WO2023083964A1 - Protéine structurale de parvovirus dirigée contre hpv bêta-et gamma - Google Patents

Protéine structurale de parvovirus dirigée contre hpv bêta-et gamma Download PDF

Info

Publication number
WO2023083964A1
WO2023083964A1 PCT/EP2022/081473 EP2022081473W WO2023083964A1 WO 2023083964 A1 WO2023083964 A1 WO 2023083964A1 EP 2022081473 W EP2022081473 W EP 2022081473W WO 2023083964 A1 WO2023083964 A1 WO 2023083964A1
Authority
WO
WIPO (PCT)
Prior art keywords
structural protein
mutated
hpv
protein
parvovirus
Prior art date
Application number
PCT/EP2022/081473
Other languages
English (en)
Inventor
Preben BRUUN-NYZELL
Jeanette PRANGSGAARD
Original Assignee
2A Pharma Ab
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 2A Pharma Ab filed Critical 2A Pharma Ab
Publication of WO2023083964A1 publication Critical patent/WO2023083964A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [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
    • 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
    • A61K2039/525Virus
    • A61K2039/5258Virus-like particles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14122New 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14123Virus like particles [VLP]
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14134Use 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14142Use of virus, viral particle or viral elements as a vector virus or viral particle as vehicle, e.g. encapsulating small organic molecule

Definitions

  • the present invention relates to a mutated parvovirus structural protein, comprising at least one insertion comprising a sequence of amino acids from amino acids from HPV16 and/or HPV31 L2 protein for use in the treatment or prevention of a beta-type or gamma-type HPV infection. Furthermore, the invention relates to multimeric structures comprising the protein, VLPs, a method of producing the mutated parvovirus structural protein and to medicaments or vaccines comprising the mutated parvovirus structural protein that may be used for treating a beta-type or gamma-type HPV infection.
  • PVs Papillomaviruses
  • Papillomaviruses are small, non-enveloped viruses with double-stranded circular DNA genomes packaged into an icosahedral capsid. PVs display strict tropism for cutaneous or mucosal stratifying epithelium. PV infections are very common, and mostly asymptomatic in individuals with an intact immune system (Beziat V; 2020).
  • HPVs human papillomaviruses
  • HPVs of the alpha-type display mucosal and cutaneous tropism.
  • the alpha-HPVs are associated with cutaneous warts (e.g. HPV2), benign mucosal diseases (e.g. HPV6, HPV13) or genital and oropharyngeal carcinomas (e.g. HPV16, HPV18).
  • the beta-, gamma-, mu- and nu-HPVs are characterized by a strict cutaneous tropism.
  • Both alpha- and beta-HPV classes comprise genotypes which are associated with low and high risks of cancer.
  • High risk alpha-HPVs are mostly associated with mucosal cancers and are known to cervix, anus, vulva, vagina, and penis.
  • the beta-type HPVs have been recognized to play an important role in inflammatory processes in cutaneous squamous cell carcinoma (cSCC) (Tampa et al. 2020).
  • Serological analysis of cSCC patients indicated an association between cSCC and HPV 8 and 17.
  • the first evidence that HPV may play a role in SCC came from observations of patients with epidermodysplasia verruciformis (EV), who are genetically predisposed to HPV infection and develop SCC harboring specific beta- HPV types (notably HPV5, HPV8, HPV20) on sites exposed to ultraviolet radiation (Tessari G, et aL, 2012).
  • EV epidermodysplasia verruciformis
  • Beta-HPV (beta- 1 , beta-2, and beta-3 species) are the most commonly identified in SCC lesions according to results obtained from both serological tests and molecular DNA detection. Serological tests identified HPV 8 (beta-1 ), HPV 15, HPV 17, HPV 38 (beta-2), HPV 49, and HPV 76 (beta-3) in patients with SCC in a higher proportion than in the control group (Tampa et al, 2016).
  • beta-type HPVs have been especially identified in the patient subgroup of organ transplant recipient.
  • Skin cancer is the most frequent malignancy in organ transplant recipients, 95% of which are nonmelanoma skin cancer (NMSC), especially squamous cell carcinoma (SCC) and basal cell carcinoma (BCC).
  • NMSC nonmelanoma skin cancer
  • SCC squamous cell carcinoma
  • BCC basal cell carcinoma
  • NMSC nonmelanoma skin cancer
  • SCC squamous cell carcinoma
  • BCC basal cell carcinoma
  • Beta- HPV DNA has been detected in SCC from transplanted patients (Tessari G, et aL 2012). Transcripts of HPV 8, 9, and 15 were found in squamous cell carcinoma, suggesting an active role of human papillomaviruses in the pathogenesis of these lesions (O'Reilly Zwald et aL, 2011 )
  • Vaccine development has entirely focused on the development of vaccines against alpha-type HPV infections.
  • Several vaccines have been developed for the protection against high risk alphatype HPV infections based in the HPV L1 protein.
  • the two first approved vaccines Cervarix and Gardasil comprise L1 -VLP.
  • the bivalent Cervarix contains VLP of high risk HPV16 and 18 and the quadrivalent Gardasil contains VLP of high risk HPV16,18, 6 and 11.
  • L1 based development strategy an additional development strategy based on neutralization epitopes found at the N-terminus of the HPV L2 protein between aa 11 -200, which is highly conserved between different alpha-HPV types was pursued.
  • L2 based vaccines using the epitope of aa 17 to 36 of HPV16 or HPV31 were investigated with regard to the induction of cross-neutralization antibodies against other alpha-HPV types (Schellenbacher et al., 2017).
  • WO 2012/031760 A1 discloses a vaccine consisting of AAV virus like particles constituted by an AAV2 VP3 protein comprising one insertion of each of aa 17 to 36 of HPV16 or HPV31. It could be shown that the vaccine induced cross-reactive antibodies against other alpha-HPV serotypes, such as HPV18, HPV45, HPV52 and HPV58, in animals. However, induction of antibodies directed against beta- or gamma-type HPVs was not shown.
  • WO 2010/118424 A1 discloses a vaccine candidate based on a combination of the HPV L1 and L2 protein in a fusion protein wherein aa 17 to 36 of HPV16 were inserted between aa 133 and 134 of the L1 protein of bovine papillomavirus (BPV) type 1 .
  • BBV bovine papillomavirus
  • This combined L1 and L2 approach induced antibodies against HPV types 16, 18, 45, 52, 58, 6, 11 , and 5.
  • Induction of antibodies against beta- and gamma-types HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV95, HPV96, HPV4, or HPV50 is not disclosed.
  • the invention relates to a mutated parvovirus structural protein comprising at least one insertion comprising a sequence of at least six consecutive amino acids from amino acids 10 to 40 of the HPV16 or HPV31 L2 protein for use in the treatment or prevention of a betatype or gamma-type HPV infection selected from a HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV95, HPV96, HPV4, and HPV50 infection.
  • a betatype or gamma-type HPV infection selected from a HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV95, HPV96, HPV4, and HPV50 infection.
  • beta-type or gamma-type HPV is selected from an infection with HPV17, HPV20, HPV24, HPV36, HPV49, HPV92, HPV96, and/or HPV50 infection.
  • amino acids sequence that is derived from a reference protein means that said the type and order of amino acids in said sequence correspond to the type and order of amino acids in the referenced protein sequence.
  • amino acid sequence does not have to be materially derived, such as for example by direct cloning steps, form the referenced sequence.
  • the mutated parvovirus structural protein according to the invention is used in the treatment or prevention of a beta-type or gamma-type HPV infection by at least two, at least three, at least four, at least five, at least six, at least seven, or at least eight HPV stereotypes selected from HPV4, HPV5, HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV95, HPV96, and HPV50.
  • an infection by at least one beta-type and at least one gamma-type HPV selected from HPV4, HPV5, HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV95, HPV96, and HPV50 is prevented or treated.
  • an infection by HPV4, HPV5, HPV20, HPV38, HPV92, and HPV95 is prevented or treated.
  • an infection by HPV5, HPV15, HPV20, HPV36, HPV38, HPV59, HPV76, and HPV92 is prevented or treated.
  • an infection by HPV4, HPV5, HPV15, HPV20, HPV36, HPV38, HPV59, HPV76, HPV92, and HPV95 is prevented or treated.
  • the least one insertion comprised in the mutated parvovirus structural protein may comprises at least 6, at least 7, at least 8, at least 9, at least 10, at least 11 , at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, most preferably at least 20 consecutive amino acids from amino acids 10 to 40 of the HPV16 or HPV31 L2 protein.
  • the insertion comprises consecutive amino acids from amino acids 17 to 36 of the HPV16 or HPV31 L2 protein.
  • the insertion comprises consecutive amino acids from amino acids 17 to 36 of the HPV16 L2 protein.
  • the HPV16 L2 protein from which the insert sequence is derived according to the present invention is the late protein L2 of HPV16 and may be characterised by the sequence according to GenBank accession number AVN82798.1.
  • the HPV31 L2 protein from which the insert sequence is derived according to the present invention is the late protein L2 of HPV31 and may be characterised by the sequence according to GenBank accession number QNM76585.1.
  • the mutated parvovirus structural protein comprises an insertion comprising an amino acids sequence selected from QLYKTCKQAGTCPPDIIPKV (SEQ ID NO: 1 ) and/or QLYQTCKAAGTCPSDVIPKI (SEQ ID NO: 2).
  • the mutated parvovirus structural protein comprises at least two insertions wherein each insertion comprises either SEQ ID: 1 or SEQ ID: 2.
  • the insertion comprises an amino acid sequence of least 20 consecutive amino acids having at least 80%, at least 85%, at least 90%, or at least 95% amino acid sequence identity in comparison to a consecutive amino acid sequence selected from amino acids 10 to 40 of the HPV16 or HPV31 L2 protein.
  • a "mutated" parvovirus structural protein within the present invention is a protein which comprises at least the afore described insertion in comparison the respective wild-type parvovirus structural protein.
  • the mutated parvovirus structural protein may comprise additional mutations, such as substitutions, insertions, and/or deletions as described in the following.
  • the amino acid sequence comprised in insertion comprises at least one mutation in comparison to SEQ ID NO: 1 and/or SEQ ID NO: 2.
  • the insertion comprising at least one mutation may have at least 80%, at least 85%, at least 90%, or at least 95% amino acid sequence identity to SEQ ID NO: 1 and/or SEQ ID NO: 2.
  • a mutation within an amino acid sequence or in a nucleotide sequence may be at least one substitution, insertion or deletion.
  • a substitution at least one amino acid or nucleotide is exchanged against another amino acid or a nucleotide in the mutated sequence in comparison to the respective wild type or comparator sequence.
  • an insertion at least one amino acid or nucleotide is inserted into the mutated sequence in comparison to the respective wild type or comparator sequence.
  • a deletion at least one amino acid or nucleotide is omitted in the mutated sequence in comparison to the respective wild type or comparator sequence.
  • substitution may be a conservative amino acid substitution in the primary sequence.
  • conservative substitution is intended to embrace the act of replacing one or more amino acids of a protein or peptide with an alternative amino acid with similar properties and which does not substantially alter the physical -chemical properties and/or structure of function of the native protein. Analogues of this type are also encompassed within the scope of this invention.
  • substitute amino acids may be selected from other members of the class to which the amino acid belongs.
  • non-polar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, glycine, proline, phenylalanine and tryptophan.
  • Polar neutral amino acids include serine, threonine, cysteine, tyrosine, asparagine and glutamine.
  • the positive charged (basic) amino acids include arginine, lysine and histidine.
  • the negative charged (acidic) amino acids include aspartic acid and glutamic acid. Examples of preferred conservative substitutions include Lys for Arg and vice versa to maintain a positive charge; Glu for Asp and vice versa to maintain a negative charge; Ser for Thr so that a free OH is maintained; and Gin for Asn to maintain a free NH 2 .
  • the parvovirus structural protein according to the invention may be derived from an adeno- associated virus (AAV), Goose parvovirus, Duck parvovirus, Snake parvovirus, feline panleukopenia virus, canine parvovirus, B19 or minute virus of mice (MVM) and may be mutated as described herein. Due to the high conservation of genome organization amongst the parvoviruses, the invention can easily be transferred to other parvovirus members.
  • structural protein according to the invention may be derived from a parvovirus that shares the general capsid assembly from viral proteins VP1 , VP2 and VP3. Structural proteins derived from these viruses are generally advantageous since they enable a virus-like particle (VLP) production only from VP3 as described below.
  • VLP virus-like particle
  • viruses of this subgroup include AAV, Goose parvovirus, Duck parvovirus, and Snake parvovirus.
  • AAV is selected from the group consisting of bovine AAV (b-AAV), canine AAV (CAAV), mouse AAV1 , caprine AAV, rat AAV, avian AAV (AAAV), AAV1 , AAV2, AAV3b, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV1 1 , AAV12, and AAV13, especially AAV2.
  • the mutated parvovirus protein is derived from AAV2.
  • the human immune system in general is well adapted to AAV2 capsid proteins as the largest fraction of the human population is infected with this virus that is not associated with any disease.
  • AAV2 as a gene therapy vector has been tested in large number of human patients and appeared not to be associated to immunological complications or other safety concerns. Accordingly, compared to other backbones aiming at put B-cell epitopes into a multimeric structure, AAV2 has the enormous advantage that the backbone itself, for most of the vaccinated humans, will not generate an unprecedented immune reaction that may cause autoimmune diseases in vaccinated humans.
  • the mutated parvovirus structural protein according to the present invention may be capable of forming a multimeric structure, wherein the insertion is located on the surface of said multimeric structure.
  • the multimeric structure may for example be a capsomer, a VLP or a virus. Epitopes presented by ordered, multivalent, highly repetitive, and often rigid structures of viruses or VLPs can lead to a strong stimulation of B-cells and the induction of robust and long-lasting antibody responses due to extensively crosslink B-cell receptors.
  • AAV derived VLPs comprising epitopes from the alpha HPV types HPV16 or HPV31 can induce cross-reactive antibodies against other alpha HPV serotypes, such as HPV18, HPV45, HPV52 and HPV58
  • the inventors surprisingly found that the respective VLPs can induce an antibody response which is cross specific against beta and gamma-HPV types, such as HPV15, HPV20, HPV36, HPV38, HPV49 and HPV92.
  • HPV15, HPV36, HPV38, and HPV49 all have a low sequence identity of 60% to 65% or lower within aa 17 to 36 of the L2 protein in comparison to the HPV16.
  • the respective sequence identity of HPV4, HPV95, and HPV50 is only 50%.
  • HPV18, HPV45, HPV52 and HPV58 have a sequence identity of 75% to 80% in comparison to aa 17 to 36 of the HPV16 L2.
  • the parvovirus mutated structural protein is a mutated VP3 protein. It was previously shown (WO 2010/099960 A2) that multimeric structures useful as vaccines can be generated based upon multimeric structures consisting essentially of VP3. The use of multimeric structures comprising only a single structural protein is generally considered advantageous, since clinical development of vaccines based on multimeric structures is simplified for products based on a single active compound/protein and being as pure as possible. With respect to e.g. VLPs this is a problem in general, as viruses are often composed of more than one protein and are capable of packaging specifically viral DNA or unspecifically DNA from the host cell.
  • VLPs that contain as few different proteins as possible and preferably no nucleic acid.
  • vaccines containing VP1 , VP2 and VP3 are generally produced in the presence of the parvoviral Rep protein.
  • Rep does not only represent a further protein that is attached to VLPs but is also held responsible for packaging of virus genomes and unspecific DNA into preformed capsids (King et aL, 2001 ).
  • Packaging of DNA is to be avoided as VLPs potentially can enter cells of a patient and thereby transfect such contaminating DNA, which may cause all sorts of unwanted effects.
  • Virus-like particles comprising a mutated parvovirus structural protein derived from VP3, which is not N-terminally extended by at least parts of the VP3 sequence, as the only structural protein may be obtained by expressing a mutated parvovirus structural protein derived from VP3 in a cell under control of a Rep-independent promoter. Additionally, a polypeptide designated “assembly activating protein” (AAP) is expressed according to methods as disclosed in Stanford et aL, 2010
  • WO 2010/099960 A2 which allows for high yields, e.g. approximately about 10 , preferably 6 7 about 10 , and more preferably about 10 virus particles to be formed per cell.
  • the mutated parvovirus structural protein derived from VP3 of a certain virus type may preferably be coexpressed with the corresponding AAP protein from said virus type (Sonntag et aL, 2010 or WO 2010/099960 A2).
  • an AAP from a closely related virus type may be used.
  • the sequence encoding AAP may be provided either in cis or in trans to assemble capsids consisting essentially of VP3.
  • Virus particle titers can be quantified from lysates of transfected cells (see above) in their undiluted form or in a dilution using a commercially available titration ELISA kit which is based on the binding of the monoclonal antibody A20 to the viral capsid in an assembled state to measure the virus particle concentration. Since the antibody A20 does not bind to the capsid of e.g. a different virus serotype, particle titers can be visualized by electron microscopy and quantified by counting. To analyze protein expression and estimate its amount cell lysates of identical portions of transfected cells can be processed for SDS-PAGE.
  • proteins Upon gel electrophoresis and transfer to a nitrocellulose membrane, proteins can be probed using binders specific to the target protein (e.g. monoclonal antibodies B1 , A69, anti-GFP). The amount of protein translation can be estimated from the amount of binders that specifically bind to the protein. These complexes can be visualized and quantified by e.g. immunohistochemical staining, immunofluorescent staining or radioactive labeling.
  • binders specific to the target protein e.g. monoclonal antibodies B1 , A69, anti-GFP.
  • the amount of protein translation can be estimated from the amount of binders that specifically bind to the protein.
  • These complexes can be visualized and quantified by e.g. immunohistochemical staining, immunofluorescent staining or radioactive labeling.
  • the virus-like particles may preferably be obtained from culture supernatant.
  • Obtaining virus-like particles from the culture supernatant advantageously supersedes the cell lysis step in the manufacturing and facilitates the purification of the particles.
  • the insertion(s) is (are) inserted into one or more positions selected from the group consisting of 1-261 , I-266, 1-381 , I-447, I-448, I-453, I-459, I- 471 , I-534, I-570, I-573, I-584, I-587, I-588, 1-591 , I-657, I-664, 1-713 and 1-716, preferably 1-261 , I-453, I-534, I-570, I-573 and I-587, more preferably I-453, I-534 and I-587, especially I-453 and I-587.
  • a position of insertion is also referred to as “insertion site” within the context of the present disclosure.
  • the used nomenclature I-### refers to the insertion site with ### naming the amino acid number relative to the VP1 protein of AAV2, however meaning that the insertion may be located directly N- or C-terminal, preferably directly C-terminal of one amino acid in the sequence of 5 Amino acids N- or C-terminal of the given AA, preferably 3, more preferably 2, especially 1 AA(s) N- or C-terminal of the given AA.
  • the corresponding insertion sites can be identified by performing an amino acid alignment or by comparison of the capsid structures, if available.
  • the amino acid position after which the insertion was introduced and which named the site is underlined. It is also possible likewise to introduce an insertion into the five directly adjacent amino acids located next to the underlined AA, because these are likewise located within a loop in the AAV2 capsid.
  • the insertion site I-587 corresponds to an insertion before and/or after one of the following Amino acids indicated by emphasis: FQSSS_TDPAT in AAV1 , LQRGN 5 87 RQAAT in AAV2, LQSSN_TAPTT in AAV3b, LQSSS_TDPAT in AAV6, LQAATAAQT in AAV7, LQQQN_TAPQI in AAV8, LQQAN_TGPIV in AAV10, NQNATLTAPIT in AAV1 1 , and NQSST TAPAT in AAV5.
  • the insertion site I-453 corresponds to an insertion directly N- or C-terminal of the following ten amino acids each, preferably directly C-terminal of the amino acid indicated by emphasis QNQSG_SAQNK in AAV1 , NTPSG453 TTTQS in AAV2, GTTSG_TTNQS in AAV3b, QNQSG.SAQNK in AAV6, SNPGGJAGNR in AAV7, GQTTG.TANTQ in AAV8, QSTGG.TQGTQ in AAV10, LSGELNQGNA in AAV11 and FVSTN_NTGGV in AAV5.
  • the parvovirus mutated structural protein of the invention comprises two or more insertions, each comprising at least one amino acid sequence of at least six consecutive amino acids from amino acids 10 to 40 of the HPV16 or HPV31 L2 protein, or specific sequences of inserts as described herein, wherein each of the inserts is inserted at a different insertion site of the parvovirus mutated structural protein, preferably wherein one insertion is at position I-587 and one is at position I-453.
  • the parvovirus mutated structural protein comprises at least one insert comprising amino acids from HPV16 L2 protein as described above and one insert comprising amino acids from HPV31 L2 as described above.
  • an insert comprising amino acids from HPV16 L2 protein is inserted at I-587 and an insert comprising amino acids from HPV31 L2 protein is inserted at I-453. More preferably, an insert comprising amino acids QLYKTCKQAGTCPPDIIPKV (SEQ ID: 1 ) is inserted at I-587 and an insert comprising amino acid QLYQTCKAAGTCPSDVIPKI (SEQ ID: 2) inserted at I-453.
  • an insert comprising amino acids QLYKTCKQAGTCPPDIIPKV (SEQ ID: 1 ) is inserted at I-453 and I-587.
  • an insert comprising amino acid QLYQTCKAAGTCPSDVIPKI (SEQ ID: 2) is inserted at I-453 and I-587.
  • the insertion may additionally preferably comprise on its N- and/or C terminus a linker sequence which preferably has a length of 2 to 10, more preferably 3 to 6 amino acids.
  • the linker comprises or consists of small neutral or polar amino acids (A, G, S, C), which support the inserted epitope to be well accessible to the immune system.
  • A, G, S, C small neutral or polar amino acids
  • C has the advantage that two Cs on both sides of the linker may be able to form a hydrogen bond. Therefore, it is envisaged that both the N-terminal and C-terminal linker contain at least one C.
  • the linker sequence(s) is (are) composed of A, G and S.
  • none of the 5 amino acids directly adjacent to the insertion is R and none of the amino acids of the linker, if present, is R.
  • R in close proximity to the insertion reduces yield of the mutated structural protein/the multimeric structures composed of the mutated structural protein during expression and purification, and therefore is preferably avoided. Accordingly, the Rs at position 585 and 588 of AAV2 have been substituted for example by A.
  • the parvovirus mutated structural protein comprises one or more additional mutations selected from an insertion, a deletion, a N- or C-terminal fusion of a heterologous amino acid sequence and a substitution, particularly a single-amino-acid exchange, or a combination of these, preferably a mutation of R585 of AAV2 and/or R588 of AAV2, especially a single-amino- acid exchange R585A of AAV2 and/or R588A of AAV2.
  • the insertion of epitopes at position I-453 of AAV2 as described in WO 2008/145401 A2 leads to the generation of an R within the linker downstream of the insertion (see example 6.4.3, page 103, lines 12 and 14) due to the generation of a useful endonuclease restriction site.
  • Parvovirus mutated structural proteins where this R was substituted for a small neutral or polar amino acid lead to considerably higher yield of VP3 only AAV virus-like particles (AAVLPs) during expression and subsequent purification. Therefore, it is preferred, that the linkers, if present, do not contain an R, especially that the linker directly downstream of the inserted epitope at I-453 does not contain an R.
  • the parvovirus mutated structural protein of the invention is an AAV2 VP3 protein having SEQ ID: 1 inserted at I-587 and SEQ ID: 2 inserted at I-453 characterized by the following sequence (AAV2-VP3-HPV(16/31 L2)):
  • the invention relates to a multimeric structure comprising parvovirus mutated structural proteins as described above, particularly comprising at least 5, preferably at least 10, more preferably at least 30, most preferably at least 60 structural protein.
  • Such multimeric structure may be a capsomer, a VLP or a virus.
  • Capsomers are multimeric subunits of a viral capsid, typically consisting of 5-6 capsid proteins (pentamers and hexamers).
  • VLPs are empty viruses, meaning that they do not comprise genetic material such as a viral genome or relevant part thereof.
  • the multimeric structures may be aggregates with amorphous structures with no symmetric order.
  • the insertion as described above, or mutants thereof, is located on the surface of the multimeric structure.
  • nucleic acid coding for a parvovirus mutated structural protein of the invention such as DNA, RNA etc.
  • the nucleic acid is an mRNA encoding.
  • Methods of designing mRNAs of proteins suitable for use as a vaccine are known in the art. Suitable modifications of an mRNA according to the invention are for example disclosed by Kim et al 2021 , which is incorporated herein by reference.
  • a further embodiment of the present invention is a vector, e.g . a virus that comprises a nucleic acid encoding the parvovirus mutated structural protein of the invention.
  • virus may be infectious or inactive, for example it may have been inactivated through standard techniques such as attenuation or irradiation.
  • the present invention is a cell comprising a nucleic acid coding for the parvovirus mutated structural protein as described above.
  • Such cell can be a bacterium, preferably E. coli, a yeast cell, preferably s. cerevisiae, hansenula polymorpha or pichia pastoris, k. lactis, an insect cell, preferably SF-9, S2, SF+ or High5, or a mammalian cell, preferably HeLa, 293, VERO, PERC6, BHK or CHO.
  • the parvovirus mutated structural proteins of the invention can be prepared by a method comprising the steps of a) producing the structural protein by cultivating the cell according to the invention under suitable conditions thereby expressing the nucleic acid of the invention, and optionally co-expressing a nucleic acid encoding an AAP, and b) optionally isolating the expressed parvovirus mutated structural protein produced in step a).
  • essentially only VP3 is expressed, leading to multimeric structures comprising essentially only VP3.
  • Expression and purification according to this method may for example be performed in accordance with Example 1 of this application.
  • Expression of parvovirus mutated structural proteins comprising an insertion and purification of the obtained AAVLPs is furthermore disclosed in WO 2012/031760 A1 , Example 1 , for mammalian cells or by WO 2010/099960 A2, Example 1 , for insect cells.
  • compositions comprising at least one parvovirus mutated structural protein according to the invention and/or a nucleic acid according to the invention, and/or preferably at least one multimeric structure according to the invention.
  • the invention relates to a parvovirus mutated structural protein according to the invention and/or a nucleic acid according to the invention, preferably a multimeric structure according to the invention, for use as a medicament. Furthermore, the invention relates to a composition comprising at least one parvovirus mutated structural protein according to the invention and/or a nucleic acid according to the invention, preferably at least one multimeric structure according to the invention, for use as a medicament.
  • the medicament may preferably be used as a vaccine comprising at least one parvovirus mutated structural protein of the invention and/or a nucleic acid of the invention, preferably at least one multimeric structure of the invention.
  • the mutated parvovirus structural protein or the medicament according to the invention may be used in the treatment or prevention of a cancerous or inflammatory disease.
  • the cancerous disease is a skin cancer, preferably a nonmelanoma skin cancer (NMSC). More preferably, the NMSC is cutaneous squamous cell carcinoma (cSCC) and/or basal cell carcinoma (BCC).
  • NMSC nonmelanoma skin cancer
  • BCC basal cell carcinoma
  • the cancer may preferably be a cancer of the oral cavity, preferably tongue cancer, nasal cancer, breast cancer, thyroid cancer.
  • the inflammatory disease is psoriasis, rheumatoid arthritis, lupus, or vitiligo.
  • HPV infections especially pose a risk for immunosuppressed subjects, as for example subjects treated with immunosuppressive therapeutic agents.
  • a subject may for example be treated with immunosuppressive therapeutic agents before or after organ transplantation.
  • subjects diagnosed with epidermodysplasia verruciformis also known as Lewandowsky-Lutz-Dysplasia or Lutz-Lewandowsky epidermodysplasia verruciformis (EV)
  • EV Lutz-Lewandowsky epidermodysplasia verruciformis
  • mutated parvovirus structural proteins comprised in VLPs are especially suitable for inducing a strong immune response and thus may in certain embodiments be suitable for treating immunosuppressed subjects, or prevent an infection of beta- or gamma-type HPV.
  • the mutated parvovirus structural protein may be used for the treatment or prevention of an HPV infection in a subject diagnosed with EV, a subject treated with an immunosuppressive therapeutic agent, or a subject that has received or will receive an organ
  • the composition, medicament encompasses pharmaceutically acceptable carriers and/or excipients.
  • the pharmaceutically acceptable carriers and/or excipients useful in this invention are conventional and may include buffers, stabilizers, diluents, preservatives, and solubilizers.
  • the nature of the carrier or excipients will depend on the particular mode of administration being employed.
  • parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol, citric acid or the like as a vehicle.
  • pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol, citric acid or the like as a vehicle.
  • conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate.
  • pharmaceutical compositions to be administered can contain minor amounts of non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, and pH buffering agents and the like, for example sodium acetate or sorbitan monolaurate.
  • the composition may comprise a buffering substance, for example sodium citrate, a salt, for example MgCls, and a surfactant, for example pluronic F-68, in addition to the parvovirus protein.
  • a buffering substance for example sodium citrate
  • a salt for example MgCls
  • a surfactant for example pluronic F-68
  • the composition comprises 20 to 250 mM sodium citrate, 1 to 5 mM MgCh, 0.0001 % to 0.01 % pluronic F-68, at a pH from 5.5 to 7.0.
  • the composition may comprise about 10OmM sodium citrate, about 2.5 mM MgCls, about 0.001 % pluronic F-68, at a pH of about 6.0.
  • the composition, medicament or vaccine may further comprise an immunostimulatory substance such as an adjuvant.
  • the adjuvant can be selected based on the method of administration and may include mineral or plant oil-based adjuvants, Montanide incomplete Seppic adjuvant such as ISA, oil in water emulsion adjuvants such as the Ribi adjuvant system, syntax adjuvant formulation containing muramyl dipeptide,.
  • the adjuvant is an oil-based adjuvant, such as ISA206 (SEPPIC, Paris, France), or ISA51 or ISA720 (SEPPIC, Paris, France).
  • the parvovirus mutated structural protein is co-formulated with at least one suitable adjuvant such as Imidazoquinolines, MPL, MDP, MALP, flagellin, LPS, LTA, or cholera toxin or derivative thereof, saponins, QS21 , ISCOMs, CFA, SAF, MF59, adamantanes, or a cytokine.
  • suitable adjuvant such as Imidazoquinolines, MPL, MDP, MALP, flagellin, LPS, LTA, or cholera toxin or derivative thereof, saponins, QS21 , ISCOMs, CFA, SAF, MF59, adamantanes, or a cytokine.
  • the immunostimulatory substance is selected from the group comprising polycationic polymers, especially polycationic peptides such as polyarginine, immunostimulatory oligodeoxynucleotides (ODNs), peptides containing at least two LysLeuLys motifs, especially KLKLLLLLKLK, neuroactive compounds, especially human growth hormone, aluminum salt adjuvants, such as aluminum hydroxide or aluminum phosphate, preferably aluminum hydroxide, adjuvants or combinations thereof.
  • polycationic polymers especially polycationic peptides such as polyarginine, immunostimulatory oligodeoxynucleotides (ODNs), peptides containing at least two LysLeuLys motifs, especially KLKLLLLLKLK, neuroactive compounds, especially human growth hormone, aluminum salt adjuvants, such as aluminum hydroxide or aluminum phosphate, preferably aluminum hydroxide, adjuvants or combinations thereof.
  • the combination is either a polycationic polymer and immunostimulatory deoxynucleotides or of a peptide containing at least two LysLeuLys motifs and immunostimulatory deoxynucleotides.
  • the polycationic polymer is a polycationic peptide.
  • the immunostimulatory substance is at least one immunostimulatory nucleic acid. Immunostimulatory nucleic acids are e.g.
  • CpG neutral or artificial cytosine-guanine dinucleotide
  • nucleic acids short stretches of nucleic acids derived from non- vertebrates or in form of short ODNs containing non -methylated CpGs in a defined base context (e.g. as described in WO 96/02555).
  • nucleic acids based on inosine and cytidine as e.g. described in WO 01/93903, or deoxynucleic acids containing deoxy-inosine and/or deoxyuridine residues (described in WO 01/93905 and WO 02/095027) may preferably be used as immunostimulatory nucleic acids in the present invention.
  • mixtures of different immunostimulatory nucleic acids are used in the present invention.
  • the aforementioned polycationic compounds may be combined with any of the immunostimulatory nucleic acids as aforementioned.
  • such combinations are according to the ones described in WO 01/93905, WO 02/32451 , WO 01/54720, WO 01/93903, WO 02/13857 and WO 02/095027.
  • composition, medicament or vaccine may not comprise a further immunostimulatory substance such as an adjuvant as described above.
  • the AAV backbone itself has a strong immune stimulatory property.
  • the structural protein, composition, medicament or vaccine according to the invention may be administered to a subject in need thereof, preferably a mammal, most preferably a human, in any conventional manner, including different routes, e.g. by intravenous, intraperitoneal, intra-lymph node, subcutaneous, intradermal, intramuscular, topical, intranasal or intrabronchial administration.
  • the composition, medicament or vaccine is administered subcutaneous or intramuscular.
  • the mutated parvovirus structural protein, composition, medicament or vaccine according to the invention may be administered to a subject which has not been infected with at least one HPV.
  • the structural protein, composition, medicament may be administered to a subject which has already been infected with an HPV, preferably with an HPV selected from HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV4, and HPV50, but has not been infected with another HPV selected from HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV4, and HPV50.
  • a broad cross- reactive immune response against different beta- and gamma-HPV types may be induced although the subject has already been infected with a specific HPV serotype.
  • the volume of each dose for administration is preferably up to about 5 ml, still more preferably between 1 ml and 3 ml, and most preferably about 2 ml.
  • the volume of the dose when intramuscular injection is the selected administration route is preferably up to about 5 ml, preferably up to 3 ml, preferably between 1 ml and 3 ml, more preferably between 0.5 ml and 2 ml, and most preferably about 1 ml.
  • the amount of vaccine in each dose should be enough to confer effective immunity HPV4, HPV5, HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV95, HPV96, and/or HPV50 and decrease the risk of developing clinical signs associated with the autoimmune disease the patient is suffering from or has a chance of developing, or prevents or reverts organ transplant rejection to a subject receiving a vaccination therewith.
  • the unit dose of protein or nucleic acid should be up to about 5 pg protein/kg body weight, more preferably between about 0.2 to about 3 pg protein/kg body weight, still more preferably between about 0.3 to about 1 .5 pg protein/kg body weight, more preferably between about 0.4 to about 0.8 pg protein/kg body weight, and still more preferably about 0.6 pg protein/kg body weight.
  • the protein may be administered at about 1 pg to about 1 mg, from about 5 pg to about 500 pg, from about 10 pg to about 250 pg, or from about 25 pg to about 100 pg .
  • the protein may be administered at a total dose of about 1 to about 50 pg, preferably about 10 to about 35 pg, more preferably about 15 to about 25 pg, most preferably about 20 pg per administration and patient.
  • Alternative preferred unit doses could be up to about 6 pg protein or nucleic acid/kg body weight, more preferably between about 0.05 to 5 pg, still more preferably between about 0.1 to 4 pg.
  • the dose is administered at least 1 time, at least 2 times, at least 3 times, at least 4 times, at least 5 times, or at least 6 times. Preferably the dose is administered 1 to 3 times, more preferably 2 to 3 times, most preferably 3 times.
  • the doses may be administered with an interval of about 20 days to about 180 days, preferably about 40 days to about 150 days, more preferably about 57 days to about 120 days between two administrations. In a preferred embodiment, the doses are administered with an interval of about 57 days between the first and the second administration and an interval of about 120 days between the second and the third administration.
  • the invention relates to a use as a vaccine, method for vaccination and/or for treating or preventing the HPV infections or diseases specified herein by administering to a patient, preferably a mammal, most preferably a human, an effective amount of a parvovirus mutated structural protein, nucleic acid, composition, medicament or vaccine according the invention.
  • the parvovirus mutated structural protein, composition or vaccine according to the invention can be used in a method of preventing or treating a HPV4, HPV5, HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV95, HPV96, and/or HPV50 infection or a cancerous or inflammatory disease preferably a skin cancer, more preferably a nonmelanoma skin cancer (NMSC), such as squamous cell carcinoma (SCC) basal cell carcinoma (BCC), cancer of the oral cavity, preferably tongue cancer, nasal cancer, breast cancer, thyroid cancer.
  • NMSC nonmelanoma skin cancer
  • SCC squamous cell carcinoma
  • BCC basal cell carcinoma
  • an "effective amount" of a parvovirus mutated structural protein, nucleic acid, composition, medicament or vaccine may be calculated as that amount capable of exhibiting an in vivo effect, e.g. preventing or ameliorating a sign or symptoms. Such amounts may be determined by one of skill in the art.
  • the invention relates to a method of generating antibodies binding to HPV15, HPV17, HPV20, HPV24, HPV36, HPV38, HPV49, HPV76, HPV92, HPV4, or HPV50 in a subject, wherein the method comprises administering a mutated parvovirus structural protein according to any of claims 1 to 10 to the subject.
  • antibodies binding to a certain HPV refers to antibodies that bind to the surface of said HPV virus.
  • a mutated parvovirus structural protein according to the invention comprising inserts derived from the alpha-types HPV16 and HPV31 induces an immune response in human subjects directed against beta- and gamma-HPV types.
  • antisera derived from such human subjects are reactive against a variety of beta- and gamma-HPV types, including HPV5, HPV15, HPV20, HPV36, HPV92, HPV76, HPV49, HPV38.
  • a virus neutralising activity of the antisera from rabbits was confirmed by pseudoviron based HPV neutralisation assays for both beta and gamma type HPVs as show in Example 3 and Figures 5 and 6.
  • the protein according to the invention induces an immune response, which is suitable for the prevention and the treatment of beta- and gamma-HPV infections.
  • the preventive properties of the antisera obtained from vaccinated subjects was further confirmed in vivo in a passive transfer animal study.
  • Figure 1 shows the detection of the average increased HPV5L2, HPV15L2, HPV20L2, HPV36L2, HPV92L2, HPV76L2, HPV49L2, HPV38L2 specific antibody titers (ng/mL) from baseline in humans vaccinated with AAVLP-HPV(16/31 L2). Data from day 57, 71 , and day 193 are shown.
  • Figure 2 shows the detection of increased HPV5L2, HPV15L2, HPV20L2, HPV36L2, and HPV76L2-specific antibody titers (ng/mL) from baseline in humans vaccinated with AAVLP- HPV(16/31 L2). Data of individual subjects from day 71 and day 193 are shown. Mean titers of each group are shown by horizontal bars with SD.
  • Figure 3 shows geometric mean titers (95%-CI) for the group receiving active drug (Active) or placebo (Placebo) for HPV16 (Figure 3A), HPV31 (Figure 3B), HPV5 (Figure 3C), HPV15 (Figure 3D), HPV20 (Figure 3E), HPV38 (Figure 3F), HPV76 ( Figure 3G).
  • Active active drug
  • placebo placebo
  • Figure 4 shows the relative treatment effect in the actively vaccinated group (Active) versus the placebo group (Placebo)
  • Figure 5 shows in vitro pseudoviron-based neutralization of beta-HPV types HPV5, HPV20, HPV38, and HPV92 by serum form AAVLP-HPV(16/31 L2) immunized rabbits.
  • Figure 6 shows in vitro pseudoviron-based neutralization of gamma-HPV types HPV4 and HPV95 by serum form AAVLP-HPV(16/31 L2) immunized rabbits.
  • Figure 7 shows an in vivo pseudovirion HPV challenge (HPV76 (A); HPV5 (B) ; HPV16 (C); HPV31 (D)) of mice where human antiserum have been passively transferred to the mice.
  • HPV76 A
  • HPV5 B
  • HPV16 C
  • HPV31 D
  • Example 1 Generation of antisera against AAVLP-HPV(16/31 L2)
  • AAVLP-HPV(16/31 L2) comprising the mutated parvovirus structural protein according to SEQ ID NO:3 may be cloned, expressed and purified in accordance with the methods disclosed by Nieto et al. 2012. Further to the purification disclosed therein, an AVB affinity column may be used according to known procedures to further improve purity.
  • NCT03929172 In a 12-month single-center, placebo controlled, double-blinded Phase 1 study (NCT03929172), twenty (20) healthy male and female subjects (>40% of each gender) were randomized to receive active drug (sixteen) or placebo (four). Subjects received a total of 3 doses of AAVLP(HPV16/31 L2) or placebo on Days 1 , 57 ( ⁇ 2 days), and 180 ( ⁇ 1 week) without adjuvant.
  • samples were allowed to clot at room temperature (for at least 30 minutes). The content of the tubes is centrifuged under refrigeration. Within 120 minutes of collection, serum samples are divided into aliquots and stored in suitably labelled tubes at - 20 ⁇ 5°C, pending assay. During the study, some samples were shipped in separate shipments with sufficient amount of dry ice to the analytical laboratory for analysis. Subsequent shipments were sent after receipt in good condition of the previous shipment at the analytical laboratory.
  • Peptides ELISAs were performed to analyze the specific immune response against beta-HPV types HPV5L2, HPV15L2, HPV17L2, HPV20L2, HPV36L2, HPV92L2, HPV96L2, HPV76L2, HPV49L2, HPV38L2 and gamma-HPV type HPV50L2.
  • Anti-beta-HPV-specific IgG-antibodies were measured by ELISA.
  • F96 streptavidin pre-coated microplates (Nunc, Thermo Scientific) were prewashed and incubated overnight at 4°C with 1 pg/well of either peptide:
  • HPV5L2 (“HIYQTCKQAGTCPPDVINKV”); HPV15L2 (“DIYRGCKQAGTCPPDVLNKV”);
  • HPV17L2 (“DIYRGCKQAGTCPPDVINKV”);
  • HPV20L2 (“NIYRTCKQAGTCPPDVINKV”);
  • HPV36L2 (“HIYQTCKQAGTCPPDWNKV”);
  • HPV50L2 (“DLYRSCLQGGDCIPDVQNKF”);
  • HPV92L2 (“NIYRTCKAAGTCPPDVVNKV”);
  • HPV96L2 (“NIYRGCKAAGTCPPDVINKV);
  • HPV76L2 (“HIYQSCKAAGTCPPDVLKNV”);
  • HPV49L2 (“NIYRTCKQAGNCPPDVVNKV”); or
  • HPV38L2 (“DIYRGCKASNTCPPDVINKV”).
  • the absorbance was measured at 450 nm using a spectrophotometer (Tecan Plate Reader).
  • a known human chimeric lgG1 HPV16 L2-specific neutralizing antibody JWW-1 , Addgene #66748, that recognizes HPV16L2 amino acid region 17-36 and has demonstrated cross-recognition to other HPV types was used as standard curve to determine the antibody titers.
  • JWW-1 A human chimeric monoclonal antibody capable of neutralizing HPV residues 17 to 36
  • Results are presented as average increase from baseline in all subjects shown in Figure 1 . Values for individual subjects are shown in Figure 2.
  • the active vaccination response shows similar patterns for HPV16 and HPV31.
  • the geometric mean titer increases from around 500 at baseline to 5000 at Day71. Prior to the booster vaccination at Day 185 the geometric mean titer has decreased to 2000 followed by an increase to 5000 at Day 194. At Day 365 the geometric mean titer has again decreased to 2000.
  • HPV5 For the rest of the HPV variants (HPV5, HPV15, HPV20, HPV38, and HPV76) a comparable pattern is seen. A clear increase from Day -1 to Day 71 , Day 194 reaching the same level as Day 71 , and ending at Day 365 somewhat above the Day -1 level. The HPV38 titer appear to be considerably higher prior to the initial vaccination a DAY -1 .
  • Geometric mean titers (95%-CI) for the group receiving active drug (Active) or placebo (Placebo) are shown in Figure 3A for HPV16, Figure 3B for HPV31 , Figure 3C for HPV5, Figure 3D for HPV15, Figure 3E for HPV20, Figure 3F for HPV38, Figure 3G for HPV76.
  • HPV16, HPV20 and HPV31 IgG-antibody levels all appear to reach a 10-fold increase compared to Day -1 at both Day 71 and Day 194.
  • the HPV5, HPV15, and HPV76 reach between a 5-fold and 10-fold increase, whereas HPV38 only reaches around a 3-fold increase. All variant but HPV38 maintains at least a 2-fold increase at Day 365. See Figure 4.
  • HPV16, HPV20, and HPV31 between 47% and 60% of the subjects reach a 10-fold increase.
  • HPV5 HPV15, and HPV76 between 27% and 36% of the subjects reach a 10-fold increase, whereas only 13% reach a 10-fold increase for HPV38.
  • Table 2 Average antibody titer against HPV50 and gamma HPV types
  • the assessment of immune response elicited by the AAVLP-HPV(16/31 L2) vaccine surprisingly demonstrated a neutralizing response against the cutaneous beta-HPV types recognizing the L2 sequences from amino acid 17-36 for the different types depicted, including HPV5L2, HPV15L2, HPV17L2, HPV20L2, HPV36L2, HPV92L2, HPV96L2, HPV76L2, HPV49L2, HPV38L2 and gamma-HPV type HPV50L2.
  • the L2 sequences from amino acid 17-36 in HPV20L2 is identical to the sequence of HPV24L2.
  • neutralizing response against HPV24L2 has been demonstrated likewise.
  • HPV-neutralizing assay Three different pseudoviron-based HPV-neutralizing assay (PBNA) have been established. L1 -, Fc- and L2-PBNA.
  • the first assay is the least sensitive method to detect the neutralizing effects of HPV L2 induced antibodies.
  • PsVs HPV pseudovirions
  • PsVs characterization a 96-well tissue culture polystyrene plate (Falcon, Germany) was prepared with 50 pl of diluted serum (in Dulbecco modified Eagle medium [DMEM] from Sigma-Aldrich, Germany), in a starting dilution of 1 :50 in the plate and then titrated out in 5 steps, 3x dilution each combined with 50 pl of diluted PsV in DMEM and incubated at room temperature for 20 min. Next, 50 pl of cell line (2.5 x 10 5 cells/ml) was added to the PsV-antibody mixture and incubated for 48 h at 37°C in a humidified incubator.
  • DMEM Dulbecco modified Eagle medium
  • the amount of secreted GLuc was determined in 10 pl of cell culture medium using the Gaussia Glow Juice kit according to the manufacturer’s instructions (PJK GmbH, Germany), in a 96-weel F-bottom LUMITRAC microplate (Greiner Bio-One, Germany). The light emissions of samples were measured in a microplate luminometer (Victor3Perkin Elmer) 15 min after substrate addition.
  • the neutralizing antibody titers are represent as the IC50 and were calculated on the GraphPad Prism 7 software.
  • the assessment of immune response elicited by AAVLP-HPV(16/31 L2) vaccine surprisingly demonstrated a neutralizing response against the cutaneous beta-HPV vectors HPV5, HPV20, HPV38, and HPV92 with IC50 levels of 60-1900 and against the gamma-HPV vectors HPV4 and HPV95 with IC50 levels of 50.
  • HPV pseudovirions with encapsulated luciferase reporter are generated by co -transfection of 293TT cells with plasmids encoding codon-modified L1 and L2 and a firefly luciferase reporter plasmid.
  • PsV are prepared by transfecting 293TT cells with a plasmid encoding for the humanized HPV L1 and L2 genes, together with a plasmid containing the gene for gLuc under the control of CMV promoter. PsVs were extracted, cleared by centrifugation at 10,000 rpm and 4°C for 10 minutes and purified by Optiprep before inoculation in mice.
  • mice Female Balb/c mice are treated with 3 mg of Depo-provera (Pfizer) 4 days prior to viral challenge.
  • Pfizer Depo-provera
  • mice One day prior to challenge, mice were injected i.p. with buffer or undiluted human sera obtained on day 1 , 71 and 193 according to Example 1 .2.
  • the mice are anesthetized and challenged with pseudovirus. Briefly, a HPV5, HPV76, HPV16, HPV31 PsV inoculum of 20 pl mixed with 20 pl of a 3% CMC preparation, based on L1 content, was used.
  • the inoculum was delivered using an M20 positive-displacement pipette in two doses; 20 pl before and 20 pl after insertion in the vagina of a Cytobrush that was turned clockwise and counterclockwise 10 times.
  • standard dissecting forceps is used to occlude the vaginal introitus to achieve maximal retention of the material while the mice recover from anesthesia.
  • the mice are again anesthetized and 20 pl of luciferin (7.8 mg/ml) was deposited in the vaginal vault.
  • Luciferase signals are acquired for 10 min with a Xenogen IVIS 100 imager, and analysis was performed with Living Image 2.0 software (Caliper Life Sciences).
  • An identical region of interest (ROI) is drawn around the luciferase signal emitted from each mouse, and the average radiance within the ROI determined.
  • Results for HPV76 and HPV5 challenge from the most responsive subject are presented in Figure 7A and 7B respectively.
  • Results for HPV16 and HPV31 challenge from the most responsive subject are presented in Figure 7C and 7D respectively.
  • anti-serum from humans vaccinated with AAVLP-HPV(16/31 L2) vaccine demonstrated protection against alpha-HPV type 16 and 31 in in vivo passive transfer studies.
  • a protection was also demonstrated against beta-HPV type 5 and 76. Mice were protected against infection with beta-HPV types 5 by day 193 and partially protected by day 71 sera.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne une protéine structurale de parvovirus muté, comprenant au moins une insertion comprenant une séquence d'acides aminés à partir d'acides aminés de la protéine L2 de l'HPV16 et/ou de l'HPV31 pour une utilisation dans le traitement ou la prévention d'une infection par HPV de type bêta ou de type gamma. En outre, l'invention concerne des structures multimères comprenant la protéine, VLP, un procédé de production de la protéine structurale de parvovirus muté et des médicaments ou des vaccins comprenant la protéine structurale de parvovirus muté qui peut être utilisée pour traiter une infection par HPV de type bêta ou de type gamma.
PCT/EP2022/081473 2021-11-11 2022-11-10 Protéine structurale de parvovirus dirigée contre hpv bêta-et gamma WO2023083964A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP21207758 2021-11-11
EP21207758.0 2021-11-11

Publications (1)

Publication Number Publication Date
WO2023083964A1 true WO2023083964A1 (fr) 2023-05-19

Family

ID=78598937

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/081473 WO2023083964A1 (fr) 2021-11-11 2022-11-10 Protéine structurale de parvovirus dirigée contre hpv bêta-et gamma

Country Status (1)

Country Link
WO (1) WO2023083964A1 (fr)

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996002555A1 (fr) 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Oligonucleotides immunomodulateurs
WO2001054720A1 (fr) 2000-01-28 2001-08-02 Cistem Biotechnologies Gmbh Composition pharmaceutique pour immunomodulation et preparation de vaccins comprenant un antigene, un oligodesoxyribonucleotide immunogene et polymere polycationique sous forme d'adjuvants
WO2001093903A1 (fr) 2000-06-08 2001-12-13 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Composition antigenique comprenant un peptide polycationique et inosine et cytosine
WO2001093905A1 (fr) 2000-06-08 2001-12-13 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Oligonucleotides synthetiques immunostimulatoires
WO2002013857A2 (fr) 2000-08-17 2002-02-21 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Vaccin comprenant au moins un antigene et un peptide antimicrobien a base de cathelicidine ou un derive de ce peptide
WO2002032451A1 (fr) 2000-10-18 2002-04-25 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Composition de vaccin
WO2002095027A2 (fr) 2001-05-21 2002-11-28 Intercell Ag Molécules oligodésoxynucléiques immunostimulatrices
WO2008145401A2 (fr) 2007-05-31 2008-12-04 Medigene Ag Protéines structurales de parvovirus mutées en tant que vaccins
WO2010099960A2 (fr) 2009-03-04 2010-09-10 Deutsches Krebsforschungszentrum Protéine activant l'assemblage (aap) et son utilisation pour la fabrication de particules de parvovirus consistant essentiellement en vp3
WO2010118424A2 (fr) 2009-04-10 2010-10-14 The Johns Hopkins University Particules ressemblant au papillomavirus (vlp) comme vaccins à large spectre contre le papillomavirus humain (vph)
WO2012031760A1 (fr) 2010-09-08 2012-03-15 Medigene Ag Protéines structurales mutées de parvovirus comprenant des épitopes de cellules b induisant une protection croisée d'une protéine l2 du hpv ainsi que produits et procédés s'y rapportant

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996002555A1 (fr) 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Oligonucleotides immunomodulateurs
WO2001054720A1 (fr) 2000-01-28 2001-08-02 Cistem Biotechnologies Gmbh Composition pharmaceutique pour immunomodulation et preparation de vaccins comprenant un antigene, un oligodesoxyribonucleotide immunogene et polymere polycationique sous forme d'adjuvants
WO2001093903A1 (fr) 2000-06-08 2001-12-13 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Composition antigenique comprenant un peptide polycationique et inosine et cytosine
WO2001093905A1 (fr) 2000-06-08 2001-12-13 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Oligonucleotides synthetiques immunostimulatoires
WO2002013857A2 (fr) 2000-08-17 2002-02-21 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Vaccin comprenant au moins un antigene et un peptide antimicrobien a base de cathelicidine ou un derive de ce peptide
WO2002032451A1 (fr) 2000-10-18 2002-04-25 Intercell Biomedizinische Forschungs- Und Entwicklungs Ag Composition de vaccin
WO2002095027A2 (fr) 2001-05-21 2002-11-28 Intercell Ag Molécules oligodésoxynucléiques immunostimulatrices
WO2008145401A2 (fr) 2007-05-31 2008-12-04 Medigene Ag Protéines structurales de parvovirus mutées en tant que vaccins
WO2010099960A2 (fr) 2009-03-04 2010-09-10 Deutsches Krebsforschungszentrum Protéine activant l'assemblage (aap) et son utilisation pour la fabrication de particules de parvovirus consistant essentiellement en vp3
WO2010118424A2 (fr) 2009-04-10 2010-10-14 The Johns Hopkins University Particules ressemblant au papillomavirus (vlp) comme vaccins à large spectre contre le papillomavirus humain (vph)
EP2416798B1 (fr) * 2009-04-10 2017-09-20 The Johns Hopkins University Particules ressemblant au papillomavirus (vlp) comme vaccins à large spectre contre le papillomavirus humain (vph)
WO2012031760A1 (fr) 2010-09-08 2012-03-15 Medigene Ag Protéines structurales mutées de parvovirus comprenant des épitopes de cellules b induisant une protection croisée d'une protéine l2 du hpv ainsi que produits et procédés s'y rapportant

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. QNM76585.1
ANDERSON, E. D ET AL.: "The Ordered and Compartment-specific Autoproteolytic Removal of the Furin Intramolecular Chaperone Is Required for Enzyme Activation", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 277, no. 15, 2002, pages 12879 - 12890
ANDERSON, E. D. ET AL.: "Activation of the furin endoprotease is a multiple-step process: Requirements for acidification and internal propeptide cleavage", EMBO JOURNAL, vol. 16, no. 7, 1997, pages 1508 - 1518, XP002136833, DOI: 10.1093/emboj/16.7.1508
BEZIAT V: "Human genetic dissection of papillomavirus-driven diseases: new insight into their pathogenesis", HUM GENET, vol. 139, no. 6-7, 20 May 2020 (2020-05-20), pages 919 - 939, XP037158320, DOI: 10.1007/s00439-020-02183-x
BUCK, C. B. ET AL.: "Efficient Intracellular Assembly of Papillomaviral Vectors", JOURNAL OF VIROLOGY, vol. 78, no. 2, 2003, pages 751 - 757, XP002395982, DOI: 10.1128/JVI.78.2.751-757.2004
CHAHOUD, J ET AL.: "Association Between β-Genus Human Papillomavirus and Cutaneous Squamous Cell Carcinoma in Immunocompetent Individuals-A Meta-analysis", JAMA DERMATOL., vol. 152, no. 12, 2016, pages 1354 - 1364
DAY, P. M. ET AL.: "A Human Papillomavirus (HPV) In Vitro Neutralization Assay That Recapitulates the In Vitro Process of Infection Provides a Sensitive Measure of HPV L2 Infection-Inhibiting Antibodies", CLINICAL AND VACCINE IMMUNOLOGY. AMERICAN SOCIETY FOR MICROBIOLOGY, vol. 19, no. 7, 2012, pages 1075 - 1082
DOORBAR, J ET AL.: "Generation of HPV Pseudovirions Using Transfection and Their Use in Neutralization Assays", HUMAN PAPILLOMAVIRUSES, 2005, pages 445 - 462, XP001539166
E. W. MARTIN: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO.
KIM SC ET AL.: "Modifications of mRNA vaccine structural elements for improving mRNA stability and translation efficiency", MOL CELL TOXICOL., vol. 20, 2021, pages 1 - 8, XP037667851, DOI: 10.1007/s13273-021-00171-4
NIETO K ET AL.: "Development of AAVLP(HPV16/31 L2) particles as broadly protective HPV vaccine candidate", PLOS ONE., vol. 7, no. 6, 2012, pages e39741
O'REILLY ZWALD F ET AL.: "Skin cancer in solid organ transplant recipients: advances in therapy and management: part I. Epidemiology of skin cancer in solid organ transplant recipients", J AM ACAD DERMATOL., vol. 65, no. 2, 2011, pages 253 - 261
SCHELLENBACHER C ET AL.: "Developments in L2-based human papillomavirus (HPV) vaccines", VIRUS RES., vol. 231, 2017, pages 166 - 175, XP029929212, DOI: 10.1016/j.virusres.2016.11.020
TAMPA, M ET AL.: "The Role of Beta-HPV Types and HPV-Associated Inflammatory Processes in Cutaneous Squamous Cell Carcinoma", J IMMUNOL RES., 2020
TESSARI G ET AL.: "Nonmelanoma skin cancer in solid organ transplant recipients: update on epidemiology, risk factors, and management", DERMATOL SURG., vol. 38, no. 10, 2012, pages 1622 - 30
WANG, J. W ET AL.: "Preparation and properties of a papillomavirus infectious intermediate and its utility for neutralization studies", VIROLOGY, vol. 449, 2013, pages 304 - 316, XP028669466, DOI: 10.1016/j.virol.2013.10.038
WANG, J. W. ET AL.: "Production of Furin-Cleaved Papillomavirus Pseudovirions and Their Use for In Vitro Neutralization Assays of L1- or L2-Specific Antibodies", CURRENT PROTOCOLS IN MICROBIOLOGY, 2015, pages 1 - 26
WATERBOER, T ET AL.: "Serological association of beta and gamma human papillomaviruses with squamous cell carcinoma of the skin", BR J DERMATOL., vol. 159, no. 2, 2008, pages 457 - 9, XP071157235, DOI: 10.1111/j.1365-2133.2008.08621.x

Similar Documents

Publication Publication Date Title
US10065994B2 (en) Norovirus derived immunogenic compositions and methods
JP5386392B2 (ja) ひとパピローマウイルス・カプシド蛋白およびウイルス様粒子の製造
US20120087937A1 (en) Novel compositions
Huber et al. Chimeric L2-based virus-like particle (VLP) vaccines targeting cutaneous human papillomaviruses (HPV)
AU2010292848B2 (en) Porcine Torque teno virus vaccines and diagnosis
WO2012031760A1 (fr) Protéines structurales mutées de parvovirus comprenant des épitopes de cellules b induisant une protection croisée d'une protéine l2 du hpv ainsi que produits et procédés s'y rapportant
KR20100029216A (ko) 고리스크군 인간 파필로마 바이러스에 대한 교차성 중화 항체를 유도하는 백신 항원
CN113913464B (zh) 一种表达载体、重组腺相关病毒及其在制备2019新型冠状病毒疫苗中的应用
CN114364692B (zh) 嵌合的乳头瘤病毒l1蛋白
CN114127098B (zh) 嵌合的人乳头瘤病毒51型l1蛋白
WO1999050424A2 (fr) Preparation et utilisation d'antigenes
JP4825958B2 (ja) 高リスク群ヒトパピローマウイルスの中和抗体を誘導する抗原
CN114174319B (zh) 嵌合的人乳头瘤病毒52型l1蛋白
CN114127100B (zh) 嵌合的人乳头瘤病毒39型l1蛋白
Remond et al. Infectious bursal disease subviral particles displaying the foot-and-mouth disease virus major antigenic site
Fraiberk et al. Exploitation of stable nanostructures based on the mouse polyomavirus for development of a recombinant vaccine against porcine circovirus 2
WO2023083964A1 (fr) Protéine structurale de parvovirus dirigée contre hpv bêta-et gamma
CN114127092B (zh) 人乳头瘤病毒多价免疫原性组合物
CN114127295B (zh) 嵌合的人乳头瘤病毒16型l1蛋白
CN114127094B (zh) 嵌合的人乳头瘤病毒58型l1蛋白
CN114127093B (zh) 嵌合的人乳头瘤病毒45型l1蛋白
CN114174320B (zh) 嵌合的人乳头瘤病毒18型l1蛋白
CN114127099B (zh) 嵌合的人乳头瘤病毒6型l1蛋白
CN114127096B (zh) 嵌合的人乳头瘤病毒31型l1蛋白
CN114127127B (zh) 嵌合的人乳头瘤病毒35型l1蛋白

Legal Events

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

Ref document number: 22821315

Country of ref document: EP

Kind code of ref document: A1