Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MODIFIED VSV-G AND VACCINES THEREOF
Document Type and Number:
WIPO Patent Application WO/2018/050738
Kind Code:
A1
Abstract:
The present invention relates to modified vesicular stomatitis virus glycoprotein (VSV-G) comprising at least one peptide, preferably an antigen or fragment thereof, nucleic acid sequence coding therefor, and vectors containing said nucleic acid sequence. The present invention also relates to vaccines and methods for the treatment of a disease or condition, in particular a cancer or an infectious disease.

Inventors:
VANDERMEULEN GAËLLE (BE)
LAMBRICHT LAURE (BE)
PRÉAT VÉRONIQUE (BE)
Application Number:
PCT/EP2017/073119
Publication Date:
March 22, 2018
Filing Date:
September 14, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV CATHOLIQUE LOUVAIN (BE)
International Classes:
A61K39/00
Domestic Patent References:
WO1995014785A11995-06-01
WO1996022378A11996-07-25
WO1994019478A11994-09-01
Foreign References:
US5882877A1999-03-16
US6013516A2000-01-11
US4861719A1989-08-29
US5278056A1994-01-11
Other References:
MAO CHIH-PING ET AL: "Combined Administration with DNA Encoding Vesicular Stomatitis Virus G Protein Enhances DNA Vaccine Potency", JOURNAL OF VIROLOGY, vol. 84, no. 5, March 2010 (2010-03-01), pages 2331 - 2339, XP002767683, ISSN: 0022-538X
ULMER ET AL., SCIENCE, vol. 259, 1993, pages 1745 - 1749
WANG ET AL., PNAS, vol. 90, 1993, pages 4156 - 4160
WANG ET AL., IMMUNOL. REV., vol. 170, 1999, pages 85 - 100
MOHAN ET AL., INDIAN. J. MED. RES., vol. 138, no. 5, 2013, pages 779 - 795
KLINMAN ET AL., J. IMMUNOL., vol. 158, no. 8, 1997, pages 3635 - 3639
LAMBRICHT ET AL., ALOL. THER., vol. 24, no. 9, 2016, pages 1686 - 1696
MARSAC ET AL., J. VIROL., vol. 76, no. 15, 2002, pages 7544 - 7553
MAO ET AL., J. VIROL., vol. 84, no. 5, 2010, pages 2331 - 2339
BATEMAN ET AL., CANCER RES., vol. 60, no. 6, 2000, pages 1492 - 1497
BATEMAN ET AL., CANCER RES., vol. 62, no. 22, 2002, pages 6566 - 6578
SCHLAKE ET AL., RNA BIOL., vol. 9, no. 11, 2012, pages 1319 - 1330
SAHIN ET AL., NAT REV DRUG DISCOV., vol. 13, no. 10, 2014, pages 759 - 780
MCNAMARA ET AL., J IMMUNOL RES., 2015, pages 794528
GUIBINGA ET AL., MOL. THER., vol. 9, no. 1, 2004, pages 76 - 84
AMMAYAPPAN ET AL., J. VIROL., vol. 87, no. 24, 2013, pages 13543 - 13555
GRIGERA ET AL., J. VIROL., vol. 70, no. 12, 1996, pages 8492 - 8501
SCHLEHUBER; ROSE, J. VIROL., vol. 78, no. 10, 2004, pages 5079 - 5087
MAO ET AL., J VIROL., vol. 84, no. 5, 2010, pages 2331 - 2339
CALAROTA; WEINER, EXPERT REV. VACCINES., vol. 3, 2004, pages S135 - S149
CALAROTA; WEINER, IMMUNOL. REV., vol. 199, 2004, pages 84 - 99
KUTZLER; WEINER, J. CLIN. INVEST., vol. 14, no. 9, 2004, pages 1241 - 1244
ARTHUR M. LESK: "Computational Molecular Biology: Sources and Methods for Sequence Analysis", 1988, OXFORD UNIVERSITY PRESS
DOUGLAS W. SMITH: "Biocomputing: Informatics and Genome Projects", 1993, ACADEMIC PRESS
HUGH G. GRIFFIN; ANNETTE M. GRIFFIN: "Computer Analysis of Sequence Data", 1994, HUMANA PRESS
GUNNAR VON HEINJE: "Sequence Analysis in Molecular Biology: Treasure Trove or Trivial Pursuit", 1987, ACADEMIC PRESS
MICHAEL GRIBSKOV; JOHN DEVEREUX: "Sequence Analysis Primer", 1991, M. STOCKTON PRESS
CARILLO ET AL., SIAAF J. APPL. MATH., vol. 48, no. 5, 1988, pages 1073 - 1082
DEVEREUX ET AL., NUCL. ACID. RES., vol. 12, no. 1, 1984, pages 387 - 395
ALTSCHUL ET AL., J. VLOL. BIOL., vol. 215, no. 3, 1990, pages 403 - 410
ALTSCHUL ET AL.: "NCB/NLM/NIH Bethesda", BLAST MANUAL
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, no. 3, 1990, pages 403 - 410
KOBER ET AL., BIOTECHNOL. BIOENG., vol. 110, 2013, pages 1164 - 1173
MORI ET AL., J. BIOSCI. BIOENG., vol. 120, no. 5, 2015, pages 518 - 525
STERN ET AL., TRENDS CELL MOL. BIO., vol. 2, 2007, pages 1 - 17
WEN ET AL., ACTA BIOCHIM BIOPHYS SIN., vol. 43, 2011, pages 96 - 102
CHEEVER ET AL., CLIN CANCER RES., vol. 15, no. 17, 2009, pages 5323 - 5337
VITA ET AL., NUCLEIC ACIDS RES., vol. 43, 2014, pages D405 - D412, Retrieved from the Internet
HIEMSTRA ET AL., PROC NATL ACAD SCI USA., vol. 94, no. 19, 16 September 1997 (1997-09-16), pages 10313 - 10318
VIGNERON ET AL., CANCER IMMUN., vol. 13, 2013, pages 15
"HIV Molecular Immunology Database", HIV.LANL.GOV., 2017, Retrieved from the Internet
"HIV Molecular Immunology 2016", 2017, LOS ALAMOS NATIONAL LABORATORY, THEORETICAL BIOLOGY AND BIOPHYSICS
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS
AUSUBEL ET AL.: "Short Protocols in Molecular Biology", 1992, JOHN WILEY AND SONS
ANGOV ET AL., BIOTECHNOL. J., vol. 6, no. 6, 2011, pages 650 - 659
SHARP; LI, NUCLEIC ACIDS RES., vol. 15, 1987, pages 1281 - 1295
SUPEK; VLAHOVICEK, BMC BIOINFORMATICS, vol. 6, 2005, pages 182
SHARP ET AL., NUCLEIC ACIDS RES., vol. 14, 1986, pages 5125 - 5143
VANDERMEULEN ET AL., MOL. THER., vol. 19, no. 11, 2011, pages 1942 - 1949
STRIBLING ET AL., PROC. NATL. ACAD. SCI. USA., vol. 189, 1992, pages 11277 - 11281
RIDELL ET AL., SCIENCE, vol. 257, 1992, pages 238 - 241
Attorney, Agent or Firm:
ICOSA (FR)
Download PDF:
Claims:
CLAIMS

1. An isolated nucleic acid sequence coding for a modified vesicular stomatitis v irus glycoprotein (VSV-G ) comprising at least one tumor antigen or fragment thereof. 2. The isolated nucleic acid sequence coding for a modified VSV-G according to claim 1, wherein said at least one tumor antigen or fragment thereof comprises at least one epitope.

3. The isolated nucleic acid sequence coding for a modified VSV-G according to claim 1 or 2, wherein said at least one tumor antigen or fragment thereof is a neoantigen.

4. The isolated nucleic acid sequence coding for a modified VSV-G according to any one of claims 1 to 3, wherein said at least one antigen or fragment thereof is inserted into VSV-G at an amino acid position selected from the group consisting of positions 18, 51 , 55, 191 , 196, 217, 368 and C -terminal, and combinations thereof, wherein position numbering is with respect to vesicular stomatitis Indiana v irus (VSIV) glycoprotein amino acid sequence.

5. A vector containing a nucleic acid sequence according to any one of claims 1 to 4.

6. A dendritic cell population transfected by a nucleic acid sequence according to any one of claims 1 to 4 or a vector according to claim 5. 7. A modified vesicular stomatitis virus glycoprotein (VSV-G ) encoded by the isolated nucleic acid sequence according to any one of claims 1 to 4 comprising at least one tumor antigen or fragment thereof.

8. A composition comprising an isolated nucleic acid sequence according to any one of claims 1 to 4. a vector according to claim 5, a dendritic cell according to claim 6, or a modified VSV-G according to claim 7.

9. A vaccine comprising an isolated nucleic acid sequence according to any one of claims 1 to 4, a vector according to claim 5, a dendritic cell according to claim 6, or a modified VSV-G according to claim 7, and optionally at least one adjuvant.

10. A modified vesicular stomatitis virus glycoprotein (VSV-G ) comprising at least one antigen or fragment thereof, a nucleic acid sequence coding therefor, a vector containing a nucleic acid sequence coding therefor, a dendritic cell population transfected by a nucleic acid sequence coding therefor, or a vaccine comprising said modified VSV-G, nucleic acid sequence, vector or dendritic cell population and optionally at least one adjuvant, for use in preventing and/or treating a disease or condition in a subject in need thereof.

1 1. The vaccine for use according to claim 10, wherein said vaccine is a polynucleotide vaccine.

12. The vaccine for use according to claim 10, wherein said vaccine is a protein vaccine. 13. The modified VSV-G, nucleic acid sequence, vector, dendritic cel l population or vaccine for use according to any one of claims 10 to 12, wherein said disease is a cancer or an infectious disease.

14. The modified VSV-G, nucleic acid sequence, vector, dendritic cell population or vaccine for use according to any one of claims 10 to 13. wherein said modified VSV-G, nucleic acid sequence, vector, dendritic cell population or vaccine is to be administered to the subject by intramuscular injection, intradermal injection, intra tumoral, injection, peritumoral injection, gene gun, electroporation or sonoporation.

15. The modified VSV-G, nucleic acid sequence, vector, dendritic cel l population or vaccine for use according to any one of claims 10 to 14, wherein said modified

VSV-G, nucleic acid sequence, vector, dendritic cell population or vaccine is to be administered before, concomitantly or after one or more checkpoint blockade antibodies.

Description:
MODIFIED VSV-G AND VACCINES THEREOF

FIELD OF INVENTION

The present invention relates to the field of methods and related compositions for the preparation and administration of vaccines, such as nucleic acid-based vaccines, for the treatment of one or more diseases.

BACKGROUND OF INVENTION

Cancer remains one of the leading causes of death in the modern world. The standard treatments currently practiced in the clinic, including surgery, radiation, and chemotherapy, have shown limited success. These therapies are usually only effective against early stage localized tumors and rarely against later staged, metastatic malignancies, leading to frequent relapses. Furthermore, various agents used in radiation and chemotherapy are damaging to normal tissues, which may lead to prominent side effects.

For a few decades, vaccines have been applied as therapeutic strategies, harnessing the power of the immune system, to activate T cells against infected cells and cancers. For example, DNA vaccines are developed against various diseases including influenza and H IV- 1 (Uimer et al, 1993. Science. 259: 1745-1749; Wang et al, 1993. PNAS. 90:41 56- 41 60). These findings, along with the discovery and identification of cancer antigens, have propelled the investigation and dev elopment of DNA vaccines against cancer (Wang et al, 1999. Immunol. Rev. 170:85-100).

DNA vaccines are more cost effectiv e compared to other vaccines, such as recombinant protein, tumor cel ls, or v iral vectors. Recent adv ancements in molecular biology and recombinant technologies along with the increasing identification of tumor antigens provide the tools for plasmid gene manipulation. Genes in DNA vaccines can be designed to encode different antigens as well as various other i m m u nomod ul atory molecules to manipulate the resulting immune responses. Despite al l the adv antages. DNA vaccines hav e had l imited success in producing therapeutic effects against most cancers due to poor immunogenicity. Various strategies have been investigated to enhance the potency of DNA v accines. PI asm ids encoding antigens have been designed to promote antigen expression and presentation. Several components derived from bacteria or viruses are able to interact with the immune system, acting as adjuvants. For example, cholera or Clostridium difficile toxins have been shown to enhance the immunogenicity of mucosal antigens (Mohan et al, 2013. Indian. J. Med. Res. 138(5): 779 795 ). Unmethylated CpG motifs that are present on bacterial DNA have a strong stimulatory influence on the immune system and can be used to modulate the immunogenicity of DNA vaccines (Klinman et al., 1997. J. Immunol. 158(8): 3635 -9). More recently, the efficacy of cancer DNA vaccine was improved by the coadministration of a pi asm id encoding I I IV- 1 Gag v iral capsid protein (Lambricht et al., 2016. Mol. Ther. 24(9): 1686-96). Vesicular stomatitis virus glycoprotein (VSV-G) has also been used as an adjuvant to enhances DNA v accine potency (Marsac et al, 2002. J. Virol. 76(15):7544-7553; Mao et al, 2010. J. Virol. 84(5):2331-2339). In addition, VSV-G has been shown as having fusogenic properties that contribute to control tumor growth and mediate cancer cells killing (Bateman et al, 2000. Cancer Res. 60(6): 1492-1497; Bateman et al, 2002. Cancer Res. 62(22):6566-6578). The poor immunogenicity of DNA vaccines has driven a shift towards mRNA vaccine, another nucleic acid-based technology with interesting properties for immunization (Schlake et al, 2012. RNA Biol. 9(11): 1319-1330; Sahin et al, 2014. Nat Rev Drug Discov. 13(10):759-80; McNamara et al, 2015. J Immunol Res. 2015:794528 ). RNA vaccines are attractiv e because they retain the same appealing characteristics as DNA vaccines but also offer some additional benefits. Unlike DNA, RNA only needs to gain entry into the cytoplasm, where translation occurs, in order to transfect a cel l. Moreover, RNA cannot integrate into the genome and therefore has no oncogenic potential.

VSV-G is frequently used for pseudotyping because viruses bearing a VSV-G envelope are able to transduce an extensive range of cel l types. To alter the tropism of viral vectors. VSV-G mutants have been constructed by inserting tumor targeting ligands (Guibinga et al, 2004. Mol Ther. 9(l):76-84; Ammayappan et al, 2013. J. Virol 87(24): 13543-13555). Modified VSV-G was also obtained to construct virus-based vaccine carrying a neutralizing epitope from H IV- 1 intended to promote generation of neutralizing antibodies (Grigera et al, 1996. J. Virol 70(12):8492-8501 ; Schichubcr and Rose, 2004. J. Virol 78( 10):5079-5087). Finally, co-administration of a plasmid coding for an antigen and a plasmid encoding VSV-G has been shown to slow down cancer progression and to prolong survival (Mao et al, 2010. J Virol 84(5): 233 1 2339).

Here, the Applicant surprisingly demonstrates that a VSV-G protein comprising epitopes inserted into specific sites retains its immunogenic properties. Consistently, the Applicant shows that administration of a nucleic acid coding for such VSV-G protein generates a strong immune response against these epitopes. In particular. DNA immunization with a VSV-G sequence comprising tumoral epitopes leads to a significant effect on tumor growth. Therefore, the present invention relates to a nucleic acid encoding a vesicular stomatitis virus glycoprotein comprising at least one heterologous peptide, such as an antigen or a fragment thereof, and uses thereof for immunization.

SUMMARY The present invention relates to an isolated nucleic acid sequence coding for a modified vesicular stomatitis virus glycoprotein (VSV-G ), comprising at least one tumor antigen or fragment thereof.

In one embodiment, the at least one tumor antigen or fragment thereof comprises at least one epitope. In one embodiment, the at least one tumor antigen or fragment thereof is a neoantigen.

In one embodiment, the at least one antigen or fragment thereof is inserted into VSV-G at an amino acid position selected from the group consisting of positions 18, 51 , 55, 191 , 196, 2 1 7, 368 and C-terminai, and combinations thereof, wherein position numbering is with respect to vesicular stomatitis Indiana v irus (VSIV) glycoprotein amino acid sequence (SEQ ID NO: 1).

The present invention further relates to a vector comprising the nucleic acid sequence of the invention. The present invention further relates to a dendritic cell population transfected by the nucleic acid of the invention or by the vector of the invention.

The present invention further relates to a modified vesicular stomatitis v irus glycoprotein (VSV-G ) encoded by the isolated nucleic acid sequence of the invention.

The present invention further relates to a composition comprising the isolated nucleic acid sequence of the invention, the vector of the invention, the dendritic cel l of the invention or the modified VSV-G of the invention.

The present invention further relates to a vaccine comprising the isolated nucleic acid sequence of the invention, the vector of the invention, the dendritic cell of the invention or the modified VSV-G of the invention, and optionally at least one adjuvant. The present invention further relates to the modified VSV-G of the invention, the nucleic acid sequence coding therefor, the vector containing the nucleic acid sequence coding therefor, the dendritic cell population transfected by the nucleic acid sequence coding therefor, or the vaccine comprising said modified VSV-G, nucleic acid sequence, vector or dendritic cel l population and optionally at least one adj uvant. for use in preventing and/or treating a disease or condition in a subject in need thereof.

In one embodiment, the vaccine for use according to the present invention is a polynucleotide vaccine. In one embodiment, the vaccine for use according to the present invention is a protein vaccine.

In one embodiment, the disease or condition is a cancer or an infectious disease. In one embodiment, the modified VSV-G of the invention, the nucleic acid sequence coding therefor, the vector containing the nucleic acid sequence coding therefor, the dendritic cel l population transfected by the nucleic acid sequence coding therefor, or the vaccine comprising said modified VSV-G, nucleic acid sequence, vector or dendritic cel l population for use according to the present invention is to be administered to the subject by intramuscular injection, intradermal injection, intratumoral injection, peritumoral injection, gene gun, electroporation or sonoporation.

In one embodiment, the modified VSV-G of the invention, the nucleic acid sequence coding therefor, the vector containing the nucleic acid sequence coding therefor, the dendritic cell population transfected by the nucleic acid sequence coding therefor, or the vaccine comprising said modified VSV-G, nucleic acid sequence, vector or dendritic cel l population for use according to the present invention is to be administered before, concomitantly or after one or more checkpoint blockade antibodies.

DEFINITIONS

In the present invention, the following terms have the following meanings: - "Peptide" refers to a linear polymer of amino acids of less than 50 amino acids linked together by peptide bonds; a "polypeptide" refers to a linear polymer of at least 50 amino acids linked together by peptide bonds; and a "protein" specifical ly refers to a functional entity formed of one or more peptides or polypeptides, and optionally of non-polypeptides cofactors. - "Signal peptide", also called signal sequence, targeting signal, local ization signal, local ization sequence, transit peptide, leader sequence or leader peptide, refers to a peptide, present at the N-terminus or at the C-terminus of a protein, used to address it to a particular cel lular compartment, such as the nucleus, the endoplasmic reticulum, the Golgi, and the like. In one embodiment, the signal peptide of the invention comprises from 4 to 35 amino acids.

"Antigen" refers to any molecule that can initiate a cel lular and/or humoral immune response in a subject, leading to the stimulation of B and/or T lymphocytes. In one embodiment, an antigen is capable of being bound by an antibody or T cell receptor. The structural aspect of an antigen, e.g., three-dimensional conformation or modification (such as, e.g., phosphorylation), that gives rise to a biological response, is referred to herein as "epitope", "antigenic determinant" or "antigen epitopic fragment".

"Neoantigen" or "neoantigenie" refers to a class of tumor antigens that arises from one or several tumor-specific mutation(s) which alter(s) the amino acid sequence of genome encoded proteins.

The terms "epitope", "antigenic determinant" and "antigen epitopic fragment" can be used interchangeably. They refer to the part of an antigen that is recognized by the immune system, specifically by antibodies, B cells or T cells. Epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein (therefore referred to as "conformational epitope"). Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas conformational epitopes are typical ly lost on treatment with denaturing solvents. They define the minimum binding site for an antibody, B cell or T cell, and thus represent the target of specificity of an antibody, B cel l or T cell.

"T-cell epitope" refers to an epitope that can be bound by MHC molecules of class I or 11 in the form of a peptide-prcsenting MHC molecule or MHC complex and then, in this form, be recognized and bound by naive T cells, cytotoxic CD8 T cells or T helper CD4 cells. T cell epitopes may be presented by MHC class I for CD8 T cell recognition (therefore referred to as CD8 T cell epitopes), by MHC class II for CD4 T cell recognition (therefore referred to as CD4 T cell epitopes or helper T cell epitopes), or by both.

"Pharmaceutically acceptable excipient" refers to an excipient that does not produce an adverse, allergic or other untoward reaction when administered to an animal, preferably a human. It includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. For human administration, preparations should meet steril ity, pyrogenicity, general safety and purity standards as required by regulatory offices, such as, for example, FDA Office or EM A. "Immunogenic composition" is a composition that comprises an antigenic molecule where administration of the composition to a subject results in the development in the subject of a humoral and/or a cellular immune response to the antigenic molecule of interest. In one embodiment, the immunogenic composition may be introduced directly into a recipient subject, such as by injection, inhalation, oral, intranasal and mucosal administration.

"Vaccme" refers to any preparation comprising substance or group of substances meant to cause the immune system of a subject to respond to pathogens, such as bacteria or viruses, or to a tumor. Prophylactic vaccines are used to prevent a subject from ever having a particular disease or to only have a mild case of the disease. Such prophylactic vaccines usually comprise the pathogen responsible for the disease, either live and weakened or killed, or components thereof, purified or recombinant. Therapeutic vaccines are intended to treat specific diseases in a subject, in particular cancer. Such therapeutic anti-cancer vaccines comprise a tumor-antigen or tumor-antigens, eliciting an immune response directed against the tumor cells.

"Adjuvant" refers to a molecule that stimulates the immune response against an antigen and/or that modulates the immune response so as to obtain the expected response. In particular, the addition of adjuvants in vaccine formulations aims to improve, accelerate, shift and/or extend the specific immune response directed against the antigen(s) comprised in the vaccine formulations. The advantages of adjuvants include enhancing the immunogenicity of antigens, changing the nature of the immune response, reducing the amount of antigen(s) required to induce an effective immunization, reducing the frequency of booster immunizations, and enhancing the immune response in the elderly and the i m m u n o c o m p ro m i sed .

"Genetic adjuvant" refers to any biological ly active factor, such as a cytokine, an interleukin, a chemokine, a ligand, and optimally combinations thereof, which is expressed by a vector, and which, when administered with a DNA vaccine encoding an antigen, enhances the antigen- specific immune response. Desirable genetic adjuvants include, but are not limited to, DNA sequences encoding: GM-CSF, interferons ( IFNs) (for example, IFN-α, IFN-β and IFN-γ). interleukins (ILs) ( for example, I L-Ιβ. IL-2, IL-10, IL-12, IL-13), TNF-a, and combinations thereof. The genetic adjuvants may also be i m munost i mu I atory polypeptide from Para pox virus, such as a polypeptide of Parapox v irus strain D 1 701 or NZ2 or Parapox i m m unosti m u l a to ty polypeptides B2WL or PP30. Still other such biologically active factors that enhance the antigen-specific immune response may be readily selected by one of skill in the art, and a suitable pi asm id vector containing the same factors constructed by known techniques (for a review on genetic adjuvant for DNA vaccines, see Caiarota & Weiner, 2004. Expert Rev. Vaccines. 3:8 135-49; Caiarota & Weiner, 2004. Immunol. Rev. 199:84-99; Kutzier & Weiner, 2004. J. Clin. Invest. 14(9): 1241-4).

