US20160279232A1 - Gene-Deleted Variant Strain Of Porcine Pseudorabies Virus, Vaccine Composition, Method Of Making The Same And Use Thereof - Google Patents

Gene-Deleted Variant Strain Of Porcine Pseudorabies Virus, Vaccine Composition, Method Of Making The Same And Use Thereof Download PDF

Info

Publication number
US20160279232A1
US20160279232A1 US15/054,404 US201615054404A US2016279232A1 US 20160279232 A1 US20160279232 A1 US 20160279232A1 US 201615054404 A US201615054404 A US 201615054404A US 2016279232 A1 US2016279232 A1 US 2016279232A1
Authority
US
United States
Prior art keywords
strain
attenuated
prv
pseudorabies virus
virus
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/054,404
Inventor
Kegong Tian
Xuke Zhang
Jinzhong Sun
Feifei Tan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pulaike Biological Engineering Co Ltd
Original Assignee
Pulaike Biological Engineering Co Ltd
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 Pulaike Biological Engineering Co Ltd filed Critical Pulaike Biological Engineering Co Ltd
Priority to US15/054,404 priority Critical patent/US20160279232A1/en
Assigned to PULIKE BIOLOGICAL ENGINEERING, INC. reassignment PULIKE BIOLOGICAL ENGINEERING, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SUN, Jinzhong, TAN, Feifei, TIAN, KEGONG, ZHANG, Xuke
Priority to US15/213,662 priority patent/US10201605B2/en
Publication of US20160279232A1 publication Critical patent/US20160279232A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • 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
    • 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
    • C12N7/04Inactivation or attenuation; Producing viral sub-units
    • 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
    • A61K2039/5252Virus inactivated (killed)
    • 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
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16634Use 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16711Varicellovirus, e.g. human herpesvirus 3, Varicella Zoster, pseudorabies
    • C12N2710/16721Viruses 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16711Varicellovirus, e.g. human herpesvirus 3, Varicella Zoster, pseudorabies
    • C12N2710/16734Use 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16711Varicellovirus, e.g. human herpesvirus 3, Varicella Zoster, pseudorabies
    • C12N2710/16761Methods of inactivation or attenuation
    • C12N2710/16762Methods of inactivation or attenuation by genetic engineering
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16711Varicellovirus, e.g. human herpesvirus 3, Varicella Zoster, pseudorabies
    • C12N2710/16771Demonstrated in vivo effect
    • 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
    • C12N2770/20011Coronaviridae
    • C12N2770/20071Demonstrated in vivo effect

