US20190177733A1 - Cancer vaccine for dogs - Google Patents

Cancer vaccine for dogs Download PDF

Info

Publication number
US20190177733A1
US20190177733A1 US16/197,563 US201816197563A US2019177733A1 US 20190177733 A1 US20190177733 A1 US 20190177733A1 US 201816197563 A US201816197563 A US 201816197563A US 2019177733 A1 US2019177733 A1 US 2019177733A1
Authority
US
United States
Prior art keywords
tert
dog
nucleic acid
sequence
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/197,563
Inventor
Pierre Langlade Demoyen
Simon Wain-Hobson
Christelle LIARD
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Invectys SAS
Original Assignee
Invectys SAS
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 Invectys SAS filed Critical Invectys SAS
Priority to US16/197,563 priority Critical patent/US20190177733A1/en
Assigned to INVECTYS reassignment INVECTYS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LANGLADE DEMOYEN, PIERRE, WAIN-HOBSON, SIMON
Publication of US20190177733A1 publication Critical patent/US20190177733A1/en
Assigned to INVECTYS reassignment INVECTYS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LIARD, Christelle
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/52Genes encoding for enzymes or proenzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001154Enzymes
    • A61K39/001157Telomerase or TERT [telomerase reverse transcriptase]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • 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
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • A61K2039/552Veterinary vaccine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • C12Y207/07049RNA-directed DNA polymerase (2.7.7.49), i.e. telomerase or reverse-transcriptase

Definitions

  • the present invention relates to cancer vaccination in dogs.
  • the incidence rate of cancers in the canine population is estimated to be between 282.2 to 958 per 100,000 dogs (Merlo et al. 2008, Vascellari et al. 2009).
  • the most frequent tumors in dogs are mammary tumors in females (70.5% of all cancers), non-Hodgkin's lymphomas (8.4% in females and 20.1% in males) and skin tumors (4% in females and 19.9% in males).
  • 50% of dogs over ten years are going to die from a cancer-related problem.
  • the panel of treatments available against veterinary cancer is substantially reduced compared with those available in human oncology.
  • Radiotherapy is another important means to treat certain types of cancers in the veterinary field. It is of particular interest for tumors which are hardly accessible for surgery like cerebral tumors (de Fornel et al. 2007). Furthermore, recent studies in humans have demonstrated that ionizing radiation (IR) could act as an immunomodulator by inducing substantial changes in the tumor microenvironment, including triggering an inflammatory process. Furthermore, the cost and the availability of the material make access to radiation therapy complicated for companion animals. Chemotherapy is more and more used in animal oncology (Marconato 2011). Taking advantages of medical advances in human cancer therapy, there are more and more molecules available like vincristine, cyclophosphamide, carboplatin or cisplatin, to treat companion animals.
  • anticancer drugs are particularly used in the treatment of tumors derived from hematopoietic tissue (lymphomas, leukemias).
  • lymphomas hematopoietic tissue
  • Chemotherapeutic agents can be particularly efficient in prolonging the life span of a cancerous animal from a few weeks to several months (the median survival time of dogs treated with the CHOP protocol is 13 months).
  • the side effects dreaded by human patients, such as vomiting, diarrhea, hair loss are usually less frequent in companion animals.
  • most of the time chemotherapy is not curative in pets and the tumor often escapes treatment.
  • TILs Tumor Infiltrating Lymphocytes
  • the tumor stroma contains many suppressive elements including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDCs); soluble factors such as interleukin 6 (IL-6), IL-10, vascular endothelial growth factor (VEGF), and transforming growth factor beta (TGF ⁇ ) that down modulate antitumor immunity (Finn 2008, Hanahan and Weinberg 2011). Consequently, the choice of a pertinent tumor associated antigen (TAA) and the bypass of cancer associated immunosuppression are two critical points for a therapeutic vaccine to succeed (Disis et al. 2009).
  • TAA tumor associated antigen
  • TAA tumor associated antigen
  • the inventors now propose a cancer vaccine strategy for dogs, based on the telomerase reverse transcriptase (TERT).
  • TERT telomerase reverse transcriptase
  • a subject of the invention is thus an immunogenic composition
  • a nucleic acid that comprises a sequence encoding (i) a dog TERT deprived of telomerase catalytic activity, or (ii) a fragment thereof.
  • the nucleic acid is preferably DNA, preferably in form of a plasmid.
  • the nucleic acid that comprises a sequence encoding a dog telomerase reverse transcriptase (TERT) deprived of telomerase catalytic activity, wherein the sequence encoding dog TERT is further deprived of a nucleolar localization signal.
  • TERT dog telomerase reverse transcriptase
  • the nucleic acid further comprises a non-dog TERT antigenic fragment.
  • a further subject of the invention is a nucleic acid that comprises a sequence encoding (i) a dog TERT deprived of telomerase catalytic activity, or (ii) a fragment thereof, and optionally further comprises a non-dog TERT antigenic fragment.
  • the immunogenic composition or the nucleic acid is useful in triggering an immune response in a dog, against cells that over-express telomerase, such as dysplasia cells or tumor cells.
  • the immunogenic composition or the nucleic acid is thus particularly useful in treating a tumor in a dog, preferably by intradermal or intramuscular route.
  • Such treatment can be referred to as an active immunotherapy or a therapeutic vaccination, as it triggers an immune response against the tumor, especially a cytotoxic CD8 T cell response, along with a specific CD4 T cell response.
  • the invention makes it possible to induce dTERT specific responses in dogs with neoplasias and so can be used for immunotherapeutic treatments of the neoplasias in a clinical setting.
  • the invention is also useful to induce dTERT specific responses in healthy dogs that could be at risk for cancer, e.g. by genetic predisposition, or in healthy dogs from a certain age (e.g. more than 10 years, preferably more than 12 years old), so as to prevent the onset of cancer.
  • the treatment of the invention may induce long term immune memory responses in healthy dogs, dogs at risk of developing a cancer and those presenting a cancer.
  • FIG. 1A shows pDUV5 nucleotide sequence (SEQ ID NO: 1) and corresponding amino acid sequence comprising dog TERT (dTERT) amino acid sequence (SEQ ID NO: 2).
  • the plasmid pDUV5 encodes a near full length dog TERT nucleotide sequence.
  • the nucleotide sequence encoding 3 key amino acids in the catalytic site of the protein have been deleted (VDD).
  • the sequence controlling the importation into the nucleoli has been deleted (nucleotide sequence encoding 47 first Amino Acids in the N-term sequence of dTERT protein).
  • the DNA sequence encoding the human ubiquitin has been added upstream the dTERT sequence. Presence of the ubiquitin protein enhances the addressing of the dTERT protein to the proteasome and increases class I presentation of derived peptides.
  • the human and dog ubiquitin sequences are identical at the protein level, there is no biological incompatibility. Downstream the dTERT sequence, the sequence of the V5 peptide of the flu was inserted to facilitate the detection of the protein.
  • FIG. 1B shows pCDT nucleotide sequence (SEQ ID NO: 3) and corresponding amino acid sequence containing cat/dog hybrid TERT amino acid sequence (SEQ ID NO:4).
  • the plasmid pCDT encodes the cat/dog hybrid TERT (hyTERT) comprising 54.4% from the cat TERT and 35.9% from the dog TERT sequence.
  • the nucleotide sequence encoding 3 key amino acids in the catalytic site of the protein have been deleted (VDD).
  • the sequence controlling the importation into the nucleoli has been depleted (nucleotide sequence encoding 47 first Amino Acids in the Nter sequence of hyTERT protein).
  • the DNA sequence encoding the human ubiquitin has been added upstream the hyTERT sequence.
  • the presence of the ubiquitin protein enhances the addressing of the hyTERT protein to the proteasome and increases class I presentation of the derived peptides. Downstream the hyTERT sequence, the sequence of the V5 peptide of the flu was inserted to facilitate the detection of the protein.
  • FIG. 2A shows a simplified map of pcDNA3.1 expression plasmid into which the dog or hybrid TERT nucleic acid sequences are cloned.
  • FIG. 2B shows dog TERT protein sequence (SEQ ID NO: 5). The region covered by the dTERT 15mer peptide pool overlapping by 11 residues (70 peptides in total) that is used for in vitro immunization studies and ELIspot assays in dog PBMCs is shown in grey.
  • FIG. 4 is a graph showing that mice immunized with pDUV5 mount specific interferon- ⁇ -secreting CD8 T-cell responses against H2 restricted dog TERT peptides.
  • mice 7 week-old C57/B16 female mice were immunized intradermally (ID) or intramuscularly (IM) (10 mice per group) with 100 ⁇ g pDUV5 plasmid at day 0 and boost 14 days later.
  • control mice received PBS via ID or IM route (6 mice per group).
  • spleens of all mice were harvested.
  • Splenocytes were Ficoll purified and stimulated in triplicates with 5 ⁇ g/mL of relevant class I peptides (p580, p621 or p98′7) for 19 hours. Spots were revealed with a biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution. Results are the mean ⁇ standard deviation. Mann Whitney non parametric test, *p-value ⁇ 0.05, **: p-value ⁇ 0.01.
  • FIGS. 5A and 5B show that ID or IM immunization of mice with pDUV5 plasmid results in a dog TERT specific cytotoxic T-lymphocyte (CTL) response measurable in vivo by elimination of transferred target cells which were pulsed with dog TERT peptides restricted to H2.
  • CTL cytotoxic T-lymphocyte
  • mice Seven week-old C57/B16 female mice were immunized intradermally (ID) or intramuscularly (IM) with 100 ⁇ g pDUV5 plasmid at day 0 and day 14 post-priming.
  • ID intradermally
  • IM intramuscularly
  • CFSE carboxyfuorescein-diacetate succinimidyl ester
  • the same number of high, medium or low CFSE labeled cells was transferred IV to vaccinated mice. After 15-18 hours, the disappearance of peptide-pulsed cells was determined by fluorescence-activated cell-sorting analysis in the spleen. The percentage of specific lysis was calculated by comparing the ratio of pulsed to un-pulsed cells in vaccinated versus control mice.
  • FIGS. 6A and 6B show IFN ⁇ +specific CD8 and CD4 T-cell responses against H2 restricted hyTERT peptides in mice immunized with pCDT.
  • mice Seven week-old female mice were immunized intradermally (ID) or intramuscularly (IM) with either 100 ⁇ g pCDT plasmid or PBS at day 0 and boost 14 days later. Ten day post-boost, spleens were harvested. Splenocytes were Ficoll-purified and stimulated in triplicates with 5 ⁇ g/mL of relevant peptides for 19 hours. Spots were revealed with a biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution.
  • Plasmid vaccinated groups were composed of five C57/B16 mice, and control groups, of three mice. Splenocytes were stimulated with class I peptides p580, p621 and p987. Results show the frequency of peptide specific IFN- ⁇ producing CD8 T cells.
  • Plasmid vaccinated groups were composed of 9 Balb/cBy mice immunized IM and 5 ID. Control groups of 8 Balb/cBy mice injected IM and 4 ID. Splenocytes were stimulated with class II peptides p951, p1105, p1106 and p1109. Results show the frequency of peptide specific IFN- ⁇ producing CD4 T cells.
  • Results are the mean ⁇ standard deviation. Mann Whitney non parametric test, *p-value ⁇ 0.05, **: p-value ⁇ 0.01.
  • FIGS. 7A and 7B show hyTERT specific cytotoxic T-lymphocyte (CTL) response in mice immunized with pCDT plasmid, measurable in vivo by elimination of transferred target cells which were pulsed with hybrid TERT peptides restricted to H2.
  • CTL cytotoxic T-lymphocyte
  • mice 7 week-old C57/B16 female mice were immunized ID or IM with 100 ⁇ g pCDT plasmid at day 0 and day 14 post-priming.
  • CFSE carboxyfluorescein-diacetate succinimidyl ester
  • FIG. 8 shows principle of in vitro immunization in dog PBMCs
  • Frozen dog PBMCs were incubated with recombinant canine GM-CSF (rcGM-CSF) and canine IL-4 (rcIL-4) or human FlT3 (hFlT3) ligand for 24h.
  • Maturation stimuli rcTNF ⁇ , hIL-7 and rcIL-1 ⁇
  • rcTNF ⁇ , hIL-7 and rcIL-1 ⁇ were then added with dTERT overlapping peptides pools for 3 days. Eleven or 18 days of culture were performed and, TERT specific T cells were then detected via an IFN- ⁇ ELISpot assay.
  • FIGS. 9A and 9B show a repertoire of dTERT specific IFN- ⁇ secreting T cells in PBMCs from a na ⁇ ve dog
  • FIGS. 10A and 10B show the kinetics of the specific IFN ⁇ T cell response against a first pool of dTERT peptides (pool 6), and a second pool of dTERT peptides (pool 19), respectively.
  • FIGS. 11A and 11B show the kinetics of the specific IFN ⁇ T cell response against pool 6 dTERT peptides, and pool 10 dTERT peptides, respectively.
  • pDUV5 DNA vaccination at days 57 and 142 show classical long term memory responses, that is rising sharply and decaying more slowly.
  • FIG. 12 shows that tumor bearing dogs and healthy dogs have dTERT specific T lymphocytes (pool 4 peptides). Peripheral blood was drawn and in vitro stimulation protocol was performed as described Martinuzzi zt al, 2011.
  • telomerase consists of an RNA template and protein components including a reverse transcriptase, designated “Telomerase Reverse Transcriptase” (TERT), which is the major determinant of telomerase activity.
  • TERT Telomerase Reverse Transcriptase
  • dog TERT refers to the TERT sequence of any domestic dog (also designated Canis familiaris or Canis lupus familiaris ).
  • a dog TERT mRNA sequence is available with NCBI accession number NM 001031630 (XM 545191).
  • Dog TERT amino acid sequence is shown as SEQ ID NO:5.
  • the invention can make also use of non-dog telomerase (TERT) sequence, which can be from any human or non-human mammal, e.g. from cat.
  • TERT non-dog telomerase
  • cat TERT refers to the TERT sequence of any domestic cat (also designated as Felis catus or Felis silvestris catus ). Partial molecular cloning of the cat TERT gene (237 bp of mRNA) has been reported by Yazawa et al. 2003. The inventors herein provide a longer sequence of Felis catus TERT. The corresponding amino acid sequence is shown as SEQ ID NO:7.
  • telomerase catalytic activity refers to the activity of TERT as a telomerase reverse transcriptase.
  • deprived of telomerase catalytic activity means that the nucleic acid sequence encodes a mutant TERT, which is inactive.
  • hybrid or “chimeric” amino acid or nucleotide sequence means that part of the sequence originates from one animal species and at least another part of the sequence is xenogeneic, i.e. it originates from at least one other animal species.
  • fragment preferably refers to fragment of at least 10 amino acids, preferably at least 20 amino acids, still preferably at least 30, 40, 50, 60, 70, 80 amino acid fragments.
  • an epitope refers to an amino acid sequence comprising one or several epitopes that induce T cell response in the animal, preferably cytotoxic T lymphocytes (CTLs).
  • An epitope is a specific site which binds to a T-cell receptor or specific antibody, and typically comprises about 3 amino acid residues to about 30 amino acid residues, preferably 8 or 9 amino acids as far as class I MHC epitopes are concerned, and preferably 11 to 25 amino acids as far as class II MHC epitopes are concerned.
  • immunogenic means that the composition or construct to which it refers is capable of inducing an immune response upon administration (preferably in a dog).
  • Immunogenic response in a subject refers to the development of a humoral immune response, a cellular immune response, or a humoral and a cellular immune response to an antigen.
  • a “humoral immune response” refers to one that is mediated by antibodies.
  • a “cellular immune response” is one mediated by T-lymphocytes. It includes the production of cytokines, chemokines and similar molecules produced by activated T-cells. Immune responses can be determined using standard immunoassays and neutralization assays for monitoring specifically the humoral immune response, which are known in the art.
  • the immune response preferably encompasses stimulation or proliferation of cytotoxic CD8 T cells and/or CD4 T cells.
  • treatment includes curative treatment. More particularly, curative treatment refers to any of the alleviation, amelioration and/or elimination, reduction and/or stabilization (e.g., failure to progress to more advanced stages) of a symptom, as well as delay in progression of the tumor or dysplasia or of a symptom thereof.
  • the term “prevention” or “preventing” refers to the alleviation, amelioration and/or elimination, reduction and/or stabilization (e.g., failure to progress to more advanced stages) of a prodrome, i.e. any alteration or early symptom (or set of symptoms) that might indicate the start of a disease before specific symptoms occur.
  • a cell that “overexpresses telomerase” refers to a cell in a subject, which either expresses telomerase, e.g. upon mutation or infection, whereas it does usually not, under normal conditions, or to a cell in a subject which expresses a higher level of telomerase (e.g. upon mutation or infection), when compared to normal conditions.
  • the cell that overexpresses telomerase shows an increase of expression of at least 5%, at least 10%, at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or more.
  • nucleic acid that comprises a sequence encoding (i) a dog telomerase reverse transcriptase (TERT) deprived of telomerase catalytic activity, or (ii) a fragment thereof.
  • the nucleic acid may be DNA or RNA, but is preferably DNA, still preferably double stranded DNA.
  • the TERT sequence is deprived of telomerase catalytic activity.
  • the sequence that encodes dog TERT contains mutations that provide inactivation of the catalytic activity.
  • the term “mutation” includes substitution of one or several amino acids, a deletion of one or several aminoacids, and/or an insertion of one of several amino acids.
  • the sequence shows a deletion, preferably a deletion of amino acids VDD, as shown on FIG. 1A or 1B .
  • the sequence encoding dog TERT can further be deprived of a nucleolar localization signal.
  • This nucleolar localization signal is correlated with the enzymatic activity of TERT. This signal corresponds to the N-terminal 47 amino acids at the N-terminus of the TERT sequence.
  • sequence encoding dog TERT is deleted of N-terminal 47 amino acids with respect to the full-length dog TERT sequence.
  • Tee TERT sequence deleted of amino acids VDD and of the N-terminal 47 amino acids is shown as SEQ ID NO: 6.
  • the nucleic acid may encode dog TERT sequence or a fragment thereof only, which preferably corresponds to at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or at least 95% of the dog TERT sequence deleted of the N-terminal 47 amino acids.
  • nucleic acid encodes dog TERT amino acid sequence comprising, or consisting of, SEQ ID NO: 5 or SEQ ID NO: 6.
  • the nucleic acid may further encode a non-dog TERT antigenic fragment.
  • This embodiment is preferred, to favor breakage of tolerance towards a self-antigen, and induce an efficient immune response along, with an immune memory response in the dog.
  • the presence of non-dog TERT fragment(s) advantageously engages certain subtypes of CD4 + T cells, providing help for antitumor immunity, and reversing potential regulation by secreting certain cytokines called Th1 cytokines.
  • the dog and non-dog TERT sequences or fragments thereof are preferably fused, to be expressed as a hybrid or chimeric protein.
  • the dog and non-dog TERT sequences or fragments thereof may be separated, but carried on the same vector, e.g. the same plasmid.
  • non-dog TERT antigenic fragment corresponds to a fragment absent or eliminated from the dog TERT sequence, to the extent it does not complement the loss of catalytic activity or the loss of the nucleolar localization signal.
  • the dog TERT sequence, or fragment thereof can represent at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or at least 95% of all TERT sequences in the nucleic acid, plasmid, or other vector.
  • the dog TERT sequence or fragment represents at least 90% of the hybrid or chimeric TERT protein.
  • the dog TERT sequence or fragment represents at least 60% of the hybrid or chimeric TERT protein.
  • the non-dog TERT antigenic fragment preferably originates from a cat TERT sequence.
  • the non-dog TERT antigenic fragment is advantageously processed by dendritic cells, thereby generating CD4 T cell help.
  • the invention employs a nucleic acid that encodes a protein sequence selected from the group consisting of SEQ ID NO: 2, 4, 5 or 6.
  • Such nucleic acid may comprise a sequence selected from the group consisting of SEQ ID NO:1 or 3, or nucleotides 241-3459 of SEQ ID NO: 1, or nucleotides 241-1413 or 241-1407 or nucleotides 3352-3456 or 3298-3456 of SEQ ID NO: 3.
  • the nucleic acid may further encode a protein which enhances the addressing of the TERT protein to the proteasome and increases class I presentation of the derived peptides.
  • Said protein may be preferably ubiquitin, or it may be any chaperon protein, e.g. calreticulin.
  • the nucleic acid is a genetic contrast comprising a polynucleotide sequence as defined herein, and regulatory sequences (such as a suitable promoter(s), enhancer(s), terminator(s), etc.) allowing the expression (e.g. transcription and translation) of the protein product in the host cell or host organism.
  • regulatory sequences such as a suitable promoter(s), enhancer(s), terminator(s), etc.
  • the genetic constructs of the invention may be DNA or RNA, and are preferably double-stranded DNA.
  • the genetic constructs of the invention may also be in a form suitable for transformation of the intended host cell or host organism, in a form suitable for integration into the genomic DNA of the intended host cell or in a form suitable for independent replication, maintenance and/or inheritance in the intended host organism.
  • the genetic constructs of the invention may be in the form of a vector, such as for example a plasmid, cosmid, YAC, a viral vector or transposon.
  • the vector may be an expression vector, i.e. a vector that can provide for expression in vitro and/or in vivo (e.g. in a suitable host cell, host organism and/or expression system).
  • a genetic construct of the invention comprises i) at least one nucleic acid of the invention; operably connected to ii) one or more regulatory elements, such as a promoter and optionally a suitable terminator; and optionally also iii) one or more further elements of genetic constructs such as 3′- or 5′-UTR sequences, leader sequences, selection markers, expression markers/reporter genes, and/or elements that may facilitate or increase (the efficiency of) transformation or integration.
  • regulatory elements such as a promoter and optionally a suitable terminator
  • further elements of genetic constructs such as 3′- or 5′-UTR sequences, leader sequences, selection markers, expression markers/reporter genes, and/or elements that may facilitate or increase (the efficiency of) transformation or integration.
  • the genetic construct can be prepared by digesting the nucleic acid polymer with a restriction endonuclease and cloning into a plasmid containing a promoter such as the SV40 promoter, the cytomegalovirus (CMV) promoter or the Rous sarcoma virus (RSV) promoter.
  • a promoter such as the SV40 promoter, the cytomegalovirus (CMV) promoter or the Rous sarcoma virus (RSV) promoter.
  • CMV cytomegalovirus
  • RSV Rous sarcoma virus
  • the TERT nucleic acid sequences are inserted into a pcDNA3.1 expression plasmid (see FIG. 2A ).
  • vectors include retroviral vectors, lentivirus vectors, adenovirus vectors, vaccinia virus vectors, pox virus vectors, adenovirus-associated vectors and measle virus vectors.
  • compositions can be prepared, comprising said nucleic acid or vector.
  • the compositions are immunogenic. They can comprise a carrier or excipients that are suitable for administration in dogs (i.e. non-toxic, and, if necessary, sterile).
  • excipients include liquid, semisolid, or solid diluents that serve as pharmaceutical vehicles, isotonic agents, stabilizers, or any adjuvant.
  • Diluents can include water, saline, dextrose, ethanol, glycerol, and the like.
  • Isotonic agents can include sodium chloride, dextrose, mannitol, sorbitol, and lactose, among others.
  • Stabilizers include albumin, among others.
  • any adjuvant known in the art may be used in the vaccine composition, including oil-based adjuvants such as Freund's Complete Adjuvant and Freund's Incomplete Adjuvant, mycolate-based adjuvants, bacterial lipopolysaccharide (LPS), peptidoglycans, proteoglycans, aluminum hydroxide, saponin, DEAE-dextran, neutral oils (such as miglyol), vegetable oils (such as arachis oil), Pluronic® polyols.
  • oil-based adjuvants such as Freund's Complete Adjuvant and Freund's Incomplete Adjuvant
  • mycolate-based adjuvants mycolate-based adjuvants
  • bacterial lipopolysaccharide (LPS) bacterial lipopolysaccharide
