Recombinant lentiviral vector for expression of a flaviviridae protein and applications thereof as a vaccine

Despres; Philippe ;   et al.

Patent Application Summary

U.S. patent application number 11/596675 was filed with the patent office on 2009-08-27 for recombinant lentiviral vector for expression of a flaviviridae protein and applications thereof as a vaccine. Invention is credited to Pierre Charneau, Philippe Despres, Marie-Pascale Frenkiel, Frederic Tangy.

Application Number20090214589 11/596675
Document ID /
Family ID34945000
Filed Date2009-08-27

United States Patent Application 20090214589
Kind Code A1
Despres; Philippe ;   et al. August 27, 2009

Recombinant lentiviral vector for expression of a flaviviridae protein and applications thereof as a vaccine

Abstract

Use of a recombinant lentiviral vector comprising a polynucleotide fragment encoding at least one protein of a virus of the family Flaviviridae or an immunogenic peptide of at least 8 amino acids of said protein, for preparing a pharmaceutical composition intended for the prevention and/or the treatment of a Flaviviridae infection in a sensitive species.


Inventors: Despres; Philippe; (La Garenne Colombes, FR) ; Charneau; Pierre; (Paris, FR) ; Tangy; Frederic; (Les Lilas, FR) ; Frenkiel; Marie-Pascale; (Levallois, FR)
Correspondence Address:
    FINNEGAN, HENDERSON, FARABOW, GARRETT & DUNNER;LLP
    901 NEW YORK AVENUE, NW
    WASHINGTON
    DC
    20001-4413
    US
Family ID: 34945000
Appl. No.: 11/596675
Filed: May 16, 2005
PCT Filed: May 16, 2005
PCT NO: PCT/IB05/01753
371 Date: August 4, 2008

Current U.S. Class: 424/204.1 ; 435/320.1; 435/325; 435/5; 435/69.1
Current CPC Class: Y02A 50/30 20180101; Y02A 50/386 20180101; C12N 15/86 20130101; C12N 2770/24134 20130101; C07K 14/005 20130101; A61P 31/12 20180101; Y02A 50/388 20180101; Y02A 50/394 20180101; C12N 2740/16043 20130101; A61K 2039/53 20130101; A61P 31/14 20180101
Class at Publication: 424/204.1 ; 435/320.1; 435/325; 435/69.1; 435/5
International Class: A61K 39/12 20060101 A61K039/12; C12N 15/867 20060101 C12N015/867; C12N 5/00 20060101 C12N005/00; C12P 21/06 20060101 C12P021/06; C12Q 1/70 20060101 C12Q001/70

Foreign Application Data

Date Code Application Number
May 17, 2004 FR 0405366

Claims



1. Use of a recombinant lentiviral vector comprising a polynucleotide fragment encoding at least one protein of a virus of the family Flaviviridae or an immunogenic peptide of at least 8 amino acids of said protein, for preparing an immunogenic composition intended for the prevention and/or the treatment of a Flaviviridae infection in a sensitive species.

2. Use according to claim 1, characterized in that said recombinant lentiviral vector is of triplex type.

3. Use according to claim 1 or claim 2, characterized in that said recombinant lentiviral vector comprises a 3' LTR in which the promoter and the activator have been deleted from the U3 region.

4. Use according to any one of claims 1 to 3, characterized in that said recombinant lentiviral vector is pseudotyped with at least one envelope protein of another virus.

5. Use according to claim 4, characterized in that said envelope protein is the vesicular stomatitis virus glycoprotein G.

6. Use according to any one of claims 1 to 5, characterized in that said polynucleotide fragment encodes at least one structural protein of a Flaviviridae or an immunogenic peptide of at least 8 amino acids of said protein.

7. Use according to claim 6, characterized in that said structural protein is a membrane protein or an envelope protein.

8. Use according to any one of claims 1 to 5, characterized in that said polynucleotide fragment encodes at least one non-structural protein of Flaviviridae or an immunogenic peptide of at least 8 amino acids of said protein.

9. Use according to any one of claims 1 to 8, characterized in that said polynucleotide fragment encodes both at least one structural protein of a Flaviviridae or an immunogenic peptide of at least 8 amino acids of said protein, and at least one non-structural protein or a fragment of at least 8 amino acids of said non-structural protein of the same Flaviviridae.

10. Use according to any one of claims 1 to 9, characterized in that said immunogenic peptide consists of the combination of immunogenic fragments of various Flaviviridae and/or of different serotypes of the same Flaviviridae.

11. Use according to any one of claims 1 to 10, characterized in that said Flaviviridae is selected from the group consisting of West Nile virus, dengue virus, yellow fever virus and hepatitis C virus.

12. Use according to any one of claims 1 to 11, characterized in that said polynucleotide fragment is selected from the group consisting of: the cDNAs encoding an E protein and, optionally, a prM or M protein, and/or a C protein, and/or a non-structural protein of West Nile virus or of dengue virus, and the cDNAs encoding one or more immunogenic peptides of at least 8 amino acids of the above proteins, the cDNAs encoding an E1 or E2 protein or an E1/E2 heterodimer, and/or a C protein according to a 0, +1 or +2 reading frame, and/or an NS3 protein of hepatitis C virus, and the cDNAs encoding one or more immunogenic peptides of at least 8 amino acids of the above proteins, the cDNAs encoding one or more different domains III (positions 295 to 394) of an E protein of dengue virus, each corresponding to one of the four types of dengue virus.

13. Use according to claim 12, characterized in that said cDNA encodes the four domains III, the sequences of which are represented by SEQ ID NOs. 1-4 in the sequence listing attached in the appendix.

14. Use according to claim 12, characterized in that said cDNA encoding a C protein according to a +1 or +2 reading frame is selected from the group consisting of the sequences SEQ ID NOs. 5 to 14.

15. Use according to any one of claims 1 to 12, characterized in that said polynucleotide is a fragment of a coding sequence of a Flaviviridae corresponding to an accession number in the NCBI database selected from the group consisting of: M23027, M19197, M93130, M14931, M12294, AF481864, M18370, X03700, U27495, M73835, M31182, M31768, J04358; M62321, D90208 and M58335.

16. Recombinant lentiviral vector as defined in any one of claims 1 to 6 and 8 to 15, this vector comprising a polynucleotide encoding at least one structural protein or a fragment of said protein and, optionally, a non-structural protein or a fragment of said protein, as defined in claim 6.

17. Recombinant vector according to claim 16, characterized in that it is of triplex type.

18. Recombinant lentiviral vector according to claim 16 or claim 17, characterized in that it comprises a cDNA selected from the group consisting of: a cDNA encoding at least one E protein and, optionally, a prM or M protein, and/or a C protein, and/or a non-structural protein of West Nile virus or of dengue virus, and a cDNA encoding one or more immunogenic peptides of at least 8 amino acids of the above proteins, a cDNA encoding an E1 or E2 protein or an E1/E2 heterodimer, and/or a C protein according to a 0, +1 or +2 reading frame and, optionally, an NS3 protein of hepatitis C virus, and a cDNA encoding one or more immunogenic peptides of at least 8 amino acids of the above proteins, and a cDNA encoding a domain III (positions 295 to 394) or several different domains III of an E protein of dengue virus, each corresponding to one of the four types of dengue virus.

19. Recombinant lentiviral vector according to claim 18, characterized in that said cDNA encoding a C protein according to a +1 or +2 reading frame is selected from the group consisting of the sequences SEQ ID NOs. 5 to 14.

20. Recombinant lentiviral vector according to claim 18, characterized in that said cDNA encodes the four domains III the sequences of which are represented by SEQ ID NOs. 1-4.

21. Recombinant lentiviral vector according to claim 18, characterized in that it comprises the cDNA encoding an E protein and a prM protein of a dengue virus or of West Nile virus.

22. Recombinant lentiviral vector according to claim 18, characterized in that it comprises the cDNA encoding a secreted form of the E protein of the IS-98-ST1 strain of West Nile virus, which vector is included in a microorganism deposited under the No. I-3076, on 27 Aug. 2003, with the Collection Nationale de Cultures de Microorganismes [National Collection of Cultures of Microorganisms], 25 rue du Docteur Roux, 757244 Paris Cedex 15.

23. Recombinant lentiviral vector according to any one of claims 16 to 22, characterized in that said polynucleotide fragment is placed under the control of the CMV promoter.

24. Immunogenic composition, characterized in that it comprises at least one recombinant lentiviral vector as defined in claims 1 to 23.

25. Composition according to claim 24, characterized in that it comprises a pharmaceutically acceptable vehicle and, optionally, a carrier substance.

26. Composition according to claim 24 or claim 25, characterized in that said recombinant lentiviral vector is of triplex type.

27. Composition according to any one of claims 24 to 26, characterized in that it comprises viral particles of said recombinant lentiviral vector, pseudotyped with an envelope protein of another virus.

28. Composition according to claim 27, characterized in that said envelope protein is the vesicular stomatitis virus glycoprotein G.

29. Composition according to claim 26, characterized in that it comprises an isolated nucleic acid molecule corresponding to the recombinant genome of said lentiviral vector of triplex type, which nucleic acid molecule comprises: (i) the regulatory sequences for encapsidation, reverse transcription and integration and the cis-active sequences for central initiation and termination of lentiviral origin and, optionally, the regulatory sequences for the Rev protein and (ii) a polynucleotide fragment encoding a Flaviviridae protein or an immunogenic peptide of at least 8 amino acids of said protein as defined above in any one of claims 1 and 6 to 15.

30. Cells, preferably eukaryotic cells, modified with a recombinant lentiviral vector as defined in claims 1 to 23.

31. Method for producing viral proteins of Flaviviridae and/or immunogenic fragments of said proteins or else viral pseudoparticles, characterized in that it comprises at least the following steps: a) culturing modified cells according to claim 30, under conditions which allow the expression of one or more viral proteins of Flaviviridae and/or of one or more of the immunogenic fragments of said proteins encoded by said recombinant lentiviral vector, and b) separating said proteins, protein fragments or viral pseudoparticles from the culture supernatant or from the cells in a).

32. Method for screening antiviral compounds, characterized in that it comprises: bringing modified eukaryotic cells according to claim 30, in particular eukaryotic cells modified with a vector comprising a cDNA encoding a non-structural protein of Flaviviridae such as NS3 or NS5, into contact with various compounds of a library to be tested, measuring the activity of said protein, in the presence or in the absence of said compounds, and selecting the compounds capable of modulating said activity.

33. Method according to claim 32, characterized in that the eukaryotic cells are selected from the group consisting of dendritic cells, neuronal cells and hepatocytes.

34. Method for diagnosing infection with a Flaviviridae, using a sample of biological fluid from an individual of a sensitive species, characterized in that it comprises at least the following steps: a) bringing said biological sample into contact with modified eukaryotic cells expressing at least one Flaviviridae antigen, as defined in claim 30, and b) revealing the antigen-antibody complexes formed in (a).

35. Method according to claim 34, characterized in that said modified eukaryotic cells in a) are permeabilized.

36. Method for diagnosing an infection with a Flaviviridae, using a sample of biological fluid from an individual of a sensitive species, characterized in that it comprises at least the following steps: a) bringing said biological sample into contact with viral pseudoparticles produced from the culture supernatant of cells modified with a lentiviral vector expressing at least one membrane protein and/or envelope protein as defined in any one of claims 7, 9 to 12, 15 to 18 and 21 to 23, and b) revealing the antigen-antibody complexes formed in (a).

37. Kit for carrying out the methods according to claims 31 to 36, characterized in that it comprises at least modified cells according to claim 30.
Description



[0001] The present invention relates to a recombinant lentiviral vector for expression of a protein of a Flaviviridae and to its applications as a vaccine intended for the prevention and/or treatment of an infection with a virus of the family Flaviviridae, in a sensitive species (host or reservoir)

[0002] The family Flaviviridae is divided up into three genera: Flavivirus, Pestivirus and Hepacivirus or hepatitis C virus; Flaviviridae represent a major human and veterinary health problem due to the large number of both human and veterinary diseases induced by Flaviviridae. Specifically, there are, for example, more than 70 species of Flavivirus, at least 50% of which are the cause of human or veterinary diseases.

[0003] Flaviviridae are small enveloped viruses. Their genome is a single-stranded RNA molecule of positive polarity, of 9.5 kb to 12.5 kb, depending on the Flaviviridae, and contains a single open reading frame flanked by two short non-coding regions at its 5' and 3' ends. This open reading frame is translated into a polyprotein, which is the precursor of the structural proteins, in its N-terminal portion, and of. the non-structural (NS) proteins, in its C-terminal portion.

[0004] More precisely: [0005] for the Flaviviruses, the genome is a single-stranded RNA molecule of positive polarity, of approximately 10-12 kbases. The genomic RNA is combined with several copies of the capsid protein C so as to form the nucleocapsid; it is surrounded by a viral envelope consisting of a double lipid layer derived from the endoplasmic reticulum (ER) membranes, in which the envelope protein E and the membrane protein M are anchored. The Flavivirus genomic RNA contains a single open reading frame of approximately 10 500 nucleotides, flanked by two short non-coding regions at its 5' and 3' ends. The genome is translated into a polyprotein of approximately 3400 amino acids, which is the precursor of the three structural proteins C, prM (intracellular precursor of M) and E, in its N-terminal portion, and of at least five non-structural (NS) proteins NS1 to NS5, in its C-terminal portion. The following structure is therefore observed: C-prM/M-E-NS1-NS2A/2B-NS3-NS4A/4B-NS5, [0006] for the Pestiviruses, the genomic RNA is longer than 12 kbases, and contains a single open reading frame translated into a polyprotein of approximately 3900 amino acids, which is the precursor of 11 to 13 pestiviral proteins, four of which are structural proteins: the following structure is observed: N.sup.pro-C.sup.ems-E1-E2-p7-NS2-NS3-NS4A/4B-NS5A/5B and [0007] for the Hepaciviruses, the genomic RNA comprises approximately 9.5 kbases, and contains a single open reading frame translated into a polyprotein of approximately 3000 amino acids, which is the precursor of the three structural proteins C, E1 and E2, in its N-terminal portion, and of at least seven non-structural (NS) proteins NS1 to NS5, in its C-terminal portion. The following structure is observed: C-E1-E2-NS1-NS2-NS3-NS4A/4B-NS5A/5B.

[0008] Many serious human and animal pathologies are induced by the viruses of this family; according to the infecting virus, the various symptoms observed are generally fever (cyclic or non-cyclic), haemorrhagic fever, diarrhoea, encephalitis, hepatitis or septic shock. More precisely, the various viruses in question are the following: [0009] Flaviviruses: the majority of Flaviviruses are transmitted to the vertebrate host by mosquitoes (Culex, Aaedes, Anopheles or Mansonia) or ticks: (i) viruses transmitted by mosquitoes: dengue virus (types 1 to 4), yellow fever virus (YFV), Japanese encephalitis virus (JEV), West Nile virus (WNV), Murray Valley encephalitis virus (MVEV), Saint-Louis encephalitis virus (SLEV) and (ii) viruses transmitted by ticks: tick-borne encephalitis virus (TBEV), Kyasanur forest disease virus, Omsk haemorrhagic fever virus and Louping ill virus. [0010] Pestiviruses: border disease virus (BDV), bovine viral diarrhoea virus (BVDV) and classical swine fever virus (CSFV) or hog cholera virus. [0011] Hepaciviruses: hepatitis C virus and hepatitis G virus.

[0012] Migratory birds can be the reservoir of some of these viruses, in particular the West Nile virus, which has also been noted to cross the species barrier, in horses and humans.