Definitions

  • This invention relates to a gene-deleted variant strain of porcine pseudorabies virus, a vaccine composition prepared therefrom, a method of making the same and a use thereof, belonging to the field of animal virology.
  • Pseudorabies also called Aujeszky's disease
  • Aujeszky's disease is an acute infectious disease caused by Suid herpesvirus 1 (SuHV1) belonging to the Alphaherpesvirinae subfamily for many kinds of livestock such as swine, cattle and sheep, as well as poultry and wild animals, with the main symptoms of fever, intense itching (except swine) and encephalomyelitis.
  • Pseudorabies in swine is found nationwide in China causing severe damages, and is one of the major diseases limiting the large-scale production of pig farms. Infection can result in abortion, stillborn or mummified fetuses in pregnant sows, and neurological signs, paralysis and a high death rate in piglets.
  • Pseudorabies virus with strong pantropic properties, neurotropic properties and latent infectivity, may establish long-term latent infection in the peripheral nervous system, and then the latently infected host starts to get sick when the latent virus is activated into the infectious virus.
  • vaccinated pigs cannot completely resist attacks by the wild virus, and still have symptoms like high fever, depression, partially or completely loss of appetite, with a infection rate of more than 30% and a mortality rate between 10% and 20%.
  • literatures in the prior art for example, Jin-mei Peng, et al., Identification and antigenic variation of new epidemiology of pseudorabies virus from swine. Chinese Journal of Preventive Veterinary Medicine, 2013, 35 (1):1-4; Tong Wu et al., Identification and Characterization of a pseudorabies virus isolated from a dead piglet born to vaccinated sow.
  • the Chinese patent application CN103756977A has disclosed a gE and gI deleted variant strain of porcine pseudorabies virus, PRV-ZJ011G strain (of which the accession number is CGMCC No. 7957) and a vaccine prepared therefrom, of which the content of virus prior to being inactivated is 10 6.0 TCID 50 . Immunization with said vaccine could provide a 100% protection rate for 5 healthy piglets at 45 days of age. Said patent application, however, cannot successfully provide an attenuated live vaccine against new PRV strains, due to both humoral and cellular immunity resulted from proliferation of attenuated live PRV vaccine in vivo.
  • the Chinese patent application CN103981153A has disclosed the construction of a gene deleted variant strain of the pseudorabies virus labeled with two fluorescent markers, wherein a variant strain of pseudorabies virus with deletion of gE, gI, US9 and TK gene is inserted with GRP gene and RFP gene as labels at the site of deleted genes through homologous recombination.
  • a gene deleted variant strain is used as a vector of antigen, we don't know if it has immunological competence itself, or how strong the virulence of the variant strain is, since there are more than 70 gene fragments for pseudorabies virus.
  • the vaccines may not be safe for 7-day-old piglets even though they are safe for 6-week-old piglets.
  • the live vaccines may spread among pigs, therefore the vaccine which is only safe for 6-week-old piglets cannot be ensured to be used clinically and a live vaccine which is safe for 7-day-old piglets, is required in the prior art to prevent effectively infection with the new variant strains of PRV.
  • the present invention provides an attenuated live strain of porcine pseudorabies virus for prevention and treatment of pseudorabies caused by mutated pseudorabies virus.
  • the main aim of present invention is to provide an attenuated strain of porcine pseudorabies virus, wherein said attenuated strain of porcine pseudorabies virus is a pseudorabies virus strain with inactivation of gI/gE/11K/28K proteins, preferably said PRV strain is a variant strain of PRV.
  • gI/gE/11K/28K proteins can be achieved by using well known methods in the art, including deletion of nucleotide sequence expressing the functional fragments of those above proteins from the gene, deletion of the whole ORF from the gene, or deletion or addition of one or more nucleotides whereby the gene cannot express functional proteins normally or the proteins expressed don't have their original function or have an extremely weak function.
  • the present invention provides an attenuated genetically engineered strain of porcine pseudorabies virus with deletion of gI/gE/11K/28K genes.
  • the whole ORF of gI/gE/11K/28K genes was deleted from the genome of said attenuated genetically engineered strain of pseudorabies virus.
  • said variant strain of pseudorabies virus is a virus strain of which gE protein has the sequence of SEQ ID NO. 5 or shares at least 95% homology to the sequence of SEQ ID NO. 5; preferably, said variant strain of pseudorabies virus is obtained through isolation, and when infection with said variant strain recurs in pigs previously immunized with attenuated gene-deleted strain of pseudorabies virus according to the prior art, the pigs still display clinical signs of infection with said variant strain, selected from high fever, depression and partial or complete loss of appetite; more preferably, said variant strain of pseudorabies virus is a variant strain of pseudorabies virus and when infection with said variant strain recurs in pigs previously immunized with attenuated strain of PRV with deletion of one or more of gE, TK and gI genes, according to the prior art, the pigs are still infected with pseudorabies, which optionally causes clinical signs of infection, selected from depression and loss of appetite among piglets at the age of
  • said variant strain of pseudorabies virus includes, but are not limited to, PRV HN1201 strain (pseudorabies virus, strain HN1201)(deposited in the China Center for Type Culture Collection on May, 20, 2013, of which the accession number is CCTCC NO. V 201311 and the address is Wuhan University, Wuhan, China); JS-2012 strain (Wu Tong, Qingzhan Zhang, Hao Zheng et al. Isolation and identification of PRV from piglets infected after immunization [J]. Chinese Journal of Animal Infectious Diseases. 2013, 21 (3): 1-7); PRV HeN1 strain (deposited in the China General Microbiological Culture Collection Center on May, 20, 2013, of which the accession number is CGMCC NO.
  • NVDC-PRV-BJ strain NVDC-PRV-HEB strain
  • NVDC-PRV-SD strain Xiuling Yu, Zhi Zhou, Dongmei Hu,et al. Pathogenic Pseudorabies Virus, China, 2012 Emerging Infectious Diseases, www.cdc.gov/eid ol. 20, No. 1, January 2014
  • PRV HN1202 strain pseudorabies virus, strain HN1202 (deposited in the China Center for Type Culture Collection on Aug. 26, 2013, of which the accession number is CCTCC NO.
  • PRV TJ strain Choun-Hua Wang Jin Yuan, Hua-Yang Qin, et al, A novel gE-deleted pseudorabies virus (PRV) provides rapid and complete protection from lethal challenge with the PRV variant emerging in Bartha-K61-vaccinated swine population in China.
  • PRV pseudorabies virus
  • PRV-ZJ01 accession number, CGMCC No. 8170, and disclosed in CN103627678A.
  • said PRV strain is HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain or PRV-ZJ01 strain.
  • said attenuated strain of porcine pseudorabies virus is an attenuated strain of porcine pseudorabies virus with further inactivation of TK protein; preferably the nucleotide sequence at the location of TK in the genome of said attenuated strain of porcine pseudorabies virus encodes and expresses the amino acid sequence shown in SEQ.NO. 4 of the sequence listing.
  • the nucleotide sequence at the location of TK in the genome of said attenuated strain of porcine pseudorabies virus is the nucleotide sequence shown in SEQ.NO. 3 of the sequence listing.
  • the present invention provides an attenuated genetically engineered strain of porcine pseudorabies virus with deletion of gI/gE/11K/28K/TK genes.
  • the term “variant strain of pseudorabies virus”, also called highly pathogenic PRV strain, refers to diseases with significant manifestations including infection among swine at any ages, horizontal transmission among swine herds, short incubation period (1 ⁇ 2 days), morbidity rates between 10% ⁇ 100%, mortality rate in pigs between 10% ⁇ 100% (mortality rate in piglets can reach up to 100%), high fever of pigs after being infected (40° C. ⁇ 42° C., lasting for more than 3 days), dyspnea, diarrhea, wheezing, coughing, sneezing, hind limb paralysis, dog sitting, suddenly falling down, convulsions, lying on their sides, opisthotonus, making strokes with their arms, and finally dying of exhaustion, and reproductive disorder symptoms caused by infection such as declined semen quality of boar, as well as abortion of pregnant sow (the abortion rate can reach up to 35%), premature birth, stillbirth, weakened piglets (weakened
  • said variant strain of pseudorabies virus obtained through isolation, and when infection with said variant strain recurs in pigs previously immunized with attenuated gene-deleted strain of pseudorabies virus according to the prior art, the pigs still display clinical signs of infection with said variant strain, selected from selected from high fever, depression and partial or complete loss of appetite.
  • said variant strain of pseudorabies virus is a virus strain of which gE protein has the sequence of SEQ ID NO. 5 or shares at least 95% homology to the sequence of SEQ ID NO. 5.
  • said variant strain of pseudorabies virus is a variant strain of pseudorabies virus wherein, when infection with said variant strain recurs in pigs previously immunized with attenuated strain of porcine pseudorabies virus with deletion of one or more of gE, TK and gI genes, according to the prior art, the pigs are still infected with pseudorabies, which optionally causes clinical signs of infection selected from depression and loss of appetite among piglets at the age of 9-10 days.
  • the term “homology” in the present invention refers to the level of similarity between two amino acid sequences or two nucleotide sequences.
  • the homology between amino acid sequences or nucleotide sequences can be calculated by any appropriate methods well known in the art, for example, the target amino acid (or nucleotide) sequence and the reference amino acid (or nucleotide) sequence is aligned, and gaps can be induced if necessary to optimize the number of the identical amino acids (or nucleotides) between two aligned sequences, and the percentage of the identical amino acids (or nucleotides) between two aligned sequences can be calculated accordingly. Alignment of amino acid (or nucleotide) sequences and calculation of homology can be achieved by software well kwon in the art.
  • BLAST which can be accessed through the website of the National Center for Biotechnology Information, NCBI, http://blast.ncbi.nlm.nih.gov/Blast.cgi or can be found in Altschul S. F. et al, J. Mol. Biol, 215:403-410 (1990); Stephen F. et al, Nucleic Acids Res., 25:3389-3402 (1997)), ClustalW2 (which can be accessed through the website of the European Bioinformatics Institute, EBI, http://www.eji.ac.uk/Toolsa/clustalw2/, or can be found in Higgins D. G.
  • gI protein is encoded by US7, and comprises 366 amino acids, with ORF located between 122298-123398.
  • GE protein is encoded by USB, and comprises 577 amino acids, with ORF located between 123502-125235.
  • 11K is encoded by US9, and comprises 98 amino acids, with ORF located between 125293-125589.
  • 28K is encoded by US2, and comprises 256 amino acids, with ORF located between 125811-126581.
  • TK also called thymidine kinase
  • UL23 UL23
  • ORF located between 59512-60474.
  • gI/gE/11K/28K and gI/gE/11K/28K/TK in the present invention refers to “gI, gE, 11K and 28K” and “gI, gE, 11K, 28K and TK”, respectively, wherein “I” in the present invention refers to “and”, for example, “inactivation of gI/gE/11K/28K proteins” refers to inactivation of gI, gE, 11K and 28K proteins.
  • PRV-gI-gE-11K-28K-TK ⁇ refers to deletion of gI, gE, 11K, 28K and TK genes.
  • said attenuated genetic strain of porcine pseudorabies virus with deletion of gI/gE/11K/28K genes is attenuated genetically engineered virus strain of PRV HN1201 strain with deletion of gI/gE/11K/28K genes.
  • said attenuated strain of porcine pseudorabies virus includes HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, with deletion of gI/gE/11K/28K.
  • said attenuated strain of porcine pseudorabies virus is PRV HN1201 strain (pseudorabies virus, strain HN1201) with deletion of gI/gE/11K/28K genes using genetic engineering, wherein said PRV HN1201 strain is deposited in the China Center for Type Culture Collection on May, 20, 2013, of which the accession number is CCTCC NO. V 201311 and the address is Wuhan University, Wuhan, China.
  • the term “attenuated” in the present invention refers to: compared with unmodified parent strain, the virulence of the gene-deleted pseudorabies virus strain is reduced, of which manifestations include reduction of numbers of dead pigs, numbers of pigs with fever, and duration of fever. If the statistically significant difference of one or more parameters for determination of severity of diseases for virus strains decreases, its virulence is attenuated.
  • Another aspect of the invention relates to a vaccine composition, wherein said vaccine composition comprises an immune amount of antigen of said attenuated strain of porcine pseudorabies virus and carrier; preferably the content of antigen of the attenuated strain of porcine pseudorabies virus is not less than 10 6.0 TCID 50 /ml.
  • the antigen of said attenuated strain of porcine pseudorabies virus is live attenuated strain of porcine pseudorabies virus; said vaccine composition further comprises a cryoprotectant.
  • said vaccine composition comprises an immune amount of attenuated live vaccine of said variant strain of pseudorabies virus with deletion of gI/gE/11K/28K genes and carrier.
  • said vaccine composition comprises an immune amount of attenuated live vaccine of said variant strain of pseudorabies virus with deletion of gI/gE/11K/28K/TK genes and carrier.
  • said vaccine composition comprises an immune amount of attenuated live vaccine of a variant strain of pseudorabies virus with deletion of gI/gE/11K/28K genes, such as HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain or PRV-ZJ01 strain and carrier.
  • a variant strain of pseudorabies virus with deletion of gI/gE/11K/28K genes such as HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain or PRV-ZJ01 strain and carrier.
  • said vaccine composition comprises an immune amount of attenuated live vaccine of variant strain of PRV strains with deletion of gI/gE/11K/28K/TK genes, such as HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain or PRV-ZJ01 strain and carrier.
  • variant strain of PRV strains with deletion of gI/gE/11K/28K/TK genes such as HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain or PRV-ZJ01 strain and carrier.
  • the antigen of said attenuated strain of porcine pseudorabies virus is attenuated live PRV strain; said vaccine composition further comprises a cryoprotectant.
  • said vaccine composition is attenuated live vaccine of the PRV strain with deletion of gI/gE/11K/28K genes.
  • one or more compounds with adjuvant activity may be added to vaccines. It does not necessarily require such an adjuvant to achieve the efficacy of the live attenuated pseudorabies virus according to the present invention, but especially for a combination vaccine comprising the live attenuated pseudorabies virus according to the present invention and antigenic materials from another pathogenic virus or microorganism (see below), it will be worth adding an adjuvant.
  • Adjuvants are non-specific stimulators of the immune system. They improve immune response of the host responding to a vaccine.
  • adjuvants known in the art is include complete/incomplete Freund's adjuvant, vitamin E, non-ionic blocking copolymers, muramyl dipeptide, ISCOMs (immune stimulating complexes, refer to, for example the European patent EP 1099 42), saponins, mineral oil, vegetable oil, and Carbopol.
  • the live attenuated vaccine according to the present invention further comprises an adjuvant.
  • compositions e.g., sorbitol, mannitol, starch, sucrose, glucose, dextran
  • proteins such as albumin or casein
  • protein-containing agents such as bovine serum or skimmed milk and buffers (e.g. phosphate buffer).
  • the vaccine is very suitable for freeze-drying. Therefore, in a more preferred form of said embodiment, the live attenuated vaccine is in a freeze-dried form.
  • said pseudorabies vaccine in the present invention can be used conjunctly with other inactivated pathogens or antigen to prepare combined vaccines or complex vacancies against various diseases including pseudorabies.
  • the term “combined vaccine” refers to a vaccine prepared with the virus mixture by mixing the pseudorabies virus in the present invention with at least one different virus.
  • the term “complex vaccine” refers to a vaccine prepared from viruses and bacterium.
  • the pseudorabies virus in the present invention can be mixed or combined with classical swine fever virus, porcine reproductive and respiratory syndrome virus, porcine circovirus and/or haemophilus parasuis and mycoplasma.
  • said vaccine further comprises inactivated pathogens or antigen, preferably, said antigen comprises antigen of classical swine fever virus, antigen of porcine reproductive and respiratory syndrome virus, antigen of porcine circovirus and/or haemophilus parasuis or antigen of mycoplasma.
  • said antigen comprises antigen of classical swine fever virus, antigen of porcine reproductive and respiratory syndrome virus, antigen of porcine circovirus and/or haemophilus parasuis or antigen of mycoplasma.
  • the attenuated virus strain in the present invention can be inserted by exogenous genes, said exogenous genes encode one or more antigens selected from a plurality of pathogens infecting pigs, said pathogens consist of porcine reproductive respiratory syndrome (PRRS) virus, porcine Influenza virus, porcine parvovirus, transmissible gastroenteritis virus, rotavirus, type 1 or type 2 porcine circovirus virus, Escherichia coli, Erysipelothrix rhusiopathiae, Bordetella bronchiseptica, Haemophilus parasuis, Mycoplasma hyopneumoniae and Streptococcus suis.
  • PRRS porcine reproductive respiratory syndrome
  • said vaccine composition may further comprise medium, adjuvants and excipients.
  • the vaccine composition according to the present invention may also comprises medium, adjuvants and/or excipients.
  • Physiological saline or distilled water can be used as medium.
  • Another aspect of the present invention relates to a method for preparing said vaccine composition, wherein said method comprises the steps: (1) said attenuated strain of porcine pseudorabies virus is amplified and cultured; and (2) a cryoprotectant is added into said attenuated strain of porcine pseudorabies virus amplified and cultured.
  • Another aspect of the present invention relates to a use of said vaccine composition for preparing medicine for treatment and prevention of pseudorabies.
  • said pseudorabies is pseudorabies caused by a variant strain of pseudorabies virus.
  • the term “diseases relating to PRV” can further refer to diseases with significant manifestations including but not limited to infection among swine at any ages, horizontal transmission among swine herds, short incubation period (1 ⁇ 2 days), morbidity rates between 10% ⁇ 100%, mortality rate in pigs between 10% ⁇ 100% (mortality rate in piglets can reach up to 100%), high fever of pigs after being infected (40° C. ⁇ 42° C., lasting for more than 3 days), dyspnea, diarrhea, wheezing, coughing, sneezing, hind limb paralysis, dog sitting, suddenly falling down, convulsions, lying on their sides, opisthotonus, making strokes with their arms, and finally dying of exhaustion, and reproductive disorder symptoms caused by infection such as declined semen quality of boar, as well as abortion of pregnant sow (the abortion rate can reach up to 35%), premature birth, stillbirth, weakened piglets (weakened piglets)
  • prevention refers to all behaviors to inhibit the infection of pseudorabies virus or delay the onset of the disease via administration of the vaccine composition according to the present invention.
  • treatment refers to all behaviors to relieve or cure the symptoms caused by infection of PRV via administration of the vaccine composition according to the present invention.
  • strain in the present invention with less virulence could provide better immune protection, induce an earlier generation of antibodies and the challenge result after immunization shows that gE antibody is still negative.
  • FIG. 1 is a schematic diagram showing construction of plasmid pUCgI/gE/11K/28KA-GFP-B;
  • FIG. 2 is a schematic diagram showing the location for deletion of gI/gE/11K/28K genes and location of homologous arms, USA and USB in the genome;
  • FIG. 3 is a gel electrophoresis pattern for comparing PCR fragments of PRV HN1201 strain before and after deletion of gI/gE/11K/28K genes through PCR method;