  • peptidoglycans peptidoglycans
  • proteoglycans peptidoglycans
  • aluminum hydroxide such as miglyol
  • saponin such as mig
  • the nucleic acid or composition can be administered directly or they can be packaged in liposomes or coated onto colloidal gold particles prior to administration.
  • Techniques for packaging DNA vaccines into liposomes are known in the art, for example from Murray, 1991.
  • techniques for coating naked DNA onto gold particles are taught in Yang, 1992, and techniques for expression of proteins using viral vectors are found in Adolph, 1996.
  • the vaccine compositions are preferably administered intradermally, subcutaneously or intramuscularly by injection or by gas driven particle bombardment, and are delivered in an amount effective to stimulate an immune response in the host organism.
  • administration comprises an electroporation step, also designated herein by the term “electrotransfer”, in addition to the injection step (as described in Mir 2008, Sardesai and Weiner 2011).
  • compositions may also be administered ex vivo to blood or bone marrow-derived cells using liposomal transfection, particle bombardment or viral transduction (including co-cultivation techniques).
  • the treated cells are then reintroduced back into the subject to be immunized.
  • a series of dosages of increasing size is administered to the corresponding species and the resulting immune response is observed, for example by detecting the cellular immune response by an IFN ⁇ Elispot assay (as described in the experimental section), by detecting CTL response using a chromium release assay or detecting CD4 T cell (helper T cell) response using a cytokine release assay.
  • the vaccination regimen comprises one to three injections, preferably repeated three or four weeks later.
  • the vaccination schedule can be composed of one or two injections followed three or four weeks later by at least one cycle of three to five injections.
  • a primer dose is composed of one to three injections, followed by at least a booster dose every year, or every two or years for instance.
  • the nucleic acid or immunogenic composition as described above is useful in a method for preventing or treating a tumor in a dog.
  • a method for preventing or treating a tumor in a dog comprises administering an effective amount of said nucleic acid or immunogenic composition in a dog in need thereof. Said nucleic acid or immunogenic composition is administered in an amount sufficient to induce an immune response in the dog.
  • the tumor may be any undesired proliferation of cells, in particular a benign tumor or a malignant tumor, especially a cancer.
  • the cancer may be at any stage of development, including the metastatic stage. However preferably the cancer has not progressed to metastases.
  • the tumor may be selected from the group consisting of bladder cancer, brain tumor, liver tumor, mammary tumors and carcinoma, mast cell tumors, malignant histiocytosis and histocytic sarcomas, squamous cell carcinomas, hemangiosarcoma, lymphoma, in particular B-cell lymphoma, melanoma, bone tumors (osteosarcoma), testicular tumors.
  • the vaccination according to the invention may be combined with conventional therapy, including chemotherapy, radiotherapy or surgery.
  • Combinations with adjuvant immunomodulating molecules such GM-CSF or IL-2 could also be useful.
  • the inventors have constructed DNA vaccines encoding an inactivated form of dog TERT and a cat/dog hybrid TERT (Example 1), and have assessed their functionality, safety and immunogenicity.
  • mice mice were found to be immunogenic and to elicit specific efficient CD8 T cells and CD4 T cells in mice (Example 3).
  • the TERT sequence is preceded by a DNA sequence encoding the human-ubiquitin.
  • the presence of the ubiquitin will increase the addressing of the TERT protein to the proteasome and increase the class I presentation pathway of TERT derived peptides.
  • TERT sequence is followed by the sequence of the influenza protein V5 to facilitate future purification or detection of the fusion protein by Western Blot or histochemistry for example.
  • the DNA sequence coding for the TERT protein has been deleted of 47 amino-acids in the N-Ter region, which encodes the nucleolar importation signal. Moreover, three amino-acids have been removed in the catalytic site of TERT (VDD), to inhibit the protein enzymatic activity.
  • pDUV5 encodes the full-length of dog TERT nucleotide sequence, depleted of the N-term 47 amino acids ( FIG. 1A ), pCDT encodes 54.4% of the cat TERT sequence and 35.9% of the dog TERT sequence ( FIG. 1B ).
  • TERT DNA sequences were synthetized from Genecust (Dudelange, Germany). Then they were cloned into the pcDNA3,1 expression plasmid provided by Life technologies SAS ( Saint-Aubin, France) using the HindIII and XbaI restriction sites (see FIG. 2A ). Plasmids were stored at ⁇ 20° C., in PBS 1 ⁇ , at a concentration of 2 mg/mL prior use. The backbone plasmid was used as empty vector for western blot and Trap-Assay experiments. It consists of the pcDNA3.1 backbone plasmid deprived of the transgene protein DNA sequence (TERT).
  • the human 293T cell line used for transfection assays and immune-fluorescence experiments were kindly provided by Pr Simon Wain-Hobson (Pasteur Institute).
  • the CrFK (Crandall-Reese feline kidney) cells used for the TRAP-assay were kindly provided by Pr J. Richardson ( Erasmus Vcierinaire de Maison Alfort). Cells were grown at 37° C., 5% CO 2- in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% heat-inactivated Fetal Calf Serum (FCS), 1% sodium-pyruvate, 1% penicillin-streptomycin pyruvate and 0.1% ⁇ -mercaptoethanol. All components of the culture medium were purchased from Life technologies SAS (Saint-Aubin, France).
  • Transfection of 293T cells were performed with either pCDT or pDUV5 plasmids using the JetPRIME® transfection kit (Polyplus-transfection SA, Illkirch, France) according to manufacturer's instruction.
  • the JetPRIME® transfection kit Polyplus-transfection SA, Illkirch, France
  • 400 000 HeLa cells or 293T cells per well were seeded in 2 mL of DMEM culture medium, and cultured 24 hours at 37° C., 5% CO 2 prior transfection.
  • 2 ⁇ g of each plasmid diluted in 200 ⁇ L of jetPRIME® buffer, or 200 ⁇ L of jetPRIME® buffer only with respectively 4 ⁇ L of jetPRIME® agent were drop onto the cells.
  • Transfection medium were removed 4 hours later and replaced by 2 mL of DMEM culture medium. Cells were put at 37° C., 5% CO 2 and recovered for analysis 24 hours later.
  • Transfected 293 T cells were lysed on ice with radioimmunoprecipitation assay (RIPA) lysis buffer (RIPA Buffer, Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France) containing protease inhibitors cocktail (Complete EDTA-free, Roche Diagnostic, Indianapolis, USA) for 10-20 minutes. Then, suspension was centrifuged 15 minutes at 14000 rpm at 4° C. in order to remove cellular debris. The supernatants were harvested and the protein concentration was measured using the Bradford method.
  • RIPA radioimmunoprecipitation assay
  • Protein samples were denatured 5 minutes at 95° C., separated on Nu-PAGE® Novex 4-12% Bis-Tris gels (Invitrogen, Carlsbad, USA) and transferred to PVDF membranes (iBlot® transfer stack, Invitrogen, Carlsbad, USA) using the iBlot® device (Invitrogen, Carlsbad, USA). The membrane was cut approximately at 60 kDa.
  • the upper part membrane was probed with an anti-V5 antibody (Invitrogen, Carlsbad, USA) while the other part was probed with an anti- ⁇ -actin antibody (Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France), then samples were revealed by an ECL (Enhanced chemiluminescence) anti-mouse Horse Radish Peroxidase (HRP) linked antibody (GE Healthcare, Vélizy, France)). Immunoblot signals were revealed using 18 ⁇ 24 films and the corresponding cassette both products purchased from GE healthcare (Buckinghamshire, UK).
  • ECL Enhanced chemiluminescence
  • HRP Horse Radish Peroxidase
  • Telomerase activity was measured by the photometric enzyme immunoassay for quantitative determination of telomerase activity, utilizing telomeric repeat amplification protocol (TRAP) (Yang et al, 2002).
  • CrFK Cell Rees Feline Kidney
  • telomerase-negative cells Yazawa et al., 2003
  • CrFK cells were transfected with a plasmid encoding the wild type human TERT (fully active).
  • Telomerase activity was calculated as suggested in the kit's manual and compared with a control template of 0.1 amol telomeric repeats, representing a relative telomerase activity (RTA) of 100. Inactivated samples and lysis buffer served as negative controls.
  • the functionality of the new plasmid constructs is shown by the presence of the plasmid encoded TERT protein in the total protein lysate of pCDT or pDUV5 transfected cells in vitro.
  • the inventors performed western-blot assays on the total protein lysate of 293T cells plasmids transfected with pCDT or pDUV5 (24 h after transfection).
  • anti-V5 antibody coupled with Horse Radish Peroxidase (HRP) was used to reveal the presence of the fusion protein of interest.
  • V5 specific-signal was detected 24 h after transfection in the protein lysate of pCDT or pDUV5 transfected cells.
  • the size of the protein band detected corresponds to the different TERT protein encoded by the plasmids which molecular weight is 123 kDa.
  • no V5 specific signal was detected in untreated or empty plasmid transfected cells.
  • the inventors demonstrated that pDUV5 and pCDT plasmids were correctly processed in vitro after transfection in mammalian cells and that the plasmid product of expression (TERT protein) was well expressed.
  • a TRAPeze assay was performed. As illustrated by FIG. 3 , protein lysates from pDUV5 or pCDT transfected cells do not exhibit any telomerase activity. As a positive control, the protein extracts from CrFk cells transfected with the native human TERT (hTERT) were used. Thus the inventors demonstrated that the TERT proteins encoded by either pCDT or pDUV5 plasmids do not express any functional enzymatic activity after in vitro transfection.
  • the inventors have further investigated the intracellular location of the two plasmid products of expression.
  • an in vitro immunofluorescence assay was performed. Briefly, 24 h after in-vitro transfection of 293T cells with either pCDT or pDUV5, an anti-V5 antibody coupled to an Alexa-Fluor labeled secondary antibody were used to detect the TERT proteins within the cells.
  • the pCDT and pDUV5 encoded TERTs were not detected inside the cell nucleoli contrary to what was observed with 293T cells transfected with the plasmid encoding the native human TERT.
  • the inventors demonstrated that after in vitro transfection with either pDUV5 and pCDT plasmids, first the TERT protein expression is excluded from the nucleoli and secondly, these products of expression do not exhibit any enzymatic activity. These two criteria establish the safety of the plasmids and favour their use for in vivo vaccination.
  • mice Female Balb/cBy and C57BL/6J mice (6-8 week old) were purchased from Janvier laboratories (Saint-Berthevin, France). Animals were housed at the Specific Pathogen Free animal facility of the Pasteur Institute. Mice were anesthetized prior to intradermal (ID) or intramuscular (IM) immunizations, with a mix solution of xylazine 2% (Rompun, Bayer Sante, Loos, France) and Ketamine 8% (Imalgen 1000, Merial, Lyon, France) in Phosphate Buffer Saline 1 ⁇ (PBS 1 ⁇ , Life technologies SAS, Saint-Aubin, France), according to individual animal weight and duration of anesthesia (intraperitoneal route). All animals were handled in strict accordance with good animal practice and complied with local animal experimentation and ethics committee guidelines of the Pasteur Institute of Paris.
  • ID intradermal
  • IM intramuscular
  • TERT peptides used in mouse studies were predicted by in-silico epitope prediction in order to bind mouse class I MHC, H2K b , H2D b or mouse class II H2-IA d using four algorithms available online:
  • H2 restricted peptides sequences determined by in silico prediction algorithms H2D b restricted TERT peptides 621-629 (RPIVNMDYI) 621 SEQ ID NO: 8 580-589 (RQLFNSVHL) 580 SEQ ID NO: 9 987-996 (TVYMNVYKI) 987 SEQ ID NO: 10
  • Intradermal (ID) immunization was performed on the lower part of the flank with Insulin specific needles (U-100, 29 G ⁇ 1/2′′ ⁇ 0.33 ⁇ 12 mm, Terumo, Belgium) after shaving. No erythema was observed after shaving, during and after immunization procedure.
  • Intramuscular immunization (IM) was performed in the anterior tibialis cranialis muscle, also using Insulin specific needles U-100. Each animal received a priming dose of either pCDT or pDUV5, independently of vaccine route, corresponding to 100 ⁇ g of DNA. All animals were boosted at day 14 post-prime using the same amount of plasmid and the same route of immunization.
  • HV-LV invasive needle electrodes
  • the muscle injection site was covered with ultrasonic gel (Labo FH, blue contact gel, NM Médical, France) and surrounded by tweezers electrodes (0.5 cm apart, tweezertrode 7 mm, BTXI45-0488, USA) and voltage was applied using the same parameters than for skin electroporation.
  • the Agilepulse® in vivo system electroporator was used for all experiments (BTX, USA).
  • PVDF microplates IFN- ⁇ Elispot kit, Diaclone, Abcyss, France, 10 ⁇ 96 tests, ref 862.031.010P
  • capture antibody anti-mouse IFN- ⁇
  • PBS 2% milk PBS 2% milk.
  • Spleens from pDNA-immunized mice were mashed and cell suspensions were filtered through a 70-mm nylon mesh (Cell Strainer, BD Biosciences, France).
  • Ficoll-purified splenocytes (Lymphocyte Separation Medium, Eurobio, France) were numerated using the Cellometer® Auto T4 Plus counter (Ozyme, France) and added to the plates in triplicates at 2 ⁇ 10 5 or 4 ⁇ 10 5 cells/well and stimulated with 5 ⁇ g/ml of dTERT or hyTERT relevant peptides or Concanavalin A (10 ⁇ g/ml), or mock stimulated with serum free culture medium. After 19 hours, spots were revealed with the biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution. Spots were counted using the Immunospot ELIspot counter and software (CTL, Germany).
  • splenocytes from na ⁇ ve C57/B16 mice were labeled in PBS 1 ⁇ containing high (5 ⁇ M), medium (1 ⁇ M) or low (0.2 ⁇ M) concentrations of CFSE (Vybrant CFDA-SE cell-tracer kit; Life technologies SAS, Saint-Aubin, France).
  • Splenocytes labeled with 5 and 1 ⁇ M CFSE were pulsed with 2 different H2 peptides at 5 ⁇ g/ml for 1 hour and 30 minutes at room temperature.
  • Peptides 987 and 621 were used for pulsing respectively CFSE high and medium labeled na ⁇ ve splenocytes.
  • CFSE low labeled splenocytes were left unpulsed.
  • the disappearance of peptide-pulsed cells was determined by comparing the ratio of pulsed (high/medium CFSE fuorescence intensity) to unpulsed (low CFSE fuorescence intensity) populations in pDNA immunized mice versus control (PBS 1 ⁇ injected) mice.
  • the percentage of specific killing per test animal was established according to the following calculation: [1 ⁇ [mean (CFSE low PBS/CFSE highi/medium PBS)/(CFSE low pDNA/CFSE high/medim pDNA)]] ⁇ 100.
  • Prism-5 software was used for data handling, analysis and graphic representations. Data are represented as the mean ⁇ standard deviation.
  • EliSPOT assays we used a Mann Whitney non parametric test, and a Kruskal-Wallis analysis with Dunn's multiple comparison test for in-vivo cytotoxicity assay. Significance was set at p-value ⁇ 0.05.
  • mice The inventors have assessed whether pDUV5 plasmid DNA plasmid was capable of eliciting efficient cellular immune responses (CD8) in mice.
  • CD8 efficient cellular immune responses
  • different groups of 9-10 C57-Bl/6 mice were injected ID or IM with pDUV5 immediately followed by electroporation. Two weeks later, mice received a boost injection with the same protocol.
  • mice spleens were harvested and the induced immune response was monitored via an IFN- ⁇ ELISPOT assay using H2 restricted peptides described in Table 1. Dog TERT peptides restricted to mouse MHC class I were predicted in silico as described in the material and methods section. As shown in FIG.
  • pDUV5 construct is able to promote the expansion of dTERT specific CD8 T-cells in mice.
  • the inventors next wanted to show that those specific T-cells exhibit a functional cytotoxic activity in vivo, which will be necessary to attack tumor cells.
  • the inventors performed in vivo cytotoxicity tests using carboxyfuorescein-diacetate suc-cinimidyl ester (CFSE)-labelled, peptide-pulsed splenocytes as target cells.
  • CFSE carboxyfuorescein-diacetate suc-cinimidyl ester
  • Target cells were splenocytes from na ⁇ ve congenic mice separately labelled with three different concentrations of CFSE and pulsed with individual peptides (p621 or p98′7) or left un-pulsed as an internal control. After 15-18 hours, spleen cells were obtained and the disappearance of peptide-pulsed cells in control versus immunized mice was quantifed by fluorescence-activated cell sorting.
  • mice develop CTLs against the 2 epitopes predicted in silico FIGS. 5A and 5B ).
  • Peptide 621 gives the strongest in vivo lysis.
  • Results were concordant with IFN- ⁇ Elispot assays ( FIG. 4 ). No significant difference was observed between the two routes of immunization.
  • mice were immunized with pCDT by ID or IM injection of the plasmid immediately followed by electroporation. Two weeks later, mice received a boost injection with the same protocol. On day 10 post-boost, mice spleens were harvested and the induced immune response was monitored via an IFN- ⁇ ELISPOT assay using H2 restricted peptides described in Table 1.
  • Hy-TERT peptides restricted to mouse MHC class I were predicted in silico as described in the material and methods section. As shown in FIG. 6A , a significant augmentation in the frequency of hyTERT specific IFN- ⁇ secreting CD8 T-cells was observed in the spleen of ID and IM vaccinated animals in comparison with control mice. This was observed for 2 out of 3 class I restricted peptides (p621 and p987, p ⁇ 0.05). No significant difference in the frequency of specific CD8 T cells was observed between IM and ID route for both peptides p921 and p987. The inventors have further investigated the hyTERT restricted CD4 T cell response.
  • Balb/C mice were immunized with pCDT by ID or IM injection immediately followed by electroporation and the CD4 specific T cell response was monitored in the spleen as described before using hyTERT IA d restricted peptides (in silico prediction). Balb/C mice were chosen because this mouse strain is known to develop good CD4 T cell responses. As shown in FIG. 6B , when performing the IFN- ⁇ ELISPOT assay, a significant augmentation in the frequency of hyTERT specific IFN- ⁇ secreting CD4 T-cells was observed in the spleen of ID and IM vaccinated Balb/C mice in comparison with control mice injected with PBS 1 ⁇ .
  • pCDT construct is able to promote the expansion of hyTERT specific CD8 and CD4 T-cells in mice.
  • the inventors next wanted to show that hyTERT specific CD8 T-cells exhibit a functional cytotoxic activity in vivo, which will be necessary to destroy tumor cells.
  • the inventors performed an in vivo cytotoxicity test using carboxyfluorescein-diacetate succinimidyl ester (CFSE)-labelled, peptide-pulsed splenocytes as target cells.
  • CFSE carboxyfluorescein-diacetate succinimidyl ester
  • Target cells were splenocytes from na ⁇ ve congenic mice separately labelled with three different concentrations of CFSE and pulsed with individual peptides (p621 or p98′7) or left un-pulsed as an internal control. After 15-18 hours, spleen cells were obtained and the disappearance of peptide-pulsed cells in control versus immunized mice was quantified by fluorescence-activated cell sorting.
  • mice develop CTLs against the 2 peptides p621 and p987 which were predicted in silico.
  • Lyophilized dTERT peptides (purity>90%) were purchased from JPT Peptide Technologies (Berlin, Germany). Each peptide was resuspended in distilled H 2 O, 5% DMSO at 2 mg/mL prior use according to supplier recommendation and kept frozen at ⁇ 20° C. before use.
  • One third of the dog TERT peptide (AA 281 to 571) was used to synthetized 70 peptides of 15 AA overlapping of 11 AA and recovering this sequence of the dog TERT as depicted in FIG. 3 .
  • Four pools of peptides were used for in vitro experiments and ELIspot assays in dogs.
  • Canine blood samples were purchased from the Bourgelat Institute (Marcy l'Etoile, France). It was taken from a healthy 4-year-old beagle dog housed, fed and cared for in accordance with institutional and ethical guidelines. Heparinized blood samples was 4 time diluted in PBS 1 ⁇ (Life technologies SAS, Saint-Aubin, France). Diluted samples were then layered on Lymphocyte Separation Medium (Eurobio, Courtaboeuf, France) and centrifuged 30 minutes at 2200 rpm (at room temperature) without break.
  • PBS 1 ⁇ Life technologies SAS, Saint-Aubin, France
  • Canine PBMCs were harvested and stored in Fetal Calf Serum (FCS, PAA Laboratories GmbH, Pashing, Austria) with 10% DMSO (Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France) in liquid nitrogen prior use.
  • FCS Fetal Calf Serum
  • DMSO Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France
  • dog frozen PBMCs were recovered, counted using the Cellometer® Auto T4 Plus counter (Ozyme, France) and plated in duplicates or triplicates at 10 6 cells/mL in 48-well flat-bottomed plates (BD, France) in AIM-V medium (Invitrogen) supplemented with either 100 ng/mL caGM-CSF and 5 ng/mL calL-4 (R&DSystems) or 50 ng/mL human FlT3L (Immunotools). Cells were cultured at 37° C., 5% CO 2 in an incubator.
  • maturation stimuli comprising the following reagent: 50 ng/mL rcTNF ⁇ , 20 ng/mL rcIL1- ⁇ (R&DSystems), 1 ng/mL hIL-7 (Miltenyi). Pools of peptides were also added. The final concentration used for each peptide was 10 ⁇ g/mL. Control wells received the cocktails of maturation cytokines only and no peptide. At day 3, culture medium was discarded and fresh AIM-V was added. Fresh AIM-V was added every 3 days until the day of testing. At either day 11 or day 18 after the beginning of culture, cells were recovered, washed in fresh AIM-V medium and used for the ELIspot assay.
  • the inventors wanted to demonstrate the existence of a pre-existing dogTERT specific T-cell repertoire in the target species, i.e dogs.
  • the inventors have investigated whether dTERT-specific T-cell responses could be enhanced in PBMCs incubated with either rcGM-CSF and rcIL-4 for 24 hours, or with hFlt3 ligand followed by maturation stimuli (rcIL1 ⁇ , rcTNF ⁇ and IL-7) and peptides stimulation for another 24 hours and 11 or 18 days of in vitro cell expansion.
  • This technique was described by Mallone and colleagues for human PBMCs and is called in vitro immunization (Martinuzzi et al. 2011). The principle of this experiment is exposed in FIG. 8 .
  • 15mer overlapping peptides recovering one third of the dog TERT protein FIG. 3
  • na ⁇ ve beagle dogs received a local anaesthetic of 2.5 mg/kg IV imalgene and 20-80 ⁇ g/kg IV dorbene 15-20 minutes before vaccination and 100-400 ⁇ g/kg IM post vaccination.
  • the dogs were injected intradermally with 400 ⁇ g of pDUV5 DNA followed by electroporation.
  • pDU5 DNA was electroporated at days 0, 29, 57 and 142.
  • Peripheral blood was drawn and mononuclear cells tested for dog telomerase specific peptides belonging either to pool 6 or pool 19 according to the method of Martinuzzi et al., 2011.
  • FIGS. 10A and 10B show that IFN ⁇ specific T cell responses were detected.
  • pDUV5 DNA vaccination at days 57 and 142 show classical long term memory responses, that is rising sharply and decaying more slowly.
  • Peripheral blood was drawn and the in vitro stimulation protocol as described in Example 4 and in Martinuzzi zt al., 2011, was performed, using pool 4 peptides.

Abstract

The present invention provides an immunogenic composition comprising a nucleic acid that comprises a sequence encoding a dog telomerase deprived of telomerase catalytic activity, or a fragment thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a Continuation of U.S. application Ser. No. 14/780,652, filed on Sep. 28, 2015, which is a U.S. National Phase application under 35 U.S.C. § 371 of International Patent Application No. PCT/EP2014/056381, filed on Mar. 28, 2014, which claims priority to European Patent Application No. EP 13305405.6, filed on Mar. 28, 2013, all of which applications are incorporated herein by reference in their entireties.
  • The present invention relates to cancer vaccination in dogs.
  • SEQUENCE LISTING
  • The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Apr. 10, 2018, is named 246393_000014_SL.txt and is 100,736 bytes in size.
  • BACKGROUND OF THE INVENTION
  • Like their human counterparts, dogs that live in developed countries have seen their life expectancy consistently prolonged. Therefore, the global burden of cancers continues to increase largely because of the aging and growing dog population.
  • The incidence rate of cancers in the canine population is estimated to be between 282.2 to 958 per 100,000 dogs (Merlo et al. 2008, Vascellari et al. 2009). The most frequent tumors in dogs are mammary tumors in females (70.5% of all cancers), non-Hodgkin's lymphomas (8.4% in females and 20.1% in males) and skin tumors (4% in females and 19.9% in males). Moreover, according to the European Society of Veterinary Oncology 50% of dogs over ten years are going to die from a cancer-related problem.
  • The panel of treatments available against veterinary cancer is substantially reduced compared with those available in human oncology.
  • Surgery remains the best way to treat animal tumors. This method presents the advantage of being accessible for many veterinarians, and, in many cases, it can be curative. However, to be curative, surgery must be bold. However in some cases the tumor is too large, too dispersed or just not accessible enough to be entirely removed. If not totally curative, surgery can still be a palliative solution to improve the animal's comfort and prolong its life expectancy.
  • Radiotherapy is another important means to treat certain types of cancers in the veterinary field. It is of particular interest for tumors which are hardly accessible for surgery like cerebral tumors (de Fornel et al. 2007). Furthermore, recent studies in humans have demonstrated that ionizing radiation (IR) could act as an immunomodulator by inducing substantial changes in the tumor microenvironment, including triggering an inflammatory process. Furthermore, the cost and the availability of the material make access to radiation therapy complicated for companion animals. Chemotherapy is more and more used in animal oncology (Marconato 2011). Taking advantages of medical advances in human cancer therapy, there are more and more molecules available like vincristine, cyclophosphamide, carboplatin or cisplatin, to treat companion animals. In the veterinary field, anticancer drugs are particularly used in the treatment of tumors derived from hematopoietic tissue (lymphomas, leukemias). For example the CHOP protocol, combining cyclophosphamide, doxorubicin, vincristine and prednisone is currently used in the treatment of numerous lymphomas (Chun 2009). Chemotherapeutic agents can be particularly efficient in prolonging the life span of a cancerous animal from a few weeks to several months (the median survival time of dogs treated with the CHOP protocol is 13 months). Interestingly, the side effects dreaded by human patients, such as vomiting, diarrhea, hair loss, are usually less frequent in companion animals. Unfortunately, most of the time chemotherapy is not curative in pets and the tumor often escapes treatment.
  • Therefore, just as in human medicine, targeted therapies are in development in veterinary medicine. Thus, some drugs are already available in the clinics like “Masitinib”, an inhibitor of the tyrosine kinase c-kit (Gentilini 2010). Other treatments, including immunotherapies, are under investigation (Manley et al. 2011). These immunotherapeutic treatments are all based on the fact that it is possible to activate the immune system of the host against cancer cells.
  • The relationship between the host immune system and cancer is dynamic and complex. Each type of tumor cell harbors a multitude of somatic mutations and epigenetically deregulated genes, the products of which are potentially recognizable as foreign antigens by immune cells (MUC-1, β-catenin, telomerase . . . ) (Fridman et al. 2012). Growing tumors contain infiltrating lymphocytes called TILs (Tumor Infiltrating Lymphocytes). These killer cells are often ineffective at tumor elimination in vivo but can exert specific functions in vitro, that is to say outside the immunosuppressive tumor microenvironment (Restifo et al. 2012). This is because the tumor stroma contains many suppressive elements including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDCs); soluble factors such as interleukin 6 (IL-6), IL-10, vascular endothelial growth factor (VEGF), and transforming growth factor beta (TGFβ) that down modulate antitumor immunity (Finn 2008, Hanahan and Weinberg 2011). Consequently, the choice of a pertinent tumor associated antigen (TAA) and the bypass of cancer associated immunosuppression are two critical points for a therapeutic vaccine to succeed (Disis et al. 2009).
  • Recent introduction of active cancer immunotherapy (also referred to cancer vaccines) in the clinical cancer practice emphasizes the role of immune responses in cancer prognosis and has led to a growing interest to extend this approach to several human and companion animal cancers (Dillman 2011, Topalian et al. 2011) (Jourdier et al. 2003).
  • In this context, there is still a need for an innovative cancer vaccine strategy for dogs, which would overcome the challenge of breaking tolerance and inducing an immune response in the animal.
  • SUMMARY OF THE INVENTION
  • The inventors now propose a cancer vaccine strategy for dogs, based on the telomerase reverse transcriptase (TERT).
  • A subject of the invention is thus an immunogenic composition comprising a nucleic acid that comprises a sequence encoding (i) a dog TERT deprived of telomerase catalytic activity, or (ii) a fragment thereof. The nucleic acid is preferably DNA, preferably in form of a plasmid.
  • In a preferred embodiment, the nucleic acid that comprises a sequence encoding a dog telomerase reverse transcriptase (TERT) deprived of telomerase catalytic activity, wherein the sequence encoding dog TERT is further deprived of a nucleolar localization signal.
  • In a particular embodiment, the nucleic acid further comprises a non-dog TERT antigenic fragment.
  • A further subject of the invention is a nucleic acid that comprises a sequence encoding (i) a dog TERT deprived of telomerase catalytic activity, or (ii) a fragment thereof, and optionally further comprises a non-dog TERT antigenic fragment.
  • The immunogenic composition or the nucleic acid is useful in triggering an immune response in a dog, against cells that over-express telomerase, such as dysplasia cells or tumor cells.
  • The immunogenic composition or the nucleic acid is thus particularly useful in treating a tumor in a dog, preferably by intradermal or intramuscular route.
  • Such treatment can be referred to as an active immunotherapy or a therapeutic vaccination, as it triggers an immune response against the tumor, especially a cytotoxic CD8 T cell response, along with a specific CD4 T cell response.
  • The invention makes it possible to induce dTERT specific responses in dogs with neoplasias and so can be used for immunotherapeutic treatments of the neoplasias in a clinical setting. The invention is also useful to induce dTERT specific responses in healthy dogs that could be at risk for cancer, e.g. by genetic predisposition, or in healthy dogs from a certain age (e.g. more than 10 years, preferably more than 12 years old), so as to prevent the onset of cancer.
  • Generally speaking, the treatment of the invention may induce long term immune memory responses in healthy dogs, dogs at risk of developing a cancer and those presenting a cancer.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1A shows pDUV5 nucleotide sequence (SEQ ID NO: 1) and corresponding amino acid sequence comprising dog TERT (dTERT) amino acid sequence (SEQ ID NO: 2).
  • The plasmid pDUV5 encodes a near full length dog TERT nucleotide sequence. The nucleotide sequence encoding 3 key amino acids in the catalytic site of the protein have been deleted (VDD). Moreover, the sequence controlling the importation into the nucleoli (Nucleolar addressing signal) has been deleted (nucleotide sequence encoding 47 first Amino Acids in the N-term sequence of dTERT protein). Moreover the DNA sequence encoding the human ubiquitin has been added upstream the dTERT sequence. Presence of the ubiquitin protein enhances the addressing of the dTERT protein to the proteasome and increases class I presentation of derived peptides. However, as the human and dog ubiquitin sequences are identical at the protein level, there is no biological incompatibility. Downstream the dTERT sequence, the sequence of the V5 peptide of the flu was inserted to facilitate the detection of the protein.
  • Nucleotides 1-6 HindIII restriction site for subcloning
  • Nucleotides 13-240 ubiquitin
  • Nucleotides 241-3459 dog TERT
  • Nucleotides 2670-2671 inactivating deletion of 9 bp encoding VDD residues
  • Nucleotides 3460-3513 influenza A A2 epitope
  • Nucleotides 3514-3555 SV5 V5 tag
  • Nucleotides 3556-3561 two stop codons
  • Nucleotides 3562-3567 Xba1 restriction site for subcloning
  • FIG. 1B shows pCDT nucleotide sequence (SEQ ID NO: 3) and corresponding amino acid sequence containing cat/dog hybrid TERT amino acid sequence (SEQ ID NO:4).
  • The plasmid pCDT encodes the cat/dog hybrid TERT (hyTERT) comprising 54.4% from the cat TERT and 35.9% from the dog TERT sequence. The nucleotide sequence encoding 3 key amino acids in the catalytic site of the protein have been deleted (VDD). Moreover, the sequence controlling the importation into the nucleoli (Nucleolar addressing signal) has been depleted (nucleotide sequence encoding 47 first Amino Acids in the Nter sequence of hyTERT protein). The DNA sequence encoding the human ubiquitin has been added upstream the hyTERT sequence. The presence of the ubiquitin protein enhances the addressing of the hyTERT protein to the proteasome and increases class I presentation of the derived peptides. Downstream the hyTERT sequence, the sequence of the V5 peptide of the flu was inserted to facilitate the detection of the protein.
  • Nucleotides 1-6 HindIII restriction site for subcloning
  • Nucleotides 13-240 ubiquitin
  • Nucleotides 241-1413 dog TERT (35.9% of TERT sequences)
  • Nucleotides 1414-3351 cat TERT (54.4% of TERT sequences)
  • Nucleotides 3352-3456 dog TERT last exon
  • Nucleotides 3457-3510 influenza A2 epitope
  • Nucleotides 3511-3552 SV5 V5 tag
  • Nucleotides 2667-2668 inactivating deletion of 9 bp encoding VDD residues
  • Nucleotides 3553-3558 two stop codons
  • Nucleotides 3559-3564 Xba1 restriction site for subcloning
  • FIG. 2A shows a simplified map of pcDNA3.1 expression plasmid into which the dog or hybrid TERT nucleic acid sequences are cloned.
  • FIG. 2B shows dog TERT protein sequence (SEQ ID NO: 5). The region covered by the dTERT 15mer peptide pool overlapping by 11 residues (70 peptides in total) that is used for in vitro immunization studies and ELIspot assays in dog PBMCs is shown in grey.
  • FIG. 3 shows that pDNA constructs are safe (Trapeze). Lysates obtained from CrFK cells transfected with hTERT, pCDT, or pDUV5 plasmids were analyzed for telomerase activity by the TRAP assay. The level of telomerase activity is shown as relative telomerase activity compared with that of control template measured in each kit and with the activity of a wild type human telomerase (hTERT). All samples at 2.1 μg protein concentration were measured in triplicate, error bars are standard error of the mean (SEM), (**P=0.0032, hTERT vs pDUV5 unpaired t test).
  • FIG. 4 is a graph showing that mice immunized with pDUV5 mount specific interferon-γ-secreting CD8 T-cell responses against H2 restricted dog TERT peptides.
  • 7 week-old C57/B16 female mice were immunized intradermally (ID) or intramuscularly (IM) (10 mice per group) with 100 μg pDUV5 plasmid at day 0 and boost 14 days later. At the same time, control mice received PBS via ID or IM route (6 mice per group). Ten days after boost, spleens of all mice were harvested. Splenocytes were Ficoll purified and stimulated in triplicates with 5 μg/mL of relevant class I peptides (p580, p621 or p98′7) for 19 hours. Spots were revealed with a biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution. Results are the mean±standard deviation. Mann Whitney non parametric test, *p-value<0.05, **: p-value<0.01.
  • FIGS. 5A and 5B show that ID or IM immunization of mice with pDUV5 plasmid results in a dog TERT specific cytotoxic T-lymphocyte (CTL) response measurable in vivo by elimination of transferred target cells which were pulsed with dog TERT peptides restricted to H2.
  • Seven week-old C57/B16 female mice were immunized intradermally (ID) or intramuscularly (IM) with 100 μg pDUV5 plasmid at day 0 and day 14 post-priming. At day 9 post-boost injection, syngeneic splenocytes, pulsed with individual dTERT peptides restricted to H2 (either p987 or p621) or left unpulsed were labeled with carboxyfuorescein-diacetate succinimidyl ester (CFSE) at three different concentrations: high=1 μM (p621), medium=0.5 μM (p98′7) and low=0.1 μM (unpulsed). The same number of high, medium or low CFSE labeled cells was transferred IV to vaccinated mice. After 15-18 hours, the disappearance of peptide-pulsed cells was determined by fluorescence-activated cell-sorting analysis in the spleen. The percentage of specific lysis was calculated by comparing the ratio of pulsed to un-pulsed cells in vaccinated versus control mice.
  • (A) Example of the in vivo CTL assay showing the elimination of target cells pulsed with p987 (medium, M)/or p621 peptide (High, H) in the spleen of mice injected via the ID route (left panel). No such disappearing is observed in control mice injected ID with PBS 1× (right panel). H=high, M=Medium, L=Low
  • (B) Percentage of specific lysis for each mouse against each individual peptide in the spleen after IM or ID vaccination with pDUV5. Horizontal bars show average percentage of lysis per peptide and per immunization route. Standard deviations are also plotted. Representative data from 2 independent experiments (n=10 individual animals/group). Kruskal-Wallis analysis with Dunn's multiple comparison test, ns: not significant. Statistical significance is set at p-value<0.05.
  • FIGS. 6A and 6B show IFNγ+specific CD8 and CD4 T-cell responses against H2 restricted hyTERT peptides in mice immunized with pCDT.
  • Seven week-old female mice were immunized intradermally (ID) or intramuscularly (IM) with either 100 μg pCDT plasmid or PBS at day 0 and boost 14 days later. Ten day post-boost, spleens were harvested. Splenocytes were Ficoll-purified and stimulated in triplicates with 5 μg/mL of relevant peptides for 19 hours. Spots were revealed with a biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution.
  • (A) Plasmid vaccinated groups were composed of five C57/B16 mice, and control groups, of three mice. Splenocytes were stimulated with class I peptides p580, p621 and p987. Results show the frequency of peptide specific IFN-γ producing CD8 T cells.
  • (B) Plasmid vaccinated groups were composed of 9 Balb/cBy mice immunized IM and 5 ID. Control groups of 8 Balb/cBy mice injected IM and 4 ID. Splenocytes were stimulated with class II peptides p951, p1105, p1106 and p1109. Results show the frequency of peptide specific IFN-γ producing CD4 T cells.
  • Results are the mean±standard deviation. Mann Whitney non parametric test, *p-value<0.05, **: p-value<0.01.
  • FIGS. 7A and 7B show hyTERT specific cytotoxic T-lymphocyte (CTL) response in mice immunized with pCDT plasmid, measurable in vivo by elimination of transferred target cells which were pulsed with hybrid TERT peptides restricted to H2.
  • 7 week-old C57/B16 female mice were immunized ID or IM with 100 μg pCDT plasmid at day 0 and day 14 post-priming. At day 9 post-boost injection, syngeneic splenocytes, pulsed with individual dTERT peptides restricted to H2 (either p987 or p621) or left unpulsed were labeled with carboxyfluorescein-diacetate succinimidyl ester (CFSE) at three different concentrations: high=1 μM (p98′7), medium=0.5 μM (p621) and low=0.1 μM (unpulsed). The same number of high, medium or low CFSE labeled cells was transferred IV to vaccinated mice. After 15-18 hours, the disappearance of peptide-pulsed cells was determined by fluorescence-activated cell-sorting analysis in the spleen. The percentage of specific lysis was calculated by comparing the ratio of pulsed to un-pulsed cells in vaccinated versus control mice.
  • (A) Example of the in vivo CTL assay showing the elimination of target cells pulsed with p621 peptide (High, H) or p987 peptide (Medium, M) in the spleen of a mouse vaccinated ID (left panel) with pCDT. No such disappearing is observed in control mice injected ID with PBS 1× (right panel).
  • (B) Percentage of specific lysis for each mouse against each individual peptide in the spleen after IM or ID vaccination with pCDT. Horizontal bars show average percentage of lysis per peptide and per immunization route. Standard deviations are also plotted. Representative data from 2 independent experiments (n=10 individual animals/group). Kruskal-Wallis analysis with Dunn's multiple comparison test, *p<0.1, ***p<0.001, ns: not significant. Statistical significance is set at p-value<0.05.
  • FIG. 8 shows principle of in vitro immunization in dog PBMCs
  • Frozen dog PBMCs were incubated with recombinant canine GM-CSF (rcGM-CSF) and canine IL-4 (rcIL-4) or human FlT3 (hFlT3) ligand for 24h. Maturation stimuli (rcTNFα, hIL-7 and rcIL-1β) were then added with dTERT overlapping peptides pools for 3 days. Eleven or 18 days of culture were performed and, TERT specific T cells were then detected via an IFN-γ ELISpot assay.
  • FIGS. 9A and 9B show a repertoire of dTERT specific IFN-γ secreting T cells in PBMCs from a naïve dog
  • Frozen PBMCs incubated during 24 hours with either rcGM-CSF and rcIL-4 or hFlT3 ligand and matured 3 day long with dTERT overlapping peptides pools and maturation cytokines (rcTNFα, hIL-7 and caIL-1β) were harvested after 11 or 18 days of culture to perform an ELISpot IFN-γ. Results show the frequency of peptide specific IFN-γ producing T cells/106 canine PBMC after 11 days (A) or 18 days (B) of culture.
  • FIGS. 10A and 10B show the kinetics of the specific IFNγ T cell response against a first pool of dTERT peptides (pool 6), and a second pool of dTERT peptides (pool 19), respectively.
  • Six naïve beagle healthy dogs were injected intradermally with 400 μg of pDUV5 DNA followed by electroporation, at days 0, 29, 57 and 142. Peripheral blood was drawn and mononuclear cells tested for dog telomerase specific peptides belonging either to pool 6 or pool 19 peptides according to the method of Martinuzzi et al., 2011. IFNγ specific T cell responses were detected by ELISPOT assay, for pool 6 and 10 dTERT peptides, all of which above baseline readings.
  • FIGS. 11A and 11B show the kinetics of the specific IFNγ T cell response against pool 6 dTERT peptides, and pool 10 dTERT peptides, respectively.
  • pDUV5 DNA vaccination at days 57 and 142 show classical long term memory responses, that is rising sharply and decaying more slowly.
  • FIG. 12 shows that tumor bearing dogs and healthy dogs have dTERT specific T lymphocytes (pool 4 peptides). Peripheral blood was drawn and in vitro stimulation protocol was performed as described Martinuzzi zt al, 2011.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • The telomerase consists of an RNA template and protein components including a reverse transcriptase, designated “Telomerase Reverse Transcriptase” (TERT), which is the major determinant of telomerase activity. Unless otherwise specified, in the present specification, the term “telomerase” refers to TERT.
  • In the present invention, the term “dog TERT” refers to the TERT sequence of any domestic dog (also designated Canis familiaris or Canis lupus familiaris). A dog TERT mRNA sequence is available with NCBI accession number NM 001031630 (XM 545191). Dog TERT amino acid sequence is shown as SEQ ID NO:5.
  • The invention can make also use of non-dog telomerase (TERT) sequence, which can be from any human or non-human mammal, e.g. from cat. The term “cat TERT” refers to the TERT sequence of any domestic cat (also designated as Felis catus or Felis silvestris catus). Partial molecular cloning of the cat TERT gene (237 bp of mRNA) has been reported by Yazawa et al. 2003. The inventors herein provide a longer sequence of Felis catus TERT. The corresponding amino acid sequence is shown as SEQ ID NO:7.
  • The “telomerase catalytic activity” refers to the activity of TERT as a telomerase reverse transcriptase. The term “deprived of telomerase catalytic activity” means that the nucleic acid sequence encodes a mutant TERT, which is inactive.
  • The term “hybrid” or “chimeric” amino acid or nucleotide sequence means that part of the sequence originates from one animal species and at least another part of the sequence is xenogeneic, i.e. it originates from at least one other animal species.
  • When referring to a protein, the term“fragment” preferably refers to fragment of at least 10 amino acids, preferably at least 20 amino acids, still preferably at least 30, 40, 50, 60, 70, 80 amino acid fragments.
  • In the context of the invention, the term “antigenic fragment” refers to an amino acid sequence comprising one or several epitopes that induce T cell response in the animal, preferably cytotoxic T lymphocytes (CTLs). An epitope is a specific site which binds to a T-cell receptor or specific antibody, and typically comprises about 3 amino acid residues to about 30 amino acid residues, preferably 8 or 9 amino acids as far as class I MHC epitopes are concerned, and preferably 11 to 25 amino acids as far as class II MHC epitopes are concerned.
  • The term “immunogenic” means that the composition or construct to which it refers is capable of inducing an immune response upon administration (preferably in a dog). “Immune response” in a subject refers to the development of a humoral immune response, a cellular immune response, or a humoral and a cellular immune response to an antigen. A “humoral immune response” refers to one that is mediated by antibodies. A “cellular immune response” is one mediated by T-lymphocytes. It includes the production of cytokines, chemokines and similar molecules produced by activated T-cells. Immune responses can be determined using standard immunoassays and neutralization assays for monitoring specifically the humoral immune response, which are known in the art. In the context of the invention, the immune response preferably encompasses stimulation or proliferation of cytotoxic CD8 T cells and/or CD4 T cells.
  • As used herein, the term “treatment” or “therapy” includes curative treatment. More particularly, curative treatment refers to any of the alleviation, amelioration and/or elimination, reduction and/or stabilization (e.g., failure to progress to more advanced stages) of a symptom, as well as delay in progression of the tumor or dysplasia or of a symptom thereof.
  • As used herein, the term “prevention” or “preventing” refers to the alleviation, amelioration and/or elimination, reduction and/or stabilization (e.g., failure to progress to more advanced stages) of a prodrome, i.e. any alteration or early symptom (or set of symptoms) that might indicate the start of a disease before specific symptoms occur. A cell that “overexpresses telomerase” refers to a cell in a subject, which either expresses telomerase, e.g. upon mutation or infection, whereas it does usually not, under normal conditions, or to a cell in a subject which expresses a higher level of telomerase (e.g. upon mutation or infection), when compared to normal conditions. Preferably the cell that overexpresses telomerase shows an increase of expression of at least 5%, at least 10%, at least 20%, 30%, 40%, 50%, 60%, 70%, 80%, or more.
  • Nucleic Acid Constructs
  • It is herein provided a nucleic acid that comprises a sequence encoding (i) a dog telomerase reverse transcriptase (TERT) deprived of telomerase catalytic activity, or (ii) a fragment thereof. The nucleic acid may be DNA or RNA, but is preferably DNA, still preferably double stranded DNA.
  • As a first safety key, the TERT sequence is deprived of telomerase catalytic activity. In a preferred embodiment, the sequence that encodes dog TERT contains mutations that provide inactivation of the catalytic activity. The term “mutation” includes substitution of one or several amino acids, a deletion of one or several aminoacids, and/or an insertion of one of several amino acids. Preferably the sequence shows a deletion, preferably a deletion of amino acids VDD, as shown on FIG. 1A or 1B.
  • As a second safety key, the sequence encoding dog TERT can further be deprived of a nucleolar localization signal. This nucleolar localization signal is correlated with the enzymatic activity of TERT. This signal corresponds to the N-terminal 47 amino acids at the N-terminus of the TERT sequence.
  • Preferably the sequence encoding dog TERT is deleted of N-terminal 47 amino acids with respect to the full-length dog TERT sequence.
  • Dog TERT sequence deleted of amino acids VDD and of the N-terminal 47 amino acids is shown as SEQ ID NO: 6.
  • In a particular embodiment, the nucleic acid may encode dog TERT sequence or a fragment thereof only, which preferably corresponds to at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or at least 95% of the dog TERT sequence deleted of the N-terminal 47 amino acids.
  • Preferably the nucleic acid encodes dog TERT amino acid sequence comprising, or consisting of, SEQ ID NO: 5 or SEQ ID NO: 6.
  • The nucleic acid may further encode a non-dog TERT antigenic fragment. This embodiment is preferred, to favor breakage of tolerance towards a self-antigen, and induce an efficient immune response along, with an immune memory response in the dog. The presence of non-dog TERT fragment(s) advantageously engages certain subtypes of CD4+ T cells, providing help for antitumor immunity, and reversing potential regulation by secreting certain cytokines called Th1 cytokines.
  • The dog and non-dog TERT sequences or fragments thereof are preferably fused, to be expressed as a hybrid or chimeric protein. Alternatively, the dog and non-dog TERT sequences or fragments thereof may be separated, but carried on the same vector, e.g. the same plasmid.
  • Preferably the non-dog TERT antigenic fragment corresponds to a fragment absent or eliminated from the dog TERT sequence, to the extent it does not complement the loss of catalytic activity or the loss of the nucleolar localization signal.
  • The dog TERT sequence, or fragment thereof, can represent at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or at least 95% of all TERT sequences in the nucleic acid, plasmid, or other vector.
  • In a preferred embodiment, the dog TERT sequence or fragment represents at least 90% of the hybrid or chimeric TERT protein.
  • In another embodiment, the dog TERT sequence or fragment represents at least 60% of the hybrid or chimeric TERT protein.
  • The non-dog TERT antigenic fragment preferably originates from a cat TERT sequence.
  • The non-dog TERT antigenic fragment is advantageously processed by dendritic cells, thereby generating CD4 T cell help.
  • In a preferred embodiment, the invention employs a nucleic acid that encodes a protein sequence selected from the group consisting of SEQ ID NO: 2, 4, 5 or 6.
  • Such nucleic acid may comprise a sequence selected from the group consisting of SEQ ID NO:1 or 3, or nucleotides 241-3459 of SEQ ID NO: 1, or nucleotides 241-1413 or 241-1407 or nucleotides 3352-3456 or 3298-3456 of SEQ ID NO: 3.
  • In a particular embodiment, the nucleic acid may further encode a protein which enhances the addressing of the TERT protein to the proteasome and increases class I presentation of the derived peptides. Said protein may be preferably ubiquitin, or it may be any chaperon protein, e.g. calreticulin.
  • Genetic Constructs, Immunogenic Compositions and Administration
  • Preferably, the nucleic acid is a genetic contrast comprising a polynucleotide sequence as defined herein, and regulatory sequences (such as a suitable promoter(s), enhancer(s), terminator(s), etc.) allowing the expression (e.g. transcription and translation) of the protein product in the host cell or host organism.
  • The genetic constructs of the invention may be DNA or RNA, and are preferably double-stranded DNA. The genetic constructs of the invention may also be in a form suitable for transformation of the intended host cell or host organism, in a form suitable for integration into the genomic DNA of the intended host cell or in a form suitable for independent replication, maintenance and/or inheritance in the intended host organism. For instance, the genetic constructs of the invention may be in the form of a vector, such as for example a plasmid, cosmid, YAC, a viral vector or transposon. In particular, the vector may be an expression vector, i.e. a vector that can provide for expression in vitro and/or in vivo (e.g. in a suitable host cell, host organism and/or expression system).
  • In a preferred but non-limiting aspect, a genetic construct of the invention comprises i) at least one nucleic acid of the invention; operably connected to ii) one or more regulatory elements, such as a promoter and optionally a suitable terminator; and optionally also iii) one or more further elements of genetic constructs such as 3′- or 5′-UTR sequences, leader sequences, selection markers, expression markers/reporter genes, and/or elements that may facilitate or increase (the efficiency of) transformation or integration.
  • In a particular embodiment, the genetic construct can be prepared by digesting the nucleic acid polymer with a restriction endonuclease and cloning into a plasmid containing a promoter such as the SV40 promoter, the cytomegalovirus (CMV) promoter or the Rous sarcoma virus (RSV) promoter. In a preferred embodiment, the TERT nucleic acid sequences are inserted into a pcDNA3.1 expression plasmid (see FIG. 2A).
  • Other vectors include retroviral vectors, lentivirus vectors, adenovirus vectors, vaccinia virus vectors, pox virus vectors, adenovirus-associated vectors and measle virus vectors.
  • Compositions can be prepared, comprising said nucleic acid or vector. The compositions are immunogenic. They can comprise a carrier or excipients that are suitable for administration in dogs (i.e. non-toxic, and, if necessary, sterile). Such excipients include liquid, semisolid, or solid diluents that serve as pharmaceutical vehicles, isotonic agents, stabilizers, or any adjuvant. Diluents can include water, saline, dextrose, ethanol, glycerol, and the like. Isotonic agents can include sodium chloride, dextrose, mannitol, sorbitol, and lactose, among others. Stabilizers include albumin, among others. Any adjuvant known in the art may be used in the vaccine composition, including oil-based adjuvants such as Freund's Complete Adjuvant and Freund's Incomplete Adjuvant, mycolate-based adjuvants, bacterial lipopolysaccharide (LPS), peptidoglycans, proteoglycans, aluminum hydroxide, saponin, DEAE-dextran, neutral oils (such as miglyol), vegetable oils (such as arachis oil), Pluronic® polyols.
  • The nucleic acid or composition can be administered directly or they can be packaged in liposomes or coated onto colloidal gold particles prior to administration. Techniques for packaging DNA vaccines into liposomes are known in the art, for example from Murray, 1991. Similarly, techniques for coating naked DNA onto gold particles are taught in Yang, 1992, and techniques for expression of proteins using viral vectors are found in Adolph, 1996.
  • For genetic immunization, the vaccine compositions are preferably administered intradermally, subcutaneously or intramuscularly by injection or by gas driven particle bombardment, and are delivered in an amount effective to stimulate an immune response in the host organism. In a preferred embodiment of the present invention, administration comprises an electroporation step, also designated herein by the term “electrotransfer”, in addition to the injection step (as described in Mir 2008, Sardesai and Weiner 2011).
  • The compositions may also be administered ex vivo to blood or bone marrow-derived cells using liposomal transfection, particle bombardment or viral transduction (including co-cultivation techniques). The treated cells are then reintroduced back into the subject to be immunized.
  • While it will be understood that the amount of material needed will depend on the immunogenicity of each individual construct and cannot be predicted a priori, the process of determining the appropriate dosage for any given construct is straightforward. Specifically, a series of dosages of increasing size, starting at about 5 to 30 μg, or preferably 20-25 μg, up to about 500 μg for instance, is administered to the corresponding species and the resulting immune response is observed, for example by detecting the cellular immune response by an IFNγ Elispot assay (as described in the experimental section), by detecting CTL response using a chromium release assay or detecting CD4 T cell (helper T cell) response using a cytokine release assay.
  • In a preferred embodiment, the vaccination regimen comprises one to three injections, preferably repeated three or four weeks later.
  • In a particular embodiment, the vaccination schedule can be composed of one or two injections followed three or four weeks later by at least one cycle of three to five injections.
  • In another embodiment, a primer dose is composed of one to three injections, followed by at least a booster dose every year, or every two or years for instance.
  • Prevention and Treatment of Tumors
  • The nucleic acid or immunogenic composition as described above is useful in a method for preventing or treating a tumor in a dog.
  • A method for preventing or treating a tumor in a dog is described, which method comprises administering an effective amount of said nucleic acid or immunogenic composition in a dog in need thereof. Said nucleic acid or immunogenic composition is administered in an amount sufficient to induce an immune response in the dog.
  • The tumor may be any undesired proliferation of cells, in particular a benign tumor or a malignant tumor, especially a cancer.
  • The cancer may be at any stage of development, including the metastatic stage. However preferably the cancer has not progressed to metastases.
  • In particular the tumor may be selected from the group consisting of bladder cancer, brain tumor, liver tumor, mammary tumors and carcinoma, mast cell tumors, malignant histiocytosis and histocytic sarcomas, squamous cell carcinomas, hemangiosarcoma, lymphoma, in particular B-cell lymphoma, melanoma, bone tumors (osteosarcoma), testicular tumors.
  • In a particular embodiment, the vaccination according to the invention may be combined with conventional therapy, including chemotherapy, radiotherapy or surgery. Combinations with adjuvant immunomodulating molecules such GM-CSF or IL-2 could also be useful.
  • The Figures and Examples illustrate the invention without limiting its scope.
  • EXAMPLES
  • The inventors have constructed DNA vaccines encoding an inactivated form of dog TERT and a cat/dog hybrid TERT (Example 1), and have assessed their functionality, safety and immunogenicity.
  • They have demonstrated that the plasmids were correctly processed in vitro after transfection in mammalian cells and that the plasmid product of expression (TERT protein) was well expressed. Moreover, no enzymatic activity was detected and TERT proteins were found excluded for the transfected cells nucleoli, which evidences safety of the constructs (Example 2).
  • Then, the plasmids were found to be immunogenic and to elicit specific efficient CD8 T cells and CD4 T cells in mice (Example 3).
  • Example 1: Construction of the DNA Plasmids
  • In all constructs, the TERT sequence is preceded by a DNA sequence encoding the human-ubiquitin. The presence of the ubiquitin will increase the addressing of the TERT protein to the proteasome and increase the class I presentation pathway of TERT derived peptides. TERT sequence is followed by the sequence of the influenza protein V5 to facilitate future purification or detection of the fusion protein by Western Blot or histochemistry for example. The DNA sequence coding for the TERT protein has been deleted of 47 amino-acids in the N-Ter region, which encodes the nucleolar importation signal. Moreover, three amino-acids have been removed in the catalytic site of TERT (VDD), to inhibit the protein enzymatic activity.
  • pDUV5 encodes the full-length of dog TERT nucleotide sequence, depleted of the N-term 47 amino acids (FIG. 1A), pCDT encodes 54.4% of the cat TERT sequence and 35.9% of the dog TERT sequence (FIG. 1B).
  • All TERT DNA sequences were synthetized from Genecust (Dudelange, Luxembourg). Then they were cloned into the pcDNA3,1 expression plasmid provided by Life technologies SAS (Saint-Aubin, France) using the HindIII and XbaI restriction sites (see FIG. 2A). Plasmids were stored at −20° C., in PBS 1×, at a concentration of 2 mg/mL prior use. The backbone plasmid was used as empty vector for western blot and Trap-Assay experiments. It consists of the pcDNA3.1 backbone plasmid deprived of the transgene protein DNA sequence (TERT).
  • Example 2: Functionality and Safety of the Plasmids 2.1. Materials and Methods Cell Culture
  • The human 293T cell line used for transfection assays and immune-fluorescence experiments were kindly provided by Pr Simon Wain-Hobson (Pasteur Institute). The CrFK (Crandall-Reese feline kidney) cells used for the TRAP-assay were kindly provided by Pr J. Richardson (Ecole Vétérinaire de Maison Alfort). Cells were grown at 37° C., 5% CO2- in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% heat-inactivated Fetal Calf Serum (FCS), 1% sodium-pyruvate, 1% penicillin-streptomycin pyruvate and 0.1% β-mercaptoethanol. All components of the culture medium were purchased from Life technologies SAS (Saint-Aubin, France).
  • Transfection Assays
  • Transfection of 293T cells were performed with either pCDT or pDUV5 plasmids using the JetPRIME® transfection kit (Polyplus-transfection SA, Illkirch, France) according to manufacturer's instruction. In a 6-well plate, 400 000 HeLa cells or 293T cells per well were seeded in 2 mL of DMEM culture medium, and cultured 24 hours at 37° C., 5% CO2 prior transfection. For each well, 2 μg of each plasmid diluted in 200 μL of jetPRIME® buffer, or 200 μL of jetPRIME® buffer only with respectively 4 μL of jetPRIME® agent were drop onto the cells. Transfection medium were removed 4 hours later and replaced by 2 mL of DMEM culture medium. Cells were put at 37° C., 5% CO2 and recovered for analysis 24 hours later.
  • Western Blots
  • Transfected 293 T cells were lysed on ice with radioimmunoprecipitation assay (RIPA) lysis buffer (RIPA Buffer, Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France) containing protease inhibitors cocktail (Complete EDTA-free, Roche Diagnostic, Indianapolis, USA) for 10-20 minutes. Then, suspension was centrifuged 15 minutes at 14000 rpm at 4° C. in order to remove cellular debris. The supernatants were harvested and the protein concentration was measured using the Bradford method. Protein samples were denatured 5 minutes at 95° C., separated on Nu-PAGE® Novex 4-12% Bis-Tris gels (Invitrogen, Carlsbad, USA) and transferred to PVDF membranes (iBlot® transfer stack, Invitrogen, Carlsbad, USA) using the iBlot® device (Invitrogen, Carlsbad, USA). The membrane was cut approximately at 60 kDa. First, the upper part membrane was probed with an anti-V5 antibody (Invitrogen, Carlsbad, USA) while the other part was probed with an anti-β-actin antibody (Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France), then samples were revealed by an ECL (Enhanced chemiluminescence) anti-mouse Horse Radish Peroxidase (HRP) linked antibody (GE Healthcare, Vélizy, France)). Immunoblot signals were reveled using 18×24 films and the corresponding cassette both products purchased from GE healthcare (Buckinghamshire, UK).
  • Immunofluorescence and Microscopy
  • Human 293T cells were seeded on 8-well Lab-Tek® chamber slides (Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France) at 20.103 cells/well in 200 μL of culture medium and incubated overnight at 37° C. The next day, culture medium was discarded. Ten μL of a mix solution containing 1 μg of either pCDT or PUF2 plasmid, 50 μL of OptiMEM (Life technologies SAS, Saint-Aubin, France) and 2.5 μL of Fugene HD (Promega France, Charbonnières-les-bains, France) were added to the corresponding chamber. As control, 20.103 HeLa cells were incubated with the 10 μL of the same mix without plasmid. Chamber slides were left in the incubator for 24 hours. Transfected 293T cells were carefully washed with PBS 1× and 200 μL 2% PFA were added to each well for 10 minutes at +4° C., in order to fix and permeabilize the cells. Then wells were washed two times with PBS 1×0.05% Tween® 20 and 293T cells were incubated 30 minutes at room temperature with 200 μL of Blocking solution (0.5% TritonX100; 3% BSA; 10% Goat Serum). Eventually, wells were incubated for 1.5 hours at room temperature with a primary mouse anti-V5 antibody (Life technologies SAS, Saint-Aubin, France) diluted in blocking solution at 1/200, with slight agitation. After three washes in PBS 1×0.05% Tween® 20, a secondary goat anti-mouse-Alexa Fluor 488® antibody (Life technologies SAS, Saint-Aubin, France) diluted in blocking solution (1/500) was put in the wells for 45 minutes at room temperature away from light and under slight agitation. Wells were washed three times with PBS 1×0.05% Tween® 20 and mounted with the Vectashield® mounting medium containing DAPI (Vector laboratories, Peterborough, UK). Slides were analyzed with a fluorescence microscope (Axio observer Z1, Carl Zeis Microlmaging GmbH, Jena, Germany) equipped with an image processing and analysis system (Axiovision, Carl Zeis Microlmaging GmbH, Jena, Germany).
  • TRAP Assay
  • Telomerase activity was measured by the photometric enzyme immunoassay for quantitative determination of telomerase activity, utilizing telomeric repeat amplification protocol (TRAP) (Yang et al, 2002). CrFK (Crandell Rees Feline Kidney) telomerase-negative cells (Yazawa et al., 2003) were transfected with plasmids encoding pDUV5, or pCDT TERT constructs. As a positive control CrFK cells were transfected with a plasmid encoding the wild type human TERT (fully active). Briefly, 24 hours after transfection, CrFK cells were harvested by mechanical scraping and then washed twice with 1 mL PBS and pelleted by centrifugation 5 minutes at 3000 g, at 4° C. Telomerase activity was assessed by TRAP-ELISA assay using the TeloTAGGG Telomerase PCR ELISAPLUS kit (Roche Diagnostics, Germany) according to the manufacturer's instructions. The protein concentration in the cell extract was measured by the Bradford method (Bio-Rad Laboratories). Three microliters of the cell extract (equivalent to 2.1, 0.21, 0.021 μg) was incubated in a Polymerase Chain reaction (PCR) mixture provided in the kit. The cycling program was performed with 30 minutes primer elongation at 25° C. and then the mixture was subjected to 30 cycles of PCR consisting of denaturation at 94° C. for 30 sec, annealing at 50° C. for 30 sec, polymerization at 72° C. for 90 sec and final extension at 72° C. for 10 minutes. 2.5 μl of amplification product was used for ELISA according to the manufacturer's instructions. The absorbance at 450 nm (with a reference of 690 nm) of each well was measured using Dynex MRX Revelation and Revelation TC 96 Well Microplate Reader.
  • Telomerase activity was calculated as suggested in the kit's manual and compared with a control template of 0.1 amol telomeric repeats, representing a relative telomerase activity (RTA) of 100. Inactivated samples and lysis buffer served as negative controls.
  • 2.2. Results
  • New TERT Encoding Plasmids are Functional In Vitro after Transfection
  • The functionality of the new plasmid constructs is shown by the presence of the plasmid encoded TERT protein in the total protein lysate of pCDT or pDUV5 transfected cells in vitro. The inventors performed western-blot assays on the total protein lysate of 293T cells plasmids transfected with pCDT or pDUV5 (24 h after transfection). As the TERT protein sequence encoded by each plasmid was tagged with the V5 protein sequence, anti-V5 antibody coupled with Horse Radish Peroxidase (HRP) was used to reveal the presence of the fusion protein of interest.
  • A highly positive V5 specific-signal was detected 24 h after transfection in the protein lysate of pCDT or pDUV5 transfected cells. The size of the protein band detected corresponds to the different TERT protein encoded by the plasmids which molecular weight is 123 kDa. Moreover no V5 specific signal was detected in untreated or empty plasmid transfected cells. The inventors demonstrated that pDUV5 and pCDT plasmids were correctly processed in vitro after transfection in mammalian cells and that the plasmid product of expression (TERT protein) was well expressed.
  • New TERT Encoding Plasmids Express a Non-Functional Enzyme of which Cellular Expression is Excluded from the Nucleoli after In Vitro Transfection
  • To test the absence of enzymatic activity, a TRAPeze assay was performed. As illustrated by FIG. 3, protein lysates from pDUV5 or pCDT transfected cells do not exhibit any telomerase activity. As a positive control, the protein extracts from CrFk cells transfected with the native human TERT (hTERT) were used. Thus the inventors demonstrated that the TERT proteins encoded by either pCDT or pDUV5 plasmids do not express any functional enzymatic activity after in vitro transfection.
  • The inventors have further investigated the intracellular location of the two plasmid products of expression. To this aim, an in vitro immunofluorescence assay was performed. Briefly, 24 h after in-vitro transfection of 293T cells with either pCDT or pDUV5, an anti-V5 antibody coupled to an Alexa-Fluor labeled secondary antibody were used to detect the TERT proteins within the cells. The pCDT and pDUV5 encoded TERTs were not detected inside the cell nucleoli contrary to what was observed with 293T cells transfected with the plasmid encoding the native human TERT.
  • To conclude, the inventors demonstrated that after in vitro transfection with either pDUV5 and pCDT plasmids, first the TERT protein expression is excluded from the nucleoli and secondly, these products of expression do not exhibit any enzymatic activity. These two criteria establish the safety of the plasmids and favour their use for in vivo vaccination.
  • Example 3: In Vivo Immune Response 3.1. Materials and Methods Mice
  • Female Balb/cBy and C57BL/6J mice (6-8 week old) were purchased from Janvier laboratories (Saint-Berthevin, France). Animals were housed at the Specific Pathogen Free animal facility of the Pasteur Institute. Mice were anesthetized prior to intradermal (ID) or intramuscular (IM) immunizations, with a mix solution of xylazine 2% (Rompun, Bayer Sante, Loos, France) and Ketamine 8% (Imalgen 1000, Merial, Lyon, France) in Phosphate Buffer Saline 1×(PBS 1×, Life technologies SAS, Saint-Aubin, France), according to individual animal weight and duration of anesthesia (intraperitoneal route). All animals were handled in strict accordance with good animal practice and complied with local animal experimentation and ethics committee guidelines of the Pasteur Institute of Paris.
  • H2 Restricted Peptides
  • TERT peptides used in mouse studies (IFNγ ELIspot) were predicted by in-silico epitope prediction in order to bind mouse class I MHC, H2Kb, H2Db or mouse class II H2-IAd using four algorithms available online:
  • Syfpeithi (http://www.syfpeithi.de/), Bimas (http://www-bimas.cit.nih.gov/), NetMHCpan and SMM (http://tools.immuneepitope.org/main/).
  • All synthetic peptides were purchased lyophilized (>90% purity) from Proimmune (Oxford, United Kingdom). Lyophilized peptides were dissolved in sterile water at 2 mg/mL and stored in 35 μL aliquots at −20° C. prior use. Details of peptides sequence and H2 restriction is shown in table 1.
  • TABLE 1
    H2 restricted peptides sequences determined by in
    silico prediction algorithms
    H2Db restricted TERT peptides
     621-629 (RPIVNMDYI)  621 SEQ ID NO: 8
     580-589 (RQLFNSVHL)  580 SEQ ID NO: 9
     987-996 (TVYMNVYKI)  987 SEQ ID NO: 10
    H2-IAd restricted TERT peptides
    1106-1121 (CLLGPLRAAKAHLSR) 1106 SEQ ID NO: 11
    1105-1120 (RCLLGPLRAAKAHLS) 1105 SEQ ID NO: 12
     951-966 (YSSYAQTSIRSSLTF)  951 SEQ ID NO: 13
    1109-1124 (GPLRAAKAHLSRQLP) 1109 SEQ ID NO: 14
  • Mice Immunization and In Vivo Electroporation
  • Intradermal (ID) immunization was performed on the lower part of the flank with Insulin specific needles (U-100, 29 G×1/2″−0.33×12 mm, Terumo, Belgium) after shaving. No erythema was observed after shaving, during and after immunization procedure. Intramuscular immunization (IM) was performed in the anterior tibialis cranialis muscle, also using Insulin specific needles U-100. Each animal received a priming dose of either pCDT or pDUV5, independently of vaccine route, corresponding to 100 μg of DNA. All animals were boosted at day 14 post-prime using the same amount of plasmid and the same route of immunization. Directly after ID vaccination, invasive needle electrodes (6×4×2, 47-0050, BTX, USA) are inserted into the skin so that the injection site is placed between the two needle rows (the two needle rows are 0.4 cm apart). Two pulses of different voltages were applied (HV-LV): HV=1125 V/cm (2 pulses, 50 μs-0.2 μs pulse interval) and LV=250 V/cm (8 pulses, 100 V-10 ms-20 ms pulse interval). Immediately after IM immunization the muscle injection site was covered with ultrasonic gel (Labo FH, blue contact gel, NM Médical, France) and surrounded by tweezers electrodes (0.5 cm apart, tweezertrode 7 mm, BTXI45-0488, USA) and voltage was applied using the same parameters than for skin electroporation. The Agilepulse® in vivo system electroporator was used for all experiments (BTX, USA).
  • For each route of immunization (IM, ID) control mice were treated with the same procedures using the same volume of PBS 1×.
  • IFN□ ELispot Assay
  • Briefly, PVDF microplates (IFN-γ Elispot kit, Diaclone, Abcyss, France, 10×96 tests, ref 862.031.010P) were coated overnight with capture antibody (anti-mouse IFN-γ) and blocked with PBS 2% milk. Spleens from pDNA-immunized mice were mashed and cell suspensions were filtered through a 70-mm nylon mesh (Cell Strainer, BD Biosciences, France). Ficoll-purified splenocytes (Lymphocyte Separation Medium, Eurobio, France) were numerated using the Cellometer® Auto T4 Plus counter (Ozyme, France) and added to the plates in triplicates at 2×105 or 4×105 cells/well and stimulated with 5 μg/ml of dTERT or hyTERT relevant peptides or Concanavalin A (10 μg/ml), or mock stimulated with serum free culture medium. After 19 hours, spots were revealed with the biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution. Spots were counted using the Immunospot ELIspot counter and software (CTL, Germany).
  • Dog TERT Peptide Pools
  • The vast majority of peptides were 15 residues long. A few are 14 amino acids long.
  • Pool 2
    (SEQ ID NO: 15)
    PQKPGAARRMRRLPA,
    (SEQ ID NO: 16)
    GAARRMRRLPARYWR,
    (SEQ ID NO: 17)
    RMRRLPARYWRMRPL,
    (SEQ ID NO: 18)
    LPARYWRMRPLFQEL,
    (SEQ ID NO: 19)
    YWRMRPLFQELLGNH,
    (SEQ ID NO: 20)
    RPLFQELLGNHARCP,
    (SEQ ID NO: 21)
    QELLGNHARCPYRAL,
    (SEQ ID NO: 22)
    GNHARCPYRALLRTH,
    (SEQ ID NO: 23)
    RCPYRALLRTHCPLR,
    (SEQ ID NO: 24)
    RALLRTHCPLRAMAA,
    (SEQ ID NO: 25)
    RTHCPLRAMAAKEGS,
    (SEQ ID NO: 26)
    PLRAMAAKEGSGNQA,
    (SEQ ID NO: 27)
    MAAKEGSGNQAHRGV,
    (SEQ ID NO: 28)
    EGSGNQAHRGVGICP,
    (SEQ ID NO: 29)
    NQAHRGVGICPLERP,
    (SEQ ID NO: 30)
    RGVGICPLERPVAAP,
    (SEQ ID NO: 31)
    ICPLERPVAAPQEQT,
    (SEQ ID NO: 32)
    PQKPGAARRMRRLPA
    Pool 4
    (SEQ ID NO: 33)
    AKLSLQELTWKMKVR,
    (SEQ ID NO: 34)
    LQELTWKMKVRDCTW,
    (SEQ ID NO: 35)
    TWKMKVRDCTWLHGN,
    (SEQ ID NO: 36)
    KVRDCTWLHGNPGAC,
    (SEQ ID NO: 37)
    CTWLHGNPGACCVPA,
    (SEQ ID NO: 38)
    HGNPGACCVPAAEHR,
    (SEQ ID NO: 39)
    GACCVPAAEHRRREE,
    (SEQ ID NO: 40)
    VPAAEHRRREEILAR,
    (SEQ ID NO: 41)
    EHRRREEILARFLVL,
    (SEQ ID NO: 42)
    REEILARFLVLVDGH,
    (SEQ ID NO: 43)
    LARFLVLVDGHIYVV,
    (SEQ ID NO: 44)
    LVLVDGHIYVVKLLR,
    (SEQ ID NO: 45)
    DGHIYVVKLLRSFFY,
    (SEQ ID NO: 46)
    YVVKLLRSFFYVTET,
    (SEQ ID NO: 47)
    LLRSFFYVTETTFQK,
    (SEQ ID NO: 48)
    FFYVTETTFQKNRLF,
    (SEQ ID NO: 49)
    TETTFQKNRLFFYRK,
    (SEQ ID NO: 50)
    FQKNRLFFYRKSVW
    Pool 6
    (SEQ ID NO: 51)
    EGGPPGTRPTTPAWH,
    (SEQ ID NO: 52)
    PGTRPTTPAWHPYPG,
    (SEQ ID NO: 53)
    PTTPAWHPYPGPQGV,
    (SEQ ID NO: 54)
    AWHPYPGPQGVPHDP,
    (SEQ ID NO: 55)
    YPGPQGVPHDPAHPE,
    (SEQ ID NO: 56)
    QGVPHDPAHPETKRF,
    (SEQ ID NO: 57)
    HDPAHPETKRFLYCS,
    (SEQ ID NO: 58)
    HPETKRFLYCSGGRE,
    (SEQ ID NO: 59)
    KRFLYCSGGRERLRP,
    (SEQ ID NO: 60)
    YCSGGRERLRPSFLL,
    (SEQ ID NO: 61)
    GRERLRPSFLLSALP,
    (SEQ ID NO: 62)
    LRPSFLLSALPPTLS,
    (SEQ ID NO: 63)
    FLLSALPPTLSGARK,
    (SEQ ID NO: 64)
    ALPPTLSGARKLVET
    Pool 10
    (SEQ ID NO: 65)
    DCTWLHGNPGACCVP,
    (SEQ ID NO: 66)
    LHGNPGACCVPAAEH,
    (SEQ ID NO: 67)
    PGACCVPAAEHRRRE,
    (SEQ ID NO: 68)
    CVPAAEHRRREEILA,
    (SEQ ID NO: 69)
    AEHRRREEILARFLV,
    (SEQ ID NO: 70)
    RREEILARFLVLVDG,
    (SEQ ID NO: 71)
    ILARFLVLVDGHIYV,
    (SEQ ID NO: 72)
    FLVLVDGHIYVVKLL,
    (SEQ ID NO: 73)
    VDGHIYVVKLLRSFF,
    (SEQ ID NO: 74)
    IYVVKLLRSFFYVTE,
    (SEQ ID NO: 75)
    KLLRSFFYVTETTFQ,
    (SEQ ID NO: 76)
    SFFYVTETTFQKNRL,
    (SEQ ID NO: 77)
    VTETTFQKNRLFFYR,
    (SEQ ID NO: 78)
    TFQKNRLFFYRKSVW
    Pool 19
    (SEQ ID NO: 79)
    QLPFNQPVRKNPSFF,
    (SEQ ID NO: 80)
    NQPVRKNPSFFLRVI,
    (SEQ ID NO: 81)
    RKNPSFFLRVIADTA,
    (SEQ ID NO: 82)
    SFFLRVIADTASCCY,
    (SEQ ID NO: 83)
    RVIADTASCCYSLLK,
    (SEQ ID NO: 84)
    DTASCCYSLLKARNA,
    (SEQ ID NO: 85)
    CCYSLLKARNAGLSL,
    (SEQ ID NO: 86)
    LLKARNAGLSLGAKG,
    (SEQ ID NO: 87)
    RNAGLSLGAKGASGL,
    (SEQ ID NO: 88)
    LSLGAKGASGLFPSE,
    (SEQ ID NO: 89)
    AKGASGLFPSEAARW,
    (SEQ ID NO: 90)
    SGLFPSEAARWLCLH,
    (SEQ ID NO: 91)
    PSEAARWLCLHAFL, 
    (SEQ ID NO: 92)
    ARWLCLHAFLLKLAH
  • In Vivo Cytotoxicity Assay
  • Briefly, for target cell preparation, splenocytes from naïve C57/B16 mice were labeled in PBS 1× containing high (5 μM), medium (1 μM) or low (0.2 μM) concentrations of CFSE (Vybrant CFDA-SE cell-tracer kit; Life technologies SAS, Saint-Aubin, France). Splenocytes labeled with 5 and 1 μM CFSE were pulsed with 2 different H2 peptides at 5 μg/ml for 1 hour and 30 minutes at room temperature. Peptides 987 and 621 were used for pulsing respectively CFSE high and medium labeled naïve splenocytes. CFSE low labeled splenocytes were left unpulsed. Each mouse previously immunized with either pCDT or pDUV5 received at day 10 post-boost injection 10′ CF SE-labeled cells of a mix containing an equal number of cells from each fraction, through the retro-orbital vein. After 15-18 hours, single-cell suspensions from spleens were analyzed by flow cytometry MACSQUANT® cytometer (Miltenyii, Germany).
  • The disappearance of peptide-pulsed cells was determined by comparing the ratio of pulsed (high/medium CFSE fuorescence intensity) to unpulsed (low CFSE fuorescence intensity) populations in pDNA immunized mice versus control (PBS 1× injected) mice. The percentage of specific killing per test animal was established according to the following calculation: [1−[mean (CFSElowPBS/CFSEhighi/mediumPBS)/(CFSElowpDNA/CFSEhigh/medimpDNA)]]×100.
  • Statistical Analysis and Data Handling
  • Prism-5 software was used for data handling, analysis and graphic representations. Data are represented as the mean±standard deviation. For statistical analyses of EliSPOT assays we used a Mann Whitney non parametric test, and a Kruskal-Wallis analysis with Dunn's multiple comparison test for in-vivo cytotoxicity assay. Significance was set at p-value<0.05.
  • 3.2. Results
  • pDUV5 Induces a Strong Cytotoxic CD8 T Cell Response after ID or IM Immunization and EP in Mice
  • The inventors have assessed whether pDUV5 plasmid DNA plasmid was capable of eliciting efficient cellular immune responses (CD8) in mice. To this aim, different groups of 9-10 C57-Bl/6 mice were injected ID or IM with pDUV5 immediately followed by electroporation. Two weeks later, mice received a boost injection with the same protocol. On day 10 post-boost, mice spleens were harvested and the induced immune response was monitored via an IFN-γ ELISPOT assay using H2 restricted peptides described in Table 1. Dog TERT peptides restricted to mouse MHC class I were predicted in silico as described in the material and methods section. As shown in FIG. 4, a significant augmentation in the frequency of dTERT specific IFN-γ secreting CD8 T-cells was observed in the spleen of ID and IM vaccinated animals in comparison with control mice. This was observed for 2 out of 3 peptides (p621 and p98′7) (p<0.05). No significant difference was observed between the 2 routes of administration.
  • pDUV5 construct is able to promote the expansion of dTERT specific CD8 T-cells in mice. The inventors next wanted to show that those specific T-cells exhibit a functional cytotoxic activity in vivo, which will be necessary to attack tumor cells. In order to measure the in vivo cytolytic strength of the CD8+ T-cell response elicited by pDUV5 immunization, the inventors performed in vivo cytotoxicity tests using carboxyfuorescein-diacetate suc-cinimidyl ester (CFSE)-labelled, peptide-pulsed splenocytes as target cells. 7 week old C57/B16 mice which received a prime and boost vaccination with pDUV5 via the ID or IM route as described before or mock-immunized with phosphate-buffered saline (PBS) were intravenously injected with 10′ target cells. Target cells were splenocytes from naïve congenic mice separately labelled with three different concentrations of CFSE and pulsed with individual peptides (p621 or p98′7) or left un-pulsed as an internal control. After 15-18 hours, spleen cells were obtained and the disappearance of peptide-pulsed cells in control versus immunized mice was quantifed by fluorescence-activated cell sorting.
  • Results show that mice develop CTLs against the 2 epitopes predicted in silico (FIGS. 5A and 5B). Peptide 621 gives the strongest in vivo lysis. Results were concordant with IFN-γ Elispot assays (FIG. 4). No significant difference was observed between the two routes of immunization.
  • pCDT Induces a Strong Cytotoxic CD8 T Cell Response Along with a Specific CD4 T Cell Response after ID or IM Immunization and Electroporation in Mice
  • In light of the importance of cytotoxic CD8 T cells in antitumor immune responses, the inventors have assessed whether plasmid pCDT was able to promote such an immune response in vivo. Thus, different groups of 9-10 C57-Bl/6 mice were immunized with pCDT by ID or IM injection of the plasmid immediately followed by electroporation. Two weeks later, mice received a boost injection with the same protocol. On day 10 post-boost, mice spleens were harvested and the induced immune response was monitored via an IFN-γ ELISPOT assay using H2 restricted peptides described in Table 1.
  • Hy-TERT peptides restricted to mouse MHC class I were predicted in silico as described in the material and methods section. As shown in FIG. 6A, a significant augmentation in the frequency of hyTERT specific IFN-γ secreting CD8 T-cells was observed in the spleen of ID and IM vaccinated animals in comparison with control mice. This was observed for 2 out of 3 class I restricted peptides (p621 and p987, p<0.05). No significant difference in the frequency of specific CD8 T cells was observed between IM and ID route for both peptides p921 and p987. The inventors have further investigated the hyTERT restricted CD4 T cell response. To this aim, 9-10 Balb/C mice were immunized with pCDT by ID or IM injection immediately followed by electroporation and the CD4 specific T cell response was monitored in the spleen as described before using hyTERT IAd restricted peptides (in silico prediction). Balb/C mice were chosen because this mouse strain is known to develop good CD4 T cell responses. As shown in FIG. 6B, when performing the IFN-γ ELISPOT assay, a significant augmentation in the frequency of hyTERT specific IFN-γ secreting CD4 T-cells was observed in the spleen of ID and IM vaccinated Balb/C mice in comparison with control mice injected with PBS 1×. This was observed for 2 out of 3 class I restricted peptides (p1106 and p1105, with respectively for p1106 p<0.05 for ID route and p<0.001 for IM route and for 1105 the difference was not significant for ID route and p<0.01 for IM route). No significant difference in the frequency of specific CD4 T cells was observed between IM and ID route for both peptides p1105 and p1106.
  • Thus, pCDT construct is able to promote the expansion of hyTERT specific CD8 and CD4 T-cells in mice. The inventors next wanted to show that hyTERT specific CD8 T-cells exhibit a functional cytotoxic activity in vivo, which will be necessary to destroy tumor cells. In order to measure the in vivo cytolytic strength of the CD8+ T-cell response elicited by pCDT immunization, the inventors performed an in vivo cytotoxicity test using carboxyfluorescein-diacetate succinimidyl ester (CFSE)-labelled, peptide-pulsed splenocytes as target cells. 7 week old C57/B16 mice which received a prime and boost vaccination with pCDT via the ID or IM route as described before or mock-immunized with phosphate-buffered saline (PBS) were intravenously injected with 107 target cells. Target cells were splenocytes from naïve congenic mice separately labelled with three different concentrations of CFSE and pulsed with individual peptides (p621 or p98′7) or left un-pulsed as an internal control. After 15-18 hours, spleen cells were obtained and the disappearance of peptide-pulsed cells in control versus immunized mice was quantified by fluorescence-activated cell sorting.
  • Results show that mice develop CTLs against the 2 peptides p621 and p987 which were predicted in silico. Peptide 987 gives the strongest in vivo lysis. Results were consistent with the ones from the IFN-γ Elispot assays (FIG. 6A). It is worth mentioning that for p621, the mean percent lysis was slightly superior when pCDT was injected via the ID route (mean ID=7.7% vs mean IM=0.2%), however, no significant difference was observed between the two routes of immunization.
  • Example 4: Dog TERT Specific T Cell Repertoire 4.1. Materials and Methods Dog TERT Peptides Library
  • Lyophilized dTERT peptides (purity>90%) were purchased from JPT Peptide Technologies (Berlin, Germany). Each peptide was resuspended in distilled H2O, 5% DMSO at 2 mg/mL prior use according to supplier recommendation and kept frozen at −20° C. before use. One third of the dog TERT peptide (AA 281 to 571) was used to synthetized 70 peptides of 15 AA overlapping of 11 AA and recovering this sequence of the dog TERT as depicted in FIG. 3. Four pools of peptides were used for in vitro experiments and ELIspot assays in dogs.
  • Canine Blood Products
  • Canine blood samples were purchased from the Bourgelat Institute (Marcy l'Etoile, France). It was taken from a healthy 4-year-old beagle dog housed, fed and cared for in accordance with institutional and ethical guidelines. Heparinized blood samples was 4 time diluted in PBS 1× (Life technologies SAS, Saint-Aubin, France). Diluted samples were then layered on Lymphocyte Separation Medium (Eurobio, Courtaboeuf, France) and centrifuged 30 minutes at 2200 rpm (at room temperature) without break. Canine PBMCs were harvested and stored in Fetal Calf Serum (FCS, PAA Laboratories GmbH, Pashing, Austria) with 10% DMSO (Sigma Aldrich chimie SARL, Saint-Quentin Fallavier, France) in liquid nitrogen prior use.
  • In Vitro Immunization Assays in Dogs PBMCs
  • On day 0, dog frozen PBMCs were recovered, counted using the Cellometer® Auto T4 Plus counter (Ozyme, France) and plated in duplicates or triplicates at 106 cells/mL in 48-well flat-bottomed plates (BD, France) in AIM-V medium (Invitrogen) supplemented with either 100 ng/mL caGM-CSF and 5 ng/mL calL-4 (R&DSystems) or 50 ng/mL human FlT3L (Immunotools). Cells were cultured at 37° C., 5% CO2 in an incubator.
  • After 24 hours (day1), maturation stimuli were added, comprising the following reagent: 50 ng/mL rcTNFα, 20 ng/mL rcIL1-β (R&DSystems), 1 ng/mL hIL-7 (Miltenyi). Pools of peptides were also added. The final concentration used for each peptide was 10 μg/mL. Control wells received the cocktails of maturation cytokines only and no peptide. At day 3, culture medium was discarded and fresh AIM-V was added. Fresh AIM-V was added every 3 days until the day of testing. At either day 11 or day 18 after the beginning of culture, cells were recovered, washed in fresh AIM-V medium and used for the ELIspot assay.
  • Briefly cells were plated with the 4 pools of peptides (5 μg/mL of each peptide) in AIMV-5 or in AIMV only. Concanavalin A (10 μg/ml) and recombinant canine IFN-γ (16 ng/mL) was used for positive control weeks. After 24 hours, spots were revealed with the biotin-conjugated detection antibody followed by streptavidin-AP and BCIP/NBT substrate solution. Spots were counted using the Immunospot ELIspot counter and software (CTL, Germany).
  • 4.2. Results
  • In order to highlight the relevance of the vaccine technology of the invention, the inventors wanted to demonstrate the existence of a pre-existing dogTERT specific T-cell repertoire in the target species, i.e dogs.
  • The inventors have investigated whether dTERT-specific T-cell responses could be enhanced in PBMCs incubated with either rcGM-CSF and rcIL-4 for 24 hours, or with hFlt3 ligand followed by maturation stimuli (rcIL1β, rcTNFα and IL-7) and peptides stimulation for another 24 hours and 11 or 18 days of in vitro cell expansion. This technique was described by Mallone and colleagues for human PBMCs and is called in vitro immunization (Martinuzzi et al. 2011). The principle of this experiment is exposed in FIG. 8. To stimulate specific T cells, 15mer overlapping peptides recovering one third of the dog TERT protein (FIG. 3) were used in pools containing 17 to 18 peptides each.
  • Eleven or 18 days after the beginning of culture, cells were subsequently transferred into dog IFN-γ ELISPOT plates for 24 hours with 5 μg/mL of each pool of peptides. The inventors have noticed a threefold increase in the frequency of dTERT specific IFN-γ secreting T cells with pool 2/rcGMCSF+rcIL-4 and a twofold increase with pool 4/FlT-3L after 11 days of culture in comparison with medium stimulated PBMCs (FIG. 9A). Moreover, a fourfold increase in the frequency of dTERT specific IFN-γ secreting T cells was observed after 18 days of culture with pool 4/Flt-3L in comparison with PBMCs stimulated with culture medium (FIG. 9B). These results demonstrate the existence of a naturally occurring repertoire of dog TERT specific IFN-γ secreting T-cell repertoire in peripheral blood of naïve experimentation dog.
  • Example 5: In Vivo Specific Cellular Immune Response in Dogs Vaccinated with pDUV5
  • Six naïve beagle dogs received a local anaesthetic of 2.5 mg/kg IV imalgene and 20-80 μg/kg IV dorbene 15-20 minutes before vaccination and 100-400 μg/kg IM post vaccination. The dogs were injected intradermally with 400 μg of pDUV5 DNA followed by electroporation. pDU5 DNA was electroporated at days 0, 29, 57 and 142. Peripheral blood was drawn and mononuclear cells tested for dog telomerase specific peptides belonging either to pool 6 or pool 19 according to the method of Martinuzzi et al., 2011.
  • FIGS. 10A and 10B show that IFNγ specific T cell responses were detected. As shown on FIGS. 11A and 11B, pDUV5 DNA vaccination at days 57 and 142 show classical long term memory responses, that is rising sharply and decaying more slowly.
  • Example 6: Specific dTERT T Cell Responses in Animals with Neoplasias
  • To show that pDUV5 DNA electroporation can induce specific dTERT T cell responses in animals with neoplasias, five pet dogs with neoplasias and three pet dogs as controls were used. The diseased animals presented with widely different tumours. See Table 2 below.
  • TABLE 2
    Data for healthy and tumor bearing dogs:
    Name of dog Dog breed Age (years) Sex Pathology
    Belka Boxer 7 F Healthy
    Choupette Jack Russel 5 F Healthy
    Tequila Rottweiler 9 F Healthy
    Bambou Labrador 8 F Mastocytoma grade II
    Lambert Labrador 12 M Tumor Hypothesis
    (liver/right adrenal)
    Maury Bernese 9 M Neoplastic process + lung
    Mountain dog metastasis
    Semelaigne Cavalier King Charles 10 F Bone tumor
    Fidji Shetland sheepdog 2.5 M Histiocytoma
  • Peripheral blood was drawn and the in vitro stimulation protocol as described in Example 4 and in Martinuzzi zt al., 2011, was performed, using pool 4 peptides.
  • As can be seen in FIG. 12, specific peptide responses well over medium controls were identified for all animals. This means that the immunological repertoire is not depleted, biased or suppressed by the neoplasias. The latter finding is particularly important for it shows that even if there was some degree of immunosuppression or excessive Treg induction in diseased dogs, the vaccination of the invention is nonetheless capable of inducing T cell responses.
  • REFERENCES
    • Adolph, K. 1996 ed. “Viral Genome Methods” CRC Press, Florida
    • de Fornel P, Delisle F, Devauchelle P, Rosenberg D. 2007. Effects of radiotherapy on pituitary corticotroph macrotumors in dogs: a retrospective study of 12 cases. Can Vet J 48: 481-486.
    • Dillman R O. 2011. Cancer Immunotherapy. Cancer Biotherapy and Radiopharmaceuticals 26: 1-64.
    • Disis M L, Bernhard H, Jaffee E M. 2009. Use of tumour-responsive T cells as cancer treatment. Lancet 373: 673-683.
    • Finn O J. 2008. Cancer immunology. N Engl J Med 358: 2704-2715.
    • Fridman W H, Pages F, Sautes-Fridman C, Galon J. 2012. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 12: 298-306.
    • Gentilini F. 2010. “Masitinib” is safe and effective for the treatment of canine mast cell tumors. J Vet Intern Med 24: 6; author reply 7.
    • Hanahan D, Weinberg R A. 2011. Hallmarks of cancer: the next generation. Cell 144: 646-674.
    • Jourdier T M, Moste C, Bonnet M C, Delisle F, Tafani J P, Devauchelle P, Tartaglia J, Moingeon P. 2003. Local immunotherapy of spontaneous feline fibrosarcomas using recombinant poxviruses expressing interleukin 2 (IL2). Gene Therapy 10: 2126-2132.
    • Manley C A, Leibman N F, Wolchok J D, Riviere I C, Bartido S, Craft D M, Bergman P J. 2011. Xenogeneic murine tyrosinase DNA vaccine for malignant melanoma of the digit of dogs. J Vet Intern Med 25: 94-99.
    • Marconato L. 2011. The staging and treatment of multicentric high-grade lymphoma in dogs: a review of recent developments and future prospects. Vet J 188: 34-38.
    • Martinez P, Blasco M A. 2011. Telomeric and extra-telomeric roles for telomerase and the telomere-binding proteins. Nature Reviews Cancer 11: 161-176.
    • Martinuzzi E, et al. 2011. acDCs enhance human antigen-specific T-cell responses. Blood 118: 2128-2137.
    • Merlo D F, et al. 2008. Cancer incidence in pet dogs: findings of the Animal Tumor Registry of Genoa, Italy. J Vet Intern Med 22: 976-984.
    • Mir L M. 2008. Application of electroporation gene therapy: past, current, and future. Methods Mol Biol 423: 3-17.
    • Murray, 1991, ed. “Gene Transfer and Expression Protocols” Humana Pres, Clifton, N.J.
    • Sardesai N Y, Weiner D B. 2011. Electroporation delivery of DNA vaccines: prospects for success. Curr Opin Immunol 23: 421-429.
    • Topalian S L, Weiner G J, Pardoll D M. 2011. Cancer Immunotherapy Comes of Age. Journal of Clinical Oncology 29: 4828-4836.
    • Vascellari M, Baioni E, Ru G, Carminato A, Mutinelli F. 2009. Animal tumour registry of two provinces in northern Italy: incidence of spontaneous tumours in dogs and cats. BMC Vet Res 5: 39.
    • Yang, 1992, “Gene transfer into mammalian somatic cells in vivo”, Crit. Rev. Biotech. 12: 335-356
    • Yang Y, Chen Y, Zhang C, Huang H, Weissman S M. 2002. Nucleolar localization of hTERT protein is associated with telomerase function. Exp Cell Res 277: 201-209.
    • Yazawa M, et al, 2003, J. Vet. Med. Sci 65(5):573-577

Claims (20)

1-19. (canceled)
20. A nucleic acid molecule comprising a sequence encoding a fusion protein which comprises a) at least one dog telomerase reverse transcriptase (TERT) fragment, and b) at least one non-dog TERT fragment;
wherein said protein (i) does not contain amino acids VDD within the TERT catalytic activity, (ii) does not contain a nucleolar localization signal sequence, and (iii) comprises an amino acid sequence which enhances the addressing of said TERT to a proteasome;
wherein the at least one dog TERT fragment represents at least 30% of the amino acid sequence of a TERT sequence;
and wherein the at least one non-dog TERT fragment is a non-dog TERT antigenic fragment that correspond to a fragment absent from said dog TERT sequence, to the extent the non-dog TERT fragment does not complement the loss of activity nor the loss of the nucleolar localization signal.
21. The nucleic acid of claim 20, wherein the nucleic acid is a DNA plasmid.
22. The nucleic acid of claim 20, wherein the amino acid sequence which enhances the addressing of said TERT to a proteasome is a sequence of ubiquitin.
23. The nucleic acid of claim 20, wherein the amino acid sequence which enhances the addressing of said TERT to a proteasome is a sequence of calreticulin.
24. The nucleic acid of claim 20, wherein the nucleolar localization signal sequence consists of 47 N-terminal amino acids of the full-length wild-type dog TERT.
25. The nucleic acid of claim 20, wherein the at least one non-dog TERT antigenic fragment originates from a cat TERT sequence.
26. The nucleic acid of claim 25, which encodes SEQ ID NO: 4.
27. The nucleic acid of claim 20, wherein the at least one dog TERT fragment represents at least 50% of all TERT sequences in the nucleic acid.
28. The nucleic acid of claim 27, wherein the at least one dog TERT fragment represents at least 70% of all TERT sequences in the nucleic acid.
29. The nucleic acid of claim 28, wherein the at least one dog TERT fragment represents at least 90% of all TERT sequences in the nucleic acid.
30. An immunogenic composition comprising the nucleic acid of claim 20 and a carrier and/or excipient.
31. A method for triggering an immune response in a dog, against cells that overexpress telomerase, wherein the method comprises administering to the dog an effective amount of the immunogenic composition of claim 30.
32. The method of claim 31, wherein said cells that overexpress telomerase are dysplasia cells or tumor cells.
33. The method of claim 31, wherein the composition is administered by intradermal or intramuscular route.
34. The method of claim 31, wherein the composition induces a long term memory immune response.
35. A method for preventing or treating a tumor in a dog, which method comprises administering to the dog an effective amount of the immunogenic composition of claim 30.
36. The method of claim 35, wherein the tumor is selected from the group consisting of bladder cancer, brain tumor, mammary tumors and carcinoma, mast cell tumors, malignant histiocytosis and histocytic sarcomas, squamous cell carcinomas, hemangiosarcoma, lymphoma, in particular B-cell lymphoma, melanoma, osteosarcoma, testicular tumors.
37. The method of claim 35, wherein the composition is administered by intradermal or intramuscular route.
38. The method of claim 35, wherein the dog is at risk of developing a tumor, or wherein the dog is healthy but is 10 years of age or more.
US16/197,563 2013-03-28 2018-11-21 Cancer vaccine for dogs Abandoned US20190177733A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/197,563 US20190177733A1 (en) 2013-03-28 2018-11-21 Cancer vaccine for dogs

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP13305405 2013-03-28
EP13305405.6 2013-03-28
PCT/EP2014/056381 WO2014154905A1 (en) 2013-03-28 2014-03-28 A cancer vaccine for dogs
US201514780652A 2015-09-28 2015-09-28
US16/197,563 US20190177733A1 (en) 2013-03-28 2018-11-21 Cancer vaccine for dogs

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2014/056381 Continuation WO2014154905A1 (en) 2013-03-28 2014-03-28 A cancer vaccine for dogs
US14/780,652 Continuation US10138488B2 (en) 2013-03-28 2014-03-28 Cancer vaccine for dogs

Publications (1)

Publication Number Publication Date
US20190177733A1 true US20190177733A1 (en) 2019-06-13

Family

ID=48083084

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/780,652 Expired - Fee Related US10138488B2 (en) 2013-03-28 2014-03-28 Cancer vaccine for dogs
US16/197,563 Abandoned US20190177733A1 (en) 2013-03-28 2018-11-21 Cancer vaccine for dogs

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/780,652 Expired - Fee Related US10138488B2 (en) 2013-03-28 2014-03-28 Cancer vaccine for dogs

Country Status (7)

Country Link
US (2) US10138488B2 (en)
EP (2) EP2978444B1 (en)
JP (2) JP6479761B2 (en)
AU (1) AU2014242916B2 (en)
CA (1) CA2908143C (en)
DK (1) DK2978444T3 (en)
WO (1) WO2014154905A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112018015893A2 (en) * 2016-02-05 2019-03-06 Inovio Pharmaceuticals Inc cancer vaccines and treatment methods using the same
EP3622078A1 (en) 2017-05-09 2020-03-18 Invectys Recombinant measles vaccine expressing htert
US20200147195A1 (en) * 2017-05-26 2020-05-14 The Wistar Institute Of Anatomy And Biology Dtert vaccines and methods of treatment using the same
JP2020522479A (en) * 2017-06-02 2020-07-30 アリゾナ ボード オブ リージェンツ オン ビハーフ オブ アリゾナ ステート ユニバーシティ Methods for making personalized cancer vaccines
EP4188426A2 (en) * 2020-07-29 2023-06-07 Evvivax S.r.l. Consensus sequence of the antigen telomerase and the use thereof in preventive and therapeutic vaccination

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69020143T2 (en) 1989-02-17 1996-01-25 Chiron Mimotopes Pty Ltd METHOD FOR USE AND PRODUCTION OF PEPTIDES.
US5840839A (en) 1996-02-09 1998-11-24 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Alternative open reading frame DNA of a normal gene and a novel human cancer antigen encoded therein
KR20000048820A (en) 1996-10-01 2000-07-25 게론 코포레이션 Telomerase reverse transcriptase
US20030143228A1 (en) 2001-10-29 2003-07-31 Baylor College Of Medicine Human telomerase reverse transcriptase as a class-II restricted tumor-associated antigen
US20040106128A1 (en) * 2002-06-27 2004-06-03 Majumdar Anish Sen Cancer vaccines containing xenogeneic epitopes of telomerase reverse transcriptase
EP1748067A1 (en) * 2005-07-29 2007-01-31 Institut Pasteur Polynucleotides encoding MHC class I-restricted hTERT epitopes, analogues thereof or polyepitopes
NZ576134A (en) 2006-10-12 2011-09-30 Angeletti P Ist Richerche Bio Telomerase reverse transcriptase fusion protein, nucleotides encoding it, and uses thereof
US20110318380A1 (en) 2008-10-01 2011-12-29 Dako Denmark A/S MHC Multimers in Cancer Vaccines and Immune Monitoring
WO2014154904A1 (en) * 2013-03-28 2014-10-02 Invectys A cancer vaccine for cats
EP3099704B1 (en) 2014-01-27 2021-03-17 Molecular Templates, Inc. Mhc class i epitope delivering polypeptides

Also Published As

Publication number Publication date
JP2019089813A (en) 2019-06-13
EP2978444A1 (en) 2016-02-03
WO2014154905A1 (en) 2014-10-02
AU2014242916B2 (en) 2018-08-02
DK2978444T3 (en) 2019-03-18
JP2016518347A (en) 2016-06-23
AU2014242916A1 (en) 2015-11-12
EP2978444B1 (en) 2018-12-05
US20160046950A1 (en) 2016-02-18
CA2908143C (en) 2021-08-31
US10138488B2 (en) 2018-11-27
JP6479761B2 (en) 2019-03-06
EP3524263A1 (en) 2019-08-14
CA2908143A1 (en) 2014-10-02

Similar Documents

Publication Publication Date Title
US20190177733A1 (en) Cancer vaccine for dogs
AU2018260898B2 (en) A cancer vaccine for cats
RU2753246C2 (en) Compositions of vaccines against neoplasia and methods for obtaining thereof
EP1931375B1 (en) Chemo-immunotherapy method
RU2367468C2 (en) Bcl-2 PROTEINS, FRAGMENTS THEREOF, AND APPLICATION THEREOF IN PATIENTS WITH MALIGNANT TUMOUR
CA2934073A1 (en) Combination therapy with neoantigen vaccine
US20190015491A1 (en) Neoepitope rna cancer vaccine
BR112016009002B1 (en) NUCLEIC ACID CONSTRUCTION AND USES OF A NUCLEIC ACID CONSTRUCTION
Thalmensi et al. A DNA telomerase vaccine for canine cancer immunotherapy
US9808504B2 (en) Immunogenic epitopes as targets for universal cancer vaccines
US10898562B1 (en) Immunotherapy against several tumors of the blood, such as acute myeloid leukemia (AML)
EP3359183B1 (en) Polyepitope constructs for use in immunotherapy

Legal Events

Date Code Title Description
AS Assignment

Owner name: INVECTYS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LANGLADE DEMOYEN, PIERRE;WAIN-HOBSON, SIMON;REEL/FRAME:049146/0556

Effective date: 20160704

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: INVECTYS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LIARD, CHRISTELLE;REEL/FRAME:050437/0806

Effective date: 20110315

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION