NZ626124B2 - Vaccines against hpv - Google Patents

Vaccines against hpv Download PDF

Info

Publication number
NZ626124B2
NZ626124B2 NZ626124A NZ62612412A NZ626124B2 NZ 626124 B2 NZ626124 B2 NZ 626124B2 NZ 626124 A NZ626124 A NZ 626124A NZ 62612412 A NZ62612412 A NZ 62612412A NZ 626124 B2 NZ626124 B2 NZ 626124B2
Authority
NZ
New Zealand
Prior art keywords
seq
amino acid
nucleic acid
acid molecule
sequence
Prior art date
Application number
NZ626124A
Other versions
NZ626124A (en
Inventor
Ali Areffard
Ole Henrik Brekke
Agnete Brunsvik Fredriksen
Mona Mari Lindeberg
Original Assignee
Vaccibody As
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 Vaccibody As filed Critical Vaccibody As
Priority claimed from PCT/EP2012/076404 external-priority patent/WO2013092875A1/en
Publication of NZ626124A publication Critical patent/NZ626124A/en
Publication of NZ626124B2 publication Critical patent/NZ626124B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6056Antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/64Medicinal preparations containing antigens or antibodies characterised by the architecture of the carrier-antigen complex, e.g. repetition of carrier-antigen units
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/892Reproductive system [uterus, ovaries, cervix, testes]
    • 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
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39575Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from other living beings excluding bacteria and viruses, e.g. protozoa, fungi, plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • C07K14/523Beta-chemokines, e.g. RANTES, I-309/TCA-3, MIP-1alpha, MIP-1beta/ACT-2/LD78/SCIF, MCP-1/MCAF, MCP-2, MCP-3, LDCF-1, LDCF-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/081Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from DNA viruses
    • C07K16/084Papovaviridae, e.g. papillomavirus, polyomavirus, SV40, BK virus, JC virus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/53Hinge
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/735Fusion polypeptide containing domain for protein-protein interaction containing a domain for self-assembly, e.g. a viral coat protein (includes phage display)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20071Demonstrated in vivo effect
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/22Vectors comprising a coding region that has been codon optimised for expression in a respective host
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof

Abstract

Disclosed is a nucleic acid molecule encoding an amino acid chain comprising (1) a signal peptide, (2) a targeting unit, (3) a dimerization motif, and (4) an antigenic unit, said targeting unit comprising an amino acid sequence having at least 80 % sequence identity to the amino acid sequence 24-93 of SEQ ID NO:1, and an antigenic unit comprising an amino acid sequence of human papillomavirus (HPV) HPV16 and/or HPV18, preferably an antigenic unit derived from early proteins E6 and/or E7 of HPV16 and/or HPV18, which amino acid chain is able to form a homodimeric protein. of SEQ ID NO:1, and an antigenic unit comprising an amino acid sequence of human papillomavirus (HPV) HPV16 and/or HPV18, preferably an antigenic unit derived from early proteins E6 and/or E7 of HPV16 and/or HPV18, which amino acid chain is able to form a homodimeric protein.

Description

VACCIN ES AGAINST H PV FIELD OF THE INVENTION The present invention relates to eutic compounds, such as vaccines against human papi||omavirus (HPV) and in particular to DNA vaccines against HPV16 and/or HPV18. The ion further relates to protein uct encoding homodimeric peptides, which peptides may be released from a DNA vaccine or used separately. Further bed are pharmaceutical formulations, host cells and methods for ing the vaccines, as well as methods for the treatment of various HPV induced diseases, such as cancers and infectious diseases by ation.
BACKGROUND OF THE INVENTION It is now well established that human papi||omavirus (HPV) is the cause of cervical cancer and other HPV—associated malignancies such as anogenital (anus, vulvar, vaginal and penile) cancers and a subset of head and neck cancers. In particular, HPV16 and HPV 18 are sible for about 70% of all cervical cancers ide.
To date, two prophylactic HPV es are on the market (Gardasil and Cervarix). The aim of the prophylactic vaccines is to induce humoral immune responses by stimulating the production of neutralizing antibodies specific for the HPV viral capsid proteins, L1 and L2.
Although the preventive vaccines are an important milestone for the control of HPV induced cervical cancer and possibly other HPV—associated malignancies, the effect of these es will not be significantly observed for 20—40 years (Ma B et al., Current Cancer Therapy Reviews, 2010). er, since the coverage of mass vaccination for the prophylactic vaccines are to date limited in addition to a substantial population worldwide that already are HPV infected, HPV—associated malignancies will continue to progress. Thus, it will be important to develop ecific therapeutic vaccines in order to reduce the mortality and morbidity of HPV—associated malignancies and its precursor lesions (Ma B et al., Current Cancer Therapy Reviews, 2010).
The development of various cancer vaccines and cancer immunotherapy strategies has throughout the last two decades expanded. Still, only one therapeutic cancer vaccine, called Provenge (Dendreon INC) has so far been approved to be applied as standard therapy for prostate cancer. y, due to ethical reasons the majority of therapeutic cancer vaccines are tested on a patient group bearing a late stage tumor. This patient group is substantially immunosuppressed meaning that the tumor cells have for long escaped the immune system and contributed to induce immunological tolerance to the tumor along carcinogenesis. In addition, the choice of antigens (tumor—specific vs. tumor—associated) applied as vaccines are critical in order to induce tumor—specific immune responses and avoid killing of healthy cells in the patients which may lead to serious adverse events. Thus, the major nges in cancer immunotherapy are to break the immunological tolerance and activate tumor—specific effector functions to recognize and kill tumor cells. gh some case s show clinical response to therapeutic cancer vaccines in late stage tumor patients, the most common primary endpoint is to observe the impact on overall survival compared to conventional therapy (surgery, chemo and radiation therapy). However, most s are either not conclusive or that they completely fail to show this. One reason for the negative results lies in the patient group carrying age tumors that are challenging to treat in the first place. A possible gy could be to include patients with early—stage tumors in therapeutic vaccine trials.
One strategy is to target pre—cancerous lesions. The nges for this strategy are mainly the lack of le biomarkers that are specifically expressed by precancerous lesions for many tissues and poor medical screening (either non—existing or that the existing method suffers from lack of sensitivity). Exceptionally, this is not the case for HPV—induced malignancies. For instance, the majority of western countries have good screening programs for cervical sia and cervical cancer by performing the papanicolaou test (Pap smear test). If there are unclear or abnormal results from Pap smear test, colposcopy will be performed (National Cervical Cancer Coalition). HPV—testing may also be recommended for some patients to detect the presence of “high—risk” HPV—type in the precancerous lesion.
Thus, HPV represents a potential biomarker for HPV—associated precancerous s, in particular cervical pithelial dysplasia (CIN).
DNA vaccines have shown increasing promise for the treatment of human diseases, in particular cancer. DNA vaccines induce strong antigen—specific immune responses and can be repeatedly administered to maintain the —specific immune responses. Such vaccines are considered to be safe and simple and cheap to produce on a large scale compared to other cancer therapeutic formats. Numerous immunotherapeutic interventions fail to induce immunological memory. Exceptionally, DNA vaccination ensures sustained release of the e product in vivo which enhances antigen—specific logical . Direct delivery of antigens to professional antigen—presenting cells (APCs) stimulates both CD4+ and CD8+ T cell immune responses in vivo. Such strong cellular immune responses have been demonstrated to specifically recognize and kill n—positive malignant cells efficiently both in vitro and in vivo.
There is still a need in the art for improved es for inducing strong and specific immune responses against HPV responsible for both ious es and cancers.
OBJECT OF THE ION It is an object of embodiments of the invention to provide specific and highly effective therapeutic compounds, such as DNA vaccines against diseases and conditions caused by HPV.
SUMMARY OF THE INVENTION It has been found by the present inventors that by combining the antigens of the early gene products E6 and E7 from HPV, such as from HPV16 and/or HPV18 with the targeting module of hMIP-lCl, therapeutic es are provided, wherein the strong immunogenic epitopes of HPV gene products are presented with high efficiency to APCs to induce a specific and strong immune response. The products according to the present invention is primarily envisioned as therapeutic nucleic acid es, such as DNA vaccines, wherein a nucleic acid uct encoding the vaccibody construct is used as the eutic compound leading to in vivo production of the protein product within the person receiving the vaccine. However, as an alternative the protein product itself may be formulated and used directly in the vaccine.
Accordingly, in a first aspect the present invention relates to a meric protein of two identical amino acid chains, each amino acid chain comprising (1) a signal peptide, (2) a targeting unit, (3) a dimerization motif, and (4) an antigenic unit, said targeting unit comprising an amino acid sequence having at least 80 % sequence identity to the amino acid sequence 24—93 of SEQ ID NO:1, and an antigenic unit comprising an amino acid sequence of human papillomavirus (HPV), such as an antigenic unit comprising an amino acid sequence of HPV16 and/or HPV18, such as an nic unit derived from early proteins E6 and/or E7 of HPV16 and/or HPV18.
In a second aspect the present invention relates to an amino acid chain comprising (1) a signal peptide, (2) a targeting unit, (3) a dimerization motif, and (4) an antigenic unit, said targeting unit comprising an amino acid sequence having at least 80 % ce identity to the amino acid sequence 24—93 of SEQ ID NO:1, and an antigenic unit comprising an amino acid sequence of human papillomavirus (HPV), such as an nic unit comprising an amino acid sequence of HPV16 and/or HPV18, such as an antigenic unit derived from early proteins E6 and/or E7 of HPV16 and/or HPV18, which amino acid chain is able to form a homodimeric n according to the invention. 2012/076404 In a third aspect the present invention relates to a nucleic acid molecule, such as a DNA, encoding an amino acid chain comprising (1) a signal peptide, (2) a targeting unit, (3) a dimerization motif, and (4) an antigenic unit, said targeting unit comprising an amino acid sequence having at least 80 % sequence identity to the amino acid sequence 24—93 of SEQ ID NO:1, and an antigenic unit comprising an amino acid sequence of human papillomavirus (HPV), such as an antigenic unit sing an amino acid sequence of HPV16 and/or HPV18, such as an antigenic unit derived from early proteins E6 and/or E7 of HPV16 and/or HPV18, which amino acid chain is able to form a homodimeric protein according to the invention.
In a further aspect the present invention relates to a homodimeric protein according to the invention, or an amino acid chain according to the invention, or the nucleic acid molecule according to the invention for use as a ment.
In a further aspect the present invention relates to a pharmaceutical composition comprising a homodimeric n according to the invention, or an amino acid chain according to the invention, or the nucleic acid molecule ing to the ion.
In a further aspect the present invention relates to a host cell comprising the nucleic acid molecule according to the invention.
In a further aspect the t invention s to a method for preparing a meric protein according to the invention, or an amino acid chain of the invention, the method comprising a) transfecting the c acid molecule according to the invention into a cell population; b) culturing the cell population; c) collecting and purifying the homodimeric protein, or amino acid chain expressed from the cell population.
In a further aspect the present invention s to a method for preparing a vaccine, such as a DNA vaccine, comprising an immunologically effective amount of a nucleic acid molecule according to the invention, the method comprising a) preparing a nucleic acid molecule according to the ion; b) dissolving the nucleic acid molecule obtained under step a) in a pharmaceutically acceptable carrier, diluent, or buffer.
In a further aspect the present invention relates to a e against HPV comprising an immunologically effective amount of a homodimeric protein according to the invention, or an amino acid chain according to the invention, or nucleic acid molecule, such as a DNA, according to the invention, wherein said vaccine is able to trigger both a T—cell— and B—cell immune response.
In a further aspect the present invention relates to a method of treating or preventing a HPV induced disease or condition, such as a cancer or an infectious e caused by HPV in a patient, the method comprising stering to the patient in need thereof, a homodimeric n according to the invention, or an amino acid chain according to the invention, or the nucleic acid molecule, such as a DNA, according to the invention.
LEGENDS TO THE FIGURE Figure 1: The overall structure of vaccibody vaccines with E7/E6 fusion antigen. Shown are both DNA and protein formats. The vaccibody consist of three functional modules; the chemokine human MIP-lCl (LD7SB) in the targeting module, hinge and CH3 sequences from human IgG3 in the zation module and full—length E7 and/or E6 fusion in the vaccine module.
Figure 2: The ted mode of action for a Vaccibody DNA vaccine against HPV —induced malignancies. Naked DNA plasmid encoding vaccibody is injected intradermal followed by electroporation. The plasmid is taken up by local cells and vaccibody proteins are produced and secreted. The actic targeting s attract CCR1 and CCR5 expressing antigen presenting cells (APC) and ensure binding and uptake into dendritic cells (DC). The DC will present antigenic peptides to CD4+ and CD8+ T cells and the CD8+ T cells will kill HPV infected and transformed cells in the cervix.
Figure 3: T s showing the number of E7 and E6 specific T cell responses as a function of different amounts of vaccine administered. C57BL/6 mice were injected i.d. with naked DNA plasmids encoding VBlOO9 and VBlOl6 and their corresponding controls followed by electroporation (Cellectis, France) on day 0 and day 7. Splenocytes were harvested at day 21 and stimulated with MHC class ricted E7 or E6 peptide for 24h. The number of IFNy secreting splenocytes was calculated by ELISPOT. (A)E7—specific responses after i.d. vaccination with 25pg of VBlOO9, control 1 (antigen alone) and pUMVC4a (empty vector).(B) E7—specific responses after i.d. vaccination with 12.5 and 1.4pg of VBlOl6, l 2 (antigen alone) and a (empty vector). (C) E6—specific responses after i.d. vaccination with 12.5 and 1.4pg of VBlOl6, control 2 (antigen alone) and pUMVC4a (empty vector).
Figure 4.Therapeutic effect of VBlOl6 shown by measured tumor volume. 6 mice were injected s.c. with 5x105 TC—1 cells at day 0. At day 3 and day 10, the mice were injected i.d. with 12.5pg naked DNA plasmids encoding VBlOl6, l 2 or empty vector followed by electroporation (Cellectis, France). The tumor sizes were measured by caliper two to three times a week and tumor volume calculated.
Figure 5.Therapeutic effect of VBlOl6 shown by measured tumor volume. C57BL/6 mice were injected s.c. in the neck area with 5x104 TC—1 cells at day 0. At day 3,7 and day 10, the mice were ed i.d. with 20pg or 2pg naked DNA plasmids encoding VBlOl6, control 2 or empty vector followed by electroporation (Cellectis, France). The tumor sizes were measured by caliper two to three times a week and tumor volume calculated.
Figure 6.Therapeutic effect of VBlOZO and VBlOZl shown by measured tumor volume.
C57BL/6 mice were injected s.c. in the thigh with 5x104 TC—1 cells at day 0. At day 3 and day , the mice were injected i.d. with 10pg naked DNA plasmids encoding VBlOl6, VBlOZO, VBlOZl or empty vector followed by electroporation ctis, France). The tumor sizes were measured by caliper two to three times a week and tumor volume calculated.
DETAILED DISCLOSURE OF THE INVENTION The ucts and DNA vaccine technology described herein by the inventors of the t invention (also referred to as body” molecules/vaccines/constructs) represents a novel vaccine strategy to induce strong and specific immune responses for both infectious diseases and cancer. The HPV E6/E7, such as HPV16 or HPV18 E6/E7 vaccine described herein may be stered as a DNA vaccine by intradermal injection, preferably followed by electroporation. This results in the uptake of the DNA—construct encoding the vaccibody— HPV16 and/or HPV18 E6/E7 e in cells at the site of injection s) including dendritic cells (Langerhans cells), leading to in vivo tion of the vaccibody—E6/E7 molecule.
The early gene products E6 and E7 from “high—risk” HPV types such as HPV16 and 18 may be responsible for transformation of the basal—epithelium cells and induction of precancerous lesions. Both proteins consist of highly immunogenic epitopes and are shown herein to induce strong immune responses g to ic eradication of “high—risk” HPV positive tumor cells both in vitro and in vivo.
The vaccibody molecule described herein is a homodimer consisting of three modules; targeting , dimerization module and the vaccine module (Figure 1). Genes encoding the three modules are genetically engineered to be expressed as one gene. When expressed in vivo, the vaccibody molecule targets antigen presenting cells (APCs) which results in an enhanced vaccine potency compared to identical, non—targeted antigens. In vivo expression of the chemokine human macrophage inflammatory protein 1 alpha (hMIP-lCl/ LD788) leads to attraction of DCs, neutrophils and other immune cells carrying the CCR1 and CCR5 receptors to the site of expression. Thus, the vaccibody molecule consisting of hMIP-lCl as the targeting WO 92875 module, will not only target the antigens to specific cells, but in addition give a response— amplifying effect (adjuvant effect) by recruiting specific immune cells to the injection site. This unique mechanism may be of great importance in a clinical g where patients can receive the vaccine without any additional adjuvants since the vaccine itself gives the adjuvant effect.
The inventors of the present invention describes herein vaccine constructs where the antigenic module consist of the E7 full length genetic sequence in fusion to the E6 full length sequence originating from the HPV16 or HPV18 subtype. The advantage of this format is that both E6 and E7 will be present in one construct and may thus be equally expressed in vivo.
Consequently, one vaccibody molecule consisting of a multi—antigenic unit may represent equal levels of E6 and E7 for the immune system. The HPV16 E6 and E7 gene products are oncogenic in their natural form. To neutralize their oncogenic properties, mutations at specific sites may be introduced in the E6 and E7 genetic sequence.
The mutations, ing ons, may be introduced at specific sites, known to t the oncogenic ties of E6 and E7, such as any one described in any of Dalal S et al., J Virol, 1996; ML'inger K et al., EMBO, 1989; wa S et al., Virology, 1995; Crook Tet al., Cell, 1991; er K et al., HPV Compendium Online, 1997 (http://www.stdgen.lanl.gov/COMPENDIUM_PDF/97PDF/3/E7.pdf); , M et al., J Virol, 2002; Nominé Y et a., Molecular Cell, 2006; Moody C et al., Nat Rev Cancer, 2010, Polakova I et al., Vaccine, 2010; Xie Q, Virologica Sinica, 2011; Mesple‘de T et al., J Virol, 2012; US 102084 and US6306397, which references are hereby incorporated by reference.
Accordingly, in some s of the ion, the constructs according to the present invention contain HPV16 E6, E7 or HPV16 E6/E7 chimeric constructs with one or more ons in either of HPV16 E6, E7 or both at a position known to inhibit the oncogenic properties as described in Dalal S et al., J Virol, 1996; Miinger K et al., EMBO, 1989; Nakagawa S et al., Virology, 1995; Crook T et al., Cell, 1991; Miinger K et al., HPV Compendium Online, 1997 (http://www.stdgen.lanl.gov/COMPENDIUM_PDF/97PDF/3/E7.pdf); Nguyen, M et al., J Virol, 2002; Nominé Y et a., Molecular Cell, 2006; Moody C et al., Nat Rev Cancer, 2010, Polakova I et al., Vaccine, 2010; Xie Q, Virologica Sinica, 2011; Mesple‘de T et al., J Virol, 2012; US 102084 or US6306397. In other aspects of the invention, the constructs according to the present invention contain HPV18 E6, E7 or HPV18 E6/E7 chimeric constructs with one or more mutations in either of HPV18 E6, E7 or both at a position known to inhibit the oncogenic properties as described in Dalal S et al., J Virol, 1996; Miinger K et al., EMBO, 1989; Nakagawa S et al., Virology, 1995; Crook T et al., Cell, 1991; Miinger K et al., HPV Compendium Online, 1997 (http://www.stdgen.lanl.gov/COMPENDIUM_PDF/97PDF/3/E7.pdf); Moody C et al., Nat Rev Cancer, 2010, US 2008/0102084 and US6306397.
WO 92875 There is a possibility that the vaccibody—moiety (targeting and dimerization modules) may eradicate the oncogenic properties of E6 and E7 wildtype proteins in the final fusion protein.
Thus, in yet another aspect of the invention is the utilization of the wildtype full—length E6 and/or E7 sequences in the ody construction.
The invention describes several variant of Vaccibody HPV therapeutic DNA vaccines all based on the overall format described in figure 1, the eutic vaccibody—HPV DNA vaccines encodes genes that are naturally expressed in humans; the targeting module genes encode the chemokine hMIP—ld, which binds to its cognate receptors, CCR1 and CCR5 sed on the cell e of APCs. The zation module genes may encode hinge regions and constant heavy chain 3, such as from human IgG3 which connects two vaccibody monomers generating a homodimer molecule. Genes ng the vaccine module for the current strategy consist of HPV, such as HPV16 and/or HPV18 E7 and E6 antigens, such as the full length HPV16 E7 and E6 ns, optionally comprising one or more mutation to inhibit the nic properties. Once administered in vivo by i.d. injection followed by electroporation, dermal cells taking up the vaccine construct will express the vaccibody—HPV molecule. The in vivo produced vaccibody es target to CCR1 and CCR5 expressed on the surface of APCs in the skin, in particular DCs. The binding of the vaccibody molecule to its cognate ors leads to internalization of the complex in the APC, degradation of the proteins into small peptides that are loaded onto MHC molecules and presented to CD4+ and CD8+ T cells to induce HPV16 E6 and E7 specific immune responses. Once stimulated and with help from ted CD4+ T cells, CD8+ T cells will target and kill HPV16 E6 and E7 sing cells (Figure 2). Such enhanced immune responses to a vaccine with a “built—in” adjuvant effect may ially overcome tumor—escape (tumor immune surveillance) by breaking immunological tolerance and efficiently kill malignant cells. The hMIP—ld targeting unit may be connected through a dimerization motif, such as a hinge , to an antigenic unit, wherein the later is in either the COOH—terminal or the NH2—terminal end. The t invention not only relates to a DNA sequence coding for this recombinant protein, but also to expression vectors comprising these DNA sequences, cell lines comprising said expression vectors, to treatment of mammals preferentially by immunization by means of Vaccibody DNA, Vaccibody RNA, or Vaccibody protein, and finally to pharmaceuticals and a kit comprising the said molecules.
The dimerization motif in the proteins according to the present invention may be constructed to include a hinge region and an immunoglobulin domain (e.g. CV3 domain), e.g. carboxyterminal C domain (CH3 domain), or a sequence that is substantially identical to said C domain. The hinge region may be Ig derived and contributes to the dimerization through the formation of an interchain covalent bond(s), e.g. disulfide bridge(s). In addition, it functions as a flexible spacer between the domains allowing the two targeting units to bind aneously to two target molecules on APC expressed with variable distances. The immunoglobulin domains contribute to homodimerization through non—covalent interactions, e.g. hydrophobic interactions. In a preferred embodiment the CH3 domain is derived from IgG. These dimerization motifs may be exchanged with other multimerization moieties (e.g. from other Ig isotypes/subclasses). Preferably the dimerization motif is derived from native human proteins, such as human IgG.
It is to be understood that the dimerization motif may have any orientation with t to nic unit and ing unit. In one embodiment the antigenic unit is in the COOH— terminal end of the dimerization motif with the targeting unit in the N—terminal end of the dimerization motif. In another embodiment the antigenic unit is in the N—terminal end of the dimerization motif with the targeting unit in the COOH—terminal end of the dimerization motif.
International application , which is hereby incorporated by nce discloses nucleic acid sequences and vectors, which may be used ing to the present invention.
The proteins according to the present ion include an antigenic unit derived from HPV, such as HPV16 E7 and E6 antigens, such as the full length HPV16 E7 and E6 antigens, as well as immunogenic fragments or variants thereof. The antigenic sequence should be of sufficient length. The minimal length of such nic unit may be around 9 amino acids. Accordingly in some embodiments, the antigenic unit derived from HPV comprises an amino acid sequence of at least 9 amino acids corresponding to at least about 27 tides in a nucleic acids sequence encoding such antigenic unit. Preferably the antigenic unit derived from HPV is considerably longer, such as the full length HPV16 E7 and E6 antigens. Diversity arises within a given HPV pe through limited nucleotide changes in the coding (at a frequency of <2%) and non—coding (at a frequency of <5%) regions (Bernard, HU et al., IntJ Cancer, 2006). Such variants phylogenetically segregate based on their geographical origin and are therefore labeled European, African, Asian, Asian—American and North American. Insertion of such sequences in a Vaccibody format might lead to activation of both arms of the immune response.
Immunization by means of Vaccibody protein, Vaccibody DNA, or Vaccibody RNA, the latter two executed e.g. by intramuscular or intradermal injection with or without a following electroporation, are all le methods according to the present ion.
As discussed above, the present invention relates to a e composition against cancer or infectious diseases caused by HPV, the vaccine composition comprising an logically ive amount of the nucleic acid encoding the molecule of the invention or degenerate variants thereof. The vaccine may be able to trigger both a T—cell— and B—cell immune response. The present invention also relates to a kit comprising Vaccibody DNA, RNA, or protein for diagnostic, medical or scientific purposes.
The invention r relates to a method of preparing the inant molecule of the invention comprising, transfecting the vector comprising the molecule of the invention into a cell population; culturing the cell population; collecting recombinant protein expressed from the cell population; and purifying the expressed protein.
The above bed nucleotide sequences may be inserted into a vector suited for gene therapy, e.g. under the control of a specific promoter, and introduced into the cells. In some embodiments the vector comprising said DNA sequence is a virus, e.g. an adenovirus, vaccinia virus or an adeno—associated virus. In some embodiments a retroviruses is used as vector.
Examples of suitable retroviruses are e.g. MoMuLV or . For the purpose of gene therapy, the DNA/RNA sequences according to the invention can also be orted to the target cells in the form of colloidal dispersions. They comprise e.g. liposomes or lipoplexes.
The present invention encompasses the use of a targeting unit as well as an antigenic unit having minimum degree of sequence identity or sequence homology with amino acid ce(s) defined herein or with a polypeptide having the specific properties defined herein. The present invention encompasses, in particular, the use of peptide variants or e units to be used in the constructs according to the present invention having a degree of ce identity with any one of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:32, or SEQ ID NO:34. Here, the term “variant” means an entity having a certain degree of sequence identity with the subject amino acid sequences or the subject nucleotide sequences, where the subject amino acid sequence preferably is SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, or SEQ ID NO:34.
In one aspect, the variant or nt amino acid ce and/or nucleotide sequence should provide and/or encode a polypeptide which retains the functional activity and/or es the activity of a polypeptide of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:32, or SEQ ID NO:34.
In the present context, a variant sequence is taken to include an amino acid sequence which may be at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99%, identical to the subject sequence. Typically, the variants used according to the present invention will comprise the same active sites etc. as the subject amino acid sequence. Although homology can also be considered in terms of similarity (i.e. amino acid residues having similar chemical properties/functions), in the context of the present invention it is red to express homology in terms of sequence identity.
Sequence identity comparisons can be conducted by eye, or more y, with the aid of readily ble ce comparison computer programs. These commercially ble computer programs use complex comparison algorithms to align two or more sequences that best reflect the ionary events that might have led to the difference(s) between the two or more sequences. ore, these algorithms operate with a scoring system rewarding alignment of identical or similar amino acids and penalising the insertion of gaps, gap extensions and alignment of non—similar amino acids. The scoring system of the comparison algorithms include: i) assignment of a penalty score each time a gap is inserted (gap penalty score), ii) assignment of a penalty score each time an ng gap is extended with an extra position (extension penalty , iii) assignment of high scores upon alignment of identical amino acids, and iv) assignment of variable scores upon alignment of non—identical amino acids.
Most alignment programs allow the gap penalties to be modified. However, it is preferred to use the default values when using such software for sequence isons.
The scores given for ent of non—identical amino acids are assigned according to a scoring matrix also called a substitution matrix. The scores ed in such substitution matrices are reflecting the fact that the likelihood of one amino acid being substituted with another during evolution varies and depends on the physical/chemical nature of the amino acid to be substituted. For example, the hood of a polar amino acid being substituted with another polar amino acid is higher compared to being substituted with a hydrophobic amino acid. Therefore, the scoring matrix will assign the highest score for identical amino acids, lower score for non—identical but similar amino acids and even lower score for non— identical non—similar amino acids. The most frequently used scoring es are the PAM matrices (Dayhoff et al. (1978), Jones et al. ), the BLOSUM matrices (Henikoff and Henikoff (1992)) and the Gonnet matrix t et al. (1992)).
Suitable computer programs for carrying out such an alignment include, but are not limited to, Vector NTI (Invitrogen Corp.) and the ClustalV, ClustalW and ClustalW2 programs (Higgins DG & Sharp PM (1988), s et al. (1992), Thompson et al. (1994), Larkin et al. (2007). A selection of different alignment tools is available from the ExPASy Proteomics server at www.expasy.org. Another example of software that can perform sequence alignment is BLAST (Basic Local Alignment Search Tool), which is available from the webpage of National Center for Biotechnology Information which can currently be found at http://www.ncbi.n|m.nih.gov/ and which was firstly described in Altschul et al. (1990) J. Mol.
Biol. 215; 403—410.
Once the software has produced an alignment, it is possible to calculate % similarity and % sequence identity. The software typically does this as part of the sequence comparison and generates a numerical .
In one embodiment, it is preferred to use the ClustalW software for performing sequence ents. Preferably, alignment with lW is med with the following parameters for pairwise alignment: Substitution matrix: Gonnet 250 Ga- o-en -enaIt : Ga- extension -ena|t : Ga- end -ena|t : ClustalW2 is for example made available on the internet by the European ormatics Institute at the EMBL—EBI webpage www.ebi.ac.uk under tools — sequence analysis — ClustalW2. Currently, the exact address of the ClustalW2 tool is www.ebi.ac.uk/Tools/clustalw2.
In r embodiment, it is preferred to use the program Align X in Vector NTI rogen) for performing sequence alignments. In one embodiment, Exp10 has been may be used with default settings: Gap opening penalty: 10 Gap extension penalty: 0.05 Gapseparation y range: 8 Score matrix: blosum62mt2 Thus, the present ion also encompasses the use of variants, fragments, and derivatives of any amino acid sequence of a protein, polypeptide, motif or domain as defined herein, ularly those of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:32, or SEQ ID NO:34.
The sequences, particularly those of variants, fragments, and derivatives of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:32, or SEQ ID NO:34, may also have deletions, insertions or substitutions of amino acid residues which produce a silent change and result in a functionally equivalent substance. Deliberate amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues as long as the secondary binding activity of the substance is retained. For example, negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; and amino acids with uncharged polar head groups having similar hydrophilicity values include leucine, cine, valine, e, alanine, asparagine, glutamine, serine, threonine, phenylalanine, and tyrosine.
The present invention also encompasses conservative substitution (substitution and ement are both used herein to mean the interchange of an existing amino acid e, with an alternative residue) that may occur i.e. or—like substitution such as basic for basic, acidic for acidic, polar for polar etc. Non—conservative substitution may also occur i.e. from one class of residue to another or alternatively involving the ion of unnatural amino acids such as ornithine nafter referred to as Z), obutyric acid ornithine nafter referred to as B), norleucine ornithine (hereinafter referred to as O), pyriylalanine, thienylalanine, ylalanine and phenylglycine.
Conservative substitutions that may be made are, for example within the groups of basic amino acids (Arginine, Lysine and Histidine), acidic amino acids (glutamic acid and aspartic acid), aliphatic amino acids (Alanine, Valine, Leucine, Isoleucine), polar amino acids (Glutamine, Asparagine, Serine, Threonine), aromatic amino acids (Phenylalanine, Tryptophan and Tyrosine), hydroxyl amino acids (Serine, Threonine), large amino acids (Phenylalanine and Tryptophan) and small amino acids (Glycine, Alanine). 2012/076404 Replacements may also be made by unnatural amino acids include; alpha* and alpha— disubstituted* amino acids, N—alkyl amino acids*, lactic acid*, halide derivatives of natural amino acids such as trifluorotyrosine*, p—Cl—phenylalanine*, p—Br—phenylalanine*, p—I— phenylalanine*, L—allyl—glycine*, B—alanine*, L—oc—amino butyric acid*, L—y—amino butyric acid*, L—oc—amino isobutyric acid*, L—e—amino caproic acid#, 7—amino heptanoic acid*, L— nine sulfone‘“, eucine*, L—norvaline*, p—nitro—L—phenylalanine*, L— hydroxyproline#, L—thioproline*, methyl derivatives of phenylalanine (Phe) such as 4—methyl— Phe*, pentamethyl—Phe*, L—Phe (4—amino)#, L—Tyr (methyl)*, L—Phe (4—isopropyl)*, L—Tic (1,2,3,4—tetrahydroisoquinoline—3—carboxyl acid)*, L—diaminopropionic acid # and L—Phe (4— benzyl)*. The notation * has been utilised for the purpose of the sion above ing to homologous or non—conservative substitution), to indicate the hydrophobic nature of the derivative whereas # has been utilised to indicate the hydrophilic nature of the derivative, #* indicates am phipathic characteristics.
Variant amino acid ces may e suitable spacer groups that may be ed between any two amino acid residues of the sequence including alkyl groups such as methyl, ethyl or propyl groups in addition to amino acid spacers such as glycine or B—alanine es.
A further form of variation, involves the presence of one or more amino acid residues in peptoid form, will be well understood by those skilled in the art. For the avoidance of doubt, “the peptoid form" is used to refer to variant amino acid residues wherein the oc—carbon substituent group is on the residue’s nitrogen atom rather than the oc—carbon. Processes for preparing peptides in the peptoid form are known in the art, for example Simon R] et al. (1992), Horwe|| DC. (1995).
In one embodiment, the variant targeting unit used in the homodimeric protein according to the t invention is variant having the sequence of amino acids at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% amino acid sequence identity therewith.
In one aspect, preferably the protein or sequence used in the present invention is in a purified form. The term “purified” means that a given component is present at a high level.
The component is desirably the predominant active component present in a composition.
A “variant” or “variants” refers to proteins, polypeptides, units, , domains or c acids. The term “variant” may be used interchangeably with the term t.” Variants include insertions, substitutions, transversions, truncations, and/or inversions at one or more locations in the amino acid or nucleotide ce, respectively. The phrases “variant polypeptide variant" and “variant enzyme" mean a polypeptide/protein that , polypeptide , has an amino acid sequence that has been modified from the amino acid sequence of SEQ ID NO: 1. The t polypeptides include a polypeptide having a certain percent, e.g., 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%, of sequence identity with the amino acid sequence of SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:32, or SEQ ID NO:34.
“Variant nucleic acids" can include sequences that are complementary to sequences that are capable of izing to the nucleotide ces ted herein. For example, a variant sequence is complementary to sequences capable of hybridizing under stringent conditions, e.g., 50°C and 0.2X SSC (1X SSC = 0.15 M NaCl, 0.015 M sodium citrate, pH 7.0), to the nucleotide sequences presented herein. More particularly, the term variant encompasses ces that are mentary to sequences that are capable of hybridizing under highly stringent conditions, e.g., 65°C and 0.1X SSC, to the nucleotide sequences presented herein.
The melting point (Tm) of a variant nucleic acid may be about 1, 2, or 30C lower than the Tm of the wild—type nucleic acid. The variant nucleic acids include a cleotide having a certain percent, e.g., 80%, 85%, 90%, 95%, or 99%, of sequence identity with the nucleic acid encoding SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:32, or SEQ ID NO:34, encoding the ric protein which can form the homodimeric protein according to invention.
A specific category of mutations are the mutations in E6 and E7: The E6 protein may be detoxified by rendering the p53 binding impossible. Five positions in the full length HPV16 E6 protein are sites for mutations for inactivation of E6 functionality, F47, L50, C63, C106 and 1128. Any amino acid substitution in these positions may lead to inactivation of E6 and induces tumor suppression. Substitutions in any one of these positions with any one different amino acid may potentially be utilized. Sites for potential mutations are shown in SEQ ID NO:22.
In the E7 protein there are conserved regions associated with oncogenic properties (see Phelps et al J. Virol. April 1992, vol. 66, no. 242; Gulliver et al J Virol. 1997, ; 71(8)) including an N—terminal Rb (retinoblastoma binding protein) binding—site motif (LXCXE) and two conserved regions 3 (upstream and downstream) with a Zn— binding motif . The preferred mutation sites in the motif are C24 and E26. Preferred sites in the two CXXC motifs are C58, C61, C91 and C94. However, any mutations in these regions can be envisaged to be substituted for the reduction of binding ons and thus abolish the oncogenic effects of E7. Sites for potential mutations are shown in SEQ ID NO:23.
Signal peptide: A signal e at the N—terminal end of the nascent polypeptide directs the molecule into the ER before transport to into the Golgi complex. The signal peptide is cleaved off by signal peptidase once it has served its purpose of targeting and importing the protein to the ER.
These signal peptides are generally between 15 and 30 amino acids, but can have more than 50 residues (Martoglio, B. et al., Trends in Cell y, 1998, Knappskog, S. et al., J Biotechnol, 2007). The native signal peptide may be replaced by signal peptides from any mammalian, prokaryotic or marine origin. Commonly used signal peptides are e.g. humanIL— 2 and human albumin due to their natural ability to secrete large amounts of protein. The choice of signal e can have a considerable impact on the amount of sized and secreted protein.
In some ments, the signal peptide used in the protein construct according to the present invention is d from a chemokine protein, such as the signal sequence of LD78beta.
In some embodiments the signal peptide is not derived from pLNOH2 (Bl—8 variable immunoglobulin leader) disclosed in the international application with International Application No: .
In some embodiments the signal peptide is not derived from an immunoglobulin gene.
The term “homodimeric protein" as used herein refers to a protein comprising two individual identical strands of amino acids, or subunits held together as a single, dimeric protein by hydrogen bonding, ionic (charged) interactions, actual covalent disulfide bonding, or some combination of these interactions.
The term “dimerization motif", as used herein, refers to the sequence of amino acids between the antigenic unit and the targeting unit comprising the hinge region and the optional second domain that may bute to the dimerization. This second domain may be an immunoglobulin domain, and optionally the hinge region and the second domain are connected through a . ingly the dimerization motif serves to t the antigenic unit and the targeting unit, but also contain the hinge region that facilitates the dimerization of the two monomeric proteins into a homodimeric protein ing to the invention.
The term “targeting unit" as used herein refers to a unit that delivers the protein with its antigen to mouse or human APC for MHC class II—restricted presentation to CD4+ T cells or for providing cross presentation to CD8+ T cells by MHC class I restriction. The targeting unit used in the constructs according to the present invention is derived from or identical to mature LD78—beta.
The term “antigenic unit" as used herein refers to any molecule, such as a e which is able to be specifically recognized by an antibody or other component of the immune system, such as a surface receptor on T—cells. Included within this tion are also immunogens that are able to induce an immune response. The terms “epitope” or “antigenic epitope" is used to refer to a distinct molecular surface, such as a lar surface provided by a short peptide sequence within an antigenic unit. In some embodiments the antigenic unit comprises two ore more antigenic epitopes. The antigenic unit used in the constructs according to the present invention is derived from or cal to the early gene products E6 and E7 from HPV, such as from HPV16 or HPV18.
The term “hinge region" refers to a peptide sequence of the homodimeric protein that facilitates the dimerization, such as through the formation of an interchain covalent bond(s), e.g. disulfide bridge(s). The hinge region may be Ig d, such as hinge exons h1+h4 of an Ig, such as IgG3.
Specific embodiments of the invention: As described above, the present invention relates to a homodimeric protein of two identical amino acid chains, each amino acid chain comprising (1) a signal peptide, (2) a targeting unit, (3) a dimerization motif, and (4) an nic unit, said targeting unit comprising an amino acid sequence having at least 80 % sequence identity to the amino acid sequence 24— 93 of SEQ ID NO:1, and an antigenic unit sing an amino acid sequence of human papillomavirus (HPV), such as an antigenic unit sing an amino acid sequence of HPV16 and/or HPV18, such as an antigenic unit derived from early proteins E6 and/or E7 of HPV16 and/or HPV18. In some embodiments according to the present invention, the targeting unit, dimerization motif and antigenic unit in the amino acid chain are in the N—terminal to C— al order of targeting unit, dimerization motif and antigenic unit.
In some embodiments, the antigenic unit used in the constructs according to the t invention is derived from HPV16, such as from early proteins E6 and/or E7.
In some embodiments, the antigenic unit used in the constructs according to the present invention is derived from E6 of HPV16.
In some embodiments, the antigenic unit used in the constructs according to the present invention is derived from E7 of HPV16.
In some embodiments, the antigenic unit used in the constructs according to the present invention is derived from HPV18, such as from early proteins E6 and/or E7.
In some embodiments, the antigenic unit used in the constructs according to the present invention is derived from E6 of HPV18.
In some embodiments, the antigenic unit used in the constructs according to the t ion is derived from E7 of HPV18.
In some ments ing to the present invention, the signal peptide consists of an amino acid sequence having at least 80 % sequence identity to the amino acid sequence 1— 23 of SEQ ID NO:1.
In some embodiments according to the present invention, the signal peptide consists of an amino acid sequence having at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99%, such as 100% sequence ty to the amino acid sequence 1—23 of SEQ ID NO:1.
In some embodiments according to the present invention, the targeting unit consists of an amino acid sequence having at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence identity to the amino acid ce 24—93 of SEQ ID NO:1.
In some embodiments according to the present invention, the dimerization motif comprises a hinge region and optionally another domain that facilitate dimerization, such as an globulin domain, optionally connected through a linker.
In some embodiments according to the present invention, the hinge region is Ig derived, such as derived from IgG3.
In some ments according to the t invention, the hinge region has the ability to form one, two, or several covalent bonds. In some embodiments ing to the present invention, the covalent bond is a disulphide bridge.
In some embodiments ing to the present invention, the immunoglobulin domain of the dimerization motif is a carboxyterminal C domain, or a sequence that is substantially identical to the C domain or a variant thereof.
In some embodiments according to the t invention, the carboxyterminal C domain is derived from IgG.
In some embodiments according to the present invention, the immunoglobulin domain of the dimerization motif has the ability to homodimerize.
In some embodiments ing to the present invention, the immunoglobulin domain has the ability to homodimerize via noncovalent interactions. In some embodiments according to the present invention, the noncovalent interactions are hydrophobic interactions.
In some embodiments according to the present invention, the dimerization domain does not se the CH2 domain.
In some embodiments according to the present invention, the zation motif consists of hinge exons hi and h4 connected through a linker to a CH3 domain of human IgG3.
In some embodiments according to the present invention, the dimerization motif consist of an amino acid sequence having at least 80 % sequence ty to the amino acid sequence 94-237 of SEQ ID NO:3.
In some embodiments according to the present invention, the linker is a G3SZG3SG linker.
In some embodiments according to the present invention, the antigenic unit and the dimerization motif is ted through a linker, such as a GLGGL linker or a GLSGL linker.
In some embodiments according to the present invention, the targeting unit consists of amino acids 24—93 of SEQ ID NO:1, or a t thereof.
In some embodiments according to the present invention, the homodimeric protein have increased affinity for any one chemokine receptor ed from CCR1, CCR3 and CCR5 as compared to the affinity of the same homodimeric protein with the targeting unit consisting of amino acids 24—93 of SEQ ID NO:1, or a variant f.
In some embodiments according to the t ion, the antigenic unit comprises an amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence identity to the amino acid sequence 243—293 of SEQ ID NO:3.
In some embodiments according to the present invention, the antigenic unit consists of an amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence identity to the amino acid sequence 243—293 of SEQ ID NO:3.
In some embodiments ing to the present invention, the antigenic unit ses one or more amino acid substitutions at a position selected from the list consisting of F47, L50, C63, C106 and 1128 of SEQ ID NO:22, or a deletion involving one or more amino acid selected from the list consisting of Y43—L50 of SEQ ID NO:22.
In some embodiments ing to the present ion, the antigenic unit comprises not more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18 or 20 amino acid substitutions and/or deletions relative to SEQ ID NO:22.
In some embodiments according to the present ion, the antigenic unit comprises the amino acid sequence 243—293 of SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, or SEQ ID NO:9, or a variant or antigenic fragment thereof.
In some embodiments according to the present invention, the antigenic unit consists of the amino acid sequence 243—293 of SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, or SEQ ID NO:9, or a variant or antigenic fragment thereof. 2012/076404 In some embodiments according to the present invention, the antigenic unit comprises an amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence identity to the amino acid sequence 243—340 of SEQ ID NO:11.
In some ments according to the present invention, the antigenic unit consists of an amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence ty to the amino acid sequence 243—340 of SEQ ID NO:11.
In some embodiments ing to the present invention, the antigenic unit comprises one or more amino acid substitutions at a position selected from the list consisting of C24, E26, C58, C61, C91, and C94 of SEQ ID NO:23, or a deletion involving one or more amino acid selected from the list consisting of L22—E26 and/or C58—C61 and/or C91—S95 of SEQ ID NO:23.
In some embodiments according to the present invention, the antigenic unit ses not more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18 or 20 amino acid substitutions and/or deletions relative to SEQ ID NO:23.
In some embodiments according to the present invention, the antigenic unit comprises the amino acid sequence 0 of SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, or SEQ ID NO:17, or a variant or antigenic fragment thereof.
In some embodiments according to the present invention, the antigenic unit consists of the amino acid sequence 243—340 of SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, or SEQ ID NO:17, or a variant or antigenic fragment thereof.
In some embodiments according to the t invention, the antigenic unit comprises an amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence identity to the amino acid sequence 243—501 of SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:32, or SEQ ID NO:34.
In some embodiments ing to the present invention, the antigenic unit consists of an amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence identity to the amino acid sequence 243—501 of SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:32, or SEQ ID NO:34.
In some embodiments according to the present invention, the antigenic unit comprising an amino acid sequence of human omavirus 16 (HPV16) derived from both early proteins E6 and E7.
In some embodiments according to the t invention, the antigenic unit comprising an amino acid sequence of human papi||omavirus 18 ) derived from both early proteins E6 and E7.
In some embodiments according to the present invention, the antigenic unit comprises one or more amino acid tutions at a position selected from the list consisting of F47, LSOG, C63, C106,1128T of SEQ ID NO:22 and C24, E26, C58, C61, C91, C94 of SEQ ID NO:23.
In some embodiments ing to the present invention, the antigenic unit comprises not more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18 or 20 amino acid substitutions and/or deletions relative to SEQ ID NO:22 and SEQ ID NO:23.
In some embodiments according to the present invention, the antigenic unit ts of the amino acid sequence 243—501 of SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:32, or SEQ ID NO:34, or a t or antigenic fragment thereof.
In some embodiments according to the present invention, the amino acid chain consists of an amino acid sequence selected from the list consisting of SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:32, and SEQ ID NO:34, or a variant or antigenic fragment thereof.
In some ments according to the present invention, the nic unit comprises an amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence identity to any one amino acid sequence ed from SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, and SEQ ID NO:25.
In some embodiments ing to the present invention, the antigenic unit consist of an amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99% sequence identity to any one amino acid sequence selected from SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, and SEQ ID NO:25.
In some embodiments the homodimeric protein according to the present invention, is in its mature form without any signal peptide sequence.
In some embodiments the nucleic acid molecule according to the present invention is human codon optimized.
It is to be understood that a human codon optimized nucleic acid molecule according to the present invention comprises one or more nucleic acid substitution as compared to the wild type sequence, which substitution provides for a codon with higher frequency of usage in human coding regions. ncy of codon usage in homo sapiens can be found at http ://biowiki.edu—wiki.org/en/codon_table In some ments the nucleic acid molecule according to the t invention is comprising any one of nucleotide sequences selected from the list consisting of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO:10, SEQ ID NO:12, SEQ ID NO:14, SEQ ID NO:16, SEQ ID NO:18, SEQ ID NO:20, SEQ ID NO:31 and SEQ ID NO:33, or a variant thereof.
In some embodiments the nucleic acid molecule ing to the present invention is comprised by a vector.
In some ments the nucleic acid molecule according to the present invention is formulated for administration to a patient to induce tion of the homodimeric n in said patient.
In some embodiments the vaccine according to the present ion further comprises a pharmaceutically acceptable carrier and/or adjuvant.
In some ments, the method of treating or preventing a HPV induced disease or condition, such as a cancer or an infectious disease caused by HPV in a patient according to the present invention comprises administering to the patient in need thereof of a nucleic acid molecule, such as a DNA, ing to the t invention with a subsequent step of electroporation. In some embodiments the administration is performed intra dermal or intra muscular.
EXAMPLE 1 Construction and expression of the vaccines.
Gene sequences were designed according to the following structure: 1: native leader sequence for human LD78 b, 2: full length LD78b sequence. 3: Human hinge—region 1 from IgG3. 4: Human hinge region 4 from IgG3. 5: Glycine— Serine linker. 6: Human CH3 domain from IgG3. 7: Glycine—Leucine linker. 8: wildtype and mutant Human papilloma virus oncogenes E6, E7 and fusion proteins of both E6 and E7 divided by a Glycine— Serine linker.
The constructs are designated according to their E6 and or E7 ition as follows: VBlOOl: Vaccibody—E6 wild type; VBlOOS: Vaccibody—E7 wild type; The mutants are designated according to the amino acid position in the corresponding native E6 or E7 sequence.
VBlOOZ: Vaccibody—E6 C63R; VBlOO3: ody—E6 C106R; v31004: Vaccibody—E6 F47R, C63R, C106R; VBlOO6: ody—E7 C24G, E26G; VBlOO7: Vaccibody—E7 C24G, E26G, C58G, C61G; VBlOOS: Vaccibody—E7 C24G, E26G, C91G, C94G; v31009: Vaccibody— E7 C24G, E26G/ E6 F47R, C63R, C106R; VBlOl6: Vaccibody— E7 C24G, E26G/ E6 C63R, C106R; VBlOZO: Vaccibody— E7 C24G, E26G/ E6 F47R, C63R, C106R human codon optimized VBlOZl: Vaccibody— E7 C24G, E26G/ E6 F47R, LSOG, C106R, 1128T human codon optimized l vaccines composed of only the antigens were included: Control 1: E7 C24G, E26G/ E6 F47R, C63R, C106R; Control 2: E7 C24G, E26G/ E6 C63R, C106R All gene sequences were ordered from Aldevron (Fargo ND, USA) or ns MWG GmbH and cloned into the expression vector pUMVC4a.
All constructs were transfected in to 293E cells and verified expression of intact vaccibody proteins were performed by dot blot and ELISA (data not shown). All amino acid sequences except for Controls 1 and 2 are shown as SEQ IDs.
EXAMPLE 2.
Immune response s VB 1009,VBlOl6, VBlOZO and VBlOZl were selected as vaccine candidates with their corresponding controls 1 and 2 respectively. As a negative control empty pUMVC4a vector was ed. , 12.5 and 1.4 pg plasmid DNA of each candidate was injected intradermal in the lower back of C57Bl/6 mice followed by electroporation, Dermavax, Cellectis , France). 7 days later the mice were boosted with similar amounts of vaccines and control plasmids. At day 21 the mice were killed and s were harvested.
The T cell responses were calculated by ELISPOT. (Figures 3 a, b and c) EXAMPLE 3. eutic effect VBlOl6, VBlOZO and VBlOZl with the corresponding controls 1 and 2 were selected as the e candidate for therapeutic vaccine studies. 5x104 or 5x105 TC—1 cells (Johns Hopkins University, Baltimore, USA, Lin KY et a/., Cancer Res, 1996) were injected in the neck or thigh region of C57Bl/6 mice. After days 3 and 10 or day 3,7 and 10, the mice were vaccinated with 2pg, 10pg, 12.5 pg or 20pg of plasmid DNA followed by electroporation, Dermavax, Cellectis France. Tumor size were measured two to three times a week up until day 49 after TC—1 cell injection (Figure 4, 5 and 6) EXAMPLE 4.
A therapeutic DNA vaccine to be used may be prepared by GMP manufacturing of the plasmid vaccine according to regulatory authorities’ guidelines, including GMP cell banking, GMP cturing of drug substance and drug product, ICH stability studies and Fill & Finish of the DNA vaccine. The DNA vaccine may be formulated by dissolving in a saline solution, such as 10nM Tris, 1mM EDTA at a concentration of 2—5 mg/ml. The vaccine may be administered either intra—dermal or muscular with or without following oporation.
SEQUENCES: C—C motif chemokine 3—like 1 precursor including signal peptide (aa 1—23 in bold) and mature peptide (LD78—beta), aa 24—93 (SEQ ID NO:1): MQVSTAALAVLLCTMALCNQVLSAPLAADTPTACCFSYTSRQIPQN FIADYFETSSQCSKPSVIFLTKR G RQVCADPSEEWVQKYVSD LELSA The specific DNA and corresponding amino acid ces of vaccibody HPV constructs: E6 g E7 single constructs: For the purpose of illustration only, the different s of the constructs are separated by an |"with the domains in the ing order: Signal peptide | human MIP-lCl | Hinge h1 | Hinge h4 | Gly—Ser Linker or Gly—Leu | hCH3 IgG3 |Gly—Ser Linker or Gly—Leu linker| wildtype or mutant full length E6 g E7. Amino acids or nucleotides in bold illustrates sites of mutations.
DNA sequence of VBlOOl (SEQ ID NO:2): ATGCAGGTCTCCAcr‘Gcr‘GCCCTTGCCG"CCTCCTCr‘GCACCATGGCTC"CTGCAACCAGGF‘CCF‘CTC" | C'T GCTGCTGACACGCCGACCGCCTGC"GCT"CAGC"ACACC"CCCGACAGA'TCCACAGAAT'T‘CA"AGC"GACTACT""G AGACGAGCAGCCAG'CC"CCAAGCCCAG"GTCA"C""CC"AACCAAGAGAGGCCGGCAGG"CTG"GC"GACCCCAG"GA GGTCCAGAAAF‘ACGTCAGF‘GACCTGGAGCF‘GAGF‘GCC | GAGCTCAAAACCCCAC'T‘GG'CACACAAC"CACAC A | GAGCCCAAA"CTr‘Gr‘GACACACCTCCCCCG"GCCCAAGG"GCCCA | GGCGGTGGAAGCAGCGGAGG"GGAAGTGGA | GGACAGCCCCGAGAACCACAGG"G"ACACCC"GCcccCA"CCCGGGAGGAGATGACCAAGAACCAGG"CAGCC"GACCT GCCTGG"CAAAGGC""CTACCCCAGCGACATCGCCGTGGAG"GGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCC"CCCA"GC"GGAC"CCGACGGCTCCW“C""CCTCTACAGCAAGCTCACCGr‘GGACAAGAGCAGGr‘GGCAGCAG GGGAACATCTF‘C"CAF‘GCF‘CCGF‘GAF‘GCATGAGGC"‘CF‘GCACAACCGCF‘TCACGCAGAAGAGCCF‘CTCCC"GTC"CCGG G"AAA| GGCCTCGGTGGCU‘G AF‘GF‘TF‘CAGGACCCACAGGAGCGACCCAGAAAG""ACCACAGF‘TAF‘GCACAGAGCF‘G CAAACAAC"A"ACA"GA"A"AA"A""AGAA"G"G"G"ACTGCAAGCAACAGT"AC"GCGACG"GAGG"A"A"GAC"T“"G CW...“CGGGAFTHAHGCA..AGHAHAHAGAGAHGGGAAHCCA..A..GC“GHAHGHGAHAAAHGHHTAAAGTHWAW.CHAA AAF‘F‘AGTGAGF‘AF‘AGACAF‘F‘A""G""‘AF‘AGF‘""‘GF‘AF‘GGAACAACA"TAGAACAGCAATACAACAAACCGF‘F‘GF‘GF‘GA" ""G""AA""AGG"G"A""AAC"G"CAAAAGCCAC"G"G"CCF‘GAAGAAAAGCAAAGACA"C"GGACAAAAAGCAAAGA" 40 r“CCAF‘AAF‘ATAAGGGG"CGGTGGACCGGTCGAF‘GF‘AF‘G"CTF‘GF‘TGCAGA"CATCAAGAACACGTAGAGAAACCCAGC" G"AA Protein sequence of VBlOOl (Homodimeric construct ing to the invention with E6, SEQ ID NO:3): Amino acid sequence 393 amino acids.
QVSTAA IAVT. ICTMAT.CNQV IS IAPTIAADTPTACCFSYTSRQIPQNFIAD QCS (PSVIFDTKRGRQVCADPS * *WVQKYVSD .4 ISAI * IKTP IG DTTHT I EPKSCDTPPPCPRCP I GGGSSGGGSG GQPRTPQVYTTIPPS Q* * TK \IQVSLTCuV <GFYPSDIAV* W * SSGQP * VVYNFTTPPM.J:DS:DGS FF IYS < .
TVDKS QWQQGVIFSCSVMHTA Q (S :8 ISPGK G IGG 'I FQDPQER PRUIPQ uCT': IQTTI-IDII QQ . |QR-T‘VY:DFAFQ:D.JCIVYR:DG\I PYAVCD (CL (FYSKISEYR-IYCYSTIYGTT F‘QQYN (P ICD . IIRCINCQ< PTICP * * (QR-I 0pa.pa.)0i QFHNI QGRWTGRC SCCQSS QT QRTTQ .* DNA sequence of VBlOOZ (SEQ ID NO:4): ATGCAGGTCTCCACTGCTGCCCTTGCCGTCCTCCTCTGCACCATGGCTCTCTGCAACCAGGTCCTCTCT I GCACCACTT GCTGCTGACACGCCGACCGCCTGCTGCTTCAGCTACACCTCCCGACAGATTCCACAGAATTTCATAGCTGACTACTTTG AGACGAGCAGCCAGTGCTCCAAGCCCAGTGTCATCTTCCTAACCAAGAGAGGCCGGCAGGTCTGTGCTGACCCCAGTGA GGAGTGGGTCCAGAAATACGTCAGTGACCTGGAGCTGAGTGCC I GAGCTCAAAACCCCACTTGGTGACACAACTCACAC A I GAGCCCAAATCTTGTGACACACCTCCCCCGTGCCCAAGGTGCCCA I GGCGGTGGAAGCAGCGGAGGTGGAAGTGGA I GGACAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGAGGAGATGACCAAGAACCAGGTCAGCCTGACCT GCCTGGTCAAAGGCTTCTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCCTCCCATGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAG GGGAACATCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCGCTTCACGCAGAAGAGCCTCTCCCTGTCTCCGG GTAAA I GGCCTCGGTGGCCTG ATGTTTCAGGACCCACAGGAGCGACCCAGAAAGTTACCACAGTTATGCACAGAGCTG CAAACAACTATACATGATATAATATTAGAATGTGTGTACTGCAAGCAACAGTTACTGCGACGTGAGGTATATGACTTTG Cmmm.—1CGGGANT“AHGCAnAGHAmAnAGAGAHGGGAAHCCAmAmGCmeACG_AGAmAAATGT.—1TAAAGTr1r1.—1Am.—1CmAA AATTAGTGAGTATAGACATTATTGTTATAGTTTGTATGGAACAACATTAGAACAGCAATACAACAAACCGTTGTGTGAT TTGTTAATTAGGTGTATTAACTGTCAAAAGCCACTGTGTCCTGAAGAAAAGCAAAGACATCTGGACAAAAAGCAAAGAT TCCATAATATAAGGGGTCGGTGGACCGGTCGATGTATGTCTTGTTGCAGATCATCAAGAACACGTAGAGAAACCCAGCT GTAA Protein sequence of VBlOOZ (Homodimeric construct according to the invention, SEQ ID NO:5): Amino acid sequence, 393 amino acids.
QVSTAA IAVT. ICTMAT.CNQV IS ADTPTACCFSYTSRQIPQNFIAD YFETSSQCS (PSVIFDTKRGRQVCADPS * *WVQKYVSD .4 ISAI * IKTP IG DTTHT I EPKSCDTPPPCPRCP I GGGSSGGGSG GQPR'I‘PQVYTTIPPS Q* * TK \IQVSLTCuV <GFYPSDIAV* W * SSGQP * \I\IYNTTPPM_JDSDGS FF IYS ( .
TVDKS QWQQGVIFSCSVMH'I‘A IHNRFTQ (S :8 ISPGKIG IGG 'I FQDPQER PR (TIPQ uCT'I‘ IQTTI {DII TCVYCKQQ . RR'I‘VYDFAF IVYR:DG\I PYAVED (CL (FYSKISEYR-IYCYSTIYGTT F‘QQYN (P ICD . IIRCINCQ ( PTICP * * (QR-I DKKQ QFHNI QGRWTGRC SCC QSS QT QRTTQ .* DNA sequence of VB 1003 (SEQ ID NO:6): ATGCAGGTCTCCACTGCTGCCCTTGCCGTCCTCCTCTGCACCATGGCTCTCTGCAACCAGGTCCTCTCT I GCACCACTT GCTGCTGACACGCCGACCGCCTGCTGCTTCAGCTACACCTCCCGACAGATTCCACAGAATTTCATAGCTGACTACTTTG AGACGAGCAGCCAGTGCTCCAAGCCCAGTGTCATCTTCCTAACCAAGAGAGGCCGGCAGGTCTGTGCTGACCCCAGTGA GGTCCAGAAATACGTCAGTGACCTGGAGCTGAGTGCC I GAGCTCAAAACCCCACTTGGTGACACAACTCACAC A I GAGCCCAAATCTTGTGACACACCTCCCCCGTGCCCAAGGTGCCCA I GGCGGTGGAAGCAGCGGAGGTGGAAGTGGA I GGACAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGAGGAGATGACCAAGAACCAGGTCAGCCTGACCT GCCTGGTCAAAGGCTTCTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCCTCCCATGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAG GGGAACATCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCGCTTCACGCAGAAGAGCCTCTCCCTGTCTCCGG GTAAA I GGCCTCGGTGGCCTG ATGTTTCAGGACCCACAGGAGCGACCCAGAAAGTTACCACAGTTATGCACAGAGCTG CAAACAACTATACATGATATAATATTAGAATGTGTGTACTGCAAGCAACAGTTACTGCGACGTGAGGTATATGACTTTG 1CGGGANT“AHGCAnAGHAmAnAGAGAHGGGAATCCAnAmGCmeATGTGAnAAATGT.—1TAAAGTHr1.—1Am.—1CmAA AATTAGTGAGTATAGACATTATTGTTATAGTTTGTATGGAACAACATTAGAACAGCAATACAACAAACCGTTGTGTGAT ATTAGGTGTATTAACCG_ACAAAAGCCACTGTGTCCTGAAGAAAAGCAAAGACATCTGGACAAAAAGCAAAGAT TCCATAATATAAGGGGTCGGTGGACCGGTCGATGTATGTCTTGTTGCAGATCATCAAGAACACGTAGAGAAACCCAGCT GTAA Protein sequence of VBlOO3 (Homodimeric construct according to the invention, SEQ ID NO:7): Amino acid sequence, 393 amino acids.
QVSTAA IAVT. ICTMAT.CNQV IS IAPTIAADTPTACCFSYTSRQIPQNFIAD YFETSSQCS (PSVIFDTKRGRQVCADPS * *WVQKYVSD .4 ISAI * IKTP IG DTTH"1 I EPKSCDTPPPCPRCP I GGGSSGGGSG GQPR'I‘PQVYTTIPPS Q* * TK \IQVSLTCuV (GFYPSDIAV* W * SSGQP * \T\IYNTTPPM_JDSDGS FF IYS ( .
TVDKS QWQQGVIFSCSVMH'I‘A IHNRFTQ (S :8 ISPGK G IGG 'I FQDPQER PRUIPQ uCT'I‘ IQTTI-IDII 'I‘CVYCKQQ . uQR'I‘VYDFAF{DDCIVYRDGV PYAVCD (CL (FYSKISEYR-IYCYSTIYGTT F‘QQYN (P ICD . IRCINEQ< PTICP * * (QR-I 0pa.pa.)0i QFHNI QGRWTGRC SCCQSS QT QRTTQ .* DNA ce of VBlOO4 (SEQ ID NO:8): ATGCAGGTCTCCACTGCTGCCCTTGCCGTCCTCCTCTGCACCATGGCTCTCTGCAACCAGGTCCTCTCTIGCACCACTT GCTGCTGACACGCCGACCGCCTGCTGCTTCAGCTACACCTCCCGACAGATTCCACAGAATT"CATAGCTGACTACTTTG AGACGAGCAGCCAGTGC"CCAAGCCCAGTGTCATCTTCCTAACCAAGAGAGGCCGGCAGGTCTGTGCTGACCCCAGTGA GGAGTGGGTCCAGAAATACGTCAG"GACCTGGAGCTGAGTGCC I GAGCTCAAAACCCCAC""""GG"“GACACAAC"1CACAC ZXI (EZXCECE(ECEZXZXZXF‘(:TF'"(}'“(}ZX(:ZX(:ZX(3(EUTCECE(3(3(3C3'"C3(3(:(31XZX(3C3"C3(3(3(:ZX ICECE(3C3C31?(}(}ZXZXC}(:ZXC}(3(3(EZX(}(}'"(}(}ZXZX(}TF(}(}ZXI GGACAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGAGGAGATGACCAAGAACCAGGTCAGCCTGACCT GCCTGGTCAAAGGCTTCTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC TCCCATGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAG GGGAACATCTTC"CATGCTCCGTGATGCATGAGGCTCTGCACAACCGCTTCACGCAGAAGAGCCTCTCCCTGTCTCCGG GTAAAIGGCCTCGGTGGCCTG ATGTT"CAGGACCCACAGGAGCGACCCAGAAAGTTACCACAGTTATGCACAGAGCTG CAAACAAC""ATACATGATATAATA"1"AGAATGTGTGTACTGCAAGCAACAGTTACTGCGACGTGAGGTATATGACT"1"G CmECGGGANT“AHGCAmAGHAmAmAGAGAHGGGAAHCCAmAmGCmeACG_AGAmAAATG".—1TAAAGTr1r1.—1Am.—1CmAA AAT""AGTGAG""ATAGACATTA"1""G"1"“ATAGT"1""GTATGGAACAACA"1TAGAACAGCAATACAACAAACCG"1"“GTGTGA"1 T""G"1""AA"1"“AGGTGTA"1"AACCG_ACAAAAGCCAC""G""GTCCTGAAGAAAAGCAAAGACATCTGGACAAAAAGCAAAGA"1 "1CCATAATATAAGGGGTCGGTGGACCGGTCGATGTATGTCTTGTTGCAGATCATCAAGAACACGTAGAGAAACCCAGC"1 GTAA Protein sequence of VBlOO4 imeric construct according to the invention, SEQ ID NO:9): Amino acid ce, 393 amino acids.
QVSTAAIAV.LCTMALCNQVISAPIAADTPTACCFSYTSQQIPQVFIAD YFETSSQCS(PSVIFDTKRGRQVCADPS**WVQKYVSDu*.SA4L<TP C) DTTHTEP(SCDTPPPCPRCPGGGSSGGGSGGQPRTPQVYTIPPSR** CDV<GFYPSDIAV*W*SSGQP*VVYNTTPPMDDSDGSFFIYS Ara A TVDKSQWQ GVIFSCSVMHTAIHNRFTQ<SISISPGKGIGGIMFQDPQ Lu (u PR<LPQLCTTIQTTI{DII :CVYCKQQIIQRTVYDFARQDJCIVYQ UC)A PYAVEDKCL{FYSKISEYRiYCYSLYGTTITQQYN<PICDIIIRCIVEQ< PLCP**KQR{ DKKQQFHNIQGRWTGRC SCCQSSQTQQETQL* DNA sequence of VBlOOS (SEQ ID NO:10): ATGCAGGTCTCCACTGCTGCCCTTGCCG"1CCTCCTCTGCACCATGGCTC"1CTGCAACCAGG"1CCTCTC"1 I GCACCACTT GCTGCTGACACGCCGACCGCCTGCTGCT"1CAGCTACACCTCCCGACAGATTCCACAGAATT"1CATAGCTGACTACTTTG AGACGAGCAGCCAGTGC"1CCAAGCCCAGTGTCA"1C"""1CCTAACCAAGAGAGGCCGGCAGGTCTGTGCTGACCCCAGTGA GGAGTGGGTCCAGAAATACGTCAGTGACCTGGAGCTGAGTGCC I GAGCTCAAAACCCCAC""""GG""GACACAAC"1CACAC A I GAGCCCAAA"1CTTGTGACACACCTCCCCCGTGCCCAAGGTGCCCA I GGCGGTGGAAGCAGCGGAGGTGGAAGTGGA I GGACAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGAGGAGATGACCAAGAACCAGG"1CAGCCTGACCT GCCTGG"1CAAAGGCT"1CTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCCTCCCATGCTGGAC"1CCGACGGCTCCT"1CTTCCTCTACAGCAAGC"1CACCGTGGACAAGAGCAGGTGGCAGCAG A"1CTTC"1CATGCTCCGTGATGCATGAGGCTC"GCACAACCGCTTCACGCAGAAGAGCCTCTCCCTGTCTCCGG GTAAA GGCCTCGGTGGCC"1G I ATGCATGGAGATACACCTACAT"1GCATGAATATATGTTAGA"1T"GCAACCAGAGACA ACTGA"1C"1CTAC"GTTATGAGCAA"1"AAATGACAGCTCAGAGGAGGAGGATGAAATAGATGG"1CCAGCTGGACAAGCAG ACAGAGCCCAT""ACAATA"1""GTAACC"1"1TTGTTGCAAGTGTGAC"1C""ACGCT"1CGG"1"GTGCGTACAAAGCAC ACACGTAGACAT"1CGTACTTTGGAAGACCTGT"AATGGGCACACTAGGAATTGTGTGCCCCA"1CTGTTC"1CAGAAACCA WO 92875 Protein sequence of VBlOOS (Homodimeric construct according to the invention with E7, SEQ ID NO:11):Anuno acm sequence,340 anuno adds.
QVS"AA .AVT. .CTMAT .CNQV AADTP’1ACCFSYTSRQI PQNFIAD YFETSSQCS (PSVIE4T <RGRQVCADPS* * SD .4 .SAI * .KTP DTT-I"1 "‘LPKSCDTPPPCPRCP IGGGSSGGGSG GQPRTPQVYTT .PPS \TQVS ITC .V DIAV*W * SSGQP *VVYN"TPPMD,S: TVD<S QWQQGNIFSCSV H'3A -IN QFTQ {S ASLSPGKI -I':‘Y .3 QPTTTDLYCYTQ .Ni 88* * *3 *IiDGPAGQAEP C {C38 ".4 QLCVQS THV:DIRTTEiJU-3T. . GT .GIVCPICSQ DNAsequenceofVBlOO6(SEQIE)NO:12y ATGCAGGTCTCCACTGCTGCCCTTGCCG"‘CCTCCTC""GCACCATGGCTC"1CTGCAACCAGGTCCTCTC"1 I GCACCACTT GACACGCCGACCGCCTGCTGCT"1CAGCTACACCTCCCGACAGA"1TCCACAGAATTTCATAGCTGACTACTTTG AGACGAGCAGCCAGTGC"1CCAAGCCCAG"1GTCATCT”CCTAACCAAGAGAGGCCGGCAGG"CTGTGCTGACCCCAGTGA GGAGTGGGTCCAGAAA"“ACGTCAG’1GACCTGGAGCTGAGTGCC I GAGCTCAAAACCCCACTTGGTGACACAAC"CACAC AIGAGCCCAAA’1CTTGTGACACACCTCCCCCGTGCCCAAGGTGCCCA I GGCGGTGGAAGCAGCGGAGGTGGAAGTGGAI GGACAGCCCCGAGAACCACAGGTGTACACCC""GCCCCCA’1CCCGGGAGGAGATGACCAAGAACCAGG"1CAGCCTGACCT GCCTGG"CAAAGGC""""CTACCCCAGCGACATCGCCGTGGAG"1GGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCCTCCCA""GCTGGAC'‘CCGACGGCTCC""CT"CCTCTACAGCAAGC"" CACCG'1GGACAAGAGCAGGTGGCAGCAG GGGAACATCTTCTCA""GCTCCGTGATGCA'‘GAGGC"C"GCACAACCGC"‘TCACGCAGAAGAGCC"1CTCCCTGTCTCCGG GTAAA GGCCTCGGTGGCC'“GIATGCA"GGAGA"ACACCTACAT"GCA"GAA"ATA"G""AGA"""1GCAACCAGAGACA ACTGATC"CTACGGATATGGACAATTAAA"‘GACAGCTCAGAGGAGGAGGA"GAAA"AGA"GG"CCAGCTGGACAAGCAG AACCGGACAGAGCCCATTACAA"1A““GHAACCHHTHGHTGCAAGHGTGACHCHACGCTHCGGHHGF1GCGTACAAAGCAC ACACGTAGACATTCGTACT"‘TGGAAGACC"GT"AA"GGGCACAC"AGGAA"TGTG"GCCCCA"C"GTTC"CAGAAACCA TAA Protein sequence of VBlOO6 (Homodimeric construct according to the invention, SEQ ID |VO:13):Anfino acm sequence,340 annno acMs.
QVS"AA .AV .T .CTMAT .CNQV .SAP .AADTPTACCFSYTS QQI PQVFIAD YFETSSQCS (PSVIEJTKRGRQVCADPS * * SD .4 .SA* T. ("PEG DTTHTEP (S C,3.—1PPPCPRCPGGGS SGGGSGGQPR'I‘PQVYT .PPSR* 4Mr1< \IQVS ITC V DIAV* W * SSGQP * TP PMD,DS:DGS FF .YSK .
TVDKS QWQQGVI FS CSVMH'3A .HN QFTQ {S .ST.SPGKG .GGT .M {GiDTP'TJ -I' .QPTT"*onygygo .NDSS * * *3 *Ii3GPAGQAEP:D QAHYNIVTFC C {CDSTD QLCVQSTHVDI RTT F‘DT. . GT .GIVCP I CSQ <P* DNAsequenceofVBlOO7(SEQIE)NO:14y ATGCAGGTCTCCACTGCTGCCCTTGCCG"‘CCTCCTC""GCACCATGGCTC"1CTGCAACCAGGTCCTCTC"1 I GCACCACTT GCTGCTGACACGCCGACCGCCTGCTGCT"1CAGCTACACCTCCCGACAGA"1TCCACAGAATTTCATAGCTGACTACTTTG AGACGAGCAGCCAGTGC"1CCAAGCCCAG"1GTCATCT”CCTAACCAAGAGAGGCCGGCAGG"CTGTGCTGACCCCAGTGA 40 GGAGTGGGTCCAGAAA"“ACGTCAG’1GACCTGGAGCTGAGTGCC I GAGCTCAAAACCCCACTTGGTGACACAAC"CACAC AIGAGCCCAAA’1CTTGTGACACACCTCCCCCGTGCCCAAGGTGCCCA I GGCGGTGGAAGCAGCGGAGGTGGAAGTGGAI GGACAGCCCCGAGAACCACAGG"G"‘ACACCC""GCCCCCA’1CCCGGGAGGAGATGACCAAGAACCAGGTCAGCCTGACCT GCCTGGTCAAAGGCTTCTACCCCAGCGACATCGCCGTGGAG"1GGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCCTCCCA""GCTGGAC'‘CCGACGGCTCC""CT"CCTCTACAGCAAGC"1CACCGTGGACAAGAGCAGGTGGCAGCAG 45 GGGAACA"C""TCTCA""GCTCCGTGATGCA'"GAGGC'‘CTGCACAACCGC"‘TCACGCAGAAGAGCC"1CTCCCTGTCTCCGG GTAAA GGCCTCGGTGGCC"‘GIATGCA’‘GGAGA"ACACCTACAT'1GCAHGAAHATAHGHHAGAHr1.—1GCAACCAGAGACA ACTGATC"CTACGGATATGGACAATTAAA"‘GACAGCTCAGAGGAGGAGGA"GAAA’1AGATGGTCCAGCTGGACAAGCAG AACCGGACAGAGCCCAT"‘ACAA"1Ar1r1Gr1AACCr""ngéTGCAAeggéeAc"‘C"ACGCT"CGG""G"1GCGTACAAAGCAC ACACGTAGACATTCGTACT"1TGGAAGACC"‘GT"1AATGGGCACAC"‘AGGAATTGTG"1GCCCCATCTGTTCTCAGAAACCA 50 TAA Protein sequence of VBlOO7 (Homodimeric construct according to the invention, SEQ ID |VO:15):Anfino acm sequence,340 annno acMs.
ALAVLLCTMALCNQVLSAPLAA:DTPTACCFSYTSRQI PQNFIAD YFETSSQCS (PSVIF.4TKRGRQVCAiDPS * WVQKYVSD .4 .SA* T.
DTT-ITEPKS CD"1PPPCPRCPGGGSSGGGSGGQPRF‘PQVYT .PPSR \TQVS_I1CLV (GFYPS:DIAV *SSGQP 4 TPPMJDSDGS FF TVD (S QWQQGVI FS CSVMH' QFTQ .S T .S PGKG Vl -IG: -I':‘Y Q .QPFT" onygygo 3GPAGQAEPD VTFE C (GDS "1.4RLCVQS THV:DIRTTEuLJ. .GIVCPI CSQ (P* DNAsequenceofVBlOOS(SEQIE)NO:16y ATGCAGGTCTCCAC'1GC'1GCCCTTGCCG'1CCTCCTC"1GCACCATGGCTC"1 CCAGG"1CC"1CTC"‘IGCACCAC"T GCTGCTGACACGCCGACCGCCTGC'1GCT"1CAGC'11ACACCT1CCCGACAGA"1TCCACAGAATF1'11CA"1AGC"1GACTACT""G AGACGAGCAGCCAG"1GC"1CCAAGCCCAG"1GTCA"1 C"1"1 C C'11AACCAAGAGAGGCCGGCAGG"1CTG"1GC"‘GACCCCAG’1GA GGAGTGGGTCCAGAAA’1ACGTCAG"1GACCTGGAGC'1GAGT1GCC I GAGCTCAAAACCCCACF1'11GG'1‘GACACAAC"CACAC CCAAA’1CT" 1GACACACCTCCCCCG"1GCCCAAGG"1GCCCA I GGAAGCAGCGGAGG"‘GGAAGTGGAI GGACAGCCCCGAGAACCACAGG' 1ACACCC"1GCCCCCA"1C CCGGGAGGAGATGACCAAGAACCAGG"1CAGCC"GACCT GCCTGG"CAAAGGC' 1CTACCCCAGCGACATCGCCGTGGAG"‘GGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCC"CCCA’1GC'1GGAC'1CCGACGGCTCC" .CT.1CCTCTACAGCAAGC"1CACCG"GGACAAGAGCAGG"1GGCAGCAG GGGAACA"C"TCTCA’1GCTCCG".GA.1GCA"1GAGGC'1CT1GCACAACCGC"‘TCACGCAGAAGAGCC"CTCCC'1GTCTCCGG GTAAA GGTGGCC‘GIATGCA’1GGAGA"1ACACCTACAT"1GCA'1GAA'1ATA" .HAGAHF 1GCAACCAGAGACA ACTGA”C"C"AC§§§' ‘GG_ACAA" 1AAA"1GACAGCTCAGAGGAGGAGGA"1GAAATAGA"GG"CCAGCTGGACAAGCAG AACCGGACAGAGCCCAT"“ACAA’ 1AACC"1"T"G"TGCAAG'1GTGAC" 1ACGCT"1CGG'11'11G"1GCG"1ACAAAGCAC ACACG"AGACATTCG'“ACT"‘TGGAAGACC’1GT"1AA"‘GGGCACAC’1AGGAA"1TGTGGGACCCA"1CGGATC"1CAGAAACCA Protein sequence of VBlOOS (Homodimeric construct according to the invention, SEQ ID |VO:17):Anfino acm sequence,340 annno acMs.
QVS'1AA .AV AT.CNQV .SAP .AADTPTACCFSYTS QQI PQVFIA: YFETS SQCS (PSVI F.4TKRGRQVCADPS * WVQKYVSD .4 .SA* T. 1PT.G DTT {'13 P (S PCPRCPGGGSSGGGSGGQPRF‘PQVYT .PPSR «Mr \TQVS ."1C .V (GFYPSDIAV * SSGQP * \IVYNTTPPM.J:DS:DGS FF .YSK TVD (S QWQQGVI FS CSVMH' . QFTQ .ST.SPGKG .GGT.1Vl-IGD" -I':‘Y Q .QP'TT'1DT IYEYEQ D 1 DGPAGQAEPDQAHYNIVTFC C (CDS "1.4 QLCVQS THVD I RTT (3‘, .GIV§P1§5o<P* Constructs with E6 and E7: For the purpose of illustration only, the different domains of the constructs are separated by an “|" with the domains in the following order: Signal peptide | human MIP-lCl | Hinge h1 | Hinge h4 | Gly—Ser Linker or Gly—Leu linker| hCH3 IgG3 |Gly—Ser Linker or u linker| E7 | r Linker or Gly—Leu linker| E6 mutant. Amino acids or nucleotides in bold illustrates sites of mutations. 4o DNAsequenceofVBlOO9(SEQIE)NO:18y ATGCAGGTCTCCAC"1GC'1GCCCTTGCCG'1CCTCCTC"1GCACCATGGCTC"1CTGCAACCAGG'11CC"1CTC"1|GCACCAC"T GCTGCTGACACGCCGACCGCCTGC"1GCT"1CAGC"1ACACC'11CCCGACAGA"1TCCACAGAAT"1"1CA"1AGC"1GACTACT""G AGACGAGCAGCCAG"1GC'11CCAAGCCCAG"1GTCA"1C"1"1CC'11AACCAAGAGAGGCCGGCAGG'1CTG"1GC"1GACCCCAG"GA GGAGTGGGTCCAGAAA"1ACGTCAG"1GACCTGGAGCT1GAGT1GCC I GAGCTCAAAACCCCAC'1'1GG"‘GACACAAC"CACAC 45 AIGAGCCCAAA’1CT" 1GACACACCTCCCCCG"1GCCCAAGG"1GCCCA I GGCGGTGGAAGCAGCGGAGG"‘GGAAGTGGAI GGACAGCCCCGAGAACCACAGG" 1ACACCC"1GCCCCCA"1C CCGGGAGGAGATGACCAAGAACCAGG"1CAGCC"GACCT GCCTGGT1CAAAGGC" 1CTACCCCAGCGACATCGCCGTGGAG"‘GGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCCr1CCCA".Gcr1GGAC"1CCGACGGCTCC" .CT.1CCTCTACAGCAAGC"1CACCG"GGACAAGAGCAGG"1GGCAGCAG GGGAACA"C"TCTCA’1GCTCCG".GA.1GCA"‘GAGGC"C"GCACAACCGC"‘TCACGCAGAAGAGCC"CTCCC'1GTCTCCGG 50 GTAAAIGGCCTCGGTGGCC‘GIATGCA’‘GGAGA’1ACACCTACAT"1GCA"1GAA'11ATA" .HAGAHF 1GCAACCAGAGACA ACTGATC"C"AC§§§' ‘GG_ACAA" "AAA"1GACAGCTCAGAGGAGGAGGA"1GAAATAGA'11GG"1 CCAGCTGGACAAGCAG AACCGGACAGAGCCCATF1ACAA" 1AACCT"1T'11G'11TGCAAG"1GTGAC"1C"1ACGCT"1CGG"-n—-.Gr1GCGTACAAAGCAC ACACGTAGACA'—1TCGTACTTTGGAAGACCTGTTAATGGGCACACTAGGAATTGTGTGCCCCATCTGTTCTCAGAAACCA GGCGGTGGAAGCAGCGGAGGTGGAAG"1GGA ATGTTTCAGGACCCACAGGAGCGACCCAGAAAGTTACCACAGTTATG CACAGAGCTGCAAACAACTA"‘ACATGA"4AmAAr4Am.4AGAAr4Gr4GTGTACTGCAAGCAACAGTTACTGCGACGTGAGGTA rfiArfiGACrfirfirfiGC""CGACGGGAT'4.4AHGCAnAGr4AmAnAGAGAr4GGGAATCCATATGCTGTACGAGATAAATGTTTAAAGT CTAAAATTAGTGAG’1A""AGACATHANqumHAHAGHr4r4GTATGGAACAACATTAGAACAGCAATACAACAAACC GmmeGmGAmmr“GTTAA"TAGGTGTATTAACCGACAAAAGCCACTGTGTCCTGAAGAAAAGCAAAGACATCTGGACAAA AAGCAAAGATTCCATAATATAAGGGGTCGGTGGACCGGTCGATGTATGTCTTGTTGCAGATCATCAAGAACACGTAGAG AAACCCAGCTG"1AA Protein sequence of VBlOO9 (Homodimeric construct according to the invention, SEQ ID |VO:19):Anfino acm sequence,501 anflno acMs.
QVSTAA IAVT . .CTMAT .C\IQV .S I APT .AADTPTACCFSYTSRQI PQNFIAD QCS (PSVI F.4T< QGRQVCADPS * * WVQKYVSD fl .SA |«.KTP.G DTT-IT EPKS CDTPPPCP QCP |GGGSSGGGSG GQPRTPQVYTT.PPSQ* * TK \IQVS ITC .V (GFYPSDIAV *W 4 SSGQP * \T\TYNTTP PM. sacs FF.YS<.
TVD<S QWQQGNI FS CSV {TA .-IN QFTQ S PGK | LGGL | {GDTPTL {TY .i3 .QPTTTD .YEYGQ .Ni388444 D «IDGPAGQAE P:U QAHYVIVTFC C (CDST.4 THVDI zanDT. GT .GIVCP I CSQ P |GGGSSGGGSG| FQDPQ':‘QP QKT.PQT.CT':' .QTTIHDI If:‘CVYCKQQT. . aTVYDFARzl4 CIVYQ DGVPYAVE:DKC.J {FYS (ISEYE {YCYST .YGTT .' UNAQQYN .
IQCIVEQ (P ICP * 4KQ Q -1 .DK {QRF {\II QGRWTGRCMS cczss QTRRETQ DNAsequenceofVBlOl6(SEQIE)NO:20y GTCTCCACTGCTGCCCTTGCCGTCCTCCTCTGCACCATGGCTCTCTGCAACCAGGTCCTCTCT I GCACCACTT GCTGCTGACACGCCGACCGCCTGC"1GCTTCAGCTACACCTCCCGACAGATTCCACAGAATTTCATAGCTGACTACTTTG AGACGAGCAGCCAGTGCTCCAAGCCCAGTGTCATCTTCCTAACCAAGAGAGGCCGGCAGGTCTGTGCTGACCCCAGTGA GGAGTGGGTCCAGAAA"‘ACGTCAG""GACCTGGAGC'‘GAGTGCC |GAGCTCAAAACCCCACTTGGTGACACAACTCACAC A|GAGCCCAAA’‘CTTGTGACACACCTCCCCCG"“GCCCAAGGTGCCCA|GGCGGTGGAAGCAGCGGAGGTGGAAGTGGA| GGACAGCCCCGAGAACCACAGGTGTACACCC"“GCCCCCATCCCGGGAGGAGATGACCAAGAACCAGGTCAGCCTGACCT GCCTGGTCAAAGGCTTCTACCCCAGCGACATCGCCGTGGAGTGGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCCTCCCA'—1GCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGGCAGCAG GGGAACATCTTCTCA"1GCTCCGTGATGCATGAGGCTCTGCACAACCGCTTCACGCAGAAGAGCCTCTCCCTGTCTCCGG GTAAA GGCCTCGGTGGCCTG |ATGCA"‘GGAGA"1ACACCTACATTGCATGAATATATGTTAGATTTGCAACCAGAGACA ACTGATCTCTACGGATATGGACAATTAAATGACAGCTCAGAGGAGGAGGATGAAATAGATGGTCCAGCTGGACAAGCAG AACCGGACAGAGCCCATTACAATATTGTAACCTTTTGTTGCAAGTGTGACTCTACGCTTCGGTTGTGCGTACAAAGCAC ACACGTAGACA'—4—4CGHACHHHGGAAGAC C'—1GTTAATGGGCACACTAGGAATTGTGTGCCCCATCTGTTCTCAGAAACCA GGCGGTGGAAGCAGCGGAGGTGGAAG"1GGA ATGTTTCAGGACCCACAGGAGCGACCCAGAAAGTTACCACAGTTATG GCTGCAAACAACTATACATGATATAATATTAGAATGTGTGTACTGCAAGCAACAGTTACTGCGACGTGAGGTA 40 rfiArfiGACrfirfirfiGC"1r1r1r1CGGGAr1r4—4Ar4GCAmAGTAmAmAGAGAHGGGAATCCATATGCTGTACGAGATAAATGTTTAAAGT .—-..—-..—-.A.—-.TCmAAAAmrqAGTGAGmAr‘AGACAT“Am“Gmr4Ar4AGr4r4r4GTATGGAACAACATTAGAACAGCAATACAACAAACC GmmeGmGAmmr“GTTAA"TAGGTGTATTAACCGACAAAAGCCACTGTGTCCTGAAGAAAAGCAAAGACATCTGGACAAA AAGCAAAGATTCCATAATATAAGGGGTCGGTGGACCGGTCGATGTATGTCTTGTTGCAGATCATCAAGAACACGTAGAG AAACCCAGCTG"1AA Protein sequence of VBlOl6 (Homodimeric construct according to the invention, SEQ ID |VO:21):Anfino acm sequence,501 anflno acMs QVSTAA .AV .T .CTMAT .C\TQV .SAP .AA:DTPTACCFSYTS QQI PQVFIAD YFETS SQCS (PSVI F.4T< QGRQVCADPS * * WVQKYVSD fl .SA 4T.(TPT.G 50 DTT-ITEP (S C: S SGGGSGGQPR':‘PQVYT .PP SR* *MT \IQVS .TC DIAV*W 4 SSGQP * \IWYNTTPPM;DSDGS {TA _ -IN QFTQ (S .ST.SPGKG .NiU 88* * *3 4 IDGPAGQA:1 T. GT .GIVCPICSQ 55 Li .QTTIHDI If:‘CVYCKQQT.
.J4AFYS (ISEYE {YCYST .YGTT .' .DK {QRF {\II QGRWTGRCMS CC SEQ ID NO:22: >tr|Q77816|Q77816_HPV16 E6 protein OS=Human papillomavirus type 16 GN=E6 PE=4 SV=1; (Underlined amino acids denotes amino acids that may be deleted; Potential amino acids that may be mutated are highlighted) MFQDPQERPRKLPQLCTELQ‘I‘I’IHDIILECVYCKQQLLRREVYDFAFRDLCIVYRDGNPYAVCDKCLKFYS HYCYSLYG'I‘I'LEQQYNKPLCDLLIRCINCQKPLCPEEKQRHLDKKQRFHNIRGRWTGRCMSCCR SSRTRRETQL SEQ ID NO:23: >sp|P03129|VE7_HPV16 Protein E7 OS=Human papillomavirus type 16 GN=E7 PE=1 SV=1; (Underlined amino acids denotes amino acids that may be deleted; Potential amino acids that may be mutated are highlighted) MHGDTPTLHEYMLDLQPETTDLYCYEQLNDSSEEEDEIDGPAGQAEPDRAHYNIVTFCCKCDSTLRLCVQ RTLEDLLMGTLGIVCPICSQKP SEQ ID NO:24: >sp|P06463|VE6_HPV18 Protein E6 OS=Human papillomavirus type 18 GN=E6 PE=1 SV=1 MARFEDPTRRPYKLPDLCTELNTSLQDIEITCVYCKTVLELTEVFEFAFKDLFVVYRDSI PHAACHKCIDFYSRIRELRHYSDSVYGDTLEKLTNTGLYNLLIRCLRCQKPLNPAEKLRH LNEKRRFHNIAGHYRGQCHSCCNRARQERLQRRRETQV SEQ ID NO:25: >sp|P06788|VE7_HPV18 Protein E7 OS=Human papillomavirus type 18 GN=E7 PE=3 SV=2 MHGPKATLQDIVLHLEPQNEIPVDLLCHEQLSDSEEENDEIDGVNHQHLPARRAEPQRHT MLCMCCKCEARIKLVVESSADDLRAFQQLFLNTLSFVCPWCASQQ SEQ ID NO:26: Hinge regions (IgG3 UH hinge), 12 amino acids: ELKTPLGD'I‘I'HT SEQ ID NO:27: Hinge region (IgG3, MH hinge, 15 amino acids): TPPPCPRCP SEQ ID NO:28: Gly—Ser Linker: GGGSSGGGSG SEQ ID NO:29: hCH3 IgG3: GQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESSGQPENNYNTI'PPMLDSDGSFFLYSKL WQQGNIFSCSVMHEALHNRFTQKSLSLSPGK SEQ ID NO:30: Linker: GLGGL SEQ ID NO:31: DNA sequence of VBlOZO: 45 ATGCAGGTCTCCAC"GC"GCCCTTGCCG"CCTCCTC"GCACCATGGCTC"CTGCAACCAGG"CC"CTC"IGCACCAC"T GCTGCTGACACGCCGACCGCCTGC"GCT"CAGC"ACACC"CCCGACAGA"TCCACAGAAT""CA"AGC"GACTACT""G GCAGCCAG"GC"CCAAGCCCAG"GTCA"C""CC"AACCAAGAGAGGCCGGCAGG"CTG"GC"GACCCCAG"GA GGAGTGGGTCCAGAAA"ACGTCAG'1GACCTGGAGC'1GAG'1GCC I GAGCTCAAAACCCCAC"1"1GG""GACACAAC"1CACAC [XI CE(ECEZXZXZX'1(:TF""(}""(}ZX(:ZX(EZX(3(EUTCECECECE(3C3""C3(3(ECEZXZX(}C3'"C3(3(3(:ZX IC3C3C:C3C31T(E(EZXZXC3(:ZXC}(3(3(EZX(}(}""(}(}Z%]X(}TF(}(}ZXI 50 GGACAGCCCCGAGAACCACAGG"G"ACACCC"GCCCCCA"CCCGGGAGGAGATGACCAAGAACCAGG"CAGCC"GACCT "CAAAGGC""CTACCCCAGCGACATCGCCGTGGAG"GGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC CACGCC"CCCA"GC"GGACTCCGACGGC"CC""C""CCTCTACAGCAAGCTCACCG"GGACAAGAGCAGG"GGCAGCAG GGGAACATCT"C"CA"GC"CCG"GA"GCATGAGGC"C"GCACAACCGCT"CACGCAGAAGAGCC"C"CCC"G"CTCCGG G"AAA|GGCCTCGGTGGCCTG/ATGCA"GGCGA"ACCCCAACAC"CCA"GAGTACATGCTGGACC""CAGCCCGAGAC 55 "ACGGA"C"G"A"GGC"A"GGGCAG""GAA"GAC"CA"C"GAGGAGGAGGACGAAA"AGACGGCCCAGCTGG"CAAGCC GAACCGGA"AGAGCCCAC"ACAACA""G"GACC""""GC"G"AAG"G"GACAGCAC"C"GAGAC"G"G"G"TCAGTCCA C"CATG"CGACA”ACGCACAT"GGAGGA"C"CC"GA"GGGAACAC"GGGAATTGTG"G"CCCA"CTG"”CCCAAAAGCC "1/GGAGGTGGAAGCAG""GGAGGCGG""""CAGGC/A""G""""CCAAGATCC""CAAGAACG""CCTCGTAAGC""GCCACAGCTG"1 G""ACCGAGCTTCAGACCACCA”TCAC&ACA""CPJ’"CC""GGAG""GCG""C""ATTGCAAACAGCAGC"CC"1""AGAAGGGAAG"1 G"ACGATTTTGCACGGAGGGACC"CTGCATCGTGTA"CGGGACGGCAATCCC"ATGCGGTACGGGA"AAATGCCTGAAG T"C"ACAGCAAAATC"CCGAGTACCGGCACTACTGC"ACTCTC"C"A"GGGACGACTCTGGAACAGCAGTACAACAAGC CCT"GTGCGA”C"GC"GATTCGC"GCATTAA"CGCCAGAAACC"C"G"GCCCAGAAGAGAAGCAAAGACACCTGGACAA GAAACAGCGA""CCACAACATCCGAGGGAGA"GGACAGGGAGG"G"A"GAGC"GCTGTCGGAGTTC"AGGACAAGGCGC GAAACCCAGC"""""GA SEQ ID NO:32: Protein sequence of VBlOZO (Homodimeric construct according to the invenUon Annno acm ce,501 annno adds: QVS'1AA IAVT. .C\IQV IS IAPTIMiDTPr‘ACCFSYTSRQIPQNFIAD YFETSSQCS {PSVIF_4T<QGRQVCADPS * *WVQKYVSD .4 ISAI * IKTP IG DTT-I"1 EPKSCDTPPPCP QCP I GGGSSGGGSG GQPR'TPQVYTTIPPS Q* * TK \TQVS 51C IV <GFYPSDIAV* W * SSGQP * VVYNr‘TPPMJ SDGSFF.YS<.
TVD (S WQQGNIFSCSVQ {TA u-IN QFTQ (S.JSLSPGK| LGGL| L -I':‘Y .i3 IQP'TTTD IYEYEQ uNDSSd * *D* IDGPAGQAEP:U QAHYVIVTFC C(CDS'TJQLCVQSTHVDI QTTF‘DT. . GT IGIVCPICSQ AP|GGGSSGGGSG| FQDPQ': QP QKTE’QTICT'iJ IQTTIHDII F‘CVYCKQQT. . N zrvyoFAgzou CIVY Q:DG\IPYAVE:DKC.J AFYS {ISEY Q-IYCYSTXGTT .'u LCD..
I QCIVEQ (P C]? * * KQQ A OK {QRF {\II QGRWTGRCMSCCQSSQTRRETQ * SEQ ID NO:33: DNA ce of VBlOZl: ATGCAGGTCTCCAC"GC"GCCCTTGCCG"CCTCCTC"GCACCATGGCTC"CTGCAACCAGG"CC"CTC"IGCACCAC"T GCTGCTGACACGCCGACCGCCTGC"GCT"CAGC"ACACC"CCCGACAGA"TCCACAGAAT""CA"AGC"GACTACT""G AGACGAGCAGCCAG"GC"CCAAGCCCAG"GTCA"C""CC"AACCAAGAGAGGCCGGCAGG"CTG"GC"GACCCCAG"GA GGAGTGGGTCCAGAAA"ACGTCAG"GACCTGGAGC"GAG"GCC|GAGCTCAAAACCCCAC"”GG"GACACAAC"CACAC AIGAGCCCAAA"CT"G"GACACACCTCCCCCG"GCCCAAGG"GCCCA|GGCGGTGGAAGCAGCGGAGG"GGAAGTGGA| GGACAGCCCCGAGAACCACAGG"G"ACACCC"GCCCCCA"CCCGGGAGGAGATGACCAAGAACCAGG"CAGCC"GACCT GCCTGG"CAAAGGC""CTACCCCAGCGACATCGCCGTGGAG"GGGAGAGCAGCGGGCAGCCGGAGAACAACTACAACAC "CCCA"GC"GGACTCCGACGGCTCC""CT"CCTCTACAGCAAGCTCACCGTGGACAAGAGCAGG"GGCAGCAG GGGAACATCTTCTCA"GCTCCG"GATGCATGAGGC"CTGCACAACCGCTTCACGCAGAAGAGCCTCTCCC"GTCTCCGG GTAAMGGCCTCGGTGGCCHEATGCATGGTGACACACCAACCCTGCACGAATACATGCTCGATCTGCAGCCAGAG ACTACCGACCTTTACGGCTATGGGCAGTTGAACGACAGCTCTGAGGAGGAGGACGAGATCGATGGTCCTGCTGGA CAAGCAGAACCAGACAGAGCCCACTACAACATCGTAACCTTTTGCTGCAAGTGTGACAGTACCCTTCGTTTGTGCG TTCAGAGCACGCATGTCGACATTCGGACACTGGAGGATCTGCTCATGGGGACTCTGGGGATTGTGTGTCCTATTTG CAGCCAGAAACQNGGCGGAGGATCTTCAGGAGGCGGGAGTGGCMJGTTCCAAGACCCTCAGGAACGCCCTCGG AAACTGCCCCAATTGTGTACTGAGCTCCAGACAACGATACACGACATAATCCTGGAGTGCGTGTATTGCAAGCAGC AGCTTCTGAGGAGGGAAGTGTACGATTTTGCCAGGAGAGATGGCTGCATTGTCTACCGAGATGGCAATCCCTATG 4o CGGTGTGTGATAAGTGTCTGAAGTTCTATTCCAAAATCAGCGAATATCGGCATTATTGCTACTCACTGTACGGAACT ACCCTCGAACAGCAGTACAACAAACCGCTCTGTGATCTGCTGATCAGATGCATCAATCGGCAGAAACCCCTTTGTC CCGAAGAGAAGCAAAGACACCTGGACAAGAAGCAGAGGTTCCACAATACCCGAGGTCGTTGGACTGGGCGCTGC TGTTGTCGCTCCTCTCGCACAAGGAGAGAGACACAACTGTGA 45 SEQ ID NO:34: Protein sequence of VBlOZl (Homodimeric construct according to the invenUon.Anfino acm sequence,501 annno adds: QVS"AA.AVL.CTMALCVQV.S|APEAADTP"ACCFSYTSRQIPQNFIAD YFETSSQCS<PSVIFuT<QGRQVCAQPSd*WVQKYVSD.*ISA|*.KTP.G 50 DTTI" EPKSCDTPPPCPQCP|GGGSSGGGSG GQPRTPQVYTLPPSQ44 TK VQVS."C.V<GFYPSDIAV*W*SSGQPdVVYN"TPPMu SDGSFF.YS<.
TVD<S WQQGNIFSCSV {TAIINQFTQ<SDSLSPGK| LGGL| {GDTPTL .QPTTTDIYGYGQINDSS***D*IDGPAGQAE@U VTFC C<cos"uzLCVQSTHVDIzTL:3L. GT.GIVCPICSQ<P|GGGSSGGGSG| 55 "11 IO U "U IO U (U "U (Ux—i' ' "U 10 _. OH 1J'.QTTIHDIIITCVYCKQQLIQQTVYDFAEQDE SEYQIYCYSEYGTTITQQYN<PLCDJL A .DK<QRF{V QGRWTGRCMSCCQSSQTRRETQ

Claims (33)

Claims
1.
A nucleic acid molecule encoding an amino acid chain comprising (1) a signal peptide, (2) a targeting unit, (3) a dimerization motif, and (4) an antigenic unit, said 5 targeting unit comprising an amino acid sequence having at least 80 % sequence identity to the amino acid sequence 24-93 of SEQ ID NO:1, and an antigenic unit comprising an amino acid sequence of human papillomavirus (HPV) HPV16 and/or HPV18, preferably an antigenic unit derived from early proteins E6 and/or E7 of HPV16 and/or HPV18, which amino acid chain is able to form a homodimeric protein. 10 2. The nucleic acid molecule according to claim 1, wherein said targeting unit, dimerization motif and antigenic unit in said amino acid chain are in the N-terminal to C-terminal order of targeting unit, dimerization motif and antigenic unit.
3. The amino acid chain according to either one of claims 1 or 2, wherein said signal peptide ts of an amino acid ce having at least 80 % sequence 15 identity to the amino acid sequence 1-23 of SEQ ID NO:1.
4. The nucleic acid molecule according to any one of claims 1-3, wherein the zation motif comprises a hinge region and optionally an immunoglobulin domain, optionally connected h a linker.
5. The nucleic acid le ing to claim 4, comprising an immunoglobulin 20 domain which is a carboxyterminal C domain, or a sequence that is substantially identical to said C domain or a variant thereof.
6. The nucleic acid molecule according to any one of claims 1-5, wherein the dimerization motif consists of hinge exons h1 and h4 connected through a linker to a CH3 domain of human IgG3. 25
7. The nucleic acid le according to any one of claims 1-6, wherein the dimerization motif consists of an amino acid ce having at least 80 % sequence identity to the amino acid sequence 94-237 of SEQ ID NO:3.
8. The nucleic acid molecule according to any one of claims 1-9, wherein said targeting unit ts of amino acids 24-93 of SEQ ID NO:1, or a variant thereof.
9. The nucleic acid molecule according to any one of claims 1-8, wherein said nic unit comprises an amino acid ce having at least 80%sequence identity to the amino acid sequence 243-293 of SEQ ID NO:3 or to the amino acid sequence 243-340 of SEQ ID NO:11. 5 10. The nucleic acid molecule according to claim 9, wherein said antigenic unit ses one or more amino acid substitutions at a position selected from the list consisting of F47, L50, C63, C106 and I128 of SEQ ID NO:22, or a deletion involving one or more amino acid selected from the list consisting of Y43-L50 of SEQ ID NO:22 or wherein said antigenic unit comprises one or more amino acid substitutions at a
10 position selected from the list consisting of C24, E26, C58, C61, C91, and C94 of SEQ ID NO:23, or a deletion involving one or more amino acid selected from the list ting of L22-E26 and/or C58-C61 and/or C91-S95 of SEQ ID NO:23.
11. The nucleic acid molecule according to any one of claims 1-10, wherein said antigenic unit comprises an amino acid sequence of human papillomavirus 16 (HPV16) 15 derived from both early proteins E6 and E7
12. The nucleic acid molecule according to any one of claims 1-11, wherein said antigenic unit comprises an amino acid sequence having at least 80% sequence identity to the amino acid sequence 1 of SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:32, or SEQ ID NO:34. 20
13. The nucleic acid molecule according to claim 12, n said antigenic unit comprises one or more amino acid substitutions at a position selected from the list consisting of F47, L50, C63, C106 and I128 of SEQ ID NO:22 and C24, E26, C58, C61, C91, C94 of SEQ ID NO:23.
14. The nucleic acid molecule ing to any one of claims 12-13, wherein said 25 antigenic unit consists of the amino acid sequence 1 of SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:32, or SEQ ID NO:34, or a variant or antigenic fragment thereof.
15. The c acid molecule according to any one of claims 1-14, wherein said amino acid chain consists of an amino acid sequence selected from the list consisting of SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO:11, SEQ ID NO:13, SEQ ID NO:15, SEQ ID NO:17, SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:32, and SEQ ID NO:34, or a variant or nic nt thereof.
16. The nucleic acid molecule according to any one of claims 1-15, wherein said amino acid chain consists of an amino acid sequence selected from the list consisting 5 of SEQ ID NO:19, SEQ ID NO:21, SEQ ID NO:32, and SEQ ID NO:34, or a variant or antigenic fragment thereof.
17. The nucleic acid molecule according to claim 1, wherein said amino acid chain consists of SEQ ID NO:32, or a variant or antigenic fragment thereof.
18. The nucleic acid molecule according to any one of claims 1-17, which nucleic 10 acid molecule is human codon optimized
19. The nucleic acid molecule according to any one of claims 1-18, which is a DNA or RNA, preferably a DNA.
20. The nucleic acid molecule ing to any one of claims 1-19 comprising a vector. 15
21. The c acid molecule according to any one of claims 1-20 formulated for administration to a patient to induce production of the dimeric protein in said patient.
22. A homodimeric protein of two amino acid chains encoded by a nucleic acid molecule according to any one of claims 1-19.
23. The homodimeric n according to claim 22, in its mature form without any 20 signal peptide sequence.
24. The homodimeric protein according to either one of claims 22 or 23, or the c acid molecule according to any one of claims 1-21, for use as a medicament.
25. A pharmaceutical composition comprising the homodimeric protein according to either one of claims 22 or 23, or the nucleic acid le according to any one of 25 claims 1-21.
26. A host cell comprising the nucleic acid molecule according to any one of claims 1-21, wherein the host cell is not a transformed host cell within a human body.
27. A method for preparing a homodimeric protein according to either one of claims 22 or 23, the method comprising a) transfecting the nucleic acid molecule ing to any one of claims 1-21 into a cell population; b) culturing the cell population; and c) collecting and ing the homodimeric protein expressed from the cell 10 population.
28. A method for preparing a vaccine comprising an immunologically effective amount of a nucleic acid molecule according to any one of claims 1-21, the method comprising a) preparing a nucleic acid molecule ing to any one of claims 1-21; 15 and b) dissolving the nucleic acid molecule obtained under step a) in a pharmaceutically acceptable carrier, diluent, or buffer.
29. A vaccine against HPV comprising an immunologically effective amount of a dimeric n according to either one of claims 22 or 23, or a c acid molecule 20 according to any one of claims 1-21, and a pharmaceutically acceptable r and/or adjuvant.
30. The vaccine according to claim 29 comprising an immunologically effective amount of a nucleic acid molecule according to any one of claims 1-21, preferably a DNA. 25
31. A meric protein according to either one of claims 22 or 23 or a nucleic acid le according to any one of claims 1-21, for use in the treatment or prevention of a HPV-induced disease or condition, such as a cancer or an infectious disease caused by HPV.
32. The homodimeric protein or the nucleic acid molecule according to claim 31, wherein the nucleic acid le, preferably a DNA, is formulated for administration to the patient through electroporation.
33. The meric protein or the nucleic acid molecule according to either one of 5 claims 31 or 32, formulated for intradermal or intramuscular administration.
NZ626124A 2011-12-21 2012-12-20 Vaccines against hpv NZ626124B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161578542P 2011-12-21 2011-12-21
US61/578,542 2011-12-21
PCT/EP2012/076404 WO2013092875A1 (en) 2011-12-21 2012-12-20 Vaccines against hpv

Publications (2)

Publication Number Publication Date
NZ626124A NZ626124A (en) 2016-07-29
NZ626124B2 true NZ626124B2 (en) 2016-11-01

Family

ID=

Similar Documents

Publication Publication Date Title
RU2644201C2 (en) Vaccines against hpv
EP2928493B1 (en) Fusion proteins for use as immunogenic enhancers for inducing antigen-specific t cell responses
AU2021232660A1 (en) HPV vaccines
JP6903120B2 (en) Compositions and Methods for Alpha Virus Vaccination
JP5345053B2 (en) Composition comprising human papillomavirus polypeptide and immunopotentiator for prevention and treatment of cervical cancer
Massa et al. Antitumor activity of DNA vaccines based on the human papillomavirus-16 E7 protein genetically fused to a plant virus coat protein
US8470372B2 (en) Material with immunogenicity
KR20050050115A (en) Dna vaccine encoding at least two nonstructural early proteins of papillomavirus
NZ626124B2 (en) Vaccines against hpv
CN105801704B (en) Construction method and application of recombinant vaccine with anti-cervical cancer cell activity
JP2024518565A (en) Vaccine formulations containing recombinant overlapping peptides and native proteins - Patents.com