CA2517258C - Recombinant measles virus comprising dengue or west nile virus polynucleotides, and their use in vaccinal and therapeutic applications - Google Patents

Recombinant measles virus comprising dengue or west nile virus polynucleotides, and their use in vaccinal and therapeutic applications Download PDF

Info

Publication number
CA2517258C
CA2517258C CA2517258A CA2517258A CA2517258C CA 2517258 C CA2517258 C CA 2517258C CA 2517258 A CA2517258 A CA 2517258A CA 2517258 A CA2517258 A CA 2517258A CA 2517258 C CA2517258 C CA 2517258C
Authority
CA
Canada
Prior art keywords
virus
thr
gly
val
leu
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.)
Expired - Lifetime
Application number
CA2517258A
Other languages
French (fr)
Other versions
CA2517258A1 (en
Inventor
Frederic Tangy
Philippe Despres
Chantal Combredet
Marie Pascale Frenkiel
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.)
Centre National de la Recherche Scientifique CNRS
Institut Pasteur de Lille
Original Assignee
Centre National de la Recherche Scientifique CNRS
Institut Pasteur de Lille
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
Priority claimed from CA002420092A external-priority patent/CA2420092A1/en
Priority claimed from CA002432738A external-priority patent/CA2432738A1/en
Application filed by Centre National de la Recherche Scientifique CNRS, Institut Pasteur de Lille filed Critical Centre National de la Recherche Scientifique CNRS
Priority to CA2517258A priority Critical patent/CA2517258C/en
Publication of CA2517258A1 publication Critical patent/CA2517258A1/en
Application granted granted Critical
Publication of CA2517258C publication Critical patent/CA2517258C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • 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

Abstract

The present invention relates to the development of viral vectors expressing different immunogens from the West Nile Encephalitis Virus (WNV) or the Dengue virus which are able to induce protective humoral and cellular immune responses against WNV or Dengue virus infections. More specifically, the present invention relates to three (3) antigens from WNV (the secreted envelope glycoprotein (E), the heterodimer glycoproteins (pre-M-E) and the NSI
protein) and from Dengue virus (the secreted envelope glycoprotein (e), the heterodimer glycoproteins (pre~- m-e) and the nsl protein) and their use in vaccinal, therapeutic and diagnostic applications.

Description

RECOMBINANT MEASLES VIRUS COMPRISING DENGUE OR WEST NILE
VIRUS POLYNUCLEOTIDES, AND THEIR USE IN VACCINAL AND
THERAPEUTIC APPLICATIONS

FIELD OF THE INVENTION
The present invention relates to West-Nile virus (WNV) and/or Dengue virus derived peptides, and more particularly to polypeptides or polynucleotides derived from WNV and/or Dengue virus polypeptides or polynucleotides and their use in the preparation of compositions and vaccines. More specifically, the present invention is concerned with compositions, vaccines and methods for providing an immune response and/or a protective immunity to animals against a West-Nile virus or a Dengue virus and methods for the diagnosis of West-Nile virus or Dengue virus infection.

BACKGROUND OF THE INVENTION

Flaviviridae are arboviruses (arthropod-borne virus) mainly transported by mosquitoes and blood-sucking ticks. They are small encapsidated viruses and their genomes consist of infectious single-stranded and linear RNA of positive polarity. In Man, flaviviruses cause deadly hemorrhagic fever or meningo-encephalitis. Yellow fever, dengue fever and japanese encephalitis are the main tropical flaviviroses. Other important human flaviviroses are Saint Louis encephalitis, tick-born European encephalitis and West Nile fever.
West Nile fever is a zoonosis associated with a fiavivirus which was first isolated in Uganda in 1937. Its transmission cycle calls for birds as the main reservoir and for blood sucking mosquitoes of the Culex genus as vectors.
Migratory viremic birds transport the virus to far-away regions where they transmit it anew to ornithophile mosquitoes of the Culex genus. Many species of mammals are permissive for the West Nile virus. Horses are particularly sensitive to the disease but do not participate in the cycle of transmission. West Nile fever is endemic in Africa, Asia, Europe and Australia. Phylogenic studies have revealed the existence of two strains of viruses : viral line 1 has a worldwide distribution, and viral line 2 is essentially African. Viral line 1 was responsible for enzooties in Romania (1996), Russia (1999), Israel (1998-2000) and more recently in North America where the virus had never been detected before 1999. The viral strains isolated during the recent epidemics in Israel and the United-States are more than 99,7 % identical. In the Middle-East and North America, where the virus has taken root, an important bird mortality rate has been observed among infected birds, notably in Corvidae. In North America, over 4000 subjects were infected with the West Nile virus, 250 of which died between the months of August and December 2002. At the present time, zoonosis is observed in all regions of the United States.
At the moment, there exists no human vaccine or specific therapy against West Nile fever.
In temperate and sub-tropical regions, human infections may occur during the fall season. When a subject is bitten by an infected mosquito, the incubation period lasts approximately one week but less than 20 % of people infected with the West Nile virus ever go on to clinical manifestations. In its benignant form, the viral infection manifests itself by an undifferentiated febrile state associated with muscular weakness, headaches and abdominal pain. In less than 1% of infected subjects, encephalitis or acute aseptic meningitis may occur. Splenomegaly, hepatitis, pancreatitis and myocarditis are also observed. Flask paralyses similar to a poliomyelitic syndrome have recently been reported, but fatal cases of viral encephalitis (5% of patients having severe neurological disorders) mainly concern fragile subjects and the aged. Inter-human transmission of the virus has also recently been observed in the United-States in subjects having undergone organ transplants or having been perfused with contaminated blood products. Infra-uterine transmission of the virus has been reported in the United-States. The development of a human vaccine against the West Nile fever is a priority in view of the fact that the zoonosis has taken root in North America and is expected to propagate in the coming months to Central America, South America and the Caribbean where dengue fever and yellow fever are already rampant.
Therefore, there is a need for West-Nile virus (WNV) and/or Dengue virus derived peptides, and more particularly to polypeptides or polynucleotides derived from WNV and/or Dengue virus polypeptides or polynucleotides and their use in the preparation of compositions and vaccines..
The present invention fulfils these needs and also other needs which will be apparent to those skilled in the art upon reading the following specification.

SUMMARY OF THE INVENTION

The present invention relates to West-Nile virus and/or Dengue virus derived polypeptides.
More specifically, one object of the invention concerns a purified polypeptide wherein it derives from a West-Nile virus antigen or a Dengue virus antigen.
Another object of the invention concerns a purified polypeptide wherein it derives from a West-Nile virus antigen, is capable of inducing a protective immune response against a West-Nile virus in an animal; wherein the West-Nile virus antigen is a heterodimer glycoprotein (PreM-E).
Another object of the invention concerns a purified polyclonal or monoclonal antibody capable of specifically binding to a polypeptide of the invention.
Another object of the invention concerns an expression vector comprising a polynucleotide sequence coding for a polypeptide as described therein.
Another object of the invention concerns a purified polynucleotide sequence coding for the polypeptide of the invention and its use for detecting the presence or absence of a West-Nile virus antigen or a Dengue virus antigen in a biological sample.
Another object of the invention concerns a purified polynucleotide sequence coding for a polypeptide as described therein.
Another object of the invention concerns a use of a polynucleotide sequence as defined therein for detecting the presence or absence of a West-Nile virus antigen in a biological sample.
A further object of the invention concerns a recombinant viral vector which is a recombinant virus comprising a polynucleotide sequence of the invention.
Another object of the invention is a recombinant measles virus capable of expressing a polypeptide of the invention or comprising, in its genome, a polynucleotide of the invention.
Yet, another object of the invention relates to a pharmaceutical composition comprising:
a) at least one component selected from the group consisting of:
- a polypeptide of the invention or a functional derivative thereof;
- an antibody of the invention;
- an expression vector of the invention;

- a polynucleotide of the invention or a fragment thereof, - a recombinant viral vector of the invention; and - a recombinant measles virus of the invention;
and b) a pharmaceutically acceptable vehicle or carrier.

Another object of the invention concerns the use of the pharmaceutical composition of the invention, as an anti-West-Nile virus and/or an anti-Dengue virus agent, or for the preparation of an anti-West-Nile virus and/or an anti-Dengue virus vaccine.
Another object of the invention concerns a use of a pharmaceutical composition as described therein, as an anti-West-Nile virus agent, or for the preparation of an anti-West-Nile virus vaccine.
Another object of the invention relates to a host cell incorporating an expression vector as defined therein or a recombinant viral vector as defined therein.
Furthermore, another object of the invention concerns a method of producing a recombinant virus for the preparation of an anti-West-Nile virus vaccine, the method comprising the steps of:
a) providing a host cell as defined therein b) placing the host cell from step a) in conditions permitting the replication of a recombinant virus capable of expressing a polypeptide of the invention; and c) isolating the recombinant virus produced in step b).

Another object of the invention concerns a µAfes.t-Mile virus neutralization assay, comprising the steps of:
a) contacting VERO cells with West-Nile virus and an antibody;
b) culturing said VERO cells under conditions which allow for West-Nile virus replication; and c) measuring reduction of West-Nile virus replication foci on said VERO
cells.

A further object of the invention is to provide a method for treating and/or preventing a WNV- or Dengue virus- associated disease or infection in an animal, the method comprising the step of administering to the animal an effective amount of at least one element selected from the group consisting of:
- a polypeptide or a functional derivative thereof as defined above;
- an antibody as defined above;
- an expression vector as defined above;
- a polynucleotide or a fragment thereof as defined above;
- a recombinant viral vector as defined above; and - a recombinant measles virus as defined above.
Another object of the invention concerns a use of an effective amount of at least one element selected from the group consisting of:
- a polypeptide as described therein or a functional derivative thereof;

5a - an antibody as described therein;
- an expression vector as described therein;
- a polynucleotide as described therein or a fragment thereof;
- a recombinant viral vector as described therein;
and - a recombinant measles virus as described therein;
for treating and/or preventing a WNV-associated disease or infection in an animal.

Another object of the invention concerns a recombinant virus which is a live attenuated or a defective measles virus comprising a polynucleotide coding for a polypeptide which is a West-Nile virus or respectively a Dengue virus antigen which is selected in the group of:
= the heterodimer glycoproteins PreM-E;
= a secreted envelope glycoprotein sE; and = a polypeptide having at least 80% identity, or 85% identity with the polypeptide having the sequence of SEQ ID NO:6, SEQ ID NO: 5 or SEQ
ID NO:8, said recombinant virus expressing said polypeptide and said polypeptide inducing a protective immune response against a West-Nile virus in an animal or respectively against a Dengue virus in an animal.

Another object of the invention concerns a pharmaceutical composition comprising :
a) at least a recombinant measles virus as defined therein; and . CA 02517258 2012-07-13 5b b) a pharmaceutically acceptable vehicle or carrier.
Another object of the invention concerns the use of a pharmaceutical composition as defined therein as an anti-West-Nile virus agent or respectively an anti-Dengue virus agent, or for the preparation of an anti-West-Nile virus vaccine or respectively an anti-Dengue virus vaccine.

Another object of the invention concerns a host cell incorporating a recombinant virus as defined as defined therein.
Another object of the invention concerns a method of producing a recombinant virus for the preparation of an anti-West-Nile virus vaccine, the method comprising the steps of:
a) providing a host cell as defined therein.
b) placing the host cell from step a) in conditions permitting the replication of a recombinant virus as defined therein, which is capable of expressing a said polypeptide; and c) isolating the recombinant virus produced in step b).
Another object of the invention concerns the use of an effective amount of at least a recombinant measles virus as defined therein, for the preparation of a medicament intended for treating and/or preventing a West-Nile virus-associated disease or respectively a Dengue virus associated disease, or infection in an animal.

Another object of the invention concerns the use of an effective amount of at least a recombinant measles virus as defined therein, for treating and/or 5c preventing a West-Nile virus-associated disease or respectively a Dengue virus associated disease, or infection in an animal.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 shows the nucleic acid sequence encoding the secreted glycoprotein E from WNV and identified as SEQ ID NO. 1.
Figure 2 shows the amino acid sequence of the secreted glycoprotein E
from WNV and identified as SEQ ID NO 5.
Figure 3 shows the nucleic acid sequence encoding the preM plus E
glycoproteins from WNV and identified as SEQ ID NO. 2.
Figure 4 shows the amino acid sequence of the preM plus E glycoproteins from WNV and identified as SEQ ID NO 6.
Figure 5 shows the nucleic acid sequence encoding the NS1 protein from WNV and identified as SEQ ID NO. 3.
Figure 6 shows the amino acid sequence of the NS1 protein from WNV and identified as SEQ ID NO 7.
Figure 7 shows the nucleic acid sequence encoding the preM-E gene from Dengue type 1 virus and identified as SEQ ID NO. 4.
Figure 8 shows the amino acid sequence of the preM-E gene from Dengue type 1 virus and identified as SEQ ID NO 8.
Figure 9 is a schematic map of the pTM-MVSchw recombinant plasmids according to preferred embodiments of the invention.
Figure 10 shows the expression of sEWNV by MVSchw-sEwNv recombinant MV in Vero cells. (A) Schematic diagram of MVschw-sEwNv and virus growth. The IS-98-ST1 cDNA coding for sEwNv was inserted into the Schwarz MV genome between the BsiW1 and Bssi-111 sites of the ATU at position 2. The MV genes are indicated: N (nucleoprotein), PVC (phosphoprotein and V, C proteins), M
(matrix), F (fusion), H (hemagglutinin), L (polymerase). T7 : T7 RNA polymerase promoter;

hh : hammerhead ribozyme, T7t : T7 RNA polymerase terminator; : hepatitis delta virus (HDV) ribozjime; ATU : additional transcription unit. (B) Growth curves of MV. Vero cells were infected with MVschw (open box) or MVSchw-sEwNv (black box) at a multiplicity of infection (m.o.i) of 0.01 TCID50/cell. At various times post-infection, infectious virus particles were titered as described in the Methods. (C) Immunofluorescence staining of sEwNv glycoprotein in syncitia of MVschw-sEwNv-infected Vero cells fixed 36 h post-infection. Cells were permeabilized (A, B) or not (C, D) with Triton X-100 and then immunostained using anti-WNV HMAF.
Magnification: x 1000. No positive signal was observed in MVschw-infected cells.
(D) Radioimmunoprecipitation (RIP) assay showing the release of sEwNv from MVschw-sEwNv-infected cells. Vero cells were infected with WNV strain IS-98-(m.o.i of 5) for 24 h, MVschw (m.o.i. of 0.1), MVschw-sEwNv (m.o.i. of 0.1) for 40 h, or mock-infected (MI). Radiolabeled supernatants and cell lysates were immunoprecipitated with specific anti-MV (a-MV) or anti-WNV (a-WNV) polyclonal antibodies. WNV E glycoprotein (open arrow head) and sEwNv (black arrow head) are shown.
Figure 11 shows anti-MVSchw-sEwNv antibodies recognizing the WNV E
glycoprotein. Vero cells were infected with WNV strain 1S-98-ST1 (WNV) or mock-infected (No virus). Labeled cell lysates were immunoprecipitated with pooled immune sera (dilution 1:100) from mice inoculated with WNV, MVSchw, MVSchw-sEwNv as described in the legend to Fig. 10D. Specific anti-lymphochoriom-mingitis virus (LCMV) antibodies were used as a negative control. WNV structural glycoproteins prM and E and non structural proteins NS3, NS5, NS2A and NS2B
are shown. p.c., post-challenge.

DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to West-Nile virus (WNV) and/or Dengue virus derived peptides, and more particularly to polypeptides or polynucleotides derived from WNV and/or Dengue virus polypeptides or polynucleotides and their use in the preparation of compositions and vaccines. More specifically, the present invention is concerned with compositions, vaccines and methods for providing an immune response and/or a protective immunity to animals against a West-Nile virus or a Dengue virus and methods for the diagnosis of West-Nile virus or Dengue virus infection.
As used herein, the term "immune response" refers to the T cell response or the increased serum levels of antibodies to an antigen, or presence of neutralizing antibodies to an antigen, such as a WNV or a Dengue virus antigen. The term "immune response" is to be understood as including a humoral response and/or a cellular response and/or an inflammatory response.
An "antigen" refers to a molecule, such as a protein or a polypeptide, containing one or more epitopes that will stimulate a host's immune system to make a humoral and/or cellular antigen-specific response. The term is also used interchangeably with "immunogen".
The term "protection" or "protective immunity" refers herein to the ability of the serum antibodies and cellular response induced during immunization to protect (partially or totally) against against a West-Nile virus or a Dengue virus.
Thus, an animal immunized by the compositions or vaccines of the invention will experience limited growth and spread of an infectious WNV or Dengue virus.
As used herein, the term "animal" refers to any animal that is susceptible to be infected by a West-Nile virus or a Dengue virus. Among the animals which are known to be potentially infected by these viruses, there are, but not limited to, humans, birds and horses.

1. P lynuci -otid- s and p lypeptides In a first embodiment, the present invention concerns a purified polypeptide characterized in that it derives from a West-Nile virus antigen or a Dengue virus antigen or functional derivative thereof. As can be appreciated, a protein/peptide is said to "derive" from a protein/peptide or from a fragment thereof when such protein/peptide comprises at least one portion, substantially similar in its sequence, to the native protein/peptide or to a fragment thereof.
The West-Nile virus antigen of the present invention is preferably selected from the group consisting of secreted envelope glycoprotein (E), heterodimer glycoproteins (PreM-E) and NS1 protein. More specifically, the secreted envelope glycoprotein (E) comprises the sequence of SEQ ID NO: 5 or a functional derivative thereof, the heterodimer glycoproteins (PreM-E) comprises the = CA 02517258 2012-07-13 sequence of SEQ ID NO: 6 or a functional derivative thereof, and the NS1 protein comprises the sequence of SEQ ID NO: 7 or a functional derivative thereof.

The Dengue virus antigen of the invention is preferably selected from the group consisting of secreted envelope glycoprotein (E), heterodimer glycoproteins (PreM-E) and NS1 protein. More specifically, the heterodimer glycoproteins (PreM-E) comprises the sequence of SEQ ID NO: 8 or a functional derivative thereof.
Accoding to a preferred embodiment, the polypeptide of the present invention has an amino acid sequence having at least 80% homology, or even preferably 85% homology to part or all of SEQ ID NO : 5, of SEQ ID NO : 6, of SEQ ID NO: 7 or of SEQ ID NO :8.
A "functional derivative", as is generally understood and used herein, refers to a protein/peptide sequence that possesses a functional biological activity that is substantially similar to the biological activity of the whole protein/peptide sequence. In other words, it refers to a polypeptide or fragment(s) thereof that substantially retain the same biological functiona as the polypeptide of SEQ
ID

Nos: 5 to 8. A functional derivative of a protein/peptide may or may not contain post-translational modifications such as covalently linked carbohydrate, if such modification is not necessary for the performance of a specific function. The term "functional derivative" is intended to the "fragments", "segments", "variants", "analogs" or "chemical derivatives" of a protein/peptide. As used herein, a protein/peptide is said to be a "chemical derivative" of another protein/peptide when it contains additional chemical moieties not normally part of the protein/peptide, said moieties being added by using techniques well known in the art. Such moieties may improve the protein/peptide solubility, absorption, bioavailability, biological half life, and the like. Any undesirable toxicity and side-effects of the protein/peptide may be attenuated and even eliminated by using such moieties.
Yet, more preferably, the polypeptide comprises an amino acid sequence substantially the same or having 100% identity with SEQ ID NO : 5, SEQ ID NO:
6, SEQ ID NO : 7, or SEQ ID NO: 8.

One can use a program such as the CLUSTAL program to compare amino acid sequences. This program compares amino acid sequences and finds the optimal alignment by inserting spaces in either sequence as appropriate. It is possible to calculate amino acid identity or homology for an optimal alignment. A
program like BLASTx will align the longest stretch of similar sequences and assign a value to the fit. It is thus possible to obtain a comparison where several regions of similarity are found, each having a different score. Both types of identity analysis are contemplated in the present invention.
As used herein, the term "polypeptide(s) " refers to any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds. "Polypeptide(s)" refers to both short chains, commonly referred to as peptides, oligopeptides and oligomers and to longer chains generally referred to as proteins. Polypeptides may contain amino acids other than the 20 gene-encoded amino acids. "Polypeptide(s)" include those modified either by natural processes, such as processing and other post-translational modifications, but also by chemical modification techniques. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature, and they are well known to those of skill in the art.
It will be appreciated that the same type of modification may be present in the same or varying degree at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains, and the amino or carboxyl termini. Modifications include, for example, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation, selenoylation, sulfation and transfer-RNA mediated addition of amino acids to proteins, such as arginylation, and ubiquitination. See, for instance:
PROTEINS--STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W.H.
Freeman and Company, New York (1993); Wold, F., Posttranslational Protein Modifications: Perspectives and Prospects, pgs. 1-12 in POSTTRANSLATIONAL
COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York (1983); Seifter et al., Meth. Enzymol. 182:626-646 (1990); and Rattan et al., Protein Synthesis: Posttranslational Modifications and Aging, Ann.
N.Y. Acad. Sci. 663: 48-62(1992). Polypeptides may be branched or cyclic, with or without branching. Cyclic, branched and branched circular polypeptides may result from post-translational natural processes and may be made by entirely synthetic methods, as well.
With respect to protein or polypeptide, the term "isolated .polypeptide" or "isolated and purified polypeptide" is sometimes used herein. This term refers primarily to a protein produced by expression of an isolated polynucleotide molecule contemplated by invention. Alternatively, this term may refer to a protein which has been sufficiently separated from other proteins with which it would naturally be associated, so as to exist in "substantially pure" form.
The term "substantially pure" refers to a preparation comprising at least 50-60% by weight the compound of interest (e. g., nucleic acid, oligonucleotide, protein, etc.). More preferably, the preparation comprises at least 75% by weight, and most preferably 90-99% by weight, the compound of interest.
Purity is measured by methods appropriate for the compound of interest (e.g. chromatographic methods, agarose or polyacrylamide gel electrophoresis, HPLC analysis, and the like).
In a second embodiment, the present invention concerns a purified polynucleotide encoding a polypeptide of the invention. Therefore, the polynucleotide of the invention has a nucleic acid sequence which is at least 65%
identical, more particularly 80% identical and even more particularly 95%
identical to part or all of any one of SEQ ID NO 1 to 4 or functional fragments thereof.

A "functional fragment", as is generally understood and used herein, refers to a nucleic acid sequence that encodes for a functional biological activity that is substantially similar to the biological activity of the whole nucleic acid sequence. In other words, it refers to a nucleic acid or fragment(s) thereof that substantially retains the capacity of encoding for a polypeptide of the invention.
The term "fragment" as used herein refer to a polynucleotide sequence (e.g., cDNA) which is an isolated portion of the subject nucleic acid constructed artificially (e.g., by chemical synthesis) or by cleaving a natural product into multiple pieces, using restriction endonucleases or mechanical shearing, or a portion of a nucleic acid synthesized by PCR, DNA polymerase or any other polymerizing technique well known in the art, or expressed in a host cell by recombinant nucleic acid technology well known to one of skill in the art.
With reference to polynucleotides of the invention, the term "isolated polynucleotide" is sometimes used. This term, when applied to DNA, refers to a DNA molecule that is separated from sequences with which it is immediately contiguous (in the 5' and 3' directions) in the naturally occurring genome of the organism from which it was derived. For example, the "isolated polynucleotide"
may comprise a DNA molecule inserted into a vector, such as a plasmid or virus vector, or integrated into the genomic DNA of a procaryote or eucaryote. An "isolated polynucleotide molecule" may also comprise a cDNA molecule.
Amino acid or nucleotide sequence "identity" and "similarity" are determined from an optimal global alignment between the two sequences being compared. An optimal global alignment is achieved using, for example, the Needleman-Wunsch algorithm (Needleman and Wunsch, 1970, J. Mol. Biol. 48:443-453). "Identity"
means that an amino acid or nucleotide at a particular position in a first polypeptide or polynucleotide is identical to a corresponding amino acid or nucleotide in a second polypeptide or polynucleotide that is in an optimal global alignment with the first polypeptide or polynucleotide. In contrast to identity, "similarity" encompasses amino acids that are conservative substitutions. A
"conservative" substitution is any substitution that has a positive score in the blosum62 substitution matrix (Hentikoff and Hentikoff, 1992, Proc. Natl. Acad.
Sci.
USA 89: 10915-10919). By the statement "sequence A is n% similar to sequence B" is meant that n% of the positions of an optimal global alignment between sequences A and B consists of identical residues or nucleotides and conservative substitutions. By the statement "sequence A is n% identical to sequence B" is meant that n% of the positions of an optimal global alignment between sequences A and B consists of identical residues or nucleotides.
As used herein, the term "polynucleotide(s)" generally refers to any polyribonucleotide or poly-deoxyribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. This definition includes, without limitation, single-and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions or single-, double- and triple-stranded regions, cDNA, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded, or triple-stranded regions, or a mixture of single-and double-stranded regions. In addition, "polynucleotide" as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules. One of the molecules of a triple-helical region often is an oligonucleotide. As used herein, the term "polynucleotide(s)" also includes DNAs or RNAs as described above that contain one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are "polynucleotide(s)" as that term is intended herein. Moreover, DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein. It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the art. "Polynucleotide(s)" embraces short polynucleotides or fragments comprising at least 6 nucleotides often referred to as oligonucleotide(s). The term "polynucleotide(s)" as it is employed herein thus embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including, for example, simple and complex cells which exhibits the same biological function as the polypeptide encoded by any one of SEQ ID
NOS.1 to 4. The term "polynucleotide(s)" also embraces short nucleotides or fragments, often referred to as "oligonucleotides", that due to mutagenesis are not 100% identical but nevertheless code for the same amino acid sequence.

2. Vectors and Cells In a third embodiment, the invention is also directed to a host, such as a genetically modified cell, comprising any of the polynucleotide sequence according to the invention and more preferably, a host capable of expressing the polypeptide encoded by this polynucleotide. Even more preferably, the present invention is concerned with a host cell that incorporates an expression vector or a recombinant viral vector as defined herein below.
The host cell may be any type of cell (a transiently-transfected mammalian cell line, an isolated primary cell, or insect cell, yeast (Saccharomyces cerevisiae, Ktuyveromyces lactis, Pichia pastoris), plant cell, microorganism, or a bacterium (such as E. co/7). The following biological deposit relating to MEF/3T3.Tet-Off/prME.WN # h2 cell line comprising an expression vector encoding for pseudo-particles of WNV strain IS-98-ST1 composed of prME complexed glycoproteins was registered at the Collection Nationale des Cultures de Microorganismes (CNCM) under accession numbers 1-3018 on May 2, 2003.
In a fourth embodiment, the invention is further directed to cloning or expression vector comprising a polynucleotide sequence as defined above.
As used herein, the term "vector" refers to a polynucleotide construct designed for transduction/transfection of one or more cell types. Vectors may be, for example, "cloning vectors" which are designed for isolation, propagation and replication of inserted nucleotides, "expression vectors" which are designed for expression of a nucleotide sequence in a host cell, or a "viral vector" which is designed to result in the production of a recombinant virus or virus-like particle, or "shuttle vectors", which comprise the attributes of more than one type of vector.
A number of vectors suitable for stable transfection of cells and bacteria are available to the public (e.g. plasmids, adenoviruses, baculoviruses, yeast baculoviruses, plant viruses, adeno-associated viruses, retroviruses, Herpes Simplex Viruses, Alphaviruses, Lentiviruses), as are methods for constructing such cell lines. It will be understood that the present invention encompasses any type of vector comprising any of the polynucleotide molecule of the invention..
According to a preferred embodiment, the vector is a recombinant viral vector which is a recombinant virus comprising a polynucleotide sequence as defined above. Preferably the recombinant virus is a live attenuated virus or a defective virus, such as a recombinant virus selected from the group consisting of measles virus, hepatitis B virus, human papillomavirus, picornaviridae and lentivirus. More preferably, the recombinant virus is a recombinant measles virus, for instance the Schwarz measles virus strain, which is capable of expressing a polypeptide as defined above or comprises, in its genome, a polynucleotide as defined above.

3. Antibodies In a fifth embodiment, the invention features purified antibodies that specifically bind to the isolated or purified polypeptide as defined above or fragments thereof. The antibodies of the invention may be prepared by a variety of methods using the polypeptides described above. For example, the West-Nile or Dengue virus antigen, or antigenic fragments thereof, may be administered to an animal in order to induce the production of polyclonal antibodies.
Alternatively, antibodies used as described herein may be monoclonal antibodies, which are prepared using hybridoma technology (see, e.g., Hammerling at al., In Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, NY, 1981).
As mentioned above, the present invention is preferably directed to antibodies that specifically bind to a West-Nile antigen or a Dengue virus antigen, or fragments thereof. In particular, the invention features "neutralizing"
antibodies.
By "neutralizing" antibodies is meant antibodies that interfere with any of the biological activities of any of the WNV antigen or Dengue virus antigen. Any standard assay known to one skilled in the art may be used to assess potentially neutralizing antibodies. Once produced, monoclonal and polyclonal antibodies are preferably tested for specific WNV or Dengue virus proteins recognition by Western blot, inirnunoprecipitation analysis or any other suitable method.
Antibodies that recognize WNV or Dengue virus proteins expressing cells and antibodies that specifically recognize WNV or Dengue virus proteins (or functional fragments thereof), such as those described herein, are considered useful to the invention. Such an antibody may be used in any standard immunodetection method for the detection, quantification, and purification of WNV
or Dengue virus proteins. The antibody may be a monoclonal or a polyclonal antibody and may be modified for diagnostic purposes. The antibodies of the invention may, for example, be used in an immunoassay to monitor WNV or Dengue virus proteins expression levels, to determine the amount of WNV or Dengue virus proteins or fragment thereof in a biological sample and evaluate the presence or not of a WNV or Dengue virus. In addition, the antibodies may be coupled to compounds for diagnostic and/or therapeutic uses such as gold particles, alkaline phosphatase, peroxidase for imaging and therapy. The antibodies may also be labeled (e.g. imrnunofluorescence) for easier detection.
With respect to antibodies of the invention, the term "specifically binds to"
refers to antibodies that bind with a relatively high affinity to one or more epitopes of a protein of interest, but which do not substantially recognize and bind molecules other than the one(s) of interest. As used herein, the term "relatively high affinity" means a binding affinity between the antibody and the protein of interest of at least 106 M-1, and preferably of at least about 107 M-1 and even more preferably 108 M-1 to 1010 M. Determination of such affinity is preferably conducted under standard competitive binding immunoassay conditions which is common knowledge to one skilled in the art. As used herein, "antibody" and "antibodies" include all of the possibilities mentioned hereinafter:
antibodies or fragments thereof obtained by purification, proteolytic treatment or by genetic engineering, artificial constructs comprising antibodies or fragments thereof and artificial constructs designed to mimic the binding of antibodies or fragments thereof. Such antibodies are discussed in Co!cher et al. (Q J Nucl Med 1998;
42:
225-241). They include complete antibodies, F(ab1)2 fragments, Fab fragments, Fv fragments, scFy fragments, other fragments, CDR peptides and mimetics. These can easily be obtained and prepared by those skilled in the art. For example, enzyme digestion can be used to obtain F(ab1)2 and Fab fragments by subjecting an IgG molecule to pepsin or papain cleavage respectively. Recombinant antibodies are also covered by the present invention.
Alternatively, the antibody of the invention may be an antibody derivative.
Such an antibody may comprise an antigen-binding region linked or not to a non-immunoglobulin region. The antigen binding region is an antibody light chain variable domain or heavy chain variable domain. Typically, the antibody comprises both light and heavy chain variable domains, that can be inserted in constructs such as single chain Fv (scFv) fragments, disulfide-stabilized Fv (dsFv) fragments, multimeric scFv fragments, diabodies, minibodies or other related forms (Colcher et al. Q J Nucl Med 1998; 42: 225-241). Such a derivatized antibody may sometimes be preferable since it is devoid of the Fc portion of the natural antibody that can bind to several effectors of the immune system and elicit an immune response when administered to a human or an animal. Indeed, derivatized antibody normally do not lead to immuno-complex disease and complement activation (type III hypersensitivity reaction).
Alternatively, a non-immunoglobulin region is fused to the antigen-binding region of the antibody of the invention. The non-immunoglobulin region is typically a non-immunoglobulin moiety and may be an enzyme, a region derived from a protein having known binding specificity, a region derived from a protein toxin or indeed from any protein expressed by a gene, or a chemical entity showing inhibitory or blocking activity(ies) against WNV or Dengue virus proteins. The two regions of that modified antibody may be connected via a cleavable or a permanent linker sequence.
Preferably, the antibody of the invention is a human or animal immunoglobulin such as IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgE or IgD carrying rat or mouse variable regions (chimeric) or CDRs (humanized or "animalized").
Furthermore, the antibody of the invention may also be conjugated to any suitable carrier known to one skilled in the art in order to provide, for instance, a specific delivery and prolonged retention of the antibody, either in a targeted local area or for a systemic application.
The term "humanized antibody" refers to an antibody derived from a non-human antibody, typically murine, that retains or substantially retains the antigen-binding properties of the parent antibody but which is less immunogenic in humans. This may be achieved by various methods including (a) grafting only the non-human CDRs onto human framework and constant regions with or without retention of critical framework residues, or (b) transplanting the entire non-human variable domains, but "cloaking" them with a human-like section by replacement of surface residues. Such methods are well known to one skilled in the art.
As mentioned above, the antibody of the invention is immunologically specific to the polypeptide of the present invention and immunological derivatives thereof. As used herein, the term "immunological derivative" refers to a polypeptide that possesses an immunological activity that is substantially similar to the immunological activity of the whole polypeptide, and such immunological activity refers to the capacity of stimulating the production of antibodies immunologically specific to the WNV or Dengue virus proteins or derivative thereof. The term "immunological derivative" therefore encompass "fragments", "segments", "variants", or "analogs" of a polypeptide.

4. Compositions and vaccines The polypeptides of the present invention, the polynucleotides coding the same, the polyclonal or monoclonal antibodies, the recombinant measles virus produced according to the invention, may be used in many ways for the diagnosis, the treatment or the prevention of WNV- or Dengue virus- associated diseases or infection.
In a sixth embodiment, the present invention relates to a composition for eliciting an immune response or a protective immunity against a WNV or a Dengue virus. According to a related aspect, the present invention relates to a vaccine for preventing and/or treating a WNV- or Dengue virus- associated disease or infection. As used herein, the term "treating" refers to a process by which the symptoms of a WNV- or Dengue virus- associated disease or infection are alleviated or completely eliminated. As used herein, the term "preventing"
refers to a process by which a WNV- or Dengue virus- associated disease or infection is obstructed or delayed. The composition or the vaccine of the invention comprises a polynucleotide, a polypeptide, an expression vector, a recombinant viral vector, a recombinant measles virus and/or an antibody as defined above and an acceptable carrier.
As used herein, the expression "an acceptable carrier" means a vehicle for containing the polynucleotide, the polypeptide, the expression vector, the recombinant viral vector, the recombinant measles virus and/or the antibody of the invention that can be injected into an animal host without adverse effects.
Suitable carriers known in the art include, but are not limited to, gold particles, sterile water, saline, glucose, dextrose, or buffered solutions. Carriers may include auxiliary agents including, but not limited to, diluents, stabilizers (i. e., sugars and amino acids), preservatives, wetting agents, emulsifying agents, pH buffering agents, viscosity enhancing additives, colors and the like.
Further agents can be added to the composition and vaccine of the invention. For instance, the composition of the invention may also comprise agents such as drugs, immunostimulants (such as a-interferon, 13-interferon, y-interferon, granulocyte macrophage colony stimulator factor (GM-CSF), macrophage colony stimulator factor (M-CSF), interleukin 2 (IL2), interleukin 12 (IL12), and CpG

oligonucleotides), antioxidants, surfactants, flavoring agents, volatile oils, buffering agents, dispersants, propellants, and preservatives. For preparing such compositions, methods well known in the ark may be used.
The amount of polynucleotide, polypeptide, expression vector, recombinant viral vector, recombinant measles virus and/or antibody present in the compositions or in the vaccines of the present invention is preferably a therapeutically effective amount. A therapeutically effective amount of the polynucleotide, the polypeptide, the expression vector, the recombinant viral vector, the recombinant measles virus and/or the antibody of the invention is that amount necessary to allow the same to perform their immunological role without causing, overly negative effects in the host to which the composition is administered. The exact amount of polynucleotide, polypeptide, expression vector, recombinant viral vector, recombinant measles virus and/or antibody to be used and the composition/vaccine to be administered will vary according to factors such as the type of condition being treated, the mode of administration, as well as the other ingredients in the composition.
5. Methods of use In a seventh embodiment, the present invention relates to methods for treating and/or preventing a WNV- or Dengue virus- associated disease or infection in an animal are provided. The method comprises the step of administering to the animal an effective amount of at least one element selected from the group consisting of:
- a polypeptide of the invention or a functional derivative thereof;
- an antibody as defined above;
- an expression vector as defined above;
- a polynucleotide of the invention or a fragment thereof, - a recombinant viral vector of the invention; and - a recombinant measles virus of the invention.

The vaccine, antibody and composition of the invention may be given to an animal through various routes of administration. For instance, the composition may be administered in the form of sterile injectable preparations, such as sterile injectable aqueous or oleaginous suspensions. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparations may also be sterile injectable solutions or suspensions in non-toxic parenterally-acceptable diluents or solvents. They may be given parenterally, for example intravenously, intramuscularly or sub-cutaneously by injection, by infusion or per os. The vaccine and the composition of the invention may also be formulated as creams, ointments, lotions, gels, drops, suppositories, sprays, liquids or powders for topical administration. They may also be administered into the airways of a subject by way of a pressurized aerosol dispenser, a nasal sprayer, a nebulizer, a metered dose inhaler, a dry powder inhaler, or a capsule. Suitable dosages will vary, depending upon factors such as the amount of each of the components in the composition, the desired effect (short or long term), the route of administration, the age and the weight of the animal to be treated. Any other methods well known in the art may be used for administering the vaccine, antibody and the composition of the invention.
The present invention is also directed to a method of producing a recombinant virus for the preparation of an anti-West-Nile virus vaccine or an anti-Dengue virus vaccine, the method comprising the steps of:
a) providing a host cell as defined above;
b) placing the host cell from step a) in conditions permitting the replication of a recombinant virus capable of expressing a polypeptide according to the invention; and c) isolating the recombinant virus produced in step b).

In a further embodiment, a West-Nile virus neutralisation assay is provided.
Accordingly, the assay comprises the steps of:
a) contacting VERO cells with West-Nile virus and an antibody;
b) culturing said VERO cells under conditions which allow for West-Nile virus replication; and c) measuring reduction of West-Nile virus replication foci on said VERO
cells.

EXAMPLES
The present invention will be more readily understood by referring to the following examples. These examples are illustrative of the wide range of applicability of the present invention and are not intended to limit its scope.
Modifications and variations can be made therein without departing from the spirit and scope of the invention. Although any methods and materials similar or equivalent to those described herein can be used in the practice for testing of the present invention, the preferred methods and materials are described.
EXAMPLE 1: CONSTRUCTION OF MEASLES VIRUSES (MV) EXPRESSING

In order to test their capacity as vaccine candidates against WNV infection, recombinant Schwarz measles viruses (MV) expressing these WNV and DEN-1 antigens were constructed. The different genes were introduced in an additional transcription unit in the Schwarz MV cDNA that the inventors previously cloned (pTM-MVSchw) (European Patent Application N 02291551.6 filed on June 20, 2002). After rescue of the different recombinant Schwarz measles viruses expressing the WNV and DEN-1 genes, their capacity to protect mice from a lethal WNV intraperitoneal challenge, and monkeys from Dengue virus infection will be tested.

MV vector Mass vaccination with live attenuated vaccines has reduced the incidence of measles and its complications dramatically since it was introduced in the 60's.
By now, the vaccine has been given to billions of people and is safe and efficacious. It induces a very efficient, life-long CD4, CD8 and humoral immunity after a single injection of 104 TCID50. Moreover, it is easy to produce, cheap, and the means to deliver it worldwide already exist. The safety of this vaccine is due to several factors: i) The stability of the MV genome which explains that reversion to pathogenicity has never been observed. ii)The impossibility for the MV genome to integrate in host chromosomes since viral replication is exclusively cytoplasmic. iii) The production of the vaccine on safe primary chick embryo fibroblastic cells.

Thus, live attenuated MV could provide a safe and efficient pediatric vaccination vector.
MV belongs to the genus Morbillivirus in the family Paramyxoviridae. The Edmonston MV was isolated in 1954 (32), serially passaged on primary human kidney and amnion cells, then adapted to chick embryo fibroblasts (CEF) to produce Edmonston A and B seeds (see (7, 8) for review). Edmonston B was licensed in 1963 as the first MV vaccine. Further passages of Edmonston A and B
on CEF produced the more attenuated Schwarz and Moraten viruses (33) whose sequences have recently been shown to be identical (34, 35). Being "reactogenic,"
Edmonston B vaccine was abandoned in 1975 and replaced by the Schwarz/Moraten vaccine. This is now the most commonly used measles vaccine (7, 8).
In a previous work, the inventors constructed an infectious cDNA from a batch of commercial Schwarz vaccine, a widely used MV vaccine (European Patent Application N 02291551.6 filed on June 20, 2002). The extremities of the cDNA were engineered in order to maximize virus yield during rescue. A
previously described helper cell-based rescue system was adapted by co-cultivating transfected cells on primary chick embryo fibroblasts, the cells used to produce the Schwarz vaccine. After two passages the sequence of the rescued virus was identical to that of the cDNA and of the published Schwarz sequence.

Two additional transcription units (ATU) were introduced in the cDNA for cloning foreign genetic material. The immunogenicity of rescued virus was studied in mice transgenic for the CD46 MV receptor and in macaques. Antibody titers in animals inoculated with low doses of the rescued virus were identical to those obtained with commercial Schwarz MV vaccine. In contrast, the immunogenicity of a previously described Edmonston strain-derived MV clone was much lower. This new molecular clone allows producing MV vaccine without having to rely on seed stocks. The ATUs, allow producing recombinant vaccines based on an approved, efficient and worldwide used vaccine strain.

EXAMPLE 2: CONSTRUCTION OF SCHWARZ MV-WNV RECOMBINANT
PLASMIDS.
1) Secreted glycoprotein E from WNV
The WNV env gene encoding the secreted form of the protein was generated by RT-PCR amplification of viral RNA purified from viral particles (WNV
IS-98-ST1 strain). The specific sequence was amplified using PfuTurbo DNA
polymerase (Stratagene) and specific primers that contain unique sites for subsequent cloning in pTM-MVSchw vector : MV-WNEnv5 5'-TATCGTACGATGAGAGTTGTGTTTGTCGTGCTA-3' (SEQ ID NO: 9) (BsiWI site underlined) and MV-WNEnv3 5'-ATAGCGCGCTTAGACAGCCTTCCCAACTGA-3' (SEQ ID NO: 10) (BssHII site underlined). A start and a stop codon were added at both ends of the gene. The whole sequence generated is 1380 nucleotides long (see Figure 1), including the start and the stop codons and respects the "rule of six", stipulating that the nucleotides number of MV genome must be divisible by 6 (28, 29). The Env protein thus generated contains its signal peptide in N-term (18 aa) and no transmembrane region. Thus, It represents amino acids 275-732 in WNV polyprotein and has the sequence shown in Figure 2.

2) preM plus E glycoproteins from WNV
The WNV gene encoding the preM plus E glycoproteins was generated by PCR amplification of plasmid pVL prM-E.55.1 (clone CNCM 1-2732 deposited on Octobre 15, 2001). This expression plasmid encodes the pre-M and E proteins of WNV (IS-98-ST1 strain). The sequence was amplified using PfuTurbo DNA
polymerase (Stratagene) and specific primers that contain unique sites for subsequent cloning in pTM-MVSchw vector : MV-WNpreME5 5'-TATCGTACGATGCAAMGAAAAGAGGAGGAAAG-3' (SEQ ID NO: 11) (BsiWI
site underlined) and MV-WNpreME3 5'-ATAGCGCGCTTAAGCGTGCACGTTCACGGAG-3' (SEQ ID NO: 12) (BssH11 site underlined). A start and a stop codon were added at both ends of the gene. The whole sequence generated is 2076 nucleotides long (see Figure 3), including th-, start and the stop codons and respects the MV "rule of six". In this construct, the C-terminus part of the C protein serves as a prM translocation signal. Both preM
and E viral glycoproteins are transmernbrane glycoproteins type I. It is presumed that WNV env preME expressing MV will produce and release multimeric forms of preM-E heterodimers exhibiting high immunogenic potential. The construct represents amino acids 302-789 in WNV polyprotein and has the sequence shown in Figure 4.

3) NS1 protein from WNV
The WNV NS1 gene was generated by RT-PCR amplification of viral RNA
purified from viral particles (WNV IS-98-ST1 strain). The specific sequence was amplified using PfuTurbo DNA polymerase (Stratagene) and specific primers: MV-WNNS15 5'- TATCGTACGATGAGGTCCATAGCTCTCACG-3' (SEQ ID NO: 13) (BsiWI site underlined) and MV-WNNS13 5'-ATAGCGCGCTCATTAGGTCTTTTCATCATGTCTC-3' (SEQ ID NO: 14) (BssHII
site underlined). A start codon was added at the 5' end and two stop codons at the 3' end of the sequence. The whole sequence is 1110 nucleotides long (see Figure 5), including the start and the two stop codons, thus respecting the "rule of six".
The NS1 protein generated contains its signal peptide sequence in N-term (23 aa).
It represents amino acids 769-1136 in WNV polyprotein and has the sequence shown in Figure 6.

4) preM-E protein from Dengue type 1 virus The Dengue virus gene encoding the preM plus E glycoproteins was generated by PCR amplification of plasmid pVL pIND/[prM+E] (clone 2) (COURAGEOT, M.-P., et al. 2000, A-glucosidase inhibitors reduce dengue virus production by affecting the initial steps of virion morphogenesis in the endoplasmic reticulum. Journal of Virology 74: 564-572). This plasmid encodes the pre-M
and E
glycoproteins of DEN-1 virus (strain FGN89). The sequence was amplified using PfuTurbo DNA polymerase (Stratagene) and specific primers that contain unique sites for subsequent cloning in pTM-MVSchw vector : MV-DEN1preME5 5'-TATCGTACGATGAACAGGAGGAAAAGATCCGTG-3' (SEQ ID NO: 15) (BsiWI
site underlined) and MV-DEN 1 preME3 5'-ATAGCGCGCTTAAACCATGACTCCTAGGTACAG-3' (SEQ ID NO: 16) (BssHII
site underlined). A start and a stop codon were added at both ends of the gene.
The whole sequence generated is 2040 nucleotides long (see Figure 7), including the start and the stop codons and respects the MV "rule of six". In this construct, the C-terminus part of the C protein serves as a preM translocation signal.
Both preM and E viral glycoproteins are transmembrane glycoproteins type I. It is presumed that DEN-1 env expressing MV will produce and realease multimeric forms of preM-E heterodimers exhibiting high immunogenic potential. The construct represents amino acids 95-773 in DEN-1 polyprotein and has the sequence shown in Figure 8.
The same immunogens can be prepared by the same way from DEN-2, DEN-3 and DEN-4 serotypes.
5) Insertion into MV Schwarz vector The different WNV and DEN-1 nucleotidic sequences were cloned in pCR2.1-TOPO plasmid (lnvitrogen) and sequenced to check that no mutations were introduced. After BsiWI/BssHII digestion of the pCR2.1-TOPO plasmids, the DNA fragments were cloned in the pTM-MVSchw vector in ATU position 2 giving plasmids : pTM-MVSchw-EnvWNV, pTM-MVSchw-preMEwnv, pTM-MVSchw-NS1WNV and pTM-MVSchw-preMEDEN-1 according to Figure 9.

EXAMPLE 3: RECOVERY OF RECOMBINANT MVSCHW-ENVWNV, MVSCHW-PREMEWNV AND MVSCHW-NS1WNV VIRUSES.
To recover recombinant Schwarz viruses from the plasmids, we used the helper-cell-based rescue system described by Radecke et al. (11) and modified by Parks et al. (30). Human helper cells stably expressing T7 RNA polymerase and measles N and P proteins (293-3-46 cells, a kind gift from MA Billeter) were transfected using the calcium phosphate procedure with pTM-MVSchw-EnvWNV, pTM-MVSchw-preMEwnv or pTM-MVSchw-NS1WNV plasmids (5 pg) and a plasmid expressing the MV polymerase L gene (pEMC-La, 20 ng, a kind gift from MA Billeter). After overnight incubation at 37 C, the transfection medium was replaced by fresh medium and a heat shock was applied (43 C for two hours) (30). After two days of incubation at 37 C, transfected cells were transferred on a CEF cells layer and incubated at 32 C in order to avoid any adaptation of the Schwarz vaccine that was originally selected on CEF cells and is currently grown on these cells for safety considerations. Infectious virus was easily recovered between 3 and 7 days following cocultivation. Syncytia appeared occasionally in CEF, but not systematically. The recombinant viruses were also rescued by the same technique after cocultivation of transfected 293-3-46 helper cells at 37 C
with primate Vero cells (african green monkey kidney). In this case, syncytia appeared systematically in all transfections after 2 days of coculture. In order to increase the yield of rescue and because these recombinant viruses will be used in mice experiments, Vero cells were used as target cells in place of the usual chick embryo fibroblasts (CEF) (European Patent Application N 02291551.6 files on June 20, 2002). Recombinant viruses were passaged two times on Vero cells.
The inventors have previously shown that two passages of the Schwarz virus on Vero cells did not change its immunogenic capacities in macaques (European Patent Application N 02291551.6 files on June 20, 2002).
The recombinant viruses were prepared as described above and the expression of the transgene in infected cells was checked by immunofluorescence. To detect WNV Envelope glycoproteins expression, immune sera from mice resistant to WNV infection were used (International Patent Application WO 02/081741).To detect NSI protein expression, the inventors used anti-NS1 Monoclonal antiobodies (International Patent Application N WO
00/75665).

EXAMPLE 4: VACCINATION AGAINST WEST-NILE VIRUS
West Nile disease has recently emerged as an important mosquito-borne flavivirus infection with numerous fatal cases of human encephalitis, thus urging to develop a safe and efficient vaccine. Measles virus (MV) vaccine, a live-attenuated RNA virus, is one of the safest and most effective human vaccine developed so far. The Schwarz vaccine strain of MV can be used as a vector to immunize against heterologous viral, thereby offering a novel and attractive vaccination strategy against West Nile virus (WNV). We evaluated the efficacy of a Schwarz measles vaccine-derived vector expressing the secreted form of the WNV
envelope E glycoprotein in a mouse model. Vaccination induced high titers of specific anti-WNV neutralizing antibodies and protection from a lethal WNV
challenge. Passive administration with antisera from immunized mice also provided protection, even after challenge with high doses of WNV. Example 4 is the first report that a live-attenuated recombinant measles virus provides efficient protective immunity against an heterologous viral disease. The induction of protective immunity shows that live attenuated-MV expressing the secreted form of the E glycoprotein is an effective vaccine against West Nile disease.
MATERIALS AND METHODS

Cells and virus. Vero-NK (African green monkey kidney) cells were maintained in DMEM Glutamax (lnvitrogen) supplemented with 5% heat-inactivated fetal bovine serum (FBS). Helper 293-3-46 cells used for viral rescue (11) (a kind gift from M. Billeter, Zurich University) were grown in DMEM/10%
FBS
and supplemented with 1.2 mg of G 418 per ml. WNV strain IS-98-ST1 (GenBank accession number AF 481864) was propagated in mosquito Aedes pseudoscutellaris AP61 cell monolayers (13). Purification on sucrose gradients, and virus titration on AP61 cells by focus immunodetection assay (FIA) were performed as previously described (13, 27).
Mouse antisera to WNV. Anti-WNV hyperimmune mouse ascitic fluid (HMAF) was obtained by repeated immunization of adult mice with WNV strain IS-98-ST1 followed by the inoculation of sarcoma 180. Mouse polyclonal anti-WNV
antibodies were obtained by immunization of adult BALB/c-MBT congenic mice with 103 FFU of IS-98-ST1 as described previously (13). The WNV-immune serum was collected one month after priming.
Construction of pTM-MVSchw-sEwNv plasmid. The plasmid pTM-MVSchw that contains an infectious MV cDNA corresponding to the anti-genome of the widely used Schwarz/Moraten MV vaccine strain has been reported elsewhere (10). Additional transcription units were introduced into the viral genome to turn it into a vector expressing foreign proteins. To construct pTM-MVSchw-sEwNv, genomic RNA of WNV was extracted from highly purified IS-98-ST1 virions and reverse transcribed using Titan One-Step RT-PCR kit (Roche Molecular Biochemicals) according to the manufacturer's instructions. An RT-PCR
fragment encoding the internal E translocation signal (prM-151 to prM-166) followed by the ectodomain and the stem region of the E protein (E-1 to E-441) was generated using the 5' primer MV-WNEnv5 5'-TATCGTACGATGAGAGTTGTGTTTGTCGTGCTA-3' (SEQ ID NO: 9) containing a BsiWI restriction site (underlined) and the 3' primer MV-WNEnv3 5'-ATAGCGCGCTTAGACAGCCTTCCCAACTGA-3' (SEQ ID NO: 10) containing a BssHII restriction site (underlined). A start and a stop codon were added at both ends of the gene. The sequence respects the rule of six , stipulating that the nucleotides number of MV genome must be multiple of 6 (28, 29). The PCR
product was directly inserted into pCR2.1-TOPO plasmid (TOPO TA cloning kit, lnvitrogen) according to the manufacturer's instructions to give TOPO-sEwNv. A
1.4-kb fragment containing truncated E protein with translocation signal sequence was excised from TOPO-sEwNv using BsiW/ and BssHII and then inserted into BsiWI/BssHII-digested pTM-MVSchw-ATU2 which contains the additional transcription unit (ATU) between the P and M genes of Schwarz MV genome (10, 11). The resulting plasmid was designated pTM-MVSchw-sEwNv (named pTM-MVSchw-EnvWVN in the previous Examples). All constructs were verified by automated sequencing.
Rescue of recombinant MVSchw-sEwNv virus from the cloned cDNA.
Rescue of recombinant Schwarz MV from the plasmid pTM-MVSchw-sEwNv was performed using the helper-cell-based rescue system described by Radecke et al.
(11) and modified by Parks et al. (30). Briefly, human helper cells stably expressing T7 RNA polymerase and measles N and P proteins (293-3-46 cells, a kind gift from MA Billeter, Zurich University) were transfected with 5 pg pTM-MVSchw-sEwNv and 0.02 pg pEMC-La expressing the MV polymerase L gene (a kind gift from MA Billeter) using the calcium phosphate procedure. After overnight incubation at 37 C, a heat shock was applied for 2 h at 43 C. After two days of incubation at 37 C, transfected cells were transferred onto a Vero cell monolayer.
Vero cells were used as target cells in place of the usual chick embryo fibroblasts (CEF) in order to increase the yield of rescued virus. The inventors have previously shown that two passages of the Schwarz virus on Vero cells did not change its immunogenicity in primates (10). Syncytia that appeared after 2-3 days of coculture were transferred to 35 mm wells of Vero cells, then expanded in and then 150-cm2 flasks in DMEM/5% FBS. When syncytia reached 80-90%
confluence (usually 36-48 h post-infection), the cells were scraped in a small volume of OptiMEM (Invitrogen) and frozen and thawed once. After low-speed centrifugation to pellet cellular debris, the supernatant, which contained virus, was stored at ¨80 C. The titers of MVSchw-sEvvNv was determined by an endpoint limit dilution assay on Vero cells. The 50% tissue culture infectious doses (TCID50) were calculated using the Karber method.
Radioimmunoprecipitation assay. Vero cells were starved for 1 h with DMEM without methionine and cysteine (ICN Biomedicals) and labeled 3 h with 250 pCi/m1 Tran36S-label (ICN Biomedicals). Cells were lysed with RIPA buffer (20 mM TrisCI, pH 8.0, 150 mM NaCI, 10 mM EDTA, 0,1%SDS, 0,5% deoxycholate, 1% Triton X-100) supplemented with a cocktail of protease inhibitors. RIP
assay was performed as previously described (31). Samples were analyzed by SDS-15%
PAGE under reducing conditions.

Mice experiments. CD46-IFNAR mice were produced as previously described (10). Adult BALB/c mice were purchased from Janvier Laboratories (Le Genest St Isle, France). Mice were housed under specific pathogen-free conditions at the Pasteur Institute. Five to 6-week-old CD46-IFNAR mice were i.p.
inoculated with 104 or 106 TCID50 of MV. Acute WNV challenge was performed by i.p. inoculation of neurovirulent WNV strain IS-98-ST1 (i.p.LD50 = 10) in Dulbecco's modified phosphate saline buffer (DPBS) supplemented with 0.2%
bovine serum albumin (BSA) pH 7.5 (Sigma Chemical Co.). The animals were monitored daily for signs of morbidity and mortality. All experiments are approved and conducted in accordance with the guidelines of the Office Laboratory Animal Care at Pasteur Institute.
Anti-WA vaccination test with antigenic bo = st. Adult CD46+/- IFN-a/(3R-1- mice were vaccinated over a four week period with the MV-WN sE virus at a dose of 104 DCIP50 (which is a dose recommended for humans) and an antigenic boost was provided by purified WNV pseudo-particles that were secreted by MEF/3T3.Tet-Off/WN prME # h2 cells.
Humoral response. To evaluate the specific antibody response in serum, mice were bled via the periorbital route at different time after inoculation.
Detection of anti-MV antibodies was performed by ELISA (Trinity Biotech, USA) as previously described (10). An anti-mouse antibody-HRP conjugate (Amersham) was used as the secondary antibody. The endpoint titer was calculated as the reciprocal of the last dilution giving a positive optical density value. The presence of anti-WNV antibodies was assessed by ELISA as previously described (13).
Briefly, microtitration plaques were coated with 106 FFU of highly purified WNV
strain IS-98-ST1 and then incubated with mouse sera dilutions. A test serum was considered positive if its optical density was twice the optical density of sera from immunized control mice.
Neutralization assay. anti-WNV neutralizing antibodies were detected by a FRNT test. Sera from each mouse group were pooled and heat-inactivated at 56 C for 30 min. Vero cells were seeded into 12-well plate (1.5 x 105 cells/well) for 24 h. Mouse serum samples were serially diluted in MEM Glutamax/2% FBS.
Dilutions (0.1 ml) were incubated at 37 C for 2 h and under gentle agitation with an equal volume of WNV strain IS-98-ST1containing ¨100 FFU. Remaining infectivity was then assayed on Vero cell monolayer overlaid with MEM Glutamax/2%FBS
containing 0.8% (w/v) carboxy methyl cellulose (BDH). After 2 days of incubation at 37 C with 5% CO2, FIA was performed with anti-WNV HMAF as previously described (27). The highest serum dilution tested that reduced the number of FFU
by at least 90% (FRNT90) was considered the end-point titer.
Passive transfer of immune ser.. Pooled immune sera were transferred into 6-week-old female BALB/c mice intraperitoneally. Mice received injection of 0.1 ml of serial dilutions of pooled serum samples in DPBS/0.2%BSA one day before WNV inoculation. The challenged mice were observed for more than 3 weeks.
DISCUSSION OF THE RESULTS
Since its introduction into the United States in 1999, West Nile virus (WNV) infection has been recognized as one of the most serious mosquito-borne disease in the Western Hemisphere, causing severe neurological disease (meningoencephalitis and poliomyelitis-like syndrome) in humans. (3). Within the last 4 years, WNV had spread through North America, Central America and the Caribbean (1, 2). It is presumed that it will reach South America in the coming years. Since 2002, the US outbreaks were characterized by an apparent increase in human disease severity with 13,000 cases and 500 deaths. Although mosquito-borne transmission of WNV predominates, WNV is also transmitted by blood transfusion, organ donations and transplacentaly to the fetus (3). Prevention of West Nile encephalitis is a new public health priority and it is imperative that a vaccine be developed (3, 4, 5). No vaccine has been approved for human use so far.
Because WNV can be transmitted across species, there is an urgent need to develop preventive strategies for humans. A rational approach should be to confer a long-term immunity in large groups of individuals, and to boost this immunity in case of WNV outbreaks. Measles virus (MV) vaccine can now be used as a vector to immunize against heterologous viral diseases, thereby offering a novel and attractive vaccination strategy against WNV. We have recently tested this vector against HIV infection (6). MV vaccine, a live-attenuated RNA
virus, is one of the safest and most effective human vaccine developed so far. It induces a very efficient, life-long immunity after a single or two injections (7, 8).
The MV
genome is very stable and reversion of vaccine strains to pathogenicity has never been observed. The Schwarz MV strain is used in two widely used measles vaccines, Attenuavax (Merck and Co. Inc., West Point, USA) and Rouvax (Aventis Pasteur, Marcy l'Etoile, France), and in the combined measles, mumps, and rubella vaccine (MMR) (9). We have recently generated an infectious cDNA for this strain (10) and introduced additional transcription units (ATU) into it for cloning foreign genes, based on the work of Radecke et al. (11). The vaccine rescued from the molecular clone was as immunogenic as the parental vaccine in primates and mice susceptible to MV infection. Thus, this approved and widely used MV
vaccine can be used as a vector to immunize individuals simultaneously against measles and other infectious diseases.
WNV is a single-stranded RNA virus of the Flaviviridae family, genus flavivirus, within the Japanese encephalitis antigenic complex (2, 3). The virion is composed of three structural proteins, designated C (core protein), M
(membrane protein) and E (envelope protein). Protein E, which is exposed on the surface of the virion, is responsible for virus attachment and virus-specific membrane fusion.
Because the E glycoprotein can potentially serve as a . major protective immunogen for a WNV vaccine (12), the inventors introduced the WNV cDNA
encoding the carboxyl-terminally truncated E glycoprotein lacking the transmembrane-anchoring region (residues E-1 to E-441, designated sEwNv hereinafter) of IS-98-ST1 strain (13) into the infectious cDNA for the Schwarz MV
vaccine (10) (Fig. 10A). WNV strain IS-98-ST1 has the same neuropathologic properties than the new variant designated Isr98/NY99 that has been responsible for the recent WNV outbreaks in North America and Middle East (13). The WNV
sequence was introduced in an ATU located between the phosphoprotein (P) and matrix (M) genes in the MV genome. The recombinant MVSchw-sEwNv virus was produced after transfection of the corresponding plasmid into human helper cells allowing the rescue of negative-stranded RNA paramyxoviruses (11), then propagation in Vero cell cultures. The growth of MVschw-sEwNv in Vero cells was only slightly delayed as compared to that of standard Schwarz MV (MVschw) (Fig.
10B). After 60 h of infection, the yield of MVschw-sEwNv was comparable to that of MVschw. The expression of sEwNv in MVschw-sEwNv-infected Vero cells was demonstrated by immunofluorescence and radioimmunoprecipitation (RIP) assays (Fig. 10C, D). At 40 h post-infection, the cell surface of MVsGhw-sEwNv-induced syncitia was clearly visualized by anti-WNV immune serum, indicating that sEwNv is transported along the compartments of the secretory pathway (Fig. 10C). RIP

analysis revealed that anti-WNV antibodies recognized sEwNv that migrated faster than authentic E glycoprotein (Fig. 10D). Interestingly, sEwNv was detected in the supernatants of MVschw-sEwNv-infected Vero cells at 40 h post-infection (Fig.
10D, panel Supernatants/ MVschw-sEwNv, lane a-WNV). Thus, MVschw-sEwNv expresses a recombinant E glycoprotein which is secreted efficiently.
lmmunoblots confirmed that sEwNv accumulated in the culture medium of MVsGhw-sEwNv-infected Vero cells (data not shown).
Genetically modified mice expressing the human CD46 MV receptor and lacking the interferon a/13 receptor (6, 14) (CD46+/- IFN-a/13 R4-, abbreviated CD46-IFNAR) that are susceptible to MV (14) were used to assess the immune response induced by MVschw-sEwNv. These mice deficient in IFN-a/I3 response raise cellular and humoral immune responses similar to those of competent mice (6, 10, 15, 16).
Two groups of six CD46-IFNAR mice were inoculated intraperitoneally (i.p.) with either 104 or 106 tissue culture infective doses (TCID50) of MVschw-sEwNv.
Each group was boosted using the same dose 1 month after the first immunization. As a control, CD46-IFNAR mice were immunized with 106 TCID50 of "empty" MVschw.
One month after the first immunization, specific anti-MV antibodies were detected in immune sera from mice inoculated with either MVschw or MVSchw-sEwNv (Table 1). Mice that received either dose of MVschw-sEwNv displayed specific anti-WNV
antibodies at a dilution of 1:3,000. One month after boosting, the titers of anti-WNV
antibodies had reached 1:30,000 to 1:200,000 (Table 1) and were highly reactive with the WNV E glycoprotein (Fig. 11). No anti-WNV antibodies were detected in the sera of any control mice (Table 1 and Fig. 11). These results show that one injection of MVschw-sEwNv induces anti-WNV antibodies, and that boosting one month after priming increases their titers 10 to 60 times.
Anti-WNV neutralizing activity was measured in MVschw-sEwNv-immune sera using a focus reduction test (FRNT90) (Table 1). As a positive control, the WNV-immune serum from immunized BALB/c-MBT congenic mice (13) gave a FRNT90 titer of 50. The immune sera from CD46-IFNAR mice inoculated with "empty" MVschw had not detectable neutralizing activity. Immunized CD46-IFNAR
mice which received 104 or 106 TCID50 of MVschw-sEwNv raised neutralizing antibodies with similar FRNT90 titers, and boosting increased their titers from 10 to 200-300. These data show that mice twice inoculated with the recombinant live-attenuated MV encoding the secreted form of the IS-98-ST1 E glycoprotein had high levels of anti-WNV antibody with neutralizing activity, regardless of the injected dose.
Because antibody-mediated immunity may be critical to protect against WNV infection (17, 18), the inventors examined if the passiv-. transfer of sera from MVschw-sEww-immunized mice can protect adult BALB/c mice from WNV
infection (Table 2). Groups of six 6-week-old BALB/c mice received i.p.
various amounts of pooled immune sera from MVschw-sEwNv-immunized CD46-IFNAR
mice collected one month after priming or boosting. One day later, the mice were challenged with 10 times the i.p. 50% lethal dose (LD50) of WNV strain IS-98-(13, 19). As a positive control, BALB/c mice that received as little as 2 pl of the WNV-immune serum were protected from the challenge (Table 2). In contrast, all mice that received 2 pl of the non-immune mouse serum or serum from "empty"
MVschw-immunized mice died within 11-12 days. Protective passive immunity was observed in all BALB/c mice following transfer of 2 pl of pooled sera from IFNAR mice immunized once with 106 TCID50 of MVSchw-sEwNv. As little as 1 pl of this antisera induced 66% protection. Passive transfer of sera collected one month after a single immunization with 104 TCID5o induced a survival rate of 50 %.
Remarkably, the administration of 1 pl of MVschw-sEwNv-immune sera collected 1 month after boosting induced 100% protection. These results indicate that a single injection of 106 TCID50 or two injections of 104 TCID50 of MVschw-sEwNv elicited protective humoral response. Because the amount of flavivirus inoculated during mosquito feeding is probably in the order of 102 to 104 infectious virus particles (1), we assessed the capacity of MVschw-sEwN-immune sera to protect against a range of 102 to 105 focus forming units (FFU) of WNV strain IS-98-ST1. Groups of six BALB/c mice were passively immunized with 2 pl of pooled immune sera collected from CD46-IFNAR mice twice inoculated with 104 TC1D50 of MVschw-sEwNv (Table 2). Survival rates of 85-100% were observed in mice that received the MVschw-sEwNv-immune serum, regardless the lethal doses of IS-98-ST1 (10 to 10,000 i.p. LD50). These data are consistent with the finding that humoral response plays a critical role in protection against WNV infection.
Mice which are completely unresponsive to IFN-ce/13 are highly susceptible to encephalitic flaviviruses (19, 20). Indeed, the inventors previously showed that WNV infection of CD46-IFNAR mice was lethal within 3 days instead of 11 days in competent mice (19). To assess whether the immunity induced by MVSchw-sEwNv could protect these compromised animals from WNV infection, three CD46-IFNAR
mice from the group that had received two injections of MVschw-sEwNv (106 TCID50), were i.p. inoculated with 100 FFU of IS-98-ST1 one month after the boost. Mice inoculated with "empty" MVschw were used as controls. The mice that had received MVschw-sEwNv survived the WNV challenge while control mice died within 3 days. MVschw-sEwNv-immunized mice were bled 3 weeks after challenge.
The FRNT90 antibody response (titer ¨ 100) was comparable to the pre-challenge response. Notably, post-challenge immune sera did not react with WNV
nonstructural proteins such as NS3 and NS5 as shown by RIP assay (Fig. 11, panel MVschw-sEwNv, lane 106 TCID5o, day 20, p.c.), suggesting that no viral replication occurred after challenge with WNV. These data show that immunizing with MVschw-sEwNv prevented WNV infection in highly susceptible animals.
The present Example shows for the first time that a live-attenuated measles vector derived from the Schwarz MV vaccine can induce a protective immunity against an heterologous lethal pathogen. These data constitute also the proof of concept that a live-attenuated Schwarz measles vaccine engineered to express the secreted form of the WNV E glycoprotein can be used as a vacoine to prevent, West Nile disease in humans. The MV vaccine vector offers several advantage over other existing viral vectors. The Schwarz MV vaccine has been used on billions of people since the sixties and shown to be safe and efficacious. It is easily produced on a large scale in most countries and can be distributed at low cost.
The MV genome is very stable and reversion to pathogenicity has never been observed (8). Moreover, MV replicates exclusively in the cytoplasm, ruling out the possibility of integration in host DNA. The MV vector has been shown to express a variety of genes, or combinations of genes, of large size over more than twelve passages (6, 16, 21, 22, 23, 24). This stability is likely due to the fact that there is little constraint on genome size for pleomorphic viruses with a helical nucleocapsid. Unlike chimeric viral vectors, the recombinant MV vector is an authentic MV expressing an additional gene. This greatly reduces the risk of changing the tropism and the pathogenicity of the original vaccine. It reduces also the risk of recombination.
The recombinant MV-WNV vaccine according to a preferred embodiment of the present invention is a promising live-attenuated vector to mass immunize children and adolescents against both measles and West Nile diseases. Although the existence of an anti-MV immunity in nearly the entire adult human population appears to restrict its use to infants, an already worthy goal, recent studies demonstrated that revaccinating already immunized children results in a boost of anti-MV antibodies (25, 26). These and other studies (Ann Arvin) demonstrated that the presence of passive MV pre-immunity (maternal antibodies) does not circumvent the replication of attenuated MV after a second injection. This opens the possibility of using the live-attenuated MV-derived vector to immunize adults.
Indeed, the inventors reported that a MV-HIV recombinant virus induced anti-HIV
neutralizing antibodies in mice and macaques even in the presence of pre-existing anti-MV immunity (6). Because of cross-species transmission, it is feared that WNV becomes a recurrent zoonosis with repeated seasonal outbreaks in humans.
The inventors propose that MVSchw-sEwNv could be used to induce long-term memory immunity in large groups of children and adults, and to boost this immunity in case of West Nile disease outbreak.

BIBLIOGRAPHY
1. D. W. C. Beasley, et al., Virology 309, 190-5 (2003).
2. M. A. Brinton, Annu. Rev. Microbiol 56, 371-402 (2002).
3. L. R. Petersen, A. A. Marlin, D. J. Gubler, JAMA 290, 524-28 (2003).
4. T. Monath, Ann.N.Y.Acad.Sci 951, 1-12 (2001).
5. T. P. Monath, J. Arroyo, F. Guirakhoo, Curr.Drug Targets Infect Disord 1, 37-50 (2001).
6. C. Lorin, et al., Journal of Virology 78, in press (2004).
7. D. Griffin, in Field's Virology, 4th Edition D. Knipe, P. Howley, Eds.
(Lippincott- Raven Publishers, Philadelphia, 2001), vol. 2, pp. 1401-1441.
8. M. Hilleman, Vaccine 20, 651-665 (2002).
9. E. Buynak, R. Weibel, W. J. JE, J. Stokes Jr, M. Hilleman, J. Am. Med.
Assoc. 207, 2259-2262 (1969).
10. C. Combredet, et al., Journal of Virology 77, 11546-11554 (2003).
11. F. Radecke, et al., Embo J 14, 5773-84. (1995).
12. T. Wang, et al., J.Immunol. 167, 5273-5277 (2001).
13. T. Mashimo, et al., Proc.NatI.Acad. Sci.USA 99, 11311-11316 (2002).
14. B. Mrkic, et al., J Virol 72, 7420-7 (1998).
15. U. Muller, et al., Science 264, 1918-21. (1994).
16. M. Singh, R. Caftaneo, M. A. Billeter, J Virol 73, 4823-8. (1999).
17. M. S. Diamond, B. Shrestha, A. Marri, D. Mahan, M. Engle, J.Virol. 77, 2578-2586 (2003).
18. M. Haley, A. S. Reiter, D. Fowler, J. Gea-Banacloche, N. P. O'Grady, Clin.

Infect. Dis. 37, 88-90 (2003).
19. M. Lucas, et al., lmmun. Cell Biol. 81, 230-236 (2003).
20. M. Lobigs, A. Mullbacher, Y. Wang, M. Pavy, E. Lee, J.Gen.Virol. 84, 567-572 (2003).
21. F. Radecke, M. Billeter, Reviews in Medical Virology 7,49-63 (1997).
22. P. Spielhofer, et al., J. Virol. 72, 21 50-21 59 (1998).

23. M. Singh, M. Billeter, J. Gen. Virol. 80, 101-6 (1999).
24. Z. Wang, et at., Vaccine 19, 2329-2336 (2001).
25. A. Dilraj, et al., Lancet 355, 798-803 (2000).
26. A. Dilraj, et al., Pediatr. Infect. Dis. J. 19, 1211-3. (2000).
27. P. Despres, M.-P. Frenkiel, V. Deubel, Virology 196, 209-219 (1993).
28. P. Calain, L. Roux, J Viral 67, 4822-30 (1993).
29. H. Schneider, K. Kaelin, M. A. Billeter, Virology 227, 314-22. (1997).
30. C. L. Parks, R. A. Lerch, P. Walpita, M. S. Sidhu, S. A. Udem, J Virol 73, 3560-6 (1999).
31. P. Despres, J. W. Griffin, D. E. Griffin, J.Virol. 69, 7345-7348 (1995).
32. Enders, J. F. & Peebles, T. C. (1954) Proc. Soc. Exp. Biol. Med. 86, 277-286.
33. Schwarz, A. (1962) Am. J. Dis. Child. 103, 216-219.
34. Parks, C. L., Lerch, R. A., Walpita, P., Wang, H. P., Sidhu, M. S. & Udem, S. A. (2001) J Virol 75, 910-20.
35. Parks, C. L., Lerch, R. A., Walpita, P., Wang, H. P., Sidhu, M. S. & Udem, S. A. (2001) J Virol 75, 921-33.
TABLE 1. Antibody response of CD46-IFNAR mice to intraperitoneal inoculation of MVSchw-sEwNy Immunizing virus MV-specific WN-specific WN-specific Ab titer 4 Ab titer 4 FRNT90 5 WNV1 (103 FFU) NT 10,000 50 MVSchw 2 (1 06 TCID50) 30,000 <10 <10 MVSChW-SEwNv 2 (104 TCID50) 15,000 3,000 10 MVSChW-SEwNv 2 (106 TCID50) 25,000 3,000 10 2 x MVSchW-SEwNv 3 (1 04 TCID5o) 90,000 30,000 200 2 x MVSchw-SEwNv 3 (1 06 TCID50) 140,000 200,000 300 1 BALB/c-MBT congenic mice were i.p. inoculated with WNV strain IS-98-ST1.
2Virus was given i.p. to CD46-IFNAR mice.
3Virus was given i.p. twice at 1 month of interval.
4Determined by ELISA on pooled heat-inactivated sera.
6The highest serum dilution that reduced the number of FFU of WNV by at least 90%.
NT: not tested TABLE 2. Protective ability of the MVSchw-sEwNv-immune serum Material used for Volume of sera WNV2 Protection M.D.O.D3 immunization transferredl (FFU) (no. surviving/ (day S.D.) (DI) no. tested) Controls 10 100 0/6 11.5 1.5 DPBS

MVSchw 5 2 100 0/6 12.0 1.5 MVSchw-sEwNv6 106 TCID50 (day 30) 2 100 6/6 1 100 4/6 11.0 1.5 104 TCID50 (day 30) 10 100 3/6 10.5 2.0 104 TCID50 (day 60) 1 100 6/6 2 1,000 6/6 2 10,000 5/6 10 2 100,000 5/6 11 1BALB/c mice received 0.1 ml of DPBS containing the indicated amount of pooled sera.
2 Mice were challenged with WNV strain IS-98-ST1 one day after passive transfer.
3 Mean day of death standard deviation.

4Immune sera from resistant BALB/c-MBT congenic mice (13) inoculated with 103 FFU of IS-98-ST1 WNV.
Immune sera from CD46-IFNAR mice collected 30 days after inoculation of MVSchw (106 TCID50).
6 Immune sera from CD46-IFNAR mice were collected 30 days after 1 injection or 60 days after 2 injections of MVSchw-sEwNv=

=

T a6Ed 08T p6ED161.1pD 61p1DDEp6e 8E66136136 8388331186 3E3183161E 6E38316386 OZT DE.I.D61.pup 1660p6Tebz p6166pp666 8833113881 3131333811 6E368E6816 09 D6EDDE0D1p6 1.DD661.1.E.61 ppl6ED6zap 8663386888 66E66E6888 86888E361E
Z <0017>
sn..1!_n alj_N-lsam <ETZ>
VNG <ZW>
9LOZ <TTZ>
Z <OW>

08ET EP1D1b1D66 pp66611Opp 1DDRD11616 656p66116p D1P65111Dp 666443bepp OZET De6p66e4D1 DEop6ulDP6 e6PD6Dbe66 8883133383 D8E3811133 688E366118 09ZT D6pApp661 Da6eeyeD66 1183383188 3186838838 8686686836 6616616818 00ZT DE18313868 661.3.1)Dppp Dpp661.1pp6 aap61pDa66 8E13638833 6638336616 OYU PD11161111 3338E31613 8316611868 D6661.6PD36 3881338638 8611831136 080T 616E316p1.a 1p1DD116pe eD611DDR66 le66DpD661 DpDp1.6eD61 lpp6611616 OZOT 61.61DpD66i pp166pDEDp 6836333138 6661131116 8831113668 eeD1161Da6 096 D661E1.ppep DEE.666pp64 1.6p361zppu pe661p6pe6 16p6e1.616E. e611apil.66 0.08 1pD61D4D6r 666p6pppep 1D6661.1pD6 8186161316 836E863833 6383833886 08L 66666 lup116Dp6p 6P6pDpp66P 6616161ppl 6up661D616 e66611DD
OZL 3133883103 8661831166 16861631ED D16611Dz1.6 3868883E86 6116138618 009 lelpp6e661. 1768881383 83E1831133 6366361331 3E31E36E31 186E666836 08V D16111.azep D661.6ep6DE 1.6ppplplpr 6p6epu611D auDDee6pu6 6eapeD66ep OZt DoulD1D61.) 36011appppo 6D61pDpDp6 lveD6ET55E ppDE06a1.1p1 1.E6E0b1.D66 09E Deu36656ap 6666pDp66a 6616e66epp eft p616161 111DopppDp 61)616Depe 00E ppolpupppl DEme6p6661. 8338633361 6361368E83 383313131E 6368316338 OtZ 1366111813 6118116836 3316686836 6133883166 3668661818 861861868P
08T 6161863183 381336883E 66ppapa6ap DzElpp6a6D 64D6ppu6D6 56 OZT 61.11p661.66 61voppD6E6 61D1616E66 pp6611alza e5p6EDEp36 p6lup6611D
09 361DEED41) 6pDplap6p3 D6616611. 143611p1D6 16D1611461 6116e6p5le T <0017>
sruo aLpi-lsam <ETZ>
VNO <ZTZ>
02E1 <TTZ>
T <OW>

O'Z 'Jan uilualPd <OLT>

91 <09T>

9Z-Z0-E001 <CST>
Z60'0Z17'Z VD <OST>

0Z-90-E00Z <CST>
8ECZE17'Z VD <OST>

9Z-Z0-t0OZ <CST>
LZOTOO/VOONVIDd <OST>

9Z-Z0-1700Z <TVT>
85Z'LTS'Z VD <0171>

9VZO-LS0000 <0E1>

SNOI1VDI1ddV DI1SONDVIO ONV DI1n3dVII3H1 '1VNIDDVA NI 3sn III3H1 ONV 'DNIOD31JOA 3H1 DNIGOD
S3N3D ONV SNI3101id S3SMIIA 31IN 153M GNV 310N3 M3N <OZT>

S)iND
JnalsPd lnlpsui <OTT>

Z 96Pd OZT E66E6eee66 ED6EE15E14 6E4E61EDED EDD6e6E666 66E6DEDEDD E61.11E3344 09 1.36613316u DED3361364 364E31331D 61EDDE6163 DIT6EEEE66 E66EDEE61E
<00t>
snuo T-adA4 an6uaa <ETz>
VNG <ZTZ>
OtOZ <TTZ>
V <OTZ>

OTTT E61EE1.DDE6 PPPP61P61P DP6P6PDPDD
0801 E6ED1E6E66 1E1.664E1.66 1161.)66D6P DP61)PPPDD P1D6D61.DPD DPDDP11DDP
OZOT D6135E66E3 6a361.551.1E EIEDEE1E511 6EEE6635E6 P6PDPDDPDD PD6DaDPOD6 096 133E661633 E3E6636436 E6E616E6la D3E31.663E1 DEE66E3336 13E11E631.1 006 DE611.E6E6E 16663366EE 6DE6661EDD D666PDDUPP PDPDP6PPDP 1666aDDE6E
0t8 66)1.UPDPD1 EEDETE6DED DE66636643 PDP)1.6PDDP 4.EE1E611DE 61.6E6E614D
08L plEE663.E6D 66661.611.33 EleD6DE6E6 133661.63Ea 64EDleuEDa bee61.66613 OZL 116ED666EE E611D6EE66 16aelE6lEe 31366E36Eu E611E6611E 13316133E6 099 16EDEDDI.E6 36611DEEDE E6EeD16136 6DEE6611ED lEbEE531.DE 61.61EE61DE

6E6616EE6E 4436Elee66 11.363634Eu 6E313E6331 61Eu66EEDD E6E6b33166 08t 1E61166161 11)DPDPPDP PDD6D1DPP6 EDDED6alle 1111E16E6E ue66661336 OZt 6EE6613651 lEEE6611EE PPP66DUDDP DADDPD1D3 6OPPP1DDPD 6PD16PPDPI.
09E 61EE666E66 EDEEE6E61.1 661631616E 1133E66461 661EE6E66E E6111134DE
00E DEE6136E6D E66EE616ED 6EE666164E EED1ED6E66 zaE6E 111 66e4D
OVZ 166361616E 66EE66uelE 3136EEE6ED 11ED1E6EED D6P13366PP DPDD6DPPP6 081 zDDDE11.E16 EEDE1663DE 661E661136 6P661.61.P61 PPDPDPZPD1 161.6E6616P
OZT E66161E6E6 1.D6P6PPD66 DD6PDITDP6 PlE0D6l616 661DPDP61D 63ED616DEE
09 616331313D 1431361311 6E66E66116 ED63131116 DED1D1D6E1 E33166E6le E <00V>
sn,n_A alA-N-lsam <ETZ>
vN0 <M>
OTTT <TTZ>
E <OTZ>

9LOZ pullD6 DpD646Dep6 163D1D1Dpa 1D1D61)146 OVOZ E66E66116E D63131.1163 ED1.31.36E1E D31E6E1E61 631361EED1 ED6b61.E661 0861 61161DD1D1 )665661D61 ap6EreppEop ulpbEapplb zepbbp66D1 161DE01D6D
OZ6T DlluD6E66E 66311616pp DaEo316136 6eE666116E D1DDED1161 6666E66116 0981 ED1E661113 E66611D6ED EDE6E66E1D 136336E13u 6E6E3636E6 6PPPD1DDDP
0081 DDEEDEz113 D6EEED5611 ED6EDETE66 1316EEDED6 611EDDED1E EpaE6EDEED
OLT Ev6E66E6ED 6661.5616E4 EDE1ED1DE6 E55144DDDE DDEE661aEE 614E613316 0Z91 6DPP1DDP6D PP611P3110 6616PD16D1. Dap100116E peD611)Dp6 6aEb6DED66 09S1 13E3E1.6E36 14EE661161. 66e66 DPD166PDPD P6PD6DDD1D P6661131.11 00ST 6EED114366 EEED116431 6D661.E1DDE EDEE666EE6 11.6E361.1Eu EEE6Eq.E6EE
OtVT 616E6E1.61.6 ER6111E316 663163E644 ETED161DED EED6EED111 lEE6616433 08ET 11EDD6E66a 3661a1D6EE DlE3613136 E666E6EEDE 31:D66611ED 6e1x616131 09Z1 3.6EE661364 6E36E6643.D DDaDDPEDaD DE5&1ED115, 616E51631E 3316611311 00ZT 63P6PPPDPP 661161DP61 P61.6)P1DP1 PD61PPDDPD E614E666ED 166DED3EE6 OKI 4613E6616E DE6166E6E6 6666 41.35EEE1DE DPDP1PD1.13 D6D6E0AZDD
0801 ZDPD1PD6PD laP6P666PD 666 P661466PDP DPDD1.DP1DP PP66DPD6Dl OZOT 6E66161.3E1 DEEDDe661E 331.611111e 336646EE6D EILEED;Ele E6E6EuE611 096 D1PDDPP6PP 66P1PP366P PDDP1)1061 DD611.1PPPD D6D61PDPDP 66e66 006 EEEDE161.11E 14E60636136 byeEDE6661. 36656EDEb6 16616E66EE DE6rD61616 0178 1111D6EDDD P630616DET PDP61PPDPD 1.D6PP6P666 1PDDP6DDD6 16D61APPP
08L DDE3313131 E636ED1633 ElD66111E1 3611E116E3 633166E6ED 66aDDEED16 OZL 6366E661.El EE61E61E6E E6161E6D1E D3P1D61?P3 P66PP1D161 PD1P1DP616 099 D6136E3E63 66EE63431.1 66114E66lb 661EDEED6u 66131616E6 6ppb611pla 009 DPE,P6PDPPD 6P61PP661.1 DD64DPPD41 D6PDP11D6P DD))661.661 1.1.1D611P1D
OtS 6163161.116 161.16P6P6P 361.PDDP3PP 36E6664136 1E66116614 E316336E36 081' 6166133361 ElE661DDDE E66E614Dle 664E31Eu6E DEEEEE1661 11E166EEDE
OZt DD66PPDDPD 6E3E661E66 11366666EE 666E 1.D1DPAPPP 6E66DPDPDP
09E 6E3616E3E6 43E3166E66 316E363E6y PD1DP7D6DP DP6PPD7PDE1 1E6EE661.E4.
00E 66ED163E1D 16ED6ED16p EEDED61661 1613E6D1ED e6Ee6EDDlE 61EE1661.36 OVZ 631613646u 33361pE64p 11DEDTElpe 1E61E63616 1E3E1E6661 61E661EED6 E aftd nLD sAl nal aLI Jill 6Jv ALE) aLI PLv ski Jql Jas sAD Ply aqd SET OET SZT
ski uLv SAD Jill dsv all JS ALE) sA1 ALE) aqd nal ALE) SAD ALE) OZI SIT OTT
usv ALD dJI ALD &iv dsv LEA LEA AP uLD 6Jv sAD LEA a4d ELY

0.1d dsv ELY 6.1V ski dsv usV s!-1-1 PLV rip ALE) law J41 dad SAD

ELv ELv ski Jql Jas nal dsv Jas LEA Jq1 PLv nal JAI sic) JAI

Jas Euv LEA nLD PIN nal usv LEA ELy ALE) law usv law law ski LEA dsv au I Jill (Lid sAl dsv sAl Jas law aLI .1141 LEA sAD Jas St OV SE
dsv ALE) nLD nal LEA nal dsv LEA di Jill ELv ALE) ...las LEA ALE) OE SZ OZ
np nal aqd dsv 6Jv usv Jas law ALE) nal sAD usv aqd Jas JAI

ST OT S
PLY cud ELV LEA nal nal nal nal LEA LEA atid LEA LEA 6Jv law S <0017>

snJo aLpi-lsam <Eu Did <ZTZ>
65.17 <ITZ>
S <OW>

0170? upa4.1661pD 1.6p6EleDDP 161.yeDp116 61pD6611.6p DEqapp616D e61p6oz11.) 0861 6D16D6D6p6 6eppllpuul 1T66611661. ppe613611D le666elpu6 6eleeee6lv 0Z61 DDE.6611Daz 46366)6eD1 1617716666 DE1ED6DDRE 661111166P pppDp1.6611 0981 uuppE661.61 Dz6Dp1.4161 6E66E66E.E 1D1)66D11D p6661pD6D) vDp6e66b1D
0081 plelD661p6 6ep6DeD6p6 6p6DDD6D1.e pApp6D116 leppp666p1 pp6e36ep66 ()VLF upp6ruDl1.b 61.36ppl.Dpp EtIllapftee Eu6166ED66 b6e16Elepl pDp1D5p5p6 0891 1661111DDE DDEP5EDE6P 611PDPED1E E7DPEPEE5P '2E7E610E11 5P1ED7D1PE

0951 5PPD61PDD6 D61P5PDPP6 EIPPE0DP1PPP 1166P3616p 1DD161Dpr6 6luD6eDDDE
0051 6p6aDb646p E5bpp6p6p1 D6pulalpp1. D66ppup645 1:26161p1p3 z6lp666epp OK pllappEll.Dp eplp661Eup pplle6ED61 EEP61DDEDE 66eD6111D1 PEDUEDE6DE
08E1 P56E7167E6 P7D1PER6ED E636666DDE 61167617PD U761EED6P6 6eu66rDED1 OZET e6661Dp16D 16p16ep66u D6pp6ppup6 1pDzAppe6 PED11E7ED1 66176111E5 0911 EEDE6EDEP6 611DE6E6UP 7331EDUED1 1766666)11 DE66117)61 DED7E113E6 00ZT Elpz11661E EDEPEDEDD1 5117661E71 EPPEE6EPE6 1EP7E611E1 76166166E6 OTT lEEz11)E66 1DO66popu6 P773E71761 DE661)E7E1 1DDDEP65DE 17E6E7E611 0801 6p3pluee66 D16Dp13313 6PPD1DDEDE p1.eppuua61 1.pppuu661e DEE6EDE1DE
OZOT 6e6DEpp666 1.66EDDeDbp Dpe6666ppe DEDD161DeD 16v1u616ED 11E;eeppll 096 Dp.epp6lx1p eD116p1p6p pp66ppb61D ETTEDp6461 61.6ppD1.1bp p1361616ip 006 64De1ppfte 66upee6611 lu1De66D61 )661pp3666 64D66p6p3p 66161166De 0178 peop634616 z6111Dep6D 6Dp6pEopv6 pp661661ae Delp66E6p6 5PEDEDEEDD
08L 161EbEED11 P677E77E77 EDEEED1P1E EUE1D6PE61 luAlbalup RD6DElaplE0 OU DD61DDDEP6 app166e663 e6pe611)1D up6lappe56 11EDPEDDEE PlEEPPEED6 099 51EDDEDDED 16761D6UE5 61E75E6611 p16616Dp66 166616Dpup 6p66ED161i 009 pHipp011bp 11Dpbe6pDp ED66EzEp66 616DB1p6D6 lEDDE051PED lEDDEDEP15 ()175 61361E6176 1)11E71111 ED1E666EPE 6E717E71E7 DlEDEE66P1 eD361RDED6 0817 P111111101 D6p1p6166D pDale66upo pppe6u631.1. D66611.Dp6p 666EEEEE) 0117 PlEPEOPPE6 6177E066PP 617177161E 661PDPEE6D DEP6PEDEUE 5E13166117 09E E66616Depu DDDD661Dua 6616DD116) EueDe6p6De 6)Depue616 6opeepp1D1 00E 1616Dp666a elpDp61.666 zppeyebuDe Dp6lpp3616 61D61De611 6Dp61e6pDD
OVZ ppb6A6p61. 0E71E5637D DD61PEEDE1 1DE61PEDED u66p6161p1 16e6v66611 081 1-e661p6D61 1.eD1D1DeD6 1.61elpeD16 166e361a1) 3p6pED1161 1D1pppl6pe Asn Ile Lys Tyr Glu val Ala Ile Phe Val His Gly Pro Thr Thr155 val Glu ser His Gly Asn Tyr Ser Thr Gin val Gly Ala Thr Gin170 Ala Gly Arg Phe Ser Ile Thr Pro Ala Ala Pro Ser Tyr Thr Leu185 Lys Leu Gly Glu Tyr Gly Glu val Thr Val Asp Cys Glu Pro Arg200 Ser Gly Ile Asp Thr Asn Ala Tyr Tyr Val Met Thr Val Gly Thr215 Lys Thr Phe Leu Val His Arg Glu Trp Phe met Asp Leu Asn Leu230 Pro Trp Ser Ser Ala Gly Ser Thr Val Trp Arg Asn Arg Glu Thr245 Leu met Glu Phe Glu Glu Pro His Ala Thr Lys Gin Ser val Ile260 Ala Leu Gly Ser Gin Glu Gly Ala Leu His Gin Ala Leu Ala Gly275 Ala Ile Pro Val Glu Phe Ser Ser Asn Thr Val Lys Leu Thr Ser290 Gly His Leu Lys Cys Arg Val Lys Met Glu Lys Leu Gin Leu Lys305 Gly Thr Thr Tyr Gly val Cys Ser Lys Ala Phe Lys Phe Leu Gly320 Thr Pro Ala Asp Thr Gly His Gly Thr Val Val Leu Glu Leu Gin335 Tyr Thr Gly Thr Asp Gly Pro Cys Lys Val Pro Ile Ser Ser Val350 Ala Ser Leu Asn Asp Leu Thr Pro Val Gly Arg Leu Val Thr Val365 Asn Pro Phe Val Ser Val Ala Thr Ala Asn Ala Lys Val Leu Ile380 Glu Leu Glu Pro Pro Phe Gly Asp Ser Tyr Ile Val Val Gly Arg395 Gly Glu Gin Gin Ile Asn His His Trp HiS Lys Ser Gly Ser Ser410 Ile Gly Lys Ala Phe Thr Thr Thr Leu Lys Gly Ala Gin Arg Leu425 Ala Ala Leu Gly Asp Thr Ala Trp Asp Phe Gly Ser Val Gly Gly440 Val Phe Thr Ser Val Gly Lys Ala Val 455 <210> 6 <211> 691 <212> PRT
<213> West-Nile virus <400> 6 Met Gin Lys Lys Arg Gly Gly Lys Thr Gly Ile Ala Val Met Ile Gly Leu Ile Ala Ser Val Gly Ala val Thr Leu Ser Asn Phe Gin Gly Lys Val Met Met Thr Val Asn Ala Thr Asp Val Thr Asp Val Ile Thr Ile Pro Thr Ala Ala Gly Lys Asn Leu Cys Ile Val Arg Ala Met Asp Val Gly Tyr Met Cys Asp Asp Thr Ile Thr Tyr Glu Cys Pro val Leu Ser Ala Gly Asn Asp Pro Glu Asp Ile Asp Cys Trp Cys Thr Lys Ser Ala Val Tyr Val Arg Tyr Gly Arg Cys Thr Lys Thr Arg His Ser Arg Arg Ser Arg Arg Ser Leu Thr Val Gin Thr His Gly Glu Ser Thr Leu Ala Asn Lys Lys Gly Ala Trp Met Asp Ser Thr Lys Ala Thr Arg Tyr Leu Val Lys Thr Glu Ser Trp Ile Leu Arg Asn Pro Gly Tyr Ala Leu val Ala Ala Val Ile Gly Trp Met Leu Gly Ser Asn Thr Met Gin Arg Val Val Phe Val Val Leu Leu Leu Leu Val Ala Pro Ala Tyr Ser Phe Asn Cys Leu Gly Met Ser Asn Arg Asp Phe Leu Glu Gly Val Ser Gly Ala Thr Trp Val Asp Leu Val Leu Glu Gly Asp Ser Cys Val Thr Ile Met Ser Lys Asp Lys Pro Thr Ile Asp Val Lys Met Met Asn Met Glu Ala Val Asn Leu Ala Glu Val Arg Ser Tyr Cys Tyr Leu Ala Thr Val Ser Asp Leu Ser Thr Lys Ala Ala Cys Pro Thr Met Gly Glu Ala His Asn Asp Lys Arg Ala Asp Pro Ala Phe Val Cys Arg Gin Gly Val Val Asp Arg Gly Trp Gly Asn Gly Cys Gly Leu Phe Gly Lys Gly Ser Ile Asp Thr Cys Ala Lys Phe Ala Cys Ser Thr Lys Ala Ile Gly Arg Thr Ile Leu Lys Glu Asn Ile Lys Tyr Glu Val Ala Ile Phe Val His Gly Pro Thr Thr Val Glu Ser His Gly Asn Tyr Ser Thr Gin val Gly Ala Thr Gin Ala Gly Arg Phe Ser Ile Thr Pro Ala Ala Pro Ser Tyr Thr Leu Lys Leu Gly Glu Tyr Gly Glu Val Thr Val Asp Cys Glu Pro Arg Ser Gly Ile Asp Thr Asn Ala Tyr Tyr Val met Thr val Gly Thr Lys Thr Phe Leu val His Arg Glu Trp Phe Met Asp Leu Asn Leu Pro Trp Ser Ser Ala Gly Ser Thr Val Trp Arg Asn Arg Glu Thr Leu Met Glu Phe Glu Glu Pro His Ala Thr Lys Gin Ser Val Ile Ala Leu Gly Ser Gin Glu Gly Ala Leu His Gin Ala Leu Ala Gly Ala Ile Pro Val Glu Phe Ser Ser Asn Thr Val Lys Leu Thr Ser Gly His Leu Lys Cys Arg val Lys Met Glu Lys Leu Gin Leu Lys Gly Thr Thr Tyr Gly Val Cys Ser Lys Ala Phe Lys Phe Leu Gly Thr Pro Ala Asp Thr Gly His Gly Thr Val Val Leu Glu Leu Gin Tyr Thr Gly Thr Asp Gly Pro Cys Lys Val Pro Ile Ser Ser Val Ala Ser Leu Asn Asp Leu Thr Pro val Gly Arg Leu Val Thr Val Asn Pro Phe Val Ser Val Ala Thr Ala Asn Ala Lys Val Leu Ile Glu Leu Glu Pro Pro Phe Gly Asp Ser Tyr Ile Val Val Gly Arg Gly Glu Gin Gin Ile Asn His His Trp His Lys Ser Gly Ser Ser Ile Gly Lys Ala Phe Thr Thr Thr Leu Lys Gly Ala Gin Arg Leu Ala Ala Leu Gly Asp Thr Ala Trp Asp Phe Gly Ser val Gly Gly Val Phe Thr Ser val Gly Lys Ala Val His Girl Val Phe Gly Gly Ala Phe Arg Ser Leu Phe Gly Gly Met Ser Trp Ile Thr Gln Gly Leu Leu Gly Ala Leu Leu Leu Trp Met Gly Ile Asn Ala Arg Asp Arg Ser Ile Ala Leu Thr Phe _ Leu Ala Val Gly Gly Val Leu Leu Phe Leu Ser Val Asn Val His680 Ala <210> 7 <211> 368 <212> PRT
<213> West-Nile virus <400> 7 Met Arg Ser Ile Ala Leu Thr Phe Leu Ala Val Gly Gly Val Leu5 Leu Phe Leu Ser Val Asn Val HiS Ala Asp Thr Gly Cys Ala Ile20 Asp Ile Ser Arg Gln Glu Leu Arg Cys Gly Ser Gly Val Phe Ile35 His Asn Asp Val Glu Ala Trp Met AS Arg Tyr Lys Tyr Tyr Pro 50 Glu Thr Pro Gin Gly Leu Ala Lys Ile Ile Gin Lys Ala His Lys Glu Gly Val Cys Gly Leu Arg Ser Val Ser Arg Leu Glu His Gin80 Met Trp Glu Ala Val Lys Asp Glu Leu Asn Thr Leu Leu Lys Glu Asn Gly Val Asp Leu Ser Val val Val Glu Lys Gin Glu Gly Met Tyr Lys Ser Ala Pro Lys Arg Leu Thr Ala Thr Thr Glu Lys Leu Glu Ile Gly Trp Lys Ala Trp Gly Lys Ser Ile Leu Phe Ala Pro Glu Leu Ala Asn Asn Thr Phe val Val Asp Gly Pro Glu Thr Lys Glu Cys Pro Thr Gin Asn Arg Ala Trp Asn Ser Leu Glu Val Glu Asp Phe Gly Phe Gly Leu Thr Ser Thr Arg Met Phe Leu Lys Val Arg Glu Ser Asn Thr Thr Glu Cys Asp Ser Lys Ile Ile Gly Thr Ala Val Lys Asn Asn Leu Ala Ile His Ser Asp Leu Ser Tyr Trp Ile Glu Ser Arg Leu Asn Asp Thr Trp Lys Leu Glu Arg Ala Val Leu Gly Glu val Lys Ser Cys Mr Trp PrO Glu Thr His Thr Leu Trp Gly Asp Gly Ile Leu Glu Ser Asp Leu Ile Ile Pro Val Thr Leu Ala Gly Pro Arg Ser Asn His Asn Arg Arg Pro Gly Tyr Lys Thr Gin Asn Gin Gly Pro Trp Asp Glu Gly Arg Val Glu Ile Asp Phe Asp Tyr Cys Pro Gly Thr Thr Val Thr Leu Ser Glu Ser Cys Gly His Arg Gly Pro Ala Thr Arg Thr Thr Thr Glu Ser Gly Lys Leu Ile Thr Asp Trp Cys Cys Arg Ser Cys Thr Leu Pro Pro Leu Arg Tyr Gin Thr Asp Ser Gly Cys Trp Tyr Gly Met Glu Ile Arg Pro Gin Arg His AS Glu Lys Thr <210> 8 <211> 679 <212> PRT
<213> Dengue type-1 virus <400> 8 Met Asn Arg Arg Lys Arg Ser Val Thr Met Leu Leu Met Leu Leu Pro Thr Val Leu Ala Phe His Leu Thr Thr Arg Gly Gly Glu Pro His Met Ile Val Ser Lys Gin Glu Arg Gly Lys Ser Leu Leu Phe Lys Thr Ser Ala Gly Val Asn Met Cys Thr Leu Ile Ala Met Asp Leu Gly Glu Leu Cys Glu Asp Thr Met Thr Tyr Lys Cys Pro Arg Ile Thr Glu Ala Glu Pro Asp Asp Val Asp Cys Trp Cys Asn Ala Thr Asp Thr Trp Val Thr Tyr Gly Thr Cys Ser Gin Thr Gly Glu His Arg Arg Asp Lys Arg Ser Val Ala Leu Ala Pro His Val Gly Leu Gly Leu Glu Thr Arg Thr Glu Thr Trp Met Ser Ser Glu Gly Ala Trp Lys Gin Ile Gin Lys Val Glu Thr Trp Ala Leu Arg His Pro Gly Phe Thr Val Ile Ala Leu Phe Leu Ala His Ala Ile Gly Thr Ser Ile Thr Gin Lys Gly Ile Ile Phe Ile Leu Leu Met Leu Val Thr Pro Ser Met Ala Met Arg Cys Val Gly Ile Gly ASn Arg , Asp Phe Val Glu Gly Leu Ser Gly Ala Thr Trp Val Asp Val Val 200 Leu Glu HIS Gly Ser Cys Val Thr Thr Met Ala Lys Asn Lys Pro 215 Thr Leu Asp Ile Glu Leu Leu Lys Thr Glu val Thr Asn Pro Ala 230 Val Leu Arg Lys Leu Cys Ile Glu Ala Lys Ile Ser Asn Thr Thr 245 Thr Asp ser Arg Cys Pro Thr Gin Gly Glu Ala Thr Leu Val Glu 260 Glu Gin Asp Ala Asn Phe Val Cys Arg Arg Thr Val val Asp Arg 275 Gly Trp Gly Asn Gly Cys Gly Leu Phe Gly Lys Gly Ser Leu Leu 290 Thr Cys Ala Lys Phe Lys Cys Val Thr Lys Leu Glu Gly Lys Ile 305 val Gin Tyr Glu Asn Leu Lys Tyr Ser Val Ile Val Thr Val His 320 Thr Gly Asp Gin His Gin val Gly Asn Glu Thr Thr Glu His Gly 335 Thr Ile Ala Thr Ile Thr Pro Gin Ala Pro Thr Ser Glu Ile Gin 350 Leu Thr Asp Tyr Gly Thr Leu Thr Leu Asp Cys Ser Pro Arg Thr 365 Gly Leu Asp Phe Asn Glu val Val Leu Leu Thr Met Lys Glu Lys 380 Ser Trp Leu Val His Lys Gin Trp Phe Leu Asp Leu Pro Leu Pro 395 Trp Thr Ser Gly Ala Ser Thr Ser Gin Glu Thr Trp ASrl Arg Gin 410 Asp Leu Leu Val Thr Phe Lys Thr Ala His Ala Lys Lys Gin Glu 425 val Val Val Leu Gly Ser Gin Glu Gly Ala Met His Thr Ala Leu 440 Thr Gly Ala Thr Glu 455 Ile Gin Thr Ser Gly Thr Thr Thr Ile Phe 460 .
Ala Gly His Leu Lys Cys Arg Leu Lys Met Asp Lys Leu Thr Leu 470 Lys Gly Met Ser Tyr Val Met Cys Thr Gly Ser Phe Lys Leu Glu 485 Lys Glu Val Ala Glu Thr Gin His Gly Thr val Leu Val Gin Val 500 Lys Tyr Glu Gly Thr Asp Ala Pro Cys Lys Ile Pro Phe Ser Thr 515 Gin AS Glu Lys Gly Val Thr Gin Asn Gly Arg Leu Ile Thr Ala 530 Asn Pro Ile Val Thr Asp Lys Glu Lys Pro Ile Asn Ile Glu Thr Glu Pro Pro Phe Gly Glu Ser Tyr Ile Ile Val Gly Ala Gly Glu Lys Ala Leu Lys Leu Ser Trp Phe Lys Lys Gly Ser Ser Ile Gly Lys Met Phe Glu Ala Ile Ala Arg Gly Ala Arg Arg Met Ala Ile Leu Gly Asp Thr Ala Trp Asp Phe Gly Ser Ile Gly Gly Val Phe Thr Ser Val Gly Lys Leu Val His Gin Val Phe Gly Thr Ala Tyr Gly Val Leu Phe Ser Gly Val Ser Trp Thr Met Lys Ile Gly Ile Gly Ile Leu Leu Thr Trp Leu Gly Leu Asn Ser Arg Ser Ala Ser Leu Ser Met Thr Cys Ile Ala Val Gly Met Val Thr Leu Tyr Leu Gly Val Met Val <210> 9 <211> 33 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Synthetic DNA

<400> 9 tatcgtacga tgagagttgt gtttgtcgtg cta 33 <210> 10 <211> 30 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Synthetic DNA

<400> 10 atagcgcgct tagacagcct tcccaactga 30 <210> 11 <211> 33 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Synthetic DNA

<400> 11 tatcgtacga tgcaaaagaa aagaggagga aag 33 <210> 12 <211> 31 <212> DNA
<213> Artificial sequence <220>
<223> Description of Artificial Sequence:Synthetic DNA

<400> 12 atagcgcgct taagcgtgca cgttcacgga g 31 <210> 13 <211> 30 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Synthetic DNA

<400> 13 tatcgtacga tgaggtccat agctctcacg 30 <210> 14 <211> 34 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Synthetic DNA

<400> 14 atagcgcgct cattaggtct tttcatcatg tctc 34 <210> 15 <211> 33 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial Sequence:Synthetic DNA

<400> 15 tatcgtacga tgaacaggag gaaaagatcc gtg 33 <210> 16 <211> 33 <212> DNA
<213> Artificial Sequence <220>
<223> Description of Artificial sequence:Synthetic DNA

<400> 16 atagcgcgct taaaccatga ctcctaggta cag 33

Claims (13)

WHAT IS CLAIMED IS:
1. A recombinant virus which is a live attenuated or a defective measles virus comprising a polynucleotide coding for a polypeptide which is a West-Nile virus or respectively a Dengue virus antigen which is selected in the group of:
= the heterodimer glycoproteins PreM-E;
= a secreted envelope glycoprotein sE; and = a polypeptide having at least 80% identity, or 85% identity with the polypeptide having the sequence of SEQ ID NO:6, SEQ ID NO: 5 or SEQ
ID NO:8, said recombinant virus expressing said polypeptide and said polypeptide inducing a protective immune response against a West-Nile virus in an animal or respectively against a Dengue virus in an animal.
2. The recombinant virus according to claim 1, wherein the polynucleotide encodes a polypeptide selected in the group of:
= the heterodimer glycoproteins PreM-E which comprises the sequence of SEQ ID NO: 6 or SEQ ID NO: 8 or has the sequence of SEQ ID NO: 6 or SEQ ID NO: 8.
= the secreted envelope protein sE which has the sequence of SEQ ID
NO: 5.
3. The recombinant virus according to any one of claims 1 to 2, wherein said polynucleotide comprises the sequence of SEQ ID NO: 2 or is the sequence of SEQ ID NO: 1 or comprises the sequence of SEQ ID NO:4.
4. The recombinant measles virus according to any one of claims 1 to 3, comprising, in its genome, said polynucleotide.
5. The recombinant measles virus according to any one of claims 1 to 4, which is derived from the Schwarz measles virus strain.
6. A pharmaceutical composition comprising :
a) at least a recombinant measles virus according to any one of claims 1 to 5 and b) a pharmaceutically acceptable vehicle or carrier.
7. The pharmaceutical composition of claim 6, inducing a protective immunity against a West-Nile virus in an animal or respectively inducing a protective immunity against a Dengue virus, in an animal.
8. Use of a pharmaceutical composition according to claim 7, as an anti-West-Nile virus agent or respectively an anti-Dengue virus agent, or for the preparation of an anti-West-Nile virus vaccine or respectively an anti-Dengue virus vaccine.
9. A host cell incorporating a recombinant virus as defined in any one of claims 1 to 5.
10. A method of producing a recombinant virus for the preparation of an anti-West-Nile virus vaccine, the method comprising the steps of:
a) providing a host cell as defined in claim 9;

b) placing the host cell from step a) in conditions permitting the replication of a recombinant virus as defined in any one of claims 1 to 6, which is capable of expressing a said polypeptide; and c) isolating the recombinant virus produced in step b).
11. The host cell according to claim 9, which is the cell line deposited at the C. N.C.M. under accession number I-3018.
12. Use of an effective amount of at least a recombinant measles virus according to any one of claims 1 to 5, for the preparation of a medicament intended for treating and/or preventing a West-Nile virus-associated disease or respectively a Dengue virus associated disease, or infection in an animal.
13. Use of an effective amount of at least a recombinant measles virus according to any one of claims 1 to 5, for treating and/or preventing a West-Nile virus-associated disease or respectively a Dengue virus associated disease, or infection in an animal.
CA2517258A 2003-02-26 2004-02-26 Recombinant measles virus comprising dengue or west nile virus polynucleotides, and their use in vaccinal and therapeutic applications Expired - Lifetime CA2517258C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA2517258A CA2517258C (en) 2003-02-26 2004-02-26 Recombinant measles virus comprising dengue or west nile virus polynucleotides, and their use in vaccinal and therapeutic applications

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CA2.420.092 2003-02-26
CA002420092A CA2420092A1 (en) 2003-02-26 2003-02-26 New dengue and west nile viruses proteins and genes coding the foregoing, and their use in vaccinal, therapeutic and diagnostic applications
CA2.432.738 2003-06-20
CA002432738A CA2432738A1 (en) 2003-02-26 2003-06-20 New dengue and west nile viruses proteins and genes coding the foregoing, and their use in vaccinal, therapeutic and diagnostic applications
CA2517258A CA2517258C (en) 2003-02-26 2004-02-26 Recombinant measles virus comprising dengue or west nile virus polynucleotides, and their use in vaccinal and therapeutic applications
PCT/IB2004/001027 WO2004076619A2 (en) 2003-02-26 2004-02-26 New dengue and west nile viruses proteins and genes coding the foregoing, and their use in vaccinal, therapeutic and diagnostic applications

Publications (2)

Publication Number Publication Date
CA2517258A1 CA2517258A1 (en) 2004-09-10
CA2517258C true CA2517258C (en) 2013-05-28

Family

ID=35415103

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2517258A Expired - Lifetime CA2517258C (en) 2003-02-26 2004-02-26 Recombinant measles virus comprising dengue or west nile virus polynucleotides, and their use in vaccinal and therapeutic applications

Country Status (1)

Country Link
CA (1) CA2517258C (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114410640B (en) * 2022-02-25 2023-07-11 世通兰达(深圳)生物科技发展有限公司 Aptamer for detecting measles virus, kit and application

Also Published As

Publication number Publication date
CA2517258A1 (en) 2004-09-10

Similar Documents

Publication Publication Date Title
US7556812B2 (en) Dengue and West Nile viruses proteins and genes coding the foregoing, and their use in vaccinal, therapeutic and diagnostic applications
US10519466B2 (en) Recombinant measles viruses expressing epitopes of antigens of RNA viruses—use for the preparation of vaccine compositions
Brandler et al. Pediatric measles vaccine expressing a dengue tetravalent antigen elicits neutralizing antibodies against all four dengue viruses
Despres et al. Live measles vaccine expressing the secreted form of the West Nile virus envelope glycoprotein protects against West Nile virus encephalitis
JP5538729B2 (en) Mock infectious flaviviruses and their use
Brandler et al. Pediatric measles vaccine expressing a dengue antigen induces durable serotype-specific neutralizing antibodies to dengue virus
EP2371966B1 (en) Recombinant lentiviral vector for expression of a Flaviviridae protein and applications thereof as a vaccine
EP1625852A1 (en) Chimeric flavivirus vaccines
KR20180137514A (en) Vaccination compositions and methods against dengue virus in children and young adults
IL202978A (en) West nile virus vaccine
Tangy et al. Live attenuated measles vaccine as a potential multivalent pediatric vaccination vector
Brandler et al. Recombinant vector derived from live attenuated measles virus: potential for flavivirus vaccines
CN106574271B (en) Chimeric west nile/dengue viruses and methods of use
CA2517258C (en) Recombinant measles virus comprising dengue or west nile virus polynucleotides, and their use in vaccinal and therapeutic applications
CA2456873A1 (en) West nile virus proteins and genes coding the foregoing and their use in vaccinal, therapeutic and diagnostic applications
TANGY et al. Sommaire du brevet 2517258
US11739348B2 (en) Recombinant vectors encoding Zika virus protein subunits
TANGY et al. Patent 2517258 Summary
Li Non-Segmented Negative-Sense RNA Viruses as Vectors for Developing Vaccine Candidates against Zika Virus Infection
Brandler et al. Pediatric Measles Vaccine Expressing a Dengue Antigen Induces Durable
Abdelgalel Expression of Dengue virus envelope glycoproteins using a Measles vaccine vector
Iyer Phylogenetics and New Approaches to Vaccine Development for West Nile

Legal Events

Date Code Title Description
EEER Examination request