AU4206489A - Recombinant vaccinia virus for prevention of disease caused by flavivirus - Google Patents

Recombinant vaccinia virus for prevention of disease caused by flavivirus

Info

Publication number
AU4206489A
AU4206489A AU42064/89A AU4206489A AU4206489A AU 4206489 A AU4206489 A AU 4206489A AU 42064/89 A AU42064/89 A AU 42064/89A AU 4206489 A AU4206489 A AU 4206489A AU 4206489 A AU4206489 A AU 4206489A
Authority
AU
Australia
Prior art keywords
virus
dengue
recombinant
flavivirus
vaccinia
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.)
Granted
Application number
AU42064/89A
Other versions
AU624863B2 (en
Inventor
Michael P. Bray
Robert M. Chanock
Ching-Juh Lai
Bangti Zhao
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.)
US Department of Commerce
Original Assignee
US Department of Health and Human Services
US Government
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 US Department of Health and Human Services, US Government filed Critical US Department of Health and Human Services
Publication of AU4206489A publication Critical patent/AU4206489A/en
Application granted granted Critical
Publication of AU624863B2 publication Critical patent/AU624863B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24141Use of virus, viral particle or viral elements as a vector
    • C12N2710/24143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2770/24011Flaviviridae
    • C12N2770/24211Hepacivirus, e.g. hepatitis C virus, hepatitis G virus
    • C12N2770/24222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Description

RECOMBINANT VACCINIA VIRUS FOR PREVENTION
OF DISEASE CAUSED BY FLAVIVIRUS
Technical Field
The present invention is related to the construction of recombinant vaccinia viruses. More particularly, the present invention is related to the construction of recombinant vaccinia viruses which are useful for the preparation of a vaccine for the prevention of diseases caused by flaviviruses, such as dengue virus, Japanese B encephalitis virus and tick-borne encephalitis virus.
Background of the Invention
Certain diseases of public health concern, such as dengue disease and encephalitis of certain types are known to be caused by viruses belonging to the flavivirus family. It has been estimated that up to 100 million illnesses occur every year in tropical areas of the world due to dengue virus alone. However, there is no effective and specific immunoprophylactic measure to control such flavivirus diseases.
SUMMARY OF THE INVENTION
It is, therefore, an object of the present invention to provide a recombinant vaccinia virus which induces an immune response against a specific flavivirus disease in a host infected with said recombinant vaccinia virus.
Other objects and advantages of the present invention will become evident from the following detailed description of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
These and other objects, features and many of the attendant advantages of the invention will be better understood upon a reading of the following detailed description when considered in connection with the accompanying drawings wherein:
Figure 1 shows schematic construction of a dengue virus-vaccinia virus recombinant plasmid. The intermediate cloning vector pscll contains interrupted thymidine kinase gene sequences (TKR amd TKL) and a bacterial β-galactosidase gene (LacZ) under the transcription control of the P11 vaccinia virus promoter. The vector DNA was partially digested with BamHI to open the BamHI site downstream of the P7.5 promoter for insertion of the 4.1 kilobase (Kb} fragment of dengue virus DNA located at the 5' end of the viral genone. This fragment of dengue cDNA contains the coding region for the three dengue structural proteins [capsid (C), pre-membrane (pre-M), and envelope glycoprotein (E)] plus the first two downstream non-structural proteins NS1 and NS2a. Recombinant plasmid containing the dengue virus DNA insert in the sense transcription orientation was selected and used for construction of the recombinant vaccinia virus.
Figures 2A and 2B demonstrate the identification of dengue virus proteins synthesized by the recombinant vaccinia virus. (Figure 2A: ) Immunoprecipitation of [35S]-methionine-labeled lysate from recombinant vaccinia virus-infected cells (multiplicity of infection, 10 PFU per cell) was carried out with one of the following specific antibodies, monoclonal antibody 1H10, specific for the envelope glycoprotein (E); rabbit antibodies prepared against dengue virus type 2 nonstructural protein l(NSι); monoclonal antibody 5C9 specific for the membrane glycoprotein precursor (preM); and polyvalent antibodies (P). The labeled precipitates were analyzed on sodium dodecyl sulfate-12% polyacrylamide gels. Also shown are the labeled dengue virus protein markers (D) obtained by immunoprecipitation with polyvalent antibodies of [35S]-methionine-labeled lysate from dengue virus-infected CV-1 cells. (Figure 2B: ) Labeled immunoprecipitates prepared as described above were analyzed by digestion with endoglycosidase H (endo H) and by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Numbers at the left of the gels indicate protein sizes in kilodaltons. DETAILED DESCRIPTION OF THE INVENTION
The above and various other objects and advantages of the present invention are achieved by a recombinant vaccinia virus containing complete coding sequence for the expression of the major specific protective flavivirus antigens in cells infected with said recombinant vaccinia virus, said antigens inducing protective immunity against infection by said flavivirus in a susceptible host.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned hereunder are incorporated herein by reference. Unless mentioned otherwise, the techniques employed herein are standard methodologies well known to one of ordinary skill in the art.
Like other members of the flavivirus family, extracellular dengue virus has a relatively simple structure. Virions contain only three virus-coded proteins, designated capsid (C) protein, membrane (M) protein, and envelope (E) glycoprotein. Intracellular virus, which is also infectious, lacks M but contains another glycoprotein, preM, from which M is derived by cleavage. Both C and M are internal proteins. The surface envelope glycoprotein is the major site responsible for neutralization of infectivity by specific antibodies. The envelope glycoprotein also exhibits hemagglutinating activity and is responsible for adsorption to the cell surface. Several nonstructural proteins have also been identified in dengue virus-infected cells. Among them is nonstructural protein NS1, which is a glycoprotein and described as a soluble complement-fixing antigen. It is believed that NS1 plays an important role in mediating immunity, since the analogous NS1 glycoprotein of yellow fever virus has been shown to be a protective antigen in mice and in primates. Furthermore, the NS1 of dengue type 2 virus has been shown to be a protective antigen.