In one embodiment, the genetic adjuvant is not encoded by the polynucleotide or vector coding for a modified VSV-G according to the invention. In another embodiment, the genetic adjuvant is encoded by the polynucleotide or vector coding for a modified VSV-G according to the inv ention. According to this embodiment. the genetic adjuvant can be under the control of its own promoter; or the genetic adjuvant can be under the control of the same promoter as the modified VSV-G according to the invention, separated therefrom by an Internal Ribosomc Entry Site (IRES).

"Dendritic cells" refers to antigen-presenting cells of the immune system which present cytoplasmic branched projections called dendrites at certain development stages. Dendritic cells have the particular function to trigger the adaptive immune response induced in response to an antigen.

"Subject" refers to an animal, preferably a mammal, more preferably a human.

In one embodiment, a subject may be a mammal . Mammals include, but are not limited to, all primates (human and non-human ), cattle ( including cows), horses, pigs, sheep, goats, dogs, cats, and any other mammal which is awaiting the receipt of, or is receiving medical care or was/is/will be the object of a medical procedure, or is monitored for the development of a disease.

In one embodiment, a subject may be a "patient", i.e., a warm-blooded animal, more preferably a human, who which is awaiting the receipt of, or is receiv ing medical care or was/is/will be the object of a medical procedure, or is monitored for the development of a disease. In one embodiment, the subject is an adult (for example a subject above the age of 18). In another embodiment, the subject is a child (for example a subject below the age of 18). In one embodiment, the subject is a male. In another embodiment, the subject is a female.

"Treating" or '"treatment" or '"alleviation" refers to both therapeutic treatment and prophylactic or preventative measures; wherein the object is to prevent or slow down (lessen) the targeted pathologic condition or disorder, such as for example a cancer or an infection. Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented. A subject or mammal is successfully "treated" for a specific disease or condition, such as for example a cancer or an infection if, after receiving a therapeutic amount of modified VSV-G, polynucleotide, composition, or vaccine according to the present invention, the patient shows observable and/or measurable reduction in or absence of one or more of the following: reduction in the number of pathogenic cells: reduction in the percent of total cells that are pathogenic; and/or relief to some extent, one or more of the symptoms associated with the specific disease or condition; reduced morbidity and mortality, and improvement in quality of life issues. The above parameters for assessing successful treatment and improvement in the disease are readily measurable by routine procedures familiar to a physician.

"About" preceding a value means plus or less 10% of said value.

DETAILED DESCRIPTION 1. Modified VSV-G

The present invention relates to a nucleic acid encoding a vesicular stomatitis virus glycoprotein (VSV-G ) comprising at least one heterologous peptide. By "heterologous peptide" is meant a peptide which is not endogenous or native to a VSV-G protein, preferably to a VSV-G wild-type protein. Therefore, in one embodiment, the present invention relates to a nucleic acid encoding a modified vesicular stomatitis virus glycoprotein (VSV-G ) comprising at least one heterologous peptide. In one embodiment, the nucleic acid of the heterologous peptide is inserted into the nucleic acid of VSV-G. Within the meaning of the present invention, the term "modified VSV-G" amounts to the equivalent terms "chimeric VSV-G" and "mutant VSV-G". All terms are used interchangeably throughout the present specification. In one embodiment, a chimeric VSV-G is a VSV-G comprising at least one heterologous peptide. In one embodiment, a mutant VSV-G is an insertion mutant, wherein at least one heterologous peptide is inserted into VSV-G. In one embodiment, the terms "modified", "chimeric" and "mutant" are appl ied in reference to a VSV-G wild-type protein.

In one embodiment, the nucleic acid encoding a modified VSV-G of the invention is an isolated nucleic acid.

The present invention further relates to a modified vesicular stomatitis virus glycoprotein (VSV-G ) comprising at least one heterologous peptide.

In one embodiment, the modified VSV-G of the invention is a recombinant modified VSV-G.

In one embodiment, the modified VSV-G of the invention is an isolated modified

VSV-G. 1.1. VSV-G

Vesicular stomatitis viruses are constitutive members of the genus Vesiculovirus of the family Rhabdoviridae. Their genome accounts for a single molecule of negative-sense RNA, that encodes five major proteins: glycoprotein (G), polymerase or large protein (L), phosphoprotein. (P), matrix protein (M) and nucleoprotein (N). The glycoprotein of the vesicular stomatitis virus (VSV-G ) is a transmembrane protein that functions as the surface coat of the wild-type viral particles. Presently, nine vesicular stomatitis virus (VSV) strains are classified in the Vesiculovirus genus: vesicular stomatitis Indiana virus (VSIV), vesicular stomatitis Alagoas virus (VSAV), Carajas virus (CJSV), Chandipura virus (CHPV), Cocal virus (COCV), Isfahan virus (ISFV), Maraba virus (MARAV), vesicular stomatitis New Jersey virus (VSNJV) and Pity virus (PIRYV). Additionally, other stains are provisionally classified in the Vesiculovirus genus: Grass carp rhabdo virus, Be An 157575 virus (BcAn 157575), Botcke virus (BTKV), Calchaqui v irus (CQIV), Eel virus American (EVA), Gray Lodge virus (GLOV), Jurona virus (JURV), Klamath virus (KLAV), Kwatta virus (KWAV), La Joya virus (LJV), Malpais Spring virus (MSPV), Mount Elgon bat virus (MEBV), Perinet virus (PERV), Pike fry rhabdovirus (PFRV), Port on virus (PORV), Radi virus (RADIV), Spring viraemia of carp virus (SVCV), Tupaia virus (TUPV), Ulcerative disease rliabdovirus (UDRV) and Yug Bogdanovac virus (YBV).

Among these strains, the protein G genes show sequence similarities. The VSV-G protein presents a N -terminal ectodomain, a transmembrane region and a C -terminal cytoplasmic tail. It is exported to the cell surface via the trans Golgi network (endoplasmic reticulum and Golgi apparatus).

Sequences alignments using MUSCLE (Multiple Sequence Comparison by Log- Expectation) are shown in Table 1 below.

Table 1 - VSV-G sequence alignments using MUSCLE.

In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G ) is VSV-G from VSIV (VSIV-G). In one embodiment, VSV-G from VSIV comprises or consists of SEQ ID NO: 1 . In one embodiment, VSV-G is a variant of SEQ I D NO: 1 . In one embodiment, a variant of SEQ I D NO: 1 is a protein having a sequence identity of at least 30%, preferably of at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99% or more with SEQ ID NO: 1.

The term "identity" or "identical", when used in a relationship between the sequences of two or more polypeptides, refers to the degree of sequence reiatedness between polypeptides, as determined by the number of matches between strings of two or more amino acid residues. "Identity" measures the percent of identical matches between the smaller of two or more sequences with gap alignments ( if any) addressed by a particular mathematical model or computer program (i.e., "algorithms"). Identity of related polypeptides can be readily calculated by known methods. Such methods include, but are not limited to, those described in Arthur M. Lesk, Computational Molecular Biology: Sources and Methods for Sequence Analysis (New- York: Oxford University Press, 1988); Douglas W. Smith, Biocomputing: Informatics and Genome Projects (New-York: Academic Press, 1993 ); Hugh G. Griffin and Annette M. Griffin, Computer Analysis of Sequence Data, Part 1 (New Jersey: Humana Press, 1994); Gunnar von Hcinje, Sequence Analysis in Molecular Biology: Treasure Trove or Trivial Pursuit (Academic Press, 1987); Michael Gribskov and John Devereux, Sequence Analysis Primer (New York: M. Stockton Press, 1991); and Caril lo et al, 1988. SI AM J. Appl. Math. 48(5): 1073-1082. Preferred methods for determining identity are designed to give the largest match between the sequences tested. Methods of determining identity are described in publicly available computer programs. Preferred computer program methods for determining identity between two sequences include the GCG program package, including GAP (Devereux et al, 1984. Nucl. Acid. Res. 12(1 Pt l):387-395; Genetics Computer Group. University of Wisconsin Biotechnology Center. Madison, WI), BLASTP, BLASTN, TBLASTN and FASTA (Aitschul et al., 1990. J. Mol. Biol. 215(3):403-410). The B LAS I X program is publicly available from the National Center for Biotechnology Information (NCBI) and other sources (BLAST Manual, Aitschul et al. NCB/NLM/NIH Bethesda, Md. 20894; Aitschul et al, 1990. J. Mol. Biol. 215(3):403-410). The well-known Smith Waterman algorithm may also be used to determine identity.

In another embodiment, a variant of SEQ I D NO: 1 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 1.

As used herein, the term, "conservative amino acid substitution" is defined herein as an amino acid exchange within one of the fol low ing five groups:

I . Small aliphatic, non polar or slightly polar residues: Ala, Ser, Thr, Pro, Giy; I I. Polar, negatively charged residues and their amides: Asp, Asn, Glu, Gin;

I I I . Polar, positively charged residues: His, Arg, Lys;

IV. Large, aliphatic, nonpolar residues: Met, Leu, He, Val, Cys;

V. Large, aromatic residues: Phe, Tyr, Ti p. As used herein, "amino acids" are represented by their full name, their three letter code or their one letter code as well known in the art. Amino acid residues in peptides are abbreviated as follows: Phenylalanine is Phe or F; Leucine is Leu or L; Isoleueine is He or I; Methionine is Met or M; Val ine is Val or V; Serine is Ser or S; Proline is Pro or P; Threonine is Thr or T; Alanine is Ala or A; Tyrosine is Tyr or Y; Histidine is His or H; Glutamine is Gin or Q; Asparagine is Asn or N; Lysine is Lys or K; Aspartic Acid is Asp or D; Glutamic Acid is Glu or E; Cysteine is Cys or C; Tryptophan is Trp or W; Arginine is Arg or R; and Glycine is Gly or G.

As used herein, the term "amino acids" includes both natural and synthetic amino acids, and both D and L amino acids. "Standard amino acid" or "naturally occurring amino acid" means any of the twenty standard L-amino acids commonly found in naturally occurring peptides. "Nonstandard amino acid residue" means any amino acid, other than the standard amino acids, regardless of whether it is prepared synthetical ly or derived from a natural source. For example, naphtlylalaninc can be substituted for tryptophan to facilitate synthesis. Other synthetic amino acids that can be substituted include, but are not limited to, L-hydroxypropyi, L-3,4-dihydroxyphenylaianyl, a-amino acids such as L-a-hydroxyiysyi and D-a-methylalanyi, L-a-mcthyialanyl, β-amino acids, and isoquinolyl .

As used herein, "amino acid" also encompasses chemically modified amino acids, including, but not l imited to, salts, amino acid derivatives (such as amides), and substitutions. Amino acids contained within the polypeptides of the present invention, and particularly at the carboxy- or amino-terminus. can be modified by methylation, amidation, acetylation or substitution with other chemical groups which can change the polypeptide's circulating half-life without adversely affecting their activity. Additionally, a disul fide l inkage may be present or absent in the polypeptides of the invention. In another embodiment, a variant of SEQ ID NO: 1 is a protein wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not ) from the sequence of SEQ ID NO: 1 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3, 4, 5, 6, 7.8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not ) is/are added.

In one embodiment, the vesicular stomatitis virus glycoprotein (VSV ' -G) is VSV-G from VSNJV (VSNJV-G). In one embodiment, VSV-G from VSNJV comprises or consists of SEQ ID NO: 2.

In one embodiment, VSV-G is a variant of SEQ ID NO: 2. In one embodiment, a variant of SEQ ID NO: 2 is a protein having a sequence identity of at least 30%, preferably of at least 35, 40, 45, 50.55, 60, 65, 70, 75.80, 85, 90.95, 96, 97, 98, 99% or more with SEQ ID NO: 2. In another embodiment, a variant of SEQ ID NO: 2 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 2. In another embodiment, a variant of SEQ ID NO: 2 is a protein wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23.24 or 25 amino acids (either contiguous or not ) from the sequence of SEQ ID NO: 2 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15.16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not) is/are added. In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G) is VSV-G from CHPV (CHPV-G). In one embodiment, VSV-G from CHPV comprises or consists of SEQ ID NO: 3.

In one embodiment, VSV-G is a variant of SEQ ID NO: 3. In one embodiment, a variant of SEQ ID NO: 3 is a protein having a sequence identity of at least 30%, preferably of at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99% or more with SEQ ID NO: 3. In another embodiment, a variant of SEQ ID NO: 3 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 3. In another embodiment, a variant of SEQ ID NO: 3 is a protein wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not ) from the sequence of SEQ ID NO: 3 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3, 4, 5, 6.7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not ) is/are added.

In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G) is VSV-G from COCV (COCV-G). In one embodiment, VSV-G from COCV comprises or consists of SEQ ID NO: 4.

In one embodiment, VSV-G is a variant of SEQ ID NO: 4. In one embodiment, a variant of SEQ ID NO: 4 is a protein having a sequence identity of at least 30%, preferably of at least 35, 40, 45, 50, 55.60.65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99% or more with SEQ ID NO: 4. In another embodiment, a variant of SEQ ID NO: 4 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 4. In another embodiment, a variant of SEQ I D NO: 4 is a protein wherein 1, 2, 3, 4, 5, 6. 7, 8, 9, 10, 11, 12.13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not) from the sequence of SEQ I D NO: 4 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3.4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16.17, 18, 19, 20.21, 22, 23, 24 or 25 amino acids (either contiguous or not ) is/are added. In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G) is VSV-G from PIRYV (PIRYV-G). In one embodiment, VSV-G from PIRYV comprises or consists of SEQ ID NO: 5.

In one embodiment, VSV-G is a variant of SEQ ID NO: 5. In one embodiment, a variant of SEQ ID NO: 5 is a protein having a sequence identity of at least 30%, preferably of at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99% or more with SEQ ID NO: 5. In another embodiment, a variant of SEQ I D NO: 5 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 5. In another embodiment, a variant of SEQ ID NO: 5 is a protein wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not) from the sequence of SEQ ID NO: 5 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not) is/are added.

In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G) is VSV-G from ISFV (ISFV-G). In one embodiment, VSV-G from ISFV comprises or consists of SEQ ID NO: 6.

In one embodiment, VSV-G is a variant of SEQ ID NO: 6. In one embodiment, a variant of SEQ I D NO: 6 is a protein having a sequence identity of at least 30%, preferably of at least 35, 40, 45, 50, 55, 60, 65, 70, 75.80, 85, 90, 95, 96, 97, 98, 99% or more with SEQ ID NO: 6. In another embodiment, a variant of SEQ ID NO: 6 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 6. In another embodiment, a variant of SEQ ID NO: 6 is a protein wherein 1, 2.3, 4.5, 6. 7, 8, 9, 10, 11, 12, 13, 14, 15.16, 17, 18, 19, 20, 21, 22, 23.24 or 25 amino acids (either contiguous or not) from the sequence of SEQ ID NO: 6 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15.16, 17, 18, 19, 20, 21.22, 23, 24 or 25 amino acids (either contiguous or not) is/are added. In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G) is VSV-G from SVCV (SVCV-G). In one embodiment, VSV-G from SVCV comprises or consists of SEQ ID NO: 7.

In one embodiment, VSV-G is a variant of SEQ ID NO: 7. In one embodiment, a variant of SEQ ID NO: 7 is a protein having a sequence identity of at least 30%, preferably of at least 35.40, 45, 50, 55, 60, 65.70, 75, 80, 85, 90, 95, 96.97, 98, 99% or more with SEQ ID NO: 7. In another embodiment, a variant of SEQ ID NO: 7 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 7. In another embodiment, a variant of SEQ ID NO: 7 is a protein wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not) from the sequence of SEQ ID NO: 7 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (cither contiguous or not) is/are added.

In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G) is VSV-G from VSAV (VSAV-G). In one embodiment, VSV-G from VSAV comprises or consists of SEQ ID NO: 54.

In one embodiment, VSV-G is a variant of SEQ ID NO: 54. In one embodiment, a variant of SEQ ID NO: 54 is a protein having a sequence identity of at least 30%, preferably of at least 35, 40, 45, 50, 55, 60, 65, 70, 75.80, 85, 90, 95, 96, 97, 98, 99% or more with SEQ ID NO: 54. In another embodiment, a variant of SEQ I D NO: 54 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 54. In another embodiment, a variant of SEQ ID NO: 54 is a protein wherein 1, 2.3, 4, 5, 6. 7, 8, 9, 10, 11, 12.13.14, 15.16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not) from the sequence of SEQ ID NO: 54 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20.21, 22, 23, 24 or 25 amino acids (either contiguous or not ) is/are added. In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G) is VSV-G from CJSV (CJSV-G). In one embodiment, VSV-G from CJSV comprises or consists of SEQ ID NO: 55.

In one embodiment, VSV-G is a variant of SEQ ID NO: 55. In one embodiment, a variant of SEQ ID NO: 55 is a protein having a sequence identity of at least 30%, preferably of at least 35.40.45, 50, 55, 60, 65.70.75, 80, 85, 90, 95, 96, 97, 98, 99% or more with SEQ ID NO: 55. In another embodiment, a variant of SEQ ID NO: 55 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 55. In another embodiment, a variant of SEQ ID NO: 55 is a protein wherein 1, 2, 3, 4.5.6, 7, 8, 9, 10, 11, 12, 13, 14.15.16.17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not ) from the sequence of SEQ ID NO: 55 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3.4, 5, 6, 7, 8, 9, 10, 11, 12.13, 14, 15, 16, 17, 18, 19, 20, 21.22, 23, 24 or 25 amino acids (either contiguous or not ) is/are added.

In one embodiment, the vesicular stomatitis virus glycoprotein (VSV-G) is VSV-G from MARAV (MARAV-G). In one embodiment, VSV-G from MARAV comprises or consists of SEQ ID NO: 56.

In one embodiment, VSV-G is a variant of SEQ ID NO: 56. In one embodiment, a variant of SEQ ID NO: 56 is a protein having a sequence identity of at least 30%, preferably of at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99% or more with SEQ ID NO: 56. In another embodiment, a variant of SEQ ID NO: 56 comprises conservative amino acid substitutions as compared to the sequence of SEQ ID NO: 56. In another embodiment, a v ariant of SEQ ID NO: 56 is a protein wherein 1, 2, 3, 4.5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19.20.21.22, 23, 24 or 25 amino acids (either contiguous or not) from the sequence of SEQ ID NO: 56 is/are absent, or substituted by any amino acid, or wherein 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12.13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 amino acids (either contiguous or not) is/are added.

The modified VSV-G of the invention may comprise naturally standard amino acids or non-standard amino acids. Polypeptide mimetics include polypeptides having the following modifications:

i) polypeptides wherein one or more of the peptidyl -C(O)NR- linkages (bonds) have been replaced by a non-peptidyl linkage such as a -Clb-carbamate linkage (-CH20C(O)NR-), a phosphonate linkage, a -(Ί h-sulfonamidc (-CH 2 -S(O) 2 NR-) linkage, a urea (-NHC(O)NH-) linkage, a -CH 2 -secondary amine linkage, or with an alkylated peptidyl linkage (-C(O)NR-) wherein R is Ci-Ct alkyl: ii) polypeptides wherein the N-terminus is derivatized to a -NR R 1 group, to a -NRC(O)R group, to a -NRC(0 )OR group, to a -NRS(O) 2 R group, to a -NHC(O)NHR group where R and R 1 are hydrogen or C1-C4 alkyl with the proviso that R and R 1 are not both hydrogen;

iii) polypeptides wherein the C terminus is derivatized to -C(O)R 2 where R 2 is selected from the group consisting of C1-C4 alkoxy and -NR 3 R 4 , where R 3 and R 4 are independently selected from the group consisting of hydrogen and C1-C4 alky I .

In one embodiment of the invention, the modified VSV-G as described herein above are modified by means wel l-known in the art, for instance by the addition of one or more functional group such as a phosphate, acetate, lipid or carbohydrate group, and/or by the addition of one or more protecting group.

For example, the modified VSV-G can be modified by the addition of one or more functional groups such as phosphate, acetate, or various lipids and carbohydrates. The modified VSV-G of the invention can also exist as protein derivatives. The term "protein derivative" refers to compound having an amino group (— NH— ), and more particularly, a peptide bond. Modified VSV-G may be regarded as substituted amides. Like the amide group, the peptide bond shows a high degree of resonance stabilization. The C— N single bond in the peptide l inkage has typical ly about 40 percent double-bond character and the C=0 double bond about 40 percent single-bond character. "Protecting groups" are those groups that prevent undesirable reactions (such as proteolysis) involving unprotected functional groups. Specific examples of amino protecting groups include formyl; trifliioroacetyl; benzyloxycarbonyl; substituted benzyloxycarbonyl such as (ortho- or para-) c h I o ro benzyl o x y c a rbo n y I and (ortho- or para-) bromobenzyloxycarbonyi; and al iphatic oxycarbonyi such as t-butoxycarbonyl and t-amiloxycarbonyl . The carboxyl groups of amino acids can be protected through conversion into ester groups. The ester groups include benzyl esters, substituted benzyl esters such as methoxybenzyi ester; aikyi esters such as cyciohexyi ester, cycloheptyl ester or t-butyi ester. The guanidino moiety may be protected by nitro; or aryisulfonyi such as tosyl, methoxybenzensulfonyl or mesitylenesulfonyi, even though it does not need a protecting group. The protecting groups of imidazole include tosy, benzyl and dinitrophenyl. The indole group of tryptophan may be protected by formyl. or may not be protected.

In one embodiment, the modified VSV-G of the invention comprises a signal peptide at the N-terminus of said modified VSV-G. In one embodiment, the modified VSV-G of the invention comprises a signal peptide at the C-terminus of said modified VSV-G.

In one embodiment, the signal peptide comprises or consists of 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 1 5, 16, 1 7, 1 8, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 3 1 , 32, 33, 34 or

35 amino acid residues. In one embodiment, the signal peptide of the modified VSV-G of the invention comprises or consists of SEQ ID NO: 52 (MKCLLYLAFLFIGVNC).

In another embodiment, the signal peptide of the modified VSV-G of the invention comprises or consists of the Gaussia princeps iuciferase signal peptide with SEQ ID NO: 53 (MGVKVLF ALICIA VAEA). In another embodiment, the signal peptide of the modified VSV-G of the invention comprises of consists of any of the signal peptides disclosed in Kober et al, 2013. Biotechnol. Bioeng. 110: 1 164-1 173; Mori et al, 2015. J. Biosci. Bioeng. 120(5):518- 525; Stern et al, 2007. Trends Cell Mol Bio. 2: 1 - 1 7; Wen et al, 2011. Acta Biochim Biophys Sin. 43:96-102. These include, without limitation:

- the signal peptide of the Mus musculus Ig kappa light chain precursor (mutant A2) comprising or consisting of SEQ ID NO: 57 (MDMRAPAGIFGFLLVLFPGYRS); the signal peptide of the Homo sapiens serum albumin preproprotein comprising or consisting of SEQ I D NO: 58 (MKWVTFI SLLFLF S SAYS) ;

the signal peptide of the Homo sapiens immunoglobulin heavy chain comprising or consisting of SEQ I D NO: 59 (MDWTWRVFCLLAVTPGAHP);

the signal peptide of the Homo sapiens immunoglobul in light chain comprising or consisting of SEQ I D NO: 60 ( M AWSPLFLTL ITHCAGSWA );

the signal peptide of the Homo sapiens azurocidin preproprotein comprising or consisting of SEQ ID NO: 61 ( MTR LT V LA L LAG L L A SS R A ); the signal peptide of the Homo sapiens Cystatin-S precursor comprising or consisting of SEQ ID NO: 62 (MARPLCTLLLLMATLAGALA);

the signal peptide of the Pseudopleuronectes americanus trypsinogen 2 precursor comprising or consisting of SEQ ID NO: 63 (MRSLVFVLLIGAAFA);

- the signal peptide of the Mesobuthus martensii potassium channel blocker comprising or consisting of SEQ ID NO: 64 (MSRLFVFILIALFLSAIIDVMS); the signal peptide of the Conus leopardus a-conotoxin ipl .3 comprising or consisting of SEQ ID NO: 65 (MGMRMMFIMFMLVVLATTVVS);

the signal peptide of the Saccharomyces cerevisiae a-gaiactosidase (mutant m3) comprising or consisting of SEQ ID NO : 66 (MRAFLFLTACISLPGVFG);

the signal peptide of the Aspergillus niger cel lula.se comprising or consisting of SEQ ID NO: 67 (MKFQSTLLLAAAAGSALA);

the signal peptide of the Nepenthes gracilis aspartic proteinase nepenthesin-1 comprising or consisting of SEQ ID NO: 68 (MASSLYSFLLALSIVYIFVAPTHS); - the signal peptide of the Nepenthes rafflesiana acid chitinase comprising or consisting of SEQ ID NO: 69 (MKTHYSSAILPILTLFVFLSINPSHG);

the signal peptide of the M28 virus K28 prepro-toxin comprising or consisting of SEQ ID NO: 70 (MESVSSLFNIFSTIMVNYKSLVLALLSVSNLKYARG);

the signal peptide of the Zygosaccharomyces bailii killer toxin zygocin precursor comprising or consisting of SEQ I D NO: 7 l ( MKAAQI I T A S I VS L I . PIYTSA ); the signal peptide of the Vibrio cholerae 0139 cholera toxin comprising or consisting of SEQ ID NO: 72 (MIKLKFG VFFT VLL S SAY A) ;

the signal peptide of the Saccharomyces cerevisiae-derived adhesion subunit of a-aggiutinin comprising or consisting of SEQ I D NO: 73 (MQLLRCFSIFSVIASVLAQELTTICEQIPSPTLESTPYSLSTTTILANGK);

- the signal peptide of the Saccharomyces cerevisiae-derived exο-1 ,3-β glucanase comprising or consisting of SEQ I D NO: 74 (MLSLKTLLCTLLTVSSVLATPVPARDPSSIQFVHEENKKRYYDYDHGSLGE); the signal peptide of the Saccharomyces cerevisiae-derived mating pheromone α-factor comprising or consisting of SEQ I D NO: 75 (MRFPSIFTAVLFAASSALAAPVNTTTEDETAQIPAEAVIGYLDLEGDFDVAV LPFSNSTNN);

the signal peptide of the Saccharomyces cerevisiae-derived chit in trans-giycosyiase comprising or consisting of SEQ I D NO: 76 (MKVLDLLTVLSASSLLSTFAAAESTATADSTTAASSTASCNPLKTTGCTPDT

ALATSFSEDFSSSSK);

the signal peptide of the Saccharomyces cerevisiae-derived phosphoiipase B comprising or consisting of SEQ I D NO: 77 (MKLQSLLVSAAVLTSLTENVNAWSPNNSYVPANVTCDDDINLVREASGLSD NETE WLKKRD A YTKE) ;

the signal peptide of the Saccharomyces cerevisiae-derived cell wall protein related to glucanases comprising or consisting of SEQ ID NO: 78 (MKLSATTLTAASLIGYSTIVSALPYAADIDTGCTTTAHGSHQHKRAVAVTY VYET VT VDKNGQT VTPTSTEAS ST VASTTTLISES S VTKS S SKVAS S SE);

the signal peptide of the Saccharomyces cerevisiae-derived phosphoiipase B comprising or consisting of SEQ I D NO: 79 ( M Q L R N I L Q A S S L I S G L S I. A A D S S STTG DG Y A PS 11 PC P S D DT S 1. V R N A S G L ST A ETD WLKKRD A YTKEALHSFLSRATSNFSDTSLLSTLFSSNSSN);

the signal peptide of the Saccharomyces cerevisiae-derived exo- l ,3-() glucanase comprising or consisting of SEQ ID NO: 80 (MISPISFLSSLLCLTYLTSALPILPKREVVTRVHTASTTNVVTDFYSTTTE); the signal peptide of the Saccharomyces cerevisiae-denved cell wail-associated protein involved in export of acctylated sterols comprising or consisting of SEQ ID NO: 81 (MLEFPISVLLGCLVAVKAQTTFPNFESDVLNEHNKFRALHV DTAPLTWSDTLATYAQNYADQYDCSGVLTHSDGPYGENLALGYTDTGAVD AWYGEISKY );

the signal peptide of the Saccharomyces cerevisiae-derived aspartic protease comprising or consisting of SEQ I D NO: 82 (MKLKT VRS AVE S SLF AS Q VLGKIIP AANKRDDD SN SKF VKLPFHKL YGD SLE NVGSDKKPEVRLLKRADGYEEIIITNQQSFYSVDLE);

the signal peptide of the Saccharomyces cerevisiae-derived cell wail mannoprotein comprising or consisting of SEQ ID NO: 83 (MVKLTSIAAGVAAIAATASATTTLAQSDERVNLVELGVYVSDIRAHLAQYY SFQVAHPTETY);

the signal peptide of the Saccharomyces cerevisiae-derived cell wall mannoprotein comprising or consisting of SEQ ID NO: 84 (MVKLTSIVAGVAAIAAGVAAAPATTTLSPSDERVNLVELGVYVSDIRAHLA EYYMFQAAHPTETY);

the signal peptide of the Saccharomyces cerevisiae-derived mating pheromone a- factor comprising or consisting of SEQ ID NO: 85 (MQPITTASTQATQKDKSSEKKDNYIIKGLFWDPACVIA);

- the signal peptide of the Saccharomyces cerevisiae-derivcd sporu I at ion -spec i fi c exo- 1 ,3-b-glucanase comprising or consisting of SEQ I D NO: 86 (MVSFRGLTTLTLLFTKLVNCNPVSTKNRDSIQFIYKEKDSIYSAINNQAINEK); the signal peptide of the Homo sapiens chymotrypsinogen comprising or consisting of SEQ ID NO: 87 (MAFLWLLSCWALLGTTFG);

- the signal peptide of the Homo sapiens inteiieukin-2 comprising or consisting of SEQ ID NO: 88 (MQLLSCIALILALV);

the signal peptide of the Homo sapiens trypsinogen-2 comprising or consisting of SEQ ID NO: 89 ( MN I . LI . I LTFVAA A VA );

the signal peptide of the Metridia longa liiciferase comprising or consisting of SEQ ID NO: 90 (MDIKVVFTLVFSALVQA);

the signal peptide of the Oikopleura dioica Oikosm 1 comprising or consisting of SEQ ID NO: 91 (MLLLSALLLGLAHGYS);

the signal peptide of the Oikopleura dioica Oikosin 2A comprising or consisting of SEQ ID NO: 92 (MKLLASVLTIAAADYACC);

- the signal peptide of the Oikopleura dioica Oikosin 3 comprising or consisting of SEQ ID NO: 93 (MKISAGLLGVALGQNEGSAEA);

the signal peptide of the Oikopleura dioica Oikosin 4A comprising or consisting of SEQ ID NO: 94 (MKLFAALSAFSASVEA);

the signal peptide of the Oikopleura dioica Oikosin 5A comprising or consisting of SEQ ID NO: 95 (MKLLCSVLLGTVFG); the signal peptide of the Oikopleura dioica Oikosin 6 A comprising or consisting of

SEQ ID NO: 96 (MKISPLLVVTAVVG);

the signal peptide of the Oikopleura dioica Oikosin 7A comprising or consisting of SEQ ID NO: 97 (MKIAATFAALASATEWQG);

- the signal peptide of the Vargula hilgendorfii lucifera.se comprising or consisting of SEQ ID NO: 98 (MKIIILSVILAYCVTDNC);

the signal peptide of the Methanococcus jannaschii Slmj 1 comprising or consisting of SEQ ID NO: 99 (MAMSLKKIGAIAVGGAMVATALASGVAA);

the signal peptide of the Hepatitis C virus serotype lb E l protein comprising or consisting of SEQ ID NO : 100 (MGCSFSIFLLALLSCLTTPASA);

the signal peptide of the Hepatitis C virus serotype lb E2 protein comprising or consisting of SEQ I D NO: 101 (MVGNWAKVLIVMLLFAGVDG);

the signal peptide of the tissue plasminogen activator comprising or consisting of SEQ ID NO: 102 (MDAMKRGLCCVLLLCGAVFVDSVTG); and

- the signal peptide comprising or consisting of SEQ I D NO: 103 (MDAMKVLLLVFVSPSQVTG).

1.2. Peptide

1.2.1. Antigen

In one embodiment, the at least one heterologous peptide of the invention is an antigen or a fragment thereof. In one embodiment, a fragment of an antigen is an epitope.

In one embodiment, the antigen is a non-self antigen, i.e., the antigen is a foreign antigen. In another embodiment, the antigen is a protein of the host, i.e., is a self- antigen.

By "non-self antigen", "heterologous antigen" or "foreign antigen" is meant a molecule or molecules which is/are not endogenous or native to a subject which is exposed to it. The foreign antigen may el icit an immune response, e.g., a humoral and/or T cel l mediated response in the mammal. Examples of foreign antigen include, but are not l imited to. proteins ( including a modi fied protein such as a glycoprotein, a mucoprotein, etc. ), nucleic acids, carbohydrates, proteoglycans, l ipids, mucin molecules, immunogenic therapeutic agents ( including proteins such as antibodies, particularly antibodies comprising non-human amino acid residues, e.g. , rodent, ch imeric human ized, and primatized antibodies), toxins (optional ly conjugated to a targeting molecule such as an antibody, wherein the targeting molecule may also be immunogenic ), gene therapy viral vectors (such as retroviruses and adenoviruses), grafts ( including antigenic components of the graft to be transplanted into the heart, lung, liver, pancreas, kidney of graft recipient and neural graft components), infectious agents (such as bacteria and virus or other organism, e.g., protists), alioantigens (i.e., an antigen that occurs in some, but not in other members of the same species) such as differences in blood types, human lymphocyte antigens (HLA), platelet antigens, antigens expressed on transplanted organs, blood components, pregnancy ( Rh ), and hemophilic factors (e.g., Factor VTfi and Factor IX ). By "self-antigen" is meant an antigen that is normal ly expressed in a body. In one embodiment, self-antigen is expressed in an organ that is the target of an autoimmune disease. In one embodiment, the self -anti gen is expressed in a pancreas, thyroid, connective tissue, kidney, lung, digestive system or nervous system. In another embodiment, self-antigen is expressed on pancreatic β cells. Examples of self-antigen include, but are not limited to, antigenic peptides of insulin, insulin β, glutamic acid decarboxylase 1 (GAD1), glutamic acid decarboxylase 65 (GAD 65 ), HSP, thyroglobul in, nuclear proteins, acetylcholine receptor, collagen, thyroid stimulating hormone receptor (TSHR), ICA5 1 2( IA-2 ) and ΙΑ-2β (phogrin ), carboxypeptida.se H, ICA69, IC A 12, thyroid peroxidase, native DNA, myelin basic protein. myel in proteolipid protein, acetylcholine receptor components, histocompatibility antigens, antigens involved in graft rejection and altered peptide ligands. 1.2.1.1. Tumor antigens

In one embodiment, the antigen is a tumor antigen, or tumor-associated antigen.

In one embodiment, the antigen is a tumor-specific antigen (TSA). In another embodiment, the antigen is a tumor-associated antigen (TAA). In another embodiment, the antigen is a cancer-germline cancer testis antigen (CTA).

In one embodiment, the tumor from which the antigen is isolated or derived is any tumor or cancer, including, but not l imited to. melanomas, squamous cell carcinoma, breast cancers, head and neck carcinomas, thyroid carcinomas, soft tissue sarcomas, bone sarcomas, testicular cancers, prostatic cancers, ovarian cancers, bladder cancers, skin cancers, brain cancers, angiosarcomas, hemangiosarcomas, mast cell tumors, primary hepatic cancers, lung cancers, pancreatic cancers, gastrointestinal cancers, renal cell carcinomas, hematopoietic neoplasias and metastatic cancers thereof.

In one embodiment, the antigen may be any tumor antigen known from the person skilled in the art. For example, the antigen is selected from the tumor T cell antigen database TANTIG EN (hup ://c vc . d fc i . h arva rd . ed u/tadb/i ndex.html ).

Examples of tumor antigens comprise those described in Table 3 of Cheever et al., 2009. Clin Cancer Res. 15(17):5323-37, including, but not limited to, WT1 , MUC1 , LMP2, HPV E6 E7, EGFRvIII, HER-2/neu, Idiotype, MAGE A3, p53 nonmutant, NY- ESO- 1 , PSMA, GD2, CEA, Melan- A/MART 1 , Ras mutant, gp l OO, p53 mutant, Proteinases (PR 1 ), bcr-abl, Tyrosinase, Survivin, PSA, hTERT, Sarcoma translocation breakpoints, EphA2, PAP, ML-IAP, AFP, EpCAM, ERG (TMPRSS2 ETS fusion gene ), NA 1 7. PAX3, ALK, Androgen receptor, Cyclin Bl , Polysialic acid, MYCN, RhoC, TRP-2, GD3, Fucosyi GM1 , Mesothelin, PSCA, MAGE Al , sLe(a), (ΎΡ Ι Β 1 , PLAC1 , GM3, BORIS, Tn, GloboH, ETV6-AML, NY-BR- 1 , RGS5, SART3, STn, Carbonic anhydrase IX. PAX 5, OY-TES 1 , Sperm protein 1 7, LCK, HMWMAA, AKAP-4, SSX2, XAGE 1 , B7H3. Legumain, Tie 2, Page4, VEGFR2, MAD-CT-1 , FAP, PDGFR-β, MAD-CT-2 and Fos-related antigen 1 . Further examples of tumor antigens include, but are not limited to.707-AP (707 alanine proline), AFP (a-fetoprotein), ART-4 (adenocarcinoma antigen recognized by T cells), BAGE (B antigen, β-catcnin/m, β-catenin. mutated), Bcr-abl (breakpoint clusterrcgion- Abelson), CA-125 (cancer antigen 125, carcinoma antigen 125, or carbohydrate antigen 125, also known as mucin 16 or MUC16), CAMEL (CTL-recognized antigen on melanoma), CAP-1 (carcinoembryonic antigen peptide- 1), CASP-8 (caspase-8), CDC27m (cell-division-cycle 27 mutated), CDK4/m (eye line-dependent kinase 4 mutated), CEA (carcinoembryonic antigen), CT (cancertestis (antigen)), Cyp-B (cyclophilin B), DAM (differentiation antigen melanoma (the epitopes of DAM-6 and DAM- 10 are equivalent, but the gene sequences are different. DAM-6 is also called MAGE-B2 and DAM- 10 is also called MAGE-Bl)), EGF-R, ELF2M (elongation factor 2 mutated), ETA (Epithelial Tumor Antigen), ETV6-AML 1 ( Ets variant gene 6 acute myeloid leukemia 1 gene ETS), G250 (glycoprotein 250), GAGE (G antigen), GnT-V ( N-acety Igl ucosam i ny I transferase V), GplOO (glycoprotein lOOkD), HAGE (helicose antigen), HER-2/neu (human epidermal receptor-2 neurological), HLA-A* 0201-R 1701 (arginine (R) to isoleucine (I) exchange at residue 170 of the a-lielix of the a2-domain in the HLA-A2 gene), F1PV-E6 (human papilloma virus E6), F1PV-E7 (human papilloma virus E7), HSP70-2M (heat shock protein 70-2 mutated), HST-2 (human signet ring tumor-2), hTERT or hTRT (human teiomerase reverse transcriptase), iCE (intestinal carboxy lesterase ), KIAA0205 (name of the gene as it appears in databases), LAGE (1, antigen). LDLR/FUT (low density lipid receptor/GDP-L-fucose: β-D- galactosida.se 2-a-L-fucosy (transferase), MAGE (melanoma antigen, including but not limited to. MAGE3, MAGEA6, MAGEA10). MART- 1 /Mel an- A (melanomaantigen recognized by T cells- 1 /Melanoma antigen A), MCI R (melanocortin 1 receptor), Myosin/m (myosin mutated). MUC1 (mucin 1), MUM-1, -2, -3 ( m e I a n o m a u b i q u i to u s mutated 1, 2, 3), NA88-A (NA cDNA clone of patient M88), NY-ESO-1 (New York - esophageous 1), PI A, PI 5 (protein 15), p 190 minor bcr-abl (protein of 190 KD bcr-abl ), Pml/RARa (promyelocytic leukaemia retinoic acid receptor a), PRAME (preferentially expressed antigen of melanoma), PSA (prostate-specific antigen), PS MA (prostate- specific membrane antigen), RAGE (renal antigen), RUT or RU2 (renalubiquitous 1 or 2), SAGE (sarcoma antigen), SART-1 or SART-3 (squamous antigenrejecting tumor 1 or 3), TEL/AM 1.1 (translocation Ets-family leukemia/acute myeloidleukemia 1), TPI/m (triosephosphate isom erase mutated ), tyrosinase. TRP- 1 (tyrosinase relatedprotein 1 , or gp75 ), TR P-2 (tyrosinase related protein 2), TRP-2/INT2 (TRP- 2/intron2 ), WT 1 (Wilms' tumor gene), and mutant oncogenic forms of p53 (TP53), p73, ras, BRAF, A PC (adenomatous polyposis coli ), mye, VH L (von 1 l ippei's Lindau protein ), Rb- 1 (retinoblastoma), Rb-2, BRCA 1 , BRCA2, AR (androgen receptor), Smad4, M DR I , Flt-3.

In a preferred embodiment, the antigen of the invention is selected from the group consisting of P I A. TRP-2, gp 100, MART- 1 Melan-A. tyrosinase, MAGE ( including, but not limited to, MAGE3, MAGEA6, MAGE A 10), NY-ESO-1 , EGF-R. PSA, PSMA. CEA, F1ER2 neu, Muc- 1 . hTERT, TRP- 1 , BCR-abl, and mutant oncogenic forms of p53 (TP53 ), p73, ras, BRAF, A PC (adenomatous polyposis coli), myc, VHL (von Hippei's Lindau protein ), Rb- 1 (retinoblastoma), Rb-2, BRCA 1 , BRCA2, AR (androgen receptor), Smad4, MDRI and Flt-3.

According to the present invention, tumor antigens include any tumor antigen as described above, in addition to any other antigen that is associated with the risk of acquiring or development of cancer or for which an immune response against such antigen can have a therapeutic benefit against a cancer. For example, a cancer antigen could include, but is not limited to, a tumor antigen, a mammalian cell molecule harboring one or more mutated amino acids, a protein normally expressed pre- or neo- natally by mammalian cells, a protein whose expression is induced by insertion of an epidemiologic agent (e.g., virus), a protein whose expression is induced by gene translocation, and a protein whose expression is induced by mutation of regulatory sequences. Some of these antigens may also serve as antigens in other types of diseases (e.g., autoimmune disease). 1.2.1.2. Neoantigens

In another embodiment, the antigen of the invention is a neoantigen.

As used herein, the term "neoantigen" is a newly formed antigen that has not been previously recognized by the immune system. Neoantigens and, by extension, neoantigenic determinants (or neoepitopes), can be formed when a protein undergoes further modification within a biochemical pathway such as glycosylation. phosphorylation or proteolysis.

Neoantigens, tumor-specific or "somatic" mutations may be identified by comparing DNA isolated from tumor versus normal sources. Preferably, any suitable sequencing-by-synthesis platform can be used to identify mutations. Four major sequencing-by-synthesis platforms are currently available: the Genome Sequencers from Roche/454 Life Sciences, the HiSeq Analyzer from Illumina/Solexa, the SOLiD system from Applied BioSystems, and the Heliscope system from Helicos Biosciences. Sequencing-by-synthesis platforms have also been described by Pacific Biosciences and VisiGen Biotechnologies. Each of these platforms can be used in the methods of the invention.

1.2.1.3. Pathogen antigens

In one aspect of the invention, the antigen of the present invention is an antigen from a pathogen (including the whole pathogen ). In a particular embodiment, the antigen is from a pathogen that is associated with (e.g., causes or contributes to) an in fectious disease.

In one embodiment, the antigen of the invention is an infectious disease antigen.

In one embodiment, antigens from an infectious disease pathogen include antigens hav ing epitopes that are recognized by T cells, antigens hav ing epitopes that are recognized by B cells, antigens that are exclusively expressed by pathogens, and antigens that are expressed by pathogens and by other cells.

In one embodiment, pathogen antigens include whole cel ls and the entire pathogen organism, as well as lysates, extracts or other fractions thereof. In some embodiments, the antigens include organisms or portions thereof which may not be ordinarily considered to be pathogenic in a subject, but against which immunization is nonetheless desired. In one embodiment, antigens include one, two or a plurality of antigens that are representative of the substantially all of the antigens present in the infectious disease pathogen against which the vaccine is to be administered. In other embodiments, antigens from two or more different strains of the same pathogen or from di fferent pathogens can be used to increase the therapeutic efficacy and/or efficiency of the vaccine.

Pathogen antigens include, but are not l imited to, antigens that are expressed by a bacterium, a v irus, a parasite or a fungus.

I n a particular embodiment, pathogen antigens of the present invention incl ude antigens which cause a chronic infectious disease in an animal. In one embodiment, a pathogen antigen of the present invention includes an antigen from a virus.

Examples of viral antigens include, but are not l imited to, env, gag, rev, tar, tat, nucleocapsid proteins and reverse transcriptase from i mmunodeficiency viruses (e.g., H IV, FIV); H BV surface antigen and core antigen; HCV antigens; influenza n ucleocapsid proteins; parainfluenza nucleocapsid proteins; human papi l loma type 1 6 E6 and E7 proteins; Epstein-Barr virus LMP-1 , LMP-2 and EBNA-2; herpes LAA and glycoprotein D; as wel l as simi lar proteins from other viruses. Particularly preferred antigens for use in the present invention incl ude, but are not l imited to. H I V- 1 gag, H IV- 1 env, H I V- 1 pol, H I V- 1 tat, H I V- 1 nef. HbsAG, HbcAg, hepatitis c core antigen, H P V E6 and E7, HSV gl ycoprotein D, and Baci l l us anthracis protective antigen .

Examples of bacterial antigens include, but are not limited to, Borrelia afzelii antigens, Borrelia garinii antigens, Brucella abortus antigens, Campylobacter jejuni antigens, Helicobacter pylori antigens, Legionella pneumophila antigens, Leptospira biflexa antigens, Mycoplasma pneumoniae antigens, Yersinia enterocolitica antigens, Chlamydia pneumoniae antigens, Chlamydia trachomatis antigens, Chlamydia abortus antigens, Clostridium difficile antigens, Neisseria gonorrhoeae antigens, Toxoplasma gondii antigens, Bordetella pertussis Filamentous Hemagglutinin (FHA), and Bordetella pertussis toxin (Pertussis Toxin, PT). Examples of fungi and parasitic antigens include, but are not l imited to. Aspergillus fumigatus antigens and Candida albicans antigens.

In another embodiment, the antigen of the invention is capable of suppressing an undesired, or harmful, immune response. In one embodiment, the immune response is caused by allergens, autoimmune antigens, inflammatory agents, antigens involved in GVHD, certain cancers, septic shock antigens, and antigens involved in transplantation rejection. Such compounds include, but are not limited to, antihistamines, cyclosporin, corticosteroids, FK506, peptides corresponding to T cell receptors involved in the production of a harmful immune response, Fas iigands (i.e. , compounds that bind to the extracellular or the cytosolic domain of cellular Fas receptors, thereby inducing apoptosis), suitable MHC complexes presented in such a way as to effect tolenzation or anet'gy, T cell receptors, and autoimmune antigens, preferably in combination with a biological response modifier capable of enhancing or suppressing cellular and Or humoral immunity. Other antigens useful in the present inv ention and combinations of antigens will be apparent to those of skill in the art. The present invention is not restricted to the use of the antigens as described above.

1.2.2. Epitope

In one embodiment, the at least one heterologous peptide of the invention is an epitope derived from an antigen as described hereinabove. Accordingly, in one embodiment, a fragment of antigen of the invention comprises or consists of an epitope or "antigen epitopic fragment". In one embodiment, a fragment of antigen of the invention comprises or consists of more than one, i.e., at least two, three, four, five or more epitopes or "antigen epitopic fragments". In one embodiment, the epitope may be any epitope known from the person skilled in the art. For example, the epitope is selected from the immune epitope database and analysis resource (Vita et al., 2014. Nucleic Acids Res. 43( Database issee):D405-12; http://wvvw.iedb.org). In one embodiment, the epitope is derived from a non-self antigen or foreign antigen as described herein above. In another embodiment, the epitope is derived from a protein of the host, i.e. , the epitope is derived from a self-antigen as described herein above.

In another embodiment, the epitope is derived from a neoantigen as described hereinabove, i.e., the epitope is a neoantigenic determinant.

In one embodiment, the epitope is a conformational epitope, i.e., is composed of discontinuous sections of the antigen's amino acid sequence. In another embodiment, the epitope is a l inear epitope, i.e. , is composed of a continuous section of the antigen's amino acid sequence.

1.2.2.1. T cell epitopes

In one embodiment, the epitope is a T cel l epitope.

1.2.2.1.1. CD8 T cell epitopes

In one embodiment, the T cell epitope is a T cell epitope presented by MHC class I molecules. In one embodiment, the epitope is a CDS T cell epitope.

Examples of CD 8 T cell epitopes include, but are not limited to epitopes from, ovalbumin (with SEQ ID NO: 1 1), P I A (with SEQ I D NO: 13), MART- 1 (with SEQ ID NO: 14), gp l OO (with SEQ ID NO: 1 5 ), tyrosinase (with SEQ I D NO: 16), gp70 (with SEQ I D NO: 133 ) and TRP2 (with SEQ I D NO: 134).

1.2.2.1.2. CD4 T cell epitopes In one embodiment, the T cell epitope is a T cell epitope presented by MHC class II molecules. In one embodiment, the epitope is a CD4 T cell epitope (or helper T cell epitope).

Examples of CD4 T cell epitopes include, but are not limited to epitopes from, ovalbumin (e.g., with SEQ I D NO: 1 2 ), pan 11 LA DR-binding epitope (PADRE) (e.g., with SEQ ID NO: 17), VI L l (e.g., with SEQ ID NO: 18), tetanus toxoid epitope (TT) (e.g., with SEQ I D NO: 19), gp100 (e.g., with SEQ I D NO: 20), HMGB1 -derived i m m u nost i m u I atory peptide hp91 (e.g. , with SEQ I D NO: 2 1 ) and NY-ESO-1 (e.g. , with SEQ ID NO: 143).

Further examples of CD4 T cell epitopes include those disclosed in Hiemstra et a!., Proc Natl Acad Sci USA . 1 997 Sep 16; 94( 19): 1 03 13 103 1 8.

A limiting factor for targeting a specific CD4 response is the large number of polymorphisms in MHC class II genes. Therefore, in one embodiment, the CD4 T cell epitope may be a universal antigenic CD4 T cell epitope. As used herein, the term "universal antigenic CD4 T cel l epitope" refers to an epitope whose amino acid sequence is derived from at least one universal antigenic (or universal immunogenic or broad range ) CD4 T ceil epitope (also cal led an immunogenic carrier peptide ), which can be presented by multiple major histocompatibility complex (MHC) haplotypes and thereby activate helper CD4 T cells, which in turn, stimulate B cel l growth and differentiation. Examples of universal antigenic CD4 T cel l epitopes include, but are not limited to, pan H I. A DR-binding epitope (PADRE) (e.g., with SEQ ID NO: 17), natural tetanus sequences, epitopes derived from tetanus toxoid (TT) (e.g., with SEQ ID NO: 19) or diphtheria toxoid (DT), VI 1. 1 (e.g. , with SEQ I D NO: 18), HMGB l -derived immunostimulatory peptide hp91 (e.g. , with SEQ ID NO: 2 1 ), NY-ESO- 1 (e.g., with SEQ ID NO: 143 ), supermotif peptides from H IV- 1 (Gag 1 71 , Gag 294, Gag 298, Pol 303, Pol 335, Pol 596, Pol 71 1 , Pol 712, Pol 758, Pol 91 5, Pol 956), and epitopes from hemagglutinin influenza virus protein.

In another embodiment, the CD4 T cell epitope may be a foreign CD4 T cell epitope, i.e. , a foreign T cell epitope which binds an MHC class 11 molecule and can be presented on the surface of an antigen presenting cel l (A PC ) bound to the MHC class 11 molecule. 1.2.2.1.3. Tumoral epitopes

In one embodiment, the epitope is able to induce an immune response against tumor antigens. Accordingly, in one embodiment, the epitope is a tumoral epitope, preferably, the epitope is a tumoral CD4 T cell epitope or a tumoral CD8 T cell epitope. In one embodiment, the tumoral T cell epitope is a tumoral T cell epitope presented by MHC class I molecules. In another embodiment, the tumoral T cell epitope is a tumoral T cell epitope presented by MHC class II molecules.

Examples of tumoral T cell epitopes comprise those described in Vigneron et a!., 2013. Cancer Immun. 13 : 15, including, but not limited to, those recited in Table 2 below: Table 2 - Examples of tumoral T cell epitopes.

1.2.2.1.4. Pathogenic epitopes

In one embodiment, the epitope is able to induce an immune response against pathogenic antigens. In one embodiment, the epitope is a pathogenic epitope; preferably, the epitope is a pathogenic T cell epitope; more preferably, the epitope is a CD4 T cell epitope or a pathogenic CD8 T cell epitope.

In one embodiment, the pathogenic T cell epitope is a pathogenic T cell epitope presented by MHC class I molecules. In another embodiment, the pathogenic T cell epitope is a pathogenic T cell epitope presented by MHC class II molecules. In one embodiment, the epitope is a bacterial T cel l epitope, a viral T cel l epitope, a parasitic T cell epitope or a fungal T cell epitope.

Examples of pathogenic T cell epitopes comprise, but are not limited to, iisterioiysin O protein of Listeria monocytogenes {e.g., with SEQ ID NO: 144), Influenza Virus Nucleoprotein (e.g., with SEQ ID NO: 145), lymphocytic choriomeningitis v irus (LCMV) glycoprotein (GP) {e.g. , with SEQ ID NO: 146 or 147) and immunodominant adeno-associated virus 2 (AAV2) {e.g., with SEQ ID NO: 148).

In one embodiment, the pathogenic T cell epitope is a HIV T cel l epitope. Examples of HIV T cell epitopes include, without limitation, those discloses on Hiv.lanl .gov. (201 7).

HIV Molecular Immunology Database. [online] Available at: https://www.hiv.lanl.gov/content/immunoiogy/index.htmi and in Yusim K, Korber BTM, Brander C, Barouch D, De Boer R. Haynes BF, Koup R, Moore JP, Walker BD and Wat kins DI (Eds.). (2017). HIV Molecular Immunology 2016. Los Alamos, New Mexico: Los Alamos National Laboratory, Theoretical Biology and Biophysics.

In one embodiment, the pathogenic T cell epitope is a hepatitis virus T cell epitope, including without l imitation, hepatitis A virus ( HAV ), hepatitis B v irus (HBV ), hepatitis C virus (HCV), hepatitis D virus (HDV), hepatitis E virus (HEV), hepatitis F virus (HFV) or hepatitis G virus (HGV). 1.2.2.1. Long CD4/CD8 epitopes

In one embodiment, the fragment of antigen according to the present invention comprises at least two epitopes.

In one embodiment, the fragment of antigen according to the present invention comprises at least two T cell epitopes, both presented by MHC class I molecules. In one embodiment, the fragment of antigen according to the present invention comprises at least two CD8 T cell epitopes.

In one embodiment, the fragment of antigen according to the present invention comprises at least two T cell epitopes, both presented by MHC class II molecules. In one embodiment, the fragment of antigen according to the present invention comprises at least two CD4 T cell epitopes.

In a preferred embodiment, the fragment of antigen according to the present inv ention comprises at least two T cell epitopes, at least one of which is presented by MHC class I molecules and at least one of which is presented by MHC class 11 molecules. In one embodiment, the fragment of antigen according to the present invention comprises at least two T cell epitopes, at least one of which is a CD4 T cell epitope and at least one of which is a CD8 T cel l epitope.

Examples of fragments of antigen comprising at least two T cel l epitopes include, but are not limited to, gp l OO (with SEQ ID NO: 22) and P I A (with SEQ ID NO: 23). In one embodiment, the fragment of antigen according to the present invention comprises more than two epitopes. In one embodiment, the fragment of antigen according to the present inv ention comprises 3, 4, 5, 6, 7, 8, 9, 10 or more epitopes.

1.2.2.2. Two or more epitopes /VSV-G

In one embodiment, the modified VSV-G of the present invention comprises more than one heterologous peptide. In a particular embodiment, the modified VSV-G of the present inv ention comprises 2, 3, 4 or more heterologous peptides. In one embodiment. the modified VSV-G of the present invention comprises a combination of heterologous peptides.

In a particular embodiment, the modified VSV-G of the present invention comprises at least two heterologous peptides. In a preferred embodiment, the modified VSV-G of the present invention comprises at least two fragments of antigens. In a preferred embodiment, the modified VSV-G of the present invention comprises at least two epitopes. In one embodiment, the at least two heterologous peptides, preferably the at least two fragments of antigens or the at least two epitopes, are identical, i.e., the share the same amino acid sequence. In another embodiment, the at least two heterologous peptides, preferably the at least two fragments of antigens or at least two epitopes, are different, i.e., they don't share the same amino acid sequence.

In a more preferred embodiment, the modified VSV-G of the present invention comprises at least one CD8 T cell epitope and at least another epitope. In a more preferred embodiment, the modified VSV-G of the present invention comprises at least one CD4 T cell epitope and at least another epitope. In an even more preferred embodiment, the modified VSV-G of the present invention comprises at least one CD8 T cell epitope and at least one CD4 T cell epitope. In an even more preferred embodiment, the modified VSV-G of the present invention comprises at least two CD4 T cell epitopes, which may be identical or different, as defined hereinabove. In an even more preferred embodiment, the modified VSV-G of the present invention comprises at least two CD8 T cell epitopes, which may be identical or different, as defined hereinabove.

In one embodiment, the modified VSV-G of the present invention comprises at least one antigen or epitopic fragment thereof, preferably an epitope, and at least one CD4 T cell epitope. In a preferred embodiment, the modified VSV-G of the present invention comprises at least one epitope, preferably a T cell epitope, and at least one CD4 T cell epitope, preferably a universal antigenic CD4 T cell epitope. 1.2.3. Length

1.2.3.1. Global

In one embodiment, the heterologous peptide or fragment thereof has a length of 4 to 50 amino acids, preferably 5 to 25 amino acids, more preferably 6 to 20 amino acids, even more preferably 8 to 18 amino acids. in one embodiment, the heterologous peptide or fragment thereof has a length of 4 to 1 0 amino acids, 4 to 1 5 amino acids, 4 to 20 amino acids, 4 to 25 amino acids or 4 to 30 amino acids.

In another embodiment, the heterologous peptide or fragment thereof has a length of 5 to 1 0 amino acids, 5 to 1 5 amino acids, 5 to 20 amino acids, 5 to 25 amino acids or

5 to 30 amino acids.

In another embodiment, the heterologous peptide or fragment thereof has a length of

6 to 10 amino acids, 6 to 1 5 amino acids, 6 to 20 amino acids, 6 to 25 amino acids or

6 to 30 amino acids.

In another embodiment, the heterologous peptide or fragment thereof has a length of

7 to 10 amino acids, 7 to 1 5 amino acids. 7 to 20 amino acids, 7 to 25 amino acids or

7 to 30 amino acids.

In another embodiment, the heterologous peptide or fragment thereof has a length of

8 to 10 amino acids, 8 to 1 5 amino acids, 8 to 20 amino acids, 8 to 25 amino acids or 8 to 30 amino acids.

In another embodiment, the heterologous peptide or fragment thereof has a length of 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 1 5, 16. 1 7, 1 8, 19, 20, 2 1 , 22, 23, 24, 25 amino acids.

1.2.3.2. Length CD4 epitopes

In one embodiment, when the heterologous peptide or fragment thereof is a CD4 T cell epitope, said heterologous peptide or fragment thereof has a length of 5 to 25 amino acids, preferably 8 to 22 amino acids, more preferably 1 0 to 20 amino acids, even more preferably 1 2 to 18 amino acids.

In another embodiment, when the heterologous peptide or fragment thereof is a CD4 T cell epitope, said heterologous peptide or fragment thereof has a length of 5 to 10 amino acids, 5 to 1 5 amino acids, 5 to 18 amino acids, 5 to 20 amino acids or 5 to 25 amino acids.

In another embodiment, when the heterologous peptide or fragment thereof is a CD4 T cell epitope, said heterologous peptide or fragment thereof has a length of 6 to 10 amino acids, 6 to 1 5 amino acids, 6 to 18 amino acids, 6 to 20 amino acids or 6 to 25 amino acids.

In another embodiment, when the heterologous peptide or fragment thereof is a CD4 T cell epitope, said heterologous peptide or fragment thereof has a length of 7 to 10 amino acids, 7 to 1 5 amino acids. 7 to 18 amino acids, 7 to 20 amino acids or 7 to 25 amino acids. In another embodiment, when the heterologous peptide or fragment thereof is a CD4 T cel l epitope, said heterologous peptide or fragment thereof has a length of 8 to 10 amino acids, 8 to 15 amino acids, 8 to 18 amino acids, 8 to 20 amino acids or 8 to 25 amino acids.

In another embodiment, when the heterologous peptide or fragment thereof is a CD4 T cel l epitope, said heterologous peptide or fragment thereof has a length of 9 to 10 amino acids, 9 to 1 5 amino acids, 9 to 1 8 amino acids, 9 to 20 amino acids or 9 to 25 amino acids.

In another embodiment, when the heterologous peptide or fragment thereof is a CD4 T cell epitope, said heterologous peptide or fragment thereof has a length of 1 0 to 1 5 amino acids, 1 0 to 18 amino acids, 10 to 20 amino acids or 10 to 25 amino acids.

In another embodiment, w hen the heterologous peptide or fragment thereof is a CD4 T cell epitope, said heterologous peptide or fragment thereof has a length of 1 1 to 1 5 amino acids, 1 1 to 18 amino acids, 1 1 to 20 amino acids or 1 1 to 25 amino acids.

I n another embodiment, when the heterologous peptide or fragment thereof is a CD4 T cell epitope, said heterologous peptide or fragment thereof has a length of 1 2 to 1 5 amino acids, 1 2 to 18 amino acids, 1 2 to 20 amino acids or 1 2 to 25 amino acids.

In another embodiment, the heterologous peptide or fragment thereof has a length of 5, 6, 7, 8, 9, 1 0. 1 1 , 1 2, 13, 1 4, 1 5, 1 6, 1 7, 18, 1 9, 20, 2 1 , 22, 23, 24, 25 amino acids.

1.2.3.3. Length CD8 epitopes I n one embodiment, when the heterologous peptide or fragment thereof is a CDS T cell epitope, said heterologous peptide or fragment thereof has a length of 3 to 20 amino acids, preferably 3 to 1 5 amino acids, more preferably 5 to 1 3 amino acids, even more preferably 7 to 1 1 amino acids.

I n one embodiment, when the heterologous peptide or fragment thereof is a CDS T cel l epitope, said heterologous peptide or fragment thereof has a length of 3 to 9 amino acids, 3 to 1 1 amino acids, 3 to 15 amino acids, 3 to 18 amino acids or 3 to 20 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a CDS T cell epitope, said heterologous peptide or fragment thereof has a length of 4 to 9 amino acids, 4 to 1 1 amino acids, 4 to 1 5 amino acids, 4 to I S amino acids or 4 to 20 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a CD8 T cell epitope, said heterologous peptide or fragment thereof has a length of 5 to 9 amino acids, 5 to 1 1 amino acids, 5 to 15 amino acids, 5 to 18 amino acids or 5 to 20 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a CD8 T cell epitope, said heterologous peptide or fragment thereof has a length of 6 to 9 amino acids, 6 to 1 1 amino acids, 6 to 1 5 amino acids, 6 to 18 amino acids or 6 to 20 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a CD8 T cell epitope, said heterologous peptide or fragment thereof has a length of 7 to 9 amino acids, 7 to 1 1 amino acids, 7 to 1 5 amino acids, 7 to 18 amino acids or 7 to 20 amino acids.

In another embodiment, when the heterologous peptide or fragment thereof is a CD8 T cel l epitope, said heterologous peptide or fragment thereof has a length of 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 1 5, 16, 17, 18, 19, 20 amino acids. 1.2.3.4. Long CD4/CD8 epitopes in one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cell epitopes, said heterologous peptide or fragment thereof has a length of 20 to 1 00 amino acids, preferably 25 to 80 amino acids, more preferably 30 to 60 amino acids, even more preferably 30 to 45 amino acids, even more preferably 35 to 40 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cell epitopes, said heterologous peptide or fragment thereof has a length of 20 to 35 amino acids, 20 to 40 amino acids, 20 to 45 amino acids,

20 to 50 amino acids or 20 to 60 amino acids. In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cell epitopes, said heterologous peptide or fragment thereof has a length of 25 to 35 amino acids, 25 to 40 amino acids, 25 to 45 amino acids, 25 to 50 amino acids or 25 to 60 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cell epitopes, said heterologous peptide or fragment thereof has a length of 30 to 35 amino acids, 30 to 40 amino acids, 30 to 45 amino acids,

30 to 50 amino acids or 30 to 60 amino acids. In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cell epitopes, said heterologous peptide or fragment thereof has a length of 31 to 35 amino acids, 3 1 to 40 amino acids, 3 1 to 45 amino acids, 3 1 to 50 amino acids or 3 1 to 60 amino acids. In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cell epitopes, said heterologous peptide or fragment thereof has a length of 32 to 35 amino acids. 32 to 40 amino acids, 32 to 45 amino acids,

32 to 50 amino acids or 32 to 60 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cel l epitopes, said heterologous peptide or fragment thereof has a length of 33 to 35 amino acids, 33 to 40 amino acids, 33 to 45 amino acids,

33 to 50 amino acids or 33 to 60 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cel l epitopes, said heterologous peptide or fragment thereof has a length of 34 to 35 amino acids, 34 to 40 amino acids, 34 to 45 amino acids,

34 to 50 amino acids or 34 to 60 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T ceil epitopes, said heterologous peptide or fragment thereof has a length of 35 to 40 amino acids, 35 to 45 amino acids, 35 to 50 amino acids or 35 to 60 amino acids.

In another embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two T cell epitopes, said heterologous peptide or fragment thereof has a length of 20, 2 1 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 3 1 , 32, 33, 34. 35, 36, 37, 38, 39, 40, 4 1 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 5 1 , 52, 53, 54, 55 amino acids.

In one embodiment, when the heterologous peptide or fragment thereof is a fragment of antigen comprising two or more T cel l epitopes, said two or more T cel l epitopes are separated by a small amino acid sequence, herein referred as to "spacer". In one embodiment, a spacer comprises between 0 and 50 amino acids, preferably between 2 and 25 amino acids, more preferably betw een 5 and 20 amino acids, more preferably between 7 and 1 5 amino acids.

In one embodiment, a spacer comprises 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 1 5, 16, 17, 18, 1 9, 20, 2 1 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49 or 50 amino acids.

