Non-human papillomaviruses for gene delivery in vitro and in vivo

Abstract
A non-human papilloma pseudovirus or virus like particle has at least one papilloma capsid protein codon-optimized for expression in eukaryotic cells or cell lines. A pharmaceutical composition includes the non-human papilloma pseudovirus or virus like particle, and a diagnostic agent, an imaging agent, and a therapeutic agent. A non-human papilloma pseudovirus or virus like particle can be used as a medicament. A method for producing a non-human papilloma pseudovirus or virus like particle involves codon-optimizing of capsid proteins of non-human papillomaviruses for expression in eukaryotic cells, synthesizing of the sequences and cloning of the synthesized sequences into expression vectors, and producing non-human papilloma pseudovirus or virus like particles. ι-carrageenan can be used as transduction enhancer for non-human papilloma pseudovirus or virus like particles in vitro.
Description
PRIORITY AND CROSS REFERENCE TO RELATED APPLICATIONS

This application is the U.S. National Phase Application under 35 U.S.C. § 371 of International Application No. PCT/EP2018/081102, filed Nov. 13, 2018, designating the U.S. and published in English as WO 2019/096796 A1 on May 23, 2019, which claims the benefit of German Application No. DE 10 2017 220 276.9, filed Nov. 14, 2017. Any and all applications for which a foreign or a domestic priority is claimed is/are identified in the Application Data Sheet filed herewith and is/are hereby incorporated by reference in their entireties under 37 C.F.R. § 1.57.


SEQUENCE LISTING IN ELECTRONIC FORMAT

The present application is being filed along with an Electronic Sequence Listing as an ASCII text file via EFS-Web. The Electronic Sequence Listing is provided as a file entitled HRZG001010APCSEQLIST.txt, created and last saved on May 13, 2020, which is 126,406 bytes in size. The information in the Electronic Sequence Listing is incorporated herein by reference in its entirety.


FIELD

The present invention relates to non-human papillomaviruses for in vitro and in vivo gene delivery.


SUMMARY

The present invention relates to a non-human papilloma pseudovirus or virus like particle comprising at least one papilloma capsid protein codon-optimized for expression in eukaryotic cells or cell lines, and to a pharmaceutical composition comprising the non-human papilloma pseudovirus or virus like particle of the invention, and an agent selected from the group consisting of: a diagnostic agent, an imaging agent, and a therapeutic agent. Moreover, the invention pertains to a non-human papilloma pseudovirus or virus like particle of the invention for use as a medicament. Further encompassed by the invention is a method for producing a non-human papilloma pseudovirus or virus like particle, comprising the steps of: a) codon-optimizing of capsid proteins of non-human papillomaviruses for expression in eukaryotic cells; b) synthesizing of the sequences and cloning of the synthesized sequences into expression vectors; and c) producing non-human papilloma pseudovirus or virus like particles. Finally, the invention relates to the use of ι-carrageenan as transduction enhancer for non-human papilloma pseudovirus or virus like particles in vitro.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows Western Blots of fractions after purification by ultracentrifugation. All produced non-human papilloma PsVs were purified by density gradient ultracentrifugation. Subsequently, the collected fractions were separated by SDS-PAGE, blotted onto a nitrocellulose membrane and probed for L1. (A) MfPV6, (B) CcrPV1, (C) CcPV1, (D) CgPV1, (E) MmPV1, (F) MfPV11, (G) PcPV1, (H) PlPV1.



FIG. 2 shows Luciferase assay 72 h after transduction of HEK293TT cells with different non-human papilloma pseudovirions (PsVs). 50.000 HEK293TT cells were transduced by adding 10 μl of fractions 3 to 8 of ultracentrifugation-purified non-human papilloma PsVs. Cell culture supernatant was used for luciferase assay 72 h after transduction. (A) Relative light units (RLU) measured after transduction with at least two independent PsV preparations. Shown are mean RLU values of the fractions yielding the highest transduction of one preparation. (B) RLU for fractions 3-8 after transduction with PcPV1 and MfPV11 PsVs, showing the typical peak of transducing PsVs between OptiPrep fractions 4 and 6.



FIG. 3 shows transmission electron microscopy of MfPV11 and PcPV1 VLPs. Purified VLPs were fixed for 24 h at room temperature with formaldehyde, contrasted with phosphotungstic acid and analyzed by transmission electron microscopy. PcPV1 (A), MfPV11 (B).



FIG. 4 shows titration of PsVs by transduction of HEK293TT cells. HEK293TT cells were transduced with indicated PsVs carrying pEGFP as reporter plasmid. 72 h after transduction, GFP-positive cells were counted and titer was calculated as transducing units per ml. PsVs. Each data point represents one PsV preparation (A). To compare the amount of transducing units with amount of particles carrying the pEGFP reporter plasmid, DNA was isolated from PsVs and quantified by qPCR. Titer is calculated as pEGFP plasmids per ml (B).



FIG. 5 shows effect of ι-carrageenan on transduction with PcPV1 and MfPV11. Different doses of ι-carrageenan were added to cell culture medium immediately before transduction of HEK293TT cells with PcPV1 PsVs carrying a G.Luc reporter plasmid. Luciferase assay was performed 72 h after transduction. Statistical analysis was performed by 2way ANOVA and Tukey's multiple comparison test (A). Additionally, HEK293TT cells were transduced with PcPV1 and MfPV11 PsVs carrying a pEGFP reporter plasmid with (+) and without (−) addition of 10 μg/ml ι-carrageenan. GFP-positive cells were counted 72 h after transduction and transducing units were calculated. Each data point represents one experiment with one PsV preparation. Statistical analysis was performed using t-test (B).



FIG. 6 shows bioluminescence imaging after intramuscular application. 0.24 of PcPV1 and MfPV11 PsVs carrying a firefly luciferase (F.Luc) reporter plasmid were added to 8000 HEK293TT cells for transduction. 72 h later, cells were lysed and lysate was used for F.Luc assay (A). 50 μl of the above analyzed PsV preparations were injected into the left thigh muscle. Bioluminescent imaging was performed approx. 3 h after injection (day 0) to test for any free F.Luc and subsequently in a weekly manner (B).





DETAILED DESCRIPTION

The use of nucleic acids as genetic vaccines has several advantages, most importantly the rapid production, simple adaptation and high stability at ambient temperature. Additionally, the vaccinee's cells themselves express the encoded antigens, which guarantees correct post-translational modifications and folding of the protein. Immunization with a DNA vaccine activates both the humoral and cellular immune response, making genetic vaccines a powerful platform. While intramuscular injection of naked DNA leads to a reasonable cellular uptake and subsequent expression in rodents, larger animals, especially non-human primates, require additional stimuli to enhance the uptake of the plasmid DNA.


One of the most powerful methods to enhance the uptake of DNA is the use of electroporation. Although quite effective, electroporation is an invasive and painful procedure, requiring local anesthesia and the presence of special equipment.


Other delivery methods include physical devices such as pressure injector, gene gun, and chemical formulations like block copolymers, cationic liposome and polyethyleneimine.


Furthermore, the application of bacteria and viruses as gene carriers has been explored, human papillomaviruses being one of them. Gene delivery using papillomaviruses does face the same issues as other more commonly used viruses, which is the problem of vector immunity. Human papillomaviruses (HPV), especially type 16, work quite well as gene delivery vehicles, however, more and more virus like particles (VLPs) of different papilloma types are being added to vaccines against HPV. Merck's “Gardasil 9”, approved by the FDA in December 2014, includes VLPs of HPV types 6, 11, 16, 18, 31, 33, 45, 52, and 58. It would consequently not be possible to apply these papillomavirus types as gene carriers in Gardasil 9 vaccinated individuals. In addition to vaccine-induced immunity against HPV, natural infection occurs quite frequently, making it difficult to reliably apply HPV for the delivery of genetic vaccines in the general population.


In view of the above, an ongoing demand exists for the development of efficient and safe means and methods for the transfer of nucleic acids by papillomaviruses.


The technical problem underlying the present invention can be seen as the provision of means and methods for complying with the aforementioned needs. The technical problem is solved by the embodiments characterized in the claims and herein below.


The present invention pertains to a non-human papilloma pseudovirus or virus-like particle comprising at least one papilloma capsid protein codon-optimized for expression in eukaryotic cells or cell lines.


The inventors explored the in vitro production of non-human papilloma virus-like particles (VLPs) and pseudovirions (PsVs) and their ability to deliver reporter plasmids in vitro and in vivo, as shown in the following Examples. Published sequences for capsid proteins L1 and L2 of ten non-human papilloma viruses indicated in the following Table 3 were codon-optimized for expression in human cell lines, synthesized and cloned into expression vectors. VLPs and PsVs were produced following the protocol by Buck et. al (C. Buck, D. Pastrana, D. Lowy, J. Schiller, in Human Papillomaviruses, C. Davy, J. Doorbar, Eds. (Humana Press, 2006), vol. 119, pp. 445-462), with slight modifications. VLPs and PsVs from Puma concolor papillomavirus 1 (PcPV1) and Macaca fascicularis papillomavirus type 11, isolate Mac1637 (MfPV11), could be identified as efficient gene vectors in vitro. Further, PsVs from Puma concolor papillomavirus 1 (PcPV1) could be found as efficient delivery vector in vivo. These findings demonstrate that it is worth exploring the wide range of non-human papilloma viruses for gene delivery, gene therapy, therapeutic, diagnostic and vaccination approaches.


Several characteristics of non-human papilloma viruses make them interesting candidates as DNA delivery vectors, such as their stability due to being non-enveloped, their ability to package foreign DNA without the need of a specific packaging sequence and—depending on the type of papillomavirus—their capability to infect mucosal tissue. Additionally, the viral capsid might provide some adjuvant effect by stimulating the innate immune system. Papillomaviruses as non-enveloped viruses are also more stable than enveloped viruses.


In 2013,260 different papillomavirus types were identified, among those 148 human (A. Rector and M. van Ranst, Animal papillomaviruses, Virology. 445, 213-223 (2013), doi:10.1016/j.virol.2013.05.007).


To date, 112 distinct non-human papillomavirus types have been genomically characterized and are available on GenBank and listed in the following Table 1 which corresponds to Table 1 of the mentioned publication by A. Rector and M. van Ranst (2013). The 112 distinct non-human papillomavirus types are distributed over 32 different genera, leaving only the genus Gammapapillomavirus, Mupapillomavirus and Nupapillomavirus to contain exclusively HPV types (see FIG. 1 of the publication by A. Rector and M. van Ranst (2013)). Within some well-studied vertebrate species, such as cynomolgus macaques, domestic cows and dogs, a multitude of different papillomavirus types have already been discovered (MfPV1 to MfPV11, BPV1 to BPV13 and CPV1 to CPV15, respectively; see Table 1 of the publication by A. Rector and M. van Ranst (2013)), indicating that also non-human vertebrate species could carry their own sets of species-specific papillomavirus types.


The term “non-human papilloma virus” as used herein refers to one of the 112 distinct non-human papillomavirus types indicated in the following Tables 1, preferably to the non-human papillomaviruses of Table 2, more preferably to the non-human papillomaviruses of Table 3.


The term “virus-like particle” (VLP) as referred to herein means a self-assembling supra-molecular structure formed by (a) viral structural protein(s). Many VLPs share the physicochemical characteristics of their parental viruses, they do not, however, carry any genetic information, including genetic information for replication. These properties can be harnessed for targeted delivery of genetic information or active drug substances to specified cell types as well as for delivery of an antigen of interest to B cells for induction of effective antibody responses. For example, VLPs can be produced experimentally by expression of capsid proteins of a virus, as detailed elsewhere herein. VLPs can be used, e.g., for gene delivery in vitro or in vivo, or for the development of pharmaceutical compositions, e.g., for diagnosis, imaging, therapy or vaccination, as set forth herein below.


The term “non-human papilloma pseudovirion or pseudovirus (PsV)” or “non-human papilloma pseudovirion or pseudovirus (PsV) particle” as used herein denotes a virus-like particle, which additionally contains plasmid-DNA packaged inside the capsid. This plasmid-DNA may code for any protein of interest, which is meant to be expressed by the target cell upon delivery by the PsV, e.g., an antigen for vaccination.


A “peptide or protein” as referred to herein relates to a molecule consisting of amino-acid residues joined by peptide bonds. Peptides, consisting of several, typically, at least 20, at least 30, at least 40, at least 50 or at least 60 amino acids that are covalently linked to each other by peptide bonds, are commonly referred to as polypeptides. Molecules consisting less than 20 amino acids covalently linked by peptide bonds are typically considered to be peptides.


The term “capsid protein” as used herein refers to the capsid protein L1 and/or L2. The papillomavirus genome is divided into an early region (E), encoding six open reading frames (ORFs) E1, E2, E4, E5, E6, and E7, that are expressed immediately after initial infection of a host cell, and a late region (L) encoding a major capsid protein L1 and a minor capsid protein L2. All viral ORFs are encoded on one DNA strand. Expression of the capsid protein L1 and/or L2 in appropriate cells allow for the formation of virus-like particles (VLPs) or non-human papilloma pseudovirions (PsVs) as defined herein. The capsid proteins L1 and/or L2 are codon-optimized for expression in eukaryotic cells or cell lines, such as human cells or cell lines, as explained elsewhere herein. The wild-type sequences for many L1 and L2 capsid proteins of non-human papilloma viruses are described in the art and available, e.g., in GenBank. For instance, Tables 1, 2 and 3 of this application provide the corresponding accession numbers of genomes of preferred non-human papilloma viruses, including the sequences coding for L1 and L2 capsid proteins. In the following Examples, capsid proteins L1 and L2 of ten non-human papilloma viruses indicated in Table 3 were codon-optimized for expression in human cell lines, synthesized and cloned into expression vectors.


The term “codon-optimized papilloma capsid protein” as used herein denotes a non-human papilloma capsid gene L1 or L2 which has been converted to a sequence having an identical translated sequence but with alternative codon usage, for expression in eukaryotic cells or cell lines, e.g., human cells or cell lines, as defined by Lathe, 1985, J. Mol. Biol. 183: p. 1-12.


Gene or codon-optimization takes advantage of the degeneracy of the genetic code. Because of degeneracy, one protein can be encoded by many alternative nucleic acid sequences. Codon preference (codon usage bias) differs in each organism, and it can create challenges for expressing recombinant proteins in heterologous expression systems, resulting in low and unreliable expression.


The methodology of codon-optimization for expression in human cells may be summarized as follows:

    • (i) Identify placement of codons for proper open reading frame.
    • (ii) Compare wild type codon for observed frequency of use by human genes.
    • (iii) If codon is not the most commonly employed, replace it with an optimal codon for high expression in human cells.
    • (iv) Repeat this procedure until the entire gene segment has been replaced.
    • (v) Inspect new gene sequence for undesired sequences generated by these codon replacements (e.g. “ATTTA” sequences, inadvertent creation of intron splice recognition sites, unwanted restriction sites, etc.) and substitute codons that eliminate these sequences.
    • (vi) Assemble synthetic gene segments and test for improved expression.


Methods and tools for codon-optimization of sequences are well described in the art; see, e.g., U.S. Pat. No. 8,326,547; Ternette N., et al., Virology Journal 2007, Expression of RNA virus proteins by RNA polymerase II dependent expression plasmids is hindered at multiple steps. https://doi.org/10.1186/1743-422X-4-51; Haas J, Park E C, Seed B: Codon usage limitation in the expression of HIV-1 envelope glycoprotein. Curr Biol 1996, 6: 315-324. 10.1016/50960-9822(02)00482-7; Wagner R, Graf M, Bieler K, Wolf H, Grunwald T, Foley P, Uberla K: Rev-independent expression of synthetic gag-pol genes of human immunodeficiency virus type 1 and simian immunodeficiency virus: implications for the safety of lentiviral vectors. Hum Gene Ther 2000, 11: 2403-2413. 10.1089/104303400750038507; or Morton C J, Cameron R, Lawrence L J, Lin B, Lowe M, Luttick A, Mason A, Kimm-Breschkin J, Parker M W, Ryan J, Smout M, Sullivan J, Tucker S P, Young P R: Structural characterization of respiratory syncytial virus fusion inhibitor escape mutants: homology model of the F protein and a syncytium formation assay. Virology 2003, 311: 275-288. 10.1016/S0042-6822(03)00115-6.


As appreciated by those skilled in the art, the use of alternative codons encoding the same protein sequence removes the constraints on expression of non-human papilloma virus capsid proteins L1 and/or L2 by eukaryotic cells or cell lines, such as human cells or cell lines.


The term “vector” as used herein encompasses preferably phage, plasmid, viral vectors such as non-human papilloma viral vectors or retroviral vectors as well as artificial chromosomes, such as bacterial or yeast artificial chromosomes. Moreover, the term also relates to targeting constructs which allow for random or site-directed integration of the targeting construct into genomic DNA. Such target constructs, in an aspect, comprise a DNA of interest of sufficient length for either homologous or heterologous recombination as known in the art. The vector, in an aspect, further comprises selectable markers for propagation and/or selection in a host cell. The vector may be incorporated into a host cell by various techniques well known in the art. For example, a plasmid vector can be introduced in a precipitate such as a calcium phosphate precipitate or rubidium chloride precipitate, or in a complex with a charged lipid or in carbon-based clusters, such as fullerens. Alternatively, a plasmid vector may be introduced by heat shock or electroporation techniques. Should the vector be a virus, it may be packaged in vitro using an appropriate packaging cell line prior to application to host cells. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host/cells.


The term “eukaryotic cell or cell line” as referred to herein means a eukaryotic cell or cell from a eukaryotic cell line susceptible to transduction or infection by non-human papilloma viruses, such as yeast (e.g., S. cerevisiae), insect cells (e.g., Sf9 or Sf21), mammalian cells (e.g., P19, NIH 3T3 or CHO cells), or human cells (e.g. HEK293, HT-1080 or HeLa cells). Eukaryotic cells or cell lines and conditions for their cultivation are well described in the art.


The term “human cell or cell line” as referred to herein means a human cell or cell from a human cell line susceptible to transduction or infection by non-human papilloma viruses. As known in the art, transduction is the process by which foreign DNA is introduced into a cell by a virus or viral vector. The term “cell” encompasses cells from human, from a variety of cell types such as, e.g., mucosal or cutaneous cells, and can be isolated from or part of a heterogeneous cell population, human tissue or organism. It is to be understood that human embryonic cells are excluded from the scope of the invention. Aspects of the present disclosure comprise, in part, a cell from an established cell line. As used herein, the term “established cell line” is synonymous with “immortal cell line,” or “transformed cell line” and refers to a cell culture of cells selected for indefinite propagation from a cell population derived from a human organism, tissue, or organ source. By definition, an established cell line excludes a cell culture of primary cells. As used herein, the term “primary cells” are cells harvested directly from fresh human tissues or organs and do not have the potential to propagate indefinitely. An established cell line can comprise a heterogeneous population of cells or a uniform population of cells. An established cell line derived from a single cell is referred to as a clonal cell line. The papilloma capsid protein referred to herein is codon-optimized for expression in eukaryotic cells or cell lines, for instance, yeast (e.g., S. cerevisiae), insect cells (e.g., Sf9 or Sf21), mammalian cells (e.g., P19, NIH 3T3 or CHO cells), or human cells (e.g. human kidney epithelial cells, HEK293, HT-1080 or HeLa cells). The human cell line can be, for example, HEK293T or HEK293FT.


The terms “comprising”, “comprises” and “comprised of” as used herein are synonymous with “including”, “includes” or “containing”, “contains”, and are inclusive or open-ended and do not exclude additional, non-recited members, elements or method steps. Evidently, the term “comprising” encompasses the term “consisting of”. More specifically, the term “comprise” as used herein means that the claim encompasses all the listed elements or method steps, but may also include additional, unnamed elements or method steps. For example, a method comprising steps a), b) and c) encompasses, in its narrowest sense, a method which consists of steps a), b) and c). The phrase “consisting of” means that the composition (or device, or method) has the recited elements (or steps) and no more. In contrast, the term “comprises” can encompass also a method including further steps, e.g., steps d) and e), in addition to steps a), b) and c).


The term “in vitro” as used herein denotes outside, or external to, the animal or human body. The term “in vitro” as used herein should be understood to include “ex vivo”. The term “ex vivo” typically refers to tissues or cells removed from an animal or human body and maintained or propagated outside the body, e.g., in a culture vessel. The term “in vivo” as used herein denotes inside, or internal to, the animal or human body. Preferably, the methods of the invention are in vitro methods.


As used herein, the term “about” when qualifying a value of a stated item, number, percentage, or term refers to a range of plus or minus 10 percent, 9 percent, 8 percent, 7 percent, 6 percent, percent, 4 percent, 3 percent, 2 percent or 1 percent of the value of the stated item, number, percentage, or term. Preferred is a range of plus or minus 10 percent.


Whether a value or number or amount or portion is statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools, e.g., determination of confidence intervals, p-value determination, Student's t-test, Mann-Whitney test, etc. Details are found in Dowdy and Wearden, Statistics for Research, John Wiley & Sons, New York 1983. Preferred confidence intervals are at least 90%, at least 95%, at least 97%, at least 98% or at least 99%. The p-values are, preferably, 0.1, 0.05, 0.01, 0.005, or 0.001. Preferably, the probability envisaged by the present invention allows that the assessment will be correct for at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95% or at least 97% of the subjects of a given cohort.


Non-human papillomavirus capsid structures, VLPs and PsVs of the invention can be produced by methods described in the art (see, e.g., C. Cerqueira et al., Efficient Production of Papillomavirus Gene Delivery Vectors in Defined In Vitro Reactions, Molecular therapy. Methods & clinical development. 5, 165-179 (2017), doi:10.1016/j.omtm.2017.04.005; Q. Zhao et al., Disassembly and reassembly of human papillomavirus virus-like particles produces more virion-like antibody reactivity, Virology journal. 9, 52 (2012), doi:10.1186/1743-422X-9-52; Buck, C., et al (2006a). Generation of HPV Pseudovirions Using Transfection and Their Use in Neutralization Assays. In Human Papillomaviruses, C. Davy and J. Doorbar, eds. (Humana Press), pp. 445-462, doi: 10.1385/1-59259-982-6:445; WO 2011/039646; U.S. Pat. Nos. 6,416,945; 6,991,795 and 7,205,126) or as demonstrated in the following Examples.


Briefly, DNA-sequences coding for non-human papillomavirus capsid proteins such as L1 and/or L2 are first codon-optimized for expression in a cell or cell line of choice such as a eukaryotic cell or cell line, and synthesized.


The codon-optimized L1 and/or L2 DNA sequences are then cloned into the multiple cloning site of an appropriate mammalian expression vector, such as the pIRES and pBApo vectors (Clontech), pVITRO and pVIVO vectors (Invivogen), the pcDNA vectors (Invitrogen), or other commercially available mammalian expression vectors well known in the art. The codon-optimized non-human papilloma L1 and/or L2 capsid DNA sequences are operatively linked to expression control sequences allowing expression in prokaryotic or eukaryotic host cells or isolated fractions thereof in the said vector. Expression of the polynucleotide encoding the mentioned codon-optimized non-human papilloma L1 and/or L2 capsid DNA sequences comprises transcription of the DNA sequences into a translatable mRNA. Regulatory elements ensuring expression in host cells are well known in the art. In an aspect, they comprise regulatory sequences ensuring initiation of transcription and/or poly-A signals ensuring termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers. Possible regulatory elements permitting expression in prokaryotic host cells comprise, e.g., the lac-, trp- or tac-promoter in E. coli, and examples for regulatory elements permitting expression in eukaryotic host cells are the AOX1- or the GAL1-promoter in yeast or the CMV-, SV40-, RSV-promoter (Rous sarcoma virus), CMV-enhancer, SV40-enhancer or a globin intron in mammalian and other animal cells. Moreover, inducible expression control sequences may be used in an expression vector referred to herein. Such inducible vectors may comprise tet or lac operator sequences or sequences inducible by heat shock or other environmental factors. Suitable expression control sequences are well known in the art. Beside elements which are responsible for the initiation of transcription such regulatory elements may also comprise transcription termination signals, such as the SV40-poly-A site or the tk-poly-A site, downstream of the polynucleotide. As mentioned above, suitable expression vectors are known in the art such as Okayama-Berg cDNA expression vector pcDV1 (Pharmacia), pBluescript (Stratagene), pCDM8, pRc/CMV, pcDNA1, pcDNA3 (Invitrogen) or pSPORT1 (Invitrogen). Methods which are well known to those skilled in the art can be used to construct and express the codon-optimized non-human papilloma L1 and/or L2 capsid proteins referred to herein; see, for example, the techniques described in Sambrook, Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory (2001) N.Y. and Ausubel, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y. (1994).


For non-human papilloma virus-like particle—(VLP) or pseudovirion—(PsV) production, appropriate eukaryotic cells or cell lines as defined herein are transfected with an expression vector which allows for expression of the codon-optimized non-human papilloma L1 and/or L2 DNA sequences. For transfection, polyethyleneimine, lipofectamine, DEAE-dextran, dendrimer, polybrene, calcium phosphate, lipofectin, DOTAP, lipofectamine, CTAB/DOPE, DOTMA, electroporation or other transfection methods described in the art can be used. A reporter gene or reporter plasmid can be used for measuring transduction efficiency. Transfection efficiency can be assessed, e.g., by various PCR methods, Western blot analysis, reporter gene assays or further methods known in the art (Molecular Cloning: A Laboratory Manual, 3rd ed., Volumes 1, 2 and 3; J. F. Sambrook and D. W. Russell, ed., Cold Spring Harbor Laboratory Press, 2001).


Harvesting of VLPs and PsVs can be performed after transfection following the standard protocol of Buck et. al (C. Buck, D. Pastrana, D. Lowy, J. Schiller, in Human Papillomaviruses, C. Davy, J. Doorbar, Eds. (Humana Press, 2006), vol. 119, pp. 445-462), optionally with modifications, as described in the following Examples. For instance, polyethyleneimine can be used for transfection, instead of lipofectamine. Moreover, Percoll can be used as ultracentrifugation medium for purification, instead of OptiPrep.


The formation of non-human papillomavirus capsid structures, VLPs and PsVs can be tested by methods known in the art (C. Cerqueira et al., Efficient Production of Papillomavirus Gene Delivery Vectors in Defined In Vitro Reactions, Molecular therapy. Methods & clinical development. 5, 165-179 (2017), doi:10.1016/j.omtm.2017.04.005; Q. Zhao et al., Disassembly and reassembly of human papillomavirus virus-like particles produces more virion-like antibody reactivity, Virology journal. 9, 52 (2012), doi:10.1186/1743-422X-9-52). For example, the produced non-human papillomavirus VLPs or PsVs can be isolated and purified by density gradient ultracentrifugation and subsequently analyzed by electron microscopy. Reporter plasmids can be used as indirect evidence for transduction efficacy, as described in the following Examples.


In some embodiments, the non-human papilloma virus-like particle of the invention may be labelled or contain other modifications which allow a detection and/or analysis of a hybridization product and/or the binding to a carrier. Labelling can be done by various techniques well known in the art and depending of the label to be used. Particularly, the non-human papilloma virus-like particle of the invention may be biotinylated in order to enable binding to a streptavidin surface or fluorescent conjugate. Exemplary labels to be used in the context of the present invention are, but are not limited to, fluorescent labels comprising, inter alia, fluorochromes such as R-phycoerythrin, Cy3, Cy5, fluorescein, rhodamin, Alexa, or Texas Red. However, the label may also be an enzyme or an antibody. It is envisaged that an enzyme to be used as a label will generate a detectable signal by reacting with a substrate. Suitable enzymes, substrates and techniques are well known in the art. An antibody to be used as label may specifically recognize a target molecule which can be detected directly (e.g., a target molecule which is itself fluorescent) or indirectly (e.g., a target molecule which generates a detectable signal, such as an enzyme). The non-human papilloma virus-like particle or pseudovirus particle of the invention may also contain 5′ restriction sites, locked nucleic acid molecules (LNA) or be part of a peptide nucleotide acid molecule (PNA). Such PNA can be, in principle, detected via the peptide part by, e. g., antibodies. It is further evident that the non-human papilloma virus-like particle of the invention can comprise an affinity tag used for purification purposes, like a His tag or FLAG tag or the like, well known in the art.


Preferred non-human papillomavirus types are listed in Tables 1, 2 and 3 below. Preferably, the non-human papilloma pseudovirus or virus-like particle of the invention is from Puma concolor papillomavirus 1 (PcPV1) or Macaca fascicularis papillomavirus type 11, isolate Mac1637 (MfPV11).


In a preferred embodiment of the non-human papilloma pseudovirus or virus-like particle of the invention, the papilloma capsid protein codon optimized for expression in eukaryotic cells or cell lines is L1 and/or L2. The non-human papilloma pseudovirus or virus-like particle of the invention can comprise the capsid protein L1, the capsid protein L2, or both the capsid protein L1 and the capsid protein L2. In a still further preferred embodiment, the non-human papilloma virus-like particle of the invention comprises the capsid protein L1; or the non-human papilloma virus-like particle or pseudovirus of the invention comprises the capsid proteins L1 and L2.


In another preferred embodiment of the non-human papilloma pseudovirus or virus-like particle of the invention, the papilloma capsid protein is from Caretta caretta papillomavirus 1, Colobus guereza papillomavirus 1, Common chimpanzee papillomavirus 1, Crocuta crocuta papillomavirus 1, Macaca fascicularis papillomavirus type 11, isolate Mac1637, Macaca fascicularis papillomavirus type 6, isolate Mac39, Procyon lotor papillomavirus 1, Puma concolor papillomavirus 1, Rhesus papillomavirus type 1b isolate Mac170 or Rousettus aegyptiacus papillomavirus type 1.


In a further preferred embodiment of the non-human papilloma virus-like particle of the invention, the codon-optimized L1 or L2 papilloma capsid protein is encoded by a nucleic acid sequence selected from the group consisting of:

    • a) a nucleic acid sequence as shown in SEQ ID Nos. 1 to 20;
    • b) a nucleic acid sequence encoding the amino acid sequence shown in SEQ ID Nos. 21 to 40; and
    • c) a nucleic acid sequence at least 40% identical to a nucleic acid sequence in a) or b).


The term “nucleic acid” or “nucleic acid sequence” as used herein refers to a single- or double-stranded DNA molecule as well as to a RNA molecule. Encompassed by the said term is genomic DNA, cDNA, hnRNA, mRNA as well as all naturally occurring or artificially modified derivatives of such molecular species. The nucleic acid may be in an aspect a linear or circular molecule. Moreover, a nucleic acid as referred to herein may comprise additional sequences required for proper transcription and/or translation such as 5′- or 3′-UTR sequences. In light of the degeneracy of the genetic code, optimized codons are used in the nucleic acid sequences referred to in this application, as described elsewhere herein. Thereby, optimal expression in, e.g., a eukaryotic cell or cell line such as a human cell or cell line can be achieved.


In addition to the aforementioned specific nucleic acid sequences SEQ ID Nos. 1 to 20 encoding the codon-optimized non-human papilloma capsid proteins L1 and L2 depicted in SEQ ID Nos. 21 to 40, the present application provides also variants of nucleic acid sequences coding for codon-optimized non-human papilloma capsid proteins L1 and L2. A variant as referred to in accordance with the present invention shall have a nucleic acid sequence which differs due to at least one amino acid substitution, deletion and/or addition wherein the amino acid sequence of the variant is still, preferably, at least 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical with the nucleic acid sequence coding for the specific codon-optimized capsid proteins mentioned above. The degree of identity between two nucleic acid or amino acid sequences can be determined by algorithms well known in the art. Preferably, the degree of identity is to be determined by comparing two optimally aligned sequences over a comparison window, where the fragment of amino acid sequence in the comparison window may comprise additions or deletions (e.g., gaps or overhangs) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment. Preferably, the alignment is over the entire nucleic acid sequences to be compared, or over the entire amino acid sequences to be compared. The percentage is calculated by determining the number of positions at which the identical nucleotides or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity. Optimal alignment of sequences for comparison may be conducted by the local homology algorithm of Smith and Waterman, by the homology alignment algorithm of Needleman and Wunsch, by the search for similarity method of Pearson and Lipman, by computerized implementations of these algorithms GAP, BESTFIT, BLAST, PASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, WI, or by visual inspection. Given that two sequences have been identified for comparison, GAP and BESTFIT are preferably employed to determine their optimal alignment and, thus, the degree of identity. Preferably, the default values of 5.00 for gap weight and 0.30 for gap weight length are used. Moreover, the variants referred to herein include fragments of the nucleic acid sequences coding for the codon-optimized capsid proteins or the aforementioned types of variants as long as these fragments have the essential immunological and biological properties as referred to herein. The variant shall be capable of forming a non-human papilloma virus-like particle or PsV as defined herein.


In a preferred embodiment, the non-human papilloma pseudovirus or virus-like particle of the invention is for gene transfer or gene delivery in vitro or in vivo.


Means and methods for adapting non-human papilloma pseudovirus or virus-like particles of the invention for gene delivery in vitro and/or in vivo are well described in the art (see, e.g., Sambrook, Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory (2001) N.Y. and Ausubel, Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y. (1994).


Briefly, the non-human papilloma pseudovirus or virus-like particle of the invention is loaded with a nucleic acid or gene of interest for gene delivery in vitro or in vivo. For instance, the gene of interest can be a reporter gene. In other cases, it can be a nucleic acid sequence to be transfected to a cell. In still further cases, it can be a nucleic acid used for gene therapy or vaccination in subjects, as defined elsewhere herein. Optimization of the transfection efficiency and optimization of the harvesting of VLPs and PsVs is within the skills of the practitioners in the field. For analysis, the VLPs and PsVs are preferably transferred to new cells. As evident to those skilled in the art, if the non-human papilloma pseudovirus or virus-like particle of the invention is loaded with a plasmid, the plasmid shall contain an eukaryotic origin of replication (SV40 origin).


Non-human papilloma pseudovirus or virus-like particles of the invention are interesting candidates as DNA delivery vectors in vitro and/or in vivo, because of their stability due to being non-enveloped, their ability to package foreign DNA without the need of a specific packaging sequence and—depending on the type of papillomavirus—their capability to infect mucosal tissue. Additionally, the viral capsid might provide some adjuvant effect by stimulating the innate immune system.


For example, human kidney epithelial cells, HEK293T or HeLa cells can be used for in vitro gene delivery. Preferably, the human cell line is HEK293T. HEK-293 cells were originally generated by treating a human embryonic kidney homogenate with sheared adenovirus DNA. The HEK293T line can also be used for production of recombinant proteins of interest. Recombinant protein production can be achieved, e.g., by placing the gene of interest under control of an appropriate promoter, such as the human elongation factor 1 alpha (EF1) housekeeping promoter.


Gene transfer in vivo can be utilized, e.g., for gene therapy or vaccination in animals, preferably mammals, even more preferred humans, as set forth elsewhere herein. Importantly, it has been found in the following Examples that mice never showed any signs of adverse reaction or inflammation at the site of infection of the non-human papilloma pseudovirus or virus-like particle of the invention.


In another preferred embodiment, the non-human papilloma pseudovirus or virus-like particle further comprises a targeting peptide. The term “targeting peptide” means a peptide sequence that serves to target or direct the non-human papilloma pseudovirus or virus-like particle to a particular location, cell type, diseased cell or tissue, or cell association. The targeting peptide is directed against a specific target molecule and allows concentration of the non-human papilloma pseudovirus or virus-like particle carrying a gene or nucleic acid of interest, or loaded with an agent such as a diagnostic agent, imaging agent or therapeutic agent as defined herein, in a particular localization within a subject. Accordingly, the targeting peptide targets the non-human papilloma pseudovirus or virus-like particle to specific tissues and/or cells in vitro and/or in vivo. Preferably, said target peptide is capable of directing the non-human papilloma pseudovirus or virus-like particle of the invention to one or more specific sites of the animal organism, mammal organism or preferably human organism, in vivo. Suitable targeting peptides and appropriate positions for including said target peptides into the codon-optimized non-human papilloma L1 or L2 capsid protein are well described in the art (see, e.g., WO 2011/039646 and references cited therein).


Preferably, the targeting peptide is capable of directing the non-human papilloma pseudovirus or virus-like particle to human cells selected from the group consisting of: liver cells, lung cells, heart cells, kidney cells, blood cells, brain cells, gut cells, stem cells, cells of the mucosa of the throat or the nose, or cancer cells.


The invention further relates to a pharmaceutical composition comprising the non-human papilloma pseudovirus or virus-like particle of the invention, and an agent selected from the group consisting of: a diagnostic agent, an imaging agent, and a therapeutic agent.


The term “pharmaceutical composition” as used herein refers to mixture comprising the non-human papilloma pseudovirus or virus-like particle of the invention, and an agent selected from the group consisting of: a diagnostic agent, an imaging agent, and a therapeutic agent. Moreover, the pharmaceutical composition of the invention may comprise further components as well such as further therapeutic or auxiliary ingredients and/or pharmaceutically acceptable carriers and/or diluents. Preferably, such further ingredients of the pharmaceutical composition of the invention can be buffers, diluents, stabilizing agents, wetting agents, pharmaceutical carriers, additional pharmaceutically active agents, release controlling agents and the like.


Preferably, the pharmaceutical composition of the present invention is to be used as a medicament. Said medicament is applied to treat and/or prevent a disease or disorder selected from the group consisting of: neurological diseases, stroke, ischemia, cancer, age-related disease, genetic disorder, allergy, auto-immune disease, an infection by bacteria, virus, fungus or parasite.


Pharmaceutical compositions comprising papilloma virus like particles are well described in the art (see, e.g., WO 2011/039646; U.S. Pat. Nos. 6,416,945; 6,991,795 and 7,205,126).


The term “diagnostic agent” as referred to herein means a compound used to detect the impaired function of a cell, tissue or body organ, or to detect abnormalities in a cell, tissue or body organ tissue structure. Diagnostic agents are well described in the literature. The term “diagnosing” as used herein refers to assessing the probability according to which a subject, preferably a human subject, is suffering or will suffer from a disease or condition referred to in this specification. As will be understood by those skilled in the art, such an assessment is usually not intended to be correct for 100% of the subjects to be diagnosed. The term, however, requires that a statistically significant portion of subjects can be correctly diagnosed to suffer from the disease or condition. Whether a portion is statistically significant can be determined by the methods described herein above. Preferably, one or more symptoms associated with a disease as referred to herein can be diagnosed by the administration of a non-human papilloma pseudovirus or virus-like particle of the invention comprising a diagnosing agent. For example, a diagnostic agent can be a labelled antibody, labeled nucleic acid such as siRNA, luciferase or other chemoluminescent agents, a biomarker or radionuclide or the like.


The term “imaging agent” as used herein denotes biomarkers for diagnosing a disease, monitoring disease progression, tracking a therapeutic response, and enhancing the knowledge of physiology and pathophysiology. For example, imaging agents include positron emission tomography and single-photon emission computed tomography tracers. Preferably, one or more symptoms associated with a disease as referred to herein can be imaged by the administration of a non-human papilloma pseudovirus or virus-like particle of the invention comprising an imaging agent.


The pharmaceutical composition comprising a non-human papilloma pseudovirus or virus-like particle of the invention and an imaging agent or a diagnostic agent can be used for imaging or diagnosing diseases or disorders in a subject as defined herein.


The term “therapeutic agent” as referred to herein is a gene, substance or compound capable of producing a curative effect in a disease state. The term “treatment or treating” as used herein denotes the improvement or even elimination of one or more symptoms associated with a disease as referred to herein, by the administration of a non-human papilloma pseudovirus or virus-like particle of the invention comprising a therapeutic agent. An improvement may also be seen as a slowing or stopping of the progression of a disease as set forth herein. The pharmaceutical composition comprising a non-human papilloma pseudovirus or virus-like particle of the invention and a therapeutic agent can be utilized for prevention or therapy of various diseases or disorders in a subject as specified elsewhere herein. The term “preventing” as used herein refers to avoiding the onset of a disease or at least one symptom thereof. The term “therapeutic agent” as used herein comprises a vaccine. A vaccine is a biological preparation that provides active acquired immunity to a particular disease. A vaccine typically contains an agent that resembles a disease-causing microorganism or virus and is often made from weakened or killed forms of the microbe or virus, its toxins or one of its surface proteins. The agent stimulates the body's immune system to recognize the agent as a threat, destroy it, and recognize and destroy any of these microorganisms or viruses that it later encounters. Vaccines can be prophylactic, i.e. they prevent or ameliorate the effects of a future infection by a natural or “wild” pathogen, or therapeutic, e.g., vaccines against cancer. The effectiveness of vaccination has been widely studied and verified; for example, the influenza vaccine, the HPV vaccine, and the chicken pox vaccine. The World Health Organization (WHO) reports that licensed vaccines are currently available for twenty-five different preventable infections, in its global vaccine action plan 2011 to 2020. The administration of vaccines is called “vaccination”. Vaccination is the most effective method of preventing infectious diseases; widespread immunity due to vaccination is largely responsible for the worldwide eradication of smallpox and the restriction of diseases such as polio, measles, and tetanus from much of the world.


According to the present invention, the pharmaceutical composition shall, preferably, comprise the non-human papilloma pseudovirus or virus-like particle of the invention, and a therapeutic agent in a therapeutically effective dose. Therapeutic efficacy and toxicity of the pharmaceutical composition of the invention can be determined by standard pharmaceutical procedures as describe elsewhere herein. The pharmaceutical composition shall further be adapted for use in treating and/or preventing a disease or disorder as described elsewhere herein. The desired mode of administration is set forth below. Formulations and preparations methods for formulating a composition as a medicament are well known in the art and include, for example, mixing, granulating, compression or dissolving the ingredients as appropriate to form the desired composition. Typically, the therapeutically active ingredients will be mixed and, preferably, combined them with a pharmaceutically acceptable carrier and/or diluent. Moreover, it is known by those skilled in the art that the formulation of a pharmaceutical composition to be used as a medicament shall take place under GMP conditions that will ensure quality, pharmaceutical security, and effectiveness of the medicament.


The term “reporter gene” as used herein means a gene that is attached to a regulatory sequence of another gene of interest in bacteria, yeast, insects, cell culture, animals, plants or human. Certain genes are chosen as reporters because the characteristics they confer on organisms expressing them are easily identified and measured, or because they are selectable markers. Reporter genes are often used as an indication of whether a certain gene has been taken up by or expressed in the cell or organism population. Commonly used reporter genes that induce visually identifiable characteristics usually involve fluorescent and luminescent proteins. Examples include the gene that encodes jellyfish green fluorescent protein (GFP), which causes cells that express it to glow green under blue light, the enzyme luciferase, which catalyzes a reaction with luciferin to produce light, and the red fluorescent protein from the gene dsRed. A common reporter in bacteria is the E. coli lacZ gene, which encodes the protein beta-galactosidase. This enzyme causes bacteria expressing the gene to appear blue when grown on a medium that contains the substrate analog X-gal. An example of a selectable-marker which is also a reporter in bacteria is the chloramphenicol acetyltransferase (CAT) gene, which confers resistance to the antibiotic chloramphenicol.


Methods for producing a non-human papilloma pseudovirus or virus-like particle of the invention, packaging it with a diagnostic agent, imaging agent, or therapeutic agent, optionally in combination with a reporter gene, and targeted delivery thereof to a subject are well described in the literature (see, e.g., Buck, C., et al (2006a). Generation of HPV Pseudovirions Using Transfection and Their Use in Neutralization Assays. In Human Papillomaviruses, C. Davy and J. Doorbar, eds. (Humana Press), pp. 445-462, doi: 10.1385/1-59259-982-6:445; U.S. Pat. Nos. 6,416,945; 6,991,795 and 7,205,126). Preferably, the non-human papilloma pseudovirus or virus-like particle of the invention, comprising a diagnostic agent, imaging agent, or therapeutic agent, optionally in combination with a reporter gene, is formulated as a pharmaceutical composition or medicament.


The term “subject” as used herein relates to insects (e.g. bees), an animal such as a rodent (mouse or rat), pet (cat, hamster, rabbit, dog), farming animal (sheep, poultry, goat, cow, horse) preferably mammal such as a non-human primate (macaque, marmoset, tamarin, spider monkey, owl monkey, vervet monkey, squirrel monkey, and baboon), and, more preferably, human.


Preferably, the therapeutic agent is selected from the group consisting of: (i) a small molecule, preferably a cytotoxic drug; (ii) a RNAi nucleic acid; (iii) a microRNA; (iv) a ribozyme; (v) an antisense nucleic acid; (vi) a morpholino; (vii) an antibody; and (viii) CRISPR/Cas. The therapeutic agent can also be a combination of two or more of the mentioned compounds, e.g. a combination of at least two cytotoxic drugs, at least a small molecule and an antibody, at least a cytotoxic drug and an antibody, at least a microRNA and antisense nucleic acid, or any further suitable combination of (i) to (viii) listed above.


The non-human papilloma pseudovirus or virus-like particle of the invention can contain a small molecule as a therapeutic agent. The term “small molecule” as used herein relates to a molecule with a low molecular weight. Typically, a small molecule is an organic compound with a molecular weight of less than 900 daltons. Small molecules include, for example, small secondary metabolites such as alkaloids, lipids, glycosides, terpenes, tetrapyrroles, phenazines, oliogonucleotides and small peptides or peptide-like molecules.


The small molecule is preferably a cytotoxic drug. The term “cytotoxic drug” as referred to herein denotes cytotoxic drugs or cytostatics (also cytotoxic chemotherapy) are drugs used to destroy proliferating cells such as cancer cells. Cytotoxic drugs inhibit cell division and in this way cause cancer cells to die. Cytotoxic drugs are transported in the bloodstream throughout the body. Cytotoxic drugs can be used to destroy tumors, boost the outcomes of surgery or radiotherapy, reduce metastases and alleviate cancer symptoms. Cytostatics can be effective outside the primary tumour and also destroy small tumors that have not been detected in tests. Cytotoxic drugs affect all dividing cells, including those of healthy tissue. But because cancer cells often divide markedly faster than normal cells, they are particularly sensitive to cytostatics. The effects on normal cells are less pronounced and healthy cells also recover faster. The role of cytotoxic drugs in cancer therapy has decreased slightly with the development of drug therapy. However, they continue to be widely used. Several types of cytotoxic drugs are used in cancer therapy that together have different kinds of effect. The most usual method is to administer a combination of several different cytotoxic drugs. The effectiveness of chemotherapy depends on the type of tumor, its composition, rate of development and proportion of cells in the distribution stage. Cytotoxic drugs are well described in the literature.


The non-human papilloma pseudovirus or virus-like particle of the invention can contain a ribozyme, as a therapeutic agent. The term “ribozyme” as referred to herein relates to an RNA molecule that is capable of catalyzing specific biochemical reactions, including cleavage and/or ligation of RNA and DNA and peptide bond formation. Methods of designing and constructing ribozymes are known in the art and include, for example, de novo rational design, oligonucleotide synthesis and in vitro-transcription. It is also known in the art that ribozymes can be stably integrated or transiently introduced into cells as part of a recombinant DNA construct such as a plasmid or vector. It will be understood that such a DNA construct may contain additional regulatory elements such as an enhancer, a constitutive or inducible promoter or a terminator.


The non-human papilloma pseudovirus or virus-like particle of the invention can contain an antibody, as a therapeutic agent. The term “antibody” as used herein, also referred to as immunoglobulin, includes both polyclonal and monoclonal antibodies, as well as fragments thereof, such as Fv, Fab and F(ab)2 fragments that are capable of binding an antigen or hapten. The present invention also includes single chain antibodies and humanized hybrid antibodies wherein amino acid sequences of a non-human donor antibody exhibiting a desired antigen-specificity are combined with sequences of a human acceptor antibody. The donor sequences will usually include at least the antigen-binding amino acid residues of the donor but may comprise other structurally and/or functionally relevant amino acid residues of the donor antibody as well. Such hybrids can be prepared by several methods well known in the art.


The non-human papilloma pseudovirus or virus-like particle of the invention can contain an inhibitory RNA molecule, as a therapeutic agent. The term “inhibitory RNA molecule” as used herein refers to an RNA molecule that inhibits gene expression in a sequence-specific manner. Inhibitory RNA molecules include, for example, small interfering RNA (siRNA), small hairpin RNAs (shRNA) and microRNA (miRNA). The inhibitory RNA molecule typically induces a process known as RNA interference (RNAi), leading to cleavage and/or translational inhibition of a target mRNA with a complementary sequence. It is known to those skilled in the art that the inhibitory RNA molecule can show perfect or imperfect base-pairing to a complementary target sequence. siRNA and shRNAs typically base-pair perfectly and induce mRNA cleavage only in a single, specific target. On the contrary, miRNAs usually have incomplete base pairing to a target and often inhibit the translation of many different mRNAs with similar sequences. An inhibitory RNA molecule may be chemically synthesized or expressed within the cell, for example by introduction of respective recombinant DNA construct. It will be understood that such a DNA construct may contain additional regulatory elements such as an enhancer, a constitutive or inducible promoter or a terminator.


The non-human papilloma pseudovirus or virus-like particle of the invention can contain an antisense oligonucleotide, as a therapeutic agent. An “antisense oligonucleotide” as used herein refers to a single strand DNA and/or RNA molecule that is capable of interfering with DNA and/or RNA processing. Antisense oligonucleotides comprise a nucleic acid sequence which is complementary to a specific RNA or DNA sequence. Typically, an antisense oligonucleotide will bind, in a sequence-specific manner, to their respective complementary oligonucleotides, DNA, or RNA, thereby interfering with DNA and/or RNA processing. It is known to those skilled in the art that antisense oligonucleotides may interfere with mRNA processing through RNase H-mediated degradation, translational arrest, modulation of splicing or they may act through steric hindrance of proteins. Means and methods for the design and synthesis of antisense oligonucleotides are well known in the art and include, for example, rational design, chemical modifications and design of antisense oligonucleotides containing locked nucleic acids (LNA) as well as solid-phase chemical synthesis. Antisense oligonucleotides can be chemically synthesized or expressed within the cell, for example by introduction of respective recombinant DNA construct. It will be understood by those skilled in the art that such a DNA construct may contain additional regulatory elements such as an enhancer, a constitutive or inducible promoter or a terminator. Preferably, the antisense oligonucleotide has a length of at least 8, at least 10, at least 12, at least 15, at least 20, at least 25, at least 30, at least 40, at least 45, at least 50 or more nucleotides. The antisense oligonucleotide may comprise deoxyribonucleotides, ribonucleotides, or a combination of both.


The non-human papilloma pseudovirus or virus-like particle of the invention can contain a morpholino, as a therapeutic agent. The term “morpholino” as used herein relates to a molecule that blocks access of other molecules to small, specific sequences of the base-pairing surfaces of a RNA. Typically, said small specific sequences have a length of about 25 nucleotides. In general, a morpholino comprises a backbone of methylenemorpholine rings and phosphorodiamidate linkages. Morpholinos are commonly also known as morpholino oligomers (MO nucleic acid analogs) and phosphorodiamidate morpholino oligomers (PMO). Morpholinos typically do not lead to degradation of their target RNA molecules, but rather act by sterical blocking, i.e. binding to a target sequence within an RNA and thereby getting in the way of molecules that may otherwise interact with said RNA.


The non-human papilloma pseudovirus or virus-like particle of the invention can contain the (CRISPR/Cas) system. The CRISPR/Cas system has been reviewed, e.g., by Horvath and Barrangou (Science. 2010 Jan. 8; 327(5962):167-70. doi: 10.1126/science.1179555). Engineered DNA-binding molecules such as the clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated proteins (Cas) (CRISPR/Cas) system have been used extensively for genome editing in cells of various types and species. The sequence-specific DNA-binding activities of these engineered DNA-binding molecules can also be utilized for other purposes, such as transcriptional activation, transcriptional repression, chromatin modification, visualization of genomic regions, and isolation of chromatin in a locus-specific manner (Fujita and Fujii, Int J Mol Sci. 2015 October; 16(10): 23143-23164; Gaj et al., Trends in Biotechnology 2013, 31, p. 397-405). For example, the CRISPR/Cas system allows editing of the mouse genome much faster than the previously used techniques and more importantly multiple mutations can be created in a single experiment (Harms et al., Curr Protoc Hum Genet. 2014; 83: 15.7.1-15.7.27.).


The disease to be imaged, diagnosed or treated by the pharmaceutical composition is preferably neurological diseases, stroke, ischemia, cancer, an age-related disease, a genetic disorder, an allergy, an auto-immune disease, an infection by bacteria, virus, fungus or parasite.


The term “cancer” as used herein relates to a disease that is characterized by an uncontrolled growth of aberrant cells. Cancer includes pre-cancerous states well as a manifested or advanced disease states. The classification of a cancer or tumor stage as well as characteristics and symptoms are known in the art and can, for example, be found in the standard text books of medicine, such as Stedman or Pschyrembl. It is understood by those skilled in the art that cancer cells may migrate from the original tumor site and spread to distant site, also known as dissemination and metastasis formation. Examples of cancers include breast cancer, colorectal cancer, pancreatic cancer, prostate cancer, ovarian cancer, cervical cancer, skin cancer, renal cancer, liver cancer, brain cancer, lymphoma, leukemia and lung cancer.


Therapeutic efficacy and toxicity of the pharmaceutical composition according to the present invention can be determined by standard pharmaceutical procedures, e.g. in experimental animals. For example, the so-called ED50 describes the dose therapeutically effective in 50% of the population and the so-called LD50 describes the dose lethal to 50% of the population. The dose ratio between therapeutic and toxic effects is the therapeutic index, and it can be expressed as the ratio, LD50/ED50. The dosage regimen will be determined by the attending physician and by clinical factors. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, age, the particular formulation of the medicament to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. Progress can be monitored by periodic assessment. Dosage recommendations shall be indicated in the prescribers or users instructions in order to anticipate dose adjustments depending on the considered recipient.


In another preferred embodiment, the therapeutic agent is a nucleic acid. Preferably, the nucleic acid is a nucleic acid for gene therapy or vaccination.


The non-human papilloma pseudovirus or virus-like particle of the invention can comprise a gene for gene therapy, as a therapeutic agent. As well known in the art, gene therapy is the insertion of genes into an individual's cells and tissues to treat a disease, and hereditary diseases in which a defective mutant allele is replaced with a functional one.


In somatic cell gene therapy, the therapeutic genes are transferred into any cell other than a gamete, germ cell, gametocyte or undifferentiated stem cell. In germline gene therapy, germ cells (sperm or egg cells) are modified by the introduction of functional genes into their genomes. Modifying a germ cell causes all the organism's cells to contain the modified gene. The change is therefore heritable and passed on to later generations. It is envisaged that the defective gene is either replaced or disrupted. Gene therapeutic approaches using the pharmaceutical composition of the invention include but are not limited to the treatment of retinal diseases Leber's congenital amaurosis, choroideremia, X-linked SCID, ADA-SCID, adrenoleukodystrophy, chronic lymphocytic leukemia (CLL), acute lymphocytic leukemia (ALL), multiple myeloma, haemophilia, peripheral artery disease, including critical limb ischemia and Parkinson's disease.


Alternatively, the non-human papilloma pseudovirus or virus-like particle of the invention can include a DNA (or RNA) for vaccination. The term “vaccine” has been defined elsewhere herein. DNA vaccination is a technique for protecting against disease by injection with genetically engineered DNA so cells directly produce an antigen, producing a protective immunological response. DNA vaccines have potential advantages over conventional vaccines, including the ability to induce a wider range of immune response types. Preferably, vaccination is for Respiratory Syncytial Virus (Grunwald T. et al., 2014. Novel vaccine regimen elicits strong airway immune responses and control of respiratory syncytial virus in nonhuman primates. doi: 10.1128/JVI.02736-13; Kohlmann, R. et al., (2009). Protective efficacy and immunogenicity of an adenoviral vector vaccine encoding the codon-optimized F protein of respiratory syncytial virus. Journal of virology 83, 12601-12610; Ternette, N. et al., (2007). Immunogenicity and efficacy of codon optimized DNA vaccines encoding the F-protein of respiratory syncytial virus. Vaccine 25, 7271-7279; Grunwald T, Ulbert S. 2015 Improvement of DNA vaccination by adjuvants and sophisticated delivery devices: vaccine-platforms for the battle against infectious diseases. doi: 10.7774/cevr.2015.4.1.1.).


The pharmaceutical composition of the invention comprising a nucleic acid for vaccination is safe enough to be administered without danger of clinical infection, do not have toxic side effects, can be administered by an effective route, are stable, and are compatible with vaccine carriers. The vaccines may be administered by a variety of routes, such as orally, parenterally, subcutaneously, mucosally, intravenously or intramuscularly. The vaccine may be used in dosage forms such as capsules, suspensions, elixirs, or liquid solutions. The vaccine may be formulated with an immunologically acceptable carrier or adjuvant, or other excipients. The vaccines are administered in therapeutically effective amounts sufficient to generate an immunologically protective response. The vaccine may be administered in single or multiple doses.


The nucleic acid can be DNA or RNA which can be wildtype, recombinant or chemically synthesized.


Suitable formulations of pharmaceutical compositions comprising the non-human papilloma pseudovirus or virus-like particle of the invention, and nucleic acid as a therapeutic agent for gene therapy or vaccination are known in the art (see, e.g., WO 2011/039646; U.S. Pat. Nos. 6,416,945; 6,991,795 and 7,205,126).


In a preferred embodiment of the pharmaceutical composition of the invention, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier.


A pharmaceutically acceptable carrier according to the present invention must be acceptable in the sense of being compatible with the other ingredients of the formulation and not being deleterious to the recipient thereof. Pharmaceutical carriers may include solid, gel, or liquid carriers such as lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid, glyceryl mono-stearate or glyceryl distearate. Moreover, further suitable carriers are known in the art and can be found for example in science text books such as Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pennsylvania.


In a still further preferred embodiment of the pharmaceutical composition of the invention, the pharmaceutical composition is to be administered by intramuscular injection or via the cutaneous route. Preferably, the pharmaceutical composition is to be administered via intramuscular injection.


As the preferred site of entry of different non-human papillomavirus genera can be either cutaneous or mucosal it is necessary to take this into account when choosing the route of administration for the pharmaceutical composition of the invention. Preferably, the pharmaceutical composition of the invention is to be administered by intramuscular injection or via the cutaneous route, even more preferred via intramuscular injection. It is known to the skilled artisan, that administration and dosage of a therapeutic agent depends on various factors such as the health state of the subject and the disease to be treated.


Moreover, the invention pertains to a non-human papilloma pseudovirus or virus-like particle of the invention for use as a medicament. Preferably, the medicament is for the treatment of a disease selected from the group consisting of Neurological diseases, stroke, ischemia, cancer, age-related disease, genetic disorder, allergy, auto-immune disease, infection by bacteria, virus, fungus or parasite.


The terms “medicament” and “pharmaceutical composition” are used interchangeably herein.


In another embodiment, the medicament is for gene therapy or vaccination.


The definition and explanations given herein above apply mutatis mutandis to the following methods and uses of the present invention.


The invention also relates to a method for treating and/or preventing a disease selected from the group consisting of: neurological diseases, stroke, ischemia, cancer, age-related disease, genetic disorder, allergy, auto-immune disease, infection by bacteria, virus, fungus or parasite in a subject in need thereof comprising administering to said subject a non-human papilloma pseudovirus or virus-like particle comprising a therapeutic agent described herein, in a therapeutically effective amount.


Furthermore, the invention relates to a method for producing a non-human papilloma pseudovirus or virus-like particle comprising the steps of:

    • a) codon-optimizing of a DNA-sequence coding for a non-human papillomavirus capsid protein L1 and/or L2, for expression in eukaryotic cells or cell lines, preferably human cells or cell lines;
    • b) synthesizing of the sequence of step a) and cloning of the synthesized sequence into an expression vector; and
    • c) transfecting of the expression vector of step b) into a cell, thereby producing non-human papilloma pseudovirus or virus-like particles.


In this method of the invention, a non-human papilloma pseudovirus or virus-like particle of the invention is produced by first codon-optimizing a DNA sequence coding for the non-human papilloma L1 and/or L2 capsid protein as defined herein, for expression in eukaryotic cells or cell lines, preferably human cells or cell lines. Thereafter, the mentioned sequence is synthesized and cloned into an expression vector, preferably a mammalian expression vectors. The expression vector is then introduced into appropriate cells such as eukaryotic cells or cell lines, preferably human cells or cell lines, for expression of the codon-optimized non-human papilloma L1 and/or L2 capsid proteins and production of the non-human papilloma virus-like particles of the invention in the mentioned cells or cell lines. Preferred non-human papillomaviruses, capsid proteins L1 and/or L2, and cells are indicated elsewhere herein.


Moreover, the invention relates to the use of ι-carrageenan as transduction enhancer for non-human papilloma pseudovirus or virus-like particles in vitro.


Unexpectedly, it has been found by the inventors that ι-carrageenan induced an increase in transduction efficiency of cells when transduced with non-human papilloma virus-like particles from PcPV1, PlPV1, CcrPV1 and MmPV1. Therefore, ι-carrageenan can be utilized for improving transduction of non-human papilloma virus-like particles, preferably from PcPV1, PlPV1, CcrPV1 and MmPV1.


All references cited in this specification are herewith incorporated by reference with respect to their entire disclosure content and the disclosure content specifically mentioned in this specification.


Tables


Table 1: Corresponds to Table 1 of the publication by A. Rector and M. van Ranst (2013).









TABLE 1







Overview of genomically characterized non-human papillomaviruses.
















Host species











taxonomic
Host

Papillomavirus
Abbreviation


Accession
Isolated



order
species

name
(previous)
Classification
# bp
no.
from
Reference





Artiodactyla
European

Alces
alces

Alces alces
AaPV1
Deltapapillo-
8095
M15953
Cutaneous
Ahola et al.



elk

papillomavirus 1
(EEPV)
mavirus 1


fibroma
(1986)



Yak

Bos

Bos grunniens
BgPV1
Deltapapillo-
7946
JX174437
Cutaneous
Zhu et al.





grunniens

papillomavirus 1

mavirus 4n


fibro-
(2013)










papilloma




Domestic

Bos
taurus

Bos taurus
BPV1
Deltapapillo-
7945
X02346nn
Cutaneous
Chen et al.



cow

papillomavirus 1

mavirus 4


fibro-
(1982)










papilloma






Bos taurus
BPV2
Deltapapillo-
7937
M20219
Cutaneous
Groffand





papillomavirus 2

mavirus 4


fibro-
Lancaster










papilloma
(unpublished)





Bos taurus
BPV3
Xipapillo-
7276
AF486184
Cutaneous
Terai et al.





papillomavirus 3

mavirus 1


papilloma
(2002)





Bos taurus
BPV4
Xipapillo-
7265
X05817
Oral/
Patel et al.





papillomavirus 4

mavirus 1


esophageal
(1987)










papilloma






Bos taurus
BPV5
Epsilonpa-
7841
AF457465
Udder
Terai et al.





papillomavirus 5

pillomavirus 1


fibro-
(2002)










papilloma






Bos taurus
BPV6
Xipapillo-
7296
AJ620208
Udder
Jarrett et al.





papillomavirus 6

mavirus 1


papilloma
(1984)





Bos taurus
BPV7
Dyoxipa-
7412
DQ217793
Teat
Ogawa et al.





papillomavirus 7

pillomavirus 1n


papilloma
(2007)










and healthy











skin






Bos taurus
BPV8
Epsilonpa-
7791
DQ098913
Cutaneous
Tomita et al.





papillomavirus 8

pilloma-


papilloma
(2007)







virus 1









Bos taurus
BPV9
Xipapillo-
7303
AB331650
Teat
Hatama et al.





papillomavirus 9

mavirus 1


papilloma
(2008)





Bos taurus
BPV10
Xipapillo-
7399
AB331651
Teat
Hatama et al.





papillomavirus 10

mavirus 1


papilloma
(2008)





Bos taurus
BPV11
Xipapillo-
7251
AB543507
Cutaneous
Hatama et al.





papillomavirus 11

mavirus 1


papilloma
(2011)





Bos taurus
BPV12
Xipapillo-
7197
JF834523
Tongue
Zhu et al.)





papillomavirus 12

mavirus 2n


epithelial
(2012










papilloma






Bos taurus
BPV13
Deltapapillo-
7961
JQ798171
Ear
Lunardi et al.





papillomavirus 13

mavirus 4n


cutaneous
(2013)










papilloma




Arabian

Camelus

Camelus
CdPV1
Deltapapillo-
7679
HQ912790
Cutaneous
Ure et al.



camel

dromedarius

dromedarius

mavirus 6n



(2011)





papillomavirus 1
CdPV2
Deltapapillo-
7906
HQ912791
fibro-
Ure et al.





Camelus

mavirus 6n


papilloma
(2011)





dromedarius




Cutaneous






papillomavirus 2




fibro-











papilloma




Domestic

Capra

Capra hircus
ChPV1
Phipapillo-
7542
DQ091200
Healthy
Van Doorslaer



goat

hircus

papillomavirus 1

mavirus 1


skin
et al. (2006)



Western

Capreolus

Capreolus
CcaPV1
Deltapa-
8032
EF680235
Cutaneous
Erdelyi et al.



roe deer

capreolus

capreolus
(RdPV1,
pillomavirus 5



(2008)





papillomavirus 1
CcPV1)








White-

Odocoileus

Odocoileus
OvPV1
Deltapa-
8374
M11910nnn
fibro-
Groff and



tailed

virginianus

virginianus
(DPV)
pilloma-


papilloma
Lancaster



deer

papillomavirus 1

virus 2


Cutaneous
(1985)










fibroma




Domestic

Ovis
aries

Ovis aries
DaPV1
Deltapa-
7761
U83594
Cutaneous
Karlis et al.



sheep

papillomavirus 1
(OvPV1)
pilloma-


fibro-
(unpublished)







virus 3


papilloma






Ovis aries
OaPV2
Deltapa-
7758
U83595
Cutaneous
Karlis et al.





papillomavirus 2
(OvPV2)
pilloma-


fibro-
(unpublished)







virus 3


papilloma






Ovies aries
OaPV3
Dyolambda-
7334
FJ796965
Squamous
Alberti et al.





papillomavirus 3

papilloma-


carcinoma
(2010)







virus 1







Reindeer

Rangifer

Rangifer
RtPV1
Deltapa-
8090
AF443292
Cutaneous
Terai et al.





tarandus

tarandus
(RPV)
pilloma-


fibro-
(2002)





papillomavirus 1

virus 1


papilloma




Domestic

Sus
scrofa

Sus scrofa
SsPV1
Dyodeltapa-
7260
EF395818
Healthy
Stevens et al.



pig

domesticus

domesticus

pilloma-


skin
(2008b)





papillomavirus 1

virus 1






Carnivora
Domestic

Canis
lupus

Canis familiaris
CPV1
Lambdapa-
8607
D55633
Oral/
Delius et al.



dog

familiaris

oral
(COPV)
pilloma-


cutaneous
(1994)



(Shar Pei)

papillomavirus

virus 2


papilloma




Domestic

Canis familiaris
CPV2
Taupapillo-
8101
AY722648
Cutaneous
Yuan et al.



dog

papillomavirus 2
(CfPV2)
mavirus 1


papilloma
(2007)



(Golden






on footpad




retriever)











Domestic

Canis familiaris
CPV3
Chipapillo-
7801
DQ295066
Malignant
Tobler et al.



dog

papillomavirus 3

mavirus 1


EV
(2006)



(Rhodesian






lesion




ridgeback)











Domestic

Canis familiaris
CPV4
Chipapillo-
7742
EF584537
Pigmented
Tobler et al.



dog

papillomavirus 4

mavirus 2


lesion
(unpublished)



(European











pug)











Domestic

Canis familiaris
CPV5
Chipapillo-
7810
FJ492743
Pigmented
Lange et al.



dog

papillomavirus 5

mavirus 1


plaque
(2009a)



Domestic

Canis familiaris
CPV6
Lambdapa-
8242
FJ492744
Inverted
Lange et al.



dog

papillomavirus 6

pilloma-


papilloma
(2009a)







virus 3







Domestic

Canis familiaris
CPV7
Taupapillo-
7955
FJ492742
In situ
Lange et al.



dog

papillomavirus 7

mavirus 1


quamous
(2009a)










cell











carcinoma




Domestic

Canis familiaris

Chipapillo-
7784
HQ262536
Pigmented
Lange et al.



dog

papillomavirus 8

mavirus 3n


plaque
(2012b)



(Dachs-

CPV8









hund-mix)











Domestic

Canis familiaris

Chipapillo-
7873
JF800656
Pigmented
Yuan et al.



dog

papillomavirus 9

mavirus 1n


plaque
(2012)



(mixed-

CPV9









breed











blood











hound)











Domestic

Canis familiaris
CPV10
Chipapillo-
7774
JF800657
Pigmented
Luff et al.



dog

papillomavirus 10

mavirus 3n


plaque
(2012)



Domestic

Canis familiaris
CPV11
Chipapillo-
7828
JF800658nn
Pigmented
Yuan et al.



dog

papillomavirus 11

mavirus 1n


plaque
(unpublished)



Domestic

Canis familiaris
CPV12
Chipapillo-
7890
JQ754321
Pigmented
Yuan et al.



dog

papillomavirus 12

mavirus 1n


plaque
(unpublished)



Domestic

Canis familiaris
CPV13
Taupapillo-
8228
JX141478
7826
Lange et al.



dog

papillomavirus 13

mavirus 2n


JQ701802
(2012a)



(mixed-











breed dog)











Domestic

Canis familiaris
CPV14
Chipapillo-
7826
JQ701802
Pigmented
Lange et al.



dog

papillomavirus 14

mavirus 3n


plaque
(2013b)



(Golden











retriever)











Domestic

Canis familiaris
CPV15
Chipapillo-
7776
JX899359
NR
Yuan et al.



dog

papillomavirus 15

mavirus 3n



(unpublished)











CcrPV1



Spotted

Crocuta

Crocuta crocuta

Lambdapa-
8344
HQ585856
Oral
Stevens et al.



hyena

crocuta

papillomavirus 1

pilloma-


papilloma
(2013)







virus n







Domestic

Felis
catus

Felis catus
FcaPV1
Lambdapa-
8300
AF480454
Cutaneous
Tachezy et al.



cat (short-

papillomavirus 1
(FdPV1)
pilloma-


papilloma
(2002a)



haired



virus 1







Persian











cat)











Domestic

Felis catus
FcaPV2
Dyothetapa-
7899
EU796884
Pigmented
Lange et al.



cat

papillomavirus 2
(FdPV2)
pilloma-


plaque
(2009b)







virus 1







Bobcat

Lynx
rufus

Lynx rufus
LrPV1
Lambdapa-
8233
AY904722
Oral
Rector et al.





papillomavirus 1

pilloma-


papilloma
(2007)







virus 1







Asiaticlion

Panthera
leo

Panthera leo
PlpPV1
Lambdapa-
8103
AY904724
Oral
Rector et al.





persica

persica

pilloma-


papilloma
(2007)





papillomavirus 1

virus 1







Raccoon

Procyon

Procyon lotor
PIPV1
Lambdapa-
8170
AY763115
Cutaneous
Rector et al.





lotor

papillomavirus 1

pilloma-


papilloma
(2005b)







virus 4







Cougar

Puma

Puma concolor
PcPV1
Lambdapa-
8321
AY904723
Oral
Rector et al.



(Florida

concolor

papillomavirus 1

pilloma-


papilloma
(2007)



panther)



virus 1







Snow

Uncia
uncia

Uncia uncia
UuPV1
Lambdapa-
8078
DQ180494
Papilloma
Rector et al.



leopard

papillomavirus 1

pillomavirus 1


lower lip
(2007)



Polar

Ursus

Ursus maritimus
UmPV1
Omegapa-
7582
EF536349
Oral
Stevens et al.



bear

maritimus

papillomavirus 1

pilloma-


papilloma
(2008a)







virus 1







California

Zalophus

Zalophus
ZcPV1
Chipapillo-
7584
HQ293213
Skin
Hoffman et al.



sealion

californianus

californianus

mavirusn


carcinoma
(unpublished)





papillomavirus 1








Cetacea
Short-

Delphinus

Delphinus
DdPV1
Upsilonpa-
7852
GU117620
Penile wart
Gottschling



beaked

delphis

delphis

pilloma-



et al. (2011)



common

papillomavirus 1

virus 1







dolphin











Harbor

Phocoena

Phocoena
PphPV1
Omikronpa-
7596
GU117621
Penile
Gottschling



porpoise

phocoena

phocoena

pilloma-


papilloma
et al. (2011)





papillomavirus 1

virus 1









Phocoena
PphPV2
Upsilonpa-
7635
GU117622
Penile
Gottschling





phocoena

pilloma-


papilloma
et al. (2011)





papillomavirus 2

virus 3









Phocoena
PphPV4
Dyopipapillo-
7348
GU117623
Penile
Gottschling





phocoena

mavirus 1


papilloma
et al. (2011)





papillomavirus 4









Burmeis-

Phocoena

Phocoena
PsPV1
Omikronpa-
7879
AJ238373
Genital
Van Bressem



ter’s

spinipinnis

spinipinnis

pilloma-


papilloma
et al. (2007)



porpoise

papillomavirus 1

virus 1







Bottlenose

Tursiops

papillomavirus 1
TtPV1
Upsilonpa-
8089
EU240894
Penilewart
Rector et al.



dolphin

truncatus



pilloma-



(2008)







virus 1









papillomavirus 2
TtPV2
Upsilonpa-
7866
AY956402
Genital
Rehtanz et al.







pilloma-


lesion
(2006)







virus 2









Tursiops truncatus
TtPV3
Upsilonpa-
7915
EU240895
Penilewart
Rector et al.





papillomavirus 3

pilloma-



(2008)







virus 1









Tursiops truncatus
TtPV4
Upsilonpa
7792
JN709469
Genital
Robles-Sikisaka





papillomavirus 4

pilloma-


lesion
et al. (2012)







virus 1n









Tursiops truncatus
TtPV5
Omikronpa-
7853
JN709470
Genital
Robles-Sikisaka





papillomavirus 5

pilloma-


lesion
et al. (2012)







virus 1n









Tursiops truncatus
TtPV6
Omikronpa-
7895
JN709471
Genital
Robles-Sikisaka





papillomavirus 6

pilloma-


lesion
et al. (2012)







virus 1n









Tursiops truncatus
TtPV7
Upsilonpa-
7783
JN709472
Normal
Robles-Sikisaka





papillomavirus 7

pilloma-


genital
et al. (2012)







virus 1n


mucosa



Chiroptera
Common

Miniopterus

Miniopterus
MscPV1
unclassified
7632
JQ814848
Pharyngeal
Wu et al.



bent-

schreibersii

schreibersii




swabor
(2012)



wingbat

papillomavirus 1




anal











swab






Miniopterus

Miniopterus
MscPV1
unclassified
7531
JQ692938
Rectalswab
Tse et al.





schreibersii

schreibersii
TT20F




(2012)





papillomavirus 1











TT20F









Rickett’s

Myotis

Myotis ricketti
MrPV1
unclassified
7339
JQ814847
Pharyngeal
Wu et al.



big-

ricketti

papillomavirus 1




swab or
(2012)



footed Bat






anal swab




Egyptian

Rousettus

Rousettus
RaPV1
Psipapillo-
7970
DQ366842
Squamous
Rector et al.



fruit bat

aegyptiacus

aegyptiacus

mavirus 1


carcinoma
(2006)





papillomavirus 1








Diproto-
Brush-

Bettongia

Bettongia
BpPV1
Dyokappapa-
7743
GU220391
Cutaneous
Bennett et al.


dontia
tailed

penicillata

penicillata

pilloma-


papilloma
(2010)



bettong

papillomavirus 1

virus 1






Erina-
European

Erinaceus

Erinaceus
EePV1
Dyoetapa-
8256
FJ379293
Facial hair
Schulz et al.


ceomorpha
hedgehog

europaeus

europaeus
(EHPV)
pilloma-


follicles
(2009a)





papillomavirus 1

virus 1






Galliformes
Yellow-

Francolinus

Francolinus
FlPV1
Dyoepsilon-
7498
EU188799
Healthy
Van Doorslaer



necked

leucoscepus

leucoscepus
(FLPV)
papilloma-


skin
et al. (2009)



Frankolin

papillomavirus 1

virus 1






agomorpha
New

Oryctolagus

Oryctolagus
OcPV1
Kappapa-
7565
AF227240
Oral
Christensen



Zealand

cuniculus

cuniculus
(ROPV)
pilloma-


papilloma
et al. (2000)



white

papillomavirus 1

virus 1







rabbit











Cottontail

Sylvilagus

Sylvilagus
SfPV1
Kappapa-
7868
K02708
Cutaneous
Giri et al.



rabbit

floridanus

floridanus
(CRPV,
pilloma-


papilloma
(1985)





papillomavirus 1
SPV)
virus 2






Passeriformes
Common

Fringilla

Fringilla coelebs
FcPV1
Etapapillo-
7729
AY057109
Cutaneous
Terai et al.



chaffinch

coelebs

papillomavirus 1
(FPV)
mavirus 1


papilloma
(2002)


Perissodactyla
Domestic

Equus
ferus

Equus caballus
EcPV1
Zetapapillo-
7610
AF498323
Cutaneous
Ghim et al.



horse

caballus

papillomavirus 1
(EQPV)
mavirus 1


papilloma
(2004)





Equus caballus
EcPV2
Dyoiotapa-
7802
EU503122nn
Equine
Scase et al.





papillomavirus 2
(EqPV2)
pilloma-


genital
(unpublished)







virus 1


neoplasia











Aural






Equus caballus
EcPV3
Dyorhopa-
7582
GU384895
plaque
Lange et al.





papillomavirus 3

pilloma-



(2011b)







virus 1n









Equus caballus
EcPV4
Dyoiotapa-
7554
JQ031032
Vulval
(Lange et al.





papillomavirus 4

pilloma-


and
2013a)







virus 2n


inguinal











plaques











Aural






Equus caballus
EcPV5
Dyoiotapa-
7519
JQ031033
plaque
Lange et al.





papillomavirus 5

pilloma-



(2013a)







virus 3n









Equus caballus
EcPV6
Dyorhopa-
7551
JQ965698
Aural
Lange et al.





papillomavirus 6

pilloma-


plaque
(2013a)







virus 1n









Equus caballus
EcPV7
Dyorhopa-
7619
JX035935
Penile
Lange et al.





papillomavirus 7

pilloma-


mass
(2013a)







virus 1n






Primates
Colobus

Colobus

Colobus guereza
CgPV1
Alphapa-
8060
GU014532nn
NR
Wood et al.



monkey

guereza

papillomavirus 1

pilloma-



(2011)







virus 14









Colobus guereza
CgPV2
Betapapillo-
7686
GU014533
Cutaneous
Wood et al.





papillomavirus 2

mavirus 1


papilloma
2011



Cyno-

Macaca

Macaca
MfPV1
Betapapillo-
7588
EF028290
Cutaneous
Joh et al.



molgus

fascicularis

fascicularis

mavirus 1


papilloma
(2009)



macaque

papillomavirus 1











Macaca
MfPV2
Betapapillo-
7632
GU014531
Cutaneous
Wood et al.





fascicularis

mavirus 6


papilloma
(2011)





papillomavirus 2











Macaca
MfPV3
Alphapapillo-
7935
EF558839
Cervical
Chen et al.





fascicularis
(RhPV-d)
mavirus 12


intra-
(2009)





papillomavirus 3




epithelial











neoplasia






Macaca
MfPV4
Alphapapillo-
7950
EF558841
Cervical
Chen et al.





fascicularis

mavirus 12


intra-
(2009)





papillomavirus 4




epithelial











neoplasia






Macaca
MfPV5
Alphapapillo-
7990
EF558843
Cervical
Chen et al.





fascicularis
(MfPV-a)
mavirus 12


intra-
(2009)





papillomavirus 5




epithelial











neoplasia






Macaca
MfPV6
Alphapapillo-
7943
EF558840
NR
Chen et al.





fascicularis

mavirus 12



(2009)





papillomavirus 6











Macaca
MIPV7
Alphapapillo-
8063
EF558838
NR
Chen et al.





fascicularis

mavirus 12



(2009)





papillomavirus 7











Macaca
MfPV8
Alphapapillo-
8001
EF558842
Cervical
Chen et al.





fascicularis
(RhPV-a)
mavirus 12


intra-
(2009)





papillomavirus 8




epithelial











neoplasia






Macaca
MfPV9
Alphapapillo-
7988
EU490516
NR
Chen et al.





fascicularis

mavirus 12



(2009)





papillomavirus 9











Macaca
MiPV10
Alphapapillo-
7920
EU490515
NR
Chen et al.





fascicularis

mavirus 12



(2009)





papillomavirus 10











Macaca
MfPV11
Alphapapillo-
8014
GQ227670
NR
Chen et al.





fascicularis
(RhPV-b)
mavirus 12



(2009)





papillomavirus 11









Rhesus

Macaca

Macaca mulatta
MmPV1
Alphapapillo-
8028
M60184
Mucosal
Ostrow et al.



macaque

mulatta

papillomavirus 1
(RhPV1)
mavirus 12


genital
(1991)










carcinoma




Rickett’s

Myotis

Myotis ricketti
MrPV1
unclassified
7339
JQ814847
Pharyngeal
Wu et al.



big-

ricketti

papillomavirus 1




swabor
(2012)



footed Bat






anal swab




Egyptian

Rousettus

Rousettus
RaPV1
Psipapillo-
7970
DQ366842
Squamous
Rector et al.



fruit bat

aegyptiacus

aegyptiacus

mavirus 1


carcinoma
(2006)





papillomavirus 1








Diprotodontia
Brush-

Bettongia

Bettongia
BpPV1
Dyokappa-
7743
GU220391
Cutaneous
Bennett et al.



tailed

penicillata

penicillata

papillo-


papilloma
(2010)



bettong

papillomavirus 1

mavirus 1






Erina-
European

Erinaceus

Erinaceus
EePV1
Dyoetapa-
8256
FJ379293
Facial hair
Schulz et al.


ceomorpha
hedgehog

europaeus

europaeus
(EHPV)
pilloma-


follicles
(2009a)





papillomavirus 1

virus 1






Galliformes
Yellow-

Francolinus

Francolinus
FlPV1
Dyoepsilon-
7498
EU188799
Healthy
Van Doorslaer



necked

leucoscepus

leucoscepus
(FLPV)
papillo-


skin
et al. (2009)



Frankolin

papillomavirus 1

mavirus 1






Lagomorpha
New

Oryctolagus

Oryctolagus
OcPV1
Kappapa-
7565
AF227240
Oral
Christensen



Zealand

cuniculus

cuniculus
(ROPV)
pilloma-


papilloma
et al. (2000)



white

papillomavirus 1

virus 1







rabbit











Cottontail

Sylvilagu

Sylvilagus
SfPV1
Kappapa-
7868
K02708
Cutaneous
Giri et al.



rabbit

sfloridanus

floridanus
(CRPV,
pilloma-


papilloma
(1985)





papillomavirus 1
SPV)
virus 2






Passeri-
Common

Fringilla

Fringilla coelebs
FcPV1
Etapapillo-
7729
AY057109
Cutaneous
Terai et al.


formes
chaffinch

coelebs

papillomavirus 1
(FPV)
mavirus 1


papilloma
(2002)


Perisso-
Domestic

Equus
ferus

Equus caballus
EcPV1
Zetapapillo-
7610
AF498323
Cutaneous
Ghim et al.


dactyla
horse

caballus

papillomavirus 1
(EQPV)
mavirus 1


papilloma
(2004)





Equus caballus
EcPV2
Dyoiotapa-
7802
EU503122nn
Equine
Scase et al.





papillomavirus 2
(EqPV2)
pilloma-


genital
(unpublished)







virus 1


neoplasia











Aural






Equus caballus
EcPV3
Dyorhopa-
7582
GU384895
plaque
Lange et al.





papillomavirus 3

pilloma-



(2011b)







virus 1n









Equus caballus
EcPV4
Dyoiotapa-
7554
JQ031032
Vulval and
(Lange et al.





papillomavirus 4

pilloma-


inguinal
2013a)







virus 2n


plaques











Aural






Equus caballus
EcPV5
Dyoiotapa-
7519
JQ031033
plaque
Lange et al.





papillomavirus 5

pilloma-



(2013a)







virus 3n









Equus caballus
EcPV6
Dyorhopa-
7551
JQ965698
Aural
Lange et al.





papillomavirus 6

pilloma-


plaque
(2013a)







virus 1n









Equus caballus
EcPV7
Dyorhopa-
7619
JX035935
Penile
Lange et al.





papillomavirus 7

pilloma-


mass
(2013a)







virus 1n






Primates
Colobus

Colobus

Colobus guereza
CgPV1
Alphapa-
8060
GU014532nn
NR
Wood et al.



monkey

guereza

papillomavirus 1

pilloma-



(2011)







virus 14









Colobus guereza
CgPV2
Betapapillo-
7686
GU014533
Cutaneous
Wood et al.





papillomavirus 2

mavirus 1


papilloma
2011



Cyno-

Macaca

Macaca
MfPV1
Betapapillo-
7588
EF028290
Cutaneous
Joh et al.



molgus

fascicularis

fascicularis

mavirus 1


papilloma
(2009)



macaque

papillomavirus 1











Macaca
MfPV2
Betapapillo-
7632
GU014531
Cutaneous
Wood et al.





fascicularis

mavirus 6


papilloma
(2011)





papillomavirus 2











Macaca
MfPV3
Alphapapillo-
7935
EF558839
Cervical
Chen et al.





fascicularis
(RhPV-d)
mavirus 12


intra-
(2009)





papillomavirus 3




epithelial











neoplasia






Macaca
MfPV4
Alphapapillo-
7950
EF558841
Cervical
Chen et al.





fascicularis

mavirus 12


intra-
(2009)





papillomavirus 4




epithelial











neoplasia






Macaca
MfPV5
Alphapapillo-
7990
EF558843
Cervical
Chen et al.





fascicularis
(MfPV-a)
mavirus 12


intra-
(2009)





papillomavirus 5




epithelial











neoplasia






Macaca
MfPV6
Alphapapillo-
7943
EF558840
NR
Chen et al.





fascicularis

mavirus 12



(2009)





papillomavirus 6











Macaca
MfPV7
Alphapapillo-
8063
EF558838
NR
Chen et al.





fascicularis

mavirus 12



(2009)





papillomavirus 7











Macaca
MfPV8
Alphapapillo-
8001
EF558842
Cervical
Chen et al.





fascicularis
(RhPV-a)
mavirus 12


intra-
(2009)





papillomavirus 8




epithelial











neoplasia






Macaca
MfPV9
Alphapapillo-
7988
EU490516
NR
Chen et al.





fascicularis

mavirus 12



(2009)





papillomavirus 9











Macaca
MfPV10
Alphapapillo-
7920
EU490515
NR
Chen et al.





fascicularis

mavirus 12



(2009)





papillomavirus 10











Macaca
MfPV11
Alphapapillo-
8014
GQ227670
NR
Chen et al.





fascicularis
(RhPV-b)
mavirus 12



(2009)





papillomavirus 11









Rhesus

Macaca

Macaca mulatta
MmPV1
Alphapapillo-
8028
M60184
Mucosal
Ostrow et al.



macaque

mulatta

papillomavirus 1
(RhPV1)
mavirus 12


genital
(1991)










carcinoma




Bonobo

Pan

Pan paniscus
PpPV1
Alphapapillo-
7902
X62844
Oral
Van Ranst





paniscus

papillomavirus 1
(PCPV)
mavirus 10


epithelial
et al. (1991)



Common

Pant

Pan troglodytes
PtPV1
Alphapapillo-
7889
AF020905
Oral
Scinicariello



chim-

roglodytes

papillomavirus 1
(CCPV)
mavirus 10


epithelial
et al.



panzee






hyperplasia
(unpublished)



Hamadryas

Papio

Papio hamadryas
PhPV1
Alphapapillo-
8008
JF304764
Cervical
Bergin



baboon

hamadryas

papillomavirus

mavirus 12n


samples
et al. (2013)





Anubis

type 1




(CIN1)



Psittaci-
African

Psittacus

Psittacus
PePV1
Thetapapillo-
7304
AF502599
Cutaneous
Tachezy


formes
gray

erithacus

erithacus
(PePV)
mavirus 1


papilloma
et al. (2002b)



Parrot

papillomavirus 1








Rodentia
Wood-

Apodemus

Apodemus
AsPV1
Pipapillo-
7589
HQ625440
Normal
Schulz



mouse

sylvaticus

sylvaticus

mavirus 2n


skin
et al. (2012)





papillomavirus 1




(ear)




North

Erethizon

Erethizon
EdPV1
Sigmapapillo-
7428
AY684126
Cutaneous
Rector



American

dorsatum

dorsatum

mavirus 1


papilloma
et al. (2005a)



porcupine

papillomavirus 1









Natal

Mastomys

Mastomys
MnPV1
Iotapapillo-
7687
U01834
Cutaneous
Tan et al.



multi-

natalensis

natalensis
(MrPV,
mavirus 1


papilloma
(1994)



mammate

papillomavirus 1
MmPV)








mouse











Southern

Mastomys

Mastomys
McPV2
Pipapillo-
7522
DQ664501
Skin
Nafz et al.



multi-

coucha

coucha

mavirus 2


carcinoma
(2008)



mammate

papillomavirus 2









mouse











Syrian

Mesocricetus

Mesocricetus
MaPV1
Pipapillo-
7647
E15111
Oral
Iwasaki et al.



golden

auratus

auratus
(HaOPV)
mavirus 1


papilloma
(unpublished);



hamster

papillomavirus 1





patent JP











1998042875-A6



Eurasian

Micromys

Micromys
MmiPV1
Pipapillo-
7393
DQ269468
Cutaneous
Van Doorslaer



harvest

minutus

minutus
(MmPV1)
mavirus 2


papilloma
et al. (2007)



mouse

papillomavirus 1









Laboratory

Mus

Mus musculus
MmuPV1
Pipapillo-
7510
GU808564
Cutaneous
Joh et al.



mouse

musculus

papillomavirus 1

mavirus 2


papilloma
(2011)



Deer

Peromyscus

Peromyscus
PmPV1
unclassified
7704
JF755418
Feces
Phan et al.



mouse

maniculatus

maniculatus





(2011)





papillomavirus 1









Norway

Rattus

Rattus norvegicus
RnPV1
Pipapillo-
7378
GQ180114
Normal
Schulz et al.



rat

norvegicus

papillomavirus 1

mavirus 2


oral
(2009b)










mucosa






Rattus norvegicus
RnPV2
Iotapapillo-
7724
HQ625441
Rectal-
Schulz et al.





papillomavirus 2

mavirus 2n


smear
(2012)


Sirenia
Florida

Trichechus

Trichechus
TmPV1
Rhopapillo-
7722
AY609301
Cutaneous
Rector et al.



manatee

manatus

manatus

mavirus 1


papilloma
(2004a)





latirostris

latirostris











papillomavirus 1











Trichechus
TmPV2
Rhopapillo-
7855
JN709473
NR
Wellehan





manatus

mavirus 1n



et al.





latirostris





(unpublished)





papillomavirus 2








Squamata
Diamond

Morelia

Morelia spilota
MsPV1
Dyomupa-
7048
HQ262535nn
Pigmented
Lange et al.



python

spilota

papillomavirus 1

pilloamvirus 1n


papilloma-
(2011a)





spilota






like lesion



Testudines
Logger-

Caretta

Caretta caretta
CcPV1
Dyozetapa-
7020
EU493092
Cutaneous
Herbst et al.



head

caretta

papillomavirus 1

pilloma-


papilloma
(2009)



sea turtle



virus 1







Green sea

Chelonia

Chelonia mydas
CmPV1
Dyozetapa-
6953
EU493091
Cutaneous
Herbst et al.



turtle

mydas

papillomavirus 1

pilloma-


fibro-
(2009)







virus 1


papilloma





NR: Not reported.



nClassification not yet approved by ICTV.




nnA revised sequence is available on the PaVE website(http://pave.niaid.nih.gov).




nnnE2 was resequenced and is available under accession number AY8032














TABLE 2





Non-human papilloma viruses and accession numbers.


Non-human papillomaviruses















 (1) Bettongia penicillata papillomavirus 1 (NC_014143)


 (2) Canine oral papillomavirus (NC_001619)


 (3) Capra hircus papillomavirus type 1 (NC_008032)


 (4) Capreolus capreolus papillomavirus 1 (NC_011051)


 (5) Caretta caretta papillomavirus 1 (NC_011530)


 (6) Cottontail Rabbit Papillomavirus (NC_001541.1)


 (7) Cottontail Rabbit Papillomavirus Papillomavirus sylvilagi a4 (AJ404003.1)


 (8) Cottontail Rabbit (Shope) Papillomavirus (K02708.1)


 (9) Cottontail Rabbit Papillomavirus Strain Hershey (JF303889.1)


(10) Cottontail Rabbit Papillomavirus Subtype b (AJ243287.1)


(11) Crocuta crocuta papillomavirus 1 (NC_018575)


(12) Deer papillomavirus (NC_001523)


(13) Equine papillomavirus 2 (NC_012123)


(14) Equus caballus papillomavirus - 1 (NC_003748)


(15) Erethizon dorsatum papillomavirus type 1 (NC_006951)


(16) Erinaceus europaeus papillomavirus (NC_011765)


(17) European Elk Papillomavirus (NC_001524.1)


(18) European hedgehog papillomavirus (NC_011765.1)


(19) Francolinus leucoscepus papillomavirus 1 (NC_013117)


(20) Mastomys natalensis papillomavirus (NC_001605)


(21) Multimammate rat papillomavirus (U01834.1)


(22) Mus musculus papillomavirus type1 (NC_014326.1)


(23) Old World harvest mouse Papillomavirus (NC_008682.1)


(24) Ovine papillomavirus type 3 isolate Sar1 (FJ796965)


(25) Phocoena spinipinnis papillomavirus (AJ238373)


(26) Phocoena spinipinnis papillomavirus (NC_003348)


(27) Phodopus sungorus papillomavirus type 1 (HG939559.1)


(28) Procyon lotor papillomavirus 1 (PlPV-1) (NC_007150)


(29) Psittacus erithacus timneh papillomavirus (NC_003973)


(30) Rabbit Oral papillomavirus (NC_002232)


(31) Rhesus Papillopmavirus Type 1 (M60184.1)


(32) Rousettus aegyptiacus papillomavirus type 1 (NC_008298.1)


(33) Rupicapra rupicapra papillomavirus 1 (NC_023895)


(34) Sus scrofa papillomavirus type 1 (NC_011280)


(35) Talpa europaea papillomavirus isolate Bruges/2009/22 (KC460987)


(36) Trichechus manatus latirostris papillomavirus 1 (NC_006563)


(37) Tursiops truncatus papillomavirus 2 (NC_008184)


(38) Ursus maritimus papillomavirus 1 (NC_010739)


(39) Western roedeer papillomavirus 1 isolate CcPV-1 (EF680235.1)


(40) Puma concolor Papillomavirus 1 (PcPV1), (AY904723)


(41) Colobus guereza (Mantelaffe) papillomavirus type 1 isolate CgPV1 (GU014532.1)


(42) Rhesus papillomavirus type 1b isolate Mac170 (EF591300.1)


(43) Macaca fascicularis papillomavirus type 6, isolate Mac39 (EF558840.1)


(44) Macaca fascicularis papillomavirus type 11, isolate Mac1637 (GQ227670.1)


(45) Common chimpanzee papillomavirus 1 (AF020905.1)
















TABLE 3







Non-human papilloma viruses used in the


Examples of the present application.









Papilloma virus
Abbreviation
GenBank-Nr.






Caretta caretta papillomavirus 1

CcPV1
NC_011530



Colobus guereza papillomavirus type 1

CgPV1
GU014532.1


Common chimpanzee papillomavirus 1
PtPV1
AF020905.1



Crocuta crocuta papillomavirus 1

CcrPV1
NC_018575



Macaca fascicularis papillomavirus

MfPV11
GQ227670.1


type 11, isolate Mac1637


Macaca fascicularis papillomavirus
MfPV6
EF558840.1


type 6, isolate Mac39



Procyon lotor papillomavirus 1

PlPV1
NC_007150



Puma concolor papillomavirus 1

PcPV1
AY904723


Rhesus papillomavirus type 1b isolate
MmPV1
EF591300.1


Mac170



Rousettus aegyptiacus papillomavirus

RaPV1
NC_008298.1


type 1









FIGURES


FIG. 1: Western Blots of fractions after purification by ultracentrifugation.


All produced non-human papilloma PsVs were purified by density gradient ultracentrifugation. Subsequently, the collected fractions were separated by SDS-PAGE, blotted onto a nitrocellulose membrane and probed for L1. (A) MfPV6, (B) CcrPV1, (C) CcPV1, (D) CgPV1, (E) MmPV1, (F) MfPV11, (G) PcPV1, (H) PlPV1.



FIG. 2: Luciferase assay 72 h after transduction of HEK293TT cells with different non-human papilloma pseudovirions (PsVs).


50.000 HEK293TT cells were transduced by adding 10111 of fractions 3 to 8 of ultracentrifugation-purified non-human papilloma PsVs. Cell culture supernatant was used for luciferase assay 72 h after transduction. (A) Relative light units (RLU) measured after transduction with at least two independent PsV preparations. Shown are mean RLU values of the fractions yielding the highest transduction of one preparation. (B) RLU for fractions 3-8 after transduction with PcPV1 and MfPV11 PsVs, showing the typical peak of transducing PsVs between OptiPrep fractions 4 and 6.



FIG. 3: Transmission electron microscopy of MfPV11 and PcPV1 VLPs.


Purified VLPs were fixed for 24 h at room temperature with formaldehyde, contrasted with phosphotungstic acid and analyzed by transmission electron microscopy. PcPV1 (A), MfPV11 (B).



FIG. 4: Titration of PsVs by transduction of HEK293TT cells.


HEK293TT cells were transduced with indicated PsVs carrying pEGFP as reporter plasmid. 72 h after transduction, GFP-positive cells were counted and titer was calculated as transducing units per ml. PsVs. Each data point represents one PsV preparation (A). To compare the amount of transducing units with amount of particles carrying the pEGFP reporter plasmid, DNA was isolated from PsVs and quantified by qPCR. Titer is calculated as pEGFP plasmids per ml (B).



FIG. 5: Effect of ι-carrageenan on transduction with PcPV1 and MfPV11.


Different doses of ι-carrageenan were added to cell culture medium immediately before transduction of HEK293TT cells with PcPV1 PsVs carrying a G.Luc reporter plasmid. Luciferase assay was performed 72 h after transduction. Statistical analysis was performed by 2way ANOVA and Tukey's multiple comparison test (A). Additionally, HEK293TT cells were transduced with PcPV1 and MfPV11 PsVs carrying a pEGFP reporter plasmid with (+) and without (−) addition of 10 μg/ml ι-carrageenan. GFP-positive cells were counted 72 h after transduction and transducing units were calculated. Each data point represents one experiment with one PsV preparation. Statistical analysis was performed using t-test (B).



FIG. 6: Bioluminescence imaging after intramuscular application.


0.2 μL of PcPV1 and MfPV11 PsVs carrying a firefly luciferase (F.Luc) reporter plasmid were added to 8000 HEK293TT cells for transduction. 72 h later, cells were lysed and lysate was used for F.Luc assay (A). 50 μl of the above analyzed PsV preparations were injected into the left thigh muscle. Bioluminescent imaging was performed approx. 3 h after injection (day 0) to test for any free F.Luc and subsequently in a weekly manner (B).


EXAMPLES

The invention will be merely illustrated by the following Examples. The said Examples shall, whatsoever, not be construed in a manner limiting the scope of the invention.


Example 1: Materials and Methods

1.1 Cells


HEK293TT cells were maintained in Dulbecco's modified Eagle medium (DMEM), supplemented with 10% fetal bovine serum (Gibco), 100 U/ml Penicillin-Streptomycin (Gibco), GlutaMAX and 25 mM glucose. The 293TT cell line was originally generated by Christopher N. Buck by transfection of 293T cells with linearized pTIH plasmid to achieve an increased expression of the SV40 t antigen (C. B. Buck, D. V. Pastrana, D. R. Lowy, J. T. Schiller, Efficient Intracellular Assembly of Papillomaviral Vectors, Journal of virology. 78, 751-757 (2004), doi:10.1128/JVI.78.2.751-757.2004). For selection, 400 μg/ml of hygromycin (Santa Cruz) were added to cell culture medium.


1.2 Plasmid Construction and Propagation


DNA-sequences coding for papillomavirus capsid proteins L1 and L2 were codon-optimized for expression in human cells and synthesized by GeneArt (Invitrogen, Regensburg, Germany). L1- and L2-sequences were cloned into the multiple cloning site of the mammalian expression vector pcDNA3.1+(Invitrogen), the two sequences being connected by an IRES-sequence. Cloning as well as plasmid propagation was performed in E. coli DH5a (New England Biolabs). Plasmids for transfection were prepared using a NucleoBond Plasmid PC500 Maxiprep Kit (Macherey Nagel). As reporter plasmids, pCMV-G.Luc (gaussia luciferase reporter), pCR-Luc3 (firefly luciferase reporter) and pEGFP (GFP reporter) were used.


1.3 Transfection


For non-human papilloma virus-like particle—(VLP) and pseudovirion—(PsV) production, approx. 18 h prior to transfection 6×106 HEK293TT cells were seeded in a 75 cm2 cell culture flask. Transfection mix was prepared by adding 19 μg of the plasmid coding for papillomavirus L1 and L2 and 19 μg of the reporter plasmid to 1 ml of DMEM. Last, 50 μl of polyethyleneimine (1 mg/ml) was added and the preparation was incubated at room temperature for 10 min. Cell culture media was changed to DMEM supplemented with 1.5% FBS and penicillin/streptomycin before adding the transfection mix. Approximately 16 h after transfection, medium was removed, cells were washed with PBS and complete cell culture medium was added.


1.4 VLP and PsV Harvest


Harvest of VLPs and PsVs was performed approximately 40 hours after transfection following the standard protocol of Buck et Al. (C. Buck, D. Pastrana, D. Lowy, J. Schiller, in Human Papillomaviruses, C. Davy, J. Doorbar, Eds. (Humana Press, 2006), vol. 119, pp. 445-462) with a few modifications. Briefly, cells lysed by adding Triton-X 100 at a final concentration of 0.5% in PBS containing an additional 9.5 mM MgCl2, 10 mM HEPES and 25 mM ammonium sulfate. DNA was digested by addition of Benzonase nuclease (final concentration ≥0.25 U/up and Plasmid Safe exonuclease (final concentration 0.01 U/μl) for 24 hours. The lysate was chilled on ice, centrifuged at 5,000 g for 10 min at 4° C. After removal of the VLP or PsV containing supernatant, the remaining cell pellet was resuspended in 100 μl of PBS containing an additional 0.8M NaCl. After initial screening experiments, PBS without additional NaCl was used for PcPV1 preparations. The resuspended cells were again centrifuged as above and supernatant was pooled with the supernatant from the first centrifugation step. This resulting clarified lysate was either stored at −80° C. or purified by ultracentrifugation. Density gradient ultracentrifugation was performed using OptiPrep (Axis-Shield) diluted with PBS+0.8M NaCl to 27%, 33%, and 39%. Gradients were cast by underlayering in a 5 ml tube (Beckman Coulter). Clarified lysate was layered on top and tubes were centrifuged in an SW 55 Ti rotor (Beckman Coulter) at 50,000 rpm (234,000 g) for 3.5 hours at 16° C. After centrifugation, the upper layer up to the transition from 27% to 33% OptiPrep was discarded and subsequently 12 fractions of 250 μl each were collected by pipetting into siliconized 1.5 ml tubes.


Alternatively to ultracentrifugation with OptiPrep, Percoll (GE Healthcare) was used for purification of MfPV11 and HPV16 PsVs. Clarified supernatant was layered on top of 4.5 ml of Percoll diluted to 58.3% with PBS+0.8M NaCl. Tubes were centrifuged in an SW 55 Ti rotor at 30,000 rpm for one hour at 16° C. After centrifugation, supernatant was removed until above the Percoll-pellet about 500 μl still remained, which were collected.


1.5 Transduction with Non-Human Papilloma PsVs


24 h prior to transduction 50,000 HEK293TT cells per well were seeded in a 24-well plate or 8000 HEK293TT cells per well were seeded in a 96-well plate. Non-human papilloma PsVs were added directly to cell culture media. When gaussia luciferase was used as reporter, 20 μl of cell culture supernatant was collected and stored at −20° C. until luciferase-assay was performed to check for free luciferase present in PsV suspension. In experiments with t-carrageenan (Sigma-Aldrich), ι-carrageenan dissolved in PBS was added to cell culture media at the indicated concentrations immediately prior to addition of PsVs. All transduction experiments were performed in triplicates.


1.6 Quantification of Transduction


Gaussia luciferase assay was performed 72 h after transduction by transferring 20 μl of cell culture supernatant into a black luciferase plate (NUNC). Substrate was prepared by 1:1000 dilution of 2 mM coelenterazine (p.j.k.) in assay buffer (1.1M NaCl, 220 mM K2HPO4/KH2PO4, 0.44 mg/ml BSA, 1.3 mM NaN3, pH 5). Centro XS3 LB 960 Microplate Luminometer (Berthold, Bad Wildbad, Germany) was used to inject 100 μl of the substrate, and measurement of relative light units was performed for 1 sec after a delay of 1 sec after injection for each individual well. Background measurements of cell culture media removed directly after transduction were subtracted from measurements 72 h after transduction. Firefly luciferase assay was performed 72 h after transduction by lysing cells for 2 mins by adding Bright-Glo reagent (Promega) and performing the measurement using a Centro XS3 LB 960 Microplate Luminometer within 5 min after addition of the reagent. When GFP was used as reporter, GFP-positive cells were counted 72 h after transduction and transducing units per ml were calculated.


1.7 Western Blot


20 μl of each fraction was mixed with β-mercaptoethanol-containing loading buffer and proteins were separated by SDS-PAGE. After blotting onto a nitrocellulose membrane, membranes were blocked with 5% non-fat dry milk in PBS-T (PBS containing 0.1% Tween-20) and incubated over night with MD2H11 antibody (from Martin Müller, DKFZ Heidelberg). MD2H11 is directed against a conserved sequence in human papillomavirus capsid protein L1. After incubation with the secondary antibody (polyclonal sheep anti-mouse IgG (H+L), peroxidase-conjugated, Jackson Immuno), the signal was detected by chemiluminescence with ECL substrate (Pierce) using an Intas chemo star system.


1.8 Extraction of DNA from Non-Human Papilloma PsVs and Quantitative PCR


10 μl of pseudovirus samples were subjected to DNA-digest with 4 units DNaseI (NEB) for 60 min at 37° C. to remove any residual DNA that may still be present. DNaseI was heat inactivated for 30 min at 75° C. before DNA was extracted using the Qiamp MinElute Virus Spin Kit (Qiagen). DNA was eluted in 100 μl, of which 5 μl were used per reaction in qPCR. QuantiNova SYBR Green PCR Kit (Qiagen) was used for PCR reaction with the following primers for GFP: 5′ ATC CTG GTC GAG CTG GAC GG 3′ (forward) (SEQ ID NO: 41) and 5′ GAC GTA GCC TTC GGG CAT GG 3′ (reverse) (SEQ ID NO: 42). In order to quantify the extracted DNA, a standard curve was created by diluting pEGFP plasmid to contain 3×105 to 30 copies per reaction.


1.9 Transmission Electron Microscopy


Density gradient ultracentrifugation purified VLPs were fixed for 24 h with 2% formaldehyde in the presence of 0.05M HEPES. Samples were contrasted with phosphotungstic acid before analysis by transmission electron microscopy.


1.10 Mice


9-12 weeks old female BALB/c mice were obtained from in-house breeding. Mice were kept in isolated ventilated cages with unrestricted access to water and rodent chow. All animal experiments were carried out in accordance with the EU Directive 2010/63/EU for animal experiments and were approved by local authorities (No.: TVV 49/15). For in vivo transduction experiments, 50 μl of non-human papilloma PsV suspension was injected into the left thigh muscle under inhalative isoflurane anesthesia.


1.11 Bioluminescence Imaging


200 μl of d-luciferin (15 mg/ml in PBS) were injected intraperitoneally after inhalative isoflurane aesthesia of the mice. 20 min after injection, luminescent images were acquired (1 min exposure, medium binning and f/1) using an IVIS SPECTRUM (Xenogen, Perkin Elmer).


1.12 Statistical Analysis


Statistical analyses were performed using GraphPad Prism6. Differences were regarded as significant for p<0.05. Statistically significant differences are indicated as follows: *=p<0.05, **=p<0.01, ***=p<0.001, ns=not significant.


Example 2: Results

2.1 Analysis of Ten Different Non-Human Papilloma PsVs


Aim of the present study was to explore the abundance of non-human (nh) papillomaviruses for their suitability to be used as gene carriers. Sequences for capsid proteins L1 and L2 of ten different papilloma viruses were identified in NCBI's GenBank database (table 3, abbreviations as published (A. Rector, M. van Ranst, Animal papillomaviruses, Virology. 445, 213-223 (2013), doi:10.1016/j.virol.2013.05.007)). Based on the successful application of HPV PsVs as gene delivery vectors (B. S. Graham et al., Mucosal delivery of human papillomavirus pseudovirus-encapsidated plasmids improves the potency of DNA vaccination, Mucosal immunology. 3, 475-486 (2010), doi:10.1038/mi.2010.31; R. C. Kines et al., Vaccination with Human Papillomavirus Pseudovirus-Encapsidated Plasmids Targeted to Skin Using Microneedles, PLoS ONE. 10, e0120797 (2015), doi:10.1371/journal.pone.0120797), half of the analyzed papilloma viruses belong to the group of alpha-papillomaviruses and naturally infect non-human primates. After synthesis of the codon-optimized DNA-sequences and cloning into pcDNA3.1+ expression vector, VLPs were produced in HEK293TT cells and purified by OptiPrep density gradient ultracentrifugation. First analyses were performed by western blotting of the resulting fractions to check for the expected pattern in which the majority of the purified VLPs would be found in fractions 4-6 due to their size and molecular weight. Eight of the ten non-human papilloma VLPs were detectable by western blot (FIG. 1) in the expected fractions. PtPV and RaPV VLPs were not detectable, but it is possible that the MD2H11 antibody does not bind to the respective L1 proteins. In order to test the functionality of the non-human papilloma PsVs as gene carriers, PsVs carrying pCMV-G.Luc as reporter plasmid were produced and used to transfect HEK293TT cells. 10 μl per fraction after ultracentrifugation were used to transduce 50,000 HEK293TT cells in a 24-well plate. Luciferase assays revealed at least some low level of luciferase expression after transduction for all tested PsVs, with the exception of PtPV. Luciferase expression—as indirect measure for transduction efficiency—differed substantially between the individual papillomavirus types (FIG. 2, A). After several PsV preparations of each non-human papillomavirus type, PcPV1 and MfPV11 proved to be the types that not only lead to the highest transduction rates, but can also be produced very reliably. Thus, in the following experiments PcPV1 and MfPV11 were analyzed further.


2.2 PcPV1 and MfPV11 as Gene Vectors


In order to confirm the formation of papillomavirus capsid structures, PcPV1 and MfPV11 VLPs were produced and purified by OptiPrep ultracentrifugation. Electron microscopic analysis revealed that PcPV1 and MfPV11 indeed form capsids resembling papilloma virions as published previously by others (C. Cerqueira et al., Efficient Production of Papillomavirus Gene Delivery Vectors in Defined In Vitro Reactions, Molecular therapy. Methods & clinical development. 5, 165-179 (2017), doi:10.1016/j.omtm.2017.04.005; Q. Zhao et al., Disassembly and reassembly of human papillomavirus virus-like particles produces more virion-like antibody reactivity, Virology journal. 9, 52 (2012), doi:10.1186/1743-422X-9-52). As transduction with gaussia luciferase reporter only provides an indirect measure of transduction efficiency, the inventors repeated transduction experiments with PcPV11 and MfPV11 PsVs carrying pEGFP as reporter to be able to determine transducing units by quantifying GFP-positive cells 72 h after transduction (FIG. 4, A). Additionally, DNA was extracted from PsVs and pEGFP was quantified by qPCR. The titer expression in pEGFP-plasmids per ml is based on the assumption that one plasmid is packaged per PsV (FIG. 4, B). Analysis by qPCR reveals a drastic difference between the amount of PsVs with a packaged reporter plasmid and the amount of transducing units, leading to the conclusion that not the number of produced particles but rather the efficiency of transduction is the limiting factor when using papilloma PsVs for gene transfer. In order to further characterize PcPV1 and MfPV11 PsVs, the inventors tested the ability of ι-carrageenan to inhibit transduction as it had been described before (C. B. Buck et al., Carrageenan is a potent inhibitor of papillomavirus infection, PLoS pathogens. 2, e69 (2006), doi:10.1371/journal.ppat.0020069). While t-carrageenan did indeed prevent transduction with MfPV11 PsVs when it was added to cell culture medium together with the PsVs, the observed effect was just contrary for PcPV1 PsVs (FIG. 5, B). When ι-carrageenan was added to PcPV1 PsVs upon transduction, the expression of the gaussia luciferase reporter protein was significantly increased (FIG. 5, A). This experiment was repeated with pEGFP as reporter plasmid and confirmed that the observed effect is due to a larger number of transduced cells and not to an increased amount of expressed reporter plasmid (FIG. 5, B). A similar ι-carrageenan induced increase in transduction efficiency was also observed for PlPV1, CcrPV1 and MmPV1 PsVs, while transduction with MfPV6 PsVs was inhibited (data not shown).


2.3 MfPV11 and PcPV1 as Gene Carriers In Vivo


It is worth noting that transduction of cell lines with PcPV1 and MfPV11 PsVs has only been moderately effective on any tested cell lines other than HEK293TT in our hands. Therefore, the most pressing question was whether transduction in vivo would be observable. As a simple way to assess gene transfer by non-human papilloma PsVs and subsequent protein expression in vivo, the inventors chose firefly luciferase (F.Luc) as reporter. MfPV11 and PcPV1 PsVs carrying an F.Luc reporter plasmid were produced without purification by ultracentrifugation, 2 μl were used to transduce HEK293TT cells and F.Luc assay was performed 72 h after transduction (FIG. 6, A). PsV preparations were then diluted such that all mice would receive the same amount of transducing units as measured by F.Luc assay in cell culture. 50 μl of PsV suspension containing theoretical 2.5×105 RLU were injected intramuscularly into the left hind leg of female Balb/c mice. Approx. 3 h after application, mice were subjected to bioluminescent imaging in order to check for any free F.Luc that may have been present in the PsV preparation (FIG. 6, B, “Day 0”). Mice were then monitored in a weekly manner by bioluminescent imaging (FIG. 6, B). Mice who had received PcPV1 PsVs showed pronounced expression of F.Luc 7 days after application, while it took 28 days until a weak F.Luc signal was detectable in mice who had received MfPV11 PsVs. The F.Luc signal in the PcPV1 group remained detectable until at least 10 weeks after application, which is when the last bioluminescent imaging was performed. Importantly, mice never showed any signs of adverse reaction or inflammation at the site of non-human papilloma PsV injection.


Example 3: Discussion

Gene-delivery based on viral vectors holds great potential to increase the applicability of genetic vaccines, which are not a competitive alternative to currently approved protein-based vaccines yet. In this study, the inventors show that it is worth exploring the wide range of non-human papilloma viruses for gene delivery, and identified PcPV1 PsVs as efficient delivery vector in vivo. Gene sequences for the capsid proteins L1 and L2 of more than one hundred animal papilloma viruses are available and can be used quite easily to produce pseudovirions, as exemplified for PcPV1 PsVs, in the present application. Since transduction in vitro and in vivo has been shown for PsVs of several human papilloma types belonging to the α-papillomaviruses, the inventors speculated that this genus might provide especially suitable candidates for gene delivery. Interestingly, this did not prove to be true. Of all tested PsVs of non-human α-papillomaviruses (CgPV1, PtPV1, MfPV6, MfPV11, MmPV1) only MfPV11 yielded good transduction rates in vitro. Upon intramuscular injection, however, MfPV11 PsVs showed only very week transduction of the reporter plasmid and subsequent expression in vivo. As the preferred site of entry of different papillomavirus genera can be either cutaneous or mucosal (E.-M. de Villiers, C. Fauquet, T. R. Broker, H.-U. Bernard, H. Zur Hausen, Classification of papillomaviruses, Virology. 324, 17-27 (2004), doi:10.1016/j.virol.2004.03.033), it might be necessary to take this into account when choosing the route of administration. Most HPV types belong to the genus of α-papillomaviruses and therefore infect the mucosa. HPV PsVs have indeed been successfully used for genital transmission in mice (C. Cerqueira et al., Efficient Production of Papillomavirus Gene Delivery Vectors in Defined In Vitro Reactions, Molecular therapy. Methods & clinical development. 5, 165-179 (2017), doi:10.1016/j.omtm.2017.04.005). Although this approach shows promise for a mucosal application of papilloma PsVs as gene vectors, intense pretreatment of the mucosa was necessary, as the intact mouse genital epithelium was found to be quite resistant to infection with HPV16 PsVs (B. S. Graham et al., Mucosal delivery of human papillomavirus pseudovirus-encapsidated plasmids improves the potency of DNA vaccination, Mucosal immunology. 3, 475-486 (2010), doi:10.1038/mi.2010.31). Intramuscular injection uses the well established and commonly accepted route for vaccine administration. The inventors show here that this form of application could be a simple alternative for the administration of papilloma PsVs for gene transfer.


Furthermore, it would be interesting to analyze the use of ι-carrageenan as additional transduction enhancer in vivo. ι-carrageenan has previously been shown to be a potent inhibitor of human papillomavirus infection by preventing the virions from binding to cells (C. B. Buck et al., Carrageenan is a potent inhibitor of papillomavirus infection, PLoS pathogens. 2, e69 (2006), doi:10.1371/journal.ppat.0020069). To the inventors' best knowledge, it has not been reported before that for certain non-human papilloma virus types the effect of ι-carrageenan can be just the opposite, leading to a significantly increased transduction in vitro. Whether this is also the case in vivo remains to be elucidated. Studies analyzing the attachment mechanisms of HPV have shown that on epithelial cells HPV uses heparan sulfate proteoglycans as primary attachment factors. This finding is supported by the fact that infection with HPV can be blocked by heparin or other sulfated polymers like carrageenan. Little is known about a potentially different entry pathway employed by animal papillomaviruses. The mechanism behind t-carrageenan mediated enhancement of transduction that the inventors observe for certain papillomavirus types is therefore unknown at this point. As many non-human papillomaviruses enter the body via the skin it might be worth exploring the cutaneous route for PsV application. In the context of genetic vaccine application, the skin is an attractive target due to the presence of large amounts of Langerhans and dendritic cells.


In conclusion, the inventors show that PcPV1 PsVs—and potentially many more non-human papilloma PsVs—effectively transduce a reporter plasmid in vivo after application into the muscle, leading to a several weeks long expression of a reporter plasmid. The vast amount of known and sequenced non-human papillomaviruses provide great potential to be explored further for the application as gene vectors.

Claims
  • 1. A non-human papilloma pseudovirus or virus-like particle comprising at least one codon-optimized nucleic acid sequence encoding a papilloma capsid protein for expression in eukaryotic cells or cell lines, wherein the codon-optimized nucleic acid sequence is as shown in SEQ ID NOS. 1 to 20.
  • 2. The non-human papilloma pseudovirus or virus-like particle of claim 1, wherein the papilloma capsid protein is L1 and/or L2.
  • 3. The non-human papilloma pseudovirus or virus-like particle of claim 1, wherein the papilloma capsid protein is from Caretta caretta papillomavirus 1, Colobus guereza papillomavirus 1, Common chimpanzee papillomavirus 1, Crocuta crocuta papillomavirus 1, Macaca fascicularis papillomavirus type 11, isolate Mac1637, Macaca fascicularis papillomavirus type 6, isolate Mac39, Procyon lotor papillomavirus 1, Puma concolor papillomavirus 1, Rhesus papillomavirus type 1b isolate Mac170 or Rousettus aegyptiacus papillomavirus type 1.
  • 4. The non-human papilloma pseudovirus or virus-like particle of claim 1, wherein the non-human papilloma virus-like particle is for gene delivery in vitro or in vivo.
  • 5. The non-human papilloma pseudovirus or virus-like particle of claim 1, wherein the non-human papilloma virus-like particle further comprises a targeting peptide.
  • 6. The non-human papilloma pseudovirus or virus-like particle of claim 5, wherein the targeting peptide is capable of directing the non-human papilloma virus-like particle to liver cells, lung cells, heart cells, kidney cells, blood cells, brain cells, gut cells, stem cells, cells of the mucosa of the throat or the nose, or cancer cells.
Priority Claims (1)
Number Date Country Kind
10 2017 220 276.9 Nov 2017 DE national
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2018/081102 11/13/2018 WO
Publishing Document Publishing Date Country Kind
WO2019/096796 5/23/2019 WO A
US Referenced Citations (4)
Number Name Date Kind
6416945 McCarthy et al. Jul 2002 B1
6991795 Lowe et al. Jan 2006 B1
7205126 Qiao et al. Apr 2007 B2
8326547 Liu et al. Dec 2012 B2
Foreign Referenced Citations (7)
Number Date Country
2017-528137 Sep 2017 JP
WO 2011039646 Apr 2011 WO
WO-2011039646 Apr 2011 WO
WO 2016038625 Mar 2016 WO
WO 2017075399 May 2017 WO
WO 2018152505 Aug 2018 WO
WO 2019096796 May 2019 WO
Non-Patent Literature Citations (41)
Entry
Pastrana et al. Cross-neutralization of cutaneous and mucosal Popillomavirus types with anti-sera to the amino terminus of L2. (2005) Virology; vol. 337; pp. 365-372 (Year: 2005).
Buck, Christopher B., et al. “Efficient intracellular assembly of papillomaviral vectors.” Journal of virology 78.2: 751-757. (Year: 2004).
Richardson, Sarah M., et al. “Gene Design: rapid, automated design of multikilobase synthetic genes.” Genome research 16.4: 550-556. (Year: 2006).
Rector, Annabel, et al. “Ancient papillomavirus-host co-speciation in Felidae.” Genome biology 8.4: 1-12. (Year: 2007).
Fujita et al., “Applications of Engineered DNA-Binding Molecules Such as TAL Proteins and the CRISPR/Cas System in Biology Research”, International Journal of Molecular Sciences, (2015); 16: 23143-23164.
Bayer et al., “Non-human Papillomaviruses for Gene Delivery in Vitro and in Vivo”, PLOS One, (2018); 13(6): e0198996.
Buck et al., “Generation of HPV Pseudovirions Using Transfection and Their Use in Neutralization Assays”, Methods in Molecular Medicine, (2005); 119: 445-462.
Cerqueira et al., “Efficient Production of Papillomavirus Gene Delivery Vectors in Defined In Vitro Reactions”, Molecular Therapy: Methods & clinical development, (2017); 5: 165-179.
Gaj et al., “ZFN,TALEN and CRISPR/Cas-Based Methods for Genome Engineering”, Trends in Biotechnology, (2013); 31(7): 397-405.
Grunwald et al., “Improvement of DNA Vaccination by Adjuvants and Sophisticated Delivery Devices: Vaccine-Platforms for the Battle against Infections Diseases”, Clinical and Experimental Vaccine Research, (2015); 4: 1-10.
Grunwald et al., “Novel Vaccine Regimen Elicits Strong Airway Immune Responses and Control of Respiratory Syncytial Virus in Nonhuman primates”, Journal of Virology, (2014); 88(8): 3997-4007.
Haas et al., “Codon Usage Limitation in the Expression of HIV-1 Envelope Glycoprotein”, Current Biology, (1996); 6(3): 315-324.
Harms et al., “Mouse Genome Editing using CRISPR/ Cas System”, Current Protocols in Human Genetics, (2014); 83: 15.7.1-15.7.27.
Herbst et al., “Genomic Characterization of Two Novel Reptilian Papillomaviruses, Chelonia Mydas Papillomavirus 1 and Caretta Caretta Papillomavirus 1”, Virology, (2009); 383: 131-135.
Kines et al., “Human Papillomavirus Capsids Preferentially Bind and Infect Tumor Cells”, International Journal of Cancer, (2016); 138: 901-911.
Kohlmann et al., “Protective Efficacy and Immunogenicity of an Adenoviral Vector Vaccine Encoding the Codon-Optimized F Protein of Respiratory Syncytial Virus”, Journal of Virology, (2009); 83: 12601-12610.
Morton et al., “Structural Characterization of Respiratory Syncytial Virus Fusion Inhibitor Escape Mutants: Homology Model of the F Protein and a Syncytium Formation Assay”, Virology, (2003); 311: 275-288.
Pastrana et al., “Cross-Neutralization of Cutaneous and Mucosal Papillomavirus Types with Anti-Sera to the Amino Terminus of L2”, Virology, (2005); 337: 365-372.
Rao et al., “Expression of Codon Optimized Major Capsid Protein (L1) of Human Papillomavirus Type 16 and 18 in Pichia Pastoris; Purification and Characterization of the Virus-like Particles”, Vaccine, (2011); 29 (43): 7326-7334.
Rector et al., “Animal Papillomaviruses”, Virology, (2013); 445, 213-223.
Ternette et al., “Immunogenicity and Efficacy of Codon Optimized DNA Vaccines Encoding the F-Protein of Respiratory Syncytial Virus”, Vaccine, (2007); 25: 7271-7279.
Ternette et al., “Expression of RNA Virus Proteins by RNA Polymerase II Dependent Expression Plasmids is Hindered at Multiple Steps”, Virology Journal, (2007); 4(51): 10.1186.
Vinzon et al., “Protective Vaccination against Papillomavirus-Induced Skin Tumors under Immunocompetent and Immunosuppressive Conditions: A Preclinical Study Using a Natural Outbred Animal Model”, PLOS Pathogens, (2014); 10(2): e1003924.
Wagner et al., “Rev-Independent Expression of Synthetic Gag-Pol Genes of Human Immunodeficiency Virus Type 1 and Simian Immunodeficiency Virus: Implications for the Safety of Lentiviral Vectors”, Human Gene Therapy, (2000); 11: 2403-2413.
Zhao et al., “Disassembly and Reassembly of Human Papillomavirus Virus-Like Particles Produces More Virion-Like Antibody Reactivity”, Virology Journal, (2012); 9(52); 1-13.
International Search Report; PCT/EP2018/081102, filed on Nov. 13, 2018, dated Feb. 25, 2019.
Bayer, L., et al., Non-Human Papillomaviruses for Gene Delivery In Vitro and In Vivo, PLOS One, vol. 13, No. 6, e0198996, 2018.
International Preliminary Report on Patentability, dated May 19, 2020, International Application No. PCT/EP2018/081102.
International Search Report & Written Opinion, dated Feb. 25, 2019, in International Application No. PCT/EP2018/081102.
Kines, R.C., et al., Human Papillomavirus Capsids Preferentially Bind and Infect Tumor Cells, International Journal of Cancer, vol. 138, No. 4, pp. 901-911, 2016.
Pastrana, D.V., et al., Cross-neutralization of cutaneous and mucosal Papillomavirus types with anti-sera to the amino terminus of L2, Virology, vol. 337, pp. 365-372, 2005.
Vinzon, S.E., et al., Protective Vaccination Against Papillomavirus-Induced Skin Tumors Under Immunocompetent and Immunosuppressive Conditions: A Preclinical Study Using A Natural Outbred Animal Model, PLoS Pathogens, vol. 10, No. 2, e1003924, 13 pages, 2014.
Buck et al., Efficient Intracellular Assembly of Papillomaviral Vectors, Journal of Virology, vol. 78, No. 2, pp. 751-757, 2004.
Buck et al., Carrageenan Is a Potent Inhibitor of Papillomavirus Infection, PLoS Pathogens, vol. 2, Issue 7, e69, pp. 0671-0680, 2006.
Graham et al., Mucosal delivery of human papillomavirus pseudovirus-encapsidated plasmids improves the potency of DNA vaccination, Mucosal immunology, vol. 3, No. 5, pp. 475-486, 2010.
Horvath et al., CRISPR/Cas, the immune system of bacteria and archaea, Science, vol. 327, pp. 167-170, 2010.
Kines et al., Vaccination with Human Papillomavirus Pseudovirus-Encapsidated Plasmids Targeted to Skin Using Microneedles, PLoS One, pp. 1-18, 2015, e0120797.
Lathe, R., Synthetic Oligonucleotide Probes Deduced from Amino Acid Sequence Data, J. Mol. Biol., vol. 183, pp. 1-12, 1985.
Office Action dated Sep. 6, 2022 in JP Application No. 2020-544150 in 9 pages.
Final Rejection dated Apr. 4, 2023 in Japanese Application No. JP 2020-544150.
Rector et al., Animal papillomaviruses, Virology, vol. 445, Nos. 1-2, pp. 213-223, 2013.
Related Publications (1)
Number Date Country
20200385718 A1 Dec 2020 US