WO2024023135A1 - Vecteurs lentiviraux pour l'expression d'antigènes du papillomavirus humain (hpv) et leur mise en oeuvre dans le traitement de cancers induits par le hpv - Google Patents

Vecteurs lentiviraux pour l'expression d'antigènes du papillomavirus humain (hpv) et leur mise en oeuvre dans le traitement de cancers induits par le hpv Download PDF

Info

Publication number
WO2024023135A1
WO2024023135A1 PCT/EP2023/070672 EP2023070672W WO2024023135A1 WO 2024023135 A1 WO2024023135 A1 WO 2024023135A1 EP 2023070672 W EP2023070672 W EP 2023070672W WO 2024023135 A1 WO2024023135 A1 WO 2024023135A1
Authority
WO
WIPO (PCT)
Prior art keywords
lentiviral vector
acid sequence
nucleic acid
protein
antigen
Prior art date
Application number
PCT/EP2023/070672
Other languages
English (en)
Inventor
Pierre Charneau
François ANNA
Fanny MONCOQ
Amandine NOIRAT
Majlessi LALEH
Laetitia DOUGUET
Ingrid FERT
Original Assignee
Theravectys
Institut Pasteur
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Theravectys, Institut Pasteur filed Critical Theravectys
Publication of WO2024023135A1 publication Critical patent/WO2024023135A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to the field of recombinant vaccine technology and relates to improvements of lentiviral vectors, which can be used as therapeutic and prophylactic vaccines.
  • the present invention relates to lentiviral vectors expressing Human Papillomavirus (HPV) antigens and to their implementation in the prevention and treatment of HPV induced cancers.
  • HPV Human Papillomavirus
  • HPV is responsible for 5.2% of cancers throughout the world (Tota et al., Prev Med 2011 Oct; 53 Suppl 1:S 12-21). More than 100 HPV types have been identified and have been classified into 3 groups depending on their association with cancer: high risk-types with a high oncogenic potential (among them, HPV type 16 (HPV- 16) and HPV type 18 (HPV- 18)), low risk-types that are associated with benign lesions (HPV-6, -11) and cutaneous types (among them, HPV-1, -2, -3, -4...) (Chen et al. Virology vol. 516 (2018): 86-101).
  • HPV associated cancer varies upon cancer type and geography, but it is estimated that 90 % of cervical, 91 % of anal, 75% of vaginal, 70% of oropharyngeal, 69% of vulvar and 63% of penile cancers are related with HPV infections (Saraiya et al., J Natl Cancer Inst. 2015 Apr 29; 107(6)).
  • HPV 16 and HPV18 are thought to be involved in 70-75% of all cervical cancers (de Sanjose et al., Eur J Cancer. 2013 Nov;49(16):3450-61). HPV16 and 18 are also largely involved in anal (91%), oropharyngeal (70%), vaginal (75%), penile (63%) and vulvar (68%) cancers (Saraiya et al., J Natl Cancer Inst. 2015 Apr 29; 107(6)).
  • a therapeutic vaccine targeting HPV16/18 could potentially be used to treat and prevent related cancers, irrespective of their location.
  • HPV Human papillomaviruses
  • E6 and E7 have been well characterized for their oncogenicity. They are known to inactivate p53 and pRb tumour suppressor proteins, thereby promoting cell proliferation. E6/E7 oncogenes are crucial for both the induction of HPV- linked malignant cell transformation and the maintenance of the oncogenic phenotype of HPV-positive cancer cells (Yim and Park, Cancer Res Treat. 2005 Dec;37(6):319-24.). The E6 and E7 proteins are expressed throughout the infection by all HPV positive cells and this observation makes them perfect targets for vaccines.
  • Recombinant viral vectors have been widely developed for vaccination purposes. Modification of viral genomes allowed the production of non-toxic and non- infectious viral particles that can be used as tools to introduce genetic material into target cells. The use of recombinant viral vectors to elicit a T-cell mediated immunity is a very promising approach for vaccination.
  • a variety of viral vectors have been evaluated for vaccination purposes including retroviruses vectors, adenoviruses vectors and vaccinia virus vectors (Milone and O’Doherty, Leukemia (2016) 32:1529-1541 and Ku et al., Expert Review of Vaccines (2021). Lentiviruses are part of the Retroviridae family, which includes the human immunodeficiency viruses (HIV).
  • Lentiviral vectors are mainly derived from HIV- 1. They have been improved in their safety by removal of the LTR U3 sequence, resulting in “self-inactivating” vectors that are entirely devoid of viral promoter and enhancer sequences. Lentiviral vectors have emerged as promising tools because they exhibit several advantages over other viral systems.
  • lentiviral vectors are not toxic and, unlike other retroviruses, are capable of transducing non-dividing cells, in particular dendritic cells (He et al. 2007, Expert Rev vaccines, 6(6):913-24), allowing a sustained antigen presentation through the endogenous pathway.
  • lentiviral vectors have the capacity to transduce non-dividing cells. Efficient transduction in non-dividing cells requires the formation of a triple-stranded DNA structure called the central DNA “flap”, which maximizes the efficiency of gene import into the nuclei of non-dividing cells, including dendritic cells (DCs) (Arhel et al., EMBO J. 2007 Jun 20; 26(12): 3025-3037) (Zennou et al., Cell. 2000 Apr 14; 101(2): 173-85).
  • DCs dendritic cells
  • DCs Dendritic cells
  • APCs antigen presenting cells
  • Mature DCs migrate to the draining lymph node and where they present the antigen-derived short peptides at the surface through Major Histocompatibility Complex (MHC) molecules.
  • MHC Major Histocompatibility Complex
  • Antigen- specific T cells present in the lymph nodes can then interact with peptide-MHC complexes through TCR (T Cell Receptor). The recognition of peptide-MHC by specific TCR in conjunction with co- stimulatory signals, initiates T cell activation (Steinman, R., Banchereau, J. Nature 449, 419-426 (2007)).
  • the aim of the present invention is to provide therapeutic and prophylactic vaccines for the prevention and treatment of HPV-induced cancers.
  • Tregs Regulatory T cells
  • the invention has for purpose to meet the above-mentioned needs.
  • Item 1 a lentiviral vector, in particular a non-integrative lentiviral vector, comprising at least four distinct nucleic acid sequences selected from the group consisting of:
  • lentiviral vectors of the invention allow for a strong therapeutic and prophylactic activity against HPV-induced tumors.
  • Item 2 The lentiviral vector according to item 1, wherein the nucleic acid sequence encoding the non-oncogenic Human papillomavirus (HPV16) protein E6 antigen encodes an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 7, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6.
  • the nucleic acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 7, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6.
  • Item 3 The lentiviral vector according to item 1 or 2, wherein the nucleic acid sequence encoding the non-oncogenic Human papillomavirus (HPV16) protein E7 antigen encodes an amino acid sequence having at least 68% sequence identity with the amino acid sequence set forth as SEQ ID NO: 16, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 14 and SEQ ID NO: 15.
  • HPV16 Human papillomavirus
  • Item 4 The lentiviral vector according to any one of items 1 to 3, wherein the nucleic acid sequence encoding the non-oncogenic Human papillomavirus (HPV18) protein E6 antigen encodes an amino acid sequence having at least 60% sequence identity with the amino acid sequence set forth as SEQ ID NO: 24, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22 and SEQ ID NO: 23.
  • the nucleic acid sequence having at least 60% sequence identity with the amino acid sequence set forth as SEQ ID NO: 24, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22 and SEQ ID NO: 23.
  • Item 5 The lentiviral vector according to any one of items 1 to 4, wherein the nucleic acid sequence encoding the non-oncogenic Human papillomavirus (HPV18) protein E7 antigen encodes an amino acid sequence having at least 83% sequence identity with the amino acid sequence set forth as SEQ ID NO: 33, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 30, SEQ ID NO: 31 and SEQ ID NO: 32.
  • the nucleic acid sequence having at least 83% sequence identity with the amino acid sequence set forth as SEQ ID NO: 33, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 30, SEQ ID NO: 31 and SEQ ID NO: 32.
  • Item 6 The lentiviral vector according to any one of items 1 to 5, wherein the at least four distinct nucleic acid sequences encoding antigens are fused together, forming a single antigenic nucleic acid sequence encoding a single antigenic fusion protein under the control of a single promoter sequence.
  • Item 7 The lentiviral vector according to any one of items 1 to 6, wherein the order of the at least four distinct nucleic acid sequences, from 5’ end to 3’ end, is selected from the group consisting of:
  • Item 8 The lentiviral vector according to any one of items 1 to 7, wherein the order of the at least four distinct nucleic acid sequences, from 5’ end to 3’ end, is (a) nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV16) protein E7 antigen - nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV16) protein E6 antigen - nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E7 antigen - nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E6 antigen.
  • HPV16 non-oncogenic Human papillomavirus
  • HPV18 non-oncogenic Human papillomavirus
  • HPV18 non-oncogenic Human papillomavirus
  • Item 9 The lentiviral vector according to any one of items 1 to 8, comprising a nucleic acid sequence which encodes an amino acid sequence having at least 90% sequence identity with the amino acid sequence set forth as SEQ ID NO: 42, the nucleic acid sequence being in particular the nucleic acid sequence SEQ ID NO: 41.
  • Item 10 The lentiviral vector according to any one of items 1 to 9, being selected from the group consisting of the non-integrative lentiviral vectors filed at the CNCM under accession numbers 1-5759, 1-5760, 1-5761 and 1-5762, and is in particular the non- integrative lentiviral vector filed at the CNCM under accession number 1-5759.
  • Item 11 The lentiviral vector according to any one of items 1 to 10, wherein the lentiviral vector comprises a MHC Class I promoter, and in particular a p2-microglobulin promoter.
  • Item 12 The lentiviral vector according to any one of items 1 to 11, wherein the lentiviral vector comprises a cPPT/CTS sequence, in particular the cPPT/CTS sequence set forth as sequence SEQ ID NO: 37.
  • Item 13 The lentiviral vector according to any one of items 1 to 12, wherein the lentiviral vector comprises a 3’ long terminal repeat (LTR) which is devoid of its U3 promoter sequence.
  • LTR long terminal repeat
  • Item 14 The lentiviral vector according to any one of items 1 to 13, wherein the lentiviral vector does not comprise a constitutive enhancer sequence.
  • Item 15 The non-integrative lentiviral vector according to any one of items 1 to 14, wherein the lentiviral vector comprises a mutant form of the woodchuck hepatitis B virus (WHV) post-transcriptional regulatory element (WPRE), and in particular having the sequence set forth as sequence SEQ ID NO: 38.
  • WV woodchuck hepatitis B virus
  • WPRE post-transcriptional regulatory element
  • Item 16 A lentiviral vector particle, in particular a non-integrative lentiviral vector particle, comprising at least one lentiviral vector as defined in any one of items 1 to 15.
  • Item 17 The lentiviral vector particle according to item 16, wherein the lentiviral vector particle comprises a functional lentiviral integrase protein.
  • Item 18 The lentiviral vector particle according to item 16 or 17, wherein the lentiviral vector particle comprises a vesicular stomatitis virus glycoprotein (VSVG), in particular a VSV-G Indiana serotype or a VSV-G New Jersey serotype.
  • VSVG vesicular stomatitis virus glycoprotein
  • Item 19 The lentiviral vector particle according to any one of items 16 to 18, wherein the lentiviral vector particle comprises HIV-1 subtype D Gag and Pol proteins.
  • Item 20 An isolated cell comprising the lentiviral vector according to any one of items 1 to 14 or the lentiviral vector particle according to any of items 16 to 19.
  • Item 21 A vaccine composition comprising a lentiviral vector according to any one of items 1 to 14, a lentiviral vector particle according to any one of items 16 to 19, or a cell according to item 19.
  • Item 22 The vaccine composition according to item 21, for use in the treatment or prevention of an HPV induced cancer, in particular selected from the group consisting of cervical cancer, vaginal cancer, vulvar cancer, penile cancer, anal cancer, oropharyngeal cancer, and metastases thereof, in particular pulmonary metastasis thereof.
  • an HPV induced cancer in particular selected from the group consisting of cervical cancer, vaginal cancer, vulvar cancer, penile cancer, anal cancer, oropharyngeal cancer, and metastases thereof, in particular pulmonary metastasis thereof.
  • Item 23 A lentiviral vector according to any one of items 1 to 15, a lentiviral vector particle according to any one of items 16 to 19, or a cell according to item 20, for use as a medicament or vaccine.
  • Item 24 The lentiviral vector, lentiviral vector particle or cell, according to item 23, for use in the treatment or prevention of an HPV induced cancer, in particular selected from the group consisting of cervical cancer, vaginal cancer, vulvar cancer, penile cancer, anal cancer, oropharyngeal cancer, and metastases thereof, in particular pulmonary metastasis thereof.
  • an HPV induced cancer in particular selected from the group consisting of cervical cancer, vaginal cancer, vulvar cancer, penile cancer, anal cancer, oropharyngeal cancer, and metastases thereof, in particular pulmonary metastasis thereof.
  • Item 25 The vaccine composition for use according to item 22, or the lentiviral vector, lentiviral vector particle or cell for use according to item 23 or 24, wherein said vaccine composition, lentiviral vector, lentiviral vector particle or cell is administered in combination with at least one immune checkpoint inhibitor, in particular at least one monoclonal antibody selected from the group consisting of anti-PD-1, anti-PD-Ll, anti- CTLA-4, anti-NKG2A, anti-TIM-3, anti-TIGIT and anti-LAG-3 monoclonal antibodies and more particularly with at least one anti-PD-1 monoclonal antibody.
  • at least one immune checkpoint inhibitor in particular at least one monoclonal antibody selected from the group consisting of anti-PD-1, anti-PD-Ll, anti- CTLA-4, anti-NKG2A, anti-TIM-3, anti-TIGIT and anti-LAG-3 monoclonal antibodies and more particularly with at least one anti-PD-1 monoclonal antibody.
  • Item 26 The vaccine composition, lentiviral vector, lentiviral vector particle or cell for use according to item 25, wherein the at least one immune checkpoint inhibitor is administered simultaneously or separately, and in particular the at least one immune checkpoint inhibitor is administered at least 2, and in particular at least 4 days after the administration of the said vaccine composition, lentiviral vector, lentiviral vector particle or cell.
  • the invention also relates to a lentiviral vector, in particular a non-integrative lentiviral vector, or a lentiviral vector particle, in particular a non-integrative lentiviral vector particle, for use for use in the treatment or prevention of an HPV induced cancer, the lentiviral vector comprising at least four distinct nucleic acid sequences selected from the group consisting of: at least one nucleic acid sequence encoding a non-oncogemc Human papillomavirus (HPV 16) protein E6 antigen, at least one nucleic acid sequence encoding a non-oncogemc Human papillomavirus (HPV 16) protein E7 antigen, at least one nucleic acid sequence encoding a non-oncogemc Human papillomavirus (HPV 18) protein E6 antigen, and
  • the lentiviral vector particle comprising at least one of the lentiviral vector; the lentiviral vector or the lentiviral vector particle being administered in combination with at least one immune checkpoint inhibitor, in particular at least one monoclonal antibody selected from the group consisting of anti-PD-1, anti-PD-Ll, anti- CTLA-4, anti-NKG2A, anti-TIM-3, anti-TIGIT and anti-LAG-3 monoclonal antibodies.
  • at least one immune checkpoint inhibitor in particular at least one monoclonal antibody selected from the group consisting of anti-PD-1, anti-PD-Ll, anti- CTLA-4, anti-NKG2A, anti-TIM-3, anti-TIGIT and anti-LAG-3 monoclonal antibodies.
  • the HPV induced cancer may be selected from the group consisting of cervical cancer, vaginal cancer, vulvar cancer, penile cancer, anal cancer, oropharyngeal cancer, and metastases thereof, in particular pulmonary metastasis thereof.
  • the nucleic acid sequence encoding the non-oncogenic Human papillomavirus (HPV 16) protein E6 antigen may encode an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 7, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6.
  • the nucleic acid sequence encoding the non-oncogenic Human papillomavirus (HPV 16) protein E7 antigen may encode an amino acid sequence having at least 68% sequence identity with the amino acid sequence set forth as SEQ ID NO: 16, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 14 and SEQ ID NO: 15.
  • the nucleic acid sequence encoding the non-oncogenic Human papillomavirus (HPV18) protein E6 antigen may encode an amino acid sequence having at least 60% sequence identity with the amino acid sequence set forth as SEQ ID NO: 24, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22 and SEQ ID NO: 23.
  • the nucleic acid sequence encoding the non-oncogenic Human papillomavirus (HPV18) protein E7 antigen may encode an amino acid sequence having at least 83% sequence identity with the amino acid sequence set forth as SEQ ID NO: 33, the nucleic acid sequence being in particular selected from the group consisting of SEQ ID NO: 30, SEQ ID NO: 31 and SEQ ID NO: 32.
  • the at least four distinct nucleic acid sequences encoding antigens may be fused together, forming a single antigenic nucleic acid sequence encoding a single antigenic fusion protein under the control of a single promoter sequence.
  • the order of the at least four distinct nucleic acid sequences, from 5’ end to 3’ end, may be selected from the group consisting of:
  • the order of the at least four distinct nucleic acid sequences, from 5’ end to 3’ end, may be (a) nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV16) protein E7 antigen - nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV16) protein E6 antigen - nucleic acid sequence encoding a non- oncogenic Human papillomavirus (HPV18) protein E7 antigen - nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E6 antigen.
  • HPV16 non-oncogenic Human papillomavirus
  • HPV18 non-oncogenic Human papillomavirus
  • HPV18 non-oncogenic Human papillomavirus
  • the lentiviral vector or lentiviral vector particle for use according to the invention may comprise a nucleic acid sequence which encodes an amino acid sequence having at least 90% sequence identity with the amino acid sequence set forth as SEQ ID NO: 42, the nucleic acid sequence being in particular the nucleic acid sequence SEQ ID NO: 41.
  • the lentiviral vector or lentiviral vector particle according for use according to the invention mays be selected from the group consisting of the non-integrative lentiviral vectors filed at the CNCM under accession numbers 1-5759, 1-5760, 1-5761 and 1-5762, and is in particular the non-integrative lentiviral vector filed at the CNCM under accession number 1-5759.
  • the lentiviral vector for use according to the invention may comprise a MHC Class I promoter, and in particular a p2-microglobulin promoter.
  • the lentiviral vector for use according to the invention may comprise a cPPT/CTS sequence, in particular the cPPT/CTS sequence set forth as sequence SEQ ID NO: 37.
  • the lentiviral vector for use according to the invention may comprise a 3’ long terminal repeat (LTR) which is devoid of its U3 promoter sequence.
  • LTR long terminal repeat
  • the lentiviral vector for use according to the invention in particular does not comprise a constitutive enhancer sequence.
  • the lentiviral vector for use according to the invention may comprise a mutant form of the woodchuck hepatitis B virus (WHV) post-transcriptional regulatory element (WPRE), and in particular having the sequence set forth as sequence SEQ ID NO: 38.
  • WV woodchuck hepatitis B virus
  • WPRE post-transcriptional regulatory element
  • the lentiviral vector particle for use according to the invention may comprise a functional lentiviral integrase protein.
  • the lentiviral vector particle for use according to the invention may comprise a vesicular stomatitis virus glycoprotein (VSVG), in particular a VSV-G Indiana serotype or a VSV-G New Jersey serotype.
  • VSVG vesicular stomatitis virus glycoprotein
  • the lentiviral vector particle for use according to the invention may comprise HIV-1 subtype D Gag and Pol proteins.
  • the lentiviral vector or lentiviral vector particle for use according to the invention may be comprised in an isolated cell.
  • the lentiviral vector or lentiviral vector particle for use according to the invention may be comprised in a vaccine composition.
  • the at least one immune checkpoint inhibitor may be selected from the group consisting of anti-PD-1, anti-NKG2A and anti-TIM-3 monoclonal antibodies.
  • the at least one immune checkpoint inhibitor may be administered simultaneously or separately, and in particular the at least one immune checkpoint inhibitor may be administered at least 2, and in particular at least 4 days after the administration of the said vaccine composition, lentiviral vector, lentiviral vector particle or cell.
  • Figure 1 demonstrates that HPV vaccines of the invention are immunogenic in vivo.
  • Mice were injected i.m. with 1X10 7 TU of lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759, the lentiviral vector filed at the CNCM under accession number 1-5760, the lentiviral vector filed at the CNCM under accession number 1-5761 or the lentiviral vector filed at the CNCM under accession number 1-5762 or 50 pL of diluent.
  • 14 days later, splenocytes were prepared and restimulated overnight for IFNg ELISPOT with 4 distinct peptide pools.
  • Abscissa from left to right: results obtained with the lentiviral vector filed at the CNCM under accession number 1-5759, the lentiviral vector filed at the CNCM under accession number 1-5760, the lentiviral vector filed at the CNCM under accession number 1-5761, the lentiviral vector filed at the CNCM under accession number 1-5762 or with 50
  • Figure 2 shows that invention vaccine fully eliminates well implanted tumors in vivo.
  • TC-1 cells were injected s.c. and tumor volume was measured every other day (caliper measurement).
  • tumor volume is 70 mm 3
  • mice were randomized and vaccinated with IxlO 8 TU i.m. of LV-GFP Indiana (as a control), Indiana lentiviral vector particles comprising 1-5759, Indiana lentiviral vector particles comprising 1-5760, Indiana lentiviral vector particles comprising 1-5761 or Indiana lentiviral vector particles comprising 1-5762.
  • Figure 3 illustrates the ability of lentiviral vector according to the invention expressing design 1-5759, 1-5760, 1-5761, 1-5762, or diluent as control, to generate a long lasting immunity to prevent relapses.
  • Mice which eliminated primary tumor were rechallenged on the other flank at day 60.
  • Control mice (untreated) were also injected s.c. in order to check on the tumor cell growth in naive mice.
  • Figure 4 represents a dose/response in mouse.
  • IxlO 6 TC-1 cells were injected in the flank of animals and tumor volume was measured twice a week (caliper measurement). When average tumor volume was 80mm 3 , mice were randomized and vaccinated with diluent (control), 1X10 7 TU of 1-5759 vaccine or IxlO 8 TU (i.m.) of 1-5759 vaccine.
  • FIG. 5 represents lymphocytic tumor infiltration after vaccination with a lentiviral vector according to the invention.
  • IxlO 6 TCI tumor cells were injected (s.c.) on the flank of animal, and the tumor volume was measured twice a week (caliper measurement).
  • mice were randomized and vaccinated with either diluent (control), or 1X10 7 TU of 1-5759 vaccine or 1X10 8 TU (i.m.) of 1-5759 vaccine.
  • tumors were collected, digested and analyzed by flow cytometry. FACS staining was performed and data were acquired on Macsquant facs according to methods well known in the art.
  • Abscissa from left to right: diluent (control); IxlO 8 TU of 1-5759 vaccine.
  • Top left Figure % CD8+ T cells (within live cells); Top right Figure: % CD4+ T cells (within live cells); Bottom Figure: % Treg cells (within live cells).
  • Figure 6 represents the ability of vectors according to the invention to eliminate well- established large tumors.
  • IxlO 6 TCI cells were injected (s.c.) on the flank of animal. When average tumor volume was around 300 mm 3 , mice were randomized and vaccinated with diluent (control), or IxlO 8 TU (i.m.) of the vaccine according to the invention comprising lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759.
  • Figure 7 depicts T cell response in human PBMC labelled with CFSE and cultured in absence (unstimulated condition) or presence of a vaccine according to the invention (1-5759).
  • CD8+ T cells and CD4+ T cells proliferation were measured by CFSE dilution) (A) and expression of CD25 activation marker (B) were increased by addition of lentiviral vectors of the invention in the culture.
  • Abscissa from left to right: Unstimulated (Unstim - control); 1-5759 vaccine.
  • Each of these antigen constructs consists in the four following sequences in various orders: a nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV16) protein E6 antigen, a nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV16) protein E7 antigen, a nucleic acid sequence encoding a non- oncogenic Human papillomavirus (HPV18) protein E6 antigen and a nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E7 antigen.
  • Figure 8 A represents the antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5759.
  • Figure 8B represents the antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5760.
  • Figure 8C represents the antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5761 and
  • Figure 8D represents the antigen construct of the lentiviral vector filed at the CNCM under accession number I- 5762.
  • ICS Intracellular Cytokine Staining
  • FIG. 9A in particular represents the cytometric gating strategy carried out on cytokine -producing CD8+ T cells and degranulation activity of the IFN-y-producing CD8 + T cells, assessed by surface CD107a staining.
  • Figure 9B represents the recapitulative frequencies of each (poly)functional cell subsets and of IFN-y + CD107a + cells within the CD8 + T subset.
  • Figure 10 depicts cytometric analysis of tumor infiltrating innate immune cells (NK) of tumor-engrafted and vaccinated with a HPV-vaccine according to the invention (1-5759), or of tumor-engrafted and non vaccinated mice (control - Ctrl Lenti). CD 11b and NKp46 were detected.
  • NK tumor infiltrating innate immune cells
  • Figure 11 represents the ability of vectors according to the invention to eliminate well- established large tumors.
  • a rechallenge of cured mice which eliminated the primary tumor (right panel) was performed on the other flank with 1.10 6 TC-1 tumor cells 119 days after the first engraftment and were maintained without any therapy.
  • Control mice (untreated - control aged-mice - left panel) were also injected s.c. in order to check on the tumor cell growth.
  • Figures 12A and 12B illustrate the synergy between a vaccination with a suboptimal dose of a vaccine according to the invention (1-5759) and an anti-PD-1 therapy (monoclonal antibody anti-PD-1).
  • Figure 12A represents the evolution of tumor volumes (mm3 -ordinate) over days post tumor engraftement in mice (DO) (abscissa). Experiments were performed on three identical groups of tumor-engrafted mice. In the first group (10 mice - group control - left panel of Figure 12A), the mice were administered a LV-empty Indiana (D13) (as a control) (day indicated by an arrow) and four days later with an anti-PD-1 (Programmed cell Death protein- 1) monoclonal antibody (D17, then D20, D22, D24, D28 and D31).
  • D13 LV-empty Indiana
  • D13 as a control
  • an anti-PD-1 Programmed cell Death protein- 1 monoclonal antibody
  • mice - group control - middle panel of Figure 12A In the second group (12 mice - group control - middle panel of Figure 12A), the mice were administered a vaccine according to the invention (1-5759) (D13) (day indicated by an arrow) and four days later with a control antibody (isotype Ctrl) (D17, then D20, D22, D24, D28 and D31).
  • a control antibody isotype Ctrl
  • mice were administered a vaccine according to the invention (D13) (1-5759) (day indicated by an arrow) and four days later with a mAb anti-PD- 1 (D17, then D20, D22, D24, D28 and D31).
  • Figure 12B represents the survival (% of mice - ordinate) of the mice of each group (group control 1 : Ctrl Lend + anti-PD-1; group control 2 : 1-5759 + Ctrl Ig; group 3: 1-5759 + anti- PD-1) over time (days - abscissa).
  • Figures 13A and 13B represent the cure of mice with pulmonary metastatic foci induced by intravenous injection of TCl-nLuc cells after a single infection of Lenti-HPV-07 vaccine.
  • Figure 13A represents the variation of luminescence values expressed as photons per second (p/s) (ordinate - Total Flux) due to nanoluciferase stably expressed by TCl-nLuc cells injected to different groups of mice overtime (abscissa - days) after i.v. injection of said cells to said mice.
  • Figure 13B represents the individual p/s values, for individual mice of the three experimental groups detailed above, at day 22 post tumor injection.
  • Ordinate luminescence values expressed as photons per second (p/s) (Total Flux). Abscissa, from left to right: group Neg Ctrl, group Ctrl Lenti then group Lenti-HPV-07.
  • Figures 14A to 14E illustrate the synergistic effect of Lenti-HPV07 and anti-NKG2A mAb in TC-1 tumor eradication.
  • Fig 14 A represents the timeline of tumor engraftment and combinatory treatment with Lenti- HPV-07 and anti-NKG2A mAb in C57BL/6 mice.
  • mice were randomized and vaccinated with the suboptimal dose of IxlO 8 TU/mouse of Lenti-HPV-07.
  • Mice were then treated 2-to-3 times a week with anti-NKG2A mAb (clone 20D5, Bioxcell) or Ig control (clone 2A3, Bioxcell).
  • Fig 14 B represents spaghetti plots of tumor growth in the two tested groups : left group Lenti-HPV-07 (1-5759) + Ctrl Ig. Right group Lenti-HPV-07 (1-5759) + anti-NKG2A. Ordinate: tumor volume (mm 3 ). Abscissa: Days post tumor engraftment.
  • Fig 14 C represents a plotted therapy response rates according to RECIST criteria in the two tested groups.
  • Abscissa left group Lenti-HPV-07 (I- 5759) + Ctrl Ig; right group Lenti-HPV-07 (1-5759) + anti-NKG2A.
  • % of therapy response Each group represents, from bottom to top, complete response, then partial response, and finally no response.
  • Fig 14 D represents the progression-free survival time of the mice in the two tested groups. Abscissa: left group Lenti-HPV-07 (1-5759) + Ctrl Ig; right group Lenti-HPV-07 (1-5759) + anti-NKG2A. Ordinate : PFS (days).
  • Fig 14 E represents the survival curves of the animals shown in Fig 14 B.
  • Abscissa days. Ordinate ; Survival (% of mice).
  • the curve having a value of about 80% at day 40 represents the group Lenti-HPV-07 (1-5759) + anti-NKG2A while the curve having a value of about 40% at day 40 represents the group Lenti-HPV-07 (1-5759) + Ctrl Ig.
  • Figures 15A to 15E illustrate the synergistic effect of Lenti-HPV07 and anti-TIM-3 mAb in TC-1 tumor eradication.
  • Figure 15 A represents the timeline of tumor engraftment and combinatory treatment with Lenti-HPV-07 and anti-TIM-3 mAb.
  • mice were randomized and vaccinated with the suboptimal dose of IxlO 8 TU/mouse of Lenti-HPV-07 or a Ctrl Lenti.
  • Fig 15 B represents spaghetti plots of tumor growth in the two tested groups : left group Lenti-HPV- 07 (1-5759) + Ctrl Ig. Right group Lenti-HPV-07 (1-5759) + anti-TIM-3. Ordinate: tumor volume (mm 3 ). Abscissa: Days post tumor engraftment.
  • Fig 15 C represents a plotted therapy response rates according to RECIST criteria in the two tested groups.
  • Abscissa from left to right groups: Ctrl Lenti + Ctrl Ig; Ctrl Lenti + anti-TIM-3; Lenti-HPV-07 (1-5759) + Ctrl Ig; Lenti-HPV-07 (1-5759) + anti-TIM-3.
  • Fig 15 D represents the progression-free survival time of the mice in the two tested groups.
  • Abscissa left group Lenti-HPV-07 (1-5759) + Ctrl Ig; right group Lenti-HPV-07 (1-5759) + anti-TIM-3.
  • Ordinate PFS (days).
  • Fig 15 E represents the survival curves of the animals shown in Fig 15 B. Abscissa: days.
  • Ordinate Survival (% of mice).
  • the curve having a value of about 75% at day 40 represents the group Lenti-HPV-07 (1-5759) + anti-TIM-3.
  • the curve having a value of about 35% at day 40 represents the group Lenti-HPV-07 (1-5759) + Ctrl Ig.
  • the curve having all its animals dead at day 33 represents the group Ctrl Lenti + anti-TIM-3.
  • the curve having all its animals dead at day 34 represents the group Ctrl Lenti + Ctrl Ig.
  • SEO ID NO: 1 is a nucleic acid sequence encoding the E6 protein from HPV 16
  • SEO ID NO: 2 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • SEO ID NO: 3 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • SEO ID NO: 4 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • SEO ID NO: 5 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • SEO ID NO: 6 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • SEO ID NO: 7 is the amino acid sequence of the E6 protein from HPV 16
  • SEO ID NO: 8 is an amino acid sequence of a non-oncogenic variant of the E6 protein from
  • SEO ID NO: 9 is an amino acid sequence of a non-oncogenic variant of the E6 protein from
  • HPV 16 is an amino acid sequence of a non-oncogenic variant of the E6 protein from is an amino acid sequence of a non-oncogenic variant of the E6 protein from is an amino acid sequence of a non-oncogenic variant of the E6 protein from
  • SEO ID NO: 13 is a nucleic acid sequence encoding the E7 protein from HPV 16
  • SEO ID NO: 14 is a nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 16
  • SEO ID NO: 15 is a nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 16
  • SEO ID NO: 16 is the amino acid sequence of the E7 protein from HPV 16
  • SEO ID NO: 17 is an amino acid sequence of a non-oncogenic variant of the E7 protein from
  • SEO ID NO: 18 is an amino acid sequence of a non-oncogenic variant of the E7 protein from
  • SEO ID NO: 19 is a nucleic acid sequence encoding the E6 protein from HPV 18
  • SEO ID NO: 20 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 18
  • SEO ID NO: 21 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 18 ID NO: 22 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 18 is a nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 18 is the amino acid sequence of the E6 protein from HPV 18 is an amino acid sequence of a non-oncogenic variant of the E6 protein from is an amino acid sequence of a non-oncogenic variant of the E6 protein from is an amino acid sequence of a non-oncogenic variant of the E6 protein from is an amino acid sequence of a non-oncogenic variant of the E6 protein from is an amino acid sequence of a non-oncogenic variant of the E6 protein from is an amino acid sequence of a non-oncogenic variant of the E6 protein from from is an amino acid sequence of a non-oncogenic variant of the E6 protein from from is an amino acid sequence
  • SEO ID NO: 29 is a nucleic acid sequence encoding the E7 protein from HPV 18
  • ID NO: 30 is a nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 18 is a nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 18 is a nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 18
  • SEO ID NO: 33 is the amino acid sequence of the E7 protein from HPV 18
  • ID NO: 34 is an amino acid sequence of a non-oncogenic variant of the E7 protein from
  • HPV 18 s an amino acid sequence of a non-oncogenic variant of the E7 protein from s an amino acid sequence of a non-oncogenic variant of the E7 protein from s a nucleic acid sequence encoding the cPPT/CTS sequence is the nucleic acid sequence encoding a mutant form of the woodchuck hepatitis B virus (WHV) post-transcriptional regulatory element (WPRE) is a synthetic E7upvi6-derived peptide containing the RAHYNIVTF H-2D b - restricted T-cell epitope ID NO: 40 is a synthetic E7HPvi6-derived peptide containing the RAHYNIVTF H-2D b - restricted T-cell epitope
  • WV woodchuck hepatitis B virus
  • WPRE post-transcriptional regulatory element
  • ID NO: 41 is the nucleic acid sequence encoding an antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5759
  • SEQ ID NO: 42 is the amino acid sequence of the antigen construct encoded by the lentiviral vector filed at the CNCM under accession number 1-5759 is the nucleic acid sequence encoding an antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5760 is the amino acid sequence of the antigen construct encoded by the lentiviral vector filed at the CNCM under accession number 1-5760 is the nucleic acid sequence encoding an antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5761 is the amino acid sequence of the antigen construct encoded by the lentiviral vector filed at the CNCM under accession number 1-5761 is the nucleic acid sequence encoding an antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5762 is the amino acid sequence of the antigen construct encoded by the lentiviral vector filed at the CNCM under accession number 1-5762 of the H 2D b -restricted T-cell epitope
  • the inventors have discovered that the administration of a lentiviral vector encoding at least four distinct human papillomavirus (HPV) antigens, and in particular at least 4 HPV antigens selected among proteins E6 and E7 of at least two different HPV subtypes, and in particular of HPV 16 and HPV 18 subtypes, to an individual in need thereof results in a high prophylactic and therapeutic activity on HPV induced cancers.
  • HPV human papillomavirus
  • Lentiviral vectors according to the invention can enable the induction of a strong, lasting and broad cell-mediated response against tumors induced by an HPV infection.
  • Lentiviral vectors according to the invention as well as lenviral vector particules comprising them, isolated cells comprising said lentiviral vectors or lentiviral vector particles and vaccine compositions comprising them are described throughout the present specification. Definitions
  • transgene means a polynucleotide that can be expressed, via recombinant techniques, in a non-native environment or heterologous cell under appropriate conditions.
  • recombinant when used in reference to a cell of the invention, indicates that the cell has been modified by the introduction of an endogenous and/or heterologous nucleic acid or protein into the cell or the alteration of a native cell or that the cell is derived from a cell so modified.
  • recombinant cells express genes or nucleic acid that are not found within the native (non-recombinant) form of the cell or express native (eg endogenous) genes at a different level than their native level or express additional or supplementary copies of native (eg endogenous) at a different level than their native level.
  • An isolated cell according to the invention is recombinant in that it comprises at least one lentiviral vector according to the invention and/or at least one lentiviral vector particle according to the invention.
  • the term “recombinant”, when used in reference to a vector, are sequences formed/obtained by technics of genetic engineering well known to the man skilled in the art.
  • polypeptide refers to a molecule comprising amino acid residues linked by peptide bonds and containing more than five amino acid residues. The amino acids are identified by either the single-letter or three-letter designations.
  • protein as used herein is synonymous with the term “polypeptide” and may also refer to two or more polypeptides. Thus, the terms “protein”, “peptide” and “polypeptide” can be used interchangeably.
  • Polypeptides may optionally be modified (e.g., glycosylated, phosphorylated, acylated, famesylated, prenylated, sulfonated, and the like) to add functionality. Polypeptides exhibiting activity may be referred to as enzymes. It will be understood that, as a result of the degeneracy of the genetic code, a multitude of nucleotide sequences encoding a given polypeptide may be produced.
  • operably linked refers to two or more nucleic acid sequence elements that are physically linked and are in a functional relationship with each other.
  • a promoter is operably linked to a coding sequence, also termed herein “antigen construct” as the promoter is able to initiate or regulate the transcription or expression of the antigen construct, in which case the antigen construct should be understood as being "under the control of” the promoter.
  • antigen construct also termed herein “antigen construct” as the promoter is able to initiate or regulate the transcription or expression of the antigen construct, in which case the antigen construct should be understood as being "under the control of” the promoter.
  • two nucleic acid sequences when operably linked, they will be in the same orientation and usually also in the same reading frame. They usually will be essentially contiguous, although this may not be required.
  • encoding or “coding for” refer to the process by which a polynucleotide, through the mechanisms of transcription and translation, produces an aminoacid sequence.
  • amino acid sequences of interest For each or the amino acid sequences of interest, reference sequences are described herein. The present description also encompasses amino acid sequences having specific percentages of amino acid identity with a reference amino acid sequence.
  • a specific nucleic acid sequence or a specific amino acid sequence which complies with, respectively, the considered nucleotide or amino acid identity should further lead to obtaining a protein (or antigen) which displays the desired biological activity.
  • the "percentage of identity" between two nucleic acid sequences or between two amino acid sequences is determined by comparing both optimally aligned sequences through a comparison window.
  • the portion of the nucleotide or amino-acid sequence in the comparison window may thus include additions or deletions (for example "gaps") as compared to the reference sequence (which does not include these additions or these deletions) so as to obtain an optimal alignment between both sequences.
  • additions or deletions for example "gaps”
  • sequence homology or “sequence identity” or “homology” or “identity” are used interchangeably herein.
  • sequences are aligned for optimal comparison purposes.
  • gaps may be introduced in any of the two sequences that are compared.
  • alignment can be carried out over the full length of the sequences being compared.
  • the alignment may be carried out over a shorter length, for example over about 20, about 50, about 100 or more nucleic acids/based or amino acids.
  • sequence identity is the percentage of identical matches between the two sequences over the reported aligned region.
  • a comparison of sequences and determination of percentage of sequence identity between two sequences can be accomplished using a mathematical algorithm.
  • the skilled person will be aware of the fact that several different computer programs are available to align two sequences and determine the identity between two sequences (Kruskal, J. B. (1983) An overview of sequence comparison In D. Sankoff and J. B. Kruskal, (ed.), Time warps, string edits and macromolecules: the theory and practice of sequence comparison, pp. 1-44 Addison Wesley).
  • the percent sequence identity between two amino acid sequences or between two nucleotide sequences may be determined using the Needleman and Wunsch algorithm for the alignment of two sequences. (Needleman, S. B. and Wunsch, C. D. (1970) J. Mol. Biol. 48, 443-453). Both amino acid sequences and nucleotide sequences can be aligned by the algorithm.
  • the Needleman-Wunsch algorithm has been implemented in the computer program NEEDLE.
  • the NEEDLE program from the EMBOSS package was used (version 2.8.0 or higher, EMBOSS: The European Molecular Biology Open Software Suite (2000) Rice, P. LongdenJ. and Bleasby,A. Trends in Genetics 16, (6) pp276 — 277, http://emboss.bioinformatics.nl/).
  • EMBOSS European Molecular Biology Open Software Suite (2000) Rice, P. LongdenJ. and Bleasby,A. Trends in Genetics 16, (6) pp276 — 277, http://emboss.bioinformatics.nl/).
  • EBLOSUM62 is used for the substitution matrix.
  • EDNAFULL is used for nucleotide sequence.
  • the optional parameters used are a gap opening penalty of 10 and a gap extension penalty of 0.5. No end gap penalty is added.
  • Yes has been indicated in response to the question “Brief identity and similarity” and “SRS pairwise” indicated as Output alignment format.
  • the percentage of sequence identity between a query sequence and a sequence of the invention is calculated as follows: Number of corresponding positions in the alignment showing an identical amino acid or identical nucleotide in both sequences divided by the total length of the alignment after subtraction of the total number of gaps in the alignment.
  • the identity defined as herein can be obtained from NEEDLE by using the NOBRIEF option and is labeled in the output of the program as "longest-identity".
  • nucleotide and amino acid sequences i.e. the percentage of sequence identity
  • sequence alignments can be determined via sequence alignments using several other art-known algorithms, preferably with the mathematical algorithm of Karlin and Altschul (Karlin & Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5877), with hmmalign (HMMER package, http://hmmer.wustl.edu/) or with the CLUSTAL algorithm (Thompson, J. D., Higgins, D. G. & Gibson, T. J. (1994) Nucleic Acids Res. 22, 4673-80) available e.g.
  • sequence matching may be calculated using e.g. BLAST, BLAT or BlastZ (or BlastX).
  • BLASTN and BLASTP programs of Altschul et al (1990) J. Mol. Biol. 215, 403-410.
  • Gapped BLAST is utilized as described in Altschul et al (1997) Nucleic Acids Res. 25, 3389-3402.
  • Sequence matching analysis may be supplemented by established homology mapping techniques like Shuffle-LAGAN (Brudno M., Bioinformatics 2003b, 19 Suppl 1: 154-162) or Markov random fields. When percentages of sequence identity are referred to in the present application, these percentages are calculated in relation to the full length of the longer sequence, if not specifically indicated otherwise.
  • % identity between two sequences is determined using CLUSTAL O (version 1.2.4).
  • non-oncogenic used herein are used in their traditional meaning, i.e. it relates to an element, in the present case to antigens, unable to cause the formation of tumors.
  • antigens implemented in the present invention have been genetically amended in order to become nononcogenic. It means, according to the usual meaning of these terms, that the nucleic acid sequences encoding the antigens implemented herein are not found in nature and are modified either by introduction or by deletion or by modification of their nucleic acid sequences, leading to encoded amino acid sequences that also do not naturally exist in nature.
  • the man skilled in the art has known for a long time a variety of means to perform a deletion, substitution or introduction in a nucleic acid sequence. As will be understood by those of skill in the art, it can moreover be advantageous to modify a coding sequence to enhance its expression in a particular host.
  • the genetic code is redundant with 64 possible codons, but most organisms typically use a subset of these codons. The codons that are utilized most often in a species are called optimal codons, and those not utilized very often are classified as rare or low-usage codons.
  • Codons can be substituted to reflect the preferred codon usage of the host, in a process sometimes called “codon optimization” or “controlling for species codon bias.” Codon optimization for other host cells can be readily determined using codon usage tables or can be performed using commercially available software, such as CodonOp (www.idtdna.com/CodonOptfrom) from Integrated DNA Technologies. Optimized coding sequences containing codons preferred by a particular prokaryotic or eukaryotic host (Murray et al, 1989 , Nucl Acids Res .
  • RNA transcripts 17: 477-508 can be prepared, for example, to increase the rate of translation or to produce recombinant RNA transcripts having desirable properties, such as a longer half-life, as compared with transcripts produced from a non-optimized sequence.
  • Translation stop codons can also be modified to reflect host preference. For example, typical stop codon for monocotyledonous plants is UGA, whereas insects and E. coli commonly use UAA as the stop codon (Dalphin et al, 1996, Nucl Acids Res. 24: 216-8).
  • a “non-integrative” lentiviral vector means that, when this lentiviral vector is in a cell, it does not integrate into the host cell genome.
  • a non-integrative lentiviral vector particle relates to a lentiviral vector particle that comprises a non-integrative lentiviral vector. It can also be termed integration-defective lentiviral vectors or non-integrating lentiviral vectors.
  • HPV-induced cancers are also known as cancers associated with HPV (Human Papillomavirus). Indeed, when HPV infections are not successfully controlled by the immune system of an infected host. When a high-risk HPV infection persists for many years, it can lead to cell changes that, if untreated, may get worse over time and become cancer.
  • the inventors have conceived novel therapeutic and prophylactic lentiviral vectors based vaccins against HPV-induced cancers.
  • the present invention relates to a lentiviral vector comprising at least four distinct nucleic acid sequences selected from a group of particular non-oncogenic HPV antigens.
  • distinct nucleic acid sequences it is meant that the at least four nucleic acid sequences comprised in the lentiviral vector are all different, i.e. that each of them is a different member of the group of particular non-oncogenic HPV antigens.
  • HPV antigens The group of non-oncogenic HPV antigens is the following:
  • E6 and E7 are thought to interfere with multiple pathways but most importantly, E6 protein expression in the cell leads to ubiquitin-mediated degradation of the tumor suppressor p53 by direct interaction with the cellular E3 ubiquitin ligase, E6AP (Huibregtse, J M et al. The EMBO journal vol. 10,13 (1991): 4129-35; Martinez-Zapien, Denise et al. Nature vol. 529,7587 (2016): 541-5) and E7 binds to Rb protein hereby disrupting the interaction between Rb and E2F and releasing E2F factor (Cassetti, M Mandarin et al. Vaccine vol. 22,3-4 (2004): 520-7).
  • E6 and E7 protein are expressed in all HPV induced cancers, it was decided to include antigens from these proteins for the two major subtypes (HPV 16 and HPV 18) in lentiviral vectors of the invention. In order to develop a vaccine from E6 and E7 antigen, it is of major importance to abolish the oncogenic risk associated to these proteins.
  • Non-oncogenic E6 and E7 proteins are thus implemented in the invention.
  • non-oncogenic E6 and E7 HPV proteins it means that their encoding sequences were modified to remove p53, Mi2b and Rb binding sites as well as PDZ binding motif.
  • partial mutation of the binding sites of the E6 and E7 HPV proteins 1 did not allow to fully abolish Rb binding, said sites were in particular fully removed from the sequences implemented in the present invention.
  • a lentiviral vector according to the invention may be single-stranded or doublestranded.
  • a lentiviral vector according to the invention may be an RNA or DNA molecule.
  • a “lentiviral vector” means a nonreplicating vector for the transduction of a host cell with a transgene comprising cis-acting lentiviral RNA or DNA sequences, and requiring essential lentiviral proteins (e.g., Gag, Pol, and/or Env) and accessory proteins (e.g, Tat, Rev) that are provided in trans.
  • the lentiviral vector lacks expression of all functional HIV proteins.
  • the lentiviral vector genome may be present in the form of an RNA or DNA molecule, depending on the stage of production or development of said retroviral vectors.
  • a lentiviral vector of the invention is a non- integrative lentiviral vector.
  • Non-integrating lentiviral vectors have been designed to mitigate the risks of potential oncogenesis linked to insertional mutagenesis events, particularly for vaccination purposes.
  • Examples of non-integrating lentiviral vectors are provided in Coutant et al., PLOS ONE 7(l l):e48644 (2012), Karwacz et al., J. Virol. 83(7):3094-3103 (2009), Negri et al., Molecular Therapy 15(9): 1716-1723 (2007); and Hu et al., Vaccine 28:6675-6683 (2010).
  • a lentiviral vector according to the invention may comprise long terminal repeats (LTRs) sequences in cis as known in the art and in particular comprise a 3’ long terminal repeat (LTR) which is devoid of its U3 promoter sequence (Miyoshi H et al, 1998, J Virol. 72(1 0):81 50-7; Zufferey et al., 1998, J V/ro/ 72(12):9873-80).
  • LTRs long terminal repeats
  • Enhancers are cis-acting sequences, which can act as transcriptional activators at a distance. They have been widely employed in viral derived vectors because they appear to be the most efficient for obtaining transgene strong expression in a variety of cell types, in particular DCs (Chinnasamy et al., 2000, Hum Gene Ther 11(13): 1901-9; Rouas et al., 2008, Cancer Gene Ther 9(9):715-24; Kimura et al., 2007, Mol Ther 15(7): 1390-9; Gruh et al., 2008, J Gene Med 10(1) 21-32).
  • transcriptional enhancer sequences should be deleted from the lentiviral vector constructs to abolish the risk of insertional mutagenesis by enhancer proximity effect.
  • This enhancer proximity effect is by far the most frequent mechanism of insertional mutagenesis and is the only effect described in human or animal cases of tumorigenic events after gene transfer.
  • a lentiviral vector according to the invention may not comprise a constitutive enhancer sequence.
  • MHC class II promoter was found not to provide sufficient transgene expression in DCs, when administered intravenously.
  • lentiviral vectors including MHC class II promoters did not provoke an immune reaction in immunocompetent C57BL/6 mice, in contrast to the immune responses observed with CMV promo ters/enhancers.
  • integration and persistent transgene expression were observed after injection in mice, the lentiviral vectors transcribed through MHC class II promoters failed to stimulate an antigen-specific CD8+ cytotoxic T-lymphocyte response, even after vaccination boost.
  • the authors of these studies therefore concluded that the use of MHC class II promoters was of interest only for applications where persistence of expression is sought as in gene replacement therapy, but not in the context of immunotherapy.
  • MHC class II promoters are expressed poorly in most cell types.
  • the MHC class II promoter is not an adequate promoter for lentiviral vectors for induction of an immune response against an antigen via IV injection.
  • the dectin-2 promoter is expressed poorly in most cell types and appears to contain an enhancer.
  • the dectin-2 promoter is not a good promoter for lentiviral vectors for safety reasons.
  • a lentiviral vector according to the invention may comprise an MHC Class I promoter, i.e. the nucleic acid sequences encoding antigens of a lentiviral vector according to the invention may be under the control of an MHC Class I promoter.
  • An appropriate MHC Class I promoter may be selected from the group consisting of a p2-microglobulin promoter, a HLA-A2 promoter, a HLA-B7 promoter, a HLA-Cw5 promoter, a HLA-E promoter or a HLA-F promoter and is more particularly a P2- microglobulin promoter.
  • MHC Class I promoters are dendritic -specific (APCs) in that expression of the promoter in BDCA+ dendritic cells is higher than the expression in kidney, smooth muscle, liver, and heart cells. They also have relatively high expression in other transduced cell types, for example, expression of the promoter in BDCA+ dendritic cells is only 12-100 times the expression of that promoter in skeletal muscle cells, in contrast to 900 times with the MHCII HLA-DRa promoter.
  • APCs dendritic -specific
  • This promoter drives in particular the transcription of the nucleic acid sequences encoding HPV antigens in a lentiviral vector of the invention.
  • Said promoter can be a naturally occurring or a synthetic MHC Class I promoter, obtained using well known molecular biological techniques.
  • a lentiviral vector according to the invention may comprise a cPPT/CTS sequence, such as described in EP2169073.
  • This cPPT/CTS sequence may in particular be the sequence set forth as sequence SEQ ID NO: 37.
  • a lentiviral vector of the invention may comprise a Woodchuck hepatitis B virus (WHV) Post-Transcriptional Regulatory Element (WPRE), which allows a more stable expression of the transgene in vivo, and in particular a mutant form of the woodchuck hepatitis B virus (WHV) post-transcriptional regulatory element (WPRE).
  • WPRE Woodchuck hepatitis B virus
  • mWPRE Woodchuck Posttranscriptional Regulatory Element
  • the mutant form of the woodchuck hepatitis B virus (WHV) post-transcriptional regulatory element (WPRE) comprised in a lentiviral vector of the invention may in particular have the nucleic acid sequence set forth as sequence SEQ ID NO: 38.
  • a lentiviral vector according to the invention and in particular a non-integrative lentiviral vector of the invention:
  • (i) comprises at least one nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV16) protein E6 antigen, at least one nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV16) protein E7 antigen, at least one nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E6 antigen, and at least one nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E7 antigen;
  • (ii) comprises a 3’ long terminal repeat (LTR) which is devoid of its U3 promoter sequence;
  • (iv) comprises an MHC Class I promoter, and in particular a p2-microglobulin promoter
  • (v) comprises a cPPT/CTS sequence, having in particular the sequence set forth as sequence SEQ ID NO: 37; and (vi) comprises a mutant form of the woodchuck hepatitis B virus (WHV) post- transcriptional regulatory element (WPRE), having in particular the nucleic acid sequence set forth as sequence SEQ ID NO: 38.
  • WV woodchuck hepatitis B virus
  • a lentiviral vector according to the invention is characterized in that it comprises:
  • the at least four, and in particular the four, distinct nucleic acid sequences encoding HPV antigens of a lentiviral vector of the invention may in particular be fused together, forming a single antigenic nucleic acid sequence encoding a single antigenic fusion protein under the control of a single promoter sequence, in particular (i) in the absence of any linking sequence (also termed spacer herein) between each of the at least four distinct nucleic acid sequences or (ii) with a linking sequence (or spacer) between at least two of the at least four distinct nucleic acid sequences, and more particularly with a a linking sequence (or spacer) between each of the at least four distinct nucleic acid sequences.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV 16) protein E6 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence having at least 80 % sequence identity with the amino acid sequence set forth as SEQ ID NO: 7.
  • an amino acid sequence having at least 80 % amino acid identity with a reference amino acid sequence encompasses amino acid sequences having at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99% amino acid identity with the said reference amino acid sequence.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV 16) protein E6 antigen may in particular have a nucleic acid sequence having at least 80% sequence identity with a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6.
  • nucleic acid sequence having at least 80% nucleotide identity with a reference nucleic acid sequence encompasses nucleic acid sequences having at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99% nucleotide identity with the said reference nucleic acid sequence.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E6 antigen may in particular have a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E6 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence having at least 80% sequence identity with an amino acid sequence selected from the group consisting of the amino acid sequences set forth as SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E6 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence selected from the group consisting of the amino acid sequences set forth as SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E7 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence having at least 68 % sequence identity with the amino acid sequence set forth as SEQ ID NO: 16.
  • an amino acid sequence having at least 68 % amino acid identity with a reference amino acid sequence encompasses amino acid sequences having at least 69 %, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99% amino acid identity with the said reference amino acid sequence.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E7 antigen may in particular have a nucleic acid sequence having at least 80% sequence identity with a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 14 and SEQ ID NO: 15.
  • nucleic acid sequence having at least 80% nucleotide identity with a reference nucleic acid sequence encompasses nucleic acid sequences having at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99% nucleotide identity with the said reference nucleic acid sequence.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E7 antigen may in particular have a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 14 and SEQ ID NO: 15.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E7 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence having at least 80% sequence identity with an amino acid sequence selected from the group consisting of the amino acid sequences set forth as SEQ ID NO: 17 and SEQ ID NO: 18.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E7 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence selected from the group consisting of the amino acid sequences set forth as SEQ ID NO: 17 and SEQ ID NO: 18.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E6 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence having at least 60 % sequence identity with the amino acid sequence set forth as SEQ ID NO: 24.
  • an amino acid sequence having at least 60 % amino acid identity with a reference amino acid sequence encompasses amino acid sequences having at least 61 %, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99% amino acid identity with the said reference amino acid sequence.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E6 antigen may in particular have a nucleic acid sequence having at least 80% sequence identity with a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22 and SEQ ID NO: 23.
  • nucleic acid sequence having at least 80% nucleotide identity with a reference nucleic acid sequence encompasses nucleic acid sequences having at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99% nucleotide identity with the said reference nucleic acid sequence.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E6 antigen may in particular have a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22 and SEQ ID NO: 23.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E6 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence having at least 80% sequence identity with an amino acid sequence selected from the group consisting of the amino acid sequences set forth as SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27 and SEQ ID NO: 28.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E6 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence selected from the group consisting of the amino acid sequences set forth as SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27 and SEQ ID NO: 28.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E7 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence having at least 83% sequence identity with the amino acid sequence set forth as SEQ ID NO: 33.
  • an amino acid sequence having at least 83% amino acid identity with a reference amino acid sequence encompasses amino acid sequences having at least 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99% amino acid identity with the said reference amino acid sequence.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E7 antigen may in particular have a nucleic acid sequence having at least 80% sequence identity with a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 30, SEQ ID NO: 31 and SEQ ID NO: 32.
  • nucleic acid sequence having at least 80% nucleotide identity with a reference nucleic acid sequence encompasses nucleic acid sequences having at least 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% and 99% nucleotide identity with the said reference nucleic acid sequence.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E7 antigen may in particular have a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 30, SEQ ID NO: 31 and SEQ ID NO: 32.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E7 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence having at least 80% sequence identity with an amino acid sequence selected from the group consisting of the amino acid sequences set forth as SEQ ID NO: 34, SEQ ID NO: 35 and SEQ ID NO: 36.
  • a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 18 (HPV18) protein E7 antigen may in particular have a nucleic acid sequence encoding an amino acid sequence selected from the group consisting of the amino acid sequences set forth as SEQ ID NO: 34, SEQ ID NO: 35 and SEQ ID NO: 36.
  • a lentiviral vector according to the invention comprises:
  • a lentiviral vector according to the invention and in particular a non-integrative lentiviral vector of the invention:
  • (i) comprises a nucleic acid sequence encoding a non-oncogenic Human papillomavirus 16 (HPV16) protein E6 antigen; a nucleic acid sequence encoding a non- oncogenic Human papillomavirus (HPV16) protein E7 antigen; a nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E6 antigen and a nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E7 antigen; the nucleic acid sequences encoding non-oncogenic HPV antigens being in particular fused together, forming a single antigenic nucleic acid sequence encoding a single antigenic fusion protein; under the control of a single promoter sequence, more particularly in the absence of any linking sequence between each of them;
  • (ii) comprises a 3’ long terminal repeat (LTR) which is devoid of its U3 promoter sequence;
  • (iv) comprises an MHC Class I promoter, and in particular a p2-microglobulin promoter
  • (v) comprises a cPPT/CTS sequence, having in particular the sequence set forth as sequence SEQ ID NO: 37;
  • (vi) comprises a mutant form of the woodchuck hepatitis B virus (WHV) post- transcriptional regulatory element (WPRE), having in particular the nucleic acid sequence set forth as sequence SEQ ID NO: 38.
  • WPRE woodchuck hepatitis B virus
  • a lentiviral vector according to the invention may more particularly comprise:
  • nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV 16) protein E6 antigen
  • the nucleic acid sequence having at least 80% sequence identity with a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6, the nucleic acid sequence being in particular selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6;
  • nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV 16) protein E7 antigen
  • nucleic acid sequence having at least 80% sequence identity with a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 14 and SEQ ID NO: 15, the nucleic acid sequence being in particular selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 14 and SEQ ID NO: 15
  • - at least one nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E6 antigen the nucleic acid sequence having at least 80% sequence identity with a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 20, SEQ ID NO: 21, SEQ ID NO: 22 and SEQ ID NO: 23, the nucleic acid sequence being in particular selected from the group consisting of the nucleic acid sequences
  • nucleic acid sequence encoding a non-oncogenic Human papillomavirus (HPV18) protein E7 antigen
  • the nucleic acid sequence having at least 80% sequence identity with a nucleic acid sequence selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 30, SEQ ID NO: 31 and SEQ ID NO: 32, the nucleic acid sequence being in particular selected from the group consisting of the nucleic acid sequences set forth as SEQ ID NO: 30, SEQ ID NO: 31 and SEQ ID NO: 32; the nucleic acid sequences encoding non-oncogenic HPV antigens being in particular fused together, forming a single antigenic nucleic acid sequence encoding a single antigenic fusion protein; under the control of a single promoter sequence, more particularly in the absence of any linking sequence between each of them.
  • the at least four, and in particular the four, distinct nucleic acid sequences encoding HPV antigens in a lentiviral vector according to the invention, and in particular a non-integrative lentiviral vector of the invention, may be in any order in the traditional 5’ to 3’ reading direction (from 5’ end to 3’ end).
  • the four distinct nucleic acid sequences encoding HPV antigens noE6-HPV16, noE7-HPV16, noE6-HPV18 and noE7-HPV18 as defined above may be in any order, in the traditional 5’ end to 3’ end reading direction, among the 24 possible combinations in lentiviral vectors according to the invention, and in particular non-integrative lentiviral vectors according to the invention.
  • the order of the at least four distinct nucleic acid sequences, from 5’ end to 3’ end is selected from the group consisting of:
  • the order of the at least four distinct nucleic acid sequences, from 5’ end to 3’ end, in a lentiviral vector according to the invention may more particularly be:
  • the order of the at least four distinct nucleic acid sequences, from 5’ end to 3’ end, in a lentiviral vector according to a lent
  • the order of the at least four distinct nucleic acid sequences, from 5’ end to 3’ end, in a lentiviral vector according to the invention is:
  • the antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5759 has the following nucleotidic sequence set forth as SEQ ID NO: 41: atgcccggagacacccccaccctgcacgaatacatgctggacctgcagcccgaaaccaccgaccccgaccgcg ctcactacaacatcgttacattctgttgtaaatgcgactccaccctgagaagatgcgtgcagtccacccacgtggacatcaggaccctgg aggacctcctcatgggaaccctgggtatcgtctgccccatcgcctcccaggctttcaggacccccaggaaggacctctcatgggaaccctgggtatcgtctgcccatcgcctcccaggctttcaggacccccaggaaggc
  • the antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5760 has the following nucleotidic sequence set forth as SEQ ID NO: 43: atgttccaggacccccaggagaggccccggaagttgccccagctgtgcaccgagctgcagaccaccatccacga catcatcctcgaatgcgtgtactgcaagcagcagctgctgaggagggaggtgtatgactttgccttcagagacggatgcattgtctaca ggaacccctacgccgtgtgcgacaaatgcctgaagttctactccaagatcagcgagtacaggcactactgctactccctgtacggcac caccctcgaacagtacaaacccctgtgtgacctcgaa
  • the antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5761 has the following nucleotidic sequence set forth as SEQ ID NO: 45: atgaggcggccctacaagctgcccgacctgtgcaccgagctgaacacctccctgcaggacatcgagatcacctg cgtgtactgcaagaccgtgctggagctgaccgaggtgttcgaccgaggtgttcgaattcgcattcaaggacggattcgtcgtgtgtatagggacagcattccac acgccctgccacaagctggagaaattgactaacaccggactgtataatctgctgatccggtgctgaggtgtcagaaggccgaga agccgaga agctgagaaggccgaga
  • the antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5762 has the following nucleotidic sequence set forth as SEQ ID NO: 47: atgggccctaaggccaccctgcaggacatcgtgctgcacttggagccccagaacgagatccccgtggacagcga ggaggagaacgacgaaatcgacggcgtgaaccaccagcacctgcccgcaagaagggccgaaccccagaggcacaccatgctctg catgtgctgcaaatgcgaggccaggatcaagctggtggtggaaagcagcgccgacgatctgagggcattccagcagctgttcctgaa caccctcttcgtgtgtgtgtgtgtgaa caccc
  • TMLCMCCKCEARIKLVVESSADDLRAFQQLFLNTLSFVCPGEPGRTIPYKLPDLCTEL NTSLQDIEITCVYCKTVLELTEVFEFAFKDGFVVYRDSIPHAACHKLEKLTNTGLYNL LIRCLRCQKAEKLRHLNEKRRFHNIAGPGDTPTLHEYMLDLQPETTDPDRAHYNIVTF CCKCDSTLRRCVQSTHVDIRTLEDLLMGTLGIVCPIASQAFQDPQERPRKLPQLCTEL QTTIHDIILECVYCKQQLLRREVYDFAFRDGCIVYRNPYAVCDKCLKFYSKISEYRHY CYSLYGTTLEQQYNKPLCDLLIRCINCQKPLRFHNIRGRWTGRCMSCCRS (SEQ ID NO: 48)
  • a lentiviral vector according to the invention may in particular comprise a nucleic acid sequence which encodes an amino acid sequence having at least 90% sequence identity with the amino acid sequence set forth as SEQ ID NO: 42, the nucleic acid sequence being in particular the nucleic acid sequence SEQ ID NO: 41.
  • amino acid sequence having at least 90% identity with a reference amino acid sequence encompasses amino acid sequences having at least 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and 100% identity with the said reference amino acid sequence.
  • a lentiviral vector according to the invention may more particularly be selected from the group consisting of lentiviral vectors filed at the CNCM under accession numbers 1-5759, 1-5760, 1-5761 and 1-5762, and is in particular lentiviral vector filed at the CNCM under accession number 1-5762.
  • a lentiviral vector according to the invention may preferably be the lentiviral vector filed at the CNCM under accession number 1-5759 and accordingly preferably comprises the nucleic acid sequence SEQ ID NO: 41.
  • Another object of the present invention relates to a lentiviral vector particle comprising at least one lentiviral vector according to the invention, and in particular at least one lentirival vector as defined above.
  • a lentiviral vector particle according to the invention which contains a lentiviral vector according to the invention, can be produced by recombinant technology known in the art upon transient transfection of cells, for example HEK 293T human cultured cells, by different DNA plasmids:
  • a packaging plasmid which expresses at least the Gag, Pol, Rev, Tat and, in some cases, structural and enzymatic proteins necessary for the packaging of the transfer construct;
  • a lentiviral vector according to the invention containing an expression cassette (antigens) and HIV cis-acting factors necessary for packaging, reverse transcription, and integration;
  • VSV.G glycoprotein of vesicular stomatitis virus
  • MHC major histocompatibility antigen-presenting cells
  • Such a method allows producing a recombinant vector particle according to the invention, comprising the following steps of: i) transfecting a suitable host cell with a lentiviral vector according to the invention; ii) transfecting said host cell with a packaging plasmid vector, containing viral DNA sequences encoding at least structural and polymerase activities of a retrovirus (preferably lentivirus);
  • packaging plasmids are for example described in the art (Dull et al., 1998, J Virol, 72(11):8463-71 ; Zufferey et al., 1998, J Virol 72(12):9873-80).
  • iii) culturing said transfected host cell in order to obtain expression and packaging of said lentiviral vector into lentiviral vector particles; and iv) harvesting the lentiviral vector particles resulting from the expression and packaging of step iii) in said cultured host cells.
  • the host cell can be further transfected with one or several envelope DNA plasmid(s) encoding viral envelope protein(s), preferably a VSV-G envelope protein.
  • the lentiviral particle vectors may also be continuously produced by cells by stably inserting the packaging genes, the proviral coding DNA, and the envelope gene into the cellular genome. This allows the continuous production of lentiviral particle vectors by the cells without the need for transient transfection.
  • a combination of these procedures can be used, with some of the DNAs/plasmids integrated into the cellular genome and others provided by transient transfection.
  • a lentiviral vector particle may be a non-integrating lentiviral vector particle.
  • Non-integrating vector particles have one or more mutations that eliminate most or all of the integrating capacity of the lentiviral vector particles.
  • a non-integrating vector particle can contain mutation(s) in the integrase encoded by the lentiviral pol gene that cause a reduction in integrating capacity.
  • a lentiviral vector particle according to the invention in particular comprises a non-integrating lentiviral vector of the invention.
  • a lentiviral vector particle according to the invention may comprise a vesicular stomatitis virus glycoprotein (VSVG), in particular a VSV-G Indiana serotype or a VSV-G New Jersey serotype.
  • VSVG vesicular stomatitis virus glycoprotein
  • pseudotyped lentiviral vector particles are more likely to escape the immune system, when this latter already developed immunity against lentiviruses. This is particularly helpful when successive injections of similar particle vectors are required for immunizing a patient against a disease.
  • the lentiviral vector particle may comprise HIV-1 Gag and Pol proteins, and in particular HIV-1 subtype D Gag and Pol proteins.
  • a further object of the present invention relates to an isolated cell comprising (i.e. transformed with) a lentiviral vector according to the invention or a lentiviral vector particle of the invention.
  • a cell according to the invention is preferably a mammalian cell, particularly a human cell. Particularly preferred are human non-dividing cells.
  • Another object of the present invention relates to a vaccine composition
  • a vaccine composition comprising a lentiviral vector according to the invention, a lentiviral vector particle according to the invention or a cell according to the invention.
  • a vaccine composition according to the invention comprises a pharmaceutically acceptable medium.
  • pharmaceutically acceptable medium any solution used to solubilize and deliver a lentiviral vector, a lentiviral vector particle or a cell according to the invention to an individual.
  • a desirable pharmaceutically acceptable carrier is saline.
  • a pharmaceutically acceptable medium includes an adjuvant.
  • An object of the present invention relates to a lentiviral vector of the invention, a lentiviral vector particle of the invention or an isolated cell of the invention for use as a medicament or vaccine.
  • an object of the present invention relates to a lentiviral vector of the invention, a lentiviral vector particle of the invention or an isolated cell of the invention, in particular in the form of a vaccine composition according to the invention, for use in the treatment or prevention of an HPV induced cancer and metastases thereof, in particular of an HPV-induced cancer.
  • HPV induced cancers are cancers induced by an infection by HPV.
  • Methods for the detection of HPV in cancers are known in the art (Aldo Venuti and Francesca Paolini; Head Neck Pathol. 2012 Jul; 6(Suppl 1): 63-74).
  • HPV induced cancers can in particular be selected from the group consisting of cervical cancer, vaginal cancer, vulvar cancer, penile cancer, anal cancer and oropharyngeal cancer.
  • Metastases of such cancers according to the invention may in particular be pulmonary metastasis.
  • Such prevention and/or treatment implies the administration of the considered active, in particular a vaccine composition of the invention as defined above, to an individual in need thereof.
  • An individual in need thereof is an animal, in particular a mammal, and may more particularly be a human being.
  • Lentiviral vectors, lentiviral vector particles, cells and vaccine compositions according to the invention are administered to an individual in need thereof by conventional methods, in dosages which are sufficient to elicit an immunological response, which can be easily determined by those skilled in the art.
  • Lentiviral vectors, lentiviral vector particles, cells and vaccine compositions according to the invention may accordingly be administered intravenously or intramuscularly as indicted below.
  • Lentiviral vectors, lentiviral vector particles, cells and vaccine compositions according to the invention may alternatively be administered intranasally. This route of administration is particularly useful in the treatment or prevention of oropharyngeal cancers and/or pulmonary metastases.
  • Lentiviral vectors, lentiviral vector particles, cells and vaccine compositions according to the invention are administered in a therapeutically effective amount, and may in particular be administered in a dose corresponding to at least 1 x 10 6 , 2 x 10 6 , 5 x 10 6 , 10 7 , 2 x 10 7 , 5 x 10 7 , 1 x 10 8 , 2 x 10 8 , 5 x 10 8 , or at least 1 x 10 9 TU (Transduction units) of lentiviral vectors according to the invention, in particular in a dose corresponding to at least 1 x 10 7 , 2 x 10 7 , 5 x 10 7 , 1 x 10 8 TU or at least 1 x 10 9 TU of lentiviral vectors according to the invention.
  • Transduction units Transduction units
  • the lentiviral vectors, lentiviral vector particles, cells and vaccine compositions according to the invention are administered in a dose corresponding to at least 1 x 10 7 TU of lentiviral vectors according to the invention, more particularly at least 1 x 10 8 TU of lentiviral vectors according to the invention and in particular at least 1 x 10 9 TU of lentiviral vectors according to the invention.
  • a “therapeutically effective amount” is for example meant the amount of a lentiviral vector or lentiviral vector particle, cell or vaccine composition according to the invention required to generate in a subject one or more of the following effects: an immune response against an HPV induced tumor; a decrease in the size of the HPV induced tumor, i.e.
  • Administration can be performed using well known routes including, for example, intravenous, intramuscular, intranasal, intraperitoneal or sub-cutaneous injection, and in particular intravenous, intranasal or intramuscular, and may be intravenous or intramuscular.
  • Lentiviral vectors, lentiviral vector particles, cells and vaccine compositions according to the invention may for example be administered in a single dose, as illustrated in the examples, or in two or more administrations. Practitioners will determine, in each case, the appropriate regimen and dosage for the administration of actives according to the invention.
  • Lentiviral vectors, lentiviral vector particles, cells and vaccine compositions according to the invention may advantageously be administered in combination with at least one immune checkpoint inhibitor (ICI).
  • ICI immune checkpoint inhibitor
  • An immune checkpoint inhibitor (ICI) may in particular be an antibody, in particular an anti-PD-1, an anti-PD-Ll (PD-1 Ligand), an anti- CTLA-4 (Cytotoxic T-Lymphocyte-Associated protein 4), an anti-NKG2A, an anti-TIM-3 (T- cell immunoglobulin and mucin-domain containing-3), an anti-TIGIT (T cell immunoreceptor with Ig and ITIM domains) or an anti-LAG-3 (Lymphocyte-activation gene 3) antibody.
  • an anti-PD-1 an anti-PD-Ll (PD-1 Ligand)
  • CTLA-4 Cytotoxic T-Lymphocyte-Associated protein 4
  • an anti-NKG2A an anti-TIM-3 (T- cell immunoglobulin and mucin-domain containing-3)
  • an anti-TIGIT T cell immunoreceptor with Ig and ITIM domains
  • an anti-LAG-3 Lymphocyte-activation gene 3
  • the at least one immune checkpoint inhibitor according to the invention may be a monoclonal antibody selected from the group consisting of anti-PD-1, anti-PD-Ll, anti- CTLA-4, anti-NKG2A, anti-TIM-3, anti-TIGIT and anti-LAG-3 monoclonal antibodies.
  • the at least one immune checkpoint inhibitor according to the invention may be a monoclonal antibody selected from the group consisting of anti-PD-1, anti-PD-Ll, anti-CTLA-4, anti-NKG2A, anti-TIM-3 and anti-TIGIT monoclonal antibodies.
  • An immune checkpoint inhibitor (ICI) may more particularly be an antibody, in particular an anti-PD-1, an anti-PD-Ll (PD-1 Ligand), an anti- CTLA-4 (Cytotoxic T-Lymphocyte-Associated protein 4), an anti-NKG2A, an anti-TIM-3 (T- cell immunoglobulin and mucin-domain containing-3), an anti-TIGIT (T cell immunoreceptor with Ig and ITIM domains) or an anti-LAG-3 (Lymphocyte-activation gene 3) antibody and even more particularly be an antibody, in particular an anti-PD-1, an anti-PD-Ll (PD-1 Ligand), an anti-CTLA-4 (Cytotoxic T-Lymphocyte-Associated protein 4), an anti-NKG2A, an anti-TIM-3 (T-cell immunoglobulin and mucin-domain containing-3) or an anti-TIGIT (T cell immunoreceptor with Ig and ITIM domains) antibody.
  • an anti-PD-1 an anti-PD-Ll (PD-1
  • the at least one immune checkpoint inhibitor according to the invention may be a monoclonal antibody selected from the group consisting of anti-PD-1, anti-PD-Ll, anti-CTLA-4, anti-NKG2A, anti-TIM-3, anti-TIGIT and anti-LAG-3 monoclonal antibodies and in particular may be a monoclonal antibody selected from the group consisting of anti-PD-1, anti-PD-Ll, anti- CTLA-4, anti-NKG2A, anti-TIM-3 and anti-TIGIT monoclonal antibodies.
  • An anti-PD-1 monoclonal antibody may for example be selected from the group consisting of Nivolumab, Pembrolizumab and Cemiplimab.
  • An anti-PD-Ll monoclonal antibody may for example be selected from the group consisting of Atezolizumab, Avelumab and Durvalumab.
  • An anti-CTLA-4 monoclonal antibody may for example be selected from the group consisting of ipilimumab, tremelimumab and quavonlimab.
  • NKG2A is an ITIM (intracytoplasmic tyrosine-based inhibitory motifs)-bearing receptor expressed at the surface of 50% of peripheral blood NK cells and 5% of human peripheral blood CD8+ T cells.
  • This cell surface molecule is expressed as a heterodimer with CD94 and interacts with the Major Histocompatibility Complex class I (MHC-I) non-classical molecules, i.e., human leukocyte antigen (HLA)-E in and murine Qa-lb. This interaction inhibits both T and NK effector anti-tumor functions (Andre et al, Cell. 2018 Dec 13; 175(7): 1731- 1743. el3).
  • MHC-I Major Histocompatibility Complex class I
  • HLA human leukocyte antigen
  • an anti-NKG2A mAb can be used as a checkpoint inhibitor and can promote anti-tumor cellular immunity by unleashing not only NK, but also CD8+ T cells in mice (Andre et al, Cell. 2018 Dec 13;175(7):1731-1743.el3).
  • An anti-NKG2A monoclonal antibody may for example be monalizumab.
  • T-cell immunoglobulin and mucin domain-3 is a negative regulatory immune checkpoint.
  • TIM-3 is expressed by various immune cells, notably T cells.
  • TIM-3 has four ligands, including galectin-9 (Gal-9), carcinoembryonic antigen cell adhesion molecule 1 (CEACAM-1), high-mobility group protein Bl (HMGB1), and phosphatidylserine (PS) (He et al, Onco Targets Ther. 2018; 11: 7005-7009).
  • TIM-3/Gal-9 can inhibit cancer immunity by negatively regulating T-cell immunity.
  • TIM-3 displays an important role in T-cell exhaustion. In cancer immunotherapy anti-TIM-3 mAb treatment displays beneficial effects comparable to those of anti-PD-1 mAb therapy.
  • An anti-TIM-3 monoclonal antibody may for example be selected from the group consisting of Sym023 and sabatolimab.
  • An anti-TIGIT monoclonal antibody may for example be tiragolumab.
  • An anti-LAG-3 monoclonal antibody may for example be relatlimab.
  • the ICI may be an anti-PD-Ll or an anti-PD-1 monoclonal antibody, and in particular be an anti-PD-1 monoclonal antibody.
  • the ICI may be selected in the group consisting of an anti-PD-Ll an anti-NKG2A, an anti-TIM-3 and an anti-PD-1 monoclonal antibody, et plustension in the group consisting of an anti-NKG2A, an anti-TIM-3 and an anti-PD-1 monoclonal antibody.
  • the vaccine composition, lentiviral vector, lentiviral vector particle or cell for use according to the invention and the immune checkpoint inhibitor may be administered simultaneously or separately.
  • a lentiviral vector according to the invention may increase the number of patients eligible for immune checkpoint inhibitor therapy, especially anti-PD-1, an anti-NKG2A or an anti-TIM-3.
  • the vaccine composition, lentiviral vector, lentiviral vector particle or cell and (ii) the immune checkpoint inhibitor may be administered at the same moment or up to the same day or couple of days. In this case, they can be administered in the same composition or in separate compositions.
  • the vaccine composition, lentiviral vector, lentiviral vector particle or cell according to the invention and (ii) the immune checkpoint inhibitor may be administered with at least several days, for example at least two days of difference.
  • the vaccine composition, lentiviral vector, lentiviral vector particle or cell and (ii) the immune checkpoint inhibitor are administered separately
  • the vaccine composition, lentiviral vector, lentiviral vector particle or cell according to the invention may be administered before the immune checkpoint inhibitor.
  • the vaccine composition, lentiviral vector, lentiviral vector particle or cell according to the invention may be administered at least 2 and in particular at least 4 days before the administration of the immune checkpoint inhibitor.
  • the immune checkpoint inhibitor may advantageously be administered at least 2 and in particular at least 4 days after the vaccine composition, lentiviral vector, lentiviral vector particle or cell according to the invention.
  • the immune checkpoint inhibitor may more particularly be administered 4 days to 1 month after the vaccine composition, lentiviral vector, lentiviral vector particle or cell according to the invention, in particular 4 days to 15 days, and more particularly 4 days to 10 days after the vaccine composition, lentiviral vector, lentiviral vector particle or cell according to the invention.
  • the vaccine composition, lentiviral vector, lentiviral vector particle or cell for use according to the invention and the immune checkpoint inhibitor may be administered by the same route or through different routes.
  • the at least one immune checkpoint inhibitor herein is administered in a therapeutically effective dose, i.e. a dose that produces the effects for which it is administered.
  • a therapeutically effective dose i.e. a dose that produces the effects for which it is administered.
  • the exact dose of immune checkpoint inhibitor will depend on the purpose of the treatment and will be ascertainable by one skilled in the art using known techniques.
  • the invention further relates to a method for the treatment and/or prevention of an HPV induced cancer in an individual in need thereof, comprising the administration to said individual of at least one lentiviral vector of the invention, lentiviral vector particle of the invention or isolated cell of the invention, in particular in the form of a vaccine composition according to the invention.
  • the invention further relates to the use of at least one lentiviral vector of the invention, lentiviral vector particle of the invention or isolated cell of the invention, in particular in the form of a vaccine composition according to the invention for the treatment and/or prevention of an HPV induced cancer in an individual in need thereof.
  • C57BL6jRj mice were purchased from Janvier Labs (Le Genest-Saint-Isle, France). All animals were maintained under specific pathogen-free conditions, and all procedures were performed according to an approved animal protocol and in accordance with recommendations for the proper use and care of laboratory animals. All animal experiments were conducted in accordance with guidelines established by the French and European regulations for the care and use of laboratory animal.
  • Anti anti-mouse H-2kb (Clone AF6-88.5), anti-CD274 (PD-Ll)-APC (Clone MIH5) and anti CD16/CD32 (Clone 2.4G2) were purchased from BD Biosciences.
  • Antibodies were mixed together with PBS containing 1% FCS (Gibco).
  • Cyclophosphamide was purchased from Sigma, resuspended in PBS (Gibco) and stored at -20 °C before use.
  • HPV-16 E6 and E7-expressing TC-1 tumor cells were generated as previously described (Lin et al. Cancer Res. 1996 Jan 1 ;56(l):21-6): Primary lung cells of C57BL6 mice were transformed with HPV-16 E6 and E7 genes and with pVEJB -expressing activated human c-Ha-ras oncogene.
  • TC-1 cell line was cultured in Glutamax RPMI medium (Gibco supplemented with 100 U/ml penicillin, 100 pg/ml streptomycin, and 10 % fetal bovine serum). Lentiviral vector construction
  • the antigen (Ag) constructs were cloned in a pFlap-B2m-Ag-WPREMutee backbone (see for example WO2016012623 for the backbone).
  • the antigen plasmid contains the cPPT/CTS sequence (sequence SEQ ID NO: 37), mandatory for transduction of non- mitotic cells.
  • the U3 promoter sequence was deleted from the 3’ long terminal repeat (LTR) to avoid vector replication.
  • the Beta-2microglobuline (P2m) promoter controls vaccine antigen expression in all transduced cells, thereby, antigen will preferentially be expressed in APC (Antigen Presenting Cells).
  • the antigen plasmid contains a mutant form of the woodchuck hepatitis B virus (WHV) post-transcriptional regulatory element (WPRE) (sequence SEQ ID NO: 38).
  • the wild-type WPRE region contains a truncated form of the WHV X protein that may have oncogenic properties (Kingsman et al. , Gene Ther. 2005 Jan;12(l):3-4).
  • the mutant form of the WPRE used in our construct precludes expression of the truncated X protein by the inclusion of point mutations within the X protein start codon. Such mutant WPRE sequence appears not to have oncogenic properties (Themis et al., Mol Ther. 2005 Oct;12(4):763-71).
  • the packaging plasmid contains the gag-pol sequences from HIV-1 subtype NDK (GenBank acc n°: A34828). The proteins nef, vif, vpr, env are not expressed. Moreover, aspartic acid (D) to valine (V) replacement at position 64 (D64V) in the HIV-1 integrase protein sequence (pol gene) is sufficient to inhibit integration without disturbing the transgene expression in vitro.
  • the lentiviral particles according to the invention are non- integrative particles.
  • Envelope plasmid pCMV-VSV-G INDco (Indiana) et pCMV-VSV-G NJco (New Jersey) vectors were constructed by subcloning the Vesicular stomatitis virus (VSV) G protein (VSV-G) Indiana (GenBank acc. n° J02428) and New Jersey (GenBank acc. n° P04882) serotype inserts into the pVAXl expression vector (Invitrogen).
  • Mammalian codon- optimized synthetic genes (GeneArt) encoding glycoproteins from the following Vesiculovirus were cloned into a pVAXl plasmid (Invitrogen): Vesicular Stomatitis Virus Indiana serotype (GenBank FW591952), New Jersey serotype (GenBank FW591956) and Cocal virus (GenBank: AF045556.1).
  • GeneArt Mammalian codon- optimized synthetic genes encoding glycoproteins from the following Vesiculovirus were cloned into a pVAXl plasmid (Invitrogen): Vesicular Stomatitis Virus Indiana serotype (GenBank FW591952), New Jersey serotype (GenBank FW591956) and Cocal virus (GenBank: AF045556.1).
  • non integrative lentiviral particles were produced by transient calcium phosphate co-transfection of HEK 293 T cells (ATCC) with 3 plasmids (The viral antigen plasmid, an envelope expression plasmid and packaging plasmid) following the method well-known in the art.
  • Culture medium is replaced by serum free medium after 24h.
  • Supernatant is harvested and clarified 48h after transfection by 2500rpm centrifugation.
  • Viral particles are concentrated by ultracentrifugation (Ih at 22000 rpm/88250g 4oC) and resuspended in preservative buffer (20mM Pipes, 75mM NaCl and 2.5% sucrose).
  • Lentiviral vector titer was determined by quantitative PCR after transduction of cells (HEK 293 T). Aphidicolin is added to HEK293T cells 24h before transduction and is maintained during the whole titration process. Cells are incubated 30 min with lysis buffer (200mM Tris, 1% NP40 and 1% Tween20), containing 50pg/ml RNase A (sigma). Proteinase K (0.2mg/ml) is added to suspension and incubated 4h at 56°C. Pairs of primers specific for RRE (element of Ag vector) and GAPDH (in host cell) are used for quantitative PCR. Titer are Titer are expressed as transduction unit (TU)/mL of vector.
  • TU transduction unit
  • lentiviral vector particules of the invention comprising functionnal lentiviral vector of the invention
  • intramuscular injection i.m. 14 days later, splenocytes were prepared and restimulated overnight for IFNg ELISPOT with 4 distinct HPV peptide pools (each peptide is 2pg/mL final).
  • Each peptide pools correspond to one of the following non-oncogenic antigen variants: non-oncogenic variant of E6 protein of HPV 16, non-oncogenic variant of E7 protein of HPV 16, non-oncogenic variant of E6 protein of HPV 17 and non-oncogenic variant of E7 protein of HPV 17. They are composed of overlapant 15 mers (with overlaps of 11 a.a.) corresponding to the full selected antigen.
  • mice were shaved with electric shaver device before injection.
  • mice were randomized and injected with the LV vaccine of the invention via intramuscular (i.m.) injection. Mice were monitored for tumor growth by measuring tumor diameter with calipers 3 times a week. Due to ethical reasons, mice with tumors >1500mm3 had to be euthanized.
  • Frozen human PBMC (StemCell) were gently thawed, stained during lOmin at 37°C with 0.5pM of CFSE (Thermofischer). Cells were then cultured in rond bottom 96 wells plate (0.2x106 cells per well) in complete RPMI : 10% FCS, lOmM Hepes (Gibco), 100 U/ml Penicilline, 100 pg/ml Streptomycine, O.lmM Non-Essential Amino acids (Gibco) and ImM Sodium Pyruvate (Gibco). After 7 days, cells are centrifuged and new complete RPMI (prewarmed) is added. After another 7 days (14 days total), cells are stained with fluorescent antibodies and data are acquired by flow cytometry (MACSQuant analyzer) Cytometric analysis of tumor immune infiltrates
  • Tumors were treated with the Mouse Tumor Dissociation kit (Miltenyi). Cell suspensions were then filtered through 70 pm -pore filters, treated with Red Blood Cell lysis buffer (Sigma), then washed and centrifuged at 1200 rpm for 5 minutes. The recovered cells were stained as follows.
  • NK To detect NK, Near IR LD (Invitrogen), Fcyll/III receptor blocking anti- CD16/CD32 (clone 2.4G2, BD Biosciences), APC-anti-CDl lb (clone N418, BD Biosciences), BV421-anti-NKp46 (clone 29A1.4, Biolegend) were used.
  • Splenocytes from immunized mice were obtained by tissue homogenization and passage through 100-pm nylon filters (Cell Strainer, BD Biosciences) and were plated at 4 x 10 6 cells/well in 24- well plates. Splenocytes were stimulated during 6h in the presence of 10 pg/mL of homologous or control peptide, 1 pg/mL of anti-CD28 (clone 37.51) and 1 pg/mL of anti-CD49d (clone 9C10-MFR4.B) mAbs (BD Biosciences). During the last 3h of incubation, cells were treated with a mixture of Golgi Plug and Golgi Stop, both from BD Biosciences.
  • PE-Cy7-anti-CD107a (clone 1D4B, BioLegend) mAb was also added to the cultures at this step. Cells were then collected, washed with PBS containing 3% FBS and 0.1% NaN3 (FACS buffer) and incubated for 25 min at 4°C with a mixture of Near IR Live/Dead (Invitrogen), Fcyll/III receptor blocking anti-CD16/CD32 (clone 2.4G2), PerCP- Cy5.5-anti-CD3s (clone 145-2C11), PE-Cy7-anti-CD4 (clone RM4-5) and BV711-anti-CD8 (clone 53-6.7) mAbs (BD Biosciences or eBioscience).
  • HPV vaccines of the invention are immunogenic in vivo
  • mice were immunized with the 4 vaccines (1 group of 5 mice per tested vaccine and control group).
  • mice were injected i.m. with 1X10 7 TU of lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759, the lentiviral vector filed at the CNCM under accession number 1-5760, the lentiviral vector filed at the CNCM under accession number 1-5761 or the lentiviral vector filed at the CNCM under accession number 1-5762 or 50 pL of diluent. 14 days later, splenocytes were prepared and restimulated overnight for IFNg ELISPOT with 4 distinct peptide pools (each peptide is 2pg/mL final). The results obtained are represented in Figure 1.
  • Example 2 HPV vaccines of the invention vaccine fully eliminates well implanted tumors in vivo
  • TC-1 tumors cells have been extensively used as a preclinical model to study HPV induced Tumors (Kim, J W et al. Gene therapy vol. 11,12 (2004): 1011-8). These lung tumor cells were modified to express E6 and E7 from HPV 16 (Lin et al. Cancer Res. 1996 Jan l;56(l):21-6).
  • TC-1 cells were injected s.c. and tumor volume was measured every other day (caliper measurement).
  • tumor volume is 70 mm 3
  • mice were randomized and vaccinated with IxlO 8 TU i.m. of LV-GFP Indiana (as a control), Indiana lentiviral vector particles comprising 1-5759, Indiana lentiviral vector particles comprising 1-5760, Indiana lentiviral vector particles comprising 1-5761 or Indiana lentiviral vector particles comprising 1-5762.
  • a rapid and very efficient elimination of tumors is observed in 100% of animals vaccinated with lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5762 and the lentiviral vector filed at the CNCM under accession number 1-5759, 87.5% of animals vaccinated with the lentiviral vector filed at the CNCM under accession number 1-5760 and 75% of animals vaccinated with the lentiviral vector filed at the CNCM under accession number 1-5761.
  • Vaccines comprising lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759 and lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5762 show equivalent tumor elimination rates (higher than 1-5760 and 1-5761) but 1-5759 vaccination allows full elimination of tumors in 37.5 days (+/- 7.4 SD) on average whereas it takes 54.7 days after I- 5762 vaccination.
  • the lentiviral vector particle comprising the lentiviral vector filed at the CNCM under accession number 1-5759 was the most efficient anti tumoral vaccine whereas the lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number I- 5760 and 1-5762 were more immunogenic vectors when measuring IFN-y production.
  • Example 3 A single HPV Vaccine of the invention administration is efficient against tumor relapse
  • Relapse is commonly observed in most cancer type and is defined as a return of the disease after a period of improvement. It is often due to a few tumor cells that survived the initial treatment and form new tumors weeks, month or even years after treatment.
  • mice who eliminated primary tumor were rechallenged on the other flank at day 60.
  • Control mice (untreated) were also injected s.c. in order to check on the tumor cell injection.
  • This Figure shows that s.c. injection of TC-1 cells in control mice allows formation of solid tumors reaching ethical limits of size in less than 30 days. Tumor growth in rechallenged mice (that previously eliminated right flank tumors after vaccination) were strongly reduced. Tumor growth was observed during the first 6 days and tumor elimination begins afterwards.
  • mice are even fully eliminated 13-16 days after implantation.
  • Single dose vaccination administered on tumor bearing mice allowed full elimination of primary tumor and a strong protection against relapse as these mice rapidly eliminated secondary tumors.
  • B. A additional experiment was performed with a rechallenge of the mice who eliminated primary tumor on the other flank with 1.10 6 TC-1 tumor cells 119 days after the first engraftment and were maintained without any therapy. Control mice (untreated) were also injected s.c. in order to check on the tumor cell injection.
  • mice All re-challenged mice were still alive 145 days after the initial tumor challenge, sustaining that a single injection of a vaccine according to the invention effectively promoted a strong antitumor memory protective immune response which efficiently shaped T-cell responses to new challenges.
  • Example 4 Therapeutic effect of anti HPV vaccine of the invention is dose
  • mice bearing TCI tumor were vaccinated with the vaccine according to the invention comprising lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759, at IxlO 8 or IxlO 7 TU/mouse.
  • IxlO 6 TC-1 cells were injected in the flank of animals and tumor volume was measured twice a week (caliper measurement).
  • tumor volume was 80mm3
  • mice were randomized and vaccinated with diluent (control), 1X10 7 TU or 1X10 8 TU (i.m.) of 1-5759 vaccine
  • a single low dose (IxlO 7 ) of a vaccine according to the invention showed a partial inhibition comparable to what is observed with 3 injections of Adenoviral vector based vaccines (Rice,AE et al. Cancer gene therapy vol. 22,9 (2015): 454-62). This suggests that a vaccine according to the invention at optimal dose would be more efficient than adenoviral plateform. Moreover, low dose efficacy would most likely be increased by a second injection of vaccine.
  • Example 5 Vaccination according to the invention increases CD4 + and CD8 + T cell infiltration and reduces T reg in the treated tumors
  • Tumor infiltration was investigated to understand further the anti-tumoral mechanisms induced after vaccination with a lentiviral vector according to the invention.
  • IxlO 6 TCI tumor cells were injected (s.c.) on the flank of animal, and the tumor volume was measured twice a week (caliper measurement).
  • mice were randomized and vaccinated with either diluent (control), or 1X10 7 TU of 1-5759 or 1X10 8 TU (i.m.) of 1-5759.
  • tumors were collected, digested and analyzed by flow cytometry. FACS staining was performed and data were acquired on Macsquant facs according to methods well known in the art.
  • Tumors from vaccinated mice were infiltrated with more CD8 + and CD4 + T cells compared to control tumors.
  • the percentage of CD8 + T cells and CD4 + T cell in the tumors are increased respectively by about 4 and 3 times.
  • the percentage of CD25+FoxP3+CD4+ Regulatory T cells (Tregs) in tumors is strongly reduced in treated animals.
  • vaccines comprising lentiviral vectors of the invention improve CD8 + T cells and CD4 + T cell recruitment to the tumor but also reduces the percentage of Tregs in the tumors.
  • Example 6 HPV vaccine of the invention fully eliminates large tumors in vivo
  • mice When average tumor volume was around 300 mm 3 , mice were randomized and vaccinated with diluent (control), or 1X10 8 TU (i.m.) of the vaccine according to the invention comprising lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759. Tumor volume was measured twice a week with a caliper.
  • Example 7 HPV vaccine of the invention induces human PBMC activation in vitro
  • human PBMC StemCell
  • CFSE fibroblast proliferation and activation were measured after 2 weeks of culture.
  • CD8 + T cells and CD4 + T cells proliferation were increased by addition of lentiviral vectors of the invention in the culture.
  • antigen presenting cells from the PBMC are able to be transduced by an HPV vaccine according to the invention and to process the antigens to activate T cells.
  • Example 8 Systemic T-cell immunity induced by HPV vaccine of the invention and phenotype of effector T cells
  • mice injected with a Ctrl Lenti (LV-empty Indiana) or with a vaccine according to the invention comprising lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759 were left untreated or were stimulated in vitro with a mixture of ETTDPDRAHYNIVTF and PDRAHYNIVTFCCKC E7upvi6-derived peptides which contain the immunodominant H-2Db-restricted RAHYNIVTF epitope (Feltkamp MC et al.. Eur J Immunol 1993;23:2242-9) and were analyzed by IntraCellular Staining (ICS) for IL-2, and TNF-a and IFN-y.
  • ICS IntraCellular Staining
  • CD8 + T-cell responses in mice vaccinated with a lentiviral vector according to the invention.
  • the functional CD8 + T-cell effectors were mainly distributed among IFN-y + (single positive), TNF-a + IFN-y + or IL-2 + IFN-y + (double positive), and IL-2 + TNF-a + IFN-y + (triple positive) subsets (voir Figure 9B).
  • the majority of IFN-y + CD8 + T cells also expressed the surface CD107a degranulating marker, showing the effector properties of these T cells (voir Figure 9B).
  • Example 9 Features of tumor cells and tumor infiltrating innate immune cells in mice vaccinated with an HPV vaccine of the invention
  • NK Natural Killer
  • Example 10 Suboptimal Lenti-HPV-07 vaccination acts synergistically with anti-PDl immunotherapy
  • the inventors further investigated the potential synergy between a suboptimal dose of Lenti-HPV-07 (1-5759) vaccination and anti-PD-1 therapy.
  • the anti-PD-1 treatment began four days (D17, i.e. 17 days after s.c. administration of the TC-1 cells to the mice) after the injection of the vaccine according to the invention comprising lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759 (D13, i.e. 13 days after s.c. administration of the TC-1 cells to the mice).
  • the vaccine according to the invention comprising lentiviral vector particles comprising the lentiviral vector filed at the CNCM under accession number 1-5759
  • D13 i.e. 13 days after s.c. administration of the TC-1 cells to the mice.
  • Several injections of anti-PD-1 were performed (D17 as mentioned above, then D20, D22, D24, D28 and D31).
  • mice were administered a LV- empty Indiana (DI 3) (as a control) and four days later with an anti-PD-1 monoclonal antibody (D17, then D20, D22, D24, D28 and D31).
  • mice were administered a vaccine according to the invention (1-5759) (D13) and four days later with a control antibody (isotype Ctrl) (D17, then D20, D22, D24, D28 and D31).
  • the mice were administered a vaccine according to the invention (D13) (1-5759) and four days later with a mAb anti-PD-1 (D17, then D20, D22, D24, D28 and D31).
  • a suboptimal dose of the vaccine which induces an insufficient antitumor T-cell response, acted synergistically with anti-PD-1 to increase the tumor regression rate.
  • Only 3 out of 12 mice treated with the suboptimal dose of vaccine of the invention alone showed partial tumor regression. Accordingly, mice survival was significantly increased in the group of combo treatment, compared to the mice treated with suboptimal dose of Lenti-HPV-07 ( Figure 12B). Therefore, a synergistic anti-tumor effect can be achieved when Lenti-HPV-07 vaccine candidate is combined with the anti-PDl checkpoint inhibitory treatment.
  • Example 11 A single injection of vaccine according to the invention cures mice with pulmonorv metastatic foci induced bv intravenous injection of TCl-nLuc cells
  • TCI parental cell line was stably transduced with an integrative lentiviral vector encoding for nanoluciferase reporter and puromycin N-acetyl-transferase (for selection), under the ubiquitin (UBC) promoter. After selection on puromycin, cells were subcloned to obtain TCl-nLuc cell line.
  • UBC ubiquitin
  • mice Six-week-old C57BL/6JRj mice, purchased from Janvier Laboratory, were intravenously injected with 150 000 TCl-nLuc cells. At day 5, mice were injected with a single dose of LIO 9 TU / mouse of Lenti-HPV-07 or Control Lenti (empty vector) via intramuscular route.
  • Bioluminescence imaging on live animals was performed using the IVIS Imaging System (IVIS Spectrum, Perkin Elmer) coupled to a charged-couple device camera. Prior to bioluminescence imaging, mice were anesthetized with 2% isoflurane in oxygen and maintained in a control flow of 1.5% isoflurane in oxygen through a nose cone during imaging.
  • the substrate furimazine (Z108) (provided by Dr. Yves Janin, Institut Pasteur) was dissolved at 2 mg/ml in acidic ethanol. Z108 was further diluted in sterile D-PBS to the desired concentration (0.4 mg/kg) prior to intravenous injection. Mice were then immediately placed in the imaging chamber and imaged. Sequential images were captured under the autoexposure settings with a maximum exposure time of 2 minutes.
  • Lenti-HPV-07 vaccine (1-5759) according to the invention completely eradicates, in 100% of animals, subcutaneously engrafted TCI tumors.
  • many cancers, including HPV-induced cancers are located in mucosal sites.
  • the present experiment evaluated the capacity of Lenti-HPV-07 to inhibit tumor growth in a mucosal site.
  • TCI cell line expressing stably the nanoluciferase reporter gene (TCl-nLuc) was developed. After TCl-nLuc intravenous injection, mice readily developed lung metastatic foci.
  • mice received a single intramuscular injection of Lenti-HPV-07 (1.109 TU / mouse) or a Control Lenti (empty vector) (Figure 13A).
  • mice having received the Lenti-HPV-07 were cured at day 22 post tumor injection, whereas pulmonary metastatic foci continued to grow in the control group.
  • the difference observed between the average of the bioluminescence signal in the two groups was largely statistically significant ( Figures 13 A and B).
  • Example 12 Lenti-HPV-07 vaccination acts synergistically with anti- NKG2A immunotherapy
  • mice treated with Lenti-HPV-07 + anti-NKG2A mAb displayed a complete or partial regression response while only 40% of mice treated with Lenti-HPV-07 alone did (Fig 14 C).
  • the anti-TIM-3 mAb treatment began four days after Lenti- HPV-07 injection.
  • Control animals received IxlO 8 TU of Lenti-HPV-07 and a control Ig (Ctrl Ig).
  • Other groups received a Ctrl Lenti + Ctrl Ig or Ctrl Lenti + anti-TIM-3 mAb.
  • mice treated with Lenti-HPV-07 + anti-TIM-3 mAb displayed a complete or partial regression response while 40% of mice treated with Lenti-HPV-07 alone did (Fig 15 C).
  • the progression-free survival (PFS) time was higher in mice that received the combination of Lenti-HPV-07 and anti-TIM-3 mAb compared to the mice treated with the Lenti-HPV-07 + Ctrl Ig even though the difference did not reach statistical significance (Fig 15 D).
  • SEQUENCES is a nucleic acid sequence encoding the E6 protein from HPV 16
  • GGTCGATGTATGTCTTGTTGCAGATCATCAAGAACACGTAGAGAAACCCAGCTGTAA is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • AGCTGCTGCAGA is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • CGGGGCAGATGGACCGGCCGGTGCATGAGCTGCTGCAGA is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16 ATGGACCCCCAAGAACGGCCCAGAAAGCTGCCCCAGCTGTGCACCGAGCTGCAGACCAC
  • CATGAGCTGCTGCAGA is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • CATGTCCTGCTGCAGG is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 16
  • GCATGTCCTGCTGCAGGTCC is the amino acid sequence of the Wild Type (WT) E6 protein from HPV 16
  • DKKQRFHNIRGRWTGRCMSCCRSSRTRRETQL is the amino acid sequence of a non oncogenic variant of the E6 protein from
  • YSKISEYRHYCYSLYGTTLEQQYNKPLCDLLIRCINCQKPLRFHNIRGRWTGRCMSCCR is the amino acid sequence of a non oncogenic variant of the E6 protein from
  • MSCCR is the amino acid sequence of a non-oncogenic variant of the E6 protein from HPV 16
  • KFYSKISEYRHYCYSLYGTTLEQQYNKPLCDLLIRCINCQKPLRFHNIRGRWTGRCMSCCR is the amino acid sequence of a non-oncogenic variant of the E6 protein from HPV 16
  • LKFYSKISEYRHYCYSLYGTTLEQQYNKPLCDLLIRCINCQKPLRFHNIRGRWTGRCMSCCR is the amino acid sequence of a non-oncogenic variant of the E6 protein from HPV 16
  • LKFYSKISEYRHYCYSLYGTTLEQQYNKPLCDLLIRCINCQKPLRFHNIRGRWTGRCMSCCRS is the nucleic acid sequence encoding the E7 protein from HPV 16 (NP-
  • AGACCTGTTAATGGGCACACTAGGAATTGTGTGCCCCATCTGTTCTCAGAAACCATAA is a nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 16
  • TCGTGTGCCCCATT is a nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 16
  • CCCTGGGTATCGTCTGCCCCATC is the amino acid sequence of the Wild Type (WT) E7 protein from HPV 16
  • STLRLCVQSTHVDIRTLEDLLMGTLGIVCPICSQKP is the amino acid sequence of a non-oncogenic variant of the E7 protein from HPV 16
  • TPTLHEYMLDLQPETTDPDRAHYNIVTFCCKCDSTLRLCVQSTHVDIRTLEDLLMGTLGIVCPI is the amino acid sequence of a non-oncogenic variant of the E7 protein from HPV 16
  • VCPI is the nucleic acid sequence encoding the E6 protein from HPV 18
  • GCTGCAACCGAGCACGACAGGAAAGACTCCAACGACGCAGAGAAACACAAGTATAA ID NO: 20 is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 18
  • AACCCCGCCGAGAAGCTGCGGCACCTGAACGAGAAGCGGAGATTCCACAATATCGCC is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 18
  • GATCTGTTCGTGGTGTACCGGGACAGCATCCCCCACGCCGCCTGCCACAAG is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 18
  • GAAACTGAGACATCTGAACGAGAAAAGGAGGTTCCACAATATTGCCGGGCACTGATAA is the nucleic acid sequence encoding a non-oncogenic variant of the E6 protein from HPV 18
  • AAGGCCGAGAAGCTGAGGCATCTGAACGAGAAAAGGAGATTCCACAATATCGCCGGACA C is the amino acid sequence of the Wild Type (WT) E6 protein from HPV 18
  • HNIAGHYRGQCHSCCNRARQERLQRRRETQV is the amino acid sequence of a non-oncogenic variant of the E6 protein from HP V 18
  • GLYNLLIRCLRCQKPLNPAEKLRHLNEKRRFHNIA is the amino acid sequence of a non-oncogenic variant of the E6 protein from HP V 18
  • PYKLPDLCTELNTSLQDIEITCVYCKTVLELTEVFEFAFKDLFVVYRDSIPHAACHK is the amino acid sequence of a non-oncogenic variant of the E6 protein from HP V 18
  • GLYNLLIRCLRCQKAEKLRHLNEKRRFHNIAGH is the amino acid sequence of a non-oncogenic variant of the E6 protein from HP V 18
  • TNTGLYNLLIRCLRCQKAEKLRHLNEKRRFHNIAGH is the nucleic acid sequence encoding the E7 protein from HPV 18
  • GTGCATCCCAGCAGTAA is the nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 18
  • ACCCTGTCCTTCGTGTGCCCTTGG is the nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 18
  • TGAACACCCTGTCCTTCGTGTGCCCCTGG is the nucleic acid sequence encoding a non-oncogenic variant of the E7 protein from HPV 18
  • CGTGTGCCCCTGG is the amino acid sequence of the Wild Type (WT) E7 protein from HPV 18
  • MCCKCEARIELVVESSADDLRAFQQLFLNTLSFVCPWCASQQ is the amino acid sequence of a non-oncogenic variant of the E7 protein from HPV 18
  • ESSADDLRAFQQLFLNTLSFVCPW is the amino acid sequence of a non-oncogenic variant of the E7 protein from HPV 18
  • VVESSADDLRAFQQLFLNTLSFVCPW is the amino acid sequence of a non-oncogenic variant of the E7 protein from HPV 18 GPKATLQDIRLEPQNEIPVDSEEENDEIDGNHQHLPARRAEPQRHTMLCMCCKCEARIKLVVE
  • SSADDLRAFQQLFLDSFVCPW is the nucleic acid sequence encoding the cPPT/CTS sequence
  • TT is the nucleic acid sequence encoding a mutant form of the woodchuck hepatitis B virus (WHV) post-transcriptional regulatory element (WPRE)
  • GCCTTCGCCCTCAGACGAGTCGGATCTCCCTTTGGGCCGCCTCCCCGC is a synthetic E7upvi6-derived peptide containing the RAHYNIVTF H-2D b - restricted T-cell epitope
  • ETTDPDRAHYNIVTF is a synthetic E7upvi6-derived peptide containing the RAHYNIVTF H-2D b - restricted T-cell epitope
  • PDRAHYNIVTFCCKC is the nucleic acid sequence encoding an antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5759
  • GAGAAAAGGAGGTTCCACAATATTGCCGGGCACTGATAA is the amino acid sequence of the antigen construct encoded by the lentiviral vector filed at the CNCM under accession number 1-5759
  • PHAACHKLEKLTNTGLYNLLIRCLRCQKAEKLRHLNEKRRFHNIAGH is the nucleic acid sequence encoding an antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5760 ATGTTCCAGGACCCCCAGGAGAGGCCCCGGAAGTTGCCCCAGCTGTGCACCGAG
  • GGCCTGATAA is the amino acid sequence of the antigen construct encoded by the lentiviral vector filed at the CNCM under accession number 1-5760
  • CCKCEARIKLVVESSADDLRAFQQLFLNTLSFVCPWA ID NO: 45 is the nucleic acid sequence encoding an antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5761
  • GTGTGCCCCATCGCCTGATAA is the amino acid sequence of the antigen construct encoded by the lentiviral vector filed at the CNCM under accession number 1-5761
  • YNIVTFCCKCDSTLRRCVQSTHVDIRTLEDLLMGTLGIVCPIA is the nucleic acid sequence encoding an antigen construct of the lentiviral vector filed at the CNCM under accession number 1-5762
  • CATGTCCTGCTGCAGGTCCTGATAA is the amino acid sequence of the antigen construct encoded by the lentiviral vector filed at the CNCM under accession number 1-5762
  • SEO ID NO: 49 is amino acid sequence of the_H-2D b -restricted T-cell epitope

Landscapes

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

Abstract

La présente invention concerne un vecteur lentiviral, en particulier un vecteur lentiviral non intégratif, ou une particule de vecteur lentiviral, en particulier une particule de vecteur lentiviral non intégratif, destiné à être utilisé dans le traitement ou la prévention d'un cancer induit par le HPV, le vecteur lentiviral comprenant au moins quatre séquences d'acide nucléique distinctes ; la particule de vecteur lentiviral comprenant au moins l'un du vecteur lentiviral ; le vecteur lentiviral ou la particule de vecteur lentiviral étant administré en combinaison avec au moins un inhibiteur de point de contrôle immunitaire.
PCT/EP2023/070672 2022-07-27 2023-07-26 Vecteurs lentiviraux pour l'expression d'antigènes du papillomavirus humain (hpv) et leur mise en oeuvre dans le traitement de cancers induits par le hpv WO2024023135A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP22306119.3 2022-07-27
EP22306119 2022-07-27

Publications (1)

Publication Number Publication Date
WO2024023135A1 true WO2024023135A1 (fr) 2024-02-01

Family

ID=83271664

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/070672 WO2024023135A1 (fr) 2022-07-27 2023-07-26 Vecteurs lentiviraux pour l'expression d'antigènes du papillomavirus humain (hpv) et leur mise en oeuvre dans le traitement de cancers induits par le hpv

Country Status (1)

Country Link
WO (1) WO2024023135A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2169073A1 (fr) 1999-10-11 2010-03-31 Institut Pasteur Vecteurs pour la préparation des compositions immunotherapeutiques
US20120315296A1 (en) * 2007-08-03 2012-12-13 Pierre Charneau Lentiviral gene transfer vectors and their medicinal applications
WO2016012623A1 (fr) 2014-07-25 2016-01-28 Theravectys Vecteurs lentiviraux pour l'expression régulée d'une molécule récepteur d'antigène chimère

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2169073A1 (fr) 1999-10-11 2010-03-31 Institut Pasteur Vecteurs pour la préparation des compositions immunotherapeutiques
US20120315296A1 (en) * 2007-08-03 2012-12-13 Pierre Charneau Lentiviral gene transfer vectors and their medicinal applications
WO2016012623A1 (fr) 2014-07-25 2016-01-28 Theravectys Vecteurs lentiviraux pour l'expression régulée d'une molécule récepteur d'antigène chimère

Non-Patent Citations (60)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. AF045556.1
"Remington's Pharmaceutical Sciences", 2003, LIPPINCOTT WILLIAMS & WILKINS
ALDO VENUTIFRANCESCA PAOLINI, HEAD NECK PATHOL, vol. 6, July 2012 (2012-07-01), pages 63 - 74
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., NUCLEIC ACIDS RES, vol. 25, 1997, pages 3389 - 3402
ARHEL ET AL., EMBO J, vol. 26, no. 12, 20 June 2007 (2007-06-20), pages 3025 - 3037
BERRAONDOPEDRO ET AL., CANCER RESEARCH, vol. 67, no. 18, 2007, pages 8847 - 55
BONKABARA ET AL., J. IMMUNOLOGY, vol. 167, 2001, pages 6893 - 6900
BORYSIEWICZ L K ET AL: "A recombinant vaccinia virus encoding human papillomavirus types 16 and 18, E6 and E7 proteins as immunotherapy for cervical cancer", THE LANCET, vol. 347, no. 9014, 20 December 2003 (2003-12-20), pages 1523 - 1527, XP085558750, ISSN: 0140-6736, DOI: 10.1016/S0140-6736(96)90674-1 *
BOURSNELL M E G ET AL: "Construction and characterisation of a recombinant vaccinia virus expressing human papillomavirus proteins for immunotherapy of cervical cancer", VACCINE, ELSEVIER, AMSTERDAM, NL, vol. 14, no. 16, 1 November 1996 (1996-11-01), pages 1485 - 1494, XP004063302, ISSN: 0264-410X, DOI: 10.1016/S0264-410X(96)00117-X *
BRUDNO M., BIOINFORMATICS, vol. 19, 2003, pages 154 - 162
CASSETTI, M CRISTINA ET AL., VACCINE, vol. 22, no. 3-4, 2004, pages 520 - 7
CAVAZZANA-CALVO ET AL., SCIENCE, vol. 288, no. 5466, 2000, pages 669 - 72
CHEN ET AL., VIROLOGY, vol. 516, 2018, pages 86 - 101
CHINNASAMY ET AL., HUM GENE THER, vol. 11, no. 13, 2000, pages 1901 - 9
COUTANT ET AL., PLOS ONE, vol. 7, no. 11, 2012, pages e48644
DALPHIN ET AL., NUCL ACIDS RES, vol. 24, 1996, pages 216 - 8
DE SANJOSE ET AL., EUR J CANCER, vol. 49, no. 16, November 2013 (2013-11-01), pages 3450 - 61
FELICIA GRASSO ET AL: "Successful therapeutic vaccination with integrase defective lentiviral vector expressing nononcogenic human papillomavirus E7 protein", INTERNATIONAL JOURNAL OF CANCER, JOHN WILEY & SONS, INC, US, vol. 132, no. 2, 28 June 2012 (2012-06-28), pages 335 - 344, XP071287079, ISSN: 0020-7136, DOI: 10.1002/IJC.27676 *
FELTKAMP MC ET AL., EUR J IMMUNOL, vol. 23, 1993, pages 2242 - 9
GHITTONI, RAFFAELLA ET AL., VIRUS GENES, vol. 40, no. 1, 2010, pages 1 - 13
GRASSO ET AL., INT J CANCER, vol. 132, no. 2, 15 January 2013 (2013-01-15), pages 335 - 44
GRUH ET AL., J GENE MED, vol. 10, no. 1, 2008, pages 21 - 32
HACEIN-BEY-ABINA ET AL., J.CLIN.INVEST., vol. 118, no. 9, 2008, pages 3132 - 3142
HE ET AL., EXPERT REV VACCINES, vol. 6, no. 6, 2007, pages 913 - 24
HE ET AL., ONCO TARGETS THER, vol. 11, 2018, pages 7005 - 7009
HU ET AL., VACCINE, vol. 28, 2010, pages 6675 - 6683
HUIBREGTSE, J M ET AL., THE EMBO JOURNAL, vol. 10, 13, 1991, pages 4129 - 35
KARLINALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 5877
KARWACZ ET AL., J. VIROL., vol. 83, no. 7, 2009, pages 3094 - 3103
KIM, J W ET AL., GENE THERAPY, vol. 11, no. 12, 2004, pages 1011 - 8
KIMURA ET AL., MOL THER, vol. 15, no. 7, 2007, pages 1390 - 9
KINGSMAN ET AL., GENE THER, vol. 12, no. l, January 2005 (2005-01-01), pages 3 - 4
KRUSKAL, J. B.: "Time warps, string edits and macromolecules: the theory and practice of sequence comparison", 1983, ADDISON WESLEY, article "An overview of sequence comparison", pages: 1 - 44
KU ET AL., EXPERT REVIEW OF VACCINES, 2021
LIN ET AL., CANCER RES., vol. 56, no. 1, 1 January 1996 (1996-01-01), pages 21 - 6
LOPES ET AL., J VIROL, vol. 82, no. 1, 2008, pages 86 - 95
MARTINEZ-ZAPIEN, DENISE ET AL., NATURE, vol. 529, no. 7587, 2016, pages 541 - 5
MILONEO'DOHERTY, LEUKEMIA, vol. 32, 2018, pages 1529 - 1541
MIYOSHI H ET AL., J VIROL, vol. 72, no. 12, 1998, pages 9873 - 80
MORROW, MATTHEW P ET AL., EXPERT REVIEW OF VACCINES, vol. 12, no. 3, 2013, pages 271 - 83
MURRAY ET AL., NUCL ACIDS RES, vol. 17, 1989, pages 477 - 508
MYERSMILLER, CABIOS, vol. 4, 1989, pages 11 - 17
NEEDLEMAN, S. B.WUNSCH, C. D., J. MOL. BIOL., vol. 48, 1970, pages 443 - 453
NEGRI ET AL., MOLECULAR THERAPY, vol. 15, no. 9, 2007, pages 1716 - 1723
PENG SHIWEN ET AL: "Development of DNA Vaccine Targeting E6 and E7 Proteins of Human Papillomavirus 16 (HPV16) and HPV18 for Immunotherapy in Combination with Recombinant Vaccinia Boost and PD-1 Antibody", MBIO, vol. 12, no. 1, 23 February 2021 (2021-02-23), US, XP093033206, ISSN: 2161-2129, Retrieved from the Internet <URL:https://journals.asm.org/doi/pdf/10.1128/mBio.03224-20> DOI: 10.1128/mBio.03224-20 *
RICE A E ET AL: "An HPV-E6/E7 immunotherapy plus PD-1 checkpoint inhibition results in tumor regression and reduction in PD-L1 expression", CANCER GENE THERAPY, NATURE PUBLISHING GROUP US, NEW YORK, vol. 22, no. 9, 1 September 2015 (2015-09-01), pages 454 - 462, XP036973509, ISSN: 0929-1903, [retrieved on 20150904], DOI: 10.1038/CGT.2015.40 *
RICE, PLONGDENJBLEASBY,A: "EMBOSS: The European Molecular Biology Open Software Suite", TRENDS IN GENETICS, vol. 16, no. 6, 2000, pages 276 - 277, XP004200114, Retrieved from the Internet <URL:http://emboss.bioinformatics.nl> DOI: 10.1016/S0168-9525(00)02024-2
RICE,AE ET AL., CANCER GENE THERAPY, vol. 22, no. 9, 2015, pages 454 - 62
ROUAS ET AL., CANCER GENE THER, vol. 9, no. 9, 2008, pages 715 - 24
SARAIYA ET AL., J NATL CANCER INST, vol. 107, no. 6, 29 April 2015 (2015-04-29)
STEINMAN, R.BANCHEREAU, J., NATURE, vol. 449, 2007, pages 419 - 426
THEMIS ET AL., MOL THER, vol. 12, no. 4, October 2005 (2005-10-01), pages 763 - 71
THOMPSON, J. D.HIGGINS, D. G.GIBSON, T. J., NUCLEIC ACIDS RES, vol. 22, 1994, pages 4673 - 80
TOMAICVJEKOSLAV, CANCERS, vol. 8, 19 October 2016 (2016-10-19), pages 10 - 95
TOTA ET AL., PREV MED, vol. 53, October 2011 (2011-10-01), pages 12 - 21
VAN MONTFOORT ET AL., CELL, vol. 175, no. 7, 13 December 2018 (2018-12-13), pages 1744 - 1755
YIMPARK, CANCER RES TREAT, vol. 37, no. 6, December 2005 (2005-12-01), pages 319 - 24
ZENNOU ET AL., CELL, vol. 101, no. 2, 14 April 2000 (2000-04-14), pages 173 - 85
ZUFFEREY ET AL., J V/RO, vol. 72, no. 12, 1998, pages 9873 - 80

Similar Documents

Publication Publication Date Title
JP6688535B2 (ja) ワクチン組成物
JP6655051B2 (ja) 発癌性ウイルスポリペプチド陽性腫瘍治療用ポリヌクレオチド
AU2018283811B2 (en) Poxvirus vectors encoding HIV antigens, and methods of use thereof
JPWO2012053646A1 (ja) ワクシニアウイルスベクターおよびセンダイウイルスベクターからなるプライム/ブーストワクチン用ウイルスベクター
CA2675257A1 (fr) Vaccin combine contre le vih et procede de primo-vaccination/rappel
WO2023084094A1 (fr) Vecteurs lentiviraux pour l&#39;expression d&#39;antigènes du papillomavirus humain (hpv) et leur mise en oeuvre dans le traitement de cancers induits par le hpv
US10052377B2 (en) Cellular vaccine and method of inducing an immune response in a subject
WO2024023135A1 (fr) Vecteurs lentiviraux pour l&#39;expression d&#39;antigènes du papillomavirus humain (hpv) et leur mise en oeuvre dans le traitement de cancers induits par le hpv
CN109923212B (zh) 用于表达乙肝病毒(hbv)抗原的慢病毒载体
JPWO2007102326A1 (ja) アデノウイルス5型/35型ベクターとワクシニアウイルスmvaベクターとの併用による強力な免疫誘導
JP2024509976A (ja) 抗原をmhc-ii経路にターゲティングし、宿主におけるcd8+及びcd4+t細胞による防御免疫を誘導するレンチウイルスベクター

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23748062

Country of ref document: EP

Kind code of ref document: A1