The dengue type 4 virus genome consists of a molecule of positive-stranded RNA 10,644 nucleotides in length. A full-length cDNA copy of the dengue type 4 virus has been prepared, and its complete nucleotide sequence determined. From such studies, it has been estimated that 96% of the dengue virus genome codes for a polyprotein which is cleaved by specific protease(s) to generate individual viral proteins. The three structural proteins, C-M-E, are located at the amino terminus, while nonstructural proteins NS1-NS2a-NS2b-NS3-NS4a-NS4b-NS5 are at the carboxy-terminus. It must be emphasized that because dengue virus gene expression involves proteolytic cleavage of the polyprotein, it cannot be assumed a priori that the expression of protective antigens such as E and NS1 from their cloned DNA sequences, would be useful in immunoprophylaxis. Such determination can be made only through actual testing. The construction of a vaccinia flavivirus recombinant is now illustrated by employing dengue virus genes as an example .
Vaccinia virus was used as a vector for construction of a live recombinant virus expressing dengue viral genes. The BglII DNA fragment (4,041 base pairs, nucleotides 88 to 4128) from the 5' terminus of dengue type 4 virus cDNA contains the coding region for the three structural proteins as well as nonstructural proteins NS1 and NS2a. This fragment was excised from the full-length dengue virus DNA copy and was inserted into the PSC11 vaccinia intermediate vector. The dengue virus DNA sequence was inserted in the BamHI site immediately downstream of the vaccinia P7.5 early-late promoter (Figure 1). In this construct, the dengue virus coding sequence was placed under the transcriptional control of the vaccinia virus P7.5 early-late promoter. The vector contained a bacterial -galactosidase gene under the control of the vaccinia virus P11 late promoter, which provided a visual selectable marker. The chimeric genes were flanked by sequences of the vaccinia virus thymidine kinase gene, which directed homologous recombination of dengue virus sequences into the vaccinia virus genome following transfection of simian CV-1 cells previously infected with wild type vaccinia virus (WR strain). Recombinant vaccinia virus harboring the dengue virus DNA insert was isolated and plaque purified two times on thymidine kinase minus (TK) cells in selective medium. Other dengue cDNA fragments, vide infra, were inserted into a vaccinia recombinant virus by the method just described. Similarly, constructs of other flavivirusvaccinia recombinants are made by inserting flavivirus cDNA following the procedure described herein for dengue virus.
Dengue virus-specific proteins synthesized during infection with the recombinant virus were initially detected by indirect immunofluorescence. CV-1 cells infected with the recombinant virus exhibited fluorescent-stainable antigens in the cytoplasm when polyvalent dengue type 4 virus hyperimmune mouse antiserum was used, although the intensity of staining was less than that observed in dengue virus-infected cells. Similar immunofluorescence was observed in CV-1 cells infected with the recombinant virus when monoclonal antibody specific to the E glycoprotein was used.
To further identify this and other dengue virus proteins, recombinant virus-infected cells were radiolabeled with [35]methionine and the cell lysate was prepared for immunoprecipitation by specific antibodies. Analysis of the labeled precipitate on sodium dodecyl sulfate-polyacrylamide gel (Figure 2A) showed that the polyvalent hyperimmune mouse antiserum precipitated three major bands with estimated molecular size 20, 40 to 46, and 55 to 60 kilodaltons (kDa), respectively. Minor bands of larger molecular size that were probably nonspecific were also observed. When an E glycoproteinspecific monoclonal antibody (1H10) was used, a 55 to 60kDa band consistent with the molecular size of the E glycoprotein was precipitated. Similarly, dengue type 2 virus NS1-specific antiserum (obtained from Dr. J. Schlesinger) precipitated a 40- to 46-kDa band, which is the predicted size for the NS1 nonstructural glycoprotein. The third major band precipitated by dengue type 4 virus hyperimmune antiserum was approximately 20 kDa which is consistent with the size of the intracellular preM glycoprotein. A similar band was precipitated by preM-specific antibodies (monoclonal antibody 2H2 or 5C9, obtained from Dr. M. K. Gentry of WRAIR).
Thus, in recombinant virus-infected cells, the three glycoproteins encoded in the cloned DNA appeared to be cleaved and modified by glycosylation in a manner similar to that observed during dengue virus infection. This suggests that the dengue viral structural proteins, as well as the NS1 nonstructural protein, were specifically processed by proteolytic cleavage of the polyprotein in the absence of dengue virus functions provided by the distal nonstructural proteins NS2b through NS5. However, as estimated by immunoprecipitation, the amount of dengue preM, E nad NS1 glycoproteins produced in recombinant virus infected cells was significantly less than in dengue type 4 virus-infected cells.
The pattern of glycosylation of the glycoproteins produced in recombinant virus-infected cells varied as indicated by their response to endoglycosidase H, which cleaves the mannose rich carbohydrate core. The preM protein band was completely sensitive to endoglycosidase H treatment, yielding a band of 17 kDa, a reduction of 3 kDA in molecular size. On the other hand, a significant portion of the carbohydrate of both the E and the NS1 glycoproteins appeared to be resistant to endoglycosidase H digestion.
Each of the dengue virus glycoproteins, i.e., preM, E and NS1, is preceded by a stretch of hydrophobic amino acids which can serve as a signal. This indicates that cellular signalase is probably responsible for proteolytic cleavage. Presumably, the cleavage mechanism which generates the three glycoproteins, also yields the capsid protein, which is located amino terminal to the preM glycoprotein.
As described herein above, initially a recombinant vaccinia virus containing a 4.0 Kb sequence from the 5' terminus of cDNA that codes for the three structural proteins [capsid (C), membrane (M), and envelope (E)] and nonstructural proteins NS1 and NS2a was constructed. Subsequently, another recombinant vaccinia virus expressing only the three structural proteins was constructed by deleting the coding sequence for nonstructural proteins. Protein analysis showed that cells infected with the first recombinant virus v(C-M-E-NS1NS2a) produced authentic pre-membrane (preM), envelope (E), and NS1 glycoproteins as detected by radioimmunoprecipitation using specific antibodies. These dengue proteins showed a glycosylation pattern similar to that found for these proteins produced during dengue virus infection. The second recombinant virus (C-M-E) produced dengue virus preM and E glycoproteins. As expected, NS1 was not synthesized by the second recombinant (Table 2).
The mouse model of dengue encephalitis was employed to evaluate protective immunity induced by these recombinants. Mice were inoculated by the intraperitoneal route with 107 plaque forming units (pfu) of recombinant vaccinia virus. Mice were reinoculated with the same dose of recombinant two weeks later. One week after the second inoculation themice were challenged with 100 LD50 of denque virus intracerebrally. The animals were then observed for signs of encephalitis and death. Ninety-one percent of mice immunized with a control vaccinia recombinant virus expressing the envelope glycoprotein of HIV died after challenge (Tables 1 and 2). In contrast, 96% of mice immunized with the vaccinia (C-M-ENS1-NS2a) recombinant that expressed the three structural proteins and NS1, survived. Also, all 15 animals immunized with the vaccinia (C-M-E) recombinant virus that expressed only the three structural proteins were completely protected. These results indicate that structural proteins alone are able to induce protective immunity. Since C and M are internal proteins, it is concluded that E is the major antigen responsible for protection conferred by the v(C-M-E) recombinant. This interpretation is supported by the complete resistance to intracerebral challenge induced by immunization with faccinia recombinants that expressed only the E glycoprotein. Data in Table 1 provide such evidence. A recombinant that expressed E including its N-terminal hydrophobic signal sequence, or a respiratory syncytial virus (RSV) G glycoprotein (amino acids 1 through 70) - E fusion protein or an influenza A virus hemagglutinin N terminal signal peptide - E fusion protein induced complete protection against dengue virus challenge.
The seroresponse of animals immunized with these recombinants was tested by radioimmunoprecipitation of labeled antigens. Each of the animals immunized with the v(C-M-E-NS1-NS2a) recombinant that expressed the three structural proteins and NS1, developed NS1 antibodies, while the amount of antibody to E was low or not detectable. The low level of E antibody response was further confirmed by other serologic tests such as virus neutralization and ELISA. Also, mice infected with the vaccinia recombinant (C-M-E) that expressed only the three structural proteins developed little or no detectable E antibody.
The nonstructural protein NS1 expressed by various vaccinia recombinant viruses was also evaluated for its antigenic properties and the most promising products were evaluated for their protective effect in mice. A total of 6 NS1 vaccinia recombinants were studied. These recombinants contained an insert in which: (i) NS1 was preceded by the structural protein sequence (C-M-E), (ii) NS1 was preceded only by its immediate upstream hydrophobic sequence or (iii) NS1 was not preceded by structural genes or its immediate upstream hydrophobic sequence. Each of these inserts terminated with the complete NS2a sequence or the N terminal 15% of the NS2a sequence. The v(C-M-E-NS1-NS2a) and v(NS1-NS2a) recombinants produced authentic NS1, whereas other recomtbinants that lacked the NS1 N terminal hydrophobic signal or lacked the complete downstream NS2a sequence yielded a NS1 product of different molecular size and/or were not glycosylated as indicated by gel analysis. From these studies it was concluded that the N-terminal signal sequence of NS1 and the complete downstream NS2a sequence are required for proper processing and proteolytic cleavage of NS1.
The above mentioned NS1 recombinants were then evaluated for their prophylactic potential in mouse protection studies as described above. The results indicated that all but one of 28 mice inoculated with vaccinia (C-M-E-NS1-NS2a) recombinant expressing the structural proteins and NS1 survived dengue challenge (Table 1). Significantly, the recombinant which expressed authentic NS1 in the absence of structural proteins, i.e., v(NS1-NS2a), induced complete resistance to lethal challenge with dengue virus (Table 2). Other vaccinia recombinants that expressed abnormal NS1 induced only partial protection. Thus, vaccination with a vaccinia recombinant that expressed (i) an upstream Nterminal RSV G sequence (amino acids 1-70) fused to NS1 or (ii) NS1 plus 15% of NS2a, induced only partial protection.
These tests demonstrate that dengue envelope (E) glycoprotein and nonstructural protein NS1 are separate protective antigens, each of which can induce complete resistance to encephalitis in the mouse model of dengue disease. These results now allow the development of vaccinia recombinants expressing E and/or NS1 glycoprotein as vaccines for protection of humans against dengue type 4 virus. Accordingly, a vaccine comprises an immunogenic amount of the recombinant vaccinia virus which induces protective immunity against a specific flavivirus in a host infected with said recombinant vaccinia virus. Conventional pharmaceutically acceptable carrier or vehicles such as non-toxic buffers, physiological saline and the like could be used together with adjuvants and booster inoculations, if necessary. Of course, the vaccine can be administered in a single or multiple dosages as indicated.
It is important to point out here that a vaccinia-dengue type 2 virus recombinant that contained the dengue sequences for C,M, and E controlled by a P11 vaccinia virus promoter was recently reported by Deubel et al, 1988 (J. Gen. Virol. 69:1921-1929) and failed to protect monkeys against dengue. In contrast, the recombinant of the present invention was efficacious in protecting the test animals against viral infection.
Since the genome organization, replication strategy and gene expression of all flaviviruses are similar, recombinant vaccinia viruses having prophylactic property against other flaviviruses are made in a manner similar to the herein illustrated vaccinia-dengue construct.
A deposit of the recombinant virus has been made at the American Type Culture Collection (ATCC), 12301 Parklawn Drive, Rockville, Maryland 20852, U.S.A., on August 19, 1988 under the accession number VR 2228. The deposit shall be viably maintained, and replaced if it becomes non-viable, for a period of 30 years from the date of the deposit, or for 5 years from the last date of request for a sample of the deposit, whichever is longer, and made available to the public without restriction in accordance with the provisions of the law. The Commissioner of Patents and Trademarks, upon request, shall have access to the deposit.
It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and the scope of the appended claims.
TABLE 1
MICE INFECTED WITH VACCINIA RECOMBINANTS EXPRESSING
DENGUE ENVELOPE (E) GLYCOPROTEIN WITH OR WITHOUT NONSTRUTCTURAL NS1 GLYCOPROTEIN DEVELOP RESISTANCE TO FATAL CHALLENGE WITH DENGUE VIRUS
NO. WHICH SURVIVED
VACONIA RECOMEXPRESSION OF NO. OF AN INTRACEREBRAL
BINANT EXPRESSING MICE CHALLENGE WITH 102 DENGUE PROTEIN(S) GLYCOAUTHENINFECTED LD50 DENGUE TYPE
SYLATED E TIC NS1 (107 PFU) 4 VIRUS
v(C-M-E-NS1-NS2A) YES YES 28 27 ( 96%) v(C-M-E-NS1-NS2A Δ)* YES NO 15 15 (100%) v(C-M-E) YES - 19 19 (100%) v(E) YES - 15 15 (100%) v(RSV G-E)+ YES - 15 15 (100%) v(FLU HA-E)++ YES - 15 15 (100%) v(HIV GP160)+++ - - 46 4 (8.7%)
*
85% OF C TERMINAL SEQUENCE OF NS2A DELETED.
**
SURVIVING MICE DEVELOPED MILD SIGNS OF ENCEPΗALITIS.
+ EXPRESSES FUSION PROTEIN OF N TERMINUS OF RSV G (AMINO ACIDS 1¬
70) PLUS E.
++ EXPRESSES FUSION PROTEIN OF INFLUENZA A VIRUS N TERMINAL SIGNAL
PEPTIDE PLUS E.
+++ CONTROL VACCINIA RECOMBINANT EXPRESSING ENVELOPE GLYCOPROTEIN OF
HIV.
TABLE 2
MICE INFECTED WITH VACCINIA RECOMBINANT EXPRESSING ONLY
DENGUE NONSTRUCTURAL PROTEIN NS1 DEVELOP RESISTANCE
TO FATAL CHALLENGE WITH DENGUE VIRUS
NO. WHICH SURVIVED AN
VACCINIA RECOMEXPRESSION OF NO. OF MICE INTRACEREBRAL CHALBINANT EXPRESSING AUTHENTIC NS1 INFECTED LENGE WITH 102 LD50 INDICATED (107 PFU) DENGUE TYPE 4 VIRUS DENGUE PROTEIN(S)
v(NS1-NS2A) YES 15 15 (100%)
v(NS1-NS2A Δ)* NO 30 20 ( 67%)
v(RSV G-NS1)+ NO 15 8 ( 54%)
v(HTV GP16O)++ - 46 4 (8.7%)
* 85% OF C TERMINAL SEQUENCE OF NS2A DELETED.
+ EXPRESSES FUSION PROTEIN OF RSV G (AMINO ACIDS 1-70) PLUS NS1.++ CONTROL VACCINIA RECOMBINANT EXPRESSING ENVELOPE GLYCOPROTElN OF
HIV.