[0013] A certain number of vaccine strategies have been proposed to date (Gould E A: Flavivirus Infections in Humans, Encyclopaedia of Life Sciences, 2001; Pugazchev K V et al., Internat. J. Parasitol, 2003, 33, 567-582; Putnak R et al., Advances in Virus Research 0.2003, 61, 445-468; Smith D B, Hepatitis C virus, Encyclopaedia of Life Sciences, 2001) and relate to: [0014] vaccines containing live attenuated viruses or inactivated viruses (Pugachev K V et al., 2003, mentioned above; Gould E A, 2001, mentioned above; Brinton M A, Annu. Rev. Microbiol., 2002, 56, 371-402; Hamers C. et al., Vet. Rec., 2003, 153, 8, 236-240; Kovacs F. et. al., Vet. Microbiol., 2003, 96, 2, 117-131); [0015] vaccines containing viral subunits; [0016] vaccines containing one or more virus-derived antigens (Wang T et al., J. Immunol., 2001, 167, 5273-5277); [0017] vaccines containing chimeric viruses (Pugachev K V et al., 2003, mentioned above); or [0018] DNA vaccines (Putnak R et al., 2003, mentioned above; Turell M J et al., Emerging Infectious Diseases, 2003, 9, 9, 1077-1081; Davis B S et al., J. Virol., 2001, 4040-4047; Pan C H et al., J. Virol., 2001, 75, 23, 11457-11463); these vaccines use various vectors. In particular, Putnak R et al., 2003, mentioned above, specifies that, for optimum expression, the most appropriate regulatory elements should be chosen (promoter and enhancer); in general, at the very least for the Flaviviruses, it is recommended to use plasmid vectors comprising a CMV promoter (plasmid pcDNA3, Invitrogen, for example) or RSV promoter and coexpressing the prM and E genes and, optionally, also at least one non-structural protein.

[0019] Taking, for example, WNV, the emergence of which in the northern hemisphere, and in particular in the United States, is quite recent, the various vaccine strategies currently proposed to combat West Nile virus infection are as follows: [0020] Japanese encephalitis virus produced in mouse brain, inactivated with formol (JE-VAX.RTM., Aventis-Pasteur; Monath et al., Curr. Drug Targets Infect. Disord., 2001, 1, 37-50); the existence of a cross protection capable of protecting humans or horses against West Nile viral infection has not been demonstrated and is controversial (Monath, A M; Trop. Med. Hyg., 2002, 66, 113-114). In addition, studies in mice have shown that cross immunity could induce brain inflammation during West Nile infection; [0021] formol-inactivated West Nile virus (International Application WO 03/061555); this vaccine proposed for the immunization of horses has been found to be devoid of any pathogenic effect and effective against West Nile virus infection in horses; however due to the low magnitude humoral response, several injections, followed by annual boosts are required; [0022] chimeric virus derived from the attenuated strain of the yellow fever virus (strain 17D; ChimeriVax.TM.-WN); more precisely, the ChimeriVax.RTM.-West Nile chimeric virus contains the prM-E cassette of WNV strain New York 1999, in the attenuated virus YV 17D (International Application WO 03/059384 and Pletnev A G et al., PNAS, 2002, 99, 5, 3036-3041; Monath T P et al., Curr. Drug Targets Disord., 2001, 1, 1, 37-50); the prM and E genes of the West Nile virus are inserted into the yellow fever virus or the dengue virus, which therefore serve as vector. The genes encoding the nucleocapsid protein and the non-structural proteins, and also the non-translated terminal regions that originate from strain 17D or from DEN4, are used for replication of the recombinant chimeric virus. The chimeric viruses replicate in the host like the 17D or DEN4 virus, but immunize specifically against the West Nile virus (Monath et al., Curr. Drug Targets Infect. Disord., mentioned above) Infection with the chimeric virus stimulates the various pathways of the immune response. In addition, the chimeric viral particles contain the complete E protein, which has redundant neutralizing epitopes. Thus, replication of the chimeric virus in the host induces high titres of neutralizing antibodies that prevent early dissemination of the virus, and the cytotoxic T immunity eliminates the virus that has succeeded in infecting cells. The post-infection memory response, which is rapid and stronger than the post-vaccine response, also contributes to the protection against West Nile infection. It has been shown that prior immunization with the 17D strain does not inhibit infection with the chimeric virus but, on the contrary, it increases the production of specific antibodies. It has also been shown, in mice and non-human primates, that the ChimeriVax.TM.-JE chimeric vaccine is less neurovirulent than the 17D strain. In addition, the genome of the chimeric virus is stable over repeated passages in vivo and in cell culture. The ChimeriVax.TM.-WN chimeric viruses are derived from a vaccine strain that has proved its innocuousness and its effectiveness in humans since it was developed for human immunization more than 65 years ago, and used in several hundred million individuals (Monath et al., Curr. Drug Targets Infect. Disord., mentioned above); however the use of chimeric live-attenuated virus raises safety concerns; non-homologous recombination between different species is possible as demonstrated by naturally occurring recombinant flaviviruses (Seligman S J and Gould E A, Lancet, 2004, 363, 2073-2075). [0023] naked DNA (Davis et al., J. Virol., 2001, 75: 4040-4047; Turell et al., Emerg. Infect. Diseases, 2003, 9, 1077-1081 and International Application WO 03/061555); the naked DNA vector used is a vector pCBWN comprising the cytomegalovirus early promoter, a sequence encoding a signal peptide, derived from the Japanese encephalitis virus, and the sequences encoding the prM and E proteins of the West Nile virus. It has been shown that a simple intramuscular injection of this plasmid induces protective immunity against West Nile infection, in mice and horses; [0024] recombinant protein E (Wang et al., J. Immunol., 2001, 167, 5273-5277); the complete E protein or the E protein from which the C-terminal region has been deleted (residues E1 to E409), expressed in the form of a fusion protein in E. coli and purified by affinity chromatography, induces the production of neutralizing antibodies directed against the E protein, in mice. The soluble E protein, from which the C-terminal region has been deleted, induces complete protection in mice, whereas only partial protection is observed with the complete E protein.

[0025] Even though most of the vaccines currently proposed are effective overall, there is nevertheless still a need for new preventive measures, and especially in the field of DNA vaccines with respect to Flaviviridae; in particular, there exists a real need for vectors that are useful both in the prevention of diseases induced by these viruses in human medicine and veterinary medicine, and in the eradication of these viruses in the reservoirs.

[0026] In fact, in the case of Hepaciviruses, for example, and more particularly of hepatitis C, trials aimed at protecting patients suffering from hepatitis C fail because the vaccinia virus is used to express the HCV viral proteins; now, this virus causes splicing which results in truncated viral proteins whose protective effectiveness is reduced (Dumonceaux J. et al., J. Virol., 2003, 77, 24, 13418-13424).

[0027] In addition, there is still a need for vaccines that only require few injections (one or two at most), so as to facilitate their use, in particular in countries where it is difficult to set up immunization programmes that are followed.

[0028] Surprisingly, the inventors have shown that a recombinant lentiviral vector for expression of at least one immunogenic protein of a virus of the family Flaviviridae effectively makes it possible to induce a strong immune response in the individual (human or animal) immunized, capable in particular of protecting said individual against infection with this virus.

[0029] The recombinant lentiviral vector was able to induce a very early, long-lasting, fully protective immune response against a high dose West Nile virus challenge.

[0030] The inventors provide the first evidence that lentiviral vectors are efficient tools for eliciting a humoral protective response against a pathogen. This broadens the applicability of lentiviral vectors as vaccination tools against pathogens like viruses of the Flaviviridae family, in which a neutralizing humoral response is one active arm of the immune system.

[0031] Consequently, a subject of the present invention is the use of a recombinant lentiviral vector comprising a polynucleotide fragment encoding at least one protein of a virus of the family Flaviviridae or an immunogenic peptide of at least 8 amino acids of said protein, for preparing an immunogenic composition intended for the prevention and/or the treatment of a Flaviviridae infection in a sensitive species.

[0032] Such a vector has a certain number of advantages and is particularly suitable for the needs disclosed above: [0033] it has an increased immunogenic capacity; consequently, it is effective after a single administration in the sensitive species. The effectiveness of this vector is related at once: (i) to its tropism for antigen-presenting cells, or APCs, such as dendritic cells, in particular when it is injected subcutaneously, (ii) to the stable integration, into the cellular genome, of the sequences of interest carried by these vectors, which allows long-lasting expression of the antigen in vivo, in particular in dendritic cells, and (iii) to its ability to stimulate the dendritic cell-dependent immune response. Thus, the duration of expression of the antigen in the dendritic cells, which is greater than that usually obtained with pulsed dendritic cells, advantageously makes it possible to do away with repeated administration of the vector, [0034] it is non-replicative; consequently, it has little or no pathogenic capacity in the sensitive species and no infectious capacity, i.e. no risk of dissemination in the environment, [0035] it is non-tumorigenic; it results in stable integration of the sequence of interest in the genome of the host cell, without causing any tumorigenic effect, [0036] it exhibits no species restriction and has a broadened cellular tropism, in particular due to the fact that it is possible to produce pseudotypes with envelope proteins from other viruses, such as the glycoproteins G of the vesicular stomatitis virus (VSV), of viruses of the family Rhabdoviridae, for instance the rabiesvirus, and of the ebola virus; consequently, it is effective for preventive and/or curative immunization in any sensitive species, and [0037] it makes it possible to do away with the use of adjuvants.

DEFINITIONS

[0037] [0038] polynucleotide fragment: the term "poly-nucleotide fragment or polynucleotide" is intended to mean a DNA or RNA fragment of at least 24 bases or base pairs, preferably from 24 to 5000 bases or base pairs, in particular a cDNA or cDNA fragment. [0039] immunogenic fragment: a peptide fragment capable of inducing a specific humoral and/or cellular response in a species sensitive to infection with a Flaviviridae. [0040] polynucleotide fragment encoding at least one protein of a Flaviviridae or an immunogenic peptide of at least 8 amino acids of said protein: a polynucleotide as defined above, encoding one or more structural or non-structural proteins and/or one or more immunogenic fragments of Flaviviridae. The open reading frame (ORF) of the Flaviviridae polyprotein and the coding sequences of the various Flaviviridae proteins which are included in said ORF are known to those skilled in the art and accessible, either in the databases, in particular that of the NCBI (http://www.ncbi.nlm.nih.gov), or in reference works, for instance Virus Taxonomy. Classification and nomenclature of viruses. Sixth report of the International Committee on taxonomy of viruses (F. A. Murphy et al., Archives of Virology Supplement 10, 1995, Springer Verlag, Vienna, New York). The invention encompasses the coding sequences of any Flaviviridae and the variants derived by mutation (insertion, deletion, substitution) of one or more nucleotides of said coding sequence or by a shift in the open reading frame of one or two nucleotides (ORF+1 and ORF+2), provided that said mutations do not substantially modify the antigenic and/or immunogenic properties of said protein or of said fragment. The invention encompasses in particular the variant polynucleotides derived from the above by mutation (insertion, deletion, substitution) of nucleotides, provided that the modified nucleotide fragments conserve, under high stringency hybridization conditions, an ability to specifically hybridize with the modified polynucleotides from which they derive. [0041] high stringency hybridization conditions: for the purpose of the present invention, the expression "high stringency hybridization conditions" is intended to mean conditions of temperature and of ionic strength that are chosen such that they make it possible to maintain the specific and selective hybridization between complementary polynucleotides. By way of illustration, high stringency conditions for the purposes of defining the polynucleotides above are advantageously as follows: the DNA-DNA or DNA-RNA hybridization is carried out in two steps: (1) prehybridization at 42.degree. C. for 3 hours in phosphate buffer (20 mM, pH 7.5) containing 5.times.SSC (1.times.SSC corresponds to a solution of 0.15 M NaCl+0.015 M sodium citrate), 50% of formamide, 7% of sodium dodecyl sulphate (SDS), 10.times.Denhardt's, 5% of dextran sulphate and 1% of salmon sperm DNA; (2) hybridization for 20 hours at 42.degree. C., followed by 2 washes of 20 minutes at 20.degree. C. in 2.times.SSC+2% SDS, 1 wash of 20 minutes at 20.degree. C. in 0.1.times.SSC+0.1% SDS. The final wash is carried out in 0.1.times.SSC+0.1% SDS for 30 minutes at 60.degree. C. [0042] sensitive species: the expression "species sensitive to infection with a Flaviviridae" is intended to mean both a host species capable of developing a pathology induced by a Flaviviridae, such as a human or non-human mammal, and a reservoir species responsible for propagation of the virus without the development of symptoms, such as in particular a bird or a reptile (crocodile). [0043] recombinant lentiviral vector: the term "recombinant lentiviral vector" is intended to mean both an isolated nucleic acid molecule corresponding to the recombinant genome of a lentiviral vector, in particular included in a plasmid (vector plasmid), and a recombinant lentiviral particle (vector particle) that includes said recombinant genome, produced in a suitable cellular system, optionally pseudotyped with envelope proteins from other viruses such as the glycoproteins G of, the vesicular stomatitis virus (VSV), of the rabiesvirus and of the ebola virus.

[0044] In accordance with the invention, said lenti-viral vector is selected from the group consisting of those derived from: HIV (human immunodeficiency virus), for example HIV-1 or HIV-2, CAEV (caprine arthritis encephalitis virus), EIAV (equine infectious anaemia virus), VMV (visna/maedi virus), SIV (simian immuno-deficiency virus) or FIV (feline immunodeficiency virus). The invention also encompasses the chimeric lentiviruses derived from at least two different lentiviruses. The choice of the lentiviral vector depends in particular on the sensitive species; for example, vectors derived from HIV are advantageously used for human immunization.

[0045] The lentiviral vectors are known to those skilled in the art; they consist of a recombinant nucleotide sequence (recombinant lentiviral genome) comprising: (i) a sequence of interest (coding sequence of Flaviviridae, in the case of the present invention) placed under the control of regulatory signals for transcription and for expression, and (ii) the regulatory sequences of lentiviral origin necessary and sufficient for encapsidation, reverse transcription and viral integration, and, optionally, regulatory sequences for the Rev protein (RRE or rev responsive element). Mention may in particular be made of lentiviral vectors derived from HIV, described by Poznansky et al. (J. Virol., 1991, 65, 532-536) and Naldini et al. (Science, 1996, 272, 263-267) or from FIV, described by Poeschla et al. (Nature Medicine, 1998, 4, 354-357), and also the minimal vectors derived from the above, as described in International Applications WO 99/32646 and WO 98/17815.

[0046] In accordance with the invention, said lenti-viral vectors are vectors capable of expressing the coding sequence(s) as defined above, in a suitable cellular system; said vector comprises an expression cassette that includes the suitable regulatory elements for transcription (promoter, enhancer, Kozak consensus sequence, polyadenylation signal, etc.) under the control of which are inserted the coding sequences as defined above; said coding sequences of interest comprise the signals required for cell transport, for instance a signal for translocation in the endoplasmic reticulum, derived in particular from the ORF preceding said coding sequence in the polyprotein of said Flaviviridae. For example, in the case of the Flaviviruses, when said coding sequence is that of the E protein or of a fragment of said protein, said signal sequence is advantageously derived from the M protein precursor (prM). Advantageously, said expression cassette comprises a strong ubiquitous promoter such as the cytomegalovirus (CMV) early promoter or an enhancer free promoter such as the elongation factor 1.alpha. (EF1.alpha.) or the phosphoglycerate (PGK) promoters.

[0047] In addition, said vector may also comprise a suicide gene such as herpes type 1 thymidine kinase (HSV 1-TK), so as to eliminate the transduced cells by treatment with the appropriate drug, for example acyclovir in the case of HSV 1-TK.

[0048] The invention encompasses simple expression vectors and multiple expression vectors that allow simultaneous expression of several coding sequences from the same promoter or from different promoters, said promoters being located in the same region or else in different regions of said expression vector.

