WO2013009717A1 - Virion derived protein nanoparticles for delivering diagnostic or therapeutic agents for the treatment of skin-related diseases - Google Patents

Virion derived protein nanoparticles for delivering diagnostic or therapeutic agents for the treatment of skin-related diseases Download PDF

Info

Publication number
WO2013009717A1
WO2013009717A1 PCT/US2012/045989 US2012045989W WO2013009717A1 WO 2013009717 A1 WO2013009717 A1 WO 2013009717A1 US 2012045989 W US2012045989 W US 2012045989W WO 2013009717 A1 WO2013009717 A1 WO 2013009717A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
composition
therapeutic agent
proteins
hpv
Prior art date
Application number
PCT/US2012/045989
Other languages
French (fr)
Inventor
Elisabet De Los Pinos
Original Assignee
Elisabet De Los Pinos
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Elisabet De Los Pinos filed Critical Elisabet De Los Pinos
Publication of WO2013009717A1 publication Critical patent/WO2013009717A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16641Use of virus, viral particle or viral elements as a vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20041Use of virus, viral particle or viral elements as a vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20041Use of virus, viral particle or viral elements as a vector
    • C12N2710/20042Use of virus, viral particle or viral elements as a vector virus or viral particle as vehicle, e.g. encapsulating small organic molecule

Definitions

  • Sequence Listing provides exemplary polynucleotide sequences of the invention. The traits associated with the used of the sequences are included in the Examples.
  • the invention relates to methods for loading protein nanoparticles with therapeutic, diagnostic or other agents, wherein the protein nanoparticles are based on viral proteins. More particularly, the present invention relates to a method for using papilloma-derived protein nanoparticles to deliver drugs to the keratinocytes and basal membrane cells for the treatment of skin-related diseases such as alopecia or patterned hair loss, non-melanoma skin cancer, psoriasis, and treatment of skin related genetic diseases (e.g. Pachyonychia Congenita and Xeroderma pigmentosum).
  • skin-related diseases such as alopecia or patterned hair loss, non-melanoma skin cancer, psoriasis
  • skin related genetic diseases e.g. Pachyonychia Congenita and Xeroderma pigmentosum.
  • the first three systems reviewed are the phagemid particles and Herpes Simplex Virus- 1 (HSV) amplicons, in which the RNAi is derived from DNA, and the SV40 in vitro- packaged vectors. These three systems are examples of "trans-packaging systems in which viral machinery and proteins have been repurposed to package genetic material other than the original viral genome" (Lund et al. 401).
  • HSV Herpes Simplex Virus- 1
  • influenza Virosome and the HVJ-E a derivative of the Hemagglutinating Virus of Japan
  • viral envelope systems in which native enveloped virus particles are inactivated and used to package other cargos
  • the object of the present invention is to overcome the shortcomings disclosed in the prior art.
  • the products of the current invention are pseudo-viruses derived from
  • papillomaviruses in particular from the genus betapapillomaviridae to deliver drugs to the keratinocytes and basal membrane cells for the treatment of skin related diseases.
  • the present in vention wil l allow for impro ved methods for administering treatment of skin diseases capable of targeting epithelial cells.
  • pseudovirions and in particular type 5 betaHPV viral shells as an ideal delivery vehicle for the delivery of drugs to the skin for the treatment of skin related diseases such as alopecia or patterned hair loss, non-melanoma skin cancer, psoriasis, and treatment of skin related genetic diseases (e.g. Pachyonychia Congenita and Xeroderma pigmentosum).
  • aspects of the invention relate to methods and compositions for producing papilloma-derived (e.g., human papilloma virus (HPV)-derived) protein nanoparticles containing one or more therapeutic or diagnostic agents.
  • aspects of the invention relate to methods and compositions for encapsulating an agent within a virus like particle (VLP) requiring an initial isolation of capsid proteins produced in a host cell system (e.g. yeast, mammalian cell, insect cell, E.coli) and subsequent reassembly in vitro.
  • a host cell system e.g. yeast, mammalian cell, insect cell, E.coli
  • FIG. 1 depicts shuttle vector information.
  • FIG. 3 shows results from in vitro reassembly of capsid protein produced in insect cell culture. DLS demonstrates presence of capsid protein in form of monomers and oligomers after harvest from nuclear fraction (left) and appearance of well formed loaded VLPs after the reassembly procedure (right).
  • FIG. 4 is a graph showing the amount of luminescence/luciferase signal measured 48 hrs after treatment of HeLa cells with loaded VLP, where luminescence is reported on a scale of 0 to 30,000 units along the y-axis.
  • FIG. 5 is a graph the same data in FIG.4, showing the amount of
  • luminescence/luciferase signal measured 48 hrs after treatment of HeLa cells with loaded VLP, where luminescence is reported on a scale of 0 to 20 units along the y-axis.
  • FIG. 6 shows a fl ow chart diagram of a preferred embodiment of the present invention for the treatment of alopecia.
  • FIG. 7 shows a flow chart diagram of a preferred embodiment of the present in vention for the treatment of non-melanoma skin cancer.
  • FIG. 8 shows a flo w chart diagram of a preferred embodiment of the present invention for the treatment of skin related genetic diseases.
  • FIG. 9 shows a flow chart diagram of a preferred embodiment of the present invention for the treatment of psoriasis.
  • SEQ ID NO: 1 shows DNA sequence for baculovirus L! X plasmid encoding HP V 16/31 L 1 (pFastBacTM).
  • SEQ ID NO: 2 shows DNA sequence for baculovirus L2 plasmid encoding
  • HPV16L2 (pFastBacTM).
  • SEQ ID NO: 3 shows forward primer DNA sequence used for generation of shE7- 1 RNA construct.
  • SEQ ID NO: 4 shows reverse primer DNA sequence used for generation of shE7- 1 RNA construct.
  • SEQ ID NO: 5 shows plasmid pl 6Ll *L2 DNA sequence encoding 16/31 L I (LI *) and L.2 human codon -optimized,
  • [0041 , 1 ] shows a sequence for the complete genome for Human
  • SEQ ID NO: 8 shows a sequence for P5 SHELL circular plasmid DS-DNA. [0037], DETAILED DESCRIPTION OF THE INVENTION [0038], Assembly Of Particles
  • the components can be associated with the nanoparticles through a linkage.
  • used as a carrier associated with it is meant that the component is carried by the nanoparticles.
  • the component can be dissolved and incorporated in the nanoparticles non-covalently.
  • aspects of the invention relate to methods and compositions for producing protein nanoparticles that contain therapeutic and/or diagnostic agents for delivery to a subject.
  • Methods and compositions have been developed for effectively encapsulating therapeutic and/or diagnostic agents within papilloma virus proteins (e.g., HPV proteins) that can be used for delivery to a subject (e.g. , a human subject).
  • papilloma virus proteins e.g., HPV proteins
  • LI and L2 capsid proteins are useful to isolate LI and L2 capsid proteins directly from host cells as opposed to disassembling VLPs that were isolated from host cells.
  • LI and L2 capsid proteins that are isolated directly from cells can be used in in vitro assembly reactions to encapsulate a therapeutic or diagnostic agent. This avoids the additional steps of isolating and disassembling VLPs. This also results in a cleaner preparation of LI and L2 proteins, because there is a lower risk of contamination with host cell material (e.g., nucleic acid, antigens or other material) that can be contained in VLPs that are isolated from cells.
  • host cell material e.g., nucleic acid, antigens or other material
  • LI and L2 capsid proteins it is useful to independently produce LI and L2 capsid proteins.
  • they can be produced from two independent nucleic acids (e.g., different vectors).
  • they can be produced in the same cell (e.g., using two different vectors within the same cell).
  • they can be produced in different cells (e.g., different host cells of the same type or different types of host cell).
  • This approach allows the ratio of LI and L2 proteins to be varied for either in vitro or intracellular assembly.
  • VLPs to be assembled (e.g., in vitro or intracellularly) with higher or lower LI to L2 ratios than in a wild type VLP.
  • H PV nanoparticles as deliver ⁇ ' vehicles for therapeutic agents.
  • a higher ratio of L,2 in the assembled structure may allow the resultant VLP to have a higher nucleic acid binding affinity and a better efficiency in delivering these intracellularly.
  • LI and L2 proteins are expressed in a host cell system (e.g., both in the same host cell or independently in different host cells). LI and/or L2 are isolated from nuclei of the host cells. In some embodiments, certain LI and/or L2 structures that are formed during cellular growth (e.g., during the fermentation process) are disrupted. Any suitable method may be used. In some embodiments, sonication may be used (e.g., nuclei may be isolated and then sonicated). Capsid proteins then may be purified using any suitable process. For example, in some embodiments, capsid proteins may be purified using chromatography.
  • Isolated capsid proteins can then be used as described herein in a cell free system to assemble together with different payloads to create superstructures that contain a drug or diagnostic agent in its interior.
  • directly isolating capsid proteins provides several benefits, In some embodiments, there is a reduced risk of encapsulating and transferring genetic information (DNA, RNA) from the host cell to the treated subject.
  • de-novo assembly of VL Ps during the assembly procedure ensures formation of a larger percentage of loaded VLPs as opposed to using already-formed VLPs for loading where a certain fraction can remain unloaded.
  • one or more therapeutic or diagnostic agents may be loaded intracellularly by expressing LI and/or L2 in the presence of intracellular levels of one or more agents of interest,
  • this method is used for encapsulating a silencing plasmid which will encode for expression of short hairpin RN A (shRNA),
  • this plasmid will have a size of 2kB-6kB.
  • any suitable size may be used.
  • a plasmid is designed to be functional within the cells of the patient or subject to be treated (to which the loaded V LP is administered). Accordingly, the plasmid will be active within the target cells resulting in knockdown of the targeted gene(s).
  • this method may be used to encapsulate short interfering RNA (siRNA) or antisense nucleic acids (DN A or RNA) transfected into the host cells (e.g., 293 cel ls or other mammalian or insect host cells) during the production of the V LPs.
  • siRNA short interfering RNA
  • DN A or RNA antisense nucleic acids
  • loaded VLPs may be produced intracellularly to provide gene silencing functions when delivered to a subject.
  • RNA interference RNA interference
  • LI and L2 proteins are expressed in a host ceil system (e.g. mammalian cells or insect cells) from independent expression nucleic acids (e.g., vectors, for example, plasmids) as opposed to both being expressed from the same nucleic acid.
  • independent expression nucleic acids e.g., vectors, for example, plasmids
  • LI and L2 proteins are expressed in a host ceil system (e.g. mammalian cells or insect cells) from independent expression nucleic acids (e.g., vectors, for example, plasmids) as opposed to both being expressed from the same nucleic acid.
  • VLP structures can be produced to fit the needs of the different classes of payloads (e.g., DNA, R A, small molecule, large molecule) both in terms of charge and other functions (e.g. DNA binding domains, VLP inner volume, endosomal release function).
  • VLPs with a higher content of L2 protein will be better to bind nucleic acids (L2 contains a DNA binding domain) whereas VLPs with a smaller content of L2 protein will be better for other small molecules.
  • VLPs with different ratios of LI :L2 protein will have different inner volumes that w r ill allow a higher concentration of drug to be encapsulated.
  • the release of pay load into the cell will also be modulated.
  • structures containing more L2 protein may have a higher ability to transfer nucleic acids intracellularly. It should be appreciated that different ratios of L1/L2 may be used, in some embodiments, ratios may be 1 :1 , 1 :2, 1 :4, 1:5, 1 :20 or 1 :100. However, other ratios may be used as aspects of the invention are not limited in this respect.
  • each separate expression nucleic acid encodes an LI (but not an L,2) or an L2 (but not an LI ) sequence operably linked to a promoter.
  • the separate expression nucleic acids may use the same or different promoters and/or other regulatory sequences and/or replication origins, and/or selectable markers.
  • the separate nucleic acids may be vectors (e.g., plasmids, or other independently replicating nucleic acids).
  • separate nucleic acids may be independently integrated into the genome of a host cell (e.g., a first nucleic acid integrated and a second nucleic acid on a vector, two different nucleic acids integrated at different positions, etc.).
  • the relative expression levels of LI and L2 may be different in different cells, different using different expression sequences, independently regulated, or a combination thereof.
  • an expression vector is used to produce a mutant LI or L2 protein.
  • a mutant HPV16L1 protein (called LI *) is expressed along with L2 in a host system (e.g., a 293 cell system). These can then be isolated and assembled as described herein to encapsulate a therapeutic or diagnostic payload (e.g. therapeutic plasmid, siRNA, small molecule drugs, etc.).
  • a therapeutic or diagnostic payload e.g. therapeutic plasmid, siRNA, small molecule drugs, etc.
  • loaded VLPs are produced using certain LI and/or L2 variant sequences that are not recognized by existing antibodies against HPV (e.g.,
  • loaded VLPs can be produced using LI and/or L2 proteins that are modified to reduce antigenicity against other HPV serotype antibodies and/or to target the loaded VLP to particular organs or tissues (e.g., lung) or cells or subcellular locations.
  • the papilloma virus particles are HPV-VLP.
  • the methods described herein utilize HPV- VLP that contain one or more naturally occurring HPV capsid proteins (e.g., LI and/or L2 capsid proteins).
  • HPV-VLP may be comprised of capsid protein oligomers or monomers.
  • VLP refers to the capsid-like structures which result upon assembly of a HPV L I capsid protein alone or in combination with a HPV L2 capsid protein.
  • VLPs are morphologically and antigenically similar to authentic virions. VLPs lack viral genetic material (e.g., viral nuclei acid), rendering the VLP non-infectious.
  • VLP may be produced in vivo, in suitable host cells, e.g., mammalian, yeast, bacterial and insect host cells.
  • a “capsomere” refers to an oligomeric configuration of LI capsid protein.
  • Capsomeres may comprise at least one L I (e.g., a pentamer of LI).
  • capsid protein refers to LI or L2 proteins that are involved in building the viral capsid structure. Capsid proteins can form oligomeric structures i.e. pentamers, trimers or be in single units as monomers.
  • a VLP can be loaded with one or more medical, diagnostic and/or therapeutic agents, or a combination of two or more thereof.
  • the methods described herein utilize HPV-VLP that contain one or more variant capsid proteins (e.g., variant LI and/or L2 capsid proteins) that have reduced or modified immunogenicity in a subject. Examples of varian t capsid proteins are described in WO 2010/120266.
  • the modification may be an amino acid sequence change that reduces or avoids neutralization by the immune system of the subject.
  • a modified HPV-VLP contains a recombinant HPV protein (e.g., a recombinant L I and/or L2 protein) that includes one or more amino acid changes that alter the immunogenicity of the protein in a subject ( . e.g., in a huma subject).
  • a modified HPV-VLP has an altered immunogenicity but retains the ability to package and deliver molecules to a subject.
  • amino acids of the viral wild-type capsid proteins such as LI and/or LI +1,2, assembling into the HPV-VLP, are mutated and/or substituted and/or deleted.
  • these amino acids are modified to enhance the positive charge of the VLP interior.
  • modifications are introduced to allow a stronger electrostatic interaction of nucleic acid molecules with one or more of the amino acids facing the interior of the VLP and/or to avoid leakage of nucleic acid molecules out of the VLP. Examples of modifications are described in WO 2010/120266. It should be appreciated that any modified HPV-VLP may be loaded with one or more agents. Such particles may be delivered to a subject without inducing an immune response that would be induced by a naturally-occurring HPV,
  • HPV- VLP loaded according to the methods described herein, are useful for delivering one or more therapeutic agents to diseased tissue (e.g., diseased mucosal tissue).
  • a diseased tissue e.g. , mucosal tissue, epithelial tissue, or endothelial tissue
  • a infected tissue e.g., infected with a virus such as HP V or HSV.
  • the mucosal tissue is cervical tissue and the disease is dysplasia or cancer (e.g., cervical dysplasia, cervical cancer, for example associated with persistent HPV infection).
  • HPV-VLP loaded according to the methods described herein, may be used to deliver compositions to other tissues (e.g. , epidermis), In some embodiments, HPV-VLP may be used to treat HPV related diseases, In some embodiments, HPV-VLP, loaded according to the methods described herein, may be used to deliver therapeutic agents to treat a disease or condition, and/or may be used to deliver diagnostic agents to diagnose other diseases or conditions. For example, quantum dots, metals, and/or other imaging agents may be loaded into VLP according to the methods described herein and delivered. In some embodiments, agents may be used to track early stage diseases (e.g. , early stage metastasis).
  • early stage diseases e.g. , early stage metastasis
  • any suitable therapeutic, diagnostic and/or other medical agent may be loaded into VLP according to the methods described herein and delivered to a subject.
  • Examples of administration of VLP to subjects are described for example in U.S. Patent No. 7,205,126, incorporated herein by reference.
  • HPV-VLPs that are modified to display epitopes from two or more different naturally-occurring HPV variants are used in the agent loading methods described herein. Such modified VLPs may be used to provide immunization against infection by any of two or more naturally occurring HPV variants.
  • HPV-VLPs comprise viral LI capsid proteins.
  • HPV-VLPs comprise viral LI capsid proteins and viral L2 capsid proteins.
  • the LI and/or L2 proteins may, in some embodiments, be wild-type viral proteins.
  • L i and/or L2 capsid proteins may be altered by mutation and/or deletion and/or insertion so that the resulting LI and/or L2 proteins comprise only 'minimal' domains essential for assembly of a VLP.
  • LI and/or L2 proteins may also be fused to other proteins and/or peptides that provide additional functionality. Examples of modifications are described for example in U.S. Patent No.
  • VLPs may be based on particles containing one or more recombinant proteins or fragments thereof (e.g. , one or more HPV membrane and/or surface proteins or fragments thereof). In some embodiments, VLPs may be based on naturally-occurring particles that are processed to incorporate one or more agents as described herein, as aspects of the invention are not limited in this respect, In certain embodiments, particles comprising one or more targeting peptides may be used. Other combinations of HPV proteins (e.g., capsid proteins) or peptides may be used as aspects of the invention are not limited in this respect.
  • viral wild-type capsid proteins are altered by mutations, insertions and deletions. All conformation-dependent type-specific epitopes identified to date are found on the HPV-VLP surface within hyper-variable loops where the amino acid sequence is highly divergent between H PV types, which are designated BC, DE, EF, FG and HI loops. Most neutralizing antibodies are generated against epitopes in these variable loops and are type-specific, with limited cross-reactivity, cross-neutralization and cross-protection. Different HPV serotypes induce antibodies directed to different type-specific epitopes and/or to different loops. Examples of variant capsid proteins are described in WO 2010/120266.
  • viral capsid proteins HPV L I and/or L2 are mutated at one or more amino acid positions located in one or more hyper-variable and/or surface- exposed loops.
  • the mutations are made at amino acid positions within the loops that are not conserved between HPV serotypes. These positions can be completely non-conserved, that is that any amino acid can be at this position, or the position can be conserved in that only conservative amino acid changes can be made.
  • LI protein and L1 ⁇ L2 protein may be produced
  • reeombraantly produced LI protein and L1+L2 protein may self-assemble to form vims-like particles (VLP).
  • VLP vims-like particles
  • Recombinant production may occur in a bacterial, insect, yeast or mammalian host system.
  • LI protein may be expressed or L1 ⁇ L2 protein may be co-expressed in the host system.
  • HPV LI and/or L2 proteins may be expressed in Spodoptera frugiperla (8f21) cells.
  • Baculoviruses encoding the LI and/or L2 gene of any HPV or recombinant versions thereof from different serotypes e.g., HPV 16, HPV18, HPV31, and HPV58
  • HPV LI and/or L,2 genes may be cloned into a plasmid, such as pFastBacl (Invitrogen).
  • Sf21 cells may be maintained in Grace's insect medium (Invitrogen) supplemented with 10% fetal calf serum (PCS, invitrogen) and infected with recombinant baculoviruses and incubated at 27°C. Three days post infection, cells can be harvested and VLP can be purified.
  • cells may be resuspended in PBS containing Nonidet P40 (0.5%), pepstatin A, and leupeptin (1 ⁇ each, Sigma Aldrich), and allowed to stand for 30 min at 4°C.
  • Nuclear lysates may then be centrifuged and pellets can be resuspended in ice cold PBS containing pepstatin A and leupeptin and then sonicated. Samples may then be loaded on a CsCl gradient and centrifuged to equilibrium (e.g., 22 h, 27,000 rpm in a SW28 rotor, 4°C).
  • CsCl gradient fractions may be investigated for density by refractonietry and for the presence of L1/L2 protein by electrophoresis in 10% sodium dodecyl sulfate-polyacryiamide gel (SDS-PAGE) and Coomassie blue staining. Positive fractions can be pooled, diluted in PBS and pelleted e.g., in a Beckman SW 28 rotor (3 h, 28,000 rpm, 4°C), After centrifugation, VLP can be resuspended in 0.15 mol/ ' L NaCl and sonicated, e.g., by one 5 second burst at 60% maximum power. Total protein content may be determined.
  • Viral capsid proteins may also be expressed using galactose-inducible Saccharomyces cerevisiae expression system, Leucine-free selective culture medium used for the propagation of yeast cultures, yeast can be induced with medium containing glucose and galactose. Cells can be harvested using filtration, After resuspension, cells may be treated with Benzonase and subsequently mechanically disrupted (e.g., using a homogenizer). Ceil lysate may be clarified using filtration.
  • An exemplary protocol can be found in Cook et al. Protein Expression and Purification 17, 477-484 (1999).
  • Transiently transfected cells e.g., 293 cells, for example 293T or 293TT cells
  • 293 cells for example 293T or 293TT cells
  • Benzonase and exonuclease V can be lysed by adding Brij58 or similar nonionic polyoxyethylene surfactant detergent, followed by benzonase and exonuclease V and incubating at 37°C for 24 h to remove unpackaged cellular and viral DNA and to allow capsid maturation.
  • the lysate can be incubated on ice with 5 M NaCl and cleared by centrifugation.
  • VLP can be collected by high-speed centrifugation.
  • Capsid proteins may also be expressed in E. coli.
  • E.coli one important potential contaminant of protein solutions is endotoxin, a lipopolysaccharide (LPS) that is a major component of the outer membrane of Gram-negative bacteria (Schadlich et /. Vaccine 27, 1 11-1522 (2009)).
  • LPS lipopolysaccharide
  • transformed BL21 bacteria may be grown in LB medium containing ImM ampiciliin and incubated with shaking at 200 rpm at 37 ' C. At an optical density (ODgoo nm) of 0.3-0.5, bacteria can be cooled down and IPTG may be added to induce protein expression. After 16-18 h bacteria may be harvested by centrifugation.
  • VLPs are loaded with the one or more therapeutic agents. After isolation of LI and L2 capsid proteins which may be in the form of monomers or oligomers, VLPs may be assembled and loaded by disassembling and reassembling LI or LI and L2 viral capsid proteins, as described herein. Salts that are useful in aiding
  • VLPs include Zn, Cu and Ni, Ru and Fe salts.
  • VLPs may be loaded with one or more therapeutic agents.
  • VLPs Loading of VLPs with agents utilizing a disassembly-reassembly method has been described previously, for example in U.S. Patent Nos: 6,416,945 and WO 2010/120266, incorporated herein by reference. Generally, these methods involve incubation of the VLP in a buffer comprising EGTA and DTT, Under these conditions, VLP completely disaggregated into structures resembling capsid proteins in monomeric or oligomeric form, A therapeutic or diagnostic agent, as described herein, may then be added and the preparation diluted in a buffer containing DMSO and CaCl 2 with or without ZnCl 2 in order to reassemble the VLP.
  • ZnCl 2 increases the reassembly of capsid proteins into VLP.
  • these reassembly methods may be used to assemble capsid proteins to form VLPs that encapsulate one or more agents without requiring an initial VLP disassembly procedure, as described herein.
  • VLP are loaded with the one or more therapeutic agents.
  • these may mixed directly after purification from the host cell with the therapeutic agent and reassembled into loaded VLPs as described herein, the preparation diluted in a buffer containing DMSO and CaCl 2 with or without ZnCl 2 in order to reassemble the VLP.
  • the presence of ZnCl 2 increases the reassembly of capsid proteins into VLP.
  • VLP loaded with agents using the methods described herein are able to deliver agent to 65%, 75%, 85%, 95%, 96%», 97%, 98%, or 99% of target cells.
  • One non imiting example of the improved method is exemplified in the Examples.
  • VLP may be loaded with a nucleic acid using a method comprising: a) contacting a preparation of capsid proteins with the nucleic acid in a reaction volume, wherein i) the ratio of capsid protein to reaction volume ranges from 0.
  • the ratio of nucleic acid to capsid protein ranges from 0.1 ⁇ g nucleic acid per 1 p.g capsid protein to 10 p.g nucleic acid per 1 ⁇ g capsid protein; and/or iii) the ratio of nucleic acid to reaction volume ranges from 0.01 ⁇ g nucleic acid per 1 ⁇ reaction volume to 10 g nucleic acid per 1 ⁇ reaction volume, and b) reassembling the capsid proteins to form a VLP, thereby
  • the ratio of HPV- capsid protein to reaction volume ranges from 0.2 ⁇ g HPV-capsid protein per 1 ⁇ reaction volume to 0.6 ⁇ g HPV-capsid protein per 1 ⁇ reaction volume.
  • the ratio of nucleic acid to HP V-capsid protein ranges from 0.5 ⁇ g nucleic acid per 1 ug HPV-capsid protein to 3.5 ⁇ nucleic acid per 1 ⁇ 3 ⁇ 4 HPV-capsid protein.
  • the ratio of nucleic acid to reaction volume ranges from 0.2 ⁇ 3 ⁇ 4 nucleic acid per 1 ⁇ reaction volume to 3 ⁇ g nucleic acid per I ⁇ reaction volume.
  • the step of dissociating the VLP or capsid protein oligomers can be carried out in a solution comprising ethylene glycol tetraacetic acid (EGTA) and dithiothreitol (DTT), wherein the concentration of EGTA ranges from 0,3 mM to 30 mM and the concentration of DTT ranges from 2 mM to 200 mM. in certain embodiments, the concentration of EGTA ranges from 1 mM to 5 mM. In certain embodiments, the concentration of DTT ranges from 5 mM to 0 mM.
  • EGTA ethylene glycol tetraacetic acid
  • DTT dithiothreitol
  • the step of reassembling of capsid proteins into a VLP can be carried out in a solution comprising dimethyl sulfoxide (DMSO), ⁇ aCL and ZnC , wherein the
  • concentration of DMSO ranges from 0.03% to 3% volume/volume
  • concentration of CaCl 2 ranges from 0.2 mM to 20 mM
  • concentration of ZnCL ranges from 0.5 ⁇ , ⁇ to 50 ⁇ .
  • concentration of DMSO ranges from 0.1% to 1% volume/volume.
  • concentration of ZnCL ranges from 1 ⁇ , ⁇ to 20 ⁇ .
  • concentration of CaCl 2 ranges from 1 mM to 10 mM.
  • the loading method is further modified to stabilize the VLP, in thai the loading reaction is dialyzed against hypertonic NaCl solution (e.g., using a NaCl concentration of about 500 mM) instead of phosphate-buffered saline (PBS), as was previously described. Surprisingly, this reduces the tendency of the loaded VLP to form larger agglomerates and precipitate.
  • the concentration ofNaCl ranges between 5 mM and 5 M. In certain embodiments, the concentration of NaCl ranges between 20 mM and 1 M.
  • VLPs can be loaded with one or more therapeutic agents according to the methods described herein.
  • the therapeutic agent is a nucleic acid molecule capable of inducing RNA interference or a nucleic acid (e.g. , plasmid or other vector) that is capable of expressing a nucleic acid molecule capable of inducing RNA interference.
  • VLP can be loaded with combinations of two or more therapeutic or diagnostic agents,
  • the therapeutic agent is an inducer of RNA interference or other inducer of gene silencing.
  • An inducer of RNA interference may be a siRNA, a shRNA, a hybrid nucleic acid molecule comprising a first part that comprises a duplex ribonucleic acid (RNA) molecule and a second part that comprises a single stranded deoxyribonucleic acid (DNA) molecule, a longer double-stranded RNA or a DNA construct for expression of siRNA or longer RNA sequences.
  • RNA duplex ribonucleic acid
  • DNA deoxyribonucleic acid
  • Other inducers of gene silencing include inducers of DNA methylation, or rihozymes, or aptamers.
  • the therapeutic agent can be a modulator of gene expression such as a PNA (Peptide Nucleic Acid).
  • RNA interference is a process whereby the introduction of double stranded RNA (dsRNA) into a cell inhibits gene expression post-transcriptionally, in a sequence dependent fashion, RNAi can be mediated by short (for example 19-25 nucleotides) dsRNAs or small interfering R ' NAs (siRN A).
  • dsRNA is cleaved in the cell to create siRNAs that are incorporated into an RNA-induced silencing complex (RISC), guiding the complex to a homologous endogenous mRNA, cleaving the mRNA transcript, and resulting in the destruction of the mRNA.
  • RISC RNA-induced silencing complex
  • dsRNA may be introduced into the cell as an isolated nucleic acid fragment or via a transgene, plasmid, or virus.
  • VLP are used to deliver dsRNA to the target cells,
  • a short hairpin RNA molecule is expressed in the cell
  • a shRNA comprises short inverted repeats separated by a small loop sequence, One inverted repeat is complimentary to the gene target.
  • the shRN A is then processed into an siRNA which degrades the target gene mRNA.
  • siiRNAs can produced within a cell with a DNA construct encoding the shRNA sequence under control of a RN A polymerase III promoter, such as the human HI, U6 or 7SK promoter.
  • the shRNA may be synthesized exogenous! ⁇ ' and introduced directly into the cell, for example through VLP deliver ⁇ '.
  • the shRNA sequence is between 40 and 100 bases in length or between 40 and 70 bases in length.
  • the stem of the hairpin are, for example, between 19 and 30 base pairs in length.
  • the stem may contain G-U pairings to stabilize the hairpin striscture.
  • siRNA sequences are selected on the basis of their homology to the target gene. Homology between two nucleotide sequences may be determined using a variety of programs including the BLAST program (Altschul et al (1990) J. Mol. Biol. 215: 403-10), or BestFit (Genetics Computer Group, 575 Science Drive, Madison, Wisconsin, USA., Wisconsin 53711). Sequence comparisons may be made using FAST A and FASTP (see Pearson & Lipman, 1988. Methods in Enzymology 183: 63-98). Tools for design and quality of siRNAs, shRNAs and/or miRNAs are known in the art.
  • siRNA sequences and scrambled siRNA sequences are for example siDirect, siSearch, SEQ2SVM, Deqor, siRNA Wizard (invivoGen).
  • the specificity can be predicted using for example SpecificityServer, miRacle.
  • Target sequences can be researched for example at HuSiDa (Human siRNA Database), and siRNAdb (a database of siRNA sequences). Sequence comparison may be made over the full length of the relevant sequence, or may more preferably be over a contiguous sequence of about or 10,15, 20,25 or 30 bases.
  • the degree of homology between the siRNA and the target gene is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%, or 100% ⁇ .
  • the siRN A may be between lObp and 30bp in length, or between 20bp and 25bp, or the siRNA is 20, 21 or 22bp in length.
  • RNAi can be detected by transfecting cultured cells with the siRNA, followed by RT-PCR of the mRNA of interest. Where R NAi is induced by the siRNA, levels of the mRNA of interest will be reduced in traiisiected ceils as compared to control cells. A reduction in protein production can be confirmed by Western blotting of cell lysates followed by probing with an antibody reactive to the protein of interest,
  • siRNA molecules may be synthesized using standard solid or solution phase synthesis techniques which are known in the art. [0093], In some embodiments, the siRNA has an overhang at one or both ends of one or more deoxythymidme bases to increase the stability of the siRN A within cells by reducing its susceptibility to degradation by nucleases,
  • the siRNA is a hybrid nucleic acid molecule comprising a first part that comprises a duplex ribonucleic acid (RNA) molecule and a second part that comprises a single stranded deoxyribonucleic acid (DNA) molecule.
  • RNA duplex ribonucleic acid
  • DNA deoxyribonucleic acid
  • Preferred targets for the RNA interference would include disease causing genes e.g.
  • the target genes would be E6 & E7 HPV viral oncogenes (e.g., E7 siRNA), In other embodiments of the invention the target genes would be p53, Sirt-1, survivin, EGFR, VEGFR, VEGF, CTNNB1 or other oncogenes.
  • Linkages between nucleotides may be phosphodiester bonds or alternatives, for example, linking groups of the formula P (O) 8, (thioate) ; P (S) 8, (dithioate); P (O) NR'2 ; P (O) R' ; P (O) OR6; CO ; or CONR'2 wherein R is H (or a salt) or alkyl ( 1-12C) and R6 is alkyl (1-9C) is joined to adjacent nucleotides through-O-or-S-.
  • Modified nucleotide bases can be used in addition to the naturally occurring bases, For example, modified bases may increase the stability of the siRNA molecule, thereby reducing the amount required for silencing,
  • the term modified nucleotide base encompasses nucleotides with a covIERly modified base and/or sugar.
  • modified nucleotides include nucleotides having sugars which are covIERly attached to low molecular weight organic groups other than a hydroxy! group at the 3'position and other than a phosphate group at the 5 'position.
  • modified nucleotides may also include ⁇ substituted sugars such as 2'- 0-methyl- ; 2-0- alkyl : 2-0-allyl; 2'-S-alkyl; 2'-S-allyl ; 2'-fiuoro- ; 2'-halo or 2; azido-ribose, carbocyclic sugar analogues a-anomeric sugars; epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, and sedoheptulose.
  • ⁇ substituted sugars such as 2'- 0-methyl- ; 2-0- alkyl : 2-0-allyl; 2'-S-alkyl; 2'-S-allyl ; 2'-fiuoro- ; 2'-halo or 2; azido-ribose, carbocyclic sugar analogues a-anomeric sugars; epimeric sugar
  • Modified nucleotides include alkylated purines and pyrimidines, acylated purines and pyrimidines, and other heterocycles. These classes of pyrimidines and purines are known in the art and include pseudoisocytosine, N4, N4- ethanocytosine, 8-hydroxy-N6-methylademne, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5 fluorouracil, 5-bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, 5- carboxymethylammomethyl uracil, dihydrouracil, inosine, N6 ⁇ isopentyl-adenine, 1- methyladenine, l-methylpseudouracil, 1- methylguanine, 2,2-dimethylguanine, 2methylad nine, 2- methylguanine, 3-methylcytosine, 5-methylcytosine, N6-
  • siRNA molecules or longer dsRNA molecules may be made recombinantly by transcription of a nucleic acid sequence, for example contained within a vector as described herein.
  • the vector may be any RNA or DNA vector,
  • the vector can be an expression vector, wherein the nucleotide sequence is operably linked to a promoter compatible with the cell.
  • promoters suitable for use in various vertebrate systems are well known in the art.
  • suitable promoters include viral promoters such as mammalian retrovirus or DN A virus promoters, e.g., MLV, CMV, RSV, SV40 IEP (immediate early promoter) and adenovirus promoters and metallothionein promoter. Strong mammalian promoters may also be used. It will be appreciated that variants of such promoters retaining substantially similar transcriptional activities may also be used.
  • the vector may have at least two promoters, one to direct expression of the sense strand and one to direct expression of the antisense strand of the dsRNA. In other embodiments, two vectors may be used, one for the sense strand and one for the antisense strand. Alternatively the vector may encode RNAs which form stem-loop structures which are subsequently cleaved by the cell to produce dsRNA,
  • the nucleic acid construct may contain a specific cellular, viral or other promoter or repressor of gene expression.
  • the promoter or repressor may be designed to reflect the context of the cell into which the constract is introduced.
  • the construct may contain a viral promoter so expression from the construct is dependent upon the presence of a viral protein, so that the construct is expressed only in viral-infected cells.
  • the construct may have a promoter or repressor specific to certain cell types or to certain developmental stages.
  • a viral promoter whic matches the disease-causing vims should be used, e.g., a HPV promoter (such as the promoter causing expression of HPV E6/E7) for HPV- infected ceils.
  • a HPV promoter such as the promoter causing expression of HPV E6/E7 for HPV- infected ceils.
  • the vector will only be expressed in the virally- infected cells.
  • nucleic acids are highly charged and do not cross ceil membranes by free diffusion.
  • the hydrophilic character and anionic backbone of nucleic acids reduces their uptake by the cells, in certain embodiments, nucleic acids (e.g., siRNA) may be loaded into VLP (e.g., HPV-VLP) to efficiently deliver them to a subject through VLP (e.g., HPV-VLP) to efficiently deliver them to a subject through
  • V LP administration of V LP
  • encapsulating the nucleic acid into a V LP increases cellular uptake, allows traversal of biological membrane barriers in vivo, and/or increases the bioavailability of the nucleic acid (e.g., siRNA).
  • the agent loaded into the VLP is an anti-viral agent, in certain embodiments, the agent is an anticancer agent, In a preferred embodiment, the anticancer agent is a taxane,
  • the therapeutic agent that may be loaded into a VLP using the methods described herein is a chemotherapeutic agent, for instance, methotrexate, vincristine, adriamycin, cisplatin, non-sugar containing chloroethylnitrosoureas, 5- fiuorouracil, mitomycin C, bleomycin, doxorubicin, dacarbazine, taxol, fragyline, Meglamine GLA, vairubicin, carmustaine and poliferposan, MMI270, BAY 12-9566, RAS famesyl transferase inhibitor, famesyl transferase inhibitor, MiV!P, MTA/LY231514,
  • chemotherapeutic agent for instance, methotrexate, vincristine, adriamycin, cisplatin, non-sugar containing chloroethylnitrosoureas, 5- fiuorouracil, mitomycin C,
  • LY264618 Lometexol. Glamolec, CI-994, TNP-470, Hycamtin/Topotecan, PKC412, Valspodar/PSC833, Novantrone/Mitroxantrone, Metaret/Suramin, Batimastat, E7070, BCH- 4556, CS-682, 9-AC, AG3340, AG3433, Incel VX-710, VX-853, ZD0101, ISI641, ODN 698, TA 2516/Marmistat, BB2516/Marmistat, CDP 845, D2163, PD183805, DX8951f, Lemonal DP 2202, FK 317, Picibanil/OK-432, AD 32/Valrubicin, Metastron/strontium derivative, Temodal/Temozolomide, Evacet liposomal doxorubicin, Y ewtaxan Paclitaxel, Taxol/Paclitaxel, Xeload/
  • Camptosar/Irinotecan Tumodex/Ralitrexed, Leustatin/Cladribine, Paxex/Paclitaxel, Doxil/liposomal doxorubicin, Caelyx/liposomal doxorubicin, Fludara/Fludarabine,
  • Aminogiutethimide Asparaginase, Busulfan, Carboplatin, Chlorambucil, Cytarabine HCI, Dactinomycin, Daunorubicin HCI, Estramustine phosphate sodium, Etoposide (VP 16-213), Floxuridine, Fluorouracil (5-FU), Flutamide, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alfa-2a, Alfa-2b, Leuprolide acetate (LHRH-releasing factor analogue),
  • Lomustine (CCNU), Mechlorethamine HCI (nitrogen mustard), Mercaptopurine, Mesna, Mitotane (o.p ' -DDD), Mitoxantrone HCI, Octreotide, Plicamycin, Procarbazine HCI, Streptozocin, Tamoxifen citrate, Thioguanine, Thiotepa, Vinblastine sulfate, Amsacrine (m ⁇ AMSA), Azacitidine, Erthropoietin, Hexamethylmelamine (HMM), Interleukin 2,
  • Mitoguazone methyl-GAG; methyl glyoxal bis-guanylhydrazone; MGBG
  • Pentostatin (2 ' deox coform cin
  • Semustine methyl-CCNU
  • Teniposide VM-26
  • Viiidesine sulfate but it is not so limited.
  • the therapeutic agent that may be loaded into a VLP using the methods described herein is an immunotherapeutic agent, for instance, Ributaxin, Herceptin, Quadramet, Panorex, IDEC-Y2B8, BEC2, C225, Oncoiym, SMART Ml 95, ATRAGEN, Ovarex, Bexxar, LDP-03, ior t6, MDX-210, MDX-1 1, MDX-22, OV103, 3622W94, anti-VEGF, Zenapax, MDX-220, MDX-447, MELIMMUNE-2, MELIMMUNE- 1, CEACIDE, Pretarget, NovoMAb-G2, TNT, Giiomab-H, GNI-250, EMD-72000,
  • an immunotherapeutic agent for instance, Ributaxin, Herceptin, Quadramet, Panorex, IDEC-Y2B8, BEC2, C225, Oncoiym, SMART Ml 95, ATRAGEN, Ovarex, Bexxar, LDP-03
  • LymphoCide CMA 676, Monopharm-C, 4B5, ior egf r3, ior c5, BABS, anti-FLK-2, MDX- 260, ANA Ab, SMART 1D10 Ab, SMART ABL 364 Ab or IrrunuRAIT-CEA, but it is not so limited.
  • the therapeutic agent that may be loaded into a VLP using the methods described herein is an antiviral agent.
  • anti-viral agents are:
  • Polysulfates PVAS
  • Polysulfonates PVS
  • Polycarboxylates Polyoxometalates
  • Chicoric acid zintevir, cosalane derivatives
  • Bicyclams i.e., AMD3100
  • T-22, T-134, ALX-40-4C CGP-64222, TAK-779
  • AZT azidothymidine
  • ddl, ddC, d4T didehydrodideoxythymidine
  • 3TC 3'-thiadideoxycytidine
  • ABC and other ddN (2',3'-dideoxynucleoside) analogs
  • Nevirapine delavirdine, efavirenz, emivirine (MKC-442), capravirine, thiocarboxanilide UC- 781 , acyclovir, valaciclovir, penciclovir, famciclovir, bromovinyldeoxyuridine
  • Bacmid/cellfectin complex and plate Incubate plates for four days in a humidified 27C tissue culture incubator. Transfer conditioned media to 30ml SF 8f9 culture. Grow culture 3-5 days. Monitor for cell viability and cell diameter using Vi-Cell. Harvest conditioned media and cell pellet when viability is less than 75%. Perform titer (BacPA RapidTiter Kit) and Western Plot analysis. Expand recombinant virus by infecting a 1 L, culture of Sf9 ceils at an MOI of 0.1 with the best expressing Baculovirus clone. Harvest conditioned media by centrifugation once viability has dropped less than 75%. Perform titer analysis using
  • the cell pellet was obtained by tangential- flow filtration, washed in PBS, re-pelleted by centrifugation, and stored at -80°C. Western blot using protein-specific antibodies for LI and L2 proteins were then used to verify the presence of the recombinant protein.
  • Bac-to-Bac selective plates Incubate plates at 37C for two. days.
  • bacmid Cellfectin complex by mixing 1 ug of bacmid that has been diluted with 100 ul of Grace's media with 6ul of cellfectin transfection reagent that has been diluted with 100 ul of Grace's media. Let complexes form for 30 minutes at room temperature. Remove media from the cells in upper left comer well, dilute bacmid cellfectin complex with SOOul of Grace's media, add transfection solution to the upper left corner well. Place plates into a humidified incubator at 27C. After five hours, remove transfection solution from the cells in the upper left corner well and add 2ml of growth media (i.e. Sf-900 II).
  • baculo viral stock For each baculo viral stock, add I ml of the PO viral stock to a 30ml culture in a 125ml shake flask of Sf9 cell at a cell density of le6 cells/ml. An additional SF is utilized as a negative control and lml of growth media added, Shaking incubator parameters are 120 rpm and 27.5C. Cultures are monitored daily with the Vi-Cell for cell density, cell viability, and diameter. In a proper infection, within 48 hours the insect ceil culture should have significantly lower cell density and cell viability and increased cell diameter. Cultures are maintained for three to five days and harvested by centrifugation (2500 x g, 10 minutes) once viability has dropped belo 75%. Transfer the conditioned media (PI) viral stock to a fresh tube and store at 4C. Reserve cell pellet for Western analysis. Determine titer for the pi viral stock using the Clontech BacPAK Rapid Titer Ket according to manufacturer's protocol.
  • baculoviral stock For the best expressing baculoviral stock (i.e. Western Analysis), add 1.5e8 pfu of PI viral stock to a II. cultureof Sf9 ceils in a3L Shake Flask at 1.5e6 ceils per ml (i.e. MOI of 0.1 ). Shaking incubator parameters are 120 rpm and 27.5C. Cultures are monitored daily with Vi- Cell for cell density, cell viability, and cell diameter. Cultures are maintained for two to five days and harvested by centrifugation (2500 x g, 10 minutes) once viability has dropped below 75%. Transfer the conditioned media (PI) viral stock to a fresh sterile bottle and store at 4C. Determine titer for the P2 viral stock using the Clontech BacPAK Rapid Titer Kit according to manufacturer's protocol.
  • Solution 3 (2 mM ( aCk 5 ⁇ CaCL, 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 434 iL dH 2 0) was then added to the above mixture and incubated at 37°C for 30 min.
  • Solution 4 (4 mM CaCi 2 , 10 ⁇ CaCL, 50 mM Tris-HCl pH 7.5, 150 mM NaCL 1224 ⁇ dH 2 0) was then added to the above mixture and incubated at 37°C for 2 hrs.
  • Capsid proteins isolated from insect cells were assembled into VLPs as described. Dynamic light scattering (DLS) demonstrates presence of capsid proteins in moiiomeric and oligomeric forms ( ⁇ 1 Onm) after harvest and prior to the loading procedure. After the reassembly in presence of the nucleic acid payload, VLPs are seen by DLS (50-70nm diameter) (FIG. 3).
  • DLS 50-70nm diameter
  • Results show functional transfer of luciferase expression.
  • VLPs were generated using different production methods to compare efficacy. Transfection of luciferase plasmid (pClucF) using standard iipofectamine transfection at various plasmid amounts (O.lng well, 1 tig/well, lOng/weil) was used to create a range of positive controls. lOng of pClucF plasmid was used without transfection reagent as a reagent/background control.
  • AB1-2 refers to HPV16L1L2 VLP generated using the methods described above, where a single plasmid like p!6sheLL (SEQ ID NO: 6) was used to co-express wildtype HPV L I and 1.2 proteins.
  • Capsid proteins were purified, as described above, from 293 cells transfected with the co-expression plasmid for LI and L2. Capsid proteins were then subjected to the following loading protocol, thereby forming loaded VLP.
  • Solution 3 (2 mM CaCl 2 , 5 ⁇ CaCl 2 , 50 mM Tris-HCl pH 7.5, 150 mM NaCL 434 iL d3 ⁇ 40) was then added to the above mixture and incubated at 37°C for 30 min.
  • Solution 4 (4 mM CaCi 2 , 10 ⁇ CaCi 2 , 50 mM Tris-HCl pH 7.5, 150 mM Nad, 1224 ⁇ dH 2 0) was then added to the above mixture and incubated at 37°C for 2 hrs.
  • Loaded VLP were then used to treat Hela cells in 96 well plates and luciferase signal was read after 48hrs (Table 2, FIGS. 4 and 5).
  • AB iuc3 and AB luc4 were produced in 293 cells after transfection with the l6sheLL plasmid as pseudovirions (PSV) already encapsulating the payload plasmid (pi iucL) (Buck, C, B., et al. (2005) Methods Mol Med., 119: 445-462). Results showed superior transfer of plasmid when the reassembly loading method was used (AB 1 -2) compared with VLPs that were loaded through packaging of plasmid in the host cells (AB luc 3 and AB luc 4).
  • PSV pseudovirions
  • Alopecia means loss of hair from the head or body and includes baldness, a term generally reserved for pattern alopecia or androgenic alopecia (AG A).
  • AGA is the most common cause of hair loss in humans, both male and female and its incidence is increasing 10% per decade.
  • DHT dihydrotestosterone
  • Men with androgenic alopecia typically have higher levels of 5-aipha-reductase, lower levels of total testosterone, higher levels of unbound/free testosterone, and higher levels of total free androgens including DHT (Denmark- Wahnefried 1997).
  • 5-aipha-reductase is responsible for converting free testosterone into DHT.
  • 5-alpha-reductase The genes for 5-alpha-reductase are known (Ellis 2005), The enzymes are present predominantly in the scalp and prostate. Levels of 5-alpha- reductase are one factor in determining levels of DHT in the scalp and drugs which interfere with 5-alpha-reductase (such as Finasteride, which inhibits the predominant type 2 isoform) have been approved by the FDA as treatments for hair loss.
  • the adverse side effects of the Minoxidil dosing include scalp irrita tion and allergic contact dermatitis.
  • the side effects associated with the DHT inhibitor, Finasteride, which when applied topically has very little penetration are more serious.
  • the compounds and formulas of U.S. Patent No. 6,268,377 which is hereby incorporated by reference, for the treatment of hyperandrogenic condition include the 5-alpha-reductase inhibitors and calcium channel blocking agents which may be combined in a single oral, topical, systemic or parenteral dosage formulation.
  • the composition has a number of side effects that include: erectile dysfunction (ED) or impotence in up to 18.5 percent of people, decreased libido up to 10 percent, abnormal ejaculation, including decreased ejaculate amount, up to 7.2 percent, and breast enlargement up to 2.2 percent. .
  • ED erectile dysfunction
  • impotence in up to 18.5 percent of people
  • libido up to 10 percent
  • abnormal ejaculation including decreased ejaculate amount, up to 7.2 percent
  • breast enlargement up to 2.2 percent.
  • Propecia blocks the fomiation of DHT which is intended to inhibit the development of male pattern baldness.
  • the ineffectiveness of Propecia in treating male patterned baldness and the adverse effects associated with the currently prescribed method of dosing and treating male pattern baldness indicate that a more effective treatment is needed.
  • a preferred method for treating alopecia preferably includes using a combination of betapapillomavirus viral shells (L1/L2) to deliver a DHT inhibitor as a therapeutic agent.
  • the method preferably includes the steps of constructing a recombinant DNA molecule which contains a sequence encoding a papillomavirus LI protein or a papillomavims L2 protein or a combination of L I and 1.2 proteins and transfecting a host cell with the recombinant DNA molecule.
  • the virus like particles may express papillomavirus L I protein or L2 protein or a combination of LI and L2 proteins in the host cell.
  • the papillomavirus virus-like particles obtained from the transfected host cell may be purified which will cause the disassembling the LI and L2 capsid proteins of the virus-like particles into smaller units.
  • it is these smaller disassembled LI and L2 capsid proteins which may be loaded with a DHT inhibitor.
  • the loaded proteins may be reassembled to form a loaded virus-like particles comprising HPV protein with the DHT inhibitor and administered to the skin of an animal or a human subject.
  • the method can be applied to enhance topical deliver ⁇ ' of Minoxidil , Finasteride or other drugs to treat AGA topically through the skin and through the hair follicle.
  • the present invention provides a method for treating alopecia 1200, which includes a first step in which a recombinant DNA molecule is contracted which contains a sequence for encoding a papillomavims LI protein or a papillomavirus 1.2 protein, or a combination of papillomavirus L I and L2 proteins 1220. Thereafter, a host cell will be transfected with the recombinant DNA molecule 1230.
  • the transfected host cell will be treated to purify the papillomavirus vims-like particles causing the LI and L2 capsid proteins to disassemble into smaller units 1240, At which time, an appropriate therapeutic agent or drag for treating AGA will be introduced into the proximity of the virus-like particle where the agent or drag for treatment will be loaded into the virus-like particles 1250. Thereafter, the loaded virus-like particles comprising HPV protein with an effective DHT inhibitor for treating alopecia may be reassembled 1260. Finally, the treatment may preferably be topically applied through the skin and the hair follicles 1270 for the treatment of alopecia ,
  • the above method further includes a step to pre-treat the area of the skin.
  • the area of the scalp to be treated for alopecia may preferably be prepared by pre-administering a delipidating substance to the region of the scalp before applying the treatment of HPV pseudo-virus particles loaded with a DHT inhibitor to the scalp of a human subject,
  • the above method also further includes another pre -treatment of the scalp.
  • the area of the scalp to be treated for alopecia may be pre -treated by micro-abrasion before applying the treatment of HPV pseudo-virus particles loaded with a DHT inhibitor to the scalp of a human subject
  • RNA short interfering nucleic acid
  • a siRNA (short interfering nucleic acid) molecule may be formulated as a composition as described in U.S. Patent Application Publication No. 200770179104 A l (which is hereby incorporated by reference herein).
  • the formulated composition which may be loaded into the betaHPV pseudovirions and transdermally administered may include such siRNA formulations which are generally referred to as "lipid nucleic acid particles" (LNP) which include membrane disruptive agents in which the siRNA molecule is complexed with a cationic lipid and helper lipid molecule; or formulated with polyethylenimine derivatives, including for example grated PEIs such as galactose PEI, cholesterol PEI, antibody derivatized PEL and polyethylene glycol PEI (PEG-PEI) and/or derivatives thereof,
  • LNP lipid nucleic acid particles
  • PEG-PEI polyethylene glycol PEI
  • a method for topically treating non-melanoma skin cancer using a combination of betapapillomavirus viral shells (L.1/L2) to deliver a siRNA against the transcription factor protein for GH2 is provided.
  • other HPV viral shells may also be used.
  • HPV5 viral shells may be used.
  • a herpes viral shell may also be use.
  • a first step in this preferred emboidement includes constructing a recombinant DNA molecule that contains a sequence encoding a papillomavirus LI protein or a papillomavirus L2 protein or a combination of LI and L2 proteins and then transfecting a host cell with the recombinant DNA molecule.
  • the virus like particles may express papillomavirus LI protein or L2 protein or a combination of L I and L2 proteins in the host cell.
  • the betapapillomavirus virus-like particles obtained from the transfected host cell may be purified which will cause the disassembling of the LI and L2 capsid proteins of the vims-like particles into smaller units.
  • L I and L2 capsid proteins which may be combmed with a siRNA against the transcription factor protein for GH2.
  • the combination of siRNA with proteins may be reassembled to form loaded vims-like particles comprising HPV protein with the siRNA against the transcription factor protein for GU2 and administered to the skin of an animal or a human subject.
  • the present invention provides a method for treating non-melanoma skin cancer 800, which includes a first step in which a recombinant DNA molecule is contracted which contains a sequence for encoding a papillomavirus LI protein or a papillomavirus L2 protein, or a combination of papillomavirus LI and L2 proteins 820. Thereafter, a host cell will be transfected with the recombinant DNA molecule 830. After which, the transfected host ceil will be treated to purify the LI and L2 capsid proteins into smaller units 840.
  • an appropriate therapeutic agent or drug for treating non-melanoma skin cancer will be introduced into the proximity of the proteins where the agent or drug for treatment will be combined with the Nanosphere particle 850. Thereafter, the particles comprisingsiRNA against the transcription factor protein for GH2 may be reassembled 860, Finally, the treatment may preferably be topically applied through the skin 870 for the treatment of non-melanoma skin cancer.
  • these smaller units will include intermediate structure and capsomers.
  • these smaller units of capsid proteins, intermediate structures and capsomers are purified and then combined with a siRNA targeting transciption factor Gli2. Thereafter, reassembly of the capsid proteins is initiated to form loaded virus-like particles which preferably include PV proteins with attached siRNA targeting transcription factor GH2.
  • a method for topically treating Pachyonychia Congenita using a combination of betapapillomavirus viral shells (L1/L2) to deliver a siRNA targeting the N17K1 mutation in the keratin 6a gene is provided,
  • the siRNA of present invention may alternatively target known mutations in the genes encoding K6b, K16 and/or K17A.
  • the present invention provides a method for treating Pachyonychia Congenita 900, which includes a first step in which a recombinant DNA molecule is contracted which contains a sequence for encoding a papillomavirus LI protein or a papillomavirus L2 protein, or a combination of papillomavirus LI and L2 proteins 920. Thereafter, a host cell will be transiected with the recombinant DNA molecule 930.
  • the transiected host cell will be treated to purify the papillomavirus virus-like particles causing the LI and L2 capsid proteins to disassemble into smaller units 940.
  • an appropriate therapeutic agent or drug for treating Pachyonychia Congenita will be introduced into the proximity of the virus-like particle where the agent or drug for treatment will be loaded into the viras-like particles 950.
  • the loaded vims-like particles enclosing siRNA targeting the N17K1 mutation in keratin 6a may be reassembled 960.
  • the treatment may preferably be topically applied through the skin 970 for the treatment of Pachyonychia Congenita.
  • Table 1 shows a list of additional genetic skin related disorders which may be considered treatable in accordance with the present invention:
  • ADULT syndrome Acro-dermato-ungual-lacrimal-tooth syndrome
  • Ankyloblepharon-ectodermal defects-cleft lip and palate syndrome (AEC syndrome) Arthrogryposis and ectodermal dysplasia
  • CHS Curly hair-ankyloblepharon-nail dysplasia
  • Oculodentodigital displasia ODDD
  • a method for topically treating Psoriasis using a combination of betapapillomavirus viral shells (L1/L2) to deliver a siRNA against cytokines is provided.
  • a first step in this preferred embodiment includes constructing a recombinant DNA molecule that contains a sequence encoding a papillomavirus LI protein or a papillomavirus L2 protein or a combination of LI and L2 proteins and then transfecting a host cell with the recombinant DNA molecule.
  • the virus like particles may express papillomavirus LI protein or L2 protein or a combination of LI and L2 proteins in the host ceil.
  • the betapapillomavirus virus-like particles obtained from the transfected host cell may be purified which will cause the disassembling of the LI and L2 capsid proteins of the viras-like particles into smaller units.
  • these smaller disassembled LI and 1.2 capsid proteins which may be loaded with a si R NA against cytokines.
  • the loaded protems may be reassembled to form loaded virus-like particles comprising HPV protein with the siRNA against cytokines and administered to the skin of an animal or a human subject.
  • the present invention provides a method for treating Psoriasis 1 100, which includes a first step in which a recombinant DNA molecule is contracted which contains a sequence for encoding a papillomavirus LI protein or a papillomavirus L2 protein, or a combination of papillomavirus LI and L2 proteins 1 120. Thereafter, a host cell will be transfected with the recombinant DN A molecule 1 130.
  • the transfected host cell will be treated to purify the papillomavirus virus-like particles causing the LI and L2 capsid proteins to disassemble into smaller units 1 140.
  • an appropriate therapeutic agent or drag for treating Psoriasis will be introduced into the proximity of the virus-like particle where die agent or drug for treatment will be loaded into the virus-like particles 1 150.
  • the loaded virus-like particles enclosing siRNA against cytokines may be reassembled 1160.
  • the treatment may preferably be topically applied through the skin 1170 for the treatment of psoriasis.
  • the RNA which inhibits the expression of cytokines is an siRNA which inhibits the expression of TNF-a for the treatment of psoriasis.
  • alternati ve viral vectors may be used in place of the betapapillomavirus.
  • alternative viral vector may include herpes virus vectors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Plant Pathology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Virology (AREA)
  • Dermatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

This invention relates to a transdermal delivery system for treating skin-related diseases employing papilloma-derived protein nanoparticles to deliver drugs to the keratinocytes and basal membrane cells for the treatment of skin-related diseases. The current invention presents an effective method for delivering small molecule nucleic acids to the epidermal cells.

Description

VIRION DERIVED PROTEIN NANOPARTICLES FOR DELIVERING
[001]. RELATED APPLICATIONS
[002]. This application claims the benefit of priority to U.S. Provisional Application 61/506,140 filed July 10, 201 1 , U.S. Non-Provisional Application No. 13/221 ,803 filed August 30, 2011, and U.S. Non-Provisional Application No. 13/253,028 filed October 4, 201 1. The disclosure of the abo ve applications are incorporated herein by reference.
[003]. REFERENCE TO SEQUENCE LISTING
[004]. The Sequence Listing provides exemplary polynucleotide sequences of the invention. The traits associated with the used of the sequences are included in the Examples.
[005]. The Sequence Listing submitted as an initial paper is named AURA_16_ST25.txt, is 45 kilobytes in size, and the Sequence Listing was created on 29 November 2011. The copies of the Sequence Listing submitted via EFS-Web as the computer readable for are hereby incorporated by reference in their entirety.
[006]. FIELD OF INVENTION
[007]. The invention relates to methods for loading protein nanoparticles with therapeutic, diagnostic or other agents, wherein the protein nanoparticles are based on viral proteins. More particularly, the present invention relates to a method for using papilloma-derived protein nanoparticles to deliver drugs to the keratinocytes and basal membrane cells for the treatment of skin-related diseases such as alopecia or patterned hair loss, non-melanoma skin cancer, psoriasis, and treatment of skin related genetic diseases (e.g. Pachyonychia Congenita and Xeroderma pigmentosum).
BACKGROUND OF THE INVENTION
[009]. Today there are a number of techniquies available for the use of virus-like particles for the deliver}' of siRNA, A number of these techniques are discussed and outlined in a recent survey by Lund et al, entitled Pseudovirions as Vehicles for the Delivery of siRNA Pseudoviral Systems which is hereby incorporated by reference. As detailed by Lund, there are at present five distinct RNAi techniques being developed and tested for the delivery of siRNA.
[0010]. The first three systems reviewed are the phagemid particles and Herpes Simplex Virus- 1 (HSV) amplicons, in which the RNAi is derived from DNA, and the SV40 in vitro- packaged vectors. These three systems are examples of "trans-packaging systems in which viral machinery and proteins have been repurposed to package genetic material other than the original viral genome" (Lund et al. 401).
[0011]. The fourth and fifth systems reviewed, the influenza Virosome and the HVJ-E, a derivative of the Hemagglutinating Virus of Japan , are described as "viral envelope systems, in which native enveloped virus particles are inactivated and used to package other cargos" (Lund et al. 412) ,
[0012], interestingly and most pertinent to the present invention, Lund details that the present methods for delivering siRNA know in the art. are limited because, "Systems such as HSV amplicons and phagemid particles are only able to deliver DNA, which is inherently more risky than siRNA oliogomers, due to the possibility of integration into the host genome and insertional mutagenesis."
[0013], Lund and other have raised additional concerns regarding the extent to which other available vectors such as the influenza virosome, HVJ-E, and S V40 in-vitro packaged pseudovirions, are inherently immunogenic.
[0014], With regards to other routes of admisitration, studies have attempted intradermal deliver}' of siRNA by injection. This method of delivery has shown effective knockdown of targeted gene expression but it is painful and the effects are localized to the injection site. Further, the delivery of siRN A through the stratum corneum is necessary but not sufficient for delivery to epidermal cells and additional steps must be taken to facilitate nucleic acid uptake by keratinocytes (and endosomal release) to allow access to the RNA-induced silencing complex. Because of these limitation, there is no existing method in the prior art which achieves both barrier disruption and intracellular delivery. [0015], Accordingly, there is an unmet need for delivery strategies that increase drug half- life, bioavailability, selectivity and targeted, sustained release of key drugs, More
specifically, there is an urgent need to deliver nucleic acid therapies to skin.
[0016], SUMMARY OF INVENTION
[0017], The object of the present invention is to overcome the shortcomings disclosed in the prior art. The products of the current invention are pseudo-viruses derived from
papillomaviruses, in particular from the genus betapapillomaviridae to deliver drugs to the keratinocytes and basal membrane cells for the treatment of skin related diseases. The present in vention wil l allow for impro ved methods for administering treatment of skin diseases capable of targeting epithelial cells.
[0018]. More specifically, the present invention describes the use of betaHPV
pseudovirions and in particular type 5 betaHPV viral shells (LI and L2) as an ideal delivery vehicle for the delivery of drugs to the skin for the treatment of skin related diseases such as alopecia or patterned hair loss, non-melanoma skin cancer, psoriasis, and treatment of skin related genetic diseases (e.g. Pachyonychia Congenita and Xeroderma pigmentosum).
[0019]. Aspects of the invention relate to methods and compositions for producing papilloma-derived (e.g., human papilloma virus (HPV)-derived) protein nanoparticles containing one or more therapeutic or diagnostic agents. In some embodiments, aspects of the invention relate to methods and compositions for encapsulating an agent within a virus like particle (VLP) requiring an initial isolation of capsid proteins produced in a host cell system (e.g. yeast, mammalian cell, insect cell, E.coli) and subsequent reassembly in vitro.
[0020]. The accompanying drawings, which are incorporated in and constitute part of the specification, illustrate various embodiments of the invention and together with the description, serve to explain the principles of the invention.
[0021 ]. Brief Description of the Sequence Listings and Drawings
[0022]. FIG. 1 depicts shuttle vector information.
[0023]. FIG. 2 depicts LI capsid protein in various fractions from insect cell culture (T=total cell lysate, C= cytoplasmid fraction, TN=total nuclear fraction, SN = soluble nuclear fraction). Harvest times after baculovirus infection indicated. [0024], FIG. 3 shows results from in vitro reassembly of capsid protein produced in insect cell culture. DLS demonstrates presence of capsid protein in form of monomers and oligomers after harvest from nuclear fraction (left) and appearance of well formed loaded VLPs after the reassembly procedure (right).
[0025], FIG. 4 is a graph showing the amount of luminescence/luciferase signal measured 48 hrs after treatment of HeLa cells with loaded VLP, where luminescence is reported on a scale of 0 to 30,000 units along the y-axis.
[0026]. FIG. 5 is a graph the same data in FIG.4, showing the amount of
luminescence/luciferase signal measured 48 hrs after treatment of HeLa cells with loaded VLP, where luminescence is reported on a scale of 0 to 20 units along the y-axis.
[0027]. FIG. 6 shows a fl ow chart diagram of a preferred embodiment of the present invention for the treatment of alopecia.
[0028], FIG. 7 shows a flow chart diagram of a preferred embodiment of the present in vention for the treatment of non-melanoma skin cancer.
[0029], FIG. 8 shows a flo w chart diagram of a preferred embodiment of the present invention for the treatment of skin related genetic diseases.
[0030]. FIG. 9 shows a flow chart diagram of a preferred embodiment of the present invention for the treatment of psoriasis.
[0031]. (SEQ ID NO: 1) shows DNA sequence for baculovirus L! X plasmid encoding HP V 16/31 L 1 (pFastBac™).
[0032]. (SEQ ID NO: 2) shows DNA sequence for baculovirus L2 plasmid encoding
HPV16L2 (pFastBac™).
[0033]. (SEQ ID NO: 3) shows forward primer DNA sequence used for generation of shE7- 1 RNA construct.
[0034]. (SEQ ID NO: 4) shows reverse primer DNA sequence used for generation of shE7- 1 RNA construct. [0035], (SEQ ID NO: 5) shows plasmid pl 6Ll *L2 DNA sequence encoding 16/31 L I (LI *) and L.2 human codon -optimized,
[0036], (SEQ ID NO: 6) shows pi 6sheLL plasmid DNA sequence.
[0041 , 1 ] (SEQ ID NO: 7) shows a sequence for the complete genome for Human
papillomavirus - 5.
[0041.2] (SEQ ID NO: 8) shows a sequence for P5 SHELL circular plasmid DS-DNA. [0037], DETAILED DESCRIPTION OF THE INVENTION [0038], Assembly Of Particles
[0039], To assemble the biological, pharmaceutical or diagnostic components to a described biological cargo-laden nanoparticles used as a carrier, the components can be associated with the nanoparticles through a linkage. By "used as a carrier associated with", it is meant that the component is carried by the nanoparticles. The component can be dissolved and incorporated in the nanoparticles non-covalently. Preferred and illustrative methods for creating, loading and assembling particles for use with the present are taught in following applications which are hereby incorporated by reference in their entirety:
PCT/US2009/00429 entitled "HVP PARTICLES AND USES THEREOF;" PCT/IB2010/002654, 1 1/24/2010 entitled "TARGETING OF PAPILLOMA VIRUS GENE DELIVERY PARTICLES;" U.S. Provisional Application No, 61/417,031 entitled "METHOD FOR LOADING HPV PARTICLES;" and U.S. Provisional Application No. 61/491 ,774 entitled "PAPILLOMA-DERIVED PROTEIN
NANOSPHERES FOR DELIVERING DIAGNOSTIC OR THERAPEUTIC AGENTS."
[0040], In some embodiments, aspects of the invention relate to methods and compositions for producing protein nanoparticles that contain therapeutic and/or diagnostic agents for delivery to a subject. Methods and compositions have been developed for effectively encapsulating therapeutic and/or diagnostic agents within papilloma virus proteins (e.g., HPV proteins) that can be used for delivery to a subject (e.g. , a human subject).
[0041]. In some embodiments, it has been discovered that it is useful to isolate LI and L2 capsid proteins directly from host cells as opposed to disassembling VLPs that were isolated from host cells. LI and L2 capsid proteins that are isolated directly from cells can be used in in vitro assembly reactions to encapsulate a therapeutic or diagnostic agent. This avoids the additional steps of isolating and disassembling VLPs. This also results in a cleaner preparation of LI and L2 proteins, because there is a lower risk of contamination with host cell material (e.g., nucleic acid, antigens or other material) that can be contained in VLPs that are isolated from cells.
[0042], In some embodiments, it has been discovered that expressing LI and/or L2 proteins intracellularly in the presence of a therapeutic or diagnostic agent can be useful in the production of a loaded VLP intracellularly tha t encapsulates the agent.
[0043]. In some embodiments, it is useful to independently produce LI and L2 capsid proteins. In some embodiments, they can be produced from two independent nucleic acids (e.g., different vectors). In some embodiments, they can be produced in the same cell (e.g., using two different vectors within the same cell). In some embodiments, they can be produced in different cells (e.g., different host cells of the same type or different types of host cell). This approach allows the ratio of LI and L2 proteins to be varied for either in vitro or intracellular assembly. This allows VLPs to be assembled (e.g., in vitro or intracellularly) with higher or lower LI to L2 ratios than in a wild type VLP. This may have benefits in the use of H PV nanoparticles as deliver}' vehicles for therapeutic agents. A higher ratio of L,2 in the assembled structure may allow the resultant VLP to have a higher nucleic acid binding affinity and a better efficiency in delivering these intracellularly.
[0044]. Capsid proteins:
[0045], In some embodiments, LI and L2 proteins are expressed in a host cell system (e.g., both in the same host cell or independently in different host cells). LI and/or L2 are isolated from nuclei of the host cells. In some embodiments, certain LI and/or L2 structures that are formed during cellular growth (e.g., during the fermentation process) are disrupted. Any suitable method may be used. In some embodiments, sonication may be used (e.g., nuclei may be isolated and then sonicated). Capsid proteins then may be purified using any suitable process. For example, in some embodiments, capsid proteins may be purified using chromatography.
[0046]. Isolated capsid proteins can then be used as described herein in a cell free system to assemble together with different payloads to create superstructures that contain a drug or diagnostic agent in its interior. [0047], It should be appreciated that directly isolating capsid proteins (as opposed to isolating and disassembling VLPS) provides several benefits, In some embodiments, there is a reduced risk of encapsulating and transferring genetic information (DNA, RNA) from the host cell to the treated subject. In certain embodiments, de-novo assembly of VL Ps during the assembly procedure ensures formation of a larger percentage of loaded VLPs as opposed to using already-formed VLPs for loading where a certain fraction can remain unloaded.
[0048]. Cellular production:
[0049]. In some embodiments, one or more therapeutic or diagnostic agents may be loaded intracellularly by expressing LI and/or L2 in the presence of intracellular levels of one or more agents of interest,
[0050]. In some embodiments, this method is used for encapsulating a silencing plasmid which will encode for expression of short hairpin RN A (shRNA), In some embodiments, this plasmid will have a size of 2kB-6kB. However, any suitable size may be used. In some embodiments, a plasmid is designed to be functional within the cells of the patient or subject to be treated (to which the loaded V LP is administered). Accordingly, the plasmid will be active within the target cells resulting in knockdown of the targeted gene(s).
[0051], In some embodiments, this method may be used to encapsulate short interfering RNA (siRNA) or antisense nucleic acids (DN A or RNA) transfected into the host cells (e.g., 293 cel ls or other mammalian or insect host cells) during the production of the V LPs.
[0052], Accordingly, loaded VLPs may be produced intracellularly to provide gene silencing functions when delivered to a subject.
[0053], It should be appreciated that there are several benefits to this method. In some embodiments, encapsulation of RNA interference (RNAi) constructs into V LPs allows for very efficient transfer of RNAi or Antisense nucleic acid into target cells,
[0054], Independent expression vectors:
[0055], In some embodiments, LI and L2 proteins are expressed in a host ceil system (e.g. mammalian cells or insect cells) from independent expression nucleic acids (e.g., vectors, for example, plasmids) as opposed to both being expressed from the same nucleic acid. [0056], it should he appreciated that the expression of LI and L2 from independent plasmids allows the relative levels of L1/L2 VLP production to be optimized for different applications and to obtain molecular structures with optimal delivery properties for different payloads. In some embodiments, a variety of VLP structures can be produced to fit the needs of the different classes of payloads (e.g., DNA, R A, small molecule, large molecule) both in terms of charge and other functions (e.g. DNA binding domains, VLP inner volume, endosomal release function). VLPs with a higher content of L2 protein will be better to bind nucleic acids (L2 contains a DNA binding domain) whereas VLPs with a smaller content of L2 protein will be better for other small molecules. VLPs with different ratios of LI :L2 protein will have different inner volumes that wrill allow a higher concentration of drug to be encapsulated. In some embodiments, the release of pay load into the cell will also be modulated. In some embodiments, structures containing more L2 protein may have a higher ability to transfer nucleic acids intracellularly. It should be appreciated that different ratios of L1/L2 may be used, in some embodiments, ratios may be 1 :1 , 1 :2, 1 :4, 1:5, 1 :20 or 1 :100. However, other ratios may be used as aspects of the invention are not limited in this respect.
[0057]. In some embodiments, each separate expression nucleic acid encodes an LI (but not an L,2) or an L2 (but not an LI ) sequence operably linked to a promoter. In some
embodiments, other suitable regulatory sequences also may be present. The separate expression nucleic acids may use the same or different promoters and/or other regulatory sequences and/or replication origins, and/or selectable markers. In some embodiments, the separate nucleic acids may be vectors (e.g., plasmids, or other independently replicating nucleic acids). In some embodiments, separate nucleic acids may be independently integrated into the genome of a host cell (e.g., a first nucleic acid integrated and a second nucleic acid on a vector, two different nucleic acids integrated at different positions, etc.). In some embodiments, the relative expression levels of LI and L2 may be different in different cells, different using different expression sequences, independently regulated, or a combination thereof.
[0058]. Variant HPV proteins having reduced immunogenicity:
[0059]. In some embodiments, an expression vector is used to produce a mutant LI or L2 protein. In some embodiments, a mutant HPV16L1 protein (called LI *) is expressed along with L2 in a host system (e.g., a 293 cell system). These can then be isolated and assembled as described herein to encapsulate a therapeutic or diagnostic payload (e.g. therapeutic plasmid, siRNA, small molecule drugs, etc.).
[0060]. In some embodiments, loaded VLPs are produced using certain LI and/or L2 variant sequences that are not recognized by existing antibodies against HPV (e.g.,
HPV16L1) that might be present in patients who have an ongoing HPV infection or who have received the vaccine. It also should be appreciated that loaded VLPs can be produced using LI and/or L2 proteins that are modified to reduce antigenicity against other HPV serotype antibodies and/or to target the loaded VLP to particular organs or tissues (e.g., lung) or cells or subcellular locations.
[0061]. Accordingly, certain aspects of the invention relate to methods for loading VLPs with therapeutic, diagnostic or other agents. In certain embodiments, the papilloma virus particles are HPV-VLP. In certain embodiments, the methods described herein utilize HPV- VLP that contain one or more naturally occurring HPV capsid proteins (e.g., LI and/or L2 capsid proteins). HPV-VLP may be comprised of capsid protein oligomers or monomers.
[0062]. A "VLP" refers to the capsid-like structures which result upon assembly of a HPV L I capsid protein alone or in combination with a HPV L2 capsid protein. VLPs are morphologically and antigenically similar to authentic virions. VLPs lack viral genetic material (e.g., viral nuclei acid), rendering the VLP non-infectious. VLP may be produced in vivo, in suitable host cells, e.g., mammalian, yeast, bacterial and insect host cells.
[0063]. A "capsomere" refers to an oligomeric configuration of LI capsid protein.
Capsomeres may comprise at least one L I (e.g., a pentamer of LI).
[0064]. A "capsid protein" refers to LI or L2 proteins that are involved in building the viral capsid structure. Capsid proteins can form oligomeric structures i.e. pentamers, trimers or be in single units as monomers.
[0065], in some embodiments, a VLP can be loaded with one or more medical, diagnostic and/or therapeutic agents, or a combination of two or more thereof. In some embodiments, the methods described herein utilize HPV-VLP that contain one or more variant capsid proteins (e.g., variant LI and/or L2 capsid proteins) that have reduced or modified immunogenicity in a subject. Examples of varian t capsid proteins are described in WO 2010/120266. The modification may be an amino acid sequence change that reduces or avoids neutralization by the immune system of the subject. In some embodiments, a modified HPV-VLP contains a recombinant HPV protein (e.g., a recombinant L I and/or L2 protein) that includes one or more amino acid changes that alter the immunogenicity of the protein in a subject (.e.g., in a huma subject). In some embodiments, a modified HPV-VLP has an altered immunogenicity but retains the ability to package and deliver molecules to a subject.
[0066]. In certain embodiments, amino acids of the viral wild-type capsid proteins, such as LI and/or LI +1,2, assembling into the HPV-VLP, are mutated and/or substituted and/or deleted. In certain embodiments, these amino acids are modified to enhance the positive charge of the VLP interior. In certain embodiments, modifications are introduced to allow a stronger electrostatic interaction of nucleic acid molecules with one or more of the amino acids facing the interior of the VLP and/or to avoid leakage of nucleic acid molecules out of the VLP. Examples of modifications are described in WO 2010/120266. It should be appreciated that any modified HPV-VLP may be loaded with one or more agents. Such particles may be delivered to a subject without inducing an immune response that would be induced by a naturally-occurring HPV,
[0067]. In certain embodiments, HPV- VLP, loaded according to the methods described herein, are useful for delivering one or more therapeutic agents to diseased tissue (e.g., diseased mucosal tissue). In some embodiments, a diseased tissue (e.g. , mucosal tissue, epithelial tissue, or endothelial tissue) may be a infected tissue (e.g., infected with a virus such as HP V or HSV). In some embodiments, the mucosal tissue is cervical tissue and the disease is dysplasia or cancer (e.g., cervical dysplasia, cervical cancer, for example associated with persistent HPV infection). In some embodiments, HPV-VLP, loaded according to the methods described herein, may be used to deliver compositions to other tissues (e.g. , epidermis), In some embodiments, HPV-VLP may be used to treat HPV related diseases, In some embodiments, HPV-VLP, loaded according to the methods described herein, may be used to deliver therapeutic agents to treat a disease or condition, and/or may be used to deliver diagnostic agents to diagnose other diseases or conditions. For example, quantum dots, metals, and/or other imaging agents may be loaded into VLP according to the methods described herein and delivered. In some embodiments, agents may be used to track early stage diseases (e.g. , early stage metastasis). It should be appreciated that any suitable therapeutic, diagnostic and/or other medical agent may be loaded into VLP according to the methods described herein and delivered to a subject. Examples of administration of VLP to subjects are described for example in U.S. Patent No. 7,205,126, incorporated herein by reference.
[0068]. in some embodiments, HPV-VLPs that are modified to display epitopes from two or more different naturally-occurring HPV variants are used in the agent loading methods described herein. Such modified VLPs may be used to provide immunization against infection by any of two or more naturally occurring HPV variants.
[0069], In some embodiments, HPV-VLPs comprise viral LI capsid proteins. In some embodiments, HPV-VLPs comprise viral LI capsid proteins and viral L2 capsid proteins. The LI and/or L2 proteins may, in some embodiments, be wild-type viral proteins. In some embodiments, L i and/or L2 capsid proteins may be altered by mutation and/or deletion and/or insertion so that the resulting LI and/or L2 proteins comprise only 'minimal' domains essential for assembly of a VLP. In some embodiments, LI and/or L2 proteins may also be fused to other proteins and/or peptides that provide additional functionality. Examples of modifications are described for example in U.S. Patent No. 6,991 ,795, incorporated herein by reference, These other proteins may be viral or non-viral and could, in some embodiments, be for example host-specific or cell type specific. It should be appreciated that VLPs may be based on particles containing one or more recombinant proteins or fragments thereof (e.g. , one or more HPV membrane and/or surface proteins or fragments thereof). In some embodiments, VLPs may be based on naturally-occurring particles that are processed to incorporate one or more agents as described herein, as aspects of the invention are not limited in this respect, In certain embodiments, particles comprising one or more targeting peptides may be used. Other combinations of HPV proteins (e.g., capsid proteins) or peptides may be used as aspects of the invention are not limited in this respect.
[0070]. In some embodiments, viral wild-type capsid proteins are altered by mutations, insertions and deletions. All conformation-dependent type-specific epitopes identified to date are found on the HPV-VLP surface within hyper-variable loops where the amino acid sequence is highly divergent between H PV types, which are designated BC, DE, EF, FG and HI loops. Most neutralizing antibodies are generated against epitopes in these variable loops and are type-specific, with limited cross-reactivity, cross-neutralization and cross-protection. Different HPV serotypes induce antibodies directed to different type-specific epitopes and/or to different loops. Examples of variant capsid proteins are described in WO 2010/120266. [0071], In certain embodiments, viral capsid proteins, HPV L I and/or L2, are mutated at one or more amino acid positions located in one or more hyper-variable and/or surface- exposed loops. The mutations are made at amino acid positions within the loops that are not conserved between HPV serotypes. These positions can be completely non-conserved, that is that any amino acid can be at this position, or the position can be conserved in that only conservative amino acid changes can be made.
[0072]. In certain embodiments, LI protein and L1÷L2 protein may be produced
recombinantly. in certain embodiments, reeombraantly produced LI protein and L1+L2 protein may self-assemble to form vims-like particles (VLP). Recombinant production may occur in a bacterial, insect, yeast or mammalian host system. LI protein may be expressed or L1÷L2 protein may be co-expressed in the host system.
[0073]. Cellular hosts that are useful for expressing and purifying HPV LI and/or L2 recombinant viral capsid proteins are known in the art. For example, HPV LI and/or L2 proteins may be expressed in Spodoptera frugiperla (8f21) cells. Baculoviruses encoding the LI and/or L2 gene of any HPV or recombinant versions thereof from different serotypes (e.g., HPV 16, HPV18, HPV31, and HPV58) may be generated as described in Touze et /., FEMS Microbiol. Lett. 2000; 189:121-7; Touze et al. J, Clin. Microbiol. 1998; 36:2046-51); and Combita et ., FEMS Microbiol. Lett. 2001; 204(1): 183-8. HPV LI and/or L,2 genes may be cloned into a plasmid, such as pFastBacl (Invitrogen). Sf21 cells may be maintained in Grace's insect medium (Invitrogen) supplemented with 10% fetal calf serum (PCS, invitrogen) and infected with recombinant baculoviruses and incubated at 27°C. Three days post infection, cells can be harvested and VLP can be purified. For example, cells may be resuspended in PBS containing Nonidet P40 (0.5%), pepstatin A, and leupeptin (1 μ^ητΐ each, Sigma Aldrich), and allowed to stand for 30 min at 4°C. Nuclear lysates may then be centrifuged and pellets can be resuspended in ice cold PBS containing pepstatin A and leupeptin and then sonicated. Samples may then be loaded on a CsCl gradient and centrifuged to equilibrium (e.g., 22 h, 27,000 rpm in a SW28 rotor, 4°C). CsCl gradient fractions may be investigated for density by refractonietry and for the presence of L1/L2 protein by electrophoresis in 10% sodium dodecyl sulfate-polyacryiamide gel (SDS-PAGE) and Coomassie blue staining. Positive fractions can be pooled, diluted in PBS and pelleted e.g., in a Beckman SW 28 rotor (3 h, 28,000 rpm, 4°C), After centrifugation, VLP can be resuspended in 0.15 mol/'L NaCl and sonicated, e.g., by one 5 second burst at 60% maximum power. Total protein content may be determined. [0074], Viral capsid proteins may also be expressed using galactose-inducible Saccharomyces cerevisiae expression system, Leucine-free selective culture medium used for the propagation of yeast cultures, yeast can be induced with medium containing glucose and galactose. Cells can be harvested using filtration, After resuspension, cells may be treated with Benzonase and subsequently mechanically disrupted (e.g., using a homogenizer). Ceil lysate may be clarified using filtration. An exemplary protocol can be found in Cook et al. Protein Expression and Purification 17, 477-484 (1999).
[0075], Buck et al. (J. Virol. 78, 751-757, 2004) reported the production of papilloma virus-like particles (VLP) and cell differentiation-independent encapsidation of genes into bovine papillomavirus (BPV) LI and L2 capsid proteins expressed in transiently transfected mammalian cells, 293TT human embryonic kidney cells, which stably express SV40 large T antigen to enhance replication of SV40 origin-containing plasmids. Pyeon et al. reported a transient transfection method that achieved the successful and efficient packaging of full- length HPV genomes into HPV16 capsids to generate vims particles (PNAS 102, 9311-9316 (2005)). Transiently transfected cells (e.g., 293 cells, for example 293T or 293TT cells) can be lysed by adding Brij58 or similar nonionic polyoxyethylene surfactant detergent, followed by benzonase and exonuclease V and incubating at 37°C for 24 h to remove unpackaged cellular and viral DNA and to allow capsid maturation. The lysate can be incubated on ice with 5 M NaCl and cleared by centrifugation. VLP can be collected by high-speed centrifugation.
[0076], Capsid proteins may also be expressed in E. coli. In E.coli, one important potential contaminant of protein solutions is endotoxin, a lipopolysaccharide (LPS) that is a major component of the outer membrane of Gram-negative bacteria (Schadlich et /. Vaccine 27, 1 11-1522 (2009)). For example, transformed BL21 bacteria may be grown in LB medium containing ImM ampiciliin and incubated with shaking at 200 rpm at 37 'C. At an optical density (ODgoo nm) of 0.3-0.5, bacteria can be cooled down and IPTG may be added to induce protein expression. After 16-18 h bacteria may be harvested by centrifugation.
Bacteria may be lysed by homogenizing, lysates may be cleared, capsid proteins purified and LPS contamination removed, using e.g., chromatographic methods, such as affinity chromatography and size exclusion chromatography. LPS contamination may also be removed using e.g., 1% Triton X-l 14, [0077], In certain embodiments, VLPs are loaded with the one or more therapeutic agents. After isolation of LI and L2 capsid proteins which may be in the form of monomers or oligomers, VLPs may be assembled and loaded by disassembling and reassembling LI or LI and L2 viral capsid proteins, as described herein. Salts that are useful in aiding
disassembly/reassembly of viral capsid proteins into VLPs, include Zn, Cu and Ni, Ru and Fe salts. In some embodiments, VLPs may be loaded with one or more therapeutic agents.
[0078]. Loading of VLPs with agents utilizing a disassembly-reassembly method has been described previously, for example in U.S. Patent Nos: 6,416,945 and WO 2010/120266, incorporated herein by reference. Generally, these methods involve incubation of the VLP in a buffer comprising EGTA and DTT, Under these conditions, VLP completely disaggregated into structures resembling capsid proteins in monomeric or oligomeric form, A therapeutic or diagnostic agent, as described herein, may then be added and the preparation diluted in a buffer containing DMSO and CaCl2 with or without ZnCl2 in order to reassemble the VLP. The presence of ZnCl2 increases the reassembly of capsid proteins into VLP. In some embodiments, one or more of these reassembly methods may be used to assemble capsid proteins to form VLPs that encapsulate one or more agents without requiring an initial VLP disassembly procedure, as described herein.
[0079]. In certain embodiments, VLP are loaded with the one or more therapeutic agents. After isolation of LI and L2 capsid proteins, these may mixed directly after purification from the host cell with the therapeutic agent and reassembled into loaded VLPs as described herein, the preparation diluted in a buffer containing DMSO and CaCl2 with or without ZnCl2 in order to reassemble the VLP. The presence of ZnCl2 increases the reassembly of capsid proteins into VLP.
[0080]. Surprisingly, it was found that certain ratios of a) Capsid protein to reaction volume, b) agent to capsid protein, and/or c) agent to reaction volume lead to agent-loaded VLP (VLP comprising entrapped agent) that exhibit superior delivery of agent to target cells when compared to agent-loaded VLP prepared using previously described methods. VLP loaded with agents using the methods described herein, in certain embodiments, are able to deliver agent to 65%, 75%, 85%, 95%, 96%», 97%, 98%, or 99% of target cells. One non imiting example of the improved method is exemplified in the Examples. [0081 ], For example, VLP may be loaded with a nucleic acid using a method comprising: a) contacting a preparation of capsid proteins with the nucleic acid in a reaction volume, wherein i) the ratio of capsid protein to reaction volume ranges from 0. 1 μg capsid protein per 1 μΐ reaction volume to 1 μ§ capsid protein per 1 μΐ reaction volume; ii) the ratio of nucleic acid to capsid protein ranges from 0.1 μg nucleic acid per 1 p.g capsid protein to 10 p.g nucleic acid per 1 μg capsid protein; and/or iii) the ratio of nucleic acid to reaction volume ranges from 0.01 μg nucleic acid per 1 μΐ reaction volume to 10 g nucleic acid per 1 μΐ reaction volume, and b) reassembling the capsid proteins to form a VLP, thereby
encapsulating the nucleic acid within the VLP. In other embodiments, the ratio of HPV- capsid protein to reaction volume ranges from 0.2 μg HPV-capsid protein per 1 μΐ reaction volume to 0.6 μg HPV-capsid protein per 1 μΐ reaction volume. In yet other embodiments, the ratio of nucleic acid to HP V-capsid protein ranges from 0.5 μg nucleic acid per 1 ug HPV-capsid protein to 3.5 μ§ nucleic acid per 1 μ¾ HPV-capsid protein. In yet other embodiments, the ratio of nucleic acid to reaction volume ranges from 0.2 μ¾ nucleic acid per 1 μΐ reaction volume to 3 μg nucleic acid per I μΐ reaction volume.
[0082], The step of dissociating the VLP or capsid protein oligomers can be carried out in a solution comprising ethylene glycol tetraacetic acid (EGTA) and dithiothreitol (DTT), wherein the concentration of EGTA ranges from 0,3 mM to 30 mM and the concentration of DTT ranges from 2 mM to 200 mM. in certain embodiments, the concentration of EGTA ranges from 1 mM to 5 mM. In certain embodiments, the concentration of DTT ranges from 5 mM to 0 mM.
[0083], The step of reassembling of capsid proteins into a VLP can be carried out in a solution comprising dimethyl sulfoxide (DMSO), ί aCL and ZnC , wherein the
concentration of DMSO ranges from 0.03% to 3% volume/volume, the concentration of CaCl2 ranges from 0.2 mM to 20 mM, and the concentration of ZnCL ranges from 0.5 μ,Μ to 50 μΜ. In certain embodiments, the concentration of DMSO ranges from 0.1% to 1% volume/volume. In certain embodiments, the concentration of ZnCL ranges from 1 μ,Μ to 20 μΜ. In certain embodiments, the concentration of CaCl2 ranges from 1 mM to 10 mM.
[0084]. In certain embodiments, the loading method is further modified to stabilize the VLP, in thai the loading reaction is dialyzed against hypertonic NaCl solution (e.g., using a NaCl concentration of about 500 mM) instead of phosphate-buffered saline (PBS), as was previously described. Surprisingly, this reduces the tendency of the loaded VLP to form larger agglomerates and precipitate. In certain embodiments, the concentration ofNaCl ranges between 5 mM and 5 M. In certain embodiments, the concentration of NaCl ranges between 20 mM and 1 M.
[0085], In some embodiments, VLPs can be loaded with one or more therapeutic agents according to the methods described herein. In certain embodiments the therapeutic agent is a nucleic acid molecule capable of inducing RNA interference or a nucleic acid (e.g. , plasmid or other vector) that is capable of expressing a nucleic acid molecule capable of inducing RNA interference. In some embodiments, VLP can be loaded with combinations of two or more therapeutic or diagnostic agents,
[0086], In some embodiments, the therapeutic agent is an inducer of RNA interference or other inducer of gene silencing. An inducer of RNA interference may be a siRNA, a shRNA, a hybrid nucleic acid molecule comprising a first part that comprises a duplex ribonucleic acid (RNA) molecule and a second part that comprises a single stranded deoxyribonucleic acid (DNA) molecule, a longer double-stranded RNA or a DNA construct for expression of siRNA or longer RNA sequences. Other inducers of gene silencing include inducers of DNA methylation, or rihozymes, or aptamers. In other embodiments, the therapeutic agent can be a modulator of gene expression such as a PNA (Peptide Nucleic Acid).
[0087]. RNA interference (RNAi) is a process whereby the introduction of double stranded RNA (dsRNA) into a cell inhibits gene expression post-transcriptionally, in a sequence dependent fashion, RNAi can be mediated by short (for example 19-25 nucleotides) dsRNAs or small interfering R'NAs (siRN A). dsRNA is cleaved in the cell to create siRNAs that are incorporated into an RNA-induced silencing complex (RISC), guiding the complex to a homologous endogenous mRNA, cleaving the mRNA transcript, and resulting in the destruction of the mRNA.
[0088], To induce RNA interference in a cell, dsRNA may be introduced into the cell as an isolated nucleic acid fragment or via a transgene, plasmid, or virus. In certain embodiments, VLP are used to deliver dsRNA to the target cells,
[0089]. In some embodiments, a short hairpin RNA molecule (shRNA) is expressed in the cell A shRNA comprises short inverted repeats separated by a small loop sequence, One inverted repeat is complimentary to the gene target. The shRN A is then processed into an siRNA which degrades the target gene mRNA. siiRNAs can produced within a cell with a DNA construct encoding the shRNA sequence under control of a RN A polymerase III promoter, such as the human HI, U6 or 7SK promoter. Alternatively, the shRNA may be synthesized exogenous!}' and introduced directly into the cell, for example through VLP deliver}'. In certain embodiments, the shRNA sequence is between 40 and 100 bases in length or between 40 and 70 bases in length. The stem of the hairpin are, for example, between 19 and 30 base pairs in length. The stem may contain G-U pairings to stabilize the hairpin striscture.
[0090]. siRNA sequences are selected on the basis of their homology to the target gene. Homology between two nucleotide sequences may be determined using a variety of programs including the BLAST program (Altschul et al (1990) J. Mol. Biol. 215: 403-10), or BestFit (Genetics Computer Group, 575 Science Drive, Madison, Wisconsin, USA., Wisconsin 53711). Sequence comparisons may be made using FAST A and FASTP (see Pearson & Lipman, 1988. Methods in Enzymology 183: 63-98). Tools for design and quality of siRNAs, shRNAs and/or miRNAs are known in the art. Web-based online software system for designing siRNA sequences and scrambled siRNA sequences are for example siDirect, siSearch, SEQ2SVM, Deqor, siRNA Wizard (invivoGen). The specificity can be predicted using for example SpecificityServer, miRacle. Target sequences can be researched for example at HuSiDa (Human siRNA Database), and siRNAdb (a database of siRNA sequences). Sequence comparison may be made over the full length of the relevant sequence, or may more preferably be over a contiguous sequence of about or 10,15, 20,25 or 30 bases. In certain embodiments, the degree of homology between the siRNA and the target gene is at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, or at least 99%, or 100%·. The siRN A may be between lObp and 30bp in length, or between 20bp and 25bp, or the siRNA is 20, 21 or 22bp in length.
[0091]. The occurrence of RNAi can be detected by transfecting cultured cells with the siRNA, followed by RT-PCR of the mRNA of interest. Where R NAi is induced by the siRNA, levels of the mRNA of interest will be reduced in traiisiected ceils as compared to control cells. A reduction in protein production can be confirmed by Western blotting of cell lysates followed by probing with an antibody reactive to the protein of interest,
[0092]. siRNA molecules may be synthesized using standard solid or solution phase synthesis techniques which are known in the art. [0093], In some embodiments, the siRNA has an overhang at one or both ends of one or more deoxythymidme bases to increase the stability of the siRN A within cells by reducing its susceptibility to degradation by nucleases,
[0094]. In some embodiments, the siRNA is a hybrid nucleic acid molecule comprising a first part that comprises a duplex ribonucleic acid (RNA) molecule and a second part that comprises a single stranded deoxyribonucleic acid (DNA) molecule. Preferred targets for the RNA interference would include disease causing genes e.g. oncogenes, inflammatory genes, regulatory genes, metabolic genes, viral genes, in one embodiment of the in vention the target genes would be E6 & E7 HPV viral oncogenes (e.g., E7 siRNA), In other embodiments of the invention the target genes would be p53, Sirt-1, survivin, EGFR, VEGFR, VEGF, CTNNB1 or other oncogenes.
[0095], Linkages between nucleotides may be phosphodiester bonds or alternatives, for example, linking groups of the formula P (O) 8, (thioate) ; P (S) 8, (dithioate); P (O) NR'2 ; P (O) R' ; P (O) OR6; CO ; or CONR'2 wherein R is H (or a salt) or alkyl ( 1-12C) and R6 is alkyl (1-9C) is joined to adjacent nucleotides through-O-or-S-.
[0096]. Modified nucleotide bases can be used in addition to the naturally occurring bases, For example, modified bases may increase the stability of the siRNA molecule, thereby reducing the amount required for silencing, The term modified nucleotide base encompasses nucleotides with a covaiently modified base and/or sugar. For example, modified nucleotides include nucleotides having sugars which are covaiently attached to low molecular weight organic groups other than a hydroxy! group at the 3'position and other than a phosphate group at the 5 'position. Thus modified nucleotides may also include ^substituted sugars such as 2'- 0-methyl- ; 2-0- alkyl : 2-0-allyl; 2'-S-alkyl; 2'-S-allyl ; 2'-fiuoro- ; 2'-halo or 2; azido-ribose, carbocyclic sugar analogues a-anomeric sugars; epimeric sugars such as arabinose, xyloses or lyxoses, pyranose sugars, furanose sugars, and sedoheptulose.
[0097]. Modified nucleotides are known in the art and include alkylated purines and pyrimidines, acylated purines and pyrimidines, and other heterocycles. These classes of pyrimidines and purines are known in the art and include pseudoisocytosine, N4, N4- ethanocytosine, 8-hydroxy-N6-methylademne, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5 fluorouracil, 5-bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, 5- carboxymethylammomethyl uracil, dihydrouracil, inosine, N6~ isopentyl-adenine, 1- methyladenine, l-methylpseudouracil, 1- methylguanine, 2,2-dimethylguanine, 2methylad nine, 2- methylguanine, 3-methylcytosine, 5-methylcytosine, N6- methyladenine, 7-methy [guanine, 5-methylammomethyl uracil, 5- methoxy amino methyl-2-thiouracil, -D- mannosylqueosine, 5- methoxycarbonyimethyluracil, 5methoxyuracil, 2 methylthio-N6- isopentenyladenine, uracil-5-oxyacetic acid methyl ester, psueouracil, 2-thiocytosine, 5- methyl-2 thiouracil, 2- thiouracil, 4-thiouracil, Smethyiuracil, N-uracil-5-oxyacetic acid methylester, uracil 5-oxyacetic acid, queosine, 2- thiocytosine, 5-propyluracil, 5- propylcytosine, 5-ethyluracil, Sethylcytosine, 5-butyluracil, 5-pentyluracil, 5-pentylcytosine, and 2,6, diaminopurine, methylpsuedouracil, 1 -methylguanine, 1 - methyl cytosine.
[0098]. In some embodiments, siRNA molecules or longer dsRNA molecules may be made recombinantly by transcription of a nucleic acid sequence, for example contained within a vector as described herein. The vector may be any RNA or DNA vector,
[0099]. The vector can be an expression vector, wherein the nucleotide sequence is operably linked to a promoter compatible with the cell. Promoters suitable for use in various vertebrate systems are well known in the art. For example, suitable promoters include viral promoters such as mammalian retrovirus or DN A virus promoters, e.g., MLV, CMV, RSV, SV40 IEP (immediate early promoter) and adenovirus promoters and metallothionein promoter. Strong mammalian promoters may also be used. It will be appreciated that variants of such promoters retaining substantially similar transcriptional activities may also be used.
[00100], In some embodiments, the vector may have at least two promoters, one to direct expression of the sense strand and one to direct expression of the antisense strand of the dsRNA. In other embodiments, two vectors may be used, one for the sense strand and one for the antisense strand. Alternatively the vector may encode RNAs which form stem-loop structures which are subsequently cleaved by the cell to produce dsRNA,
[00101], The nucleic acid construct may contain a specific cellular, viral or other promoter or repressor of gene expression. The promoter or repressor may be designed to reflect the context of the cell into which the constract is introduced. For example, the construct may contain a viral promoter so expression from the construct is dependent upon the presence of a viral protein, so that the construct is expressed only in viral-infected cells. Similarly, the construct may have a promoter or repressor specific to certain cell types or to certain developmental stages. For example, where the vector is for use in viraliy infected cell such as cells infected with HPV, a viral promoter whic matches the disease-causing vims should be used, e.g., a HPV promoter (such as the promoter causing expression of HPV E6/E7) for HPV- infected ceils. In suc embodiments, the vector will only be expressed in the virally- infected cells.
[00102], Nucleic acids are highly charged and do not cross ceil membranes by free diffusion. The hydrophilic character and anionic backbone of nucleic acids such as, for example, siRNAs reduces their uptake by the cells, in certain embodiments, nucleic acids (e.g., siRNA) may be loaded into VLP (e.g., HPV-VLP) to efficiently deliver them to a subject through
administration of V LP, In certain embodiments, encapsulating the nucleic acid into a V LP increases cellular uptake, allows traversal of biological membrane barriers in vivo, and/or increases the bioavailability of the nucleic acid (e.g., siRNA).
[00103], in some embodiments, the agent loaded into the VLP is an anti-viral agent, in certain embodiments, the agent is an anticancer agent, In a preferred embodiment, the anticancer agent is a taxane,
[00104], In certain embodiments, the therapeutic agent that may be loaded into a VLP using the methods described herein is a chemotherapeutic agent, for instance, methotrexate, vincristine, adriamycin, cisplatin, non-sugar containing chloroethylnitrosoureas, 5- fiuorouracil, mitomycin C, bleomycin, doxorubicin, dacarbazine, taxol, fragyline, Meglamine GLA, vairubicin, carmustaine and poliferposan, MMI270, BAY 12-9566, RAS famesyl transferase inhibitor, famesyl transferase inhibitor, MiV!P, MTA/LY231514,
LY264618 Lometexol. Glamolec, CI-994, TNP-470, Hycamtin/Topotecan, PKC412, Valspodar/PSC833, Novantrone/Mitroxantrone, Metaret/Suramin, Batimastat, E7070, BCH- 4556, CS-682, 9-AC, AG3340, AG3433, Incel VX-710, VX-853, ZD0101, ISI641, ODN 698, TA 2516/Marmistat, BB2516/Marmistat, CDP 845, D2163, PD183805, DX8951f, Lemonal DP 2202, FK 317, Picibanil/OK-432, AD 32/Valrubicin, Metastron/strontium derivative, Temodal/Temozolomide, Evacet liposomal doxorubicin, Y ewtaxan Paclitaxel, Taxol/Paclitaxel, Xeload/Capecitabine, Furtulon/Doxifluridine, Cyclopax/oral paclitaxel. Oral Taxoid, SPU-077/Cisplatin, HMR 1275/FlavopiridoL CP-358 (774)/EGFR, CP-609 (754)/RAS oncogene inhibitor, BMS- 182751 /oral platinum, UFT(Tegafur/XJracil),
Ergamisol/Levamisole, Eniiuracil/776C85/5FU enhancer, Campto/Levamisole,
Camptosar/Irinotecan, Tumodex/Ralitrexed, Leustatin/Cladribine, Paxex/Paclitaxel, Doxil/liposomal doxorubicin, Caelyx/liposomal doxorubicin, Fludara/Fludarabine,
Pharmarubicin/Epirubicin, DepoCyt, ZD 1839, LU 79553/Bis-Naphtalimide, LU
103793/Dolastam, Caetyx/liposomal doxorubicin, Gemzar/Gemcitabine, ZD 0473/Anormed, Y 1 16, Iodine seeds, CDK.4 and CDK2 inhibitors, PARP inhibitors, D4809/Dexifosamide, Ifes/Mesnex/Ifosamide, Vumon/Teniposide, Parapiatin/Carboplatin, Plantinol/cisplatin, Vepeside/Etoposide, ZD 9331, Taxotere/Docetaxel, prodrug of guanine arabinoside, Taxane Analog, nitrosoureas, alkylating agents such as melphelan and cyclophosphamide,
Aminogiutethimide, Asparaginase, Busulfan, Carboplatin, Chlorambucil, Cytarabine HCI, Dactinomycin, Daunorubicin HCI, Estramustine phosphate sodium, Etoposide (VP 16-213), Floxuridine, Fluorouracil (5-FU), Flutamide, Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alfa-2a, Alfa-2b, Leuprolide acetate (LHRH-releasing factor analogue),
Lomustine (CCNU), Mechlorethamine HCI (nitrogen mustard), Mercaptopurine, Mesna, Mitotane (o.p'-DDD), Mitoxantrone HCI, Octreotide, Plicamycin, Procarbazine HCI, Streptozocin, Tamoxifen citrate, Thioguanine, Thiotepa, Vinblastine sulfate, Amsacrine (m~ AMSA), Azacitidine, Erthropoietin, Hexamethylmelamine (HMM), Interleukin 2,
Mitoguazone (methyl-GAG; methyl glyoxal bis-guanylhydrazone; MGBG), Pentostatin (2 ' deox coform cin), Semustine (methyl-CCNU), Teniposide (VM-26) or Viiidesine sulfate, but it is not so limited.
[00105], In certain embodiments, the therapeutic agent that may be loaded into a VLP using the methods described herein is an immunotherapeutic agent, for instance, Ributaxin, Herceptin, Quadramet, Panorex, IDEC-Y2B8, BEC2, C225, Oncoiym, SMART Ml 95, ATRAGEN, Ovarex, Bexxar, LDP-03, ior t6, MDX-210, MDX-1 1, MDX-22, OV103, 3622W94, anti-VEGF, Zenapax, MDX-220, MDX-447, MELIMMUNE-2, MELIMMUNE- 1, CEACIDE, Pretarget, NovoMAb-G2, TNT, Giiomab-H, GNI-250, EMD-72000,
LymphoCide, CMA 676, Monopharm-C, 4B5, ior egf r3, ior c5, BABS, anti-FLK-2, MDX- 260, ANA Ab, SMART 1D10 Ab, SMART ABL 364 Ab or IrrunuRAIT-CEA, but it is not so limited.
[00106]. In certain embodiments, the therapeutic agent that may be loaded into a VLP using the methods described herein is an antiviral agent. Examples of anti-viral agents are:
Polysulfates (PVAS), Polysulfonates (PVS), Polycarboxylates, Polyoxometalates, Chicoric acid, zintevir, cosalane derivatives, Bicyclams (i.e., AMD3100), T-22, T-134, ALX-40-4C, CGP-64222, TAK-779, AZT (azidothymidine), ddl, ddC, d4T (didehydrodideoxythymidine), 3TC (3'-thiadideoxycytidine), ABC, and other ddN (2',3'-dideoxynucleoside) analogs, Nevirapine, delavirdine, efavirenz, emivirine (MKC-442), capravirine, thiocarboxanilide UC- 781 , acyclovir, valaciclovir, penciclovir, famciclovir, bromovinyldeoxyuridine (BVDU, brivudin), Cidofovir, Adefovir dipivoxil, Tenofovir disoproxil, Ribavirin, valacyclovir, ganciclovir, formivirsen, foscarnet, EICAR (5-ethytiyl- 1 - P-D-ribofuranosyiimidazole-4- carboxamide), Mycophenolic acid, Neplanocin A, 3-deazanepla.nocin A, 6 -C- methylnepianocin A, DHCeA (9-(/m/¾s,-2Vn¾«s-3'-dihydroxycyclopent-4'-enyl)adenine), or c3DHCeA (9-(rr «5-2V-r i^is'-3'-dihydroxycyi^opent-4'-enyl)-3-deazaadenine), as described, for example, in De Clercq J Pharmacol Exp Ther 2001 , 297: 1-10, incorporated by reference herein, but it is not so limited.
[00107]. Aspects of the invention are not limited in its appl ication to the detail s of construction and the arrangement of components set forth in the preceding description or illustrated in the examples or in the drawings. Aspects of the invention are capable of other embodiments and of being practiced or of being carried out in various ways. Also, the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of "including," "comprising," or "having,''' "containing," "involving," and variations thereof herein, is meant to encompass the items listed thereafter and equivalents thereof as well as additional items.
[00108]. EXAMPLES
[00109]. EXAMPLE 1
[001 10]. Production and purification of capsid proteins in host cells and in vitro reassembly into VLPs
[001 1 1], Suspension cultures of Sf9 insect cells were maintained in serum- free Sf-900™ II medium (Invitrogen, Lide Technologies) and expanded from shake flasks to WAVE bioreactors™ (GE Healthcare Lifesciences). Approximately 2L of shake flask culture was utilized to seed the 10L WAVE bioreactors™ at an initial density of 4x105 cells/ml.
[001 12]. Once the actively growing culture reached a density between 1 .5 - 2 x 1.0° cells ml, it was infected with a recombinant baculoviras stock for HPV 16L 1 or HPV 16/3 1 mutant and a HPV1 6L2 at an MOI of 5. Recombinant baculoviras stocks were produced, as described herein (Table 1). [00113]. According the present invention, an overview of an exemplary protocol for generating Baculovirus generation and preparing a higti-titer stock preparation is described as follows. Transform DHlOBac Competent Cells with pFastBac construct and heat shock the mixture. Serial dilute the cells using SOC medium to 1 : 10, 1 : 100 andhlOOO dilutions. Grow cultures for 4 hours at 37C at 250 rpm. Streak the 1 : 10, 1 : 100 and 1 : 1000 dilutions onto selective plates of LB-Agar/Kan/Tet/Gent/X-ga /PTG. Incubate plates for 48 hours at 37C. Select three white colonies. Grow each culture O/N at 37C at 250 rpm in LB plus Kan, Gent. & Tel. Harvest cell pellets by centrifugation and Isolate recombinant Bacmid by alkaline lysis method. Determine Bacmid concentration by 260:280. Tranfect Sf9 cells with
Bacmid/cellfectin complex and plate. Incubate plates for four days in a humidified 27C tissue culture incubator. Transfer conditioned media to 30ml SF 8f9 culture. Grow culture 3-5 days. Monitor for cell viability and cell diameter using Vi-Cell. Harvest conditioned media and cell pellet when viability is less than 75%. Perform titer (BacPA RapidTiter Kit) and Western Plot analysis. Expand recombinant virus by infecting a 1 L, culture of Sf9 ceils at an MOI of 0.1 with the best expressing Baculovirus clone. Harvest conditioned media by centrifugation once viability has dropped less than 75%. Perform titer analysis using
RapidTiter Kit.
[001 14]. To generate the recombinant baculovirus for HPV 16/31 LI production, the pFastBac™ plasmid (Invitrogen, Life Technologies) (FIG. 1) containing 16/31 LI DNA sequence (SEQ ID NO: 1) was used. To generate the recombinant baculovims for HPV16L2 production, the pFastBac™ plasmid containing L2 DNA sequence (SEQ ID NO: 2) was used. During recombinant protein production, the bioreactor was monitored daily for cell count, viability, ceil size and pH. Seventy-two hours post-infection, the cell pellet was obtained by tangential- flow filtration, washed in PBS, re-pelleted by centrifugation, and stored at -80°C. Western blot using protein-specific antibodies for LI and L2 proteins were then used to verify the presence of the recombinant protein.
[001 15]. Following verification of expression, purification of HPV capsomeres produced above was performed. Cells were thawed on ice and then resuspended in ice-cold lysis buffer (PBS plus 0.5% Nonidet™ P-40 (Shell Chemical Co.)) at a ratio of 10 ml of buffer per gram of cell Table 1
Figure imgf000024_0001
Use pFastbac Dual construct generated at DNA2.0 to transform DHl OBac cells by heat shock method (i.e. Ing, pFactbac construct in 100 ul of cells. Incubate for 30 minutes on ice. Heat at 42C for 45 seconds. Chill on ice for two minutes). Grow cultures at 37C, 225 rpm in SOC media for four hours. Prepare 1 ; 10, 1 : 100, and 1 : 1000 dilutions of culture. Plate dilutions on
Bac-to-Bac selective plates. Incubate plates at 37C for two. days.
Purify Recombinant Bacmid
Select three well defined white colonies from the Bac-to-Bac selective plates and culture the cells in selective LB media overnight. Collect bacterial ceils by centrifugation (14Kxg. 3 minutes). Resuspend cell pellets in PI buffer. Lyse cells by the addition of an equal volume of P2 buffer. Incubate at room temperature for five minutes. Precipitate genomic D A and protein by addition of a half colume of P3 bugger and incubation on ice for five minutes. Remove precipitated contaminants by centrifugation (14K x g; 10 minutes) and reserve supernatant. Precipitate the bacmid by addition of an equal volume of Isopropanol followed by an overnight incubation at 20C. Pellet bacmid by centrifugation. Wash pelleted bacmid with 70% ethanol. Let pellet air dry. Resuspend pellet in TE. Determine yield and purity by
OD260-OD280.
Transfect Sf Cells With Recombinant Bacmid
For each bacmid prepare a 6-well plate with Ix20e6 cells per well in standard growth media
(i.e. Sf-900 II). Allow cells to attach to the plate for at least 1 hour. In a BSC, prepare bacmid Cellfectin complex by mixing 1 ug of bacmid that has been diluted with 100 ul of Grace's media with 6ul of cellfectin transfection reagent that has been diluted with 100 ul of Grace's media. Let complexes form for 30 minutes at room temperature. Remove media from the cells in upper left comer well, dilute bacmid cellfectin complex with SOOul of Grace's media, add transfection solution to the upper left corner well. Place plates into a humidified incubator at 27C. After five hours, remove transfection solution from the cells in the upper left corner well and add 2ml of growth media (i.e. Sf-900 II). Return plates to the humidified incubator. Check cells daily under a microscope to confirm transfection (cells should not grow as fast as control cells and should increase in diameter, and eventually the cells should show signs of lysing). After four days, harvest P0 viral stock (i.e. conditioned media from upper left comer well).
Ampllfy po BacuIoviraI stock:
For each baculo viral stock, add I ml of the PO viral stock to a 30ml culture in a 125ml shake flask of Sf9 cell at a cell density of le6 cells/ml. An additional SF is utilized as a negative control and lml of growth media added, Shaking incubator parameters are 120 rpm and 27.5C. Cultures are monitored daily with the Vi-Cell for cell density, cell viability, and diameter. In a proper infection, within 48 hours the insect ceil culture should have significantly lower cell density and cell viability and increased cell diameter. Cultures are maintained for three to five days and harvested by centrifugation (2500 x g, 10 minutes) once viability has dropped belo 75%. Transfer the conditioned media (PI) viral stock to a fresh tube and store at 4C. Reserve cell pellet for Western analysis. Determine titer for the pi viral stock using the Clontech BacPAK Rapid Titer Ket according to manufacturer's protocol.
Expand PI Baculoviral Stock
For the best expressing baculoviral stock (i.e. Western Analysis), add 1.5e8 pfu of PI viral stock to a II. cultureof Sf9 ceils in a3L Shake Flask at 1.5e6 ceils per ml (i.e. MOI of 0.1 ). Shaking incubator parameters are 120 rpm and 27.5C. Cultures are monitored daily with Vi- Cell for cell density, cell viability, and cell diameter. Cultures are maintained for two to five days and harvested by centrifugation (2500 x g, 10 minutes) once viability has dropped below 75%. Transfer the conditioned media (PI) viral stock to a fresh sterile bottle and store at 4C. Determine titer for the P2 viral stock using the Clontech BacPAK Rapid Titer Kit according to manufacturer's protocol.
paste. Resuspended cells were then incubated on ice for 15 min. After chemical lysis, nuclei were isolated by centrifugation (3000 x g for 15 min) and then resuspended in ice-cold PBS without detergent. Capsid proteins were then solubilized from the isolated nuclei with three 15 s bursts of a somcator at 50% maximal power. Insoluble material was then clarified by centrifugation (1000 x g for 10 min) and the resulting supernatant was diafiltered into TMAE: buffer by TFF using a 100 kDa molecular weight cut-off filter. Western Blot was used to demonstrate that the majority of the capsid proteins was localized in the nuclear fraction. (FIG. 2)
[001 16]. Capsid proteins were then loaded onto a TMAE column, washed, and eluted using a linear salt gradient. Early fractions containing the proteins of interest were then pooled, dialyzed into disassociation buffer, and concentrated to a final concentration of lmg/ml.
[001 17]. Purified capsid proteins were then assembled in a cell free system together with a plasmid (pENTR™/XJ6 plasmid (Invitrogen, Life Technologies)) expressing an shRNA construct containing the short hairpin R A sequence generated using primer sequences (SEQ ID NO: 3 and SEQ ID NO: 4) to create VLP encapsulating the shRNA using the following loading protocol.
[001 18]. Loading Protocol
[001 19]. In a clean 15 ml conical tube the following reagents were added and incubated at 37°C for 30 min: 200 iig of capsomere protein; 100 g pENTRTM/U6/shRNA plasmid; 0.5 μΐ DMSO; and 15 μΐ Solution 2 (1 0 niM Tris-HCl pH7.5, 450 mM NaCL 330 μΐ df !,< )).
brought up to a total volume of 1 50 μΐ.
[00120]. Solution 3 (2 mM ( aCk 5 μΜ CaCL, 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 434 iL dH20) was then added to the above mixture and incubated at 37°C for 30 min.
[00121], Solution 4 (4 mM CaCi2, 10 μΜ CaCL, 50 mM Tris-HCl pH 7.5, 150 mM NaCL 1224 μΐ dH20) was then added to the above mixture and incubated at 37°C for 2 hrs.
[00122], The mixture was then dialyzed in IX PBS at 4°C overnight.
[00123]. EXAM PLE 2
[00124], Production of mutant L I * and L2 capsid proteins in mammalian cell system [00125]. Similarly to Example 1 described above, a mammalian culture system is used to produce mutant LI * (16/31 ) and L2 capsid proteins. Plasmids containing human-optimized codon sequences are used for this purpose (SEQ ID NO: 5), and a general protocol is followed (Buck, C. B., et ai. (2005) Methods MoL Pied., 119: 445-462, which reference is incorporated herein).
[00126]. EXAMPLE 3
[00127], Assembly into VLPs from capsid proteins
[00128]. Capsid proteins isolated from insect cells were assembled into VLPs as described. Dynamic light scattering (DLS) demonstrates presence of capsid proteins in moiiomeric and oligomeric forms (<1 Onm) after harvest and prior to the loading procedure. After the reassembly in presence of the nucleic acid payload, VLPs are seen by DLS (50-70nm diameter) (FIG. 3).
[00129]. EXAM PLE 4
[00130], Functional transfer of luciferase expression
[00131], Results show functional transfer of luciferase expression. VLPs were generated using different production methods to compare efficacy. Transfection of luciferase plasmid (pClucF) using standard iipofectamine transfection at various plasmid amounts (O.lng well, 1 tig/well, lOng/weil) was used to create a range of positive controls. lOng of pClucF plasmid was used without transfection reagent as a reagent/background control.
[00132]. AB1-2 refers to HPV16L1L2 VLP generated using the methods described above, where a single plasmid like p!6sheLL (SEQ ID NO: 6) was used to co-express wildtype HPV L I and 1.2 proteins.
[00133]. Capsid proteins were purified, as described above, from 293 cells transfected with the co-expression plasmid for LI and L2. Capsid proteins were then subjected to the following loading protocol, thereby forming loaded VLP.
[00134]. Loading Protocol
[001.35], In a clean .1 ml conical tube the following reagents were added and incubated at 37°C for 30 min: 200 ug of capsid proteins, 100 ^ig pClucF, 0.5 μΐ DMSO, 15 μΐ Solution 2 (150 mM Tris-HCl pH7.5, 450 mM NaCL 330 μΐ dH20), brought up to a total volume of 150 μΐ
[00136], Solution 3 (2 mM CaCl2, 5 μΜ CaCl2, 50 mM Tris-HCl pH 7.5, 150 mM NaCL 434 iL d¾0) was then added to the above mixture and incubated at 37°C for 30 min.
[00137]. Solution 4 (4 mM CaCi2, 10 μΜ CaCi2, 50 mM Tris-HCl pH 7.5, 150 mM Nad, 1224 μΐ dH20) was then added to the above mixture and incubated at 37°C for 2 hrs.
[00138], The mixture was then dialyzed in IX PBS at 4°C overnight.
[00139], Loaded VLP were then used to treat Hela cells in 96 well plates and luciferase signal was read after 48hrs (Table 2, FIGS. 4 and 5).
[00140], AB iuc3 and AB luc4 were produced in 293 cells after transfection with the l6sheLL plasmid as pseudovirions (PSV) already encapsulating the payload plasmid (pi iucL) (Buck, C, B., et al. (2005) Methods Mol Med., 119: 445-462). Results showed superior transfer of plasmid when the reassembly loading method was used (AB 1 -2) compared with VLPs that were loaded through packaging of plasmid in the host cells (AB luc 3 and AB luc 4).
[00141 ]. Table 2
Figure imgf000029_0001
AB luc3 5769.104355 1 178.278814
AB luc 4 5487.777321 1115.096887 pClucF 1 .639379622 0.218550273
[00142]. Table 3
Figure imgf000030_0001
Grace's Insect Cell Culture Gibco 11595-030 Medium
BacPak Rapid Titer Kit Clontech 631406
Mouse anti-6XHis antibody Clontech 631212
Qdot 800 goat anti-mouse igG Invitrogen Q1107 P conjugate
Acetone J.T.Baker 9002-03
Formaldehyde VWR VW3408-1
Dimethylformamide Sigma-Aidrich 319937
[00143], Table 4
Item Manufacturer/Model Equipment #
Microbial Biosaiety Cabinet Forma Scientific/ 1184 PB0138
Shaking Microbial Incubator NBS/PsycroTherm PB0045
Microcentrifuge Eppendorf/5415D PB0159
UV/Vis Spectrophotometer Agilent 8453 PB0090
Insect Biosafety Cabinet Baker Co./ SterilGARD III 5007-0000
Humidified Incubator Forma S ci entific/3326 PB0013
Microscope Olympus/ 1X70 PB0075
Shaking Insect Incubator NBS/Innova 4000 PB0044
Cell Analyzer Beckman Coulter/Vi-Cell XR PB0085
Table Top Centrifuge Beckman Allegra X-15R PB0160
Western Imaging Station Li-Cor/Odyssey PB0073
[001441. EXAMPLE 5 [00145]. Topical Therapeutic Treatment Of Alopecia With BetaHPV Pseudovirions
[00146]. Alopecia means loss of hair from the head or body and includes baldness, a term generally reserved for pattern alopecia or androgenic alopecia (AG A). AGA is the most common cause of hair loss in humans, both male and female and its incidence is increasing 10% per decade. Researchers have determined that pattern hair loss is related to hormones called androgens, particularly an androgen called dihydrotestosterone (DHT). Men with androgenic alopecia typically have higher levels of 5-aipha-reductase, lower levels of total testosterone, higher levels of unbound/free testosterone, and higher levels of total free androgens including DHT (Denmark- Wahnefried 1997). 5-aipha-reductase is responsible for converting free testosterone into DHT. The genes for 5-alpha-reductase are known (Ellis 2005), The enzymes are present predominantly in the scalp and prostate. Levels of 5-alpha- reductase are one factor in determining levels of DHT in the scalp and drugs which interfere with 5-alpha-reductase (such as Finasteride, which inhibits the predominant type 2 isoform) have been approved by the FDA as treatments for hair loss.
[00147], The adverse side effects of the Minoxidil dosing include scalp irrita tion and allergic contact dermatitis. The side effects associated with the DHT inhibitor, Finasteride, which when applied topically has very little penetration are more serious. The compounds and formulas of U.S. Patent No. 6,268,377 which is hereby incorporated by reference, for the treatment of hyperandrogenic condition, include the 5-alpha-reductase inhibitors and calcium channel blocking agents which may be combined in a single oral, topical, systemic or parenteral dosage formulation. The composition has a number of side effects that include: erectile dysfunction (ED) or impotence in up to 18.5 percent of people, decreased libido up to 10 percent, abnormal ejaculation, including decreased ejaculate amount, up to 7.2 percent, and breast enlargement up to 2.2 percent. .
[00148], Propecia blocks the fomiation of DHT which is intended to inhibit the development of male pattern baldness. However, the ineffectiveness of Propecia in treating male patterned baldness and the adverse effects associated with the currently prescribed method of dosing and treating male pattern baldness indicate that a more effective treatment is needed.
[00149], In accordance with a preferred embodiment of the present invention, a preferred method for treating alopecia preferably includes using a combination of betapapillomavirus viral shells (L1/L2) to deliver a DHT inhibitor as a therapeutic agent. In accordance with the present invention, the method preferably includes the steps of constructing a recombinant DNA molecule which contains a sequence encoding a papillomavirus LI protein or a papillomavims L2 protein or a combination of L I and 1.2 proteins and transfecting a host cell with the recombinant DNA molecule. Preferably, the virus like particles may express papillomavirus L I protein or L2 protein or a combination of LI and L2 proteins in the host cell. Thereafter, the papillomavirus virus-like particles obtained from the transfected host cell may be purified which will cause the disassembling the LI and L2 capsid proteins of the virus-like particles into smaller units. Preferably, it is these smaller disassembled LI and L2 capsid proteins which may be loaded with a DHT inhibitor. Next, the loaded proteins may be reassembled to form a loaded virus-like particles comprising HPV protein with the DHT inhibitor and administered to the skin of an animal or a human subject. The method can be applied to enhance topical deliver}' of Minoxidil , Finasteride or other drugs to treat AGA topically through the skin and through the hair follicle.
[00150], With reference now to FIG. 6, a method in accordance with an embodiment of the present invention, a preferred method of treating al opecia, will now be disc ussed. As shown in FIG. 6, the present invention provides a method for treating alopecia 1200, which includes a first step in which a recombinant DNA molecule is contracted which contains a sequence for encoding a papillomavims LI protein or a papillomavirus 1.2 protein, or a combination of papillomavirus L I and L2 proteins 1220. Thereafter, a host cell will be transfected with the recombinant DNA molecule 1230. Afterwhich, the transfected host cell will be treated to purify the papillomavirus vims-like particles causing the LI and L2 capsid proteins to disassemble into smaller units 1240, At which time, an appropriate therapeutic agent or drag for treating AGA will be introduced into the proximity of the virus-like particle where the agent or drag for treatment will be loaded into the virus-like particles 1250. Thereafter, the loaded virus-like particles comprising HPV protein with an effective DHT inhibitor for treating alopecia may be reassembled 1260. Finally, the treatment may preferably be topically applied through the skin and the hair follicles 1270 for the treatment of alopecia ,
[00151]. As further shown in FIG. 6 in accordance with another preferred embodiment of the present invention, the above method further includes a step to pre-treat the area of the skin. In step 1280, the area of the scalp to be treated for alopecia may preferably be prepared by pre-administering a delipidating substance to the region of the scalp before applying the treatment of HPV pseudo-virus particles loaded with a DHT inhibitor to the scalp of a human subject,
[00152], As further shown in FIG, 6, in accordance with another preferred embodiment of the present invention, the above method also further includes another pre -treatment of the scalp. In step 1290, the area of the scalp to be treated for alopecia may be pre -treated by micro-abrasion before applying the treatment of HPV pseudo-virus particles loaded with a DHT inhibitor to the scalp of a human subject
[00153], The current invention can also be applied for hair removal. In one embodiment of the invention, a siRNA (short interfering nucleic acid) molecule may be formulated as a composition as described in U.S. Patent Application Publication No. 200770179104 A l (which is hereby incorporated by reference herein). In this embodiment, the formulated composition which may be loaded into the betaHPV pseudovirions and transdermally administered may include such siRNA formulations which are generally referred to as "lipid nucleic acid particles" (LNP) which include membrane disruptive agents in which the siRNA molecule is complexed with a cationic lipid and helper lipid molecule; or formulated with polyethylenimine derivatives, including for example grated PEIs such as galactose PEI, cholesterol PEI, antibody derivatized PEL and polyethylene glycol PEI (PEG-PEI) and/or derivatives thereof,
[00154], EXAMPLE 6
[00155], In accordance with a first preferred embodiment of the present invention, a method for topically treating non-melanoma skin cancer using a combination of betapapillomavirus viral shells (L.1/L2) to deliver a siRNA against the transcription factor protein for GH2 is provided. According to alternative embodiments, other HPV viral shells may also be used. For instance, according to a further preferred embodiment, HPV5 viral shells may be used. Two sequence listings for preferred embodiments of H VPS viral shells are provided as SEQ.ID.NO. 7 and SEQ.ID.NO. 8. Alternatively, a herpes viral shell may also be use.
[00156], A first step in this preferred emboidement includes constructing a recombinant DNA molecule that contains a sequence encoding a papillomavirus LI protein or a papillomavirus L2 protein or a combination of LI and L2 proteins and then transfecting a host cell with the recombinant DNA molecule. Preferably, the virus like particles may express papillomavirus LI protein or L2 protein or a combination of L I and L2 proteins in the host cell. Next, the betapapillomavirus virus-like particles obtained from the transfected host cell may be purified which will cause the disassembling of the LI and L2 capsid proteins of the vims-like particles into smaller units. Preferably, it is these smaller disassembled L I and L2 capsid proteins which may be combmed with a siRNA against the transcription factor protein for GH2. Next the combination of siRNA with proteins may be reassembled to form loaded vims-like particles comprising HPV protein with the siRNA against the transcription factor protein for GU2 and administered to the skin of an animal or a human subject.
[00157]. With reference nowr to FIG. 7, a method in accordance with an embodiment of the present invention will now be discussed. As shown in FIG. 7, the present invention provides a method for treating non-melanoma skin cancer 800, which includes a first step in which a recombinant DNA molecule is contracted which contains a sequence for encoding a papillomavirus LI protein or a papillomavirus L2 protein, or a combination of papillomavirus LI and L2 proteins 820. Thereafter, a host cell will be transfected with the recombinant DNA molecule 830. After which, the transfected host ceil will be treated to purify the LI and L2 capsid proteins into smaller units 840. At which time, an appropriate therapeutic agent or drug for treating non-melanoma skin cancer will be introduced into the proximity of the proteins where the agent or drug for treatment will be combined with the Nanosphere particle 850. Thereafter, the particles comprisingsiRNA against the transcription factor protein for GH2 may be reassembled 860, Finally, the treatment may preferably be topically applied through the skin 870 for the treatment of non-melanoma skin cancer.
[00158]. In some embodiments, when the LI and L2 capsid protems disassemble into smaller units as described in FIG. 7, 840, these smaller units will include intermediate structure and capsomers. Preferably, these smaller units of capsid proteins, intermediate structures and capsomers are purified and then combined with a siRNA targeting transciption factor Gli2. Thereafter, reassembly of the capsid proteins is initiated to form loaded virus-like particles which preferably include PV proteins with attached siRNA targeting transcription factor GH2.
[00159]. EXAMPLE 7
[00160]. In accordance with a first preferred embodiment of the present invention, a method for topically treating Pachyonychia Congenita using a combination of betapapillomavirus viral shells (L1/L2) to deliver a siRNA targeting the N17K1 mutation in the keratin 6a gene is provided, According to further preferred embodiments, the siRNA of present invention may alternatively target known mutations in the genes encoding K6b, K16 and/or K17A.
Examples of sequences which can be used to inhibit the K6a, K6b, K.16 and/or K.17A are discussed in U.S. Patent No. 7,723,314 to jasper (including sequence listings) which is hereby incorporated by reference herein.
[00161], With reference now to FIG. 8, a method in accordance with an embodiment of the present invention will no be discussed. As shown in FIG. 8, the present invention provides a method for treating Pachyonychia Congenita 900, which includes a first step in which a recombinant DNA molecule is contracted which contains a sequence for encoding a papillomavirus LI protein or a papillomavirus L2 protein, or a combination of papillomavirus LI and L2 proteins 920. Thereafter, a host cell will be transiected with the recombinant DNA molecule 930. After which, the transiected host cell will be treated to purify the papillomavirus virus-like particles causing the LI and L2 capsid proteins to disassemble into smaller units 940. At which time, an appropriate therapeutic agent or drug for treating Pachyonychia Congenita will be introduced into the proximity of the virus-like particle where the agent or drug for treatment will be loaded into the viras-like particles 950. Thereafter, the loaded vims-like particles enclosing siRNA targeting the N17K1 mutation in keratin 6a may be reassembled 960. Finally, the treatment may preferably be topically applied through the skin 970 for the treatment of Pachyonychia Congenita.
[00162]. Table 1 shows a list of additional genetic skin related disorders which may be considered treatable in accordance with the present invention:
[00163]. Table 1
Acro-dermato-ungual-lacrimal-tooth syndrome (ADULT syndrome)
Ankyloblepharon-ectodermal defects-cleft lip and palate syndrome (AEC syndrome) Arthrogryposis and ectodermal dysplasia
Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy syndrome (APECED) Basan syndrome
Bullous congenital ichthyosiform erythroderma
Cartilage-hair hypoplasia syndrome
Chanarin-Dorfman syndrome
CHILD
Cleft lip/palate-ectodermal dysplasia syndrome
Clouston syndrome
Cockayne syndrome Congenital hypotrichosis with juvenile macular dystrophy
Congenital insensitivity to pain with anhidrosis
Corneodermatoosseous syndrome
Cranioectodermal syndrome
Cronkhite-Canada syndrome
Curly hair-ankyloblepharon-nail dysplasia (CHA.NDS)
Cutis lax a, hereditary
Darier disease
Dyskeratosis congenita
Ectodermal dysplasia, Margarita Island type
Ectodermal dysplasia, pure hair and nail type
Ectodennal dysplasia, skin fragility syndrome (McGrath syndrome)
Ectodermal dysplasia, with ectrodactyly and macular dystrophy
Ectrodactyly-ectodermal dysplasia-cleft lip/palate syndrome (EEC syndrome)
Ehlers-Danlos syndrome, arthrochalasis type
Ehlers-Danlos syndrome, classic type
Ehlers-Danlos syndrome, dermatosparaxis type
Ehlers-Danlos syndrome, hypermobility type
Ehlers-Danlos syndrome, kyphoscoliotic type
Ehlers-Danlos syndrome, unclassified variants
Ehlers-Danlos syndrome, vascular type
Ellis-va Creveld syndrome
Epidermolysis bullosa dystrophic, dominant
Epidermolysis bullosa dystrophic, Hallopeau-Siemens
Epidermolysis bullosa dystrophic, non-Hallopeau-Siemens
Epidermolysis bullosa dystrophic, pretibialis & pruriginosa
Epidermolysis bullosa junctional, Herlitz
Epidermolysis bullosa junctional, non~Heriitz
Epidermolysis bullosa junctional, with pyloric atresia
Epidermolysis bullosa simplex, Dowlmg-Meara
Epidermolysis bullosa simplex, Koebner
Epidermolysis bullosa simplex, Weber-Cockayne
Epidermolysis bullosa simplex, with mottled pigmentation
Epidermolysis bullosa simplex, with muscular dystrophy
Erythrokeratodermia variabilis
Focal dermal hypoplasia syndrome
Growth retardation-alopecia-pseudoanodontia-optic atrophy (GAPO syndrome) Hailey-Hailey disease
Hailerman-Streiff syndrome
Harlequin type ichthyosis congenita
Heimler syndrome Hypohidrotic ectodermal dysplasia
Hypohidrotic ectodermal dysplasia with hypothyroidism and agenesis of the corpus callosum
Hypohidrotic ectodermal dysplasia, with hypothyroidism and ciliary dyskinesia
Hypohidrotic ectodermal dysplasia, with immune deficiency
Hypohidrotic ectodermal dysplasia, with immune deficiency, osteopetrosis and lymphoedema
Ichthyosis of Siemens
Incontinentia pigmenti
Johanson-Blizzard syndrome
Johnson neuroectodermal syndrome
Keratitis-ichthyosis-deafness syndrome
Kindler syndrome
Lamellar iehthyosis/Non-bullous congenital ichthyosiform erythroderma
Limb-mammary syndrome
Lipoid proteinosis
Marshall syndrome
Myoepithelial dysplasia, hereditary'
Mutilating Vohwinkel palmo-plantar keratoderma without deafness
Mutilating Vohwinkel palmo-plantar keratoderma with deafness
Naegeli syndrome
Netherton syndrome
Oculodentodigital displasia (ODDD)
Odontoonychodermal dysplasia
Odontotrichomelic syndrome
Onychotrichodyspiasia and neutropenia
Orofaciodigital syndrome, type I (OFDS1)
Pachyonychia congenita type 1
Pachyonychia congenita type 2
Pseudoxanthoma Elasti cum
Rapp-Hodgkin syndrome
Refsum disease
Rothmund-Thomson syndrome
Sabinas brittle hair and mental deficiency syndrome
Scalp-ear-nipple syndrome
Schopf-Schulz-Passarge syndrome
Sjogren Larsson syndrome
Taurodontia, absent teeth and sparse hair
Trichodental dysplasia
Trichodentoosseous syndrome
Trichothiodystrophy Ulnar mammary syndrome
Weyer acrofacial dysostosis
Witkop syndrome
Xeroderma pigmentosum
X-linked recessive ichthyosis
[00164]. EXAMPLE 8
[00165]. In accordance with a first preferred embodiment of the present invention, a method for topically treating Psoriasis using a combination of betapapillomavirus viral shells (L1/L2) to deliver a siRNA against cytokines is provided.
[00166]. A first step in this preferred embodiment includes constructing a recombinant DNA molecule that contains a sequence encoding a papillomavirus LI protein or a papillomavirus L2 protein or a combination of LI and L2 proteins and then transfecting a host cell with the recombinant DNA molecule. Preferably, the virus like particles may express papillomavirus LI protein or L2 protein or a combination of LI and L2 proteins in the host ceil. Next, the betapapillomavirus virus-like particles obtained from the transfected host cell may be purified which will cause the disassembling of the LI and L2 capsid proteins of the viras-like particles into smaller units. Preferably, it is these smaller disassembled LI and 1.2 capsid proteins which may be loaded with a si R NA against cytokines. Next the loaded protems may be reassembled to form loaded virus-like particles comprising HPV protein with the siRNA against cytokines and administered to the skin of an animal or a human subject.
[00167], With reference now to FIG. 9, a method in accordance with an embodiment of the present invention will now be discussed. As shown in FIG . 9, the present invention provides a method for treating Psoriasis 1 100, which includes a first step in which a recombinant DNA molecule is contracted which contains a sequence for encoding a papillomavirus LI protein or a papillomavirus L2 protein, or a combination of papillomavirus LI and L2 proteins 1 120. Thereafter, a host cell will be transfected with the recombinant DN A molecule 1 130. After which, the transfected host cell will be treated to purify the papillomavirus virus-like particles causing the LI and L2 capsid proteins to disassemble into smaller units 1 140. At which time, an appropriate therapeutic agent or drag for treating Psoriasis will be introduced into the proximity of the virus-like particle where die agent or drug for treatment will be loaded into the virus-like particles 1 150. Thereafter, the loaded virus-like particles enclosing siRNA against cytokines may be reassembled 1160. Finally, the treatment may preferably be topically applied through the skin 1170 for the treatment of psoriasis.
[00168], Preferably, according to one aspect of the present invention, the RNA which inhibits the expression of cytokines is an siRNA which inhibits the expression of TNF-a for the treatment of psoriasis.
[00169], While the above descriptions regarding the present invention contains much specificity, these should not be construed as limitations on the scope, but rather as examples. Many other variations are possible. Accordingly, the scope should be determined not by the embodiments illustrated, but by the appended claims and their legal equivalents. For example, alternati ve viral vectors may be used in place of the betapapillomavirus. For example, alternative viral vector may include herpes virus vectors.

Claims

The claims defining the invention are as follows:
1. A composition for transdermal drug delivery comprising a HPV protein and a therapeutic agent to treat a skin related disease,
2. The composition of claim 1, wherein the therapeutic agent is a small molecule.
3. The composition of claim 2, wherein the therapeutic agent is a nucleic acid.
4. The composition of claim 3, wherein the therapeutic agent is comprised of siRNA.
5. The composition of claim 3, wherein the therapeutic agent is comprised of a DHT inhibitor.
6. The composition of claim 5, wherein the DHT inhibitor is NAi.
7. The composition of claim 2, wherein the therapeutic agent inhibits cell proliferation.
8. The composition of claim 3, wherein the therapeutic agent inhibits the Hedgehog Pathway.
9. The composition of claim 8, wherein the nucleic acid comprises siRN A targeting the transcription factor Gii2.
10. The composition of claim 4, wherein the siRNA targets the NT7 1 mutation in keratin 6a.
11. The composition of claim 3, wherein the therapeutic agent is comprised of a DNA plasmid, wherein the DNA plasmid further comprises a sequence of NER enzymes,
12. The composition of claim 11, wherein the NER enzymes are SPC, SPA, ERCC2, ERCC3, or POCH.
13. The composition of claim 12, wherein the enzyme is Bacteriophage T4 endonuclease V (T4N5).
14. The composition of claim 4, wherein the therapeutic agent comprises siRNA which inhibits the expression of TNF-a,
15. A method for treating a skin related disease using a combination of betapapillomavirus viral shells (L1/L2) to deliver a therapeutic agent, the method comprising essentially the steps of: constructing a recombinant DN A molecule that contains a sequence encoding a papillomavirus LI protein or a papillomavirus L2 protein or a combination of LI and L2 proteins; transfectmg a host cell with the recombinant DNA molecule; expressing papillomavirus LI protein or L2 protein or a combination of L I and L2 proteins in the host cell; obtaining the papillomavirus virus-like particles from the transfected host cell; purifiying the vims-like particles; disassembling the LI and L2 capsid proteins of the vims-like particles into smaller units; loading said disassembled L I and L2 capsid proteins with the therapeutic agent; reassembling the loaded proteins to form a loaded virus-like Particles comprising HPV protein with the therapeutic agent; and administering said loaded vims-like paxticles to the skin of an animal or a human subject.
PCT/US2012/045989 2011-07-10 2012-07-09 Virion derived protein nanoparticles for delivering diagnostic or therapeutic agents for the treatment of skin-related diseases WO2013009717A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201161506140P 2011-07-10 2011-07-10
US61/506,140 2011-07-10
US201113221803A 2011-08-30 2011-08-30
US13/221,803 2011-08-30
US201113253028A 2011-10-04 2011-10-04
US13/253,028 2011-10-04

Publications (1)

Publication Number Publication Date
WO2013009717A1 true WO2013009717A1 (en) 2013-01-17

Family

ID=47506445

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/045989 WO2013009717A1 (en) 2011-07-10 2012-07-09 Virion derived protein nanoparticles for delivering diagnostic or therapeutic agents for the treatment of skin-related diseases

Country Status (1)

Country Link
WO (1) WO2013009717A1 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
US10588984B2 (en) 2013-09-18 2020-03-17 Aura Biosciences, Inc. Virus-like particle conjugates for diagnosis and treatment of tumors
US10596275B2 (en) * 2012-02-07 2020-03-24 Aura Biosciences, Inc. Virion-derived nanospheres for selective delivery of therapeutic and diagnostic agents to cancer cells
US10688172B2 (en) 2009-04-13 2020-06-23 INSERM (Institut National de la Santé et de la Recherche Médicale) HPV particles and uses thereof
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5302389A (en) * 1992-08-17 1994-04-12 Board Of Regents, The University Of Texas System Method for treating UV-induced suppression of contact hypersensitivity by administration of T4 endonuclease
US20040071666A1 (en) * 2002-05-24 2004-04-15 The Regents Of The University Of Michigan Compositions and methods related to graft-versus-host disease
US20070141009A1 (en) * 2003-01-03 2007-06-21 Shaharyar Khan Sirna mediated post-transriptional gene silencing of genes involved in alopecia
US20090181908A1 (en) * 2006-12-29 2009-07-16 Roger Louis Kaspar Methods and compositions for treating keratin hyperproliferative disorders
WO2010120266A1 (en) * 2009-04-13 2010-10-21 Inserm, Institut National De La Sante Et De La Recherche Medicale Hpv particles and uses thereof
US20110039909A1 (en) * 2008-10-15 2011-02-17 Wisconsin Alumni Research Foundation Methods and materials for reducing gli2 expression

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5302389A (en) * 1992-08-17 1994-04-12 Board Of Regents, The University Of Texas System Method for treating UV-induced suppression of contact hypersensitivity by administration of T4 endonuclease
US20040071666A1 (en) * 2002-05-24 2004-04-15 The Regents Of The University Of Michigan Compositions and methods related to graft-versus-host disease
US20070141009A1 (en) * 2003-01-03 2007-06-21 Shaharyar Khan Sirna mediated post-transriptional gene silencing of genes involved in alopecia
US20090181908A1 (en) * 2006-12-29 2009-07-16 Roger Louis Kaspar Methods and compositions for treating keratin hyperproliferative disorders
US20110039909A1 (en) * 2008-10-15 2011-02-17 Wisconsin Alumni Research Foundation Methods and materials for reducing gli2 expression
WO2010120266A1 (en) * 2009-04-13 2010-10-21 Inserm, Institut National De La Sante Et De La Recherche Medicale Hpv particles and uses thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
AHMAD ET AL.: "Mislocalization of XPF-ERCC1 nuclease contributes to reduced DNA repair in XP -F patients.", PLOS. GENET., vol. 6, no. 3, 5 March 2010 (2010-03-05), pages 1 - 11 *
DATABASE GENBANK 5 April 2007 (2007-04-05), STRAUSBERG ET AL.: "Keratin 6A [Homo sapiens].", retrieved from http://www.ncbi.nlm.nih.gov/protein/AAI39754.1 accession no. A139754. *

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10688172B2 (en) 2009-04-13 2020-06-23 INSERM (Institut National de la Santé et de la Recherche Médicale) HPV particles and uses thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8680069B2 (en) 2011-12-16 2014-03-25 Moderna Therapeutics, Inc. Modified polynucleotides for the production of G-CSF
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US10596275B2 (en) * 2012-02-07 2020-03-24 Aura Biosciences, Inc. Virion-derived nanospheres for selective delivery of therapeutic and diagnostic agents to cancer cells
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US10588984B2 (en) 2013-09-18 2020-03-17 Aura Biosciences, Inc. Virus-like particle conjugates for diagnosis and treatment of tumors
US11110181B2 (en) 2013-09-18 2021-09-07 Aura Biosciences, Inc. Virus-like particle conjugates for diagnosis and treatment of tumors
US11806406B2 (en) 2013-09-18 2023-11-07 Aura Biosciences, Inc. Virus-like particle conjugates for diagnosis and treatment of tumors
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
WO2015051214A1 (en) 2013-10-03 2015-04-09 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof

Similar Documents

Publication Publication Date Title
WO2013009717A1 (en) Virion derived protein nanoparticles for delivering diagnostic or therapeutic agents for the treatment of skin-related diseases
US20130012566A1 (en) Virion Derived Protein Nanoparticles For Delivering Diagnostic Or Therapeutic Agents For The Treatment of Alopecia
US10596275B2 (en) Virion-derived nanospheres for selective delivery of therapeutic and diagnostic agents to cancer cells
US10688172B2 (en) HPV particles and uses thereof
JP7330590B2 (en) Nucleic acid vaccine for varicella zoster virus (VZV)
US20230114180A1 (en) Respiratory syncytial virus vaccine
US20200289638A1 (en) Zika virus rna vaccines
KR20180096592A (en) Respiratory syncytial virus vaccine
US20110052496A1 (en) Hollow nanoparticles and uses thereof
WO2018170245A1 (en) Broad spectrum influenza virus vaccine
KR20180096591A (en) A broad-spectrum influenza virus vaccine
JP2018531996A (en) Human cytomegalovirus vaccine
WO2017015463A2 (en) Infectious disease vaccines
CN117247954A (en) Circular RNA vaccine, novel circular RNA and preparation method thereof

Legal Events

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

Ref document number: 12810964

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205 DATED 22-05-2014)

122 Ep: pct application non-entry in european phase

Ref document number: 12810964

Country of ref document: EP

Kind code of ref document: A1