1.3. Insertion method

In one embodiment, peptides of the invention are inserted into VSV-G by recombinant DNA methods. Nucleic acids of the present invention can be readily prepared by the skilled person using techniques known in the art (for example, see Sambrook et al., Molecular Cloning: A Laboratory Manual. New-York: Cold Spring Harbor Laboratory Press, 1989; Ausubei et al., Short Protocols in Molecular Biology. New- York: John Wiley and Sons, 1992 ). For example, the modified sequence of VSV-G is obtained by arti ficial gene synthesis. This al lows an adaptation of codon usage for a better expression of the sequence (Angov et al., 2011. Biotechnol. J. 6(6): 650-659). The optimized sequence is then subcioned into an expression vector. In another example, a synthetic nucleic acid sequence or vector containing a nucleic acid sequence encoding a peptide to be inserted into VSV-G is specifically designed to include restriction endonuciease sites matched to a specified endonuclease-cut nucleic acid sequence encoding VSV-G or to a specified endonuclease-cut nucleic acid sequence previously added into the VSV-G sequence. Where a desirable VSV-G insertion site contains a single, unique restriction endonuciease site, the peptide's nucleic acid sequence is preferably engineered to include matched restriction sites at both ends of the sequence. In this manner, the sequence encoding the peptide is inserted into the VSV-G sequence w ithout removal of any VSV-G-encoding nucleotides. Care is taken to match the peptide-encoding nucleic acid sequence to be inserted with the reading frame of the VSV-G sequence so that normal expression of the encoded VSV-G with the encoded peptide of interest is achieved. Modified VSV-G can also result from Gibson assembly cloning where multiple DNA fragments can be assembled, regardless of fragment length or end compatibil ity. In one embodiment, at least one heterologous peptide or antigen fragment is inserted into VSV-G at any VSV-G permissive insertion site, preferably at a VSV-G permissive epitope insertion site.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G, in highly variable regions. In one embodiment, said highly variable regions are defined on the basis of sequence alignments of VSV-G from various strains. These highly variable regions can undergo sequence modifications without affecting the stabil ity and/or function of the protein. In one embodiment, said highly variable regions are regions which are exposed at the surface of the protein. In one embodiment, said highly variable regions are regions comprised in exposed turns, including a-turns, β- turns, γ-turns, δ-turns, π- turns, ω-turns, loops and/or hairpins. Suitable regions for inserting the at least one heterologous peptide or fragment thereof can be determined by methods known from the skilled person, using for example protein structure prediction software and/or loop modeling software. In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G, at its C-tcrminal extremity, i.e. , after the last amino acid residue of its sequence.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from vesicular stomatitis Indiana virus (VSIV ) (SEQ ID NO: 1) within region(s) selected from the group consisting of:

Region 1 : amino acid residues 1 to 19 of SEQ ID NO: 1 ;

Region 2: amino acid residues 42 to 61 of SEQ I D NO: 1 ;

Region 3 : amino acid residues 184 to 233 of SEQ I D NO: 1 ;

Region 4: amino acid residues 253 to 268 of SEQ I D NO: 1 ;

- Region 5 : amino acid residues 270 to 289 of SEQ I D NO: 1 :

Region 6: amino acid residues 362 to 372 of SEQ I D NO: 1 ; and

Region 7: after amino acid residue 5 1 1 , i.e. , at the C-terminal extremity of

SEQ ID NO: 1. In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from vesicular stomatitis New Jersey virus (VSNJV) (SEQ I D NO: 2) within region(s) selected from the group consisting of:

- Region 1 : amino acid residues I to 19 of SEQ I D NO: 2;

- Region 2: amino acid residues 42 to 6 1 of SEQ I D NO: 2;

- Region 3 : amino acid residues 1 84 to 233 of SEQ I D NO: 2;

- Region 4: amino acid residues 253 to 272 of SEQ I D NO: 2;

- Region 5 : amino acid residues 274 to 293 of SEQ I D NO: 2;

- Region 6: amino acid residues 366 to 376 of SEQ I D NO: 2; and

- Region 7: after amino acid residue 517, i.e. , at the C-terminal extremity of SEQ ID NO:2.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from Chandipura v irus (CHPV) (SEQ ID NO: 3) within region(s) selected from the group consisting of:

- Region 1 : amino acid residues 1 to 24 of SEQ ID NO: 3;

- Region 2: amino acid residues 47 to 66 of SEQ I D NO: 3;

- Region 3 : amino acid residues 189 to 237 of SEQ I D NO: 3;

- Region 4: amino acid residues 257 to 276 of SEQ I D NO: 3;

- Region 5 : amino acid residues 278 to 297 of SEQ I D NO: 3;

- Region 6: amino acid residues 370 to 381 of SEQ I D NO: 3; and

Region 7: after amino acid residue 530, i.e. , at the C-terminal extremity of SEQ ID NO:3.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from Cocal v irus (COCV) (SEQ I D NO: 4 ) w ithin region(s) selected from the group consisting of:

- Region 1 : amino acid residues 1 to 20 of SEQ ID NO: 4;

- Region 2: amino acid residues 43 to 62 of SEQ ID NO: 4;

- Region 3 : amino acid residues 185 to 234 of SEQ I D NO: 4;

- Region 4: amino acid residues 254 to 269 of SEQ I D NO: 4;

- Region 5 : amino acid residues 271 to 290 of SEQ I D NO: 4; Region 6: amino acid residues 363 to 373 of SEQ ID NO: 4; and

Region 7: after amino acid residue 512, i.e. , at the C -terminal extremity of

SEQ ID NO:4.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from Piry virus (PI RYV ) (SEQ I D NO: 5) within region(s) selected from the group consisting of:

Region 1 : amino acid residues 1 to 2 1 of SEQ I D NO: 5;

Region 2 : amino acid residues 44 to 63 of SEQ I D NO: 5;

Region 3 : amino acid residues 186 to 233 of SEQ I D NO: 5;

- Region 4: amino acid residues 253 to 272 of SEQ ID NO: 5;

Region 5 : amino acid residues 274 to 293 of SEQ I D NO: 5;

Region 6: amino acid residues 366 to 377 of SEQ I D NO: 5; and

Region 7: after amino acid residue 529, i.e. , at the C -terminal extremity of

SEQ I D NO:5. In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from Isfahan virus ( ISFV ) (SEQ I D NO: 6) within rcgion(s) selected from the group consisting of:

Region 1 : amino acid residues 1 to 23 of SEQ I D NO: 6;

Region 2: amino acid residues 46 to 65 of SEQ I D NO: 6;

- Region 3 : amino acid residues 188 to 236 of SEQ I D NO: 6;

Region 4: amino acid residues 256 to 275 of SEQ I D NO: 6;

Region 5 : amino acid residues 277 to 296 of SEQ I D NO: 6;

Region 6: amino acid residues 369 to 380 of SEQ I D NO: 6; and

Region 7: after amino acid residue 523, i.e. , at the ('-terminal extremity of SEQ I D NO:6.

In one embodiment, the at least one heterologous pept ide or fragment thereof is inserted into VSV-G from Spring viraemia of carp virus (SVCV) (SEQ I D NO: 7) within region(s) selected from the group consisting of:

Region 1 : amino acid residues 1 to 20 of SEQ I D NO: 7;

- Region 2: amino acid residues 44 to 63 of SEQ I D NO: 7; - Region 3 : amino acid residues 1 86 to 235 of SEQ ID NO: 7;

- Region 4: amino acid residues 254 to 270 of SEQ ID NO: 7;

- Region 5 : amino acid residues 272 to 291 of SEQ I D NO: 7;

- Region 6: amino acid residues 364 to 374 of SEQ I D NO: 7; and

- Region 7: after amino acid residue 509, i.e. , at the C-terminal extremity of SEQ I D NO:7.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from Alagoas virus (VSAV ) (SEQ I D NO: 54 ) within rcgion(s ) selected from the group consisting of:

- Region 1 : amino acid residues 1 to 20 of SEQ ID NO: 54;

- Region 2: amino acid residues 43 to 62 of SEQ I D NO: 54;

- Region 3: amino acid residues 185 to 234 of SEQ ID NO: 54:

- Region 4: amino acid residues 254 to 269 of SEQ I D NO: 54;

- Region 5 : amino acid residues 271 to 290 of SEQ I D NO: 54;

- Region 6: amino acid residues 363 to 373 of SEQ I D NO: 54; and

Region 7: after amino acid residue 51 1 , i.e. , at the C-terminal extremity of SEQ I D NO:54.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from Carajas virus (CJSV) (SEQ ID NO: 55 ) within rcgion(s) selected from the group consisting of:

- Region 1 : amino acid residues 1 to 24 of SEQ I D NO: 55;

- Region 2: amino acid residues 47 to 66 of SEQ I D NO: 55;

- Region 3 : amino acid residues 189 to 238 of SEQ I D NO: 55;

- Region 4 : amino acid residues 258 to 277 of SEQ I D NO: 55;

- Region 5 : amino acid residues 279 to 298 of SEQ I D NO: 55;

- Region 6: amino acid residues 371 to 381 of SEQ I D NO: 55; and

Region 7: after amino acid residue 523, i.e. , at the C-terminal extremity of SEQ ID NO: 55. In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from Maraba virus (MARAV) (SEQ ID NO: 56) within region(s) selected from the group consisting of:

Region 1 : amino acid residues 1 to 19 of SEQ I D NO: 56;

- Region 2 : amino acid residues 42 to 61 of SEQ I D NO: 56;

- Region 3 : amino acid residues 184 to 233 of SEQ I D NO: 56;

Region 4: amino acid residues 253 to 268 of SEQ I D NO: 56;

- Region 5 : amino acid residues 270 to 289 of SEQ I D NO: 56;

Region 6: amino acid residues 362 to 372 of SEQ I D NO: 56; and

- Region 7: after amino acid residue 512, i.e. , at the C-terminal extremity of SEQ I D NO:56.

In another embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from a virus strain classified or provisionally classified in the Vesiculovirus genus such as Chandipura virus (CHPV), Cocai virus (COCV), Indiana virus (VSIV), Isfahan virus (ISFV), New Jersey virus (VSNJV ), Pity virus (PIRYV ), Grass carp rhabdov irus, BeAn 1 57575 virus ( Be An 1 57575 ), Boteke virus (BTKV), Calchaqui virus (CQIV), Eel virus American (EVA), Gray Lodge virus (GLOV), Jurona virus (JURV), Klamath virus (K1.AV ). Kvvatta virus (KWAV), La Joya virus (LJV), Malpais Spring virus (MSPV), Mount Elgon bat virus (MEBV), Peri net virus (PERV), Pike fry rhabdovirus (PFRV), Porton virus (PORV), Radi virus ( RADIV), Spring viraemia of carp vims (SVCV), Tupaia virus (TUPV), Ulcerative disease rhabdovirus (UDRV) and Yug Bogdanovac virus (YBV). The at least one heterologous peptide or fragment thereof is inserted in positions that are readily selected by the one skilled in the art. As used hereafter, and unless indicated otherwise, the positions into which the heterologous peptide(s) is/are inserted are defined by the amino acid residue directly after the insertion site. In other words, insertion position 18 corresponds to the region between amino acid residues 1 7 and 18.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from vesicular stomatitis Indiana virus (VSIV ) (SEQ ID NO: 1 ) at a VSV-G amino acid position selected from the group comprising or consisting of positions 13, 14, 15, 16, 1 7, 18, 19, 20, 21 , 22, 23, 46 ,47, 48, 49, 50, 5 1 , 52, 53. 54, 55, 56, 57, 58, 59, 60, 186, 187, 188, 189, 190, 191 , 192, 193, 1 94, 195, 196, 197, 198, 199, 200, 201 . 202, 2 12, 2 1 3, 2 14, 2 15, 2 16, 2 17, 218, 2 19, 220, 22 1 , 222. 363, 364, 365, 366, 367, 368, 369, 370, 371 , 372, 373 and C -terminal extremity, and combinations thereof.

In one embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G from vesicular stomatitis Indiana virus (VSIV) (SEQ ID NO: 1) at a VS V -G amino acid position selected from the group comprising or consisting of positions 1 8, 5 1 , 55, 1 91 , 1 96, 2 17, 368 and C -terminal extremity, and combinations thereof.

In a preferred embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G amino acid positions 18 and/or 191 with respect to SEQ ID NO: 1 . In other words, in a preferred embodiment, the nucleic acid sequence encoding the heterologous peptide is inserted into the nucleic acid sequence encoding VSV-G such that the expressed modified VSV-G will include the heterologous peptide inserted at VSV-G amino acid position 18 and/or 191 with respect to SEQ I D NO: 1 .

In another preferred embodiment, the at least one heterologous peptide or fragment thereof is inserted into VSV-G at the C -terminal extremity of VSV-G. In a particular embodiment, more than one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G amino acid positions 18 with respect to SEQ I D NO: 1 . In a particular embodiment, more than one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G amino acid positions 5 1 with respect to SEQ I D NO: 1. In a particular embodiment, more than one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G amino acid positions 55 with respect to SEQ I D NO: 1 . In a particular embodiment, more than one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G amino acid positions 191 with respect to SEQ ID NO: 1. In a particular embodiment, more than one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G amino acid positions 196 with respect to SEQ ID NO: 1. In a particular embodiment, more than one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G amino acid positions 217 with respect to SEQ ID NO: 1. In a particular embodiment, more than one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G amino acid positions 368 with respect to SEQ ID NO: 1 . In a particular embodiment, more than one heterologous peptide or fragment thereof is inserted into VSV-G at VSV-G C-terminal extremity.

Techniques to determine amino acid positions in a VSV-G other than VSV-G from vesicular stomatitis Indiana virus (VSIV ) ( SEQ I D NO: 1) into which at least one heterologous peptide or fragment thereof can be inserted are well-known in the art. In one embodiment, multiple heterologous peptides may be inserted into VSV-G, e.g., at more than one site in VSV-G, preferably at two or more sites. In one embodiment, the modified VSV-G of the invention comprises multiple copies of the same heterologous peptide. In another embodiment, the modified VSV-G of the invention comprises one copy of different heterologous peptides. In still another embodiment, the modified VSV-G of the invention comprises one or more copies of different heterologous peptides.

2. Polynucleotide

A second aspect of the inv ention relates to a polynucleotide, or a nucleic acid sequence, coding for a modified VSV-G according to the inv ention. A "coding sequence" or a sequence "encoding" a modified VSV-G, is a nucleotide sequence that, when expressed, results in the production of that modified VSV-G, i.e., the nucleotide sequence encodes an amino acid sequence for that modified VSV-G. In one embodiment, the coding sequence includes a start codon (usually ATG) and a stop codon. In one embodiment, the polynucleotide or nucleic acid sequence is an isolated polynucleotide or an isolated nucleic acid sequence. In one embodiment, polynucleotides or nucleic acids of the invention may be obtained by conventional methods well known to those skilled in the art. Typically, said polynucleotide or nucleic acid is a DNA or RNA molecule, which may be included in a suitable vector, such as a piasmid, cosmid, episome, artificial chromosome, phage or viral vector.

In one embodiment, the polynucleotide or nucleic acid of the invention is a DNA molecule. In another embodiment, the polynucleotide or nucleic acid of the invention is a RNA molecule. In a particular embodiment, the polynucleotide or nucleic acid of the invention is a mRNA molecule. In one embodiment, the codon usage bias of the polynucleotide or nucleic acid of the invention is optimized. As used herein, the term "codon usage bias" refers to the high- frequency preferential use of a particular codon (as opposed to other, synonymous codons) coding for an amino acid within a giv en organism, tissue or cell. A codon usage bias may be expressed as a quantitative measurement of the rate at which a particular codon is used in the genome of a particular organism, tissue or cell, for example, when compared to other codons that encode the same amino acid. Various methods are known to those of skill in the art for determining codon usage bias. In some embodiments, codon usage bias may be determined by the codon adaptation index (CAI) method, which is essentially a measurement of the distance of a gene's codon usage to the codon usage of a predefined set of highly-expressed genes (Sharp and Li, 1987. Nucleic Acids Res. 15: 1281-95). Alternative methods for determining a codon usage bias include MILC (Mea.su re Independent of Length and Composition) (Supek and Vlahovicek, 2005. BMC Bioinformatics . 6: 182) and relative synonymous codon usage (RSCU), which is the observ ed frequency of a particular codon divided by the frequency expected from equal usage of all the synonymous codons for that amino acid (Sharp et al, 1986. Nucleic Acids Res. 14:5125-43). RSCU values close to 1.0 indicate a lack of bias for the particular codon, whereas departure from 1.0 reflects codon usage bias.

In one embodiment, one or more polynucleotides are inserted ex vivo into dendritic cells, such that one or more selected heterologous peptides, preferably antigens, are presented in effective amounts on the surface of the dendritic cells. By "effective amount" is meant that presentation is sufficient to enable the dendritic cells to provoke an immune response.

Techniques for nucleic acid manipulation are well known. Reagents useful in applying such techniques, such as restriction enzymes and the like, are widely known in the art and commercially available from a number of vendors.

Polynucleotides encoding the desired heterologous peptides, preferably antigens, for presentation in the dendritic cells are preferably recombinant expression vectors in which high levels of expression may occur. In one embodiment, the vectors may also contain polynucleotide sequences encoding selected class I and class 11 MHC molecules, costimulation and other immunoregulatory molecules, ABC transporter proteins, including the TAP l and TAP2 proteins. In one embodiment, the vectors may also contain at least one positive marker that enables the selection of dendritic cells carrying the inserted nucleic acids. Expression of the polynucleotide of interest after transfection into dendritic cells may be confirmed by immunoassays or biological assays. For example, expression of introduced polynucleotides into cells may be confirmed by detecting the binding to the cells of labeled antibodies specific for the antigens of interest using assays well known in the art such as FACS (Fluorescent Activated Cell Sorting) or ELISA (enzyme-linked i m m u noa bsorben t assay) or by simply by staining (e.g. , with β-gal ) and determining cell counts.

T cell activ ation may be detected by various known methods, including measuring changes in the proliferation of T cel ls, killing of target cells, tetramer staining, and secretion of certain regulatory factors, such as lymphokines, expression of mRNA of certain immunoregulatory molecules, or a combination of these. 3. Vector

Therefore, a further object of the present invention relates to a vector or a pi asm id in which a polynucleotide of the invention is associated with suitable elements for controlling transcription (in particular promoter, enhancer and, optional ly, terminator) and, optionally translation.

The present invention also relates to the recombinant vectors into which a polynucleotide in accordance with the invention is inserted. These recombinant vectors may, for example, be cloning vectors, or expression vectors.

The terms "vector", "cloning vector" and "expression vector" mean the vehicle by which the polynucleotide of the invention may be introduced into a host cell, so as to transform the host and promote expression (e.g., transcription and translation) of the polynucleotide.

Any expression vector for animal cell may be used, as long as a polynucleotide encoding a modified VSV-G of the inv ention can be inserted and expressed. Examples of suitable v ectors include, but are not l imited to, pVAX2, pAGE107, pAGE103, pHSG274, pKCR, pSGl β d2-4 and the like.

Other examples of plasmids include replicating plasmids comprising an origin of repl ication, or integrative plasmids, such as for instance pUC, pcDNA, pBR, and the like. In one embodiment, the vector is devoid of antibiotic resistance gene. For example, selection is based either on the complementation of auxotrophic strain, toxin-antitoxin systems, operator-repressor titration, RNA markers, or on the ov erexpression of a growth essential gene. M inicircles or any other method that allow remov ing of the antibiotic resistance gene from the initial vector can also be used (Vandermeulcn et al., 201 1 . Mol. Ther. 19( 1 1 ): 1 942-49).

In one embodiment, the polynucleotide of the inv ention is l igated into an expression vector which has been specifically optimized for polynucleotide vaccinations. Elements include but are not l imited to a transcriptional promoter, immunogenic epitopes. additional cistrons encoding immunoenhancing or immunomodulatory genes (such as ubiquitin ), with their own promoters, transcriptional terminator, bacterial origin of replication, antibiotic resistance gene or another selection marker, and CpG sequences to stimulate innate immunity, al l of which are well known to those skilled in the art. Optionally, the vector may comprise internal ribosome entry sites (IRES).

In one embodiment, the vector comprises tissue-specific promoters or enhancers to limit expression of the polynucleotide to a particular tissue type.

For example, the muscle creatine kinase (MCK) enhancer element may be desirable to limit expression of the polynucleotide to a particular tissue type. Myocytes are terminal ly differentiated cells which do not divide. Integration of foreign DNA into chromosomes appears to require both cell division and protein synthesis. Thus, limiting protein expression to non-dividing cells such as myocytes may be preferable.

A further example includes k era t i nocy te-spec i fi c promoters, melanocyte-specific promoters and dermal papilla-specific promoters, such as for instance: keratin (including keratin 5 ( K.5 ) and keratin 14 (K.14 ) promoters for the basal layer of skin; keratin 1 (Kl) and keratin 10 (K 10) promoters for the suprabasal layer of skin), loricrin, involucrin, transglutaminase I, E-cadherin, elastin, fi laggrin, a 1 collagen, corn i fin β, mCCIO or meianocortin 1 receptor (MCR1) promoters.

In one embodiment, tissue- or cell-specific promoters may be used to target the expression of the modified VSV-G to antigen-presenting cells.

Examples of other eukaryotic transcription promoters include, but are not limited to, the Rous sarcoma virus (RSV ) promoter, the simian virus 40 (SV40) promoter, the human elongation factor- 1 a (EF-lα) promoter and the human ubiquitin C (UbC) promoter.

Suitable vectors include any pi asm id DNA construct encoding a polynucleotide of the invention, operatively linked to a eukaryotic promoter. Examples of such vectors include the pCMV series of expression vectors, commercially available from Stratagene (La Jolla, Calif); or the pcDNA or pREP series of expression vectors by Invitrogen Corporation (Carlsbad, Cal if ). In another embodiment, the vector is a viral v ector. In one embodiment, v iral vectors include adenoviral, retrov iral, herpes virus and AAV vectors. Such recombinant viruses may be produced by techniques known in the art, such as by transfecting packaging cells or by transient transfection with helper plasmids or viruses. Typical examples of virus packaging cells include PA317 cells, PsiCRIP cells, GPenv + cells, 293 cells, and the like. Detailed protocols for producing such replication-defectiv e recombinant viruses may be found for instance in WO 1 995014785, WO 1996022378, US5.882.877, US6.013.5 16, US4.861 .71 9, US5,278.056 and WO 1 994019478.

4. Host cell / dendritic cell Another object of the inv ention is also a prokaryotic or eukaryotic host cell genetical ly transformed with at least one polynucleotide according to the invention.

The term "transformation" means the introduction of a "foreign " (i.e., extrinsic or extracellular) gene. DNA or RNA sequence ( including plasmids and viral vectors), to a host cel l, so that the host cel l will express the introduced gene or sequence to produce a desired substance, typically a protein coded by the introduced gene or sequence. A host cel l that receives and expresses introduced DNA or RNA has been "transformed".

Preferably, for expressing and producing the proteins, and in particular the modified VSV-G according to the inv ention, eukaryotic cells, in particular mammalian cel ls, and more particularly human cel ls, will be chosen. Typically, cell l ines such as CHO, BHK.-2 1 , COS-7, C 1 27, PER.C6 or HEK293 could be used, for their ability to process to the right post-translational modifications of the derivatives.

The construction of expression vectors in accordance with the invention, and the transformation of the host cells can be carried out using conventional molecular biology techniques. The modified VSV-G of the inv ention, can, for example, be obtained by culturing genetically transformed cells in accordance with the invention and recov ering the deriv ative expressed by said cell, from the culture. They may then, if necessary, be purified by conventional procedures, known in themselves to those skilled in the art, for example by fractionated precipitation, in particular ammonium sulphate precipitation, electrophoresis, gel filtration, affinity chromatography, etc.

In particular, conventional methods for preparing and purifying recombinant proteins may be used for producing the modified VSV-G in accordance with the invention. The present invention further relates to a dendritic cell transfected by polynucleotide(s) of the invention, i.e., a dendritic cell in which one or more polynucleotides according to the invention are inserted into.

Another object of the invention is a dendritic cel l population transfected by a nucleic acid sequence or a v ector according to the invention. 5. Composition

The present invention also relates to a composition comprising, consisting essentially of or consisting of a modified VSV-G. polynucleotide, vector or cel l of the invention.

As used herein, the expression "consist essentially of means that the composition to which it refers does not comprise any other active ingredient, i.e. , an ingredient responsible for a physiologic or therapeutic response, other than the modified VSV-G, polynucleotide, vector or cel l of the invention.

The present invention further relates to a pharmaceutical composition comprising, consisting essentially of or consisting of a modified VSV-G, polynucleotide, vector or cell of the inv ention and at least one pharmaceutically acceptable excipient. As used herein, the term "pharmaceutical composition" includes v eterinary composition.

The present invention also relates to an immunogenic composition comprising, consisting essentially of or consisting of a modified VSV-G, polynucleotide, vector or cel l of the invention. 6. V accine

The present invention also relates to a vaccine comprising the nucleic acid sequence coding for a modified VSV-G according to the invention, the vector comprising the nucleic acid sequence coding for a modified VSV-G according to the invention, the host cel l genetically transformed with the nucleic acid sequence coding for a modified VSV-G according to the invention or the modified VSV-G according to the invention.

In one embodiment, the vaccine of the invention is a prophylactic vaccine.

By "prophylactic vaccine" is meant that the vaccine is to be administered before definitive clinical signs, diagnosis or identification of the disease. According to this embodiment, the vaccine is to be administered to prevent the disease.

If the vaccine appears to induce an effective, but short-lived immune response, prophylactic vaccines may also be designed to be used as booster vaccines. Such booster v accines are given to indiv iduals who hav e previously received a v accination, with the intention of prolonging the period of protection. In another embodiment, the vaccine is a therapeutic vaccine, i.e., is to be administered after first clinical signs, diagnosis or identification of the disease. According to this embodiment, the vaccine is to be administered to treat the disease.

6.1. Polynucleotide vaccine

In one embodiment, the vaccine is a polynucleotide vaccine. Immunization with polynucleotide is also referred to as "genetic immunization", "RNA immunization" or "DNA immunization".

Accordingly, in one embodiment, the vaccine of the invention comprises a polynucleotide encoding, or a nucleic acid sequence coding for, a modified VSV-G according to the invention. In one embodiment, the vaccine of the invention is a DNA-based vaccine. Accordingly, in one embodiment, the vaccine of the invention comprises a DNA molecule encoding a modified VSV-G according to the invention.

In another embodiment, the vaccine of the invention is a RNA-based vaccine. Accordingly, in one embodiment, the vaccine of the invention comprises a RNA molecule, preferably a mRNA molecule, encoding a modified VSV-G according to the invention.

In one embodiment, the vaccine of the invention expresses more than one modified VSV-G. Accordingly, in one embodiment, the vaccine of the invention expresses two modified VSV-G or more. In a particular embodiment, the vaccine of the invention expresses two modified VSV-G or more, wherein said modified VSV-G are different.

According to this embodiment, the polynucleotide vaccine of the invention may comprise two polynucleotides encoding, or two nucleic acid sequences coding for, two different modified VSV-G. Still according to this embodiment, the protein vaccine of the invention may comprise two different modified VSV-G.

In a preferred embodiment, the vaccine of the invention expresses a first modified VSV-G and a second modified VSV-G wherein the first modified VSV-G comprises a CD8 T cell epitope and wherein the second modified VSV-G comprises a CD4 T cell epitope. The present invention further relates to a combination of:

(a) a first modified VSV-G, polynucleotide, vector, composition, cell or vaccine comprising a first heterologous peptide or a first combination of more than one heterologous peptide or nucleic acid sequence thereof; and

(b) a second modified VSV-G, polynucleotide, vector, composition, cell or vaccine comprising a second heterologous peptide or a second combination of more than one heterologous peptide or nucleic acid sequence thereof;

wherein said first heterologous peptide or combination of more than one heterologous peptide or nucleic acid sequence thereof and said second heterologous peptide or combination of more than one heterologous peptide or nucleic acid sequence thereof are different.

In one embodiment, said first heterologous peptide or nucleic acid sequence thereof is a CD8 T cell epitope and said second heterologous peptide or nucleic acid sequence thereof is a CD4 T cell epitope.

In one embodiment, said first and/or second modified VSV-G, polynucleotide, vector, composition, cell or vaccine may further comprise a universal antigenic CD4 T cell epitope or nucleic acid sequence thereof.

6.2. Protein vaccine In another embodiment, the vaccine of the invention is a protein vaccine. Accordingly, in one embodiment, the vaccine of the invention comprises a modi fied VSV-G according to the invention. In another embodiment, the vaccine of the invention comprises two modi fied VSV-G or more. In a particular embodiment, the vaccine of the invention comprises two modified VSV-G or more, wherein said modified VSV-G arc different.

In a preferred embodiment, the vaccine of the invention comprises a first modified VSV-G and a second modified VSV-G wherein the first modified VSV-G comprises a CD8 T cell epitope and wherein the second modified VSV G comprises a CD4 T cell epitope. In one embodiment, the vaccine of the present invention is used in a prime-boost strategy to induce robust and long-lasting immune response to the peptide. Priming and boosting vaccination protocols based on repeated injections of the same antigenic construct are well known and result in strong CTL responses. In general, the first dose may not produce protective immunity, but only "primes" the immune system.. A protective immune response develops after the second or third dose.

In one embodiment, the vaccine of the invention is used in a conventional prime-boost strategy, in which the same vaccine is to be administered to the subject in multiple doses. In a preferred embodiment, the vaccine is used in one or more inoculations. These boosts are performed according to conventional techniques, and can be further optimized empirically in terms of schedule of administration, route of administration, choice of adjuvant, dose, and potential sequence when administered with another vaccine, therapy or homologous vaccine.

In another embodiment, the vaccine of the present invention is used in a prime-boost strategy using an alternative administration of modified VSV-G comprising xenoantigen and autoantigen or fragment thereof, or of polynucleotides encoding modified VSV-G comprising xenoantigen and autoantigen or fragment thereof. Specifically, according to the present invention, the subject is first treated, or "primed", with a vaccine encoding an antigen of foreign origin or fragment thereof (a '"xenoantigen"). Subsequently, the subject is then treated with another v accine encoding an antigen or fragment thereof which is corresponding to the xenoantigen, but is of self-origin ("autoantigen"). This way. the immune reaction to the antigen is boosted. The boosting step may be repeated one or more times .

6.3. Excipients

In one embodiment, vaccines of the present invention are formulated with pharmaceutical ly acceptable carriers or excipients such as water, saline, dextrose, glycerol, and the l ike, as wel l as combinations thereof. In one embodiment, vaccines may aiso contain auxiliary substances such as wetting agents, emulsifying agents, buffers, adjuvants, and the l ike.

In another embodiment, excipient for use in the polynucleotide vaccines of the present invention is a polymer such as a cationic polymer or a non-ionic polymer ( including but not l imited to: polyoxyethyiene (POE), p o I y o x y p ro p y I e n e (POP), polyethyleneglyeol (PEG), linear or branched polyethylenimine (PEI)). In another embodiment, polymers can form block copolymers, for instance, a POE-POP-POE block copolymer. As used herein, the term "polyplex" refers to po I y m er-po I y n u c I eo t i d e or copolymer- polynucleotide complexes. In another embodiment, the polynucleotide vaccines are formulated with cationic lipids. Optional ly, lipids can be mannolysated. As used herein, the term "lipoplex" refers to lipid-polynucleotide or I i poso m e-po I y n u c I cot i d e complexes.

In one embodiment, iipoplexes are further complexed with polymers or copolymers to form tertiary complexes. These tertiary complexes have enhanced in vivo delivery and transfection capacities of the polynucleotide to the targeted cells, and thereby, facilitate enhanced immune responses.

In one embodiment, carries for use in the polynucleotide vaccines of the present invention are nanoparticles. These include but are not limited to: nano -emulsions, dendrirners, nano-goid, iipid-based nanoparticles, liposomes, drug-carrier conjugates, antibody-drug complexes, and magnetic nanoparticles.

6.4. Adjuvants

In one embodiment, the polynucleotide vaccine of the present invention is formulated with at least one adjuvant which may increase immunogenicity of the polynucleotide vaccines of the present invention. It is w ithin the purview of the skilled artisan to utilize available adjuvants which may increase the immune response of the polynucleotide vaccines of the present invention in comparison to administration of a non-adjuvanted polynucleotide vaccine.

In some embodiments, the adjuvant is selected from the group consisting of a-interferon, γ-interferon, platelet derived growth factor (PDGF), TNF-a, TNF-β, GM- CSF, epidermal growth factor ( EOF), H IV- 1 gag, cutaneous T cel l-attracting chemokine (CTACK), epithelial thymus-expressed chemokine (TECK), mucosae-associated epithelial chemokine (MEC), 11.-2. I E- 12, I I.- 1 5, IL-28, MHC, CD80, CD86 including I L- 1 5 having the signal sequence deleted and optionally including the signal peptide from IgE. Other genes which may be useful adjuvants include those encoding: MCP-I, MIP-loc, MIP-I p, IL-8, R ANTES, L-selectin, P-selectin, E-selectin, CD34, GlyCAM- 1 , MadCAM-1 , LEA- I. VI A- 1, Mac- 1 , pl50.95, PECAM, ICAM-I, ICAM-2, ICAM-3, CD2, LFA-3, M-CSF, G-CSE, 11. -4, mutant forms of IL-18, CD40, CD40L, vascular growth factor, fibroblast growth factor. I L-7. nerve growth factor, vascular endothelial growth factor. Fas, TNF receptor. Fit, Apo- 1 , p55, WSL-I, DR3, TRAM P, Apo-3, AIR, LARD, NGRF, DR4, DR5, KI LLER, TRA I L-R2, TRICK 2, DR6, Caspase ICE, Fos, c-jun, Sp-I, Ap-I, Ap-2, p38, p65Rel, MyD88, IRAK, TRAF6, IkB, Inactive NIK, SAP K, SAP-I, .INK, interferon response genes, NFkB, Bax, TRAIL, TR A I I . tec, TRAlLrecDRC5 , TRAI L-R3, TRAI L-R4, RANK, RANK LIGAND, Ox40, Ox40 LIGAND, NKG2D, MICA, MICB, NKG2A, NKG2B, NKG2C, NKG2E, NKG2F, TAP 1 , TAP2, functional fragments and combinations thereof.

In some preferred embodiments, the adjuvant is selected from the group consisting of a-interferon, γ-intcrfcron, 11.-2, IL-8, I L- 12, I I - 1 5, I L- 1 8, I L-28, MCP-I, M lP-la, M I P-Ip, RANTES, RANK, RANK LIGAND, Ox40, Ox40 LIGAND, CTACK, TECK, MEC, functional fragments and combinations thereof.

In some preferred embodiments, the adjuvant is selected from the group consisting of a-interferon, γ-interferon, I L-2, I L- 1 2, functional fragments and combinations thereof In another embodiment, adjuvant for use in the polynucleotide vaccines of the present invention is mineral-based compounds such as one or more forms of an aluminum phosphate-based adjuvant, or one or more forms of a calcium phosphate.

In another embodiment, adjuvant is saponin, monophosphoryl l ipid A or other compounds that can be used to increase immunogenicity of the polynucleotide vaccine. In one embodiment, the polynucleotide vaccine of the present invention is formulated with at least one genetic adjuvant which may increase immunogenicity of the polynucleotide vaccines of the present invention. It is within the purview of the skilled artisan to util ize available genetic adjuvants which may increase the immune response of the polynucleotide vaccines of the present invention in comparison to administration of a non-adjuvanted polynucleot ide vaccine.

As used herein, genetic adjuvants refer to immunomodulatory molecules encoded by a plasmid vector. They stimulate the innate immune system to trigger appropriate dendritic cell maturation and thereby a robust, specific, and long-lasting adaptive immune response. I m m u nomod u 1 atoty molecules include cytokines, chemokines, or immune stimulatory molecules, such as toll-like receptor agonists or interferon regulatory factors.

In one embodiment, the genetic adjuvant is not encoded by the polynucleotide or vector coding for a modified VSV-G according to the invention. In another embodiment, the genetic adjuv ant is encoded by the polynucleotide or vector coding for a modified VSV-G according to the invention. According to this embodiment, the genetic adjuvant can be under the control of its own promoter:, or the genetic adjuvant can be under the control of the same promoter as the modified VSV-G according to the invention, separated therefrom by an Internal Ribosome Entry Site (IRES).

In some embodiments, the genetic adjuv ant is selected from the group consisting of a-interferon, γ-interferon, platelet derived growth factor (PDGF), TNF-a, TNF-β, GM- CSF, epidermal growth factor (EGF), Ll IV- 1 gag, cutaneous T cell-attracting chemokine (CTACK), epithelial thymus-expressed chemokine (TECK), mucosae-associated epithelial chemokine (MEC), IL-2, IL-12, I L- 1 5, IL-28, MHC, CD80, CD86 including I L- 1 5 having the signal sequence deleted and optionally including the signal peptide from IgE. Other genes which may be useful adjuvants include, without limitation, those encoding MCP-I, MIP-loc, MIP-I p. 11.-8, RANTES, L-seiectin, P-selectin, E-selectin, CD34, GlyCAM- 1 , MadCAM-1 , I. FA- I, VI. A- 1, Mac- 1 , pi50.95, PEC AM, ICAM-I. ICAM-2, ICAM-3, CD2, LFA-3, M-CSF, G-CSF, IL-4, mutant forms of I L- 1 8, CD40, CD40L, vascular growth factor, fibroblast growth factor, I L-7, nerv e growth factor, vascular endothelial growth factor, Fas, TNF receptor. Fit, Apo- 1 , p55, WSL-I, DR3, TRAMP, Apo-3, AIR, LARD, NGRF, DR4, DR5, KI LLER, TRAI L-R2, TRICK.2, DR6, Caspase ICE, Fos, c-jun, Sp-I, Ap-I, Ap-2, p38, p65Rei, MyD88, I RAK, TRAF6, IkB, Inactive NIK, SAP K, SAP-L JNK, interferon response genes, NFkB, Bax, TRA I L, TRAI Lrec, TRAI LrecDRCS, TRAI L-R3, TRA I L-R4, RANK, RANK LIGAND, Ox40, 0x40 LIGAND, NKG2D, M ICA. MICB, NKG2A, NKG2B, NKG2C, NKG2E, NKG2F, TAP 1 , TAP2, functional fragments and combinations thereof.

In some preferred embodiments, the genetic adjuv ant is selected from the group consisting of a-interferon, γ-interferon, I L-2, I I. -8, I L- 1 2, I L- 1 5, I L- 1 8, I L-28, MCP-I, MIP-Ia, MIP-Ip, RANTES. RANK, RANK LIGAND, Ox40, Ox40 LIGAND, CTACK, TECK, MEC, functional fragments and combinations thereof.

In some preferred embodiments, the genetic adjuvant is selected from the group consisting of a-interferon, γ-interferon, IL-2, 11.- 1 2, functional fragments and combinations thereof.

Examples of other adjuvants include, but are not limited to, particle bombardment using DNA-coated or RNA-coated gold beads; co-administration of polynucleotide vaccines with piasmid DNA expressing cytokines, chemokines, or costimulatory molecules.

7. Use A further object of the present invention relates to a modified VSV-G, polynucleotide, vector, composition, cell or vaccine according to the invention for use in the prevention or treatment of, or for use in preventing or treating, a disease or condition.

In one embodiment, the modified VSV-G, polynucleotide, vector, composition, cell or vaccine according to the invention is for use in the prevention or treatment of, or for use in preventing or treating, a cancer or an infectious disease.

In a particular embodiment, the modified VSV-G, polynucleotide, vector, composition, cell or vaccine according to the invention is used to provide long term inhibition of tumor growth in a subject.

According to an embodiment of the invention, dendritic cells transfected by polynucleotides of the invention are used to activate T cells in vitro. T cells or a subset of T cells can be obtained from various lymphoid tissues. Examples of such tissues include, but are not limited to, spleens, lymph nodes and peripheral blood.

The cells can be co-cultured with transfected dendritic cells as a mixed T cell population or as a purified T cell subset. For instance, it may be desired to culture purified CD8+ T cells with antigen transfected dendritic cells, as early elimination of CD4+ T cells may prevent the overgrowth of CD4+ cells in a mixed culture of both CD8+ and CD4+ T cells. T cell purification may be achieved by positive or negative selection, including, but not l imited to. the use of antibodies directed to CD2, CD3, CD4, CDS, and CD8. On the other hand, it may be desired to use a mixed population of

CD4+ and CD8+ T cells to elicit a specific response encompassing both a cytotoxic and Th immune response. In one embodiment, after activation in vitro, the T cells may be administered to a subject in a dose sufficient to induce or enhance an immune response to the selected antigen expressed by the dendritic cells of the invention.

8. Administration / doses

In one embodiment, the composition or vaccine of the invention is to be administered ex vivo or in vivo.

Ex vivo administration refers to performing part of the regulatory step outside of the subject, such as administering a composition of the present invention to a population of cells, preferably dendritic cells, removed from a subject under conditions such that the modified VSV-G, polynucleotide or vaccine is loaded into the cell, and returning the cells to the subject.

In one embodiment, the composition or vaccine of the invention may be administered to a subject, or returned to a subject, by any suitable mode of administration.

In one embodiment, the admin istration is systemic, mucosal and/or proximal to the location of the target site (e.g. , near a tumor). The preferred routes of administration will be apparent to those of skill in the art, depending on the type of condition to be prevented or treated, the antigen used and/or the target cell population or tissue.

Preferred methods of administration include, but arc not limited to, electroporation or sonoporation. Administration by electroporation involves the application of a pulsed electric field to create transient pores in the cellular membrane without causing permanent damage to the cel l. Administration by sonoporation involves the application of pulsed ultrasonic frequencies to create transient pores in the cellular membrane without causing permanent damage to the cell. It thereby al lows for the introduction of exogenous molecules. By adjusting the electrical pulse and/or the ultrasonic frequencies, nucleic acid molecules can travel through passageways or pores in the cel l that are created during the procedure. Other preferred methods of administration include, but are not limited to, intravenous administration, intraperitoneal administration, intramuscular administration, intranodal administration, intracoronary administration, intraarterial administration (e.g., into a carotid artery), subcutaneous administration, intradermal administration, transdermal del ivery, intratiimoral administration, peritumoral administration, intratracheal administration, subcutaneous administration. intraarticular administration, intraventricular administration, inhalation (e.g. , aerosol ), intracranial, intraspinal, intraocular, aural, intranasal, oral, pulmonary administration, impregnation of a catheter, and direct injection into a tissue. In some embodiments, administration may be a combination of two or more of the v arious routes of administration. Particularly preferred routes of administration include, but are not l imited to, electroporation, sonoporation, intravenous, intraperitoneal, subcutaneous, intratiimoral, peritumoral, intradermal, intranodal, intramuscular, transdermal, inhaled, intranasal, oral, intraocular, intraarticular, intracranial and intraspinal.

Parenteral delivery includes, without limitation, electroporation, sonoporation, intratiimoral. peritumoral, intradermal, intramuscular, intraperitoneal, intrapleural, intrapulmonary, intravenous, subcutaneous, atrial catheter and v enal catheter routes.

Aural deliv ery includes, without limitation, ear drops, intranasal del iv ery can include nose drops or intranasal injection, and intraocular delivery can include eye drops.

Aerosol ( inhalation ) delivery can also be performed using methods standard in the art (see, for example, Stribling et al, 1992. Proc. Natl. Acad. Sci. USA. 189: 1 1277-1 1281). For example, in one embodiment, a composition or vaccine of the invention can be formulated into a composition suitable for nebulized delivery using a suitable inhalation device or nebulizer. Oral del ivery includes, without l imitation, solids and liquids that can be taken through the mouth, and is useful in the development of mucosal immunity and since compositions comprising yeast vehicles can be easily prepared for oral delivery, for example, as tablets or capsules, as wel l as being formulated into (bod and beverage products.

Other routes of adm inistration that modulate mucosal immunity are useful in the treatment of viral infections, epithelial cancers, immunosuppressive disorders and other diseases affecting the epithelial region. Such routes include bronchial, intradermal, intramuscu lar, intranasal, other inhalatory, rectal, subcutaneous, topical , transdermal, vaginal and urethral routes.

In one embodiment, the composition or vaccine may be administered to the subject by intramuscular injection, intradermal injection, gene gun, electroporation. or biojector. I n a more preferred embodiment, the composition or vaccine is to be administered by electroporation. preferably by intramuscular or intradermal electroporation. Electroporation uses pulsed electric currents to open pores in cell membranes (a process cal led permeabi l ization ) and al lows the injected polynucleotide to be taken up by cells and immune cells residing in the tissue.

I n one embodiment, the polynucleotide is formulated as l ipoplex (cationic l iposome-

DNA complex), polyplex (cationic polymer-DNA complex), or protein-DNA. complex. In one embodiment, the composition or vaccine of the present invention is to be administered before symptoms appear, i.e. , the composition or vaccine of the present invention is to be adm inistered prophy lactical ly .

In one embodiment, the composition or vaccine of the present invention is to be administered after symptoms appear, i.e., the composition or vaccine of the present invention is to be administered therapeutically.

According to the present invention, an effective adm in istration protocol (i.e. , administering a composition or vaccine in an effective manner) comprises suitable dose parameters and modes of administration that result in el icitation of an immune response in a subject that has a disease or condition, or that is at risk of contracting a disease or condition, preferably so that the subject is protected from the disease.

Effective dose parameters can be determined using methods standard in the art for a particular disease. Such methods include, but are not limited to, determination of survival rates, side effects (i.e., toxicity) and progression or regression of disease.

In particular, the effectiveness of dose parameters of a therapeutic composition of the present invention when treating cancer can be determined by assessing response rates. Such response rates refer to the percentage of treated patients in a population of patients that respond with either partial or complete remission. Remission can be determined by, for example, measuring tumor size or microscopic examination for the presence of cancer cells in a tissue sample.

According to the present invention, a suitable single dose size is a dose that is capable of eliciting an antigen-specific immune response in a subject when administered once or more times over a suitable time period. Doses can vary depending upon the disease or condition being treated. In the treatment of cancer, for example, a therapeutic effective amount can be dependent upon whether the cancer being treated is a primary tumor or a metastatic form of cancer. One of skills in the art can readily determine prophylactic or therapeutic effective amounts for administration based on the size of a subject and the route of administration. In one embodiment, a prophylactic or therapeutic effective amount of the composition or vaccine of the invention is from about 0.5 pg to about 5 mg per kilogram body weight of the subject being administered the composition or vaccine. In a preferred embodiment, a prophylactic or therapeutic effective amount of the composition or vaccine of the invention is from about 0. 1 tig to about 1 mg per kilogram body weight of the subject, preferably from about 1 tig to about 100 pμegr kilogram body weight of the subject, preferably from about 1 0 tig to about 75 tig per kilogram body weight of the subject, preferably about 50 [ig per kilogram body weight of the subject.

When T cel ls or dendritic cel ls are administered to a subject, the cel ls may be administered (with or without adjuvant ) parenterally ( including, for example. intravenous, intraperitoneal. intramuscular. intradermal. and subcutaneous administration ). Alternatively, the cel ls may be administered local ly by direct injection into a tumor or infected tissue.

Adjuvants include any known pharmaceutically acceptable carrier. Parenteral vehicles for use as pharmaceutical carriers include, but are not limited to, sodium chloride solution. Ringer's dextrose, dextrose and sodium chloride, and lactated Ringer's. Other adjuvants may be added as desired such as antimicrobials.

As an example. T cells may be administered by intravenous infusion, at doses of about 10 8 to 1 0" cells 'm 2 of body surface area (see, e.g. , Ridel I et al, 1992. Science. 257:238- 241). Infusion can be repeated at desired intervals, for example, monthly. Recipients are monitored during and after T cel l infusions for any evidence of adverse effects.

According to a preferred embodiment, the T cells are obtained from the same subject from whom the dendritic cells were obtained.

According to another embodiment, the T cel ls are obtained from a subject and the dendritic cel ls, which are used to stimulate the T cells, are obtained from an H LA- matched healthy donor (e.g. , a sibling), or vice versa.

According to yet another embodiment, both the T cel ls and the dendritic cel ls are obtained from an H LA-matched healthy donor. This embodiment may be particularly advantageous, for example, when the subject is a late stage cancer patient who has been treated with radiation and/or chemotherapy agents and may not be able to provide sufficient or efficient dendritic or T cel ls.

According to another embodiment of the invention, dendritic ceils isolated from a subject are cultured, transfected in vitro and administered back to the subject to stimulate an immune response, including T cell activation. As such, the dendritic cel ls constitute a vaccine and/or immunotherapeutic agent.

As an example, dendritic cel ls presenting antigen are administered, via intravenous infusion, at a dose of, for example, about 10 5 to 10 8 cells. According to an embodiment, dendritic cel ls presenting antigen are administered at a dose from about 0.5 x 10 6 to about 40 χ 10 7 dendritic cells per administration, preferably from about l x lO 6 to about 20 χ 10 7 dendritic cel ls per administration, more preferably from about 10 χ 10 6 to about 1 x 10 7 dendritic cells per administration.

In one embodiment, infusion can be repeated at desired intervals based upon the subject's immune response.

When vaccines of the invention arc used in a prime-boost strategy, "boosters" of the vaccine are preferably administered when the immune response against the peptide, preferably antigen, has waned or as needed to provide an immune response or induce a memory response against a particular peptide, preferably antigen. Boosters can be administered from, about 1 week to several years after the original administration. In one embodiment, an administration schedule is one in which from about 0.5 pg to about 5 mg of a vaccine per kilogram body weight of the subject is to be administered from about one to about 4 times over a time period of from about 1 month to about 6 months.

It will be obvious to one of skills in the art that the number of doses administered to a subject is dependent upon the extent of the disease and the response of said subject to the treatment. For example, a large tumor may require more doses than a smaller tumor, and a chronic disease may require more doses than an acute disease. In some cases, however, a subject having a large tumor may require few er doses than a patient with a smaller tumor, if the subject with the large tumor responds more favorably to the composition or vaccine than the subject w ith the smaller tumor. Thus, it is within the scope of the present invention that a suitable number of doses includes any number required to treat a giv en disease.

9. Diseases

9.1. Cancer In one embodiment, the disease or condition which may be prevented or treated with the modified VSV-G, polynucleotide, vector, composition, cell or vaccine according to the inv ention is a cancer. As used herein, the term "cancer" includes, but is not l imited to. solid tumors and blood borne tumors. The term cancer includes diseases of the skin, tissues, organs, bone, cartilage, blood, and vessels.

In one embodiment, the cancer is a primary cancer. In another embodiment, the cancer is a metastatic cancer. A metastatic cancer is a cancer that has spread from its primary origin to another part of the body, also referred to as "late stage cancer" or "advanced stage cancer". In some embodiments, advanced stage cancer includes stages 3 and 4 cancers. Cancers are ranked into stages depending on the extent of their growth and spread through the body; stages correspond with severity. Determining the stage of a given cancer helps doctors to make treatment recommendations, to form a l ikely outcome scenario for what will happen to the patient (prognosis), and to communicate effectively with other doctors.

Examples of cancer include, but are not l imited to, melanomas, squamous cel l carcinoma, breast cancers, head and neck carcinomas, thyroid carcinomas, soft tissue sarcomas, bone sarcomas, testicular cancers, prostatic cancers, ovarian cancers, bladder cancers, skin cancers, brain cancers, angiosarcomas, hemangio sarcomas, mast cell tumors, hepatic cancers, lung cancers, pancreatic cancers, gastrointestinal cancers, renal cell carcinomas, hematopoietic neoplasias and metastatic cancers thereof.

In a particular embodiment, cancer is selected from the group comprising or consisting of melanomas, prostatic cancers, ovarian cancers, brain cancers, lung cancers and others.

Preferably, expression of the tumor antigen in a tissue of a subject, i.e. , an animal or a human, that has cancer produces a result selected from the group of al lev iation of the cancer, reduction of a tumor associated w ith the cancer, el imination of a tumor associated with the cancer, prevention of metastatic cancer, prevention of the cancer and stimulation of effector cel l immunity against the cancer. 9.2. Infections diseases

In one embodiment, the disease or condition which may be prevented or treated with the modified VSV-G, polynucleotide, vector, composition, cell or vaccine according to the invention is an infectious disease. In one embodiment, the infectious disease is selected from the group consisting of viral, bacterial, fungal and parasitic infection.

Examples of infectious virus include, but are not limited to, Retroviridae (e.g., human immunodeficiency viruses, such as H IV- 1 , also referred to as HTLV-III, LAV or HTLV-I I I/LAV. or H IV-I I I; and other isolates, such as HIV-LP); Picornaviridae (e.g., polio viruses, hepatitis A virus; enteroviruses, human coxsackie viruses, rhinoviruses, echoviruses); Calciviridac (e.g. , strains that cause gastroenteritis); Togaviridae (e.g., equine encephalitis v iruses, rubella viruses); Flav iridae (e.g. , dengue viruses, encephalitis viruses, yellow fever viruses); Coronav iridac (e.g., coronaviruses); Rhabdoviridae (e.g. , vesicular stomatitis viruses, rabies viruses); Filov iridae (e.g., cbola viruses); Paramyxoviridae (e.g., parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus); O rt h o m y x o v iridac (e.g., influenza viruses); Bungaviridae (e.g., Hantaan viruses, bunga viruses, phleboviruses and Nairo viruses); Arena viridae (hemorrhagic fever virus); Reov iridae (e.g., reoviruses, orbiviruses and rotaviruses); Birna viridae; Hepadnaviridac ( Hepatitis B virus); Parvoviridac (parvoviruses); Papovaviridae (papil loma viruses, polyoma viruses); Adenoviridae (most adenov iruses); Herperviridae (herpes simplex virus (HSV) 1 and 2, varicella zoster virus, cytomegalovirus (CMV), herpes viruses); Poxv iridae (variola viruses, vaccinia viruses, pox viruses); and Iridov iridae (e.g. , African swine fever virus); and unclassified viruses (e.g., the etiological agents of Spongiform encephalopathies, the agent of delta hepatitides (thought to be a defective satell ite of hepatitis B virus), the agents of non-A, non-B hepatitis (class 1— internal ly transmitted; class 2— parenterally transmitted (i.e., Hepatitis C); Norwalk and related viruses, and astro viruses).

Examples of infectious bacteria include, but are not limited to, Helicobacter pylons, Boreliai burgdorferi, Legionella pneumophilia, Mycobacteria sps (e.g. , M. tuberculosis, M. avium, M. Intracellulare, M. kansaii, M gordonae), Staphylococcus aureus, Neisseria gonorrhoeae, Neisseria meningitidis, Listeria monocytogenes, Streptococcus pyogenes (Group A Streptococcus), Streptococcus agalactiae (Group B Streptococcus), Streptococcus (viridans group), Streptococcus faecaiis, Streptococcus bovis, Streptococcus (anaerobic sps.), Streptococcus pneumoniae, pathogenic Campylobacter sp., Enterococcus sp., Haemophilus influenzae, Bacillus antracis, corynebacterium diphtheriae, corynebacterium sp., Erysipelothrix rhusiopathiae, Clostridium perfringers, Clostridium tetani, Enterobacter erogenes, Klebsiella pneuomiae, Pasturella multicoda, Bacter aides sp., Fusobacterium nucleatum, Sreptobacillus moniliformis, Treponema pallidium, Treponema pertenue, Leptospira, and Actinomeyces israelii.

Examples of infectious fungi include, but are not limited to, Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, Candida albicans. Other infectious organisms (i.e. , protists) include, but are not limited to, Plasmodium falciparum and Toxoplasma gondii.

10. Subject

In one embodiment, the subject is susceptible or suspected of having a disease or condition, preferably a cancer or an infectious disease. in one embodiment, the subject is at risk of developing a disease or condition, preferably a cancer or an infectious disease.

Examples of risks of developing a cancer include, but are not limited to, age, alcohol, exposure to cancer-causing substances, chronic inflammation, diet, hormones, familial cancer predisposition, genetic cancer predisposition, immunosuppression, infectious agents, obesity, exposure to radiation, exposure to sunlight, tobacco and the like. Examples of risks of developing an infectious disease include, but are not limited to, exposure to bacteria, viruses, fungi, and parasites (for instance by indirect contact, insect bites or food contamination ); having certain types of cancer or H IV; taking of steroids; implanted medical devices; malnutrition; extremes of age and the like. In another embodiment, the subject suffers from a disease or condition, preferably a cancer or an infectious disease.

In one embodiment, the subject was not treated prev iously with another treatment for the disease or condition. In another embodiment, the subject previously receiv ed one, two or more other treatments for the disease or condition. In one embodiment, the subject previously received one or more other treatments for the disease or condition, but was unresponsive or did not respond adequately to these treatments, which means that there is no or too low therapeutic benefit induced by these treatments. In one embodiment, the subject is an animal, preferably a mammal .

In a further embodiment, said mammal is a domestic animal. As used herein, the term "domestic animal" refers to any of various animals domesticated so as to live and breed in a tame (as opposed to wild) condition. Domestic animals include, but are not limited to, cattle ( including cows), horses, pigs, sheep, goats, dogs, cats, and any other mammal which is awaiting the receipt of, or is receiving medical care or was/is/will be the object of a medical procedure, or is monitored for the development of a disease.

In another embodiment, said mammal is a primate. As used herein, the term "primate" includes non-human primates such as lemurs, galagos, lorisids, tarsiers, monkeys, apes; and human primates, i.e. , human. In one embodiment, the subject of the invention is young. As used herein, the term "young" means that the subject is at most 20 years old, at most 15 or 10 years old if the subject is a human; or has an equivalent age according to the specie if the subject is a non-human animal.

In one embodiment, the subject is a child. As used herein, the term, "child" refers to a human being (person) during the period between birth and puberty. By "puberty" it means the time in which sexual and physical characteristics mature person because of hormonal changes. In a particular embodiment, the present invention child is considered a person of up to 14 years (inclusive). In one embodiment, the subject is a male. In another embodiment, the subject is a female. In one embodiment, the subject is a man. In another embodiment, the subject is a woman.

11. Method Another object of the present invention is a method for preventing and/or treating a disease or a condition comprising administering a modified VSV-G, polynucleotide, vector, composition, cell or vaccine of the invention to a subject in need thereof.

In a particular embodiment, the method of the invention is for preventing and/or treating a cancer in a subject in need thereof, comprising administering a modified VSV-G, polynucleotide, vector, composition, cell or vaccine of the invention to said subject.

In another particular embodiment, the method of the invention is for preventing and/or treating an infectious disease in a subject in need thereof, comprising administering a modified VSV-G, polynucleotide, vector, composition, cel l or vaccine of the invention to said subject. In one embodiment, the method comprises administering a modified VSV-G, polynucleotide, vector, composition, cell or vaccine of the invention before symptoms appear. According to this embodiment, the method may be a prophylactic method.

In another embodiment, the method comprises administering a modified VSV-G, polynucleotide, vector, composition, cell or vaccine of the invention after first symptoms appear. According to this embodiment, the method may be a therapeutic method.

In one embodiment, the method of the invention is combined with other prophylactic and/or therapeutic approaches to enhance the efficacy of the method. For example, in the treatment of cancer, the modified VSV-G, polynucleotide, vector, composition, cel l or vaccine of the invention may be administered after surgical resect ion of a tumor from the subject. In another embodiment, the modified VSV ' -G, polynucleotide, vector, composition, cel l or vaccine of the invention may be admin istered in combination with another therapeutic molecule, such as chemotherapeutic agents, anti-angiogenesis agents, checkpoint blockade antibodies or other molecules that reduce immune-suppression; or in combination with another antitumor treatment, such as radiation therapy, hormonal therapy, targeted therapy or immunotherapy.

In a particular embodiment, the modified VSV-G, polynucleotide, vector, composition, cell or vaccine of the invention is to be administered in combination with antibodies.

Examples of antibodies which may be co-administered include, but are not limited to, antibodies anti-PD-1 (e.g., nivolumab, pidiiizumab and MK-3475), antibodies anti-PD- I . 1 (e.g., BMS-936559, MEDI4736 and MPDL33280A), antibodies anti-CTLA4 (e.g., ipilimumab and tremelimumab ), antibodies anti-OX40, antibodies anti-4- 1 BB, antibodies anti-CD47, antibodies anti-KIR, antibodies anti-CD40, antibodies anti-LAG-3 and combinations thereof. In a particular embodiment, the modified VSV-G, polynucleotide, v ector, composition, cel l or vaccine of the invention is to be administered in combination with stimulating factors. Example of stimulating factors which may be co-administered include, but are not limited to, granulocyte -macrophage colony-stimulating factor (GM-CSF) (e.g., sargramostim or molgramostim ). Another object of the present invention is a method for inducing in a subject a protectiv e immune response comprising administering a modified VSV-G, polynucleotide, v ector, composition, cell or vaccine of the invention to a subject in need thereof.

In one embodiment, the method of the invention is for inducing in a subject a protective immune response against cancer. In another embodiment, the method of the invention is for inducing in a subject a protective immune response against a pathogen. 11.1. Personalized treatment

The present invention also relates to a personalized method for treating a disease or condition, preferably a cancer, in a subject (i.e., a human being or a non-human animal) in need thereof comprising administering a modified VSV-G, polynucleotide, vector, cell, composition or vaccine as described herein above.

In one embodiment, the personalized method for treating a cancer in a subject in need thereof comprises the steps of:

a) providing a sample of a tumor from a subject;

b) identifying at least one neoantigen;

c) preparing a composition comprising the at least one neoantigen inserted into VSV-G; and

d) administering the composition to the subject.

In one embodiment, the personalized method for treating a cancer in a subject in need thereof comprises the steps of:

a) providing a sample of a tumor from a subject;

b) identifying at least one neoantigen;

c) preparing a composition comprising a polynucleotide encoding a modified VSV-G into which a polynucleotide encoding the at least one neoantigen is inserted; and

d) administering the composition to the subject.

Any cell type or tissue may be utilized to obtain nucleic acid samples for use in the sequencing methods described herein. In a preferred embodiment, the DNA or RNA sample is obtained from a sample of a tumor from a subject or a bodily fluid, e.g., blood, obtained by known techniques (e.g., venipuncture), saliva, sweat, urine, feces, vomit, breast milk and semen. Alternatively, nucleic acid tests can be performed on dry samples (e.g., hair or skin).

Methods for identifying neoantigens are well-known from the person skilled in the art. For example, tumor sample from a subject and normal tissue may be subjected to whole-exome sequencing and RNA-Seq to identify expressed nonsynonymous somatic mutations. These mutations may be pipel ined into an epitope prediction algorithm (such as for example IEDB, EpiBot, EpiToolK.it ) to prioritize a list of candidate antigens and/or may be expressed as minigenes used for the identification and expansion of mutant n eoa n t i ge n -s pec i fi c autologous T cells isolated from blood or tumor of the same subject. Ex vzvo-expanded T cells may be then infused back into the cancer patient.

Preferably, any suitable sequencing-by-synthesis platform can be used to identify mutations. Four major sequencing-by-synthesis platforms are currently available: the Genome Sequencers from Roche/454 Life Sciences, the HiSeq Analyzer from l llumina, Solexa, the SOLiD system from Appl ied BioSystems, and the Heliscope system from Helicos Biosciences. Sequencing-by-synthesis platforms have also been described by Pacific Biosciences and VisiGen Biotechnologies. Each of these platforms can be used in the methods of the invention. A variety of methods are available for detecting the presence of a particular mutation or al lele in an individual's DNA or RNA.

Examples of such methods include, but are not limited to, dynamic al lele-specific hybridization (DASH), microplate array diagonal gel electrophoresis (MADGE), pyrosequencing, oligonucleotide-specific ligation, the TaqMan system as well as various DNA "chip" technologies such as the Affymetrix SNP chips. These methods require amplification of the target genetic region, typical ly by PGR.

Examples that eliminate the need for PGR include methods based on the generation of small signal molecules by invasive cleavage fol lowed by mass spectrometry or immobilized padlock probes and rolling-circle amplification. Alternatively, expressed mutations predicted to form neoantigens by MHC class I epitope-binding algorithms may be confirmed and then used to generate neoantigen vaccines. Tumor-specific neoantigens may also be identified using MHC multimers to identify neoantigen-specific T cell responses. For example, high throughput analysis of 11 eoan t i gen-spec i fi c T cell responses in patient samples may be performed using MHC tetramer-based screening techniques.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 is a set of graphs showing the effect of pTOP-OVA CD8 prophylactic intramuscular immunization on the anti-tumor activity. (A) Tumor growth follow-up after challenge. The tumor size was measured three times a week with an electronic digital caliper. Tumor volume was calculated as the length x width x height ( in mm 3 ). (B) Surv ival rates monitoring after chal lenge. The asterisks indicate significant differences compared with naive mice (**P<0.01) (n=6) (Comparison of survival curves, Mantel-Cox test).

Figure 2 is a set of graphs showing the effect of pTOP-OVA CD8 therapeutic intratumoral immunization on the anti-tumor activity. (A) Tumor growth follow -up after challenge. The tumor size was measured three times a week with an electronic digital cal iper. Tumor volume was calculated as the length x width x height (in mm 3 ). (B) Survival rates monitoring after challenge. The asterisks indicate significant differences compared with naive mice (***P<0.001) (n=6) (Comparison of survival curves, Mantel- Cox test).

Figure 3 is a set of graphs showing the effect of restriction sites addition around the inserted epitope sequence, for prophylactic intramuscular immunization. (A) Tumor growth follow -up after chal lenge. The tumor size was measured three times a week with an electronic digital caliper. Tumor v olume was calculated as the length x width x height (in mm 3 ). (B) Survival rates monitoring after chal lenge. The asterisks indicate significant differences compared with naive mice ( **P<0.01 ) (n=6) (Comparison of survival curves, Mantel-Cox test).

Figure 4 is a set of graphs showing the effect of pTOP l -OVA CD8 and pTOP l - OVA CD4 prophylactic intramuscular immunization on the anti-tumor activity. (A) Tumor growth fol low-up after challenge. The tumor size was measured three times a week with an electronic digital caliper. Tumor volume was calculated as the length x width x height (in mm 3 ). (B) Survival rates monitoring after challenge. The asterisks indicate significant differences compared with naive mice (***P<0.001) (n=6) (Comparison of survival curves, Mantel-Cox test).

Figure 5 is a set of graphs showing the effect of pTOP l -OVA CD8 and pTOPl- OVA CD4 therapeutic intramuscular immunization on the anti-tumor activity. (A) and (C) Tumor growth follow-up after challenge. (B) and (D) Survival rates monitoring after challenge. Survival curves were compared with a Mantel-Cox test. The asterisks indicate significant differences compared with naive mice ( ***P<0.001 ) (n=10 and n=6 respectiv ely).

Figure 6 is a graph showing the effect of co-delivery of pTOP l -OVA CD4 with pTOP l -OVA CDS on the cytotoxic T cell response. Percentages of OVA target cell killing were compared and the asterisks indicate significant differences (***P<0.001) (n=5) (Student's T-test).

Figure 7 is a graph showing an ΟΊ I I proliferation assay and effect of immunization with MHC class II restricted epitope inserted in pTOPl . The percentages of cell division were compared by Student's T-test (***p<0.001) (n=5).

Figure 8 is a set of graphs showing OTI proliferation assay and the effect of immunization with MHC class I restricted epitope inserted in pTOP l . The graph shows the percentages of cell division. The asterisks indicate significant differences ( ***P< 0.001 ) (n=5) (Student's T-test).

Figure 9 is a set of graphs showing the effect of pTOPl intramuscular therapeutic immunization in combination with immune checkpoint blockade (ICB) therapy. (A) Tumor growth fol low -up after challenge. Tumor volume was calculated as the length x width x height (in mm 3 ). (B) Survival rates monitoring after challenge. The asterisks indicate significant differences between curves (*P<0.05; ***P<0.001 ) (n=6) (Comparison of survival curves, Mantel-Cox test). Figure 10 is a set of graphs showing the effect of pTOPl- OVA CD4( 1 8 ) OVA_CD8( 1 91 ) and pTOP 1 gp 1 00 CD4( 1 8 ) TRP2 CD8( 191 ) therapeutic intramuscular immunization on the anti-tumor activ ity. (A) Tumor growth fol low-up after chal lenge. The tumor size was measured three times a week with an electronic digital caliper. Tumor volume was calculated as the length x width x height (in mm 3 ). (B) Survival rates monitoring after challenge. The asterisks indicate significant differences compared with naive mice (**P<0.01 ; ***P<0.001 ) (n=6) (Comparison of survival curves. Mantel -Cox test).

Figure 11 is a set of graphs showing the effect of pTOP 1 -PADRE( 1 8 ) P 1 A CD8( 1 91 ) prophylactic intramuscular immunization on the anti-tumor activity. (A) Tumor growth follow -up after challenge. The tumor size was measured three times a week with an electronic digital cal iper. Tumor v olume was calculated as the length x width x height (in mm 3 ). (B) Survival rates monitoring after challenge. The asterisks indicate significant differences compared with naive mice (**P<0.01) (n=6) (Comparison of survival curves, Mantel-Cox test).

Figure 12 is a graph showing the effect of pTOP 1 -PADRE( 1 8 ) P 1 A CD8( 191 ) therapeutic intramuscular immunization on the anti-tumor activity. It indicates survival rate monitoring after challenge. The asterisk indicates significant differences compared with naive mice ( *P<0.05 ) (n=6) (Comparison of survival curves, Mantel-Cox test). Figure 13 is a set of graphs showing the effect of pTOPl- PADRE( 1 8 ) AH 1 A5 CD8( 191 ) prophylactic intramuscular immunization on the antitumor activ ity. (A) Tumor growth follow -up after challenge. The tumor size was measured three times a w eek with an electronic digital cal iper. Tumor volume was calculated as the length x width x height (in mm " ). (B) Survival rates monitoring after challenge. The asterisks indicate significant differences compared with naive mice (***P<0.001) (n=6) (Comparison of survival curves, Mantel-Cox test).

Figure 14 is a set of graphs show ing the effect of pTOP l - PADRE( 1 8 ) TRP2 CD8( 191 ) prophylactic intramuscular immunization on the antitumor activ ity. (A) Tumor grow th fol low-up after challenge. The tumor size was measured three times a week with an electronic digital caliper. Tumor volume was calculated as the length x width x height ( in mm 3 ). (B) Survival rates monitoring after challenge. The asterisks indicate significant differences compared with naive mice (***P<0.001) (n=6) (Comparison of survival curves, Mantel-Cox test ). Figure 15 is graph showing the effect of pTOP 1 -gp 100 CD4( 1 8 ) OVA CD8( 1 91 ) and pTOP l gp l OO LP ( 1 8 ) OVA CD8( 1 9 l ) therapeutic intramuscular immunization on the an ti -tumor activity. It indicates survival rates monitoring after chal lenge. The asterisks indicate significant differences compared with naive mice (*P<0.05; **P<0.01 ) (n=6) (Comparison of survival curves, Mantel-Cox test).

EXAMPLES

The present invention is further illustrated by the fol lowing examples.

Materials and Methods

Material Plasmids

Codon-optimized gene sequences of VSV-G (pTOP), VSV-G-OVA CD8 (pTOP- OVA CD8 ) and VSV-G-RS (with restriction sites, pTOPl) were designed using GeneOptimizer and obtained by standard gene synthesis from GeneArt® (Thermo Fisher Scientific, Waltham, MA, US ). These sequences were subcloned in the pVAX2 vector using cohesive-ends cloning. The pVAX2 vector consists of a pVAX 1 plasmid ( Invitrogen, Carlsbad, CA) in which the promoter was replaced by the pCMVP plasmid promoter (Clontech, Palo Alto, CA ). The plasmids were prepared using the EndoFree Plasmid Giga Kit (Qiagen, Venlo, Netherlands) according to the manufacturer ' s protocol. Plasmid dilutions were performed in Dulbecco's Phosphate Buffered Saline (l x) (PBS) (Life Technologies, Carlsbad, CA, US). The quality of the purified plasmid was assessed by the ratio of optical densities (260 nm 280 nm ) and by 0.5% agarose gel electrophoresis. DNA concentration was determined by optical density at 260 nm. The plasmids were stored at -20 C. VSV-G sequences cloned in pVAX2

Vesicular stomatitis Indiana virus glycoprotein G (VSV-G ) (SEQ ID NO: 1 , encoded by SEQ ID NO: 10).

Plasmid nomenclature: pVAX2-VSVG (pTOP).

