WO2013017570A1 - Novel prrs virus inducing type i interferon in susceptible cells - Google Patents

Novel prrs virus inducing type i interferon in susceptible cells Download PDF

Info

Publication number
WO2013017570A1
WO2013017570A1 PCT/EP2012/064893 EP2012064893W WO2013017570A1 WO 2013017570 A1 WO2013017570 A1 WO 2013017570A1 EP 2012064893 W EP2012064893 W EP 2012064893W WO 2013017570 A1 WO2013017570 A1 WO 2013017570A1
Authority
WO
WIPO (PCT)
Prior art keywords
virus
prrs virus
prrs
nspl
seq
Prior art date
Application number
PCT/EP2012/064893
Other languages
French (fr)
Inventor
Andreas GALLEI
Original Assignee
Boehringer Ingelheim Vetmedica Gmbh
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 Boehringer Ingelheim Vetmedica Gmbh filed Critical Boehringer Ingelheim Vetmedica Gmbh
Priority to EP12741333.4A priority Critical patent/EP2737059A1/en
Publication of WO2013017570A1 publication Critical patent/WO2013017570A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • A61K2039/552Veterinary vaccine
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/10011Arteriviridae
    • C12N2770/10021Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/10011Arteriviridae
    • C12N2770/10022New 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/10011Arteriviridae
    • C12N2770/10034Use 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/10011Arteriviridae
    • C12N2770/10061Methods of inactivation or attenuation
    • C12N2770/10062Methods of inactivation or attenuation by genetic engineering

Definitions

  • the present invention belongs to the field of vaccines and medicaments for the prophylaxis and treatment of infectious diseases.
  • it relates to attenuated live viruses useful as vaccine or medicament for preventing or treating Porcine Reproductive and Respiratory Syndrome (PRRS), a viral disease affecting swine.
  • PRRS Porcine Reproductive and Respiratory Syndrome
  • PRRSV Porcine reproductive and respiratory syndrome virus
  • PRRSV is a member of the virus family Arteriviridae and belongs, together with the Coronaviridae, to the virus order Nidoviraies.
  • PRRSV is an enveloped virus with a single-stranded, positive-sense RNA genome of about 15 kilobases comprising nine open reading frames (ORFs), namely ORF1a, ORFl ab, ORF2a, ORF 2ab, and ORFs 3 through ORF7.
  • ORFs 1 a and 1 ab encode large polyproteins that are processed into the viral nonstructural proteins (nsp) by auto- and transcleavages of viral proteases nspl , nsp2, and nsp4 (Snijder and Meulenberg, 1998).
  • nsp viral nonstructural proteins
  • the North American (US) prototype strain is VR-2332, while the European (EU) prototype strain is Lelystad virus.
  • PRRSV is considered one of the economically most important infectious agents in pigs causing late-term reproductive failure in sows and respiratory disease in growing pigs.
  • PRRSV infection is complicated by secondary bacterial infections being attributed to the immunosuppressive nature of the virus. Also, PRRSV viremia lasts for weeks, and virus then still can be detected in lymphoid organs for several months, demonstrating difficulties or failure of the host's immune response to clear the virus (Allende et al., 2000).
  • the specific immune response to PRRSV infection is characterized by delayed induction of neutralizing antibodies (Lopez and Osorio, 2004) and short cell-mediated immune response (Xiao et al., 2004). It is commonly accepted that these effects can in part be attributed, along with presentation of decoy epitopes (Ostrowski et al., 2002; Ansari et al., 2006) and glycan shielding of viral envelope proteins (Ansari et al., 2006), to the viral inhibition of the host's innate immune system.
  • PRRSV infection does not or only weakly or delayedly induce production of type I interferon (IFN) (interferon a and interferon ⁇ ; (Miller et al., 2004)) or type II IFN, (interferon ⁇ ; (Meier et al., 2003)) in susceptible cell lines (swine pumonary alveolar macrophages, monkey kidney cells MARC-145) and/or pigs (Buddaert et al., 1998).
  • IFN interferon
  • IFN interferon ⁇
  • type II IFN interferon ⁇
  • susceptible cell lines swine pumonary alveolar macrophages, monkey kidney cells MARC-145
  • pigs Buddaert et al., 1998.
  • nsps PRRSV nonstructural proteins
  • nspl is located at the N-terminus of the PRRSV ORF1a-derived polyprotein 1 a and is processed into two multifunctional subunits, nspl a and ⁇ ⁇ , each of which contains a papain-like cystein protease (PCP) domain essential for self-release from the viral polyprotein (den Boon et al., 1995; Chen et al., 2010).
  • PCP papain-like cystein protease
  • ⁇ ⁇ consists of an N-terminal domain (NTD), a linker domain (LKD), the PCP domain (PCP beta), and a C-terminal extension (CTE); (Xue et al., 2010), see Figure 2.
  • nspl ⁇ -mediated cleavage of nspl from nsp2 occurs at site WYG/AGR for PRRSV US strains (Kroese et al., 2008) or is predicted at site WYG/AAG for PRRSV EU strains (Chen et al., 2010), while ⁇ ⁇ / ⁇ ⁇ cleavage occurs at site ECAM/AxVYD for PRRSV US strains or is predicted at site EEAH/SxVYR for PRRSV EU strains (Chen et al., 2010).
  • PRRSV nspl and/or its autocleavage-derived subunits nspla and/or ⁇ ⁇ inhibit type I IFN production by interfering with IFN transcription (Song et al., 2010; Kim et al., 2010; Chen et al., 2010; Beura et al., 2010).
  • ⁇ ⁇ interferes with the cellular response to interferon (interferon signaling); (Chen et al . , 201 0).
  • PRRSV infection inhibits IFNa and/or IFNp production in PRRSV infected cells in vitro (Kim et al., 2010; Beura et al., 2010), the subcellular localization of nspl (subunits) was determined (Song et al., 2010; Chen et al., 2010), and mechanistic aspects of type I IFN inhibition that were obtained by others from single protein expression experiments were confirmed in cells infected with PRRSV (Shi et al., 2010).
  • nspl mutagenesis study based on nspl protein expression investigated effects on viral IFN inhibition (Shi et al., 2011 b), showing that mutations that inactivated papain-like cysteine protease activity of nspla made nspl lose its IFN antagonism activity, whereas mutations that inactivated papain-like cysteine protease activity of ⁇ ⁇ did not influence the IFN antagonism activity of nspl .
  • a viable PRRSV strain that induces IFN production and/or does not interfere with IFN signaling after infection of susceptible cells and/or the host in particular a PRRSV strain comprising a genomic mutation in its nspl gene, has not been described yet.
  • PRRS virus inducing the IFN response of a cell, in particular of a host cell, wherein said PRRS virus may serve as an effective vaccine or medicament for the prophylaxis or treatment of the Porcine reproductive and respiratory syndrome in swine.
  • the invention is based on the suprising finding of a Porcine Reproductive and Respiratory Syndrome (PRRS) virus which is able to induce the interferon type I response of a cell, in particular of a host cell.
  • PRRS Porcine Reproductive and Respiratory Syndrome
  • one aspect of the invention concerns a PRRS virus (PRRSV) which is able to induce the interferon type I production and secretion by a cell infected by said virus, wherein the PRRS virus according to the invention in particular is able to induce or induces the interferon type I production and secretion by an interferon competent cell infected by said virus.
  • PRRSV PRRS virus
  • interferon type I As used herein, it is understood that the terms "interferon type I”, “IFN type I”, “type I interferon” and “type I IFN” are equivalent.
  • Interferon type I production and secretion by a cell particularly means that interferon type I is made and released by a cell in response to the infection of the PRRS virus according to the invention.
  • interferon type I is preferably interferon-a and/or interferon- ⁇ .
  • the infection of a cell by the PRRS virus according to the invention in particular includes attachment of the virus to the cell, entry of the virus into the cell, disassembly of the virion, and preferably replication and transcription of the viral genome, expression of viral proteins, and assembly and release of new infectious viral particles.
  • the cell is a primary or secondary susceptile cell, preferably a mammalian cell, in particular a porcine or a simian cell, more preferably said cell is a porcine macrophage or a MA-104 cell or a MARC-145 cell or a Vero cell.
  • the PRRS virus according to the invention is able to induce in vitro and/or in vivo the interferon type I production and secretion by a cell infected by said virus.
  • the PRRS virus according to the invention is a live PRRS virus and/or a modified- live PRRS virus and/or an attenuated PRRS virus.
  • attenuated PRRS virus is in particular directed to a PRRS virus which is attenuated in vitro and/or in vivo, more particular in susceptible cell lines and/or the host.
  • host is in particular directed to animals infectable with PRRS virus, in particular swine, more particular pigs, such as domestic pigs.
  • attenuated particularly relates to a reduced virulence of a pathogen, in particular of a wild type PRRS virus, wherein “virulence” is understood to be the degree of pathogenicity, and wherein “pathogenicity” is directed to the ability of the pathogen to produce clinical symptoms in the host, such as elevated body temperature.
  • the PRRS virus according to the invention has or shows increased sensitivity to type I INF when compared to wild type PRRSV, wherein the term "sensitivity to type I INF" is understood as reduced viral infectivity when IFN is present, preferably in a sufficient amount for significantly reducing viral infectivity of wild type PRRSV, in the medium surrounding the virus at the time of infection.
  • wild type PRRS virus or wild type PRRSV, respectively, as used herein, is in particular directed to an infectious pathogenic PRRS virus, which is particularly capable of causing PRRS in swine.
  • wild type PRRS virus is directed to a PRRS virus whose genome comprises a RNA sequence or consists of a RNA polynucleotide, wherein said RNA sequence or RNA polynucleotide is a RNA copy of a polynucleotide selected from the group consisting of SEQ ID NO:13, SEQ ID NO:14 and SEQ ID NO:15.
  • the PRRS virus according to the invention is a PRRS virus mutant, in particular comprising, in comparison with the genome of a wild type PRRSV strain, a mutation in a gene encoding a protein of said virus.
  • the PRRS virus according to the invention comprises a mutation in the gene encoding the nspl protein of said virus.
  • the invention preferably concerns a PRRS virus which is able to induce the interferon type I production and secretion by an infected cell as a result of a mutation in the gene encoding the nspl protein, wherein said mutation is preferably a mutation as mentioned hereinafter.
  • the invention particulalry concerns a PRRS virus comprising, in comparison with the genome of a wild type strain, a mutation in the gene encoding the nspl protein.
  • the mutation as described herein is preferably a mutation in comparison with the sequence of the corresponding wild type gene encoding the nspl protein.
  • the invention is thus preferably directed to a PRRS virus, in particular a wild type PRRS virus, wherein a mutation, preferably a mutation as mentioned hereinafter, has been implemented in the genome of said virus resulting in a non-natural nspl protein or in the lack of nspl protein of said virus.
  • the mutation as described herein may be implemented to any PRRS virus, such as to a PRRS virus selected from the group consisting of wild type PRRS virus, attenuated PRRS virus, modified-life PRRS virus, PRRS virus mutant and combinations thereof.
  • the mutation as described herein may be implemented to an attenuated and/or live PRRS virus, for example to a PRRS virus selected from the group of the PRRS virus strains that have been deposited on 27 October 2004 with the European Collection of Cell Cultures (ECACC), Porton Down, Salisbury, Wiltshire, SP4 OJG, Great Britain, under the Accession Numbers ECACC 04102703, ECACC 04102702, and ECACC 04102704.
  • the mutation as described herein can be combined with one or more other mutations, preferably one ore more other attenuating mutations, in a PRRS virus.
  • PPRRS virus in particular equivalent with “PRRS virus strain”.
  • mutation in the context of the invention is understood as a change in a genomic sequence, in particular in the RNA sequence of a PRRS virus. Since viruses that use RNA as their genetic material have rapid mutation rates, the term “mutation”, as mentioned herein, is particularly directed to a genetically engineered change in a genomic sequence, such as by site directed mutagenesis, which in particular results in a virus growing to titers significantly lower than wild type PRRS virus in interferon competent cells and/or in the infected host, when propagated under the same conditions. Moreover, in another preferred embodiment the mutation described herein can also be caused by natural mutation and subsequent isolation of the PRRS virus according to the invention, wherein said isolated virus includes the mutation described herein.
  • the mutation comprises or consists of one or more point mutations and/or one or more genomic deletions and/or one or more insertions.
  • nspl protein is directed to the PRRSV nonstructural protein 1.
  • the nspl protein is preferably a polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:1 and SEQ ID NO:2 if the PRRS virus according to the invention is a genotype I PRRS virus, or the nspl protein is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide set forth in SEQ ID NO:3 if the PRRS virus according to the invention is a genotype II PRRS virus.
  • the PRRS virus according to the invention preferably also may comprise a mutation in the gene encoding the nspl protein selected from the group consisting of SEQ ID NOs 1-3.
  • the PRRS virus according to the invention is a genotype I PRRS virus or a genotype II PRRS virus. It is further understood that the terms “genotype I” and “genotype M” are equivalent to the terms “genotype 1” and “genotype 2” or to the terms “type 1” and “type 2", as frequently used in the literature in the context of PRRSV.
  • Sequence identity in the context of the invention is understood as being based on pairwise determined similarity between nucleotide or protein sequences.
  • the determination of percent identity between two sequences is preferably accomplished using a mathematical algorithm, in particular the well-known Smith-Waterman algorithm (Smith and Waterman, M. S. (1981 ) J Mol Biol, 147(1 ):195-197).
  • percent sequence identity of an amino acid sequence is determined using the Smith-Waterman homology search algorithm using an affine 6 gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix 62.
  • the Smith-Waterman homology search algorithm is taught in Smith and Waterman (1981 ) Adv. Appl. Math 2:482-489, herein incorporated by reference.
  • a variant may, for example, differ from the reference nspl , nspi a, ⁇ ⁇ or NTD molecule by as few as 1 to 15 amino acid residues, as few as 1 to 10 amino acid residues, such as 6- 10, as few as 5, as few as 4, 3, 2, or even 1 amino acid residue.
  • percent identity of a nucleotide sequence is determined using the Smith-Waterman homology search algorithm using a gap open penalty of 25 and a gap extension penalty of 5.
  • sequence identity can be performed using, for example, the DeCypher Hardware Accelerator from TimeLogic Version G, or the sequence identity is determined with the software CLC MAIN WORKBENCH 4.1.1 (CLC BIO).
  • the mutation in the PRRS virus according to the invention comprises or consists of one or more point mutations and/or one or more genomic deletions and/or one or more insertions.
  • the PRRS virus according to the invention comprises a mutation in the gene sequences encoding the nspl a subunit of the nspl protein and/or the nspl ⁇ subunit of the nspl protein of said virus.
  • nspla is thus directed to the PRRSV nspla subunit of the nspl protein, and the term " ⁇ ⁇ ”, as used herein, is hence directed to the PRRSV ⁇ ⁇ subunit of the nspl protein.
  • the nspl a is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:4 and SEQ ID NO:5 if the PRRS virus according to the invention is a genotype I PRRS virus, or the nspla is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide set forth in SEQ ID NO:6 if the PRRS virus according to the invention is a genotype II PRRS virus.
  • the nsp1 is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:7 and SEQ ID NO:8 if the PRRS virus according to the invention is a genotype I PRRS virus, or the ⁇ ⁇ is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide set forth in SEQ ID NO:9 if the PRRS virus according to the invention is a genotype II PRRS virus.
  • the PRRS virus according to the invention comprises a mutation in the gene sequence encoding the N-terminal domain (NTD) of the ⁇ ⁇ of said virus.
  • NTD N-terminal domain
  • the NTD is preferably a polypeptide or has a polypeptide sequence, respectively, having at least 60%, particularly at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide or a polypeptide sequence, respectively, selected from the group consisting of SEQ ID NO:10 and SEQ ID NO:1 1 if the PRRS virus according to the invention is a genotype I PRRS virus, or the NTD is preferably a polypeptide or has polypeptide sequence, respectively, having at least 60%, particularly at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide or the polypeptide sequence, respectively, set forth in SEQ ID NO:12 if the PRRS virus according to the invention is a genotype II PRRS virus.
  • the NTD comprising the mutation has the sequence selected from the group consisting of the sequences
  • the PRRS virus according to the invention is a genotype I PRRS virus
  • the NTD comprising the mutation preferably has the sequence selected from the group consisting of the sequences
  • the PRRS virus according to the invention preferably comprises a gene sequence encoding a NTD selected from the sequences
  • the PRRS virus according to the invention is a genotype I PRRS virus
  • the PRRS virus according to the invention preferably comprises a gene sequence encoding a NTD selected from the sequences
  • the NTD comprising the mutation may, for instance, have the sequence selected from the group consisting of the sequences
  • the PRRS virus according to the invention is a genotype I PRRS virus
  • NTD comprising the mutation may, for instance, have the sequence selected from the group consisting of the sequences
  • the PRRS virus according to the invention may, for instance, comprise a gene sequence encoding a NTD selected from the sequences
  • the PRRS virus according to the invention is a genotype I PRRS virus
  • the PRRS virus according to the invention may, for instance, comprise a gene sequence encoding a NTD selected from the sequences
  • the PRRS virus according to the invention is a genotype II PRRS virus.
  • a genotype I PRRS virus is in particular a virus whose genome comprises a gene sequence coding for a polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:7 and SEQ ID NO:8, or is in particular a virus whose genome comprises a gene sequence coding for a polypeptide having at least 60%, particularly at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO: 10 and SEQ ID NO:11.
  • the genotype I PRRS virus comprises or consists of a RNA polynucleotide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a RNA polynucleotide complementary to a polynucleotide selected from the group consisting of SEQ ID NO:13 and SEQ ID NO:14.
  • a genotype II PRRS virus is in particular a virus whose genome comprises a gene sequence coding for a polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:3, SEQ ID NO:6 and SEQ ID NO:9, or is in particular a virus whose genome comprises a gene sequence coding for a polypeptide having at least 60%, particularly at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide set forth in SEQ ID NO:12.
  • the genotype II PRRS virus comprises or consists of a RNA polynucleotide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a RNA polynucleotide complementary to the polynucleotide set forth in SEQ ID NO: 15.
  • the N-terminal domain (NTD) of ⁇ ⁇ starts with the N-terminus of ⁇ ⁇ , preferably starts with the amino acid sequence SXXY if the PRRS virus is a genotype I PRRS virus or with the amino acid sequence AXVY if the PRRS virus is a genotype II PRRS virus, and the NTD ends with the serine residue (S) within the amino sequence SFP of the ⁇ ⁇ .
  • NTD domain of a genotype II PRRS virus it is in particular referred to the publication Xue et al. 2010.
  • Said amino acid sequences SXXY or AXVY and SFP are especially conserved in the ⁇ ⁇ of wild type PRRS viruses and are thus preferably used amino acid sequence motifs for defining the NTD domain according to the invention.
  • the PRRS virus according to the invention thus comprises a mutation in the gene sequence coding for the NTD, wherein the NTD preferably starts with motif SXXY for PRRS virus type 1 strains or with motif AXVY for PRRS virus type 2 strains and ends with the serine residue (S) within the conserved SFP motif of the ⁇ ⁇ subunit.
  • S serine residue
  • the mutation thus comprises a deletion or replacement of a nucleotide triplett coding for an amino acid residue located within the first 24 N-terminal amino acid residues of the NTD sequence.
  • the mutation described herein comprises or consistis of a deletion or replacement of 2 to 7 or more, preferably consecutive, nucleotide tripletts each coding for an amino acid residue located within the first 24 N-terminal amino acid residues of the NTD sequence.
  • the mutation as mentioned herein, comprises the deletion or replacement of a nucleotide triplett coding for an amino acid residue with a charged, preferably positively charged, side chain, in particular coding for an arginine residue.
  • the mutation mentioned herein preferably consists of a deletion of 2, 3, 4, 5, 6 or 7 consecutive nucleotide tripletts coding for the respective number (2, 3, 4, 5, 6 or 7) of consecutive amino acid residues located within the first 24 N-terminal amino acid residues of the NTD sequence, wherein this mutation comprises the deletion of a nucleotide triplett coding for an amino acid residue with a charged, preferably positively charged, side chain, in particular coding for an arginine residue.
  • the mutation preferably comprises or consists of a deletion or replacement of the nucleotide triplett coding for the first arginine residue (R) located at least 21 amino acid residues in C-terminal direction from the N-terminal amino acid residue of the ⁇ ⁇ NTD and/or the mutation, as mentioned herein, comprises a deletion or replacement of the nucleotide triplett coding for the arginine residue (R) of the nspl amino acid sequence RXMW if the PRRS virus is a genotype I PRRS virus or with the amino acid sequence RGG of said virus if the PRRS virus is a genotype II PRRS virus and/or the mutation, as mentioned herein, comprises or consists of a deletion or replacement of the nucleotide triplett coding for the arginine residue (R) of the amino acid sequence RMM or RGG of the nspl protein.
  • the phrase "replacement of a nucleotide tiplett” is understood as being a replacement of a nucleotide triplett of the wild type sequence by another nucleotide triplett coding for a different amino acid residue than the wild type sequence.
  • the phrase "deletion of nucleotide triplett” in the context of the invention is directed to a deletion of a nucleotide triplett resulting in the deletion of an amino aicd residue in comparison with the wild type sequence.
  • said mutation further comprises or additionally consists of a deletion or replacement of one nucleotide triplett coding for the amino acid residue flanking said arginine residue (R) in N-terminal direction, wherein said mutation preferably comprises or consists of a deletion or replacement of two consecutive nucleotide tripletts coding for the amino acid residues PR. More particular, said mutation further comprises or additionally consists of a deletion or replacement of two, three, four, five, six, seven or more consecutive nucleotide tripletts coding for two, three, four, five, six, seven or more amino acid residues flanking said arginine residue (R) in N-terminal direction.
  • said mutation comprises or consists of a deletion or replacement of three consecutive nucleotide tripletts coding for the amino acid sequence RXR or APR of the ⁇ ⁇ , or wherein said mutation comprises a deletion or replacement of four consecutive nucleotide tripletts coding for the amino acid residues GRXR or WAPR of the ⁇ ⁇ .
  • the PRRS virus according to the invention comprises in one embodiment a gene coding for a nspl protein selected from the group consisting of SEQ ID NO: 16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO: 19 and SEQ ID NO:20, or comprises in another embodiment a gene sequence coding for a ⁇ ⁇ selected from the group consisiting of SEQ ID NO:21 , SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24 and SEQ ID NO:25, or comprises in a further embodiment a gene sequence coding for a NTD selected from the group consisting of SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29 and SEQ ID NO:30.
  • the mutation as described herein comprises or consists of a deletion of the gene sequence encoding the (whole) nspl protein, encoding the (whole) nspla subunit, encoding the (whole) ⁇ ⁇ subunit, or encoding a part, preferably at least seven consecutive amino acid residues, of or the (whole) NTD domain of the ⁇ ⁇ subunit.
  • Yet another aspect of the invention is directed to a polynucleotide comprising or consisting of the genome of the PPRS virus according to the invention.
  • the invention is also directed to a virus particle, wherein said virus particle comprises a polynucleotide which comprises or consists of the genome of the PPRS virus according to the invention or which comprises or consists of a DNA copy of the PPRS virus according to the invention. Still further, the present invention provides a DNA-Vector comprising a copy of, or a cDNA sequence complementary to, respectively, a polynucleotide which comprises or consists of the genome of the PPRS virus according to the invention.
  • the DNA vector comprises a polynucleotide sequence selected from the group consisting of SEQ ID NO:31 , SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34 and SEQ ID NO:35.
  • the present invention provides a cell comprising a polynucleotide, which comprises or consists of the genome of the PPRS virus according to the invention or which comprises or consists of a DNA copy of the PPRS virus according to the invention.
  • the PRRS virus according to the invention, or the genome of the PRRS virus according to the invention respectively, comprises a RNA sequence or consists of a RNA polynucleotide, wherein said RNA sequence or RNA polynucleotide is a RNA copy of a polynucleotide selected from the group consisting of SEQ ID NO:31 , SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34 and SEQ ID NO:35.
  • Another aspect of the invention concerns the PRRS virus according to the invention for use as vaccine or medicament, in particular for use in the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome preferably in swine.
  • the present invention further provides a composition containing the PRRS virus according to the invention for use as a vaccine or as a medicament, in particular for use in the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome preferably in swine.
  • said vaccine or medicament is administered in two doses or preferebly in one dose to an animal in need thereof.
  • prophylaxis or therapy it is meant that the prophylaxis is to administer a drug, in particular a vaccine, before exposure to PRRSV and the treatment is to administer a drug, in particular a medicament, after infection with PRRSV or onset of PRRS.
  • the vaccine as mentioned herein, is a live vaccine and/or a modified live vaccine and/or an attenuated live or attenuated modified live vaccine.
  • the strains of the PRRS virus according to the invention can be grown and harvested by methods known in the art, e.g. by propagating in suitable cells like the simian cell line MA- 104, Vera cells, or porcine alveolar macrophages.
  • vaccines according to the present invention are modified live vaccines comprising one or more of these strains alive in a suitable carrier, but inactivated virus may also be used to prepare killed vaccine (KV).
  • Modified live vaccines (MLV) are typically formulated to allow administration of 10 1 to 10 7 viral particles per dose, preferably 10 3 to 10 6 particles per dose, more preferably 10 4 to 10 6 particles per dose (4.0-6.0 log 0 TCID 50 ).
  • KV may be formulated based on a pre-inactivation titre of 10 3 to 10 10 viral particles per dose.
  • the vaccine may comprise a pharmaceutically acceptable carrier, for example a physiological salt-solution.
  • the vaccine may or may not comprise an adjuvant.
  • An example of a suitable adjuvant is a-tocopherol acetate which can be obtained under the trade name Diluvac Forte®.
  • alum based adjuvants may be used.
  • a further aspect of the invention is thus directed to the use of the PRRS virus according to the invention for the preparation of a vaccine or medicament for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome preferably in swine.
  • a vaccine according to the present invention may be presented in form of a freeze-dried preparation of the live virus, to be reconstituted with a solvent, to result in a solution for injection.
  • the solvent may e.g. be water, physiological saline, or buffer, or an adjuvanting solvent.
  • the solvent may contain adjuvants, for example a-tocopherol acetate.
  • the reconstituted vaccine may then be injected into a pig, for example as an intramuscular or intradermal injection into the neck.
  • the present invention therefore is a product, comprising in separate containers a freeze-dried composition of the virus, and a solvent for reconstitution, and optionally further containing a leaflet or label comprising instructions of use.
  • Yet another aspect of the invention thus concerns a method for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome comprising administering the PRRS virus according to the invention to an animal, preferably to a swine, more preferably to a pig, in particular preferably to a piglet or a sow.
  • a vaccine according to the present invention may not only comprise the PRRS virus according to the invention as antigen, but may include further components active against PRRS or other porcine viral or bacterial diseases, like porcine circovirus, classical swine fever virus or mycoplasma hyorhinis. Therefore, the invention further relates to a vaccine as described, characterised in that it contains at least one further antigen active against a porcine disease which is not PRRS.
  • Still another aspect of the invention thus concerns a medicament or vaccine comprising the PRRS virus according to the invention.
  • Still a further aspect of the invention is directed to a method of preparing a vaccine or medicament, in particular for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome preferably in swine, said method comprising the cultivation of the PRRS virus according to the invention in cell culture.
  • the invention comprises a method of producing the PRRS virus according to the invention, said method comprising the mutagenesis of the gene coding for the nspl protein in the genome of a PRRS virus, and wherein the mutation, as described herein, is in particular introduced by genetic engineering in the genome of said virus, preferably by site directed mutagenesis.
  • the above-mentioned DNA-vector is used and/or said methods comprise the detection of interferon type I, in particular interferon type I secretion, and/or of mRNA coding for interferon type I.
  • Said detection in particular comprises the detection of interferon type I and/or of mRNA coding for interferon type I by a bioassay, wherein said bioassay is preferably selected from the group consisting of ELISA, PCR, GLISA, IFA, biosensoric measurement, Surface Plasmon Resonance (SPR) measurement, selective media , lateral flow, biochip measurement, immunomagnetic separation , electrochemiluminescence, chromogenic media, immunodiffusion, DNA hybridization, staining, colorimetric detection, luminescence, and combinations thereof.
  • a bioassay is preferably selected from the group consisting of ELISA, PCR, GLISA, IFA, biosensoric measurement, Surface Plasmon Resonance (SPR) measurement, selective media , lateral flow, biochip measurement, immunomagnetic separation , electrochemiluminescence, chromogenic media, immunodiffusion, DNA hybridization, staining, colorimetric detection, luminescence, and combinations thereof.
  • the invention provides an immunogenic composition containing the PRRS virus according to the invention.
  • immunogenic composition in particular refers to a composition that will elicit an immune response in an animal that has been exposed to the composition.
  • An immune response may include induction of antibodies and/or induction of a T-cell response.
  • an "immune response” includes but is not limited to one or more of the following effects: the production or activation of antibodies, B cells, helper T cells, suppressor T cells, and/or cytotoxic T cells, directed specifically to an antigen or antigens included in the composition or vaccine of interest.
  • the host will display either a therapeutic or a protective immunological (memory) response such that resistance to new infection will be enhanced and/or the clinical severity of the disease reduced.
  • a protective immunological (memory) response such that resistance to new infection will be enhanced and/or the clinical severity of the disease reduced.
  • Such protection will be demonstrated by either a reduction in number or severity of, or lack of one or more of the symptoms associated with the infection of the pathogen, in the delay of onset of viremia, in a reduced viral persistence, in a reduction of the overall viral load and/or in a reduction of viral excretion.
  • an “immune response” in particular means but is not limited to the development in a subset of a cellular and/or antibody-mediated immune response to the composition or vaccine of interest.
  • the immunogenic composition of the invention is preferably a vaccine or medicament.
  • viremia is particularly understood as a condition in which PRRS virus particles circulate in the bloodstream of an animal.
  • animal is in particular directed to swine, more particular to a pig, preferably a domestic pig.
  • the immunogenic composition of the invention comprises an amount of 10 1 to 10 7 viral particles of the PRRS virus according to the invention per dose, preferably 10 3 to 10 6 particles per dose, more preferably 10 4 to 10 6 particles per dose.
  • the immunogenic composition of the invention comprises an amount of the PRRS virus according to the invention which is equivalent to a virus titre of at least about 10 3 TCID 50 /mL per dose, preferably between 10 3 to 10 5 TCID 50 /mL per dose
  • the immunogenic composition of the invention further contains one or more pharmaceutically acceptable carriers or excipients.
  • Said one or more pharmaceutically acceptable carriers or excipients are preferably selected from the group consisting of solvents, dispersion media, adjuvants, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, and adsorption delaying agents.
  • the present invention is further directed to the immunogenic composition of the invention for use in a method for
  • the term "inducing an immune response" to an antigen or composition is the development of a humoral and/or cellular immune response in an animal to an antigen present in the composition of interest.
  • prevention means, but is not limited to a process which includes the administration of a PRRSV antigen, namely of the PRRSV according to the invention which is included in the composition of the invention, to an animal, wherein said PRRSV antigen, when administered to said animal elicits or is able to elicit an immune response in said animal against PRRSV.
  • PRRS induction or onset of the disease process
  • “reduction of PRRS” or “reduction of clinical symptoms associated with PRRSV infection” means, but is not limited to, reducing the number of infected subjects in a group, reducing or eliminating the number of subjects exhibiting clinical symptoms of infection, or reducing the severity of any clinical symptoms that are present in the subjects, in comparison to wild-type infection.
  • these clinical symptoms are reduced in subjects receiving the composition of the present invention by at least 10% in comparison to subjects not receiving the composition and may become infected. More preferably, clinical symptoms are reduced in subjects receiving the composition of the present invention by at least 20%, preferably by at least 30%, more preferably by at least 40%, and even more preferably by at least 50%.
  • the elevated body temperature usually associated with the administration of an attenuated PRRSV vaccine to an animal is reduced in subjects receiving the composition of the present invention by at least 10% in comparison to subjects receiving a conventional attenuated PRRSV vaccine. More preferably, the elevated body temperature usually associated with the administration of an attenuated PRRSV vaccine is reduced in subjects receiving the composition of the present invention by at least 20%, preferably by at least 30%, more preferably by at least 40%, and even more preferably by at least 50%.
  • a further advantage of the PRRS virus according to the invention is in particular the effect that its administration to an animal, e.g. for the prophylaxis or treatment of PRRS, results in a reduced increase of body temperature in the animal in comparision with a conventional attenuated PPRSV vaccine.
  • inventions are in particular directed to any attenuated PRRSV useful as a vaccine, wherein said PRRSV does not have the characteristic features of the PRRS virus of the present invention, in particular is not able to induce the interferon type I production and secretion by a cell infected by said PRRSV.
  • subject in particular relates to an animal.
  • body temperature refers to the approximate average normal, internal temperature of an animal, for example about 38.5-39°C in pigs, whereas the body temperature associated with a PRRSV infection may be elevated up to 41 °C in pigs.
  • the term "reduction of PRRSV viremia” means, but is not limited to, the reduction of PRRS virus entering the bloodstream of an animal, wherein the viremia level, i.e. the number of PRRSV RNA copies per ml_ of blood serum or the number of plaque forming colonies per deciliter of blood serum, is reduced in the blood serum of subjects receiving the composition of the present invention by at least 50% in comparison to subjects not receiving the composition and may become infected. More preferably the viremia level is reduced in subjects receiving the composition of the present invention by at least 90%, preferably by at least 99.9%, more preferably by at least 99.99%, and even more preferably by at least 99.999%.
  • the present invention further relates to the use of the PRRS virus according to the invention or of the immunogenic composition of the invention for the preparation of a vaccine or medicament for
  • the vaccine or medicament is to be administered in two doses or preferably in a single dose to an animal.
  • the present invention also provides a method of preparing an immunogenic composition, preferably a vaccine or medicament, more preferably a vaccine or medicament for
  • said method comprises the step of mixing the PRRS virus according to the invention with one or more pharmaceutically acceptable carriers or excipients, and wherein said one or more pharmaceutically acceptable carriers or excipients are preferably selected from the group consisting of solvents, dispersion media, adjuvants, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, and adsorption delaying agents.
  • the present invention further provides a method for
  • said method comprises the step of administering the immunogenic composition of the invention to an animal in need thereof, and wherein preferably the immunogenic composition and/or the vaccine is administered in two doses or, more preferably, in a single dose.
  • the deletions are located in the predicted ⁇ ⁇ portion of nspl ( Figure 2).
  • the deletion site for all vaccine candidates is located in the N-terminal domain (NTD) of ⁇ ⁇ and overlaps with aminoacids P23R24 of GD-XH ⁇ ⁇ ; ( Figure 2).
  • virus titers of the obtained virus stocks were determined for each vaccine candidate by serial virus titrations on 96-well plates containing MA104 cells followed by PRRSV-specific immunofluorescence analyses six to seven days post infection. Unlike experience with titrations of virus stocks from parental PRRSV LoN94-13 (data not shown), the first serial dilutions of vaccine candidates delta nspl XVII-1 and delta nspl XVIII-12 did not demonstrate a cytopathic effect and virus plaque formation, while at higher dilutions of the virus stocks a cytopathic effect was detectable.
  • a commercial ELISA specific for human IFN (Invitrogen) was used.
  • MA104 cells are epithelial Green Monkey kidney cells. According to the ELISA manufacturer, this Invitrogen ELISA is also suited for the detection of primate IFN other than human.
  • 100 ⁇ served as assay input, while a virus stock from parental strain LoN94-13, cell culture medium , and medium from noninfected cells served as controls.
  • a calibration curve was included using a positive control of the ELISA manufacturer. All samples were measured in duplicates.
  • virus stocks of the vaccine candidates contained considerable levels of type I IFN, while the virus stock of the parental virus showed IFN levels as low as the negative controls ( Figure 4).
  • cells infected for the time course experiment summarized in Figure 5 not only produced considerable amounts of ⁇ , but at the end of the experiment at six days post infection, cells infected with vaccine candidates delta nspl XVII-1 and delta nspl XVIII- 12 showed signs of recovery from usually lytical PRRSV infection. While cells infected with parental strain LoN94-13 were fully lysed, cells infected with the vaccine candidates grew in a partially (delta nspl XVIII-12) or completely intact monolayer (delta nspl XVII-1 ). For the latter, only weak signs of a PRRSV-induced cytopathic effect were still detectable.
  • the interferon production of infected cells together with the observed sensitivity of vaccine candidates to type I IFN correlated with partial or almost complete recovery of infected cells over time. It is reasonable to expect that type I IFN induction by the vaccine candidates together with their increased sensitivity to type I IFN will contribute to a significantly attenuated viral phenotype in the natural host.
  • expected features of the vaccine candidates' attenuation in pigs include stimulation of the innate and specific immunity, both humoral and cellular, and less shedding and/or shortened viremia of the vaccine viruses.
  • Mean body temperatures were determined for each animal for the time after vaccination but before challenge using measured body temperature data from all timepoints from day 0 through day 20. Subsequently, mean body temperatures were determined for each group ( Figure 7, blue (left-hand) columns). Error bars indicate standard deviations, respectively. Following the same procedure, mean body temperatures and standard deviations were determined for all groups for the time after challenge using measured body temperature data from all timepoints from day 22 through 31 ( Figure 7, brown (right-hand) columns).
  • the nspl group showed a significant reduction in mean body temperature of more than 0.2 °C when compared to the WT group.
  • the considerable reduction in increase of body temperature compared to WT demonstrates that the described mutation in the genome of delta nspl XVIII-12 has significantly reduced virulence of the WT parental strain LoN94-13 in the infected animal.
  • the significant rise in the mean body temperature of the Ch control group from before challenge to after challenge of 0.4°C demonstrates virulence of the heterologous EU PRRSV challenge strain.
  • the mean temperature of the WT group after challenge was slightly lower than before challenge, but not significantly reduced (Figure 7).
  • the invention described here represents the first known viable PRRSV (EU) strains that contain mutations (deletions) in the nspl gene ( ⁇ ⁇ ) that induce type I IFN (IFN ) production in susceptible cells (MA104) and that show increased sensitivity to type I IFN (IFNp).
  • the animal data demonstrates that (i) vaccine candidate delta nspl XVIII-12 is significantly attenuated in the host when compared to its parental PRRSV strain LoN94-13 and that (ii) vaccine candidate delta nspl XVIII-12 confers significant protection from signs of disease induced by challenge with a heterologous PRRSV strain while parental strain LoN94-1 3 does not.
  • the described vaccine candidates or the described mutations therein, either alone or combined with other attenuating mutations may serve as promising life attenuated PRRSV vaccines. LIST OF FIGURES
  • Figure 1 Sequence alignment of ⁇ ⁇ protein sequences from parental EU PRRSV strain LoN94-13 and from vaccine candidates.
  • Figure 2 Sequence alignment of ⁇ ⁇ proteins from PRRSV strains.
  • Figure 3 PRRSV-specific immunofluorescence of virus stock titrations.
  • Figure 4 Virus stocks of vaccine candidates contain IFN3.
  • Figure 5 Time course of IFN induction after infection of MA104 cells.
  • Figure 6 delta nspl mutants show increased sensitivity to type I IFN.
  • Figure 7 Mean body temperatures of vaccinated groups before and after challenge.
  • SEQ ID NO:1 corresponds to LoN94-13 complete nspl protein
  • SEQ ID NO:2 corresponds to Lelystad virus complete nspl protein
  • SEQ ID NO:3 corresponds to VR2332 complete nspl protein
  • SEQ ID NO:4 corresponds to LoN94-13 complete nspl Alpha
  • SEQ ID NO:5 corresponds to Lelystad virus complete nspl Alpha
  • SEQ ID NO:6 corresponds to VR2332 complete nspl Alpha
  • SEQ ID NO:7 corresponds to LoN94-13 complete nspl Beta
  • SEQ ID NO:8 corresponds to Lelystad virus complete nspl Beta
  • SEQ ID NO:9 corresponds to VR2332 complete nspl Beta
  • SEQ ID NO: 10 corresponds to LoN94-13 nspl Beta NTD
  • SEQ ID NO:1 1 corresponds to Lelystad virus nspl Beta NTD
  • SEQ ID NO:12 corresponds to VR2332 nspl Beta NTD
  • SEQ ID NO:13 corresponds to LoN94-13 complete viral cDNA insert
  • SEQ ID NO:14 corresponds to Lelystad virus complete genome
  • SEQ ID NO: 15 corresponds to VR2332 complete genome
  • SEQ ID NO:16 corresponds to delta nspl IX-10 complete nspl protein sequence
  • SEQ ID NO:17 corresponds to delta nspl XVII-1 complete nspl protein sequence
  • SEQ ID NO:18 corresponds to delta nspl XVIII-12 complete nspl protein sequence
  • SEQ ID NO:19 corresponds to delta nspl XIX-2 complete nspl protein sequence
  • SEQ ID NO:20 corresponds to delta nspl XX-9 complete nspl protein sequence
  • SEQ ID NO:21 corresponds to delta nspl IX-10 complete nspl Beta protein sequence
  • SEQ ID NO 22 corresponds to delta nsp XVII- 1 complete nspl Beta protein sequence
  • SEQ ID NO 23 corresponds to delta nsp XVIII- 12 complete nspl Beta protein sequence
  • SEQ ID NO 24 corresponds to delta nsp XIX- 2 complete nspl Beta protein sequenc
  • SEQ ID NO 25 corresponds to delta nsp XX- 9 complete nspl Beta protein sequence
  • SEQ ID NO 26 corresponds to delta nsp IX-10 nspl Beta NTD protein sequence
  • SEQ ID NO 27 corresponds to delta nsp XVII- 1 nspl Beta NTD protein sequence
  • SEQ ID NO 28 corresponds to delta nsp XVIII- 12 nspl Beta NTD protein sequence
  • SEQ ID NO 29 corresponds to delta nsp XIX- 2 nspl Beta NTD protein sequence
  • SEQ ID NO 30 corresponds to delta nsp XX- 9 nspl Beta NTD protein sequence
  • SEQ ID NO 31 corresponds to delta nsp IX-10 complete viral cDNA insert sequence
  • SEQ ID NO 32 corresponds to delta nsp XVII- 1 complete viral cDNA insert sequence
  • SEQ ID NO 33 corresponds to delta nsp XVIII- 12 complete viral cDNA insert sequence
  • SEQ ID NO 34 corresponds to delta nsp XIX- 2 complete viral cDNA insert sequence
  • SEQ ID NO 35 corresponds to delta nsp XX-9 complete viral cDNA insert sequence
  • SEQ ID NO 36 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion o1 2 aa),
  • SEQ ID NO 37 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion of 3 aa),
  • SEQ ID NO 38 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 4 aa),
  • SEQ ID NO 39 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 5 aa),
  • SEQ ID NO 40 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion o1 6 aa),
  • SEQ ID NO 41 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 7 aa),
  • SEQ ID NO 42 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 2 aa),
  • SEQ ID NO 43 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion of 3 aa),
  • SEQ ID NO 44 corresponds to a (type 2) nspl Beta NTD with mutal tion (deletion o1 4 aa),
  • SEQ ID NO 45 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion o1 5 aa),
  • SEQ ID NO 46 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 6 aa),
  • SEQ ID NO 47 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 7 aa),
  • SEQ ID NO 48 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion of 2 aa),
  • SEQ ID NO 49 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion o1 3 aa),
  • SEQ ID NO 50 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 4 aa),
  • SEQ ID NO 51 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 5 aa),
  • SEQ ID NO 52 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion of 6 aa),
  • SEQ ID NO 53 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion o1 7 aa),
  • SEQ ID NO 54 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion of 2 aa),
  • SEQ ID NO 55 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 3 aa),
  • SEQ ID NO 56 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 4 aa),
  • SEQ ID NO 57 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 5 aa)
  • SEQ ID NO:58 corresponds to a (type 2) nspl Beta NTD with mutation (deletion of 6 aa)
  • SEQ ID NO:59 corresponds to a (type 2) nspl Beta NTD with mutation (deletion of 7 aa).
  • Porcine reproductive and respiratory syndrome virus description of persistence in individual pigs upon experimental infection. J. Virol. 74, 10834-10837. Ansari, I.H., Kwon, B., Osorio, F.A., Pattnaik, A.K., 2006. Influence of N-linked glycosylation of porcine reproductive and respiratory syndrome virus GP5 on virus infectivity, antigenicity, and ability to induce neutralizing antibodies. J. Virol. 80, 3994-4004.
  • Porcine reproductive and respiratory syndrome virus nonstructural protein 1 eta modulates host innate immune response by antagonizing IRF3 activation. J.
  • Nonstructural Protein 1 Papain-Like Cysteine Protease Was Necessary for Porcine Reproductive and Respiratory Syndrome Virus Nonstructural Protein 1 to Inhibit Interferon-beta Induction. DNA Cell Biol. 30, 355-362. Snijder, E.J., Meulenberg, J. J., 1998. The molecular biology of arteriviruses. J. Gen. Virol. 79 ( Pt 5), 961-979.

Landscapes

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

Abstract

The present invention relates to the field of attenuated live viruses useful as vaccine or medicament for preventing or treating Porcine Reproductive and Respiratory Syndrome (PRRS) in swine, and is based on the suprising finding of a PRRS virus which is able to induce the interferon type I response of a cell infected by said virus. In one embodiment, the PRRS virus according to the invention is a PRRS virus mutant comprising, in comparison with the genome of a wild type strain, a mutation in the gene encoding the non structural protein 1 (nsp1) of said virus.

Description

NOVEL PRRS VIRUS INDUCING TYPE I INTERFERON IN
SUSCEPTIBLE CELLS
FIELD OF THE INVENTION
The present invention belongs to the field of vaccines and medicaments for the prophylaxis and treatment of infectious diseases. In particular, it relates to attenuated live viruses useful as vaccine or medicament for preventing or treating Porcine Reproductive and Respiratory Syndrome (PRRS), a viral disease affecting swine.
BACKGROUND OF THE INVENTION
Porcine reproductive and respiratory syndrome virus (PRRSV) is a member of the virus family Arteriviridae and belongs, together with the Coronaviridae, to the virus order Nidoviraies. PRRSV is an enveloped virus with a single-stranded, positive-sense RNA genome of about 15 kilobases comprising nine open reading frames (ORFs), namely ORF1a, ORFl ab, ORF2a, ORF 2ab, and ORFs 3 through ORF7. ORFs 1 a and 1 ab encode large polyproteins that are processed into the viral nonstructural proteins (nsp) by auto- and transcleavages of viral proteases nspl , nsp2, and nsp4 (Snijder and Meulenberg, 1998). There are two distinct viral PRRSV genotypes causing similar clinical symptoms that diverge by about 40 % on nucleotide sequence level, genotype I (EU) and genotype II (US). The North American (US) prototype strain is VR-2332, while the European (EU) prototype strain is Lelystad virus. PRRSV is considered one of the economically most important infectious agents in pigs causing late-term reproductive failure in sows and respiratory disease in growing pigs. Often, PRRSV infection is complicated by secondary bacterial infections being attributed to the immunosuppressive nature of the virus. Also, PRRSV viremia lasts for weeks, and virus then still can be detected in lymphoid organs for several months, demonstrating difficulties or failure of the host's immune response to clear the virus (Allende et al., 2000).
The specific immune response to PRRSV infection is characterized by delayed induction of neutralizing antibodies (Lopez and Osorio, 2004) and short cell-mediated immune response (Xiao et al., 2004). It is commonly accepted that these effects can in part be attributed, along with presentation of decoy epitopes (Ostrowski et al., 2002; Ansari et al., 2006) and glycan shielding of viral envelope proteins (Ansari et al., 2006), to the viral inhibition of the host's innate immune system. It has been demonstrated that PRRSV infection does not or only weakly or delayedly induce production of type I interferon (IFN) (interferon a and interferon β; (Miller et al., 2004)) or type II IFN, (interferon γ; (Meier et al., 2003)) in susceptible cell lines (swine pumonary alveolar macrophages, monkey kidney cells MARC-145) and/or pigs (Buddaert et al., 1998).
Thus, there is a need for novel vaccines and medications effecting a rapid induction of neutralizing antibodies and interferon responses for the prophylaxis and treatment of PRRSV infection.
DESCRIPTION OF THE INVENTION
IFNs play an important role in establishing an effective adaptive immune response against viral infections, and many viruses therefore have developed strategies and counteractions against onset of the host's innate immune system (Haller and Weber, 2009). In the interest to identify the anticipated PRRSV IFN antagonist(s), extensive screening analyses based on cell lines stably expressing genes of interest or on cells transfected with protein-expressing plasmids have identified several PRRSV nonstructural proteins (nsps) including nspl (see below), nsp2 (Beura et al., 2010; Li et al., 2010), nsp4 (Beura et al., 2010), and nspl 1 (Beura et al., 2010; Shi et al., 2011a) to be involved in blocking the induction of type I IFN. nspl is located at the N-terminus of the PRRSV ORF1a-derived polyprotein 1 a and is processed into two multifunctional subunits, nspl a and ηβρΐ β, each of which contains a papain-like cystein protease (PCP) domain essential for self-release from the viral polyprotein (den Boon et al., 1995; Chen et al., 2010). nspl a contains an N-terminal zinc- finger domain and the PCPa protease domain, while ηερΐ β contains PCP . For both nspl subunits, nspla and ηερΐ β, the three-dimensional crystal structure has been resolved (Sun et al., 2009; Xue et al., 2010). According to these analyses, ηερΐ β consists of an N-terminal domain (NTD), a linker domain (LKD), the PCP domain (PCP beta), and a C-terminal extension (CTE); (Xue et al., 2010), see Figure 2. C-terminal, nspl β-mediated cleavage of nspl from nsp2 occurs at site WYG/AGR for PRRSV US strains (Kroese et al., 2008) or is predicted at site WYG/AAG for PRRSV EU strains (Chen et al., 2010), while ηερΐ α/ηερΐ β cleavage occurs at site ECAM/AxVYD for PRRSV US strains or is predicted at site EEAH/SxVYR for PRRSV EU strains (Chen et al., 2010). Several studies on protein level demonstrated to the mechanistic detail that PRRSV nspl and/or its autocleavage-derived subunits nspla and/or ηερΐ β inhibit type I IFN production by interfering with IFN transcription (Song et al., 2010; Kim et al., 2010; Chen et al., 2010; Beura et al., 2010). In addition, it has been demonstrated that ηερΐ β interferes with the cellular response to interferon (interferon signaling); (Chen et al . , 201 0). Moreover, it was demonstrated that PRRSV infection inhibits IFNa and/or IFNp production in PRRSV infected cells in vitro (Kim et al., 2010; Beura et al., 2010), the subcellular localization of nspl (subunits) was determined (Song et al., 2010; Chen et al., 2010), and mechanistic aspects of type I IFN inhibition that were obtained by others from single protein expression experiments were confirmed in cells infected with PRRSV (Shi et al., 2010). Finally, a nspl mutagenesis study based on nspl protein expression investigated effects on viral IFN inhibition (Shi et al., 2011 b), showing that mutations that inactivated papain-like cysteine protease activity of nspla made nspl lose its IFN antagonism activity, whereas mutations that inactivated papain-like cysteine protease activity of ηερΐ β did not influence the IFN antagonism activity of nspl .
However, a viable PRRSV strain that induces IFN production and/or does not interfere with IFN signaling after infection of susceptible cells and/or the host, in particular a PRRSV strain comprising a genomic mutation in its nspl gene, has not been described yet.
It is thus an aim of the present invention to make available such a viable PRRS virus inducing the IFN response of a cell, in particular of a host cell, wherein said PRRS virus may serve as an effective vaccine or medicament for the prophylaxis or treatment of the Porcine reproductive and respiratory syndrome in swine.
The solution to the above technical problem is achieved by the description and the embodiments characterized in the claims.
Thus, the invention in its different aspects and embodiments is implemented according to the claims.
The invention is based on the suprising finding of a Porcine Reproductive and Respiratory Syndrome (PRRS) virus which is able to induce the interferon type I response of a cell, in particular of a host cell. Hence, one aspect of the invention concerns a PRRS virus (PRRSV) which is able to induce the interferon type I production and secretion by a cell infected by said virus, wherein the PRRS virus according to the invention in particular is able to induce or induces the interferon type I production and secretion by an interferon competent cell infected by said virus.
As used herein, it is understood that the terms "interferon type I", "IFN type I", "type I interferon" and "type I IFN" are equivalent.
Interferon type I production and secretion by a cell, as described herein, particularly means that interferon type I is made and released by a cell in response to the infection of the PRRS virus according to the invention. In this regard, interferon type I, as mentioned herein, is preferably interferon-a and/or interferon-β.
The infection of a cell by the PRRS virus according to the invention in particular includes attachment of the virus to the cell, entry of the virus into the cell, disassembly of the virion, and preferably replication and transcription of the viral genome, expression of viral proteins, and assembly and release of new infectious viral particles.
The cell, as mentioned herein, is a primary or secondary susceptile cell, preferably a mammalian cell, in particular a porcine or a simian cell, more preferably said cell is a porcine macrophage or a MA-104 cell or a MARC-145 cell or a Vero cell.
It is further understood that the PRRS virus according to the invention is able to induce in vitro and/or in vivo the interferon type I production and secretion by a cell infected by said virus.
Preferably, the PRRS virus according to the invention is a live PRRS virus and/or a modified- live PRRS virus and/or an attenuated PRRS virus. The term "attenuated PRRS virus", as described herein, is in particular directed to a PRRS virus which is attenuated in vitro and/or in vivo, more particular in susceptible cell lines and/or the host.
The term "host", as used herein, is in particular directed to animals infectable with PRRS virus, in particular swine, more particular pigs, such as domestic pigs. As mentioned herein, "attenuated" particularly relates to a reduced virulence of a pathogen, in particular of a wild type PRRS virus, wherein "virulence" is understood to be the degree of pathogenicity, and wherein "pathogenicity" is directed to the ability of the pathogen to produce clinical symptoms in the host, such as elevated body temperature.
In particular preferably, the PRRS virus according to the invention has or shows increased sensitivity to type I INF when compared to wild type PRRSV, wherein the term "sensitivity to type I INF" is understood as reduced viral infectivity when IFN is present, preferably in a sufficient amount for significantly reducing viral infectivity of wild type PRRSV, in the medium surrounding the virus at the time of infection.
The term "wild type PRRS virus" or "wild type PRRSV", respectively, as used herein, is in particular directed to an infectious pathogenic PRRS virus, which is particularly capable of causing PRRS in swine. In one particular preferred embodiment, the term "wild type PRRS virus" is directed to a PRRS virus whose genome comprises a RNA sequence or consists of a RNA polynucleotide, wherein said RNA sequence or RNA polynucleotide is a RNA copy of a polynucleotide selected from the group consisting of SEQ ID NO:13, SEQ ID NO:14 and SEQ ID NO:15. In one embodiment, the PRRS virus according to the invention is a PRRS virus mutant, in particular comprising, in comparison with the genome of a wild type PRRSV strain, a mutation in a gene encoding a protein of said virus.
In a preferred embodiment, the PRRS virus according to the invention comprises a mutation in the gene encoding the nspl protein of said virus. Thus, the invention preferably concerns a PRRS virus which is able to induce the interferon type I production and secretion by an infected cell as a result of a mutation in the gene encoding the nspl protein, wherein said mutation is preferably a mutation as mentioned hereinafter.
Hence, the invention particulalry concerns a PRRS virus comprising, in comparison with the genome of a wild type strain, a mutation in the gene encoding the nspl protein. Accordingly, the mutation as described herein is preferably a mutation in comparison with the sequence of the corresponding wild type gene encoding the nspl protein.
The invention is thus preferably directed to a PRRS virus, in particular a wild type PRRS virus, wherein a mutation, preferably a mutation as mentioned hereinafter, has been implemented in the genome of said virus resulting in a non-natural nspl protein or in the lack of nspl protein of said virus.
In the context of the invention it is understood, that the mutation as described herein may be implemented to any PRRS virus, such as to a PRRS virus selected from the group consisting of wild type PRRS virus, attenuated PRRS virus, modified-life PRRS virus, PRRS virus mutant and combinations thereof. In one example, the mutation as described herein may be implemented to an attenuated and/or live PRRS virus, for example to a PRRS virus selected from the group of the PRRS virus strains that have been deposited on 27 October 2004 with the European Collection of Cell Cultures (ECACC), Porton Down, Salisbury, Wiltshire, SP4 OJG, Great Britain, under the Accession Numbers ECACC 04102703, ECACC 04102702, and ECACC 04102704. Hence, the mutation as described herein can be combined with one or more other mutations, preferably one ore more other attenuating mutations, in a PRRS virus.
In the context of the invention, the term "PPRRS virus" is in particular equivalent with "PRRS virus strain".
The term "mutation" in the context of the invention is understood as a change in a genomic sequence, in particular in the RNA sequence of a PRRS virus. Since viruses that use RNA as their genetic material have rapid mutation rates, the term "mutation", as mentioned herein, is particularly directed to a genetically engineered change in a genomic sequence, such as by site directed mutagenesis, which in particular results in a virus growing to titers significantly lower than wild type PRRS virus in interferon competent cells and/or in the infected host, when propagated under the same conditions. Moreover, in another preferred embodiment the mutation described herein can also be caused by natural mutation and subsequent isolation of the PRRS virus according to the invention, wherein said isolated virus includes the mutation described herein.
Preferably, the mutation, as described herein, comprises or consists of one or more point mutations and/or one or more genomic deletions and/or one or more insertions.
The term "nspl protein", as used herein, is directed to the PRRSV nonstructural protein 1.
The nspl protein is preferably a polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:1 and SEQ ID NO:2 if the PRRS virus according to the invention is a genotype I PRRS virus, or the nspl protein is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide set forth in SEQ ID NO:3 if the PRRS virus according to the invention is a genotype II PRRS virus.
In one aspect, the PRRS virus according to the invention preferably also may comprise a mutation in the gene encoding the nspl protein selected from the group consisting of SEQ ID NOs 1-3.
It is hence understood that the PRRS virus according to the invention is a genotype I PRRS virus or a genotype II PRRS virus. It is further understood that the terms "genotype I" and "genotype M" are equivalent to the terms "genotype 1" and "genotype 2" or to the terms "type 1" and "type 2", as frequently used in the literature in the context of PRRSV.
Sequence identity in the context of the invention is understood as being based on pairwise determined similarity between nucleotide or protein sequences. The determination of percent identity between two sequences is preferably accomplished using a mathematical algorithm, in particular the well-known Smith-Waterman algorithm (Smith and Waterman, M. S. (1981 ) J Mol Biol, 147(1 ):195-197). For purposes of the present invention, percent sequence identity of an amino acid sequence is determined using the Smith-Waterman homology search algorithm using an affine 6 gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix 62. The Smith-Waterman homology search algorithm is taught in Smith and Waterman (1981 ) Adv. Appl. Math 2:482-489, herein incorporated by reference. A variant may, for example, differ from the reference nspl , nspi a, ηερΐ β or NTD molecule by as few as 1 to 15 amino acid residues, as few as 1 to 10 amino acid residues, such as 6- 10, as few as 5, as few as 4, 3, 2, or even 1 amino acid residue. Alternatively, percent identity of a nucleotide sequence is determined using the Smith-Waterman homology search algorithm using a gap open penalty of 25 and a gap extension penalty of 5. Such a determination of sequence identity can be performed using, for example, the DeCypher Hardware Accelerator from TimeLogic Version G, or the sequence identity is determined with the software CLC MAIN WORKBENCH 4.1.1 (CLC BIO).
The term "having 100% sequence identity", as used herein, is understood to be equivalent to the term "being identical". ln another preferred embodiment, the mutation in the PRRS virus according to the invention comprises or consists of one or more point mutations and/or one or more genomic deletions and/or one or more insertions.
In a further preferred embodiment, the PRRS virus according to the invention comprises a mutation in the gene sequences encoding the nspl a subunit of the nspl protein and/or the nspl β subunit of the nspl protein of said virus.
The term "nspla", as mentioned herein, is thus directed to the PRRSV nspla subunit of the nspl protein, and the term "ηερΐ β", as used herein, is hence directed to the PRRSV ηερΐ β subunit of the nspl protein.
The nspl a is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:4 and SEQ ID NO:5 if the PRRS virus according to the invention is a genotype I PRRS virus, or the nspla is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide set forth in SEQ ID NO:6 if the PRRS virus according to the invention is a genotype II PRRS virus.
The nsp1 is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:7 and SEQ ID NO:8 if the PRRS virus according to the invention is a genotype I PRRS virus, or the ηερΐ β is preferably a polypeptide having at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide set forth in SEQ ID NO:9 if the PRRS virus according to the invention is a genotype II PRRS virus.
In a particular preferred embodiment, the PRRS virus according to the invention comprises a mutation in the gene sequence encoding the N-terminal domain (NTD) of the ηερΐ β of said virus. The term "NTD", as mentioned herein, is thus directed to the N-terminal domain of the PRRSV ηερΐ β subunit.
The NTD is preferably a polypeptide or has a polypeptide sequence, respectively, having at least 60%, particularly at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide or a polypeptide sequence, respectively, selected from the group consisting of SEQ ID NO:10 and SEQ ID NO:1 1 if the PRRS virus according to the invention is a genotype I PRRS virus, or the NTD is preferably a polypeptide or has polypeptide sequence, respectively, having at least 60%, particularly at least 70%, preferably at least 80 %, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide or the polypeptide sequence, respectively, set forth in SEQ ID NO:12 if the PRRS virus according to the invention is a genotype II PRRS virus.
In a preferred embodiment, the NTD comprising the mutation has the sequence selected from the group consisting of the sequences
SXXYXXXXXVXFXDXXXXGXXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXXYXXXXXVXFXDXXXXGX WXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXXYXXXXXVXFXDXXXXXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXXYXXXXXVXFXDXXXXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXXYXXXXXVXFXDXXXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXX YXXXXX VX FX DXXMWXXXSXXSXXL EX LPXXLXXXXXXLX RS ,
as set forth in SEQ ID NOs:36-41 , if the PRRS virus according to the invention is a genotype I PRRS virus,
or the NTD comprising the mutation preferably has the sequence selected from the group consisting of the sequences
AXVYDIGXXAVMXVAXXXXSWAGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYDIGXXAVMXVAXXXXSWGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYDIGXXAVMXVAXXXXSGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYDIGXXAVMXVAXXXXGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYDIGXXAVMXVAXXXGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYDIGXXAVMXVAXXGGXXXXFXXXXXXLXLXAXXXXXS,
as set forth in SEQ ID NOs:42-47, if the PRRS virus according to the invention is a genotype II PRRS virus. Thus, the PRRS virus according to the invention preferably comprises a gene sequence encoding a NTD selected from the sequences
SXXYXXXXXVXFXDXXXXGXX WXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXXYXXXXXVXFXDXXXXGXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXXYXXXXXVXFXDXXXXXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXXYXXXXXVXFXDXXXXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS,
SXXYXXXXXVXFXDXXXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS, SXXYXXXXXVXFXDXXMWXXXSXXSXXLEXLPXXLXXXXXXLXRS,
as set forth in SEQ ID NOs:36-41 , if the PRRS virus according to the invention is a genotype I PRRS virus,
or the PRRS virus according to the invention preferably comprises a gene sequence encoding a NTD selected from the sequences
AXVYD IGXXAV XVAXXXXSWAGGXXXXFXXXXXXLXLXAXXXXXS ,
AXVYDIGXXAVMXVAXXXXSWGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYDIGXXAVMXVAXXXXSGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYDIGXXAVMXVAXXXXGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYDIGXXAVMXVAXXXGGXXXXFXXXXXXLXLXAXXXXXS,
AXVYD IGXXAVMXVAXXGGXXXX FXXXXXXLXLXAXXXXXS ,
as set forth in SEQ ID NOs:42-47, if the PRRS virus according to the invention is a genotype II PRRS virus. In one exemplary embodiment, the NTD comprising the mutation may, for instance, have the sequence selected from the group consisting of the sequences
SSVYRWKKFWFTDSSXNGRMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSSXNGMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSSXNMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSSXMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSSMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSMMWTPESDDSAXLEXLPPELERQVEILIRS,
as set forth in SEQ ID NOs:48-53, if the PRRS virus according to the invention is a genotype I PRRS virus,
or the NTD comprising the mutation may, for instance, have the sequence selected from the group consisting of the sequences
ATV Y D I GXXAVM YVAXX K VS W AGGX EVKFEXVPXE L KLXA N R LXTS ,
ATVYDIGXXAVMYVAXXKVSWGGXEVKFEXVPXELKLXANRLXTS,
ATVYDIGXXAVMYVAXXKVSGGXEVKFEXVPXELKLXANRLXTS,
ATVYDIGXXAVMYVAXXKVGGXEVKFEXVPXELKLXANRLXTS,
ATVYDIGXXAVMYVAXXKGGXEVKFEXVPXELKLXANRLXTS,
ATVYDIGXXAVMYVAXXGGXEVKFEXVPXELKLXANRLXTS,
as set forth in SEQ ID NOs:54-59, if the PRRS virus according to the invention is a genotype II PRRS virus. Thus, in one exemplary embodiment, the PRRS virus according to the invention may, for instance, comprise a gene sequence encoding a NTD selected from the sequences
SSVYRWKKFWFTDSSXNGRMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSSXNGMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSSXNMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSSXMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSSMMWTPESDDSAXLEXLPPELERQVEILIRS,
SSVYRWKKFWFTDSMMWTPESDDSAXLEXLPPELERQVEILIRS,
as set forth in SEQ ID NOs:48-53, if the PRRS virus according to the invention is a genotype I PRRS virus,
or the PRRS virus according to the invention may, for instance, comprise a gene sequence encoding a NTD selected from the sequences
ATVYDIGXXAVMYVAXXKVSWAGGXEVKFEXVPXELKLXANRLXTS,
ATVYDIGXXAVMYVAXXKVSWGGXEVKFEXVPXELKLXANRLXTS,
ATVYDIGXXAVMYVAXXKVSGGXEVKFEXVPXELKLXANRLXTS,
ATVYDIGXXAVMYVAXXKVGGXEVKFEXVPXELKLXANRLXTS,
ATVYD IGXXAVM YVAXXKGGXEVKFEXVPXELKLXAN RLXTS ,
ATVYDIGXXAVMYVAXXGGXEVKFEXVPXELKLXANRLXTS,
as set forth in SEQ ID NOs:54-59, if the PRRS virus according to the invention is a genotype II PRRS virus.
It is thus understood that a genotype I PRRS virus, as mentioned herein, is in particular a virus whose genome comprises a gene sequence coding for a polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:7 and SEQ ID NO:8, or is in particular a virus whose genome comprises a gene sequence coding for a polypeptide having at least 60%, particularly at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO: 10 and SEQ ID NO:11.
Preferably, the genotype I PRRS virus, as mentioned herein, comprises or consists of a RNA polynucleotide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a RNA polynucleotide complementary to a polynucleotide selected from the group consisting of SEQ ID NO:13 and SEQ ID NO:14.
It is further understood that a genotype II PRRS virus, as mentioned herein, is in particular a virus whose genome comprises a gene sequence coding for a polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NO:3, SEQ ID NO:6 and SEQ ID NO:9, or is in particular a virus whose genome comprises a gene sequence coding for a polypeptide having at least 60%, particularly at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with the polypeptide set forth in SEQ ID NO:12.
Preferably, the genotype II PRRS virus, as mentioned herein, comprises or consists of a RNA polynucleotide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a RNA polynucleotide complementary to the polynucleotide set forth in SEQ ID NO: 15.
In the following, the single letter code for amino acids is used for amino acid sequences, where additionally X signifies any genetically encoded amino acid residue.
Within the context of the invention it is in particular understood that the N-terminal domain (NTD) of ηερΐ β starts with the N-terminus of ηερΐ β, preferably starts with the amino acid sequence SXXY if the PRRS virus is a genotype I PRRS virus or with the amino acid sequence AXVY if the PRRS virus is a genotype II PRRS virus, and the NTD ends with the serine residue (S) within the amino sequence SFP of the ηερΐ β. Regarding the NTD domain of a genotype II PRRS virus it is in particular referred to the publication Xue et al. 2010.
Said amino acid sequences SXXY or AXVY and SFP are especially conserved in the ηερΐ β of wild type PRRS viruses and are thus preferably used amino acid sequence motifs for defining the NTD domain according to the invention.
In a particular preferred embodiment, the PRRS virus according to the invention thus comprises a mutation in the gene sequence coding for the NTD, wherein the NTD preferably starts with motif SXXY for PRRS virus type 1 strains or with motif AXVY for PRRS virus type 2 strains and ends with the serine residue (S) within the conserved SFP motif of the ηερΐ β subunit. According to the invention, it is in particular preferred if said mutation is located within the part of the gene sequence coding for the NTD, where the amino acid stretches folding into β sheet secondary structures are encoded.
In one preferred aspect, the mutation, as described herein, thus comprises a deletion or replacement of a nucleotide triplett coding for an amino acid residue located within the first 24 N-terminal amino acid residues of the NTD sequence. In particular, it is preferred if the mutation described herein comprises or consistis of a deletion or replacement of 2 to 7 or more, preferably consecutive, nucleotide tripletts each coding for an amino acid residue located within the first 24 N-terminal amino acid residues of the NTD sequence. In another preferred aspect, the mutation, as mentioned herein, comprises the deletion or replacement of a nucleotide triplett coding for an amino acid residue with a charged, preferably positively charged, side chain, in particular coding for an arginine residue.
Examplarily, the mutation mentioned herein preferably consists of a deletion of 2, 3, 4, 5, 6 or 7 consecutive nucleotide tripletts coding for the respective number (2, 3, 4, 5, 6 or 7) of consecutive amino acid residues located within the first 24 N-terminal amino acid residues of the NTD sequence, wherein this mutation comprises the deletion of a nucleotide triplett coding for an amino acid residue with a charged, preferably positively charged, side chain, in particular coding for an arginine residue.
The mutation, as described herein, preferably comprises or consists of a deletion or replacement of the nucleotide triplett coding for the first arginine residue (R) located at least 21 amino acid residues in C-terminal direction from the N-terminal amino acid residue of the ηερΐ β NTD and/or the mutation, as mentioned herein, comprises a deletion or replacement of the nucleotide triplett coding for the arginine residue (R) of the nspl amino acid sequence RXMW if the PRRS virus is a genotype I PRRS virus or with the amino acid sequence RGG of said virus if the PRRS virus is a genotype II PRRS virus and/or the mutation, as mentioned herein, comprises or consists of a deletion or replacement of the nucleotide triplett coding for the arginine residue (R) of the amino acid sequence RMM or RGG of the nspl protein. According to the invention, the phrase "replacement of a nucleotide tiplett" is understood as being a replacement of a nucleotide triplett of the wild type sequence by another nucleotide triplett coding for a different amino acid residue than the wild type sequence. The phrase "deletion of nucleotide triplett" in the context of the invention is directed to a deletion of a nucleotide triplett resulting in the deletion of an amino aicd residue in comparison with the wild type sequence.
In particular, said mutation further comprises or additionally consists of a deletion or replacement of one nucleotide triplett coding for the amino acid residue flanking said arginine residue (R) in N-terminal direction, wherein said mutation preferably comprises or consists of a deletion or replacement of two consecutive nucleotide tripletts coding for the amino acid residues PR. More particular, said mutation further comprises or additionally consists of a deletion or replacement of two, three, four, five, six, seven or more consecutive nucleotide tripletts coding for two, three, four, five, six, seven or more amino acid residues flanking said arginine residue (R) in N-terminal direction. Preferably, said mutation comprises or consists of a deletion or replacement of three consecutive nucleotide tripletts coding for the amino acid sequence RXR or APR of the ηερΐ β, or wherein said mutation comprises a deletion or replacement of four consecutive nucleotide tripletts coding for the amino acid residues GRXR or WAPR of the ηερΐ β. As result, the PRRS virus according to the invention comprises in one embodiment a gene coding for a nspl protein selected from the group consisting of SEQ ID NO: 16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO: 19 and SEQ ID NO:20, or comprises in another embodiment a gene sequence coding for a ηερΐ β selected from the group consisiting of SEQ ID NO:21 , SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24 and SEQ ID NO:25, or comprises in a further embodiment a gene sequence coding for a NTD selected from the group consisting of SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29 and SEQ ID NO:30.
In yet another preferred embodiment, the mutation, as described herein comprises or consists of a deletion of the gene sequence encoding the (whole) nspl protein, encoding the (whole) nspla subunit, encoding the (whole) ηερΐ β subunit, or encoding a part, preferably at least seven consecutive amino acid residues, of or the (whole) NTD domain of the ηερΐ β subunit.
Yet another aspect of the invention is directed to a polynucleotide comprising or consisting of the genome of the PPRS virus according to the invention.
Further, the invention is also directed to a virus particle, wherein said virus particle comprises a polynucleotide which comprises or consists of the genome of the PPRS virus according to the invention or which comprises or consists of a DNA copy of the PPRS virus according to the invention. Still further, the present invention provides a DNA-Vector comprising a copy of, or a cDNA sequence complementary to, respectively, a polynucleotide which comprises or consists of the genome of the PPRS virus according to the invention.
In one exemplary and non-limiting example the DNA vector, as mentioned herein, comprises a polynucleotide sequence selected from the group consisting of SEQ ID NO:31 , SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34 and SEQ ID NO:35.
Also, the present invention provides a cell comprising a polynucleotide, which comprises or consists of the genome of the PPRS virus according to the invention or which comprises or consists of a DNA copy of the PPRS virus according to the invention. In one exemplary and non-limiting example the PRRS virus according to the invention, or the genome of the PRRS virus according to the invention, respectively, comprises a RNA sequence or consists of a RNA polynucleotide, wherein said RNA sequence or RNA polynucleotide is a RNA copy of a polynucleotide selected from the group consisting of SEQ ID NO:31 , SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34 and SEQ ID NO:35.
Another aspect of the invention concerns the PRRS virus according to the invention for use as vaccine or medicament, in particular for use in the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome preferably in swine. The present invention further provides a composition containing the PRRS virus according to the invention for use as a vaccine or as a medicament, in particular for use in the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome preferably in swine.
In a preferred aspect, said vaccine or medicament is administered in two doses or preferebly in one dose to an animal in need thereof. By the term "prophylaxis or therapy", as mentioned herein, it is meant that the prophylaxis is to administer a drug, in particular a vaccine, before exposure to PRRSV and the treatment is to administer a drug, in particular a medicament, after infection with PRRSV or onset of PRRS. In particular, the vaccine, as mentioned herein, is a live vaccine and/or a modified live vaccine and/or an attenuated live or attenuated modified live vaccine.
The strains of the PRRS virus according to the invention can be grown and harvested by methods known in the art, e.g. by propagating in suitable cells like the simian cell line MA- 104, Vera cells, or porcine alveolar macrophages. Preferably, vaccines according to the present invention are modified live vaccines comprising one or more of these strains alive in a suitable carrier, but inactivated virus may also be used to prepare killed vaccine (KV). Modified live vaccines (MLV) are typically formulated to allow administration of 101 to 107 viral particles per dose, preferably 103 to 106 particles per dose, more preferably 104 to 106 particles per dose (4.0-6.0 log 0 TCID50). KV may be formulated based on a pre-inactivation titre of 103 to 1010 viral particles per dose. The vaccine may comprise a pharmaceutically acceptable carrier, for example a physiological salt-solution. The vaccine may or may not comprise an adjuvant. An example of a suitable adjuvant is a-tocopherol acetate which can be obtained under the trade name Diluvac Forte®. Alternatively, for example alum based adjuvants may be used.
A further aspect of the invention is thus directed to the use of the PRRS virus according to the invention for the preparation of a vaccine or medicament for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome preferably in swine. A vaccine according to the present invention may be presented in form of a freeze-dried preparation of the live virus, to be reconstituted with a solvent, to result in a solution for injection. The solvent may e.g. be water, physiological saline, or buffer, or an adjuvanting solvent. The solvent may contain adjuvants, for example a-tocopherol acetate. The reconstituted vaccine may then be injected into a pig, for example as an intramuscular or intradermal injection into the neck. For intramuscular injection, a volume of 2 ml may be applied, for an intradermal injection it is typically 0.2 ml. In a further aspect, the present invention therefore is a product, comprising in separate containers a freeze-dried composition of the virus, and a solvent for reconstitution, and optionally further containing a leaflet or label comprising instructions of use. Yet another aspect of the invention thus concerns a method for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome comprising administering the PRRS virus according to the invention to an animal, preferably to a swine, more preferably to a pig, in particular preferably to a piglet or a sow. A vaccine according to the present invention may not only comprise the PRRS virus according to the invention as antigen, but may include further components active against PRRS or other porcine viral or bacterial diseases, like porcine circovirus, classical swine fever virus or mycoplasma hyorhinis. Therefore, the invention further relates to a vaccine as described, characterised in that it contains at least one further antigen active against a porcine disease which is not PRRS.
Still another aspect of the invention thus concerns a medicament or vaccine comprising the PRRS virus according to the invention. Still a further aspect of the invention is directed to a method of preparing a vaccine or medicament, in particular for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome preferably in swine, said method comprising the cultivation of the PRRS virus according to the invention in cell culture.
Moreover, the invention comprises a method of producing the PRRS virus according to the invention, said method comprising the mutagenesis of the gene coding for the nspl protein in the genome of a PRRS virus, and wherein the mutation, as described herein, is in particular introduced by genetic engineering in the genome of said virus, preferably by site directed mutagenesis.
Preferably, for any of the aforementioned methods, the above-mentioned DNA-vector is used and/or said methods comprise the detection of interferon type I, in particular interferon type I secretion, and/or of mRNA coding for interferon type I. Said detection in particular comprises the detection of interferon type I and/or of mRNA coding for interferon type I by a bioassay, wherein said bioassay is preferably selected from the group consisting of ELISA, PCR, GLISA, IFA, biosensoric measurement, Surface Plasmon Resonance (SPR) measurement, selective media , lateral flow, biochip measurement, immunomagnetic separation , electrochemiluminescence, chromogenic media, immunodiffusion, DNA hybridization, staining, colorimetric detection, luminescence, and combinations thereof.
In yet a further aspect, the invention provides an immunogenic composition containing the PRRS virus according to the invention. As used herein, the term "immunogenic composition" in particular refers to a composition that will elicit an immune response in an animal that has been exposed to the composition. An immune response may include induction of antibodies and/or induction of a T-cell response. Usually, an "immune response" includes but is not limited to one or more of the following effects: the production or activation of antibodies, B cells, helper T cells, suppressor T cells, and/or cytotoxic T cells, directed specifically to an antigen or antigens included in the composition or vaccine of interest. Preferably, the host will display either a therapeutic or a protective immunological (memory) response such that resistance to new infection will be enhanced and/or the clinical severity of the disease reduced. Such protection will be demonstrated by either a reduction in number or severity of, or lack of one or more of the symptoms associated with the infection of the pathogen, in the delay of onset of viremia, in a reduced viral persistence, in a reduction of the overall viral load and/or in a reduction of viral excretion.
Thus, an "immune response" in particular means but is not limited to the development in a subset of a cellular and/or antibody-mediated immune response to the composition or vaccine of interest.
In one aspect, the immunogenic composition of the invention is preferably a vaccine or medicament.
As used herein, the term "viremia" is particularly understood as a condition in which PRRS virus particles circulate in the bloodstream of an animal.
The term "animal", as mentioned herein, is in particular directed to swine, more particular to a pig, preferably a domestic pig.
In a preferred aspect, the immunogenic composition of the invention comprises an amount of 101 to 107 viral particles of the PRRS virus according to the invention per dose, preferably 103 to 106 particles per dose, more preferably 104 to 106 particles per dose. In another preferred aspect, the immunogenic composition of the invention comprises an amount of the PRRS virus according to the invention which is equivalent to a virus titre of at least about 103 TCID50/mL per dose, preferably between 103 to 105 TCID50/mL per dose
In yet another preferred aspect, the immunogenic composition of the invention further contains one or more pharmaceutically acceptable carriers or excipients. Said one or more pharmaceutically acceptable carriers or excipients are preferably selected from the group consisting of solvents, dispersion media, adjuvants, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, and adsorption delaying agents.
The present invention is further directed to the immunogenic composition of the invention for use in a method for
- inducing an immune response against PRRSV, or
- the prevention or reduction of PRRS, or
- the prevention or reduction of PRRSV viremia and/or
- preventing or reducing clinical symptoms, in particular the elevated body temperature, associated with PRRSV infection, or
- the prevention or reduction of the elevated body temperature associated with the administration of an attenuated PRRSV vaccine to an animal.
As used herein, the term "inducing an immune response" to an antigen or composition is the development of a humoral and/or cellular immune response in an animal to an antigen present in the composition of interest.
The term "prevention" or "reduction" or "preventing" or "reducing", respectively, as used herein, means, but is not limited to a process which includes the administration of a PRRSV antigen, namely of the PRRSV according to the invention which is included in the composition of the invention, to an animal, wherein said PRRSV antigen, when administered to said animal elicits or is able to elicit an immune response in said animal against PRRSV. Altogether, such treatment results in reduction of the clinical symptoms of PRRS or of symptoms associated with PRRSV infection, respectively. More specifically, the term "prevention" or "preventing, as used herein, means generally a process of prophylaxis in which an animal is exposed to the immunogenic composition of the present invention prior to the induction or onset of the disease process (PRRS).
Herein, "reduction of PRRS" or "reduction of clinical symptoms associated with PRRSV infection" means, but is not limited to, reducing the number of infected subjects in a group, reducing or eliminating the number of subjects exhibiting clinical symptoms of infection, or reducing the severity of any clinical symptoms that are present in the subjects, in comparison to wild-type infection. For example, it should refer to any reduction of pathogen load, pathogen shedding, reduction in pathogen transmission, or reduction of any clinical sign symptomatic of PRRSV infection, in particular of elevated body temperature. Preferably these clinical symptoms are reduced in subjects receiving the composition of the present invention by at least 10% in comparison to subjects not receiving the composition and may become infected. More preferably, clinical symptoms are reduced in subjects receiving the composition of the present invention by at least 20%, preferably by at least 30%, more preferably by at least 40%, and even more preferably by at least 50%.
Also, the elevated body temperature usually associated with the administration of an attenuated PRRSV vaccine to an animal is reduced in subjects receiving the composition of the present invention by at least 10% in comparison to subjects receiving a conventional attenuated PRRSV vaccine. More preferably, the elevated body temperature usually associated with the administration of an attenuated PRRSV vaccine is reduced in subjects receiving the composition of the present invention by at least 20%, preferably by at least 30%, more preferably by at least 40%, and even more preferably by at least 50%.
Hence, a further advantage of the PRRS virus according to the invention is in particular the effect that its administration to an animal, e.g. for the prophylaxis or treatment of PRRS, results in a reduced increase of body temperature in the animal in comparision with a conventional attenuated PPRSV vaccine.
The term "conventional attenuated PRRSV vaccine", as mentioned herein, is in particular directed to any attenuated PRRSV useful as a vaccine, wherein said PRRSV does not have the characteristic features of the PRRS virus of the present invention, in particular is not able to induce the interferon type I production and secretion by a cell infected by said PRRSV.
The term "subject", as mentioned herein, in particular relates to an animal.
The term "body temperature", as used herein, in particular refers to the approximate average normal, internal temperature of an animal, for example about 38.5-39°C in pigs, whereas the body temperature associated with a PRRSV infection may be elevated up to 41 °C in pigs.
The term "reduction of PRRSV viremia" means, but is not limited to, the reduction of PRRS virus entering the bloodstream of an animal, wherein the viremia level, i.e. the number of PRRSV RNA copies per ml_ of blood serum or the number of plaque forming colonies per deciliter of blood serum, is reduced in the blood serum of subjects receiving the composition of the present invention by at least 50% in comparison to subjects not receiving the composition and may become infected. More preferably the viremia level is reduced in subjects receiving the composition of the present invention by at least 90%, preferably by at least 99.9%, more preferably by at least 99.99%, and even more preferably by at least 99.999%.
The present invention further relates to the use of the PRRS virus according to the invention or of the immunogenic composition of the invention for the preparation of a vaccine or medicament for
- inducing an immune response against PRRSV, or
- treating or preventing PRRS, or
- preventing or reducing PRRSV viremia and/or
- preventing or reducing clinical symptoms, in particular the elevated body temperature, associated with PRRSV infection, or
- the prevention or reduction of the elevated body temperature associated with the administration of an attenuated PRRSV vaccine to an animal.
In particular, it is preferred if the vaccine or medicament, as mentioned herein, is to be administered in two doses or preferably in a single dose to an animal.
The present invention also provides a method of preparing an immunogenic composition, preferably a vaccine or medicament, more preferably a vaccine or medicament for
- inducing an immune response against PRRSV, or
- treating or preventing PRRS, or - preventing or reducing PRRSV viremia and/or
- preventing or reducing clinical symptoms, in particular the elevated body temperature, associated with PRRSV infection, or
- the prevention or reduction of the elevated body temperature associated with the administration of an attenuated PRRSV vaccine to an animal, wherein said method comprises the step of mixing the PRRS virus according to the invention with one or more pharmaceutically acceptable carriers or excipients, and wherein said one or more pharmaceutically acceptable carriers or excipients are preferably selected from the group consisting of solvents, dispersion media, adjuvants, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, and adsorption delaying agents.
In another aspect, the present invention further provides a method for
- inducing an immune response against PRRSV, or
- treating or preventing PRRS, or
- preventing or reducing PRRSV viremia and/or
- preventing or reducing clinical symptoms, in particular the elevated body temperature, associated with PRRSV infection, or
- the prevention or reduction of the elevated body temperature associated with the administration of an attenuated PRRSV vaccine to an animal,
wherein said method comprises the step of administering the immunogenic composition of the invention to an animal in need thereof, and wherein preferably the immunogenic composition and/or the vaccine is administered in two doses or, more preferably, in a single dose.
EXAMPLES
In the examples five viable, genetically designed PRRSV mutant strains are described which are based on infectious EU PRRSV cDNA clone LoN94-13. These strains, delta nspl IX-10, delta nspl XVII-1 , delta nspl XVIII-12, delta nspl XIX-2 and delta nspl XX-9 (henceforth refered to as vaccine candidates), harbor genomic deletions of two, three, four, five, or six codons in their predicted nspl genes, respectively, resulting in deletions of two (motif P21 R22), three (motif R20P21 R22), four (motif G19R20P21 R22), five (motif N18G19R20P21 R22), or six (motif (P17N18G19R20P21 R22) amino acids in their predicted ηερΐ β proteins, respectively (Figure 1 ).
Based on sequence alignments of parental strain LoN94-13 with PRRSV US and EU reference strains VR-2332 and Lelystad virus as well as with strain GD-XH, the deletions are located in the predicted ηερΐ β portion of nspl (Figure 2). In more detail, the deletion site for all vaccine candidates is located in the N-terminal domain (NTD) of ηερΐ β and overlaps with aminoacids P23R24 of GD-XH ηερΐ β; (Figure 2).
After transfection of synthetic transcripts of the vaccine candidates into BHK21 cells and transfer of cell culture supernatant from transfected BHK21 cells onto PRRSV-susceptible MA104 cells, plaque formation typical for PRRSV infection occurred (data not shown). PRRSV-specificity and viability for each of the vaccine strains then was demonstrated by subsequent cell culture passages on MA104 cells and PRRSV-specific immunofluorescence using monoclonal antibody SDOW17 (Rural Technologies); data not shown.
After endpoint dilution and generation of virus stocks each derived from material of a single virus plaque, virus titers of the obtained virus stocks were determined for each vaccine candidate by serial virus titrations on 96-well plates containing MA104 cells followed by PRRSV-specific immunofluorescence analyses six to seven days post infection. Unlike experience with titrations of virus stocks from parental PRRSV LoN94-13 (data not shown), the first serial dilutions of vaccine candidates delta nspl XVII-1 and delta nspl XVIII-12 did not demonstrate a cytopathic effect and virus plaque formation, while at higher dilutions of the virus stocks a cytopathic effect was detectable. Moreover, when respective titrations were investigated by immunofluorescence, cell culture wells of the first serial dilutions were negative for PRRSV infection for both vaccine candidates, while wells infected with higher dilutions of the virus stocks showed PRRSV-specific immunofluorescence, respectively (Figure 3).
To determine whether the prepared vaccine candidate virus stocks contained type I IFN, a commercial ELISA specific for human IFN (Invitrogen) was used. MA104 cells are epithelial Green Monkey kidney cells. According to the ELISA manufacturer, this Invitrogen ELISA is also suited for the detection of primate IFN other than human. For each vaccine candidate's virus stock, 100μΙ served as assay input, while a virus stock from parental strain LoN94-13, cell culture medium , and medium from noninfected cells served as controls. For quantification of the obtained results, a calibration curve was included using a positive control of the ELISA manufacturer. All samples were measured in duplicates. Unlike the negative controls, virus stocks of the vaccine candidates contained considerable levels of type I IFN, while the virus stock of the parental virus showed IFN levels as low as the negative controls (Figure 4).
To confirm the results obtained and to assess kinetics of type I IFN production in cells infected with the vaccine candidates, a time course experiment was performed using MA104 cells infected at a multiplicity of infection (MOI) of 0.001 , respectively. Parental strain LoN94- 13 served as negative control. While there were only very little and unaltered levels of type I IFN near background detectable for infection with parental strain LoN94-13, vaccine candidates delta nspl XVII-1 and delta nspl XVIII-12 induced considerable and increasing amounts of up to about 18 I.U. IFN per 25 μΙ sample volume from two days post infection on (Figure 5). It was experimentally assessed whether vaccine candidates containing genomic deletions in nspl demonstrate an increased sensitivity to type I IFN (Figure 6). 5x105 MA104 cells were seeded into a well of a six-well plate and were either not infected (n.inf.) or infected with 800 infectious virus particles of one of the virus strains given on top, repectively. Cells then were either inoculated with 120 I.U. human IFN (+IFN , bottom row), respectively, or not (-IFN , top row). Three days post infection, immunofluorescence analysis specific for the PRRSV capsid protein was performed using monoclonal antibody SDOW17 (Rural Technologies). The total numbers of foci of PRRSV-infected cells per well are given below, respectively. This experiment demonstrated that inoculation with type I IFN reduced the number of PRRSV infection events in cells after inoculation with a defined number of infectious virus particles, reflecting reduced viral infectivity of PRRSV when IFN was added. This reduction was 80-fold for wild type virus LoN94-13 (Figure 6). In addition, for infection with wild type virus, foci of infected cells were smaller than in the well not inoculated with IFN (Figure 6). However, for vaccine strains delta nspl XVII-1 and delta nspl XVIII-12, viral infectivity was reduced to zero when INF was added (Figure 6).
Thus, these vaccine candidates not only induce production of type I IFN in infected cells (Figures 4 and 5), but also demonstrate increased sensitivity to type I INF when compared to wild type PRRSV (Figure 6). This is reflected by their dramatically reduced viral infectivity when IFN is present.
Interestingly, the cells infected for the time course experiment summarized in Figure 5 not only produced considerable amounts of ΙΡΝβ, but at the end of the experiment at six days post infection, cells infected with vaccine candidates delta nspl XVII-1 and delta nspl XVIII- 12 showed signs of recovery from usually lytical PRRSV infection. While cells infected with parental strain LoN94-13 were fully lysed, cells infected with the vaccine candidates grew in a partially (delta nspl XVIII-12) or completely intact monolayer (delta nspl XVII-1 ). For the latter, only weak signs of a PRRSV-induced cytopathic effect were still detectable. Thus, the interferon production of infected cells together with the observed sensitivity of vaccine candidates to type I IFN correlated with partial or almost complete recovery of infected cells over time. It is reasonable to expect that type I IFN induction by the vaccine candidates together with their increased sensitivity to type I IFN will contribute to a significantly attenuated viral phenotype in the natural host. In particular, expected features of the vaccine candidates' attenuation in pigs include stimulation of the innate and specific immunity, both humoral and cellular, and less shedding and/or shortened viremia of the vaccine viruses. To assess whether the PRRSV vaccine candidates are attenuated in the host, an animal experiment in piglets was performed as described in the following.
Three groups, each of ten animals, were infected at study day 0 either with wild-type parental EU PRRSV strain Lon94-13 (WT group), or with delta nspl XVIII-12 (nspl group), or were not infected (Ch control group). Infection was applied by intramuscular injection to the neck at dosages of 106'56 TCID50 for LoN94-13 or 106'6 TCID50 for delta nspl XVIII-12, respectively. 21 days post vaccination, all animals were challenged with a virulent EU PRRSV strain being heterologous to LoN94-13 by intramuscular injection and intranasel inoculation at a total dosage of 3x106,52 TCID50. Animals were kept until the end of the experiment at day 31 , ten days after challenge, and body temperatures were measured for all animals at days 0 (1 and 4 hours post vacciantion), 1 , 3, 5, 8, 10, 12, 14, 18, 20, 22, 24, 26, 28, and 31.
Mean body temperatures were determined for each animal for the time after vaccination but before challenge using measured body temperature data from all timepoints from day 0 through day 20. Subsequently, mean body temperatures were determined for each group (Figure 7, blue (left-hand) columns). Error bars indicate standard deviations, respectively. Following the same procedure, mean body temperatures and standard deviations were determined for all groups for the time after challenge using measured body temperature data from all timepoints from day 22 through 31 (Figure 7, brown (right-hand) columns).
Significant(ly) in the context of the following means either (i) p-values of 0.05 or lower as determined by the Dunnett test and obtained from comparing the nspl group with either the WT or the Ch control group for either of the two time periods investigated (before and after challenge) or (ii) p-values of 0.05 or lower when comparing the mean temperature change within a group and between the two time periods investigated (before and after challenge). When comparing the determined mean body temperatures for the time after vaccination but before challenge (blue columns) in between the three groups, animals from the WT group demonstrated a rise in body temperature of more than 0.4 °C when compared to animals from the noninfected Ch control group, thus demonstrating virulence of LoN94-13 in the infected host.
In contrast, the nspl group showed a significant reduction in mean body temperature of more than 0.2 °C when compared to the WT group.Thus, since vaccination dosages were the same for the WT and the nspl group, the considerable reduction in increase of body temperature compared to WT demonstrates that the described mutation in the genome of delta nspl XVIII-12 has significantly reduced virulence of the WT parental strain LoN94-13 in the infected animal. The significant rise in the mean body temperature of the Ch control group from before challenge to after challenge of 0.4°C demonstrates virulence of the heterologous EU PRRSV challenge strain. The mean temperature of the WT group after challenge was slightly lower than before challenge, but not significantly reduced (Figure 7). In contrast, the mean body temperature of the nspl group after challenge was significantly reduced by almost 0.2 °C when compared to that before challenge. Moreover, the body temperature of the nspl group after challenge was significantly reduced by almost 0.4 °C when compared to the Ch control group after challenge. Also, mean body temperature of the nspl group after challenge was significantly lower that that of the WT group after challenge by more than 0.2°C. Taken together, this demonstrates that a measurable and significant degree of protection from signs of disease induced by the applied challenge virus was conferred to pigs by vaccination with delta nspl XVIII-12. Since parental PRRSV strain LoN94-13 did not confer significant protection, it is evident that the described mutation in the genome of delta nspl XVIII-12 is causative for the observed significant protective technical effect.
Analogous experiments, wherein the vaccination was performed with lower amounts (105 TCID50) of delta nspl XVIII-12 showed results similar to the above described results (data not shown). Thus, in practice, a preferred amount of 103 to 105 TCID50 is sufficient for vaccination.
Taken together, the invention described here represents the first known viable PRRSV (EU) strains that contain mutations (deletions) in the nspl gene (ηερΐ β) that induce type I IFN (IFN ) production in susceptible cells (MA104) and that show increased sensitivity to type I IFN (IFNp). Moreover, the animal data demonstrates that (i) vaccine candidate delta nspl XVIII-12 is significantly attenuated in the host when compared to its parental PRRSV strain LoN94-13 and that (ii) vaccine candidate delta nspl XVIII-12 confers significant protection from signs of disease induced by challenge with a heterologous PRRSV strain while parental strain LoN94-1 3 does not. Thus, the described vaccine candidates or the described mutations therein, either alone or combined with other attenuating mutations, may serve as promising life attenuated PRRSV vaccines. LIST OF FIGURES
Figure 1 : Sequence alignment of ηερΐ β protein sequences from parental EU PRRSV strain LoN94-13 and from vaccine candidates.
Figure 2: Sequence alignment of ηερΐ β proteins from PRRSV strains.
Figure 3: PRRSV-specific immunofluorescence of virus stock titrations.
(A) delta nspl XVII-1 ; (B) delta nspl XVIII-12.
Black, negative fluorescence; grey, few positive cells; light green, foci of positive cells; dark green, complete cell monolayer positive.
Figure 4: Virus stocks of vaccine candidates contain IFN3.
Figure 5: Time course of IFN induction after infection of MA104 cells.
Figure 6: delta nspl mutants show increased sensitivity to type I IFN.
Figure 7: Mean body temperatures of vaccinated groups before and after challenge. In the sequence listing:
SEQ ID NO:1 corresponds to LoN94-13 complete nspl protein,
SEQ ID NO:2 corresponds to Lelystad virus complete nspl protein,
SEQ ID NO:3 corresponds to VR2332 complete nspl protein,
SEQ ID NO:4 corresponds to LoN94-13 complete nspl Alpha,
SEQ ID NO:5 corresponds to Lelystad virus complete nspl Alpha,
SEQ ID NO:6 corresponds to VR2332 complete nspl Alpha,
SEQ ID NO:7 corresponds to LoN94-13 complete nspl Beta,
SEQ ID NO:8 corresponds to Lelystad virus complete nspl Beta,
SEQ ID NO:9 corresponds to VR2332 complete nspl Beta,
SEQ ID NO: 10 corresponds to LoN94-13 nspl Beta NTD,
SEQ ID NO:1 1 corresponds to Lelystad virus nspl Beta NTD,
SEQ ID NO:12 corresponds to VR2332 nspl Beta NTD,
SEQ ID NO:13 corresponds to LoN94-13 complete viral cDNA insert,
SEQ ID NO:14 corresponds to Lelystad virus complete genome,
SEQ ID NO: 15 corresponds to VR2332 complete genome,
SEQ ID NO:16 corresponds to delta nspl IX-10 complete nspl protein sequence,
SEQ ID NO:17 corresponds to delta nspl XVII-1 complete nspl protein sequence,
SEQ ID NO:18 corresponds to delta nspl XVIII-12 complete nspl protein sequence,
SEQ ID NO:19 corresponds to delta nspl XIX-2 complete nspl protein sequence,
SEQ ID NO:20 corresponds to delta nspl XX-9 complete nspl protein sequence,
SEQ ID NO:21 corresponds to delta nspl IX-10 complete nspl Beta protein sequence, SEQ ID NO 22 corresponds to delta nsp XVII- 1 complete nspl Beta protein sequence,
SEQ ID NO 23 corresponds to delta nsp XVIII- 12 complete nspl Beta protein sequence,
SEQ ID NO 24 corresponds to delta nsp XIX- 2 complete nspl Beta protein sequenc,
SEQ ID NO 25 corresponds to delta nsp XX- 9 complete nspl Beta protein sequence,
SEQ ID NO 26 corresponds to delta nsp IX-10 nspl Beta NTD protein sequence,
SEQ ID NO 27 corresponds to delta nsp XVII- 1 nspl Beta NTD protein sequence,
SEQ ID NO 28 corresponds to delta nsp XVIII- 12 nspl Beta NTD protein sequence,
SEQ ID NO 29 corresponds to delta nsp XIX- 2 nspl Beta NTD protein sequence,
SEQ ID NO 30 corresponds to delta nsp XX- 9 nspl Beta NTD protein sequence,
SEQ ID NO 31 corresponds to delta nsp IX-10 complete viral cDNA insert sequence,
SEQ ID NO 32 corresponds to delta nsp XVII- 1 complete viral cDNA insert sequence,
SEQ ID NO 33 corresponds to delta nsp XVIII- 12 complete viral cDNA insert sequence,
SEQ ID NO 34 corresponds to delta nsp XIX- 2 complete viral cDNA insert sequence,
SEQ ID NO 35 corresponds to delta nsp XX-9 complete viral cDNA insert sequence,
SEQ ID NO 36 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion o1 2 aa),
SEQ ID NO 37 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion of 3 aa),
SEQ ID NO 38 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 4 aa),
SEQ ID NO 39 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 5 aa),
SEQ ID NO 40 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion o1 6 aa),
SEQ ID NO 41 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 7 aa),
SEQ ID NO 42 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 2 aa),
SEQ ID NO 43 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion of 3 aa),
SEQ ID NO 44 corresponds to a (type 2) nspl Beta NTD with mutal tion (deletion o1 4 aa),
SEQ ID NO 45 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion o1 5 aa),
SEQ ID NO 46 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 6 aa),
SEQ ID NO 47 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 7 aa),
SEQ ID NO 48 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion of 2 aa),
SEQ ID NO 49 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion o1 3 aa),
SEQ ID NO 50 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 4 aa),
SEQ ID NO 51 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion ol 5 aa),
SEQ ID NO 52 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion of 6 aa),
SEQ ID NO 53 corresponds to a (type 1 ) nspl Beta NTD with muta tion (deletion o1 7 aa),
SEQ ID NO 54 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion of 2 aa),
SEQ ID NO 55 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 3 aa),
SEQ ID NO 56 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 4 aa),
SEQ ID NO 57 corresponds to a (type 2) nspl Beta NTD with muta tion (deletion ol 5 aa), SEQ ID NO:58 corresponds to a (type 2) nspl Beta NTD with mutation (deletion of 6 aa), SEQ ID NO:59 corresponds to a (type 2) nspl Beta NTD with mutation (deletion of 7 aa).
Reference List:
Allende, R., Laegreid, W.W., Kutish, G.F., Galeota, J.A., Wills, R.W., Osorio, F.A., 2000.
Porcine reproductive and respiratory syndrome virus: description of persistence in individual pigs upon experimental infection. J. Virol. 74, 10834-10837. Ansari, I.H., Kwon, B., Osorio, F.A., Pattnaik, A.K., 2006. Influence of N-linked glycosylation of porcine reproductive and respiratory syndrome virus GP5 on virus infectivity, antigenicity, and ability to induce neutralizing antibodies. J. Virol. 80, 3994-4004.
Beura, L.K., Sarkar, S.N., Kwon, B., Subramaniam, S., Jones, C, Pattnaik, A. K., Osorio, F.A., 2010. Porcine reproductive and respiratory syndrome virus nonstructural protein 1 eta modulates host innate immune response by antagonizing IRF3 activation. J.
Virol. 84, 1574-1584.
Buddaert, W., Van, R.K., Pensaert, M., 1998. In vivo and in vitro interferon (IFN) studies with the porcine reproductive and respiratory syndrome virus (PRRSV). Adv. Exp. Med. Biol. 440, 461-467. Chen, Z., Lawson, S., Sun, Z., Zhou, X., Guan, X., Christopher-Hennings, J., Nelson, E.A., Fang, Y., 2010. Identification of two auto-cleavage products of nonstructural protein 1 (nspl ) in porcine reproductive and respiratory syndrome virus infected cells: nspl function as interferon antagonist. Virology 398, 87-97. den Boon, J.A., Faaberg, K.S., Meulenberg, J.J ., Wassenaar, A.L., Plagemann, P.G., Gorbalenya, A.E., Snijder, E.J., 1995. Processing and evolution of the N-terminal region of the arterivirus replicase ORF1 a protein: identification of two papainlike cysteine proteases. J. Virol. 69, 4500-4505.
Haller, O., Weber, F., 2009. The interferon response circuit in antiviral host defense. Verh. K.
Acad. Geneeskd. Belg. 71 , 73-86. Kim, O., Sun, Y., Lai, F.W., Song, C, Yoo, D., 2010. Modulation of type I interferon induction by porcine reproductive and respiratory syndrome virus and degradation of CREB- binding protein by non-structural protein 1 in MARC-145 and HeLa cells. Virology 402, 315-326.
Kroese, M.V., Zevenhoven-Dobbe, J.C., Bos-de Ruijter, J.N., Peeters, B.P., Meulenberg, J. J., Cornelissen, L.A., Snijder, E.J., 2008. The nsplalpha and nspl papain-like autoproteinases are essential for porcine reproductive and respiratory syndrome virus RNA synthesis. J. Gen. Virol. 89, 494-499.
Li, H., Zheng, Z., Zhou, P., Zhang, B., Shi,∑., Hu, Q., Wang, H., 2010. The cysteine protease domain of porcine reproductive and respiratory syndrome virus non-structural protein 2 antagonizes interferon regulatory factor 3 activation. J. Gen. Virol. 91 , 2947-2958.
Lopez, O.J., Osorio, F.A., 2004. Role of neutralizing antibodies in PRRSV protective immunity. Vet. Immunol. Immunopathol. 102, 155-163.
Meier, W.A., Galeota, J., Osorio, F.A., Husmann, R.J., Schnitzlein, W.M., Zuckermann, F.A., 2003. Gradual development of the interferon-gamma response of swine to porcine reproductive and respiratory syndrome virus infection or vaccination. Virology 309, 18-31.
Miller, L.C., Laegreid, W.W., Bono, J.L., Chitko-McKown, C.G., Fox, J.M., 2004. Interferon type I response in porcine reproductive and respiratory syndrome virus-infected MARC-145 cells. Arch. Virol. 149, 2453-2463.
Ostrowski, M. , Galeota, J.A., Jar, A.M. , Piatt, K. B. , Osorio, F.A. , Lopez, O.J . , 2002.
Identification of neutralizing and nonneutralizing epitopes in the porcine reproductive and respiratory syndrome virus GP5 ectodomain. J. Virol. 76, 4241-4250.
Shi , X. , Wang, L. , Li, X. , Zhang, G. , Guo, J . , Zhao, D. , Chai, S. , Deng, R. , 201 1 a.
Endoribonuclease activities of porcine reproductive and respiratory syndrome virus nsp1 1 was essential for nsp11 to inhibit IFN-beta induction. Mol. Immunol. 48, 1568- 1572.
Shi, X., Wang, L, Zhi, Y., Xing, G., Zhao, D., Deng, R., Zhang, G., 2010. Porcine reproductive and respiratory syndrome virus (PRRSV) could be sensed by professional beta interferon-producing system and had mechanisms to inhibit this action in MARC-145 cells. Virus Res. 153, 151-156. Shi, X., Zhang, G., Wang, L, Li, X., Zhi, Y., Wang, F., Fan, J., Deng, R., 2011 b. The Nonstructural Protein 1 Papain-Like Cysteine Protease Was Necessary for Porcine Reproductive and Respiratory Syndrome Virus Nonstructural Protein 1 to Inhibit Interferon-beta Induction. DNA Cell Biol. 30, 355-362. Snijder, E.J., Meulenberg, J. J., 1998. The molecular biology of arteriviruses. J. Gen. Virol. 79 ( Pt 5), 961-979.
Song, C, Krell, P., Yoo, D., 2010. Nonstructural protein l alpha subunit-based inhibition of NF-kappaB activation and suppression of interferon-beta production by porcine reproductive and respiratory syndrome virus. Virology 407, 268-280. Sun, Y., Xue, F., Guo, Y., Ma, M., Hao, N., Zhang, X.C. , Lou, Z., Li, X. , Rao, Z., 2009.
Crystal structure of porcine reproductive and respiratory syndrome virus leader protease Nsp1 alpha. J. Virol. 83, 10931-10940.
Xiao, Z., Batista, L., Dee, S., Halbur, P., Murtaugh, M.P., 2004. The level of virus-specific T- cell and macrophage recruitment in porcine reproductive and respiratory syndrome virus infection in pigs is independent of virus load. J. Virol. 78, 5923-5933.
Xue, F., Sun, Y., Yan, L, Zhao, C, Chen, J., Bartlam, M., Li, X., Lou, Z., Rao, Z., 2010. The crystal structure of porcine reproductive and respiratory syndrome virus nonstructural protein Nspl beta reveals a novel metal-dependent nuclease. J. Virol. 84, 6461-6471.

Claims

Claims
A Porcine Reproductive and Respiratory Syndrome (PRRS) virus which is able to induce the interferon type I production and secretion by a cell infected by said virus.
The PRRS virus of claim 1 , wherein said virus comprises a mutation in the gene encoding the nspl protein of said virus.
The PRRS virus of claim 1 or 2, wherein said virus comprises a mutation in the gene sequences encoding the nspla subunit and/or the ηερΐ β subunit of the nspl protein (nspl a and/or nspl β) of said virus.
The PRRS virus of any one of claims 1 to 3, wherein said virus comprises a mutation in the gene sequence encoding the N-terminal domain (NTD) of the ηερΐ β subunit of the nspl protein of said virus.
The PRRS virus of claim 4, wherein the NTD starts with the N-terminus of ηερΐ β, preferably with the amino acid sequence SXXY if the PRRS virus is a genotype I PRRS virus or with the amino acid sequence AXVY if the PRRS virus is a genotype II PRRS virus and ends with the serine residue (S) within the amino acid sequence SFP of ηερΐ β.
The PRRS virus of any one of claims 2 to 5, wherein said mutation comprises one or more point mutations and/or one or more genomic deletions and/or one or more insertions.
7. The PRRS virus of any one of claims 2 to 6, wherein said mutation comprises a
deletion or replacement of a nucleotide triplett coding for an amino acid residue located within the first 24 N-terminal amino acid residues of the NTD sequence.
8. The PRRS virus of any one of claims 2 to 7 a, wherein said mutation comprises or consistis of a deletion or replacement of 2 to 7 or more, preferably consecutive, nucleotide tripletts each coding for an amino acid residue located within the first 24 N- terminal amino acid residues of the NTD sequence.
9. The PRRS virus of any one of claims 2 to 8, wherein said mutation comprises the deletion or replacement of a nucleotide triplett coding for an amino acid residue with a charged, preferably positively charged, side chain.
10. The PRRS virus of any one of claims 2 to 9, wherein said mutation comprises a
deletion or replacement of the nucleotide triplett coding for the first arginine residue (R) located at least 21 amino acid residues in C-terminal direction from the N-terminal amino acid residue of the ηερΐ β or the NTD.
1 1. The PRRS virus of any one of claims 2 to 10, wherein said mutation comprises a deletion or replacement of the nucleotide triplett coding for the arginine residue (R) of the nspl amino acid sequence RXMW if the PRRS virus is a genotype I PRRS virus or with the amino acid sequence RGG of said virus if the PRRS virus is a genotype II PRRS virus.
12. The PRRS virus of any one of claims 2 to 1 1 , wherein said mutation comprises a deletion or replacement of the nucleotide triplett coding for the arginine residue (R) of the sequence RMM or RGG of the nspl protein of said virus.
13. The PRRS virus of any one of claims 10 to 12, wherein said mutation comprises a deletion or replacement of one nucleotide triplett coding for the amino acid residue flanking said arginine residue (R) in N-terminal direction.
14. The PRRS virus of claim 13, wherein said mutation comprises a deletion or
replacement of two consecutive nucleotide tripletts coding for the amino acid residues PR.
15. The PRRS virus of any one of claims 10 to 14, wherein said mutation further
comprises a deletion or replacement of two, three, four, five, six, seven or more consecutive nucleotide tripletts coding for two, three, four, five, six, seven or more amino acid residues flanking said arginine residue (R) in N-terminal direction.
16. The PRRS virus of claim 15, wherein said mutation comprises a deletion or
replacement of three consecutive nucleotide tripletts coding for the amino acid sequence RXR or APR of the ηερΐ β subunit, or wherein said mutation comprises a deletion or replacement of four consecutive nucleotide tripletts coding for the amino acid residues GRXR or WAPR of the ηερΐ β subunit, wherein X is a genetically encoded amino acid residue.
17. The PRRS virus of any one of claims 2 to 16, wherein the mutation comprises a deletion of the gene sequence encoding the (whole) nspl protein, encoding the (whole) nspla subunit, encoding the (whole) ηερΐ β subunit, or encoding a part, preferably seven or more amino acid residues, of or the (whole) NTD domain of the ηερΐ β subunit.
18. The PRRS virus of any one of claims 2 to 17, wherein the nspl protein is a
polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NOs 1-3.
19. The PRRS virus of any one of claims 3 to 17, wherein the nspla subunit is a
polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NOs 4-6.
20. The PRRS virus of any one of claims 3 to 17, wherein the nspl β subunit is a
polypeptide having at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide selected from the group consisting of SEQ ID NOs 7-9.
21. The PRRS virus of any one of claims 4 to 17, wherein the NTD domain of the nspl β subunit has a polypeptide sequence or is a polypeptide having at least 60%, in particular at least 70%, preferably at least 80%, more preferably at least 90%, still more preferably at least 95% or in particular 100% sequence identity with a polypeptide sequence or a polypeptide selected from the group consisting of SEQ ID NOs 10-12.
22. The PRRS virus of any one of claims 1 to 21 , wherein said PPRS virus comprises a gene sequence coding for a polypeptide sequence or a polypeptide selected from the group consisting of SEQ ID NOs:36-47.
23. The PRRS virus of any one of claims 1 to 22, wherein said PPRS virus comprises a gene sequence coding for a polypeptide sequence or a polypeptide selected from the group consisting of SEQ ID NOs:48-59.
24. The PRRS virus of any one of claims 1 to 23, wherein said PPRS virus comprises a gene sequence coding for a polypeptide sequence or a polypeptide selected from the group consisting of SEQ ID NOs:16-30.
25. The PRRS virus of any one of claims 1 to 24, wherein said PPRS virus comprises a RNA sequence complementary to a polynucleotide selected from the group consisting of SEQ ID NOs: 31-35.
26. The PRRS virus of any one of claims 1 to 25, wherein said PPRS virus has increased sensitivity to interferon type I.
27. The PRRS virus of any one of claims 1 to 26, wherein said PPRS virus is a live PRRS virus.
28. The PRRS virus of any one of claims 1 to 27 wherein said PPRS virus is a modified- live PRRS virus.
29. The PRRS virus of any one of claims 1 to 28, wherein said PRRS virus is a PRRS virus mutant.
30. The PRRS virus of any one of claims 1 to 29, wherein said PRRS virus is attenuated PRRS virus.
31. The PRRS virus of any one of claims 1 to 30, wherein said PPRS virus is a genotype I or a genotype II PRRS virus.
32. The PRRS virus of any one of claims 1 to 31 , wherein said interferon type I is
Interferon-a and/or lnterferon-β.
33. The PRRS virus of any one of claims 1 to 32, wherein said cell is a mammalian cell, and wherein said mammalian cell is preferably a cell of a primary or secondary cell line and/or a host cell.
34. The PRRS virus of claim 33, wherein said mammalian cell is a porcine cell or a simian cell.
35. The PRRS virus of claim 34, wherein said porcine cell is a porcine macrophage or wherein said simian cell is a MA-104 or a MARC-145 cell.
36. The PRRS virus of any one of claims 1 to 35 for use as vaccine or medicament, peferably for use as live vaccine.
37. The PRRS virus of any one of claims 1 to 36 for use in the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome, preferably in swine.
38. Use of the PRRS virus of any one of claims 1 to 37 for the preparation of a vaccine or medicament for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome, preferably in swine
39. Method for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome comprising administering the PRRS virus of any one of claims 1 to 37 to an animal, preferably to a swine, more preferably to a pig, in particular preferably to a piglet or a sow.
40. Polynucleotide comprising the genome of the PPRS virus of any one of claims 1 to 37.
41. DNA-Vector comprising a copy of the polynucleotide of claim 40.
42. A virus particle comprising the polynucleotide of claim 40 or 41.
43. A cell comprising the polynucleotide of claim 40 or the DNA-vector of claim 41.
44. Medicament or vaccine comprising the PRRS virus of of any one of claims 1 to 37.
45. Method of preparing a vaccine or medicament, in particular for the prophylaxis or treatment of Porcine Reproductive and Respiratory Syndrome in swine, comprising the cultivation of the PRRS virus of any one of claims 1 to 37 in cell culture.
46. Method of producing the PRRS virus of any one of claims 1 to 37, comprising the mutagenesis of the gene coding for the nspl protein in the genome of a PRRS virus, preferably in the genome of a wild type PRRS virus.
47. The method of claim 46, wherein the mutation according to any one claims 2 to 17 is introduced in the genome of said virus, preferably by site directed mutagenesis.
48. The method of any one of claims 45 to 47, wherein the vector of claim 41 is used.
49. The method of any one of claims 45 to 48, comprising the detection of interferon type I, preferably interferon type I secretion, and/or of mRNA coding for interferon type I.
50. The method of claim 49, wherein the interferon type I and/or the mRNA coding for interferon type I is detected by a bioassay, wherein said bioassay is preferably selected from the group consisting of ELISA, PCR, GLISA, IFA, biosensoric measurement, Surface Plasmon Resonance (SPR) measurement, selective media, lateral flow, biochip measurement, immunomagnetic separation,
electrochemiluminescence, chromogenic media, immunodiffusion, DNA hybridization, staining, colorimetric detection, luminescence, and combinations thereof.
51. An immunogenic composition containing the PRRS virus of any one of claims 1 to 37.
52. The immunogenic composition of claim 51 , comprising an amount of 101 to 107 viral particles per dose, preferably 103 to 106 particles per dose, more preferably 104 to 106 particles per dose, or comprising an amount of said PRRS virus which is equivalent to a virus titre of at least about 103 TCID50/mL per dose, preferably between 103 to 105 TCID50/mL per dose.
53. The immunogenic composition of claim 51 or 52, further containing one or more pharmaceutically acceptable carriers or excipients, wherein said one or more pharmaceutically acceptable carriers or excipients are preferably selected from the group consisting of solvents, dispersion media, adjuvants, stabilizing agents, diluents, preservatives, antibacterial and antifungal agents, isotonic agents, and adsorption delaying agents.
54. The immunogenic composition of any one of claims 51 to 53 for use as a vaccine or as a medicament.
55. The immunogenic composition of any one of claims 51 to 54 for use in a method for inducing an immune response against PRRSV or for the prevention or reduction of PRRS or for the prevention or reduction of PRRSV viremia and/or of clinical symptoms, in particular the elevated body temperature, associated with PRRSV infection or for the prevention or reduction of the elevated body temperature associated with the administration of an attenuated PRRSV vaccine to an animal.
56. Use of the PRRS virus of any one of claims 1 to 37 or of the immunogenic composition of any one of claims 51 to 54, in particular the use of claim 38, for the preparation of a vaccine or medicament for inducing an immune response against PRRSV or for treating or preventing PRRS or for preventing or reducing PRRSV viremia and/or clinical symptoms, in particular the elevated body temperature, associated with PRRSV infection or for the prevention or reduction of the elevated body temperature associated with the administration of an attenuated PRRSV vaccine to an animal.
57. A method for inducing an immune response against PRRSV or for treating or
preventing PRRS or for preventing or reducing PRRSV viremia and/or clinical symptoms, in particular the elevated body temperature, associated with PRRSV infection or for the prevention or reduction of the elevated body temperature associated with the administration of an attenuated PRRSV vaccine to an animal, comprising the step of administering the immunogenic composition according to any one of claims 51 to 54 to an animal in need thereof.
58. The method of claim 57, wherein the immunogenic composition is administered in a single dose or in two doses.
PCT/EP2012/064893 2011-07-29 2012-07-30 Novel prrs virus inducing type i interferon in susceptible cells WO2013017570A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP12741333.4A EP2737059A1 (en) 2011-07-29 2012-07-30 Novel prrs virus inducing type i interferon in susceptible cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP11176023.7 2011-07-29
EP11176019.5 2011-07-29
EP11176019 2011-07-29
EP11176023 2011-07-29

Publications (1)

Publication Number Publication Date
WO2013017570A1 true WO2013017570A1 (en) 2013-02-07

Family

ID=46582718

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2012/064888 WO2013017568A1 (en) 2011-07-29 2012-07-30 INFECTIOUS cDNA CLONE OF EUROPEAN PRRS VIRUS AND USES THEREOF
PCT/EP2012/064893 WO2013017570A1 (en) 2011-07-29 2012-07-30 Novel prrs virus inducing type i interferon in susceptible cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/064888 WO2013017568A1 (en) 2011-07-29 2012-07-30 INFECTIOUS cDNA CLONE OF EUROPEAN PRRS VIRUS AND USES THEREOF

Country Status (3)

Country Link
US (1) US9315781B2 (en)
EP (2) EP2737059A1 (en)
WO (2) WO2013017568A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015092058A1 (en) 2013-12-20 2015-06-25 Boehringer Ingelheim Vetmedica Gmbh Prrs virus variant, european prrs virus cdna clone, and uses thereof
CN106367529A (en) * 2016-08-30 2017-02-01 中国农业科学院兰州兽医研究所 RT-LAMP kit for detecting American type highly-pathogenic porcine reproductive and respiratory syndrome by adopting rapid developing one-step method
WO2018024677A1 (en) 2016-08-05 2018-02-08 Hipra Scientific, S.L.U. PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME VIRUS cDNA CLONE AND USES THEREOF
EP3119429A4 (en) * 2014-03-21 2018-04-04 Nutech Ventures A non-naturally occuring porcine reproductive and respiratory syndrome virus (prrsv) and methods of using

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2675475T3 (en) 2011-02-17 2015-12-31 Boehringer Ingelheim Vetmedica Gmbh Novel european prrsv strain
US9187731B2 (en) * 2011-07-29 2015-11-17 Boehringer Ingelheim Vetmedica Gmbh PRRS virus inducing type I interferon in susceptible cells
US9315781B2 (en) * 2011-07-29 2016-04-19 Boehringer Ingelheim Vetmedica Gmbh Infectious CDNA clone of european PRRS virus and uses thereof
ES2872500T3 (en) * 2014-07-21 2021-11-02 Fatro Spa Attenuated strain of PRRS and potential use in immunizing preparations
EP4021577A1 (en) * 2019-08-29 2022-07-06 Elanco Us Inc. Porcine reproductive and respiratory syndrome vaccine virus

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002060921A2 (en) * 2000-11-09 2002-08-08 Board Of Trustees Of The University Of Illinois Enhancement of immune response to vaccine by interferon alpha

Family Cites Families (103)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3137631A (en) 1959-12-01 1964-06-16 Faberge Inc Encapsulation in natural products
US3080291A (en) 1960-06-10 1963-03-05 Jensen Salsberg Lab Inc Serial passage of distemper virus in tissue cultures of chick embryo and canine tissue and vaccine therefrom
US3959457A (en) 1970-06-05 1976-05-25 Temple University Microparticulate material and method of making such material
US4015100A (en) 1974-01-07 1977-03-29 Avco Everett Research Laboratory, Inc. Surface modification
US4122167A (en) 1977-02-09 1978-10-24 Merck & Co., Inc. Respiratory synctial vaccine
US4205060A (en) 1978-12-20 1980-05-27 Pennwalt Corporation Microcapsules containing medicament-polymer salt having a water-insoluble polymer sheath, their production and their use
US4224412A (en) 1979-05-01 1980-09-23 Dorofeev Viktor M Living virus culture vaccine against canine distemper and method of preparing same
US4554159A (en) 1981-11-12 1985-11-19 Institute Merieux Vaccine and method of immunizing against herpes simplex virus (types 1 and 2)
US4452747A (en) 1982-03-22 1984-06-05 Klaus Gersonde Method of and arrangement for producing lipid vesicles
US4468346A (en) 1983-10-27 1984-08-28 The United States Of America As Represented By The Secretary Of Agriculture Monoclonal antibodies to porcine immunoglobulins
DE3405100A1 (en) 1984-02-14 1985-08-14 Drägerwerk AG, 2400 Lübeck PT CATALYST ON A CARRIER AS AN AIR PURIFIER
US4744933A (en) 1984-02-15 1988-05-17 Massachusetts Institute Of Technology Process for encapsulation and encapsulated active material system
US5008050A (en) 1984-06-20 1991-04-16 The Liposome Company, Inc. Extrusion technique for producing unilamellar vesicles
US4921706A (en) 1984-11-20 1990-05-01 Massachusetts Institute Of Technology Unilamellar lipid vesicles and method for their formation
US4606940A (en) 1984-12-21 1986-08-19 The Ohio State University Research Foundation Small particle formation and encapsulation
US5206163A (en) 1985-07-08 1993-04-27 Chiron Corporation DNA encoding bovine diarrhea virus protein
US4753884A (en) 1986-01-28 1988-06-28 Novagene, Inc. Pseudorabies virus mutants, vaccines containing same, methods for the production of same and methods for the use of same
JPS62198626A (en) 1986-02-26 1987-09-02 Biseibutsu Kagaku Kenkyusho:Kk Vaccine for preventing infectious disease of virus of hemagglutinating encephalomyelitis
FR2602791B1 (en) 1986-08-18 1988-11-10 Ministere Agri Direction Quali METHOD FOR CULTURING TURKEY INFECTIOUS RHINOTRACHEITIS VIRUS, AND VACCINE PREPARED FROM THE VIRUS OBTAINED THEREBY
NZ222465A (en) 1986-11-07 1992-11-25 Pasteur Institut Nanb (non a, non b hepatitis viral) antigen
US5009956A (en) 1987-02-24 1991-04-23 Univ Minnesota Phospholipase A2-resistant liposomes
DE3833925A1 (en) 1988-03-11 1989-09-21 Inst Angewandte Biotechnologie PROCESS AND MANUFACTURE OF VIRUS AND VIRAL ANTIGEN AND DEVICE THEREFOR
US5213759A (en) 1988-05-05 1993-05-25 Elopak Systems A.G. Sterilization
US4927637A (en) 1989-01-17 1990-05-22 Liposome Technology, Inc. Liposome extrusion method
US4944948A (en) 1989-02-24 1990-07-31 Liposome Technology, Inc. EGF/Liposome gel composition and method
US5132117A (en) 1990-01-11 1992-07-21 Temple University Aqueous core microcapsules and method for their preparation
AU7007491A (en) 1990-02-02 1991-08-08 Schweiz. Serum- & Impfinstitut Bern Cdna corresponding to the genome of negative-strand rna viruses, and process for the production of infectious negative-strand rna viruses
CA2103460C (en) 1991-06-06 2000-09-26 Gert Wensvoort Causative agent of the mystery swine disease, vaccine compositions and diagnostic kits
US6042830A (en) 1992-08-05 2000-03-28 Boehringer Ingelheim Vetmedica, Inc. Viral agent associated with mystery swine disease
US6080570A (en) 1991-08-26 2000-06-27 Boehringer Ingelheim Vetmedica, Inc. Method of producing a vaccine for Swine Infertility and Respiratory Syndrome
US6982160B2 (en) 1991-08-26 2006-01-03 Boehringer Ingelheim Vetmedica, Inc. Immunogenic compositions that include SIRS virus
US5846805A (en) 1991-08-26 1998-12-08 Boehringer Ingelheim Animal Health, Inc. Culture of swine infertility and respiratory syndrome virus in simian cells
ATE144144T1 (en) 1991-08-26 1996-11-15 James Edward Collins VACCINE AGAINST AND DIAGNOSTIC METHOD FOR SWINE INFERTILITY AND RESPIRATORY SYNDROME (SUAS)
CA2076744C (en) 1991-08-26 2000-06-27 Danny W. Chladek Viral agent associated with mystery swine disease
WO1993006211A1 (en) 1991-09-16 1993-04-01 Collins James E Vaccine for mystery swine disease and method for diagnosis thereof
EP0610250B2 (en) 1991-10-14 2008-10-29 Intervet International BV Porcine reproductive respiratory syndrome (prrs) vaccine and diagnostic
FR2686097B1 (en) 1992-01-14 1994-12-30 Rhone Merieux PREPARATION OF ANTIGENS AND MYSTERY DISEASE VIRUS VACCINES, ANTIGENS AND VACCINES OBTAINED FOR THE PREVENTION OF THIS DISEASE.
US5338543A (en) 1992-02-27 1994-08-16 Ambico, Inc. Thimerosal inactivated mycoplasma hyopneumoniae vaccine
TW289731B (en) 1992-07-09 1996-11-01 Akzo Nv
US6380376B1 (en) 1992-10-30 2002-04-30 Iowa State University Research Foundation Proteins encoded by polynucleic acids of porcine reproductive and respiratory syndrome virus (PRRSV)
US5695766A (en) 1992-10-30 1997-12-09 Iowa State University Research Foundation Highly virulent porcine reproductive and respiratory syndrome viruses which produce lesions in pigs and vaccines that protect pigs against said syndrome
US6773908B1 (en) 1992-10-30 2004-08-10 Iowa State University Research Foundation, Inc. Proteins encoded by polynucleic acids of porcine reproductive and respiratory syndrome virus (PRRSV)
US6592873B1 (en) 1992-10-30 2003-07-15 Iowa State University Research Foundation, Inc. Polynucleic acids isolated from a porcine reproductive and respiratory syndrome virus (PRRSV) and proteins encoded by the polynucleic acids
US6251397B1 (en) 1992-10-30 2001-06-26 Iowa State University Research Foundation, Inc. Proteins encoded by polynucleic acids isolated from a porcine reproductive and respiratory syndrome virus and immunogenic compositions containing the same
US5419907A (en) 1992-11-10 1995-05-30 Iowa State University Research Foundation, Inc. Pathogenic porcine respiratory coronavirus
CZ289743B6 (en) 1993-02-08 2002-03-13 Bayer Corporation Porcine reproductive and respiratory syndrome virus (PRRSV) isolate, process for growing PRRSV, tissue culture, process for preparing a vaccine and the vaccine containing the PRRSV isolate
ES2074950B1 (en) 1993-09-17 1996-03-16 Iberica Cyanamid VACCINE FOR THE PREVENTION OF REPRODUCTIVE AND RESPIRATORY DISEASE OF THE SOW.
EP0659885A1 (en) 1993-12-21 1995-06-28 Akzo Nobel N.V. Vaccine against viruses associated with antibody-dependent-enhancement of viral infectivity
DE4407489A1 (en) 1994-03-07 1995-09-14 Bayer Ag Vaccine for the prevention of respiratory and reproductive diseases of the pig
ES2165405T3 (en) 1994-04-11 2002-03-16 Akzo Nobel Nv EUROPEAN VACCINE BREEDS OF THE SWINE REPRODUCTIVE AND RESPIRATORY SYNDROME VIRUS (PRRSV).
CA2187768A1 (en) 1994-04-15 1995-10-26 Moser, Charlotte A. Aqueous solvent encapsulation method, apparatus and microcapsules
GB2289279B (en) 1994-05-13 1998-09-16 Iberica Cyanamid Diagnostic kits and vaccines containing recombinant PRRSV proteins
DE69535061T2 (en) 1994-08-05 2007-01-25 Regents Of The University Of Minnesota, Minneapolis VIRAL NUCLEOTIDE SEQUENCE VR-2332 AND USE METHOD
ATE268783T1 (en) 1995-03-14 2004-06-15 Akzo Nobel Nv EXPRESSION IN THE SAME CELL OF POLYPEPTIDES FROM SWINE REPRODUCTIVE AND RESPIRATORY SYNDROME
US5690940A (en) 1995-06-21 1997-11-25 Regents Of The University Of Minnesota Low pathogencity PRRS live virus vaccines and methods of preparation thereof
ES2102971B1 (en) 1996-01-25 1998-03-01 Hipra Lab Sa NEW ATTENUATED STRAIN OF THE VIRUS CAUSING THE RESPIRATORY AND REPRODUCTIVE SYNDROME (PRRS), THE VACCINES AND DIAGNOSTIC MEDIA OBTAINABLE WITH THE SAME AND THE PROCEDURES FOR ITS OBTAINING.
JP3135069B2 (en) 1996-03-01 2001-02-13 シェーリング コーポレイション Swine reproductive and respiratory syndrome vaccine
US6015663A (en) 1996-03-01 2000-01-18 The United States Of America As Represented By The Secretary Of Agriculture Restriction enzyme screen for differentiating porcine reproductive and respiratory syndrome virus strains
US5866401A (en) 1996-03-01 1999-02-02 Schering Corporation Porcine reproductive and respiratory syndrome vaccine
US5976537A (en) 1996-07-02 1999-11-02 The United States Of America As Represented By The Secretary Of Agriculture Porcine reproductive and respiratory syndrome vaccine
PT835930E (en) 1996-10-09 2001-06-29 Akzo Nobel Nv EUROPEAN VACCINE VIRUSES OF PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME VIRUS (PRRSV)
US20040224327A1 (en) 1996-10-30 2004-11-11 Meulenberg Johanna Jacoba Maria Infectious clones of RNA viruses and vaccines and diagnostic assays derived thereof
EP0839912A1 (en) 1996-10-30 1998-05-06 Instituut Voor Dierhouderij En Diergezondheid (Id-Dlo) Infectious clones of RNA viruses and vaccines and diagnostic assays derived thereof
WO1998035023A1 (en) 1997-02-07 1998-08-13 Origen, Inc. Method for growing porcine reproductive and respiratory syndrome virus for use as vaccines and diagnostic assays
DE69837992T2 (en) 1997-05-06 2008-03-20 Boehringer Ingelheim Vetmedica Gmbh PRRSV ANTIGENS BASED ON PEPTIDE SEQUENCES OF THE PRRS VIRUS FOR USE AS A VACCINE AND FOR DIAGNOSTIC TESTS
CA2283422A1 (en) 1997-06-04 1998-12-10 Calgene, Llc Production of polyunsaturated fatty acids by expression of polyketide-like synthesis genes in plants
AU7960598A (en) 1997-06-05 1998-12-21 Origen, Inc. Recombinant porcine reproductive and respiratory syndrome virus (prrsv) for use as a vaccine
US7211379B2 (en) 1997-10-03 2007-05-01 Merial Sas Prevention of myocarditis, abortion and intrauterine infection associated with porcine circovirus-2
US6391314B1 (en) 1997-10-03 2002-05-21 Merial Porcine circoviruses vaccines diagnostic reagents
NZ501264A (en) 1998-12-22 2001-09-28 Pfizer Prod Inc Polynucleotide DNA sequence encoding an infectious RNA molecule encoding a North American PRRS
US7618797B2 (en) 1998-12-22 2009-11-17 Pfizer Inc Infectious cDNA clone of North American porcine reproductive and respiratory syndrome (PRRS) virus and uses thereof
US7691389B2 (en) 1998-12-22 2010-04-06 Pfizer Inc Infectious cDNA clone of north american porcine reproductive and respiratory syndrome (PRRS) virus and uses thereof
US7132106B2 (en) 1998-12-22 2006-11-07 Pfizer Inc. Infectious cDNA clone of North American porcine reproductive and respiratory syndrome (PRRS) virus and uses thereof
FR2789695B1 (en) 1999-02-11 2003-03-07 Merial Sas VIRAL VACCINES AND VACCINES BASED ON RECOMBINANT AND REPLICATIVE SWINE ADENOVIRUSES
EP1157121B1 (en) 1999-03-08 2013-04-24 Boehringer Ingelheim Vetmedica GmbH Prrsv replicon
MXPA01010681A (en) 1999-04-22 2003-08-20 Us Agriculture Porcine reproductive and respiratory syndrome vaccine, based on isolate ja-142.
US20040213805A1 (en) 1999-10-12 2004-10-28 Verheije Monique Helene Deletions in arterivirus replicons
US20020012670A1 (en) 2000-01-26 2002-01-31 Knut Elbers Recombinant attenuation of porcine reproductive and respiratory syndrome (PRRSV)
DK1255815T3 (en) 2000-02-08 2006-10-30 Univ Minnesota Porcine reproductive and respiratory syndrome virus for methods of use
EP1156111A1 (en) 2000-05-19 2001-11-21 Stichting Dienst Landbouwkundig Onderzoek Chimeric arterivirus-like particles
US7018638B2 (en) 2000-12-19 2006-03-28 Wyeth Mycoplasma hyopneumoniae bacterin vaccine
EP1397498A1 (en) 2001-05-21 2004-03-17 ID-Lelystad, Instituut voor Dierhouderij en Diergezondheid B.V. Delections in arterivirus replicons
US7279166B2 (en) 2001-12-12 2007-10-09 Virginia Tech Intellectual Properties, Inc. Chimeric infectious DNA clones, chimeric porcine circoviruses and uses thereof
US6841364B2 (en) 2002-01-22 2005-01-11 Protatek International, Inc. Infectious cDNA clones of porcine reproductive and respiratory syndrome virus and expression vectors thereof
ATE557082T1 (en) 2002-04-05 2012-05-15 Boehringer Ingelheim Vetmed SEQUENCE POSITIONS FOR ADJUSTMENT IN PRRSV
US7335361B2 (en) 2003-06-09 2008-02-26 Animal Technology Institute Taiwan Fusion antigen used as vaccine
US7722878B2 (en) 2004-06-17 2010-05-25 Boehringer Ingelheim Vetmedica, Inc. PRRSV subunit vaccines
AR049924A1 (en) 2004-06-18 2006-09-13 Univ Minnesota IDENTIFICATION OF VIRALLY INFECTED AND VACCINATED ORGANISMS
US20060063151A1 (en) 2004-09-21 2006-03-23 Michael Roof Porcine reproductive and respiratory syndrome isolates and methods of use
US7632636B2 (en) 2004-09-21 2009-12-15 Boehringer Ingelheim Vetmedica, Inc. Porcine reproductive and respiratory syndrome isolates and methods of use
AU2006205852A1 (en) 2005-01-13 2006-07-20 Boehringer Ingelheim Vetmedica Gmbh Improved PRRS vaccines
CA2894069C (en) 2005-06-24 2019-02-26 Regents Of The University Of Minnesota Prrs viruses, infectious clones, mutants thereof, and methods of use
CN106075422B (en) 2005-11-29 2021-05-28 衣阿华州立大学研究基金公司 Identification of protective antigenic determinants of Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) and uses thereof
US8563313B2 (en) 2007-02-13 2013-10-22 Washington State University Research Foundation Macrophage cell-lines for propagation of porcine reproductive and respiratory syndrome virus
WO2008121958A1 (en) 2007-04-02 2008-10-09 Boehringer Ingelheim Vetmedica, Inc. Use of prrsv vaccines to reduce pcv2 viremia
US20100003278A1 (en) 2008-07-03 2010-01-07 Boehringer Ingelheim Vetmedica, Inc. Immunological Compositions Effective for Lessening the Severity or Incidence of PRRSV Signs and Methods of Use Thereof
WO2010025109A1 (en) 2008-08-25 2010-03-04 Boehringer Ingelheim Vetmedia, Inc. Vaccine against highly pathogenic porcine reproductive and respiratory syndrome (hp prrs)
US20100129398A1 (en) 2008-11-26 2010-05-27 Boehringer Ingelheim Vetmedica, Inc. Materials and Methods for Control of Porcine Reproductive and Respiratory Syndrome
ES2742262T3 (en) 2010-04-16 2020-02-13 Univ Gent Vaccine with cross protection for swine respiratory and reproductive syndrome virus
US8728487B2 (en) 2011-01-20 2014-05-20 Hua Wu Attenuated live vaccine for prevention of porcine reproductive and respiratory syndrome
US9187731B2 (en) 2011-07-29 2015-11-17 Boehringer Ingelheim Vetmedica Gmbh PRRS virus inducing type I interferon in susceptible cells
US9315781B2 (en) * 2011-07-29 2016-04-19 Boehringer Ingelheim Vetmedica Gmbh Infectious CDNA clone of european PRRS virus and uses thereof
US8906385B2 (en) 2011-12-01 2014-12-09 University Of Maryland, College Park Interferon-inducing porcine reproductive and respiratory syndrome virus isolate

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002060921A2 (en) * 2000-11-09 2002-08-08 Board Of Trustees Of The University Of Illinois Enhancement of immune response to vaccine by interferon alpha

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
ALLENDE, R.; LAEGREID, W.W.; KUTISH, G.F.; GALEOTA, J.A.; WILLS, R.W.; OSORIO, F.A.: "Porcine reproductive and respiratory syndrome virus: description of persistence in individual pigs upon experimental infection", J. VIROL., vol. 74, 2000, pages 10834 - 10837
ANSARI, I.H.; KWON, B.; OSORIO, F.A.; PATTNAIK, A.K.: "Influence of N-linked glycosylation of porcine reproductive and respiratory syndrome virus GP5 on virus infectivity, antigenicity, and ability to induce neutralizing antibodies", J. VIROL., vol. 80, 2006, pages 3994 - 4004, XP002436516, DOI: doi:10.1128/JVI.80.8.3994-4004.2006
BEURA, L.K., SARKAR, S.N., KWON, B., SUBRAMANIAM, S., JONES, C., PATTNAIK, A.K., OSORIO, F.A.: "Porcine reproductive and respiratory syndrome virus nonstructural protein 1beta modulates host innate immune response by antagonizing IRF3 activation", J. VIROL., vol. 84, 2010, pages 1574 - 1584, XP055025626 *
BEURA, L.K.; SARKAR, S.N.; KWON, B.; SUBRAMANIAM, S.; JONES, C.; PATTNAIK, A.K.; OSORIO, F.A.: "Porcine reproductive and respiratory syndrome virus nonstructural protein 1 beta modulates host innate immune response by antagonizing RF3 activation", J. VIROL., vol. 84, 2010, pages 1574 - 1584, XP055025626, DOI: doi:10.1128/JVI.01326-09
BUDDAERT, W; VAN, R.K.; PENSAERT, M.: "In vivo and in vitro interferon (IFN) studies with the porcine reproductive and respiratory syndrome virus (PRRSV", ADV. EXP. MED. BIOL., vol. 440, 1998, pages 461 - 467, XP008006545
CHEN Z ET AL: "Identification of two auto-cleavage products of nonstructural protein 1 (nsp1) in porcine reproductive and respiratory syndrome virus infected cells: nsp1 function as interferon antagonist", VIROLOGY, ACADEMIC PRESS,ORLANDO, US, vol. 398, no. 1, 1 March 2010 (2010-03-01), pages 87 - 97, XP026894720, ISSN: 0042-6822, [retrieved on 20091216], DOI: 10.1016/J.VIROL.2009.11.033 *
CHEN, Z.; LAWSON, S.; SUN, Z.; ZHOU, X.; GUAN, X.; CHRISTOPHER-HENNINGS, J.; NELSON, E.A.; FANG, Y: "Identification of two auto-cleavage products of nonstructural protein 1 (nsp1) in porcine reproductive and respiratory syndrome virus infected cells: nsp1 function as interferon antagonist", VIROLOGY, vol. 398, 2010, pages 87 - 97, XP026894720, DOI: doi:10.1016/j.virol.2009.11.033
DEN BOON, J.A; FAABERG, K.S.; MEULENBERG, J.J.; WASSENAAR, A.L.; PLAGEMANN, P.G.; GORBALENYA, A.E.; SNIJDER, E.J.: "Processing and evolution of the N-terminal region of the arterivirus replicase ORF1a protein: identification of two papainlike cysteine proteases", J. VIROL., vol. 69, 1995, pages 4500 - 4505
HALLER, 0.; WEBER, F.: "The interferon response circuit in antiviral host defense", VERH. K. ACAD. GENEESKD. BELG, vol. 71, 2009, pages 73 - 86
KIM, O.; SUN, Y.; LAI, F.W.; SONG, C.; YOO, D: "Modulation of type I interferon induction by porcine reproductive and respiratory syndrome virus and degradation of CREB- binding protein by non-structural protein 1 in MARC-145 and HeLa cells", VIROLOGY, vol. 402, 2010, pages 315 - 326, XP027053570
KIMMAN T G ET AL: "Challenges for porcine reproductive and respiratory syndrome virus (PRRSV) vaccinology", VACCINE, ELSEVIER LTD, GB, vol. 27, no. 28, 8 June 2009 (2009-06-08), pages 3704 - 3718, XP026134046, ISSN: 0264-410X, [retrieved on 20090503], DOI: 10.1016/J.VACCINE.2009.04.022 *
KROESE, M.V.; ZEVENHOVEN-DOBBE, J.C; BOS-DE RUIJTER, J.N.; PEETERS, B.P.; MEULENBERG, J.J.; CORNELISSEN, L.A.; SNIJDER, E.J.: "The nsplalpha and nsp1 papain-like autoproteinases are essential for porcine reproductive and respiratory syndrome virus RNA synthesis", J. GEN. VIROL., vol. 89, 2008, pages 494 - 499, XP002548874, DOI: doi:10.1099/vir.0.83253-0
LI, H.; ZHENG, Z.; ZHOU, P.; ZHANG, B.; SHI, Z.; HU, Q.; WANG, H.: "The cysteine protease domain of porcine reproductive and respiratory syndrome virus non-structural protein 2 antagonizes interferon regulatory factor 3 activation", J. GEN. VIROL., vol. 91, 2010, pages 2947 - 2958
LOPEZ, O.J.; OSORIO, F.A.: "Role of neutralizing antibodies in PRRSV protective immunity", VET. IMMUNOL. IMMUNOPATHOL., vol. 102, 2004, pages 155 - 163, XP004613286, DOI: doi:10.1016/j.vetimm.2004.09.005
MEIER, W.A; GALEOTA, J.; OSORIO, F.A.; HUSMANN, R.J.; SCHNITZLEIN, W.M; ZUCKERMANN, F.A.: "Gradual development of the interferon-gamma response of swine to porcine reproductive and respiratory syndrome virus infection or vaccination", VIROLOGY, vol. 309, 2003, pages 18 - 31
MILLER, L.C.; LAEGREID, W.W.; BONO, J.L.; CHITKO-MCKOWN, C.G.; FOX, J.M.: "Interferon type I response in porcine reproductive and respiratory syndrome virus-infected MARC-145 cells", ARCH. VIROL., vol. 149, 2004, pages 2453 - 2463, XP002548876, DOI: doi:10.1007/s00705-004-0377-9
OSTROWSKI, M.; GALEOTA, J.A.; JAR, A.M.; PLATT, K.B.; OSORIO, F.A.; LOPEZ, O.J.: "Identification of neutralizing and nonneutralizing epitopes in the porcine reproductive and respiratory syndrome virus GP5 ectodomain", J. VIROL., vol. 76, 2002, pages 4241 - 4250, XP002412599, DOI: doi:10.1128/JVI.76.9.4241-4250.2002
RUI LUO: "Antiviral activity of type I and type III interferons against porcine reproductive and respiratory syndrome virus (PRRSV)", ANTIVIRAL RESEARCH, vol. 91, 4 May 2011 (2011-05-04), pages 99 - 101, XP055025675 *
See also references of EP2737059A1 *
SHI, X.; WANG, L.; ZHI, Y; XING, G.; ZHAO, D.; DENG, R; ZHANG, G.: "Porcine reproductive and respiratory syndrome virus (PRRSV) could be sensed by professional beta interferon-producing system and had mechanisms to inhibit this action in MARC-145 cells", VIRUS RES., vol. 153, 2010, pages 151 - 156
SHI, X.; ZHANG, G.; WANG, L.; LI, X.; ZHI, Y.; WANG, F.; FAN, J.; DENG, R: "The Nonstructural Protein 1 Papain-Like Cysteine Protease Was Necessary for Porcine Reproductive and Respiratory Syndrome Virus Nonstructural Protein 1 to Inhibit Interferon-beta Induction", DNA CELL BIOL., vol. 30, 2011, pages 355 - 362
SHI, X; WANG, L.; LI, X.; ZHANG, G.; GUO, J; ZHAO, D.; CHAI, S.; DENG, R.: "Endoribonuclease activities of porcine reproductive and respiratory syndrome virus nsp11 was essential for nsp11 to inhibit IFN-beta induction", MOL. IMMUNOL., vol. 48, 2011, pages 1568 - 1572, XP028225648, DOI: doi:10.1016/j.molimm.2011.03.004
SMITH; WATERMAN, ADV. APPL. MATH, vol. 2, 1981, pages 482 - 489
SMITH; WATERMAN, M. S., J MOL BIOL, vol. 147, no. 1, 1981, pages 195 - 197
SNIJDER, E.J; MEULENBERG, J.J.: "The molecular biology of arteriviruses", J. GEN. VIROL, vol. 79, no. 5, 1998, pages 961 - 979, XP000791110
SONG, C.; KRELL, P.; YOO, D.: "Nonstructural protein 1alpha subunit-based inhibition of NF-kappaB activation and suppression of interferon-beta production by porcine reproductive and respiratory syndrome virus", VIROLOGY, vol. 407, 2010, pages 268 - 280
SUN, Y.; XUE, F.; GUO, Y.; MA, M.; HAO, N.; ZHANG, X.C.; LOU, Z.; LI, X.; RAO, Z.: "Crystal structure of porcine reproductive and respiratory syndrome virus leader protease Nsp1alpha", J. VIROL., vol. 83, 2009, pages 10931 - 10940
XIAO, Z.; BATISTA, L.; DEE, S; HALBUR, P.; MURTAUGH, M.P.: "The level of virus-specific T-cell and macrophage recruitment in porcine reproductive and respiratory syndrome virus infection in pigs is independent of virus load", J. VIROL., vol. 78, 2004, pages 5923 - 5933
XUE, F.; SUN, Y.; YAN, L.; ZHAO, C.; CHEN, J.; BARTLAM, M.; LI, X.; LOU, Z.; RAO, Z.: "The crystal structure of porcine reproductive and respiratory syndrome virus nonstructural protein Nspl beta reveals a novel metal-dependent nuclease", J. VIROL., vol. 84, 2010, pages 6461 - 6471

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015092058A1 (en) 2013-12-20 2015-06-25 Boehringer Ingelheim Vetmedica Gmbh Prrs virus variant, european prrs virus cdna clone, and uses thereof
EP3591042A1 (en) 2013-12-20 2020-01-08 Boehringer Ingelheim Vetmedica GmbH Prrs virus variant, european prrs virus cdna clone, and uses thereof
EP3119429A4 (en) * 2014-03-21 2018-04-04 Nutech Ventures A non-naturally occuring porcine reproductive and respiratory syndrome virus (prrsv) and methods of using
WO2018024677A1 (en) 2016-08-05 2018-02-08 Hipra Scientific, S.L.U. PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME VIRUS cDNA CLONE AND USES THEREOF
CN106367529A (en) * 2016-08-30 2017-02-01 中国农业科学院兰州兽医研究所 RT-LAMP kit for detecting American type highly-pathogenic porcine reproductive and respiratory syndrome by adopting rapid developing one-step method
CN106367529B (en) * 2016-08-30 2019-07-26 中国农业科学院兰州兽医研究所 A kind of RT-LAMP kit of the detection of quick colour-developing one-step method american type high-pathogenicity porcine reproductive and respiration syndrome

Also Published As

Publication number Publication date
EP2737058A1 (en) 2014-06-04
WO2013017568A1 (en) 2013-02-07
EP2737059A1 (en) 2014-06-04
US9315781B2 (en) 2016-04-19
US20140314808A1 (en) 2014-10-23

Similar Documents

Publication Publication Date Title
US9187731B2 (en) PRRS virus inducing type I interferon in susceptible cells
US10639364B2 (en) PRRS virus variant, european PRRS virus cDNA clone, and uses thereof
Kimman et al. Challenges for porcine reproductive and respiratory syndrome virus (PRRSV) vaccinology
US7608272B2 (en) Methods and compositions for vaccination of animals with PRRSV antigens with improved immunogenicity
WO2013017570A1 (en) Novel prrs virus inducing type i interferon in susceptible cells
TW201319083A (en) Novel PRRS virus inducing type I interferon in susceptible cells
KR101253692B1 (en) Permissive Cell Lines to Porcine Reproductive and Respiratory Syndrome Virus
Sun Study of a Synthetic Porcine Reproductive and Respiratory Syndrome Virus Strain as a Vaccine Candidate
EA044385B1 (en) PRRS VIRUS VARIANT, EUROPEAN TYPE PRRS VIRUS cDNA CLONE AND THEIR APPLICATION
MX2008003046A (en) Methods and compositions for vaccination of animals with prrsv antigens with improved immunogenicity

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12741333

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2012741333

Country of ref document: EP