  • FIG. 4 is a schematic diagram showing the location for deletion of TK gene and location of homologous arms of TKA and TKB in the genome;
  • FIG. 5 is a gel electrophoresis pattern for comparing PCR fragments of PRV HN1201 strain before and after deletion of TK gene through PCR method.
  • SEQ ID NO. 1 is the nucleotide sequence of gI/gE/11K/28K genes in the PRV HN1201 strain.
  • SEQ ID NO. 2 is the nucleotide sequence of TK gene in the PRV HN1201 strain.
  • SEQ ID NO. 3 is the nucleotide sequence of location for deletion of TK gene in the PRV HN1201 strain.
  • SEQ ID NO. 4 is the amino acid sequence of location for deletion of TK gene in the PRV HN1201 strain.
  • SEQ ID NO. 5 is the amino acid sequence of gE in the PRV HN1201 strain.
  • the term “per pig” refers to the amount of vaccine each pig injected.
  • TCID 50 refers to 50% tissue culture infective dose, a way to represent viral infectivity.
  • MEM liquid medium is prepared with MEM dry powdered medium purchased from Life Technologies, Corp. according to the instruction.
  • Dulbecco's Modified Eagle's Medium (DMEM) in the present invention is prepared with reference to the preparation method from Appendix A of GB/T18641-2002 Diagnostic Techniques for Aujeszk's Disease.
  • PBS Phosphate Buffer Saline
  • 0.01 mM pH 7.4 PBS as used in the present invention is prepared as described in Molecular cloning: Laboratory manuals, 3 rd edition.
  • the PRV HN1201 strain (pseudorabies virus, strain HN1201) used in the embodiments is deposited in the China Center for Type Culture Collection on May, 20, 2013, of which the accession number is CCTCC NO. V 201311 and the address is Wuhan University, Wuhan, China.
  • the PRV HN1202 strain (pseudorabies virus, strain HN1202) used in the embodiments is deposited in the China Center for Type Culture Collection on Aug. 26, 2013, of which the accession number is CCTCC NO. V 201335 and the address is Wuhan University, Wuhan, China.
  • PRV is the abbreviation for the term Pseudorabies virus.
  • the description of the present invention is further provided with examples of PRV HN1201 strain, NVDC-PRV-BJ strain, NVDCPRV-HEB strain, NVDC-PRV-SD strain and HN1202 strain.
  • FIG. 1 is a schematic diagram showing construction of the transfer vector
  • FIG. 2 shows the location of the homologous arms, USA and USB in the genome.
  • the upstream and downstream primers for the homologous arm USA at the left side are, respectively:
  • USAF CCG GAATTC TCGTCGTGGGCATCGTCATCAT (the underline portion refers to the EcoR I cutting site) USAR: CTA TCTAGA ataacttcgtataatgtatgctatacgaagttat CGGTACTGCGGAGGCTACGGAC (the underline portion refers to the Xba I cutting site, lowercase letters refer to the loxp site
  • the upstream and downstream primers for the homologous arm USB at the right side are, respectively:
  • USBF ACAT GCATGC ataacttcgtatagcatacattatacgaagttat ACGGCAGGATCTCTCCGCGTCCC (the underline portion refers to the SphI cutting site, lowercase letters refer to the loxp site)
  • USBR CCC AAGCTT AGGAGGGGGCGGGGAGCGCGAGC (the underline portion refers to the Hind III cutting site)
  • Vero cells were transfected with PRV HN1201, and part of supernatant was harvested when the cytopathic effect of cells reached to 80%, for extracting genomic DNA of virus by using Geneaid Viral Nucleic Acid Extraction kit as the template for amplification of the homologous arms.
  • PRV HN1201 DNA 1 ⁇ L PrimeSTAR 0.5 ⁇ L 2*primeSTAR GC buffer 25 ⁇ L dNTP(25 mM) 4 ⁇ L Upstream primer 0.5 ⁇ L downstream primer 0.5 ⁇ L Water Used for adjusting to a final volume of 50 ⁇ L
  • pUCUSA and USB was digested by both of SalI and HindIII, and the target fragments were recovered, linked by T4 DNA ligase, and the product was transformed into DH5 ⁇ .
  • the transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified by sequencing after enzyme digestion, and the correct plasmid after identification was named pUCUSAB.
  • the pAcGFP-C1 plasmid (purchased from Clontech, Catalog No. 632470) was digested by Bgl II and Sma I, and the linearized vector was recovered, linked by T4 DNA Ligase after filling-in with DNA Polymerase I Large (Klenow) Fragment, and transformed into the competent cell DH5 ⁇ to obtain MCS deleted GFP plasmid, named pAcGFP ⁇ MCS.
  • the primers for amplifying GFP were designed according to the sequence of pAcGFP-C 1 vector.
  • Upstream primer CMVU ACGC GTCGAC TAGTTATTAATAGTAATCAATTACG (the underline portion refers to the SalI cutting site.)
  • Downstream primer SV40R ACAT GCATGC CTAGAATGCAGTGAAAAAAATGC ((the underline portion refers to the Sph I cutting site.)
  • GFP gene was amplified with pAcGFP ⁇ MCS plasmid as the template, of which the system and condition is as follows:
  • a target band was recovered by electrophoresis on agarose gel for further linking.
  • GFP was digested with both of Sal and Sph I, and the target fragments were recovered, linked to pUCUSB plasmid which had been through the same double enzyme digestion, and the product was transformed into the competent cell DH5 ⁇ .
  • the transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified by sequencing after enzyme digestion, and the correct plasmid after identification was named pUCUSA-GFP-B.
  • a purified recombinant virus rPRV-GFP-US ⁇ which was free of wild-type virus HN1201 and with deletion of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) was obtained.
  • pBS185 plasmid expressing Cre enzyme purchased from addgene, Cre enzyme recognizes loxP sites at downstream of USA and upstream of USB, wherein USA and USB are homology arms, and deletes sequence between two loxp sites
  • genomic DNA of recombinant virus rPRV-GFP-US ⁇ was co-transfected into vero cells, with the results showing relatively obvious cytopathic effect and more single fluorescence 24 h after transfection.
  • PO virus harvested was inoculated for plaque screening; fluorescence-negative plaque was picked for the next round of purification. After 2 rounds of screening and purification, a fluorescence-negative virus was obtained, and named vPRV-US ⁇ .
  • PCR identification result after extraction and purification of viral genomic DNA showed deletion of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment, and indicated that GFP label gene had been deleted.
  • the result showed a successful purification of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment-deleted virus containing no GFP label gene.
  • the viral genome of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment-deleted virus and wild-type virus, was extracted and identified by PCR, with the following primers:
  • the size of PCR amplification product of the wild-type virus was 6286 bp
  • the size of PCR amplification fragment of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment-deleted virus was 1960 bp.
  • PCR assay result confirmed that ORF of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment had been completely missing.
  • TKA and TKB were cloned into pUC19 vector and named pUCTKAB.
  • GFP gene was cloned into pUCTKAB, to obtain a transfer vector for recombinant virus which was called pUCTKA-GFP-B.
  • the homologous arms in the transfer vector are sequences of two sides of TK, therefore the recombinant virus obtained after recombination, was TK gene deleted.
  • FIG. 4 shows the location of homologous arms, TKA and TKB in the genome.
  • Two pairs of primers were designed for amplifying the homologous arms at two sides of TK gene according to the gene sequence of HN1201 virus:
  • the upstream and downstream primers for the homologous arm TKA at the left side are, respectively:
  • TKAF CCG GAATTC GTAGTGCCGGTTGCCCACGTACA (the underline portion refers to the EcoR I cutting site)
  • TKAR CTAG TCTAGA ataacttcgtatagtacacattatacgaagttat CGCTCAGGCTGCCGTTCTGC (the underline portion refers to the Xba I cutting site, lowercase letters refer to the loxp site)
  • the upstream and downstream primers for the homologous arm TKB at the right side are, respectively:
  • TKBF ACAT GCATGC ataacttcgtataatgtgtactatacgaagttat AACGACGACGGCGTGGGAGG (the underline portion refers to the SphI cutting site, lowercase letters refer to the loxp site)
  • TKBR CCC AAGCTT AGGGCGACGGCGAAGAAGAGC (the underline portion refers to the Hind III cutting site)
  • Vero cells were transfected with PRV HN1201, and part of supernatant was harvested when the cytopathic effect of cells reached to 80%, for extracting genomic DNA of virus by using Geneaid Viral Nucleic Acid Extraction kit as the template for amplification of the homologous arms.
  • TKA and TKB were amplified through PCR method by using TAKARA PrimeSTAR, of which the system and condition is as follows:
  • PRV HN1201 DNA 1 ⁇ L PrimeSTAR 0.5 ⁇ L 2*primeSTAR GC buffer 25 ⁇ L dNTP(25 mM) 4 ⁇ L Upstream primer 0.5 ⁇ L downstream primer 0.5 ⁇ L Water Used for adjusting to a final volume of 50 ⁇ L
  • TKA and TKB fragments amplified by PCR were separated by electrophoresis on agarose gel, and the target fragments were recovered with TIANGEN Gel Recovery Kit.
  • TKA fragment and pUC19 vector was digested with both of EcoR I and XbaI, and the target fragments were recovered, linked by T4 DNA ligase, and the product transformed into DH5 ⁇ . The transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified after enzyme digestion, and the correct plasmid after identification was named pUCTKA.
  • pUCTKA and TKB was digested with both of SalI and HindIII, and the target fragments were recovered, linked by T4 DNA ligase, and the product transformed into DH5 ⁇ .
  • the transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified by sequencing after enzyme digestion, and the correct plasmid after identification was named pUCTKAB.
  • the pAcGFP-C1 plasmid (purchased from Clontech, Catalog No. 632470) was digested with Bgl II and Sma I, and the linearized vector was recovered, linked by T4 DNA Ligase after filling-in with DNA Polymerase I Large (Klenow) Fragment, and the product was transformed into the competent cell DH5 ⁇ to obtain MCS deleted GFP plasmid, named pAcGFP ⁇ MCS.
  • the primers for amplifying GFP were designed according to the sequence of pAcGFP-C1 vector.
  • Upstream primer CMVU ACGC GTCGAC TAGTTATTAATAGTAATCAATTACG (the underline portion refers to the SalI cutting site.)
  • Downstream primer SV40R ACAT GCATGC CTAGAATGCAGTGAAAAAAATGC ((the underline portion refers to the Sph I cutting site.)
  • GFP gene was amplified with the template of pAcGFP ⁇ MCS plasmid, of which the system and condition is as follows:
  • a target band was recovered by electrophoresis on agarose gel for further linking.
  • GFP was digested with both of Sal and Sph I, and the target fragments were recovered, linked to pUCTKAB plasmid which had been through the same double enzyme digestion, and the linked product was transformed into the competent cell DH5 ⁇ .
  • the transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified by sequencing after enzyme digestion, and the correct plasmid after identification was named pUCTKA-GFP-B.
  • PO recombinant virus named rPRV-GFP-gI-gE-11K-28K-TK ⁇ , was collected 36-48 h after transfection, or until the cytopathic effect was visible and infected cells exhibited fluorescence.
  • the pBS185 plasmid expressing Cre enzyme (purchased from addgene, Cre enzyme recognizes mutated loxP sites at downstream of TKA and upstream of TKB, wherein TKA and TKB are homology arms, and deletes sequence between two loxp sites) and genomic DNA of recombinant virus rPRV-GFP-gI-gE-11K-28K-TK ⁇ was co-transfected into vero cells, with the results showing relatively obvious cytopathic effect and more single fluorescence 24 h after transfection. After serial dilution, P0 virus harvested was inoculated for plaque screening; fluorescence-negative plaque was picked for the next round of purification.
  • the primers used for identifying deletion of gI/gE/11K/28K were the same as above.
  • the viral genome of gI/gE/11K/28K/TK-deleted virus and wild-type virus was extracted and identified by PCR, with the following primers:
  • the size of PCR amplification product of the wild-type virus was 1566 bp, the size of PCR amplification fragment of TK deleted virus was 742 bp (refer to FIG. 5 ).
  • PRV HN1201 strain with deletion of gI/gE was prepared by reference to the method in Example 1 of CN103756977A.
  • 25 7-day-old piglets which were negative for pseudorabies antibodies and pseudorabies antigen were randomly divided into 5 groups (A, B, C, D and blank control group), each with 5 piglets. Grouping conditions and challenge conditions are shown in Table 1.
  • the virus seed of PRV HN1201 strain with deletion of gI/gE/11K/28K prepared in Example 1, PRV HN1201 strain with deletion of gI/gE/11K/28K/TK prepared in Example 2 and PRV HN1201 strain with deletion of gI/gE prepared in Example 3 was diluted at 5 ⁇ 10 4 fold, and then inoculated into a monolayer of ST cell. After 1 h adhesion, 1000 ml of DMEM medium containing fetal calf serum was added into ST cell, which was then placed at 37° C. in a roller bottle with a rotation speed of 6 rph. The cell medium containing viruses was harvested when the cytopathic effect of cells reached to 80%; the viruses were harvested after 2 times of freezing-thawing the medium and the virus titer was assessed. The virus solution was preserved at low temperature.
  • sucrose and 8 g of gelatin was added into every 100 ml of deionized water, and the solution was autoclaved (under 121° C. for 30 min) after fully melted.
  • the virus solution prepared and preserved in Example 5.1 was mixed with the protective agent prepared and preserved in Example 5.2 at a volume ratio of 1:1, distributed into sterilized bottles, each of which containing 2.6 ml and the mixed virus solution was freeze-dried.
  • the vaccine was tested and determined to be free of contamination of bacterium and exogenous viruses and the content of virus was consistent with that before freeze-drying.
  • the batch number of PRV HN1201 strain with deletion of gI/gE/11K/28K prepared in Example 1, PRV HN1201 strain with deletion of gI/gE/11K/28K/TK prepared in Example 2 and PRV HN1201 strain with deletion of gI/gE prepared in Example 3 were 20140501, 20140502 and 20140503, respectively.
  • the vaccine control group was inoculated with the live PRV vaccine, Bartha K-61strain purchased from HIPRA, Spain, Batch No. 42RH, at the dosage from the protocol.
  • the blank control group was inoculated with 1 mL/piglet of DMEM medium.
  • the piglets were challenged with 1 ⁇ 10 7.0 TCID 50 /piglet of PRV HN1201 strain on day 28 after immunization. After challenge, the body temperature of piglets was determined daily, and in the meanwhile clinical signs and death status were observed (The results are shown in Table 3), the blood of piglets in all the experimental groups and control groups was collected respectively before challenge.
  • the piglets were challenged with 1 ⁇ 10 7.0 TCID 50 /piglet (1 ml/piglet) of PRV HN1201 strain on day 28 after immunization. After challenge, the body temperature of piglets was determined daily, and in the meanwhile clinical signs and death status were observed (The results are shown in Table 5).
  • gI/gE/11K/28K genes and gI/gE/11K/28K/TK genes were deleted from the parent strains, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain and NVDC-PRV-SD strain (Xiuling Yu, Zhi Zhou, Dongmei Hu,et al. Pathogenic Pseudorabies Virus, China, 2012 Emerging Infectious Diseases, www.cdc.gov/eid ol. 20, No. 1, January 2014) (the applicant promises to open it to public for 20 year from the patent application date according to provisions of Guidelines for Patent Examination), HN1202 strain (deposited in the China Center for Type Culture Collection on Aug.
  • the names of the attenuated strains obtained were NVDC-PRV-BJ with deletion of gI/gE/11K/28K/TK, NVDCPRV-HEB with deletion of gI/gE/11K/28K/TK, NVDC-PRV-SD with deletion of gI/gE/11K/28K/TK, and PRVHN1202 with deletion of gI/gE/11K/28K/TK.
  • the deletion of genes was verified through comparison of PCR results with that of parent strains respectively.
  • Example 7 Each attenuated vaccine strains prepared in Example 7 was proliferated according to the method from Example 5.1, mixed with the protective agent (prepared by adding 40 g of sucrose and 8 g of gelatin into every 100 ml of deionized water, and autoclaved (under 121° C. for 30 min) after fully melted) at a volume ratio of 1:1 and the mixed vaccine compositions were freeze-dried.
  • the protective agent prepared by adding 40 g of sucrose and 8 g of gelatin into every 100 ml of deionized water, and autoclaved (under 121° C. for 30 min) after fully melted
  • the batch numbers of NVDC-PRV-BJ strain with deletion of gI/gE/11K/28K/TK, NVDCPRV-HEB strain with deletion of gI/gE/11K/28K/TK, NVDC-PRV-SD strain with deletion of gI/gE/11K/28K/TK and PRV HN1201 strain with deletion of gI/gE/11K/28K/TK were Q01, Q02, Q03 and Q04, respectively.
  • Pathogenicity test was conducted according to the method in Example 4, in which the piglets were randomly divided into 5 groups, each with 5 piglets, inoculated with 1 ml (10 7.0 TCID 50 /ml) of NVDC-PRV-BJ strain with deletion of gI/gE/11K/28K/TK, NVDC-PRV-HEB strain with deletion of gI/gE/11K/28K/TK, NVDC-PRV-SD strain with deletion of gI/gE/11K/28K/TK, and PRV HN1202 strain with deletion of gI/gE/11K/28K/TK by intranasal instillation, respectively.
  • the results showed that all the piglets were alive in each group, with normal body temperature and no clinical signs. It proved that the virulence of mutated PRV strain was reduced through deletion of gI/gE/11K/28K/TK genes.
  • Example 8 Immunogenicity assay of the vaccines prepared in Example 8 was conducted according to the method and dose in Example 6, in the meanwhile the piglets in the vaccine control group were inoculated with the live PRV vaccine, HB-98 strain Batch No. 1308011-1 (purchased from China Animal Husbandry Industry Co., Ltd. Chengdu Medical Equipments Factory). The piglets were challenged with 1 ⁇ 10 7.0 TCID 50 /piglet of PRV HN1201 strain on day 28 after immunization. After challenge, the body temperature of piglets was determined daily, and in the meanwhile clinical signs and death status were observed (the results are shown in Table 6).
  • the piglets exhibited substantially no clinical signs, indicating excellent immune protection of the PRV vaccines relative to live vaccines in the prior art.
  • Groups 1-3 were injected with the vaccine prepared in Example 5,which is PRV HN1201 strain with deletion of gI/gE/11K/28K/TK, with Batch No. 20140502, the live PRV vaccine Bartha K-61 strain, with Batch No. 66KR, purchased from HIPRA, Spain, and the live PRV vaccine, K-61, with Batch No. 195-B59B purchased from Boehringer Ingelheim (US) respectively.
  • All the dose for immunization is 1 ml/piglet (for commercial vaccine, 1 piglet dosage/piglet, according to protocols; the PRV HN1201 with deletion of gI/gE/11K/28K/TK vaccine, 10 6.0 TCID 50 /piglet).
  • the blank control group was inoculated with 1 mL/piglet of DMEM medium.
  • the blood of piglets was collected on day 8, 10, 12, 14 and 21 after immunization, and gB antibody was determined according to the protocol of gB ELISA antibody detection kit (purchased from Biochek, Batch No. F S5763, Expiry Date: Jan. 7, 2015) after the serum was separated. The detailed results of detection are shown in Table 7 below.
  • the antibody test results showed that, all gB antibodies turned positive on day 12 after immunization with PRV HN1201 strain with deletion of gI/gE/11K/28K/TK, while not all the gB antibodies had turned positive on day 21 after immunization with the two control vaccine. It showed that PRV HN1201 strain with deletion of gI/gE/11K/28K/TK could provide earlier immune protection.
  • Groups 1-3 were injected with the vaccine prepared in Example 5,which is PRV HN1201 strain with deletion of gI/gE/11K/28K/TK, with Batch No. 20140502, the live PRV vaccine, Bartha K-61 strain, with Batch No. 66KR, purchased from HIPRA, Spain, and the live PRV vaccine, K-61, with Batch No. 195-B59B purchased from Boehringer Ingelheim (US).
  • Example 5 is PRV HN1201 strain with deletion of gI/gE/11K/28K/TK, with Batch No. 20140502, the live PRV vaccine, Bartha K-61 strain, with Batch No. 66KR, purchased from HIPRA, Spain, and the live PRV vaccine, K-61, with Batch No. 195-B59B purchased from Boehringer Ingelheim (US).
  • All the dose for immunization is 1 ml/piglet (for commercial vaccine, 1 piglet dosage/piglet, according to protocols; the PRV HN1201 with deletion of gI/gE/11K/28K/TK vaccine, 10 6.0 TCID 50 /piglet).
  • the piglets were challenged with 10 7.0 TCID 50 /piglet, 1 ml/piglet of PRV HN1201 strain on day 21 after immunization.
  • the blood of piglets was collected daily continuously from day 7 to day 14 after challenge, and gE antibody was determined according to the protocol of gE ELISA antibody detection kit (purchased from IDEXX Co., Batch No. AK650, Expiry Date: Jun. 13, 2015) after the serum was separated.
  • the vaccine strains in the present invention has a better immunogenicity than commercial vaccine in the prior art, and after immunization therewith a faster generation of the antibody can be achieved, and the effective amplification of virus in the body of pigs can be blocked, and gE antibody is negative.

Landscapes

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

Abstract

The present invention provides an attenuated strain of porcine pseudorabies virus (PRV), in which said attenuated strain of PRV is a variant strain of PRV with inactivation of gI/gE/11K/28K proteins. In addition, the present invention also provides a vaccine composition comprising the attenuated strain of PRV as an antigen, a preparation method and use thereof. Proved by immunogenicity and pathogenicity testing of the live vaccine, said live PRV vaccine can provide a good protection for pigs with no clinical signs observed, indicating excellent immune protection.

Description

  • This application is a continuation of U.S. Patent Application No. 14/901981, filed Dec. 29, 2015, which is a National Stage of PCT/CN2015/070221, filed Jan. 6, 2015, which claims the benefit of priority to CN 201410418379.8, filed Aug. 22, 2014.
  • FIELD OF THE INVENTION
  • This invention relates to a gene-deleted variant strain of porcine pseudorabies virus, a vaccine composition prepared therefrom, a method of making the same and a use thereof, belonging to the field of animal virology.
  • BACKGROUND
  • Pseudorabies, also called Aujeszky's disease, is an acute infectious disease caused by Suid herpesvirus 1 (SuHV1) belonging to the Alphaherpesvirinae subfamily for many kinds of livestock such as swine, cattle and sheep, as well as poultry and wild animals, with the main symptoms of fever, intense itching (except swine) and encephalomyelitis. Pseudorabies in swine is found nationwide in China causing severe damages, and is one of the major diseases limiting the large-scale production of pig farms. Infection can result in abortion, stillborn or mummified fetuses in pregnant sows, and neurological signs, paralysis and a high death rate in piglets. Pseudorabies virus (PRV) with strong pantropic properties, neurotropic properties and latent infectivity, may establish long-term latent infection in the peripheral nervous system, and then the latently infected host starts to get sick when the latent virus is activated into the infectious virus.
  • According to recent researches, there are reports of new features of pseudorabies, of which the significant manifestations include that infection among swine at any ages, horizontal transmission among swine herds, short incubation period (1˜2 days), morbidity rates between 10%˜100%, mortality rate in pigs between 10%˜100% (mortality rate in piglets can reach up to 100%), high fever in pigs after being infected (40° C.˜42° C., lasting for more than 3 days), dyspnea, diarrhea, wheezing, coughing, sneezing, hind limb paralysis, dog sitting, suddenly falling down, convulsions, lying on their sides, opisthotonus, making strokes with their arms, and finally dying of exhaustion, and the infection also can cause reproductive disorder symptoms such as declined semen quality of boar, as well as abortion of pregnant sow (the abortion rate can reach up to 35%), premature birth, stillbirth, weakened piglets (weakened piglets die by 14 days of age), etc. By means of prior art, vaccinated pigs cannot completely resist attacks by the wild virus, and still have symptoms like high fever, depression, partially or completely loss of appetite, with a infection rate of more than 30% and a mortality rate between 10% and 20%. According to literatures in the prior art, for example, Jin-mei Peng, et al., Identification and antigenic variation of new epidemiology of pseudorabies virus from swine. Chinese Journal of Preventive Veterinary Medicine, 2013, 35 (1):1-4; Tong Wu et al., Identification and Characterization of a pseudorabies virus isolated from a dead piglet born to vaccinated sow. Chinese Journal of Animal infectious diseases, 2013, 21 (3):1-7; Yu et al., Pathogenic Pseudorabies Virus, China, 2012. Emerging infectious Diseases. 2014, 20 (1):102-104; An et al., Pseudorabies virus variant in Bartha-K61-vaccinated pigs, China, 2012. Emerging infectious Diseases. 2013. 19 (11): 1749-1755, there is no vaccines capable of solving the pseudorabies caused by variant strains of porcine pseudorabies virus in the prior art.
  • The Chinese patent application CN103756977A has disclosed a gE and gI deleted variant strain of porcine pseudorabies virus, PRV-ZJ011G strain (of which the accession number is CGMCC No. 7957) and a vaccine prepared therefrom, of which the content of virus prior to being inactivated is 106.0TCID50. Immunization with said vaccine could provide a 100% protection rate for 5 healthy piglets at 45 days of age. Said patent application, however, cannot successfully provide an attenuated live vaccine against new PRV strains, due to both humoral and cellular immunity resulted from proliferation of attenuated live PRV vaccine in vivo.
  • The Chinese patent application CN103981153A has disclosed the construction of a gene deleted variant strain of the pseudorabies virus labeled with two fluorescent markers, wherein a variant strain of pseudorabies virus with deletion of gE, gI, US9 and TK gene is inserted with GRP gene and RFP gene as labels at the site of deleted genes through homologous recombination. Whereas in this patent application said a gene deleted variant strain is used as a vector of antigen, we don't know if it has immunological competence itself, or how strong the virulence of the variant strain is, since there are more than 70 gene fragments for pseudorabies virus.
  • It has been disclosed by Chun-Hua Wang et al. (Chun-Hua Wang, Jin Yuan, Hua-Yang Qin, et al, A novel gE-deleted pseudorabies virus (PRV) provides rapid and complete protection from lethal challenge with the PRV variant emerging in Bartha-K61-vaccinated swine population in China, Vaccine 32 (2014)3379-3385) that 6-week-old piglets injected with PRVTJ-gElive vaccines got a complete protection from challenge and didn't exhibit fever. Whereas as is known to those skilled in the art, the virulence of the PRV live vaccines depends on the age of piglets. Also according to the general experience in the art, the vaccines may not be safe for 7-day-old piglets even though they are safe for 6-week-old piglets. The live vaccines may spread among pigs, therefore the vaccine which is only safe for 6-week-old piglets cannot be ensured to be used clinically and a live vaccine which is safe for 7-day-old piglets, is required in the prior art to prevent effectively infection with the new variant strains of PRV.
  • SUMMARY OF INVENTION
  • In order to solve the deficiency of the prior art, the present invention provides an attenuated live strain of porcine pseudorabies virus for prevention and treatment of pseudorabies caused by mutated pseudorabies virus.
  • The main aim of present invention is to provide an attenuated strain of porcine pseudorabies virus, wherein said attenuated strain of porcine pseudorabies virus is a pseudorabies virus strain with inactivation of gI/gE/11K/28K proteins, preferably said PRV strain is a variant strain of PRV.
  • The inactivation of gI/gE/11K/28K proteins can be achieved by using well known methods in the art, including deletion of nucleotide sequence expressing the functional fragments of those above proteins from the gene, deletion of the whole ORF from the gene, or deletion or addition of one or more nucleotides whereby the gene cannot express functional proteins normally or the proteins expressed don't have their original function or have an extremely weak function.
  • As an embodiment of the present invention, the present invention provides an attenuated genetically engineered strain of porcine pseudorabies virus with deletion of gI/gE/11K/28K genes.
  • As an embodiment of the present invention, the whole ORF of gI/gE/11K/28K genes was deleted from the genome of said attenuated genetically engineered strain of pseudorabies virus.
  • As an embodiment of the present invention, said variant strain of pseudorabies virus is a virus strain of which gE protein has the sequence of SEQ ID NO. 5 or shares at least 95% homology to the sequence of SEQ ID NO. 5; preferably, said variant strain of pseudorabies virus is obtained through isolation, and when infection with said variant strain recurs in pigs previously immunized with attenuated gene-deleted strain of pseudorabies virus according to the prior art, the pigs still display clinical signs of infection with said variant strain, selected from high fever, depression and partial or complete loss of appetite; more preferably, said variant strain of pseudorabies virus is a variant strain of pseudorabies virus and when infection with said variant strain recurs in pigs previously immunized with attenuated strain of PRV with deletion of one or more of gE, TK and gI genes, according to the prior art, the pigs are still infected with pseudorabies, which optionally causes clinical signs of infection, selected from depression and loss of appetite among piglets at the age of 9-10 days.
  • Most preferably, said variant strain of pseudorabies virus, includes, but are not limited to, PRV HN1201 strain (pseudorabies virus, strain HN1201)(deposited in the China Center for Type Culture Collection on May, 20, 2013, of which the accession number is CCTCC NO. V 201311 and the address is Wuhan University, Wuhan, China); JS-2012 strain (Wu Tong, Qingzhan Zhang, Hao Zheng et al. Isolation and identification of PRV from piglets infected after immunization [J]. Chinese Journal of Animal Infectious Diseases. 2013, 21 (3): 1-7); PRV HeN1 strain (deposited in the China General Microbiological Culture Collection Center on May, 20, 2013, of which the accession number is CGMCC NO. 6656 and has been disclosed in the patent application CN102994458A); NVDC-PRV-BJ strain, NVDC-PRV-HEB strain and NVDC-PRV-SD strain (Xiuling Yu, Zhi Zhou, Dongmei Hu,et al. Pathogenic Pseudorabies Virus, China, 2012 Emerging Infectious Diseases, www.cdc.gov/eid ol. 20, No. 1, January 2014); PRV HN1202 strain (pseudorabies virus, strain HN1202) (deposited in the China Center for Type Culture Collection on Aug. 26, 2013, of which the accession number is CCTCC NO. V 201335 and the address is Wuhan University, Wuhan, China); PRV TJ strain (Chun-Hua Wang Jin Yuan, Hua-Yang Qin, et al, A novel gE-deleted pseudorabies virus (PRV) provides rapid and complete protection from lethal challenge with the PRV variant emerging in Bartha-K61-vaccinated swine population in China. Vaccine. 32 (2014) 3379-3385); a variant strain of pseudorabies virus PRV-ZJ01 (with the accession number, CGMCC No. 8170, and disclosed in CN103627678A).
  • As an embodiment of the present invention, said PRV strain is HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain or PRV-ZJ01 strain.
  • As an embodiment of the present invention, said attenuated strain of porcine pseudorabies virus is an attenuated strain of porcine pseudorabies virus with further inactivation of TK protein; preferably the nucleotide sequence at the location of TK in the genome of said attenuated strain of porcine pseudorabies virus encodes and expresses the amino acid sequence shown in SEQ.NO. 4 of the sequence listing.
  • As a preferred embodiment of the present invention, the nucleotide sequence at the location of TK in the genome of said attenuated strain of porcine pseudorabies virus is the nucleotide sequence shown in SEQ.NO. 3 of the sequence listing.
  • As a preferred embodiment of the present invention, the present invention provides an attenuated genetically engineered strain of porcine pseudorabies virus with deletion of gI/gE/11K/28K/TK genes.
  • As used herein, the term “variant strain of pseudorabies virus”, also called highly pathogenic PRV strain, refers to diseases with significant manifestations including infection among swine at any ages, horizontal transmission among swine herds, short incubation period (1˜2 days), morbidity rates between 10%˜100%, mortality rate in pigs between 10%˜100% (mortality rate in piglets can reach up to 100%), high fever of pigs after being infected (40° C.˜42° C., lasting for more than 3 days), dyspnea, diarrhea, wheezing, coughing, sneezing, hind limb paralysis, dog sitting, suddenly falling down, convulsions, lying on their sides, opisthotonus, making strokes with their arms, and finally dying of exhaustion, and reproductive disorder symptoms caused by infection such as declined semen quality of boar, as well as abortion of pregnant sow (the abortion rate can reach up to 35%), premature birth, stillbirth, weakened piglets (weakened piglets die by 14 days of age), etc. Preferably, said variant strain of pseudorabies virus obtained through isolation, and when infection with said variant strain recurs in pigs previously immunized with attenuated gene-deleted strain of pseudorabies virus according to the prior art, the pigs still display clinical signs of infection with said variant strain, selected from selected from high fever, depression and partial or complete loss of appetite. Preferably, said variant strain of pseudorabies virus is a virus strain of which gE protein has the sequence of SEQ ID NO. 5 or shares at least 95% homology to the sequence of SEQ ID NO. 5. More preferably, said variant strain of pseudorabies virus is a variant strain of pseudorabies virus wherein, when infection with said variant strain recurs in pigs previously immunized with attenuated strain of porcine pseudorabies virus with deletion of one or more of gE, TK and gI genes, according to the prior art, the pigs are still infected with pseudorabies, which optionally causes clinical signs of infection selected from depression and loss of appetite among piglets at the age of 9-10 days. The term “homology” in the present invention refers to the level of similarity between two amino acid sequences or two nucleotide sequences. The homology between amino acid sequences or nucleotide sequences can be calculated by any appropriate methods well known in the art, for example, the target amino acid (or nucleotide) sequence and the reference amino acid (or nucleotide) sequence is aligned, and gaps can be induced if necessary to optimize the number of the identical amino acids (or nucleotides) between two aligned sequences, and the percentage of the identical amino acids (or nucleotides) between two aligned sequences can be calculated accordingly. Alignment of amino acid (or nucleotide) sequences and calculation of homology can be achieved by software well kwon in the art. Examples of such software include, but is not limited to, BLAST (which can be accessed through the website of the National Center for Biotechnology Information, NCBI, http://blast.ncbi.nlm.nih.gov/Blast.cgi or can be found in Altschul S. F. et al, J. Mol. Biol, 215:403-410 (1990); Stephen F. et al, Nucleic Acids Res., 25:3389-3402 (1997)), ClustalW2 (which can be accessed through the website of the European Bioinformatics Institute, EBI, http://www.eji.ac.uk/Toolsa/clustalw2/, or can be found in Higgins D. G. et al, Methods in Enzymology, 266:383-402 (1996); Larkin M. A. et al, Bioinformatics (Oxford, England), 23 (21):2947-8 (2007)), and TCoffee (which can be accessed through the website of the Swiss Institute of Bioinformatics, SIB, http://tcoffee.vital-it.ch/cgi-bin/Tcoffee/tcoffee_cgi/index.cgi, or can be found in, Poirot O. et al, Nucleic Acids Res., 31 (13):3503-6 (2003); Notredame C. et al, J. Mol. Biol, 302 (1):205-17(2000)) etc. It is all within the knowledge scope of a person skilled in the art that when using the software to do sequence alignment, he can use the default parameters provided by the software or adjust the parameters provided by the software according to the actual condition.
  • The term “gI protein” is encoded by US7, and comprises 366 amino acids, with ORF located between 122298-123398.
  • The term “gE protein” is encoded by USB, and comprises 577 amino acids, with ORF located between 123502-125235.
  • The term “11K” is encoded by US9, and comprises 98 amino acids, with ORF located between 125293-125589.
  • The term “28K” is encoded by US2, and comprises 256 amino acids, with ORF located between 125811-126581.
  • The term “TK”, also called thymidine kinase, is encoded by UL23, and comprises 320 amino acids, with ORF located between 59512-60474.
  • The term “gI/gE/11K/28K” and “gI/gE/11K/28K/TK” in the present invention refers to “gI, gE, 11K and 28K” and “gI, gE, 11K, 28K and TK”, respectively, wherein “I” in the present invention refers to “and”, for example, “inactivation of gI/gE/11K/28K proteins” refers to inactivation of gI, gE, 11K and 28K proteins.
  • Unless otherwise stated, the term “PRV-gI-gE-11K-28K-TK−” in the present invention refers to deletion of gI, gE, 11K, 28K and TK genes.
  • As a preferred embodiment of the present invention, said attenuated genetic strain of porcine pseudorabies virus with deletion of gI/gE/11K/28K genes is attenuated genetically engineered virus strain of PRV HN1201 strain with deletion of gI/gE/11K/28K genes.
  • As a preferred embodiment of the present invention, said attenuated strain of porcine pseudorabies virus includes HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, with deletion of gI/gE/11K/28K.
  • As a preferred embodiment of the present invention, said attenuated strain of porcine pseudorabies virus is PRV HN1201 strain (pseudorabies virus, strain HN1201) with deletion of gI/gE/11K/28K genes using genetic engineering, wherein said PRV HN1201 strain is deposited in the China Center for Type Culture Collection on May, 20, 2013, of which the accession number is CCTCC NO. V 201311 and the address is Wuhan University, Wuhan, China.
  • As used herein, the term “attenuated” in the present invention refers to: compared with unmodified parent strain, the virulence of the gene-deleted pseudorabies virus strain is reduced, of which manifestations include reduction of numbers of dead pigs, numbers of pigs with fever, and duration of fever. If the statistically significant difference of one or more parameters for determination of severity of diseases for virus strains decreases, its virulence is attenuated.
  • Another aspect of the invention relates to a vaccine composition, wherein said vaccine composition comprises an immune amount of antigen of said attenuated strain of porcine pseudorabies virus and carrier; preferably the content of antigen of the attenuated strain of porcine pseudorabies virus is not less than 106.0TCID50/ml.
  • As a preferred embodiment of the present invention, the antigen of said attenuated strain of porcine pseudorabies virus is live attenuated strain of porcine pseudorabies virus; said vaccine composition further comprises a cryoprotectant.
  • As an embodiment of the present invention, said vaccine composition comprises an immune amount of attenuated live vaccine of said variant strain of pseudorabies virus with deletion of gI/gE/11K/28K genes and carrier.
  • As a preferred embodiment of the present invention, said vaccine composition comprises an immune amount of attenuated live vaccine of said variant strain of pseudorabies virus with deletion of gI/gE/11K/28K/TK genes and carrier.
  • As a preferred embodiment of the present invention, said vaccine composition comprises an immune amount of attenuated live vaccine of a variant strain of pseudorabies virus with deletion of gI/gE/11K/28K genes, such as HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain or PRV-ZJ01 strain and carrier.
  • As a preferred embodiment of the present invention, said vaccine composition comprises an immune amount of attenuated live vaccine of variant strain of PRV strains with deletion of gI/gE/11K/28K/TK genes, such as HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain or PRV-ZJ01 strain and carrier.
  • Preferably, the antigen of said attenuated strain of porcine pseudorabies virus is attenuated live PRV strain; said vaccine composition further comprises a cryoprotectant.
  • As an embodiment of the present invention, said vaccine composition is attenuated live vaccine of the PRV strain with deletion of gI/gE/11K/28K genes.
  • Optionally, one or more compounds with adjuvant activity may be added to vaccines. It does not necessarily require such an adjuvant to achieve the efficacy of the live attenuated pseudorabies virus according to the present invention, but especially for a combination vaccine comprising the live attenuated pseudorabies virus according to the present invention and antigenic materials from another pathogenic virus or microorganism (see below), it will be worth adding an adjuvant. Adjuvants are non-specific stimulators of the immune system. They improve immune response of the host responding to a vaccine. Examples of adjuvants known in the art is include complete/incomplete Freund's adjuvant, vitamin E, non-ionic blocking copolymers, muramyl dipeptide, ISCOMs (immune stimulating complexes, refer to, for example the European patent EP 1099 42), saponins, mineral oil, vegetable oil, and Carbopol.
  • Therefore, in a preferred form of said embodiment, the live attenuated vaccine according to the present invention further comprises an adjuvant.
  • Other examples of pharmaceutically acceptable carriers or diluents can be used in the present invention, include stabilizers such as SPGA, carbohydrates (e.g., sorbitol, mannitol, starch, sucrose, glucose, dextran), proteins such as albumin or casein, protein-containing agents such as bovine serum or skimmed milk and buffers (e.g. phosphate buffer).
  • Especially when such stabilizers are added to the vaccine, the vaccine is very suitable for freeze-drying. Therefore, in a more preferred form of said embodiment, the live attenuated vaccine is in a freeze-dried form.
  • In addition, said pseudorabies vaccine in the present invention can be used conjunctly with other inactivated pathogens or antigen to prepare combined vaccines or complex vacancies against various diseases including pseudorabies. As used herein, the term “combined vaccine” refers to a vaccine prepared with the virus mixture by mixing the pseudorabies virus in the present invention with at least one different virus. The term “complex vaccine” refers to a vaccine prepared from viruses and bacterium. For example, the pseudorabies virus in the present invention can be mixed or combined with classical swine fever virus, porcine reproductive and respiratory syndrome virus, porcine circovirus and/or haemophilus parasuis and mycoplasma.
  • As an embodiment of the present invention, said vaccine further comprises inactivated pathogens or antigen, preferably, said antigen comprises antigen of classical swine fever virus, antigen of porcine reproductive and respiratory syndrome virus, antigen of porcine circovirus and/or haemophilus parasuis or antigen of mycoplasma.
  • As an embodiment of the present invention, the attenuated virus strain in the present invention can be inserted by exogenous genes, said exogenous genes encode one or more antigens selected from a plurality of pathogens infecting pigs, said pathogens consist of porcine reproductive respiratory syndrome (PRRS) virus, porcine Influenza virus, porcine parvovirus, transmissible gastroenteritis virus, rotavirus, type 1 or type 2 porcine circovirus virus, Escherichia coli, Erysipelothrix rhusiopathiae, Bordetella bronchiseptica, Haemophilus parasuis, Mycoplasma hyopneumoniae and Streptococcus suis.
  • Preferably, said vaccine composition may further comprise medium, adjuvants and excipients.
  • The vaccine composition according to the present invention may also comprises medium, adjuvants and/or excipients. Physiological saline or distilled water can be used as medium.
  • Another aspect of the present invention relates to a method for preparing said vaccine composition, wherein said method comprises the steps: (1) said attenuated strain of porcine pseudorabies virus is amplified and cultured; and (2) a cryoprotectant is added into said attenuated strain of porcine pseudorabies virus amplified and cultured.
  • Another aspect of the present invention relates to a use of said vaccine composition for preparing medicine for treatment and prevention of pseudorabies.
  • As an embodiment of the present invention, said pseudorabies is pseudorabies caused by a variant strain of pseudorabies virus.
  • As used herein, the term “diseases relating to PRV” can further refer to diseases with significant manifestations including but not limited to infection among swine at any ages, horizontal transmission among swine herds, short incubation period (1˜2 days), morbidity rates between 10%˜100%, mortality rate in pigs between 10%˜100% (mortality rate in piglets can reach up to 100%), high fever of pigs after being infected (40° C.˜42° C., lasting for more than 3 days), dyspnea, diarrhea, wheezing, coughing, sneezing, hind limb paralysis, dog sitting, suddenly falling down, convulsions, lying on their sides, opisthotonus, making strokes with their arms, and finally dying of exhaustion, and reproductive disorder symptoms caused by infection such as declined semen quality of boar, as well as abortion of pregnant sow (the abortion rate can reach up to 35%), premature birth, stillbirth, weakened piglets (weakened piglets die by 14 days of age), etc. The differences between above described symptoms and symptoms caused by infection of regular pseudorabies virus in the prior art are: in adult pigs (whose weight is above 50 kg), high fever of infected pigs (40° C.˜42° C., lasting for more than 3 days), dyspnea, diarrhea, wheezing, coughing, sneezing, hind limb paralysis, dog sitting, suddenly falling down, convulsions, lying on their sides, opisthotonus, making strokes with their arms, and finally dying of exhaustion; sudden incidence of pseudorabies in newborn piglets and piglets below the age of 4 weeks, further resulting in massive death with a mortality of more than 90%; main manifestations in infected piglets including increased body temperature over 41° C., completely loss of appetite, obvious neurological signs and diarrhea; and in piglets just before or after being weaned, mainly respiratory symptoms, such as dyspnea, coughing and runny noses, etc.
  • As used herein, the term “prevention” refers to all behaviors to inhibit the infection of pseudorabies virus or delay the onset of the disease via administration of the vaccine composition according to the present invention. The term “treatment” refers to all behaviors to relieve or cure the symptoms caused by infection of PRV via administration of the vaccine composition according to the present invention.
  • ADVANTAGES OF THE PRESENT INVENTION
  • The strain in the present invention with less virulence could provide better immune protection, induce an earlier generation of antibodies and the challenge result after immunization shows that gE antibody is still negative.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a schematic diagram showing construction of plasmid pUCgI/gE/11K/28KA-GFP-B;
  • FIG. 2 is a schematic diagram showing the location for deletion of gI/gE/11K/28K genes and location of homologous arms, USA and USB in the genome;
  • FIG. 3 is a gel electrophoresis pattern for comparing PCR fragments of PRV HN1201 strain before and after deletion of gI/gE/11K/28K genes through PCR method;
  • FIG. 4 is a schematic diagram showing the location for deletion of TK gene and location of homologous arms of TKA and TKB in the genome;
  • FIG. 5 is a gel electrophoresis pattern for comparing PCR fragments of PRV HN1201 strain before and after deletion of TK gene through PCR method.
  • SEQUENCE LISTING
  • SEQ ID NO. 1 is the nucleotide sequence of gI/gE/11K/28K genes in the PRV HN1201 strain.
  • SEQ ID NO. 2 is the nucleotide sequence of TK gene in the PRV HN1201 strain.
  • SEQ ID NO. 3 is the nucleotide sequence of location for deletion of TK gene in the PRV HN1201 strain.
  • SEQ ID NO. 4 is the amino acid sequence of location for deletion of TK gene in the PRV HN1201 strain.
  • SEQ ID NO. 5 is the amino acid sequence of gE in the PRV HN1201 strain.
  • DETAILED DESCRIPTION
  • The description of the present invention is further provided as follows with reference to the specific embodiments, and features and advantages of the present invention will become more apparent from the following description. However, these embodiments are only exemplary, but not forming any limitation to the scope of the present invention. It should be understood by a person skilled in the art that modifications or alternatives to details and forms of the technical solution of the present invention without deviation from the spirit and scope of the present invention will be allowed, while those modification and alternatives should all fall within the scope of the present invention.
  • In the invention, the term “per pig” refers to the amount of vaccine each pig injected.
  • In the invention, the term “TCID50” refers to 50% tissue culture infective dose, a way to represent viral infectivity.
  • Minimum Essential Medium (MEM) liquid medium is prepared with MEM dry powdered medium purchased from Life Technologies, Corp. according to the instruction.
  • Dulbecco's Modified Eagle's Medium (DMEM) in the present invention is prepared with reference to the preparation method from Appendix A of GB/T18641-2002 Diagnostic Techniques for Aujeszk's Disease.
  • In the present invention, the term “PBS” is the abbreviation for Phosphate Buffer Saline, and 0.01 mM pH 7.4 PBS as used in the present invention is prepared as described in Molecular cloning: Laboratory manuals, 3rd edition.
  • The PRV HN1201 strain (pseudorabies virus, strain HN1201) used in the embodiments is deposited in the China Center for Type Culture Collection on May, 20, 2013, of which the accession number is CCTCC NO. V 201311 and the address is Wuhan University, Wuhan, China.
  • The PRV HN1202 strain (pseudorabies virus, strain HN1202) used in the embodiments is deposited in the China Center for Type Culture Collection on Aug. 26, 2013, of which the accession number is CCTCC NO. V 201335 and the address is Wuhan University, Wuhan, China.
  • PRV is the abbreviation for the term Pseudorabies virus.
  • In the following specific embodiments, the description of the present invention is further provided with examples of PRV HN1201 strain, NVDC-PRV-BJ strain, NVDCPRV-HEB strain, NVDC-PRV-SD strain and HN1202 strain.
  • EXAMPLE 1 Preparation of PRV HN1201 Strain With Deletion of gI/gE/11K/28K 1.1 Construction of a Transfer Vector for Recombinant PRV HN1201GFP Virus
  • According to the sequence of US segment (gI/gE/11K/28K) to be deleted, the homologous arms were designed at its two ends, called USA and USB, respectively. USA and USB were cloned into pUC19 vector and named pUCUSAB. Then GFP gene was cloned into pUCUSAB, to obtain a transfer vector for recombinant virus which was called pUCUSA-GFP-B. The homologous arms in the transfer vector are sequences of two sides of US, therefore the recombinant virus obtained after recombination, was US segment deleted, which comprised gI/gE/11K/28K. FIG. 1 is a schematic diagram showing construction of the transfer vector, and FIG. 2 shows the location of the homologous arms, USA and USB in the genome.
  • 1.1.1 Amplification and Cloning of the Homologous Recombinant Arms 1.1.1.1 Design of Primers and Preparation of Templates
  • Two pairs of primers were designed for amplifying the homologous arms at two sides of segment to be deleted according to the gene sequence of HN1201 virus:
  • The upstream and downstream primers for the homologous arm USA at the left side are, respectively:
  • USAF:
    CCGGAATTCTCGTCGTGGGCATCGTCATCAT
    (the underline portion refers to the EcoR I
    cutting site)
    USAR:
    CTATCTAGAataacttcgtataatgtatgctatacgaagttat
    CGGTACTGCGGAGGCTACGGAC
    (the underline portion refers to the Xba I
    cutting site, lowercase letters refer to the
    loxp site
  • The upstream and downstream primers for the homologous arm USB at the right side are, respectively:
  • USBF:
    ACATGCATGCataacttcgtatagcatacattatacgaagttat
    ACGGCAGGATCTCTCCGCGTCCC
    (the underline portion refers to the SphI cutting
    site, lowercase letters refer to the loxp site)
    USBR:
    CCCAAGCTTAGGAGGGGGCGGGGAGCGCGAGC
    (the underline portion refers to the Hind III
    cutting site)
  • Vero cells were transfected with PRV HN1201, and part of supernatant was harvested when the cytopathic effect of cells reached to 80%, for extracting genomic DNA of virus by using Geneaid Viral Nucleic Acid Extraction kit as the template for amplification of the homologous arms.
  • 1.1.1.2 Amplification and Cloning of the Homologous Arms, USA and USB
  • USA and USB were amplified through PCR method by using TAKARA PrimeSTAR, of which the system and condition is as follows:
  • PRV HN1201 DNA 1 μL
    PrimeSTAR 0.5 μL
    2*primeSTAR GC buffer 25 μL
    dNTP(25 mM) 4 μL
    Upstream primer 0.5 μL
    downstream primer 0.5 μL
    Water Used for adjusting to a final
    volume of 50 μL
  • 98° C. 2 min
    98° C. 10 s
    68° C. 1 min } 30 cycles
    15 s
    68° C. 5 min
  • USA and USB fragments amplified by PCR were separated by electrophoresis on agarose gel, and the target fragments were recovered with TIANGEN Gel Recovery Kit. USA fragment and pUC19 vector was digested by both of EcoR I and XbaI, and the target fragments were recovered, connected by T4 DNA ligase, and the product was transformed into DH5α. The transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified using enzyme digestion, and the correct plasmid after identification was named pUCUSA. pUCUSA and USB was digested by both of SalI and HindIII, and the target fragments were recovered, linked by T4 DNA ligase, and the product was transformed into DH5α. The transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified by sequencing after enzyme digestion, and the correct plasmid after identification was named pUCUSAB.
  • 1.1.3 Amplification of Label Gene GFP 1.1.2.1 Removal of Multiple Cloning Site (MCS) of GFP Vector pAcGFP-C1
  • The pAcGFP-C1 plasmid (purchased from Clontech, Catalog No. 632470) was digested by Bgl II and Sma I, and the linearized vector was recovered, linked by T4 DNA Ligase after filling-in with DNA Polymerase I Large (Klenow) Fragment, and transformed into the competent cell DH5α to obtain MCS deleted GFP plasmid, named pAcGFPΔMCS.
  • 1.1.2.2 Amplification of GFP Gene
  • The primers for amplifying GFP were designed according to the sequence of pAcGFP-C 1 vector.
  • Upstream primer
    CMVU:
    ACGCGTCGACTAGTTATTAATAGTAATCAATTACG
    (the underline portion refers to the SalI
    cutting site.)
    Downstream primer
    SV40R:
    ACATGCATGCCTAGAATGCAGTGAAAAAAATGC
    ((the underline portion refers to the Sph I
    cutting site.)
  • GFP gene was amplified with pAcGFPΔMCS plasmid as the template, of which the system and condition is as follows:
  • pAcGFPΔMCS 1 μL
    primeSTAR 0.5 μL
    2*primeSTAR GC buffer 25 μL
    dNTP(25 mM) 4 μL
    Upstream primer CMVU 0.5 μL
    Downstream primer SV40R 0.5 μL
    Water Used for adjusting to a final
    volume of 50 μL
  • 94° C. 2 min
    94° C. 30 s
    60° C. 30 s 30 cycles {close oversize brace}
    72° C. 2 min
    72° C. 5 min
  • A target band was recovered by electrophoresis on agarose gel for further linking.
  • 1.1.3 Linking of GFP Label Gene and pUCUSAB
  • GFP was digested with both of Sal and Sph I, and the target fragments were recovered, linked to pUCUSB plasmid which had been through the same double enzyme digestion, and the product was transformed into the competent cell DH5α. The transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified by sequencing after enzyme digestion, and the correct plasmid after identification was named pUCUSA-GFP-B.
  • 1.2 Acquisition of Recombinant Virus Containing GFP 1.2.1 Acquisition of Recombinant Virus Through Co-Transfection of Vero Cells With the Transfer Vector and HN1201 DNA
  • Co-transfection of vero cells was conducted by using lipofectin technique, wherein 3 μg PRV-HN1201 viral genomic DNA and 5 μg the transfer vector pUCUSA-GFP-B was transfected, in accordance with procedures of Lipofectamine 2000 Protocol (Invitrogen, Catalog No. 11668030). Cells were incubated at 37° C. in an incubator containing 5% CO2. The supernatant of cell culture, i.e. P0 recombinant virus, named rPRV-GFP-US−, was collected 36-48h after transfection, or until the cytopathic effect was visible and infected cells exhibited fluorescence.
  • 1.2.2 Plaque Purification of Recombinant Viruses
  • When infected with the obtained PO recombinant virus rPRV-GFP-US−, vero cells infected were covered with 2% agarose with low melting point. After 48h when the cytopathic effect became apparent and infected cells exhibited obvious fluorescence, a plaque with a green fluorescence was picked and freeze-thawed 3 times in −70° C., inoculated at 10-fold serial dilutions into vero cells previously laid in six-well plates. Such plaque with a green fluorescence was continued to be picked for purification. After 8 rounds of plaques purification, a purified recombinant virus rPRV-GFP-US−which was free of wild-type virus HN1201 and with deletion of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) was obtained.
  • 1.3 Deletion of GFP Label Gene in the gI/gE/US9/US2 (i.e. gI/gE/11K/28K) Segment-Deleted Recombinant Virus
  • pBS185 plasmid expressing Cre enzyme (purchased from addgene, Cre enzyme recognizes loxP sites at downstream of USA and upstream of USB, wherein USA and USB are homology arms, and deletes sequence between two loxp sites) and genomic DNA of recombinant virus rPRV-GFP-US− was co-transfected into vero cells, with the results showing relatively obvious cytopathic effect and more single fluorescence 24 h after transfection. After serial dilution, PO virus harvested was inoculated for plaque screening; fluorescence-negative plaque was picked for the next round of purification. After 2 rounds of screening and purification, a fluorescence-negative virus was obtained, and named vPRV-US−. PCR identification result after extraction and purification of viral genomic DNA, showed deletion of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment, and indicated that GFP label gene had been deleted. The result showed a successful purification of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment-deleted virus containing no GFP label gene.
  • 1.4 Confirmation of PRV HN1201 Strain With Deletion of US Segment
  • The viral genome of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment-deleted virus and wild-type virus, was extracted and identified by PCR, with the following primers:
  • USDCF:
    TACATCGTCGTGCTCGTCTTTGGC
    USDCR:
    AGCTCGTGCGTCTCGGTGGTG
  • The size of PCR amplification product of the wild-type virus was 6286 bp, the size of PCR amplification fragment of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment-deleted virus was 1960 bp.
  • PCR assay result confirmed that ORF of gI/gE/US9/US2 (i.e. gI/gE/11K/28K) segment had been completely missing.
  • EXAMPLE 2 Preparation of PRV HN1201 Strain With Deletion of gI/gE/11K/28K/TK 2.1 Construction of a Transfer Vector for Recombinant PRV HN1201GFP Virus
  • According to the sequence of TK gene to be deleted, the homologous arms at its two ends were designed, called TKA and TKB, respectively. TKA and TKB were cloned into pUC19 vector and named pUCTKAB. Then GFP gene was cloned into pUCTKAB, to obtain a transfer vector for recombinant virus which was called pUCTKA-GFP-B. The homologous arms in the transfer vector are sequences of two sides of TK, therefore the recombinant virus obtained after recombination, was TK gene deleted. FIG. 4 shows the location of homologous arms, TKA and TKB in the genome.
  • 2.1.1 Amplification and Cloning of the Homologous Recombinant Arms 2.1.1.1 Design of Primers and Preparation of Template
  • Two pairs of primers were designed for amplifying the homologous arms at two sides of TK gene according to the gene sequence of HN1201 virus:
  • The upstream and downstream primers for the homologous arm TKA at the left side are, respectively:
  • TKAF:
    CCGGAATTCGTAGTGCCGGTTGCCCACGTACA
    (the underline portion refers to the EcoR I
    cutting site)
    TKAR:
    CTAGTCTAGAataacttcgtatagtacacattatacgaagttat
    CGCTCAGGCTGCCGTTCTGC
    (the underline portion refers to the Xba I
    cutting site, lowercase letters refer to the
    loxp site)
  • The upstream and downstream primers for the homologous arm TKB at the right side are, respectively:
  • TKBF:
    ACATGCATGCataacttcgtataatgtgtactatacgaagttat
    AACGACGACGGCGTGGGAGG
    (the underline portion refers to the SphI
    cutting site, lowercase letters refer to the
    loxp site)
    TKBR:
    CCCAAGCTTAGGGCGACGGCGAAGAAGAGC
    (the underline portion refers to the Hind III
    cutting site)
  • Vero cells were transfected with PRV HN1201, and part of supernatant was harvested when the cytopathic effect of cells reached to 80%, for extracting genomic DNA of virus by using Geneaid Viral Nucleic Acid Extraction kit as the template for amplification of the homologous arms.
  • 2.1.1.2 Amplification and Cloning of the Homologous Arms, TKA and TKB
  • TKA and TKB were amplified through PCR method by using TAKARA PrimeSTAR, of which the system and condition is as follows:
  • PRV HN1201 DNA 1 μL
    PrimeSTAR 0.5 μL
    2*primeSTAR GC buffer 25 μL
    dNTP(25 mM) 4 μL
    Upstream primer 0.5 μL
    downstream primer 0.5 μL
    Water Used for adjusting to a final
    volume of 50 μL
  • 98° C. 2 min
    98° C. 10 s
    68° C. 1 min } 30 cycles
    15 s
    68° C. 5 min
  • TKA and TKB fragments amplified by PCR were separated by electrophoresis on agarose gel, and the target fragments were recovered with TIANGEN Gel Recovery Kit. TKA fragment and pUC19 vector was digested with both of EcoR I and XbaI, and the target fragments were recovered, linked by T4 DNA ligase, and the product transformed into DH5α. The transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified after enzyme digestion, and the correct plasmid after identification was named pUCTKA. pUCTKA and TKB was digested with both of SalI and HindIII, and the target fragments were recovered, linked by T4 DNA ligase, and the product transformed into DH5α. The transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified by sequencing after enzyme digestion, and the correct plasmid after identification was named pUCTKAB.
  • 2.1.2 Amplification of Label Gene GFP 2.1.2.1 Removal of Multiple Cloning Site (MCS) of GFP Vector pAcGFP-C1
  • The pAcGFP-C1 plasmid (purchased from Clontech, Catalog No. 632470) was digested with Bgl II and Sma I, and the linearized vector was recovered, linked by T4 DNA Ligase after filling-in with DNA Polymerase I Large (Klenow) Fragment, and the product was transformed into the competent cell DH5α to obtain MCS deleted GFP plasmid, named pAcGFPΔMCS.
  • 2.1.2.2 Amplification of GFP Gene
  • The primers for amplifying GFP were designed according to the sequence of pAcGFP-C1 vector.
  • Upstream primer
    CMVU:
    ACGCGTCGACTAGTTATTAATAGTAATCAATTACG
    (the underline portion refers to the SalI
    cutting site.)
    Downstream primer
    SV40R:
    ACATGCATGCCTAGAATGCAGTGAAAAAAATGC
    ((the underline portion refers to the Sph I
    cutting site.)
  • GFP gene was amplified with the template of pAcGFPΔMCS plasmid, of which the system and condition is as follows:
  • pAcGFPΔMCS 1 μL
    PrimeSTAR 0.5 μL
    2*primeSTAR GC buffer 25 μL
    dNTP(25 mM) 4 μL
    Upstream primer CMVU 0.5 μL
    Downstream primer SV40R 0.5 μL
    Water Used for adjusting to a final
    volume of 50 μL
  • 94° C. 2 min
    94° C. 30 s
    60° C. 30 s
    72° C. 2 min 30 cycles {close oversize brace}
    72° C. 5 min
  • A target band was recovered by electrophoresis on agarose gel for further linking.
  • 2.1.3 Linking of GFP Label Gene and pUCTKAB
  • GFP was digested with both of Sal and Sph I, and the target fragments were recovered, linked to pUCTKAB plasmid which had been through the same double enzyme digestion, and the linked product was transformed into the competent cell DH5α. The transformation mix was spread onto plates containing ampicillin, and incubated at 37° C. overnight. A single colony was picked to extract the plasmid and the plasmid was identified by sequencing after enzyme digestion, and the correct plasmid after identification was named pUCTKA-GFP-B.
  • 2.2 Acquisition of Recombinant Virus Containing GFP 2.2.1 Acquisition of Recombinant Virus Through Co-Transfection of Vero Cells With the Transfer Vector and vPRV-gI-gE-11K-28K-DNA
  • Co-transfection of vero cells was conducted by using lipofectin technique, wherein 3 μg vPRV-gI-gE-11K-28K− viral genomic DNA and 5 μg the transfer vector pUCTKA-GFP-B was transfected, in accordance with procedures of Lipofectamine 2000 Protocol (Invitrogen, Catalog No. 11668030). Cells were incubated at 37 ° C. in an incubator containing 5% CO2. The supernatant of cell culture, i.e. PO recombinant virus, named rPRV-GFP-gI-gE-11K-28K-TK−, was collected 36-48 h after transfection, or until the cytopathic effect was visible and infected cells exhibited fluorescence.
  • 2.2.2 Plaque Purification of Recombinant Virus rPRV-GFP-gI-gE-11K-28K-TK−
  • When infected with the obtained PO recombinant virus rPRV-GFP-gI-gE-11K-28K-TK−, vero cells infected were covered with 2% agarose with low melting point. After 48 h when the cytopathic effect became apparent and infected cells exhibited obvious fluorescence, a plaque with a green fluorescence was picked and freeze-thawed 3 times in −70 ° C., inoculated at 10-fold serial dilutions into vero cells previously laid in six-well plates. Such plaque with a green fluorescence was continued to be picked for purification. After 11 rounds of plaques purification, a purified recombinant virus rPRV-GFP-gI-gE-11K-28K-TK− which was free of PRV-gI-gE-11K-28K-TK− and with deletion of five genes was obtained.
  • 2.3 Deletion of GFP Label Gene in gI/gE/11K/28K/TK Deleted Recombinant Virus
  • The pBS185 plasmid expressing Cre enzyme (purchased from addgene, Cre enzyme recognizes mutated loxP sites at downstream of TKA and upstream of TKB, wherein TKA and TKB are homology arms, and deletes sequence between two loxp sites) and genomic DNA of recombinant virus rPRV-GFP-gI-gE-11K-28K-TK− was co-transfected into vero cells, with the results showing relatively obvious cytopathic effect and more single fluorescence 24 h after transfection. After serial dilution, P0 virus harvested was inoculated for plaque screening; fluorescence-negative plaque was picked for the next round of purification. After 2 rounds of screening and purification, a fluorescence-negative virus was obtained, and named PRV-gI-gE-11K-28K-TK−. PCR identification result after extraction and purification of viral genomic DNA, showed deletion of TK gene, and also indicated that GFP label gene had been deleted. The result showed a successful purification of gI-gE-11K-28K-TK− deleted virus containing no GFP label gene.
  • 2.4 Confirmation of PRV HN1201 Strain With Deletion of gI/gE/11K/28K/TK
  • The primers used for identifying deletion of gI/gE/11K/28K were the same as above.
  • The viral genome of gI/gE/11K/28K/TK-deleted virus and wild-type virus, was extracted and identified by PCR, with the following primers:
    • TKDCF: cctacggcaccggcaagagca
    • TKDCR: cgcccagcgtcacgttgaagac
  • The size of PCR amplification product of the wild-type virus was 1566 bp, the size of PCR amplification fragment of TK deleted virus was 742 bp (refer to FIG. 5).
  • EXAMPLE 3 Preparation of PRV HN1201 Strain With Deletion of gI/gE
  • PRV HN1201 strain with deletion of gI/gE was prepared by reference to the method in Example 1 of CN103756977A.
  • EXAMPLE 4 Pathogenicity Test of Gene-Deleted PRV Strain
  • 25 7-day-old piglets which were negative for pseudorabies antibodies and pseudorabies antigen were randomly divided into 5 groups (A, B, C, D and blank control group), each with 5 piglets. Grouping conditions and challenge conditions are shown in Table 1.
  • TABLE 1
    Grouping of animals in the pathogenicity test
    Group Strain used for inoculation Dose
    A PRV HN1201 strain with inoculated with 1 ml
    deletion of gI/gE/11K/28K (107.0 TCID50/ml)/piglet by
    prepared in Example 1 intranasal instillation
    B PRV HN1201 strain with inoculated with 1 ml
    deletion of (107.0 TCID50/ml)/piglet by
    gI/gE/11K/28K/TK intranasal instillation
    prepared in Example 2
    C PRV HN1201 strain with inoculated with 1 ml
    deletion of gI/gE prepared (107.0 TCID50/ml)/piglet by
    in Example 3 intranasal instillation
    D PRV HN1201 strain inoculated with 1 ml
    (107.0 TCID50/ml)/piglet by
    intranasal instillation
    Blank DMEM medium inoculated with 1 ml/piglet
    control by intranasal instillation
  • After inoculation of virus, the temperature of piglets was determined daily, and clinical signs and death status were observed. The results are shown in Table 2.
  • TABLE 2
    Pathogenicity of different genes-deleted
    PRV HN1201 strains in 7-day-old piglets
    Group Number Clinical signs Death status
    A A1 Normal body temperature, no clinical Survived
    signs
    A2 Body temperature increased for 1 day, Survived
    no clinical signs
    A3 Normal body temperature, no clinical Survived
    signs
    A4 Normal body temperature, no clinical Survived
    signs
    A5 Body temperature increased for 1 day, Survived
    no clinical signs
    B B1 Normal body temperature, no clinical Survived
    signs
    B2 Normal body temperature, no clinical Survived
    signs
    B3 Normal body temperature, no clinical Survived
    signs
    B4 Normal body temperature, no clinical Survived
    signs
    B5 Normal body temperature, no clinical Survived
    signs
    C C1 Body temperature increased for 1 day, Survived
    slightly depression, loss of appetite
    C2 Body temperature increased for 1 day, Survived
    slightly depression, loss of appetite
    C3 Body temperature increased for 1 day, Survived
    slightly depression, loss of appetite
    C4 Body temperature increased for 1 day, Survived
    slightly depression, loss of appetite
    C5 Body temperature increased for 1 day, Survived
    slightly depression, loss of appetite
    D D1 Body temperature increased for 3 days, Died on day
    depression, completely loss of 3 after
    appetite, neurological signs such as challenge
    staying lying, dyspnea, trembling,
    convulsions, turning around, and
    making strokes with their arms
    D2 Body temperature increased for 4 days, Died on day
    depression, completely loss of 4 after
    appetite, staying lying, dyspnea, challenge
    trembling and convulsions.
    D3 Body temperature increased for 4 days, Died on day
    depression, completely loss of 4 after
    appetite, neurological signs such as challenge
    staying lying, dyspnea, trembling,
    convulsions, turning around, and
    making strokes with their arms
    D4 Body temperature increased for 4 days, Died on day
    depression, completely loss of 4 after
    appetite, neurological signs such as challenge
    staying lying, dyspnea, trembling,
    convulsions, turning around, and
    making strokes with their arms
    D5 Body temperature increased for 4 days, Died on day
    depression, completely loss of 4 after
    appetite, neurological signs such as challenge
    staying lying, dyspnea, trembling,
    convulsions, and making strokes with
    their arms
    Blank K1 Normal Survived
    control K2 Normal Survived
    K3 Normal Survived
    K4 Normal Survived
    K5 Normal Survived
  • It showed in the results that inoculation with PRV HN1201 strain in 7-day-old piglets could lead to 100% death (5/5) of inoculated piglets, while the virulence of PRV HN1201 strain with deletion of gI/gE/11K/28K was significantly decreased, which could only make the temperature of 2 piglets increased, without any clinical signs. Inoculation with PRV HN1201 strain with deletion of gI/gE in 7-day-old piglets could still lead to common clinical signs such as increased body temperature and depression etc., indicating remaining virulence; while PRV HN1201 strain with deletion of gI/gE/11K/28K/TK gene had completely lost its virulence.
  • EXAMPLE 5 Preparation of the Live Gene-Deleted PRV Vaccines 5.1 Proliferation of Vaccine Virus
  • The virus seed of PRV HN1201 strain with deletion of gI/gE/11K/28K prepared in Example 1, PRV HN1201 strain with deletion of gI/gE/11K/28K/TK prepared in Example 2 and PRV HN1201 strain with deletion of gI/gE prepared in Example 3 was diluted at 5×104 fold, and then inoculated into a monolayer of ST cell. After 1 h adhesion, 1000 ml of DMEM medium containing 2% fetal calf serum was added into ST cell, which was then placed at 37° C. in a roller bottle with a rotation speed of 6 rph. The cell medium containing viruses was harvested when the cytopathic effect of cells reached to 80%; the viruses were harvested after 2 times of freezing-thawing the medium and the virus titer was assessed. The virus solution was preserved at low temperature.
  • 5.2 Preparation of a Protective Agent
  • 40 g of sucrose and 8 g of gelatin was added into every 100 ml of deionized water, and the solution was autoclaved (under 121° C. for 30 min) after fully melted.
  • 5.3 Preparation of Vaccine Virus Suspension
  • The virus solution prepared and preserved in Example 5.1 was mixed with the protective agent prepared and preserved in Example 5.2 at a volume ratio of 1:1, distributed into sterilized bottles, each of which containing 2.6 ml and the mixed virus solution was freeze-dried. The vaccine was tested and determined to be free of contamination of bacterium and exogenous viruses and the content of virus was consistent with that before freeze-drying. The batch number of PRV HN1201 strain with deletion of gI/gE/11K/28K prepared in Example 1, PRV HN1201 strain with deletion of gI/gE/11K/28K/TK prepared in Example 2 and PRV HN1201 strain with deletion of gI/gE prepared in Example 3 were 20140501, 20140502 and 20140503, respectively.
  • EXAMPLE 6 Immunogenicity Assay of the Live Gene-Deleted PRV Vaccines
  • 12 9-day-old piglets which were negative for PRV antibodies and PRV antigens were randomly divided into 5 groups, each with 5 piglets, and the piglets were injected with the vaccines prepared in Example 5 according to Table 3. The vaccine control group was inoculated with the live PRV vaccine, Bartha K-61strain purchased from HIPRA, Spain, Batch No. 42RH, at the dosage from the protocol. The blank control group was inoculated with 1 mL/piglet of DMEM medium. The piglets were challenged with 1□107.0TCID50/piglet of PRV HN1201 strain on day 28 after immunization. After challenge, the body temperature of piglets was determined daily, and in the meanwhile clinical signs and death status were observed (The results are shown in Table 3), the blood of piglets in all the experimental groups and control groups was collected respectively before challenge.
  • TABLE 3
    Grouping of animals in the pathogenicity test
    Group Vaccines injected Dose
    Group I Batch No. inoculated with 1 ml
    20140501 106.0 TCID50/piglet by
    intramuscular injection
    Group II Batch No. inoculated with 1 ml
    20140502 106.0 TCID50/piglet by
    intramuscular injection
    Group III Batch No. inoculated with 1 ml
    20140503 106.0 TCID50/piglet by
    intramuscular injection
    Vaccine control Live PRV inoculated with 2 ml
    group 2 vaccine 106.0 TCID50/piglet by
    intramuscular injection
    Blank control DMEM inoculated with 1 mL/piglet
    group medium by intramuscular injection
  • The piglets were challenged with 1□107.0TCID50/piglet (1 ml/piglet) of PRV HN1201 strain on day 28 after immunization. After challenge, the body temperature of piglets was determined daily, and in the meanwhile clinical signs and death status were observed (The results are shown in Table 5).
  • TABLE 5
    clinical status and challenge status for piglets challenged
    after immunization with live PRV vaccines
    clinical signs and death
    Group status Rate of protection
    Group I Normal body temperature, 100% (5/5)
    normal appetite, no
    abnormal clinical signs,
    survived
    Group II Normal body temperature, 100% (5/5)
    normal appetite, no
    abnormal clinical signs,
    survived
    Group III After immunization, body 100% (5/5)
    temperature increased,
    slightly depression and
    loss of appetite. After
    challenge, normal body
    temperature, normal
    appetite, no abnormal
    clinical signs, survived
    Vaccine Body temperature of three  80% (4/5)
    control piglets increased for 7-10
    group days, depression, loss of
    appetite, one died.
    Blank Body temperature of three  0% (0/5)
    control piglets increased for 7-10
    group days, depression in all
    piglets, partially or
    completely loss of
    appetite, significant
    clinical signs, two piglets
    died on day 4 after
    challenge, and all died
    within 5 days after
    challenge.
  • The result from Table 5 indicated that immunizing piglets with the gene-deleted PRV vaccines prepared in example 5 can blocked virus infection (i.e. displaying clinical signs), and provide 100% (5/5) protection rate for piglets, while all the piglets in the blank control group died by day 5 after challenge, therefore the PRV vaccines in three experimental groups can provide excellent protection, showing excellent immune protection and safety; meanwhile it indicated that either deletion of gI/gE/11K/28K or deletion of gI/gE/11K/28K/TK for PRV strain would not affect the immunogenicity. For the vaccine group with only deletion of gI/gE, the clinical signs such as increased body temperature could not be avoided, while the vaccine still possessed good immunogenicity. Whereas the commercial vaccines in the prior art cannot provide a full protection to pigs.
  • EXAMPLE 7 Construction of Gene-Deleted Variant Strains of NVDC-PRV-BJ Strain, NVDCPRV-HEB Strain and NVDC-PRV-SD Strain, HN1202 PRV Variant Strain
  • gI/gE/11K/28K genes and gI/gE/11K/28K/TK genes were deleted from the parent strains, NVDC-PRV-BJ strain, NVDC-PRV-HEB strain and NVDC-PRV-SD strain (Xiuling Yu, Zhi Zhou, Dongmei Hu,et al. Pathogenic Pseudorabies Virus, China, 2012 Emerging Infectious Diseases, www.cdc.gov/eid ol. 20, No. 1, January 2014) (the applicant promises to open it to public for 20 year from the patent application date according to provisions of Guidelines for Patent Examination), HN1202 strain (deposited in the China Center for Type Culture Collection on Aug. 26, 2013, of which the accession number is CCTCC NO. V 201335 and the address is Wuhan University, Wuhan, China), according to methods in Example 1 and 2. The names of the attenuated strains obtained were NVDC-PRV-BJ with deletion of gI/gE/11K/28K/TK, NVDCPRV-HEB with deletion of gI/gE/11K/28K/TK, NVDC-PRV-SD with deletion of gI/gE/11K/28K/TK, and PRVHN1202 with deletion of gI/gE/11K/28K/TK. The deletion of genes was verified through comparison of PCR results with that of parent strains respectively.
  • EXAMPLE 8 Preparation of Vaccine Compositions of the Attenuated Variant Strains of NVDC-PRV-BJ Strain, NVDC-PRV-HEB Strain and NVDC-PRV-SD Strain, HN1202 PRV Strain
  • Each attenuated vaccine strains prepared in Example 7 was proliferated according to the method from Example 5.1, mixed with the protective agent (prepared by adding 40 g of sucrose and 8 g of gelatin into every 100 ml of deionized water, and autoclaved (under 121° C. for 30 min) after fully melted) at a volume ratio of 1:1 and the mixed vaccine compositions were freeze-dried. The batch numbers of NVDC-PRV-BJ strain with deletion of gI/gE/11K/28K/TK, NVDCPRV-HEB strain with deletion of gI/gE/11K/28K/TK, NVDC-PRV-SD strain with deletion of gI/gE/11K/28K/TK and PRV HN1201 strain with deletion of gI/gE/11K/28K/TK were Q01, Q02, Q03 and Q04, respectively.
  • EXAMPLE 9 Pathogenicity Test of the Virus Strains Prepared in Example 7
  • Pathogenicity test was conducted according to the method in Example 4, in which the piglets were randomly divided into 5 groups, each with 5 piglets, inoculated with 1 ml (107.0TCID50/ml) of NVDC-PRV-BJ strain with deletion of gI/gE/11K/28K/TK, NVDC-PRV-HEB strain with deletion of gI/gE/11K/28K/TK, NVDC-PRV-SD strain with deletion of gI/gE/11K/28K/TK, and PRV HN1202 strain with deletion of gI/gE/11K/28K/TK by intranasal instillation, respectively. The results showed that all the piglets were alive in each group, with normal body temperature and no clinical signs. It proved that the virulence of mutated PRV strain was reduced through deletion of gI/gE/11K/28K/TK genes.
  • EXAMPLE 10 Immunogenicity Assay of the Vaccines Prepared in Example 8
  • Immunogenicity assay of the vaccines prepared in Example 8 was conducted according to the method and dose in Example 6, in the meanwhile the piglets in the vaccine control group were inoculated with the live PRV vaccine, HB-98 strain Batch No. 1308011-1 (purchased from China Animal Husbandry Industry Co., Ltd. Chengdu Medical Equipments Factory). The piglets were challenged with 1□107.0TCID50/piglet of PRV HN1201 strain on day 28 after immunization. After challenge, the body temperature of piglets was determined daily, and in the meanwhile clinical signs and death status were observed (the results are shown in Table 6).
  • TABLE 6
    clinical status and challenge status for piglets challenged
    after immunization with live PRV vaccines
    clinical signs and death Rate of
    Group Vaccines status protection
    Group IV Q01 Normal body 100% (5/5)
    temperature, normal
    appetite, no abnormal
    clinical signs, survived
    Group V Q02 Normal body 100% (5/5)
    temperature, normal
    appetite, no abnormal
    clinical signs, survived
    Group VI Q03 Normal body 100% (5/5)
    temperature, normal
    appetite, no abnormal
    clinical signs, survived
    Group VII Q04 Normal body 100% (5/5)
    temperature, normal
    appetite, no abnormal
    clinical signs, survived
    Vaccine the live PRV Body temperature of five  80% (4/5)
    control vaccine, HB- piglets increased for 7-10
    group 98 strain days, loss of appetite, one
    Batch No. piglet died and four
    1308011-1 survived.
    Blank DMEM Body temperature of all  0% (0/5)
    control medium piglets increased,
    group depression in all piglets,
    partially or completely
    loss of appetite,
    significant clinical signs,
    two piglets died on day 4
    after challenge, and all
    died within 5 days after
    challenge.
  • The result from Table 6 indicated that immunizing piglets with the PRV vaccines prepared in Example 8 can block virus infection (i.e. displaying clinical signs), and provide 100% (5/5) protection rate for piglets, while the vaccine control group can only provide 80% (4/5) protection rate for piglets, and all the piglets in the blank control group died by day 5 after challenge, therefore the PRV vaccines of the present invention can provide excellent protection. In addition, the piglets exhibited substantially no clinical signs, indicating excellent immune protection of the PRV vaccines relative to live vaccines in the prior art.
  • EXAMPLE 11 Monitoring of gB Antibodies After Immunization With Different Strain Vaccines
  • 15 piglets at the age of around 13 days which were negative for PRV antigens and PRV antibodies were randomly divided into 5 groups, each with 5 piglets. Groups 1-3 were injected with the vaccine prepared in Example 5,which is PRV HN1201 strain with deletion of gI/gE/11K/28K/TK, with Batch No. 20140502, the live PRV vaccine Bartha K-61 strain, with Batch No. 66KR, purchased from HIPRA, Spain, and the live PRV vaccine, K-61, with Batch No. 195-B59B purchased from Boehringer Ingelheim (US) respectively. All the dose for immunization is 1 ml/piglet (for commercial vaccine, 1 piglet dosage/piglet, according to protocols; the PRV HN1201 with deletion of gI/gE/11K/28K/TK vaccine, 106.0TCID50/piglet). The blank control group was inoculated with 1 mL/piglet of DMEM medium. The blood of piglets was collected on day 8, 10, 12, 14 and 21 after immunization, and gB antibody was determined according to the protocol of gB ELISA antibody detection kit (purchased from Biochek, Batch No. F S5763, Expiry Date: Jan. 7, 2015) after the serum was separated. The detailed results of detection are shown in Table 7 below.
  • TABLE 7
    Results of detection of gB antibodies of piglets after immunization.
    Before Day 8 after Day 10 after
    immunization immunization immunization
    Group No. of piglet OD405nm S/P OD405nm S/P OD405nm S/P
    PRV HN1201 1# 0.184 0.025 0.398 0.469 0.439 0.555
    strain with 2# 0.170 −0.004 0.369 0.409 0.453 0.584
    deletion of 3# 0.172 0.000 0.263 0.189 0.360 0.390
    gI/gE/11K/28K/ 4# 0.181 0.019 0.320 0.307 0.494 0.669
    TK vaccine 5# 0.182 0.021 0.339 0.347 0.400 0.474
    with Batch No.
    20140502
    Bartha K-61 6# 0.177 0.010 0.223 0.106 0.243 0.147
    7# 0.176 0.008 0.256 0.174 0.286 0.237
    8# 0.167 −0.010 0.224 0.108 0.246 0.154
    9# 0.186 0.029 0.221 0.102 0.219 0.098
    10# 0.175 0.006 0.242 0.145 0.277 0.218
    K-61 11# 0.162 −0.019 0.195 0.059 0.185 0.035
    12# 0.16 −0.023 0.174 0.009 0.192 0.052
    13# 0.167 −0.007 0.182 0.028 0.218 0.113
    14# 0.16 −0.023 0.199 0.068 0.201 0.073
    15# 0.17 0.000 0.219 0.115 0.225 0.129
    Day 12 after Day 14 after Day 21 after
    immunization immunization immunization
    Group No. of piglet OD405nm S/P OD405nm S/P OD405nm S/P
    PRV HN1201 1# 0.471 0.621 0.678 1.051 1.069 1.863
    strain with 2# 0.510 0.702 0.631 0.953 0.984 1.686
    deletion of 3# 0.453 0.584 0.496 0.673 0.619 0.928
    gI/gE/11K/28K/ 4# 0.596 0.881 0.687 1.070 0.844 1.396
    TK vaccine, 5# 0.602 0.893 0.547 0.779 0.690 1.076
    Batch No.
    20140502
    Bartha K-61 6# 0.275 0.214 0.290 0.245 0.570 0.827
    7# 0.302 0.270 0.317 0.301 0.418 0.511
    8# 0.283 0.231 0.309 0.285 0.315 0.297
    9# 0.211 0.081 0.223 0.106 0.316 0.299
    10# 0.272 0.208 0.299 0.264 0.486 0.652
    K-61 11# 0.239 0.162 0.274 0.244 0.314 0.338
    12# 0.205 0.082 0.211 0.096 0.277 0.251
    13# 0.248 0.183 0.25 0.188 0.449 0.655
    14# 0.256 0.202 0.285 0.27 0.321 0.354
    15# 0.28 0.258 0.3 0.305 0.385 0.505
    Note:
    evaluation criteria: negative, S/P value ≦0.499; positive, S/P value ≧0.500.
  • In conclusion, the antibody test results showed that, all gB antibodies turned positive on day 12 after immunization with PRV HN1201 strain with deletion of gI/gE/11K/28K/TK, while not all the gB antibodies had turned positive on day 21 after immunization with the two control vaccine. It showed that PRV HN1201 strain with deletion of gI/gE/11K/28K/TK could provide earlier immune protection.
  • EXAMPLE 12 Monitoring of gE Antibodies After Immunization With Four Genes Deleted Strain Vaccine and Challenge
  • 15 piglets at the age of around 13 days which were negative for PRV antigens and PRV antibodies were randomly divided into 3 groups, each with 5 piglets. Groups 1-3 were injected with the vaccine prepared in Example 5,which is PRV HN1201 strain with deletion of gI/gE/11K/28K/TK, with Batch No. 20140502, the live PRV vaccine, Bartha K-61 strain, with Batch No. 66KR, purchased from HIPRA, Spain, and the live PRV vaccine, K-61, with Batch No. 195-B59B purchased from Boehringer Ingelheim (US). All the dose for immunization is 1 ml/piglet (for commercial vaccine, 1 piglet dosage/piglet, according to protocols; the PRV HN1201 with deletion of gI/gE/11K/28K/TK vaccine, 106.0TCID50/piglet). The piglets were challenged with 107.0TCID50/piglet, 1 ml/piglet of PRV HN1201 strain on day 21 after immunization. The blood of piglets was collected daily continuously from day 7 to day 14 after challenge, and gE antibody was determined according to the protocol of gE ELISA antibody detection kit (purchased from IDEXX Co., Batch No. AK650, Expiry Date: Jun. 13, 2015) after the serum was separated. The results showed that gE antibody was still negative (If the value of S/N is less or equal to 0.60, the sample should be determined as PRV gE antibody positive) on Day 14 after challenge when the piglets were immunized with the vaccine prepared in Example 5, PRV HN1201 with deletion of gI/gE/11K/28K/TK with Batch No. 20140502, while gE antibody became positive at different level when the piglets were immunized with the two commercial vaccines. The detailed results of deletion are shown in Table 8 below.
  • TABLE 8
    Results of detection of gE antibody of piglets after immunization.
    No Before Day 7 after Day 8 after Day 9 after Dayb10 after
    of challenge challenge challenge challenge challenge
    Group piglet OD650nm S/N OD650nm S/N OD650nm S/N OD650nm S/N OD650nm S/N
    PRV 1# 1.024 1.041 0.917 0.932 0.956 0.972 0.860 0.874 0.863 0.877
    HN1201 2# 1.006 1.008 0.979 0.980 0.931 0.932 0.889 0.890 0.780 0.781
    with 3# 1.070 1.072 0.990 0.991 1.007 1.009 0.970 0.971 0.929 0.930
    deletion 4# 1.052 1.054 0.795 0.796 0.899 0.872 0.972 0.943 1.000 0.970
    of 5# 0.969 0.970 0.912 0.913 0.915 0.916 0.922 0.923 0.683 0.684
    gI/gE/11K/
    28K/TK,
    vaccine
    with
    Batch
    No.
    20140502
    Bartha 6# 1.045 1.078 0.634 0.654 0.684 0.706 0.587 0.606 0.518 0.535
    K-61 7# 1.063 1.097 0.788 0.813 0.758 0.782 0.664 0.685 0.612 0.632
    8# 1.008 1.040 0.897 0.926 0.857 0.884 0.784 0.809 0.783 0.808
    9# 1.017 1.050 0.720 0.743 0.637 0.657 0.599 0.618 0.467 0.482
    10# 0.987 1.019 0.871 0.899 0.701 0.723 0.656 0.677 0.655 0.676
    K-61 11# 0.905 0.934 0.946 0.976 0.698 0.720 0.643 0.664 0.618 0.638
    12# 1.024 1.057 0.898 0.927 0.773 0.798 0.688 0.710 0.760 0.784
    13# 1.030 1.063 0.957 0.928 0.965 0.936 0.913 0.886 0.732 0.710
    14# 0.963 0.934 0.757 0.734 0.899 0.872 0.972 0.943 1.000 0.970
    15# 0.944 0.916 0.747 0.725 0.591 0.573 0.543 0.527 0.531 0.515
    Day 11 after Day 12 after Day 13 after Day 14 after
    No of challenge challenge challenge challenge
    Group piglet OD650nm S/N OD650nm S/N OD650nm S/N OD650nm S/N
    PRV 1# 0.884 0.898 0.877 0.891 0.871 0.885 0.880 0.894
    HN1201 2# 0.854 0.855 0.780 0.781 0.793 0.794 0.732 0.733
    with 3# 0.907 0.908 0.905 0.906 0.904 0.905 1.067 1.069
    deletion 4# 0.965 0.936 0.864 0.838 0.997 0.967 0.929 0.901
    of 5# 0.623 0.624 0.718 0.719 0.784 0.785 0.718 0.719
    gI/gE/11K/
    28K/
    TK
    vaccine,
    with
    Batch
    No.
    20140502
    Bartha 6# 0.552 0.570 0.482 0.497 0.463 0.478 0.456 0.471
    K-61 7# 0.664 0.685 0.533 0.550 0.499 0.515 0.478 0.493
    8# 0.749 0.773 0.647 0.668 0.700 0.722 0.753 0.777
    9# 0.450 0.464 0.410 0.423 0.432 0.446 0.433 0.447
    10# 0.633 0.653 0.699 0.721 0.684 0.706 0.676 0.698
    K-61 11# 0.568 0.586 0.472 0.487 0.472 0.487 0.449 0.463
    12# 0.745 0.769 0.659 0.680 0.659 0.680 0.714 0.737
    13# 0.785 0.761 0.678 0.658 0.505 0.490 0.425 0.412
    14# 0.965 0.936 0.864 0.838 0.997 0.967 0.929 0.901
    15# 0.578 0.561 0.528 0.512 0.457 0.443 0.398 0.386
  • The above results indicated that the vaccine strains in the present invention has a better immunogenicity than commercial vaccine in the prior art, and after immunization therewith a faster generation of the antibody can be achieved, and the effective amplification of virus in the body of pigs can be blocked, and gE antibody is negative.
  • Those are only preferred embodiments of the present invention as described above, which cannot be used to limit the present invention. Any change, substitution or modification etc., which are within the spirit and principle of the invention, should be included within the scope of protection of the present invention.