MKCLLYLAFLFIGVNCKFTIVFPHNQKGNWKNVPSNYHYCPSSSDLNWHNDLI G T A 10 V K M P K S 11 K A I Q A D G W M C H ASK W V" IT C D F R W Y G P K Y I TQ SI RS FT P S V E

QCKESIEQTKQGTWLNPGFPPQSCGYATVTDAEAVIVQVTPHHVLVDEYTGEW VDSQFINGKCSNYICPTVHNSTTWHSDYKVKGLCDSNLISMDITFFSEDGELSSL GKEGTGFRSNYFAYETGGKACKMQYCKHWGVRLPSGVWFEMADKDLFAAAR FPECPEGSSISAPSQTSVDVSLIQDVERILDYSLCQETWSKIRAGLPISPVDLSYLA PKNPGTGPAFTIINGTLKYFETRYIRVDIAAPILSRMVGMISGTTTERELWDDWA PYEDVEIGPNGVLRTSSGYKFPLYMIGHGMLDSDLHLSSKAQVFEHPHIQDAAS QLPDDESLFFGDTGLSKNPIELVEGWFSSWKSSIASFFFIIGLIIGLFLVLRVGIHLC IKLKHTKKRQIYTDIEMNRLGK.

- VSV-G (SEQ I D NO: 1) containing SIINFEKL sequence (OVA CDS, SEQ I D NO: 1 1) at position 191 (SEQ ID NO: 8).

Plasmid nomenclature: pV AX2-VSVG-OV A CD8 (pTOP-OVA CD8 ).

MKCLLYLAFLFIGVNCKFTIVFPHNQKGNWKNVPSNYHYCPSSSDLNWHNDLI GTAIQVKMPKSHKAIQADGWMCHASKWVTTCDFRWYGPKYITQSIRSFTPSVE QCKESIEQTKQGTWLNPGFPPQSCGYATVTDAEAVIVQVTPHHVLVDEYTGEW VDSQFINGKCSNYICPTVHNSTTWHSDYKVKSIINFEKLGLCDSNLISMDITFFS EDGELSSLGKEGTGFRSNYFAYETGGICACKMQYCKHWGVRLPSGVWFEMAD KDLFAAARFPECPEGSSISAPSQTSVDVSLIQDVERILDYSLCQETWSKIRAGLPI SPVDLSYLAPKNPGTGPAFTIINGTLKYFETRYIRVDIAAPILSRMVGMISGTTTE REL WDD WAP YED VEIGPNG VLRT S S G YKFPL YMIGHGMLD SDLHL S SKAQ VFE HPHIQDAASQLPDDESLFFGDTGLSKNPIELVEGWFSSWKSSIASFFFIIGLIIGLFL VLRVGIHLCIKLKHTKKRQIYTDIEMNRLGK.

(in bold underlined is the OVA CDS sequence, SEQ ID NO: 1 1). - VSV-G (SEQ I D NO: 1) containing restriction sites (RS ) at position 1 91

(SEQ ID NO: 9).

Piasmid nomenclature: pVAX2-VSVG-RS (pTOPl).

MKCLLYLAFLFIGVNCKFTIVFPHNQKGNWKNVPSNYHYCPSSSDLNWHNDLI GTAIQVKMPKSHKAIQADGWMCHASKWVTTCDFRWYGPKYITQSIRSFTPSVE QCKESIEQTKQGTWLNPGFPPQSCGYATVTDAEAVIVQVTPHHVLVDEYTGEW VDSQFINGKCSNYICPTVHNSTTWHSDYKVKTSEFGLCDSNLISMDITFFSEDGE LSSLGKEGTGFRSNYFAYETGGKACKMQYCKHWGVRLPSGVWFEMADKDLF AAARFPECPEGSSISAPSQTSVDVSLIQDVERILDYSLCQETWSKIRAGLPISPVD LS YLAPKNPGTGPAFTIINGTLKYFETRYIRVDIAAPILSRMVGMISGTTTERELW DDWAPYEDVEIGPNGVLRTSSGYKFPLYMIGHGMLDSDLHLSSKAQVFEHPHI QDAASQLPDDESLFFGDTGLSKNPIELVEGWFSSWKSSIASFFFIIGLIIGLFLVLR VGIHLCIKLKHTKKRQIYTDIEMNRLGK.

(in bold underlined are the SpeVEcoBl restriction sites). Peptide insertion in pTOPl

To insert epitopes in position 1 91 of VSV-G (SEQ ID NO: 1), into the pTOPl vector, cohesive-ends cloning was used, p V A X 2 - V S V G - R S was opened using Spel and EcoEl and two complementary and overlapping phosphorylated oligonucleotides were incorporated. Multiple plasmids w ere obtained by varying the sequence of the oligonucleotides which w ere ordered from Eurogentec (Seraing, Belgium ) or IDT-DNA (Leuven, Belgium). For peptide insertion in position 18 of pTOPl , Gibson Assembly Cloning Kit (New England Bio Labs Inc. ) with gBlocks gene fragments was used according to the manufacturer instructions. A Hindlll restriction site was added for allowing easy peptide modification at the position 18. Plasmids w ere then purified, characterized and stored as explained here above. Table 3 - Peptides inserted in pTOP-1 by cohesive-ends cloning at position 191 of SEQ ID NO: 1. Peptide sequence, name and function are described.

Table 4 - Peptides inserted in pTOPl by gB locks cloning at position 18 of SEQ ID NO: 1. Peptide sequence, name and function are described.

List of constructs

Table 5 - List of chimeric VSV-G used in the present invention. Given are their amino acid sequence ID and nucleic acid sequence ID.

Cell culture

B 16F 1 0-OVA, a melanoma cel l l ine from C57BL 6 mice that stably expresses ovalbumin, was cultured in MEM medium supplemented with GlutaMAX with 10% I BS, 1 00 iig/ml, streptomycin and 100 U/mL penicill in ( Life Technologies, Carlsbad, CA, US).

B 16F10, a melanoma cel l l ine from C57BL/6 mice, was cultured in MEM medium supplemented with GlutaMAX with 10% FBS, 100 ng ml, streptomycin and 100 U/mL penicillin (Life Technologies, Carlsbad, CA, US).

CT26, a colon carcinoma cell l ine from BALB/C mice, was cultured in DM EM with 10% FBS, 1 00 μg/mL streptomycin and 100 U/mL penicill in, and supplemented with L-glutamate and pyruvate ( Life Technologies, Carlsbad, CA, US).

P815, a mastocytoma ceil l ine from DBA/2 mice, was cultured in DM EM with 10% FBS, 100 pg ml , streptomycin and 100 U/mL penicill in ( Life Technologies, Carlsbad, CA, US ). Animals

Six to eight-week-old C57BL/6, BA LB/C and DBA/2 female mice were obtained from

Janvier Labs (Le Genest Saint Isle, FR) and housed in a minimal disease facility with ad libitum access to food and water.

For tumor implantation and electroporation, the mice were anaesthetized by intraperitoneal ( ip ) injection of 150 μ L of a solution of 10 mg 'm L ketaminc and 1 mg ml, xyiazine. The ethical committee for Animal Care and Use of the Medical Sector of the Universite Cathol ique de Louvain approved our experimental protocols

(UCL/MD/201 1/007 and UCL/MD/2016/001). Methods

Immunization

After removing the hair using a rodent shaver (AgnTho's, Lidingo, Sweden ), 1 μg or 50μg of piasmid were injected, diluted in 30 ill, of PBS, into the left tibial cranial muscle. Immediately after injection, the leg was placed between 4-mm-spaced. plate electrodes ( BTX Caliper Electrodes), and 8 square-wave electric pulses (80 V, 20 ms, 2 Hz) were delivered by a Gemini System generator ( BTX; both from VWR International, Leuven, Belgium ). A conductive gel was used to ensure electrical contact with the skin (Aquasonic 1 00; Parker Laboratories, Inc., Fairfield, NJ, USA ). For prophylactic vaccination experiments, two boosts (i.e. , second and third administrations of the vaccine) were similarly applied two and four weeks after the priming.

For therapeutic vaccination experiments, the treatment started two days after the injection of the tumor cells and the two boosts were delivered every week. Alternatively, plasmids were injected and electroporated into the tumors when they reached a size in-between 30 and 50 mm 3 . This treatment was then repeated after two days.

For the study of the OT-I and OT-II proliferation, plasmids were injected into ears and 2-mm-spaced electrodes were applied to deliver 10 square-wave electric pulses ( 100 V. 20 ms, 1 Hz).

Tumor implantation

1 x 10 5 B16F10-OVA or B16F10 cells, diluted in 100 μΐ, PBS, were injected subcutaneously into the right flank of each C57BL/6.

1 x 10 6 CT26 cells, diluted in 100 μL PBS, were injected subcutaneously into the right flank of each BA LB/C .

1 x 10 6 P815 cells, diluted in 1 00 μL PBS, were injected subcutaneously into the right flank of each DBA/2.

Tumor cells were implanted two days before the first plasmid administration or two weeks after the last administration for therapeutic and prophylactic DNA immunization studies, respectively. The tumor size was measured three times a week with an electronic digital cal iper. Tumor volume was calculated as the length x width x height

(in mm 3 ). The mice were sacrificed when the volume of the tumor reached 1500 mm 3 or when they were in poor condition and expected to die shortly.

Administration of immune checkpoint blockade (ICB) antibodies

For administration of 1CB, mice received 100 Lig of InVivoMAb anti-mouse CTLA-4 (CD 152) clone 9D9 and 100 μg of InVivoMAb anti-mouse PD-1 (CD279) clone 29F. 1 A 12, both from BioXceil (CT, US ) by intraperitoneal injection in 200 id, of PBS at day 3, 6 and 9 fol low ing implantation of the B 16F 10-OVA cel ls.

OT-I and OT-II proliferation

T cells were isolated from spleen and lymph nodes of transgenic OT-I and OT-II mice using CDS + and CD4+ Tcell isolation kit 11 mouse (Miltenyi Biotec, The Netherlands). Subsequently the T cells were labeled with CFSE (carboxy fluorescein diacetate suecinimidyl ester; Molecular probes) by incubating 50 χ 10 6 cells/mL with 5 μΜ CFSE for 7 minutes at 37°C. The reaction was blocked by adding ice-cold PBS (Lonza, Belgium ) + 10% serum. 2x l 0 6 OT-I or OT-I I cel ls were injected into the tail vein of C57BL/6 mice. They were treated 2 days later by plasmid injection and electroporation. M ice were sacrificed 4 days later to col lect the draining lymph nodes for single cel l suspension preparation. Flow cytometric measurement was performed after staining w ith aqua live dead ( Inv itrogcn, Belgium), CD 19 APC-Cy7, CDS PerCP (all BD Biosciences), dextramer SIINFEKL H-2kb PE ( Immudex, Denmark). In vivo killing assay

Splenocytes from naive mice were pulsed with SIINFEKL peptide or with an irrelevant peptide (40μg in 40 ml, PBS) for one hour at 37°C. Subsequently, these pulsed splenocytes were washed and respectively stained with high (5 μΜ, hi) or low (0,5 μΜ, low ) CFSE concentration. The two populations of splenocytes were mixed in a 1 : 1 ratio, and 10 7 splenocytes were intravenously injected into immunized mice two weeks after the last booster immunization. Two days after transfer, the spleens of the host mice were isolated and analyzed by flow cytometry after staining with α-F4/80 (BD Biosciences, San Diego, CA, USA) to exclude auto-fiuorescent macrophages. The percentage antigen-specific kill ing was determined using the fol lowing formula:

Example 1: The effect of pTOP-OVA_CD8( 191 ) prophylactic intramuscular immunization on the anti-tumor activity B 16 melanoma is a spontaneous melanoma derived from C57BL/6 mice. The most commonly used variant is B 16F10, which is highly aggressive and will metastasize from a primary subcutaneous site to the lungs, as well as colonize lungs upon intravenous (iv) injection.

C57BL/6 mice were immunized in a regimen of one prime and two boosts at a 2-week interval with the pTOP-OVA C D8( 191 ) plasmid (1 tig). Two weeks after the last vaccination, they were challenged with B 16F 10-OVA. cells. This B 16F 10-OVA. cel l line is a stable trans fectant derived from B 16F10 melanoma that stably expresses chicken ov albumin.

Tumor growth, and mouse survival were assessed for three months. Inoculation of B 16F 1 0-OVA. cel ls induced tumors that grow rapidly and killed naive mice. How ev er, prophylactic immunization by intramuscular eiectroporation of a plasmid encoding VSV-G containing a tumor model CD8 T cell epitope delayed tumor growth and improved mice survival ( Figure 1). Example 2: The effect of pTOP-OVA CD8( 191 ) therapeutic intratumoral immunization on the anti-tumor activity

C57BL/6 mice were chal lenged with B16F10-OVA cells. When tumor reached between 30 and 50 mm 3 , mice were immunized twice with a two-day interval with the pTOP-OVA CD8( 1 91 ) plasmid, the pTOP control plasmid (expressing VSV-G of SEQ I D NO: 1 without inserted peptide) or the empty pVAX2 (pEmpty ) plasmid (50 iig each).

Therapeutic immunization by intratumoral electroporation of a plasmid encoding VSV-G containing a tumor model CDS T cell epitope delays tumor growth ( Figure 2). Example 3: The effect of restriction sites addition around the inserted epitope sequence on vaccine efficacy

C57BL/6 mice were immunized in a regimen of one prime and two boosts at a 2-week interval with the pTOP-OVA CD8( 191 ) plasmid or the pTOP l -OVA CD8( 1 91 ) plasmid (1μg each ). Two weeks after the last vaccination, they were chailenged with B 16F10-OVA cells. Tumor growth and mouse surv ival were assessed.

The addition of Spel and EcoBJ restriction sites introduce amino acids TS and EF around the inserted epitope. This result showed that adding these amino acids around the T cel l epitope does not alter vaccine efficacy ( Figure 3).

Example 4: The effect of pTOPl-OVA CD8( 191 ) and pTOPl-OVA C D4( 191 ) prophylactic intramuscular immunization on the anti-tumor activity

Insertion of a CD8 T cell epitope in VSV-G is necessary to observe anti-tumor efficacy. There is no anti-tumor effect fol lowing pTOP and pTOP 1 -OVA CD4( 191 ) del ivery. Prophylactic immunization by intramuscular electroporation of two pTOP l plasmids containing respectively OVA CDS and OVA CD4 T cell epitopes improve protection against tumor challenge as compared to pTOP 1 -OVA CD8( 1 91 ) alone. The tumor grow th delay and mice survival are improved when the helper epitope is co-delivered with the MHC class I restricted epitope (Figure 4). Example 5: The effect of pTOPl-OVA CD8(191) and pTOPl-OVA CD4(191) therapeutic intramuscular immunization on the anti-tumor activity

C57BL/6 mice were challenged with B 16F 10-OVA cells. Two days later, they were immunized in a regimen of one prime and two boosts at a 1-week interval with 1 oμfg the pTOPl-OVA CD8(191) alone or combined with 1μg of the pTOPl- OVA CD4( 191 ) plasmid. Tumor growth and mouse survival were assessed.

Therapeutic immunization by intramuscular electroporation of two pTOPl plasmids containing respectively CD8 and CD4 T cell epitopes improves protection against tumor challenge. Two separate experiments have been performed. First, it was shown that therapeutic immunization with pTOP 1 -OVA CD8( 191) tends to improve protection against challenge (but the effect is not significant). Second, the combination of pTOPl- OVA CD4(191) and pTOPl-OVA CD8(191) drastically improved mice survival and delayed tumor growth (Figure 5).

Example 6: The effect of co-delivery of pTOPl-OVA C 1)4(191) with pTOP- OV A CD8(191) on the cytotoxic T cell response

C57BL/6 mice were immunized in a regimen of one prime and two boosts at a 2-week interval with 1 μg of the pTOP 1 -OVA CD8( 19l) plasmid alone or combined with 1 iig of the pTOP 1 -OVA CD4( 191) plasmid. The percentage of antigen specific killing was analyzed by in vivo cytotoxic assay. Immunized mice were adoptively transferred with two populations of labelled spienocytes: MHC-I OVA peptide-pulsed-target cells and a MHC-I irrelevant-peptide-pulsed cells. Two days after transfer, the specific killing of target cells was obtained by comparing the relative decrease of the two populations.

An in vivo killing assay demonstrated that co-deliv ery of pTOP 1 -OVA_CD8( 191) and pTOP 1 -OVA CD4( 191) improves the cytotoxic T cell response to the vaccine antigen as compared to delivery of pTOPl-OVA CD8(191 ) alone (Figure 6).

Example 7: OT-II proliferation assay

The effect of immunization with MHC class II-restricted epitope inserted in pTOPl on the CD4+ T cell response has been demonstrated using OT-II cells. T cells were isolated from spleen and lymph nodes of transgenic OT-II mice, labeled with CFSE and adoptively transferred to C57BL 6 mice. Mice were immunized two days later with 1 μg of pTOP l -OVA CD40 91 ) or 1 μg of pTOP l -OVA CD8( 191 ). Mice were sacrificed four days later and labelled T cell proliferation was assessed. The insertion of MHC class Il-restricted epitopes in VSV-G-induced CD4+ T cell response, whereas MHC class I-restricted epitopes are unable to induce helper response ( Figure 7).

Example 8: OT-I proliferation assay

The effect of immunization with MHC class I-restricted epitope inserted in pTOPl on the CD8+ T cell response has been demonstrated using OT-I cells. T cells were isolated from spleen and lymph nodes of transgenic OT-I mice, labeled with CFSE and adoptively transferred to receptor C57BL 6 mice. Mice were immunized two days later by eiectroporation of pTOP l -OVA CD4( 191 ) (1 μg) or pTOP l -OVA CD8( 191 ) (1 fig). Mice were sacrificed four days later and labelled T cel l proliferation was assessed.

The insertion of MHC class I-restricted epitopes in VSV-G induced CD8+ T cell response, whereas MHC class Il-restricted epitopes are unable to induce CD8+ T cell response (Figure 8).

Example 9: The effect of pTOPl immunization in combination with immune checkpoint blockade (ICB) therapy

C57BL 6 mice were challenged with B 1 6F 10-OVA cells. Two days later, they were immunized in a regimen of one prime and two boosts at a 1-week interval. On day 3, 6 and 9 fol lowing challenge, the ICB treatments were given. Mice received either

(1) both pTOP l -OVA CD8( 19 I ) (1 iig) and pTOP l -OVA CD4( 191 ) (1 iig) piasmids;

(2) a cocktail of anti-PD-1 and anti-CTLA-4 antibodies [ICB group]; or

(3) a combination of the two piasmids (1 μg each) and the antibodies cocktail

[combination group]. Tumor growth and mice surv ival were assessed following challenge.

Efficacy of pTOPl is further enhanced by combination with immune checkpoint blockade therapy. These results demonstrated that the combinatory treatment has a synergic effect compared to treatments alone. Indeed, survival, tumor growth and tumor volume observed after the combinatory treatment are better than the sum of effects obtained after separate treatments (Figure 9).

Example 10: The effect of p TO P 1 -OVA C D4( 18)_0 V A C D8( 191 ) and pTOPl- gp 100 C D4( 18) TR P2 C D8( 191 ) therapeutic intramuscular immunization on the anti-tumor activity C57BL 6 mice were challenged with B 16F 10-OVA. cells. Two days later, they were immunized in a regimen of one prime and two boosts at a 1-week interv al with 1 ^tg of the pTOP 1 -OVA CD4( 1 8 ) OVA CD8( 191 ) plasmid or 1 μg of the pTOPl- gp 100 CD4( 1 8 ) TRP2 CD8( 191 ) plasmid. Tumor growth and mouse survival were assessed. Therapeutic immunization by intramuscular eiectroporation of pTOPl- OVA CD4( 1 8 ) OVA CD8( 191 ) plasmid or pTOP 1 -gp 1 00 CD4( 1 8 ) TRP2 CD8( 191 ) was able to significantly delay tumor growth. There was no statistical di fference between the two vaccines ( Figure 10).

Example 11 : The effect of p TO P 1 - P A DR E( 18)_P 1 A C D8( 191 ) prophylactic intramuscular immunization on the anti-tumor activity

DBA/2 mice were immunized in a regimen of one prime and two boosts at a 2-week interval with the pTOP 1 -PADRE( 1 8 ) P 1 A CD8( 191 ) plasmid (1 μg). Two weeks after the last v accination, they were challenged with P8 1 5 cells. Tumor growth and mouse surviv al were assessed for two months. Inoculation of P8 1 5 cells induced tumors that grow rapidly and kil led naive mice. However, prophylactic immunization by intramuscular eiectroporation of a plasmid encoding VSV-G containing a tumor model CD8 T cel l epitope and a universal antigenic

CD4 T cell epitope delayed tumor growth and improved mice survival (Figure 11). Example 12: The effect of pTO P 1 - PA D R F( 18)_P 1 A C D8( 191 ) therapeutic intramuscular immunization on the anti-tumor activity

DBA/2 mice were challenged with PS 1 5 cells. Two days later, they were immunized in a regimen of one prime and two boosts one and two weeks later with the pTOPl- PADRE( I S ) P I A CDS( 1 91 ) plasm id (1 Lig). M ice surv ival was assessed for two months.

Therapeutic immunization by intramuscular electroporation of pTOPl- PADR E( 1 S ) P 1 A CDS( 1 91 ) plasm id was able to significantly delay tumor growth. (Figure 12). Example 13: The effect of pTOPl -PADRE( 18)_AH 1 A5_CD8( 191 ) prophylactic intramuscular immunization on the anti-tumor activity

BALB/C mice were immunized in a regimen of one prime and two boosts at a 2-week interv al with the pTOP 1 -PADRE( I S ) AH 1 A5 CDS( 1 9 l ) plasmid (1 iig). Two weeks after the last vaccination, they were challenged with CT26 cells. Tumor growth and mouse survival were assessed for two months.

Inoculation of CT26 cells induced tumors that grow rapidly and killed naive mice. However, prophylactic immunization by intramuscular electroporation of a plasmid encoding VSV-G containing a tumor model CDS T cell epitope and a universal antigenic CD4 T cell epitope delayed tumor growth (Figure 13). Example 14: The effect of pTO P 1 - PA DR E( 18)_TR P2 C D8( 191 ) prophylactic intramuscular immunization on the anti-tumor activity

BALB/C mice were immunized in a regimen of one prime and two boosts at a 2-week interv al with the pTOP 1 -PADR E( 1 S ) TRP2 CDS( 191 ) plasmid (1 ). Tμwgo weeks after the last v accination, they were challenged with B16F10 cei ls. Tumor growth and mouse surviv al were assessed for two months.

Inoculation of B16F10 cells induced tumors that grow rapidly and killed naive mice. However, prophylactic immunization by intramuscular electroporation of a plasmid encoding VSV-G containing a tumor model CD8 T cel l epitope and a universal antigenic CD4 T cell epitope delayed tumor growth and improved mice survival ( Figure 14).

Example 15: The effect of pTO P 1 -gp 100_C D4( 18)_0 V A C D8( 191 ) and pTOPl - gp 100 L P( 18) OVA C D8( 191 ) therapeutic intramuscular immunization on the anti-tumor activity

C57BL 6 mice were challenged with B 1 6F 10-OVA cel ls. Two days later, they were immunized in a regimen of one prime and two boosts at a 1-week interval with 1 oμfg the pTOP I -gp 1 00 CD4( 1 8 ) OVA CD8( 191 ) plasmid or 1 μg of the pTOP l - gp 1 00 LP( 1 8 ) OVA CD8( 191 ) plasmid. Tumor growth and mouse survival were assessed.

Therapeutic immunization by intramuscular electroporation of pTOP l - gp 100 CD4( 1 8 ) OVA CD8( 191 ) plasmid or pTOP 1 -gp 100 LP( 1 8 ) OVA CD8( 191 ) was able to significantly delay tumor growth. There was no statistical di fference between the two vaccines ( Figure 15).