invention include a polynucleotide sequence encoding a polypeptide of the invention, e.g., encoding an amino acid sequence or unique subsequence selected from the group consisting of SEQ ID NOs:2-l 1 or an artificial conservative variation thereof.
  • a nucleic acid of the invention optionally comprises at least one artificially mutated nucleotide, e.g., at least one artificially deleted, inserted, and/or substituted nucleotide.
  • mutation of the polynucleotide sequence results in alteration of an encoded amino acid sequence.
  • At least one polypeptide encoded by the nucleic acid comprises at least one deleted, inserted, and/or substituted amino acid residue (e.g., at least one conservatively or non-conservatively substituted amino acid residue).
  • the mutated nucleotide can be located in an ORF encoding a polypeptide selected from SEQ ID NOs:2-12.
  • Another class of embodiments provides vectors comprising the nucleic acids of the invention. Yet another class of embodiments provides a host cell into which such a vector has been introduced. Another class of embodiments provides methods of producing a recombinant respiratory syncytial virus. In the methods, such a host cell is cultured in a suitable culture medium under conditions permitting expression of the nucleic acid, and the recombinant respiratory syncytial virus is isolated from the host cell and/or the medium. Recombinant RSV produced according to these methods form another feature of the invention, as do recombinant RSV comprising a nucleic acid of the invention. A related class of embodiments provides methods of producing an isolated or recombinant polypeptide.
  • a host cell comprising a vector that includes a nucleic acid of the invention is cultured in a suitable culture medium under conditions permitting expression of the nucleic acid, and the polypeptide is isolated from the host cell and or the medium.
  • Polypeptides produced according to these methods form another feature of the invention, as do polypeptides comprising an amino acid sequence or subsequence that is encoded by a nucleic acid of the invention.
  • One aspect of the invention provides isolated or recombinant polypeptides comprising an amino acid sequence of the invention.
  • an isolated or recombinant polypeptide comprising an amino acid sequence selected from the group consisting of SEQ IP NOs:2-l 1 is a favored embodiment of the invention.
  • An isolated or recombinant polypeptide comprising a unique amino acid subsequence comprising at least 7 (e.g., at least 8, at least 10, at least 20, at least 50, or more) contiguous amino acid residues of any one of SEQ IP NOs:2-ll is another favored embodiment.
  • amino acid sequences or subsequences of the invention are also amino acid sequences of the invention, as are amino acid sequences that are substantially identical to an amino acid sequence of the invention.
  • amino acid sequence or subsequence that is specifically bound by an antibody that specifically binds to an amino acid sequence or subsequence encoded by SEQ ID NO:l, wherein said antibody does not specifically bind to an amino acid sequence or subsequence encoded by SEQ ID NO: 13 or SEQ ID NO: 14, is an amino acid sequence of the invention.
  • a polypeptide of the invention optionally comprises at least one artificially altered amino acid, e.g., at least one deleted, inserted, and/or substituted amino acid.
  • at least one deleted, inserted, and/or substituted amino acid e.g., one class of embodiments provides an isolated or recombinant polypeptide comprising the amino acid sequence of SEQ ID NO: 12 with a deletion of residues 164-197, or an artificial conservative variation thereof.
  • Immunogenic compositions comprising an immunologically effective amount of a recombinant respiratory syncytial virus, polypeptide, and/or nucleic acid of the invention form another aspect of the invention.
  • another feature of the invention provides methods for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus.
  • an immunologically effective amount of a respiratory syncytial virus, polypeptide, and/or nucleic acid of the invention is administered to the individual in a physiologically acceptable carrier.
  • Figure 1 schematically illustrates assembly of the full-length antigenomic
  • FIG. 1 schematically illustrates the cloning of pB-L, the assembly of the L coding region from three subclones. Positions of various subclones and primers used to assemble pB-L are indicated.
  • Figure 3 schematically illustrates cloning of the 5' portion of the RSV 9320 antigenome. Positions of various subclones and primers are indicated.
  • Figure 4 presents line graphs illustrating the growth of wild type 9320
  • Vero cells Panel A
  • HEp-2 cells Panel B
  • Vero or HEp-2 cells were infected with each virus at an m.o.i of 0.1 and incubated at 35°C for 5 days.
  • the viruses released into the culture supernatants at each day were titrated in Vero cells by plaque assay.
  • a virus includes a plurality of viruses
  • a host cell includes mixtures of host cells, and the like.
  • amino acid sequence is a polymer of amino acid residues (a protein, polypeptide, etc.) or a character string representing an amino acid polymer, depending on context.
  • a "polynucleotide sequence” or “nucleotide sequence” is a polymer of nucleotides (an oligonucleotide, a DNA, a nucleic acid, etc.) or a character string representing a nucleotide polymer, depending on context. From any specified polynucleotide sequence, either the given nucleic acid or the complementary polynucleotide sequence (e.g., the complementary nucleic acid) can be determined.
  • a “subsequence” is any portion of an entire sequence, up to and including the complete sequence. Typically, a subsequence comprises less than the full-length sequence.
  • a “unique subsequence” is a subsequence that is not found in any previously determined RSV polynucleotide or polypeptide sequence (e.g., the A2, BI, and B18537 sequences listed and/or referenced herein).
  • an "artificial mutation” is a mutation introduced by human intervention, e.g., under laboratory conditions.
  • an "artificially mutated” nucleotide is a nucleotide that has been mutated as a result of human intervention
  • an "artificially altered” amino acid residue is a residue that has been altered as a result of human intervention
  • an “artificial conservative variation” is a conservative variation that has been produced by human intervention.
  • a wild type virus e.g., one circulating naturally among human hosts
  • other parental strain of virus can be "artificially mutated” using recombinant DNA techniques to alter the viral genome (e.g., the viral genome can be altered by in vitro mutagenesis, or by exposing it to a chemical, ionizing radiation, or the like and then performing in vitro or in vivo selection for a temperature sensitive, cold sensitive, or otherwise attenuated strain of virus).
  • a wild type protein can be "artificially altered” by artificially mutating the gene encoding that protein.
  • variants refers to an amino acid sequence that is altered by one or more amino acids with respect to a reference sequence.
  • the variant can have "conservative" changes, wherein a substituted amino acid has similar structural or chemical properties, e.g., replacement of leucine with isoleucine.
  • a variant can have "nonconservative” changes, e.g., replacement of a glycine with a tryptophan.
  • Analogous minor variation can also include amino acid deletion or insertion, or both.
  • Guidance in determining which amino acid residues can be substituted, inserted, or deleted without eliminating biological or immunological activity can be found using computer programs well known in the art, for example, DNASTAR software. Examples of conservative substitutions are also described below.
  • nucleic acid or “polynucleotide” encompasses any physical string of monomer units that can be corresponded to a string of nucleotides, including a polymer of nucleotides (e.g., a typical DNA or RNA polymer), PNAs, modified oligonucleotides (e.g., oligonucleotides comprising bases that are not typical to biological RNA or DNA in solution, such as 2'-O-methylated oligonucleotides), and the like.
  • a nucleic acid can be e.g., single-stranded or double-stranded. Unless otherwise indicated, a particular nucleic acid sequence of this invention encompasses complementary sequences, in addition to the sequence explicitly indicated.
  • genes include coding sequences and/or the regulatory sequences required for their expression.
  • the term “gene” applies to a specific genomic sequence, as well as to a cDNA or an mRNA encoded by that genomic sequence.
  • Genes also 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.
  • “Expression of a gene” or “expression of a nucleic acid” means transcription of DNA into RNA (optionally including modification of the RNA, e.g., splicing), translation of RNA into a polypeptide (possibly including subsequent modification of the polypeptide, e.g., posttranslational modification), or both transcription and translation, as indicated by the context.
  • vector refers to the means by which a nucleic acid 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.
  • the vectors of the present invention are plasmids.
  • An "expression vector” is a vector, such as a plasmid, which is capable of promoting expression as well as replication of a nucleic acid incorporated therein.
  • the nucleic acid to be expressed is “operably linked” to a promoter and/or enhancer, and is subject to transcription regulatory control by the promoter and/or enhancer.
  • host cell means a cell which contains a heterologous nucleic acid, such as a vector, and 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.
  • Exemplary host cells in the context of the invention include HEp-2 cells and Vero cells.
  • the term "introduced" when referring to a heterologous or isolated nucleic acid refers to the transfer of a nucleic acid into a eukaryotic or prokaryotic cell where the nucleic acid can be incorporated into the genome of the cell (e.g., chromosome, plasmid, plastid or mitochondrial DNA), converted into an autonomous replicon, or transiently expressed (e.g., transfected mRNA).
  • the term includes such methods as “infection,” “transfection,” “transformation” and “transduction.”
  • methods can be employed to introduce nucleic acids into host cells, including electroporation, calcium phosphate precipitation, lipid mediated transfection (lipofection), etc.
  • ORF is a possible translational reading frame of DNA or RNA (e.g., of a gene), which is capable of being translated into a polypeptide. That is, the reading frame is not interrupted by stop codons.
  • ORF does not necessarily indicate that the polynucleotide is, in fact, translated into a polypeptide.
  • a "polypeptide” is a polymer comprising two or more amino acid residues
  • polypeptide e.g., a peptide or a protein
  • the polymer can optionally comprise modifications such as glycosylation or the like.
  • the amino acid residues of the polypeptide can be natural or non- natural and can be unsubstituted, unmodified, substituted or modified.
  • the term "recombinant” indicates that the material (e.g., a virus, a nucleic acid, or a protein) 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.
  • a "recombinant nucleic acid” is one that is made by recombining nucleic acids, e.g., during cloning, DNA shuffling or other procedures, or by chemical or other mutagenesis
  • a "recombinant polypeptide” or “recombinant protein” is a polypeptide or protein which is produced by expression of a recombinant nucleic acid
  • a "recombinant virus”, e.g., a recombinant respiratory syncytial virus is produced by the expression of a recombinant nucleic acid.
  • isolated refers to a biological material, such as a virus, a nucleic acid or a protein, which is substantially free from components that normally accompany or interact with it in its naturally occurring environment.
  • the isolated material optionally comprises material not found with the material in its natural environment, e.g., a cell. For example, if the material is in its natural environment, such as a cell, the material has been placed at a location in the cell (e.g., genome or genetic element) not native to a material found in that environment.
  • a naturally occurring nucleic acid e.g., a coding sequence, a promoter, an enhancer, etc.
  • a locus of the genome e.g., a vector, such as a plasmid or virus vector, or amplicon
  • Such nucleic acids are also referred to as "heterologous" nucleic acids.
  • An isolated virus for example, is in an environment (e.g., a cell culture system, or purified from cell culture) other than the native environment of wild-type virus (e.g., the nasopharynx of an infected individual).
  • chimeric or “chimera” when referring to a virus, indicates that the virus includes genetic and/or polypeptide components derived from more than one parental viral strain or source.
  • chimeric or “chimera” when referring to a viral protein, indicates that the protein includes polypeptide components (i.e., amino acid subsequences) derived from more than one parental viral strain or source.
  • An RSV "having an attenuated phenotype" or an "attenuated” RSV exhibits a substantially lower degree of virulence as compared to a wild-type virus (e.g., one circulating naturally among human hosts).
  • An attenuated RSV typically exhibits a slower growth rate and or a reduced level of replication (e.g., a peak titer, e.g., in cell culture, in an animal model of infection, or in a human vacinee's nasopharynx, that is at least about ten fold, preferably at least about one hundred fold, less than that of a wild-type RSV).
  • An "immunologically effective amount" of RSV is an amount sufficient to enhance an individual's (e.g., a human's) own immune response against a subsequent exposure to RSV.
  • Levels of induced immunity can be monitored, e.g., by measuring amounts of neutralizing secretory and/or serum antibodies, e.g., by plaque neutralization, complement fixation, enzyme-linked immunosorbent, or microneutralization assay.
  • a "protective immune response" against RSV refers to an immune response exhibited by an individual (e.g., a human) that is protective against serious lower respiratory tract disease (e.g., pneumonia and/or bronchiolitis) when the individual is subsequently exposed to and/or infected with wild-type RSV.
  • the wild-type (e.g., naturally circulating) RSV can still cause infection, particularly in the upper respiratory tract (e.g., rhinitis), but it can not cause a serious infection.
  • the protective immune response results in detectable levels of host engendered serum and secretory antibodies that are capable of neutralizing virus of the same strain and/or subgroup (and possibly also of a different, non-vaccine strain and/or subgroup) in vitro and in vivo.
  • an "antibody” is a protein comprising one or more polypeptides substantially or partially encoded by immunoglobulin genes or fragments of immunoglobulin genes.
  • the recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.
  • a typical immunoglobulin (antibody) structural unit comprises a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light” (about 25 kD) and one "heavy” chain (about 50-70 kD).
  • the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively.
  • Antibodies exist as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases.
  • pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)'2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond.
  • the F(ab)'2 may be reduced under mild conditions to break the disulfide linkage in the hinge region thereby converting the (Fab')2 dimer into a Fab' monomer.
  • the Fab' monomer is essentially a Fab with part of the hinge region (see, Fundamental Immunology, W.E. Paul, ed., Raven Press, N.Y. (1999), for a more detailed description of other antibody fragments).
  • antibody includes antibodies or fragments either produced by the modification of whole antibodies or synthesized de novo using recombinant DNA methodologies.
  • Antibodies include, e.g., polyclonal antibodies, monoclonal antibodies, multiple or single chain antibodies, including single chain Fv (sFv or scFv) antibodies in which a variable heavy and a variable light chain are joined together (directly or through a peptide linker) to form a continuous polypeptide, and humanized or chimeric antibodies.
  • an "antigenome” is a polynucleotide that is complementary (typically, perfectly complementary) to a single-stranded viral (e.g., RSV) genome. Since RSV is a negative-sense RNA virus, the genome is the “antisense” strand, and the antigenome is the “sense” strand that corresponds to mRNA.
  • the present invention provides the complete polynucleotide sequence of human RSV subgroup B strain 9320.
  • the sequence of a B9320 antigenomic cDNA is listed as SEQ ID NO:l.
  • the RSV genome is an RNA with a polynucleotide sequence complementary to that of SEQ ID NO:l.
  • the B9320 genome comprises 10 transcriptional units encoding 11 proteins.
  • NSl is listed as SEQ ID NO:2, NS2 as SEQ ID NO:3, N as SEQ ID NO:4, P as SEQ ID NO:5, M as SEQ ID NO:6, SH as SEQ ID NO:7, G as SEQ ID NO: 12, F as SEQ ID NO:8, M2-1 as SEQ ID NO:9, M2-2 as SEQ ID NO: 10, and L as SEQ ID NO:ll.
  • the invention provides isolated or recombinant polynucleotides and polypeptides comprising the novel B9320 sequences.
  • Recombinant RSV comprising the nucleic acids and or polypeptides (e.g., attenuated recombinant RSV suitable for use in attenuated live viral vaccines) are also provided.
  • One aspect of the present invention provides isolated or recombinant nucleic acids comprising a polynucleotide sequence of the invention.
  • Polynucleotide sequences of the invention include the polynucleotide sequence represented by SEQ ID NO: 1, with the caveat that SEQ ID NOs: 14-19, representing limited subsequences of RSV B9320, have been previously described (e.g., in GenBank accession numbers M73544 and S75820; Jin et al. (1998) Virology 251:206-214; and Cheng et al. (2001) Virology 283:59-68).
  • an isolated or recombinant nucleic acid comprising the polynucleotide sequence of SEQ ID NO:l or a complementary polynucleotide sequence thereof is a favored embodiment of the invention.
  • An isolated or recombinant nucleic acid comprising at least one unique polynucleotide subsequence of SEQ ID NO:l (e.g., a unique coding subsequence) or a complementary polynucleotide sequence thereof, with the proviso that the unique polynucleotide subsequence includes at least one subsequence not included in SEQ ID NOs: 14-19 or a complementary polynucleotide sequence thereof, is another favored embodiment.
  • the unique polynucleotide subsequence can, for example, comprise at least 10 contiguous nucleotides of SEQ ID NO:l or a complementary polynucleotide sequence thereof (e.g., at least 20 contiguous nucleotides, at least 50 contiguous nucleotides, at least 100 contiguous nucleotides, at least 500 contiguous nucleotides, or even at least 1000 contiguous nucleotides).
  • polynucleotide sequences that are highly related structurally and/or functionally are polynucleotides of the invention.
  • polynucleotide sequences of the invention include a polynucleotide sequence that hybridizes under stringent conditions over substantially the entire length of the polynucleotide sequence of SEQ ID NO: 1 (or a complementary sequence thereof) with at least 2x a signal to noise ratio (e.g., at least 5x or at least lOx the signal to noise ratio) that the polynucleotide sequence hybridizes to the polynucleotide sequence of SEQ ID NO: 13 or a complementary polynucleotide sequence thereof.
  • a signal to noise ratio e.g., at least 5x or at least lOx the signal to noise ratio
  • Polynucleotide sequences of the invention also include a polynucleotide sequence that hybridizes under stringent conditions over substantially the entire length of a polynucleotide subsequence comprising at least 100 contiguous nucleotides of SEQ ID NO:l or its complementary sequence (e.g., a unique subsequence) with at least 2x a signal to noise ratio (e.g., at least 5x or at least lOx the signal to noise ratio) that the polynucleotide sequence hybridizes to the corresponding subsequence of SEQ ID NO: 13 or a complementary polynucleotide sequence thereof (or, optionally, the corresponding subsequence of a genome of another naturally occurring respiratory syncytial virus or a complementary polynucleotide sequence thereof).
  • a signal to noise ratio e.g., at least 5x or at least lOx the signal to noise ratio
  • polynucleotide sequences of the invention include a polynucleotide sequence encoding an amino acid sequence or unique subsequence selected from the group consisting of SEQ ID NOs:2-ll or an artificial conservative variation thereof.
  • polynucleotide sequences that are substantially identical to a polynucleotide of the invention can be used in the compositions and methods of the invention.
  • Substantially identical or substantially similar polynucleotide sequences are defined as polynucleotide sequences that are identical, on a nucleotide by nucleotide basis, with at least a subsequence of a reference polynucleotide, e.g., selected from SEQ ID NO:l.
  • Such polynucleotides can include, e.g., insertions, deletions, and substitutions relative to SEQ ID NO:l.
  • isolated or recombinant nucleic acids comprising polynucleotide sequences (or subsequences) having greater than 97.8% sequence identity to SEQ ID NO:l or a complementary polynucleotide sequence thereof, as determined by BLASTN using default parameters, with the proviso that the polynucleotide sequence includes at least one subsequence not selected from SEQ IP NOs: 14-19, are favored embodiments of the invention.
  • the polynucleotide sequences (or subsequences) can be at least 98.5% (e.g., at least 99.0%, at least 99.5%, or more) identical to SEQ IP NO:l or a complementary polynucleotide sequence thereof.
  • the nucleic acids of the invention can be, e.g., single-stranded or double- stranded, and can be, e.g., a PNA (e.g., a cPNA), an RNA, or an artificial nucleic acid (e.g., a peptide nucleic acid).
  • SEQ IP NO: 1 presents the PNA sequence of the antigenomic B9320 cPNA; however, it will be understood that the complementary genomic polynucleotide sequence can readily be determined from SEQ ID NO:l and that U in an RNA sequence corresponds to T in a DNA sequence.
  • Nucleic acids of the invention include nucleic acids encoding polypeptides of the invention.
  • the nucleic acid comprises at least one unique polynucleotide subsequence of SEQ ID NO:l (or a complementary polynucleotide sequence thereof) encoding at least 20 contiguous amino acid residues of any one of SEQ ID NOs:2-12 (e.g., at least 50, at least 100, at least 200, or more contiguous amino acid residues).
  • the unique polynucleotide subsequence comprises at least one complete ORF, preferably at least one complete ORF encoding a polypeptide selected from among SEQ ID NOs: 2-12.
  • the nucleic acid comprises a plurality of complete open reading frames.
  • a nucleic acid of the invention optionally comprises at least one artificially mutated nucleotide, e.g., at least one artificially deleted, inserted, and/or substituted nucleotide (e.g., in a noncoding region, e.g., a C to G change at the fourth position of the antigenomic sequence, and or in a coding region).
  • the nucleic acid can comprise a plurality of artificially mutated nucleotides.
  • the artificially mutated nucleotide(s) can be introduced by site-directed mutagenesis, chemical mutagenesis, or the like.
  • mutation of the polynucleotide sequence results in alteration of an encoded amino acid sequence.
  • at least one polypeptide encoded by the nucleic acid comprises at least one deleted, inserted, and or substituted amino acid residue (e.g., at least one conservatively or non-conservatively substituted amino acid residue).
  • the mutated nucleotide can be located in an ORF encoding a polypeptide selected from SEQ ID NOs:2-12.
  • the at least one artificially mutated nucleotide is located in the open reading frame encoding the polypeptide of SEQ ID NO: 12.
  • the artificially mutated nucleotide(s) can comprise, e.g., a deletion, e.g., a deletion resulting in a deletion of one or more amino acid residues from the G protein encoded by SEQ ID NO: 12 (e.g., a deletion of residues 164-197), or a deletion resulting in a deletion of the open reading frame encoding G.
  • the at least one artificially mutated nucleotide is located in the open reading frame encoding the polypeptide of SEQ ID NO: 10.
  • the artificially mutated nucleotide(s) can comprise, e.g., a deletion, e.g., a deletion resulting in a deletion of one or more amino acid residues from the M2-2 protein encoded by SEQ ID NO: 10, or a deletion resulting in a deletion of the open reading frame encoding M2-2.
  • at least one of the nucleotides encoding amino acid residue 1, amino acid residue 4 and or amino acid residue 10 of M2-2 can be mutated (e.g., substituted or deleted, e.g., forcing use of the second and or third start codon and resulting in a deletion of amino acid residues 1-3 or 1-9 of M2-2).
  • the nucleic acids of the invention include chimeric nucleic acids, for example, a nucleic acid comprising at least one subsequence of SEQ ID NO:l or a complementary polynucleotide sequence thereof and at least one polynucleotide subsequence from a different strain of virus.
  • the subsequence of SEQ ID NO:l is preferably a unique polynucleotide subsequence that comprises at least 10 contiguous nucleotides of SEQ ID NO:l or its complement and that includes at least one subsequence not included in SEQ ID NOs: 14-19 or a complementary polynucleotide sequence thereof.
  • the different strain of virus can be, e.g., a different strain of human RSV (e.g., A2, BI, or the like) or a different species of virus (e.g., another paramyxovirus, e.g., pneumonia virus of mice, bovine RSV, or metapneumo virus).
  • chimeric nucleic acids can, for example, encode chimeric proteins and/or chimeric viruses (e.g., for use in vaccines to induce a protective immune response against one or more strains of RSV and/or another virus).
  • the nucleic acid comprises at least one complete open reading frame of SEQ ID NO:l and at least one complete open reading frame of the different strain of virus.
  • Another class of embodiments provides vectors comprising the nucleic acids of the invention. Yet another class of embodiments provides a host cell into which such a vector has been introduced.
  • RSV B9320 polypeptides and variants thereof for example, a polypeptide comprising an amino acid sequence or subsequence that is encoded by a nucleic acid of the invention, with the proviso that the amino acid sequence or subsequence is not encoded by SEQ ID NO: 14.
  • One general class of embodiments provides isolated or recombinant polypeptides comprising an amino acid sequence of the invention.
  • an isolated or recombinant polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:2-l 1 is a favored embodiment of the invention.
  • An isolated or recombinant polypeptide comprising a unique amino acid subsequence comprising at least 7 (e.g., at least 8, at least 10, at least 20, at least 50, or more) contiguous amino acid residues of any one of SEQ IP NOs:2-ll is another favored embodiment.
  • Artificial conservative variations of amino acid sequences or subsequences of the invention are also amino acid sequences of the invention.
  • Such polypeptides are optionally immunogenic.
  • amino acid sequences that are substantially identical to an amino acid sequence of the invention can be used in the compositions and methods of the invention.
  • Substantially identical or substantially similar polypeptide sequences are defined as amino acid sequences that are identical, on an amino acid by amino acid basis, with at least a subsequence of a reference polypeptide, e.g., selected from among SEQ ID NOs:2-ll.
  • Such amino acid sequences can include, e.g., insertions, deletions, and substitutions relative to SEQ ID NOs:2-ll.
  • an isolated or recombinant polypeptide comprising an amino acid sequence that is greater than 99.3% identical to SEQ ID NO:2, an amino acid sequence that is greater than 98.4% identical to SEQ ID NO:3, an amino acid sequence that is greater than 99.7% identical to SEQ ID NO:4, an amino acid sequence that is greater than 98.3% identical to SEQ ID NO:5, an amino acid sequence that is greater than 99.6% identical to SEQ ID NO:6, an amino acid sequence that is greater than 97.0% identical to SEQ ID NO:7, an amino acid sequence that is greater than 99.3% identical to SEQ ID NO:8, an amino acid sequence that is greater than 99.5% identical to SEQ ID NO:9, an amino acid sequence that is greater than 96.4% identical to SEQ ID NO: 10, or an amino acid sequence that is greater than 99.2% identical to SEQ ID NO: 11, as determined by BLASTP using default parameters, is a favored embodiment of the invention.
  • the isolated or recombinant polypeptide can comprise an amino acid sequence (or subsequence) that is at least 99.5% identical to SEQ ID NO:2, at least 98.6% identical to SEQ ID NO:3, at least 99.9% identical to SEQ ID NO:4, at least 98.5% identical to SEQ ID NO:5, at least 99.8% identical to SEQ ID NO:6, at least 97.2% identical to SEQ ID NO:7, at least 99.5% identical to SEQ ID NO:8, at least 99.7% identical to SEQ ID NO:9, at least 96.6% identical to SEQ ID NO: 10, or at least 99.4% identical to SEQ ID NO: 11, as determined by BLASTP using default parameters.
  • a polypeptide of the invention optionally comprises at least one artificially altered amino acid, e.g., at least one deleted, inserted, and/or substituted amino acid.
  • the polypeptide can comprise a plurality of artificially altered amino acids.
  • One class of embodiments provides an isolated or recombinant polypeptide comprising the amino acid sequence of SEQ ID NO: 12 with a deletion of residues 164-197, or an artificial conservative variation thereof.
  • Methods of producing isolated or recombinant polypeptides form another aspect of the invention.
  • a host cell into which a vector e.g., an expression vector, e.g., an expression vector comprising one or more ORFs, or a vector comprising an entire viral genome or antigenome
  • a vector e.g., an expression vector, e.g., an expression vector comprising one or more ORFs, or a vector comprising an entire viral genome or antigenome
  • the polypeptide is then isolated from the host cell and/or the medium.
  • the polypeptide can be purified from the host cell and/or the medium such that the resulting purified polypeptide is enriched at least 5x as compared to its initial state.
  • polypeptides produced according to the methods described herein are also features of the invention.
  • Such polypeptides can comprise, e.g., subsequences (e.g., unique subsequences, immunogenic subsequences, etc.) of SEQ ID NOs:2-ll from a few amino acids (e.g., 7 or more, 10 or more, or 20 or more) up to the full length proteins.
  • a variety of methods for determining relationships (e.g., identity, similarity and/or homology) between two or more sequences, such as SEQ ID NO:l and SEQ ID NO.T3, are available and well known in the art.
  • the methods include manual alignment, computer assisted sequence alignment, and combinations thereof.
  • a number of algorithms (which are generally computer implemented) for performing sequence alignment are widely available, or can be produced by one of skill. These methods include, e.g., the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math. 2:482; the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443; the search for similarity method of Pearson and Lipman (1988) Proc. Natl. Acad. Sci.
  • HSPs high scoring sequence pairs
  • the word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always ⁇ 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • W wordlength
  • E expectation
  • BLASTP BLAST Protein
  • W wordlength
  • E expectation
  • BLOSUM62 scoring matrix see, Henikoff and Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915.
  • the BLAST algorithm performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Nat'l. Acad. Sci. USA 90:5873-5787).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • a nucleic acid is considered similar to a reference sequence (and, therefore, in this context, homologous) if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, or less than about 0.01, and or even less than about 0.001.
  • PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments. It can also plot a tree showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng and Doolittle (1987) J. Mol. Evol. 35:351-360. The method used is similar to the method described by Higgins and Sharp (1989) CAB/OS5:151-153. The program can align, e.g., up to 300 sequences of a maximum length of 5,000 letters. The multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences.
  • This cluster can then be aligned to the next most related sequence or cluster of aligned sequences.
  • Two clusters of sequences can be aligned by a simple extension of the pairwise alignment of two individual sequences. The final alignment is achieved by a series of progressive, pairwise alignments.
  • the program can also be used to plot a dendogram or tree representation of clustering relationships. The program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison.
  • nucleic acids can also be evaluated by "hybridization" between single stranded (or single stranded regions of) nucleic acids with complementary or partially complementary polynucleotide sequences.
  • Hybridization is a measure of the physical association between nucleic acids, typically, in solution, or with one of the nucleic acid strands immobilized on a solid support, e.g., a membrane, a bead, a chip, a filter, etc.
  • Nucleic acid hybridization occurs based on a variety of well characterized physico-chemical forces, such as hydrogen bonding, solvent exclusion, base stacking and the like. Numerous protocols for nucleic acid hybridization are well known in the art.
  • Conditions suitable for obtaining hybridization are selected according to the theoretical melting temperature (T m ) between complementary and partially complementary nucleic acids.
  • T m the theoretical melting temperature between complementary and partially complementary nucleic acids.
  • the T m is the temperature at which the duplex between the hybridizing nucleic acid strands is 50% denatured. That is, the T m corresponds to the temperature corresponding to the midpoint in transition from helix to random coil; it depends on length, nucleotide composition, and ionic strength for long stretches of nucleotides.
  • unhybridized nucleic acids can be removed by a series of washes, the stringency of which can be adjusted depending upon the desired results.
  • Low stringency washing conditions e.g., using higher salt and lower temperature
  • Higher stringency conditions e.g., using lower salt and higher temperature that is closer to the T m
  • lower the background signal typically with primarily the specific signal remaining. See, also, Rapley, R. and Walker, J.M. eds., Molecular Biomethods Handbook (Humana Press, Inc. 1998).
  • Stringent hybridization wash conditions or “stringent conditions” in the context of nucleic acid hybridization experiments, such as Southern and northern hybridizations, are sequence dependent, and are different under different environmental parameters. An extensive guide to the hybridization of nucleic acids is found in Tijssen (1993), supra, and in Hames and Higgins 1 and Hames and Higgins 2, supra.
  • An example of stringent hybridization conditions for hybridization of complementary nucleic acids which have more than 100 complementary residues on a filter in a Southern or northern blot is 2x SSC, 50% formamide at 42°C, with the hybridization being carried out overnight (e.g., for approximately 20 hours).
  • stringent wash conditions is a 0.2x SSC wash at 65°C for 15 minutes (see Sambrook, supra for a description of SSC buffer). Often, the wash determining the stringency is preceded by a low stringency wash to remove signal due to residual unhybridized probe.
  • An example low stringency wash is 2x SSC at room temperature (e.g., 20°C for 15 minutes).
  • a signal to noise ratio of at least 2x (or higher, e.g., at least 5x, lOx, 20x, 50x, lOOx, or more) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization.
  • Detection of at least stringent hybridization between two sequences in the context of the present invention indicates relatively strong structural similarity to, e.g., the nucleic acids of the present invention provided in the sequence listings herein.
  • highly stringent hybridization and wash conditions are selected to be about 5° C or less lower than the thermal melting point (T m ) for the specific sequence at a defined ionic strength and pH (as noted below, highly stringent conditions can also be referred to in comparative terms).
  • T m thermal melting point
  • target sequences that are closely related or identical to the nucleotide sequence of interest e.g., "probe”
  • T m thermal melting point
  • the hybridization and wash conditions are gradually increased (e.g., by increasing temperature, decreasing salt concentration, increasing detergent concentration and/or increasing the concentration of organic solvents, such as formamide, in the hybridization or wash), until a selected set of criteria are met.
  • the hybridization and wash conditions are gradually increased until a probe comprising one or more polynucleotide sequences of the invention, e.g., sequences or unique subsequences selected from SEQ ID NO:l and/or complementary polynucleotide sequences, binds to a perfectly matched complementary target (again, a nucleic acid comprising one or more nucleic acid sequences or subsequences selected from SEQ ID NO:l and/or complementary polynucleotide sequences thereof), with a signal to noise ratio that is at least 2x (and optionally 5x, lOx, or lOOx or more) as high as that observed for hybridization of the probe to an unmatched target (e.g., a polynucleotide sequence comprising the corresponding one or more sequences or subsequences selected from SEQ ID NO: 13 and/or complementary polynucleotide sequences thereof), as desired.
  • a probe comprising one or more polynucleo
  • target nucleic acids can be obtained; such target nucleic acids are also a feature of the invention.
  • target nucleic acids include sequences that hybridize under stringent conditions to a unique oligonucleotide probe corresponding to any of the polynucleotides of the invention, e.g., SEQ ID NO:l.
  • Nucleic acids which do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides which they encode are substantially identical. This occurs, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code.
  • polypeptides of the invention provide a variety of new polypeptide sequences
  • the polypeptides also provide new structural features which can be recognized, e.g., in immunological assays.
  • the generation of antibodies or antisera which specifically bind the polypeptides of the invention, as well as the polypeptides which are bound by such antibodies or antisera, ⁇ and the antibodies or antisera themselves, are a feature of the invention.
  • proteins of the invention can also be identified by immunoreactivity; e.g., a polypeptide of the invention can include an amino acid sequence or subsequence that is specifically bound by an antibody that specifically binds to an amino acid sequence or subsequence encoded by SEQ ID NO:l, wherein the antibody does not specifically bind to an amino acid sequence or subsequence encoded by SEQ ID NO: 13 or SEQ ID NO: 14 (or, optionally, to an amino acid sequence or subsequence encoded by the genome of another naturally occurring respiratory syncytial virus).
  • an immunoassay to identify a polypeptide of the invention uses a polyclonal antiserum which was raised against one or more of the RSV 9320 polypeptides of the invention (e.g., a polypeptide comprising SEQ ID NOs:2-ll or SEQ ID NO: 12 with a deletion of residues 164-197), or a substantial subsequence thereof (i.e., at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 98% or more of the full length sequence provided).
  • a polyclonal antiserum which was raised against one or more of the RSV 9320 polypeptides of the invention (e.g., a polypeptide comprising SEQ ID NOs:2-ll or SEQ ID NO: 12 with a deletion of residues 164-197), or a substantial subsequence thereof (i.e., at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 98% or more of the full length sequence
  • the full set of potential polypeptide immunogens derived from one or more of the RSV 9320 polypeptides of the invention are collectively referred to below as "the immunogenic polypeptides.”
  • the resulting antisera is optionally selected to have low cross-reactivity against the control RSV BI polypeptides and/or other known, e.g., naturally occurring, RSV polypeptides, and any such cross-reactivity is removed by immunoabsorption with one or more of the control RSV polypeptides, prior to use of the polyclonal antiserum in the immunoassay.
  • one or more of the immunogenic polypeptides is produced and purified as described herein.
  • recombinant protein can be produced in a mammalian cell line.
  • An inbred strain of mice (used in this assay because results are more reproducible due to the virtual genetic identity of the mice) is immunized with the immunogenic polypeptide(s) in combination with a standard adjuvant, such as Freund's adjuvant, and a standard mouse immunization protocol (see Harlow and Lane (1988) Antibodies, A Laboratory Manual Cold Spring Harbor Press, New York, for a standard description of antibody generation, immunoassay formats and conditions that can be used to determine specific immunoreactivity).
  • one or more synthetic or recombinant polypeptides derived from the sequences disclosed herein is conjugated to a carrier protein and used as an immunogen.
  • Polyclonal sera are collected and titered against the immunogenic poly ⁇ eptide(s) in an immunoassay, for example, a solid phase immunoassay with one or more of the immunogenic polypeptides immobilized on a solid support.
  • Polyclonal antisera with a titer of 10 6 or greater are selected, pooled and subtracted with the control RSV polypeptides to produce subtracted pooled titered polyclonal antisera.
  • the subtracted pooled titered polyclonal antisera are tested for cross reactivity against the control RSV polypeptides.
  • Preferably at least two of the immunogenic RSV 9320 polypeptides are used in this determination, preferably in conjunction with at least two of the control RSV polypeptides, to identify antibodies which are specifically bound by the immunogenic polypeptides(s).
  • discriminatory binding conditions are determined for the subtracted titered polyclonal antisera which result in at least about a 5-10 fold higher signal to noise ratio for binding of the titered polyclonal antisera to the immunogenic RSV 9320 polypeptides as compared to binding to the control RSV polypeptides. That is, the stringency of the binding reaction is adjusted by the addition of non-specific competitors, such as albumin or non-fat dry milk, or by adjusting salt conditions, temperature, or the like. These binding conditions are used in subsequent assays for determining whether a test polypeptide is specifically bound by the pooled subtracted polyclonal antisera.
  • a test polypeptide which shows at least a 2-5x higher signal to noise ratio than the control polypeptides under discriminatory binding conditions, and at least about a V_ signal to noise ratio as compared to the immunogenic polypeptide(s), shares substantial structural similarity or homology with the immunogenic polypeptide(s) as compared to the control RSV polypeptides, and is, therefore, a polypeptide of the invention.
  • immunoassays in the competitive binding format are used for detection of a test polypeptide.
  • cross-reacting antibodies are removed from the pooled antisera mixture by immunoabsorption with the control RSV polypeptides.
  • the immunogenic polypeptide(s) are then immobilized to a solid support which is exposed to the subtracted pooled antisera.
  • Test proteins are added to the assay to compete for binding to the pooled subtracted antisera.
  • test protein(s) The ability of the test protein(s) to compete for binding to the pooled subtracted antisera as compared to the immobilized protein(s) is compared to the ability of the immunogenic polypeptide(s) added to the assay to compete for binding (the immunogenic polypeptides compete effectively with the immobilized immunogenic polypeptides for binding to the pooled antisera).
  • the percent cross-reactivity for the test proteins is calculated, using standard calculations.
  • the ability of the control proteins to compete for binding to the pooled subtracted antisera is determined as compared to the ability of the immunogenic polypeptide(s) to compete for binding to the antisera. Again, the percent cross-reactivity for the control polypeptides is calculated, using standard calculations. Where the percent cross-reactivity is at least 5-10x as high for the test polypeptides, the test polypeptides are said to specifically bind the pooled subtracted antisera, and are, therefore, polypeptides of the invention.
  • the immunoabsorbed and pooled antisera can be used in a competitive binding immunoassay as described herein to compare any test polypeptide to the immunogenic polypeptide(s).
  • the two polypeptides are each assayed at a wide range of concentrations and the amount of each polypeptide required to inhibit 50% of the binding of the subtracted antisera to the immobilized protein is determined using standard techniques. If the amount of the test polypeptide required is less than twice the amount of the immunogenic polypeptide that is required, then the test polypeptide is said to specifically bind to an antibody generated to the immunogenic polypeptide, provided the amount is at least about 5-1 Ox as high as for a control polypeptide.
  • the pooled antisera is optionally fully immunosorbed with the immunogenic polype ⁇ tide(s) (rather than the control polypeptides) until little or no binding of the resulting immunogenic polypeptide subtracted pooled antisera to the immunogenic polypeptide(s) used in the immunoabsorption is detectable.
  • This fully immunosorbed antisera is then tested for reactivity with the test polypeptide. If little or no reactivity is observed (i.e., no more than 2x the signal to noise ratio observed for binding of the fully immunosorbed antisera to the immunogenic polypeptide), then the test polypeptide is specifically bound by the antisera elicited by the immunogenic protein.
  • any of a variety of nucleic acid sequences encoding polypeptides and/or viruses of the invention are optionally produced, some which can bear lower levels of sequence identity to the RSV nucleic acid and polypeptide sequences in the figures.
  • the following provides a typical codon table specifying the genetic code, found in many biology and biochemistry texts.
  • the codon table shows that many amino acids are encoded by more than one codon.
  • the codons AGA, AGG, CGA, CGC, CGG, and CGU all encode the amino acid arginine.
  • the codon can be altered to any of the corresponding codons described above without altering the encoded polypeptide. It is understood that U in an RNA sequence corresponds to T in a DNA sequence.
  • a nucleic acid sequence corresponding to the amino acid sequence FEV is TTTGAAGTG.
  • a silent variation of this sequence includes TTCGAGGTA (also encoding FEV).
  • Such "silent variations” are one species of “conservatively modified variations", discussed below.
  • each codon in a nucleic acid except ATG, which is ordinarily the only codon for methionine, and TTG, which is ordinarily the only codon for tryptophan
  • TTG which is ordinarily the only codon for tryptophan
  • each silent variation of a nucleic acid which encodes a polypeptide is implicit in any described sequence. The invention, therefore, explicitly provides each and every possible variation of a nucleic acid sequence encoding a polypeptide of the invention that could be made by selecting combinations based on possible codon choices.
  • Constantly modified variations or, simply, “conservative variations” of a particular nucleic acid sequence or polypeptide are those which encode identical or essentially identical amino acid sequences.
  • One of skill will recognize that individual substitutions, deletions or additions which alter, add or delete a single amino acid or a small percentage of amino acids (typically less than 5%, more typically less than 4%, 2% or 1%) in an encoded sequence are “conservatively modified variations” where the alterations result in the deletion of an amino acid, addition of an amino acid, or substitution of an amino acid with a chemically similar amino acid.
  • “conservatively substituted variations" of a polypeptide sequence of the present invention include substitutions of a small percentage, typically less than 5%, more typically less than 2% or 1%, of the amino acids of the polypeptide sequence, with a conservatively selected amino acid of the same conservative substitution group.
  • M2-1 polypeptide in SEQ ID NO:9 will contain "conservative substitutions", e.g., according to the six groups defined above, in up to about 10 residues (i.e., about 5% of the amino acids) in the full-length polypeptide.
  • nucleic acid molecule which do not alter the encoded activity of a nucleic acid molecule, such as the addition or deletion of a non-functional sequence, an epitope tag, a polyhistidine tag, GFP, or the like, is a conservative variation of the basic nucleic acid or polypeptide.
  • nucleic acid constructs which are disclosed yield a functionally identical construct.
  • silent substitutions i.e., substitutions in a nucleic acid sequence which do not result in an alteration in an encoded polypeptide
  • conserve amino acid substitutions in which one or a few amino acids in an amino acid sequence are substituted with different amino acids with highly similar properties, are also readily identified as being highly similar to a disclosed construct.
  • conservative variations of each disclosed or claimed virus, nucleic acid or protein are a feature of the present invention.
  • Such conservative (e.g., silent) variations can be used, e.g., to produce antibodies for detection of or immunoprotection against RSV.
  • viral 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 2003) John Wiley & Sons, New York (“Ausubel”); Sambrook et al. Molecular Cloning - A Laboratory Manual (3rd Ed.), Vol. 1- 3, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 2001 (“Sambrook”), and Berger and Kimmel Guide to Molecular Cloning Techniques, Methods in Enzymology volume 152 Academic Press, Inc., San Diego, CA (“Berger”).
  • RNA polymerase mediated techniques e.g., NASBA
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • NASBA RNA polymerase mediated techniques
  • polynucleotides of the invention can be synthesized utilizing various solid-phase strategies including mononucleotide- and/or trinucleotide-based phosphoramidite coupling chemistry.
  • 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 Enzvmol 211:3.
  • any nucleic acid can be custom ordered from any of a variety of commercial sources, such as The Midland Certified Reagent Company (www.mcrc.com), The Great American Gene Company (www.genco.com), ExpressGen, Inc. (www.expressgen.com), QIAGEN (http://oligos.qiagen.com), and many others.
  • substitutions of selected amino acid residues in viral polypeptides can be accomplished by, e.g., site directed mutagenesis.
  • viral polypeptides with amino acid substitutions functionally correlated with desirable phenotypic characteristic e.g., an attenuated phenotype, cold adaptation and or temperature sensitivity
  • desirable phenotypic characteristic e.g., an attenuated phenotype, cold adaptation and or temperature sensitivity
  • a viral nucleic acid segment e.g., a cDNA
  • Methods for site directed mutagenesis are well known in the art, and are described, e.g., in Ausubel, Sambrook, and Berger, supra.
  • kits for performing site directed mutagenesis are commercially available, e.g., the ExSite and ChameleonTM site directed mutagenesis kits (Stratagene, La Jolla), and can be used according to the manufacturer's instructions to introduce, e.g., one or more nucleotide substitutions specifying one or more amino acid substitutions into an RSV polynucleotide.
  • mutagenesis Various types are optionally used in the present invention, e.g., to modify nucleic acids and encoded polypeptides and/or viruses to produce conservative or non-conservative variants. Any available mutagenesis procedure can be used. Such mutagenesis procedures optionally include selection of mutant nucleic acids and polypeptides for one or more activity of interest.
  • Procedures that can be used include, but are not limited to: site-directed point mutagenesis, random point mutagenesis, in vitro or in vivo homologous recombination (DNA shuffling), mutagenesis using uracil containing templates, oligonucleotide-directed mutagenesis, phosphorothioate-modified DNA mutagenesis, mutagenesis using gapped duplex DNA, point mismatch repair, mutagenesis using repair-deficient host strains, restriction-selection and restriction-purification, deletion mutagenesis, mutagenesis by total gene synthesis, double-strand break repair, and many others known to persons of skill.
  • mutagenesis can be guided by information known about the naturally occurring molecule or altered or mutated naturally occurring molecules, e.g., sequence, sequence comparisons, physical properties, crystal structure or the like.
  • modification is essentially random (e.g., as in classical DNA shuffling).
  • the present invention includes recombinant constructs incorporating one or more of the nucleic acid sequences described above.
  • constructs include a vector, for example, a plasmid, a cosmid, a phage, a virus, a bacterial artificial chromosome (BAG), a yeast artificial chromosome (YAC), etc., into which one or more of the polynucleotide sequences of the invention, for example, SEQ ID NO:l or subsequences thereof, e.g., including at least one ORF selected from SEQ ID NO:l, has been inserted, in a forward or reverse orientation.
  • a vector for example, a plasmid, a cosmid, a phage, a virus, a bacterial artificial chromosome (BAG), a yeast artificial chromosome (YAC), etc.
  • the inserted nucleic acid can include all or part of at least one of the polynucleotide sequences of the invention.
  • the vector is selected based on the characteristics, e.g., size of the selected polynucleotide sequence, and on the intended use, e.g., expression, amplification, etc.
  • the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art and are commercially available.
  • polypeptide (or peptide) expression products e.g., selected from SEQ ID NOs:2-ll.
  • Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, pseudorabies, adenovirus, adeno-associated virus, retroviruses, and many others, as well as viral amplicon vectors.
  • Any vector that is capable of introducing genetic material into a cell, and, if replication is desired, which is replicable in the relevant host, can be used.
  • the polynucleotide sequence commonly a subsequence of SEQ ID NO: 1, e.g., comprising an ORF, such as an ORF encoding a polypeptide (or peptide) selected from among SEQ ID NOs: 2-11 (or variants thereof, e.g., conservative variations thereof), is physically arranged in proximity and orientation to an appropriate transcription control sequence (promoter, and optionally, one or more enhancers) to direct mRNA synthesis.
  • an appropriate transcription control sequence promoter, and optionally, one or more enhancers
  • a subsequence of SEQ ID NO.T e.g., encoding a polypeptide selected from a subsequence of one of SEQ ID NOs:2-ll
  • an expression vector e.g., encoding a polypeptide selected from a subsequence of one of SEQ ID NOs:2-ll
  • the polynucleotide sequence of interest is operably linked to an appropriate transcription control sequence.
  • promoters include: LTR or SV40 promoter, E. coli lac or trp promoter, phage lambda P JL promoter, and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses.
  • the expression vector typically also contains a ribosome binding site for translation initiation, and a transcription terminator.
  • the vector optionally includes appropriate sequences for amplifying expression.
  • the expression vectors optionally comprise one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells, such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E. coli.
  • additional translation specific initiation signals can improve the efficiency of translation.
  • These signals can include, e.g., an ATG initiation codon and adjacent sequences.
  • full-length cDNA molecules or chromosomal segments including a coding sequence incorporating, e.g., a polynucleotide sequence of the invention, a translation initiation codon and associated sequence elements are inserted into the appropriate expression vector simultaneously with the polynucleotide sequence of interest. In such cases, additional translational control signals are not required.
  • exogenous translational control signals including an ATG initiation codon is provided for expression of the relevant sequence.
  • the initiation codon is put in the correct reading frame to ensure transcription of the polynucleotide sequence of interest.
  • 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 (Scharf D et al. (1994) Results Probl Cell Differ 20:125-62; Bittner et al. (1987) Methods in Enzymol 153:516-544).
  • the present invention also relates to the introduction of vectors incorporating the polynucleotides of the invention (e.g., polynucleotides including all or part of one or more ORFs selected from SEQ ID NO: 1) into host cells and the production of polypeptides of the invention, e.g., one or more polypeptide selected from SEQ ID NOs:2-ll or subsequences thereof (e.g., unique subsequences thereof) by recombinant techniques.
  • polypeptides of the invention e.g., polypeptide selected from SEQ ID NOs:2-ll or subsequences thereof (e.g., unique subsequences thereof) by recombinant techniques.
  • Recombinant and/or isolated polypeptides encoded by the polynucleotides of the invention e.g., SEQ ID NOs:2-ll, or subsequences (e.g., unique subsequences) thereof are used, for example, as antigens to produce antibodies in animal or human subjects.
  • antigenic polypeptides (or peptides) corresponding to all or part of a polypeptide represented by SEQ ID NOs:2-ll can be injected into an experimental animal to produce antibodies specific for one or more strains of RSV, as further described in the section entitled "Antibodies" below.
  • the antigenic polypeptides can be administered, e.g., as a vaccine, to human subjects to elicit an immune response specific for one or more strains of RSV.
  • an elicited immune response can be a protective immune response.
  • host cells are genetically engineered (e.g., transduced, transformed or transfected) with a vector, such as an expression vector, of this invention.
  • a vector such as an expression vector
  • the vector can be in the form of a plasmid, a viral particle, a phage, etc.
  • appropriate expression hosts include: bacterial cells, such as E.
  • coli coli, Streptomyces, and Salmonella typhimurium
  • fungal cells such as Saccharomyces cerevisiae, Pichia pastoris, and Neurospora crassa
  • insect cells such as Drosophila and Spodopterafrugiperda
  • mammalian cells such as 3T3, COS, CHO, BHK, HEK 293 or Bowes melanoma
  • plant cells etc.
  • the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants, or amplifying the inserted polynucleotide sequences.
  • the culture conditions such as temperature, pH and the like, are typically those previously used with the host cell selected for expression, and will be apparent to those skilled in the art and in the references cited herein, including, e.g., Freshney (1994) Culture of Animal Cells, a Manual of Basic Technique, third edition, Wiley- Liss, New York and the references cited therein.
  • Freshney (1994) Culture of Animal Cells, a Manual of Basic Technique, third edition, Wiley- Liss, New York and the references cited therein.
  • details regarding cell culture can be found in Payne et al.
  • a number of expression vectors can be selected depending upon the use intended for the expressed product. For example, when large quantities of a polypeptide or fragments thereof are needed for the production of antibodies, vectors which direct high level expression of fusion proteins that are readily purified are favorably employed. Such vectors include, but are not limited to, multifunctional E.
  • coli cloning and expression vectors such as BLUESCRIPT (Stratagene), in which the coding sequence of interest, e.g., a polynucleotide of the invention as described above, can be ligated into the vector in-frame with sequences for the amino-terminal translation initiating Methionine and the subsequent 7 residues of beta-galactosidase producing a catalytically active beta galactosidase fusion protein; pIN vectors (Van Heeke and Schuster (1989) J Biol Chem 264:5503-5509); pET vectors (Novagen, Madison WI); and the like.
  • BLUESCRIPT Stratagene
  • yeast Saccharomyces cerevisiae a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH can be used for production of the desired expression products.
  • constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH.
  • Exemplary vectors include those derived from SV40, retroviruses, bovine papilloma virus, vaccinia virus, other herpesviruses and adenovirus.
  • Such suitable mammalian expression vectors optionally contain a promoter to mediate transcription of foreign DNA sequences and, optionally, an enhancer.
  • Suitable promoters are known in the art and include viral promoters such as those from SV40, cytomegalovirus (CMV), Rous sarcoma virus (RSV), adenovirus (ADV), and bovine papilloma virus (BPV).
  • an enhancer is any regulatory DNA sequence that can stimulate transcription up to 1000-fold when linked to endogenous or heterologous promoters, with synthesis beginning at the normal mRNA start site. Enhancers are also active when placed upstream or downstream from the transcription initiation site, in either normal or flipped orientation, or at a distance of more than 1000 nucleotides from the promoter. See, e.g., Maniatis (1987) Science 236:1237 and Alberts (1989) Molecular Biology of the Cell, 2nd Ed. (or later). Enhancers derived from viruses may be particularly useful, because they typically have a broader host range.
  • Examples include the SV40 early gene enhancer (see Dijkema (1985) EMBO J. 4:761) and the enhancer/promoters derived from the long terminal repeat (LTR) of the RSV (see Gorman (1982) Proc. Natl. Acad. Sci. 79:6777) and from human cytomegalovirus (see Boshart (1985) Cell 41:521). Additionally, some enhancers are regulatable and become active only in the presence of an inducer, such as a hormone or metal ion (see Sassone-Corsi and Borelli (1986) Trends Genet.
  • an inducer such as a hormone or metal ion
  • the expression vector can and will typically also include a termination sequence and poly(A) addition sequences which are operably linked to the heterologous coding sequence.
  • Sequences that cause amplification of the gene may also be desirably included in the expression vector or in another vector that is co-translated with the expression vector, as are sequences which encode selectable markers.
  • Selectable markers for mammalian cells include, for example, thymidine kinase, dihydrofolate reductase (together with methotrexate as a DHFR amplifier), aminoglycoside phosphotransferase, hygromycin B phosphotransferase, asparagine synthetase, adenosine deaminase, metallothionien, and antibiotic resistant genes such as neomycin.
  • Mammalian cell lines available as hosts for expression are known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC) as well as primary cultured cells and established cell lines, including but not limited to Vero, HEp-2, 3T3, COS, CHO, HeLa, BHK, MDCK, 293, WI38, Hep G2, MRC-5, and many others.
  • ATCC American Type Culture Collection
  • Host cells containing the vectors (e.g., expression vectors) described above are also a feature of the invention.
  • the host cell can be a eukaryotic cell, such as a mammalian cell, a yeast cell, or a plant cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, electroporation, encapsulation of the polynucleotide(s) in liposomes, direct microinjection of the DNA into nuclei, or other common techniques (see, e.g., Davis, L., Dibner, M., and Battey, I. (1986) Basic Methods in Molecular Biology).
  • a host cell strain is optionally chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein in the desired fashion.
  • modifications of the protein include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation.
  • Post-translational processing which cleaves a precursor form into a mature form of the protein is sometimes important for correct insertion, folding and/or function.
  • Different host cells have specific cellular machinery and characteristic mechanisms for such post-translational activities and can be chosen to ensure the correct modification and processing of the introduced, foreign protein.
  • Host cells transformed with a nucleotide sequence encoding a polypeptide of the invention are optionally cultured under conditions suitable for the expression and recovery of the encoded protein from cell culture.
  • the protein or fragment thereof produced by a recombinant cell can be secreted, membrane-bound, or contained intracellularly, depending on the sequence and/or the vector used.
  • the selected promoter is induced if necessary by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period.
  • appropriate means e.g., temperature shift or chemical induction
  • the secreted polypeptide product is then recovered from the culture medium.
  • cells can be harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
  • Eukaryotic or microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, or other methods, which are well know to those skilled in the art.
  • Expressed polypeptides can be recovered and purified from recombinant cell cultures by any of a number of methods well known in the art, including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography (e.g., using any of the tagging systems noted herein), hydroxylapatite chromatography, and lectin chromatography. Protein refolding steps can be used, as desired, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed in the final purification steps.
  • HPLC high performance liquid chromatography
  • cell-free transcription translation systems can be employed to produce polypeptides comprising an amino acid sequence or subsequence encoded by the polynucleotides of the invention.
  • a number of suitable in vitro transcription and translation systems are commercially available. A general guide to in vitro transcription and translation protocols is found in Tymms (1995) In vitro Transcription and Translation Protocols: Methods in Molecular Biology Volume 37, Garland Publishing, NY. Cell free transcription/translation systems can be particularly useful for the production of polypeptides, including proteins for administration to human subjects.
  • polypeptides, or subsequences thereof can be produced manually or by using an automated system, by direct peptide synthesis using solid-phase techniques (see, e.g., Stewart et al. (1969) Solid-Phase Peptide Synthesis, WH Freeman Co, San Francisco; Merrifield J (1963) J. Am. Chem. Soc. 85:2149-2154).
  • automated systems include the Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer, Foster City, CA).
  • subsequences can be chemically synthesized separately, and combined using chemical methods to provide full- length polypeptides.
  • Expressed polypeptides of the invention can contain one or more modified amino acid.
  • the presence of modified amino acids can be advantageous in, for example, (a) increasing polypeptide serum half-life, (b) reducing polypeptide antigenicity or (c) increasing polypeptide storage stability.
  • Amino acid(s) are modified, for example, co- translationally or post-translationally during recombinant production (e.g., N-linked glycosylation at N-X-S/T motifs during expression in mammalian cells) or are modified by synthetic means (e.g., via PEGylation).
  • Non-limiting examples of a modified amino acid include a glycosylated amino acid, a sulfated amino acid, a prenlyated (e.g., farnesylated, geranylgeranylated) amino acid, an acetylated amino acid, an acylated amino acid, a PEG-ylated amino acid, a biotinylated amino acid, a carboxylated amino acid, a phosphorylated amino acid, and the like, as well as amino acids modified by conjugation to, e.g., lipid moieties or other organic derivatizing agents.
  • References adequate to guide one of skill in the modification of amino acids are replete throughout the literature. Example protocols are found in Walker (1998) Protein Protocols on CD-ROM Human Press, Towata, NJ. ANTIBODIES
  • polypeptides of the invention can be used to produce antibodies specific for the polypeptides comprising amino acid sequences or subsequences encoded by the polynucleotides of the invention.
  • Antibodies specific for antigenic peptides encoded by, e.g., SEQ ID NO:l (e.g., SEQ ID NOs:2-ll), and related variant polypeptides are useful, e.g., for diagnostic and therapeutic purposes, e.g., related to the activity, distribution, and expression of target polypeptides.
  • Antibodies specific for the polypeptides of the invention can be generated by methods well known in the art. Such antibodies can include, but are not limited to, polyclonal, monoclonal, chimeric, humanized, single chain, Fab fragments and fragments produced by an Fab expression library.
  • Polypeptides do not require biological activity for antibody production.
  • polypeptide or oligopeptide is antigenic.
  • Peptides used to induce specific antibodies typically have an amino acid sequence of at least about 5 amino acids, and often at least 10 or 20 amino acids.
  • Short stretches of a polypeptide, e.g., encoded by a polynucleotide of the invention such a sequence selected from SEQ ID NO:l (such as a polypeptide selected from among SEQ ID NOs:2-ll) can optionally be fused with another protein, such as keyhole limpet hemocyanin (KLH), and antibodies produced against the fusion protein or polypeptide.
  • KLH keyhole limpet hemocyanin
  • humanized antibodies are desirable for certain therapeutic applications (e.g., administration of an antibody or anitserum specific for one or more strains of RSV to provide passive immunity to a subject, e.g., a human, to prevent or decrease the severity of RSV disease).
  • a subject e.g., a human
  • humanized antibodies are desirable.
  • Detailed methods for preparation of humanized antibodies can be found in USPN 5,482,856. Additional details on humanization and other antibody production and engineering techniques can be found in Borrebaeck (ed) (1995) Antibody Engineering, 2 nd Edition Freeman and Company, NY (Borrebaeck); McCafferty et al.
  • novel nucleic acid sequences of the present invention can be used in diagnostic assays to detect RSV in a sample, to detect RSV B9320-like sequences, and to detect strain differences in clinical isolates of RSV using either chemically synthesized or recombinant RSV B9320 polynucleotide fragments, e.g., selected from SEQ ID NO:l.
  • fragments of the novel B9320 sequences comprising at least between 10 and 20 nucleotides can be used as primers to amplify nucleic acids using polymerase chain reaction (PCR) methods well known in the art (e.g., reverse transcription- PCR) and as probes in nucleic acid hybridization assays to detect target genetic material such as RSV RNA in clinical specimens.
  • PCR polymerase chain reaction
  • the novel RSV B9320 polynucleotide sequences can be used in their entirety or as fragments to detect the presence of RNA sequences or transcription products in cells, tissues, samples and the like using hybridization techniques known in the art or in conjunction with one of the methods discussed herein.
  • the probes can be either DNA or RNA molecules, such as fragments of viral RNA, isolated restriction fragments of cloned DNA, cDNAs, amplification products, transcripts, and oligonucleotides, and can vary in length from oligonucleotides as short as about 10 nucleotides in length to viral RNA fragments or cDNAs in excess of one or more kilobases.
  • a probe of the invention includes a polynucleotide sequence or subsequence (e.g., a unique subsequence) selected from SEQ ID NO:l or sequences complementary thereto.
  • the polynucleotide sequence (or subsequence) is selected from a portion of SEQ ID NO:l that includes at least a subsequence that is not included in SEQ ID NOs: 14-19.
  • polynucleotide sequences that are variants of one of the above designated sequences are used as probes. Most typically, such variants include one or a few nucleotide variations.
  • pairs (or sets) of oligonucleotides can be selected, in which the two (or more) polynucleotide sequences are substantially identical variants of each other, wherein one polynucleotide sequence or set corresponds identically to a first viral strain (e.g., B9320) and the other sequence(s) or set(s) correspond identically to additional viral strains (e.g., BI, A2, etc.).
  • a first viral strain e.g., B9320
  • additional viral strains e.g., BI, A2, etc.
  • Such pairs of oligonucleotide probes are particularly useful, for example, in the context of an allele specific hybridization experiment to determine the identity of an RSV virus or viral nucleic acid, e.g., for diagnostic or monitoring purposes.
  • probes are selected that are more or less divergent, that is probes that are at least about 70% (or 80%, 90%, 95%, 98%, or 99%) identical are selected.
  • the probes of the invention can also be used to identify additional useful polynucleotide sequences (such as to characterize additional strains of RSV) according to procedures routine in the art.
  • one or more probes, as described above are utilized to screen libraries of expression products or cloned viral nucleic acids (i.e., expression libraries or genomic libraries) to identify clones that include sequences identical to, or with significant sequence identity to SEQ ID NO:l.
  • each of these identified sequences can be used to make probes, including pairs or sets of variant probes as described above.
  • computer assisted bioinformatic approaches e.g., BLAST and other sequence homology search algorithms, and the like, can also be used for identifying related polynucleotide sequences.
  • the probes of the invention are particularly useful for detecting the presence and for determining the identity of RSV nucleic acids in cells, tissues or other biological samples (e.g., a nasal wash or bronchial lavage).
  • the probes of the invention are favorably utilized to determine whether a biological sample, such as a subject (e.g., a human subject) or model system (such as a cultured cell sample) has been exposed to, or become infected with, RSV.
  • Detection of hybridization of the selected probe to nucleic acids originating in (e.g., isolated from) the biological sample or model system is indicative of exposure to or infection with the virus (or a related virus) from which the probe polynucleotide is selected.
  • a polynucleotide sequence that hybridizes preferentially to a subsequence of SEQ ID NO:l as compared to the corresponding subsequence of SEQ ID NO: 13 (or the genome of another naturally occurring RSV strain) can be used to distinguish RSV B 9320 from RSV BI (or another RSV strain).
  • probe design is influenced by the intended application. For example, where several allele-specific probe-target interactions are to be detected in a single assay, e.g., on a single DNA chip, it is desirable to have similar melting temperatures for all of the probes. Accordingly, the length of the probes are adjusted so that the melting temperatures for all of the probes on the array are closely similar (it will be appreciated that different lengths for different probes may be needed to achieve a particular T m where different probes have different GC contents). Although melting temperature is a primary consideration in probe design, other factors are optionally used to further adjust probe construction, such as selecting against primer self-complementarity and the like.
  • probes that are polypeptides, peptides or antibodies are favorably utilized.
  • polypeptides, polypeptide fragments and peptides encoded by or having subsequences encoded by the polynucleotides of the invention e.g., SEQ ID NO:l
  • SEQ ID NO:l are favorably used to identify and isolate antibodies or other binding proteins, e.g., from phage display libraries, combinatorial libraries, polyclonal sera, and the like.
  • Antibodies specific for a polypeptide subsequence encoded by any subsequence (e.g., unique subsequence) or ORF of SEQ ID NO:l are likewise valuable as probes for evaluating expression products, e.g., from cells or tissues.
  • suitable polypeptide sequences are selected from among the amino acid sequences represented by SEQ ID NOs:2-ll.
  • antibodies are particularly suitable for evaluating expression of proteins comprising amino acid subsequences encoded by SEQ ID NO:l (e.g., SEQ ID NOs:2-ll), e.g., in a sample from a subject infected with or exposed to RSV.
  • Antibodies can be directly labeled with a detectable reagent as described below, or detected indirectly by labeling of a secondary antibody specific for the heavy chain constant region (i.e., isotype) of the specific antibody. Additional details regarding production of specific antibodies are provided above in the section entitled "Antibodies.”
  • nucleic acid and polypeptide (or peptide or antibody) probes of the invention include: 1) fluorescence (using, e.g., fluorescein, Cy-5, rhodamine or other fluorescent tags); 2) isotopic methods, e.g., using end-labeling, nick translation, random priming, or PCR to incorporate radioactive isotopes into the probe polynucleotide/oligonucleotide; 3) chemifluorescence, e.g., using alkaline phosphatase and the substrate AttoPhos (Amersham) or other substrates that produce fluorescent products; 4) chemiluminescence (e.g., using either horseradish peroxidase and/or alkaline phosphatase with substrates that produce photons as breakdown products; kits providing reagents and protocols are available from such commercial sources as Amersham, Boehringer-Mannheim, and Life Technologies/G
  • a probe can be labeled with any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical, chemical or other available means.
  • useful labels in the present invention include spectral labels such as fluorescent dyes (e.g., fluorescein isothiocyanate, Texas red, rhodamine, and the like), radiolabels (e.g., 3 H, 125 1, 35 S, 14 C, 32 P, 33 P, etc.), enzymes (e.g., horse-radish peroxidase, alkaline phosphatase, etc.), spectral colorimetric labels such as colloidal gold or colored glass or plastic (e.g.
  • the label may be coupled directly or indirectly to a component of the detection assay (e.g., a probe, such as an oligonucleotide, isolated DNA, amplicon, restriction fragment, or the like) according to methods well known in the art.
  • a component of the detection assay e.g., a probe, such as an oligonucleotide, isolated DNA, amplicon, restriction fragment, or the like
  • a detector which monitors a probe-target nucleic acid hybridization is adapted to the particular label which is used.
  • Typical detectors include spectrophotometers, phototubes and photodiodes, microscopes, scintillation counters, cameras, film and the like, as well as combinations thereof. Examples of suitable detectors are widely available from a variety of commercial sources known to persons of skill. Commonly, an optical image of a substrate comprising a nucleic acid array with particular set of probes bound to the array is digitized for subsequent computer analysis.
  • radiolabeled nucleotides Because incorporation of radiolabeled nucleotides into nucleic acids is straightforward, this detection represents one favorable labeling strategy.
  • exemplary technologies for incorporating radiolabels include end-labeling with a kinase or phosphatase enzyme, nick translation, incorporation of radio-active nucleotides with a polymerase and many other well known strategies.
  • Fluorescent labels are desirable, having the advantage of requiring fewer precautions in handling, and being amenable to high-throughput visualization techniques.
  • Preferred labels are typically characterized by one or more of the following: high sensitivity, high stability, low background, low environmental sensitivity and high specificity in labeling.
  • Fluorescent moieties which are incorporated into the labels of the invention, are generally are known, including Texas red, fluorescein isothiocyanate, rhodamine, etc.
  • Many fluorescent tags are commercially available from SIGMA chemical company (Saint Louis, MO), Molecular Probes (Eugene, OR), R&D systems (Minneapolis, MN), Pharmacia LKB Biotechnology (Piscataway, NJ), CLONTECH Laboratories, Inc.
  • moieties such as digoxygenin and biotin, which are not themselves fluorescent but are readily used in conjunction with secondary reagents, i.e., anti-digoxygenin antibodies, avidin (or streptavidin), that can be labeled, are suitable as labeling reagents in the context of the probes of the invention.
  • the label is coupled directly or indirectly to a molecule to be detected (a product, substrate, enzyme, or the like) according to methods well known in the art.
  • a molecule to be detected a product, substrate, enzyme, or the like
  • Non radioactive labels are often attached by indirect means.
  • a ligand molecule e.g., biotin
  • a nucleic acid such as a probe, primer, amplicon, or the like.
  • the ligand then binds to an anti-ligand (e.g., streptavidin) molecule which is either inherently detectable or covalently bound to a signal system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound.
  • a signal system such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound.
  • ligands and anti-ligands can be used. Where a ligand has a natural anti-ligand, for example, biotin, thyroxine, and cortisol, it can be used in conjunction with labeled anti-ligands. Alternatively, any haptenic or antigenic compound can be used in combination with an antibody.
  • Labels can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorophore or chromophore.
  • Enzymes of interest as labels will primarily be hydrolases, particularly phosphatases, esterases and glycosidases, or oxidoreductases, particularly peroxidases.
  • Fluorescent compounds include fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, etc.
  • Chemiluminescent compounds include luciferin, and 2,3-dihydrophthalazinediones, e.g., luminol. Means of detecting labels are well known to those of skill in the art.
  • means for detection include a scintillation counter or photographic film as in autoradiography.
  • typical detectors include microscopes, cameras, phototubes and photodiodes and many other detection systems which are widely available.
  • Negative strand RNA viruses can be genetically engineered and recovered using a recombinant reverse genetics approach (USPN 5,166,057 to Palese et al). Although this method was originally applied to engineer influenza viral genomes (Luytjes et al. (1989) Cell 59:1107-1113; Enami et al. (1990) Proc. Natl. Acad. Sci. USA 92:11563- 11567), it has been successfully applied to a wide variety of segmented and nonsegmented negative strand RNA viruses, e.g., rabies (Schnell et al. (1994) EMBO J. 13: 4195-4203); VSV (Lawson et al.
  • recombinant RSV incorporating the nucleic acids of this invention are generated, for example, in a suitable cell line (e.g., Vero or Hep-2 cells) by transfection of the cells with an antigenomic cDNA.
  • a suitable cell line e.g., Vero or Hep-2 cells
  • the antigenomic cDNA is flanked by a T7 RNA polymerase promoter and a hepatitis delta virus ribozyme plus the T7 transcriptional terminator.
  • Plasmids expressing the viral N, P, and L proteins are also introduced into the cells, and a T7 RNA polymerase is typically expressed in the transfected cells (e.g., by infection of the cells with a modified vaccinia virus Ankara expressing T7 RNA polymerase).
  • Recombinant RSV can also be produced by infection of suitable cells with previously isolated recombinant virus. Techniques for propagation, separation from host cell cellular components, and/or further purification of RSV are well known to those skilled in the art.
  • Methods of producing recombinant RSV are a feature of the invention.
  • a host cell into which a vector comprising a nucleic acid of the invention has been introduced is cultured in a suitable culture medium under conditions permitting expression of the nucleic acid (e.g., coexpression of RSV N, P, and L and optionally M2-1 and/or T7 RNA polymerase).
  • the recombinant respiratory syncytial virus is then isolated from the host cell and/or the medium.
  • the nucleic acid comprises an entire RSV genome or antigenome.
  • the nucleic acid can comprise a portion of an RSV genome or antigenome (e.g., one or more open reading frames encoding proteins to be assembled with an RSV genome from another source to form the recombinant virus).
  • Recombinant RSV e.g., attenuated recombinant RSV
  • recombinant RSV e.g., attenuated recombinant RSV
  • comprising one or more nucleic acids and/or polypeptides of the invention are also a feature of the invention, as are recombinant RSV (e.g., attenuated recombinant RSV) comprising one or more nucleic acids and/or polypeptides of the invention.
  • a recombinant virus e.g., recombinant RSV
  • Suitable host cells for the replication of RSV include, e.g., Vero cells and HEp-2 cells.
  • cells are cultured in a standard 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 C0 2 concentration suitable for maintaining neutral buffered pH (e.g., at pH between 7.0 and 7.2).
  • 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, ⁇ - mercaptoethanol, and the like.
  • antibiotics to prevent bacterial growth
  • additional nutrients such as L-glutamine, sodium pyruvate, non-essential amino acids
  • additional supplements to promote favorable growth characteristics, e.g., trypsin, ⁇ - mercaptoethanol, and the like.
  • Cells for production of RSV can be cultured in serum-containing or serum free medium. In some cases, e.g., for the preparation of purified viruses, it is desirable to grow the host cells in serum free conditions. For example, cells can be grown to the desired density in serum-containing medium, infected, and then maintained in serum-free medium.
  • Cells can be cultured in small scale, e.g., less than 25 ml medium, 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.
  • a single liter of medium can include more than 20 million microcarrier beads providing greater than 8000 square centimeters of growth surface.
  • Bioreactors are available in volumes from under 1 liter to in excess of 100 liters, e.g., Cyto3 Bioreactor (Osmonics, Minnetonka, MN); NBS bioreactors (New Brunswick Scientific, Edison, N.J.); and laboratory and commercial scale bioreactors from B. Braun Biotech International (B. Braun Biotech, Melsoder, Germany).
  • vectors into host cells e.g., vectors incorporating RSV polynucleotides, are introduced
  • vectors e.g., plasmids
  • host cells e.g., Vero cells or Hep-2 cells
  • electroporation can be employed to introduce vectors incorporating RSV genome segments into host cells.
  • Attenuated RSV e.g. those described herein comprising all or part of SEQ
  • ID NO:l or variations thereof can be tested in in vitro and in vivo models to confirm adequate attenuation, genetic stability, and or immunogenicity for vaccine use.
  • in vitro assays e.g., replication in cultured cells
  • the virus can be tested, e.g., for genetic stability, temperature sensitivity of virus replication and or a small plaque phenotype.
  • RSV can be further tested in animal models of infection.
  • primate e.g., chimpanzee, African green monkey
  • rodent e.g., cotton rat
  • One aspect of the invention provides immunogenic compositions (e.g., vaccines) comprising an immunologically effective amount of a recombinant RSV of the invention (e.g., an attenuated live recombinant RSV), an immunologically effective amount of a polypeptide of the invention, and or an immunologically effective amount of a nucleic acid of the invention.
  • a recombinant RSV of the invention e.g., an attenuated live recombinant RSV
  • an immunologically effective amount of a polypeptide of the invention e.g., an attenuated live recombinant RSV
  • a nucleic acid of the invention e.g., nucleic acid of the invention.
  • a related aspect of the invention provides methods for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus.
  • an immunologically effective amount of a recombinant RSV of the invention, an immunologically effective amount of a polypeptide of the invention, and/or an immunologically effective amount of a nucleic acid of the invention is administered to the individual in a physiologically acceptable carrier.
  • the RSV, polypeptides, and nucleic acids 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 RSV.
  • the carrier or excipient is a pharmaceutically acceptable carrier or excipient, such as sterile water, aqueous saline solution, aqueous buffered saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, ethanol, or combinations thereof.
  • sterile water sterile water, aqueous saline solution, aqueous buffered saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, ethanol, or combinations thereof.
  • sterile water such as sterile water, aqueous saline solution, aqueous buffered saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, ethanol, or combinations thereof.
  • 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, oral, topical, etc.
  • the resulting aqueous solutions can e.g., be packaged for use as is or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration
  • the RSV (or RSV components) of the invention are administered in a quantity sufficient to stimulate an immune response specific for one or more strains of RSV (e.g., an immunologically effective amount of RSV or an RSV component is administered).
  • administration of RSV elicits a protective immune response.
  • Dosages and methods for eliciting a protective anti -viral immune response, adaptable to producing a protective immune response against RSV are known to those of skill in the art. See, e.g., USPN 5,922,326; Wright et al. (1982) Infect. Immun. 37:397-400; Kim et al. (1973) Pediatrics 52:56-63; and Wright et al. (1976) J.
  • virus can be provided in the range of about 10 -10 pfu (plaque forming units) per dose administered (e.g., 10 4 -10 5 pfu per dose administered).
  • the dose will be adjusted based on, e.g., age, physical condition, body weight, sex, diet, mode and time of administration, and other clinical factors.
  • the prophylactic vaccine formulation can be systemically administered, e.g., by subcutaneous or intramuscular injection using a needle and syringe or a needleless injection device.
  • the vaccine formulation is administered intranasally, e.g., by drops, aerosol (e.g., large particle aerosol (greater than about 10 microns)), or spray into the upper respiratory tract.
  • aerosol e.g., large particle aerosol (greater than about 10 microns)
  • spray into the upper respiratory tract e.g., by drops, aerosol (e.g., large particle aerosol (greater than about 10 microns)
  • intranasal administration confers the added benefit of eliciting mucosal immunity at the site of entry of the virus.
  • attenuated live virus vaccines are often preferred, e.g., an attenuated, cold adapted and or temperature sensitive recombinant RSV, e.g., a chimeric recombinant RSV.
  • killed virus vaccines nucleic acid vaccines, and/or polypeptide subunit vaccines, for example, can be used, as suggested by Walsh et al. (1987) J. Infect. Pis. 155:1198-1204 and Murphy et al. (1990) Vaccine 8:497-502.
  • the attenuated recombinant RSV of this invention as used in a vaccine is sufficiently attenuated such that symptoms of infection, or at least symptoms of serious infection, will not occur in most individuals immunized (or otherwise infected) with the attenuated RSV.
  • viral components e.g., the nucleic acids or polypeptides herein
  • serious infection is not typically an issue.
  • the attenuated RSV (or RSV components of the invention) can still be capable of producing symptoms of mild illness (e.g., mild upper respiratory illness) and/or of dissemination to unvaccinated individuals.
  • virulence is sufficiently abrogated such that severe lower respiratory tract infections do not typically occur in the vaccinated or incidental host.
  • While stimulation of a protective immune response with a single dose is preferred, additional dosages can be administered, by the same or different route, to achieve the desired prophylactic effect.
  • multiple administrations may be required to elicit sufficient levels of immunity.
  • Administration can continue at intervals throughout childhood, as necessary to maintain sufficient levels of protection against wild-type RSV infection.
  • adults who are particularly susceptible to repeated or serious RSV infection such as, for example, health care workers, day care workers, family members of young children, the elderly, and individuals with compromised cardiopulmonary function may require multiple immunizations to establish and/or maintain protective immune responses.
  • Levels of induced immunity can be monitored, for example, by measuring amounts of neutralizing secretory and serum antibodies, and dosages adjusted or vaccinations repeated as necessary to elicit and maintain desired levels of protection.
  • an immune response can be stimulated by ex vivo or in vivo targeting of dendritic cells with virus.
  • proliferating dendritic cells are exposed to viruses in a sufficient amount and for a sufficient period of time to permit capture of the RSV antigens by the dendritic cells.
  • the cells are then transferred into a subject to be vaccinated by standard intravenous transplantation methods.
  • the formulation for prophylactic administration of the RSV also contains one or more adjuvants for enhancing the immune response to the RSV antigens.
  • Suitable adjuvants include, for example: complete Freund's adjuvant, incomplete Freund's adjuvant, 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), Cot ⁇ nebacterium parvum, and the synthetic adjuvant QS-21.
  • prophylactic vaccine administration of RSV can 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 RSV, or can be administered separately. Either the protein or an expression vector encoding the protein can be administered to produce an immunostimulatory effect.
  • vaccination of an individual with an attenuated RSV of a particular strain of a particular subgroup can induce cross-protection against RSV of different strains and/or subgroups
  • cross-protection can be enhanced, if desired, by vaccinating the individual with attenuated RSV from at least two strains, e.g., each of which represents a different subgroup.
  • the attenuated RSV vaccines of this invention can optionally be combined with vaccines that induce protective immune responses against other infectious agents.
  • kits of the invention contains one or more nucleic acid, polypeptide, antibody, or cell line described herein. Most often, the kit contains a diagnostic nucleic acid or polypeptide (e.g., an antibody or a probe, e.g., as a cDNA microarray packaged in a suitable container) or other nucleic acid such as one or more expression vector.
  • the kit typically further comprises one or more additional reagents, e.g., substrates, labels, primers, tubes and/or other accessories, reagents for collecting samples, buffers, hybridization chambers, cover slips, etc.
  • the kit optionally further comprises an instruction set or user manual detailing preferred methods of using the kit components.
  • the present invention provides digital systems, e.g., computers, computer readable media and integrated systems comprising character strings corresponding to the sequence information herein for the polypeptides and nucleic acids herein, including, e.g., those sequences listed herein and the various silent substitutions and conservative substitutions thereof.
  • Integrated systems can further include, e.g., gene synthesis equipment for making genes and/or peptide synthesis equipment for making polypeptides corresponding to the character strings.
  • a system of the invention can include the foregoing software having the appropriate character string information, e.g., used in conjunction with a user interface (e.g., a GUI in a standard operating system such as a Windows, Macintosh or LINUX system) to manipulate strings of characters corresponding to the sequences herein.
  • a user interface e.g., a GUI in a standard operating system such as a Windows, Macintosh or LINUX system
  • specialized alignment programs such as BLAST can also be incorporated into the systems of the invention for alignment of nucleic acids or proteins (or corresponding character strings).
  • Systems in the present invention typically include a digital computer with data sets entered into the software system comprising any of the sequences herein.
  • the computer can be, e.g., a PC (Intel x86 or Pentium chip- compatible DOSTM, OS2TM WINDOWSTM WINDOWS NTTM, WINDOWS95TM, WINDOWS98TM LINUX based machine, a MACINTOSHTM, Power PC, or a UNIX based (e.g., SUNTM work station) machine) or other commercially common computer which is known to one of skill.
  • Software for aligning or otherwise manipulating sequences is available, or can easily be constructed by one of skill using a standard programming language such as Visualbasic, Fortran, Basic, Java, or the like.
  • Any controller or computer optionally includes a monitor which is often a cathode ray tube ("CRT") display, a flat panel display (e.g., active matrix liquid crystal display, liquid crystal display), or others.
  • Computer circuitry is often placed in a box which includes numerous integrated circuit chips, such as a microprocessor, memory, interface circuits, and others.
  • the box also optionally includes a hard disk drive, a floppy disk drive, a high capacity removable drive such as a writeable CD-ROM, and other common peripheral elements.
  • Inputting devices such as a keyboard or mouse optionally provide for input from a user and for user selection of sequences to be compared or otherwise manipulated in the relevant computer system.
  • the computer typically includes appropriate software for receiving user instructions, either in the form of user input into a set parameter fields, e.g., in a GUI, or in the form of preprogrammed instructions, e.g., preprogrammed for a variety of different specific operations.
  • the software then converts these instructions to appropriate language, e.g., for instructing the operation of equipment, e.g., gene and or peptide synthesis equipment, to carry out the desired operation.
  • the software can also include output elements for controlling nucleic acid synthesis (e.g., based upon a sequence or an alignment of a sequences herein) or other operations.
  • the present invention provides system kits embodying the methods, composition, systems and apparatus herein.
  • System kits of the invention optionally comprise one or more of the following: (1) an apparatus, system, system component or apparatus component as described herein; (2) instructions for practicing the methods described herein, and/or for operating the apparatus or apparatus components herein and/or for using the compositions herein.
  • the present invention provides for the use of any apparatus, apparatus component, composition or kit herein, for the practice of any method or assay herein, and/or for the use of any apparatus or kit to practice any assay or method herein.
  • ATCC Type Culture Collection
  • MEM minimal essential medium
  • RSV A2 was obtained from the ATCC and grown in
  • Opti-MEM medium Modified vaccinia virus Ankara expressing bacteriophage T7 RNA polymerase (MVA-T7; Sutter et al. (1995) FEBS Lett. 371:9-12 and Wyatt et al. (1995)
  • Virology 210:202-205) was provided by Pr. Bernard Moss and amplified in CEK cells.
  • Recombinant fowlpox virus expressing the T7 RNA polymerase (FPV-T7; Britton et al. (1996) "Expression of bacteriophage T7 RNA polymerase in avian and mammalian cells by a recombinant fowlpox virus" J Gen Virol 77:963-7) was obtained from Dr. Michael Skinner and grown in CEK cells.
  • RSV B9320 was grown in Vero cells and viral RNA was extracted from virus purified by ultracentrifugation from infected cell culture supernatant. 9320 genome sequences were obtained by sequencing DNA fragments generated by RT-PCR; the cDNA full length clone was also sequenced for comparison. All sequencing was done by
  • RSV 9320 protein expression plasmids [0188] The 9320 N, P, and L protein coding regions were each cloned into a pCITE vector (Novagen, Madison, WI) under control of a T7 RNA polymerase promoter, to produce expression plasmids pB-N, pB-P, and pB-L.
  • pB-N The N gene was amplified by RT-PCR from 9320 RNA extracted from virus particles purified by ultracentrifugation, using primers XC19 (5'- GATCCCATGGCTCTTAGCAAAGTCAAG-3' containing Nco I site, SEQ ID NO:20) and XC020 (5'-GTACGGATCCGTTGACTTATTTGCCCCGTAT-3' containing Bamffl site, SEQ ID NO:21), and cloned between the Ncol and Bamffl sites of pCITE2a 3a (Novagen) under the control of T7 promoter.
  • This N protein expression plasmid was designated as AD740.
  • pB-P The P gene was amplified by RT-PCR using primers XC17 (5 -
  • pB-L The L gene was cloned from three cDNA subclones obtained by RT-
  • GCTTGGCCATAACGATTCTATATCATCC-3', SEQ ID NO:24) and XC014 (5'- GGTAGTATAATGTTGTGCACTTTTAG-3', SEQ ID NO:25) were used to amplify 9320 L from nt8511 to nt!1685 and the cDNA was cloned into T/A vector (Invitrogen, pCR 2.1) to generate subclone AD762.
  • GGTCACGATTTACAAGATAAGCTCC-3', SEQ ID NO:26) andXC007 (5'- CAGATCCTTTTAACTTGCTACCTAGGCACA-3' SEQ ID NO:27) were used to amplify nt 11686 to ntl4495 and the BamH I to Avr II fragment was cloned into the T/A vector as AD763.
  • CTTACGTGTGCCTAGGTAGCAAG-3', SEQ ID NO:28) and XC010 (5'- ACGAGAAAAAAAGTGTCAAAAACTAATGTCTCG, SEQ ID NO:29) were used to amplify 9320 nt 14495 to 15225, producing a first PCR product.
  • RBZ ribozyme cleavage sequence
  • T70 T7 terminator sequence
  • the three subclones were verified by sequence analysis. To join the three L subclones together, the Avrll and Kpn I fragment was removed from AD764 and cloned into AD763 and the larger clone was designated as AD767.
  • the Bamffl to Notl restriction fragment from ntl!685 to the sequence downstream of the T7 terminator was removed from AD767 and cloned into pCITE2a/3a vector under the control of the T7 promoter and the plasmid was designated as AD766.
  • the Bamffl fragment from nt 8511 to nt 11685 was removed from clone AD762 and inserted into the BamH I site of AD766.
  • the second Bamffl site at position of nt 11685 was then knocked out by site-directed mutagenesis and the clone is designated as pB-L (AD778).
  • Xma I, Avr II, Sac I, BamH I, and Bssffll The Xma I-Avr II cDNA fragment (BI) containing the T7 RNA polymerase promoter proximal to the 5' antigenomic sense DNA was joined with the Avr Il-Sac I cDNA fragment (B2) to form the B7 fragment (through a
  • the B7 fragment was used to replace the corresponding region in a full-length RSV A2-B9320 chimera containing the G, F and M2 sequence (B3).
  • the Sac I restriction site at nt 2310 in the resulting pUC-B8 was mutated without affecting the coding sequence of the SH gene.
  • the L gene fragment (B10) was assembled from B4 and B9 fragments.
  • the hepatitis delta virus ribozyme (RBZ) and the T7 RNA polymerase terminator sequence was amplified from RSV A2 antigenomic cDNA (Jin et al. (1998)
  • the BamH I site at nt position 11685 was deleted from B10 cDNA by mutagenesis without affecting the protein coding sequence.
  • the L gene (B10) was cloned into the chimeric clone that contained the 9320 B8 fragment and A2 L to replace the A2-L sequence through the BamH I and Not I restriction sites.
  • the antigenomic cDNA clone (Bll) encoding the complete RSV 9320 genome was designated ⁇ B9320C4.
  • an antigenomic cDNA clone containing a single C to G change at the fourth position of the leader sequence was also obtained by mutagenesis and designated pB9320G4.
  • the PCR fragment was cloned between the Xma I and Sad sites of the modified pET vector (pET21b was cut with BspEI, and the pET21b was ligated with a polylinker with Xmal, Smal, Sad, Mscl, Bamffl, Spel, Pmll, and Bssffll restriction sites to produce the modified pET-21b vector) and designated as AD803.
  • a second PCR DNA that contained nt 2106 to 4494 was obtained by RT/PCR using XC034 (5'-GTGTGGTCCTAGGCAATGCAGCAG-3', SEQ ID NO.-34) and XC032 (5'-GACACAGCATGATGGTAGAGCTCTATGTG-3', SEQ ID NO:35) as primers and was cloned into the Sac I site of the pET vector for sequence analysis. This Sad fragment was then moved to AD803 through the Sac I site in AD803 (producing B7).
  • the Sac I site at position nt 2310 was removed by mutagenesis and the resulting clone was designated as AD816.
  • the Xma I to Sac I fragment from AD816 was released to replace the corresponding region of the A2 sequence in the chimeric cDNA AD379 that had the G and F genes of 9320 in place of A2 G and F genes (in a pUC19 backbone, Cheng et al. (2001) Virology 283:59-68). This clone was designated as AD827.
  • RT-PCR using XC063 (5'-GCTAAGTGAACATAAAACATTCTGTAAC-3', SEQ ID NO:36) as RT primer and XC006 (5'-CCATTAATAATGGGATCCATTTTGTC-3' with Sad site, SEQ ID NO:37) and XC062 (5'-CACATAGAGCTCTACCATCATGCTGTGTC- 3' with BamH I site, SEQ ID NO:38) as PCR primers and was initially cloned into pET vector as AP835 for sequence analysis. The Sad to BamH I fragment from AD835 was then moved to AD827 and the clone was designated as AD848.
  • the recombinant 9320 cDNA has the following genetic tags that are different from wild-type 9320 virus.
  • the enclosed sequence, SEQ ID NO:l is the wild type RSV 9320 strain and does not reflect the recombinant DNA sequence.
  • Sac I sites at nt 2310, 10376 and 14951 were removed without changing the protein coding sequences, using the following primers: Sad at 2310 nt, XC049 (5'- GATGATGTAGAGCTTTAAGTTAAC-3', SEQ ID NO: 45) and XC050 (5'- GTTAACTTAAAGCTCTACATCATC-3", SEQ ID NO: 46); Sad at 10376 nt, XC088 (5'- CTAACTGGTAAAGAAAGAGAGCTTAGTGTAGGTAGAATGTTTGC-3', SEQ ID NO: 47) and XC089 (5'--
  • a BamH I site at nt 11685 was removed using primers XC067 (5'- CATTAATGAGGGACCCACAGGCTTTAG-3', SEQ IP NO:39) and XC068 (5'- CTAAAGCCTGTGGGTCCCTCATTAATG-3', SEQ IP NO:40).
  • XC067 5'- CATTAATGAGGGACCCACAGGCTTTAG-3', SEQ IP NO:39
  • XC068 5'- CTAAAGCCTGTGGGTCCCTCATTAATG-3', SEQ IP NO:40.
  • Sac I site at nt 4477 was added using XC032 (5'-GACACAGCATGATGGTAGAGCTCTATGTG, SEQ
  • G gene deletion mutants Two mutants were constructed to determine if the G gene of RSV B9320 strain is dispensable, e.g., for viral replication in tissue culture and/or an animal host. In one mutant, the entire open reading frame of the G gene was removed from the 9320 cDNA. In the other mutant, the region encoding the cysteine noose and heparin binding sites of G was removed from the 9320 cDNA.
  • deletion mutagenesis was performed on a pET-S/B cPNA subclone that contained sequences of the 9320 G, F and M2 genes using a pair of PCR primers flanking the G open reading frame in opposite orientations (5'-GATCCCATACTAATAATTCATCATTATG-3', SEQ IP NO:51, and 5'-AGCAGAGAACCGTGATCTATCAAGCAAG-3', SEQ IP NO:52) using the ExSite PCR-based Site-Pirected Mutagenesis Kit (Stratagene, La Jolla, CA). The deletion was confirmed by restriction enzyme digestion and nucleotide sequencing analysis. The Sac I-BaniH I fragment containing only the F and M2 genes was introduced into pB9320G4, and the antigenomic cPNA was designated pB9320 ⁇ G.
  • a small cPNA fragment of nt 5179-5280 nt was deleted from the 9320 G gene using primers XC079 (5 * -GTAATCATCTTTTGGTTTTTTTGGTGG-3', SEQ IP NO:53) and XC080 (5'-CCAACCATCAAACCCACAAACAAACCAACCGTC-3' 5 SEQ IP NO:54).
  • the cPNA containing the desired deletion was then removed from the subclone by digestion with Sac I and Bamffl and shuffled into the full-length 9320 antigenomic cPNA.
  • the resulting antigenomic cPNA was designated pB9320 ⁇ HBS.
  • HEp-2 cells were infected with MVA-T7 at an m.o.i. of 5.0 and transfected with 0.4 ⁇ g of pB-N (pN), 0.4 ⁇ g of pB-P (pP), 0.2 ⁇ g of pB-L
  • LipofectamineTM 2000 (Invitrogen, Carlsbad, CA). In some transfection reactions, 0.2 ⁇ g pRSV-M2-l (encoding the RSV A2 M2-1 protein) was also included. Alternatively, Vero cells were infected with FPV-T7 (Britton et al. (1996) J Gen Virol 77:963-7) at an m.o.i. of
  • the culture supernatant was used to infect Vero cells to amplify the recovered viruses.
  • the culture supernatant was harvested and virus-infected cells were identified by immunostaining using polyclonal anti-
  • RSV A2 serum (Biogenesis, Singer, NH).
  • the recombinant virus from the culture supernatant was plaque purified and amplified in Vero cells.
  • Replication of rg9320C4, rg9320G4 and rg9320 ⁇ G in Vero and HEp-2 cells was compared with replication of wild type 9320. Vero or HEp-2 cell monolayers in 6-well plates were infected with each virus in duplicate at an m.o.i. of 0.1. After 1 hr adsorption at room temperature, the infected cells were washed with PBS three times and incubated with
  • Vero or HEp-2 cells were infected with virus at an m.o.i of 5.0 and the infected cells were lysed in Laemmli protein sample buffer (Bio-Rad, Hercules, CA). The cell lysates were electrophoresed on 12% polyacrylamide gels containing 0.1% SPS, and then transferred to a nylon membrane. The blots were incubated with either polyclonal anti-
  • RS V A2 serum or a mixture of four monoclonal antibodies against the G protein of RSV B strain 2434PB3, 2218BP5, 2218AE7 and 2218PG7 obtained from Pr. Gregory Storch
  • HRP horseradish peroxidase
  • the RSV 9320 genome contains 15,225 nucleotides and shares 97.8% and
  • the A2 genome contains 15,222 nucleotides; the BI genome contains 15,225 nucleotides (Karron et al. (1997) Proc Natl Acad Sci USA 94:13961-13966).
  • 9320 contains 10 transcriptional units encoding 11 proteins in the order of NS1/NS2/N/P/M/SH G F/M2-1/M2-2/L.
  • NSl is listed as SEQ ID NO:2, NS2 as SEQ IP NO:3, N as SEQ IP NO:4, P as SEQ IP NO:5, M as SEQ IP NO:6, SH as SEQ IP NO:7, G as SEQ IP NO: 12, F as SEQ IP NO:8, M2-1 as SEQ IP NO:9, M2-2 as SEQ IP NO:10, and L as SEQ IP NO:ll.
  • Table 3 lists the size of each of the 11 proteins for the 9320, B 1 , and A2 strains.
  • Table 4 lists the length of the intergenic regions for the three RSV strains.
  • the SH, G and M2-2 proteins display the greatest differences between A2 and 9320, while the other proteins have an amino acid identity greater than 86%.
  • M2-1 function was probably supplied by cryptic expression of the M2-1 protein from the transfected full-length antigenomic cPNA (Collins et al. (1999) Virology 259:251-
  • pB9320C4 or ⁇ B9320G4 was transfected into MVA-T7 infected HEp-2 cells or FPV-T7 infected Vero cells together with the 9320 N, P and L expression plasmids with or without the RSV A2 M2-1 plasmid.
  • Table 6 lists the recovery efficiency of recombinant RSV in the presence or absence of the M2-1 expression plasmid.
  • FPV-T7 infected Vero cells or MVA-T7 infected HEp-2 cells were transfected with p9320C4 or p9320G4 in triplicate wells together with N, P, L expression plasmids with or without M2-1.
  • the culture supematants were titrated on Vero cells and the plaque numbers in pfu/ml from each well are shown. The average plaque number is given in parentheses.
  • rg9320G4 was rescued more efficiently than rg9320 in both the FPV-T7 infected Vero cells and the MVA-T7 infected HEp-2 cells. Inclusion of the M2-1 expression plasmid slightly increased rescue efficiency in both cell types.
  • the vaccine is typically produced from a qualified cell line that is free of any adventitious agents.
  • the HEp-2 cells currently used for recovery of infectious RSV A2 e.g., Collins et al. (1995) Proc. Natl. Acad. Sci. USA 92:11563- 11567 and Jin et al. (1998) Virology 251:206-214) are not suitable for vaccine production. Vero cells were therefore explored as the cell substrate for recovering 9320 virus from its cDNA. It was very difficult to recover virus from MVA-T7-infected Vero cells.
  • FPV-T7 has been shown to have a less cytopathic effect in infected Vero cells and thus result in more efficient virus rescue of other viruses (Britton et al. (1996) J Gen Virol 77:963-7 and Das et al. (2000) "Improved technique for transient expression and negative strand virus rescue using fowlpox T7 recombinant virus in mammalian cells" J Virol Meth 89:119-127).
  • the recombinant RSV described here were more efficiently recovered from FPV- T7 infected Vero cells than from MVA-T7 infected cells, which may pave the way for recovering RSV vaccine candidates for clinical studies.
  • rg9320C4 and rg9320G4 were plaque purified and amplified in Vero cells, both reaching a titer of 1 x 10 7 pfu/ml.
  • a recombinant virus with the G gene deleted from rg9320G4 was also obtained and the virus was designated rg9320 ⁇ G.
  • the identity of the recombinant viruses generated from cDNA was analyzed by RT/PCR of each viral RNA using primer pairs spanning the introduced marker sites or the G deletion region. Digestion of the RT/PCR DNA product from nt 2104 to nt 3096 by the Sac I restriction enzyme showed that the Sac I site was present in 9320 virus but not in the recombinant viruses rg9320C4 and rg9320 ⁇ G. Digestion of the RT/PCR product from nt 11593 to nt 11822 by BamH I confirmed that the BamH I site in the recombinant viruses was also abolished.
  • RT/PCR using a pair of primers spanning the G gene confirmed that the G gene was deleted from rg9320 ⁇ G, since its DNA product was approximately lkb shorter than that of rg9320. Sequencing of the G deletion junction region confirmed the expected deleted sequence.
  • Replication of recombinant 9320 and its G deletion mutant were compared with replication of the biologically derived 9320 strain.
  • Vero or HEp-2 cells were infected with 9320, rg9320C4, rg9320G4 or rg9320 ⁇ G at an m.o.i. of 0.1, and the accumulated level of viruses released into the culture supernatant at each day was titrated in Vero cells.
  • Figure 4 Panel A the growth kinetics of rg9320C4, rg9320G4 and rg9320 ⁇ G were very similar to those of 9320 in Vero cells, reaching peak titers of 6.0 logio pfu/ml at 96 hours post infection.
  • the G protein enhances virus binding to target cells, but it has no role in virus penetration once virus has attached the cells (Techaarpornkul et al. (2001) J Virol 75:6825- 34).
  • rA2 ⁇ G replicated as well as wt A2 strain, indicating an alternative pathway might be present in Vero cells for efficient virus entry that is independent of the G protein (Teng et al. (2001) Virology 289:283-96).
  • rg9320 ⁇ G replicated efficiently in Vero cells; however, in contrast to rA2 ⁇ G, its replication was only slightly reduced in HEp-2 cells.
  • rg9320 ⁇ HBS a recombinant 9320 virus with a partial deletion of G (the cysteine noose and heparin binding sites) was also recovered from cPNA as described above. Like rg9320 ⁇ G, replication of rg9320 ⁇ HBS in Vero cells is not significantly impaired.
  • Subgroup B RSV typically replicates better in cotton rats than in mice.
  • Vero cells Table 7 lists the mean viral titer for each recombinant virus.
  • rg9320C4 and rg9320G4 replicated to a titer of 3.1 and 3.0 log 10 pfu/g, respectively, in the lungs of cotton rats.
  • Both G deletion mutants (rg9320 ⁇ G and rg9320 ⁇ HBS) replicated poorly, indicating G deletion affected RSV 9320 replication in the animal host.
  • rg9320 ⁇ G and rg9320 ⁇ HBS are thus potential candidates for live attenuated RSV vaccines.
  • RSV vaccine would preferably provide protection against both subgroup
  • RSV vaccines developed in the past have been based on the RSV A2 strain. However, it remains to be determined whether an RSV vaccine based solely on a subgroup A strain would provide sufficient immunity to both RSV subgroups. Although recombinant technology has been employed to express subgroup B RSV antigens in the A2 strain, including the replacement of the G and F genes by those of BI (Whitehead et al.
  • RSV B9320 can optionally be attenuated by methods used to attenuate subgroup A strains.
  • the methods that have been used to attenuate RSV subgroup A RSV include, e.g., mutagenesis of the viral internal proteins (Lu et al. (2002) "Identification of temperature- sensitive mutations in the phosphoprotein of respiratory syncytial virus that are likely involved in its interaction with the nucleoprotein" J Virol 76:2871-2880; Lu et al.
  • G gene e.g., as described herein, are attenuated 9320 vaccine candidates.
  • Lys Leu lie Leu Leu Thr Asn Ala Leu Ala Lys Ala Ala lie His Thr 35 40 45
  • Val Cys Pro Asp Asn Asn lie Val Val Lys Ser Asn Phe Thr Thr Met 65 70 75 80
  • Lys lie Leu Lys Asp Ala Gly Tyr His Val Lys Ala Asn Gly Val Asp 85 90 95
  • Val Leu Thr Leu Ser Ser Leu Thr Ser Glu lie Gin Val Asn lie Glu 115 120 125
  • aactattata aacttaatac ttatccatct 10020 ctacttgaaa tcacagaaaa tgatttgatt attttatcag gattgcggtt ctatcgtgaa 10080 tttcatctgc ctaaaaaagt ggatcttgaa atgataataa atgacaaagc catttcacct 10140 ccaaaagatc taatatggac tagttttcct agaaattaca tgccatcaca tatacaaaat 10200 tatatagaac atgaaaagtt gaagttctct gaaagcgaca gatcaagaag agtactagag 10260 tattacttga gagataataa attcaatgaa tgcgatctat acaattgtgt
  • oligonucleotide primer Bglllsite, RSV B 9320 G
  • oligonucleotide primer Bglllsite, RSV B 9320 G
  • oligonucleotide primer BamHI site, RSV 9320 G

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pulmonology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Agricultural Chemicals And Associated Chemicals (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The complete polynucleotide sequence of the human respiratory syncytial virus subgroup B strain 9320 genome is provided. Proteins encoded by this polynucleotide sequence are also provided. Isolated or recombinant RSV (e.g. attenuated recombinant RSV), nucleic acids, and polypeptides, e.g., comprising mutations in the attachment protein G, are also provided, as are immunogenic compositions comprising such isolated or recombinant RSV, nucleic acids, and polypeptides, Related methods are also described.

Description

COMPOSITIONS AND METHODS INVOLVING RESPIRATORY SYNCYTIAL
VIRUS SUBGROUP B STRAIN 9320
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application is a non-provisional utility patent application claiming priority to and benefit of the following prior provisional patent applications: USSN 60/458,331, filed March 28, 2003, entitled "Compositions and Methods Involving Respiratory Syncytial Virus Subgroup B Strain 9320" by Xing Cheng, et al., and USSN 60/508,320, filed October 3, 2003, entitled "Compositions and Methods Involving Respiratory Syncytial Virus Subgroup B Strain 9320" by Xing Cheng, et al., each of which is incorporated herein by reference in its entirety for all purposes.
FIELD OF THE INVENTION
[0002] The present invention is in the field of virology. More specifically, the invention relates to human respiratory syncytial virus, including the diagnosis, treatment, and prevention of human RSV infections.
BACKGROUND OF THE INVENTION
[0003] Human Respiratory Syncytial Virus (RSV) is the leading cause of hospitalization for viral respiratory tract disease (e.g., bronchiolitis and pneumonia) in infants and young children worldwide, as well as a significant source of morbidity and mortality in immunocompromised adults and in the elderly (see, e.g., Shay et al. (1999) "Bronchiolitis-associated hospitalizations among US children, 1980-1996" JAMA 282:1440-1446, Falsey et al. (1995) "Respiratory syncytial virus and influenza A infections in the hospitalized elderly" J Infect Pis 172:389-394, Falsey et al. (1992) "Viral respiratory infections in the institutionalized elderly: clinical and epidemiologic findings" J Am Geriatr Soc 40: 115-119, Falsey and Walsh (1998) "Relationship of serum antibody to risk of respiratory syncytial virus infection in elderly adults" J Infect is 177:463-466, Hall et al. (1986) "Respiratory syncytial viral infection in children with compromised immune function" N End J Med 315:77-81, and Harrington et al. (1992) "An outbreak of respiratory syncytial virus in a bone marrow transplant center" J Infect Pis 165:987-993). To date, no vaccines have been approved which are able to prevent the diseases associated with RSV infection. RSV is an enveloped virus that has a single-stranded negative sense non- segmented RNA genome, and it is classified in the Pneumovirus genus of the Paramyxoviridae family (Collins et al. (2001) Respiratory syncytial virus, pp. 1443-1485. In; Knipe and Howley (eds.) Fields Virology vol. 1. Lippincott, Williams and Wilkins, Philadelphia; Lamb and olakofsky (2001) Paramyxoviridae: the viruses and their replication pp. 1305-1340. In; Knipe and Howley (eds.) Fields Virology vol. 1. Lippincott, Williams and Wilkins, Philadelphia). Human RSV is classified into two subgroups, subgroups A and B, based on antigenic diversity and nucleotide sequence divergence. For example, the attachment protein G is most divergent and the fusion protein F is relatively conserved between the two subgroups.
[0004] Considerable progress has been made towards understanding the molecular biology of subgroup A RSV; however, much less information is available for subgroup B RSV. Most work to date has focused on subgroup A strains. For example, RSV strain A2 has been sequenced. The genome of the A2 strain RSV is 15,222 nt in length and contains 10 transcriptional units that encode 11 proteins (NS1, NS2, N, P, M, SH, G, F, M2-1, M2-2, and L). The genome is tightly bound by the N protein to form the nucleocapsid, which is the template for the viral RNA polymerase comprising the N, P and L proteins (Grosfeld et al. (1995) J. Virol. 69:5677-5686; Yu et al. (1995) J. Virol. 69:2412-2419). Each transcription unit is flanked by a highly conserved 10-nt gene-start (GS) signal, at which mRNA synthesis begins, and ends with a semiconserved 12- to 13-nt gene-end (GE) signal that directs polyadenylation and release of mRNAs (Harmon et al. (2001) J. Virol. 75:36-44; Kuo et al. (1996) J. Virol. 70:6892-6901). Transcription of RSV genes is sequential, and there is a gradient of decreasing mRNA synthesis due to transcription attenuation (Barik (1992) J. Virol. 66:6813-6818; Pickens et al. (1984) J. Virol. 52:364-369). The viral RNA polymerase must first terminate synthesis of the upstream message in order to initiate synthesis of the downstream mRNA.
[0005] The nucleocapsid protein (N), phosphoprotein (P), and large polymerase protein (L) constitute the minimal components for viral RNA replication and transcription in vitro (Grosfield et al. (1995) J. Virol. 69:5677-5686; Yu et al. (1995) J. Virol. 69:2412- 2419). The N protein associates with the genomic RNA to form the nucleocapsid, which serves as the template for RNA synthesis. The L protein is a multifunctional protein that contains RNA-dependent RNA polymerase catalytic motifs and is also probably responsible for capping and polyadenylation of viral mRNAs. However, the L protein alone is not sufficient for the polymerase function; the P protein is also required. Transcription and replication of RSV RNA are also modulated by the M2-1, M2-2, NSl, and NS2proteins that are unique to the pneumo viruses. M2-1 is a transcription antitermination (or elongation) factor required for processive RNA synthesis and transcription read-through at gene junctions, essential for RNA transcription and virus replication (Collins et al. (1996) "Transcription elongation factor of respiratory syncytial virus, a nonsegmented negative- strand RNA virus" Proc Natl Acad Sci USA 93:81-85; Hardy and Wertz (2000) "The Cys3- Hisl motif of the respiratory syncytial virus M2-1 protein is essential for protein function" J Virol 74:5880-5885; Tang et al. (2001) "Requirement of cysteines and length of the human respiratory syncytial virus M2-1 protein for protein function and virus viability" J Virol 75:11328-11335; Collins et al. (2001) in P. M. Knipe et al. (eds.), Fields Virology. 4th ed. Lippincott, Philadelphia; Hardy et al. (1999) J. Virol. 73:170-176; and Hardy andWertz (1998) J. Virol. 72:520-526). M2-2, though not essential for virus replication in tissue culture, is involved in the switch between viral RNA transcription and replication (Bermingham and Collins (1999) Proc. Natl. Acad. Sci. USA 96:11259-11264; Jin et al. (2000) J. Virol. 74:74-82). NSl and NS2 have been shown to inhibit minigenome synthesis in vitro (Atreya et al. (1998) J. Virol. 72:1452-1461).
[0006] NSl, NS2, SH, M2-2 and G are accessory proteins that can be deleted from the RSV A2 strain without affecting virus viability (Bermingham and Collins (1999) Proc. Natl. Acad. Sci. USA 96:11259-11264; Jin et al. (2000) J. Virol. 74:74-82; Jin et al. (2000) "Recombinant respiratory syncytial viruses with deletions in the NSl, NS2, SH, and M2-2 genes are attenuated in vitro and in vivo" Virology 273:210-218; Bukreyev et al. (1997) "Recombinant respiratory syncytial virus from which the entire SH gene has been deleted grows efficiently in cell culture and exhibits site- specific attenuation in the respiratory tract of the mouse" J Virol 71:8973-8982: Teng and Collins (1999) "Altered growth characteristics of recombinant respiratory syncytial viruses which do not produce NS2 protein" J Virol 73:466-473; Teng et al. (2000) "Recombinant respiratory syncytial virus that does not express the NSl or M2-2 protein is highly attenuated and immunogenic in chimpanzees" J Virol 74:9317-9321; Karron et al. (1997) "Respiratory syncytial virus (RSV) SH and G proteins are not essential for viral replication in vitro: clinical evaluation and molecular characterization of a cold-passaged, attenuated RSV subgroup B mutant" Proc Natl Acad Sci USA 94:13961-13966). However, except for the SH deletion mutant, most of the gene deletion mutants do not replicate as well as the wild type RSV either in tissue culture or in animal hosts.
[0007] The G and F proteins are the two major surface antigens that elicit anti-RSV neutralizing antibodies to provide protective immunity against RSV infection and reinfection. High levels of circulating antibodies correlate with protection against RSV infections or reduction of disease severity (Crowe (1999) Microbiol. Immunol. 236:191- 214). As noted, two antigenic RSV subgroups (A and B) have been recognized based on virus antigenic and sequence divergence (Anderson et al. (1985) J. Infect. Pis. 151:626-633; Mufson et al. (1985) J. Gen. Virol. 66:2111-2124). By using a reciprocal cross- neutralization assay, it has been determined that the F proteins between the two subgroups are 50% related and the G proteins are only 1-7% related (reviewed by Collins et al. (2001) "Respiratory syncytial virus" In: P.M. Knipe et al. (Ed) Fields Virology, pp. 1443-1485, Vol. 1, Lippincott Williams & Wilkins, Philadelphia). This antigenic diversity may be partly responsible for repeated RSV infection. The antigenic diversity of these two RSV subgroups enables viruses from both subgroups to circulate concurrently in a community, and the prevalence of each subgroup can alternate during successive years. Epidemic studies of RSV infection in children have suggested that naturally acquired infection elicits a relatively higher protection against disease caused by the homologous subgroup virus (Mclntosh and Chanock (1990) "Respiratory syncytial virus" In: P.M. Knipe et al. (Ed) Second Edition Virology, pp.1045-1072, Raven Press, Ltd., New York). The immunity induced by RSV infection is transient and subsequent reinfection can occur. However, RSV reinfection usually does not cause severe disease. An RSV vaccine is therefore typically targeted to provide protection against severe lower respiratory disease caused by RSV subgroup A and B viruses.
[0008] Efforts to produce a safe and effective RSV vaccine have focused on the administration of purified viral antigen or the development of live attenuated RSV for intranasal administration. For example, a formalin-inactivated virus vaccine not only failed to provide protection against RSV infection, but was shown to exacerbate symptoms during subsequent infection by the wild-type virus in infants (Kapi ian et al. (1969) Am. J. Epidemiol. 89:405-421; Chin et al. (1969) Am. J. Epidemiol. 89:449-63). More recently, efforts have been aimed towards developing live attenuated temperature-sensitive mutants by chemical mutagenesis or cold passage of the wild-type RSV (Crowe et al. (1994) Vaccine 12:691-9). Typically, the virus candidates have been either underattenuated or overattenuated (Kim et al. (1973) Pediatrics 52:56-63; Wright et al. (1976) J. Pediatrics 88:931-6), and some of the candidates were genetically unstable, resulting in the loss of the attenuated phenotype (Hodges et al. (1974) Proc Soc. Exp. Bio. Med. 145:1158-64). To date, no live attenuated vaccine has been brought to market.
[0009] Characterization of additional strains of RSV, particularly from subgroup B, will assist in production of effective vaccines (e.g., regions of homology or identity between strains can indicate functionally conserved regions that can be targeted by mutagenesis). Although short regions of various subgroup B strains have been sequenced (e.g., B9320 protein G, SEQ ID NO: 14, from GenBank accession number M73544; a B9320 intergenic region, SEQ IP NO: 15, from GenBank accession number S75820; B9320 G and F gene start and end sequences, SEQ IP NOs:16-19, from Jin et al. (1998) Virology 251:206-214 and Cheng et al. (2001) Virology 283:59-68; and various B18537 coding and intergenic regions, GenBank accession numbers P00334, P00392-P00397, P00736, D01042, and M17213), only one subgroup B strain, strain BI, has been sequenced in its entirety (SEQ IP NO: 13, from GenBank accession number AF013254).
[0010] Accordingly, this invention presents the complete polynucleotide sequence of human RSV subgroup B strain 9320. Polypeptides encoded by the B9320 genome are also provided, as are other benefits which will become apparent upon review of the disclosure.
SUMMARY OF THE INVENTION
[0011] The present invention provides the complete polynucleotide sequence of human respiratory syncytial virus subgroup B strain 9320. Amino acid sequences of proteins encoded by the B9320 genome are also provided. The invention provides isolated or recombinant nucleic acids and polypeptides comprising the novel B9320 sequences. Isolated or recombinant RSV comprising the nucleic acids and polypeptides of the invention (e.g., attenuated recombinant RSV) are also provided, as are immunogenic compositions including such nucleic acids, polypeptides, and RSV that are suitable for use as vaccines. Recovery of infectious recombinant 9320 viruses from cPNAs is described.
[0012] In a first aspect, the present invention provides isolated or recombinant nucleic acids comprising a polynucleotide sequence of the invention. Thus, for example, an isolated or recombinant nucleic acid comprising the polynucleotide sequence of SEQ ID NO: 1 or a complementary polynucleotide sequence thereof is a favored embodiment of the invention. An isolated or recombinant nucleic acid comprising at least one unique polynucleotide subsequence of SEQ ID NO:l or a complementary polynucleotide sequence thereof, with the proviso that the unique polynucleotide subsequence includes at least one subsequence not included in SEQ ID NOs: 14-19 or a complementary polynucleotide sequence thereof, is another favored embodiment. The unique polynucleotide subsequence can, for example, comprise at least 10 contiguous nucleotides of SEQ ID NO:l or a complementary polynucleotide sequence thereof (e.g., at least 20 contiguous nucleotides, at least 50 contiguous nucleotides, at least 100 contiguous nucleotides, at least 500 contiguous nucleotides, or even at least 1000 contiguous nucleotides). In some embodiments, the polynucleotide subsequence includes at least one compete open reading frame (ORF) of SEQ ED NO-.l.
[0013] In addition to the sequences explicitly provided in the accompanying sequence listing, polynucleotide sequences that are highly related structurally and/or functionally (e.g., as defined by hybridization and or sequence identity) are polynucleotides of the invention. For example, a polynucleotide sequence that is greater than 97.8% identical to SEQ ID NO:l or a complementary polynucleotide sequence thereof, as determined by BLASTN using default parameters, is a polynucleotide of the invention. As another example, a polynucleotide sequence that hybridizes under stringent conditions over substantially the entire length of a polynucleotide subsequence comprising at least 100 contiguous nucleotides of SEQ I NO:l or a complementary polynucleotide sequence thereof, wherein the polynucleotide sequence hybridizes to the polynucleotide subsequence of SEQ IP NO:l or the complementary polynucleotide sequence thereof under said stringent conditions with at least 2x a signal to noise ratio that the polynucleotide sequence hybridizes to a corresponding polynucleotide subsequence of SEQ ID NO: 13 or a complementary polynucleotide sequence thereof, is a polynucleotide sequence of the invention. Similarly, polynucleotide sequences of the invention include a polynucleotide sequence encoding a polypeptide of the invention, e.g., encoding an amino acid sequence or unique subsequence selected from the group consisting of SEQ ID NOs:2-l 1 or an artificial conservative variation thereof. [0014] A nucleic acid of the invention optionally comprises at least one artificially mutated nucleotide, e.g., at least one artificially deleted, inserted, and/or substituted nucleotide. In certain embodiments, mutation of the polynucleotide sequence results in alteration of an encoded amino acid sequence. Thus, in one class of embodiments, at least one polypeptide encoded by the nucleic acid comprises at least one deleted, inserted, and/or substituted amino acid residue (e.g., at least one conservatively or non-conservatively substituted amino acid residue). For example, the mutated nucleotide can be located in an ORF encoding a polypeptide selected from SEQ ID NOs:2-12.
[0015] Another class of embodiments provides vectors comprising the nucleic acids of the invention. Yet another class of embodiments provides a host cell into which such a vector has been introduced. Another class of embodiments provides methods of producing a recombinant respiratory syncytial virus. In the methods, such a host cell is cultured in a suitable culture medium under conditions permitting expression of the nucleic acid, and the recombinant respiratory syncytial virus is isolated from the host cell and/or the medium. Recombinant RSV produced according to these methods form another feature of the invention, as do recombinant RSV comprising a nucleic acid of the invention. A related class of embodiments provides methods of producing an isolated or recombinant polypeptide. In the methods, a host cell comprising a vector that includes a nucleic acid of the invention is cultured in a suitable culture medium under conditions permitting expression of the nucleic acid, and the polypeptide is isolated from the host cell and or the medium. Polypeptides produced according to these methods form another feature of the invention, as do polypeptides comprising an amino acid sequence or subsequence that is encoded by a nucleic acid of the invention.
[0016] One aspect of the invention provides isolated or recombinant polypeptides comprising an amino acid sequence of the invention. Thus, for example, an isolated or recombinant polypeptide comprising an amino acid sequence selected from the group consisting of SEQ IP NOs:2-l 1 is a favored embodiment of the invention. An isolated or recombinant polypeptide comprising a unique amino acid subsequence comprising at least 7 (e.g., at least 8, at least 10, at least 20, at least 50, or more) contiguous amino acid residues of any one of SEQ IP NOs:2-ll is another favored embodiment. Artificial conservative variations of amino acid sequences or subsequences of the invention are also amino acid sequences of the invention, as are amino acid sequences that are substantially identical to an amino acid sequence of the invention. For example, an amino acid sequence that is greater than 99.3% identical to SEQ ID NO:2, greater than 98.4% identical to SEQ ID NO:3, greater than 99.7% identical to SEQ IP NO:4, greater than 98.3% identical to SEQ IP NO:5, greater than 99.6% identical to SEQ IP NO:6, greater than 97.0% identical to SEQ ID NO:7, greater than 99.3% identical to SEQ ID NO:8, greater than 99.5% identical to SEQ ID NO:9, greater than 96.4% identical to SEQ ID NO: 10, or greater than 99.2% identical to SEQ ID NO: 11, as determined by BLASTP using default parameters, is an amino acid sequence of the invention. Similarly, an amino acid sequence or subsequence that is specifically bound by an antibody that specifically binds to an amino acid sequence or subsequence encoded by SEQ ID NO:l, wherein said antibody does not specifically bind to an amino acid sequence or subsequence encoded by SEQ ID NO: 13 or SEQ ID NO: 14, is an amino acid sequence of the invention.
[0017] A polypeptide of the invention optionally comprises at least one artificially altered amino acid, e.g., at least one deleted, inserted, and/or substituted amino acid. For example, one class of embodiments provides an isolated or recombinant polypeptide comprising the amino acid sequence of SEQ ID NO: 12 with a deletion of residues 164-197, or an artificial conservative variation thereof.
[0018] Immunogenic compositions comprising an immunologically effective amount of a recombinant respiratory syncytial virus, polypeptide, and/or nucleic acid of the invention form another aspect of the invention. Similarly, another feature of the invention provides methods for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus. In the methods, an immunologically effective amount of a respiratory syncytial virus, polypeptide, and/or nucleic acid of the invention is administered to the individual in a physiologically acceptable carrier.
BRIEF DESCRIPTION OF THE DRAWINGS [0019] Figure 1 schematically illustrates assembly of the full-length antigenomic
RSV 9320 cDNA. Positions of various subclones used to assemble the full length antigenomic cDNA are indicated. The cDNA fragments obtained by RT PCR were ligated through the indicated restriction enzyme sites. The fourth residue of the leader sequence at the antigenomic sense was either C or G as indicated. [0020] Figure 2 schematically illustrates the cloning of pB-L, the assembly of the L coding region from three subclones. Positions of various subclones and primers used to assemble pB-L are indicated.
[0021] Figure 3 schematically illustrates cloning of the 5' portion of the RSV 9320 antigenome. Positions of various subclones and primers are indicated.
[0022] Figure 4 presents line graphs illustrating the growth of wild type 9320
(diamonds), rg9320C4 (squares), rg9320G4(triangles), and rg9320ΔG (circles) in Vero cells (Panel A) and HEp-2 cells (Panel B). Vero or HEp-2 cells were infected with each virus at an m.o.i of 0.1 and incubated at 35°C for 5 days. The viruses released into the culture supernatants at each day were titrated in Vero cells by plaque assay.
DEFINITIONS
[0023] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. The following definitions supplement those in the art and are directed to the current application and are not to be imputed to any related or unrelated case, e.g., to any commonly owned patent or application. Although any methods and materials similar or equivalent to those described herein can be used in the practice for testing of the present invention, the preferred materials and methods are described herein. Accordingly, the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting.
[0024] As used in this specification and the appended claims, the singular forms "a,"
"an" and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a virus" includes a plurality of viruses; reference to "a host cell" includes mixtures of host cells, and the like.
[0025] An "amino acid sequence" is a polymer of amino acid residues (a protein, polypeptide, etc.) or a character string representing an amino acid polymer, depending on context.
[0026] A "polynucleotide sequence" or "nucleotide sequence" is a polymer of nucleotides (an oligonucleotide, a DNA, a nucleic acid, etc.) or a character string representing a nucleotide polymer, depending on context. From any specified polynucleotide sequence, either the given nucleic acid or the complementary polynucleotide sequence (e.g., the complementary nucleic acid) can be determined.
[0027] A "subsequence" is any portion of an entire sequence, up to and including the complete sequence. Typically, a subsequence comprises less than the full-length sequence. A "unique subsequence" is a subsequence that is not found in any previously determined RSV polynucleotide or polypeptide sequence (e.g., the A2, BI, and B18537 sequences listed and/or referenced herein).
[0028] An "artificial mutation" is a mutation introduced by human intervention, e.g., under laboratory conditions. Thus, an "artificially mutated" nucleotide is a nucleotide that has been mutated as a result of human intervention, an "artificially altered" amino acid residue is a residue that has been altered as a result of human intervention, and an "artificial conservative variation" is a conservative variation that has been produced by human intervention. For example, a wild type virus (e.g., one circulating naturally among human hosts) or other parental strain of virus can be "artificially mutated" using recombinant DNA techniques to alter the viral genome (e.g., the viral genome can be altered by in vitro mutagenesis, or by exposing it to a chemical, ionizing radiation, or the like and then performing in vitro or in vivo selection for a temperature sensitive, cold sensitive, or otherwise attenuated strain of virus). As another example, a wild type protein can be "artificially altered" by artificially mutating the gene encoding that protein.
[0029] The term "variant" with respect to a polypeptide refers to an amino acid sequence that is altered by one or more amino acids with respect to a reference sequence. The variant can have "conservative" changes, wherein a substituted amino acid has similar structural or chemical properties, e.g., replacement of leucine with isoleucine. Alternatively, a variant can have "nonconservative" changes, e.g., replacement of a glycine with a tryptophan. Analogous minor variation can also include amino acid deletion or insertion, or both. Guidance in determining which amino acid residues can be substituted, inserted, or deleted without eliminating biological or immunological activity can be found using computer programs well known in the art, for example, DNASTAR software. Examples of conservative substitutions are also described below.
[0030] The term "nucleic acid" or "polynucleotide" encompasses any physical string of monomer units that can be corresponded to a string of nucleotides, including a polymer of nucleotides (e.g., a typical DNA or RNA polymer), PNAs, modified oligonucleotides (e.g., oligonucleotides comprising bases that are not typical to biological RNA or DNA in solution, such as 2'-O-methylated oligonucleotides), and the like. A nucleic acid can be e.g., single-stranded or double-stranded. Unless otherwise indicated, a particular nucleic acid sequence of this invention encompasses complementary sequences, in addition to the sequence explicitly indicated.
[0031] The term "gene" is used broadly to refer to any nucleic acid associated with a biological function. Thus, genes include coding sequences and/or the regulatory sequences required for their expression. The term "gene" applies to a specific genomic sequence, as well as to a cDNA or an mRNA encoded by that genomic sequence. Genes also 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.
[0032] "Expression of a gene" or "expression of a nucleic acid" means transcription of DNA into RNA (optionally including modification of the RNA, e.g., splicing), translation of RNA into a polypeptide (possibly including subsequent modification of the polypeptide, e.g., posttranslational modification), or both transcription and translation, as indicated by the context.
[0033] The term "vector" refers to the means by which a nucleic acid 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. Most commonly, the vectors of the present invention are plasmids.
[0034] An "expression vector" is a vector, such as a plasmid, which is capable of promoting expression as well as replication of a nucleic acid incorporated therein. Typically, the nucleic acid to be expressed is "operably linked" to a promoter and/or enhancer, and is subject to transcription regulatory control by the promoter and/or enhancer.
[0035] The term "host cell" means a cell which contains a heterologous nucleic acid, such as a vector, and 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. Exemplary host cells in the context of the invention include HEp-2 cells and Vero cells.
[0036] The term "introduced" when referring to a heterologous or isolated nucleic acid refers to the transfer of a nucleic acid into a eukaryotic or prokaryotic cell where the nucleic acid can be incorporated into the genome of the cell (e.g., chromosome, plasmid, plastid or mitochondrial DNA), converted into an autonomous replicon, or transiently expressed (e.g., transfected mRNA). The term includes such methods as "infection," "transfection," "transformation" and "transduction." In the context of the invention a variety of methods can be employed to introduce nucleic acids into host cells, including electroporation, calcium phosphate precipitation, lipid mediated transfection (lipofection), etc.
[0037] An "open reading frame" or "ORF" is a possible translational reading frame of DNA or RNA (e.g., of a gene), which is capable of being translated into a polypeptide. That is, the reading frame is not interrupted by stop codons. However, it should be noted that the term ORF does not necessarily indicate that the polynucleotide is, in fact, translated into a polypeptide.
[0038] A "polypeptide" is a polymer comprising two or more amino acid residues
(e.g., a peptide or a protein). The polymer can optionally comprise modifications such as glycosylation or the like. The amino acid residues of the polypeptide can be natural or non- natural and can be unsubstituted, unmodified, substituted or modified.
[0039] The term "recombinant" indicates that the material (e.g., a virus, a nucleic acid, or a protein) 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. For example, a "recombinant nucleic acid" is one that is made by recombining nucleic acids, e.g., during cloning, DNA shuffling or other procedures, or by chemical or other mutagenesis; a "recombinant polypeptide" or "recombinant protein" is a polypeptide or protein which is produced by expression of a recombinant nucleic acid; and a "recombinant virus", e.g., a recombinant respiratory syncytial virus, is produced by the expression of a recombinant nucleic acid.
[0040] The term "isolated" refers to a biological material, such as a virus, a nucleic acid or a protein, which is substantially free from components that normally accompany or interact with it in its naturally occurring environment. The isolated material optionally comprises material not found with the material in its natural environment, e.g., a cell. For example, if the material is in its natural environment, such as a cell, the material has been placed at a location in the cell (e.g., genome or genetic element) not native to a material found in that environment. For example, a naturally occurring nucleic acid (e.g., a coding sequence, a promoter, an enhancer, etc.) becomes isolated if it is introduced by non- naturally occurring means to a locus of the genome (e.g., a vector, such as a plasmid or virus vector, or amplicon) not native to that nucleic acid. Such nucleic acids are also referred to as "heterologous" nucleic acids. An isolated virus, for example, is in an environment (e.g., a cell culture system, or purified from cell culture) other than the native environment of wild-type virus (e.g., the nasopharynx of an infected individual).
[0041] The term "chimeric" or "chimera," when referring to a virus, indicates that the virus includes genetic and/or polypeptide components derived from more than one parental viral strain or source. Similarly, the term "chimeric" or "chimera," when referring to a viral protein, indicates that the protein includes polypeptide components (i.e., amino acid subsequences) derived from more than one parental viral strain or source.
[0042] An RSV "having an attenuated phenotype" or an "attenuated" RSV exhibits a substantially lower degree of virulence as compared to a wild-type virus (e.g., one circulating naturally among human hosts). An attenuated RSV typically exhibits a slower growth rate and or a reduced level of replication (e.g., a peak titer, e.g., in cell culture, in an animal model of infection, or in a human vacinee's nasopharynx, that is at least about ten fold, preferably at least about one hundred fold, less than that of a wild-type RSV).
[0043] An "immunologically effective amount" of RSV is an amount sufficient to enhance an individual's (e.g., a human's) own immune response against a subsequent exposure to RSV. Levels of induced immunity can be monitored, e.g., by measuring amounts of neutralizing secretory and/or serum antibodies, e.g., by plaque neutralization, complement fixation, enzyme-linked immunosorbent, or microneutralization assay.
[0044] A "protective immune response" against RSV refers to an immune response exhibited by an individual (e.g., a human) that is protective against serious lower respiratory tract disease (e.g., pneumonia and/or bronchiolitis) when the individual is subsequently exposed to and/or infected with wild-type RSV. In some instances, the wild-type (e.g., naturally circulating) RSV can still cause infection, particularly in the upper respiratory tract (e.g., rhinitis), but it can not cause a serious infection. Typically, the protective immune response results in detectable levels of host engendered serum and secretory antibodies that are capable of neutralizing virus of the same strain and/or subgroup (and possibly also of a different, non-vaccine strain and/or subgroup) in vitro and in vivo.
[0045] As used herein, an "antibody" is a protein comprising one or more polypeptides substantially or partially encoded by immunoglobulin genes or fragments of immunoglobulin genes. The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively. A typical immunoglobulin (antibody) structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kD) and one "heavy" chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively. Antibodies exist as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)'2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond. The F(ab)'2 may be reduced under mild conditions to break the disulfide linkage in the hinge region thereby converting the (Fab')2 dimer into a Fab' monomer. The Fab' monomer is essentially a Fab with part of the hinge region (see, Fundamental Immunology, W.E. Paul, ed., Raven Press, N.Y. (1999), for a more detailed description of other antibody fragments). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such Fab' fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term antibody, as used herein, includes antibodies or fragments either produced by the modification of whole antibodies or synthesized de novo using recombinant DNA methodologies. Antibodies include, e.g., polyclonal antibodies, monoclonal antibodies, multiple or single chain antibodies, including single chain Fv (sFv or scFv) antibodies in which a variable heavy and a variable light chain are joined together (directly or through a peptide linker) to form a continuous polypeptide, and humanized or chimeric antibodies.
[0046] An "antigenome" is a polynucleotide that is complementary (typically, perfectly complementary) to a single-stranded viral (e.g., RSV) genome. Since RSV is a negative-sense RNA virus, the genome is the "antisense" strand, and the antigenome is the "sense" strand that corresponds to mRNA.
[0047] A variety of additional terms are defined or otherwise characterized herein.
DETAILED DESCRIPTION
[0048] The present invention provides the complete polynucleotide sequence of human RSV subgroup B strain 9320. The sequence of a B9320 antigenomic cDNA is listed as SEQ ID NO:l. As will be evident, the RSV genome is an RNA with a polynucleotide sequence complementary to that of SEQ ID NO:l.
[0049] The B9320 genome comprises 10 transcriptional units encoding 11 proteins.
Amino acid sequences of the proteins are also provided: NSl is listed as SEQ ID NO:2, NS2 as SEQ ID NO:3, N as SEQ ID NO:4, P as SEQ ID NO:5, M as SEQ ID NO:6, SH as SEQ ID NO:7, G as SEQ ID NO: 12, F as SEQ ID NO:8, M2-1 as SEQ ID NO:9, M2-2 as SEQ ID NO: 10, and L as SEQ ID NO:ll.
[0050] The invention provides isolated or recombinant polynucleotides and polypeptides comprising the novel B9320 sequences. Recombinant RSV comprising the nucleic acids and or polypeptides (e.g., attenuated recombinant RSV suitable for use in attenuated live viral vaccines) are also provided.
POLYNUCLEOTIDEg OF THE INVENTION
[0051] One aspect of the present invention provides isolated or recombinant nucleic acids comprising a polynucleotide sequence of the invention. Polynucleotide sequences of the invention include the polynucleotide sequence represented by SEQ ID NO: 1, with the caveat that SEQ ID NOs: 14-19, representing limited subsequences of RSV B9320, have been previously described (e.g., in GenBank accession numbers M73544 and S75820; Jin et al. (1998) Virology 251:206-214; and Cheng et al. (2001) Virology 283:59-68). Thus, for example, an isolated or recombinant nucleic acid comprising the polynucleotide sequence of SEQ ID NO:l or a complementary polynucleotide sequence thereof is a favored embodiment of the invention. An isolated or recombinant nucleic acid comprising at least one unique polynucleotide subsequence of SEQ ID NO:l (e.g., a unique coding subsequence) or a complementary polynucleotide sequence thereof, with the proviso that the unique polynucleotide subsequence includes at least one subsequence not included in SEQ ID NOs: 14-19 or a complementary polynucleotide sequence thereof, is another favored embodiment. The unique polynucleotide subsequence can, for example, comprise at least 10 contiguous nucleotides of SEQ ID NO:l or a complementary polynucleotide sequence thereof (e.g., at least 20 contiguous nucleotides, at least 50 contiguous nucleotides, at least 100 contiguous nucleotides, at least 500 contiguous nucleotides, or even at least 1000 contiguous nucleotides).
[0052] In addition to the sequences explicitly provided in the accompanying sequence listing, polynucleotide sequences that are highly related structurally and/or functionally are polynucleotides of the invention. Thus, polynucleotide sequences of the invention include a polynucleotide sequence that hybridizes under stringent conditions over substantially the entire length of the polynucleotide sequence of SEQ ID NO: 1 (or a complementary sequence thereof) with at least 2x a signal to noise ratio (e.g., at least 5x or at least lOx the signal to noise ratio) that the polynucleotide sequence hybridizes to the polynucleotide sequence of SEQ ID NO: 13 or a complementary polynucleotide sequence thereof. Polynucleotide sequences of the invention also include a polynucleotide sequence that hybridizes under stringent conditions over substantially the entire length of a polynucleotide subsequence comprising at least 100 contiguous nucleotides of SEQ ID NO:l or its complementary sequence (e.g., a unique subsequence) with at least 2x a signal to noise ratio (e.g., at least 5x or at least lOx the signal to noise ratio) that the polynucleotide sequence hybridizes to the corresponding subsequence of SEQ ID NO: 13 or a complementary polynucleotide sequence thereof (or, optionally, the corresponding subsequence of a genome of another naturally occurring respiratory syncytial virus or a complementary polynucleotide sequence thereof).
[0053] Similarly, polynucleotide sequences of the invention include a polynucleotide sequence encoding an amino acid sequence or unique subsequence selected from the group consisting of SEQ ID NOs:2-ll or an artificial conservative variation thereof.
[0054] In addition to the polynucleotide sequences of the invention, e.g., listed in
SEQ ID NO:l, polynucleotide sequences that are substantially identical to a polynucleotide of the invention can be used in the compositions and methods of the invention. Substantially identical or substantially similar polynucleotide sequences are defined as polynucleotide sequences that are identical, on a nucleotide by nucleotide basis, with at least a subsequence of a reference polynucleotide, e.g., selected from SEQ ID NO:l. Such polynucleotides can include, e.g., insertions, deletions, and substitutions relative to SEQ ID NO:l. For example, isolated or recombinant nucleic acids comprising polynucleotide sequences (or subsequences) having greater than 97.8% sequence identity to SEQ ID NO:l or a complementary polynucleotide sequence thereof, as determined by BLASTN using default parameters, with the proviso that the polynucleotide sequence includes at least one subsequence not selected from SEQ IP NOs: 14-19, are favored embodiments of the invention. For example, the polynucleotide sequences (or subsequences) can be at least 98.5% (e.g., at least 99.0%, at least 99.5%, or more) identical to SEQ IP NO:l or a complementary polynucleotide sequence thereof.
[0055] The nucleic acids of the invention can be, e.g., single-stranded or double- stranded, and can be, e.g., a PNA (e.g., a cPNA), an RNA, or an artificial nucleic acid (e.g., a peptide nucleic acid). SEQ IP NO: 1 presents the PNA sequence of the antigenomic B9320 cPNA; however, it will be understood that the complementary genomic polynucleotide sequence can readily be determined from SEQ ID NO:l and that U in an RNA sequence corresponds to T in a DNA sequence.
[0056] Nucleic acids of the invention include nucleic acids encoding polypeptides of the invention. In one general class of embodiments, the nucleic acid comprises at least one unique polynucleotide subsequence of SEQ ID NO:l (or a complementary polynucleotide sequence thereof) encoding at least 20 contiguous amino acid residues of any one of SEQ ID NOs:2-12 (e.g., at least 50, at least 100, at least 200, or more contiguous amino acid residues). In one class of embodiments, the unique polynucleotide subsequence comprises at least one complete ORF, preferably at least one complete ORF encoding a polypeptide selected from among SEQ ID NOs: 2-12. In some embodiments, the nucleic acid comprises a plurality of complete open reading frames.
[0057] A nucleic acid of the invention optionally comprises at least one artificially mutated nucleotide, e.g., at least one artificially deleted, inserted, and/or substituted nucleotide (e.g., in a noncoding region, e.g., a C to G change at the fourth position of the antigenomic sequence, and or in a coding region). For example, the nucleic acid can comprise a plurality of artificially mutated nucleotides. The artificially mutated nucleotide(s) can be introduced by site-directed mutagenesis, chemical mutagenesis, or the like.
[0058] In certain embodiments, mutation of the polynucleotide sequence results in alteration of an encoded amino acid sequence. Thus, in one class of embodiments, at least one polypeptide encoded by the nucleic acid comprises at least one deleted, inserted, and or substituted amino acid residue (e.g., at least one conservatively or non-conservatively substituted amino acid residue). For example, the mutated nucleotide can be located in an ORF encoding a polypeptide selected from SEQ ID NOs:2-12. Thus, in one class of example embodiments, the at least one artificially mutated nucleotide is located in the open reading frame encoding the polypeptide of SEQ ID NO: 12. The artificially mutated nucleotide(s) can comprise, e.g., a deletion, e.g., a deletion resulting in a deletion of one or more amino acid residues from the G protein encoded by SEQ ID NO: 12 (e.g., a deletion of residues 164-197), or a deletion resulting in a deletion of the open reading frame encoding G. In another class of example embodiments, the at least one artificially mutated nucleotide is located in the open reading frame encoding the polypeptide of SEQ ID NO: 10. The artificially mutated nucleotide(s) can comprise, e.g., a deletion, e.g., a deletion resulting in a deletion of one or more amino acid residues from the M2-2 protein encoded by SEQ ID NO: 10, or a deletion resulting in a deletion of the open reading frame encoding M2-2. As another example, at least one of the nucleotides encoding amino acid residue 1, amino acid residue 4 and or amino acid residue 10 of M2-2 can be mutated (e.g., substituted or deleted, e.g., forcing use of the second and or third start codon and resulting in a deletion of amino acid residues 1-3 or 1-9 of M2-2). [0059] The nucleic acids of the invention include chimeric nucleic acids, for example, a nucleic acid comprising at least one subsequence of SEQ ID NO:l or a complementary polynucleotide sequence thereof and at least one polynucleotide subsequence from a different strain of virus. The subsequence of SEQ ID NO:l is preferably a unique polynucleotide subsequence that comprises at least 10 contiguous nucleotides of SEQ ID NO:l or its complement and that includes at least one subsequence not included in SEQ ID NOs: 14-19 or a complementary polynucleotide sequence thereof. The different strain of virus can be, e.g., a different strain of human RSV (e.g., A2, BI, or the like) or a different species of virus (e.g., another paramyxovirus, e.g., pneumonia virus of mice, bovine RSV, or metapneumo virus). Such chimeric nucleic acids can, for example, encode chimeric proteins and/or chimeric viruses (e.g., for use in vaccines to induce a protective immune response against one or more strains of RSV and/or another virus). For example, in certain embodiments, the nucleic acid comprises at least one complete open reading frame of SEQ ID NO:l and at least one complete open reading frame of the different strain of virus.
[0060] Another class of embodiments provides vectors comprising the nucleic acids of the invention. Yet another class of embodiments provides a host cell into which such a vector has been introduced.
POLYPEPTIDES OF THE INVENTION
[0061] One aspect of the present invention provides RSV B9320 polypeptides and variants thereof, for example, a polypeptide comprising an amino acid sequence or subsequence that is encoded by a nucleic acid of the invention, with the proviso that the amino acid sequence or subsequence is not encoded by SEQ ID NO: 14.
[0062] One general class of embodiments provides isolated or recombinant polypeptides comprising an amino acid sequence of the invention. Thus, for example, an isolated or recombinant polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:2-l 1 is a favored embodiment of the invention. An isolated or recombinant polypeptide comprising a unique amino acid subsequence comprising at least 7 (e.g., at least 8, at least 10, at least 20, at least 50, or more) contiguous amino acid residues of any one of SEQ IP NOs:2-ll is another favored embodiment. Artificial conservative variations of amino acid sequences or subsequences of the invention are also amino acid sequences of the invention. Such polypeptides are optionally immunogenic.
[0063] In addition to the amino acid sequences of the invention, e.g., listed in SEQ
ID NOs:2-ll, amino acid sequences that are substantially identical to an amino acid sequence of the invention can be used in the compositions and methods of the invention. Substantially identical or substantially similar polypeptide sequences are defined as amino acid sequences that are identical, on an amino acid by amino acid basis, with at least a subsequence of a reference polypeptide, e.g., selected from among SEQ ID NOs:2-ll. Such amino acid sequences can include, e.g., insertions, deletions, and substitutions relative to SEQ ID NOs:2-ll. For example, an isolated or recombinant polypeptide comprising an amino acid sequence that is greater than 99.3% identical to SEQ ID NO:2, an amino acid sequence that is greater than 98.4% identical to SEQ ID NO:3, an amino acid sequence that is greater than 99.7% identical to SEQ ID NO:4, an amino acid sequence that is greater than 98.3% identical to SEQ ID NO:5, an amino acid sequence that is greater than 99.6% identical to SEQ ID NO:6, an amino acid sequence that is greater than 97.0% identical to SEQ ID NO:7, an amino acid sequence that is greater than 99.3% identical to SEQ ID NO:8, an amino acid sequence that is greater than 99.5% identical to SEQ ID NO:9, an amino acid sequence that is greater than 96.4% identical to SEQ ID NO: 10, or an amino acid sequence that is greater than 99.2% identical to SEQ ID NO: 11, as determined by BLASTP using default parameters, is a favored embodiment of the invention. For example, the isolated or recombinant polypeptide can comprise an amino acid sequence (or subsequence) that is at least 99.5% identical to SEQ ID NO:2, at least 98.6% identical to SEQ ID NO:3, at least 99.9% identical to SEQ ID NO:4, at least 98.5% identical to SEQ ID NO:5, at least 99.8% identical to SEQ ID NO:6, at least 97.2% identical to SEQ ID NO:7, at least 99.5% identical to SEQ ID NO:8, at least 99.7% identical to SEQ ID NO:9, at least 96.6% identical to SEQ ID NO: 10, or at least 99.4% identical to SEQ ID NO: 11, as determined by BLASTP using default parameters.
[0064] A polypeptide of the invention optionally comprises at least one artificially altered amino acid, e.g., at least one deleted, inserted, and/or substituted amino acid. For example, the polypeptide can comprise a plurality of artificially altered amino acids. [0065] One class of embodiments provides an isolated or recombinant polypeptide comprising the amino acid sequence of SEQ ID NO: 12 with a deletion of residues 164-197, or an artificial conservative variation thereof.
[0066] Methods of producing isolated or recombinant polypeptides form another aspect of the invention. In the methods, a host cell into which a vector (e.g., an expression vector, e.g., an expression vector comprising one or more ORFs, or a vector comprising an entire viral genome or antigenome) comprising a nucleic acid of the invention has been introduced is cultured in a suitable culture medium under conditions permitting expression of the nucleic acid. The polypeptide is then isolated from the host cell and/or the medium. For example, the polypeptide can be purified from the host cell and/or the medium such that the resulting purified polypeptide is enriched at least 5x as compared to its initial state. Polypeptides produced according to the methods described herein are also features of the invention. Such polypeptides can comprise, e.g., subsequences (e.g., unique subsequences, immunogenic subsequences, etc.) of SEQ ID NOs:2-ll from a few amino acids (e.g., 7 or more, 10 or more, or 20 or more) up to the full length proteins.
DETERMINING SEQUENCE RELATIONSHIPS
[0067] A variety of methods for determining relationships (e.g., identity, similarity and/or homology) between two or more sequences, such as SEQ ID NO:l and SEQ ID NO.T3, are available and well known in the art. The methods include manual alignment, computer assisted sequence alignment, and combinations thereof. A number of algorithms (which are generally computer implemented) for performing sequence alignment are widely available, or can be produced by one of skill. These methods include, e.g., the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math. 2:482; the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443; the search for similarity method of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. (USA) 85:2444; and or by computerized implementations of these algorithms (e.g., GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package Release 7.0, Genetics Computer Group, 575 Science Dr., Madison, WI).
[0068] For example, software for performing sequence identity (and sequence similarity) analysis using the BLAST algorithm is described in Altschul et al. (1990) J. Mol. Biol. 215:403-410. This software is publicly available, e.g., through the National Center for Biotechnology Information on the world wide web at ncbi.nlm.nih.gov. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold. These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always < 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, an expectation (E) of 10, a cutoff of 100, M=5, N=-4, and a comparison of both strands. For amino acid sequences, the BLASTP (BLAST Protein) program uses as defaults a wordlength (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix (see, Henikoff and Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915).
[0069] Additionally, the BLAST algorithm performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Nat'l. Acad. Sci. USA 90:5873-5787). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence (and, therefore, in this context, homologous) if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, or less than about 0.01, and or even less than about 0.001.
Another example of a useful sequence alignment algorithm is PILEUP. PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments. It can also plot a tree showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng and Doolittle (1987) J. Mol. Evol. 35:351-360. The method used is similar to the method described by Higgins and Sharp (1989) CAB/OS5:151-153. The program can align, e.g., up to 300 sequences of a maximum length of 5,000 letters. The multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences. This cluster can then be aligned to the next most related sequence or cluster of aligned sequences. Two clusters of sequences can be aligned by a simple extension of the pairwise alignment of two individual sequences. The final alignment is achieved by a series of progressive, pairwise alignments. The program can also be used to plot a dendogram or tree representation of clustering relationships. The program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison.
[0071] An additional example of an algorithm that is suitable for multiple DNA, or amino acid, sequence alignments is the CLUSTALW program (Thompson, J. D. et al. (1994) Nucl. Acids. Res. 22: 4673-4680). CLUSTALW performs multiple pairwise comparisons between groups of sequences and assembles them into a multiple alignment based on homology. Gap open and Gap extension penalties can be, e.g., 10 and 0.05 respectively. For amino acid alignments, the BLOSUM algorithm can be used as a protein weight matrix. See, e.g., Henikoff and Henikoff (1992) Proc. Natl. Acad. Sci. USA 89: 10915-10919.
NUCLEIC ACID HYBRIDIZATION
[0072] Similarity between nucleic acids can also be evaluated by "hybridization" between single stranded (or single stranded regions of) nucleic acids with complementary or partially complementary polynucleotide sequences. Hybridization is a measure of the physical association between nucleic acids, typically, in solution, or with one of the nucleic acid strands immobilized on a solid support, e.g., a membrane, a bead, a chip, a filter, etc. Nucleic acid hybridization occurs based on a variety of well characterized physico-chemical forces, such as hydrogen bonding, solvent exclusion, base stacking and the like. Numerous protocols for nucleic acid hybridization are well known in the art. An extensive guide to the hybridization of nucleic acids is found in Tijssen (1993) Laboratory Techniques in Biochemistry and Molecular Biology— Hybridization with Nucleic Acid Probes, part I, chapter 2, "Overview of principles of hybridization and the strategy of nucleic acid probe assays," (Elsevier, New York), as well as in Ausubel et al. Current Protocols in Molecular Biology (supplemented through 2003) John Wiley & Sons, New York ("Ausubel"); Sambrook et al. Molecular Cloning - A Laboratory Manual (3rd Ed.), Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 2001 ("Sambrook"), and Berger and Kimmel Guide to Molecular Cloning Techniques, Methods in Enzymology volume 152 Academic Press, Inc., San Diego, CA ("Berger"). Hames and Higgins (1995) Gene Probes ! IRL Press at Oxford University Press, Oxford, England (Hames and Higgins 1) and Hames and Higgins (1995) Gene Probes 2, IRL Press at Oxford University Press, Oxford, England (Hames and Higgins 2) provide details on the synthesis, labeling, detection and quantification of DNA and RNA, including oligonucleotides.
[0073] Conditions suitable for obtaining hybridization, including differential hybridization, are selected according to the theoretical melting temperature (Tm) between complementary and partially complementary nucleic acids. Under a given set of conditions, e.g., solvent composition, ionic strength, etc., the Tm is the temperature at which the duplex between the hybridizing nucleic acid strands is 50% denatured. That is, the Tm corresponds to the temperature corresponding to the midpoint in transition from helix to random coil; it depends on length, nucleotide composition, and ionic strength for long stretches of nucleotides.
[0074] After hybridization, unhybridized nucleic acids can be removed by a series of washes, the stringency of which can be adjusted depending upon the desired results. Low stringency washing conditions (e.g., using higher salt and lower temperature) increase sensitivity, but can produce nonspecific hybridization signals and high background signals. Higher stringency conditions (e.g., using lower salt and higher temperature that is closer to the Tm) lower the background signal, typically with primarily the specific signal remaining. See, also, Rapley, R. and Walker, J.M. eds., Molecular Biomethods Handbook (Humana Press, Inc. 1998).
[0075] "Stringent hybridization wash conditions" or "stringent conditions" in the context of nucleic acid hybridization experiments, such as Southern and northern hybridizations, are sequence dependent, and are different under different environmental parameters. An extensive guide to the hybridization of nucleic acids is found in Tijssen (1993), supra, and in Hames and Higgins 1 and Hames and Higgins 2, supra. [0076] An example of stringent hybridization conditions for hybridization of complementary nucleic acids which have more than 100 complementary residues on a filter in a Southern or northern blot is 2x SSC, 50% formamide at 42°C, with the hybridization being carried out overnight (e.g., for approximately 20 hours). An example of stringent wash conditions is a 0.2x SSC wash at 65°C for 15 minutes (see Sambrook, supra for a description of SSC buffer). Often, the wash determining the stringency is preceded by a low stringency wash to remove signal due to residual unhybridized probe. An example low stringency wash is 2x SSC at room temperature (e.g., 20°C for 15 minutes).
[0077] In general, a signal to noise ratio of at least 2x (or higher, e.g., at least 5x, lOx, 20x, 50x, lOOx, or more) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization. Detection of at least stringent hybridization between two sequences in the context of the present invention indicates relatively strong structural similarity to, e.g., the nucleic acids of the present invention provided in the sequence listings herein.
[0078] For purposes of the present invention, generally, "highly stringent" hybridization and wash conditions are selected to be about 5° C or less lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH (as noted below, highly stringent conditions can also be referred to in comparative terms). Target sequences that are closely related or identical to the nucleotide sequence of interest (e.g., "probe") can be identified under stringent or highly stringent conditions. Lower stringency conditions are appropriate for sequences that are less complementary.
[0079] For example, in determining stringent or highly stringent hybridization (or even more stringent hybridization) and wash conditions, the hybridization and wash conditions are gradually increased (e.g., by increasing temperature, decreasing salt concentration, increasing detergent concentration and/or increasing the concentration of organic solvents, such as formamide, in the hybridization or wash), until a selected set of criteria are met. For example, the hybridization and wash conditions are gradually increased until a probe comprising one or more polynucleotide sequences of the invention, e.g., sequences or unique subsequences selected from SEQ ID NO:l and/or complementary polynucleotide sequences, binds to a perfectly matched complementary target (again, a nucleic acid comprising one or more nucleic acid sequences or subsequences selected from SEQ ID NO:l and/or complementary polynucleotide sequences thereof), with a signal to noise ratio that is at least 2x (and optionally 5x, lOx, or lOOx or more) as high as that observed for hybridization of the probe to an unmatched target (e.g., a polynucleotide sequence comprising the corresponding one or more sequences or subsequences selected from SEQ ID NO: 13 and/or complementary polynucleotide sequences thereof), as desired. Preferably, the sequences or subsequences are selected from a portion of SEQ ID NO:l that includes at least a subsequence that is not included in SEQ ID NOs: 14-19
[0080] Using the polynucleotides of the invention, or subsequences thereof, novel target nucleic acids can be obtained; such target nucleic acids are also a feature of the invention. For example, such target nucleic acids include sequences that hybridize under stringent conditions to a unique oligonucleotide probe corresponding to any of the polynucleotides of the invention, e.g., SEQ ID NO:l.
[0081] Higher ratios of signal to noise can be achieved by increasing the stringency of the hybridization conditions such that ratios of about 15x, 20x, 30x, 50x or more are obtained. The particular signal will depend on the label used in the relevant assay, e.g., a fluorescent label, a colorimetric label, a radioactive label, or the like.
[0082] Nucleic acids which do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides which they encode are substantially identical. This occurs, e.g., when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code.
DEFINING PROTEINS BY IMMUNOREACTIVITY
[0083] Because the polypeptides of the invention provide a variety of new polypeptide sequences, the polypeptides also provide new structural features which can be recognized, e.g., in immunological assays. The generation of antibodies or antisera which specifically bind the polypeptides of the invention, as well as the polypeptides which are bound by such antibodies or antisera,^ and the antibodies or antisera themselves, are a feature of the invention.
[0084] Thus, the proteins of the invention can also be identified by immunoreactivity; e.g., a polypeptide of the invention can include an amino acid sequence or subsequence that is specifically bound by an antibody that specifically binds to an amino acid sequence or subsequence encoded by SEQ ID NO:l, wherein the antibody does not specifically bind to an amino acid sequence or subsequence encoded by SEQ ID NO: 13 or SEQ ID NO: 14 (or, optionally, to an amino acid sequence or subsequence encoded by the genome of another naturally occurring respiratory syncytial virus).
[0085] Methods of producing antibodies, performing immunoassays, and the like are well known in the art. See e.g., the section entitled "Antibodies" below and references therein.
[0086] In one typical format, an immunoassay to identify a polypeptide of the invention uses a polyclonal antiserum which was raised against one or more of the RSV 9320 polypeptides of the invention (e.g., a polypeptide comprising SEQ ID NOs:2-ll or SEQ ID NO: 12 with a deletion of residues 164-197), or a substantial subsequence thereof (i.e., at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 98% or more of the full length sequence provided). The full set of potential polypeptide immunogens derived from one or more of the RSV 9320 polypeptides of the invention are collectively referred to below as "the immunogenic polypeptides." The resulting antisera is optionally selected to have low cross-reactivity against the control RSV BI polypeptides and/or other known, e.g., naturally occurring, RSV polypeptides, and any such cross-reactivity is removed by immunoabsorption with one or more of the control RSV polypeptides, prior to use of the polyclonal antiserum in the immunoassay.
[0087] In order to produce antisera for use in an immunoassay, one or more of the immunogenic polypeptides is produced and purified as described herein. For example, recombinant protein can be produced in a mammalian cell line. An inbred strain of mice (used in this assay because results are more reproducible due to the virtual genetic identity of the mice) is immunized with the immunogenic polypeptide(s) in combination with a standard adjuvant, such as Freund's adjuvant, and a standard mouse immunization protocol (see Harlow and Lane (1988) Antibodies, A Laboratory Manual Cold Spring Harbor Press, New York, for a standard description of antibody generation, immunoassay formats and conditions that can be used to determine specific immunoreactivity). Alternatively, one or more synthetic or recombinant polypeptides derived from the sequences disclosed herein is conjugated to a carrier protein and used as an immunogen.
[0088] Polyclonal sera are collected and titered against the immunogenic polyρeptide(s) in an immunoassay, for example, a solid phase immunoassay with one or more of the immunogenic polypeptides immobilized on a solid support. Polyclonal antisera with a titer of 106 or greater are selected, pooled and subtracted with the control RSV polypeptides to produce subtracted pooled titered polyclonal antisera.
[0089] The subtracted pooled titered polyclonal antisera are tested for cross reactivity against the control RSV polypeptides. Preferably at least two of the immunogenic RSV 9320 polypeptides are used in this determination, preferably in conjunction with at least two of the control RSV polypeptides, to identify antibodies which are specifically bound by the immunogenic polypeptides(s).
[0090] In this comparative assay, discriminatory binding conditions are determined for the subtracted titered polyclonal antisera which result in at least about a 5-10 fold higher signal to noise ratio for binding of the titered polyclonal antisera to the immunogenic RSV 9320 polypeptides as compared to binding to the control RSV polypeptides. That is, the stringency of the binding reaction is adjusted by the addition of non-specific competitors, such as albumin or non-fat dry milk, or by adjusting salt conditions, temperature, or the like. These binding conditions are used in subsequent assays for determining whether a test polypeptide is specifically bound by the pooled subtracted polyclonal antisera. In particular, a test polypeptide which shows at least a 2-5x higher signal to noise ratio than the control polypeptides under discriminatory binding conditions, and at least about a V_ signal to noise ratio as compared to the immunogenic polypeptide(s), shares substantial structural similarity or homology with the immunogenic polypeptide(s) as compared to the control RSV polypeptides, and is, therefore, a polypeptide of the invention.
[0091] In another example, immunoassays in the competitive binding format are used for detection of a test polypeptide. For example, as noted, cross-reacting antibodies are removed from the pooled antisera mixture by immunoabsorption with the control RSV polypeptides. The immunogenic polypeptide(s) are then immobilized to a solid support which is exposed to the subtracted pooled antisera. Test proteins are added to the assay to compete for binding to the pooled subtracted antisera. The ability of the test protein(s) to compete for binding to the pooled subtracted antisera as compared to the immobilized protein(s) is compared to the ability of the immunogenic polypeptide(s) added to the assay to compete for binding (the immunogenic polypeptides compete effectively with the immobilized immunogenic polypeptides for binding to the pooled antisera). The percent cross-reactivity for the test proteins is calculated, using standard calculations. [0092] In a parallel assay, the ability of the control proteins to compete for binding to the pooled subtracted antisera is determined as compared to the ability of the immunogenic polypeptide(s) to compete for binding to the antisera. Again, the percent cross-reactivity for the control polypeptides is calculated, using standard calculations. Where the percent cross-reactivity is at least 5-10x as high for the test polypeptides, the test polypeptides are said to specifically bind the pooled subtracted antisera, and are, therefore, polypeptides of the invention.
[0093] In general, the immunoabsorbed and pooled antisera can be used in a competitive binding immunoassay as described herein to compare any test polypeptide to the immunogenic polypeptide(s). In order to make this comparison, the two polypeptides are each assayed at a wide range of concentrations and the amount of each polypeptide required to inhibit 50% of the binding of the subtracted antisera to the immobilized protein is determined using standard techniques. If the amount of the test polypeptide required is less than twice the amount of the immunogenic polypeptide that is required, then the test polypeptide is said to specifically bind to an antibody generated to the immunogenic polypeptide, provided the amount is at least about 5-1 Ox as high as for a control polypeptide.
[0094] As a final determination of specificity, the pooled antisera is optionally fully immunosorbed with the immunogenic polypeρtide(s) (rather than the control polypeptides) until little or no binding of the resulting immunogenic polypeptide subtracted pooled antisera to the immunogenic polypeptide(s) used in the immunoabsorption is detectable. This fully immunosorbed antisera is then tested for reactivity with the test polypeptide. If little or no reactivity is observed (i.e., no more than 2x the signal to noise ratio observed for binding of the fully immunosorbed antisera to the immunogenic polypeptide), then the test polypeptide is specifically bound by the antisera elicited by the immunogenic protein.
SEQUENCE VARIATIONS
Silent Variations [0095] Due to the degeneracy of the genetic code, any of a variety of nucleic acid sequences encoding polypeptides and/or viruses of the invention are optionally produced, some which can bear lower levels of sequence identity to the RSV nucleic acid and polypeptide sequences in the figures. The following provides a typical codon table specifying the genetic code, found in many biology and biochemistry texts.
Table 1
Codon Table
Amino acids Codon
Alanine Ala A GCA GCC GCG GCU
Cysteine Cys C UGC UGU
Aspartic acid Asp D GAC GAU
Glutamic acid Glu E GAA GAG
Phenylalanine Phe F UUC UUU
Glycine Gly G GGA GGC GGG GGU
Histidine His H CAC CAU
Isoleucine He I AUA AUC AUU
Lysine Lys K AAA AAG
Leucine Leu L UUA UUG CUA CUC CUG CUU
Methionine Met M AUG
Asparagine Asn N AAC AAU
Proline Pro P CCA CCC CCG CCU
Glutamine Gin Q CAA CAG
Arginine Arg R AGA AGG CGA CGC CGG CGU
Serine Ser S AGC AGU UCA UCC UCG UCU
Threonine Thr T ACA ACC ACG ACU
Valine Val V GUA GUC GUG GUU
Tryptophan Tip W UGG
Tyrosine Tyr Y UAC UAU
[0096] The codon table shows that many amino acids are encoded by more than one codon. For example, the codons AGA, AGG, CGA, CGC, CGG, and CGU all encode the amino acid arginine. Thus, at every position in the nucleic acids of the invention where an arginine is specified by a codon, the codon can be altered to any of the corresponding codons described above without altering the encoded polypeptide. It is understood that U in an RNA sequence corresponds to T in a DNA sequence.
[0097] As an example, a nucleic acid sequence corresponding to the amino acid sequence FEV (residues 164-166 of SEQ ID NO: 12) is TTTGAAGTG. A silent variation of this sequence includes TTCGAGGTA (also encoding FEV).
[0098] Such "silent variations" are one species of "conservatively modified variations", discussed below. One of skill will recognize that each codon in a nucleic acid (except ATG, which is ordinarily the only codon for methionine, and TTG, which is ordinarily the only codon for tryptophan) can be modified by standard techniques to encode a functionally identical polypeptide. Accordingly, each silent variation of a nucleic acid which encodes a polypeptide is implicit in any described sequence. The invention, therefore, explicitly provides each and every possible variation of a nucleic acid sequence encoding a polypeptide of the invention that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code (e.g., as set forth in Table 1, or as is commonly available in the art) as applied to the nucleic acid sequence encoding an RSV polypeptide of the invention. All such variations of every nucleic acid herein are specifically provided and described by consideration of the sequence in combination with the genetic code. One of skill is fully able to make these silent substitutions using the methods herein.
Conservative Variations [0099] "Conservatively modified variations" or, simply, "conservative variations" of a particular nucleic acid sequence or polypeptide are those which encode identical or essentially identical amino acid sequences. One of skill will recognize that individual substitutions, deletions or additions which alter, add or delete a single amino acid or a small percentage of amino acids (typically less than 5%, more typically less than 4%, 2% or 1%) in an encoded sequence are "conservatively modified variations" where the alterations result in the deletion of an amino acid, addition of an amino acid, or substitution of an amino acid with a chemically similar amino acid.
[0100] Conservative substitution tables providing functionally similar amino acids are well known in the art. Table 2 sets forth six groups which contain amino acids that are "conservative substitutions" for one another. Alternative conservative substitution charts are available in the art and can be used in a similar manner.
Table 2
Conservative Substitution Groups
1 Alanine (A) Serine (S) Threonine (T)
2 Aspartic acid (D) Glutamic acid (E)
3 Asparagine (N) Glutarnine (Q)
4 Arginine (R) Lysine (K)
5 Isoleucine (I) Leucine (L) Methionine (M) Valine (V)
6 Phenylalanine (F) Tyrosine (Y) Tryptophan (W)
[0101] Thus, "conservatively substituted variations" of a polypeptide sequence of the present invention include substitutions of a small percentage, typically less than 5%, more typically less than 2% or 1%, of the amino acids of the polypeptide sequence, with a conservatively selected amino acid of the same conservative substitution group.
[0102] For example, a conservatively substituted variation of the RSV strain B9320
M2-1 polypeptide in SEQ ID NO:9 will contain "conservative substitutions", e.g., according to the six groups defined above, in up to about 10 residues (i.e., about 5% of the amino acids) in the full-length polypeptide.
[0103] In a further example, if conservative substitutions were localized in the region corresponding to amino acids 10-12 of RSV 9320 M2-1 (EJR), examples of conservatively substituted variations of this region include conservative substitutions of DLK or DMR (or any others that can be made according to Table 2) for EIR.
[0104] Listing of a protein sequence herein, in conjunction with the above substitution table, provides an express listing of all conservatively substituted proteins.
[0105] Finally, the addition or deletion of sequences which do not alter the encoded activity of a nucleic acid molecule, such as the addition or deletion of a non-functional sequence, an epitope tag, a polyhistidine tag, GFP, or the like, is a conservative variation of the basic nucleic acid or polypeptide.
[0106] One of skill will appreciate that many conservative variations of the nucleic acid constructs which are disclosed yield a functionally identical construct. For example, as discussed above, owing to the degeneracy of the genetic code, "silent substitutions" (i.e., substitutions in a nucleic acid sequence which do not result in an alteration in an encoded polypeptide) are an implied feature of every nucleic acid sequence which encodes an amino acid. Similarly, "conservative amino acid substitutions," in which one or a few amino acids in an amino acid sequence are substituted with different amino acids with highly similar properties, are also readily identified as being highly similar to a disclosed construct. Such conservative variations of each disclosed or claimed virus, nucleic acid or protein are a feature of the present invention. Such conservative (e.g., silent) variations can be used, e.g., to produce antibodies for detection of or immunoprotection against RSV.
PRODUCTION OF VIRAL NUCLEIC ACIDS
[0107] In the context of the invention, viral (e.g., RSV) 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 2003) John Wiley & Sons, New York ("Ausubel"); Sambrook et al. Molecular Cloning - A Laboratory Manual (3rd Ed.), Vol. 1- 3, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 2001 ("Sambrook"), and Berger and Kimmel Guide to Molecular Cloning Techniques, Methods in Enzymology volume 152 Academic Press, Inc., San Diego, CA ("Berger").
[0108] 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 polynucleotides of the invention, are found in Mullis et al. (1987) U.S. Patent No. 4,683,202; PCR Protocols A Guide to Methods and Applications (Innis et al. eds) Academic Press Inc. San Diego, CA (1990) ("Innis"); Arnheim and Levinson (1990) C&EN 36: The Journal Of NM 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: Lomell 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.
[0109] 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 Enzvmol 211:3.
[0110] 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 (www.mcrc.com), The Great American Gene Company (www.genco.com), ExpressGen, Inc. (www.expressgen.com), QIAGEN (http://oligos.qiagen.com), and many others.
[0111] 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 and or temperature sensitivity, can be produced by introducing specific mutations into a viral nucleic acid segment (e.g., a cDNA) encoding the polypeptide. Methods for site directed mutagenesis are well known in the art, and are described, e.g., in Ausubel, Sambrook, and Berger, supra. Numerous kits for performing site directed mutagenesis are commercially available, e.g., the ExSite and Chameleon™ site directed mutagenesis kits (Stratagene, La Jolla), and can be used according to the manufacturer's instructions to introduce, e.g., one or more nucleotide substitutions specifying one or more amino acid substitutions into an RSV polynucleotide.
[0112] Various types of mutagenesis are optionally used in the present invention, e.g., to modify nucleic acids and encoded polypeptides and/or viruses to produce conservative or non-conservative variants. Any available mutagenesis procedure can be used. Such mutagenesis procedures optionally include selection of mutant nucleic acids and polypeptides for one or more activity of interest. Procedures that can be used include, but are not limited to: site-directed point mutagenesis, random point mutagenesis, in vitro or in vivo homologous recombination (DNA shuffling), mutagenesis using uracil containing templates, oligonucleotide-directed mutagenesis, phosphorothioate-modified DNA mutagenesis, mutagenesis using gapped duplex DNA, point mismatch repair, mutagenesis using repair-deficient host strains, restriction-selection and restriction-purification, deletion mutagenesis, mutagenesis by total gene synthesis, double-strand break repair, and many others known to persons of skill. In one embodiment, mutagenesis can be guided by information known about the naturally occurring molecule or altered or mutated naturally occurring molecules, e.g., sequence, sequence comparisons, physical properties, crystal structure or the like. In another class of embodiments, modification is essentially random (e.g., as in classical DNA shuffling).
[0113] Several of these procedures are set forth in Sambrook and Ausubel, herein.
Additional information on these procedures is found in the following publications and the references cited therein: Arnold (1993) "Protein engineering for unusual environments" Current Opinion in Biotechnology 4:450-455; Bass et al. (1988) "Mutant Trp repressors with new DNA-binding specificities" Science 242:240-245; Botstein and Shortle (1985) "Strategies and applications of in vitro mutagenesis" Science 229:1193-1201; Carter et al. (1985) "Improved oligonucleotide site-directed mutagenesis using M13 vectors" Nucl. Acids Res. 13: 4431-4443; Carter (1986) "Site-directed mutagenesis" Biochem. J. 237:1-7; Carter (1987) "Improved oligonucleotide-directed mutagenesis using M13 vectors" Methods in Enzymol. 154: 382-403; Dale et al. (1996) "Oligonucleotide-directed random mutagenesis using the phosphorothioate method" Methods Mol. Biol. 57:369-374; Eghtedarzadeh and Henikoff (1986) "Use of oligonucleotides to generate large deletions" Nucl. Acids Res. 14: 5115; Fritz et al. (1988) "Oligonucleotide-directed construction of mutations: a gapped duplex DNA procedure without enzymatic reactions in vitro" Nucl. Acids Res. 16: 6987-6999; Grundstrδm et al. (1985) "Oligonucleotide-directed mutagenesis by microscale 'shot-gun' gene synthesis" Nucl. Acids Res. 13: 3305-3316; Kunkel (1987) "The efficiency of oligonucleotide directed mutagenesis" in Nucleic Acids and Molecular Biology (Eckstein, F. and Lilley, D.M.J. eds., Springer Verlag, Berlin)); Kunkel (1985) "Rapid and efficient site-specific mutagenesis without phenotypic selection" Proc. Natl. Acad. Sci. USA 82:488-492; Kunkel et al. (1987) "Rapid and efficient site-specific mutagenesis without phenotypic selection" Methods in Enzymol. 154, 367-382; Kramer et al. (1984) "The gapped duplex DNA approach to oligonucleotide-directed mutation construction" Nucl. Acids Res. 12: 9441-9456; Kramer and Fritz (1987) "Oligonucleotide- directed construction of mutations via gapped duplex DNA" Methods in Enzymol. 154:350- 367; Kramer et al. (1984) "Point Mismatch Repair" CeU 38:879-887; Kramer et al. (1988) "Improved enzymatic in vitro reactions in the gapped duplex DNA approach to oligonucleotide-directed construction of mutations" Nucl. Acids Res. 16: 7207; Ling et al. (1997) "Approaches to DNA mutagenesis: an overview" Anal Biochem. 254(2): 157-178; Lorimer and Pastan (1995) Nucleic Acids Res. 23, 3067-8; Mandecki (1986) "Oligonucleotide-directed double-strand break repair in plasmids of Escherichia coli: a method for site-specific mutagenesis" Proc. Natl. Acad. Sci. USA 83:7177-7181; Nakamaye and Eckstein (1986) "Inhibition of restriction endonuclease Nci I cleavage by phosphorothioate groups and its application to oligonucleotide-directed mutagenesis" Nucl. Acids Res. 14: 9679-9698; Nambiar et al. (1984) "Total synthesis and cloning of a gene coding for the ribonuclease S protein" Science 223: 1299-1301; Sakamar and Khorana (1988) "Total synthesis and expression of a gene for the a-subunit of bovine rod outer segment guanine nucleotide-binding protein (transducin)" Nucl. Acids Res. 14: 6361-6372; Sayers et al. (1988) "Y-T Exonucleases in phosphorothioate-based oligonucleotide-directed mutagenesis" Nucl. Acids Res. 16:791-802: Sayers et al. (1988) "Strand specific cleavage of phosphorothioate-containing DNA by reaction with restriction endonucleases in the presence of ethidium bromide" Nucl. Acids Res. 16: 803-814; Sieber et al.(2001) Nature Biotechnology 19:456-460; Smith (1985) "In vitro mutagenesis" Ann. Rev. Genet. 19:423- 462; Methods in Enzymol. 100: 468-500 (1983); Methods in Enzymol. 154: 329-350 (1987); Stemmer (1994) Nature 370, 389-91; Taylor et al. (1985) "The use of phosphorothioate-modified DNA in restriction enzyme reactions to prepare nicked DNA" Nucl. Acids Res. 13: 8749-8764; Taylor et al. (1985) "The rapid generation of oligonucleotide-directed mutations at high frequency using phosphorothioate-modified DNA" Nucl. Acids Res. 13: 8765-8787; Wells et al. (1986) "Importance of hydrogen-bond formation in stabilizing the transition state of subtilisin" Phil. Trans. R. Soc. Lond. A 317: 415-423; Wells et al. (1985) "Cassette mutagenesis: an efficient method for generation of multiple mutations at defined sites" Gene 34:315-323; Zoller and Smith (1982) "Oligonucleotide-directed mutagenesis using M13-derived vectors: an efficient and general procedure for the production of point mutations in any DNA fragment" Nucleic Acids Res. 10:6487-6500; Zoller and Smith (1983) "Oligonucleotide-directed mutagenesis of DNA fragments cloned into M13 vectors" Methods in Enzymol. 100:468-500; and Zoller and Smith (1987) "Oligonucleotide-directed mutagenesis: a simple method using two oligonucleotide primers and a single-stranded DNA template" Methods in Enzymol. 154:329-350. Additional details on many of the above methods can be found in Methods in Enzymology Volume 154, which also describes useful controls for trouble-shooting problems with various mutagenesis methods.
VECTORS. PROMOTERS AND EXPRESSION SYSTEMS
[0114] The present invention includes recombinant constructs incorporating one or more of the nucleic acid sequences described above. Such constructs include a vector, for example, a plasmid, a cosmid, a phage, a virus, a bacterial artificial chromosome (BAG), a yeast artificial chromosome (YAC), etc., into which one or more of the polynucleotide sequences of the invention, for example, SEQ ID NO:l or subsequences thereof, e.g., including at least one ORF selected from SEQ ID NO:l, has been inserted, in a forward or reverse orientation. For example, the inserted nucleic acid can include all or part of at least one of the polynucleotide sequences of the invention. Typically the vector is selected based on the characteristics, e.g., size of the selected polynucleotide sequence, and on the intended use, e.g., expression, amplification, etc. In a preferred embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art and are commercially available.
[0115] The polynucleotides of the present invention can be included in any one of a variety of vectors suitable for generating sense or antisense RNA, and optionally, polypeptide (or peptide) expression products, e.g., selected from SEQ ID NOs:2-ll. Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, pseudorabies, adenovirus, adeno-associated virus, retroviruses, and many others, as well as viral amplicon vectors. Any vector that is capable of introducing genetic material into a cell, and, if replication is desired, which is replicable in the relevant host, can be used.
[0116] In an expression vector, the polynucleotide sequence, commonly a subsequence of SEQ ID NO: 1, e.g., comprising an ORF, such as an ORF encoding a polypeptide (or peptide) selected from among SEQ ID NOs: 2-11 (or variants thereof, e.g., conservative variations thereof), is physically arranged in proximity and orientation to an appropriate transcription control sequence (promoter, and optionally, one or more enhancers) to direct mRNA synthesis. For example, a subsequence of SEQ ID NO.T, e.g., encoding a polypeptide selected from a subsequence of one of SEQ ID NOs:2-ll, can be inserted into an expression vector to produce antigenic peptide for the production of antibodies, e.g., for diagnostic or therapeutic purposes. That is, the polynucleotide sequence of interest is operably linked to an appropriate transcription control sequence. Examples of such promoters include: LTR or SV40 promoter, E. coli lac or trp promoter, phage lambda PJL promoter, and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses. The expression vector typically also contains a ribosome binding site for translation initiation, and a transcription terminator. The vector optionally includes appropriate sequences for amplifying expression. In addition, the expression vectors optionally comprise one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells, such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E. coli.
[0117] Where translation of polypeptide encoded by a nucleic acid comprising a polynucleotide sequence of the invention is desired, additional translation specific initiation signals can improve the efficiency of translation. These signals can include, e.g., an ATG initiation codon and adjacent sequences. In some cases, for example, full-length cDNA molecules or chromosomal segments including a coding sequence incorporating, e.g., a polynucleotide sequence of the invention, a translation initiation codon and associated sequence elements are inserted into the appropriate expression vector simultaneously with the polynucleotide sequence of interest. In such cases, additional translational control signals are not required. However, in cases where only a polypeptide coding sequence, e.g., encoding an amino acid sequence selected from among SΕQ ID NOs:2-ll or a portion thereof, is inserted, exogenous translational control signals, including an ATG initiation codon is provided for expression of the relevant sequence. The initiation codon is put in the correct reading frame to ensure transcription of the polynucleotide sequence of interest. 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 (Scharf D et al. (1994) Results Probl Cell Differ 20:125-62; Bittner et al. (1987) Methods in Enzymol 153:516-544).
POLYPEPTIDEPRODUCTIONANDRECOVERY
[0118] The present invention also relates to the introduction of vectors incorporating the polynucleotides of the invention (e.g., polynucleotides including all or part of one or more ORFs selected from SEQ ID NO: 1) into host cells and the production of polypeptides of the invention, e.g., one or more polypeptide selected from SEQ ID NOs:2-ll or subsequences thereof (e.g., unique subsequences thereof) by recombinant techniques. Recombinant and/or isolated polypeptides encoded by the polynucleotides of the invention, e.g., SEQ ID NOs:2-ll, or subsequences (e.g., unique subsequences) thereof are used, for example, as antigens to produce antibodies in animal or human subjects. For example, antigenic polypeptides (or peptides) corresponding to all or part of a polypeptide represented by SEQ ID NOs:2-ll can be injected into an experimental animal to produce antibodies specific for one or more strains of RSV, as further described in the section entitled "Antibodies" below. Additionally, the antigenic polypeptides can be administered, e.g., as a vaccine, to human subjects to elicit an immune response specific for one or more strains of RSV. For example, such an elicited immune response can be a protective immune response.
[0119] To produce the polypeptides of the invention, host cells are genetically engineered (e.g., transduced, transformed or transfected) with a vector, such as an expression vector, of this invention. As described above, the vector can be in the form of a plasmid, a viral particle, a phage, etc. Examples of appropriate expression hosts include: bacterial cells, such as E. coli, Streptomyces, and Salmonella typhimurium; fungal cells, such as Saccharomyces cerevisiae, Pichia pastoris, and Neurospora crassa; insect cells such as Drosophila and Spodopterafrugiperda; mammalian cells such as 3T3, COS, CHO, BHK, HEK 293 or Bowes melanoma; plant cells, etc.
[0120] The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants, or amplifying the inserted polynucleotide sequences. The culture conditions, such as temperature, pH and the like, are typically those previously used with the host cell selected for expression, and will be apparent to those skilled in the art and in the references cited herein, including, e.g., Freshney (1994) Culture of Animal Cells, a Manual of Basic Technique, third edition, Wiley- Liss, New York and the references cited therein. In addition to Sambrook, Berger and Ausubel, details regarding cell culture can be found in Payne et al. (1992) Plant Cell and Tissue Culture in Liquid Systems John Wiley & Sons, Inc. New York, NY; Gamborg and Phillips (eds) (1995) Plant Cell, Tissue and Organ Culture; Fundamental Methods Springer Lab Manual, Springer- Verlag (Berlin Heidelberg New York) and Atlas and Parks (eds) The Handbook of Microbiological Media (1993) CRC Press, Boca Raton, FL.
[0121] In bacterial systems, a number of expression vectors can be selected depending upon the use intended for the expressed product. For example, when large quantities of a polypeptide or fragments thereof are needed for the production of antibodies, vectors which direct high level expression of fusion proteins that are readily purified are favorably employed. Such vectors include, but are not limited to, multifunctional E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene), in which the coding sequence of interest, e.g., a polynucleotide of the invention as described above, can be ligated into the vector in-frame with sequences for the amino-terminal translation initiating Methionine and the subsequent 7 residues of beta-galactosidase producing a catalytically active beta galactosidase fusion protein; pIN vectors (Van Heeke and Schuster (1989) J Biol Chem 264:5503-5509); pET vectors (Novagen, Madison WI); and the like.
[0122] Similarly, in the yeast Saccharomyces cerevisiae a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase and PGH can be used for production of the desired expression products. For reviews, see Berger, Ausubel, and, e.g., Grant et al. (1987) Methods in Enzymology 153:516-544.
[0123] Vectors suitable for replication in mammalian cells are also known in the art.
Exemplary vectors include those derived from SV40, retroviruses, bovine papilloma virus, vaccinia virus, other herpesviruses and adenovirus. Such suitable mammalian expression vectors optionally contain a promoter to mediate transcription of foreign DNA sequences and, optionally, an enhancer. Suitable promoters are known in the art and include viral promoters such as those from SV40, cytomegalovirus (CMV), Rous sarcoma virus (RSV), adenovirus (ADV), and bovine papilloma virus (BPV).
[0124] The optional presence of an enhancer, combined with the promoter described above, will typically increase expression levels. An enhancer is any regulatory DNA sequence that can stimulate transcription up to 1000-fold when linked to endogenous or heterologous promoters, with synthesis beginning at the normal mRNA start site. Enhancers are also active when placed upstream or downstream from the transcription initiation site, in either normal or flipped orientation, or at a distance of more than 1000 nucleotides from the promoter. See, e.g., Maniatis (1987) Science 236:1237 and Alberts (1989) Molecular Biology of the Cell, 2nd Ed. (or later). Enhancers derived from viruses may be particularly useful, because they typically have a broader host range. Examples include the SV40 early gene enhancer (see Dijkema (1985) EMBO J. 4:761) and the enhancer/promoters derived from the long terminal repeat (LTR) of the RSV (see Gorman (1982) Proc. Natl. Acad. Sci. 79:6777) and from human cytomegalovirus (see Boshart (1985) Cell 41:521). Additionally, some enhancers are regulatable and become active only in the presence of an inducer, such as a hormone or metal ion (see Sassone-Corsi and Borelli (1986) Trends Genet. 2:215); Maniatis (1987) Science 236:1237), In addition, the expression vector can and will typically also include a termination sequence and poly(A) addition sequences which are operably linked to the heterologous coding sequence. [0125] Sequences that cause amplification of the gene may also be desirably included in the expression vector or in another vector that is co-translated with the expression vector, as are sequences which encode selectable markers. Selectable markers for mammalian cells are known in the art, and include, for example, thymidine kinase, dihydrofolate reductase (together with methotrexate as a DHFR amplifier), aminoglycoside phosphotransferase, hygromycin B phosphotransferase, asparagine synthetase, adenosine deaminase, metallothionien, and antibiotic resistant genes such as neomycin.
[0126] Mammalian cell lines available as hosts for expression are known in the art and include many immortalized cell lines available from the American Type Culture Collection (ATCC) as well as primary cultured cells and established cell lines, including but not limited to Vero, HEp-2, 3T3, COS, CHO, HeLa, BHK, MDCK, 293, WI38, Hep G2, MRC-5, and many others.
[0127] Host cells containing the vectors (e.g., expression vectors) described above are also a feature of the invention. The host cell can be a eukaryotic cell, such as a mammalian cell, a yeast cell, or a plant cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, electroporation, encapsulation of the polynucleotide(s) in liposomes, direct microinjection of the DNA into nuclei, or other common techniques (see, e.g., Davis, L., Dibner, M., and Battey, I. (1986) Basic Methods in Molecular Biology).
[0128] A host cell strain is optionally chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein in the desired fashion. Such modifications of the protein include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation. Post-translational processing which cleaves a precursor form into a mature form of the protein is sometimes important for correct insertion, folding and/or function. Different host cells have specific cellular machinery and characteristic mechanisms for such post-translational activities and can be chosen to ensure the correct modification and processing of the introduced, foreign protein.
[0129] Host cells transformed with a nucleotide sequence encoding a polypeptide of the invention are optionally cultured under conditions suitable for the expression and recovery of the encoded protein from cell culture. The protein or fragment thereof produced by a recombinant cell can be secreted, membrane-bound, or contained intracellularly, depending on the sequence and/or the vector used.
[0130] Following transduction of a suitable host cell line or strain and growth of the host cells to an appropriate cell density, the selected promoter is induced if necessary by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period. The secreted polypeptide product is then recovered from the culture medium. Alternatively, cells can be harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification. Eukaryotic or microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, or other methods, which are well know to those skilled in the art.
[0131] Expressed polypeptides can be recovered and purified from recombinant cell cultures by any of a number of methods well known in the art, including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography (e.g., using any of the tagging systems noted herein), hydroxylapatite chromatography, and lectin chromatography. Protein refolding steps can be used, as desired, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed in the final purification steps. In addition to the references noted above, a variety of purification methods are well known in the art, including, e.g., those set forth in Deutscher, Methods in Enzymology Vol. 182: Guide to Protein Purification, Academic Press, Inc. NY. (1990); Sandana (1997) Bioseparation of Proteins, Academic Press, Inc.; Bollag et al. (1996) Protein Methods, 2nd Edition Wiley- Liss, NY; Walker (1996) The Protein Protocols Handbook Humana Press, NJ; Harris and Angal (1990) Protein Purification Applications: A Practical Approach IRL Press at Oxford, Oxford, U.K.; Scopes (1993) Protein Purification: Principles and Practice 3rd Edition Springer Verlag, NY; Janson and Ryden (1998) Protein Purification: Principles, High Resolution Methods and Applications, Second Edition Wiley- VCH, NY; and Walker (1998) Protein Protocols on CD-ROM Humana Press, NJ.
[0132] Alternatively, cell-free transcription translation systems can be employed to produce polypeptides comprising an amino acid sequence or subsequence encoded by the polynucleotides of the invention. A number of suitable in vitro transcription and translation systems are commercially available. A general guide to in vitro transcription and translation protocols is found in Tymms (1995) In vitro Transcription and Translation Protocols: Methods in Molecular Biology Volume 37, Garland Publishing, NY. Cell free transcription/translation systems can be particularly useful for the production of polypeptides, including proteins for administration to human subjects.
[0133] In addition, the polypeptides, or subsequences thereof, e.g., subsequences comprising antigenic peptides, can be produced manually or by using an automated system, by direct peptide synthesis using solid-phase techniques (see, e.g., Stewart et al. (1969) Solid-Phase Peptide Synthesis, WH Freeman Co, San Francisco; Merrifield J (1963) J. Am. Chem. Soc. 85:2149-2154). Exemplary automated systems include the Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer, Foster City, CA). If desired, subsequences can be chemically synthesized separately, and combined using chemical methods to provide full- length polypeptides.
MODIFIED AMINO ACIDS
[0134] Expressed polypeptides of the invention can contain one or more modified amino acid. The presence of modified amino acids can be advantageous in, for example, (a) increasing polypeptide serum half-life, (b) reducing polypeptide antigenicity or (c) increasing polypeptide storage stability. Amino acid(s) are modified, for example, co- translationally or post-translationally during recombinant production (e.g., N-linked glycosylation at N-X-S/T motifs during expression in mammalian cells) or are modified by synthetic means (e.g., via PEGylation).
[0135] Non-limiting examples of a modified amino acid include a glycosylated amino acid, a sulfated amino acid, a prenlyated (e.g., farnesylated, geranylgeranylated) amino acid, an acetylated amino acid, an acylated amino acid, a PEG-ylated amino acid, a biotinylated amino acid, a carboxylated amino acid, a phosphorylated amino acid, and the like, as well as amino acids modified by conjugation to, e.g., lipid moieties or other organic derivatizing agents. References adequate to guide one of skill in the modification of amino acids are replete throughout the literature. Example protocols are found in Walker (1998) Protein Protocols on CD-ROM Human Press, Towata, NJ. ANTIBODIES
[0136] The polypeptides of the invention can be used to produce antibodies specific for the polypeptides comprising amino acid sequences or subsequences encoded by the polynucleotides of the invention. Antibodies specific for antigenic peptides encoded by, e.g., SEQ ID NO:l (e.g., SEQ ID NOs:2-ll), and related variant polypeptides are useful, e.g., for diagnostic and therapeutic purposes, e.g., related to the activity, distribution, and expression of target polypeptides.
[0137] Antibodies specific for the polypeptides of the invention can be generated by methods well known in the art. Such antibodies can include, but are not limited to, polyclonal, monoclonal, chimeric, humanized, single chain, Fab fragments and fragments produced by an Fab expression library.
[0138] Polypeptides do not require biological activity for antibody production.
However, the polypeptide or oligopeptide is antigenic. Peptides used to induce specific antibodies typically have an amino acid sequence of at least about 5 amino acids, and often at least 10 or 20 amino acids. Short stretches of a polypeptide, e.g., encoded by a polynucleotide of the invention such a sequence selected from SEQ ID NO:l (such as a polypeptide selected from among SEQ ID NOs:2-ll) can optionally be fused with another protein, such as keyhole limpet hemocyanin (KLH), and antibodies produced against the fusion protein or polypeptide.
[0139] Numerous methods for producing polyclonal and monoclonal antibodies are known to those of skill in the art, and can be adapted to produce antibodies specific for the polypeptides or peptides of the invention. See, e.g., Coligan (1991) Current Protocols in Immunology Wiley/Greene, NY; and Harlow and Lane (1989) Antibodies: A Laboratory Manual Cold Spring Harbor Press, NY; Stites et al. (eds.) Basic and Clinical Immunology (4th ed.) Lange Medical Publications, Los Altos, CA, and references cited therein; Goding (1986) Monoclonal Antibodies: Principles and Practice (2d ed.) Academic Press, New York, NY; Fundamental Immunology, e.g., 4th Edition (or later),W.E. Paul (ed.), Raven Press, N . (1998); and Kohler and Milstein (1975) Nature 256: 495-497. Other suitable techniques for antibody preparation include selection of libraries of recombinant antibodies in phage or similar vectors. See, Huse et al. (1989) Science 246: 1275-1281; and Ward, et al. (1989) Nature 341: 544-546. Specific monoclonal and polyclonal antibodies and antisera will usually bind with a KD of at least about 0.1 μM, preferably at least about 0.01 μM or better, and most typically and preferably, 0.001 μM or better.
[0140] For certain therapeutic applications (e.g., administration of an antibody or anitserum specific for one or more strains of RSV to provide passive immunity to a subject, e.g., a human, to prevent or decrease the severity of RSV disease), humanized antibodies are desirable. Detailed methods for preparation of humanized antibodies can be found in USPN 5,482,856. Additional details on humanization and other antibody production and engineering techniques can be found in Borrebaeck (ed) (1995) Antibody Engineering, 2nd Edition Freeman and Company, NY (Borrebaeck); McCafferty et al. (1996) Antibody Engineering, A Practical Approach IRL at Oxford Press, Oxford, England (McCafferty), and Paul (1995) Antibody Engineering Protocols Humana Press, Towata, NJ (Paul). Additional details regarding specific procedures can be found, e.g., in Ostberg et al. (1983) Hvbridoma 2: 361-367, Ostberg, USPN 4,634,664, and Engelman et al. USPN 4,634,666.
DIAGNOSTIC ASSAYS
[0141] The novel nucleic acid sequences of the present invention can be used in diagnostic assays to detect RSV in a sample, to detect RSV B9320-like sequences, and to detect strain differences in clinical isolates of RSV using either chemically synthesized or recombinant RSV B9320 polynucleotide fragments, e.g., selected from SEQ ID NO:l. For example, fragments of the novel B9320 sequences (SEQ ID NO:l) comprising at least between 10 and 20 nucleotides can be used as primers to amplify nucleic acids using polymerase chain reaction (PCR) methods well known in the art (e.g., reverse transcription- PCR) and as probes in nucleic acid hybridization assays to detect target genetic material such as RSV RNA in clinical specimens.
[0142] The novel RSV B9320 polynucleotide sequences can be used in their entirety or as fragments to detect the presence of RNA sequences or transcription products in cells, tissues, samples and the like using hybridization techniques known in the art or in conjunction with one of the methods discussed herein. The probes can be either DNA or RNA molecules, such as fragments of viral RNA, isolated restriction fragments of cloned DNA, cDNAs, amplification products, transcripts, and oligonucleotides, and can vary in length from oligonucleotides as short as about 10 nucleotides in length to viral RNA fragments or cDNAs in excess of one or more kilobases. For example, in some embodiments, a probe of the invention includes a polynucleotide sequence or subsequence (e.g., a unique subsequence) selected from SEQ ID NO:l or sequences complementary thereto. Preferably the polynucleotide sequence (or subsequence) is selected from a portion of SEQ ID NO:l that includes at least a subsequence that is not included in SEQ ID NOs: 14-19. Alternatively, polynucleotide sequences that are variants of one of the above designated sequences are used as probes. Most typically, such variants include one or a few nucleotide variations. For example, pairs (or sets) of oligonucleotides can be selected, in which the two (or more) polynucleotide sequences are substantially identical variants of each other, wherein one polynucleotide sequence or set corresponds identically to a first viral strain (e.g., B9320) and the other sequence(s) or set(s) correspond identically to additional viral strains (e.g., BI, A2, etc.). Such pairs of oligonucleotide probes are particularly useful, for example, in the context of an allele specific hybridization experiment to determine the identity of an RSV virus or viral nucleic acid, e.g., for diagnostic or monitoring purposes. In other applications, probes are selected that are more or less divergent, that is probes that are at least about 70% (or 80%, 90%, 95%, 98%, or 99%) identical are selected.
[0143] The probes of the invention, e.g., as exemplified by unique subsequences selected from SEQ ID NO:l, can also be used to identify additional useful polynucleotide sequences (such as to characterize additional strains of RSV) according to procedures routine in the art. In one set of preferred embodiments, one or more probes, as described above, are utilized to screen libraries of expression products or cloned viral nucleic acids (i.e., expression libraries or genomic libraries) to identify clones that include sequences identical to, or with significant sequence identity to SEQ ID NO:l. In turn, each of these identified sequences can be used to make probes, including pairs or sets of variant probes as described above. It will be understood that in addition to such physical methods as library screening, computer assisted bioinformatic approaches, e.g., BLAST and other sequence homology search algorithms, and the like, can also be used for identifying related polynucleotide sequences.
[0144] The probes of the invention are particularly useful for detecting the presence and for determining the identity of RSV nucleic acids in cells, tissues or other biological samples (e.g., a nasal wash or bronchial lavage). For example, the probes of the invention are favorably utilized to determine whether a biological sample, such as a subject (e.g., a human subject) or model system (such as a cultured cell sample) has been exposed to, or become infected with, RSV. Detection of hybridization of the selected probe to nucleic acids originating in (e.g., isolated from) the biological sample or model system is indicative of exposure to or infection with the virus (or a related virus) from which the probe polynucleotide is selected. For example, a polynucleotide sequence that hybridizes preferentially to a subsequence of SEQ ID NO:l as compared to the corresponding subsequence of SEQ ID NO: 13 (or the genome of another naturally occurring RSV strain) can be used to distinguish RSV B 9320 from RSV BI (or another RSV strain).
[0145] It will be appreciated that probe design is influenced by the intended application. For example, where several allele-specific probe-target interactions are to be detected in a single assay, e.g., on a single DNA chip, it is desirable to have similar melting temperatures for all of the probes. Accordingly, the length of the probes are adjusted so that the melting temperatures for all of the probes on the array are closely similar (it will be appreciated that different lengths for different probes may be needed to achieve a particular Tm where different probes have different GC contents). Although melting temperature is a primary consideration in probe design, other factors are optionally used to further adjust probe construction, such as selecting against primer self-complementarity and the like.
[0146] In other circumstances, e.g., relating to functional attributes of cells or organisms expressing the polynucleotides and polypeptides of the invention, probes that are polypeptides, peptides or antibodies are favorably utilized. For example, polypeptides, polypeptide fragments and peptides encoded by or having subsequences encoded by the polynucleotides of the invention, e.g., SEQ ID NO:l, are favorably used to identify and isolate antibodies or other binding proteins, e.g., from phage display libraries, combinatorial libraries, polyclonal sera, and the like.
[0147] Antibodies specific for a polypeptide subsequence encoded by any subsequence (e.g., unique subsequence) or ORF of SEQ ID NO:l are likewise valuable as probes for evaluating expression products, e.g., from cells or tissues. For example, suitable polypeptide sequences are selected from among the amino acid sequences represented by SEQ ID NOs:2-ll. In addition, antibodies are particularly suitable for evaluating expression of proteins comprising amino acid subsequences encoded by SEQ ID NO:l (e.g., SEQ ID NOs:2-ll), e.g., in a sample from a subject infected with or exposed to RSV. Antibodies can be directly labeled with a detectable reagent as described below, or detected indirectly by labeling of a secondary antibody specific for the heavy chain constant region (i.e., isotype) of the specific antibody. Additional details regarding production of specific antibodies are provided above in the section entitled "Antibodies."
LABELINGANDDETECTINGPROBEg
[0148] Numerous methods are available for labeling and detection of the nucleic acid and polypeptide (or peptide or antibody) probes of the invention. These include: 1) fluorescence (using, e.g., fluorescein, Cy-5, rhodamine or other fluorescent tags); 2) isotopic methods, e.g., using end-labeling, nick translation, random priming, or PCR to incorporate radioactive isotopes into the probe polynucleotide/oligonucleotide; 3) chemifluorescence, e.g., using alkaline phosphatase and the substrate AttoPhos (Amersham) or other substrates that produce fluorescent products; 4) chemiluminescence (e.g., using either horseradish peroxidase and/or alkaline phosphatase with substrates that produce photons as breakdown products; kits providing reagents and protocols are available from such commercial sources as Amersham, Boehringer-Mannheim, and Life Technologies/Gibco BRL); and, 5) colorimetric methods (again using, e.g., horseradish peroxidase and/or alkaline phosphatase with substrates that produce a colored precipitate; kits are available from Life Technologies/Gibco BRL, and Boehringer-Mannheim). Other methods for labeling and detection will be readily apparent to one skilled in the art.
[0149] More generally, a probe can be labeled with any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical, chemical or other available means. Useful labels in the present invention include spectral labels such as fluorescent dyes (e.g., fluorescein isothiocyanate, Texas red, rhodamine, and the like), radiolabels (e.g., 3H, 1251, 35S, 14C, 32P, 33P, etc.), enzymes (e.g., horse-radish peroxidase, alkaline phosphatase, etc.), spectral colorimetric labels such as colloidal gold or colored glass or plastic (e.g. polystyrene, polypropylene, latex, etc.) beads. The label may be coupled directly or indirectly to a component of the detection assay (e.g., a probe, such as an oligonucleotide, isolated DNA, amplicon, restriction fragment, or the like) according to methods well known in the art. As indicated above, a wide variety of labels may be used, with the choice of label depending on sensitivity required, ease of conjugation with the compound, stability requirements, available instrumentation, and disposal provisions. In general, a detector which monitors a probe-target nucleic acid hybridization is adapted to the particular label which is used. Typical detectors include spectrophotometers, phototubes and photodiodes, microscopes, scintillation counters, cameras, film and the like, as well as combinations thereof. Examples of suitable detectors are widely available from a variety of commercial sources known to persons of skill. Commonly, an optical image of a substrate comprising a nucleic acid array with particular set of probes bound to the array is digitized for subsequent computer analysis.
[0150] Because incorporation of radiolabeled nucleotides into nucleic acids is straightforward, this detection represents one favorable labeling strategy. Exemplary technologies for incorporating radiolabels include end-labeling with a kinase or phosphatase enzyme, nick translation, incorporation of radio-active nucleotides with a polymerase and many other well known strategies.
[0151] Fluorescent labels are desirable, having the advantage of requiring fewer precautions in handling, and being amenable to high-throughput visualization techniques. Preferred labels are typically characterized by one or more of the following: high sensitivity, high stability, low background, low environmental sensitivity and high specificity in labeling. Fluorescent moieties, which are incorporated into the labels of the invention, are generally are known, including Texas red, fluorescein isothiocyanate, rhodamine, etc. Many fluorescent tags are commercially available from SIGMA chemical company (Saint Louis, MO), Molecular Probes (Eugene, OR), R&D systems (Minneapolis, MN), Pharmacia LKB Biotechnology (Piscataway, NJ), CLONTECH Laboratories, Inc. (Palo Alto, CA), Chem Genes Corp., Aldrich Chemical Company (Milwaukee, WI), Glen Research, Inc., GEBCO BRL Life Technologies, Inc. (Gaithersberg, MD), Fluka Chemica- Biochemika Analytika (Fluka Chemie AG, Buchs, Switzerland), and Applied Biosystems (Foster City, CA) as well as other commercial sources known to one of skill. Similarly, moieties such as digoxygenin and biotin, which are not themselves fluorescent but are readily used in conjunction with secondary reagents, i.e., anti-digoxygenin antibodies, avidin (or streptavidin), that can be labeled, are suitable as labeling reagents in the context of the probes of the invention.
[0152] The label is coupled directly or indirectly to a molecule to be detected (a product, substrate, enzyme, or the like) according to methods well known in the art. As indicated above, a wide variety of labels are used, with the choice of label depending on the sensitivity required, ease of conjugation of the compound, stability requirements, available instrumentation, and disposal provisions. Non radioactive labels are often attached by indirect means. Generally, a ligand molecule (e.g., biotin) is covalently bound to a nucleic acid such as a probe, primer, amplicon, or the like. The ligand then binds to an anti-ligand (e.g., streptavidin) molecule which is either inherently detectable or covalently bound to a signal system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound. A number of ligands and anti-ligands can be used. Where a ligand has a natural anti-ligand, for example, biotin, thyroxine, and cortisol, it can be used in conjunction with labeled anti-ligands. Alternatively, any haptenic or antigenic compound can be used in combination with an antibody. Labels can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorophore or chromophore. Enzymes of interest as labels will primarily be hydrolases, particularly phosphatases, esterases and glycosidases, or oxidoreductases, particularly peroxidases. Fluorescent compounds include fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, etc. Chemiluminescent compounds include luciferin, and 2,3-dihydrophthalazinediones, e.g., luminol. Means of detecting labels are well known to those of skill in the art. Thus, for example, where the label is a radioactive label, means for detection include a scintillation counter or photographic film as in autoradiography. Where the label is optically detectable, typical detectors include microscopes, cameras, phototubes and photodiodes and many other detection systems which are widely available.
PRODUCTION OF RECOMBINANT VIRUS
[0153] Negative strand RNA viruses can be genetically engineered and recovered using a recombinant reverse genetics approach (USPN 5,166,057 to Palese et al). Although this method was originally applied to engineer influenza viral genomes (Luytjes et al. (1989) Cell 59:1107-1113; Enami et al. (1990) Proc. Natl. Acad. Sci. USA 92:11563- 11567), it has been successfully applied to a wide variety of segmented and nonsegmented negative strand RNA viruses, e.g., rabies (Schnell et al. (1994) EMBO J. 13: 4195-4203); VSV (Lawson et al. (1995) Proc. Natl. Acad. Sci. USA 92: 4477-4481); measles virus (Radecke et al.(1995) EMBO J. 14:5773-5784); rinderpest virus (Baron and Barrett (1997) J.Virol. 71: 1265-1271); human parainfluenza virus (Hoffman and Banerjee (1997) J. Virol. 71: 3272-3277; Dubin et al. (1997) Virology 235:323-332); SV5 (He et al. (1997) Virology 237:249-260); canine distemper virus (Gassen et al. (2000) J. Virol. 74:10737-44); and Sendai virus (Park et al. (1991) Proc. Natl. Acad. Sci. USA 88: 5537-5541; Kato et al. (1996) Genes to Cells 1:569-579).
[0154] Recently, a system for producing recombinant subgroup A RSV (e.g., attenuated recombinant RSV suitable for vaccine production) has been described by the inventors and coworkers in WO 02/44334 by Jin et al. entitled "Recombinant RSV virus expression systems and vaccines," the disclosure of which is incorporated herein in its entirety. Rescue of subgroup A RSV has also been described, e.g., in Jin et al. (1998) Virology 251:206-214 and Collins et al. (1995) Proc. Natl. Acad. Sci. USA 92:11563- 11567. (See also e.g., Jin et al. (2000) J. Virol. 74:74-82; Jin et al. (2000) Virology 273:210-218; Cheng et al. (2001) Virology 283:59-68; Tang et al. (2001) J. Virol. 75:11328-11335; US patent application 60/444,287 (filed on January 31, 2003) by Jin et al. entitled "Functional mutations in respiratory syncytial virus"; and US patent application 10/672,302 (filed on September 26, 2003) by Jin et al. entitled "Functional mutations in respiratory syncytial virus.") Rescue of subgroup B RSV is briefly described below and in the examples herein.
[0155] In brief, recombinant RSV incorporating the nucleic acids of this invention are generated, for example, in a suitable cell line (e.g., Vero or Hep-2 cells) by transfection of the cells with an antigenomic cDNA. Typically, the antigenomic cDNA is flanked by a T7 RNA polymerase promoter and a hepatitis delta virus ribozyme plus the T7 transcriptional terminator. Plasmids expressing the viral N, P, and L proteins (and optionally also the M2-1 protein) are also introduced into the cells, and a T7 RNA polymerase is typically expressed in the transfected cells (e.g., by infection of the cells with a modified vaccinia virus Ankara expressing T7 RNA polymerase). Recombinant RSV can also be produced by infection of suitable cells with previously isolated recombinant virus. Techniques for propagation, separation from host cell cellular components, and/or further purification of RSV are well known to those skilled in the art.
[0156] Methods of producing recombinant RSV are a feature of the invention. In the methods, a host cell into which a vector comprising a nucleic acid of the invention has been introduced is cultured in a suitable culture medium under conditions permitting expression of the nucleic acid (e.g., coexpression of RSV N, P, and L and optionally M2-1 and/or T7 RNA polymerase). The recombinant respiratory syncytial virus is then isolated from the host cell and/or the medium. Typically, the nucleic acid comprises an entire RSV genome or antigenome. Alternatively, the nucleic acid can comprise a portion of an RSV genome or antigenome (e.g., one or more open reading frames encoding proteins to be assembled with an RSV genome from another source to form the recombinant virus).
[0157] Recombinant RSV (e.g., attenuated recombinant RSV) produced according to the methods described herein are also a feature of the invention, as are recombinant RSV (e.g., attenuated recombinant RSV) comprising one or more nucleic acids and/or polypeptides of the invention.
CELL CULTURE
[0158] Typically, propagation of a recombinant virus (e.g., recombinant RSV) is accomplished in the media compositions in which the host cell is commonly cultured. Suitable host cells for the replication of RSV include, e.g., Vero cells and HEp-2 cells. Typically, cells are cultured in a standard 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 C02 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.
[0159] 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. Additionally, variations in such procedures adapted to the present invention are readily determined through routine experimentation.
[0160] Cells for production of RSV can be cultured in serum-containing or serum free medium. In some cases, e.g., for the preparation of purified viruses, it is desirable to grow the host cells in serum free conditions. For example, cells can be grown to the desired density in serum-containing medium, infected, and then maintained in serum-free medium. Cells can be cultured in small scale, e.g., less than 25 ml medium, 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 is often 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, MN); NBS bioreactors (New Brunswick Scientific, Edison, N.J.); and laboratory and commercial scale bioreactors from B. Braun Biotech International (B. Braun Biotech, Melsungen, Germany).
[0161] Other useful references, e.g. for cell isolation and culture (e.g., of bacterial cells containing recombinant nucleic acids, e.g., for subsequent nucleic acid isolation) include Freshney (1994) Culture of Animal Cells, a Manual of Basic Technique, third edition, Wiley- Liss, New York and the references cited therein; Payne et al. (1992) Plant Cell and Tissue Culture in Liquid Systems John Wiley & Sons, Inc. New York, NY; Gamborg and Phillips (eds) (1995) Plant Cell, Tissue and Organ Culture: Fundamental Methods Springer Lab Manual, Springer- Verlag (Berlin Heidelberg New York) and Atlas and Parks (eds) The Handbook of Microbiological Media (1993) CRC Press, Boca Raton, FL.
Introduction of vectors into host cells [0162] Vectors, e.g., vectors incorporating RSV polynucleotides, are introduced
(e.g., transfected) into host cells according to methods well known in the art for introducing heterologous nucleic acids into eukaryotic cells, including, e.g., calcium phosphate co- precipitation, electroporation, microinjection, lipofection, and transfection employing polyamine transfection reagents. For example, vectors, e.g., plasmids, can be transfected into host cells, e.g., Vero cells or Hep-2 cells, using the transfection reagent LipofectACE or Lipofectamine 2000 (Invitrogen) according to the manufacturer's instructions. Alternatively, electroporation can be employed to introduce vectors incorporating RSV genome segments into host cells.
MODEL SYSTEMS
[0163] Attenuated RSV, e.g. those described herein comprising all or part of SEQ
ID NO:l or variations thereof, can be tested in in vitro and in vivo models to confirm adequate attenuation, genetic stability, and or immunogenicity for vaccine use. In in vitro assays, e.g., replication in cultured cells, the virus can be tested, e.g., for genetic stability, temperature sensitivity of virus replication and or a small plaque phenotype. RSV can be further tested in animal models of infection. A variety of animal models, e.g., primate (e.g., chimpanzee, African green monkey) and rodent (e.g., cotton rat), are known in the art, as described briefly herein and in USPN 5,922,326 to Murphy et al. (July 13, 1999) entitled "Attenuated respiratory syncytial virus compositions"; USPN 4,800,078; Meignier et al. eds. (1991) Animal Models of Respiratory Syncytial Virus Infection, Merieux Foundation Publication; Prince et al. (1985) Virus Res. 3:193-206; Richardson et al. (1978) J. Med. Virol. 3:91-100; Wright et al. Infect. Immun. (1982) 37:397-400; and Crowe et al. (1993) Vaccine 11:1395-1404.
METHODS AND COMPOSITIONS FOR PROPHYLACTIC ADMINISTRATION
OF VACCINES
[0164] One aspect of the invention provides immunogenic compositions (e.g., vaccines) comprising an immunologically effective amount of a recombinant RSV of the invention (e.g., an attenuated live recombinant RSV), an immunologically effective amount of a polypeptide of the invention, and or an immunologically effective amount of a nucleic acid of the invention.
[0165] A related aspect of the invention provides methods for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus. In the methods, an immunologically effective amount of a recombinant RSV of the invention, an immunologically effective amount of a polypeptide of the invention, and/or an immunologically effective amount of a nucleic acid of the invention is administered to the individual in a physiologically acceptable carrier.
[0166] The RSV, polypeptides, and nucleic acids 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 RSV. Typically, the carrier or excipient is a pharmaceutically acceptable carrier or excipient, such as sterile water, aqueous saline solution, aqueous buffered saline solutions, aqueous dextrose solutions, aqueous glycerol solutions, ethanol, 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, oral, topical, etc. The resulting aqueous solutions can e.g., be packaged for use as is or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration
[0167] Generally, the RSV (or RSV components) of the invention are administered in a quantity sufficient to stimulate an immune response specific for one or more strains of RSV (e.g., an immunologically effective amount of RSV or an RSV component is administered). Preferably, administration of RSV elicits a protective immune response. Dosages and methods for eliciting a protective anti -viral immune response, adaptable to producing a protective immune response against RSV, are known to those of skill in the art. See, e.g., USPN 5,922,326; Wright et al. (1982) Infect. Immun. 37:397-400; Kim et al. (1973) Pediatrics 52:56-63; and Wright et al. (1976) J. Pediatr. 88:931-936. For example, virus can be provided in the range of about 10 -10 pfu (plaque forming units) per dose administered (e.g., 104 -105 pfu per dose administered). Typically, the dose will be adjusted based on, e.g., age, physical condition, body weight, sex, diet, mode and time of administration, and other clinical factors. The prophylactic vaccine formulation can be systemically administered, e.g., by subcutaneous or intramuscular injection using a needle and syringe or a needleless injection device. Preferably, the vaccine formulation is administered intranasally, e.g., by drops, aerosol (e.g., large particle aerosol (greater than about 10 microns)), or spray into the upper respiratory tract. While any of the above routes of delivery results in a protective systemic immune response, intranasal administration confers the added benefit of eliciting mucosal immunity at the site of entry of the virus. For intranasal administration, attenuated live virus vaccines are often preferred, e.g., an attenuated, cold adapted and or temperature sensitive recombinant RSV, e.g., a chimeric recombinant RSV. As an alternative or in addition to attenuated live virus vaccines, killed virus vaccines, nucleic acid vaccines, and/or polypeptide subunit vaccines, for example, can be used, as suggested by Walsh et al. (1987) J. Infect. Pis. 155:1198-1204 and Murphy et al. (1990) Vaccine 8:497-502.
[0168] Typically, the attenuated recombinant RSV of this invention as used in a vaccine is sufficiently attenuated such that symptoms of infection, or at least symptoms of serious infection, will not occur in most individuals immunized (or otherwise infected) with the attenuated RSV. In embodiments in which viral components (e.g., the nucleic acids or polypeptides herein) are used as vaccine or immunogenic components, serious infection is not typically an issue. In some instances, the attenuated RSV (or RSV components of the invention) can still be capable of producing symptoms of mild illness (e.g., mild upper respiratory illness) and/or of dissemination to unvaccinated individuals. However, virulence is sufficiently abrogated such that severe lower respiratory tract infections do not typically occur in the vaccinated or incidental host.
[0169] While stimulation of a protective immune response with a single dose is preferred, additional dosages can be administered, by the same or different route, to achieve the desired prophylactic effect. In neonates and infants, for example, multiple administrations may be required to elicit sufficient levels of immunity. Administration can continue at intervals throughout childhood, as necessary to maintain sufficient levels of protection against wild-type RSV infection. Similarly, adults who are particularly susceptible to repeated or serious RSV infection, such as, for example, health care workers, day care workers, family members of young children, the elderly, and individuals with compromised cardiopulmonary function may require multiple immunizations to establish and/or maintain protective immune responses. Levels of induced immunity can be monitored, for example, by measuring amounts of neutralizing secretory and serum antibodies, and dosages adjusted or vaccinations repeated as necessary to elicit and maintain desired levels of protection.
[0170] Alternatively, an immune response can be stimulated by ex vivo or in vivo targeting of dendritic cells with virus. For example, proliferating dendritic cells are exposed to viruses in a sufficient amount and for a sufficient period of time to permit capture of the RSV antigens by the dendritic cells. The cells are then transferred into a subject to be vaccinated by standard intravenous transplantation methods. [0171] Optionally, the formulation for prophylactic administration of the RSV also contains one or more adjuvants for enhancing the immune response to the RSV antigens. Suitable adjuvants include, for example: complete Freund's adjuvant, incomplete Freund's adjuvant, 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), Cotγnebacterium parvum, and the synthetic adjuvant QS-21.
[0172] If desired, prophylactic vaccine administration of RSV can 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 RSV, or can be administered separately. Either the protein or an expression vector encoding the protein can be administered to produce an immunostimulatory effect.
[0173] Although vaccination of an individual with an attenuated RSV of a particular strain of a particular subgroup can induce cross-protection against RSV of different strains and/or subgroups, cross-protection can be enhanced, if desired, by vaccinating the individual with attenuated RSV from at least two strains, e.g., each of which represents a different subgroup. Similarly, the attenuated RSV vaccines of this invention can optionally be combined with vaccines that induce protective immune responses against other infectious agents.
KITS AND REAGENTS
[0174] The present invention is optionally provided to a user as a kit. For example, a kit of the invention contains one or more nucleic acid, polypeptide, antibody, or cell line described herein. Most often, the kit contains a diagnostic nucleic acid or polypeptide (e.g., an antibody or a probe, e.g., as a cDNA microarray packaged in a suitable container) or other nucleic acid such as one or more expression vector. The kit typically further comprises one or more additional reagents, e.g., substrates, labels, primers, tubes and/or other accessories, reagents for collecting samples, buffers, hybridization chambers, cover slips, etc. The kit optionally further comprises an instruction set or user manual detailing preferred methods of using the kit components.
DIGITAL SYSTEMS
[0175] The present invention provides digital systems, e.g., computers, computer readable media and integrated systems comprising character strings corresponding to the sequence information herein for the polypeptides and nucleic acids herein, including, e.g., those sequences listed herein and the various silent substitutions and conservative substitutions thereof. Integrated systems can further include, e.g., gene synthesis equipment for making genes and/or peptide synthesis equipment for making polypeptides corresponding to the character strings.
[0176] Various methods known in the art can be used to detect homology or similarity between different character strings, or can be used to perform other desirable functions such as to control output files, provide the basis for making presentations of information including the sequences and the like. Examples include BLAST, discussed supra. Computer systems of the invention can include such programs, e.g., in conjunction with one or more data file or data base comprising a sequence as noted herein.
[0177] Thus, different types of homology and similarity of various stringency and length can be detected and recognized in the integrated systems herein. For example, many homology determination methods have been designed for comparative analysis of sequences of biopolymers, for spell-checking in word processing, and for data retrieval from various databases. With an understanding of double-helix pair-wise complement interactions among 4 principal nucleobases in natural polynucleotides, models that simulate annealing of complementary homologous polynucleotide strings can also be used as a foundation of sequence alignment or other operations typically performed on the character strings corresponding to the sequences herein (e.g., word-processing manipulations, construction of figures comprising sequence or subsequence character strings, output tables, etc.).
[0178] Thus, standard desktop applications such as word processing software (e.g.,
Microsoft Word™ or Corel WordPerfect™) and database software (e.g., spreadsheet software such as Microsoft Excel™, Corel Quattro Pro™, or database programs such as Microsoft Access™ or Paradox™) can be adapted to the present invention by inputting a character string corresponding to one or more polynucleotides and polypeptides of the invention (either nucleic acids or proteins, or both). For example, a system of the invention can include the foregoing software having the appropriate character string information, e.g., used in conjunction with a user interface (e.g., a GUI in a standard operating system such as a Windows, Macintosh or LINUX system) to manipulate strings of characters corresponding to the sequences herein. As noted, specialized alignment programs such as BLAST can also be incorporated into the systems of the invention for alignment of nucleic acids or proteins (or corresponding character strings).
[0179] Systems in the present invention typically include a digital computer with data sets entered into the software system comprising any of the sequences herein. The computer can be, e.g., a PC (Intel x86 or Pentium chip- compatible DOS™, OS2™ WINDOWS™ WINDOWS NT™, WINDOWS95™, WINDOWS98™ LINUX based machine, a MACINTOSH™, Power PC, or a UNIX based (e.g., SUN™ work station) machine) or other commercially common computer which is known to one of skill. Software for aligning or otherwise manipulating sequences is available, or can easily be constructed by one of skill using a standard programming language such as Visualbasic, Fortran, Basic, Java, or the like.
[0180] Any controller or computer optionally includes a monitor which is often a cathode ray tube ("CRT") display, a flat panel display (e.g., active matrix liquid crystal display, liquid crystal display), or others. Computer circuitry is often placed in a box which includes numerous integrated circuit chips, such as a microprocessor, memory, interface circuits, and others. The box also optionally includes a hard disk drive, a floppy disk drive, a high capacity removable drive such as a writeable CD-ROM, and other common peripheral elements. Inputting devices such as a keyboard or mouse optionally provide for input from a user and for user selection of sequences to be compared or otherwise manipulated in the relevant computer system.
[0181] The computer typically includes appropriate software for receiving user instructions, either in the form of user input into a set parameter fields, e.g., in a GUI, or in the form of preprogrammed instructions, e.g., preprogrammed for a variety of different specific operations. The software then converts these instructions to appropriate language, e.g., for instructing the operation of equipment, e.g., gene and or peptide synthesis equipment, to carry out the desired operation. [0182] The software can also include output elements for controlling nucleic acid synthesis (e.g., based upon a sequence or an alignment of a sequences herein) or other operations.
[0183] In an additional aspect, the present invention provides system kits embodying the methods, composition, systems and apparatus herein. System kits of the invention optionally comprise one or more of the following: (1) an apparatus, system, system component or apparatus component as described herein; (2) instructions for practicing the methods described herein, and/or for operating the apparatus or apparatus components herein and/or for using the compositions herein. In a further aspect, the present invention provides for the use of any apparatus, apparatus component, composition or kit herein, for the practice of any method or assay herein, and/or for the use of any apparatus or kit to practice any assay or method herein.
EXAMPLES
[0184] The following sets forth a series of experiments that demonstrate construction of RSV B9320 cDNAs and recovery of recombinant RSV from the cDNAs. It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. Accordingly, the following examples are offered to illustrate, but not to limit, the claimed invention.
MATERIALS AND METHODS
Cells and Viruses
[0185] Monolayer cultures of HEp-2 and Vero cells (obtained from the American
Type Culture Collection, ATCC) were maintained in minimal essential medium (MEM) containing 5% fetal bovine serum. RSV A2 was obtained from the ATCC and grown in
Vero cells in Opti-MEM. RSV subgroup B strain 9320, originally isolated in Massachusetts in 1977 (Hierholzer and Hirsch (1979) J. Infect. Pis. 140:826-828), was obtained from the
ATCC and grown in Vero cells in Opti-MEM. Infected cells were maintained in serum-free
Opti-MEM medium. Modified vaccinia virus Ankara expressing bacteriophage T7 RNA polymerase (MVA-T7; Sutter et al. (1995) FEBS Lett. 371:9-12 and Wyatt et al. (1995)
Virology 210:202-205) was provided by Pr. Bernard Moss and amplified in CEK cells. Recombinant fowlpox virus expressing the T7 RNA polymerase (FPV-T7; Britton et al. (1996) "Expression of bacteriophage T7 RNA polymerase in avian and mammalian cells by a recombinant fowlpox virus" J Gen Virol 77:963-7) was obtained from Dr. Michael Skinner and grown in CEK cells.
Sequencing
[0186] RSV B9320 was grown in Vero cells and viral RNA was extracted from virus purified by ultracentrifugation from infected cell culture supernatant. 9320 genome sequences were obtained by sequencing DNA fragments generated by RT-PCR; the cDNA full length clone was also sequenced for comparison. All sequencing was done by
Sequetech, Mountain View CA (www.sequetech.com).
Sequence Analysis
[0187] Sequence analysis was performed with Vector NTI version 6.0 and 8.0
ContigExpress and AlignX (Informax, Inc., www.informaxinc.com). Pairwise nucleic acid or polypeptide sequence alignments were performed with Vector NTI AlignX using default parameters set by the provider.
Construction of full-length cDNA of RSV subgroup B9320 strain and recovery of infectious respiratory syncytial virus from cDNA
Construction of RSV 9320 protein expression plasmids [0188] The 9320 N, P, and L protein coding regions were each cloned into a pCITE vector (Novagen, Madison, WI) under control of a T7 RNA polymerase promoter, to produce expression plasmids pB-N, pB-P, and pB-L.
[0189] pB-N: The N gene was amplified by RT-PCR from 9320 RNA extracted from virus particles purified by ultracentrifugation, using primers XC19 (5'- GATCCCATGGCTCTTAGCAAAGTCAAG-3' containing Nco I site, SEQ ID NO:20) and XC020 (5'-GTACGGATCCGTTGACTTATTTGCCCCGTAT-3' containing Bamffl site, SEQ ID NO:21), and cloned between the Ncol and Bamffl sites of pCITE2a 3a (Novagen) under the control of T7 promoter. This N protein expression plasmid was designated as AD740.
[0190] pB-P: The P gene was amplified by RT-PCR using primers XC17 (5 -
GATCCCATGGAGAAGTTTGCACCTG-3' with Nco I site, SEQ ID NO:22) and XC018 (5'-GTACGGATCCTGAGTGAGTTGATCACTG-3' with BamH I site, SEQ ID NO:23) and cloned between the Ncol and Bamffl sites of pCHE2a/3a. This clone was designated as AD741.
[0191] pB-L: The L gene was cloned from three cDNA subclones obtained by RT-
PCR and the clone was assigned as AD778.
[0192] To obtain one subclone, primers XC003 (5'
GCTTGGCCATAACGATTCTATATCATCC-3', SEQ ID NO:24) and XC014 (5'- GGTAGTATAATGTTGTGCACTTTTAG-3', SEQ ID NO:25) were used to amplify 9320 L from nt8511 to nt!1685 and the cDNA was cloned into T/A vector (Invitrogen, pCR 2.1) to generate subclone AD762.
[0193] To obtain the second subclone, primers XC011 (5'-
GGTCACGATTTACAAGATAAGCTCC-3', SEQ ID NO:26) andXC007 (5'- CAGATCCTTTTAACTTGCTACCTAGGCACA-3' SEQ ID NO:27) were used to amplify nt 11686 to ntl4495 and the BamH I to Avr II fragment was cloned into the T/A vector as AD763.
[0194] To obtain the third subclone, primers XC009 (5 -
CTTACGTGTGCCTAGGTAGCAAG-3', SEQ ID NO:28) and XC010 (5'- ACGAGAAAAAAAGTGTCAAAAACTAATGTCTCG, SEQ ID NO:29) were used to amplify 9320 nt 14495 to 15225, producing a first PCR product. To add the ribozyme cleavage sequence (RBZ) and T7 terminator sequence (T70), a second PCR product was obtained using XC015 (5'-
GTTTTTGACACTTTTTTTCTCGTGGCCGGCATGGTCCCAGCC-3', SEQ ID NO:30) and XC016 (5'-GATCTAGAGCTCCAAGCTTGCGGCCGCGTCGAC-3' containing the Kpn I site, SEQ ID NO:31) as primers and ρRSV-A2 full-length antigenomic cDNA (Jin et al. (1998) Virology 251:206-214) as template. Since primers XC010 and XC015 contained overlapping sequences, these two PCR products were annealed, extended and amplified by PCR using XC009 and XC016 as primers. The cDNA was cloned into the T/A vector and designated as AD764.
[0195] The three subclones were verified by sequence analysis. To join the three L subclones together, the Avrll and Kpn I fragment was removed from AD764 and cloned into AD763 and the larger clone was designated as AD767. The Bamffl to Notl restriction fragment from ntl!685 to the sequence downstream of the T7 terminator was removed from AD767 and cloned into pCITE2a/3a vector under the control of the T7 promoter and the plasmid was designated as AD766. The Bamffl fragment from nt 8511 to nt 11685 was removed from clone AD762 and inserted into the BamH I site of AD766. The second Bamffl site at position of nt 11685 was then knocked out by site-directed mutagenesis and the clone is designated as pB-L (AD778).
[0196] The functions of the pB-N, pB-P, and pB-L expression plasmids were examined by the RSV minigenome assay (as described in, e.g., Tang et al. (2002) Virology 302:207-216). A level of the CAT reporter gene similar to those of A2 expression plasmids was detected in cells transfected with pB-N, pB-P, pB-L and pRSVCAT minigenome, indicating all three of these plasmids are functional.
Assembly of full-length antigenomic cDNA of RSV 9320 strain [0197] An antigenomic cDNA spanning the entire RSV 9320 genome was assembled by sequential ligation of RSV cDNA fragments with the indicated unique restriction sites (Figure 1). In brief, six cDNA fragments (B1-B6) were generated from
9320 viral RNA by RT-PCR using the pfu polymerase (Stratagene, La Jolla, CA) and cloned into a modified pET vector containing the RSV 9320 unique restriction enzyme sites
(Xma I, Avr II, Sac I, BamH I, and Bssffll). The Xma I-Avr II cDNA fragment (BI) containing the T7 RNA polymerase promoter proximal to the 5' antigenomic sense DNA was joined with the Avr Il-Sac I cDNA fragment (B2) to form the B7 fragment (through a
Sad site, as described below). The B7 fragment was used to replace the corresponding region in a full-length RSV A2-B9320 chimera containing the G, F and M2 sequence (B3).
The Sac I restriction site at nt 2310 in the resulting pUC-B8 was mutated without affecting the coding sequence of the SH gene. The L gene fragment (B10) was assembled from B4 and B9 fragments. The hepatitis delta virus ribozyme (RBZ) and the T7 RNA polymerase terminator sequence was amplified from RSV A2 antigenomic cDNA (Jin et al. (1998)
"Recombinant human respiratory syncytial virus (RSV) from cDNA and construction of subgroup A and B chimeric RSV" Virology 251:206-214) and ligated to the trailer sequence through PCR (B6). Ligation of B5 and B6 fragments were mediated through the Avr II and
Not I restriction sites. The BamH I site at nt position 11685 was deleted from B10 cDNA by mutagenesis without affecting the protein coding sequence. The L gene (B10) was cloned into the chimeric clone that contained the 9320 B8 fragment and A2 L to replace the A2-L sequence through the BamH I and Not I restriction sites. The antigenomic cDNA clone (Bll) encoding the complete RSV 9320 genome was designated ρB9320C4. In addition, an antigenomic cDNA clone containing a single C to G change at the fourth position of the leader sequence was also obtained by mutagenesis and designated pB9320G4. A more detailed description of the cDNA construction follows.
[0198] Positions of various primers and subclones used in cloning the full length cDNA are illustrated in Figures 1-3. The 3' genome was amplified by RT-PCR using PFU polymerase with primer V1964 (5'-
GGGTACCCCCGGGTAATACGACTCACTATAGGGACGGGAAAAAATG-3' containing Xma I restriction enzyme, the T7 promoter and the leader sequences, SEQ ID NO:32) and XC051 (5 '-GTTAACTTAGAGCTCT AC ATCATC-3' containing the Sac I restriction site present in 9320 genome at position of nt 2310, SEQ ID NO:33). The PCR fragment was cloned between the Xma I and Sad sites of the modified pET vector (pET21b was cut with BspEI, and the pET21b was ligated with a polylinker with Xmal, Smal, Sad, Mscl, Bamffl, Spel, Pmll, and Bssffll restriction sites to produce the modified pET-21b vector) and designated as AD803. A second PCR DNA that contained nt 2106 to 4494 was obtained by RT/PCR using XC034 (5'-GTGTGGTCCTAGGCAATGCAGCAG-3', SEQ ID NO.-34) and XC032 (5'-GACACAGCATGATGGTAGAGCTCTATGTG-3', SEQ ID NO:35) as primers and was cloned into the Sac I site of the pET vector for sequence analysis. This Sad fragment was then moved to AD803 through the Sac I site in AD803 (producing B7). In order to ligate the cDNA encoding NSl, NS2, N, P, M and SH genes with the rest of the 9320 cDNA, the Sac I site at position nt 2310 was removed by mutagenesis and the resulting clone was designated as AD816. The Xma I to Sac I fragment from AD816 was released to replace the corresponding region of the A2 sequence in the chimeric cDNA AD379 that had the G and F genes of 9320 in place of A2 G and F genes (in a pUC19 backbone, Cheng et al. (2001) Virology 283:59-68). This clone was designated as AD827.
[0199] The cDNA containing the G, F, and M2 genes of 9320 strain was derived by
RT-PCR using XC063 (5'-GCTAAGTGAACATAAAACATTCTGTAAC-3', SEQ ID NO:36) as RT primer and XC006 (5'-CCATTAATAATGGGATCCATTTTGTC-3' with Sad site, SEQ ID NO:37) and XC062 (5'-CACATAGAGCTCTACCATCATGCTGTGTC- 3' with BamH I site, SEQ ID NO:38) as PCR primers and was initially cloned into pET vector as AP835 for sequence analysis. The Sad to BamH I fragment from AD835 was then moved to AD827 and the clone was designated as AD848.
[0200] The Bamffl and Notl fragment from pB-L (AD869 encoding 9320 L with the two Sac I sites at positions of nt 10376 and nt 14951 knocked-out) was swapped into AD848 to complete the assembly of a full length antigenomic cDNA of RSV 9320.
[0201] Three mutations introduced by PCR during the cloning process were corrected by site-directed mutagenesis in their respective subclones, as follows. To reverse a His to Asn change in L at amino acid position 209 , site-directed mutagenesis was performed to correct the His with primers XC081 (5'-
CATGGTTAATACACTGGTTCAATTTATATACA-3', SEQ ID NO.-41) and XC082 (5'- TGTATATAAATTGAACCAGTGTATTAACCATG-3', SEQ ID NO:42). To correct an Arg to Lys change in N at amino acid position 194 (nt 1748), site directed mutagenesis was performed with primers XC086 (5'-
GTCTTAAAAAACGAAATAAAACGCTACAAGGGCCTCATACC-3', SEQ ID NO:43) and XC087 (5'GGTATGAGGCCCTTGTAGCGTTTTATTTCGTTTTTTAAGAC-3', SEQ ID NO:44). A Ser to Asn change in NSl at amino acid position 108 was not corrected.
[0202] The recombinant 9320 cDNA has the following genetic tags that are different from wild-type 9320 virus. (The enclosed sequence, SEQ ID NO:l, is the wild type RSV 9320 strain and does not reflect the recombinant DNA sequence.) First, Sac I sites at nt 2310, 10376 and 14951 were removed without changing the protein coding sequences, using the following primers: Sad at 2310 nt, XC049 (5'- GATGATGTAGAGCTTTAAGTTAAC-3', SEQ ID NO: 45) and XC050 (5'- GTTAACTTAAAGCTCTACATCATC-3", SEQ ID NO: 46); Sad at 10376 nt, XC088 (5'- CTAACTGGTAAAGAAAGAGAGCTTAGTGTAGGTAGAATGTTTGC-3', SEQ ID NO: 47) and XC089 (5'-
GCAAACATTCTACCTACACTAAGCTCTCTTTCTTTACCAGTTAG-3', SEQ ID NO: 48); and Sad at 14951 nt, XC090 (5'-
GTTTAACAACCAATGAGCTTAAAAAGCTGATTAAAATTAC-3', SEQ ID NO:49) and XC091 (5'-GTAATTTTAATCAGCTTTTTAAGCTCATTGGTTGTTAAAC-3', SEQ ID NO:50). The removal of the two Sac I sites at positions nt 10376 and 14951 in the L gene from AD864 to generate AD869 did not alter the amino acid sequence of L. Second, a BamH I site at nt 11685 was removed using primers XC067 (5'- CATTAATGAGGGACCCACAGGCTTTAG-3', SEQ IP NO:39) and XC068 (5'- CTAAAGCCTGTGGGTCCCTCATTAATG-3', SEQ IP NO:40). Third, a Sac I site at nt 4477 was added using XC032 (5'-GACACAGCATGATGGTAGAGCTCTATGTG, SEQ
ID NO:35).
[0203] It was previously reported that changing the C at the fourth nucleotide position of the leader region of RSV A2 to a G increased the promoter strength, resulting in increased transcription/replication of an RSV minigenome (Collins et al. (1993) "Rescue of a 7502-nucleotide (49.3% of full-length) synthetic analog of respiratory syncytial virus genomic RNA" Virology 195:252-256) and higher virus recovery efficiency (Jin et al. (1998) Virology 251:206-214). Thus, as noted, a 9320 cDNA with a C4 to G change in the leader sequence at the antigenomic sense was made to increase the promoter strength, and the resulting clone was designated as AD897 (pB9320G4).
Construction of G gene deletion mutants [0204] Two mutants were constructed to determine if the G gene of RSV B9320 strain is dispensable, e.g., for viral replication in tissue culture and/or an animal host. In one mutant, the entire open reading frame of the G gene was removed from the 9320 cDNA. In the other mutant, the region encoding the cysteine noose and heparin binding sites of G was removed from the 9320 cDNA.
[0205] For the RSV A2 strain, the G protein has been shown to be dispensable for virus replication in vitro (Techaarpornkul et al. (2001) "Functional analysis of recombinant respiratory syncytial virus deletion mutants lacking the small hydrophobic and/or attachment glycoprotein gene" J Virol 75:6825-34 and Teng et al. (2001) "Contribution of the respiratory syncytial virus G glycoprotein and its secreted and membrane-bound forms to virus replication in vitro and in vivo" Virology 289:283-96). However, rA2ΔG (RSV A2 lacking G) replicated poorly in HEp-2 cells, and its replication was attenuated in the respiratory tracts of mice. Previously, it was also shown that a cold-adapted RSV BI strain, cρ-52, had both the SH and G genes deleted (Karron et al. (1997) Proc Natl Acad Sci USA 94:13961-13966). However, this deletion mutant replicated poorly and was over-attenuated in animals and in humans. To determine whether the G protein was also dispensable for 9320 replication in vitro, an antigenomic cDNA was constructed in which the entire G gene (957 nt) including the gene start and gene end sequences was deleted from the cDNA and the new SH-F intergenic region contained 75 nt. [0206] To construct the 9320 antigenomic cPNA that had the G gene deleted, deletion mutagenesis was performed on a pET-S/B cPNA subclone that contained sequences of the 9320 G, F and M2 genes using a pair of PCR primers flanking the G open reading frame in opposite orientations (5'-GATCCCATACTAATAATTCATCATTATG-3', SEQ IP NO:51, and 5'-AGCAGAGAACCGTGATCTATCAAGCAAG-3', SEQ IP NO:52) using the ExSite PCR-based Site-Pirected Mutagenesis Kit (Stratagene, La Jolla, CA). The deletion was confirmed by restriction enzyme digestion and nucleotide sequencing analysis. The Sac I-BaniH I fragment containing only the F and M2 genes was introduced into pB9320G4, and the antigenomic cPNA was designated pB9320ΔG.
[0207] To delete only the cysteine noose and heparin binding sites of G (amino acids 164-197), a small cPNA fragment of nt 5179-5280 nt was deleted from the 9320 G gene using primers XC079 (5*-GTAATCATCTTTTGGTTTTTTTGGTGG-3', SEQ IP NO:53) and XC080 (5'-CCAACCATCAAACCCACAAACAAACCAACCGTC-3'5 SEQ IP NO:54). The cPNA containing the desired deletion was then removed from the subclone by digestion with Sac I and Bamffl and shuffled into the full-length 9320 antigenomic cPNA. The resulting antigenomic cPNA was designated pB9320ΔHBS.
Recovery of recombinant viruses from cPNAs [0208] Recovery of 9320 viruses (rg9320C4, rg9320G4, rg9320ΔG and rg9320ΔHBS) by reverse genetics (rg) was performed as described previously (Jin et al.
(1998) Virology 251:206-214). Briefly, HEp-2 cells were infected with MVA-T7 at an m.o.i. of 5.0 and transfected with 0.4 μg of pB-N (pN), 0.4 μg of pB-P (pP), 0.2 μg of pB-L
(pL), and 0.8 μg of pB9320C4, pB9320G4, pB9320ΔG or pB9320ΔHBS by
Lipofectamine™ 2000 (Invitrogen, Carlsbad, CA). In some transfection reactions, 0.2 μg pRSV-M2-l (encoding the RSV A2 M2-1 protein) was also included. Alternatively, Vero cells were infected with FPV-T7 (Britton et al. (1996) J Gen Virol 77:963-7) at an m.o.i. of
1.0 for 1 hr and transfected with the PNAs as above. Transfected cells were incubated at
35°C. Three days after transfection, the culture supernatant was used to infect Vero cells to amplify the recovered viruses. Six days after infection, the culture supernatant was harvested and virus-infected cells were identified by immunostaining using polyclonal anti-
RSV A2 serum (Biogenesis, Kingston, NH). The recombinant virus from the culture supernatant was plaque purified and amplified in Vero cells. Replication of rg9320C4. rg9320G4. rg9320ΔG in tissue culture [0209] Replication of rg9320C4, rg9320G4 and rg9320ΔG in Vero and HEp-2 cells was compared with replication of wild type 9320. Vero or HEp-2 cell monolayers in 6-well plates were infected with each virus in duplicate at an m.o.i. of 0.1. After 1 hr adsorption at room temperature, the infected cells were washed with PBS three times and incubated with
2 ml of OptiMEM at 35°C. At 24 hr intervals, aliquots of 250 μl of culture supernatant were removed and stored at -80°C prior to virus titration. Each aliquot taken was replaced with the same amount of fresh media. The virus titer was determined by plaque assay on
Vero cells using an overlay consisting of 1% methylcellulose and IX MEM/L15 (JRH
Bioscience, Lenexa, KS) containing 2% FBS. After incubation at 35°C for 6 days, the monolayers were fixed with methanol and plaques enumerated by immunostaining.
Western Blotting analysis of virus infected cells [0210] Vero or HEp-2 cells were infected with virus at an m.o.i of 5.0 and the infected cells were lysed in Laemmli protein sample buffer (Bio-Rad, Hercules, CA). The cell lysates were electrophoresed on 12% polyacrylamide gels containing 0.1% SPS, and then transferred to a nylon membrane. The blots were incubated with either polyclonal anti-
RS V A2 serum or a mixture of four monoclonal antibodies against the G protein of RSV B strain (2434PB3, 2218BP5, 2218AE7 and 2218PG7) obtained from Pr. Gregory Storch
(Storch et al. (1991) "Antigenic and genomic diversity within group A respiratory syncytial virus" J Infect Pis 163:858-861). Viral proteins were visualized by incubation with horseradish peroxidase (HRP)-conjugated secondary antibodies followed by chemiluminescent detection (Amersham Biosciences, Piscataway, NJ).
RESULTS AND DISCUSSION
[0211] We have described the construction of a full-length antigenomic cDNA derived from RSV subgroup B9320 strain and recovery of infectious virus from the cPNA. The antigenomic sequence (complementary to the wild-type RSV 9320 genome and not including the changes introduced into our recombinant RSV) is listed as SEQ IP NO:l. The sequence is being deposited in GenBank (accession number AY353550).
[0212] The RSV 9320 genome contains 15,225 nucleotides and shares 97.8% and
86% identity compared to RSV BI and A2 strains, respectively. As noted previously, the A2 genome contains 15,222 nucleotides; the BI genome contains 15,225 nucleotides (Karron et al. (1997) Proc Natl Acad Sci USA 94:13961-13966). Like the RSV A2 strain, 9320 contains 10 transcriptional units encoding 11 proteins in the order of NS1/NS2/N/P/M/SH G F/M2-1/M2-2/L. Amino acid sequences of the proteins are also provided: NSl is listed as SEQ ID NO:2, NS2 as SEQ IP NO:3, N as SEQ IP NO:4, P as SEQ IP NO:5, M as SEQ IP NO:6, SH as SEQ IP NO:7, G as SEQ IP NO: 12, F as SEQ IP NO:8, M2-1 as SEQ IP NO:9, M2-2 as SEQ IP NO:10, and L as SEQ IP NO:ll.
[0213] Table 3 lists the size of each of the 11 proteins for the 9320, B 1 , and A2 strains.
Table 3
Figure imgf000070_0001
[0214] Table 4 lists the length of the intergenic regions for the three RSV strains.
Table 4
Figure imgf000070_0002
[0215] Table 5 lists the percentage amino acid sequence identity between strains
B9320 and BI, 9320 and A2, and A2 and BI for each protein. The SH, G and M2-2 proteins display the greatest differences between A2 and 9320, while the other proteins have an amino acid identity greater than 86%.
Table 5
Figure imgf000071_0001
Recovery of infectious 9320 from cPNA [0216] A reverse genetics system for the A2 strain of subgroup A RSV was established several years ago (Collins et al. (1995) Proc. Natl. Acad. Sci. USA 92:11563-
11567 and Jin et al. (1998) Virology 251:206-214). However, a system for recovery of subgroup B RSV has not previously been available. Herein we describe construction of an antigenomic cPNA derived from RSV subgroup B9320 and recovery of infectious RSV from cPNA. Similar to the A2 strain, rescue of 9320 depends on the expression of the viral polymerase proteins N, P and L. The M2-1 expression plasmid is not required for RSV
9320 recovery in either the FPV-T7 infected Vero or the MVA-T7 infected HEp-2 cells.
However, M2-1 function was probably supplied by cryptic expression of the M2-1 protein from the transfected full-length antigenomic cPNA (Collins et al. (1999) Virology 259:251-
255). The establishment of the reverse genetics system for the 9320 strain should greatly aid studies of viral protein structure and function of this divergent RSV subgroup.
[0217] Recovery of infectious RSV requires co-transfection of a minimum of three plasmids encoding the N, P and L proteins (Jin et al. (1998) Virology 251:206-214). In addition, the elongation function of M2-1 is also required for the virus recovery from cPNA (Collins et al. (1999) "Support plasmids and support proteins required for recovery of recombinant respiratory syncytial virus" Virology 259:251-255 and Collins et al. (1995) "Production of infectious human respiratory syncytial virus from cloned cDNA confirms an essential role for the transcription elongation factor from the 5' proximal open reading frame of the M2 mRNA in gene expression and provides a capability for vaccine development" Proc. Natl. Acad. Sci. USA 92:11563-11567). The expression plasmids encoding the 9320 N, P, and L proteins were constructed and their functions were examined in the RSV minigenome assay using the pRSV/CAT replicon that contained the negative sense CAT gene flanked by the leader and trailer sequences derived from RSV A2 strain (Tang et al. (2001) J Virol 75:11328-11335). The minigenome assay indicated that the 9320 N, P and L expression plasmids functioned as well as those of RSV A2 strain.
[0218] To recover virus from RSV 9320 cDNA, pB9320C4 or ρB9320G4 was transfected into MVA-T7 infected HEp-2 cells or FPV-T7 infected Vero cells together with the 9320 N, P and L expression plasmids with or without the RSV A2 M2-1 plasmid. Several days after inoculation of Vero cells with the transfected culture supernatant, syncytia formation was observed in the infected Vero cells and virus infection was confirmed by immunostaining.
[0219] Table 6 lists the recovery efficiency of recombinant RSV in the presence or absence of the M2-1 expression plasmid. FPV-T7 infected Vero cells or MVA-T7 infected HEp-2 cells were transfected with p9320C4 or p9320G4 in triplicate wells together with N, P, L expression plasmids with or without M2-1. Three days after transfection, the culture supematants were titrated on Vero cells and the plaque numbers in pfu/ml from each well are shown. The average plaque number is given in parentheses.
Figure imgf000072_0001
[0220] As shown in Table 6, rg9320G4 was rescued more efficiently than rg9320 in both the FPV-T7 infected Vero cells and the MVA-T7 infected HEp-2 cells. Inclusion of the M2-1 expression plasmid slightly increased rescue efficiency in both cell types.
[0221] For vaccine production, the vaccine is typically produced from a qualified cell line that is free of any adventitious agents. The HEp-2 cells currently used for recovery of infectious RSV A2 (e.g., Collins et al. (1995) Proc. Natl. Acad. Sci. USA 92:11563- 11567 and Jin et al. (1998) Virology 251:206-214) are not suitable for vaccine production. Vero cells were therefore explored as the cell substrate for recovering 9320 virus from its cDNA. It was very difficult to recover virus from MVA-T7-infected Vero cells. FPV-T7 has been shown to have a less cytopathic effect in infected Vero cells and thus result in more efficient virus rescue of other viruses (Britton et al. (1996) J Gen Virol 77:963-7 and Das et al. (2000) "Improved technique for transient expression and negative strand virus rescue using fowlpox T7 recombinant virus in mammalian cells" J Virol Meth 89:119-127). Similarly, the recombinant RSV described here were more efficiently recovered from FPV- T7 infected Vero cells than from MVA-T7 infected cells, which may pave the way for recovering RSV vaccine candidates for clinical studies.
[0222] rg9320C4 and rg9320G4 were plaque purified and amplified in Vero cells, both reaching a titer of 1 x 107 pfu/ml. A recombinant virus with the G gene deleted from rg9320G4 was also obtained and the virus was designated rg9320ΔG.
[0223] The identity of the recombinant viruses generated from cDNA was analyzed by RT/PCR of each viral RNA using primer pairs spanning the introduced marker sites or the G deletion region. Digestion of the RT/PCR DNA product from nt 2104 to nt 3096 by the Sac I restriction enzyme showed that the Sac I site was present in 9320 virus but not in the recombinant viruses rg9320C4 and rg9320ΔG. Digestion of the RT/PCR product from nt 11593 to nt 11822 by BamH I confirmed that the BamH I site in the recombinant viruses was also abolished. RT/PCR using a pair of primers spanning the G gene confirmed that the G gene was deleted from rg9320ΔG, since its DNA product was approximately lkb shorter than that of rg9320. Sequencing of the G deletion junction region confirmed the expected deleted sequence.
[0224] The lack of G protein expression in rg9320ΔG infected cells was also confirmed by Western blotting analysis using monoclonal antibodies against the G protein of subgroup B RSV (Storch et al. (1991) J Infect Pis 163:858-861). The G protein was not expressed in rg9320ΔG-infected cells. Western blotting using a polyclonal antibody against RSV revealed an equivalent level of other viral proteins (F, N and P) synthesized by rg9320ΔG compared with rg9320G4.
Replication of the G deletion mutant in tissue culture
[0225] Replication of recombinant 9320 and its G deletion mutant were compared with replication of the biologically derived 9320 strain. Vero or HEp-2 cells were infected with 9320, rg9320C4, rg9320G4 or rg9320ΔG at an m.o.i. of 0.1, and the accumulated level of viruses released into the culture supernatant at each day was titrated in Vero cells. As shown in Figure 4 Panel A, the growth kinetics of rg9320C4, rg9320G4 and rg9320ΔG were very similar to those of 9320 in Vero cells, reaching peak titers of 6.0 logio pfu/ml at 96 hours post infection. In HEp-2 cells (Figure 4 Panel B), rg9320C4 grew similarly to 9320 whereas rg9320G4 grew slightly slower. Replication of rg9320ΔG was reduced by 5- fold compared to rg9320G4.
[0226] The plaque morphology of rg9320ΔG was compared to that of rg9320G4 in
Vero and HEp-2 cells. The spread of rg9320ΔG in Vero and HEp-2 cells in liquid medium was similar to that of rg9320G4. rg9320ΔG also formed plaques of similar size in both the Vero and HEp-2 cells. These data indicated that the deletion of G from 9320 did not have significant impact on virus replication in Vero and Hep-2 cells.
[0227] We have demonstrated that the G protein is not essential for RSV 9320 replication in tissue culture cells. However, deletion of G from RSV A2 strain severely affected virus replication in HEp-2 cells; rA2ΔG had a reduction of more than 3.0 log10 and did not form distinct plaques in HEp-2 cells (Teng et al. (2001) Virology 289:283-96). A study by Techaarpornkul et al. (Techaarpornkul et al. (2001) J Virol 75:6825-34) showed that rA2ΔG had 1-2 logs lower titer in HEp-2 cells and formed smaller plaques in HEp-2 cells. The G protein enhances virus binding to target cells, but it has no role in virus penetration once virus has attached the cells (Techaarpornkul et al. (2001) J Virol 75:6825- 34). In Vero cells, rA2ΔG replicated as well as wt A2 strain, indicating an alternative pathway might be present in Vero cells for efficient virus entry that is independent of the G protein (Teng et al. (2001) Virology 289:283-96). Like rA2ΔG, rg9320ΔG replicated efficiently in Vero cells; however, in contrast to rA2ΔG, its replication was only slightly reduced in HEp-2 cells. The reduction of rg9320ΔG virus in HEp-2 cells was 5-fold compared to the parental rg9320G4 vims and the plaque size reduction in HEp-2 cells was also less apparent. Therefore, 9320 appears to be less dependent on the G protein for infectivity in HEp-2 cells than RSV A2.
[0228] rg9320ΔHBS, a recombinant 9320 virus with a partial deletion of G (the cysteine noose and heparin binding sites) was also recovered from cPNA as described above. Like rg9320ΔG, replication of rg9320ΔHBS in Vero cells is not significantly impaired.
Virus replication in cotton rats [0229] Subgroup B RSV typically replicates better in cotton rats than in mice.
Therefore, in vivo replication of recombinant viruses rg9320C4, rg9320G4, rg9320ΔG and rg9320ΔHBS was examined in cotton rats (Sigmodon hispidus). Cotton rats in groups of five were inoculated intranasally with 150 μl of inoculum containing 106 pfu of the specified virus per animal. Animals were sacrificed four days after inoculation. Lung tissues were harvested and homogenized, and virus titer was determined by plaque assay on
Vero cells. Table 7 lists the mean viral titer for each recombinant virus. rg9320C4 and rg9320G4 replicated to a titer of 3.1 and 3.0 log10 pfu/g, respectively, in the lungs of cotton rats. Both G deletion mutants (rg9320ΔG and rg9320ΔHBS) replicated poorly, indicating G deletion affected RSV 9320 replication in the animal host. rg9320ΔG and rg9320ΔHBS are thus potential candidates for live attenuated RSV vaccines.
Figure imgf000075_0001
Recombinant 9320 and vaccines [0230] An RSV vaccine would preferably provide protection against both subgroup
A and subgroup B RSV infection, which tend to circulate concurrently in communities. Most RSV vaccines developed in the past have been based on the RSV A2 strain. However, it remains to be determined whether an RSV vaccine based solely on a subgroup A strain would provide sufficient immunity to both RSV subgroups. Although recombinant technology has been employed to express subgroup B RSV antigens in the A2 strain, including the replacement of the G and F genes by those of BI (Whitehead et al. (1999) "Replacement of the F and G proteins of respiratory syncytial virus (RSV) subgroup A with those of subgroup B generates chimeric live attenuated RSV subgroup B vaccine candidates" J Virol 73:9773-9780) or 9320 (Cheng et al. (2001) "Chimeric subgroup A respiratory syncytial virus with the glycoproteins substituted by those of subgroup B and RSV without the M2-2 gene are attenuated in African green monkeys" Virology 283:59-68) or the insertion of the 9320 G gene in the A2 strain (Jin et al. (1998) Virology 251:206- 214), subgroup B RSV vaccine development currently lags significantly behind subgroup A vaccine development. Availability of the subgroup B RSV rescue system described herein permits manipulation of the subgroup B RSV genome, e.g., for vaccine development. For example, RSV B9320 can optionally be attenuated by methods used to attenuate subgroup A strains. The methods that have been used to attenuate RSV subgroup A RSV include, e.g., mutagenesis of the viral internal proteins (Lu et al. (2002) "Identification of temperature- sensitive mutations in the phosphoprotein of respiratory syncytial virus that are likely involved in its interaction with the nucleoprotein" J Virol 76:2871-2880; Lu et al. (2002) "The major phosphorylation sites of the respiratory syncytial virus phosphoprotein are dispensable for virus replication in vitro" J Virol 76:10776-10784; Tang et al. (2002) "Clustered charged-to-alanine mutagenesis of human respiratory syncytial virus L polymerase generated temperature-sensitive viruses" Virology 302(207-216); and Tang et al. (2001) "Requirement of cysteines and length of the human respiratory syncytial virus M2-1 protein for protein function and virus viability" J Virol 75:11328-11335), deletion of the accessory genes (Bermingham and Collins (1999) Proc. Natl. Acad. Sci. USA 96:11259- 11264; Jin et al. (2000) J. Virol. 74:74-82; Jin et al. (2000) Virology 273:210-218; Bukreyev et al. (1997) J Virol 71:8973-8982: Teng and Collins (1999) J Virol 73:466-473); and introduction of attenuating mutations from different strains to fine-tune the level of attenuation of a vaccine strain (Firestone et al. (1996) "Nucleotide sequence analysis of the respiratory syncytial virus subgroup A cold-passaged (cp) temperature sensitive (ts) cpts- 248/404 live attenuated virus vaccine candidate" Virology 225:419-422 and Whitehead et al. (1999) "Addition of a missense mutation present in the L gene of respiratory syncytial virus (RSV) cpts530/1030 to RSV vaccine candidate cpts248/404 increases its attenuation and temperature sensitivity" J Virol 73:871-877). As one example, from the gene deletion approach, it appears that rA2ΔM2-2 exhibits some desired features for further clinical evaluation (Cheng et al. (2001) Virology 283:59-68; Jin et al. (2003) "Evaluation of recombinant respiratory syncytial virus gene deletion mutants in African green monkeys for their potential as live attenuated vaccine candidates" Vaccine 21:3647-3652; and Teng et al. (2000) J Virol 74:9317-9321); 9320 with a deletion of the M2-2 gene is thus an attenuated 9320 vaccine candidate. Similarly, forcing use of the second or third start codon of the A2 M2-2 mRNA (instead of the first or all three start codons) results in a decrease in M2-2 activity (US patent application 10/672,302 (attorney docket number 26-000320US), filed on September 26, 2003, Jin et al. entitled "Functional mutations in respiratory syncytial virus"). 9320 with a mutation (e.g., substitution or deletion) in the first, second, and/or third start codon of M2-2 is thus an attenuated 9320 vaccine candidate.
[0231] As noted, recombinant 9320 viruses with complete or partial deletions of the
G gene, e.g., as described herein, are attenuated 9320 vaccine candidates.
[0232] 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 compositions and techniques described above can be used in various combinations. All publications, patents, patent applications, and/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, and/or other document were individually indicated to be incorporated by reference for all purposes.
SEQUENCELISTING
<110> Medlm une Vaccines, Inc. Cheng, Xing Park, Hy n J Jin, Hong
<120> COMPOSITIONS AND METHODS INVOLVING RESPIRATORY SYNCYTIAL VIRUS SUBGROUP B STRAIN 9320
<130> 26-003820US
<160> 54
<170> Patentln version 3.1
<210> 1
<211> 15225
<212> DNA
<213> respiratory syncytial virus B 9320
<400> 1 acgcgaaaaa atgcgtacta caaacttgca cattcgaaaa aaatggggca aataagaatt 60 tgataagtgt tatttaagtc taaccttttc aatcagaaat ggggtgcaat tcattgagca 120 tgataaaggt tagattacaa aatttatttg acaatgacga agtagcattg ttaaaaataa 180 catgttatac tgacaaatta attcttctga ccaatgcatt agccaaagca gcaatacata 240 caattaaatt aaacggcata gtttttatac atgttataac aagcagtgaa gtgtgccctg 300 ataacaatat tgtagtgaaa tctaacttta caacaatgcc aatattacaa aacggaggat 360 acatatggga attgattgag ttgacacact gctctcaatt aaatggtcta atggatgata 420 attgtgaaat caaattttct aaaagactaa gtgactcagt aatgactgat tatatgaatc 480 aaatatctga tttacttggg cttgatctca attcatgaat tgtgtttagt ctaattcaat 540 agacatgtgt ttattaccat tttagttaat ataaaaactc atcaaagaga aatggggcaa 600 ataaactcac ctaatcagtc aaatcatgag cactacaaat aacaacacta ctatgcaaag 660 attgatgatc acagacatga gacccctgtc gatggaatca ataataacat ctctcaccaa 720 agaaatcata acacacaaat tcatatactt gataaacaat gaatgtattg taagaaaact 780 tgatgaaaga caagctacat tcacattcct agtcaattat gagatgaagc tactacacaa 840 agtagggagt accaaatata agaaatacac tgaatataat acaaaatatg gcactttccc 900 tatgcctata tttatcaatc atggcgggtt tctagaatgt attggcatta agcctacaaa 960 acacactcct ataatataca aatatgacct caacccgtaa attccaacaa aaaactaacc 1020 catccaaact aagctattcc ttaaataaca gtgctcaaca gttaagaagg ggctaatcca 1080 ttttagtaat taaaaataaa ggtaaagcca ataacataaa ttggggcaaa tacaaagatg 1140 gctcttagca aagtcaagtt aaatgataca ttaaataagg atcagctgct gtcatctagc 1200 aaatacacta ttcaacgtag tacaggagat aatattgaca ctcccaatta tgatgtgcaa 1260 aaacacttaa acaaactatg tggtatgcta ttaatcactg aagatgcaaa tcataaattc 1320 acaggattaa taggtatgtt atatgctatg tccaggttag gaagggaaga cactataaag 1380 atacttaaag atgctggata tcatgttaaa gctaatggag tagatataac aacatatcgt 1440 caagatataa atggaaagga aatgaaattc gaagtattaa cattatcaag cttgacatca 1500 gaaatacaag tcaatattga gatagaatct agaaagtcct acaaaaaaat gctaaaagag 1560 atgggagaag tggctccaga atataggcat gattctccag actgtgggat gataatactg 1620 tgtatagctg cacttgtaat aaccaaatta gcagcaggag atagatcagg tcttacagca 1680 gtaattagga gggcaaacaa tgtcttaaaa aacgaaataa aacgctacaa gggcctcata 1740 ccaaaggata tagctaacag tttttatgaa gtgtttgaaa aacaccctca tcttatagat 1800 gtttttgtgc actttggcat tgcacaatca tccacaagag ggggtagtag agttgaagga 1860 atctttgcag gattatttat gaatgcctat ggttcagggc aagtaatgct aagatgggga 1920 gttttagcca aatctgtaaa aaatatcatg ctaggacatg ctagtgtcca ggcagaaatg 1980 gagcaagttg tggaagtcta tgagtatgca cagaagttgg gaggagaagc tggattctac 2040 catatattga acaatccaaa agcatcattg ctgtcattaa ctcaatttcc taacttctca 2100 agtgtggtcc taggcaatgc agcaggtcta ggcataatgg gagagtatag aggtacacca 2160 agaaaccagg atctttatga tgcagccaaa gcatatgcag agcaactcaa agaaaatgga 2220 gtaataaact acagtgtatt agacttaaca gcagaagaat tggaggccat aaagcatcaa 2280 ctcaacccca aagaagatga tgtagagctc taagttaaca aaaaatacgg ggcaaataag 2340 tcaacatgga gaagtttgca cctgaatttc atggagaaga tgcaaataac aaagctacca 2400 aattcctaga atcaataaag ggcaagttcg catcatccaa agatcctaag aagaaagata 2460 gcataatatc tgttaactca atagatatag aagtaactaa agagagcccg ataacatctg 2520 gcaccaacat caacaatcca acaagtgaag ctgacagtac cccagaagcc aaaaccaact 2580 acccaagaaa acccctagta agcttcaaag aagatctcac cccaagtgac aacccctttt 2640 ctaagttgta caaagaaaca atagaaacat ttgataacaa tgaagaagaa tctagctact 2700 catatgaaga aataaatgat caaacaaatg acaacattac agcaagacta gatagaattg 2760 atgaaaaatt aagtgaaata ttaggaatgc tccatacatt agtagttgca agtgcaggac 2820 ccacttcagc tcgcgatgga ataagagatg ctatggttgg tctaagagaa gaaatgatag 2880 aaaaaataag agcggaagca ttaatgacca atgataggtt agaggctatg gcaagactta 2940 ggaatgagga aagcgaaaaa atggcaaaag acacctcaga tgaagtgtct ctcaatccaa 3000 cttccaaaaa attgagtgac ttgctggaag acaacgatag tgacaatgat ctatcacttg 3060 atgatttttg atcagtgatc aactcactca gcaatcaaca acatcaataa gacagacatc 3120 aatccattga atcaactgcc agaccgaaca aacaaacgtt catcagcaga accaccaacc 3180 aatcaatcaa ccaattgatc aatcagcaac ctaacaaaat taacaatata gtaacaaaaa 3240 aagaacaaga tggggcaaat atggaaacat acgtgaacaa gcttcacgaa ggctccacat 3300 acacagcagc tgttcagtac aatgttctag aaaaagatga tgatcctgca tcactaacaa 3360 tatgggtgcc tatgttccag tcatctgtgc cagcagactt gctcataaaa gaacttgcaa 3420 gcatcaacat actagtgaag cagatctcta cgcccaaagg accttcacta cgagtcacga 3480 tcaactcaag aagcgctgtg ctggcacaaa tgcccagtaa ttttatcata agtgcaaatg 3540 tatcattaga tgaaagaagc aaattagcat atgatgtaac tacaccttgt gaaatcaaag 3600 catgcagtct aacatgctta aaagtaaaaa gtatgctaac tacagtcaaa gatcttacca 3660 tgaaaacatt caaccccact catgagatta ttgctctatg tgaatttgaa aatattatga 3720 catcaaaaag agtaataata ccaacctatc taagatcaat tagtgtcaaa aacaaggacc 3780 tgaactcact agaaaatata gcaaccaccg aattcaaaaa tgctatcacc aatgcgaaaa 3840 ttattcccta tgcaggatta gtattagtta tcacagttac tgacaataaa ggagcattca 3900 aatatatcaa gccacagagt caatttatag tagatcttgg agcctaccta gaaaaagaga 3960 gcatatatta tgtgactaca aattggaagc atacagctac acgtttttca atcaaaccac 4020 tagaggatta aacttaatta tcaacgctaa atgacaggtc cacatatatc ctcaaactac 4080 acactatatc caaacatcat gaacatctac actacacact tcatcacaca aaccaatccc 4140 acttaaaatc caaaatcact tccagccact atctgctaga cctagagtgc gaataggtaa 4200 ataaaaccaa aatatggggt aaatagacat tagttagagt tcaatcaatc tcaacaacca 4260 tttatactgc taattcaata catatactat aaatttcaaa atgggaaata catccatcac 4320 aatagaattc actagcaaat tttggcctta ttttacacta atacatatga tcttaactct 4380 aatctcttta ctaattataa tcactattat gattgcaata ctaaataagc taagtgaaca 4440 taaaacattc tgtaacaaaa ctcttgaact aggacagatg tatcaaatca acacatagtg 4500 ttctaccatc atgctgtgtc aaattataat cctgtatatg taaacaaaca aatccaatct 4560 tctcacagag tcatggtggc gcaaagccac gccaactatc atggtagcat agagtagtta 4620 tttaaaaatt aacataatga tgaattatta gtatgggatc aaaaacaaca ttggggcaaa 4680 tgcaaccatg tccaaacaca agagtcaacg cactgccagg actctagaaa agacctggga 4740 tactcttaat catctaattg taatatcctc ttgtttatac agactaaacc taaaatctat 4800 agcacaaata gcactatcag ttttggcaat gataatctca acctctctca taattgcagc 4860 cataatattc atcatctctg ccaatcacaa agttacacta acaacggtta cagttcaaac 4920 aataaaaaac cacactgaaa aaaacatcac cacctacctt actcaagtct caccagaaag 4980 ggttagctca tccatacaac ctacaaccac atcaccaatc cacacaaatt cagctacaat 5040 atcaccaaat acaaaatcag aaacacacca tacaacaaca caagccaaaa gcagaatcac 5100 cacttcaaca cagaccaaca agccaagcac aaaatcacgt tcaaaaaatc caccaaaaaa 5160 accaaaagat gattaccatt ttgaagtgtt caattttgtt ccctgtagta tatgtggcaa 5220 caatcaactt tgcaaatcca tctgcaaaac aataccaagc aacaaaccaa agaaaaaacc 5280 aaccatcaaa cccacaaaca aaccaaccgt caaaaccaca aacaaaagag acccaaaaac 5340 accagccaaa atgatgaaaa aagaaaccac caccaaccca acaaaaaaac caaccctcaa 5400 gaccacagaa ggagacacca gcacctcaca atccactgtg ctcgacacaa ccacatcaaa 5460 acacacaatc caacagcaat ccctccactc aatcacctcc gaaaacacac ccaactccac 5520 acaaataccc acagcaaccg aggcctccac atcaaattct acttaaaaaa cctagtcaca 5580 tgcttagtta ttcaaaaact acatcttagc agagaaccgt gatctatcaa gcaagaatga 5640 aattaaacct ggggcaaata accatggagt tgctgatcca caggtcaagt gcaatcttcc 5700 taactcttgc tattaatgca ttgtacctca cctcaagtca gaacataact gaggagtttt 5760 accaatcgac atgtagtgca gttagcagag gttattttag tgctttaaga acaggttggt 5820 ataccagtgt tataacaata gaattaagta atataaaaga aaccaaatgc aatggaactg 5880 acactaaagt aaaacttata aaacaagaat tagataagta taagaatgca gtaacagaat 5940 tacagctact tacgcaaaac acgccagctg ccaacaaccg ggccagaaga gaagcaccac 6000 agtacatgaa ctacacaatc aataccacta aaaacctaaa cgtatcaata agcaagaaga 6060 ggaaacgaag atttctggga ttcttgttag gtgtaggatc tgcaatagca agtggtatag 6120 ctgtatccaa agttctacac cttgaaggag aagtgaacaa aatcaaaaat gctttgttgt 6180 ctacaaacaa agctgtagtc agtctatcaa atggggtcag tgttttaacc agcaaagtgt 6240 tagatctcaa gagttacata aataaccaat tattacccat agtaaatcaa cagagctgtc 6300 gcatctccaa cattgaaaca gttatagaat tccagcagaa gaacagcaga ttgttggaaa 6360 tcaccagaga atttagtgtc aatgcaggtg taacaacacc tttaagcact tacatgttaa 6420 caaacagtga gttactatca ttgatcaatg atatgcctat aacaaatgat cagaaaaaat 6480 taatgtcaag caatgtccag atagtaaggc aacaaagtta ttctatcatg tctataataa 6540 aggaagaagt ccttgcatat gttgtacagc tacctatcta tggtgtaata gatacacctt 6600 gctggaaatt acacacatca cctctatgca ccaccaacat caaagaagga tcaaatattt 6660 gtttaacaag gactgataga ggatggtatt gtgataatgc aggatcagta tccttcttcc 6720 cacaggctga cacttgcaaa gtgcagtcca atcgagtatt ttgtgacact atgaacagtt 6780 tgacattacc aagtgaagtc agcctttgta acactgacat attcaattcc aagtatgact 6840 gcaaaatcat gacttcaaaa acagacataa gcagctcagt aattacttct cttggagcta 6900 tagtgtcatg ctatggtaaa actaaatgca ctgcatccaa taaaaatcgt gggattataa 6960 agacattttc taatggttgt gactatgtgt caaacaaagg agtagatact gtgtcagtgg 7020 gcaacacttt atactatgta aacaagctgg aaggcaaaaa cctttatgta aaaggggaac 7080 ctataataaa ttactatgat cctctagtgt ttccttctga tgagtttgat gcatcaatat 7140 ctcaagtcaa tgaaaaaatc aatcaaagtt tagcttttat acgtagatct gatgaattac 7200 tacataatgt aaatactggc aaatctacta caaatattat gataaccaca atcattatag 7260 taatcattgt agtattgtta tcattaatag ctattggttt actgttgtat tgcaaagcta 7320 aaaacacacc agttacacta agcaaagacc aactaagtgg aatcaacaat attgcattca 7380 gcaaatagac aaaaaaccac ttgatcatgt ttcaacaaca atctgctgac caccaatccc 7440 aaatcaactt aacaacaaat atttcaacat catagcacag gctgaatcat ttcctcacat 7500 catgctacct acacaactaa gctagatcct taactcatag ttacataaaa acctcaagta 7560 tcacaatcaa acactaaatc gacacatcat tcacaaaatt aacaactggg gcaaatatgt 7620 cgcgaagaaa tccttgtaaa tttgagatta gaggtcattg cttgaatggt agaagatgtc 7680 actacagtca taattatttt gaatggcctc ctcatgcatt actagtgagg caaaacttca 7740 tgttaaacaa gatacttaag tcaatggaca aaagcataga cactttgtcg gaaataagtg 7800 gagctgctga actggataga acagaagaat atgctcttgg tatagttgga gtgctagaga 7860 gttacatagg atctataaac aacataacaa aacaatcagc atgtgttgct atgagtaaac 7920 ttcttattga gatcaacagt gatgacatta aaaaactgag agataatgaa gaacccaatt 7980 cacctaagat aagagtgtac aatactgtta tatcatacat tgagagcaat agaaaaaaca 8040 acaagcaaac catccatctg ctcaaaagac taccagcaga tgtgctgaag aagacaataa 8100 agaacacatt agatatccac aaaagcataa ccataagcaa cccaaaagag tcaaccgtga 8160 atgatcaaaa tgaccaaacc aaaaataatg atattaccgg ataaatatcc ttgtagtata 8220 tcatccatac tgatttcaag tgaaagcatg gttgccacat tcaatcacaa aaacatatta 8280 caatttaacc ataaccattt ggataaccac cagtgtttat taaatcatat atttgatgaa 8340 attcattgga cacctaaaaa cttattagat accactcaac aatttctcca acatcttaac 8400 atccctgaag atatatatac agtatatata ttagtgtcat aatgcttgac cataacgatc 8460 ttatatcatc caaccataaa actatcataa taaggttatg ggacaaaatg gatcccatta 8520 ttaatggaaa ctctgctaat gtgtatctaa ctgatagtta tctaaaaggt gttatctctt 8580 tttcagaatg taatgcttta gggagttacc tttttaacgg cccttatctt aaaaatgatt 8640 acactaactt aattagtaga caaagcccac tactagagca tatgaatcta aaaaaactaa 8700 ctataacaca gtcattaata tctagatatc ataaaggtga actgaaatta gaagaaccaa 8760 cttatttcca gtcattactt atgacatata aaagtatgtc ctcgtctgaa caaattgcta 8820 caactaactt acttaaaaaa ataatacgaa gagctataga aataagtgat gtaaaggtgt 8880 acgccatctt gaataaacta ggactaaagg aaaaggacag agttaagccc aacaataatt 8940 caggtgatga aaactcagtt cttacaacca taattaaaga tgatatactt tcggctgtgg 9000 aaaacaatca atcatataca aattcagaca aaaatcactc agtgaaccaa aatatcacta 9060 tcaaaacaac actcttgaaa aaattgatgt gttcaatgca acatcctcca tcatggttaa 9120 tacactggtt caatttatat acaaaattaa ataacatatt aacacaatat cgatcaaatg 9180 aggtaaaaag tcatgggttt atattaatag ataatcaaac tttaagtggt tttcagttta 9240 ttttaaatca atatggttgt attgtttatc ataaaggact taaaaaaatc acaactacta 9300 cttacaatca atttttgaca tggaaagaca tcagccttag cagattaaat gtttgcttaa 9360 ttacttggat aagtaattgt ttaaatacat taaataaaag cttagggctg agatgtggat 9420 tcaataatgt tgtgttatca caattatttc tttatggaga ttgtatactg aaattatttc 9480 ataatgaagg cttctacata ataaaagaag tagagggatt tattatgtct ttaattctaa 9540 acataacaga agaagatcaa tttaggacac gattttataa cagcatgcta aataacatca 9600 cagatgcagc tattaaggct caaaaaaacc tactatcaag agtatgtcac actttattgg 9660 acaagacagt gtctgataat atcataaatg gtaaatggat aatcctatta agtaaatttc 9720 ttaaattgat taagcttgca ggtgataata atctcaataa cttgagtgag ctatattttc 9780 tcttcagaat ctttggacat ccaatggtcg atgaaagaca agcaatggat gctgtaagaa 9840 ttaactgtaa tgaaactaag ttctacttat taagtagtct aagtacgtta agaggtgctt 9900 tcatttatag aatcataaaa gggtttgtaa atacctacaa cagatggccc actttaagga 9960 atgctattgt tctacctcta agatggttga actattataa acttaatact tatccatctc 10020 tacttgaaat cacagaaaat gatttgatta ttttatcagg attgaggttc tatcgtgagt 10080 ttcatctgcc taaaaaagtg gatcttgaaa tgataataaa tgacaaagcc atttcacctc 10140 caaaagatct aatatggact agttttccca gaaattacat gccatcacat atacaaaatt 10200 atatagaaca tgaaaagttg aagttctctg aaagcgacag atcaagaaga gtactagagt 10260 attacttgag agataataaa ttcaatgaat gcgatctata caattgtgtg gtcaatcaaa 10320 gctatctcaa caactctaac cacgtggtat cactaactgg taaagaaaga gagctcagtg 10380 taggtagaat gtttgctatg caaccaggta tgtttaggca aattcaaatc ttagcagaga 10440 aaatgatagc cgaaaatatt ttacaattct tccctgagag tttgacaaga tatggtgatc 10500 tagagcttca aaagatatta gaattaaaag caggaataag caacaaatca aatcgttata 10560 atgataacta caacaattat atcagtaaat gttctatcat tacagacctt agcaaattca 10620 atcaagcatt tagatatgaa acatcatgta tctgcagtga tgtattagat gaactgcatg 10680 gagtacaatc actgttctct tggttgcatt taacaatacc tcttgtcaca ataatatgta 10740 catatagaca tgcacctcct ttcataaagg atcatgttgt taatctgaat gaagttgatg 10800 aacaaagtgg attatacaga tatcatatgg gtggtattga gggctggtgt caaaaactgt 10860 ggaccattga agctatatca ttattagatc taatatccct caaagggaaa ttctctatca 10920 cagctctaat aaatggtgat aatcagtcaa ttgatataag taaaccagtt agacttatag 10980 agggtcagac ccatgctcaa gcagattatt tgttagcatt aaatagcctt aaattgctat 11040 ataaagagta tgcaggcata ggccataagc tcaagggaac agaaacctat atatcccgag 11100 atatgcaatt catgagcaaa acaatccagc acaatggagt gtactatcca gccagtatca 11160 aaaaagtcct gagagtaggt ccatggataa atacaatact tgatgatttt aaagttagtt 11220 tagaatctat aggcagctta acacaggagt tagaatacag aggagaaagc ttattatgca 11280 gtttaatatt tagaaacatt tggttataca atcaaattgc tttgcaactc cgaaatcatg 11340 cattatgtca caataagcta tatttagata tattgaaagt attaaaacac ttaaaaactt 11400 tttttaatct tgatagtatc gatatggcat tatcattgta tatgaatttg cctatgctgt 11460 ttggtggtgg tgatcctaat ttgttatatc gaagctttta tagaagaact ccagacttcc 11520 ttacagaagc tatagtacat tcagtgtttg tgttgagcta ttatactggt cacgatttac 11580 aagataagct ccaggatctt ccagatgata gactgaacaa attcttgaca tgtatcatca 11640 catttgataa aaatcccaat gccgagtttg taacattaat gagggatcca caggctttag 11700 ggtctgaaag gcaagctaaa attactagtg agattaatag attagcagta acggaagtct 11760 taagtatagc tccaaacaaa atattttcta aaagtgcaca acattatact accactgaga 11820 ttgatctaaa tgatattatg caaaatatag aaccaactta ccctcatgga ttaagagttg 11880 tttatgaaag tttacctttt tataaagcag aaaaaatagt taatcttata tcaggaacaa 11940 aatccataac taatatactt gaaaaaacat cagcaataga tacaactgat attaataggg 12000 ctactgatat gatgaggaaa aatataactt tacttataag gatacttcca ctagattgta 12060 acaaagacaa aagagagtta ttaagtttag a.aaatcttag tataactgaa ttaagcaagt 12120 atgtaagaga aagatcttgg tcgttatcca atatagtagg agtaacatcg ccaagtatta 12180 tgttcacaat ggacattaaa tatacaacta gcactatagc cagtggtata attatagaaa 12240 aatataatgt taatagttta actcgtggtg aaagaggacc tactaagcca tgggtaggtt 12300 catctacgca ggagaaaaaa acaatgccag tgtataatag acaagtttta accaaaaagc 12360 aaagag'acca aatagattta ttagcaaaat tagactgggt atatgcatcc atagacaaca 12420 aagatgaatt catggaagaa ctgagtactg gaacacttgg attgtcatat gaaaaagcca 12480 aaaaattgtt tccacaatat ctaagtgtca attatttaca ccgcttaaca gtcagtagta 12540 ggccatgtga attccctgca tcaataccag cttatagaac aacaaattat catttcgata 12600 ctagtcctat caatcatgta ttaacagaaa agtatggaga tgaagatatc gacatagtgt 12660 ttcaaaattg cataagtttt ggtcttagcc taatgtcggt tgtggaacaa ttcacaaaca 12720 tatgtcctaa tagaattatt ctcataccga agctgaatga gatacatttg atgaaacctc 12780 ctatatttac aggagatgtt gatatcatca aattgaagca agtgatacaa aaacagcaca 12840 tgttcctacc agataaaata agtttaaccc aatatgtaga attattccta agtaacaaag 12900 cacttaaatc tggatctcac atcaactcta atttaatatt agtacataaa atgtctgatt 12960 attttcataa tgattatatt ttaagtacta atttagctgg acattggatt ctgattattc 13020 aacttatgaa agattcaaaa ggtatttttg aaaaagattg gggagagggg tatataactg 13080 atcatatgtt cattaatttg aatgttttct ttaatgctta taagacttat ttgctatgtt 13140 ttcataaagg ttatggtaaa gcaaaattag aatgtgatat gaacacttca gatcttcttt 13200 gtgttttgga gttaatagac agtagctact ggaaatctat gtctaaagtt ttcctagaac 13260 agaaagtcat aaaatacata gtcaatcaag acacaagttt gcatagaata aaaggttgtc 13320 atagttttaa gttgtggttt ttaaaacgcc ttaataatgc taaatttacc gtatgccctt 13380 gggttgttaa catagattat cacccaacac acatgaaagc tatattatct tacatagatt 13440 tagttagaat ggggttaata aatgtagata aattaaccat taaaaataaa aacaaattca 13500 atgatgaatt ttacacatca aatctctttt atattagtta taacttttca gacaacactc 13560 atttgctaac aaaacaaata agaattgcta attcagaatt agaaaataat tataacaaac 13620 tatatcaccc aaccccagaa actttagaaa atatgtcatt aattcctgtt aaaagtaaca 13680 atagtaacaa acctaaatct tgtataagtg gaaataccga atctatgatg acgtcaacat 13740 tctccaataa aatgcatatt aaatcttcca ctgttaccac aagattaaac tatagcaaac 13800 aagacttgta caatttattt ccaattgttg tgatagacag gattatagat cattcaggca 13860 atacagcaaa atccaaccaa ctttacacca ccacttcaca tcagacatct ttagtaagga 13920 atagtgcatc actttattgc atgcttcctt ggcatcatgt caatagattt aactttgtat 13980 ttagttccac aggatgcaag atcagtatag agtatatttt aaaagatctt aagattaagg 14040 accccagttg tatagcattc ataggtgaag gagctggtaa cttattatta cgtacggtag 14100 tagaacttca tccagacata agatacattt acagaagttt aaaagattgc aatgatcata 14160 gtttacctat tgaatttcta aggttataca acgggcatat aaacatagat tatggtgaga 14220 atttaaccat tcctgctaca gatgcaacta ataacattca ttggtcttat ttacatataa 14280 aatttgcaga acctattagc atctttgtct gcgatgctga attacctgtt acagccaatt 14340 ggagtaaaat tataattgaa tggagtaagc atgtaagaaa gtgcaagtac tgttcctctg 14400 taaatagatg cattttaatt gcaaaatatc atgctcaaga tgatattgat ttcaaattag 14460 ataacattac tatattaaaa acttatgtgt gcctaggtag caagttaaaa ggatctgaag 14520 tttacttagt ccttacaata ggcccttcaa atatacttcc tgtttttaat gttgtgcaaa 14580 atgctaaatt gattctttca agaactaaaa atttcattat gcctaaaaaa actgacaaag 14640 aatctatcga tgcaaatatt aaaagcttaa tacctttcct ttgttaccct ataacaaaaa 14700 aaggaattaa gacttcattg tcaaaattga agagtgtagt taatggagat atattatcat 14760 attctatagc tggacgtaat gaagtattca gcaacaagct tataaaccac aagcatatga 14820 atatcctaaa atggctagat catgttttaa actttagatc aactgaactt aattacaatc 14880 atttat'atat gatagagtcc acatatcctt acttaagtga attgttaaat agtttaacaa 14940 ccaatgagct caaaaagctg attaaaatta caggtagtgt actatacaac cttcccaatg 15000 aacagtaact taaaatatca ttaacaagtt tggtcaaatt tagatgctaa cacatcatta 15060 tattatagtt attaaaaaat atgcaaactt ttcaataatt tagcatattg attccaaaat 15120 tatctatttt ggtcttaagg ggttaaataa aaatctaaaa ctaacaatta tacatgtgca 15180 tttacaacac aacgagacat tagtttttga cacttttttt ctcgt 15225 <210> 2
<211> 139
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 2
Met Gly Cys Asn Ser Leu Ser Met lie Lys Val Arg Leu Gin Asn Leu 1 5 10 15
Phe Asp Asn Asp Glu Val Ala Leu Leu Lys lie Thr Cys Tyr Thr Asp 20 25 30
Lys Leu lie Leu Leu Thr Asn Ala Leu Ala Lys Ala Ala lie His Thr 35 40 45
lie Lys Leu Asn Gly lie Val Phe lie His Val lie Thr Ser Ser Glu 50 55 60
Val Cys Pro Asp Asn Asn lie Val Val Lys Ser Asn Phe Thr Thr Met 65 70 75 80
Pro lie Leu Gin Asn Gly Gly Tyr lie Trp Glu Leu lie Glu Leu Thr 85 90 95
His Cys Ser Gin Leu Asn Gly Leu Met Asp Asp Asn Cys Glu lie Lys 100 105 110
Phe Ser Lys Arg Leu Ser Asp Ser Val Met Thr Asp Tyr Met Asn Gin 115 120 125
lie Ser Asp Leu Leu Gly Leu Asp Leu Asn Ser 130 135
<210> 3
<211> 124
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 3
Met Ser Thr Thr Asn Asn Asn Thr Thr Met Gin Arg Leu Met lie Thr 1 5 10 15
Asp Met Arg Pro Leu Ser Met Glu Ser lie lie Thr Ser Leu Thr Lys 20 25 30
Glu lie lie Thr His Lys Phe lie Tyr Leu lie Asn Asn Glu Cys lie 35 40 45
Val Arg Lys Leu Asp Glu Arg Gin Ala Thr Phe Thr Phe Leu Val Asn 50 55 60
Tyr Glu Met Lys Leu Leu His Lys Val Gly Ser Thr Lys Tyr Lys Lys 65 70 75 80
Tyr Thr Glu Tyr Asn Thr Lys Tyr Gly Thr Phe Pro Met Pro lie Phe 85 90 95
lie Asn His Gly Gly Phe Leu Glu Cys lie Gly lie Lys Pro Thr Lys 100 105 110
His Thr Pro lie lie Tyr Lys Tyr Asp Leu Asn Pro 115 120
<210> 4
<211> 391
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 4
Met Ala Leu Ser Lys Val Lys Leu Asn Asp Thr Leu Asn Lys Asp Gin 1 5 10 15
Leu Leu Ser Ser Ser Lys Tyr Thr lie Gin Arg Ser Thr Gly Asp Asn 20 25 30
lie Asp Thr Pro Asn Tyr Asp Val Gin Lys His Leu Asn Lys Leu Cys 35 40 45
Gly Met Leu Leu lie Thr Glu Asp Ala Asn His Lys Phe Thr Gly Leu 50 55 60
lie Gly Met Leu Tyr Ala Met Ser Arg Leu Gly Arg Glu Asp Thr lie 65 70 75 80
Lys lie Leu Lys Asp Ala Gly Tyr His Val Lys Ala Asn Gly Val Asp 85 90 95
lie Thr Thr Tyr Arg Gin Asp lie Asn Gly Lys Glu Met Lys Phe Glu 100 105 110
Val Leu Thr Leu Ser Ser Leu Thr Ser Glu lie Gin Val Asn lie Glu 115 120 125
He Glu Ser Arg Lys Ser Tyr Lys Lys Met Leu Lys Glu Met Gly Glu 130 135 140
Val Ala Pro Glu Tyr Arg His Asp Ser Pro Asp Cys Gly Met He He 145 150 155 160
Leu Cys He Ala Ala Leu Val He Thr Lys Leu Ala Ala Gly Asp Arg 165 170 175
Ser Gly Leu Thr Ala Val He Arg Arg Ala Asn Asn Val Leu Lys Asn 180 185 190
Glu He Lys Arg Tyr Lys Gly Leu He Pro Lys Asp He Ala Asn Ser 195 200 205
Phe Tyr Glu Val Phe Glu Lys His Pro His Leu He Asp Val Phe Val 210 215 220
His Phe Gly He Ala Gin Ser Ser Thr Arg Gly Gly Ser Arg Val Glu 225 230 235 240
Gly He Phe Ala Gly Leu Phe Met Asn Ala Tyr Gly Ser Gly Gin Val 245 250 255
Met Leu Arg Trp Gly Val Leu Ala Lys Ser Val Lys Asn He Met Leu 260 265 270
Gly His Ala Ser Val Gin Ala Glu Met Glu Gin Val Val Glu Val Tyr 275 280 285
Glu Tyr Ala Gin Lys Leu Gly Gly Glu Ala Gly Phe Tyr His He Leu 290 295 300
Asn Asn Pro Lys Ala Ser Leu Leu Ser Leu Thr Gin Phe Pro Asn Phe 305 310 315 320
Ser Ser Val Val Leu Gly Asn Ala Ala Gly Leu Gly He Met Gly Glu 325 330 335
Tyr Arg Gly Thr Pro Arg Asn Gin Asp Leu Tyr Asp Ala Ala Lys Ala 340 345 350 Tyr Ala Glu Gin Leu Lys Glu Asn Gly Val He Asn Tyr Ser Val Leu 355 360 365
Asp Leu Thr Ala Glu Glu Leu Glu Ala He Lys His Gin Leu Asn Pro 370 375 380
Lys Glu Asp Asp Val Glu Leu 385 390
<210> 5
<211> 241
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 5
Met Glu Lys Phe Ala Pro Glu Phe His Gly Glu Asp Ala Asn Asn Lys 1 5 10 15
Ala Thr Lys Phe Leu Glu Ser He Lys Gly Lys Phe Ala Ser Ser Lys 20 25 30
Asp Pro Lys Lys Lys Asp Ser He He Ser Val Asn Ser He Asp He 35 40 45
Glu Val Thr Lys Glu Ser Pro He Thr Ser Gly Thr Asn He Asn Asn 50 55 60
Pro Thr Ser Glu Ala Asp Ser Thr Pro Glu Ala Lys Thr Asn Tyr Pro 65 70 75 80
Arg Lys Pro Leu Val Ser Phe Lys Glu Asp Leu Thr Pro Ser Asp Asn 85 90 95
Pro Phe Ser Lys Leu Tyr Lys Glu Thr He Glu Thr Phe Asp Asn Asn 100 105 110
Glu Glu Glu Ser Ser Tyr Ser Tyr Glu Glu He Asn Asp Gin Thr Asn 115 120 125
Asp Asn He Thr Ala Arg Leu Asp Arg He Asp Glu Lys Leu Ser Glu 130 135 140
He Leu Gly Met Leu His Thr Leu Val Val Ala Ser Ala Gly Pro Thr 145 150 155 160 Ser Ala Arg Asp Gly He Arg Asp Ala Met Val Gly Leu Arg Glu Glu 165 170 175
Met He Glu Lys He Arg Ala Glu Ala Leu Met Thr Asn Asp Arg Leu 180 185 190
Glu Ala Met Ala Arg Leu Arg Asn Glu Glu Ser Glu Lys Met Ala Lys 195 200 205
Asp Thr Ser Asp Glu Val Ser Leu Asn Pro Thr Ser Lys Lys Leu Ser 210 215 220
Asp Leu Leu Glu Asp Asn Asp Ser Asp Asn Asp Leu Ser Leu Asp Asp 225 230 235 240
Phe
<210> 6
<211> 256
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 6
Met Glu Thr Tyr Val Asn Lys Leu His Glu Gly Ser Thr Tyr Thr Ala 1 5 10 15
Ala Val Gin Tyr Asn Val Leu Glu Lys Asp Asp Asp Pro Ala Ser Leu 20 25 30
Thr He Trp Val Pro Met Phe Gin Ser Ser Val Pro Ala Asp Leu Leu 35 40 45
He Lys Glu Leu Ala Ser He Asn He Leu Val Lys Gin He Ser Thr 50 55 60
Pro Lys Gly Pro Ser Leu Arg Val Thr He Asn Ser Arg Ser Ala Val 65 70 75 80
Leu Ala Gin Met Pro Ser Asn Phe He He Ser Ala Asn Val Ser Leu 85 90 95
Asp Glu Arg Ser Lys Leu Ala Tyr Asp Val Thr Thr Pro Cys Glu He 100 105 110 Lys Ala Cys Ser Leu Thr Cys Leu Lys Val Lys Ser Met Leu Thr Thr 115 120 125
Val Lys Asp Leu Thr Met Lys Thr Phe Asn Pro Thr His Glu He He 130 135 140
Ala Leu Cys Glu Phe Glu Asn He Met Thr Ser Lys Arg Val He He 145 150 155 160
Pro Thr Tyr Leu Arg Ser He Ser Val Lys Asn Lys Asp Leu Asn Ser 165 170 175
Leu Glu Asn He Ala Thr Thr Glu Phe Lys Asn Ala He Thr Asn Ala 180 185 190
Lys He He Pro Tyr Ala Gly Leu Val Leu Val He Thr Val Thr Asp 195 200 205
Asn Lys Gly Ala Phe Lys Tyr He Lys Pro Gin Ser Gin Phe He Val 210 215 220
Asp Leu Gly Ala Tyr Leu Glu Lys Glu Ser He Tyr Tyr Val Thr Thr 225 230 235 240
Asn Trp Lys His Thr Ala Thr Arg Phe Ser He Lys Pro Leu Glu Asp 245 250 255
<210> 7
<211> 65
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 7
Met Gly Asn Thr Ser He Thr He Glu Phe Thr Ser Lys Phe Trp Pro 1 5 10 15
Tyr Phe Thr Leu He His Met He Leu Thr Leu He Ser Leu Leu He 20 25 30
He He Thr He Met He Ala He Leu Asn Lys Leu Ser Glu His Lys 35 40 45
Thr Phe Cys Asn Lys Thr Leu Glu Leu Gly Gin Met Tyr Gin He Asn 50 55 60 Thr 65
<210> 8
<211> 574
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 8
Met Glu Leu Leu He His Arg Ser Ser Ala He Phe Leu Thr Leu Ala 1 5 10 15
He Asn Ala Leu Tyr Leu Thr Ser Ser Gin Asn He Thr Glu Glu Phe 20 25 30
Tyr Gin Ser Thr Cys Ser Ala Val Ser Arg Gly Tyr Phe Ser Ala Leu 35 40 45
Arg Thr Gly Trp Tyr Thr Ser Val He Thr He Glu Leu Ser Asn He 50 55 60
Lys Glu Thr Lys Cys Asn Gly Thr Asp Thr Lys Val Lys Leu He Lys 65 70 75 80
Gin Glu Leu Asp Lys Tyr Lys Asn Ala Val Thr Glu Leu Gin Leu Leu 85 90 95
Thr Gin Asn Thr Pro Ala Ala Asn Asn Arg Ala Arg Arg Glu Ala Pro 100 105 110
Gin Tyr Met Asn Tyr Thr He Asn Thr Thr Lys Asn Leu Asn Val Ser 115 120 125
He Ser Lys Lys Arg Lys Arg Arg Phe Leu Gly Phe Leu Leu Gly Val 130 135 140
Gly Ser Ala He Ala Ser Gly He Ala Val Ser Lys Val Leu His Leu 145 150 155 160
Glu Gly Glu Val Asn Lys He Lys Asn Ala Leu Leu Ser Thr Asn Lys 165 170 175
Ala Val Val Ser Leu Ser Asn Gly Val Ser Val Leu Thr Ser Lys Val 180 185 190 Leu Asp Leu Lys Ser Tyr He Asn Asn Gin Leu Leu Pro He Val Asn 195 200 205
Gin Gin Ser Cys Arg He Ser Asn He Glu Thr Val He Glu Phe Gin 210 215 220
Gin Lys Asn Ser Arg Leu Leu Glu He Thr Arg Glu Phe Ser Val Asn 225 230 235 240
Ala Gly Val Thr Thr Pro Leu Ser Thr Tyr Met Leu Thr Asn Ser Glu 245 250 255
Leu Leu Ser Leu He Asn Asp Met Pro He Thr Asn Asp Gin Lys Lys 260 265 270
Leu Met Ser Ser Asn Val Gin He Val Arg Gin Gin Ser Tyr Ser He 275 280 285
Met Ser He He Lys Glu Glu Val Leu Ala Tyr Val Val Gin Leu Pro 290 295 300
He Tyr Gly Val He Asp Thr Pro Cys Trp Lys Leu His Thr Ser Pro 305 310 315 320
Leu Cys Thr Thr Asn He Lys Glu Gly Ser Asn He Cys Leu Thr Arg 325 330 335
Thr Asp Arg Gly Trp Tyr Cys Asp Asn Ala Gly Ser Val Ser Phe Phe 340 345 350
Pro Gin Ala Asp Thr Cys Lys Val Gin Ser Asn Arg Val Phe Cys Asp 355 360 365
Thr Met Asn Ser Leu Thr Leu Pro Ser Glu Val Ser Leu Cys Asn Thr 370 375 380
Asp He Phe Asn Ser Lys Tyr Asp Cys Lys He Met Thr Ser Lys Thr 385 390 395 400
Asp He Ser Ser Ser Val He Thr Ser Leu Gly Ala He Val Ser Cys 405 410 415
Tyr Gly Lys Thr Lys Cys Thr Ala Ser Asn Lys Asn Arg Gly He He 420 425 430 Lys Thr Phe Ser Asn Gly Cys Asp Tyr Val Ser Asn Lys Gly Val Asp 435 440 445
Thr Val Ser Val Gly Asn Thr Leu Tyr Tyr Val Asn Lys Leu Glu Gly 450 455 460
Lys Asn Leu Tyr Val Lys Gly Glu Pro He He Asn Tyr Tyr Asp Pro 465 470 475 480
Leu Val Phe Pro Ser Asp Glu Phe Asp Ala Ser He Ser Gin Val Asn 485 490 495
Glu Lys He Asn Gin Ser Leu Ala Phe He Arg Arg Ser Asp Glu Leu 500 505 510
Leu His Asn Val Asn Thr Gly Lys Ser Thr Thr Asn He Met He Thr 515 520 525
Thr He He He Val He He Val Val Leu Leu Ser Leu He Ala He 530 535 540
Gly Leu Leu Leu Tyr Cys Lys Ala Lys Asn Thr Pro Val Thr Leu Ser 545 550 555 560
Lys Asp Gin Leu Ser Gly He Asn Asn He Ala Phe Ser Lys 565 570
<210> 9
<211> 195
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 9
Met Ser Arg Arg Asn Pro Cys Lys Phe Glu He Arg Gly His Cys Leu 1 5 10 15
Asn Gly Arg Arg Cys His Tyr Ser His Asn Tyr Phe Glu Trp Pro Pro 20 25 30
His Ala Leu Leu Val Arg Gin Asn Phe Met Leu Asn Lys He Leu Lys 35 40 45
Ser Met Asp Lys Ser He Asp Thr Leu Ser Glu He Ser Gly Ala Ala 50 55 60 Glu Leu Asp Arg Thr Glu Glu Tyr Ala Leu Gly He Val Gly Val Leu 65 70 75 80
Glu Ser Tyr He Gly Ser He Asn Asn He Thr Lys Gin Ser Ala Cys 85 90 95
Val Ala Met Ser Lys Leu Leu He Glu He Asn Ser Asp Asp He Lys 100 105 110
Lys Leu Arg Asp Asn Glu Glu Pro Asn Ser Pro Lys He Arg Val Tyr 115 120 125
Asn Thr Val He Ser Tyr He Glu Ser Asn Arg Lys Asn Asn Lys Gin 130 135 140
Thr He His Leu Leu Lys Arg Leu Pro Ala Asp Val Leu Lys Lys Thr 145 150 155 160
He Lys Asn Thr Leu Asp He His Lys Ser He Thr He Ser Asn Pro 165 170 175
Lys Glu Ser Thr Val Asn Asp Gin Asn Asp Gin Thr Lys Asn Asn Asp 180 185 190
He Thr Gly 195
<210> 10
<211> 93
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 10
Met He Lys Met Thr Lys Pro Lys He Met He Leu Pro Asp Lys Tyr 1 5 10 15
Pro Cys Ser He Ser Ser He Leu He Ser Ser Glu Ser Met Val Ala 20 25 30
Thr Phe Asn His Lys Asn He Leu Gin Phe Asn His Asn His Leu Asp 35 40 45
Asn His Gin Cys Leu Leu Asn His He Phe Asp Glu He His Trp Thr 50 55 60 Pro Lys Asn Leu Leu Asp Thr Thr Gin Gin Phe Leu Gin His Leu Asn 65 70 75 80
He Pro Glu Asp He Tyr Thr Val Tyr He Leu Val Ser 85 90
<210> 11
<211> 2166
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 11
Met Asp Pro He He Asn Gly Asn Ser Ala Asn Val Tyr Leu Thr Asp 1 5 10 15
Ser Tyr Leu Lys Gly Val He Ser Phe Ser Glu Cys Asn Ala Leu Gly 20 25 30
Ser Tyr Leu Phe Asn Gly Pro Tyr Leu Lys Asn Asp Tyr Thr Asn Leu 35 40 45
He Ser Arg Gin Ser Pro Leu Leu Glu His Met Asn Leu Lys Lys Leu 50 55 60
Thr He Thr Gin Ser Leu He Ser Arg Tyr His Lys Gly Glu Leu Lys 65 70 75 80
Leu Glu Glu Pro Thr Tyr Phe Gin Ser Leu Leu Met Thr Tyr Lys Ser 85 90 95
Met Ser Ser Ser Glu Gin He Ala Thr Thr Asn Leu Leu Lys Lys He 100 105 110
He Arg Arg Ala He Glu He Ser Asp Val Lys Val Tyr Ala He Leu 115 120 125
Asn Lys Leu Gly Leu Lys Glu Lys Asp Arg Val Lys Pro Asn Asn Asn 130 135 140
Ser Gly Asp Glu Asn Ser Val Leu Thr Thr He He Lys Asp Asp He 145 150 155 160
Leu Ser Ala Val Glu Asn Asn Gin Ser Tyr Thr Asn Ser Asp Lys Asn 165 170 175 His Ser Val Asn Gin Asn He Thr He Lys Thr Thr Leu Leu Lys Lys 180 185 190
Leu Met Cys Ser Met Gin His Pro Pro Ser Trp Leu He His Trp Phe 195 200 205
Asn Leu Tyr Thr Lys Leu Asn Asn He Leu Thr Gin Tyr Arg Ser Asn 210 215 220
Glu Val Lys Ser His Gly Phe He Leu He Asp Asn Gin Thr Leu Ser 225 230 235 240
Gly Phe Gin Phe He Leu Asn Gin Tyr Gly Cys He Val Tyr His Lys 245 250 255
Gly Leu Lys Lys He Thr Thr Thr Thr Tyr Asn Gin Phe Leu Thr Trp 260 265 270
Lys Asp He Ser Leu Ser Arg Leu Asn Val Cys Leu He Thr Trp He 275 280 285
Ser Asn Cys Leu Asn Thr Leu Asn Lys Ser Leu Gly Leu Arg Cys Gly 290 295 300
Phe Asn Asn Val Val Leu Ser Gin Leu Phe Leu Tyr Gly Asp Cys He 305 310 315 320
Leu Lys Leu Phe His Asn Glu Gly Phe Tyr He He Lys Glu Val Glu 325 330 335
Gly Phe He Met Ser Leu He Leu Asn He Thr Glu Glu Asp Gin Phe 340 345 350
Arg Thr Arg Phe Tyr Asn Ser Met Leu Asn Asn He Thr Asp Ala Ala 355 360 365
He Lys Ala Gin Lys Asn Leu Leu Ser Arg Val Cys His Thr Leu Leu 370 375 380
Asp Lys Thr Val Ser Asp Asn He He Asn Gly Lys Trp He He Leu 385 390 395 400
Leu Ser Lys Phe Leu Lys Leu He Lys Leu Ala Gly Asp Asn Asn Leu 405 410 415 Asn Asn Leu Ser Glu Leu Tyr Phe Leu Phe Arg He Phe Gly His Pro 420 425 430
Met Val Asp Glu Arg Gin Ala Met Asp Ala Val Arg He Asn Cys Asn 435 440 445
Glu Thr Lys Phe Tyr Leu Leu Ser Ser Leu Ser Thr Leu Arg Gly Ala 450 455 460
Phe He Tyr Arg He He Lys Gly Phe Val Asn Thr Tyr Asn Arg Trp 465 470 475 480
Pro Thr Leu Arg Asn Ala He Val Leu Pro Leu Arg Trp Leu Asn Tyr 485 490 495
Tyr Lys Leu Asn Thr Tyr Pro Ser Leu Leu Glu He Thr Glu Asn Asp 500 505 510
Leu He He Leu Ser Gly Leu Arg Phe Tyr Arg Glu Phe His Leu Pro 515 520 525
Lys Lys Val Asp Leu Glu Met He He Asn Asp Lys Ala He Ser Pro 530 535 540
Pro Lys Asp Leu He Trp Thr Ser Phe Pro Arg Asn Tyr Met Pro Ser 545 550 555 560
His He Gin Asn Tyr He Glu His Glu Lys Leu Lys Phe Ser Glu Ser 565 570 575
Asp Arg Ser Arg Arg Val Leu Glu Tyr Tyr Leu Arg Asp Asn Lys Phe 580 585 590
Asn Glu Cys Asp Leu Tyr Asn Cys Val Val Asn Gin Ser Tyr Leu Asn 595 600 605
Asn Ser Asn His Val Val Ser Leu Thr Gly Lys Glu Arg Glu Leu Ser 610 615 620
Val Gly Arg Met Phe Ala Met Gin Pro Gly Met Phe Arg Gin He Gin 625 630 635 640
He Leu Ala Glu Lys Met He Ala Glu Asn He Leu Gin Phe Phe Pro 645 650 655
Glu Ser Leu Thr Arg Tyr Gly Asp Leu Glu Leu Gin Lys He Leu Glu 660 665 670
Leu Lys Ala Gly He Ser Asn Lys Ser Asn Arg Tyr Asn Asp Asn Tyr 675 680 685
Asn Asn Tyr He Ser Lys Cys Ser He He Thr Asp Leu Ser Lys Phe 690 695 700
Asn Gin Ala Phe Arg Tyr Glu Thr Ser Cys He Cys Ser Asp Val Leu 705 710 715 720
Asp Glu Leu His Gly Val Gin Ser Leu Phe Ser Trp Leu His Leu Thr 725 730 735
He Pro Leu Val Thr He He Cys Thr Tyr Arg His Ala Pro Pro Phe 740 745 750
He Lys Asp His Val Val Asn Leu Asn Glu Val Asp Glu Gin Ser Gly 755 760 765
Leu Tyr Arg Tyr His Met Gly Gly He Glu Gly Trp Cys Gin Lys Leu 770 775 780
Trp Thr He Glu Ala He Ser Leu Leu Asp Leu He Ser Leu Lys Gly 785 790 795 800
Lys Phe Ser He Thr Ala Leu He Asn Gly Asp Asn Gin Ser He Asp 805 810 815
He Ser Lys Pro Val Arg Leu He Glu Gly Gin Thr His Ala Gin Ala 820 825 830
Asp Tyr Leu Leu Ala Leu Asn Ser Leu Lys Leu Leu Tyr Lys Glu Tyr 835 840 845
Ala Gly He Gly His Lys Leu Lys Gly Thr Glu Thr Tyr He Ser Arg 850 855 860
Asp Met Gin Phe Met Ser Lys Thr He Gin His Asn Gly Val Tyr Tyr 865 870 875 880 Pro Ala Ser He Lys Lys Val Leu Arg Val Gly Pro Trp He Asn Thr 885 890 895
He Leu Asp Asp Phe Lys Val Ser Leu Glu Ser He Gly Ser Leu Thr 900 905 910
Gin Glu Leu Glu Tyr Arg Gly Glu Ser Leu Leu Cys Ser Leu He Phe 915 920 925
Arg Asn He Trp Leu Tyr Asn Gin He Ala Leu Gin Leu Arg Asn His 930 935 940
Ala Leu Cys His Asn Lys Leu Tyr Leu Asp He Leu Lys Val Leu Lys 945 950 955 960
His Leu Lys Thr Phe Phe Asn Leu Asp Ser He Asp Met Ala Leu Ser 965 970 975
Leu Tyr Met Asn Leu Pro Met Leu Phe Gly Gly Gly Asp Pro Asn Leu 980 985 990
Leu Tyr Arg Ser Phe Tyr Arg Arg Thr Pro Asp Phe Leu Thr Glu Ala 995 1000 1005
He Val His Ser Val Phe Val Leu Ser Tyr Tyr Thr Gly His Asp 1010 1015 1020
Leu Gin Asp Lys Leu Gin Asp Leu Pro Asp Asp Arg Leu Asn Lys 1025 1030 1035
Phe Leu Thr Cys He He Thr Phe Asp Lys Asn Pro Asn Ala Glu 1040 1045 1050
Phe Val Thr Leu Met Arg Asp Pro Gin Ala Leu Gly Ser Glu Arg 1055 1060 1065
Gin Ala Lys He Thr Ser Glu He Asn Arg Leu Ala Val Thr Glu 1070 1075 1080
Val Leu Ser He Ala Pro Asn Lys He Phe Ser Lys Ser Ala Gin 1085 1090 1095
His Tyr Thr Thr Thr Glu He Asp Leu Asn Asp He Met Gin Asn 1100 1105 1110 He Glu Pro Thr Tyr Pro His Gly Leu Arg Val Val Tyr Glu Ser 1115 1120 1125
Leu Pro Phe Tyr Lys Ala Glu Lys He Val Asn Leu He Ser Gly 1130 1135 1140
Thr Lys Ser He Thr Asn lie Leu Glu Lys Thr Ser Ala He Asp 1145 1150 1155
Thr Thr Asp He Asn Arg Ala Thr Asp Met Met Arg Lys Asn He 1160 1165 1170
Thr Leu Leu He Arg He Leu Pro Leu Asp Cys Asn Lys Asp Lys 1175 1180 1185
Arg Glu Leu Leu Ser Leu Glu Asn Leu Ser He Thr Glu Leu Ser 1190 1195 1200
Lys Tyr Val Arg Glu Arg Ser Trp Ser Leu Ser Asn He Val Gly 1205 1210 1215
Val Thr Ser Pro Ser He Met Phe Thr Met Asp He Lys Tyr Thr 1220 1225 1230
Thr Ser Thr He Ala Ser Gly He He He Glu Lys Tyr Asn Val 1235 1240 1245
Asn Ser Leu Thr Arg Gly Glu Arg Gly Pro Thr Lys Pro Trp Val 1250 1255 1260
Gly Ser Ser Thr Gin Glu Lys Lys Thr Met Pro Val Tyr Asn Arg 1265 1270 1275
Gin Val Leu Thr Lys Lys Gin Arg Asp Gin He Asp Leu Leu Ala 1280 1285 1290
Lys Leu Asp Trp Val Tyr Ala Ser He Asp Asn Lys Asp Glu Phe
1295 1300 1305
Met Glu Glu Leu Ser Thr Gly Thr Leu Gly Leu Ser Tyr Glu Lys 1310 1315 1320
Ala Lys Lys Leu Phe Pro Gin Tyr Leu Ser Val Asn Tyr Leu His 1325 1330 1335 Arg Leu Thr Val Ser Ser Arg Pro Cys Glu Phe Pro Ala Ser He 1340 1345 1350
Pro Ala Tyr Arg Thr Thr Asn Tyr His Phe Asp Thr Ser Pro He 1355 1360 1365
Asn His Val Leu Thr Glu Lys Tyr Gly Asp Glu Asp He Asp He 1370 1375 1380
Val Phe Gin Asn Cys He Ser Phe Gly Leu Ser Leu Met Ser Val 1385 1390 1395
Val Glu Gin Phe Thr Asn He Cys Pro Asn Arg He He Leu He 1400 1405 1410
Pro Lys Leu Asn Glu He His Leu Met Lys Pro Pro He Phe Thr 1415 1420 1425
Gly Asp Val Asp He He Lys Leu Lys Gin Val He Gin Lys Gin 1430 1435 1440
His Met Phe Leu Pro Asp Lys He Ser Leu Thr Gin Tyr Val Glu 1445 1450 1455
Leu Phe Leu Ser Asn Lys Ala Leu Lys Ser Gly Ser His He Asn 1460 1465 1470
Ser Asn Leu He Leu Val His Lys Met Ser Asp Tyr Phe His Asn 1475 1480 1485
Asp Tyr He Leu Ser Thr Asn Leu Ala Gly His Trp He Leu He 1490 1495 1500
He Gin Leu Met Lys Asp Ser Lys Gly He Phe Glu Lys Asp Trp 1505 1510 1515
Gly Glu Gly Tyr He Thr Asp His Met Phe He Asn Leu Asn Val 1520 1525 1530
Phe Phe Asn Ala Tyr Lys Thr Tyr Leu Leu Cys Phe His Lys Gly 1535 1540 1545
Tyr Gly Lys Ala Lys Leu Glu Cys Asp Met Asn Thr Ser Asp Leu 1550 1555 1560
Leu Cys Val Leu Glu Leu He Asp Ser Ser Tyr Trp Lys Ser Met 1565 1570 1575
Ser Lys Val Phe Leu Glu Gin Lys Val He Lys Tyr He Val Asn 1580 1585 1590
Gin Asp Thr Ser Leu His Arg He Lys Gly Cys His Ser Phe Lys 1595 1600 1605
Leu Trp Phe Leu Lys Arg Leu Asn Asn Ala Lys Phe Thr Val Cys 1610 1615 1620
Pro Trp Val Val Asn He Asp Tyr His Pro Thr His Met Lys Ala 1625 1630 1635
He Leu Ser Tyr He Asp Leu Val Arg Met Gly Leu He Asn Val 1640 1645 1650
Asp Lys Leu Thr He Lys Asn Lys Asn Lys Phe Asn Asp Glu Phe 1655 1660 1665
Tyr Thr Ser Asn Leu Phe Tyr He Ser Tyr Asn Phe Ser Asp Asn 1670 1675 1680
Thr His Leu Leu iThr Lys Gin He Arg He Ala Asn Ser Glu Leu 1685 1690 1695
Glu Asn Asn Tyr Asn Lys Leu Tyr His Pro Thr Pro Glu Thr Leu 1700 1705 1710
Glu Asn Met Ser Leu He Pro Val Lys Ser Asn Asn Ser Asn Lys 1715 1720 1725
Pro Lys Ser Cys He Ser Gly Asn Thr Glu Ser Met Met Thr Ser 1730 1735 1740
Thr Phe Ser Asn Lys Met His He Lys Ser Ser Thr Val Thr Thr 1745 1750 1755
Arg Leu Asn Tyr Ser Lys Gin Asp Leu Tyr Asn Leu Phe Pro He 1760 1765 1770 Val Val He Asp Arg He He Asp His Ser Gly Asn Thr Ala Lys 1775 1780 1785
Ser Asn Gin Leu Tyr Thr Thr Thr Ser His Gin Thr Ser Leu Val 1790 1795 1800
Arg Asn Ser Ala Ser Leu Tyr Cys Met Leu Pro Trp His His Val 1805 1810 1815
Asn Arg Phe Asn Phe Val Phe Ser Ser Thr Gly Cys Lys He Ser 1820 1825 1830
He Glu Tyr He Leu Lys Asp Leu Lys He Lys Asp Pro Ser Cys 1835 1840 1845
He Ala Phe He Gly Glu Gly Ala Gly Asn Leu Leu Leu Arg Thr 1850 1855 1860
Val Val Glu Leu His Pro Asp He Arg Tyr He Tyr Arg Ser Leu 1865 1870 1875
Lys Asp Cys Asn Asp His Ser Leu Pro He Glu Phe Leu Arg Leu 1880 1885 1890
Tyr Asn Gly His He Asn He Asp Tyr Gly Glu Asn Leu Thr He 1895 1900 1905
Pro Ala Thr Asp Ala Thr Asn Asn He His Trp Ser Tyr Leu His 1910 1915 1920
He Lys Phe Ala Glu Pro He Ser He Phe Val Cys Asp Ala Glu 1925 1930 1935
Leu Pro Val Thr Ala Asn Trp Ser Lys He He He Glu Trp Ser 1940 1945 1950
Lys His Val Arg Lys Cys Lys Tyr Cys Ser Ser Val Asn Arg Cys 1955 1960 1965
He Leu He Ala Lys Tyr His Ala Gin Asp Asp He Asp Phe Lys 1970 1975 1980
Leu Asp Asn He Thr He Leu Lys Thr Tyr Val Cys Leu Gly Ser 1985 1990 1995 Lys Leu Lys Gly Ser Glu Val Tyr Leu Val Leu Thr He Gly Pro 2000 2005 2010
Ser Asn He Leu Pro Val Phe Asn Val Val Gin Asn Ala Lys Leu 2015 2020 2025
He Leu Ser Arg Thr Lys Asn Phe He Met Pro Lys Lys Thr Asp 2030 2035 2040
Lys Glu Ser He Asp Ala Asn He Lys Ser Leu He Pro Phe Leu 2045 2050 2055
Cys Tyr Pro He Thr Lys Lys Gly He Lys Thr Ser Leu Ser Lys 2060 2065 2070
Leu Lys Ser Val Val Asn Gly Asp He Leu Ser Tyr Ser He Ala 2075 2080 2085
Gly Arg Asn Glu Val Phe Ser Asn Lys Leu He Asn His Lys His 2090 2095 2100
Met Asn He Leu Lys Trp Leu Asp His Val Leu Asn Phe Arg Ser 2105 2110 2115
Thr Glu Leu Asn Tyr Asn His Leu Tyr Met He Glu Ser Thr Tyr 2120 2125 2130
Pro Tyr Leu Ser Glu Leu Leu Asn Ser Leu Thr Thr Asn Glu Leu 2135 2140 2145
Lys Lys Leu He Lys He Thr Gly Ser Val Leu Tyr Asn Leu Pro 2150 2155 2160
Asn Glu Gin 2165
<210> 12
<211> 292
<212> PRT
<213> respiratory syncytial virus B 9320
<400> 12
Met Ser Lys His Lys Ser Gin Arg Thr Ala Arg Thr Leu Glu Lys Thr 1 5 10 15 Trp Asp Thr Leu Asn His Leu He Val He Ser Ser Cys Leu Tyr Arg 20 25 30
Leu Asn Leu Lys Ser He Ala Gin He Ala Leu Ser Val Leu Ala Met 35 40 45
He He Ser Thr Ser Leu He He Ala Ala He He Phe He He Ser 50 55 60
Ala Asn His Lys Val Thr Leu Thr Thr Val Thr Val Gin Thr He Lys 65 70 75 80
Asn His Thr Glu Lys Asn He Thr Thr Tyr Leu Thr Gin Val Ser Pro 85 90 95
Glu Arg Val Ser Ser Ser He Gin Pro Thr Thr Thr Ser Pro He His 100 105 110
Thr Asn Ser Ala Thr He Ser Pro Asn Thr Lys Ser Glu Thr His His 115 120 125
Thr Thr Thr Gin Ala Lys Ser Arg He Thr Thr Ser Thr Gin Thr Asn 130 135 140
Lys Pro Ser Thr Lys Ser Arg Ser Lys Asn Pro Pro Lys Lys Pro Lys 145 150 155 160
Asp Asp Tyr His Phe Glu Val Phe Asn Phe Val Pro Cys Ser He Cys 165 170 175
Gly Asn Asn Gin Leu Cys Lys Ser He Cys Lys Thr He Pro Ser Asn 180 185 190
Lys Pro Lys Lys Lys Pro Thr He Lys Pro Thr Asn Lys Pro Thr Val 195 200 205
Lys Thr Thr Asn Lys Arg Asp Pro Lys Thr Pro Ala Lys Met Met Lys 210 215 220
Lys Glu Thr Thr Thr Asn Pro Thr Lys Lys Pro Thr Leu Lys Thr Thr 225 230 235 240
Glu Gly Asp Thr Ser Thr Ser Gin Ser Thr Val Leu Asp Thr Thr Thr 245 250 255 Ser Lys His Thr He Gin Gin Gin Ser Leu His Ser He Thr Ser Glu 260 265 270
Asn Thr Pro Asn Ser Thr Gin He Pro Thr Ala Thr Glu Ala Ser Thr 275 280 285
Ser Asn Ser Thr 290
<210> 13
<211> 15225
<212> DNA
<213> respiratory syncytial virus B 1
<400> 13 acgcgaaaaa atgcgtacta caaacttgca cattcggaaa aaatggggca aataagaatt 60 tgataagtgc tatttaagtc taaccttttc aatcagaaat ggggtgcaat tcactgagca 120 tgataaaggt tagattacaa aatttatttg acaatgacga agtagcattg ttaaaaataa 180 catgttatac tgacaaatta attcttctga ccaatgcatt agccaaagca gcaatacata 240 caattaaatt aaacggtata gtttttatac atgttataac aagcagtgaa gtgtgccctg 300 ataacaacat tgtagtaaaa tctaacttta caacaatgcc aatattacaa aacggaggat 360 acatatggga attgattgag ttgacacact gctctcaatt aaacggtcta atggatgata 420 attgtgaaat caaattttct aaaagactaa gtgactcagt aatgactaat tatatgaatc 480 aaatatctga tttacttggg cttgatctca attcatgaat tatgtttagt ctaactcaat 540 agacatgtgt ttattaccat tttagttaat ataaaaactc atcaaaggga aatggggcaa 600 ataaactcac ctaatcaatc aaactatgag cactacaaat gacaacacta ctatgcaaag 660 attaatgatc acggacatga gacccctgtc gatggattca ataataacat ctctcaccaa 720 agaaatcatc acacacaaat tcatatactt gataaacaat gaatgtattg taagaaaact 780 tgatgaaaga caagctacat ttacattctt agtcaattat gagatgaagc tactgcacaa 840 agtagggagt accaaataca agaaatacac tgaatataat acaaaatatg gcactttccc 900 catgcctata tttatcaatc atggcgggtt tctagaatgt attggcatta agcctacaaa 960 acacactcct ataatataca aatatgacct caacccgtaa attccaacaa aaaaaaccaa 1020 cccaaccaaa ccaagctatt cctcaaacaa caatgctcaa tagttaagaa ggagctaatc 1080 cgttttagta attaaaaata aaagtaaagc caataacata aattggggca aatacaaaga 1140 tggctcttag caaagtcaag ttaaatgata cattaaataa ggatcagctg ctgtcatcca 1200 gcaaatacac tattcaacgt agtacaggag ataatattga cactcccaat tatgatgtgc 1260 aaaaacacct aaacaaacta tgtggtatgc tattaatcac tgaagatgca aatcataaat 1320 tcacaggatt aataggtatg ttatatgcta tgtccaggtt aggaagggaa gacactataa 1380 agatacttaa agatgctgga tatcatgtta aagctaatgg agtagatata acaacatatc 1440 gtcaagatat aaatggaaag gaaatgaaat tcgaagtatt aacattatca agcttgacat 1500 cagaaataca agtcaatatt gagatagaat ctagaaaatc ctacaaaaaa atgctaaaag 1560 agatgggaga agtggctcca gaatataggc atgattctcc agactgtggg atgataatac 1620 tgtgtatagc agcacttgta ataaccaaat tagcagcagg agacagatca ggtcttacag 1680 cagtaattag gagggcaaac aatgtcttaa aaaatgaaat aaaacgctac aagggtctca 1740 taccaaagga tatagctaac agtttttatg aagtgtttga aaaacaccct catcttatag 1800 atgtttttgt gcactttggc attgcacaat catcaacaag agggggtagt agagttgaag 1860 gaatctttgc aggattgttt atgaatgcct atggttcagg gcaagtaatg ctaagatggg 1920 gagttttagc caaatctgta aaaaatatca tgctaggtca tgctagtgtc caggcagaaa 1980 tggagcaagt tgtggaagtc tatgagtatg cacagaagtt gggaggagaa gctggattct 2040 accatatatt gaacaatcca aaagcatcat tgctgtcatt aactcaattt cctaacttct 2100 caagtgtggt cctaggcaat gcagcaggtc taggcataat gggagagtat agaggtacgc 2160 caagaaacca ggatctttat gatgcagcca aagcatatgc agagcaactc aaagaaaatg 2220 gagtaataaa ctacagtgta ttagacttaa cagcagaaga attggaagcc ataaagaatc 2280 aactcaaccc taaagaagat gatgtagagc tttaagttaa caaaaaatac ggggcaaata 2340 agtcaacatg gagaagtttg cacctgaatt tcatggagaa gatgcaaata acaaagctac 2400 caaattccta gaatcaataa agggcaagtt cgcatcatcc aaagatccta agaagaaaga 2460 tagcataata tctgttaact caatagatat agaagtaacc aaagagagcc cgataacatc 2520 tggcaccaac atcatcaatc caacaagtga agccgacagt accccagaaa ccaaagccaa 2580 ctacccaaga aaacccctag taagcttcaa agaagatctc accccaagtg acaacccttt 2640 ttctaagttg tacaaagaaa caatagaaac atttgataac aatgaagaag aatctagcta 2700 ctcatatgaa gagataaatg atcaaacaaa tgacaacatt acagcaagac tagatagaat 2760 tgatgaaaaa ttaagtgaaa tattaggaat gctccataca ttagtagttg caagtgcagg 2820 acccacttca gctcgcgatg gaataagaga tgctatggtt ggtctgagag aagaaatgat 2880 agaaaaaata agagcggaag cattaatgac caatgatagg ttagaggcta tggcaagact 2940 taggaatgag gaaagcgaaa aaatggcaaa agacacctca gatgaagtgc ctcttaatcc 3000 aacttccaaa aaattgagtg acttgttgga agacaacgat agtgacaatg atctgtcact 3060 tgatgatttt tgatcagtga tcaactcact cagcaatcaa caacatcaat aaaacagaca 3120 tcaatccatt gaatcaactg ccagaccgaa caaacaaatg tccgtcagcg gaaccaccaa 3180 ccaatcaatc aaccaactga tccatcagca acctgacgaa attaacaata tagtaacaaa 3240 aaaagaacaa gatggggcaa atatggaaac atacgtgaac aagcttcacg aaggctccac 3300 atacacagca gctgttcagt acaatgttct agaaaaagat gatgatcctg catcactaac 3360 aatatgggtg cctatgttcc agtcatctgt accagcagac ttgctcataa aagaacttgc 3420 aagcatcaac atactagtga agcagatctc tacgcccaaa ggaccttcac tacgagtcac 3480 gattaactca agaagtgctg tgctggctca aatgcctagt aatttcatca taagcgcaaa 3540 tgtatcatta gatgaaagaa gcaaattagc atatgatgta actacacctt gtgaaatcaa 3600 agcatgcagt ctaacatgct taaaagtgaa aagtatgtta actacagtca aagatcttac 3660
I, catgaagaca ttcaacccca ctcatgagat cattgctcta tgtgaatttg aaaatattat 3720 gacatcaaaa agagtaataa taccaaccta tctaagacca attagtgtca aaaacaagga 3780 tctgaactca ctagaaaaca tagcaaccac cgaattcaaa aatgctatca ccaatgcgaa 3840 aattattccc tatgctggat tagtattagt tatcacagtt actgacaata aaggagcatt 3900 caaatatatc aagccacaga gtcaatttat agtagatctt ggtgcctacc tagaaaaaga 3960 gagcatatat tatgtgacta ctaattggaa gcatacagct acacgttttt caatcaaacc 4020 actagaggat taaatttaat tatcaacact gaatgacagg tccacatata tcctcaaact 4080 acacactata tccaaacatc atgaacatct acactacaca cttcatcaca caaaccaatc 4140 ccactcaaaa tccaaaatca ctaccagcca ctatctgcta gacctagagt gcgaataggt 4200 aaataaaacc aaaatatggg gtaaatagac attagttaga gttcaatcaa tctcaacaac 4260 catttatacc gccaattcaa tacatatact ataaatctta aaatgggaaa tacatccatc 4320 acaatagaat tcacaagcaa attttggccc tattttacac taatacatat gatcttaact 4380 ctaatctctt tactaattat aatcactatt atgattgcaa tactaaataa gctaagtgaa 4440 cataaaacat tctgtaacaa tactcttgaa ctaggacaga tgcatcaaat caacacatag 4500 tgctctacca tcatgctgtg tcaaattata atcctgtata tataaacaaa caaatccaat 4560 cttctcacag agtcatggtg tcgcaaaacc acgccaacta tcatggtagc atagagtagt 4620 tatttaaaaa ttaacataat gatgaattat tagtatggga tcaaaaacaa cattggggca 4680 aatgcaacca tgtccaaaca caagaatcaa cgcactgcca ggactctaga aaagacctgg 4740 gatactctca atcatctaat tgtaatatcc tcttgtttat acagattaaa tttaaaatct 4800 atagcacaaa tagcactatc agttctggca atgataatct caacctctct cataattgca 4860 gccataatat tcatcatctc tgccaatcac aaagttacac taacaacggt cacagttcaa 4920 acaataaaaa accacactga aaaaaacatc accacctacc ttactcaagt cccaccagaa 4980 agggttagct catccaaaca acctacaacc acatcaccaa tccacacaaa ttcagccaca 5040 acatcaccca acacaaagtc agaaacacac cacacaacag cacaaaccaa aggcagaacc 5100 accacctcaa cacagaccaa caagccgagc acaaaaccac gcctaaaaaa tccaccaaaa 5160 aaaccaaaag atgattacca ttttgaagtg ttcaacttcg ttccctgtag tatatgtggc 5220 aacaatcaac tttgcaaatc catctgtaaa acaataccaa gcaacaaacc aaagaagaaa 5280 ccaaccatca aacccacaaa caaaccaacc accaaaacca caaacaaaag agacccaaaa 5340 acaccagcca aaacgacgaa aaaagaaact accaccaacc caacaaaaaa accaaccctc 5400 acgaccacag aaagagacac cagcacctca caatccactg tgctcgacac aaccacatta 5460 gaacacacaa tccaacagca atccctccac tcaaccaccc ccgaaaacac acccaactcc 5520 acacaaacac ccacagcatc cgagccctct acatcaaatt ccacccaaaa tacccaatca 5580 catgcttagt tattcaaaaa ctacatctta gcagaaaacc gtgacctatc aagcaagaac 5640 gaaattaaac ctggggcaaa taaccatgga gctgctgatc cacaggttaa gtgcaatctt 5700 cctaactctt gctattaatg cattgtacct cacctcaagt cagaacataa ctgaggagtt 5760 ttaccaatcg acatgtagtg cagttagcag aggttatttt agtgctttaa gaacaggttg 5820 gtataccagt gtcataacaa tagaattaag taatataaaa gaaaccaaat gcaatggaac 5880 tgacactaaa gtaaaactta taaaacaaga attagataag tataagaatg cagtgacaga 5940 attacagcta cttatgcaaa acacaccagc tgccaacaac cgggccagaa gagaagcacc 6000 acagtatatg aactatacaa tcaataccac taaaaaccta aatgtatcaa taagcaagaa 6060 gaggaaacga agatttctgg gcttcttgtt aggtgtagga tctgcaatag caagtggtat 6120 agctgtatcc aaagttctac accttgaagg agaagtgaac aagatcaaaa atgctttgtt 6180 atctacaaac aaagctgtag tcagtctatc aaatggggtc agtgttttaa ccagcaaagt 6240 gttagatctc aagaattaca taaataacca attattaccc atagtaaatc aacagagctg 6300 tcgcatctcc aacattgaaa cagttataga attccagcag aagaacagca gattgttgga 6360 aatcaacaga gaattcagtg tcaatgcagg tgtaacaaca cctttaagca cttacatgtt 6420 aacaaacagt gagttactat cattga.tcaa tgatatgcct ataacaaatg atcagaaaaa 6480 attaatgtca agcaatgttc agatagtaag gcaacaaagt tattctatca tgtctataat 6540 aaaggaagaa gtccttgcat atgttgtaca gctacctatc tatggtgtaa tagatacacc 6600 ttgctggaaa ttacacacat cacctctatg caccaccaac atcaaagaag gatcaaatat 6660 ttgtttaaca aggactgata gaggatggta ttgtgataat gcaggatcag tatccttctt 6720 tccacaggct gacacttgta aagtacagtc caatcgagta ttttgtgaca ctatgaacag 6780 tttgacatta ccaagtgaag tcagcctttg taacactgac atattcaatt ccaagtatga 6840 ctgcaaaatt atgacatcaa aaacagacat aagcagctca gtaattactt ctcttggagc 6900 tatagtgtca tgctatggta aaactaaatg cactgcatcc aacaaaaatc gtgggattat 6960 aaagacattt tctaatggtt gtgactatgt gtcaaacaaa ggagtagata ctgtgtcagt 7020 gggcaacact ttatactatg taaacaagct ggaaggcaag aacctttatg taaaagggga 7080 acctataata aattactatg accctctagt gtttccttct gatgagtttg atgcatcaat 7140 atctcaagtc aatgaaaaaa tcaatcaaag tttagctttt attcgtagat ctgatgaatt 7200 actacataat gtaaatactg gcaaatctac tacaaatatt atgataacta caattattat 7260 agtaatcatt gtagtattgt tatcattaat agctattggt ttgctgttgt attgcaaagc 7320 caaaaacaca ccagttacac taagcaaaga ccaactaagt ggaatcaata atattgcatt 7380 cagcaaatag acaaaaaacc acctgatcat gtttcaacaa cagtctgctg atcaccaatc 7440 ccaaatcaac ccataacaaa cacttcaaca tcacagtaca ggctgaatca tttcttcaca 7500 tcatgctacc cacacaacta agctagatcc ttaactcata gttacataaa aacctcaagt 7560 atcacaatca aacactaaat caacacatca ttcacaaaat taacagctgg ggcaaatatg 7620 tcgcgaagaa atccttgtaa atttgagatt agaggtcatt gcttgaatgg tagaagatgt 7680 cactacagtc ataattactt tgaatggcct cctcatgcct tactagtgag gcaaaacttc 7740 atgttaaaca agatactcaa gtcaatggac aaaagcatag acactttgtc tgaaataagt 7800 ggagctgctg aactggacag aacagaagaa tatgctcttg gtatagttgg agtgctagag 7860 agttacatag gatctataaa caacataaca aaacaatcag catgtgttgc tatgagtaaa 7920 cttcttattg agatcaatag tgatgacatt aaaaagctga gagataatga agaacccaat 7980 tcacctaaga taagagtgta caatactgtt atatcataca ttgagagcaa tagaaaaaac 8040 aacaagcaaa caatccatct gctcaaaaga ctaccagcag acgtgctgaa gaagacaata 8100 aaaaacacat tagatatcca caaaagcata atcataagca acccaaaaga gtcaaccgtg 8160 aatgatcaaa atgaccaaac caaaaataat gatattaccg gataaatatc cttgtagtat 8220 atcatccata ttgatttcaa gtgaaagcat gattgctaca ttcaatcata aaaacatatt 8280 acaatttaac cataaccatt tggataacca ccagcgttta ttaaataata tatttgatga 8340 aattcattgg acacctaaaa acttattaga tgccactcaa caatttctcc aacatcttaa 8400 catccctgaa gatatatata caatatatat attagtgtca taatgcttgg ccataacgat 8460 tctatatcat ccaaccataa aactatctta ataaggttat gggacaaaat ggatcccatt 8520 attaatggaa actctgctaa tgtgtatcta actgatagtt atttaaaagg tgttatctct 8580 ttttcagaat gtaatgcttt agggagttac ctttttaacg gcccttatct caaaaatgat 8640 tacaccaact taattagtag acaaagtcca ctactagagc atatgaatct taaaaaacta 8700 actataacac agtcattaat atctagatat cataaaggtg aactgaaatt agaagaacca 8760 acttatttcc agtcattact tatgacatat aaaagcatgt cctcgtctga acaaattgct 8820 acaactaact tacttaaaaa aataatacga agagctatag aaataagtga tgtaaaggtg 8880 tacgccatct tgaataaact aggactaaag gaaaaggaca gagttaagcc caacaataat 8940 tcaggtgatg aaaactcagt acttacaact ataattaaag atgatatact ttcggctgtg 9000 gaaagcaatc aatcatatac aaattcagac aaaaatcact cagtaaatca aaatatcact 9060 atcaaaacaa cactcttgaa aaaattgatg tgttcaatgc aacatcctcc atcatggtta 9120 atacactggt tcaatttata tacaaaatta aataacatat taacacaata tcgatcaaat 9180 gaggtaaaaa gtcatgggtt tatattaata gataatcaaa ctttaagtgg ttttcagttt 9240 attttaaatc aatatggttg tatcgtttat cataaaggac tcaaaaaaat cacaactact 9300 acttacaatc aatttttaac atggaaagac atcagcctta gcagattaaa tgtttgctta 9360 attacttgga taagtaattg tttgaataca ttaaataaaa gcttagggct gagatgtgga 9420 ttcaataatg ttgtgttatc acaattattt ctttatggag attgtatact gaaattattt 9480 cataatgaag gcttctacat aataaaagaa gtagagggat ttattatgtc tttaattcta 9540 aacataacag aagaagatca atttaggaaa cgattttata atagcatgct aaataacatc 9600 acagatgcag ctattaaggc tcaaaagaac ctactatcaa gggtatgtca cactttatta 9660 gacaagacag tgtctgataa tatcataaat ggtaaatgga taatcctatt aagtaaattt 9720 cttaaattga ttaagcttgc aggtgataat aatctcaata atttgagtga gctatatttt 9780 ctcttcagaa tctttggaca tccaatggtt gatgaaagac aagcaatgga tgctgtaaga 9840 attaactgta atgaaactaa gttctactta ttaagtagtc taagtacgtt aagaggtgct 9900 ttcatttata gaatcataaa agggtttgta aatacctaca acagatggcc cactttaagg 9960 aatgctattg tcctacctct aagatggtta. aactattata aacttaatac ttatccatct 10020 ctacttgaaa tcacagaaaa tgatttgatt attttatcag gattgcggtt ctatcgtgaa 10080 tttcatctgc ctaaaaaagt ggatcttgaa atgataataa atgacaaagc catttcacct 10140 ccaaaagatc taatatggac tagttttcct agaaattaca tgccatcaca tatacaaaat 10200 tatatagaac atgaaaagtt gaagttctct gaaagcgaca gatcaagaag agtactagag 10260 tattacttga gagataataa attcaatgaa tgcgatctat acaattgtgt agtcaatcaa 10320 agctatctca acaactctaa tcacgtggta tcactaactg gtaaagaaag agagctcagt 10380 gtaggtagaa tgtttgctat gcaaccaggt atgtttaggc aaatccaaat cttagcagag 10440 aaaatgatag ccgaaaatat tttacaattc ttccctgaga gtttgacaag atatggtgat 10500 ctagagcttc aaaagatatt agaattaaaa gcaggaataa gcaacaagtc aaatcgttat 10560 aatgataact acaacaatta tatcagtaaa tgttctatca ttacagatct tagcaaattc 10620 aatcaagcat ttagatatga aacatcatgt atctgcagtg atgtattaga tgaactgcat 10680 ggagtacaat ctctgttctc ttggttgcat ttaacaatac ctcttgtcac aataatatgt 10740 acatatagac atgcacctcc tttcataaag gatcatgttg ttaatcttaa tgaagttgat 10800 gaacaaagtg gattatacag atatcatatg ggtggtattg agggctggtg tcaaaaactg 10860 tggaccattg aagctatatc attattagat ctaatatctc tcaaagggaa attctctatc 10920 acagctctga taaatggtga taatcagtca attgatataa gtaaaccagt tagacttata 10980 gagggtcaga cccatgctca agcagattat ttgttagcat taaatagcct taaattgcta 11040 tataaagagt atgcaggtat aggccataag cttaagggaa cagagaccta tatatcccga 11100 gatatgcagt tcatgagcaa aacaatccag cacaatggag tgtactatcc agccagtatc 11160 aaaaaagtcc tgagagtagg tccatggata aatacaatac ttgatgattt taaagttagt 11220 ttagaatcta taggtagctt aacacaggag ttagaataca gaggggaaag cttattatgc 11280 agtttaatat ttaggaacat ttggttatac aatcaaattg ctttgcaact ccgaaatcat 11340 gcattatgta acaataagct atatttagat atattgaaag tattaaaaca cttaaaaact 11400 ttttttaatc ttgatagtat cgatatggcg ttatcattgt atatgaattt gcctatgctg 11460 tttggtggtg gtgatcctaa tttgttatat cgaagctttt ataggagaac tccagacttc 11520 cttacagaag ctatagtaca ttcagtgttt gtgttgagct attatactgg tcacgattta 11580 caagataagc tccaggatct tccagatgat agactgaaca aattcttgac atgtgtcatc 11640 acattcgata aaaatcccaa tgccgagttt gtaacattga tgagggatcc acaggcgtta 11700 gggtctgaaa ggcaagctaa aattactagt gagattaata gattagcagt aacagaagtc 11760 ttaagtatag ctccaaacaa aatattttct aaaagtgcac aacattatac taccactgag 11820 attgatctaa atgacattat gcaaaatata gaaccaactt accctcatgg attaagagtt 11880 gtttatgaaa gtctaccttt ttataaagca gaaaaaatag ttaatcttat atcaggaaca 11940 aaatccataa ctaatatact tgaaaaaaca tcagcaatag atacaactga tattaatagg 12000 gctactgata tgatgaggaa aaatataact ttacttataa ggatacttcc actagattgt 12060 aacaaagaca aaagagagtt attaagttta gaaaatctta gtataactga attaagcaag 12120 tatgtaagag aaagatcttg gtcattatcc aatatagtag gagtaacatc gccaagtatt 12180 atgttcacaa tggacattaa atatacaact agcactatag ccagtggtat aattatagaa 12240 aaatataatg ttaatagttt aactcgtggt gaaagaggac ctactaagcc atgggtaggt 12300 tcatctacgc aggagaaaaa aacaatgcca gtgtacaata gacaagtttt aaccaaaaag 12360 caaagagacc aaatagattt attagcaaaa ttagactggg tatatgcatc catagacaac 12420 aaagatgaat tcatggaaga actgagtact ggaacacttg gactgtcata tgaaaaagcc 12480 aaaaagttgt ttccacaata tctaagtgtc aattatttac accgtttaac agtcagtagt 12540 agaccatgtg aattccctgc atcaatacca gcttatagaa caacaaatta tcatttcgat 12600 actagtccta tcaatcatgt attaacagaa aagtatggag atgaagatat cgacattgtg 12660 tttcaaaatt gcataagttt tggtcttagc ctgatgtcgg ttgtggaaca attcacaaac 12720 atatgtccta atagaattat tctcataccg aagctgaatg agatacattt gatgaaacct 12780 cctatattta caggagatgt tgatatcatc aagttgaagc aagtgataca aaaacagcat 12840 atgttcctac cagataaaat aagtttaacc caatatgtag aattattcct aagtaacaaa 12900 gcacttaaat ctggatctaa catcaattct aatttaatat tagtacataa aatgtctgat 12960 tattttcata atgcttatat tttaagtact aatttagctg gacattggat tctaattatt 13020 caacttatga aagattcaaa aggtattttt gaaaaagatt ggggagaggg gtacataact 13080 gatcatatgt tcattaattt gaatgttttc tttaatgctt ataagactta tttgctatgt 13140 tttcataaag gttatggtaa agcaaaatta gaatgtgata tgaacacttc agatcttctt 13200 tgtgttttgg agttaataga cagtagctac tggaaatcta tgtctaaagt tttcctagaa 13260 caaaaagtca taaaatacat agtcaatcaa gacacaagtt tgcatagaat aaaaggctgt 13320 cacagtttta agttgtggtt tttaaaacgc cttaataatg ctaaatttac cgtatgccct 13380 tgggttgtta acatagatta tcacccaaca catatgaaag ctatattatc ttacatagat 13440 ttagttagaa tggggttaat aaatgtagat aaattaacca ttaaaaataa aaacaaattc 13500 aatgatgaat tttacacatc aaatctcttt tacattagtt ataacttttc agacaacact 13560 catttgctaa caaaacaaat aagaattgct aattcagaat tagaagataa ttataacaaa 13620 ctatatcacc caaccccaga aactttagaa aatatatcat taattcctgt taaaagtaat 13680 aatagtaaca aacctaaatt ttgtataagt ggaaataccg aatctataat gatgtcaaca 13740 ttctctaata aaatgcatat taaatcttcc actgttacca caagattcaa ttatagcaaa 13800 caagacttgt acaatttatt tccaaatgtt gtgatagaca ggattataga tcattcaggt 13860 aatacagcaa aatctaacca actttacatc accacttcac atcagacatc tttagtaagg 13920 aatagtgcat cactttattg catgcttcct tggcatcatg tcaatagatt taactttgta 13980 tttagttcca caggatgcaa gatcagtata gagtatattt taaaagatct taagattaag 14040 gaccccagtt gtatagcatt cataggtgaa ggagctggta acttattatt acgtacggta 14100 gtagaacttc atccagacat aagatacatt tacagaagtt taaaagattg caatgatcat 14160 agtttaccta ttgaatttct aagattatac aacgggcata taaacataga ttatggtgag 14220 aatttaacca ttcctgctac agatgcaact aataacattc attggtctta tttacatata 14280 aaatttgcag aacctattag catctttgtc tgcgatgctg aattacctgt tacagccaat 14340 tggagtaaaa ttataattga atggagtaag catgtaagaa agtgcaagta ctgttcttct 14400 gtaaatagat gcattttaat cgcaaaatat catgctcaag atgatattga tttcaaatta 14460 gataacatta ctatattaaa aacttacgtg tgcctaggta gcaagttaaa aggatctgaa 14520 gtttacttag tccttacaat aggccctgca aatatacttc ctgtttttga tgttgtgcaa 14580 aatgctaaat tgattttttc aagaactaaa aatttcatta tgcctaaaaa aactgacaag 14640 gaatctatcg atgcaaatat taaaagctta atacctttcc tttgttaccc tataacaaaa 14700 aaaggaatta agacttcatt gtcaaaattg aagagtgtag ttaatgggga tatattatca 14760 tattctatag ctggacgtaa tgaagtattc agcaacaagc ttataaacca caagcatatg 14820 aatatcctaa aatggctaga tcatgtttta aattttagat cagctgaact taattacaat 14880 catttataca tgatagagtc cacatatcct tacttaagtg aattgttaaa tagtttaaca 14940 accaatgagc tcaagaaact gattaaaata acaggtagtg tactatacaa ccttcccaac 15000 gaacagtaac ttaaaatatc attaacaagt ttggtcaaat ttagatgcta acacatcatt 15060 atattatagt tattaaaaaa tatgcaaact tttcaataat ttagcttact gattccaaaa 15120 ttatcatttt atttttaagg ggttgaataa aagtctaaaa ctaacaatga tacatgtgca 15180 tttacaacac aacgagacat tagtttttga cacttttttt ctcgt 15225
<210> 14 <211> 868 <212> DNA
<213> respiratory syncytial virus B 9320
<400> 14 agtcaacgca ctgccaggac tctagaaaag acctgggata ctcttaatca tctaattgta 60 atatcctctt gtttatacag actaaaccta aaatctatag cacaaatagc actatcagtt 120 ttggcaatga taatctcaac ctctctcata attgcagcca taatattcat catctctgcc 180 aatcacaaag ttacactaac aacggttaca gttcaaacaa taaaaaacca cactgaaaaa 240 aacatcacca cctaccttac tcaagtctca ccagaaaggg ttagctcatc catacaacct 300 acaaccacat caccaatcca cacaaattca gctacaatat caccaaatac aaaatcagaa 360 acacaccata caacaacaca agccaaaagc agaatcacca cttcaacaca gaccaacaag 420 ccaagcacaa aatcacgttc aaaaaatcca ccaaaaaaac caaaagatga ttaccatttt 480 gaagtgttca attttgttcc ctgtagtata tgtggcaaca atcaactttg caaatccatc 540 tgcaaaacaa taccaagcaa caaaccaaag aaaaaaccaa ccatcaaacc cacaaacaaa 600 ccaaccgtca aaaccacaaa caaaagagac ccaaaaacac cagccaaaat gatgaaaaaa 660 gaaaccacca ccaacccaac aaaaaaacca accctcaaga ccacagaagg agacaccagc 720 acctcacaat ccactgtgct cgacacaacc acatcaaaac acacaatcca acagcaatcc 780 ctccactcaa tcacctccga aaacacaccc aactccacac aaatacccac agcaaccgag 840 gcctccacat caaattctac ttaaaaaa 868
<210> 15
<211> 218
<212> DNA
<213> respiratory syncytial virus B 9320
<400> 15 attggcatta agcctacaaa acacactcct ataatataca aatatgacct caacccgtaa 60 attccaacaa aaaactaacc catccaaact aagctattcc ttaaataaca gtgctcaaca 120 gttaagaagg ggctaatcca ttttagtaat taaaaataaa ggtaaagcca ataacataaa 180 ttggggcaaa tacaaagatg gctcttagca aagtcaag 218
<210> 16
<211> 35
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer; Bglllsite, RSV B 9320 G
<400> 16 gatatcaaga tctacaataa cattggggca aatgc 35
<210> 17
<211> 31
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer; Bglllsite, RSV B 9320 G
<400> 17 gctaagagat ctttttgaat aactaagcat g 31
<210> 18
<211> 36
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer; BamHIsite, RSV B 9320
<400> 18 atcaggatcc acaataacat tggggcaaat gcaacc 36
<210> 19
<211> 36
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer; BamHI site, RSV 9320 G
<400> 19 ctggcattcg gatccgtttt atgtaactat gagttg
<210> 20
<211> 27
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 20 gatcccatgg ctcttagcaa agtcaag 27
<210> 21
<211> 31
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer <400> 21 gtacggatcc gttgacttat ttgccccgta t 31
<210> 22
<211> 25
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 22 gatcccatgg agaagtttgc acctg 25
<210> 23
<211> 28
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 23 gtacggatcc tgagtgagtt gatcactg 28
<210> 24
<211> 28
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 24 gcttggccat aacgattcta tatcatcc
<210> 25
<211> 26
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 25 ggtagtataa tgttgtgcac ttttag
<210> 26
<211> 25
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer <400> 26 ggtcacgatt tacaagataa gctcc 25
<210> 27
<211> 30
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 27 cagatccttt taacttgcta cctaggcaca 30
<210> 28
<211> 23
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 28 cttacgtgtg cctaggtagc aag 23
<210> 29
<211> 33
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 29 acgagaaaaa aagtgtcaaa aactaatgtc teg 33
<210> 30
<211> 42
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 30 gtttttgaca ctttttttct cgtggccggc atggtcccag cc 42
<210> 31
<211> 33
<212> DNA
<213> Artificial
<220> <223> oligonucleotide primer
<400> 31 gatctagagc tccaagcttg cggccgcgtc gac 33
<210> 32
<211> 46
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 32 gggtaccccc gggtaatacg actcactata gggacgggaa aaaatg
<210> 33
<211> 24
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 33 gttaacttag agctctacat catc 24
<210> 34
<211> 24
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 34 gtgtggtcct aggcaatgca gcag 24
<210> 35
<211> 29
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 35 gacacagcat gatggtagag ctctatgtg 29
<210> 36
<211> 28
<212> DNA
<213> Artificial <220>
<223> oligonucleotide primer
<400> 36 gctaagtgaa cataaaacat tctgtaac 28
<210> 37
<211> 26
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 37 ccattaataa tgggatccat tttgtc
<210> 38
<211> 29
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 38 cacatagagc tctaccatca tgctgtgtc 29
<210> 39
<211> 27
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 39 cattaatgag ggacccacag gctttag 27
<210> 40
<211> 27
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 40 ctaaagcctg tgggtccctc attaatg 27
<210> 41
<211> 32
<212> DNA
<213> Artificial <220>
<223> oligonucleotide primer
<400> 41 catggttaat acactggttc aatttatata ca 32
<210> 42
<211> 32
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 42 tgtatataaa ttgaaccagt gtattaacca tg 32
<210> 43
<211> 41
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 43 gtcttaaaaa acgaaataaa acgctacaag ggcctcatac c 41
<210> 44
<211> 41
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 44 ggtatgaggc ccttgtagcg ttttatttcg ttttttaaga c 41
<210> 45
<211> 24
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 45 gatgatgtag agctttaagt taac 24
<210> 46 <211> 24 <212> DNA <213> Artificial
<220>
<223> oligonucleotide primer
<400> 46 gttaacttaa agctctacat catc 24
<210> 47
<211> 44
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 47 ctaactggta aagaaagaga gcttagtgta ggtagaatgt ttgc 44
<210> 48
<211> 44
<212> DNA
<213> Artificial
<220>
<223> oligonucle
<400> 48 gcaaacattc tacctacact aagctctctt tctttaccag ttag 44
<210> 49
<211> 40 r<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 49 gtttaacaac caatgagctt aaaaagctga ttaaaattac 40
<210> 50
<211> 40
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 50 gtaattttaa tcagcttttt aagctcattg gttgttaaac 40
<210> 51 <211> 23 <212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 51 cggtctaatg gatgataatt gtg 23
<210> 52
<211> 23
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 52 atgaagctac tgcacaaagt agg 23
<210> 53
<211> 27
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 53 gtaatcatct tttggttttt ttggtgg 27
<210> 54
<211> 33
<212> DNA
<213> Artificial
<220>
<223> oligonucleotide primer
<400> 54 ccaaccatca aacccacaaa caaaccaacc gtc 33

Claims

CLAIMSWHAT IS CLAIMEP IS:
1. An isolated or recombinant nucleic acid comprising a polynucleotide sequence selected from the group consisting of:
(a) SEQ ID NO:l or a complementary polynucleotide sequence thereof;
(b) a polynucleotide sequence that is greater than 97.8% identical to SEQ IP NO:l or a complementary polynucleotide sequence thereof, as determined by BLASTN using default parameters;
(c) a polynucleotide sequence that hybridizes under stringent conditions over substantially the entire length of a polynucleotide subsequence comprising at least 100 contiguous nucleotides of SEQ IP NO:l or a complementary polynucleotide sequence thereof, wherein the polynucleotide sequence hybridizes to the polynucleotide subsequence of SEQ IP NO: 1 or the complementary polynucleotide sequence thereof under said stringent conditions with at least 2x a signal to noise ratio that the polynucleotide sequence hybridizes to a corresponding polynucleotide subsequence of SEQ IP NO: 13 or a complementary polynucleotide sequence thereof;
(d) a polynucleotide sequence comprising at least one unique polynucleotide subsequence comprising at least 10 contiguous nucleotides of SEQ IP NO:l or a complementary polynucleotide sequence thereof, with the proviso that the unique polynucleotide subsequence includes at least one subsequence not included in SEQ IP NOs: 14-19 or a complementary polynucleotide sequence thereof; and,
(e) a polynucleotide sequence encoding an amino acid sequence or unique subsequence selected from the group consisting of SEQ IP NOs:2-ll or an artificial conservative variation thereof.
2. The nucleic acid of claim 1, wherein the nucleic acid is selected from the group consisting of a PNA, an RNA, and an artificial nucleic acid.
3. The nucleic acid of claim 2, wherein the nucleic acid is a cPNA.
4. The nucleic acid of claim 1, wherein the polynucleotide sequence of (b) is at least 98.5% identical to SEQ IP NO: 1 or a complementary polynucleotide sequence thereof, as determined by BLASTN using default parameters.
5. The nucleic acid of claim 1, wherein the polynucleotide sequence of (c) hybridizes to the polynucleotide subsequence of SEQ ID NO:l or the complementary polynucleotide sequence thereof under said stringent conditions with at least 5x the signal to noise ratio that the polynucleotide sequence of (c) hybridizes to the polynucleotide subsequence of SEQ ID NO: 13 or the complementary polynucleotide sequence thereof.
6. The nucleic acid of claim 1, comprising at least one artificially mutated nucleotide.
7. The nucleic acid of claim 6, wherein the at least one artificially mutated nucleotide comprises one or more of: a deleted nucleotide, an inserted nucleotide, or a substituted nucleotide.
8. The nucleic acid of claim 6, comprising a plurality of artificially mutated nucleotides.
9. The nucleic acid of claim 6, wherein the artificially mutated nucleotide is introduced by site-directed mutagenesis.
10. The nucleic acid of claim 6, wherein at least one polypeptide encoded by the nucleic acid comprises at least one deleted, inserted, or substituted amino acid residue.
11. The nucleic acid of claim 10, wherein the polypeptide comprises at least one conservatively substituted amino acid residue.
12. The nucleic acid of claim 6, wherein the at least one artificially mutated nucleotide is located in the open reading frame encoding the polypeptide of SEQ ID NO: 12.
13. The nucleic acid of claim 12, wherein the at least one artificially mutated nucleotide comprises a deletion.
14. The nucleic acid of claim 13, wherein the open reading frame encoding the polypeptide of SEQ ID NO: 12 is deleted.
15. The nucleic acid of claim 13, wherein the nucleotides encoding amino acid residues 164-197 of SEQ ID NO: 12 are deleted.
16. The nucleic acid of claim 6, wherein the at least one artificially mutated nucleotide is located in the open reading frame encoding the polypeptide of SEQ ID NO: 10.
17. The nucleic acid of claim 16, wherein the at least one artificially mutated nucleotide comprises a deletion.
18. The nucleic acid of claim 17, wherein the open reading frame encoding the polypeptide of SEQ ID NO: 10 is deleted.
19. The nucleic acid of claim 16, wherein at least one of the nucleotides encoding amino acid residue 1, amino acid residue 4, amino acid residue 10, or a combination thereof, of SEQ ID NO: 10 is mutated.
20. The nucleic acid of claim 1, wherein the unique polynucleotide subsequence of (d) encodes at least 20 contiguous amino acid residues of any one of SEQ ID NOs:2-12.
21. The nucleic acid of claim 20, wherein the unique polynucleotide subsequence of (d) encodes at least 50 contiguous amino acid residues of any one of SEQ ID NOs:2-12.
22. The nucleic acid of claim 21, wherein the unique polynucleotide subsequence of (d) encodes at least 100 contiguous amino acid residues of any one of SEQ ID NOs:2-12.
23. The nucleic acid of claim 22, wherein the unique polynucleotide subsequence of (d) encodes at least 200 contiguous amino acid residues of any one of SEQ ID NOs:2-12.
24. The nucleic acid of claim 1, wherein the unique polynucleotide subsequence of (d) comprises at least one complete open reading frame.
25. The nucleic acid of claim 24, wherein the complete open reading frame encodes a polypeptide selected from the group consisting of SEQ ID NOs: 2-12.
26. The nucleic acid of claim 24, comprising a plurality of complete open reading frames.
27. The nucleic acid of claim 1, wherein the nucleic acid of (d) further comprises at least one polynucleotide subsequence from a different strain of virus.
28. The nucleic acid of claim 27, wherein the different strain of virus is a different strain of human RSV.
29. The nucleic acid of claim 27, wherein the different strain of virus is a different species of virus.
30. The nucleic acid of claim 27, wherein the nucleic acid comprises at least one complete open reading frame of SEQ ID NO: 1 and at least one complete open reading frame of the different strain of virus.
31. A vector comprising the nucleic acid of claim 1.
32. A host cell into which the vector of claim 31 has been introduced.
33. A method of producing a recombinant respiratory syncytial virus, the method comprising: culturing the host cell of claim 32 in a suitable culture medium under conditions permitting expression of the nucleic acid; and, isolating the recombinant respiratory syncytial virus from the host cell and/or the medium.
34. A recombinant respiratory syncytial virus produced according to the method of claim 33.
35. A recombinant respiratory syncytial virus comprising the nucleic acid of claim 1.
36. The recombinant respiratory syncytial virus of claim 35, wherein the nucleic acid comprises at least one artificially mutated nucleotide.
37. The recombinant respiratory syncytial virus of claim 36, wherein the open reading frame encoding the polypeptide of SEQ ID NO: 12 is deleted.
38. The recombinant respiratory syncytial virus of claim 36, wherein the open reading frame encoding the polypeptide of SEQ ID NO: 10 is deleted.
39. An immunogenic composition comprising an immunologically effective amount of the recombinant respiratory syncytial virus of claim 35.
40. A method for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus, the method comprising administering to the individual an immunologically effective amount of the recombinant respiratory syncytial virus of claim 35 in a physiologically acceptable carrier.
41. A method of producing an isolated or recombinant polypeptide, the method comprising: culturing the host cell of claim 32 in a suitable culture medium under conditions permitting expression of the nucleic acid; and, isolating the polypeptide from the host cell and/or the medium.
42. A polypeptide produced according to the method of claim 41.
43. A polypeptide comprising an amino acid sequence or subsequence that is encoded by the nucleic acid of claim 1, with the proviso that the amino acid sequence or subsequence is not encoded by SEQ ID NO: 14.
44. An immunogenic composition comprising an immunologically effective amount of the polypeptide of claim 43.
45. A method for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus, the method comprising administering to the individual an immunologically effective amount of the polypeptide of claim 43 in a physiologically acceptable carrier.
46. An isolated or recombinant polypeptide comprising an amino acid sequence selected from the group consisting of:
(a) an amino acid sequence selected from the group consisting of SEQ ID NOs:2-ll;
(b) a unique amino acid subsequence comprising at least 7 contiguous amino acid residues of any one of SEQ ID NOs:2-ll;
(c) an amino acid sequence or subsequence corresponding to an artificial conservative variation of an amino acid sequence or subsequence of (a) or (b);
(d) an amino acid sequence that is greater than 99.3% identical to SEQ ID NO:2, an amino acid sequence that is greater than 98.4% identical to SEQ ID NO:3, an amino acid sequence that is greater than 99.7% identical to SEQ ID NO:4, an amino acid sequence that is greater than 98.3% identical to SEQ ID NO: 5, an amino acid sequence that is greater than
99.6% identical to SEQ ID NO:6, an amino acid sequence that is greater than 97.0% identical to SEQ ID NO:7, an amino acid sequence that is greater than 99.3% identical to SEQ ID NO:8, an amino acid sequence that is greater than 99.5% identical to SEQ ID NO:9, an amino acid sequence that is greater than 96.4% identical to SEQ ID NO: 10, or an amino acid sequence that is greater than 99.2% identical to SEQ ID NO: 11, as determined by BLASTP using default parameters; and, (e) an amino acid sequence or subsequence that is specifically bound by an antibody that specifically binds to an amino acid sequence or subsequence encoded by SEQ ID NO:l, wherein said antibody does not specifically bind to an amino acid sequence or subsequence encoded by SEQ ID NO: 13 or SEQ ID NO: 14.
47. The polypeptide of claim 46, wherein the amino acid sequence of (d) is at least 99.5% identical to SEQ ID NO:2, at least 98.6% identical to SEQ ID NO:3, at least 99.9% identical to SEQ ID NO:4, at least 98.5% identical to SEQ ID NO:5, at least 99.8% identical to SEQ ID NO:6, at least 97.2% identical to SEQ ID NO:7, at least 99.5% identical to SEQ ID NO:8, at least 99.7% identical to SEQ ID NO:9, at least 96.6% identical to SEQ ID NO: 10, or at least 99.4% identical to SEQ ID NO: 11, as determined by BLASTP using default parameters.
48. The polypeptide of claim 46, comprising at least one artificially altered amino acid.
49. The polypeptide of claim 48, wherein the at least one artificially altered amino acid comprises one or more of: a deleted amino acid, an inserted amino acid, or a substituted amino acid.
50. The polypeptide of claim 48, comprising a plurality of artificially altered amino acids.
51. The polypeptide of claim 46, wherein the unique amino acid subsequence of (b) or the artificial conservative variation of (c) is immunogenic.
52. An antibody specific for the polypeptide of claim 51.
53. An immunogenic composition comprising an immunologically effective amount of the polypeptide of claim 46.
54. A method for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus, the method comprising administering to the individual an immunologically effective amount of the polypeptide of claim 46 in a physiologically acceptable carrier.
55. A recombinant respiratory syncytial virus comprising the polypeptide of claim 46.
56. The recombinant respiratory syncytial virus of claim 55, wherein the polypeptide comprises at least one artificially altered amino acid.
57. An immunogenic composition comprising an immunologically effective amount of the recombinant respiratory syncytial virus of claim 55.
58. A method for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus, the method comprising administering to the individual an immunologically effective amount of the recombinant respiratory syncytial virus of claim 55 in a physiologically acceptable carrier.
59. A nucleic acid encoding the polypeptide of claim 46.
60. An isolated or recombinant polypeptide comprising the amino acid sequence of SEQ ID NO: 12 with a deletion of residues 164-197, or an artificial conservative variation thereof.
61. An immunogenic composition comprising an immunologically effective amount of the polypeptide of claim 60.
62. A method for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus, the method comprising administering to the individual an immunologically effective amount of the polypeptide of claim 60 in a physiologically acceptable carrier.
63. An isolated or recombinant respiratory syncytial virus comprising the polypeptide of claim 60.
64. A method for stimulating the immune system of an individual to produce a protective immune response against respiratory syncytial virus, the method comprising administering to the individual an immunologically effective amount of the respiratory syncytial virus of claim 63 in a physiologically acceptable carrier.
65. A nucleic acid encoding the polypeptide of claim 60.
PCT/US2004/009438 2003-03-28 2004-03-26 Compositions and methods involving respiratory syncytial virus subgroup b strain 9320 WO2004087062A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP04749465A EP1613345B1 (en) 2003-03-28 2004-03-26 Compositions and methods involving respiratory syncytial virus subgroup b strain 9320
AT04749465T ATE469170T1 (en) 2003-03-28 2004-03-26 COMPOSITIONS AND METHODS USING THE RESPIRATORY SYNCYTIAL VIRUS SUBGROUP B STRAIN 9320
DE602004027362T DE602004027362D1 (en) 2003-03-28 2004-03-26 COMPOSITIONS AND METHODS WITH THE RESPIRATORY SYNCYTIAL VIRUS SUB-GROUP B STAMM 9320
CA2520671A CA2520671C (en) 2003-03-28 2004-03-26 Compositions and methods involving respiratory syncytial virus subgroup b strain 9320
JP2006509391A JP4783726B2 (en) 2003-03-28 2004-03-26 Compositions and methods related to respiratory syncytial virus subgroup B strain 9320
AU2004226369A AU2004226369A1 (en) 2003-03-28 2004-03-26 Compositions and methods involving respiratory syncytial virus subgroup B strain 9320

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US45833103P 2003-03-28 2003-03-28
US60/458,331 2003-03-28
US50832003P 2003-10-03 2003-10-03
US60/508,320 2003-10-03

Publications (2)

Publication Number Publication Date
WO2004087062A2 true WO2004087062A2 (en) 2004-10-14
WO2004087062A3 WO2004087062A3 (en) 2005-03-31

Family

ID=33135096

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/009438 WO2004087062A2 (en) 2003-03-28 2004-03-26 Compositions and methods involving respiratory syncytial virus subgroup b strain 9320

Country Status (8)

Country Link
US (3) US7572904B2 (en)
EP (1) EP1613345B1 (en)
JP (1) JP4783726B2 (en)
AT (1) ATE469170T1 (en)
AU (1) AU2004226369A1 (en)
CA (1) CA2520671C (en)
DE (1) DE602004027362D1 (en)
WO (1) WO2004087062A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2885139A1 (en) * 2005-04-29 2006-11-03 Agronomique Inst Nat Rech PREPARATION OF SOLUBLE COMPLEXES PROTEIN N-PROTEIN P TRUNCATED OR SOLUBLE N-PROTEIN OF A VIRUS OF THE FAMILY OF PARAMYXOVIRIDAE AND THEIR VACCINE USES
FR2899902A1 (en) * 2006-04-18 2007-10-19 Agronomique Inst Nat Rech N-PROTEIN FUSION PROTEINS OF A PARAMYXOVIRIDAE-PROTEIN FAMILY VIRUS OF INTEREST AND USES THEREOF FOR VACCINATION AND INTRA-CELL VECTORIZATION
WO2010053883A1 (en) * 2008-11-05 2010-05-14 Merck Sharp & Dohme Corp. Live, attentuated respiratory syncytial virus

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1572108A4 (en) * 2002-09-27 2008-09-17 Medimmune Vaccines Inc Functional mutations in respiratory syncytial virus
ATE469170T1 (en) * 2003-03-28 2010-06-15 Medimmune Llc COMPOSITIONS AND METHODS USING THE RESPIRATORY SYNCYTIAL VIRUS SUBGROUP B STRAIN 9320
US20070190560A1 (en) * 2006-02-13 2007-08-16 Olga Ornatsky Element-tagged olignucleotide gene expression analysis
US20100040606A1 (en) * 2006-03-06 2010-02-18 Symphogen A/S Recombinant polyclonal antibody for treatment of respiratory syncytial virus infections
CN101679513A (en) * 2007-03-06 2010-03-24 西福根有限公司 Recombinant antibodies for treatment of respiratory syncytial virus infections
WO2009012155A2 (en) * 2007-07-13 2009-01-22 Medimmune, Llc Preparation of negative-stranded rna viruses by electroporation
US8354230B2 (en) * 2007-12-21 2013-01-15 Quest Diagnostics Investments Inc. Multiplex detection assay for influenza and RSV viruses
GB201019240D0 (en) 2010-11-15 2010-12-29 Vib Vzw Respiratory syncytical virus vaccine
US10227569B2 (en) 2011-04-12 2019-03-12 Emory University Respiratory syncytial virus expression vectors
EP2970981B1 (en) 2013-03-14 2020-10-28 Emory University Recombinant rsv with silent mutations, vaccines, and methods related thereto
US11235050B2 (en) 2015-10-29 2022-02-01 Emory University Chimeric RSV, immunogenic compositions, and methods of use

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4800078A (en) * 1987-05-28 1989-01-24 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Immunotherapeutic method of treating respiratory disease by intranasal administration of Igb
US5840520A (en) * 1989-08-28 1998-11-24 Aviron Recombinant negative strand RNA virus expression systems
GB9200117D0 (en) 1992-01-06 1992-02-26 Connaught Lab Production of recombinant chimeric proteins for vaccine use
IL105456A (en) * 1992-04-21 1996-12-05 American Home Prod Attenuated respiratory syncytial virus vaccine compositions
US20030054505A1 (en) * 1997-09-26 2003-03-20 Hong Jin Recombinant rsv expression systems and vaccines
US6713066B1 (en) 1996-07-15 2004-03-30 The United States Of America As Represented By The Department Of Health And Human Services Production of attenuated respiratory syncytial virus vaccines involving modification of M2 ORF2
BR9712138A (en) 1996-09-27 2000-01-18 American Cyanamid Co Isolated RNA virus, vaccine, process to immunize an individual to induce protection against a non-segmented, negative, single-stranded RNA virus of the mononegaviral order and to produce RNA virus, isolated nucleic acid molecule and composition.
US6017694A (en) 1997-12-19 2000-01-25 American Cyanamid Company Methods of screening for modulators of respiratory syncytial virus matrix protein interaction
EP1064358A2 (en) * 1998-03-26 2001-01-03 American Cyanamid Company Mutations responsible for attenuation in measles virus or human respiratory syncytial virus subgroup b
JP4268407B2 (en) 2000-11-28 2009-05-27 アヴィロン,インコーポレーテッド Recombinant RSV virus expression system and vaccine
US8927206B2 (en) * 2001-01-19 2015-01-06 Vironovative B.V. Virus causing respiratory tract illness in susceptible mammals
EP1572108A4 (en) 2002-09-27 2008-09-17 Medimmune Vaccines Inc Functional mutations in respiratory syncytial virus
ATE469170T1 (en) * 2003-03-28 2010-06-15 Medimmune Llc COMPOSITIONS AND METHODS USING THE RESPIRATORY SYNCYTIAL VIRUS SUBGROUP B STRAIN 9320

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1613345A4 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2885139A1 (en) * 2005-04-29 2006-11-03 Agronomique Inst Nat Rech PREPARATION OF SOLUBLE COMPLEXES PROTEIN N-PROTEIN P TRUNCATED OR SOLUBLE N-PROTEIN OF A VIRUS OF THE FAMILY OF PARAMYXOVIRIDAE AND THEIR VACCINE USES
WO2006117456A1 (en) * 2005-04-29 2006-11-09 Institut National De La Recherche Agronomique (Inra) Preparation of soluble n-protein/truncated p-protein complexes or n-proteins soluble in a virus of the paramyxoviridae family and use thereof in vaccines
US8071334B2 (en) 2005-04-29 2011-12-06 Institut National De La Recherche Agronomique Preparation of soluble N-protein/truncated P-protein complexes of the paramyxoviridae family
FR2899902A1 (en) * 2006-04-18 2007-10-19 Agronomique Inst Nat Rech N-PROTEIN FUSION PROTEINS OF A PARAMYXOVIRIDAE-PROTEIN FAMILY VIRUS OF INTEREST AND USES THEREOF FOR VACCINATION AND INTRA-CELL VECTORIZATION
WO2007119011A2 (en) * 2006-04-18 2007-10-25 Institut National De La Recherche Agronomique (Inra) N protein fusion proteins of a virus in the paramyxoviridae-protein of interest family
WO2007119011A3 (en) * 2006-04-18 2008-03-06 Agronomique Inst Nat Rech N protein fusion proteins of a virus in the paramyxoviridae-protein of interest family
WO2010053883A1 (en) * 2008-11-05 2010-05-14 Merck Sharp & Dohme Corp. Live, attentuated respiratory syncytial virus
US20110212130A1 (en) * 2008-11-05 2011-09-01 Yagodich Mary K Live, attenuated respiratory syncytial virus
AU2009311287B2 (en) * 2008-11-05 2013-02-07 Merck Sharp & Dohme Corp. Live, attentuated respiratory syncytial virus
AU2009311287C1 (en) * 2008-11-05 2015-02-19 Merck Sharp & Dohme Corp. Live, attentuated respiratory syncytial virus
US9011876B2 (en) 2008-11-05 2015-04-21 Merck Sharp & Dohme Corp. Live, attenuated respiratory syncytial virus

Also Published As

Publication number Publication date
CA2520671A1 (en) 2004-10-14
ATE469170T1 (en) 2010-06-15
DE602004027362D1 (en) 2010-07-08
US20090285853A1 (en) 2009-11-19
US20120263744A1 (en) 2012-10-18
CA2520671C (en) 2014-03-18
JP4783726B2 (en) 2011-09-28
EP1613345A2 (en) 2006-01-11
AU2004226369A1 (en) 2004-10-14
US20040224309A1 (en) 2004-11-11
US7572904B2 (en) 2009-08-11
US8163530B2 (en) 2012-04-24
JP2006524053A (en) 2006-10-26
EP1613345A4 (en) 2007-01-10
WO2004087062A3 (en) 2005-03-31
EP1613345B1 (en) 2010-05-26

Similar Documents

Publication Publication Date Title
US8163530B2 (en) Nucleic acids encoding respiratory syncytial virus subgroup B strain 9320
US5993824A (en) Production of attenuated respiratory syncytial virus vaccines from cloned nucleotide sequences
JP5743794B2 (en) Live attenuated respiratory syncytial virus
US6689367B1 (en) Production of attenuated chimeric respiratory syncytial virus vaccines from cloned nucleotide sequences
CA2302867A1 (en) Attenuated respiratory syncytial viruses
KR100746979B1 (en) Production of attenuated chimeric respiratory syncytial virus vaccines from cloned nucleotide sequences
JP2011250798A5 (en)
JP2012507302A5 (en)
CA2380108C (en) Recombinant rsv virus expression systems and vaccines
CA2430115C (en) Recombinant rsv virus expression systems and vaccines
EP2396033B1 (en) Influenza hemagglutinin and neuraminidase variants
US20120308602A1 (en) Recombinant RSV Virus Expression Systems And Vaccines
CN109310752B (en) Recombinant respiratory syncytial virus strains with mutations in the M2-2ORF that provide a range of attenuated phenotypes
CN106119287B (en) A kind of recombinant vector and method for expressing respiratory syncystial virus F protein
KR20230021115A (en) Live attenuated respiratory syncytial virus (LIVE ATTENUATED RESPIRATORY SYNCYTIAL VIRUS)
AU5592201A (en) Production of attenuated respiratory syncytial virus vaccines from cloned nucleotide sequences
AU5591601A (en) Production of attenuated respiratory syncytial virus vaccines from cloned nucleotide sequences

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

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

Ref document number: 2004226369

Country of ref document: AU

Ref document number: 2006509391

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2520671

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2004226369

Country of ref document: AU

Date of ref document: 20040326

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004226369

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2004749465

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004749465

Country of ref document: EP