[0049] According to an advantageous embodiment of said use, said recombinant lentiviral vector is of triplex type.

[0050] The vectors of triplex type are in particular described in Zennou et al., Cell, 2000, 101, 173-185 and in International Applications WO 99/55892, WO 01/27304 and WO 01/27300.

[0051] The triplex vectors are characterized in that they comprise a DNA region capable of forming a triplex (or DNA trimer) during viral reverse transcription. This triplex DNA region consists of a cis-active region for central initiation, or polypurine tract (cPPT), and a cis-active region for termination (CTS), said regions making it possible to initiate the transcription of a +strand whose synthesis is initiated by the PTT region present at the centre of the genome of the lentivirus, and to interrupt the transcription of a +strand whose synthesis is initiated at a 3' PPT site upstream of the retroviral LTR. The presence of this triplex DNA region in the lentiviral vectors notably improves the transduction of genes in mitotic or non-mitotic cells, by stimulating the rate of nuclear import of the vector.

[0052] According to another advantageous embodiment of said use, said recombinant lentiviral vector comprises a 3' LTR in which the promoter and the activator have been deleted from the U3 region; this deletion provides additional safety features.

[0053] According to another advantageous embodiment of said use, said recombinant lentiviral vector is pseudotyped with at least one envelope protein of another virus, preferably the vesicular stomatitis virus (VSV) glycoprotein G; the VSV glycoprotein G advantageously makes it possible to obtain high titres of vector particles and to produce vector particles having a broad cellular tropism, capable of transducing in particular antigen-presenting cells such as dendritic cells, in any vertebrate species: humans or animals including horses, fowl, and zoo animals at risk.

[0054] In accordance with the invention, said Flaviviridae is chosen from a Flavivirus, a Pestivirus or a Hepacivirus, as specified above.

[0055] According to yet another advantageous embodiment of said use, said Flaviviridae is selected from the group consisting of the West Nile virus, dengue virus, yellow fever virus and hepatitis C virus.

[0056] In accordance with the invention, said polynucleotide, in particular a cDNA or a cDNA fragment of Flaviviridae encodes: (i) one or more different structural proteins (C, prM, M, E, E1, E2), and/or (ii) one or more different non-structural (NS) proteins, and/or (iii) one or more different immunogenic fragments of said proteins, said proteins or their fragments being derived either from the same Flaviviridae (monovalent vaccine) or from various Flaviviridae and/or from different serotypes or different types of the same Flaviviridae, for preparing polyvalent vaccines.

[0057] Said cDNA can also derive from a coding sequence of a Flaviviridae by a shift in the open reading frame of one or two nucleotides (ribosomal frameshifting). Such cDNAs are known to those skilled in the art, in particular for the C protein of the hepatitis C virus (Xu et al., EMBO, 2001, 20, 3840-3848; Roussel et al., J. Gen. Virol., 2003, 84, 1751-1759; Vassilaki et al., J. Biol. Chem., 2003, 278, 40503-40513; International Application WO 99/63941).

[0058] According to yet another advantageous embodiment of said use, said polynucleotide is a fragment of a coding sequence of Flaviviridae corresponding to the accession number in the NCBI database listed in Table 1:

TABLE-US-00001 TABLE 1 Coding sequences of Flaviviridae NCBI accession FLAVIVIRIDAE number Sequence description Flavivirus M23027 5' cDNA sequence of the poly- protein of the dengue virus type 1 Flavivirus M19197 DNA equivalent of the genome of the dengue virus type 2 Flavivirus M93130 DNA equivalent of the genome of the dengue virus type 3 Flavivirus M14931 DNA equivalent of the genome of the dengue virus type 4 Flavivirus M12294 DNA equivalent of the genome of the West Nile virus Flavivirus AF481864 DNA equivalent of the genome of the IS-98-ST1 strain of the West Nile virus Flavivirus M18370 DNA equivalent of the genome of the Japanese encephalitis virus Flavivirus X03700 cDNA of the polyprotein of the yellow fever virus (vaccination strain 17D) Flavivirus U27495 DNA equivalent of the genome of the Neudoerfl virus of the tick-borne encephalitis virus complex (TBE complex) Flavivirus M73835 cDNA of the structural proteins of the Langat virus (TBE complex) Pestivirus M31182 DNA equivalent of the genome of the BVD virus Pestivirus M31768 DNA equivalent of the genome of the Brescia strain of the CSF virus (hog cholera virus) Pestivirus J04358 DNA equivalent of the genome of the Alfort strain of the CSF virus (hog cholera virus) Hepacivirus M62321 cDNA of the polyprotein of the hepatitis C virus type 1 (HCV-1) Hepacivirus D90208 cDNA of the polyprotein of the hepatitis C virus Hepacivirus M58335 Complete cDNA of the polyprotein of the hepatitis C virus

[0059] The positions of the coding sequences of the various Flaviviridae proteins are indicated in the sequences corresponding to the accession numbers listed in Table 1, which correspond to the cDNAs of the polyprotein or to the DNA equivalents of the Flaviviridae genome.

[0060] According to yet another advantageous embodiment of said use, said polynucleotide fragment is selected from:

[0061] a) the cDNAs encoding an E protein and, optionally, a prM or M protein, and/or a C protein, and/or a non-structural protein of West Nile virus or of dengue virus, and the cDNAs encoding one or more immunogenic peptides of at least 8 amino acids of the above proteins,

[0062] b) the cDNAs encoding an E1 or E2 protein or an E1/E2 heterodimer, and/or a C protein according to a 0, +1 or +2 reading frame, and/or an NS3 protein of hepatitis C virus, and the cDNAs encoding one or more immunogenic peptides of at least 8 amino acids of the above proteins, and

[0063] c) the cDNAs encoding one or more different domains III (positions 295 to 394) of an E protein of dengue virus, each corresponding to one of the four types of dengue virus (types 1 to 4 or DEN-1 to DEN-4), preferably a cDNA encoding the four domains III (DEN-1 to DEN-4), the sequences of which are represented by SEQ ID NOs. 1-4 in the sequence listing attached in the appendix.

[0064] According to an advantageous provision of said use, said cDNA encoding a C protein according to a +1 or +2 reading frame is selected from the group consisting of the sequences SEQ ID NOs. 5 to 14.

[0065] In accordance with the invention, said membrane proteins (prM or M) and/or envelope proteins (E, E1, E2) are expressed by the recombinant lentiviral vector as defined above, either in membrane form, located in the plasma membrane, at the surface of the cells, or in secreted form, i.e. exported from the cell, to the extracellular medium.

[0066] In addition, when the Flavivirus prM and E proteins are expressed simultaneously in the cells transduced by the recombinant vector (in vitro or in vivo), they assemble as viral pseudoparticles (or virus-like particles, VLPs) that are secreted into the extracellular medium. Such particles are particularly immunogenic and induce the production of neutralizing antibodies.

[0067] The cDNA encoding said membrane form comprises the sequence encoding the mature protein, preceded by a sequence encoding a signal peptide for translocation in the endoplasmic reticulum, which sequence includes a translation initiation codon (ATG) at its 5' end. In the case of the Flaviviruses, said signal sequence is advantageously derived from the M protein precursor (prM). The cDNA encoding said secreted form comprises the sequence encoding a truncated mature protein, from which the membrane anchoring region has been deleted and which is preceded by a signal peptide as defined above.

[0068] For example, in the case of the West Nile virus: [0069] the mature E protein corresponds to positions 291 to 791 of the polyprotein sequence, with reference to the Genbank sequence AAL87234'; the corresponding nucleotide sequence is located from positions 967 to 2469 in the sequence of the genome of the West Nile virus, with reference to the Genbank sequence AF481864; [0070] a truncated mature E protein from which the membrane anchoring region has been deleted corresponds in particular to positions 291 to 732 of the sequence of the polyprotein of the West Nile virus, with reference to the Genbank sequence AAL87234; the corresponding nucleotide sequence is located from positions 967 to 2292 in the sequence of the genome of the West Nile virus, with reference to the Genbank sequence AF481864; [0071] the internal signal peptide derived from the M protein precursor corresponds to positions 275 to 290 of the sequence of the polyprotein, with reference to the Genbank sequence AAL87234; the corresponding nucleotide sequence is located from positions 919 to 966 in the sequence of the genome of West Nile virus, with reference to the Genbank sequence AF481864.

[0072] Thus, the cDNAs encoding the membrane form of the E protein, the secreted form of the E protein and the prM and E proteins of the West Nile virus correspond, respectively, to positions 919 to 2469, 919 to 2292 and 399 to 2469 in the sequence of the genome of said virus as defined above.

[0073] A subject of the present invention is also a recombinant lentiviral vector comprising a polynucleotide fragment encoding at least one structural protein of a Flaviviridae or an immunogenic peptide of at least 8 amino acids of said protein; in addition, as specified above in the context of the use of such vectors, said vector advantageously also comprises a cDNA encoding one or more non-structural proteins and/or one or more immunogenic fragments of said proteins. Said polynucleotide fragment is in particular selected from the sequences as defined above. Advantageously, said recombinant lentiviral vector is a vector of triplex type. In addition, said recombinant lentiviral vector can advantageously comprise a 3' LTR in which the promoter and the activator has been deleted from the U3 region. It is preferably a vector that is pseudotyped with at least one envelope protein of another virus, preferably the vesicular stomatitis virus (VSV) glycoprotein G.

[0074] According to an advantageous embodiment of said vector, it comprises the cDNA encoding at least one E protein and, optionally, a prM or M protein, and/or a C protein, and/or a non-structural protein of West Nile virus or of dengue virus, or the cDNA encoding one or more immunogenic peptides of at least 8 amino acids of the above proteins.

[0075] According to another advantageous embodiment of said vector, it comprises the cDNA encoding an E1 or E2 protein or an E1/E2 heterodimer, and/or a C protein according to a 0, +1 or +2 reading frame and, optionally, an NS3 protein of hepatitis C virus, or the cDNA encoding one or more immunogenic peptides of at least 8 amino acids of the above proteins.

[0076] According to an advantageous provision of said vector, said cDNA encoding a C protein according to a +1 or +2 reading frame is selected from the group consisting of the sequences SEQ ID NOs. 5 to 14.

[0077] According to yet another advantageous embodiment of said vector, it comprises the cDNA encoding a domain III (positions 295 to 394) or several different domains III of an E protein of dengue virus, each corresponding to one of the four types of dengue virus (types 1 to 4 or DEN-1 to DEN-4), preferably it comprises a cDNA encoding the four domains III (DEN-1 to DEN-4) the sequences of which are represented by SEQ ID NOs. 1-4 in the sequence listing attached in the appendix.

[0078] According to yet another advantageous embodiment of said vector, it is a vector plasmid called pTRIP.DELTA.U3.CMV-sE (WNV), comprising the cDNA encoding a secreted form of the E protein of the IS-98-ST1 strain of West Nile virus, which vector is included in a microorganism deposited under the No. I-3076, on 27 Aug. 2003, with the Collection Nationale de Cultures de Microorganismes [National Collection of Cultures of Microorganisms], 25 rue du Docteur Roux, 75724 Paris Cedex 15.

[0079] The invention encompasses the vector plasmids as defined above and the vector particles derived from the above vector particles, in particular the vector particles pseudotyped with at least one envelope protein of another virus, such as in particular the vesicular stomatitis virus (VSV) glycoprotein G.

[0080] The recombinant lentiviral vectors as defined above are prepared by conventional methods, that are known in themselves, and according to standard protocols such as those described in Current Protocols in Molecular Biology (Frederick M. AUSUBEL, 2000, Wiley and son Inc., Library of Congress, USA).

[0081] More precisely, the polynucleotide fragments can be obtained either by amplification of a matrix consisting of a genomic RNA or an mRNA of a Flaviviridae or else a cDNA or a DNA fragment derived from the above, by PCR or RT-PCR using primers specific for the genome of a virus of the family Flaviviridae, or by digestion of the Flaviviridae cDNA using a restriction enzyme, or alternatively by total or partial chemical synthesis.

[0082] The polynucleotide fragment thus obtained is cloned into a vector plasmid containing the lentiviral vector genome, so as to produce a recombinant vector plasmid.

[0083] The particles of the recombinant lentiviral vector (vector particles) are produced by cotransfection of cells with the recombinant vector plasmid as defined above, an encapsidation plasmid that provides, in trans, the structural proteins and the enzymes of the viral particle and, optionally, a plasmid for expression of the envelope glycoprotein of a virus such as VSV, for the production of pseudotyped particles.

[0084] A subject of the present invention is also an immunogenic composition, characterized in that it comprises at least one recombinant vector as defined above.

[0085] According to an advantageous embodiment of said composition, it comprises a pharmaceutically acceptable vehicle and, optionally, a carrier substance.

[0086] The pharmaceutically acceptable vehicles and the carrier substances are those conventionally used.

[0087] The carrier substances are advantageously selected from the group consisting of unilamellar liposomes, multilamellar liposomes, saponin micelles or solid microspheres of a saccharide or auriferous nature.

[0088] According to another advantageous embodiment of said composition, it comprises particles of said recombinant lentiviral vector (vector particles), preferably pseudotyped with an envelope protein of another virus, preferably with the vesicular stomatitis virus glycoprotein G.

[0089] According to yet another advantageous embodiment of said composition, it comprises a recombinant lentiviral vector of triplex type as defined above.

[0090] According to an advantageous provision of said composition, it comprises an isolated nucleic acid molecule corresponding to the recombinant genome of said recombinant lentiviral vector of triplex type, which nucleic acid molecule comprises: (i) the regulatory sequences for encapsidation, reverse transcription and integration and the cis-active sequences for central initiation (or polypurine tract cPPT) and termination (CTS) of lentiviral origin and, optionally, the regulatory sequences for the Rev protein (RRE or Rev Responsive Element) and (ii) a polynucleotide fragment encoding a Flaviviridae protein or an immunogenic peptide of at least 8 amino acids of said protein as defined above.

[0091] In accordance with the invention, said vector of triplex type comprises an expression cassette that includes the suitable regulatory elements for transcription (promoter, enhancer, Kozak consensus sequence, polyadenylation signal, etc.) under the control of which are inserted the coding sequences as defined above, and said coding sequences of interest optionally comprise the signals required for cellular transport, as defined above.

[0092] The immunogenic or vaccine compositions according to the invention can be administered generally (orally, intramuscularly, subcutaneously, intraperitonealy or intravenously), locally (nasally, other mucosal routes) or by a combination of these routes, in a sensitive species as defined above (human or non-human mammalian host, or reservoir (birds, reptiles)).

[0093] Preferably, they are administered subcutaneously in order to target antigen-presenting cells such as dendritic cells, so as to obtain prolonged expression of the antigen in these cells.

[0094] Alternatively, the immunogenic or vaccine compositions according to the invention are used to modify autologous cells of a host species, in particular antigen-presenting cells such as dendritic cells. The modified cells are then re-administered to the host; such a use is particularly advantageous for the treatment of an infection with a Flaviviridae in a human or non-human host mammal.

[0095] The dose of vector varies according to the route of administration, and also according to the nature and the weight of the species to be treated (human or animal).

[0096] A subject of the present invention is also cells modified with a recombinant vector as defined above. Preferably, said cells are eukaryotic cells that are stably modified with said recombinant vector; such cells that stably express at least one protein or one antigenic peptide of Flaviviridae are useful: [0097] for producing particles of said recombinant lentiviral vector (vector particles),

[0098] for producing recombinant viral proteins of Flaviviridae, immunogenic fragments of said proteins, and viral pseudoparticles of the type of those of Flaviviridae (VLPs or virus-like particles), derived from the envelope proteins and/or membrane proteins of Flaviviridae, in particular from the Flavivirus prM and E proteins; the pseudoparticles are advantageously used as a reagent for diagnosing a Flaviviridae infection by immunocapture of the specific immunoglobulins present in the biological fluids of infected individuals, [0099] for screening antiviral compounds, and [0100] as a diagnostic reagent.

[0101] In accordance with the invention, it is possible to produce viral proteins of Flaviviridae and/or immunogenic fragments of said proteins or else viral pseudoparticles, in accordance with the following steps:

[0102] a) culturing modified cells as defined above, under conditions which allow the expression of one or more viral proteins of Flaviviridae and/or one or more of the immunogenic fragments of said proteins encoded by said recombinant lentiviral vector, and

[0103] b) separating said proteins, protein fragments or pseudoparticles from the culture supernatant or from said cells in a), by any suitable means.

[0104] In accordance with this method, the purification of viral protein(s) or of fragment(s) can be carried out, from a culture supernatant, or from lysates of the cells modified with a recombinant vector as defined above, by conventional techniques such as: [0105] affinity chromatography: a tag, such as a nucleotide sequence encoding a polyhistidine tail is then introduced into the vector, and the protein is purified on nickel-gel (agarose or the like) columns; [0106] immunoaffinity chromatography: the viral sequence of interest is fused, at the C-terminal or at the N-terminal, with a nucleotide sequence encoding a peptide epitope, also comprising a site for cleavage by an enzyme, such as thrombin, in order to subsequently separate the epitope sequence from the protein; useful epitopes are, for example, the C9 epitope (TETSQVAPA) (Mirzabekov T. et al., J. Biol. Chem., 1999, 274, 28745-28750) or the myc epitope. The expressed protein is purified on an affinity column to which an antibody specific for said epitope has been attached (1D14 for the C9 epitope or 9.sup.E10 for the myc epitope) and the protein of interest is separated by means of cleavage with thrombin; [0107] precipitation with a precipitating agent such as polyethylene glycol, and then centrifugation in order to recover the protein in the pellet.

[0108] In accordance with this method, the purification of the particles of the type of those of Flaviviridae is carried out, from a culture supernatant from cells modified with a recombinant vector as defined above, by conventional techniques such as: [0109] precipitation with a precipitating agent such as polyethylene glycol, and then centrifugation in order to recover the pseudoparticles in the pellet, and [0110] continuous or discontinuous gradient centrifugation, in particular on a sucrose gradient.

[0111] A subject of the present invention is also a method for screening antiviral compounds, characterized in that it comprises: [0112] bringing eukaryotic cells modified with a recombinant vector as defined above, and in particular with a vector comprising a cDNA encoding a non-structural protein of Flaviviridae such as NS3 (helicase or protease) or NS5 (polymerase), into contact with various compounds of a library to be tested, and [0113] measuring, by any suitable means, the activity (helicase, protease, polymerase) of said protein, in the presence or in the absence of said compounds, and [0114] selecting the compounds capable of modulating (activating or inhibiting) said activity.

[0115] This activity is evaluated by conventional methods known to those skilled in the art, such as those described in particular in Borowski et al., Acta Biochimica Polonica, 2002, 49, 597-614; Steffens et al., J. Gen. Virol., 1999, 80, 2583-2590; Ryan et al., J. Gen. Virol., 1998, 79, 947-959; Bretner et al., Antivir. Chem. Chemother., 2004, 15, 35-42.

[0116] Preferably, the screenings are carried out on specific target tissues, and in particular on dendritic cells, neuronal cells or hepatocytes.

[0117] A subject of the present invention is also a method for diagnosing infection with a Flaviviridae, using a sample of biological fluid from an individual of a sensitive species, characterized in that it comprises at least the following steps:

[0118] a) bringing said biological sample into contact with modified eukaryotic cells expressing at least one Flaviviridae antigen (C, E, E1, E2, prM, M, NS (in particular NS1)) as defined above, optionally permeabilized,

[0119] b) revealing, by any suitable means, the antigen-antibody complexes formed in (a), for example by EIA, ELISA or RIA, or by immunofluorescence.

[0120] A subject of the present invention is also a method for diagnosing infection with a Flaviviridae using a sample of biological fluid from an individual of a sensitive species, characterized in that it comprises at least the following steps:

[0121] a) bringing said biological sample into contact with viral pseudoparticles produced from the culture supernatant of cells modified with a lentiviral vector expressing at least one membrane protein and/or envelope protein as defined above, and

[0122] b) revealing, by any suitable means, the antigen-antibody complexes formed in (a), for example by EIA, ELISA or RIA, or by immunofluorescence.

[0123] A subject of the present invention is also a kit for carrying out the methods as defined above, characterized in that it comprises at least modified cells as defined above.

[0124] A subject of the present invention is also a method of immunization against a Flaviviridae, characterized in that it comprises a single administration of a recombinant vector as defined above, preferably subcutaneously.

[0125] Besides the above provisions, the invention also comprises other provisions, which will emerge from the following description that refers to examples of preparation of the recombinant vector according to the present invention and of use of said vector for immunization, and derived modified cells for the production of proteins, and also to the attached drawings in which:

[0126] FIG. 1 is a diagrammatic representation of the vector plasmid pTRIP.DELTA.U3CMV-sE(WNV) corresponding to the sequence SEQ ID NO. 15, containing the cDNA (SEQ ID NO. 16) encoding the truncated E protein (E1-411) of the West Nile virus (SEQ ID NO. 17).

[0127] FIG. 2 illustrates the analysis by ELISA and by means of a neutralization assay, of the sera from mice immunized with a single intraperitoneal injection of 1 .mu.g of TRIP.DELTA.U3CMV-sE(WNV) vector particles.

[0128] FIG. 3 represents the immunoprecipitation of the lysates of VERO cells infected with West Nile virus, with the sera from the mice immunized with 1 .mu.g of TRIP.DELTA.U3CMV-sE(WNV) vector particles, by comparison with control sera.

Lanes 1 to 10: lysates of VERO cells infected with West Nile virus were precipitated with the following sera:

[0129] lane 1: serum at D14 post-immunization with the TRIP.DELTA.U3CMV-GFP vector,

[0130] lane 2: serum at D23 post-immunization with the TRIP.DELTA.U3CMV-GFP vector,

[0131] lane 3: polyclonal anti-West Nile virus (strain IS-98-ST1) ascites,

[0132] lane 4: non-immune serum,

[0133] lane 5: serum at D14 post-immunization with the TRIP.DELTA.U3CMV-sE(WNV) vector,

[0134] lane 6: serum at D23 post-immunization with the TRIP.DELTA.U3CMV-sE(WNV) vector,

[0135] lane 7: serum at D22 post-challenge (10 LD.sub.50 of the IS-98-ST1 strain) from the mice immunized for 14 days with the TRIP.DELTA.U3CMV-sE(WNV) vector,

[0136] lane 8: serum at D30 post-challenge (10 LD.sub.50 of the IS-98-ST1 strain) from the mice immunized for 14 days with the TRIP.DELTA.U3CMV-sE(WNV) vector,

[0137] lane 9: serum at D22 post-challenge (100 LD.sub.50 of the IS-98-ST1 strain) from the mice immunized for 30 days with the TRIP.DELTA.U3CMV-sE(WNV) vector,

[0138] lane 10: serum from mice immunized with the lymphocytic choriomeningitis virus.

Lanes 11 and 12: lysates of non-infected VERO cells were precipitated with the following sera:

[0139] lane 11: polyclonal anti-West Nile virus (IS-98-ST1 strain) ascites,

[0140] lane 12: serum at D22 post-challenge (100 LD.sub.50 of the IS-98-ST1 strain) from the mice immunized for 30 days with the TRIP.DELTA.U3CMV-sE(WNV) vector.

[0141] FIG. 4 represents the survival curve for the mice immunized intraperitoneally and then challenged by the same route, either 2 weeks after immunization, with 10 LD.sub.50 of the IS-98-ST1 strain (A), or 4 weeks after immunization, with 100 LD.sub.50 of the IS-98-ST1 strain (B). .cndot.: control mice inoculated with DPBS. .tangle-solidup.: control mice immunized with 1 .mu.g of TRIP.DELTA.U3CMV-EGFP vector particles. .box-solid.: mice immunized with 1 .mu.g of TRIP.DELTA.U3CMV-sE(WNV) vector particles.

[0142] FIG. 5 illustrates the purification of viral pseudoparticles from the supernatant of eukaryotic cells transduced with a recombinant lentiviral vector expressing the prM and E proteins of the West Nile virus.

[0143] FIG. 6 illustrates the detection of anti-WNV-sE antibodies in sera from TRIP.DELTA.U3.CMV-sE(WNV) vaccinated 129 mice. Radio-labeled cell lysates from WNV infected Vero cells were immunoprecipitated with pooled immune sera from lentiviral vector vaccinated 129 mice. (A) Pre-WNV challenge sera. (B) Post-challenge sera. HMAF=Hyperimmune Mouse Ascitic Fluid. Control sera=non immune sera. Antisera to MV=antisera to Measle Virus. TRIP/WNsE=TRIP.DELTA.U3.CMVsE(WNV). TRIP/GFP=TRIP.DELTA.U3.CMV-GFP.

[0144] FIG. 7 illustrates the analysis by flow cytometry of the effect of heat treatment on recombinant lentiviral vector transduction efficiency. 293T cells were incubated with TRIP.DELTA.U3.CMV-GFP vector particles which have been heat-inactivated for 10 min at 70.degree. C. (heated TRIP/GFP), or not inactivated (TRIP/GFP). Non-infected 293T cells (Mock) were used as control. At 48 h, the GFP fluorescence intensity was measured; the percentage of GFP positive cells is indicated.

EXAMPLE 1

Preparation of the TRIP.DELTA.U3.CMV-sE(WNV) Recombinant Vector

[0145] 1) Construction of the pTRIP.DELTA.U3.CMV-sE(WNV) Vector Plasmid

[0146] A cDNA representing the nucleotide sequence from positions 967 to 2292 of the genome of the IS-98-ST1 strain of the West Nile virus (application FR 01 04599 and Genbank AF481864), corresponding to the amino acids from positions 291 to 732 of the polyprotein (application FR 01 04599 and Genbank AAL87234), was amplified by polymerase chain reaction (PCR) using the sense primer: 5'-TATCGTACGATGAGAGTTGTGTTTGTCGTGCTA-3' (SEQ ID NO. 18), containing the BsiW I site in the underlined form) and the antisense primer: 5'-ATAGCGCGCTTAGACAGCCCTTCCCAACTGA-3' (SEQ ID NO. 19), containing the BssH II site in the underlined form. This cDNA, corresponding to the sequence SEQ ID NO. 16 in the sequence listing attached in the appendix, is bordered, in the 5' position, by a BsiW I site and, in the 3' position, by a BssH II site. The sequence SEQ ID. NO. 16 contains, successively from 5' to 3': an ATG, the sequence encoding the signal peptide derived from the M protein precursor (prM 151-166) and the sequence encoding a truncated E protein (E 1-441), from which the membrane anchoring region has been deleted. It encodes an E protein which is secreted into the extracellular medium (sE protein); the signal peptide derived from the prM protein is used for translocation of the E protein in the endoplasmic reticulum and for its transport, in secretion vesicles, to the plasma membrane, where it is released into the extracellular medium.

[0147] The lentiviral vector plasmid pTRIP.DELTA.U3.CMV-EGFP (application WO 01/27302) was digested so as to excise the EGFP gene, and then the linearized plasmid was ligated with a linker containing the BsiW I and BssH II sites, so as to give the plasmid called pTRIP.DELTA.U3.CMV-BsiW I-BssH II. The 1.4 kb BsiW I-BssH II fragment of the cDNA obtained above, that includes the sE protein construct, was cloned into the same sites of the plasmid pTRIP.DELTA.U3.CMV-BsiW I-BssH II, to give the recombinant lentiviral vector plasmid called pTRIP.DELTA.U3.CMV-sE (WNV) or pTRIP.DELTA.U3.CMV-sE (WNV) (FIG. 1 and SEQ ID NO. 15). A culture of E. coli bacteria transformed with the pTRIP.DELTA.U3.CMV-sE (WNV) vector plasmid was deposited, under the No. I-3076, on 27 Aug. 2003, with the Collection Nationale de Cultures de Microorganismes [National Collection of Cultures of Microorganisms], 25 rue du Docteur Roux, 75724 Paris Cedex 15.

[0148] The conformity of the pTRIP.DELTA.U3.CMV-sE (WNV) recombinant vector plasmid was verified by enzyme restriction and by sequencing of the insert corresponding to the sE protein construct.

[0149] The sequence of the 1.4 kb BsiW I-BssH II insert corresponds to the nucleotide sequence SEQ ID NO. 16 in the sequence listing attached in the appendix; it encodes a secreted E protein, called sE, corresponding to the amino acid sequence SEQ ID NO. 17 in the sequence listing attached in the appendix.

2) Preparation of Viral Particles of the TRIP.DELTA.U3.CMV-sE (WNV) Vector, Pseudotyped with the Vesicular Stomatitis Virus Envelope Glycoprotein (VSV-G)

[0150] Human fibroblastic 293T cells (ATCC) are grown in Dulbecco's modified Eagle medium (DMEM) Glutamax (GIBCO) supplemented with 10% Fetal Calf Serum (FCS). The viral particles of the TRIP.DELTA.U3.CMV-sE (WNV) vector, pseudotyped with the vesicular stomatitis virus envelope glycoprotein (VSV-G), also called TRIP.DELTA.U3.CMV-sE (WNV) vector particles, are produced by calcium phosphate cotransfection of the 293T cell line with the pTRIP.DELTA.U3.CMV-sE (WNV) vector plasmid as defined above, an encapsidation plasmid that provides, in trans, the structural proteins and the enzymes of the viral particle (pCMV.DELTA.R8.2: Naldini et al., Science, 1996, 272, 263-267; pCMV.DELTA.R8.91 or p8.7: Zufferey et al., Nat. Biotechnol., 1997, 15, 871-877) and a plasmid for expression of the VSV virus envelope glycoprotein (pHCMV-G: Yee et al., P.N.A.S., 1994, 91, 9564-9568), as described in Zennou et al., Cell., 2000, 101, 173-185).

3) Expression of the Secreted Version of the E Glycoprotein of WNV (WNV-sE) by the Recombinant TRIP.DELTA.U3.CMV-sE (WNV) Vector

[0151] Expression of WNV-sE in lentiviral vector transduced 293T cells was examined by indirect immunofluorescence. Briefly, human 293T cells cultured on 8-chamber Glass-Labteks (NUNC) were transduced with TRIP.DELTA.U3.CMV-sE (WNV) vector. After 48 h, cells were fixed with 3% paraformaldehyde (PFA) in PBS for 20 min and permeabilized with 0.1% Triton X-100 in PBS for 4 min. Cells were incubated with anti-WNV HMAF at a 1:100 dilution in PBS for 1 h. After blocking with 0.2% BSA in PBS, cells were further incubated with a Cy3-conjugated anti-mouse IgG antibody (AMERSHAM PHARMACIA) at a 1:500 dilution in PBS 0.2% BSA. Cell nuclei were visualized with DAPI. The slides were examined using a Zeiss Axioplan microscope with ApoTome system.

[0152] At 48 h post-transduction, a high fraction of cells were immunostained. Immunostaining pattern suggests that WNV-sE migrated through the secretory pathway.

4) Titration of the Recombinant TRIP.DELTA.U3.CMV-sE (WNV) Vector

4.1) Material and Methods

a) p24 Antigen Titration by ELISA

[0153] Quantification of p24 antigen content of concentrated vector particles was done with a commercial HIV-1 p24 ELISA kit (PERKIN ELMER LIFESCIENCES).

b) Quantitative PCR

[0154] Primers and probes were synthesized by PROLIGO. For detection of the U5-R sequences in the lentiviral vector, primers and probes used (Brussel A and Sonigo P, J. Virol., 2003, 77, 10119-10124), were as follows (SEQ ID NO: 20 to 27):

TABLE-US-00002 probes (3'fluorescein(PITC) or phosphorylated (P)) LTR-FL: 5'-CACAACAGACGGGCACACACTACTTGA-FITC-3' LTR-LC: 5'-RED640-CACTCAAGGCAAGCTTTATTGAGGC-P-3' primers AA55M: 5'-GCTAGAGATTTTCCACACTGACTAA-3' M667: 5'-GGCTAACTAGGGAACCCACTG-3'. For detection of CD3, the sequences of primers and probes were as follows: probes CD3-P1: 5'-GGCTGAAGGTTAGGGATACCAATATTCCTGTCTC- FITC-3', CD3-P2: 5'RED705-CTAGTGATGGGCTCTTCCCTTGAGCCCTTC- P-3' primers CD3-in-F: 5'-GGCTATCATTCTTCTTCAAGGTA-3' CD3-in-R: 5'-CCTCTCTTCAGCCATTTAAGTA-3'.

[0155] Genomic DNA from approximately 3.10.sup.6 lentiviral vector transduced 293T cells was isolated 48 h after transduction using QIAamp.RTM. DNA Blood Mini Kit (QIAGEN). For real-time PCR analysis, 5 .mu.L of DNA were mixed with 15 .mu.L of a PCR master mix consisting of 1.times. Jumpstart.TM. Taq ReadyMix.TM. (SIGMA), 1.9 mM MgCl.sub.2, 1.5 .mu.M of forward and reverse primers (AA55M/M667 or CD3-in-F/CD3-in-R), 200 nM of the probes (LTR-FL/LTR-LC or CD3-P1/CD3-P2) and, 1.5 units of Taq DNA Polymerase (Invitrogen). Amplifications were performed using one cycle of 95.degree. C. for 3 min, and 40 cycles of 95.degree. C. for 5 s, 55.degree. C. for 15 s and 72.degree. C. for 10 s. To take into account the possible plasmid contamination of vector stocks, DNA from 293T cells transduced with heat-inactivated (10 min at 70.degree. C.) vector was always tested in parallel. For negative controls 5 .mu.L of genomic DNA from untransduced cells was used. Each DNA sample was tested in duplicate and the mean values are reported. Ten-fold serial dilutions of known concentration of the plasmid pTripCD3, containing the relevant sequences U5-R and CD3, were amplified in parallel with DNA samples to generate a standard curve.

[0156] The total number of vector copies per cell was calculated by normalizing the number of U5-R copies to the number of 293T cells, as quantified by the copy number of CD3 molecules on the same genomic DNA sample, and then subtracting the number of copies obtained for the heat-inactivated vector-transduced cells.

4.2) Results

[0157] The number of physical particles of the vector stock used in this study was first evaluated using a commercially available ELISA assay against the p24 HIV-1 capsid protein. The determined concentration was 58 ng of p24 per microliter.

[0158] The vector stock actual titer was calculated on the basis of the transfer of vector DNA to the target cell, using a quantitative PCR assay. The quantification of both a vector specific sequence (U5) and a cellular locus (CD3) gives the average DNA vector copy number per cell. This allows the calculation, after transduction with a defined concentration of vector particles, of the titer of the vector preparation. The TRIP.DELTA.U3.CMV-sE vector stock used in this study was titrated in human 293T cells at 5.2.times.10.sup.7 transduction units (TU) per ml. In other words, 1 ng of p24 antigen from this TRIP.DELTA.U3.CMV-sE vector preparation can transduce 900 human 293T cells.

[0159] For simplicity reasons, in the following sections, the quantity of vector particles used will be expressed as ng of p24 antigen.

EXAMPLE 2

Analysis of the Immunogenic Capacity of the TRIP.DELTA.U3.CMV-sE Vector in BALB/c Mice

1) Materials and Methods

1.1) Immunization/Vaccination Protocol

[0160] Six-week-old BALB/c mice (2 groups of 6 mice; Janvier breeding colony) were inoculated intraperitoneally with 0.1 ml of Dulbecco's PBS (DPBS) containing 1 .mu.g of TRIP.DELTA.U3.CMV-sE vector particles prepared as described in Example 1. The animals were given a single vaccine injection.

[0161] The control groups were inoculated, under the same conditions, either with 1 .mu.g of TRIP.DELTA.U3.CMV-GFP vector particles prepared in a similar manner to the TRIP.DELTA.U3.CMV-sE (WNV) vector particles (2 groups of 3 mice), or DPBS buffer alone (2 groups of 3 mice).

[0162] The mouse sera were taken 14 days (D.sub.14) and 23 days (D.sub.23) after the vaccine injection and heat-inactivated for 30 min at 56.degree. C. before measurement of the antibody response.

1.2) West Nile Virus Strain, Purification and Titering

[0163] The West Nile virus strain used is the IS-98-ST1 strain, described in application FR 01 04599; it is produced on Aedes mosquito cells (AP61 line) and purified according to the protocol described by Despres et al., Virol., 1993, 196, 209-219. More precisely, AP61 cells are infected, at a multiplicity of infection of 0.4, with the IS-98-ST1 strain of the West Nile virus. Three days after infection, the viral particles present in the culture supernatant are precipitated with PEG 6000 (7%), and then purified on a discontinuous 30-60% sucrose gradient and on a linear 10-50% sucrose gradient. The virions thus obtained are conserved at -80.degree. C. in sucrose (30%).

[0164] The West Nile virus is titered by means of a Focus ImmunoAssay (FIA) on AP61 cells, and the infectious titre is expressed as focus-forming units (FFU.sub.AP61/ml), according to the protocol described by Despres et al., mentioned above.

[0165] The infectious titres of the purified viral preparations are approximately 10.sup.10 FFU.sub.AP61/ml.

1.3) Anti-WNV Hyperimmune Ascitic Fluid

[0166] 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 WNV-resistant BALB/c-MBT congenic mice with 10.sup.3 FFU of IS-98-ST1 as described previously (Mashimo et al., PNAS, 2002, 99, 11311-11316). The WNV-immune serum was collected one month after priming.

1.4) ELISA

[0167] The anti-E total antibodies titres are measured by ELISA according to the protocol described in Mashimo et al., PNAS, 2002, 99, 11311-11316, using, as antigen, WN IS-98-ST1 virions purified on a sucrose gradient as described in paragraph 1.2 (10.sup.6 FFU.sub.AP61 per 96-well microplate). Peroxidase-conjugated anti-mouse immunoglobulin (H+ L) (JACKSON IMMUNO RESEARCH) at a 1:4000 dilution, peroxidase-conjugated anti-mouse IgM (.mu.-chain specific) (SIGMA) at a 1:20,000 or peroxidase-conjugated anti-mouse IgG (.gamma.-chain specific) (Sigma) at a 1:20,000 dilution were used as secondary antibodies. The titres are determined by means of the final dilution of serum that corresponds to the optical density (OD) value which is at least twice that of the serum from the control animals, as defined above. The anti-E IgG and IgM antibodies are also measured using an already described isotype specific ELISA (Despres P et. al., J. Infect. Dis., 2005, 191, 207-214).

1.5) Immunoprecipitation (RIP Assay)

[0168] The experimental protocol is as described in Despres et al. (J. Virol., 1995, 69, 7345-7348). More precisely, VERO cells are infected with the IS-98-ST1 strain of the West Nile virus, at the multiplicity of infection of 5 FFU.sub.AP61/cell. Twenty hours after infection, the cell proteins are labelled with Tran.sup.35Slabel (ICN; 100 .mu.Ci/ml) for 3 hours. After three washes with cold PBS, cells are lysed in RIPA buffer (50 mM Tris-Cl, pH 8.0, 150 mM NaCl, 10 mM EDTA, 0.1% SDS, 0.5% sodium deoxycholate, 1% Triton X-100, supplemented with 25 .mu.g/ml aprotinin (SIGMA) for 10 min at +4.degree. C. The cell lysates are then clarified by centrifugation at 10,000 rpm for 5 min at +4.degree. C. The lysates are incubated with the sera to be tested at the final dilution of 1:100, in the presence of Protein A Sepharose. The immunoprecipitates are then analysed in an SDS-15% PAGE gel, under non-reducing conditions, and revealed by autoradiography.

1.6) Neutralization Test

[0169] The neutralizing activity of the sera from the immunized mice, with respect to the IS-98-ST1 strain of the West Nile virus, was measured by the reduction in viral replication foci on VERO cells (ATCC). More precisely, serial dilutions of the sera inactivated for 30 minutes at 56.degree. C. (0.1 ml) are incubated in the presence of an inoculum of the IS-98-ST1 strain of the West Nile virus (100 FFU.sub.AP61 in 0.1 ml). VERO cells (1.5.times.10.sup.5 cells per well of a 12-well plate) are then infected with the mixture for two hours at 37.degree. C., and the viral replication foci are counted two days after infection. The neutralizing antibody titre of the sera, called TNRF90 (Test for Neutralization by 90% Reduction in viral replication Foci), is determined by virtue of the final dilution of the serum that neutralizes at least 90 of the 100 FFUs of viruses inoculated in each well.

2) Results

[0170] 2.1) Analysis by ELISA of the Reactivity of the Sera from the Immunized Animals, with Respect to West Nile Virus

[0171] The production of antibodies directed against the E protein of West Nile virus was verified by ELISA assay carried out of the mouse sera taken 14 and 23 days after the injection of TRIP.DELTA.U3.CMV-sE (WNV) vector particles, using purified West Nile virus as antigen.

[0172] The results given in FIG. 2 show that the specific antibody titre of the sera from the mice immunized with the TRIP.DELTA.U3.CMV-sE (WNV) vector particles is 1/10 000 and 1/20 000, respectively, 14 days and 23 days after the vaccine injection.

2.2) Analysis by Immunoprecipitation of the Specificity of the Sera from the Immunized Animals

[0173] The specificity of the sera from the animals immunized with the TRIP.DELTA.U3.CMV-sE vector was verified by immunoprecipitation. The sera from the mice immunized with the TRIP.DELTA.U3.CMV-sE vector react with the envelope protein E of West Nile virus; the reactivity is stronger at D.sub.23 than at D.sub.14 after the vaccine injection (FIG. 3).

2.3) Analysis of the Neutralizing Activity of the Sera from the Immunized Animals, with Respect to West Nile Virus

[0174] The neutralizing activity of the sera from the mice immunized with a single injection of TRIP.DELTA.U3.CMV-sE (WNV) vector particles, with respect to West Nile virus, was verified experimentally by measuring the reduction in viral replication foci (TNRF90) on VERO cells. The titres at D.sub.14 and D.sub.23 after vaccine injection are, respectively, 10 and 20 (FIG. 2).

EXAMPLE 3

Analysis of the Protective Capacity of the TRIP.DELTA.U3.CMV-sE (WNV) Vector in BALB/c Mice

[0175] The protective role of the anti-E protein, antibodies produced after immunization of mice with TRIP.DELTA.U3.CMV-sE (WNV) vector particles was tested in the murine model of WNV-associated encephalitis (Deubel et al., Ann. N.Y. Acad. Sci., 2001, 951, 195-206; Mashimo et al., 2002, precited; International Application WO 02/081511; Ceccaldi et al., FEMS Microbiol. Lett., 2004, 233, 1-6). Thus, mice were challenged by intraperitoneal inoculation of 10 LD.sub.50 (dose that is lethal in 50% of the mice) or 100 LD.sub.50 of the highly neuroinvasive and neurovirulent IS-98-ST1 strain of West Nile virus.

[0176] More precisely, two challenge protocols were used: (i) the first group of 6 mice immunized as described in Example 2 received 10 LD.sub.50 of the IS-98-ST1 strain, 15 days after the vaccine injection (D15); (ii) the second group of 6 mice immunized as described in Example 2 received 100 LD.sub.50 of the IS-98-ST1 strain, 30 days after the vaccine injection (D30). The challenge virus is diluted in DPBS (pH 7.5), supplemented with 0.2% of bovine serum albumin (Sigma); 1 LD.sub.50 corresponds to 10 FFU.sub.AP61/ml.

[0177] The survival curve for the first group of mice (FIG. 4A) shows that all the control mice, inoculated with DPBS or with the TRIP.DELTA.U3.CMV-EGFP vector, die 13 days after the inoculation of the viral challenge dose.

[0178] On the other hand, the 6 mice immunized with the TRIP.DELTA.U3.CMV-sE (WNV) vector are resistant to the lethal dose and showed no morbidity.

[0179] Twenty-two days after the challenge, the resistant mice have anti-West Nile virus antibody titres (1.7.+-.0.1, dilution 1:10.sup.4), by ELISA, that are greater than those obtained before the challenge. The sera from the challenged mice react strongly with the E protein of West Nile virus (FIG. 3) and the neutralizing antibodies have a titre of 100, 1 month after the challenge.

[0180] The survival curve for the second group of mice (FIG. 4B) shows that all the control mice, inoculated with DPBS or with the TRIP.DELTA.U3.CMV-EGFPs vector (DPBS), die within 9 days following the inoculation of the viral challenge dose. On the other hand, the 6 mice immunized with the TRIP.DELTA.U3.CMV-sE (WNV) vector are resistant to the lethal dose and show no morbidity. As in the first group of mice, the sera from the challenged mice react strongly with the E protein of West Nile virus (FIG. 3) and the neutralizing antibodies have a titre of 100, 1 month after the challenge.

[0181] In addition, the absence of reactivity of the antibodies from challenged mice, with respect to the non-structural proteins of West Nile virus (FIG. 3), suggest that the protective immunity induced by the TRIP.DELTA.U3.CMV-sE (WNV) vector is sufficient to prevent infection with the challenge virus.

[0182] The results show that a single injection of a small amount of TRIP.DELTA.U3.CMV-sE (WNV) vector particles in adult mice induces, after two weeks of immunization, the production of neutralizing antibodies and confers protective immunity against a lethal challenge with West Nile virus inoculated peripherally.

EXAMPLE 4

Preparation of Viral Pseudoparticles Using the TRIP.DELTA.U3.CMV-prM-E (WNV) Recombinant Vector

1) Preparation of the TRIP.DELTA.U3.CMV-prM-E (WNV) Vector

[0183] A recombinant HIV vector of triplex type, comprising a cDNA encoding the prM and E proteins of the IS-98-ST1 strain of West Nile virus, corresponding to positions 399 to 2469 of the sequence of the genome (application FR 01 04599 and Genbank AF481864), was constructed as described in Example 1. Stable lines transduced with the TRIP.DELTA.U3.CMV-prM-E (WNV) recombinant vector were obtained as described in Example 1.

2) Preparation of Viral Pseudoparticles or VLPs

[0184] The culture supernatant of the cells transduced with the TRIP.DELTA.U3.CMV-prM-E (WNV) vector is harvested, and precipitated with PEG 6000 (Fluka, 0.7% W/V) 4 to 5 hours at 4.degree. C., with gentle agitation. The precipitate obtained is centrifuged for 30 minutes at 9000 rpm at 4.degree. C., and the pellet containing the VLPs is taken up in 4 ml of TNE (20 mM Tris-HCl, pH 8.0; 150 mM NaCl; 2 mM EDTA) and deposited onto a discontinuous sucrose gradient (20%-60% sucrose in 1.times.TNE). The gradient is centrifuged at 39 000 rpm for 2 h, and the opalescent band at the 20-60% interface is harvested, deposited on a linear gradient (11-55% sucrose in 1.times.TNE) and centrifuged at 35 000 rpm for 16 h. The gradient fractions are collected (11 fractions of 0.5 ml) and then analyzed by ELISA using an anti-WNV immune serum (1:20), by SDS-PAGE gel electrophoresis and Coomassie blue staining, and by Western blotting using an anti-WNV immune serum. The results of the ELISA, given in FIG. 5, indicate the presence of purified VLPs in fractions 6 to 10 of the gradient.