Claims (17)

What is claimed is:
1. An attenuated strain of porcine pseudorabies virus comprising a first attenuated porcine pseudorabies virus strain, wherein the first attenuated strain of porcine pseudorabies comprises a porcine pseudorabies virus strain with inactivation of gI/gE/11K/28K protein.
2. The attenuated strain of porcine pseudorabies virus of claim 1, wherein the first attenuated porcine pseudorabies virus strain is a variant strain of pseudorabies virus.
3. The attenuated strain of porcine pseudorabies virus of claim 1, wherein a whole ORF is deleted from a gI/gE/11K/28K gene of the first attenuated porcine pseudorabies virus strain.
4. The attenuated strain of porcine pseudorabies virus of claim 2, wherein the variant strain of pseudorabies virus comprises a virus strain in which gE protein has 95% or greater homology with SEQ ID NO. 05.
5. The attenuated strain of porcine pseudorabies virus of claim 4, wherein said variant strain of pseudorabies virus is obtained through isolation, and when infection with said variant strain occurs in pigs previously immunized with a second attenuated gene deleted porcine pseudorabies virus strain, wherein the second attenuated gene deleted porcine pseudorabies virus strain is prepared according to the prior art, the pigs display clinical signs of infection selected from the group consisting of high fever, depression, and partial or complete loss of appetite.
6. The attenuated strain of porcine pseudorabies virus of claim 5, wherein when exposed to said variant strain of pseudorabies virus, pigs previously immunized with the second attenuated porcine pseudorabies virus strain develop a pseudorabies infection, and wherein the second attenuated porcine pseudorabies vaccine is a prior art strain with deletion of one or more of the group consisting of gE, TK and gI genes.
7. The attenuated strain of claim 6, wherein the pseudorabies infection comprises clinical signs of infection selected from the group consisting of depression and loss of appetite among piglets at the age of 9-10 days.
8. The attenuated strain of porcine pseudorabies virus of claim 1, wherein the first porcine pseudorabies virus strain is selected from the group consisting of an HN1201 strain, HN1202 strain, JS-2012 strain, PRV HeN1 strain, NVDC-PRV-BJ strain, NVDCPRV-HEB strain or NVDC-PRV-SD strain, PRV TJ strain, and PRV-ZJ01 strain.
9. The attenuated strain of porcine pseudorabies virus of claim 1, wherein the first attenuated strain of porcine pseudorabies virus strain comprises a gene variation resulting in inactivation of TK protein.
10. The attenuated strain of porcine pseudorabies virus of claim 9, wherein the gene variation comprises a nucleotide sequence located at a site corresponding to a TK gene of the first attenuated porcine pseudorabies virus strain, and wherein the nucleotide sequence encodes for SEQ ID NO. 4.
11. A vaccine composition, comprising an immunizing amount of an antigen of an attenuated strain of porcine pseudorabies and a carrier, wherein the attenuated strain of porcine pseudorabies comprises a genetic variation resulting in inactivation of gI/gE/11K/28K protein.
12. The vaccine composition of claim 11, wherein the immunizing amount comprises at least 106.0TCID50/ml of the attenuated strain of porcine pseudorabies virus.
13. The vaccine composition of claim 11, wherein the antigen comprises a live attenuated strain of porcine pseudorabies virus, and wherein the vaccine composition further comprises a cryoprotectant.
14. The vaccine composition of claim 11, further comprising an inactivated pathogen or antigen.
15. The vaccine composition of claim 14, wherein the inactivated pathogen or antigen is selected from the group consisting classical swine fever virus, antigen of porcine reproductive and respiratory syndrome virus, antigen of porcine circovirus, antigen of haemophilus parasuis, and antigen of mycoplasma.
16. A method of treating and preventing pseudorabies infection, comprising immunizing a pig with an antigen comprising a porcine pseudorabies virus strain with inactivation of gI/gE/11K/28K protein.
17. The method of claim 16, wherein the porcine pseudorabies virus strain comprises a virus strain in which gE protein has 95% or greater homology with SEQ ID NO. 05.
US15/054,404 2014-08-22 2016-02-26 Gene-Deleted Variant Strain Of Porcine Pseudorabies Virus, Vaccine Composition, Method Of Making The Same And Use Thereof Abandoned US20160279232A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/054,404 US20160279232A1 (en) 2014-08-22 2016-02-26 Gene-Deleted Variant Strain Of Porcine Pseudorabies Virus, Vaccine Composition, Method Of Making The Same And Use Thereof
US15/213,662 US10201605B2 (en) 2014-08-22 2016-07-19 Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN201410418379.8 2014-08-22
CN201410418379.8A CN104250640A (en) 2014-08-22 2014-08-22 Porcine pseudorabies virus gene deletion strain, vaccine composition, preparation method and application thereof
PCT/CN2015/070221 WO2016026264A1 (en) 2014-08-22 2015-01-06 Porcine pseudorabies virus gene deletion strain, vaccine composition, and preparation method therefor and application thereof
US14/901,981 US10548972B2 (en) 2014-08-22 2015-01-06 Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof
US15/054,404 US20160279232A1 (en) 2014-08-22 2016-02-26 Gene-Deleted Variant Strain Of Porcine Pseudorabies Virus, Vaccine Composition, Method Of Making The Same And Use Thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2015/070221 Continuation WO2016026264A1 (en) 2014-08-22 2015-01-06 Porcine pseudorabies virus gene deletion strain, vaccine composition, and preparation method therefor and application thereof
US14/901,981 Continuation US10548972B2 (en) 2014-08-22 2015-01-06 Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/213,662 Continuation US10201605B2 (en) 2014-08-22 2016-07-19 Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof

Publications (1)

Publication Number Publication Date
US20160279232A1 true US20160279232A1 (en) 2016-09-29

Family

ID=52185894

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/901,981 Active US10548972B2 (en) 2014-08-22 2015-01-06 Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof
US15/054,404 Abandoned US20160279232A1 (en) 2014-08-22 2016-02-26 Gene-Deleted Variant Strain Of Porcine Pseudorabies Virus, Vaccine Composition, Method Of Making The Same And Use Thereof
US15/213,662 Active US10201605B2 (en) 2014-08-22 2016-07-19 Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/901,981 Active US10548972B2 (en) 2014-08-22 2015-01-06 Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/213,662 Active US10201605B2 (en) 2014-08-22 2016-07-19 Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof

Country Status (7)

Country Link
US (3) US10548972B2 (en)
EP (1) EP3012322B1 (en)
JP (1) JP6368725B2 (en)
CN (2) CN104250640A (en)
DK (1) DK3012322T3 (en)
TW (1) TW201612316A (en)
WO (1) WO2016026264A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109750005A (en) * 2018-12-26 2019-05-14 河南农业大学 A kind of porcine pseudorabies recombinant virus and its construction method
WO2022221612A1 (en) * 2021-04-16 2022-10-20 Zortis Services Llc Pseudorabies virus vaccine

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104250640A (en) 2014-08-22 2014-12-31 普莱柯生物工程股份有限公司 Porcine pseudorabies virus gene deletion strain, vaccine composition, preparation method and application thereof
CN111560355A (en) * 2015-03-20 2020-08-21 普莱柯生物工程股份有限公司 Porcine pseudorabies virus weakening method, porcine pseudorabies virus weakening virus strain, porcine pseudorabies virus vaccine composition and application of porcine pseudorabies virus weakening virus strain
CN106282132A (en) * 2015-05-11 2017-01-04 中国农业科学院上海兽医研究所 The low virulent strain of Pseudorabies virus variant and application thereof
CN106310248B (en) * 2015-06-18 2019-12-20 普莱柯生物工程股份有限公司 Application of porcine pseudorabies virus live vaccine composition in treating porcine pseudorabies
CN108251382B (en) * 2016-12-29 2021-08-20 普莱柯生物工程股份有限公司 Porcine pseudorabies virus weakening method, porcine pseudorabies virus weakening virus strain, porcine pseudorabies virus vaccine composition and application of porcine pseudorabies virus weakening virus strain
CN107828741B (en) * 2017-11-07 2021-01-12 江苏省农业科学院 Pseudorabies virus double-gene deletion attenuated strain and application thereof
CN108553641A (en) * 2018-05-15 2018-09-21 贵州都匀市黔昌畜牧发展有限责任公司 A kind of pseudoabies immunization method
CN109337874B (en) * 2018-12-10 2020-05-12 畜科生物工程有限公司 Recombinant porcine pseudorabies virus, application thereof and recombinant porcine pseudorabies live vaccine
CN109609468B (en) * 2018-12-10 2020-06-30 畜科生物工程有限公司 Six-gene-deleted porcine pseudorabies virus, porcine pseudorabies vaccine and preparation method thereof
CN109750007A (en) * 2019-02-01 2019-05-14 华南农业大学 The Pseudorabies virus gE/gI deletion mutation strain and its building and application of double expression gC gene
CN110241090B (en) * 2019-05-07 2023-10-13 江苏南农高科技股份有限公司 Method for producing porcine pseudorabies virus antigen by full suspension cell culture
CN110257345B (en) * 2019-07-18 2021-07-16 河南农业大学 Porcine pseudorabies double-gene deletion mutation virus strain and construction method thereof
CN110527669B (en) * 2019-09-06 2021-07-06 中牧实业股份有限公司 Porcine pseudorabies virus gene deletion strain and construction method and application thereof
CN112831477A (en) * 2019-11-25 2021-05-25 中国农业科学院哈尔滨兽医研究所(中国动物卫生与流行病学中心哈尔滨分中心) Recombinant pseudorabies virus strain expressing classical swine fever virus E2 protein and porcine circovirus Cap protein as well as construction method and application thereof
CN111154733A (en) * 2020-01-09 2020-05-15 天津瑞普生物技术股份有限公司 Pseudorabies virus attenuated strain and application thereof
CN111748529B (en) * 2020-06-19 2022-01-11 国药集团动物保健股份有限公司 Porcine pseudorabies virus strain and application thereof
CN112342201B (en) * 2020-11-04 2023-01-03 武汉科前生物股份有限公司 Porcine pseudorabies attenuated strain prepared through CRISPR/Cas9 and application thereof
CN112501133B (en) * 2020-12-01 2022-05-24 山东信得科技股份有限公司 Pseudorabies virus QD strain three-gene deletion weakening strain
CN113444697A (en) * 2021-04-19 2021-09-28 温氏食品集团股份有限公司 Recombinant porcine pseudorabies virus and preparation method and application thereof
CN113957171A (en) * 2021-10-26 2022-01-21 苏州药明检测检验有限责任公司 Method for determining pseudorabies virus titer by tissue half infection method staining
CN116064557A (en) * 2022-09-15 2023-05-05 华中农业大学 Application of preparation for activating pig OGFOD2 gene expression in preparation of pig anti-pseudorabies virus infection medicine
CN116790509B (en) * 2023-06-28 2024-02-02 中国农业科学院哈尔滨兽医研究所(中国动物卫生与流行病学中心哈尔滨分中心) Monoclonal hybridoma cell strain secreting anti-porcine pseudorabies virus gB protein antibody and application thereof
CN117384863B (en) * 2023-10-18 2024-04-05 武汉科前生物股份有限公司 Porcine pseudorabies virus natural passage attenuated variant JS18-150 and application thereof
CN117126818B (en) * 2023-10-25 2024-02-02 江西农业大学 Method for constructing gE gene deletion PRV strain by utilizing ABE and application
CN117106736B (en) * 2023-10-25 2024-02-20 江西农业大学 Method for constructing three-gene deletion PRV strain by using CBE and application

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5674500A (en) * 1992-07-09 1997-10-07 Akzo Nobel N.V. Vaccines against Aujeszky's disease and other animal diseases containing pseudorabies virus mutants
CN103627678A (en) * 2013-12-19 2014-03-12 姜平 Porcine pseudorabies virus (PRV) variant PRV-ZJ01 and application thereof
US20140093535A1 (en) * 2011-05-27 2014-04-03 Sinovet (Beijing) Biotechnology Co., Ltd Combined vaccines for prevention of porcine virus infections
CN103923884A (en) * 2014-02-21 2014-07-16 普莱柯生物工程股份有限公司 Porcine pseudorabies virus gene deletion strain, vaccine composition, and preparation method and application of vaccine composition

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE8205892D0 (en) 1982-10-18 1982-10-18 Bror Morein IMMUNOGENT MEMBRANE PROTEIN COMPLEX, SET FOR PREPARATION AND USE THEREOF
US4514497B1 (en) * 1983-12-30 1998-02-24 Novagene Inc Modified live pseudorabies viruses
FR2781159B1 (en) 1998-07-06 2000-10-06 Merial Sas CIRCOVIRUS VACCINE AND PIG PARVOVIRUS
CN100354414C (en) * 2005-09-29 2007-12-12 华中农业大学 Pseudo-rabies gE/gI-gene loss poison strain, killed vaccine containing it and use
EP2018182A4 (en) 2006-05-19 2010-10-06 Inst Medical W & E Hall Immunogenic compositions
CN101186902B (en) * 2006-07-04 2010-11-10 四川农业大学 Pseudorabies virus SA215, pseudorabies virus polygene deletion bacterin and preparation method thereof
KR20100094587A (en) 2007-12-21 2010-08-26 와이어쓰 엘엘씨 Methods and compositions for immunizing pigs against porcine circovirus
WO2009143865A1 (en) 2008-05-30 2009-12-03 Xigen S.A. Use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of various diseases
CN102994458B (en) 2012-11-26 2014-04-02 中国农业科学院哈尔滨兽医研究所 Porcine pseudorabies virus virulent strain, and gene deletion vaccine strain thereof and applications thereof
CN103756977B (en) 2013-12-11 2016-01-06 姜平 Porcine pseudorabies variant gE and gI genetically deficient virus strain and application thereof
CN103981153B (en) 2014-05-29 2017-07-07 中国兽医药品监察所 The structure of pseudorabies virus double fluorescence labeling deleted virus
CN104250640A (en) * 2014-08-22 2014-12-31 普莱柯生物工程股份有限公司 Porcine pseudorabies virus gene deletion strain, vaccine composition, preparation method and application thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5674500A (en) * 1992-07-09 1997-10-07 Akzo Nobel N.V. Vaccines against Aujeszky's disease and other animal diseases containing pseudorabies virus mutants
US20140093535A1 (en) * 2011-05-27 2014-04-03 Sinovet (Beijing) Biotechnology Co., Ltd Combined vaccines for prevention of porcine virus infections
CN103627678A (en) * 2013-12-19 2014-03-12 姜平 Porcine pseudorabies virus (PRV) variant PRV-ZJ01 and application thereof
CN103923884A (en) * 2014-02-21 2014-07-16 普莱柯生物工程股份有限公司 Porcine pseudorabies virus gene deletion strain, vaccine composition, and preparation method and application of vaccine composition

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
Brideau et al., “The Us9 Gene Product of Pseudorabies Virus, an Alphaherpesvirus, Is a Phosphorylated, Tail-Anchored Type II Membrane Protein," Journal of Virology Vol. 72, No. 6: 4560-4570 (1998) *
Dong et al., "An Overview of Live Attenuated Recombinant Pseudorabies Viruses for Use as Novel Vaccines," Journal of Immunology Research 2014;2014;824630 (2014) *
Google English translation of CN 103627678 A (2013) *
Google English translation of CN 103923884 A (2014) *
Lomniczi et al., “Deletions in the Genomes of Pseudorabies Virus Vaccine Strains and Existence of Four Isomers of the Genomes,” Journal of Virology, Vol. 49, No. 3: 970-979 (1984) *
Luo et al., “Pathogenicity and genomic characterization of a pseudorabies virus variant isolated from Bartha-K-61-vaccinated swine population in China,” Veterinary Microbiology 174: 107-115 (2014) *
Maresch et al., “Oral immunization of wild boar and domestic pigs with attenuated live vaccine protects against Pseudorabies virus infection,” Veterinary Microbiology 151: 20-25 (2012) *
Wang et al., “A novel gE-deleted pseudorabies virus (PRV) provides rapid and complete protection from lethal challenge with the PRV variant emerging in Bartha-K61-vaccinated swine population in China," Vaccine 32: 3379 (2014) *
Wu et al., “Emergence of virulent pseudorabies virus infection in northern China," J. Vet. Sci 14(3): 363-365 (2013) *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109750005A (en) * 2018-12-26 2019-05-14 河南农业大学 A kind of porcine pseudorabies recombinant virus and its construction method
WO2022221612A1 (en) * 2021-04-16 2022-10-20 Zortis Services Llc Pseudorabies virus vaccine

Also Published As

Publication number Publication date
US10548972B2 (en) 2020-02-04
US20160317650A1 (en) 2016-11-03
CN104862286B (en) 2018-09-11
TW201612316A (en) 2016-04-01
JP2016531545A (en) 2016-10-13
US20160281065A1 (en) 2016-09-29
EP3012322B1 (en) 2019-08-28
DK3012322T3 (en) 2019-10-07
US10201605B2 (en) 2019-02-12
EP3012322A1 (en) 2016-04-27
WO2016026264A1 (en) 2016-02-25
CN104250640A (en) 2014-12-31
CN104862286A (en) 2015-08-26
EP3012322A4 (en) 2016-07-20
JP6368725B2 (en) 2018-08-01

Similar Documents

Publication Publication Date Title
US10201605B2 (en) Gene-deleted variant strain of porcine pseudorabies virus, vaccine composition, method of making the same and use thereof
US9650424B2 (en) Porcine pseudorabies virus, vaccine composition and preparation method and use thereof
US9801936B2 (en) Method of attenuating porcine pseudorabies virus, attenuated strains of porcine pseudorabies virus, vaccine composition and use thereof
US10449246B2 (en) Subunit vaccine of porcine pseudorabies virus and preparation method
JP6096176B2 (en) Combination vaccine for the prevention of swine virus infection
CN104248762B (en) A kind of pig epidemic diarrhea vaccine combination and its preparation method and application
CN105018433B (en) Porcine pseudorabies virus gene-deleted strain, vaccine composition and its preparation method and application
JP2017511328A (en) Porcine epidemic diarrhea virus vaccine
CA3046684A1 (en) Porcine coronavirus vaccines
CN106148287B (en) Porcine epidemic diarrhea virus strain and vaccine composition, preparation method and application thereof
CN110387355B (en) Recombinant porcine pseudorabies virus strain and vaccine for expressing porcine circovirus Cap protein gene as well as preparation method and application thereof
CN111491663A (en) Swine influenza A virus vaccine
CN108251382B (en) Porcine pseudorabies virus weakening method, porcine pseudorabies virus weakening virus strain, porcine pseudorabies virus vaccine composition and application of porcine pseudorabies virus weakening virus strain
CN105018436B (en) Porcine pseudorabies virus gene-deleted strain, vaccine composition and its preparation method and application
EP3897710A1 (en) Prime-boost vaccination regimen

Legal Events

Date Code Title Description
AS Assignment

Owner name: PULIKE BIOLOGICAL ENGINEERING, INC., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TIAN, KEGONG;ZHANG, XUKE;SUN, JINZHONG;AND OTHERS;REEL/FRAME:037837/0381

Effective date: 20151224

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION