EP1503792A2 - Attenuation du metapneumovirus - Google Patents

Attenuation du metapneumovirus

Info

Publication number
EP1503792A2
EP1503792A2 EP03730063A EP03730063A EP1503792A2 EP 1503792 A2 EP1503792 A2 EP 1503792A2 EP 03730063 A EP03730063 A EP 03730063A EP 03730063 A EP03730063 A EP 03730063A EP 1503792 A2 EP1503792 A2 EP 1503792A2
Authority
EP
European Patent Office
Prior art keywords
virus
region
vaccine
modification
rsv
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
EP03730063A
Other languages
German (de)
English (en)
Inventor
Clive J. Naylor
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lohmann Animal Health GmbH and Co KG
Original Assignee
Lohmann Animal Health GmbH and Co KG
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 Lohmann Animal Health GmbH and Co KG filed Critical Lohmann Animal Health GmbH and Co KG
Publication of EP1503792A2 publication Critical patent/EP1503792A2/fr
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/155Paramyxoviridae, e.g. parainfluenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1013Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1019Tetrapeptides with the first amino acid being basic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • 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
    • C12N2760/18311Metapneumovirus, e.g. avian pneumovirus
    • C12N2760/18322New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18311Metapneumovirus, e.g. avian pneumovirus
    • C12N2760/18334Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18311Metapneumovirus, e.g. avian pneumovirus
    • C12N2760/18361Methods of inactivation or attenuation
    • C12N2760/18362Methods of inactivation or attenuation by genetic engineering
    • 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
    • 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
    • C12N2760/18534Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18511Pneumovirus, e.g. human respiratory syncytial virus
    • C12N2760/18561Methods of inactivation or attenuation
    • C12N2760/18562Methods of inactivation or attenuation by genetic engineering

Definitions

  • the present invention relates to a vaccine against members of the genus metapneumovirus or RSV, the vaccine being an attenuated live vaccine.
  • the vaccine is directed against metapneumovirus of avian or human origin.
  • the present invention also relates to a method for the production of a vaccine which is directed against members of the genus Metapneumovirus or RSV. Furthermore, the present invention relates to an attenuated living virus belonging to the genus metapneumovirus. It also relates to a host cell comprising such a virus. The present invention is also directed to a specific DNA or cDNA or RNA sequence as well as a vector or a plasmid comprising the DNA, cDNA or RNA sequence.
  • the present invention relates to an F protein which is modified and the use of the F protein to produce a vaccine for preventing a disorder or disease caused by viruses belonging to the genus metapneumovirus or RSV.
  • the Paramyxovirinae and Pneumovirinae subfamilies of the Paramyxoviridae family contain several important pathogens for humans and animals.
  • the Pneumovirinae are taxonomically divided into the Pneumovirus and the Metapneumovirus genus.
  • the human respiratory syncytic virus the type of pneumovirus genus, which is also described below, is the only and most important trigger of diseases of the lower respiratory tract in infancy and early childhood worldwide.
  • Other members of the pneumovirus genus include the bovine respiratory syncytic virus, the ovine respiratory syncytic virus, and the pneumonia virus in mice.
  • the avian pneumovirus APV is also further described below, and was previously known as Turkey Rhinotracheitis Virus (TRTV). It is considered an etiological agent of upper RTI and was previously considered the only member of the recently created genus metapneumovirus.
  • TRTV Turkey Rhinotracheitis Virus
  • the Paramyxoviridae family comprises a large number of different viruses which are involved in different, more or less serious disorders or diseases in both humans and animals.
  • the Paramyxoviridae contain a single molecule of single-stranded negative RNA as a genome and are all associated with acute respiratory disease of the host (Pringe, CR (1987) in Molecular Basis of Virus Disease, ed. WC Russell and JW Almond, Society for General Microbiology, Vol 40, pp. 91-138, Cambridge University Press).
  • the respiratory syncytic virus RS virus
  • the human RS virus is the main cause of severe diseases of the lower respiratory tract in children, which leads to annual epidemics worldwide (Chanock et al., 1991, in “Viral Infections of Humans", pages 525-544, ed. AS Evans, plenum Press). Although there is limited nucleotide or amino acid sequence homology between the pneumoviruses to which the RS virus belongs and other Paramyxoviridae, there are important structural features such as the main nucleoprotein (N) and the F protein (F) conserved in proteins with similar functions in a wide range of viruses (Barr et al., 1991, J. of Gen. Virol. 72, 677-685; Chambers et al., 1990, J. of Gen. Virol.
  • RNA synthesis methods are carried out by the helionucleocapsid complex, which is formed by an association of the genomic RNA, the nucleoprotein, a phosphoprotein (P) and the polymerase (L) protein.
  • the function of the nucleoprotein complex depends on the presence of all three proteins that interact with each other as well as with the genomic RNA and possibly with other virus and / or cell components (Barik, 1992, J. of Virol. 66, 6813-6818).
  • the maturation of the virion depends on a number of interactions between the ribonucleoprotein complex, the matrix (M) protein and the virus glycoproteins that are embedded in the modified cell membrane.
  • NS1 and NS2 are also involved in the virion structure and presumably in the assembly, but in a way that is still poorly understood.
  • the function of the pneumovirus proteins referred to as NS1 and NS2 is not known and does not appear to form part of the virus particle (Collins, 1991, in The Paramyxoviruses, pages 103-162, ed. DW Kingsbury, Plenum Press).
  • the APV virus is a member of the metapneumovirus genus. It triggers turkey rhinotracheitis, which is an acute respiratory disease in turkeys. It was first described in South Africa in the late 1970s and later in Europe and other parts of the world. The disease has been one of the main causes of economic losses in the turkey industry in recent years. In Europe, APV serotypes A and B are generally found, while similar infections have been found in Colorado in the United States. The APV strain isolated from it is the so-called Colorado or C strain. Another strain of APV was isolated from ducks in France.
  • APV avian metapneumovirus
  • F fusion protein
  • G glycoproteins
  • the original APV was classified into two subspecies. Originally v / ar APV from South Africa and Great Britain of subspecies A and the other European APV strains belonged to subtype B. As stated above, subtype C is now also known, in addition to presumably other subtypes that are non-A non- B are.
  • the avian metapneumovirus can be cultivated in tracheal ring cultures, where it exerts a ciliostatic effect. Furthermore, it can be found in embryonated eggs and others. Cell cultures are cultivated. The American strains were also cultivated in HEF (hen embryo fibroblasts), Vero cells and embryonated eggs.
  • turkey rhinotracheitis correspond to those of an acute rhinotracheitis infection. Two to three days after infection, the animals were apathetic and showed coughing, sneezing, and shaking their heads. If the infection is not complicated, the animals are normal seven to eight days after infection.
  • the clinical symptoms in hens correspond to those in turkeys, but are usually less severe. If E. coli or other bacteria are also involved, high losses can occur and the actual period of the disease can be lengthened unpleasantly. However, APV rarely leads to the "swollen head syndrome" with massive losses in chickens. It is probably more important for the continuous losses due to lower clinical illnesses. After infection with conventional APV, hens and turkeys produce neutralizing antibodies in the serum ELISA are detectable. These antibodies do not appear to have any real relevance for the control of rhinotracheitis.
  • Maternal antibodies are not effective for chicks against early infection with viral APV. Circulating antibodies, however, appear to be important for protecting the gonads in the event of infection in older animals.
  • Virus-specific IgA and IgG antibodies with a virus-neutralizing effect appear in the tear fluid after infections with virulent APV. To date, no therapy is known for the treatment of APV infections. With the exception of the USA, attempts are being made to control APV by vaccination.
  • Paramyxoviridae Another important member of Paramyxoviridae is the newly discovered human metapneumovirus, which has been isolated from young children with respiratory disorders. This virus was isolated from 28 young children in the Netherlands and was identified as a new member of the genus metapneumovirus based on virological data, sequence homology and gene constellation (Van Den Hoogen et al., Nature, Medicine, Vol. 7, No. 6, June 2001, pages 719-724). Until the discovery of this new metapneumovirus, APV was regarded as the only member of the recently named genus metapneumovirus as described above (Virus Taxonomy, Seventh Report of the International Committee on Taxonomy of Viruses (ed.
  • Live vaccines are usually used as a spray or eye drops for chicks around day 1. Some manufacturers recommend repeating immunization with live vaccines one or more times. The live vaccines that are currently on the market are produced from different strains of different species and are more or less attenuated. Inactivated vaccines are sometimes also used for older animals, although even in this case they are usually first vaccinated with a live vaccine.
  • the live vaccines are subtypes A or B. Results from different experimental studies show that there is good cross-reactivity between the different strains, but homologous protection appears to be more complete.
  • the human metapneumovirus there is no vaccine at all, as it was only recently discovered. However, particularly with regard to the human metapneumovirus, it would be particularly desirable to obtain a vaccine that provides good and effective protection against the virus that leads to serious illnesses in children.
  • a vaccine is provided against members of the genus metapneumovirus and RSV as well as against viruses which have a significant genetic homology in the area of the F protein to members of the genus metapneumovirus, characterized in that it comprises a virus or part of a virus, the virus being modified in the region aa 293-296 of the amino acid sequence of the F protein (fusion protein) compared to the wild type virus, or in a region which shows the same function as aa 293-296.
  • part of a virus it is meant that the vaccine should comprise at least that part of the virus which comprises the modification in region aa 293-296 of the F protein or a functionally similar region and all other components of the virus which are necessary so that the virus can act as a vaccine, ie it produces immunogenicity but is not virulent.
  • the generation of such a virus is within the general specialist knowledge of a person skilled in the art. The invention is further described according to the figures included here, the figures representing the following:
  • Fig. 1 Exemplary sequence changes during attenuation and reversion
  • Fig. 4 changes at positions aa 293-296 of the fusion protein
  • the usual cleavage site of the fusion protein F e.g. common to A-type APVs and other APVs, as well as other members of Paramyxoviridae, has a motif with basic amino acids located approximately at aa 99-109, and the F protein, when cleaved by proteases, separates into Fl and F2, exposing the fusion-related domain. Therefore, the provision of an increased number of basic amino acids in region aa 293-296, which could be a second cleavage site or a functionally corresponding site, could lead to a similar cleavage site for proteases.
  • FIG. 1 shows a possibility of exemplary sequence changes during attenuation and reversion of viruses to virus vaccines and back to virulent viruses.
  • the abbreviations mean the following:
  • N nucleocapsid
  • Gen approximately 1,200 bases
  • M2 2nd matrix
  • Gen about 600 bases
  • G glycoprotein
  • Gen approximately 1,100 bases
  • M2 is believed to be a transcription elongation factor that is believed to be essential for virus rescue. It contains a second reading frame that does not appear to be expressed.
  • the G protein could be responsible for attachment to a target cell receptor. It is highly glycosylated and highly variable between different strains. L-polymerase affects the transcription and replication of the genome. N, P and L combine to provide the minimal replication unit called the nucleocapsid.
  • the fusion protein is responsible for the fusion of the virus with the target cell. It is translated as F0 and is cleaved by cellular proteases at aa positions 99-102 (RRRR) to form Fl (containing the fusion-related domain, membrane-bound) and F2 (which remains attached to Fl by disulphide bridges between cysteine cleavage residues).
  • RRRR cellular proteases at aa positions 99-102
  • F2 which remains attached to Fl by disulphide bridges between cysteine cleavage residues.
  • the potential additional cleavage site aa 293-296 in APV and hMPV and aa 323-328 in RSV
  • the virus arrangement is the wild-type virus arrangement.
  • the virus arrangement which is shown in white, is the virus arrangement of an attenuated virus, while on the right side the virus arrangement is the arrangement of a virus that has returned to virulence.
  • Sequence changes that can occur during attenuation and reversion and that are shown in FIG. 1 are only exemplary.
  • a modification in region aa 293-296 or a region with the same functionality leads to the generation of an attenuated virus which can be used as a vaccine against members of the genus metapneumovirus or RSV.
  • the present inventors have found that a modification in this region will produce an attenuated virus that has lost its virulent ability while still providing complete immunogenicity.
  • the modification in this region will also reduce the risk of returning to virulence.
  • Table 1 is provided, which provides an overview of the 20 amino acids, their single letter code (SLC) and their corresponding DNA codons.
  • Serine S 0 glycos TCT, TCC, TCA, TCG, AGT, AGC
  • the region aa 293-296 is located in the F protein, namely the fusion protein, from viruses of the genus Metapneumovirus and with RSV.
  • the location of the aa 293-296 region can be derived, for example, from SEQ ID NOS 28 to 34.
  • the present inventors were able to show that when the codons encoding the amino acids in the aa 293-296 region or a region with the same functionality as aa 323-328 were modified in RSV of the F gene, attenuated Virus strains could be generated that retained their immunogenicity, but lost their virulence.
  • APV type A RKEK (SEQ ID No 24), RKKE, REEK APV type B RHER (SEQ ID No 25)
  • APV type C SGKD (SEQ ID No 26) human metapneumovirus: SGKK (SEQ ID No 27)
  • TTNTKE SEQ ID No 77
  • the human metapneumovirus is meant as defined according to the article by Van den Hoogen et al. , Nature, Medicine, Vol. 7, No. 6, June 2001, pages 719-724. It appears likely that the hydrophilic region is needed to add function and structure to the functional protein. Therefore, the presence of basic amino acids should favor cleavage by serine proteases; Accordingly, a suitable modification of the four amino acids in the region aa 293-296 in APV or hMPV as well as a suitable modification of aa 323-328 in RSV should have a universal attenuating effect.
  • the virus described above is an attenuated live virus.
  • Live attenuated viruses are known in the art and their preparation can be easily accomplished by one of ordinary skill in the art.
  • the live attenuated virus needs a modification in region aa 293-296 of the amino acid sequence of the fusion protein compared to the wild type or in a region with the same functionality, e.g. aa 323-328 in RSV.
  • the attenuation of the virus occurs through the modification in the region (s) above.
  • known attenuation methods that are already known for these viruses can be used.
  • the modification comprises stabilizing region aa 293-296 or a region with the same functionality as aa 323-328 in RSV in the virus.
  • stabilization is intended to describe a situation in which the amino acids at position aa 293-296 or a region with the same functionality as aa 323-328 in RSV are coded by codons which cannot easily return to the wild type. Such stabilization is preferably achieved by substitution of codons which code for amino acids in the region Codons that need more mutations to return to the wild type. Such stabilization is exemplified as follows:
  • the goal is to replace the amino acid glutamic acid (E) with a basic amino acid.
  • Glutamic acid is encoded by the DNA codon GAA or GAG.
  • the amino acid lysine (K) is chosen to replace glutamic acid, the following situation arises: Lysine is encoded by the DNA codon AAA or AAG. If the DNA codon AAA is chosen for lysine, a mutation is required to become the DNA codon GAA, i.e. Glutamic acid to return. The same applies if the DNA codon AAG is selected which needs a mutation to return to the DNA codon GAG, which in turn codes for glutamic acid.
  • the stabilization is achieved by substitution of codons which code for the amino acid in the region, preferably the acidic amino acids in this region, with codons which are less likely to mutate into a codon which codes for glutamic acid.
  • the present inventors have found that the presence of glutamic acid in region aa 293-296 or a region with the same functionality, such as aa 323-328 in RSV of the F protein of viruses, reduces or prevents attenuation and increases virulence. There is no reference in any prior art document relating to this discovery.
  • Glutamic acid like other acidic amino acids, appears to contribute to the virulence of the virus when located in region aa 293-296 or a region with the same functionality as aa 323-328 in RSV, the F protein.
  • this enhancement of virulence with simultaneous weakening or elimination of attenuation could be attributed to a situation in which hydrophilic regions are needed to impart a functional structure to the F protein, while the presence of basic amino acids would favor serine protease cleavage.
  • the modification in region aa 293-296 or a region with the same functionality as aa 323-328 in RSV, the F protein of viruses, preferably the genus metapneumovirus comprises the substitution of at least one non-basic amino acid a basic amino acid. Even more preferably, at least two non-basic amino acids are replaced by basic amino acids. Even more preferably, at least three non-basic amino acids are replaced by basic amino acids. According to a particularly preferred embodiment, the amino acids in region aa 293-296 of the F protein of hMPV or APV are modified such that all four amino acids are basic amino acids.
  • all six amino acids of region aa 323-328 are basic amino acids in RSV.
  • the modification can comprise the addition of at least one amino acid.
  • This addition of at least one amino acid can be carried out in addition to the substitution mentioned above.
  • the addition of at least one relates Amino acid the addition of at least one basic amino acid, preferably arginine, lysine and / or histidine, particularly preferably arginine or lysine.
  • the modification can also include the deletion of at least one amino acid.
  • This at least one deleted amino acid is preferably an acidic amino acid.
  • the modification comprises the substitution of at least one glutamic acid residue by at least one basic amino acid.
  • the basic amino acid is preferably selected from the following group consisting of arginine, lysine and / or histidine. The use of arginine and / or lysine is particularly preferred.
  • region aa 293-296 of the wild-type virus has a sequence as shown in one of SEQ ID NO 24 to 27.
  • SEQ ID NO 24 represents, for example, the amino acids in region aa 293-296 of the wild-type APV-A-Starnms No. 8544, namely
  • SEQ ID NO 25 represents the typical amino acids in region aa 293-296 of APV-B viruses, namely
  • SEQ ID NO 26 represents, for example, the typical amino acids in positions aa 293-296 of viruses of the APV-C type, namely
  • SEQ ID NO 27 represents the amino acids in positions aa 293-296 of human metapneumovirus, namely
  • wild type virus refers to those viruses that do not have mutations in positions 293-296 or a region with the same functionality (e.g. aa 323-328 in RSV) of the F protein.
  • a wild-type virus is a virulent virus.
  • Preferred viruses which can be wild-type viruses in the sense of the present invention, are human metapneumovirus, APV virus of types A, B, C or non-A, non-B or RS virus.
  • the aa 293-296 region of the attenuated virus preferably has a sequence as shown in one of SEQ ID NOs 1 to 23.
  • SEQ ID NO 1 to 23 can also be selected to form part of region aa 323-328 in the RS virus.
  • SEQ ID NO 1 is an exemplary amino acid sequence at position aa 293-296, which, for example, makes an attenuated strain usable as a vaccine for e.g. Strain No. 8544 (APV type A) can be provided.
  • RRRR could also provide, for example, an attenuated human metapneumovirus or an attenuated APV virus of the B or C type or non-A-non-B type.
  • the F protein as disclosed according to the present invention By modifying the amino acids or the codons coding for the amino acids at positions aa 293-296 or in a region with the same functionality, e.g. aa 323-328 in RSV, the F protein as disclosed according to the present invention, it is possible to provide a live attenuated virus which is useful as a vaccine.
  • region aa 293-296 of the wild-type virus has the amino acid sequence as shown in SEQ ID NO 24, the region of the attenuated virus having a sequence as shown in SEQ ID NO 1.
  • Such an embodiment would be an attenuated life virus for APV type A, e.g. Provide strain no. 8544.
  • region aa 293-296 of the wild-type virus has the sequence shown in SEQ ID NO 27, the region of the attenuated virus having the sequence shown in SEQ ID NO 1, 2, 10 or 21.
  • Such a modification in region aa 293-296 would provide, for example, a live attenuated virus for the human metapneumovirus.
  • the preferred sequences, shown in SEQ ID NO 1 to 23, may also include one or more histidines to replace either lysine or arginine. The above is mutatis mutandis also applicable to region aa 323-328 of RSV.
  • the present modification becomes in region aa 293-296 or in a region with the same functionality as aa 323-328 in RSV of the F protein a supply of attenuated live viruses, for example of the genus Metapneumovirus or RSV, which have the common feature of an F protein with a substantial genetic homology between the sequences of the respective F proteins in these viruses.
  • the vaccine provided according to the present invention is effective against the human metapneumovirus.
  • the metapneumovirus avian metapneumovirus, in particular APV, is further preferred.
  • the present invention is not limited to metapneumovirus, but is applicable to all viruses which have a clear genetic homology, e.g. more than 50%, preferably more than 65%, particularly preferably more than 75% sequence homology, determined based on
  • the attenuated live virus is preferably formulated with a suitable excipient and / or carrier and / or adjuvant.
  • the virus is a live attenuated virus formulated with an appropriate amount of interleukin-6 (IL-6).
  • IL-6 interleukin-6
  • Interleukin-6 formulation is particularly preferred when the virus is an avian metapneumovirus.
  • the virus is an attenuated virus and is formulated with a suitable amount of interleukin-12 (IL-12) and / or interleukin-18 ( ⁇ L-18).
  • the attenuated virus is preferably formulated with interleukin-12 and / or interleukin-18 if the virus is a human metapneumovirus or RSV.
  • an appropriate amount in the context of the present invention means an amount that when is formulated with the attenuated virus according to the present invention, provides the desired effect, but does not adversely affect the usefulness of the live attenuated virus as a vaccine.
  • Suitable amounts can be determined by those skilled in the art and will depend on the attenuated virus used and the subject to be treated, e.g. in terms of age, weight, physical condition and the disease to be treated.
  • a method for the production of a vaccine according to the present invention directed e.g. against members of the genus metapneumovirus includes the following steps:
  • the modification preferably relates to a modification as defined above, in particular in the appended claims 2 to 16.
  • the virus is more preferably a virus selected from the group as defined above, in particular according to the appended claims 17 to 19.
  • step b) affects region aa 293-296 if APV or hMPV is affected and affects region aa 323-328 if RSV is affected.
  • provision of a modification is preferably obtained as follows:
  • Changes are made in the fusion protein sequence, for example by PCR amplification, using primers that have been changed compared to the original sequence.
  • the sequence of primers 3.82 Sst neg and 3.82 Sst pos follow the embodiment sequence, except for the substitution of agg aaa aag aaa by agg aga cgc cgc.
  • Two PCRs, one from the leader side up to 3.82 Sst neg and the other from 3.82 Sst pos to a position downstream (trailer direction) are performed. Specifically, the PCR from LTZ 3.
  • lXho + to 3.82Sst- (designated as 3a) will have the changed sequence (change from aa, coding for RRRR) and will also contain a Sstll RE site (together with the neighboring gg, directly downstream) / up from this sequence, [Sstll recognition site ccgcgg]).
  • the PCR between the primers 3.82 Sstll pos and LTZ 4.6 Sal- (referred to as 3b) produces the same changes in the adjacent fragment, therefore when the two PCR products are cut with SstII and ligated together, the product becomes the have the original LTZ 3. lXho + to LTZ 4.6 Sal sequence, except for the change indicated above.
  • 3a is cloned first (bluntly), then - after checking the sequence - PCR 3b is added (after both the plasmid and 3b have been cut with Sstll and Sall).
  • the method can also be used for much longer PCRs using the same primers (3.82 Sst neg and 3.82 Sst pos) and their ligation will result in DNA that Sstll can be cut and then ligated together to go directly for virus rescue to be used, thereby avoiding any stages of cloning. In this way, it should be possible to generate rapidly attenuated viruses from either field isolates or RNA extracts.
  • FIGS. 2 and 3 A preferred cloning strategy is given in FIGS. 2 and 3.
  • the genome fragments were originally cloned in TWF 18 (LTZ T7 1 to LTZ 9 + 10 HDVR).
  • Each cloned area (starting with LTZ T7 1 and ending with LTZ 9 + 10 HDVR) was digested with Xhol and Sall, then each was sequentially cloned into CTPE.
  • ligation of the cut Sall (plasmid) and Xhol (leader end of the LTZ fragment) sites at the leader of the fragment resulted in the intragome sequence that is resistant to both enzymes, while a combination of two Sall cut ends at the trailer end ensured that Sall could still be there to be able to accept the next fragment. Therefore, after the addition of each genome area and subsequent cloning, the plasmid was digested again with Insert Sall and the next area (cut out of TWF with Xhol and Sall) was cloned into it.
  • a vaccine comprising a mixture of two or more live attenuated viruses, whereby one or more of the live attenuated viruses according to the present invention are obtained, i.e. a modification in the amino acid sequence at positions aa 293-296 or in a region with the same functionality as aa 323-328 in RSV of the F protein.
  • the present invention also relates to an attenuated live virus belonging to the genus metapneumovirus or RSV or to a virus which has a significant genetic homology in the F protein with viruses of the genus metapneumovirus, which is characterized in that it is a modification in region aa 293 -296 or a region with the same functionality, such as aa 323-328 in RSV, the F protein. Both the modification and the virus are as defined above.
  • the present invention also relates to a host cell comprising a virus that is attenuated by modification as described above.
  • the present invention also relates to a DNA or cDNA sequence as defined in one of SEQ ID NO 28 to 34. All these sequences 28 to 34 are total length sequences of the F protein of human metapneumovirus, comprising one suitable modification in region aa 293-296, which provide an attenuated live human metapneumovirus.
  • the present invention also relates to RNA sequences corresponding to the DNA sequences v / ie defined in SEQ ID NO 28 to 34.
  • the present invention also relates to a vector as well as a plasmid comprising the DNA, cDNA or RNA sequence as defined above.
  • the present invention further relates to a live attenuated virus obtainable by the method as described above.
  • the present invention also relates to an F protein of a member of the genus metapneumovirus or RSV or a virus which shares a significant genetic homology in the F protein range with a member of the genus metapneumovirus, characterized in that it comprises a modification or modifications as defined above , ie a modification (modifications) of the amino acids at positions 293-296 or in a region with the same functionality, e.g. aa 323-328 at RSV.
  • the modifications (modifications) that may be contained in the F protein according to the present invention are defined above.
  • the present invention also relates to the use of the F protein as defined above or an attenuated live virus as defined above for the production of a vaccine for the prevention of a disorder or disease caused by a virus as defined above.
  • the F protein or live attenuated virus of the present invention can be used to prepare a vaccine for preventing a disorder or disease caused by any of the following viruses: APV type A, B, C or non-A, non-B human metapneumovirus RS virus.
  • the leader area PCR used the APV-Lead primed RT reaction as a template and the PCR primers were APV-Lead ext (5 'ACGAGAAAAAAACGCATTCAAGCAGGTTCT3') (SEQ ID NO 37) and LTZ 8.2 sal neg (5 'GGGTATCTATGATGGTCGACAGATGTG3') (SE ID NO 38).
  • the trailer area used M2Start + primed RT reaction as a template and PCR primers were LT7
  • PCR primers included the sequence modifications that introduced restriction endonuclease recognition sites or other changes to DNA extremities and 3 'extremities, while, with the exception of LTZ 3, the encoded protein sequence remained unchanged.
  • the T7 promoter was added to the viral leader sequence in LTZ 1T7, a shortened form of the human RNA polymerase 1 promoter (pol 1) was added to the viral leader sequence in LTZ 1 pol, the region which encoded aa 293-296 of the fusion protein was changed so that RKKK became RRRR in LTZ 3 and in LTZ 10 HDVR the hepatitis delta virus ribozyme was added to the viral trailer area (LTZ 9 + 10).
  • the changes in the F protein gene sequence increased the number of mutations needed to allow the sequence to mutate into a sequence encoding acidic amino acids, as detailed in Figure 4.
  • each blunt-ended PCR product was ligated into the purpose-built plasmid pTWF 18, which was a pUC 18-derived plasmid in which the Sall site had been changed to EcoRI.
  • the plasmid was copied and modified using a modifying primer (pl8-eco420- 5 'TAG AAT TCA CCT GCA GGC ATG C3' (SEQ ID NO 60) in a pfu-based PCR (cycle 94 ° C 5s, 60 ° C 20s, 68 ° C 3 Min.
  • LTZ regions were ligated into the smal site of pTWFl ⁇ (Bioline QS ligase, 14 ° C. overnight). The ligation mixtures were used to transform competent DH5alpha cells. All bacterial cultivation work with plasmids containing the LTZ genome or portions thereof was done at 30 ° C based on stability considerations. LTZ 3 was cloned in half in pUC 18. The 5 'half (3a, antigenome sense) was amplified and modified using the primers LTZ 3.1 Xho + and F 3.82 Sst neg (see Table 2) in a PCR of 30 cycles (94 ° C 5s, 45 ° C 20s, 68 ° C 2 min.
  • the low copy plasmid pCTPE was a modification of pOLTV5 (Peeters et al (1999), J. Virol. 23, 5001-5009), in which the cloning efficiency was achieved by removing HDVR, T7 terminator and the remaining partial lac z genome regions using a Similar approach to that used to generate pTWFl ⁇ was strengthened.
  • both PCR primers were modifying (V5 630BE + 5 'CGG ATA TCC ACA GGA TCC GGG GAT AAC GC3' (SEQ ID NO 62) and V5 190 Barn- 5 'CGA GAT CCT CGA GCC GGA TCC TC3' (SEQ ID NO 63) and introduced new EcoRV blunt-ended sites flanked on each side by BamHI sites All growth media contained 15 ⁇ g / ml kanamycin (Gibco, Invirogen, Paisley, UK).
  • LTZ T71 was cloned into the EcoRV site of pCTPE, producing pCTPE-LTZT71 and the overall sequence of the insert was confirmed. It was then digested with Sall, in which LTZ2, cut from pTWFl ⁇ (Xhol Sall double digest), was ligated and cloned (DH5alpha (Invitrogen), kanamycin plates and broth). The orientation was checked by comparing a BamHl digest with a 3amHl, Sall double digest. The plasmid with the correctly oriented insert (pCTPE-LTZT71, 2) was cut with Sall only, and Xhol and Sall digested LTZ 3 RRRR were added and ligated as before.
  • Nucleocapsid (N), phospho (P) and matrix 2 (M2) sequences from strain LTZ1 were copied and cloned.
  • the RNA was extracted (Rnease, Qiagen), which was then reverse transcribed and amplified by RT-PCR (Superscript 2 Invitrogen; BioX-Act, Bioline; cycle of 94 ° C 5s, 50 ° C 20s, 68 ° C 2 min. [increasing 10s per cycle after cycle 5], repeated 29 times) using primers (see below) that introduced the T7 promoter sequence just before the start codon of each gene and proceeded beyond each stop codon.
  • T7 pre-fixed genes were cloned into the smal site of pUC18 using a procedure identical to that used for TWF above. PCR products without a T7 promoter were cloned into the smal site of pTarget (mammalian expression vector, Promega, Southhampton, UK).
  • the viral polymerase gene (L) was cloned into regions in the EcoRV site of pCTPE using the sequential approach used for the complete viral genome. In this order T7 L start, LTZ 6 + 7, LTZ8, LTZ 9 + 10 were ligated in pCTPE.
  • LTZ T7 L start was a PCR product (30 cycles 94 ° C 5s, 60 ° C 20s, 68 ° C 2 min. [Increasing 10s per cycle after cycle 5] repeated 29 times using Pfu [Stratagene]) from the 12 cycle trailer PCR described above using the primers T7-L (Table 3) and LTZ 7.6Xho- (Table 2).
  • LTZ T7start was ligated into the CPTE EcoRV site while the following areas of pTWF18 (Sall and Xhol) were cut and ligated into the new unique Sall site that was introduced at each stage of the cloning. The orientation was checked as with the full length genome.
  • the L gene was also cloned into pTarget in a manner similar to that described for the starter sequence (generated using the primers L Start Xho + (Table 2) and LTZ 7.6Xho (Table 2)) and this was not a T7 promoter upstream.
  • the primers used were as follows: leader area, T7-APV lead 1 and F 3.82 sst pos (5 'CCA CTC TGT AGG AGA CGC CGC GGC AAT TAT GCT TG3' (SEQ ID NO 75); central area F 3.82 sst neg (5 1 CAA GCA TAA TTG CCG CGG CGT CTC CTA CAG AGT GG3 ') (SEQ ID NO 76) and LTZ 8.2 Sal and the trailer area used LTZ 8.2 Xho + and APV trail ext.
  • Vero cells (70% confluent) in 35 mm wells were washed once with 1.0 ml Optimem 1, then combined with chickenpox T7 polymerase combined with an MOI of 0.2. After an incubation period of 1 hour, the medium was removed and the cells were washed with 1 ml Optimem 1, then with 2 ml.
  • the supernatant was collected, passed through a 0.2 ⁇ m filter and used to inoculate fresh Vero cells. CPE was observed and the cells were stained for the presence of TRT antigen using indirect immunofluorescent staining. The virus was confirmed to be derived from the full-length copy made by sequencing PCR copies from the unique RRRR regions of the F gene and other Sall / Xhol junction regions.
  • Vero cells (70% confluent) or CEFs in 35 mm wells were washed once with 1.0 ml Optimem 1; then 2 ml of MEM (5%) FCS was added.
  • the DNA / Fugene 6 (Boehringer Mannheim) complex was prepared by the four cloned carrier protein genes N, P, M2 (each 0.5 ⁇ g), L (50 ⁇ g) in pTarget, poll full-length genome (1 ⁇ g, cloned in pCPTE) and 10 ul Fugene 6 (Boehringer Mannheim) were mixed and dissolved in 300 ul dMEM. After complete mixing, this mixture was added dropwise to cells with gentle shaking.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pulmonology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Communicable Diseases (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne un vaccin agissant à l'encontre de membres de l'espèce du métapneumovirus ou du virus respiratoire syncytial ou d'un virus présentant une homologie générique significative dans la protéine F avec des membres de l'espèce du métapneumovirus, le vaccin étant un vaccin vivant atténué. Ledit vaccin est particulièrement indiqué pour lutter contre le métapneumovirus d'origine aviaire ou humaine.
EP03730063A 2002-05-16 2003-05-16 Attenuation du metapneumovirus Ceased EP1503792A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
DE10221836 2002-05-16
DE10221836A DE10221836A1 (de) 2002-05-16 2002-05-16 Attenuierung von Metapneumovirus
PCT/EP2003/005187 WO2003097089A2 (fr) 2002-05-16 2003-05-16 Attenuation du metapneumovirus

Publications (1)

Publication Number Publication Date
EP1503792A2 true EP1503792A2 (fr) 2005-02-09

Family

ID=29413891

Family Applications (1)

Application Number Title Priority Date Filing Date
EP03730063A Ceased EP1503792A2 (fr) 2002-05-16 2003-05-16 Attenuation du metapneumovirus

Country Status (12)

Country Link
US (1) US20060002958A1 (fr)
EP (1) EP1503792A2 (fr)
JP (1) JP2005538702A (fr)
CN (1) CN1652819A (fr)
AU (1) AU2003240668A1 (fr)
BR (1) BR0310046A (fr)
CA (1) CA2484295A1 (fr)
DE (1) DE10221836A1 (fr)
IL (1) IL164698A0 (fr)
MX (1) MXPA04011226A (fr)
PL (1) PL372529A1 (fr)
WO (1) WO2003097089A2 (fr)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101921732A (zh) 2001-01-19 2010-12-22 维洛诺瓦蒂夫公司 在易感哺乳动物中引起呼吸道疾病的病毒
US8715922B2 (en) 2001-01-19 2014-05-06 ViroNovative Virus causing respiratory tract illness in susceptible mammals
WO2003072720A2 (fr) 2002-02-21 2003-09-04 Medimmune Vaccines, Inc. Systemes d'expression de virus recombinant parainfluenza et vaccins comprenant des antigenes heterologues derives de metapneumovirus
JP2007525178A (ja) 2003-04-25 2007-09-06 メッドイミューン バクシーンズ,インコーポレイティド メタニューモウイルス株、そのワクチン製剤における用途、抗原性配列の発現のためのベクターとしての用途、並びにウイルス増殖方法
ES2386272T3 (es) * 2004-09-09 2012-08-16 Novartis Vaccines And Diagnostics Gmbh Reducción de riesgos iatrogénicos potenciales asociados a las vacunas antigripales
WO2010077712A1 (fr) * 2008-12-09 2010-07-08 Novavax, Inc. Particule de type viral du virus syncytial respiratoire bovin (vlps)
JP5796011B2 (ja) * 2009-06-24 2015-10-21 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム ワクチン
US9512178B2 (en) * 2012-07-11 2016-12-06 Research Foundation For Mental Hygiene, Inc. Neurogenic brain-derived neurotrophic factor peptides
WO2014208821A1 (fr) * 2013-06-27 2014-12-31 건국대학교기술지주 주식회사 Nouveau métapneumovirus aviaire et vaccin correspondant
KR101560337B1 (ko) 2013-06-27 2015-10-19 주식회사 카브 신규한 조류메타뉴모바이러스 및 그 백신
SI3718565T1 (sl) 2015-10-22 2022-08-31 Modernatx, Inc. Cepiva za respiratorni virus
EP3551193A4 (fr) 2016-12-08 2020-08-19 Modernatx, Inc. Vaccins à acide nucléique contre des virus respiratoires
RU2656188C1 (ru) * 2017-05-03 2018-06-04 Общество с ограниченной ответственностью "Анкрим" (ООО "Анкрим") Синтетическое анальгетическое средство пептидной природы и способ его применения
FR3084079A1 (fr) * 2018-07-23 2020-01-24 Universite Claude Bernard Lyon 1 Nouvelle souche virale attenuee et son utilisation en tant que vaccin
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6083925A (en) * 1995-06-07 2000-07-04 Connaught Laboratories Limited Nucleic acid respiratory syncytial virus vaccines
CN101921732A (zh) * 2001-01-19 2010-12-22 维洛诺瓦蒂夫公司 在易感哺乳动物中引起呼吸道疾病的病毒

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE PUB MED 29 November 2007 (2007-11-29), MOK, H. ET AL. *
GULATI B R ET AL: "Protective efficacy of high-passage avian pneumovirus (APV/MN/turkey/1-a/97) in Turkeys", AVIAN DISEASES, vol. 45, no. 4, July 2001 (2001-07-01) - September 2001 (2001-09-01), pages 593 - 597, XP002908367 *

Also Published As

Publication number Publication date
BR0310046A (pt) 2005-02-15
CA2484295A1 (fr) 2003-11-27
WO2003097089A3 (fr) 2004-02-05
JP2005538702A (ja) 2005-12-22
CN1652819A (zh) 2005-08-10
WO2003097089A2 (fr) 2003-11-27
MXPA04011226A (es) 2005-06-20
US20060002958A1 (en) 2006-01-05
IL164698A0 (en) 2005-12-18
DE10221836A1 (de) 2003-12-04
AU2003240668A1 (en) 2003-12-02
PL372529A1 (en) 2005-07-25

Similar Documents

Publication Publication Date Title
DE60124098T2 (de) Modifiziertes Morbillivirus V Proteine
DE69936585T2 (de) Newcastle-krankheitsvirus infektiöser klone, impfstoffe und diagnosetest
DE69532369T2 (de) Rekombinantes infektiöses nicht-in-Segmente-geteiltes, negativ-Strange-RNS-Virus
EP0912724B1 (fr) Production de vaccins a base de virus respiratoire syncytial attenue, a partir de sequences nucleotidiques clonees
EP1503792A2 (fr) Attenuation du metapneumovirus
CN100354425C (zh) 减毒的人-牛嵌合呼吸道合胞病毒疫苗的生产
DE60010358T2 (de) Infectiösenbursalkrankheitsvirus (IBDV) die an Zellkultur angepasst sind
EP1851239B1 (fr) Virus arn a replication deficiente en tant que vaccins
Collins et al. Rational design of live-attenuated recombinant vaccine virus for human respiratory syncytial virus by reverse genetics
DE60033939T2 (de) Rekombinante parainfluenzavirus vakzine, die durch deletion oder unterdrückung eines nicht wesentlichen gens abgeschwächt ist
EP2102345B1 (fr) Proteine f du virus respiratoire syncytial (rsv), et son utilisation
KR19990063811A (ko) 클론된 뉴클레오타이드 서열로부터 감염성 호흡장애 바이러스의생산 방법
DE69532588T2 (de) Verfahren zur verbesserung der immunogenität einer immunogenen zusammensetzung oder eines haptens und verwendung zur herstellung von impfstoffen
KR20020013526A (ko) 클로닝된 뉴클레오티드 서열로부터의 약독화된 키메릭호흡기 신시티움 바이러스 백신의 제조
DE69625356T2 (de) cDNA KLON FÜR DEN SÜDAFRIKANISCHEN ARBOVIRUS NO.86
EP2836231B1 (fr) Vaccin vivant atténué et de stabilite genetique contre le virus respiratoire syncytial et sa production
DE60129860T2 (de) Rekombinante rsv-virus-expressionssysteme und -impfstoffe
EP1276853B1 (fr) Proteines ns de pneumovirus antagonisant la reponse d'interferon (ifn)
Linné et al. The molecular biology of the porcine paramyxovirus LPMV
DE69838097T2 (de) Infektiöser klon für menschlichen parainfluenzavirus-typ
DE60011963T2 (de) Rekombinantes Newcastle-Krankheitsvirus zur in ovo Impfung
DE60120840T2 (de) Impfstoffe basierend auf Mutanten des Virus der infektiösen Bursitis
EP3497206A1 (fr) Variantes du virus de la bronchite infectieuse (bi) et compositions, utilisations et procédés les concernant
EP0859831B1 (fr) Production de virus syncytial respiratoire infectieux a partir de sequences de nucleotides clones
Brown Towards the development of novel ELISAS for avian pneumovirus (APV) serology

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20041013

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

17Q First examination report despatched

Effective date: 20050411

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20050411

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20090922