Claims (6)

WHAT IS CLAIMED IS:
1. A recombinant vaccinia virus containing complete coding sequence for the expression of flavivirus antigenic proteins.
2. The virus of claim 1 wherein said flavivirus is a dengue type 4 virus.
3. The virus of claim 1 wherein said flavivirus is a Japanese B or tick-borne encephalitis virus.
4. The virus of claim 1 wherein said antigenic protein is selected from the group consisting of a part or whole of an envelope glycoprotein, capsid protein, pre-matrix protein, NS1, NS2 nonstructural protein and a combination thereof.
5. A pharmaceutical composition, comprising an immunogenic amount of the virus of claim 1 in a pharmaceutically acceptable carrier.
6. A method for inducing protective immunity against a flavivirus disease, comprising inoculating a host susceptible to flavivirus with an immunogenic amount of the virus of claim 1 in a single or multiple dosages.
AU42064/89A 1988-08-20 1989-08-21 Recombinant vaccinia virus for prevention of disease caused by flavivirus Ceased AU624863B2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23920588A 1988-08-20 1988-08-20
US239205 1988-08-20

Publications (2)

Publication Number Publication Date
AU4206489A true AU4206489A (en) 1990-03-23
AU624863B2 AU624863B2 (en) 1992-06-25

Family

ID=22901085

Family Applications (1)

Application Number Title Priority Date Filing Date
AU42064/89A Ceased AU624863B2 (en) 1988-08-20 1989-08-21 Recombinant vaccinia virus for prevention of disease caused by flavivirus

Country Status (5)

Country Link
EP (1) EP0431064A4 (en)
JP (1) JPH03503364A (en)
AU (1) AU624863B2 (en)
IL (1) IL91304A0 (en)
WO (1) WO1990001946A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU667836B2 (en) * 1991-09-19 1996-04-18 United States Of America, As Represented By The Secretary, Department Of Health And Human Services, The Chimeric and/or growth-restricted flaviviruses

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7767449B1 (en) 1981-12-24 2010-08-03 Health Research Incorporated Methods using modified vaccinia virus
US6184024B1 (en) 1988-07-14 2001-02-06 The United States Of America As Represented By The Department Of Health And Human Services Chimeric and/or growth-restricted flaviviruses
US6676936B1 (en) 1988-07-14 2004-01-13 The United States Of America As Represented By The Department Of Health And Human Services. Chimeric and/or growth-restricted flaviviruses
HUT54306A (en) * 1989-06-06 1991-02-28 Immuno Ag Process for producing proteins, vaccines and nucleic acids
MY109299A (en) * 1990-08-15 1996-12-31 Virogenetics Corp Recombinant pox virus encoding flaviviral structural proteins
US6117640A (en) * 1995-05-02 2000-09-12 The United States Of America As Represented By The Secretary Of The Army Recombinant vaccine made in E. coli against dengue virus
ES2184869T3 (en) * 1995-05-24 2003-04-16 Hawaii Biotech Group SUB-UNIT VACCINE AGAINST INFECTION WITH FLAVIVIRUS.
ID19548A (en) * 1996-09-24 1998-07-23 Bavarian Nordic Res Inst As RECOMBINED MVA VIRUSES EXPRESSING FEATURES ANTIGENS AND FEVER USING THEMS IN VACCINES
PL192957B1 (en) * 1997-02-28 2006-12-29 Acambis Inc Chimeric flaviviral vaccines
IL139844A0 (en) 1998-06-04 2002-02-10 Us Gov Health & Human Serv Nucleic acid vaccines for prevention of flavivirus infection
US7227011B2 (en) 1998-06-04 2007-06-05 United States Of America As Represented By The Secretary Of The Department Of Health And Human Services, Centers For Disease Control And Prevention Nucleic acid vaccines for prevention of flavivirus infection
EP2278012B1 (en) 2000-02-16 2015-08-26 The Government of the United States of America, as represented by the Secretary, Department of Health & Human Services Immunogenic Dengue 2 virus chimeras
WO2003048184A2 (en) * 2001-12-04 2003-06-12 Bavarian Nordic A/S Flavivirus ns1 subunit vaccine
CA2432738A1 (en) 2003-02-26 2004-08-26 Philippe Despres New dengue and west nile viruses proteins and genes coding the foregoing, and their use in vaccinal, therapeutic and diagnostic applications
WO2014150939A2 (en) 2013-03-15 2014-09-25 Takeda Vaccines, Inc Compositions and methods for dengue virus chimeric constructs in vaccines
US11426461B2 (en) 2018-09-05 2022-08-30 Takeda Vaccines, Inc. Methods for preventing dengue and hepatitis A
US11464815B2 (en) 2018-09-05 2022-10-11 Takeda Vaccines, Inc. Dengue vaccine unit dose and administration thereof
JP2021536463A (en) 2018-09-05 2021-12-27 タケダ ワクチン, インコーポレイテッドTakeda Vaccines, Inc. Dengue vaccine unit dose and its administration
EP3939607A4 (en) * 2019-03-14 2023-01-25 Tokyo Metropolitan Institute of Medical Science Dengue virus vaccine

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH084508B2 (en) * 1987-09-16 1996-01-24 国立予防衛生研究所長 Recombinant vaccinia virus

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU667836B2 (en) * 1991-09-19 1996-04-18 United States Of America, As Represented By The Secretary, Department Of Health And Human Services, The Chimeric and/or growth-restricted flaviviruses

Also Published As

Publication number Publication date
WO1990001946A1 (en) 1990-03-08
AU624863B2 (en) 1992-06-25
JPH03503364A (en) 1991-08-01
EP0431064A4 (en) 1991-11-13
IL91304A0 (en) 1990-03-19
EP0431064A1 (en) 1991-06-12

Similar Documents

Publication Publication Date Title
AU624863B2 (en) Recombinant vaccinia virus for prevention of disease caused by flavivirus
Zhao et al. Expression of dengue virus structural proteins and nonstructural protein NS1 by a recombinant vaccinia virus
JP3531934B2 (en) Chimeric and / or growth-restricted flavivirus
JP4500049B2 (en) Flavivirus NS1 subunit vaccine
JP4504464B2 (en) Chimeric flavivirus vaccine
Falgout et al. Immunization of mice with recombinant vaccinia virus expressing authentic dengue virus nonstructural protein NS1 protects against lethal dengue virus encephalitis
Chen et al. Screening of protective antigens of Japanese encephalitis virus by DNA immunization: a comparative study with conventional viral vaccines
US5494671A (en) C-terminally truncated dengue and Japanese encephalitis virus envelope proteins
Bray et al. Genetic determinants responsible for acquisition of dengue type 2 virus mouse neurovirulence
US8088391B2 (en) West nile virus vaccine
KR20010022452A (en) Recombinant dimeric envelope vaccine against flaviviral infection
Bonaldo et al. Surface expression of an immunodominant malaria protein B cell epitope by yellow fever virus
Jan et al. Increased immunogenicity and protective efficacy in outbred and inbred mice by strategic carboxyl-terminal truncation of Japanese encephalitis virus envelope glycoprotein
Chia et al. Fragment of Japanese encephalitis virus envelope protein produced in Escherichia coli protects mice from virus challenge
US6676936B1 (en) Chimeric and/or growth-restricted flaviviruses
ES2637629T3 (en) Induction of an immune response against dengue virus using a sensitization-enhancement approach
Hahn et al. Expression of the structural proteins of dengue 2 virus and yellow fever virus by recombinant vaccinia viruses
WO1992003161A1 (en) Flavivirus envelope proteins with increased immunogenicity for use in immunization against virus infection
Venugopal et al. Immunity to St. Louis encephalitis virus by sequential immunization with recombinant vaccinia and baculovirus derived PrM/E proteins
AU674134B2 (en) Recombinant viruses comprising artificial proteolytic cleavage site
Kinney The structural genes of Venezuelan equine encephalitis virus: molecular cloning, sequencing and expression in recombinant vaccinia virus
WO1991002819A1 (en) Recombinant japanese encephalitis molecules and methods of use therefor
WGGA et al. 465 JMENTATION PAGE] _