EXAMPLE 5

Analysis of the Immunogenic and Protective Capacity of the TRIP.DELTA.U3.CMV-sE Vector in 129 Mice

1) Materials and Methods

1.1) Immunization/Vaccination Protocol

[0185] Six to eight week old 129 mice (six groups of six mice) were intraperitoneally (i.p.) inoculated with varying doses of TRIP.DELTA.U3.CMV-sE (WNV) vector particles prepared as described in Example 1, diluted in 0.1 ml Dulbecco's PBS (DPBS; pH 7.5) supplemented with 0.2% bovine serum albumin (BSA).

[0186] The animals were given a single vaccine injection.

[0187] The control groups were inoculated, under the same conditions, with 500 ng p24 antigen equivalent of TRIP.DELTA.U3.CMV-GFP vector particles prepared in a similar manner to the TRIP.DELTA.U3.CMV-sE (WNV) vector particles (one group of six mice), or DPBS buffer alone (one group of six mice).

[0188] Mice were bled periorbitally at 6, 13, 20 or 27 days post-immunization (D.sub.6, D.sub.13, D.sub.20, D.sub.27) and pooled sera were heat-inactivated for 30 min at 56.degree. C. before measurement of anti-WNV total antibodies, IgG and IgM, and in vitro neutralizing activity, as described in example 2.

[0189] WNV challenge was performed by i.p. inoculation of neurovirulent WNV strain IS-98-ST1, prepared as described in example 2. Animals were subsequently challenged i.p. with 1.000 LD.sub.50 (i.p. LD.sub.50=10 FFU) of WNV strain IS-98-ST1 at 7 or 14 days post immunization. The challenged mice were monitored daily for signs of morbidity or mortality, for up to 21 days.

1.2) Flow Cytometry Assay

[0190] 293T cells cultured on 25 cm2 flasks were transduced with TRIP.DELTA.U3.CMV-GFP vector particles which have been, either heat-inactivated for 0.10 min at 70.degree. C., or untreated (positive control). At 48 h, cells were detached, washed and fixed with 2% PFA. The GFP fluorescence intensity was measured by FACSscan and analyzed with CellQuest software.

2) Results

[0191] In order to take into account inter-individual immune response variability, 129 mice which are less congenic than BALB/c were selected for assessing the humoral immune response induced by the lentiviral vector expressing WNV-sE.

2.1) Strong Antibody Responses Following Intraperitoneal Injection of TRIP.DELTA.U3.CMV-sE Vector Particles.

[0192] In 129 adult mice immunized with a single dose of TRIP.DELTA.U3.CMV-sE (WNV) vector particles equivalent to 500 ng of p24 antigen, total antibodies against WNV were detectable as early as 6 days post immunization, although present at low concentration. By comparison, no anti-WNV antibodies were detected in sera of TRIP.DELTA.U3.CMV-GFP immunized mice. As expected at this time point, the humoral response corresponded to IgM and not IgG antibodies. Total antibody responses increased 10-fold to reach a plateau at day 13, and were then maintained over time. At these later time points (day 13, 20, 27), IgM antibody disappeared, to be replaced by IgG (Table 2).

TABLE-US-00003 TABLE 2 Antibody response of mice to inoculation with TRIP.DELTA.U3.CMV-sE (WNV) Immunizing vector.sup.a, WNV WNV IgM WNV IgG day of antibody antibody antibody Anti-WNV bleeding titer.sup.b titer.sup.b titer.sup.b FRNT90.sup.c TRIP.DELTA.U3.CMV- GFP Day 27 <100 <100 <100 <10 TRIP.DELTA.U3.CMV- sE (WNV) Day 06 3 000 300 <100 10 Day 13 30 000 <100 1 000 10 Day 20 30 000 <100 1 000 10 Day 27 30 000 <100 1 000 20 .sup.aGroups of adult 129 mice were inoculated i.p. with a quantity of lentiviral vector particles corresponding to 500 ng of p24 antigen .sup.bDetermined by ELISA on pooled heat-inactivated sera .sup.cFRNT: Focus Reduction Neutralization Test: the highest serum dilution that reduced the number of FFU of WNV by least 90%.

[0193] These antibodies were reactive with WNV E-glycoprotein from IS-98-ST1 infected Vero cell lysates as demonstrated by RIP assay (FIG. 6A). A focus reduction neutralization test (FRNT) showed that sera from TRIP.DELTA.U3'.CMV-sE (WNV) immunized mice contained detectable levels of WNV neutralizing antibodies as early as 6-days post-immunization (Table 2). Together these data show that an early and specific anti-WNV antibody immune response is mounted in mice immunized with TRIP.DELTA.U3.CMV-sE (WNV) vector particles.

2.2) Early Protection Conferred to Mice by TRIP.DELTA.U3.CMV-sE (WNV) Vaccination Against a High-Dose WNV Challenge.

[0194] Mice immunized with a single dose of TRIP.DELTA.U3.CMV-sE (WNV) vector particles equivalent to 500 ng of p24 antigen were fully protected against a high viral challenge as early as 7 days post-immunization, since no morbidity or mortality were observed in this group (Table 3).

TABLE-US-00004 TABLE 3 Rapid protection by TRIP.DELTA.U3.CMV-sE (WNV)against WNV infection Immunizing Protection.sup.b Post-challenge vector.sup.a, (n.sup.o of surviving/n.sup.o WNV antibody day of challenge of infected) titer.sup.c DPBS Day 7 0/2 ND Day 14 0/2 ND TRIP.DELTA.U3.CMV-GFP Day 7 0/2 ND Day 14 0/2 ND TRIP.DELTA.U3.CMV-sE (WNV) Day 7 6/6 200 000 Day 14 6/6 300 000 .sup.aGroups of adult 129 mice were inoculated i.p. with a single dose of lentiviral vector particles corresponding to 500 ng of p24 antigen or with DPBS. .sup.bAt day of challenge, mice were inoculated i.p. with 1,000 i.p. LD50 of WNV strain IS-98-ST1. Survivals were recorded for 21 days. .sup.cDetermined by ELISA on pooled heat-inactivated sera. ND: not determined

[0195] The infectious virus dose used in the viral challenge was selected to correspond to the maximal viral inoculum that can be transmitted by a mosquito bite. This dose is estimated to correspond to 10,000 in vitro FFU (Despres et al., J. Infect. Dis., 2005, 191, 207-214; Mashimo et al., 2002, precited), itself corresponding to 1000 in vivo-LD.sub.50 by the intraperitoneal route.

[0196] All mice immunized with the control vector TRIP.DELTA.U3.CMV-GFP or with DPBS died within 11 days of challenge (Table 3). Interestingly, total antibodies against WNV increased by a factor of ten after challenge, suggesting that an effective secondary response was mounted in TRIP.DELTA.U3.CMV-sE (WNV) immunized mice (Table 3). Equivalent results were obtained in BALB/c mice. These results indicate that TRIP.DELTA.U3.CMV-sE (WNV) vaccination confers a very quick, fully protective immune response against a high WNV challenge. This could be of major importance in the context of an outbreak where protection of sensitive species is an emergency.

2.3) The Immunity Conferred by the Lentiviral Vector Vaccine is Sterilizing.

[0197] To address whether or not WNV primo-infection can take place in vaccinated animals upon challenge, in other words, whether the elicited immune response confers sterilizing protective immunity, RIP assays were performed on pooled sera from immunized mice, collected before and at 21 days after WNV challenge. Sera obtained at day 13, 20 and 27 post-immunization with a single dose of TRIP.DELTA.U3.CMV-sE (WNV) vector particles equivalent to 500 ng of p24 antigen, reacted with the E protein of WNV. However, sera obtained from day 6 post-immunization did not react with this protein (FIG. 6A). Since RIP assays are not capable of detecting IgM, this is consistent with the ELISA results that show that at 6 days p.i. only IgM and not IgG antibodies against WNV are present. Sera from TRIP.DELTA.U3.CMV-GFP immunized mice did not react with WNV E protein.

[0198] Interestingly, no antibodies against any viral protein other than WNV E were detected in post-challenge sera from TRIP.DELTA.U3.CMV-sE (WNV) vaccinated mice (FIG. 6B). This absence of antibodies against WNV non-structural proteins, strongly suggests that no viral replication took place in all TRIP.DELTA.U3.CMV-sE (WNV) vaccinated mice. Thus, TRIP.DELTA.U3.CMV-sE (WNV) vaccination confers full sterilizing immunity to mice.

[0199] This could represent an important advantage if the vaccine were to be used for bird-immunization. Indeed, while horses, humans and other mammals are believed to be dead-end hosts of WNV infection, birds are known to be amplifying hosts and participate in the maintenance of an epidemic (Dauphin et al., Comp. Immunol. Microbiol. Infect. Dis., 2004, 27, 343-355).

2.4) Protection Provided by a Single Immunization of TRIP.DELTA.U3.CMV-sE (WNV) is Long Lasting.

[0200] In order to determine whether a single immunization with the TRIP.DELTA.U3.CMV-sE (WNV) lentiviral vector based vaccine has the potential to elicit long-term protective immunity against WNV, pooled sera from the 129 immunized mice were tested by ELISA and FRNT, three months after the injection of the TRIP.DELTA.U3.CMV-sE (WNV) vaccine.

[0201] Antibody levels in mice immunized with a single dose of TRIP.DELTA.U3.CMV-sE (WNV) vector particles equivalent to 500 ng of p24 antigen, were still remarkably high 3 months post injection (1:30,000), and neutralizing antibodies persisted (Table 4).

TABLE-US-00005 TABLE 4 Long-term protection by TRIP/sE.sub.WNV against WNV infection WNV Anti- Protection.sup.d WNV Anti- antibody WNV n.sup.o of antibody WNV Immu- titer.sup.b FRNT90.sup.c surviving/ titer.sup.b FRNT90.sup.c nizing (pre- (pre- n.sup.o of (post- (post- vector.sup.a challenge) challenge) infected challenge) challenge) TRIP.DELTA.U3. <100 <10 0/3 ND ND CMV- GFP TRIP.DELTA.U3. 30 000 20 13/13 500 000 400 CMV-sE (WNV) .sup.ang to 500 ng of p24 antigen. .sup.bDetermined by ELISA on pooled heat-inactivated sera. .sup.cFRNT: Focus Reduction Neutralization Test:: the highest serum dilution that reduced the number of FFU of WNV by least 90%. .sup.dMice were inoculated i.p. with 1000 LD.sub.50 of WNV strain IS-98-ST1, three months post immunization. Survival was recorded for 21 days.

[0202] Neither morbidity nor mortality was observed in mice immunized with TRIP.DELTA.U3.CMV-sE (WNV) and subsequently challenged i.p. with a 1000 LD.sub.50 dose of IS-98-ST1 WNV, whereas all control mice died (Table 4). Total antibody titers as well as neutralizing antibodies increased after challenge, suggesting that an effective secondary response was mounted in mice immunized with a TRIP.DELTA.U3.CMV-sE (WNV) three month earlier (Table 4). This shows that a single immunization with the WNV-sE coding lentiviral vector is enough to provide for a long lasting protective immunity in mice.

2.5) A Single Minute Dose of TRIP.DELTA.U3.CMV-sE (WNV) is Enough to Confer Full and Rapid Protection.

[0203] To calculate the minimal dose of vector required to achieve full protective immunity, several groups of 129 mice were immunized i.p. with decreasing doses of TRIP.DELTA.U3.CMV-sE (WNV) or a 500 ng dose of TRIP.DELTA.U3.CMV-GFP vector particles as a control. Seven days later, all mice were challenged with 1000 LD.sub.50 IS-98-ST1. As expected, all mice that received the control vector died within 11-13 days of challenge. Results showed that the minimal dose of TRIP.DELTA.U3.CMV-sE (WNV) required for full protection of mice was a vector particle quantity equivalent to 50 ng of p24 antigen (Table 5).

TABLE-US-00006 TABLE 5 Dose-dependent protection by TRIP/sE.sub.WNV against WNV infection Protection.sup.b Post-challenge Immunizing vector.sup.a, n.sup.o of surviving/n.sup.o WNV antibody dose (ng of p24) of infected titer.sup.c TRIP.DELTA.U3.CMV-GFP 500 0/6 ND Heat-inactivated TRIP.DELTA.U3.CMV-sE (WNV).sup.d 50 0/6 ND TRIP.DELTA.U3.CMV-sE (WNV) 500 6/6 200 000 150 6/6 300 000 50 12/12 300 000 15 5/6 300 000 5 2/5 200 000 1.5 11/12 ND .sup.aGroups of adult 129 mice were inoculated i.p. with a single dose of lentiviral vector particles. .sup.bMice were inoculated i.p. with 1,000 i.p. LD50 of WNV strain IS-98-ST1 one week after priming. Survival was recorded for 21 days. .sup.cDetermined by ELISA on pooled heat-inactivated sera. .sup.dLentiviral vector particles were heat-inactivated for 10 min at 70.degree. C.

[0204] Lower doses conferred only partial protection thus allowing to calculate the 50% protective dose to be the vector particle equivalent of 6.2 ng of p24 antigen. Of note, these dose-protection experiments were performed in the most stringent challenge conditions, with an early challenge at day 7 post-vaccination and with a high virus challenge inoculum (1000 LD.sub.50). Owing that total antibody concentrations increases by a ten-fold factor between day 7 and 15, it is probable that the 50% protective dose would have been even lower than 6.2 ng if calculated only one week later. Immune sera from mice that received the vector particle equivalent of 50 ng of p24 of TRIP.DELTA.U3.CMV-sE (WNV) had no detectable anti-WNV antibodies. Given that such low amount of TRIP.DELTA.U3.CMV-sE confers full protection one week after priming, one might predict that the lentiviral vector based vaccine must generate signals that initiate innate immunity to WNV.

[0205] Furthermore, it is important to note that the dose required for full protective immunity could have been sub-evaluated due to the model used. Indeed, it has been shown that mice cells have a lower permissivity to lentiviral vector transduction than other mammal cells, including human cells (Giannini et al., Hepatology, 2003, 38, 114-122; Nguyen et al., Mol. Ther., 2002, 6, 199-209). Avian cells show a better permissivity to transduction than murine cells allowing to predict that minute lentiviral vector vaccine doses would be effective in fowl.

[0206] In order to make sure that the protection obtained was specifically due to the actual vector-mediated expression of the WNV-sE antigen and not to residual WNV-sE protein or vector plasmid DNA contaminating the vector stock. Thus, mice were immunized with heat-inactivated (10 min at 70.degree. C.) TRIP.DELTA.U3.CMV-sE (WNV) vector particles, a treatment that abrogates transduction (FIG. 7). After WNV challenge, all mice injected with the heat-inactivated TRIP.DELTA.U3.CMV-sE (WNV) died (Table 5). It is therefore unlikely that free naked DNA plays a role in protection.

[0207] In addition, by virtue of the ubiquitous tropism of the VSV-G envelope used for pseudotyping the vector particles the lentiviral vector vaccine can theoretically be used, with no modification, in any vertebrate species, including humans and animals like horses, fowl, and zoo mammals at risk.

[0208] These results demonstrate that a minute dose of vector particles is enough to achieve quick and fully protective immunity in mice. This makes this candidate vaccine interestingly cost-effective, and allows the set up of protocols for mass vaccination (for instance via aerosols), in poultry stock or horse breeding farms.

Sequence CWU 1

1

271100PRTFlavivirus sp. 1Lys Gly Met Ser Tyr Val Met Cys Thr Gly Ser Phe Lys Leu Glu Lys1 5 10 15Glu Val Ala Glu Thr Gln His Gly Thr Val Leu Val Gln Val Lys Tyr 20 25 30Glu Gly Thr Asp Ala Pro Cys Lys Ile Pro Phe Ser Thr Gln Asp Glu 35 40 45Lys Gly Val Thr Gln Asn Gly Arg Leu Ile Thr Ala Asn Pro Ile Val 50 55 60Thr Asp Lys Glu Lys Pro Ile Asn Ile Glu Thr Glu Pro Pro Phe Gly65 70 75 80Glu Ser Tyr Ile Ile Val Gly Ala Gly Glu Lys Ala Leu Lys Leu Ser 85 90 95Trp Phe Lys Arg 1002100PRTFlavivirus sp. 2Lys Gly Met Ser Tyr Ser Met Cys Thr Gly Lys Phe Lys Val Val Lys1 5 10 15Glu Ile Ala Glu Thr Gln His Gly Thr Ile Val Ile Arg Val Gln Tyr 20 25 30Glu Gly Asp Gly Ser Pro Cys Lys Thr Pro Phe Glu Ile Met Asp Leu 35 40 45Glu Lys Arg His Val Leu Gly Arg Leu Thr Thr Val Asn Pro Ile Val 50 55 60Thr Glu Lys Asp Ser Pro Val Asn Ile Glu Ala Glu Pro Pro Phe Gly65 70 75 80Asp Ser Tyr Ile Ile Gly Val Gly Glu Pro Gly Gln Leu Lys Leu Asp 85 90 95Trp Phe Lys Lys 1003100PRTFlavivirus sp. 3Lys Gly Met Ser Tyr Ala Met Cys Leu Asn Thr Phe Val Leu Lys Lys1 5 10 15Glu Ser Glu Glu Thr Gln His Gly Thr Ile Leu Val Lys Val Glu Tyr 20 25 30Lys Gly Glu Asp Ala Pro Cys Lys Ile Pro Phe Ser Thr Glu Asp Gly 35 40 45Gln Gly Lys Ala His Asn Gly Arg Leu Ile Thr Ala Asn Pro Val Val 50 55 60Thr Lys Lys Glu Lys Pro Val Asn Ile Glu Ala Glu Pro Pro Phe Gly65 70 75 80Glu Ser Asn Ile Val Ile Gly Ile Gly Asp Lys Ala Leu Lys Ile Asn 85 90 95Trp Tyr Arg Lys 100499PRTFlavivirus sp. 4Lys Gly Met Ser Tyr Thr Met Cys Ser Gly Lys Phe Ser Ile Asp Lys1 5 10 15Glu Met Ala Glu Thr Gln His Gly Thr Thr Val Val Lys Val Lys Tyr 20 25 30Glu Gly Ala Gly Ala Pro Cys Lys Val Ile Glu Ile Arg Asp Val Asn 35 40 45Lys Glu Lys Val Val Gly Arg Ile Ile Ser Ser Thr Pro Leu Ala Glu 50 55 60Asn Thr Asn Ser Val Thr Asn Ile Glu Leu Glu Arg Pro Leu Gly Asp65 70 75 80Ser Tyr Ile Val Ile Val Gly Gly Asn Ser Ala Leu Thr Leu His Trp 85 90 95Phe Arg Lys598PRTHepatitis C virus 5Ala Gln Ile Leu Asn Leu Lys Glu Lys Pro Asn Val Thr Pro Thr Ala1 5 10 15Ala His Arg Thr Leu Ser Ser Arg Val Ala Val Arg Ser Leu Ala Glu 20 25 30Phe Thr Cys Cys Arg Ala Gly Ala Pro Asp Trp Val Cys Ala Arg Leu 35 40 45Gly Arg Leu Pro Ser Gly Arg Ser Leu Val Glu Gly Ala Ser Leu Ser 50 55 60Pro Arg Ile Ala Gly Pro Arg Ala Gly Pro Gly Leu Ser Pro Gly Thr65 70 75 80Leu Gly Pro Ser Met Ala Met Arg Val Ala Gly Gly Gln Asp Gly Ser 85 90 95Cys Pro698PRTHepatitis C virus 6Ala Arg Ile Leu Asn Leu Lys Glu Lys Pro Asn Val Thr Pro Thr Val1 5 10 15Ala His Arg Thr Ser Ser Ser Arg Val Ala Val Arg Ser Leu Val Glu 20 25 30Phe Thr Cys Cys Arg Ala Gly Ala Leu Asp Trp Val Cys Ala Arg Arg 35 40 45Gly Arg Leu Pro Ser Gly Arg Asn Leu Glu Val Asp Val Ser Leu Ser 50 55 60Pro Arg His Val Gly Pro Arg Ala Gly Pro Gly Leu Ser Pro Gly Thr65 70 75 80Leu Gly Pro Ser Met Ala Met Arg Val Ala Gly Gly Arg Asp Gly Ser 85 90 95Cys Leu798PRTHepatitis C virus 7Ala His Phe Leu Asn Leu Lys Glu Lys Pro Lys Glu Thr Pro Ser Val1 5 10 15Ala His Arg Thr Ser Ser Ser Arg Val Ala Asp Arg Ser Leu Val Glu 20 25 30Tyr Thr Cys Cys Arg Ala Gly Ala His Asp Trp Val Cys Ala Arg Arg 35 40 45Val Lys Leu Leu Asn Gly His Ser Leu Ala Asp Asp Asp Ser Leu Ser 50 55 60Pro Arg Arg Val Gly Ala Lys Ala Gly Pro Gly Leu Ser Pro Gly Thr65 70 75 80Leu Gly Pro Ser Met Val Thr Arg Ala Ala Gly Gln Gly Gly Gly Ser 85 90 95Cys Pro898PRTHepatitis C virus 8Ala Gln Ile Leu Asn Leu Lys Glu Lys Pro Lys Glu Thr Gln Thr Ala1 5 10 15Ala His Arg Thr Leu Ser Ser Arg Val Ala Val Arg Ser Leu Ala Glu 20 25 30Phe Thr Cys Cys Arg Ala Gly Ala Pro Gly Trp Val Cys Ala Arg Gln 35 40 45Gly Arg Leu Leu Ser Asp Pro Ser Arg Val Asp Asp Ala Ser Pro Ser 50 55 60Arg Lys Ile Gly Ala Pro Pro Ala Ser Pro Gly Glu Ser Gln Asp Ile65 70 75 80Leu Gly Pro Cys Thr Glu Thr Arg Val Ala Ala Gly Arg Val Gly Ser 85 90 95Cys Pro998PRTHepatitis C virus 9Ala Arg Ile Leu Asn Leu Lys Glu Lys Pro Asn Val Thr Pro Thr Ala1 5 10 15Ala His Arg Thr Leu Ser Ser Arg Val Ala Ala Arg Ser Leu Ala Glu 20 25 30Phe Thr Cys Cys Arg Ala Gly Ala Pro Glu Trp Val Cys Ala Arg Arg 35 40 45Gly Arg Leu Pro Ser Gly Arg Asn Leu Ala Gly Gly Val Ser Leu Phe 50 55 60Pro Arg Pro Ala Asp Pro Arg Glu Gly Pro Gly Arg Ser Pro Gly Thr65 70 75 80Leu Gly Pro Ser Met Ala Thr Arg Ala Val Gly Gly Arg Asp Pro Ser 85 90 95Cys Pro1098PRTHepatitis C virus 10Ala Gln Ile Leu Asn Leu Lys Glu Lys Pro Lys Glu Thr Pro Thr Val1 5 10 15Ala His Lys Thr Leu Ser Phe Arg Ala Ala Ala Arg Ser Leu Ala Glu 20 25 30Tyr Thr Cys Cys Arg Ala Gly Ala Pro Gly Trp Val Cys Ala Arg Gln 35 40 45Gly Arg Leu Arg Ser Gly Pro Ser His Val Glu Gly Ala Ser Pro Ser 50 55 60Leu Arg Ile Gly Ala Pro Leu Ala Asn Pro Gly Glu Asn Gln Asp Thr65 70 75 80Pro Gly Pro Tyr Thr Gly Met Arg Asp Ser Ala Gly Gln Asp Arg Ser 85 90 95Cys Pro1198PRTHepatitis C virus 11Ala Gln Ile Gln Asn Pro Lys Asp Lys Pro Lys Glu Thr Pro Thr Val1 5 10 15Ala His Arg Thr Ser Ser Ser Arg Ala Val Val Arg Ser Trp Val Glu 20 25 30Tyr Thr Cys Cys Arg Ala Gly Ala Leu Asp Trp Val Cys Ala Arg Leu 35 40 45Gly Arg Leu Pro Asn Gly Pro Ser Pro Glu Ala Gly Val Ser Pro Phe 50 55 60Gln Arg Leu Ala Ala Arg Arg Ala Val Pro Gly Val Ser Leu Gly Thr65 70 75 80His Gly Pro Cys Met Gly Met Arg Ala Ala Gly Gly Gln Gly Gly Ser 85 90 95Cys Pro1298PRTHepatitis C virus 12Ala Arg Ile Leu Asn Leu Lys Glu Lys Pro Asn Val Thr Pro Thr Ala1 5 10 15Ala Gln Trp Thr Leu Ser Ser Arg Val Val Ala Arg Ser Leu Ala Glu 20 25 30Phe Thr Cys Cys Arg Ala Gly Ala Pro Asp Trp Val Cys Ala Arg Leu 35 40 45Gly Arg Leu Arg Ser Gly Arg Asn Leu Val Glu Asp Ala Asn Leu Ser 50 55 60Pro Arg Arg Val Asp Pro Arg Glu Gly Pro Gly His Asn Gln Asp Ile65 70 75 80His Gly Leu Phe Thr Val Met Arg Val Val Gly Gly Gln Asp Gly Ser 85 90 95Cys Pro1398PRTHepatitis C virus 13Ala Gln Ile Leu Asn Leu Lys Glu Lys Pro Asn Val Thr Pro Thr Ala1 5 10 15Ala His Arg Thr Ser Ser Ser Arg Ala Val Val Arg Ser Leu Val Glu 20 25 30Phe Thr Cys Cys Arg Ala Gly Ala Pro Gly Trp Val Cys Ala Arg Leu 35 40 45Gly Arg Leu Pro Ser Gly Arg Asn Leu Val Glu Gly Asp Asn Leu Ser 50 55 60Pro Arg Phe Ala Gly Pro Arg Ala Gly Pro Gly Leu Ser Pro Gly Thr65 70 75 80Leu Gly Pro Ser Met Ala Met Arg Val Trp Gly Gly Gln Asp Gly Ser 85 90 95Cys His14160PRTHepatitis C virus 14Ala Arg Ile Leu Asn Leu Lys Lys Lys Thr Asn Val Thr Pro Thr Val1 5 10 15Ala His Arg Thr Ser Ser Ser Arg Val Ala Val Arg Ser Leu Val Glu 20 25 30Phe Thr Cys Cys Arg Ala Gly Ala Leu Asp Trp Val Cys Ala Arg Arg 35 40 45Glu Arg Leu Pro Ser Gly Arg Asn Leu Glu Val Asp Val Ser Leu Ser 50 55 60Pro Arg Leu Val Gly Pro Arg Ala Gly Pro Gly Leu Ser Pro Gly Thr65 70 75 80Leu Gly Pro Ser Met Ala Met Arg Ala Ala Gly Gly Arg Asp Gly Ser 85 90 95Cys Leu Pro Val Ala Leu Gly Leu Ala Gly Ala Pro Gln Thr Pro Gly 100 105 110Val Gly Arg Ala Ile Trp Val Arg Ser Ser Ile Pro Leu Arg Ala Ala 115 120 125Ser Pro Thr Ser Trp Gly Thr Tyr Arg Ser Ser Ala Pro Leu Leu Glu 130 135 140Ala Leu Pro Gly Pro Trp Arg Met Ala Ser Gly Phe Trp Lys Thr Ala145 150 155 160154555DNAartificial sequenceplasmid 15tggaagggct aattcactcc caacgaagac aagatatcct tgatctgtgg atctaccaca 60cacaaggcta cttccctgat tagcagaact acacaccagg gccagggatc agatatccac 120tgacctttgg atggtgctac aagctagtac cagttgagcc agagaagtta gaagaagcca 180acaaaggaga gaacaccagc ttgttacaac ctgtgagcct gcatgggatg gatgacccgg 240agagagaagt gttagagtgg aggtttgaca gccgcctagc atttcatcac ggtggcccga 300gagctgcatc cggagtactt caagaactgc tgatatcgag cttgctacaa gggactttcc 360gctgggggac tttccaggga ggcgtggcct gggcgggact ggggagtggc gagccctcag 420atcctgcata taagcagctg ctttttgcct gtactgggtc tctctggtta gaccagatct 480gagcctggga gctctctggc taactaggga acccactgct taagcctcaa taaagcttgc 540cttgagtgct tcaagtagtg tgtgcccgtc tgttgtgtga ctctggtaac tagagatccc 600tcagaccctt ttagtcagtg tggaaaatct ctagcagtgg cgcccgaaca gggacttgaa 660agcgaaaggg aaaccagagg agctctctcg acgcaggact cggcttgctg aagcgcggaa 720ttccgcgcca cggcaagagg cgaggggcgg cgactggtga gtacgccaaa aattttgact 780agcggaggct agaaggagag agatgggtgc gagagcgtca gtattaagcg ggggagaatt 840agatcgcgat gggaaaaaat tcggttaagg ccagggggaa agaaaaaata taaattaaaa 900catatagtat gggcaagcag ggagctagaa cgattcgcag ttaatcctgg cctgttagaa 960acatcagaag gctgtagaca aatactggga cagctacaac catcccttca gacaggatca 1020gaagaactta gatcattata taatacagta gcaaccctct attgtgtgca tcaaaggata 1080gagataaaag acaccaagga agctttagac aagatagagg aagagcaaaa caaaagtaag 1140accaccgcac agcaagcggc cgctgatctt cagacctgga ggaggagata tgagggacaa 1200ttggagaagt gaattatata aatataaagt agtaaaaatt gaaccattag gagtagcacc 1260caccaaggca aagagaagag tggtgcagag agaaaaaaga gcagtgggaa taggagcttt 1320gttccttggg ttcttgggag cagcaggaag cactatgggc gcagcgtcaa tgacgctgac 1380ggtacaggcc agacaattat tgtctggtat agtgcagcag cagaacaatt tgctgagggc 1440tattgaggcg caacagcatc tgttgcaact cacagtctgg ggcatcaagc agctccaggc 1500aagaatcctg gctgtggaaa gatacctaaa ggatcaacag ctcctgggga tttggggttg 1560ctctggaaaa ctcatttgca ccactgctgt gccttggaat gctagttgga gtaataaatc 1620tctggaacag atttggaatc acacgacctg gatggagtgg gacagagaaa ttaacaatta 1680cacaagctta atacactcct taattgaaga atcgcaaaac cagcaagaaa agaatgaaca 1740agaattattg gaattagata aatgggcaag tttgtggaat tggtttaaca taacaaattg 1800gctgtggtat ataaaattat tcataatgat agtaggaggc ttggtaggtt taagaatagt 1860ttttgctgta ctttctatag tgaatagagt taggcaggga tattcaccat tatcgtttca 1920gacccacctc ccaaccccga ggggacccga caggcccgaa ggaatagaag aagaaggtgg 1980agagagagac agagacagat ccattcgatt agtgaacgga tctcgacggt atcgccgaat 2040tcacaaatgg cagtattcat ccacaatttt aaaagaaaag gggggattgg ggggtacagt 2100gcaggggaaa gaatagtaga cataatagca acagacatac aaactaaaga attacaaaaa 2160caaattacaa aaattcaaaa ttttcgggtt tattacaggg acagcagaga tccactttgg 2220ggcgataagc ttgggagttc cgcgttacat aacttacggt aaatggcccg cctggctgac 2280cgcccaacga cccccgccca ttgacgtcaa taatgacgta tgttcccata gtaacgccaa 2340tagggacttt ccattgacgt caatgggtgg agtatttacg gtaaactgcc cacttggcag 2400tacatcaagt gtatcatatg ccaagtacgc cccctattga cgtcaatgac ggtaaatggc 2460ccgcctggca ttatgcccag tacatgacct tatgggactt tcctacttgg cagtacatct 2520acgtattagt catcgctatt accatggtga tgcggttttg gcagtacatc aatgggcgtg 2580gatagcggtt tgactcacgg ggatttccaa gtctccaccc cattgacgtc aatgggagtt 2640tgttttggca ccaaaatcaa cgggactttc caaaatgtcg taacaactcc gccccattga 2700cgcaaatggg cggtaggcgt gtacggtggg aggtctatat aagcagagct cgtttagtga 2760accgtcagat cgcctggaga cgccatccac gctgttttga cctccataga agacaccgac 2820tctagaggac gtacgatgag agttgtgttt gtcgtgctat tgcttttggt ggccccagct 2880tacagcttca actgccttgg aatgagcaac agagacttct tggaaggagt gtctggagca 2940acatgggtgg atttggttct cgaaggcgac agctgcgtga ctatcatgtc taaggacaag 3000cctaccatcg atgtgaagat gatgaatatg gaggcggtca acctggcaga ggtccgcagt 3060tattgctatt tggctaccgt cagcgatctc tccaccaaag ctgcgtgccc gaccatggga 3120gaagctcaca atgacaaacg tgctgaccca gcttttgtgt gcagacaagg agtggtggac 3180aggggctggg gcaacggctg cggattattt ggcaaaggaa gcattgacac atgcgccaaa 3240tttgcctgct ctaccaaggc aataggaaga accatcttga aagagaatat caagtacgaa 3300gtggccattt ttgtccatgg accaactact gtggagtcgc acggaaacta ctccacacag 3360gttggagcca ctcaggcagg gagattcagc atcactcctg cggcgccttc atacacacta 3420aagcttggag aatatggaga ggtgacagtg gactgtgaac cacggtcagg gattgacacc 3480aatgcatact acgtgatgac tgttggaaca aagacgttct tggtccatcg tgagtggttc 3540atggacctca acctcccttg gagcagtgct ggaagtactg tgtggaggaa cagagagacg 3600ttaatggagt ttgaggaacc acacgccacg aagcagtctg tgatagcatt gggctcacaa 3660gagggagctc tgcatcaagc tttggctgga gccattcctg tggaattttc aagcaacact 3720gtcaagttga cgtcgggtca tttgaagtgt agagtgaaga tggaaaaatt gcagttgaag 3780ggaacaacct atggcgtctg ttcaaaggct ttcaagtttc ttgggactcc cgcagacaca 3840ggtcacggca ctgtggtgtt ggaattgcag tacactggca cggatggacc ttgcaaagtt 3900cctatctcgt cagtggcttc attgaacgac ctaacgccag tgggcagatt ggtcactgtc 3960aacccttttg tttcagtggc cacggccaac gctaaggtcc tgattgaatt ggaaccaccc 4020tttggagact catacatagt ggtgggcaga ggagaacaac agatcaatca ccattggcac 4080aagtctggaa gcagcattgg caaagccttt acaaccaccc tcaaaggagc gcagagacta 4140gccgctctag gagacacagc ttgggacttt ggatcagttg gaggggtgtt cacctcagtt 4200gggaaggctg tctaatgcgc gcggtacctt taagaccaat gacttacaag gcagctgtag 4260atcttagcca ctttttaaaa gaaaaggggg gactggaagg gctaattcac tcccaacgaa 4320gacaagatcg tcgagagatg ctgcatataa gcagctgctt tttgcttgta ctgggtctct 4380ctggttagac cagatctgag cctgggagct ctctggctaa ctagggaacc cactgcttaa 4440gcctcaataa agcttgcctt gagtgcttca agtagtgtgt gcccgtctgt tgtgtgactc 4500tggtaactag agatccctca gaccctttta gtcagtgtgg aaaatctcta gcagt 4555161393DNAFlavivirus sp.CDS(7)..(1386) 16cgtacg atg aga gtt gtg ttt gtc gtg cta ttg ctt ttg gtg gcc cca 48 Met Arg Val Val Phe Val Val Leu Leu Leu Leu Val Ala Pro 1 5 10gct tac agc ttc aac tgc ctt gga atg agc aac aga gac ttc ttg gaa 96Ala Tyr Ser Phe Asn Cys Leu Gly Met Ser Asn Arg Asp Phe Leu Glu15 20 25 30gga gtg tct gga gca aca tgg gtg gat ttg gtt ctc gaa ggc gac agc 144Gly Val Ser Gly Ala Thr Trp Val Asp Leu Val Leu Glu Gly Asp Ser 35 40 45tgc gtg act atc atg tct aag gac aag cct acc atc gat gtg aag atg 192Cys Val Thr Ile Met Ser Lys Asp Lys Pro Thr Ile Asp Val Lys Met 50 55 60atg aat atg gag gcg gtc aac ctg gca gag gtc cgc agt tat tgc tat 240Met Asn Met Glu Ala Val Asn Leu Ala Glu Val Arg Ser Tyr Cys Tyr 65 70 75ttg gct acc gtc agc gat ctc tcc acc aaa gct gcg tgc ccg acc atg 288Leu Ala Thr Val Ser Asp Leu Ser Thr Lys Ala Ala Cys Pro Thr Met 80 85 90gga gaa gct cac aat gac aaa cgt gct gac cca gct ttt gtg tgc aga 336Gly Glu Ala His Asn Asp Lys Arg Ala Asp Pro Ala Phe Val Cys Arg95 100 105 110caa gga gtg gtg gac agg ggc tgg ggc aac ggc tgc gga tta ttt ggc 384Gln Gly Val Val Asp Arg Gly Trp Gly Asn Gly Cys Gly Leu Phe Gly 115 120 125aaa gga agc att gac aca tgc gcc aaa ttt gcc tgc tct acc aag gca 432Lys Gly Ser Ile Asp Thr Cys Ala Lys Phe Ala Cys Ser Thr Lys Ala 130 135 140ata gga aga acc atc ttg aaa gag aat atc aag tac

gaa gtg gcc att 480Ile Gly Arg Thr Ile Leu Lys Glu Asn Ile Lys Tyr Glu Val Ala Ile 145 150 155ttt gtc cat gga cca act act gtg gag tcg cac gga aac tac tcc aca 528Phe Val His Gly Pro Thr Thr Val Glu Ser His Gly Asn Tyr Ser Thr 160 165 170cag gtt gga gcc act cag gca ggg aga ttc agc atc act cct gcg gcg 576Gln Val Gly Ala Thr Gln Ala Gly Arg Phe Ser Ile Thr Pro Ala Ala175 180 185 190cct tca tac aca cta aag ctt gga gaa tat gga gag gtg aca gtg gac 624Pro Ser Tyr Thr Leu Lys Leu Gly Glu Tyr Gly Glu Val Thr Val Asp 195 200 205tgt gaa cca cgg tca ggg att gac acc aat gca tac tac gtg atg act 672Cys Glu Pro Arg Ser Gly Ile Asp Thr Asn Ala Tyr Tyr Val Met Thr 210 215 220gtt gga aca aag acg ttc ttg gtc cat cgt gag tgg ttc atg gac ctc 720Val Gly Thr Lys Thr Phe Leu Val His Arg Glu Trp Phe Met Asp Leu 225 230 235aac ctc cct tgg agc agt gct gga agt act gtg tgg agg aac aga gag 768Asn Leu Pro Trp Ser Ser Ala Gly Ser Thr Val Trp Arg Asn Arg Glu 240 245 250acg tta atg gag ttt gag gaa cca cac gcc acg aag cag tct gtg ata 816Thr Leu Met Glu Phe Glu Glu Pro His Ala Thr Lys Gln Ser Val Ile255 260 265 270gca ttg ggc tca caa gag gga gct ctg cat caa gct ttg gct gga gcc 864Ala Leu Gly Ser Gln Glu Gly Ala Leu His Gln Ala Leu Ala Gly Ala 275 280 285att cct gtg gaa ttt tca agc aac act gtc aag ttg acg tcg ggt cat 912Ile Pro Val Glu Phe Ser Ser Asn Thr Val Lys Leu Thr Ser Gly His 290 295 300ttg aag tgt aga gtg aag atg gaa aaa ttg cag ttg aag gga aca acc 960Leu Lys Cys Arg Val Lys Met Glu Lys Leu Gln Leu Lys Gly Thr Thr 305 310 315tat ggc gtc tgt tca aag gct ttc aag ttt ctt ggg act ccc gca gac 1008Tyr Gly Val Cys Ser Lys Ala Phe Lys Phe Leu Gly Thr Pro Ala Asp 320 325 330aca ggt cac ggc act gtg gtg ttg gaa ttg cag tac act ggc acg gat 1056Thr Gly His Gly Thr Val Val Leu Glu Leu Gln Tyr Thr Gly Thr Asp335 340 345 350gga cct tgc aaa gtt cct atc tcg tca gtg gct tca ttg aac gac cta 1104Gly Pro Cys Lys Val Pro Ile Ser Ser Val Ala Ser Leu Asn Asp Leu 355 360 365acg cca gtg ggc aga ttg gtc act gtc aac cct ttt gtt tca gtg gcc 1152Thr Pro Val Gly Arg Leu Val Thr Val Asn Pro Phe Val Ser Val Ala 370 375 380acg gcc aac gct aag gtc ctg att gaa ttg gaa cca ccc ttt gga gac 1200Thr Ala Asn Ala Lys Val Leu Ile Glu Leu Glu Pro Pro Phe Gly Asp 385 390 395tca tac ata gtg gtg ggc aga gga gaa caa cag atc aat cac cat tgg 1248Ser Tyr Ile Val Val Gly Arg Gly Glu Gln Gln Ile Asn His His Trp 400 405 410cac aag tct gga agc agc att ggc aaa gcc ttt aca acc acc ctc aaa 1296His Lys Ser Gly Ser Ser Ile Gly Lys Ala Phe Thr Thr Thr Leu Lys415 420 425 430gga gcg cag aga cta gcc gct cta gga gac aca gct tgg gac ttt gga 1344Gly Ala Gln Arg Leu Ala Ala Leu Gly Asp Thr Ala Trp Asp Phe Gly 435 440 445tca gtt gga ggg gtg ttc acc tca gtt ggg aag gct gtc taa tgcgcgc 1393Ser Val Gly Gly Val Phe Thr Ser Val Gly Lys Ala Val 450 45517459PRTFlavivirus sp. 17Met Arg Val Val Phe Val Val Leu Leu Leu Leu Val Ala Pro Ala Tyr1 5 10 15Ser Phe Asn Cys Leu Gly Met Ser Asn Arg Asp Phe Leu Glu Gly Val 20 25 30Ser Gly Ala Thr Trp Val Asp Leu Val Leu Glu Gly Asp Ser Cys Val 35 40 45Thr Ile Met Ser Lys Asp Lys Pro Thr Ile Asp Val Lys Met Met Asn 50 55 60Met Glu Ala Val Asn Leu Ala Glu Val Arg Ser Tyr Cys Tyr Leu Ala65 70 75 80Thr Val Ser Asp Leu Ser Thr Lys Ala Ala Cys Pro Thr Met Gly Glu 85 90 95Ala His Asn Asp Lys Arg Ala Asp Pro Ala Phe Val Cys Arg Gln Gly 100 105 110Val Val Asp Arg Gly Trp Gly Asn Gly Cys Gly Leu Phe Gly Lys Gly 115 120 125Ser Ile Asp Thr Cys Ala Lys Phe Ala Cys Ser Thr Lys Ala Ile Gly 130 135 140Arg Thr Ile Leu Lys Glu Asn Ile Lys Tyr Glu Val Ala Ile Phe Val145 150 155 160His Gly Pro Thr Thr Val Glu Ser His Gly Asn Tyr Ser Thr Gln Val 165 170 175Gly Ala Thr Gln Ala Gly Arg Phe Ser Ile Thr Pro Ala Ala Pro Ser 180 185 190Tyr Thr Leu Lys Leu Gly Glu Tyr Gly Glu Val Thr Val Asp Cys Glu 195 200 205Pro Arg Ser Gly Ile Asp Thr Asn Ala Tyr Tyr Val Met Thr Val Gly 210 215 220Thr Lys Thr Phe Leu Val His Arg Glu Trp Phe Met Asp Leu Asn Leu225 230 235 240Pro Trp Ser Ser Ala Gly Ser Thr Val Trp Arg Asn Arg Glu Thr Leu 245 250 255Met Glu Phe Glu Glu Pro His Ala Thr Lys Gln Ser Val Ile Ala Leu 260 265 270Gly Ser Gln Glu Gly Ala Leu His Gln Ala Leu Ala Gly Ala Ile Pro 275 280 285Val Glu Phe Ser Ser Asn Thr Val Lys Leu Thr Ser Gly His Leu Lys 290 295 300Cys Arg Val Lys Met Glu Lys Leu Gln Leu Lys Gly Thr Thr Tyr Gly305 310 315 320Val Cys Ser Lys Ala Phe Lys Phe Leu Gly Thr Pro Ala Asp Thr Gly 325 330 335His Gly Thr Val Val Leu Glu Leu Gln Tyr Thr Gly Thr Asp Gly Pro 340 345 350Cys Lys Val Pro Ile Ser Ser Val Ala Ser Leu Asn Asp Leu Thr Pro 355 360 365Val Gly Arg Leu Val Thr Val Asn Pro Phe Val Ser Val Ala Thr Ala 370 375 380Asn Ala Lys Val Leu Ile Glu Leu Glu Pro Pro Phe Gly Asp Ser Tyr385 390 395 400Ile Val Val Gly Arg Gly Glu Gln Gln Ile Asn His His Trp His Lys 405 410 415Ser Gly Ser Ser Ile Gly Lys Ala Phe Thr Thr Thr Leu Lys Gly Ala 420 425 430Gln Arg Leu Ala Ala Leu Gly Asp Thr Ala Trp Asp Phe Gly Ser Val 435 440 445Gly Gly Val Phe Thr Ser Val Gly Lys Ala Val 450 4551833DNAARTIFICIAL SEQUENCEprimer 18tatcgtacga tgagagttgt gtttgtcgtg cta 331931DNAARTIFICIAL SEQUENCEprimer 19atagcgcgct tagacagccc ttcccaactg a 312027DNAARTIFICIAL SEQUENCELTR-FL probe 20cacaacagac gggcacacac tacttga 272125DNAARTIFICIAL SEQUENCELTR-LC probe 21cactcaaggc aagctttatt gaggc 252225DNAARTIFICIAL SEQUENCEAA55M primer 22gctagagatt ttccacactg actaa 252321DNAARTIFICIAL SEQUENCEM667 primer 23ggctaactag ggaacccact g 212434DNAARTIFICIAL SEQUENCECD3-P1 probe 24ggctgaaggt tagggatacc aatattcctg tctc 342530DNAARTIFICIAL SEQUENCECD3-P2 probe 25ctagtgatgg gctcttccct tgagcccttc 302623DNAARTIFICIAL SEQUENCECD3-in-F primer 26ggctatcatt cttcttcaag gta 232722DNAARTIFICIAL SEQUENCECD3-in-R primer 27cctctcttca gccatttaag ta 22

* * * * *

References


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed